Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PHTHALAZINONE COMPOUNDS AS PARP7 INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2023/192989
Kind Code:
A2
Abstract:
The present disclosure relates to phthalazinone compounds and related compounds and their use in treating a disease or condition responsive to inhibition of PARP7.

Inventors:
JOHNSON MICHAEL G (US)
SPELLMEYER DAVID C (US)
NG RAYMOND A (US)
LAPOINTE DAVID (US)
DENG JINXIA N (US)
COHEN MICHAEL S (US)
RODRIGUEZ KELSIE M (US)
SUNDALAM SUNIL K (US)
SANDERSON DANIEL J (US)
PELLETIER GUILLAUME (CA)
WINTER DANA K (CA)
NOVOSELTSEVA POLINA (CA)
ZHOU YUCHEN (CA)
Application Number:
PCT/US2023/065214
Publication Date:
October 05, 2023
Filing Date:
March 31, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
AZKARRA THERAPEUTICS INC (US)
UNIV OREGON HEALTH & SCIENCE (US)
International Classes:
C07D487/02; C07D237/32
Attorney, Agent or Firm:
YIN SOWATZKE, Tina et al. (US)
Download PDF:
Claims:
CLAIMS 1. A compound, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is of Formula I: wherein: X1 is -N- or -CR1a2-; X2 is -N- or -CR1a4-; R1a1, R1a2, R1a3, and R1a4 are independently H, halo, -OH, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1-6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO2-cycloalkyl, -NR1L1R1L2, or C2-6alkynyl-NR1L3R1L4; wherein if X1 is -CR1a2- and X2 is -CR1a4-, then at least one of R1a1, R1a2, R1a3, and R1a4 is not H; wherein each C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1- 6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO2- cycloalkyl of R1a1, R1a2, R1a3, and R1a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2; R1L1, R1L2, R1L3, and R1L4 are independently H, C1-6alkyl, C2-6alkynyl, or cycloalkyl, wherein each C1-6alkyl, C2-6alkynyl, or cycloalkyl of R1L1, R1L2, R1L3, and R1L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1-6alkyl, C1-6haloalkyl, cycloalkyl, and halocycloalkyl; X3 is -O-, -NR1b1-, or -CR1b2R1b3-, wherein R1b1, R1b2, and R1b3 are independently H or C1-6alkyl; bond marked 2B is to D3, D4, D5, or D6; R3f1 is H or C1-6alkyl; X4 is -O-, -NR3f2-, or -CR3f3R3f4-, wherein R3f2, R3f3, and R3f4 are independently selected from H and C1-6alkyl; R1c1, R1c2, R1c3, R1c4, R1c5, R1c6, R1c7, and R1c8 are independently H, halo, -CN, -SO2CH3, -SO2NH2, wherein a bond marked 2B is to D3, D4, D5, or D6; X5 is -CH(R7g3)- or -CH(R7g4)CH2N(R7g5)-; R5g1, R7g2, R7g3, R7g4, and R7g5 are independently H or C1-6alkyl; B3, B4, B5, and B6 are independently a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18- membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B3, B4, B5, and B6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1- 6alkyl, and oxo; provided that B4 is not D3, D4, D5, D6, and D7 are independently C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, - O-aryl, -O-heteroaryl, -C(O)-C1-6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)- heteroaryl, -N(R1D1)(R1D2), -C(O)N(R1D3)(R1D4), or -N(R1D5)C(O)R1D6; wherein the C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1- 6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D3, D4, D5, D6, and D7 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, - CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, C1-6alkyl-OH, -O-C1-6haloalkyl, C1-6alkyl-O- C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R1D10)2, wherein each R1D10 is independently H or C1-6alkyl; R1D1, R1D3, and R1D5 are independently H or C1-6alkyl; R1D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R1D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R1D11)2, wherein each R1D11 is independently H or C1-6alkyl; and R1D4 and R1D6 are independently C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R1D4 and R1D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1- 6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R1D12)2, wherein each R1D12 is independently H or C1-6alkyl; provided that 1a2 , then R is not F, if X3 is -O-, B4 is and D4 is -C(O)-aryl, th 1a4 en R is not Cl, and if D4 is and R1c2 is F, then R1a2 is not F. 2. The compound of claim 1, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is of Formula II: wherein: X1 is -N- or -CR2a2-; X2 is -N- or -CR2a4-; R2a1, R2a2, R2a3, and R2a4 are independently H, halo, -OH, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1-6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO2-cycloalkyl, -NR2L1R2L2, or C2-6alkynyl-NR2L3R2L4; wherein if X1 is -CR2a2- and X2 is -CR2a4-, then at least one of R2a1, R2a2, R2a3, and R2a4 is not H; wherein each C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1- 6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO2- cycloalkyl of R2a1, R2a2, R2a3, and R2a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2; R2L1, R2L2, R2L3, and R2L4 are independently H, C1-6alkyl, C2-6alkynyl, or cycloalkyl, wherein each C1- 6alkyl, C2-6alkynyl, or cycloalkyl of R2L1, R2L2, R2L3, and R2L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1-6alkyl, C1-6haloalkyl, cycloalkyl, and halocycloalkyl; X3 is -O-, -NR2b1-, or -CR2b2R2b3-, wherein R2b1, R2b2, and R2b3 are independently H or C1-6alkyl; bond marked 2B is to D4, D5, or D6; X5 is -CH(R7g3)- or -CH(R7g4)CH2N(R7g5)-; R5g1, R7g2, R7g3, R7g4, and R7g5 are independently H or C1-6alkyl; B4, B5, and B6 are independently a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18- membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B4, B5, and B6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1- 6alkyl, and oxo; provided that B4 is not D4, D5, D6, and D7 are independently C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O- aryl, -O-heteroaryl, -C(O)-C1-6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R2D1)(R2D2), -C(O)N(R2D3)(R2D4), or -N(R2D5)C(O)R2D6; wherein the C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1- 6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D4, D5, D6, and D7 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, C1-6alkyl-OH, -O-C1-6haloalkyl, C1-6alkyl-O-C1- 6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R2D10)2, wherein each R2D10 is independently H or C1-6alkyl; R2D1, R2D3, and R2D5 are independently H or C1-6alkyl; R2D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R2D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R2D11)2, wherein each R2D11 is independently H or C1-6alkyl; and R2D4 and R2D6 are independently C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R2D4 and R2D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1- 6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R2D12)2, wherein each R2D12 is independently H or C1-6alkyl; provided that if B4 is and D4 is -CHF 2a2 2, -CF2CH3, or -CF2CF3, then R is not F, if X3 is -O-, B4 is and D4 i 2a4 s -C(O)-aryl, then R is not Cl, and if D4 is and R2c2 is F, then R2a2 is not F. 3. The compound of claim 1, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is of Formula III: wherein: X1 is -N- or -CR3a2-; X2 is -N- or -CR3a4-; R3a1, R3a2, R3a3, and R3a4 are independently H, halo, -OH, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1-6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO2-cycloalkyl, -NR3L1R3L2, or C2-6alkynyl-NR3L3R3L4 ; wherein if X1 is -CR3a2- and X2 is -CR3a4-, then at least one of R3a1, R3a2, R3a3, and R3a4 is not H; wherein each C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1- 6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO2- cycloalkyl of R3a1, R3a2, R3a3, and R3a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2; R3L1, R3L2, R3L3, and R3L4 are independently H, C1-6alkyl, C2-6alkynyl, or cycloalkyl, wherein each C1-6alkyl, C2-6alkynyl, or cycloalkyl of R3L1, R3L2, R3L3, and R3L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1-6alkyl, C1-6haloalkyl, cycloalkyl, and halocycloalkyl; X3 is -O-, -NR3b1-, or -CR3b2R3b3-, wherein R3b1, R3b2, and R3b3 are independently H or C1-6alkyl; R3f1 is H or C1-6alkyl; X4 is -O-, -NR3f2-, or -CR3f3R3f4-, wherein R3f2, R3f3, and R3f4 are independently selected from H and C1-6alkyl; B3 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B3 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C1-6alkyl, and oxo; D3 is C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1-6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R3D1)(R3D2), -C(O)N(R3D3)(R3D4), or -N(R3D5)C(O)R3D6; wherein the C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1- 6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D3 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1- 6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, C1-6alkyl-OH, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl- O-C1-6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R3D10)2, wherein each R3D10 is independently H or C1- 6alkyl; R3D1, R3D3, and R3D5 are independently H or C1-6alkyl; R3D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R3D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R3D11)2, wherein each R3D11 is independently H or C1-6alkyl; and R3D4 and R3D6 are independently C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R3D4 and R3D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1- 6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R3D12)2, wherein each R3D12 is independently H or C1-6alkyl. 4. The compound of claim 1 or 2, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is of Formula IV: wherein: X1 is -N- or -CR4a2-; X2 is -N- or -CR4a4-; R4a1, R4a2, R4a3, and R4a4 are independently H, halo, -OH, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1-6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO2-cycloalkyl, -NR4L1R4L2, or C2-6alkynyl-NR4L3R4L4 ; wherein if X1 is -CR4a2- and X2 is -CR4a4-, then at least one of R4a1, R4a2, R4a3, and R4a4 is not H; wherein each C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1- 6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO2- cycloalkyl of R4a1, R4a2, R4a3, and R4a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2; R4L1, R4L2, R4L3, and R4L4 are independently H, C1-6alkyl, C2-6alkynyl, or cycloalkyl, wherein each C1-6alkyl, C2-6alkynyl, or cycloalkyl of R4L1, R4L2, R4L3, and R4L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1-6alkyl, C1-6haloalkyl, cycloalkyl, and halocycloalkyl; X3 is -O-, -NR4b1-, or -CR4b2R4b3-, wherein R4b1, R4b2, and R4b3 are independently H or C1-6alkyl; alo, -CN, -SO2CH3, -SO2NH2, or -NHSO2CH3; B4 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B4 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C1-6alkyl, and oxo; provided that B4 is not D4 is C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1-6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R4D1)(R4D2), -C(O)N(R4D3)(R4D4), or -N(R4D5)C(O)R4D6; wherein the C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1- 6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D4 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1- 6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, C1-6alkyl-OH, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl- O-C1-6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R4D10)2, wherein each R4D10 is independently H or C1- 6alkyl; R4D1, R4D3, and R4D5 are independently H or C1-6alkyl; R4D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R4D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R4D11)2, wherein each R4D11 is independently H or C1-6alkyl; and R4D4 and R4D6 are independently C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R4D4 and R4D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1- 6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R4D12)2, wherein each R4D12 is independently H or C1-6alkyl; provided that , then R4a2 is not F, if X3 is -O-, B4 is and D4 is -C(O)-aryl, then R4a4 is not Cl, and if D4 is and R4c2 is F, then R4a2 is not F. 5. The compound of claim 1 or 2, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is of Formula V: wherein: X1 is -N- or -CR5a2-; X2 is -N- or -CR5a4-; R5a1, R5a2, R5a3, and R5a4 are independently H, halo, -OH, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1-6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO2-cycloalkyl, -NR5L1R5L2, or C2-6alkynyl-NR5L3R5L4 ; wherein if X1 is -CR5a2- and X2 is -CR5a4-, then at least one of R5a1, R5a2, R5a3, and R5a4 is not H; wherein each C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1- 6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO2- cycloalkyl of R5a1, R5a2, R5a3, and R5a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2; R5L1, R5L2, R5L3, and R5L4 are independently H, C1-6alkyl, C2-6alkynyl, or cycloalkyl, wherein each C1-6alkyl, C2-6alkynyl, or cycloalkyl of R5L1, R5L2, R5L3, and R5L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1-6alkyl, C1-6haloalkyl, cycloalkyl, and halocycloalkyl; X3 is -O-, -NR5b1-, or -CR5b2R5b3-, wherein R5b1, R5b2, and R5b3 are independently H or C1-6alkyl; A5 is

R5g1 is H or C1-6alkyl; B5 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B5 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C1-6alkyl, and oxo; D5 is C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1-6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R5D1)(R5D2), -C(O)N(R5D3)(R5D4), or -N(R5D5)C(O)R5D6; wherein the C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1- 6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D5 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1- 6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, C1-6alkyl-OH, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl- O-C1-6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R5D10)2, wherein each R5D10 is independently H or C1- 6alkyl; R5D1, R5D3, and R5D5 are independently H or C1-6alkyl; R5D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R5D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R5D11)2, wherein each R5D11 is independently H or C1-6alkyl; and R5D4 and R5D6 are independently C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R5D4 and R5D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1- 6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R5D12)2, wherein each R5D12 is independently H or C1-6alkyl. 6. The compound of claim 1 or 2, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is of Formula VI:

X1 is -N- or -CR6a2-; X2 is -N- or -CR6a4-; R6a1, R6a2, R6a3, and R6a4 are independently H, halo, -OH, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1-6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO2-cycloalkyl, -NR6L1R6L2, or C2-6alkynyl-NR6L3R6L4 ; wherein if X1 is -CR6a2- and X2 is -CR6a4-, then at least one of R6a1, R6a2, R6a3, and R6a4 is not H; wherein each C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1- 6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO2- cycloalkyl of R6a1, R6a2, R6a3, and R6a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2; R6L1, R6L2, R6L3, and R6L4 are independently H, C1-6alkyl, C2-6alkynyl, or cycloalkyl, wherein each C1-6alkyl, C2-6alkynyl, or cycloalkyl of R6L1, R6L2, R6L3, and R6L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1-6alkyl, C1-6haloalkyl, cycloalkyl, and halocycloalkyl; X3 is -O-, -NR6b1-, or -CR6b2R6b3-, wherein R6b1, R6b2, and R6b3 are independently H or C1-6alkyl; o B6 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B6 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C1-6alkyl, and oxo; D6 is C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1-6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R6D1)(R6D2), -C(O)N(R6D3)(R6D4), or -N(R6D5)C(O)R6D6; wherein the C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1- 6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D6 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1- 6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, C1-6alkyl-OH, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl- O-C1-6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R6D10)2, wherein each R6D10 is independently H or C1- 6alkyl; R6D1, R6D3, and R6D5 are independently H or C1-6alkyl; R6D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R6D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R6D11)2, wherein each R6D11 is independently H or C1-6alkyl; and R6D4 and R6D6 are independently C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R6D4 and R6D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1- 6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R6D12)2, wherein each R6D12 is independently H or C1-6alkyl. 7. The compound of claim 1 or 2, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is of Formula VII: X1 is -N- or -CR7a2-; X2 is -N- or -CR7a4-; R7a1, R7a2, R7a3, and R7a4 are independently H, halo, -OH, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1-6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO2-cycloalkyl, -NR7L1R7L2, or C2-6alkynyl-NR7L3R7L4; wherein if X1 is -CR7a2- and X2 is -CR7a4-, then at least one of R7a1, R7a2, R7a3, and R7a4 is not H; wherein each C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1- 6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO2- cycloalkyl of R7a1, R7a2, R7a3, and R7a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2; R7L1, R7L2, R7L3, and R7L4 are independently H, C1-6alkyl, C2-6alkynyl, or cycloalkyl, wherein each C1-6alkyl, C2-6alkynyl, or cycloalkyl of R7L1, R7L2, R7L3, and R7L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1-6alkyl, C1-6haloalkyl, cycloalkyl, and halocycloalkyl; X3 is -O-, -NR7b1-, or -CR7b2R7b3-, wherein R7b1, R7b2, and R7b3 are independently H or C1-6alkyl; 2NH2, or -NHSO2CH3; X5 is -CH(R7g3)- or -CH(R7g4)CH2N(R7g5)-; R7g2, R7g3, R7g4, and R7g5 are independently H or C1-6alkyl; D7 is C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1-6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R7D1)(R7D2), -C(O)N(R7D3)(R7D4), or -N(R7D5)C(O)R7D6; wherein the C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1- 6alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D7 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1- 6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, C1-6alkyl-OH, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl- O-C1-6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R7D10)2, wherein each R7D10 is independently H or C1- 6alkyl; R7D1, R7D3, and R7D5 are independently H or C1-6alkyl; R7D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R7D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R7D11)2, wherein each R7D11 is independently H or C1-6alkyl; and R7D4 and R7D6 are independently C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R7D4 and R7D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1- 6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R7D12)2, wherein each R7D12 is independently H or C1-6alkyl. 8. The compound of any one of claims 1-7, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein X1 is -CR1a2-, -CR2a2-, -CR3a2-, -CR4a2-, -CR5a2-, -CR6a2-, or -CR7a2-. 9. The compound of any one of claims 1-8, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein X2 is -CR1a4-, -CR2a4-, -CR3a4-, -CR4a4-, -CR5a4-, -CR6a4-, or -CR7a4-. 10. The compound of any one of claims 1, 2, 4, 8, and 9, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a2, R2a2, or R4a2 is H, Cl, Br, I, -OH, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1-6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO2-cycloalkyl, -NR1L1R1L2, C2-6alkynyl-NR1L3R1L4, -NR2L1R2L2, C2- 6alkynyl-NR2L3R2L4, -NR4L1R4L2, or C2-6alkynyl-NR4L3R4L4; wherein each C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1- 6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO2- cycloalkyl of R1a2, R2a2, or R4a2 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2. 11. The compound of any of claims 1, 2, 4, or 8-10, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a4, R2a4, or R4a4 is H, F, Br, I, -OH, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1-6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO2-cycloalkyl, -NR1L1R1L2, C2-6alkynyl-NR1L3R1L4, -NR2L1R2L2, C2- 6alkynyl-NR2L3R2L4, -NR4L1R4L2, or C2-6alkynyl-NR4L3R4L4; wherein each C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1- 6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO2- cycloalkyl of R1a4, R2a4, or R4a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2. 12. The compound of any one of claims 1-11, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein one, two, three, or four of R1a1, R1a2, R1a3, and R1a4, R2a1, R2a2, R2a3, and R2a4, R3a1, R3a2, R3a3, and R3a4, R4a1, R4a2, R4a3, and R4a4, R5a1, R5a2, R5a3, and R5a4, R6a1, R6a2, R6a3, and R6a4, or R7a1, R7a2, R7a3, and R7a4 is not H. 13. The compound of any one of claims 1-12, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a3, R2a3, R3a3, R4a3, R5a3, R6a3, or R7a3 is not H. 14. The compound of any one of claims 1-13, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein one, two, three, or four of R1a1, R1a2, R1a3, and R1a4, R2a1, R2a2, R2a3, and R2a4, R3a1, R3a2, R3a3, and R3a4, R4a1, R4a2, R4a3, and R4a4, R5a1, R5a2, R5a3, and R5a4, R6a1, R6a2, R6a3, and R6a4, or R7a1, R7a2, R7a3, and R7a4 is F or Cl. 15. The compound of any one of claims 1-14, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a1, R2a1, R3a1, R4a1, R5a1, R6a1, or R7a1 is F or Cl. 16. The compound of any one of claims 1-9 and 11-15, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a2, R2a2, R3a2, R4a2, R5a2, R6a2, or R7a2 is F or Cl. 17. The compound of any one of claims 1-16, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a3, R2a3, R3a3, R4a3, R5a3, R6a3, or R7a3 is F or Cl. 18. The compound of any one of claims 1-17, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein one, two, three, or four of R1a1, R1a2, R1a3, and R1a4, R2a1, R2a2, R2a3, and R2a4, R3a1, R3a2, R3a3, and R3a4, R4a1, R4a2, R4a3, and R4a4, R5a1, R5a2, R5a3, and R5a4, R6a1, R6a2, R6a3, and R6a4, or R7a1, R7a2, R7a3, and R7a4 is -OH. 19. The compound of any one of claims 1-16 and 18, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a3, R2a3, R3a3, R4a3, R5a3, R6a3, or R7a3 is -OH. 20. The compound of any one of claims 1-19, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein one, two, three, or four of R1a1, R1a2, R1a3, and R1a4, R2a1, R2a2, R2a3, and R2a4, R3a1, R3a2, R3a3, and R3a4, R4a1, R4a2, R4a3, and R4a4, R5a1, R5a2, R5a3, and R5a4, R6a1, R6a2, R6a3, and R6a4, or R7a1, R7a2, R7a3, and R7a4 is -O-C1-6alkyl or -O-cycloalkyl, wherein the -O-C1-6alkyl or -O-cycloalkyl is unsubstituted or substituted with halo or -CN. 21. The compound of any of claims 1-16, 18, or 20, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a3, R2a3, R3a3, R4a3, R5a3, R6a3, or R7a3 is -O-C1-6alkyl or -O-cycloalkyl, wherein the -O-C1-6alkyl or -O-cycloalkyl is unsubstituted or substituted with halo or -CN. 22. The compound of any one of claims 1-21, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein one, two, three, or four of R1a1, R1a2, R1a3, and R1a4, R2a1, R2a2, R2a3, and R2a4, R3a1, R3a2, R3a3, and R3a4, R4a1, R4a2, R4a3, and R4a4, R5a1, R5a2, R5a3, and R5a4, R6a1, R6a2, R6a3, and R6a4, or R7a1, R7a2, R7a3, and R7a4 is -O-C2-6alkynyl, wherein the -O-C2-6alkynyl is unsubstituted or substituted with halo. 23. The compound of any of claims 1-16, 18, 20, or 22, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a3, R2a3, R3a3, R4a3, R5a3, R6a3, or R7a3 is -O-C2-6alkynyl, wherein the -O- C2-6alkynyl is unsubstituted or substituted with halo. 24. The compound of any one of claims 1-23, or a stereoisomer or a pharmaceutically acceptable salt

1 1 1 2 1 1 4 2 1 2 2 2 2 4 1 2 25. The compound of any of claims 1-16, 18, 20, 22, or 24, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a3, R2a3, R3a3, R4a3, R5a3, R6a3, or R7a3 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O-heteroaryl is unsubstituted or substituted with halo. 26. The compound of any one of claims 1-25, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein two of R1a1, R1a2, R1a3, and R1a4, R2a1, R2a2, R2a3, and R2a4, R3a1, R3a2, R3a3, and R3a4, R4a1, R4a2, R4a3, and R4a4, R5a1, R5a2, R5a3, and R5a4, R6a1, R6a2, R6a3, and R6a4, or R7a1, R7a2, R7a3, and R7a4 is not H. 27. The compound of any one of claims 1-26, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a1, R2a1, R3a1, R4a1, R5a1, R6a1, or R7a1 is F or Cl and R1a2, R2a2, R3a2, R4a2, R5a2, R6a2, or R7a2 is F or Cl. 28. The compound of any of claims 1-16, 18, 20-22, 24, or 26 or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1a2, R2a2, R3a2, R4a2, R5a2, R6a2, or R7a2 is F or Cl and R1a3, R2a3, R3a3, R4a3, R5a3, R6a3, or R7a3 is -O-C1-6alkyl or -O-cycloalkyl, wherein the -O-C1-6alkyl or -O-cycloalkyl is unsubstituted or substituted with halo. 29. The compound of any one of claims 1-28, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein X3 is -O-. 30. The compound of any one of claims 1-28, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein X3 is -CH2-. 31. The compound of any of claims 1, 3, or 8-30, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein X4 is -O-. 32. The compound of any of claims 1, 3, or 8-30, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein X4 is -NR3f2-. 33. The compound of any of claims 1, 3, or 8-30, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein X4 is -CR3f3R3f4-. 34. The compound of any of claims 1, 2 or 4-30, or a stereoisomer or a pharmaceutically acceptable

35. The compound of any of claims 1, 2, or 4-34, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein one or more of X10, X11, and X12 is -N-. 36. The compound of any of claims 1, 2, 4-30, or 34, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein X10 is -CR1c1-, -CR2c1-, -CR4c1-, -CR5c1-, -CR6c1-, or -CR7c1-, X11 is - CR1c2-, -CR2c2-, -CR4c2-, -CR5c2-, -CR6c2-, or -CR7c2-, and X12 is -CR1c3-, -CR2c3-, -CR4c3-, -CR5c3-, -CR6c3-, or -CR7c3-. 37. The compound of any of claims 1, 2, 4-30, or 34-36, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein one, two, three, or four of R1c1, R1c2, R1c3, and R1c4, R2c1, R2c2, R2c3, and R2c4, R4c1, R4c2, R4c3, and R4c4, R5c1, R5c2, R5c3, and R5c4, R6c1, R6c2, R6c3, and R6c4, or R7c1, R7c2, R7c3, and R7c4 is not H. 38. The compound of any of claims 1, 2, 4-30, or 34-37 or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1c2, R2c2, R4c2, R5c2, R6c2, or R7c2 is F and R1c1, R1c3, and R1c4, R2c1, R2c3, and R2c4, R4c1, R4c3, and R4c4, R5c1, R5c3, and R5c4, R6c1, R6c3, and R6c4, or R7c1, R7c3, and R7c4 are H. 39. The compound of any of claims 1, 2, 4-30, or 34-36, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R1c1, R1c2, R1c3, and R1c4, R2c1, R2c2, R2c3, and R2c4, R4c1, R4c2, R4c3, and R4c4, R5c1, R5c2, R5c3, and R5c4, R6c1, R6c2, R6c3, and R6c4, or R7c1, R7c2, R7c3, and R7c4 are H. 40. The compound of any of claims 1, 2, or 4-30, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein A1, A2, A4, A5, A6, or A7 is , or 41. The compound of any of claims 1-6 or 8-40, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein B3, B4, B5, or B6 is a 3-membered monocyclic heterocyclediyl, a 4-membered monocyclic heterocyclediyl, a 5-membered monocyclic heterocyclediyl comprising 2 or more N, a 6- membered monocyclic heterocyclediyl comprising 2 or more N, a 7-membered monocyclic heterocyclediyl, an 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3-membered monocyclic heterocyclediyl, 4-membered monocyclic heterocyclediyl, 5- membered monocyclic heterocyclediyl, 6-membered monocyclic heterocyclediyl, 7-membered monocyclic heterocyclediyl, 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B3, B4, B5, or B6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1- 6alkyl, and oxo.

42. The compound of any of claims 1-6 or 8-40, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein B3, B4, B5, or B6 is a 3 to 8-membered monocyclic heterocyclediyl, wherein the 3 to 8-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C1-6alkyl, and oxo. 43. The compound of any of claims 1-6 or 8-42, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein B3, B4, B5, or B6 is a 4-membered monocyclic heterocyclediyl, wherein the 4- membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C1-6alkyl, and oxo. 44. The compound of any of claims 1-6 or 8-43, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein . 45. The compound of any of claims 1-6, 8-40 or 42, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein B3, B4, B5, or B6 is a 6-membered monocyclic heterocyclediyl, wherein the 6-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C1-6alkyl, and oxo. 46. The compound of any of claims 1-6, 8-42 or 45, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein B3, B4, B5, or B6 is . 47. The compound of any of claims 1-6 or 8-42, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein 48. The compound of any of claims 1-6 or 8-41, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein B3, B4, B5, or B6 is a 7 to 18-membered polycyclic heterocyclediyl, wherein the 7 to 18-membered polycyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C1-6alkyl, and oxo. 49. The compound of any of claims 1-6, 8-41, or 48, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein B3, B4, B5, or B6 is . 50. The compound of any of claims 1-6, 8-41, or 48, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein 51. The compound of any of claims 1-6, 8-41, or 48, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein . 52. The compound of any of claims 1-6 or 8-41, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein B3, B4, B5, or B6 is a 7 to 18-membered spirocyclic heterocyclediyl, wherein the 7 to 18-membered spirocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C1-6alkyl, and oxo. 53. The compound of any of claims 1-6 or 8-52, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B3, B4, B5, or B6 comprises one or more N. 54. The compound of any of claims 1-6, 8-43, or 45-54, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B3, B4, B5, or B6 comprises two or more N. 55. The compound of any of claims 1, 5, 8-30, or 34-54, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R5g1 is H. 56. The compound of any of claims 1, 5, 8-30, or 34-54, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R5g1 is -CH3. 57. The compound of any of claims 1, 7, 8-30, or 34-54, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R7g2 is H. 58. The compound of any of claims 1, 7, 8-30, or 34-54, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein R7g2 is -CH3. 59. The compound of any of claims 1, 7, 8-30, 34-54, 57, or 58 or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein X5 is -CH(R7g3)-, wherein R7g3 is H or C1-6alkyl. 60. The compound of any of claims 1, 7, 8-30, 34-54, 57, or 58, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein X5 is -CH(R7g4)CH2N(R7g5)-, wherein R7g4 and R7g5 are independently H or C1-6alkyl. 61. The compound of any one of claims 1-60, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is C1-6alkyl, wherein the C1-6alkyl is unsubstituted or substituted with halo. 62. The compound of any one of claims 1-60, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is cycloalkyl, wherein the cycloalkyl is unsubstituted or substituted with halo. 63. The compound of any one of claims 1-60, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is aryl, wherein the aryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, - OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)-cycloalkyl, - C(O)NH2, -C(O)NHCH3, and -C(O)N(CH3)2. 64. The compound of any of claims 1-60 or 63, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is a monocyclic 6-membered aryl, wherein the monocyclic 6- membered aryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1- 6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)-cycloalkyl, -C(O)NH2, -C(O)NHCH3, and - C(O)N(CH3)2. 65. The compound of any one of claims 1-60, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is heteroaryl, wherein the heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1- 6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1- 6haloalkyl, -C(O)-cycloalkyl, -C(O)NH2, -C(O)NHCH3, and -C(O)N(CH3)2. 66. The compound of any of claims 1-60 or 65, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is a monocyclic 5 or 6-membered heteroaryl comprising one or more N, wherein the monocyclic 5 or 6-membered heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, - OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)-cycloalkyl, - C(O)NH2, -C(O)NHCH3, and -C(O)N(CH3)2. 67. The compound of any one of claims 1-60, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is -O-C1-6alkyl or C1-6alkyl-O-C1-6alkyl, wherein the -O-C1-6alkyl or C1-6alkyl-O-C1-6alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)-cycloalkyl, -C(O)NH2, -C(O)NHCH3, and - C(O)N(CH3)2. 68. The compound of any one of claims 1-60, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, - CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1- 6alkyl-O-C1-6haloalkyl, -C(O)-cycloalkyl, -C(O)NH2, -C(O)NHCH3, and -C(O)N(CH3)2. 69. The compound of any one of claims 1-60, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is -C(O)-C1-6alkyl, -C(O)-cycloalkyl, or -C(O)-heterocyclyl, wherein the -C(O)-C1-6alkyl, -C(O)-cycloalkyl, or -C(O)-heterocyclyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, -C(O)NH2, -C(O)NHCH3, and -C(O)N(CH3)2. 70. The compound of any one of claims 1-60, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is -C(O)-aryl or -C(O)-heteroaryl, wherein the -C(O)-aryl or - C(O)-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1- 6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)-cycloalkyl, -C(O)NH2, -C(O)NHCH3, and - C(O)N(CH3)2. 71. The compound of any one of claims 1-60, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is -N(R1D1)(R1D2), -N(R2D1)(R2D2), -N(R3D1)(R3D2), -N(R4D1)(R4D2), -N(R5D1)(R5D2), -N(R6D1)(R6D2), or -N(R7D1)(R7D2). 72. The compound of any one of claims 1-60, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is -C(O)N(R1D3)(R1D4), -C(O)N(R2D3)(R2D4), -C(O)N(R3D3)(R3D4), -C(O)N(R4D3)(R4D4), -C(O)N(R5D3)(R5D4), -C(O)N(R6D3)(R6D4), or -C(O)N(R7D3)(R7D4). 73. The compound of any one of claims 1-60, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein D3, D4, D5, D6, or D7 is -N(R1D5)C(O)R1D6-, -N(R2D5)C(O)R2D6-, -N(R3D5)C(O)R3D6-, - N(R4D5)C(O)R4D6, -N(R5D5)C(O)R5D6-, -N(R6D5)C(O)R6D6-, or -N(R7D5)C(O)R7D6-. 74. The compound of claim 1, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of O NH N Me O Me N N H N N N CF3 , , O 75. The compound of any one of claims 1-7, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of O M O

76. The compound of any one of claims 1, 2, or 7, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of 77. The compound of any one of claims 1, 2, or 6, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of

78. The compound of any one of claims 1, 2, or 5, or a stereoisomer or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the group consisting of

. 79. The compound of any one of claims 1-78, wherein the compound selectively inhibits PARP7. 80. The compound of any one of claims 1-79, or a stereoisomer or a pharmaceutically acceptable salt thereof, for use in medicine. 81. The compound of any one of claims 1-79, or a stereoisomer or a pharmaceutically acceptable salt thereof, for use in the treatment of a disorder that is responsive to inhibition of PARP7. 82. The compound of any one of claims 1-79, or a stereoisomer or a pharmaceutically acceptable salt thereof, for use in the manufacture of a medicament for the treatment of a disorder that is responsive to inhibition of PARP7. 83. The compound of claim 81 or 82, wherein the disorder is selected from the group consisting of cancer, cardiovascular disorder, neurological disorder, inflammatory disorder, autoimmune disorder, and infectious disease. 84. The compound of any one of claims 81-83, wherein the disorder is cancer, and the cancer is of solid organ origin or of hematopoietic origin. 85. The compound of any one of claims 81-83, wherein the disorder is cancer, the cancer is of solid organ origin, and the solid organ is selected from the group consisting of the brain, breast, colon, endometrium, esophagus, head and neck, upper gastrointestinal tract, respiratory tract, lung, kidney, liver, lower gastrointestinal tract, small intestine, large intestine, ovary, pancreas, prostate, stomach, testes, and urinary tract. 86. The compound of any one of claims 81-83, wherein the disorder is cancer, and the cancer is leukemia or lymphoma. 87. The compound of claim 86, wherein the leukemia is acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), or chronic myelogenous leukemia (CML). 88. The compound of claim 86, wherein the lymphoma is Hodgkin's lymphoma , non-Hodgkin's lymphoma, multiple myeloma, B-cell lymphoma, diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic lymphoma (CLL), T-cell lymphoma, hairy cell lymphoma, or Burkett's lymphoma. 89. The compound of any one of claims 81-83, wherein the disorder is cancer and the cancer is selected from the group consisting of cancer of the bladder, bone cancer, cancer of the cervix, cancer of the epithelium, cancer of the gallbladder, cancer of the rectum, skin cancer, thyroid cancer, and cancer of the uterus.

90. A pharmaceutical composition comprising a compound of any one of claims 1-89, or a stereoisomer or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. 91. Use of a compound of any one of claims 1-89, or a stereoisomer or a pharmaceutically acceptable salt thereof, in the treatment of a disorder that is responsive to inhibition of PARP7. 92. A method of treating a disorder in a subject in need thereof, wherein the disorder is mediated by PARP7, comprising administering to the subject a compound of any one of claims 1-89, or a stereoisomer or a pharmaceutically acceptable salt thereof.

Description:
PHTHALAZINONE COMPOUNDS AS PARP7 INHIBITORS CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of U.S. Provisional Application Ser. No.63/326,484, filed on April 1, 2022 and entitled “Phthalazinone Compounds as PARP7 Inhibitors,” the entire contents of which are incorporated herein by reference. FIELD OF DISCLOSURE [0002] The present disclosure relates to compounds comprising a phthalazinone core, their use for modulating or inhibiting Poly (ADP-ribose) polymerase 7 (PARP7), and their use in pharmaceutical formulations. STATEMENT OF GOVERNMENT INTEREST [0003] This invention was made with government support under 2R01NS088629 awarded by the National Institutes of Health. The government has certain rights in this invention. BACKGROUND [0004] In humans there are 17 members of the PARP family of enzymes that catalyze the transfer of ADP-ribose from nicotinamide adenine dinucleotide (NAD + ) to amino acids on protein targets of post- translational modification (PTM). PARP7 is a member of the PARP family that catalyzes PTM known as mono-ADP-ribosylation (MARylation) as opposed to the poly-ADP-ribosylation (PARylation) effected by other PARPs such as PARP1 and PARP2. Multiple independent lines of evidence point to PARP7 catalytic activity as a regulator of interferon signaling. In mouse embryonic fibroblasts (MEFs), knockout of PARP7 increases the type I interferon, interferon-beta (IFN-β), and synergizes with pattern recognition receptor (PRR) ligands (e.g.3pRNA, agonist for RIG-1) to induce IFN-β production in cells. IFN-β has antitumor effects where it plays a role in dendritic cell (DC) driven T cell responses to various cancers. Hence, in cancers that overexpress PARP7 or that otherwise have dysregulated PARP7 activity, inhibition of PARP7 may increase IFN-β in the presence of PRR ligands, which could lead to immunogenic cell death and long-term protective antitumor immunity. Therefore, modulating or inhibiting PARP is a potential therapeutic approach for treating disorders such as cancer. Thus, there is a need for compounds that can modulate or inhibit PARP. SUMMARY [0005] The present disclosure provides a compound of Formula I: and stereoisomers and pharmaceutically acceptable salts thereof, wherein the constituent members are defined herein throughout the disclosure. [0006] The present disclosure further provides a pharmaceutical composition comprising a compound of the present disclosure, and stereoisomers and pharmaceutically acceptable salts thereof, and a pharmaceutically acceptable carrier. [0007] The present disclosure further provides a compound of the present disclosure, and stereoisomers and pharmaceutically acceptable salts thereof, for use in the treatment of a disorder that is responsive to inhibition of PARP7. [0008] The present disclosure further provides for use of a compound of the present disclosure, and stereoisomers and pharmaceutically acceptable salts thereof, in the treatment of a disorder that is responsive to inhibition of PARP7. [0009] The present disclosure further provides a compound of the present disclosure, and stereoisomers and pharmaceutically acceptable salts thereof, for use in the manufacture of a medicament for the treatment of a disorder that is responsive to inhibition of PARP7. [0010] The present disclosure further provides a method of treating a disorder in a subject in need thereof, wherein the disorder is mediated by PARP7, comprising administering to the subject a compound of the present disclosure. DESCRIPTION OF THE FIGURES [0011] The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee. The present application can be understood by reference to the following description taking in conjunction with the accompanying figures. [0012] FIGS.1A-E provide an overview of the rational design of Example 10, a potent membrane- permeable PARP7 inhibitor. FIG.1A provides chemical structures of Phthal01, Example 10, and RBN- 2397. Example 10 contains a propynyl group (shown in red) at the C-6 of the phthalazinone scaffold designed to impart PARP7-selectivity by interacting with a unique hydrophobic cavity in PARP7. FIG. 1B provides results of a family-wide PARP inhibitor screening assay that reveals that Example 10 is selective for PARP7. Selectivity data (IC 50 values) is represented as a heat map (red = more potent; and blue = less potent). IC 50 values for the data provided in the heat map can be found in Table 4 and individual dose response curves in FIGS.11 and 12. FIG.1C provides fit homology model of Example 10 (orange) bound to PARP7 and shows the propynyl group occupying the hydrophobic cavity formed by ile631 and hydrophobic amino acids in the D-loop (cyan). The induced fit homology model of RBN-2397 (steel blue) shows that the methyl substituent only partially occupies the hydrophobic cavity. FIG.1D provides data showing Example 10 inhibits GFP-PARP7 auto-MARylation in a dose-dependent manner in HEK 293T cells. GFP-PARP7 levels and MARylation were detected by Western blot analysis using an anti-GFP antibody and an anti-M/PAR antibody, respectively. Replicates shown in FIG.8. FIG.1E provides quantification of replicates in FIG.1D and FIG.8 using BioRad ImageLab software. Curves fit in Graphpad Prism with four parameter nonlinear regression. [0013] FIGS.2A-D demonstrate inhibition of PARP7 catalytic activity by Example 10 derepresses AHR ligand-mediated gene transcription in mouse embryonic fibroblasts (MEFs). FIG.2A provides CYP1A1 mRNA levels assessed by qPCR after treatment of wild type (WT) MEFs with increasing concentrations of Example 10 in the presence (gray bars) or absence (black bars) of the AHR agonist TCDD (1 nM). FIG.2B provides the same as in FIG.2A except with PARP7 -/- MEFs. FIG.2C provides CYP1A1 mRNA levels assessed by qPCR after treatment of wild type (WT) MEFs with increasing concentrations of TCDD in the presence (gray bars) or absence (black bars) of 100 nM Example 10. FIG. 2D provides the same as in FIG.2C, except with PARP7 -/- MEFs. [0014] FIGS.3A-C demonstrate Example 10 enhances 3pRNA- and cGAMP-stimulated IFN-β transcription. FIG.3A shows MEFs were treated with 100 nM Example 10 (black bar) or DMSO (white bar) for 24 h. IFN-β mRNA levels were measured using quantitative RT-PCR. IFN-β levels were normalized to expression of TBP mRNA levels. FIG.3B shows MEFs were treated with the RIG-I ligand 3pRNA (100 ng/ml) in the presence (black bar) or absence (gray bar) of 100 nM Example 10 for 4 h. IFN- β mRNA levels were assessed as in FIG.3A. FIG.3C shows MEFs were treated with the STING ligand cGAMP (5 mg/ml) in the presence (black bar) or absence (gray bar) of 100 nM Example 10 for 4 h. IFN-β mRNA levels were assessed as in FIG.3A. [0015] FIGS.4A-E demonstrate inhibition of PARP7 catalytic activity induces a STING-dependent type I interferon response and PARP7 protein accumulation in CT-26 cells. FIG.4A demonstrate Example 10 and RBN-2397 increase STAT1 and phosphor-Tyr701-STAT1 (pSTAT1) in a dose- dependent manner. CT-26 cells were treated with increasing concentrations of either Example 10 or RBN- 2397 for 16 h. FIG.4B provides of Western blots in FIG.4A. Quantification was performed using BioRad ImageLab software. Curves were fit using four parameter nonlinear regression in GraphPad Prism. STAT1 and pSTAT1 levels were normalized to tubulin. Data was from 3 biological replicates. FIG.4C demonstrates Example 10 and RBN-2397 induced STING degradation and signaling, but not to the same extent as a STING agonist. Treatment was as in FIG.4A. Note: tubulin blot was the same as in FIG.4A since samples were run on the same nitrocellulose membrane for western blot analysis. SE = short exposure; LE = long exposure. FIG.4D demonstrates Example 10 and RBN-2397 increase PARP7 levels in a dose-dependent manner. Treatment was as in FIG.4A. Note: tubulin blot is the same as in FIG.4A since samples were run on the same nitrocellulose membrane for western blot analysis. FIG.4E demonstrates quantification of Western blots in FIG.4D. Quantification was performed using BioRad ImageLab software. Curves fitting was done using a four parameter nonlinear regression in GraphPad Prism. PARP7 levels were normalized to tubulin. Data was from 3 biological replicates. FIG.4F demonstrates Example 10 and RBN-2397 increase IFN-β levels in a STING-dependent manner. CT-26 cells were treated with 300 nM Example 10 (dark gray) or 300 nM RBN-2397 (light gray) in the presence or absence of the covalent STING inhibitor H-151 (500 nM) for 16 h. IFN-β levels were determined by ELISA (PBL Assay Science). Statistical analysis was performed as one-way ANOVA in GraphPad Prism. **** = p<0.001. FIG.4G demonstrates Example 10 and RBN-2397 increase type I interferon reporter levels in a dose dependent manner. CT-26 cells stably expressing an IRSE luciferase reporter were treated with increasing concentrations of Example 10 or RBN-2397 for 16 h. Luciferase levels were measured using a luciferase assay reagent (Promega). [0016] FIGS.5A-D demonstrate PARP7 catalytic activity controls its protein abundance in the nucleus. FIG.5A demonstrates inhibition of PARP7 with either Example 10 or RBN-2397 increases nuclear PARP7 protein levels. HiBiT-PARP7 knockin (KI) CT-26 cells were treated with 300 nM KMR, 300 nM RBN-2397, or DMSO for 18 h. WT CT-26 cells were used as a control. HiBiT-PARP7 levels were determined by immunofluorescence staining with an anti-HiBiT antibody followed by an anti-mouse AlexaFluor647 antibody. Nuclei were stained with DAPI. Scale bar = 25 µm. FIG.5B provides quantification of DAPI and AlexaFluor647 fluorescence in FIG.5A across linear trace of nuclei. Replicate traces across RBN-2397 and Example 10 treated cells presented in FIG.11. FIG.5C demonstrates PARP7 levels were only detected in nucleus of CT-26 cells treated with PARP7 inhibitors. CT-26 cells were treated with 300 nM Example 10 or 300 nM RBN-2397 for 18 h. Cells were fractioned and PARP7 levels were detected by Western blot using an anti-PARP7 antibody. An anti-PARP-1 antibody was used as a nuclear marker whereas an anti-GAPDH was used as a cytosolic maker. FIG.5D demonstrates increases in nuclear PARP7 levels were be detected within 30 min of PARP7 inhibitor treatment. HeLa cells overexpressing GFP-PARP7 and mRuby2-Nup90 (nuclear pore marker) were treated with 300 nM Example 10, 300 nM RBN-2397, or DMSO and live cell images were taken at 0, 30, and 120 min. Representative images shown. Scale bar = 20 µm. [0017] FIG.6A provides individual dose response curves for Example 10 across the PARP family. FIG.6B further provides individual dose response curves for Example 10 across the PARP family. [0018] FIG.7A provides individual dose response curves for RBN-2397 across the PARP family. FIG.7B further provides individual dose response curves for RBN-2397 across the PARP family. [0019] FIG.8 provides replicate blots of FIG.1D. HEK 293T cells were transfected with GFP-PARP7 and treated with increasing concentrations of Example 10 PARP7 for 18 h. GFP-PARP7 auto- MARylation and GFP-PARP7 protein levels were determined by Western blot using specific antibodies. [0020] FIG.9 demonstrates PARP7 inhibition decreases viability in NCI-H1373 cells. Cell viability in NCI-H1373 cells was measured by Cell Titer-Glo assay following a 6 d treatment with increasing concentrations of Example 10 or RBN-2397. Curves were fit using three parameter a nonlinear regression in GraphPad Prism. [0021] FIG.10A-D demonstrates PARP7 inhibition, but not Type I interferons drives PARP7 protein accumulation and IFN-I signaling in a time-dependent manner. FIG.10A demonstrates Example 10 and RBN-2397 increase STAT1 and pSTAT1 in a time-dependent manner. CT-26 cells were treated with 300 nM Example 10 or RBN-2397 and cells were harvested for western blot analysis at the indicated times. FIG.10B demonstrates Example 10 and RBN-2397 decrease STING levels and increase pIRF3 levels in a time-dependent manner. Treatment was same as in FIG.10A. Note: tubulin blot is the same as in FIG. 10A since samples were run on the same nitrocellulose membrane for western blot analysis. FIG.10C demonstrates Example 10 and RBN-2397 increase PARP7 levels in a time-dependent manner. Treatment was same as in FIG.10A. Note: tubulin blot is the same as in FIG.10A since samples were run on the same nitrocellulose membrane for western blot analysis. FIG.10D shows results of CT-26 cells treated with indicated compounds, IFN-β (100 U/ml), or DMSO for 18 h. Proteins were detected by Western blot using specific antibodies. [0022] FIG.11 provides replicate cell traces from HiBiT-PARP7 CT-26 cells in FIG.4 treated with 0.3 µM RBN-2397 or Example 10. Traces were drawn and measured with ImageJ software. Plots were generated in GraphPad Prism. [0023] FIG.12 provides synthesis of Example 10 (a) NBS, AIBN, CCl 4 reflux; (b) PPh 3 , THF reflux; (c) 1) 2-fluoro-5-formylbenzonitrile, TEA, DCM 2) Hydrazine hydrate, H 2 O, EtOH, DMF reflux; (d) 1) KOH, EtOH, ddH 2 O, 2) HCl; (e) 6-(piperizino)pyridine-3-carbonitrile, propanephosphonic acid anhydride (T3P®), DIPEA, DMF; (f) tributyl(1-propynyl)tin, Pd(PPh 3 ) 4 , toluene reflux. DETAILED DESCRIPTION [0024] The following description sets forth numerous exemplary configurations, methods, parameters, and the like. It should be recognized, however, that such description is not intended as a limitation on the scope of the present disclosure, but is instead provided as a description of exemplary embodiments. [0025] As used herein, the terms “including,” “containing,” and “comprising” are used in their open, non-limiting sense. [0026] The articles “a” and “an”, as used herein, refer to one or more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” refers to one element or more than one element. [0027] To provide a more concise description, some of the quantitative expressions given herein are not qualified with the term “about”. It is understood that, whether the term “about” is used explicitly or not, every quantity given herein is meant to refer to the actual given value, and it is also meant to refer to the approximation to such given value that would reasonably be inferred based on the ordinary skill in the art, including equivalents and approximations due to the experimental and/or measurement conditions for such given value. Whenever a yield is given as a percentage, such yield refers to a mass of the entity for which the yield is given with respect to the maximum amount of the same entity that could be obtained under the particular stoichiometric conditions. Concentrations that are given as percentages refer to mass ratios, unless indicated differently. [0028] “Alkyl”, as used herein, refers to an unbranched or branched saturated hydrocarbon chain. Alkyl can be used alone, or as part of another radical, such as -O-alkyl. In some embodiments, alkyl as used herein has 1 to 20 carbon atoms ((C 1 - 20 )alkyl), 1 to 12 carbon atoms ((C 1 - 12 )alkyl), 1 to 10 carbon atoms ((C 1 - 10 )alkyl), 1 to 8 carbon atoms ((C 1 - 8 )alkyl), 1 to 6 carbon atoms ((C 1-6 )alkyl), 1 to 4 carbon atoms ((C 1 - 4 )alkyl), or 1 to 3 carbon atoms ((C 1 - 3 )alkyl). Examples of alkyl groups include methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, pentyl, 2-pentyl, isopentyl, neopentyl, hexyl, 2-hexyl, 3- hexyl, and 3-methyl pentyl. When an alkyl residue having a specific number of carbons is named, all geometric isomers having that number of carbons may be encompassed. Thus, for example, “butyl” can include n-butyl, sec-butyl, isobutyl and t-butyl, and ‘‘propyl” can include n-propyl and isopropyl. [0029] “Alkenyl”, as used herein, refers to an unbranched or branched hydrocarbon chain. The “alkenyl” group contains at least one double bond. The double bond of an alkenyl group can be unconjugated or conjugated to another group. The alkenyl may be branched or straight. In some embodiments, alkenyl as used herein has 2 to 20 carbon atoms ((C 2 - 20 )alkenyl), 2 to 12 carbon atoms ((C 2 - 12 )alkenyl), 2 to 10 carbon atoms ((C 2 - 10 )alkenyl), 2 to 8 carbon atoms ((C 2 - 8 )alkenyl), 2 to 6 carbon atoms ((C 2-6 )alkenyl, 2 to 4 carbon atoms ((C 2 - 4 )alkenyl), or 2 to 3 carbon atoms ((C 2 - 3 )alkenyl). Examples of alkenyl groups include, but are not limited to, ethylenyl, vinyl, allyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl, 2-ethylhexenyl, 2-propyl-2-butenyl, 4-(2-methyl-3-butene)-pentenyl and the like. When an alkenyl residue having a specific number of carbons is named, all geometric isomers and all E-Z isomers having that number of carbons may be encompassed. [0030] “Alkynyl”, as used herein, refers to an unbranched or branched unsaturated hydrocarbon chain. The “alkynyl” group contains at least one triple bond. The alkynyl may be branched or straight. The triple bond of an alkynyl group can be unconjugated or conjugated to another group. In some embodiments, alkynyl as used herein has 2 to 50 carbon atoms ((C 2 - 50 )alkynyl), 2 to 20 carbon atoms ((C 2 - 20 )alkynyl), 2 to 12 carbon atoms ((C 2 - 12 )alkynyl), 2 to 10 carbon atoms ((C 2 - 10 )alkynyl), 2 to 8 carbon atoms ((C 2 - 8 )alkynyl), 2 to 6 carbon atoms ((C 2-6 )alkynyl, 2 to 4 carbon atoms ((C 2 - 4 )alkynyl), or 2 to 3 carbon atoms ((C 2 - 3 )alkynyl). Examples of alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, pentynyl, hexynyl, methylpropynyl, 4-methyl-1-butynyl, 4-propyl-2-pentynyl, 4-butyl-2-hexynyl and the like. When an alkynyl residue having a specific number of carbons is named, all geometric isomers having that number of carbons may be encompassed. [0031] “Cycloalkyl”, as used herein, refers to a saturated or partially saturated, monocyclic, fused or spiro polycyclic, carbocycle having from 3 to 18 carbon atoms per ring. The cycloalkyl ring or carbocycle may be unsubstituted or substituted by one or more substituents, e.g., 1 to 5 substituents, at any point of attachment. The substituents can themselves be unsubstituted or substituted. Examples of cycloalkyl groups include, without limitations, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptanyl, cyclooctanyl, norboranyl, norborenyl, bicyclo[2.2.2]octanyl, bicyclo[2.2.2]octenyl, decahydronaphthalenyl, octahydro-1H-indenyl, cyclopentenyl, cyclohexenyl, cyclohexa-1,4-dienyl, cyclohexa-1,3-dienyl, 1,2,3,4-tetrahydronaphthalenyl, octahydropentalenyl, 3a,4,5,6,7,7a-hexahydro-1H- indenyl, 1,2,3,3a-tetrahydropentalenyl, bicyclo[3.1.0]hexanyl, bicyclo[2.1.0]pentanyl, spiro[3.3]heptanyl, bicyclo[2.2.1]heptanyl, bicyclo[2.2.1]hept-2-enyl, bicyclo[2.2.2]octanyl, 6-methylbicyclo[3.1.1]heptanyl, 2,6,6-trimethylbicyclo[3.1.1]heptanyl, and derivatives thereof. [0032] “Cycloalkenyl”, as used herein, refers to a partially saturated, monocyclic or fused or spiro polycyclic carbocycle having from 3 to 18 carbon atoms per ring and containing at least one double bond. The cycloalkenyl ring may be unsubstituted or substituted by one or more substituents, e.g., 1 to 5 substituents, at any point of attachment. The substituents can themselves be unsubstituted or substituted. [0033] “Heterocycle”, “heterocyclyl”, or “heterocyclediyl”, as used herein, refers to a saturated or partially unsaturated and non-aromatic monocyclic or fused polycyclic or spiro polycyclic ring structure of 4- to- 18 atoms containing carbon and heteroatoms taken from oxygen, nitrogen, or sulfur wherein there is not delocalized π-electrons (aromaticity) shared among all ring carbons or heteroatoms. A heterocyclyl ring structure attaches to a single point of a moiety of the formulae described herein, while a heterocyclediyl ring structure attaches to two points of a moiety or moieties of formulae described herein. The heterocycle, heterocyclyl, or heterocyclediyl ring structure may be unsubstituted or substituted by one or more substituents. The substituents can themselves be unsubstituted or substituted. Examples of heterocycle, heterocyclyl, or heterocyclediyl rings include, but are not limited to, oxetanyl, azetidinyl, tetrahydrofuranyl, pyrrolidinyl, oxazolinyl, oxazolidinyl, thiazolinyl, thiazolidinyl, pyranyl, thiopyranyl, tetrahydropyranyl, dioxalinyl, piperidinyl, morpholinyl, thiomorpholinyl, thiomorpholinyl S-oxide, thiomorpholinyl S-dioxide, piperazinyl, azepinyl, oxepinyl, diazepinyl, tropanyl, homotropanyl, dihydrothiophen-2(3H)-onyl, tetrahydrothiophene 1,1-dioxide, 2,5-dihydro-1H-pyrrolyl, imidazolidin-2- one, pyrrolidin-2-one, dihydrofuran-2(3H)-one, 1,3-dioxolan-2-one, isothiazolidine 1,1-dioxide, 4,5- dihydro-1H-imidazolyl, 4,5-dihydrooxazolyl, oxiranyl, pyrazolidinyl, 4H-1,4-thiazinyl, thiomorpholinyl, 1,2,3,4-tetrahydropyridinyl, 1,2,3,4-tetrahydropyrazinyl, 1,3-oxazinan-2-one, tetrahydro-2H-thiopyran 1,1-dioxide, 7-oxabicyclo[2.2.1]heptanyl, 1,2-thiazepane 1,1-dioxide, octahydro-2H-quinolizinyl, 1,3- diazabicyclo[2.2.2]octanyl, 2,3-dihydrobenzo[b][1,4]dioxine, 3-azabicyclo[3.2.1]octanyl, 8- azaspiro[4.5]decane, 8-oxa-3-azabicyclo[3.2.1]octanyl, 2-azabicyclo[2.2.1]heptane, 2,8- diazaspiro[5.5]undecanyl, 2-azaspiro[5.5]undecanyl, 3-azaspiro[5.5]undecanyl, decahydroisoquinolinyl, 1-oxa-8-azaspiro[4.5]decanyl, 8-azabicyclo[3.2.1]octanyl, 1,4’-bipiperidinyl, azepanyl, 8-oxa-3- azabicyclo[3.2.1]octanyl, 3,4-dihydro-2H-benzo[b][1,4]oxazinyl, 5,6,7,8-tetrahydroimidazo[1,2- a]pyridinyl, 1,4-diazepanyl, phenoxathiinyl, benzo[d][1,3]dioxolyl, 2,3-dihydrobenzofuranyl, 2,3- dihydrobenzo[b][1,4] dioxinyl, 4-(piperidin-4-yl)morpholinyl, 3-azaspiro[5.5]undecanyl, decahydroquinolinyl, piperazin-2-one, 1-(pyrrolidin-2-ylmethyl)pyrrolidinyl, 1,3’-bipyrrolidinyl, and 6,7,8,9-tetrahydro-1H,5H-pyrazolo[1,2-a][1,2]diazepinyl. [0034] “Aryl”, as used herein, refers to a monocyclic or polycyclic group having at least one hydrocarbon aromatic ring wherein all of the ring atoms of the at least one hydrocarbon aromatic ring are carbon. Aryl may include groups with a single aromatic ring (e.g., phenyl) and multiple fused aromatic rings (e.g., naphthyl, anthryl). Aryl may further include groups with one or more aromatic hydrocarbon rings fused to one or more non-aromatic hydrocarbon rings (e.g., fluorenyl; 2,3-dihydro-1H-indene; 1,2,3,4-tetrahydronaphthalene). In certain embodiments, aryl includes groups with an aromatic hydrocarbon ring fused to a non-aromatic ring wherein the non-aromatic ring comprises at least one ring hetero atom independently selected from the group consisting of N, O, and S. For example, in some embodiments, aryl includes groups with a phenyl ring fused to a non-aromatic ring, wherein the non- aromatic ring comprises at least one ring hetero atom independently selected from the group consisting of N, O, and S (e.g., chromane; thiochromane; 2,3-dihydrobenzofuran; indoline). In some embodiments, aryl as used herein has from 6 to 14 carbon atoms ((C 6 -C 14 )aryl), or 6 to 10 carbon atoms ((C 6 -C 10 )aryl). Where the aryl includes fused rings, the aryl may connect to one or more substituents or moieties of the formulae described herein through any atom of the fused ring for which valency permits. [0035] “Heteroaryl”, as used herein, refers to a monocyclic or polycyclic group comprising at least one aromatic ring, wherein the aromatic ring comprises at least one ring heteroatom independently selected from the group consisting of N, O, and S. The heteroaryl group may comprise 5, 6, 7, 8, 9, 10, 11, 12, or more ring atoms, where ring atoms refer to the sum of carbon and heteroatoms in the one or more rings (e.g., be a 5-membered, 6-membered, 7-membered, 8-membered, 9-membered, 10-membered, 11- membered, or 12-membered heteroaryl). In some embodiments, heteroaryl includes groups with an aromatic ring that comprises at least one ring heteroatom independently selected from the group consisting of N, O, and S, (e.g., pyridinyl, pyrazinyl, furanyl, thiophenyl). In certain embodiments, heteroaryl includes polycyclic groups with an aromatic ring comprising at least one ring heteroatom, fused to a non-aromatic hydrocarbon ring (e.g., 5,6,7,8-tetrahydroquinolinyl; 4,5,6,7- tetrahydroisobenzofuranyl). In some embodiments, heteroaryl includes polycyclic groups with an aromatic ring comprising at least one ring heteroatom fused to an aromatic hydrocarbon ring (e.g., quinolinyl, quinoxalinyl, benzothiazolyl). In still further embodiments, heteroaryl includes polycyclic groups with two fused aromatic rings, wherein each ring comprises at least one ring heteroatom (e.g., naphthyridinyl). Heteroaryl may include groups comprising 1 to 5 ring heteroatoms, 1 to 4 ring heteroatoms, 1 to 3 ring heteroatoms, 1 or 2 ring heteroatoms, or 1 ring heteroatom, wherein each ring heteroatom is independently selected from the group consisting of N, O, and S. In one example, a heteroaryl has 3 to 8 ring carbon atoms, with 1 to 3 ring heteroatoms independently selected from N, O, and S. Examples of heteroaryl groups include, without limitations, pyridyl, pyridazinyl, pyrimidinyl, benzothiazolyl, and pyrazolyl. [0036] As used herein, the term “substituted” means that the specified group or moiety bears one or more suitable substituents wherein the substituents may connect to the specified group or moiety at one or more positions. For example, an aryl substituted with a cycloalkyl may indicate that the cycloalkyl connects to one atom of the aryl with a bond or by fusing with the aryl and sharing two or more common atoms. [0037] As used herein, the term “unsubstituted” means that the specified group bears no substituents. [0038] “Amino”, as used herein, means a substituent containing at least one nitrogen atom. For example, NH 2 , -NH(alkyl) or alkylamino, -N(alkyl) 2 or dialkylamino, amide, carboxamide, urea, and sulfamide are included in the term “amino”. [0039] “Cyano”, as used herein, refers to a substituent having a carbon atom joined to a nitrogen atom by a triple bond, [0040] “Hydroxyl” or “hydroxy”, as used herein, refers to an OH group. [0041] “Halogen” or “halo”, as used herein, refers to fluoro, chloro, bromo, or iodo radicals. [0042] “Haloalkyl,” as used herein, refers to an alkyl group substituted with one or more halogen. [0043] “Halocycloalkyl”, as used herein, refers to a cycloalkyl group substituted with one or more halogen. [0044] “Haloaryl”, as used herein, refers to an aryl group substituted with one or more halogen. [0045] “Oxo”, as used herein, refers to an “=O” group. [0046] It should be understood that when a range of values is listed, it is intended to encompass each value and sub-range within the range. For example, “C 1-6 alkyl” (which may also be referred to as C1-C6 alkyl, C 1- C 6 alkyl, or C 1-6 alkyl) is intended to encompass C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1-6 , C 1-5 , C 1-4 , C 1-3 , C 1-2 , C 2-6 , C 2-5 , C 2-4 , C 2-3 , C 3-6 , C 3-5 , C 3-4 , C 4-6 , C 4-5 , and C 5-6 alkyl. [0047] It should also be noted that any carbon as well as heteroatom with unsatisfied valences in the text, schemes, examples and tables herein is assumed to have the sufficient number of hydrogen atom(s) to satisfy the valences. [0048] As used herein, references to hydrogen may also refer to a deuterium substitution if desired. The term “deuterium” as used herein means a stable isotope of hydrogen having odd numbers of protons and neutrons. [0049] Compounds of the various Formulae and stereoisomers and pharmaceutically acceptable salts thereof may exist in their tautomeric form (for example, as an amide or imino ether). All such tautomeric forms are contemplated herein as part of the present disclosure. [0050] It should be understood that all isomeric forms are included within the present disclosure, including mixtures thereof. If the compound contains a double bond, the substituent may be in the E or Z configuration. If the compound contains a disubstituted cycloalkyl the cycloalkyl substituent may have a cis- or trans- configuration. [0051] The compounds of the various Formulae may contain asymmetric or chiral centers, and, therefore, exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the various Formulae as well as mixtures thereof, including racemic mixtures, form part of the present disclosure. In some embodiments, isomers of the compounds herein are stereoisomers. In addition, the present disclosure embraces all geometric and positional isomers. For example, if a compound of the various Formulae incorporates a double bond or a fused ring, both the cis- and trans- forms, as well as mixtures, are embraced within the scope of the present disclosure. Each compound herein disclosed includes all the enantiomers that conform to the general structure of the compound. The compounds may be in a racemic or enantiomerically pure form, or any other form in terms of stereochemistry. The assay results may reflect the data collected for the racemic form, the enantiomerically pure form, or any other form in terms of stereochemistry. [0052] Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher’s acid chloride), separating the diastereomers, and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Also, some of the compounds of the various Formulae may be atropisomers (e.g., substituted biaryls) and are considered as part of the present disclosure. Enantiomers can also be separated by use of a chiral HPLC column. [0053] In some embodiments, the compounds of Formulae I, II, III, IV, V, VI, and VII and pharmaceutically acceptable salts thereof are enantiomers. In some embodiments, the compounds and pharmaceutically acceptable salts thereof are the (S)-enantiomer. In other embodiments the compounds and pharmaceutically acceptable salts thereof are the (R)-enantiomer. In some embodiments, the compounds and pharmaceutically acceptable salts thereof are the (+) enantiomer or (-) enantiomer. [0054] Some embodiments are directed to isotopically-labelled compounds of the present disclosure which are identical to those recited herein but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the present disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, such as 2 H (or D), 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl, respectively. [0055] Certain isotopically-labelled compounds of the various Formulae (e.g., those labeled with 3 H and 14 C) are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon- 14 (i.e., 14 C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Isotopically labelled compounds of the various Formulae can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below by substituting an appropriate isotopically labelled reagent for a non- isotopically labelled reagent. [0056] In some embodiments, the compound comprises at least one deuterium atom. For example, one or more hydrogen atoms in a compound of the present disclosure can be replaced or substituted by deuterium. In some embodiments, the compound comprises two or more deuterium atoms. In some embodiments, the compound comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 deuterium atoms. [0057] The compounds of Formulae I, II, III, IV, V, VI, and VII may form salts which are also within the scope of the present disclosure. Reference to a compound of the Formula herein is understood to include reference to salts thereof, unless otherwise indicated. [0058] The present disclosure is directed to compounds as described herein and stereoisomers and pharmaceutically acceptable salts thereof. The present disclosure is also directed to pharmaceutical compositions comprising one or more compounds as described herein and stereoisomers and pharmaceutically acceptable salts thereof . [0059] “Pharmaceutically acceptable”, as used herein, refers to that which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and not biologically or otherwise undesirable, and includes that which is acceptable for veterinary use as well as human pharmaceutical use. For example, provided herein is a pharmaceutical composition comprising a compound of Formulae I, II, III, IV, V, VI, or VII and stereoisomers and pharmaceutically acceptable salts thereof, and a pharmaceutically acceptable excipient. [0060] “Pharmaceutically acceptable salt”, as used herein, refers to a salt which is generally safe, non- toxic and not biologically or otherwise undesirable, and includes that which is acceptable for veterinary use as well as human pharmaceutical use. Such salts may include acid addition salts and base addition salts. Acid addition salts may be formed with inorganic acid such as, but not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like; or an organic acid such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo-glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-1,5-disulfonic acid, naphthalene-2-sulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, p-toluenesulfonic acid, trifluoroacetic acid, or undecylenic acid. Salts derived from inorganic bases may include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, and aluminum salts. Salts derived from organic bases may include, but are not limited to, salts of primary, secondary, or tertiary amines; substituted amines including naturally occurring substituted amines; cyclic amines; ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, or N-ethylpiperidine. [0061] The term “carrier”, as used herein, encompasses carriers, excipients, and diluents and refers to a material, composition, or vehicle such as a liquid or solid filler, diluent, excipient, solvent, or encapsulating material involved in carrying or transporting a pharmaceutical agent from one organ, or portion of the body, to another organ, or portion of the body, of a subject. Excipients should be selected on the basis of compatibility and the release profile properties of the desired dosage form. Exemplary carrier materials include, e.g., binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, spray-dried dispersions, and the like. [0062] “Pharmaceutically compatible carrier materials” may include, e.g., acacia, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, maltodextrin, glycerine, magnesium silicate, sodium caseinate, soy lecithin, sodium chloride, tricalcium phosphate, dipotassium phosphate, sodium stearoyl lactylate, carrageenan, monoglyceride, diglyceride, pregelatinized starch, and the like. See, e.g., Hoover, John E., Remington’s Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa.1975. [0063] “Solvate”, as used herein, refers to a complex of variable stoichiometry formed by a solute and solvent. Such solvents for the purpose of the present disclosure may not interfere with the biological activity of the solute. Examples of suitable solvents include, but are not limited to, water, methanol, ethanol, and acetic acid. Solvates wherein water is the solvent molecule are typically referred to as hydrates. Hydrates include compositions containing stoichiometric amounts of water, as well as compositions containing variable amounts of water. Compounds [0064] The present disclosure provides a compound of Formula I: and stereoisomers and pharmaceutically acceptable salts thereof, wherein: X 1 is -N- or -CR 1a2 -; X 2 is -N- or -CR 1a4 -; R 1a1 , R 1a2 , R 1a3 , and R 1a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 1L1 R 1L2 , or C 2-6 alkynyl-NR 1L3 R 1L4 ; wherein if X 1 is -CR 1a2 - and X 2 is -CR 1a4 -, then at least one of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 ; are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 1L1 , R 1L2 , R 1L3 , and R 1L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 1b1 -, or -CR 1b2 R 1b3 -, wherein R 1b1 , R 1b2 , and R 1b3 are independently H or C 1-6 alkyl; , wherein a bond marked 1A is to X 3 and a bond marked 2B is to D3, D4, D5, or D6; R 3f1 is H or C 1-6 alkyl; X 4 is -O-, -NR 3f2 -, or -CR 3f3 R 3f4 -, wherein R 3f2 , R 3f3 , and R 3f4 are independently selected from H and C 1-6 alkyl; A1 is alo, -CN, -SO 2 CH 3 , -SO 2 NH 2 , bond marked 2B is to D3, D4, D5, or D6; X 5 is -CH(R 7g3 )- or -CH(R 7g4 )CH 2 N(R 7g5 )-; R 5g1 , R 7g2 , R 7g3 , R 7g4 , and R 7g5 are independently H or C 1-6 alkyl; B3, B4, B5, and B6 are independently a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18- membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B3, B4, B5, and B6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1- 6 alkyl, and oxo; provided that B4 is not D3, D4, D5, D6, and D7 are independently C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, - O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)- heteroaryl, -N(R 1D1 )(R 1D2 ), -C(O)N(R 1D3 )(R 1D4 ), or -N(R 1D5 )C(O)R 1D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D3, D4, D5, D6, and D7 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, - CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O- C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D10 ) 2 , wherein each R 1D10 is independently H or C 1-6 alkyl; R 1D1 , R 1D3 , and R 1D5 are independently H or C 1-6 alkyl; R 1D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 1D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 1D11 ) 2 , wherein each R 1D11 is independently H or C 1-6 alkyl; and R 1D4 and R 1D6 are independently C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 1D4 and R 1D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D12 ) 2 , wherein each R 1D12 is independently H or C 1-6 alkyl; provided that , 1a2 then R is not F, if and R 1c2 is F, then R 1a2 is not F. [0065] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, X 1 is -CR 1a2 -. In some embodiments, X 2 is -CR 1a4 -. In some embodiments, X 1 is - N-. In some embodiments, X 2 is -N-. In some embodiments, X 1 is -CR 1a2 -. In some embodiments, X 1 is - CR 1a2 - and X 2 is -N-. In some embodiments, X 1 is -N- and X 2 is -CR 1a4 -. [0066] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, R 1a2 is H, Cl, Br, I, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 1L1 R 1L2 , or C 2-6 alkynyl-NR 1L3 R 1L4 , wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 -cycloalkyl of R 4a2 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . [0067] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, R 1a4 is H, F, Br, I, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 1L1 R 1L2 , or C 2-6 alkynyl-NR 1L3 R 1L4 , wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 -cycloalkyl of R 1a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . [0068] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is not H. In some embodiments, R 1a1 is not H. In some embodiments, R 1a2 is not H. In some embodiments R 1a3 is not H. In some embodiments, R 1a4 is not H. [0069] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is halo. In some embodiments, R 1a1 is halo. In some embodiments, R 1a2 is halo. In some embodiments, R 1a3 is halo. In some embodiments, R 1a4 is halo. In some embodiments, the halo is F, Cl, or Br. In some embodiments, the halo is Cl. In some embodiments, the halo is F. In some embodiments, R 1a1 is F. In some embodiments, R 1a3 is F. In some embodiments, R 1a3 is F. [0070] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is -OH. In some embodiments, R 1a1 is - OH. In some embodiments, R 1a2 is -OH. In some embodiments, R 1a3 is -OH. In some embodiments, R 1a4 is -OH. [0071] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is C 1-6 alkyl. In some embodiments, R 1a1 is C 1-6 alkyl. In some embodiments, R 1a2 is C 1-6 alkyl. In some embodiments, R 1a3 is C 1-6 alkyl. In some embodiments, R 1a4 is C 1-6 alkyl. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, R 1a1 , R 1a2 , R 1a3 , or R 1a4 is -CH 3 , -CH 2 CH 3 , -CH 2 CF 3 , -CF 3 , - [0072] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is C 2-6 alkenyl. In some embodiments, R 1a1 is C 2-6 alkenyl. In some embodiments, R 1a2 is C 2-6 alkenyl. In some embodiments, R 1a3 is C 2-6 alkenyl. In some embodiments, R 1a4 is C 2-6 alkenyl. In some embodiments, the C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkenyl is substituted with halo. In some embodiments, R 1a1 , R 1a2 , R 1a3 , or R 1a4 is or . [0073] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is C 2-6 alkynyl. In some embodiments, R 1a1 is C 2-6 alkynyl. In some embodiments, R 1a2 is C 2-6 alkynyl. In some embodiments, R 1a3 is C 2-6 alkynyl. In some embodiments, R 1a4 is C 2-6 alkynyl. In some embodiments, the C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C2-6alkynyl is substituted with halo. In some embodiments, the C2-6alkynyl is substituted with cycloalkyl. In some embodiments, R 1a1 , R 1a2 , R 1a3 , or R 1a4 is , [0074] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is heterocyclyl. In some embodiments, R 1a1 is heterocyclyl. In some embodiments, R 1a2 is heterocyclyl. In some embodiments, R 1a3 is heterocyclyl. In some embodiments, R 1a4 is heterocyclyl. In some embodiments, the heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the heterocyclyl is substituted with halo. In some embodiments, the heterocyclyl is substituted with -OH. In some embodiments, R 1a1 , R 1a2 , R 1a3 , or R 1a4 is , . [0075] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is aryl or heteroaryl. In some embodiments, R 1a1 is aryl or heteroaryl. In some embodiments, R 1a2 is aryl or heteroaryl. In some embodiments, R 1a3 is aryl or heteroaryl. In some embodiments, R 1a4 is aryl or heteroaryl. In some embodiments, the aryl or heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, R 1a1 , R 1a2 , R 1a3 , or R 1a4 is . [0076] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is -O-C 1-6 alkyl. In some embodiments, R 1a1 is -O-C 1-6 alkyl. In some embodiments, R 1a2 is -O-C 1-6 alkyl. In some embodiments, R 1a3 is -O-C 1- 6 alkyl. In some embodiments, R 1a4 is -O-C 1-6 alkyl. In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, - OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl -O-C 1 6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 1-6 alkyl is substituted with halo. In some embodiments, the -O-C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, R 1a1 , R 1a2 , R 1a3 , or R 1a4 is -OCH3, -OCHF2, -OCH2CF3, [0077] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is -O-C 2-6 alkenyl. In some embodiments, R 1a1 is -O-C 2-6 alkenyl. In some embodiments, R 1a2 is -O-C 2-6 alkenyl. In some embodiments, R 1a3 is -O-C 2- 6 alkenyl. In some embodiments, R 1a4 is -O-C 2-6 alkenyl. In some embodiments, the -O-C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 2-6 alkenyl is substituted with halo. [0078] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is -O-C 2-6 alkynyl. In some embodiments, R 1a1 is -O-C 2-6 alkynyl. In some embodiments, R 1a2 is -O-C 2-6 alkynyl. In some embodiments, R 1a3 is -O-C 2- 6 alkynyl. In some embodiments, R 1a4 is -O-C 2-6 alkynyl. In some embodiments, the -O-C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 2-6 alkynyl is substituted with halo. In some embodiments, R 1a1 , R 1a2 , R 1a3 , [0079] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is -O-cycloalkyl. In some embodiments, R 1a1 is -O-cycloalkyl. In some embodiments, R 1a2 is -O-cycloalkyl. In some embodiments, R 1a3 is -O- cycloalkyl. In some embodiments, R 1a4 is cycloalkyl. In some embodiments, the -O-cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-cycloalkyl is substituted with halo. In some embodiments, the -O- cycloalkyl is substituted with C 1-6 haloalkyl. In some embodiments, R 1a1 , R 1a2 , R 1a3 , or R 1a4 is , . [0080] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is -O-heterocyclyl. In some embodiments, R 1a1 is -O-heterocyclyl. In some embodiments, R 1a2 is -O-heterocyclyl. In some embodiments, R 1a3 is -O- heterocyclyl. In some embodiments, R 1a4 is -O-heterocyclyl. In some embodiments, the -O-heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heterocyclyl is substituted with halo. [0081] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is -O-aryl. In some embodiments, R 1a1 is - O-aryl. In some embodiments, R 1a2 is -O-aryl. In some embodiments, R 1a3 is -O-aryl. In some embodiments, R 1a4 is -O-aryl. In some embodiments, the -O-aryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-aryl is substituted with halo. In some embodiments, the -O-aryl is substituted with C 1-6 alkyl. In some embodiments, the -O-aryl is substituted with -C(O)NH 2 . In some embodiments, the aryl of is a 6 membered monocyclic aryl. In some embodiments, R 1a3 is -O-aryl wherein the aryl is an unsubstituted 6- membered monocyclic aryl. In some embodiments, R 1a1 , [0082] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is -O-heteroaryl. In some embodiments, R 1a1 is -O-heteroaryl. In some embodiments, R 1a2 is -O-heteroaryl. In some embodiments, R 1a3 is -O- heteroaryl. In some embodiments, R 1a4 is -O-heteroaryl. In some embodiments, the -O-heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heteroaryl is substituted with halo. In some embodiments, R 1a1 , R 1a2 , R 1a3 , . [0083] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is -SO 2 -cycloalkyl. In some embodiments, R 1a1 is -SO 2 -cycloalkyl. In some embodiments, R 1a2 is -SO 2 -cycloalkyl. In some embodiments, R 1a3 is -SO 2 -cycloalkyl. In some embodiments, R 1a4 is -SO 2 -cycloalkyl. In some embodiments, the -SO 2 -cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -SO 2 -cycloalkyl is substituted with halo. In some embodiments, [0084] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is -NR 1L1 R 1L2 . In some embodiments, R 1a1 is -NR 1L1 R 1L2 . In some embodiments, R 1a2 is -NR 1L1 R 1L2 . In some embodiments, R 1a3 is -NR 1L1 R 1L2 . In 1 a4 1 some embodiments, R is -NR L1 R 1L2 . In some embodiments, R 1a1 , R 1a2 , R 1a3 , or R 1a4 is , , [0085] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, R 1L1 and R 1L2 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 1L1 is H. In some embodiments, R 1L1 is H and R 1L2 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 1L1 is -CH 3 . In some embodiments, R 1L1 is -CH 3 and R 1L2 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 1L1 is H or -CH 3 and R 1L2 is C 1-6 alkyl. In some embodiments, R 1L1 is H or -CH 3 and R 1L2 is C 2-6 alkynyl. In some embodiments, R 1L1 is H or -CH 3 and R 1L2 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C 1-6 alkyl is substituted with cycloalkyl. [0086] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is C 2-6 alkynyl-NR 1L3 R 1L4 . In some embodiments, R 1a1 is C 2-6 alkynyl-NR 1L3 R 1L4 . In some embodiments, R 1a2 is C 2-6 alkynyl-NR 1L3 R 1L4 . In some embodiments, R 1a3 is C 2-6 alkynyl-NR 1L3 R 1L4 . In some embodiments, R 1a4 is C 2-6 alkynyl-NR 1L3 R 1L4 . [0087] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, R 1L3 and R 1L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 1L3 is H. In some embodiments, R 1L3 is H and R 1L4 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 1L3 is -CH 3 . In some embodiments, R 1L3 is -CH 3 and R 1L4 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 1L3 is C 1-6 alkyl and R 1L4 is C 1-6 alkyl. In some embodiments, R 1L3 is H or -CH 3 and R 1L4 is C 1-6 alkyl. In some embodiments, R 1L3 is H or -CH 3 and R 1L4 is C 2-6 alkynyl. In some embodiments, R 1L3 is H or -CH 3 and R 1L4 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, [0088] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, two or more of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is not H. In some embodiments, R 1a3 is not H and one of R 1a1 , R 1a2 , or R 1a4 is not H. In some embodiments, R 1a2 is F or Cl and R 1a3 is -O-C 1-6 alkyl or -O-cycloalkyl, wherein the -O-C 1-6 alkyl or -O-cycloalkyl is unsubstituted or substituted with halo or - CN. In some embodiments, R 1a1 is not H and R 1a2 is not H. In some embodiments, R 1a1 is F or Cl and R 1a2 is F or Cl. [0089] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, X 3 is -O-. In some embodiments, X 3 is -NR 1b1 -. In some embodiments, X 3 is -NH- . In some embodiments, X 3 is -N(CH 3 )-. In some embodiments, X 3 is -CR 1b2 R 1b3 -. In some embodiments, X 3 is -CH(CH 3 )-. In some embodiments, X 3 is -CD 2 -. In some embodiments, X 3 is -CH 2 -. [0090] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, X 4 is -O-. In some embodiments, X 4 is -NR 3f2 -. In some embodiments, X 4 is -NH- . In some embodiments, X 4 is -N(CH 3 )-. In some embodiments, X 4 is -CR 3f3 R 3f4 -. In some embodiments, X 4 is -CH(CH 3 )-. In some embodiments, X 4 is -CH 2 -. [0091] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, some embodiments, one or more of X 10 , X 11 , and X 12 is -N-. In some embodiments, X 10 is -N-. In some embodiments, X 11 is -N-. In some embodiments, X 12 is -N- . In some embodiments, X 10 is -CR 1c1 -. In some embodiments, X 11 is -CR 1c2 -. In some embodiments, X 12 is -CR 1c3 . In some embodiments, X 10 is -CR 1c1 -, X 11 is -CR 1c2 -, and X 12 is -CR 1c3 -. [0092] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 1c1 , R 1c2 , R 1c3 , and R 1c4 is not H. In some embodiments, one or more of R 1c1 , R 1c2 , R 1c3 , and R 1c4 is -CN. In some embodiments, one or more of R 1c1 , R 1c2 , R 1c3 , and R 1c4 is independently Cl, F, or Br. In some embodiments, two or more of R 1c1 , R 1c2 , R 1c3 , and R 1c4 are independently Cl, F, or Br. In some embodiments, R 1c2 is F and R 1c1 , R 1c3 , and R 1c4 are H. In some embodiments, R 1c1 , R 1c2 , R 1c3 , and R 1c4 are H. [0093] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof,

[0094] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, B3, B4, B5, or B6 is a 3-membered monocyclic heterocyclediyl, a 4-membered monocyclic heterocyclediyl, a 5-membered monocyclic heterocyclediyl comprising 2 or more N, a 6- membered monocyclic heterocyclediyl comprising 2 or more N, a 7-membered monocyclic heterocyclediyl, an 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl, wherein the 3-membered monocyclic heterocyclediyl, 4-membered monocyclic heterocyclediyl, 5-membered monocyclic heterocyclediyl, 6- membered monocyclic heterocyclediyl, 7-membered monocyclic heterocyclediyl, 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B3, B4, B5, or B6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18-membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered polycyclic heterocyclediyl. In some embodiments, the 7 to 18-membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered spirocyclic heterocyclediyl. [0095] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, B3, B4, B5, or B6 is a 3 to 8-membered monocyclic heterocyclediyl, wherein the 3 to 8-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 3 to 8- membered monocyclic heterocyclediyl is a 3, 4, 5, 6, 7, or 8-membered monocyclic heterocyclediyl. In some embodiments, B3, B4, B5, or B6 is a 4-membered monocyclic heterocyclediyl, wherein the 4- membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B3, B4, B5, or B6 is In some embodiments, B3, B4, B5, or B6 is a 6-membered monocyclic heterocyclediyl, wherein the 6-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B3, B4, B5, or B6 is . In some embodiments, B3, B4, B5, or B6 is a 7- membered monocyclic heterocyclediyl, wherein the 7-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1- 6 alkyl, and oxo. In some embodiments, [0096] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, B3, B4, B5, or B6 is a 7 to 18-membered polycyclic heterocyclediyl, wherein the 7 to 18-membered polycyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18- membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered polycyclic heterocyclediyl. In some embodiments, B3, B4, B5, or B6 is some embodiments, B3, B4, B5, or B6 is . In some embodiments B3, B4, B5, or B6 is [0097] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, B3, B4, B5, or B6 is a 7 to 18-membered spirocyclic heterocyclediyl, wherein the 7 to 18-membered spirocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18- membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered spirocyclic heterocyclediyl. In some embodiments, [0098] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B3, B4, B5, or B6 comprises one or more N. In some embodiments, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B3, B4, B5, or B6 comprises two or more N. [0099] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, R 5g1 is H. In some embodiments, R 5g1 is C 1-6 alkyl. In some embodiments, R 5g1 is - CH 3 . [0100] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, R 7g2 is H. In some embodiments, R 7g2 is C 1-6 alkyl. In some embodiments, R 7g2 is - CH 3 . In some embodiments, R 7g2 is -CH 2 CH 3 . [0101] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, X 5 is -CH(R 7g3 )-, wherein R 7g3 is H or C 1-6 alkyl. In some embodiments, X 5 is - CH 2 -. In some embodiments, X 5 is -CH(CH 3 )-. [0102] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, X 5 is -CH(R 7g4 )CH 2 N(R 7g5 )-, wherein R 7g4 and R 7g5 are independently H or C 1- 6 alkyl. In some embodiments, R 7g4 is H and R 7g5 is C 1-6 alkyl. In some embodiments, R 7g5 is -CH 3 . In some embodiments, R 7g5 is -CH 2 CH 3 . In some embodiments, R 7g4 is H and R 7g5 is H. [0103] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is C 1-6 alkyl, wherein the C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D10 ) 2 . In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -OH. In some embodiments, the C1-6alkyl is unsubstituted or substituted with -O-C1-6alkyl. [0104] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is cycloalkyl, wherein the cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D10 ) 2 . In some embodiments, the cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D3, D4, D5, D6, or D7 is . [0105] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is aryl, wherein the aryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D10 ) 2 . In some embodiments, the aryl is a monocyclic 6- membered aryl. In some embodiments, the aryl is substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments, the aryl is substituted with -CN. [0106] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is heteroaryl, wherein the heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0107] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is a monocyclic 5 or 6-membered heteroaryl comprising one or more N, wherein the monocyclic 5 or 6-membered heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . In some embodiments, the monocyclic 5 or 6- membered heteroaryl is substituted with -CN. In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CF 3 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -OCH 3 . [0108] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is -O-C 1-6 alkyl, wherein the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D10 ) 2 . In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with halo. [0109] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O- heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1- 6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D10 ) 2 . In some embodiments, the -O-aryl or -O-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -O-aryl or -O-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0110] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is -C(O)-C 1-6 alkyl, wherein the -C(O)-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D10 ) 2 . In some embodiments, the - C(O)-C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the -C(O)-C 1-6 alkyl is unsubstituted or substituted with -CN. In some embodiments, D3, D4, D5, D6, or D7 is [0111] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is -C(O)-cycloalkyl, wherein the -C(O)-cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D10 ) 2 . In some embodiments, the - C(O)-cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D3, D4, D5, D6, or D7 is . [0112] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is -C(O)-heterocyclyl, wherein the -C(O)-heterocyclyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D10 )2. In some embodiments, the - C(O)-heterocyclyl is unsubstituted or substituted with halo. In some embodiments, D3, D4, D5, D6, or D7 [0113] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is -C(O)-aryl or -C(O)-heteroaryl, wherein the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1- 6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D10 ) 2 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CF 3 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0114] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is -N(R 1D1 )(R 1D2 ). In some embodiments, D3, D4, D5, D6, [0115] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, R 1D1 is H. In some embodiments, R 1D1 is C 1-6 alkyl. In some embodiments, R 1D2 is aryl. In some embodiments, R 1D2 is heteroaryl. In some embodiments, the heteroaryl is a 6-membered heteroaryl. [0116] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is -C(O)N(R 1D3 )(R 1D4 ). In some embodiments, D3, D4, [0117] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, R 1D3 is H. In some embodiments, R 1D3 is C 1-6 alkyl. In some embodiments, R 1D4 is C 1-6 alkyl. In some embodiments, R 1D3 is H and R 1D4 is C 1-6 alkyl. [0118] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, D3, D4, D5, D6, or D7 is -N(R 1D5 )C(O)R 1D6 . In some embodiments, D3, D4, D5, [0119] In some embodiments of the compounds of Formula I and stereoisomers and pharmaceutically acceptable salts thereof, R 1D5 is H. In some embodiments, R 1D5 is C 1-6 alkyl. In some embodiments, R 1D6 is C 1-6 alkyl. In some embodiments, R 1D6 is cycloalkyl. In some embodiments, R 1D6 is heterocyclyl. In some embodiments, R 1D5 is H and R 1D6 is C 1-6 alkyl, cycloalkyl, or heterocyclyl. [0120] In some embodiments, the compound is of Formula II: and stereoisomers and pharmaceutically acceptable salts thereof, wherein: X 1 is -N- or -CR 2a2 -; X 2 is -N- or -CR 2a4 -; R 2a1 , R 2a2 , R 2a3 , and R 2a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 2L1 R 2L2 , or C 2-6 alkynyl-NR 2L3 R 2L4 ; wherein if X 1 is -CR 2a2 - and X 2 is -CR 2a4 -, then at least one of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2; R 2L1 , R 2L2 , R 2L3 , and R 2L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1- 6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 2L1 , R 2L2 , R 2L3 , and R 2L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 2b1 -, or -CR 2b2 R 2b3 -, wherein R 2b1 , R 2b2 , and R 2b3 are independently H or C 1-6 alkyl; bond marked 2B is to D4, D5, or D6; X 5 is -CH(R 7g3 )- or -CH(R 7g4 )CH 2 N(R 7g5 )-; R 5g1 , R 7g2 , R 7g3 , R 7g4 , and R 7g5 are independently H or C 1-6 alkyl; B4, B5, and B6 are independently a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18- membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B4, B5, and B6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1- 6 alkyl, and oxo; provided that B4 is not D4, D5, D6, and D7 are independently C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O- aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 2D1 )(R 2D2 ), -C(O)N(R 2D3 )(R 2D4 ), or -N(R 2D5 )C(O)R 2D6 ; wherein the C1-6alkyl, cycloalkyl, aryl, heteroaryl, -O-C1-6alkyl, -O-aryl, -O-heteroaryl, -C(O)-C1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D4, D5, D6, and D7 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D10 ) 2 , wherein each R 2D10 is independently H or C 1-6 alkyl; R 2D1 , R 2D3 , and R 2D5 are independently H or C 1-6 alkyl; R 2D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 2D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 2D11 ) 2 , wherein each R 2D11 is independently H or C 1-6 alkyl; and R 2D4 and R 2D6 are independently C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 2D4 and R 2D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D12 ) 2 , wherein each R 2D12 is independently H or C 1-6 alkyl; provided that , the 2a2 n R is not F, if and R 2c2 is F, then R 2a2 is not F. [0121] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, X 1 is -CR 2a2 -. In some embodiments, X 2 is -CR 2a4 -. In some embodiments, X 1 is - N-. In some embodiments, X 2 is -N-. In some embodiments, X 1 is -CR 2a2 - and X 2 is -CR 2a4 -. In some embodiments, X 1 is -CR 2a2 - and X 2 is -N-. In some embodiments, X 1 is -N- and X 2 is -CR 2a4 -. [0122] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, R 2a2 is H, Cl, Br, I, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 2L1 R 2L2 , or C 2-6 alkynyl-NR 2L3 R 2L4 , wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 -cycloalkyl of R 2a2 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . [0123] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, R 2a4 is H, F, Br, I, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 2L1 R 2L2 , or C 2-6 alkynyl-NR 2L3 R 2L4 , wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 -cycloalkyl of R 2a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2. [0124] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is not H. In some embodiments, R 2a1 is not H. In some embodiments, R 2a2 is not H. In some embodiments, R 2a3 is not H. In some embodiments, R 2a4 is not H. [0125] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is halo. In some embodiments, R 2a1 is halo. In some embodiments, R 2a2 is halo. In some embodiments, R 2a3 is halo. In some embodiments, R 2a4 is halo. In some embodiments, the halo is F, Cl, or Br. In some embodiments, the halo is Cl. In some embodiments, the halo is F. In some embodiments, R 2a1 is F. In some embodiments, R 2a3 is F. In some embodiments, R 2a3 is F. [0126] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is -OH. In some embodiments, R 2a1 is - OH. In some embodiments, R 2a2 is -OH. In some embodiments, R 2a3 is -OH. In some embodiments, R 2a4 is -OH. [0127] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is C 1-6 alkyl. In some embodiments, R 2a1 is C 1-6 alkyl. In some embodiments, R 2a2 is C 1-6 alkyl. In some embodiments, R 2a3 is C 1-6 alkyl. In some embodiments, R 2a4 is C 1-6 alkyl. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, R 2a1 , R 2a2 , R 2a3 , or R 2a4 is -CH 3 , -CH 2 CH 3 , -CH 2 CF 3 , -CF 3 , - [0128] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is C 2-6 alkenyl. In some embodiments, R 2a1 is C 2-6 alkenyl. In some embodiments, R 2a2 is C 2-6 alkenyl. In some embodiments, R 2a3 is C 2-6 alkenyl. In some embodiments, R 2a4 is C 2-6 alkenyl. In some embodiments, the C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2. In some embodiments, the C 2-6 alkenyl is substituted with halo. In some embodiments, R 2a1 , R 2a2 , R 2a3 , or R 2a4 is or . [0129] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is C2-6alkynyl. In some embodiments, R 2a1 is C 2-6 alkynyl. In some embodiments, R 2a2 is C 2-6 alkynyl. In some embodiments, R 2a3 is C 2-6 alkynyl. In some embodiments, R 2a4 is C 2-6 alkynyl. In some embodiments, the C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkynyl is substituted with halo. In some embodiments, the C 2-6 alkynyl is substituted with cycloalkyl. In some embodiments, R 2a1 , R 2a2 , R 2a3 , or R 2a4 is [0130] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is cycloalkyl. In some embodiments, R 2a1 is cycloalkyl. In some embodiments, R 2a2 is cycloalkyl. In some embodiments, R 2a3 is cycloalkyl. In some embodiments, R 2a4 is cycloalkyl. In some embodiments, the cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the cycloalkyl is substituted with halo. In some embodiments, R 2a1 , R 2a2 , R 2a3 , or R 2a4 is . [0131] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is heterocyclyl. In some embodiments, R 2a1 is heterocyclyl. In some embodiments, R 2a2 is heterocyclyl. In some embodiments, R 2a3 is heterocyclyl. In some embodiments, R 2a4 is heterocyclyl. In some embodiments, the heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the heterocyclyl is substituted with halo. In some embodiments, the heterocyclyl is substituted with -OH. In some embodiments, R 2a1 , R 2a2 , R 2a3 , or R 2a4 is , [0132] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is aryl or heteroaryl. In some embodiments, R 2a1 is aryl or heteroaryl. In some embodiments, R 2a2 is aryl or heteroaryl. In some embodiments, R 2a3 is aryl or heteroaryl. In some embodiments, R 2a4 is aryl or heteroaryl. In some embodiments, the aryl or heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, R 2a1 , R 2a2 , R 2a3 , or R 2a4 is . [0133] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is -O-C 1-6 alkyl. In some embodiments, R 2a1 is -O-C 1-6 alkyl. In some embodiments, R 2a2 is -O-C 1-6 alkyl. In some embodiments, R 2a3 is -O-C 1- 6 alkyl. In some embodiments, R 2a4 is -O-C 1-6 alkyl. In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, - OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 1-6 alkyl is substituted with halo. In some embodiments, the -O-C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, R 2a1 , R 2a2 , R 2a3 , or R 2a4 is -OCH 3 , -OCHF 2 , -OCH 2 CF 3 , [0134] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is -O-C 2-6 alkenyl. In some embodiments, R 2a1 is -O-C 2-6 alkenyl. In some embodiments, R 2a2 is -O-C 2-6 alkenyl. In some embodiments, R 2a3 is -O-C 2- 6 alkenyl. In some embodiments, R 2a4 is -O-C 2-6 alkenyl. In some embodiments, the -O-C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 2-6 alkenyl is substituted with halo. [0135] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is -O-C 2-6 alkynyl. In some embodiments, R 2a1 is -O-C 2-6 alkynyl. In some embodiments, R 2a2 is -O-C 2-6 alkynyl. In some embodiments, R 2a3 is -O-C 2- 6 alkynyl. In some embodiments, R 2a4 is -O-C 2-6 alkynyl. In some embodiments, the -O-C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 2-6 alkynyl is substituted with halo. In some embodiments, R 2a1 , R 2a2 , R 2a3 , [0136] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is -O-cycloalkyl. In some embodiments, R 2a1 is -O-cycloalkyl. In some embodiments, R 2a2 is -O-cycloalkyl. In some embodiments, R 2a3 is -O- cycloalkyl. In some embodiments, R 2a4 is cycloalkyl. In some embodiments, the -O-cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-cycloalkyl is substituted with halo. In some embodiments, the -O- cycloalkyl is substituted with C 1-6 haloalkyl. In some embodiments, R 2a1 , R 2a2 , R 2a3 , or R 2a4 is , [0137] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is -O-heterocyclyl. In some embodiments, R 2a1 is -O-heterocyclyl. In some embodiments, R 2a2 is -O-heterocyclyl. In some embodiments, R 2a3 is -O- heterocyclyl. In some embodiments, R 2a4 is -O-heterocyclyl. In some embodiments, the -O-heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heterocyclyl is substituted with halo. [0138] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is -O-aryl. In some embodiments, R 2a1 is - O-aryl. In some embodiments, R 2a2 is -O-aryl. In some embodiments, R 2a3 is -O-aryl. In some embodiments, R 2a4 is -O-aryl. In some embodiments, the -O-aryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-aryl is substituted with halo. In some embodiments, the -O-aryl is substituted with C 1-6 alkyl. In some embodiments, the -O-aryl is substituted with -C(O)NH 2 . In some embodiments, the aryl of is a 6 membered monocyclic aryl. In some embodiments, R 2a3 is -O-aryl wherein the aryl is an unsubstituted 6- membered monocyclic aryl. In some embodiments, R 2a1 , R 2a2 , R 2a3 , or R 2a4 is , [0139] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is -O-heteroaryl. In some embodiments, R 2a1 is -O-heteroaryl. In some embodiments, R 2a2 is -O-heteroaryl. In some embodiments, R 2a3 is -O- heteroaryl. In some embodiments, R 2a4 is -O-heteroaryl. In some embodiments, the -O-heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heteroaryl is substituted with halo. In some embodiments, R 2a1 , R 2a2 , R 2a3 , . [0140] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is -SO 2 -cycloalkyl. In some embodiments, R 2a1 is -SO 2 -cycloalkyl. In some embodiments, R 2a2 is -SO 2 -cycloalkyl. In some embodiments, R 2a3 is -SO2-cycloalkyl. In some embodiments, R 2a4 is -SO2-cycloalkyl. In some embodiments, the -SO 2 -cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -SO 2 -cycloalkyl is substituted with halo. In some embodiments, [0141] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is -NR 2L1 R 2L2 . In some embodiments, R 2a1 is -NR 2L1 R 2L2 . In some embodiments, R 2a2 is -NR 2L1 R 2L2 . In some embodiments, R 2a3 is -NR 2L1 R 2L2 . In embodiments, R 2a4 is -NR 2L1 R 2L2 . In some embodiments, R 2a some 1 , R 2a2 , R 2a3 , or R 2a4 is , , [0142] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, R 2L1 and R 2L2 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 2L1 is H. In some embodiments, R 2L1 is H and R 2L2 is C1-6alkyl, C2-6alkynyl, or cycloalkyl. In some embodiments, R 2L1 is -CH 3 . In some embodiments, R 2L1 is -CH 3 and R 2L2 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 2L1 is H or -CH 3 and R 2L2 is C 1-6 alkyl. In some embodiments, R 2L1 is H or -CH 3 and R 2L2 is C 2-6 alkynyl. In some embodiments, R 2L1 is H or -CH 3 and R 2L2 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C1-6alkyl is substituted with cycloalkyl. [0143] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is C 2-6 alkynyl-NR 2L3 R 2L4 . In some embodiments, R 2a1 is C 2-6 alkynyl-NR 2L3 R 2L4 . In some embodiments, R 2a2 is C 2-6 alkynyl-NR 2L3 R 2L4 . In some embodiments, R 2a3 is C 2-6 alkynyl-NR 2L3 R 2L4 . In some embodiments, R 2a4 is C 2-6 alkynyl-NR 2L3 R 2L4 . [0144] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, R 2L3 and R 2L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 2L3 is H. In some embodiments, R 2L3 is H and R 2L4 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 2L3 is -CH 3 . In some embodiments, R 2L3 is -CH 3 and R 2L4 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 2L3 is C 1-6 alkyl and R 2L4 is C 1-6 alkyl. In some embodiments, R 2L3 is H or -CH 3 and R 2L4 is C 1-6 alkyl. In some embodiments, R 2L3 is H or -CH 3 and R 2L4 is C 2-6 alkynyl. In some embodiments, R 2L3 is H or -CH 3 and R 2L4 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, [0145] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, two or more of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is not H. In some embodiments, R 2a3 is not H and one of R 2a1 , R 2a2 , or R 2a4 is not H. In some embodiments, R 2a2 is F or Cl and R 2a3 is -O-C 1-6 alkyl or -O-cycloalkyl, wherein the -O-C 1-6 alkyl or -O-cycloalkyl is unsubstituted or substituted with halo or - CN. In some embodiments, R 2a1 is not H and R 2a2 is not H. In some embodiments, R 2a1 is F or Cl and R 2a2 is F or Cl. [0146] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, X 3 is -O-. In some embodiments, X 3 is -NR 2b1 -. In some embodiments, X 3 is -NH- . In some embodiments, X 3 is -N(CH 3 )-. In some embodiments, X 3 is -CR 2b2 R 2b3 -. In some embodiments, X 3 is -CH(CH 3 )-. In some embodiments, X 3 is -CD 2 -. In some embodiments, X 3 is -CH 2 -. [0147] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, some embodiments, one o 10 11 12 r more of X , X , and X is -N-. In some embodiments, X 10 is -N-. In some embodiments, X 11 is -N-. In some embodiments, X 12 is -N- . In some embodiments, X 10 is -CR 2c1 -. In some embodiments, X 11 is -CR 2c2 -. In some embodiments, X 12 is -CR 2c3 -. In some embodiments, X 10 is -CR 2c1 -, X 11 is -CR 2c2 -, and X 12 is -CR 2c3 -. [0148] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 2c1 , R 2c2 , R 2c3 , and R 2c4 is not H. In some embodiments, one or more of R 2c1 , R 2c2 , R 2c3 , and R 2c4 is -CN. In some embodiments, one or more of R 2c1 , R 2c2 , R 2c3 , and R 2c4 is independently Cl, F, or Br. In some embodiments, two or more of R 2c1 , R 2c2 , R 2c3 , and R 2c4 are independently Cl, F, or Br. In some embodiments, R 2c2 is F and R 2c1 , R 2c3 , and R 2c4 are H. In some embodiments, R 2c1 , R 2c2 , R 2c3 , and R 2c4 are H. [0149] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically [0150] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, B4, B5, or B6 is a 3-membered monocyclic heterocyclediyl, a 4-membered monocyclic heterocyclediyl, a 5-membered monocyclic heterocyclediyl comprising 2 or more N, a 6- membered monocyclic heterocyclediyl comprising 2 or more N, a 7-membered monocyclic heterocyclediyl, an 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl, wherein the 3-membered monocyclic heterocyclediyl, 4-membered monocyclic heterocyclediyl, 5-membered monocyclic heterocyclediyl, 6- membered monocyclic heterocyclediyl, 7-membered monocyclic heterocyclediyl, 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B4, B5, or B6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18-membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered polycyclic heterocyclediyl. In some embodiments, the 7 to 18-membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered spirocyclic heterocyclediyl. [0151] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, B4, B5, or B6 is a 3 to 8-membered monocyclic heterocyclediyl, wherein the 3 to 8-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 3 to 8- membered monocyclic heterocyclediyl is a 3, 4, 5, 6, 7, or 8-membered monocyclic heterocyclediyl. In some embodiments, B4, B5, or B6 is a 4-membered monocyclic heterocyclediyl, wherein the 4-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B4, B5, or B6 is or . In some embodiments, B4, B5, or B6 is a 6-membered monocyclic heterocyclediyl, wherein the 6-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B4, B5, or B6 is . In some embodiments, B4, B5, or B6 is a 7-membered monocyclic heterocyclediyl, wherein the 7-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B4, B5, or B6 is . [0152] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, B4, B5, or B6 is a 7 to 18-membered polycyclic heterocyclediyl, wherein the 7 to 18-membered polycyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18- membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered polycyclic heterocyclediyl. In some embodiments, B4, B5, or B6 is some embodiments, B4, B5, or B6 is . In some embodiments B4, B5, or B6 is or e [0153] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, B4, B5, or B6 is a 7 to 18-membered spirocyclic heterocyclediyl, wherein the 7 to 18-membered spirocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18- membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered spirocyclic heterocyclediyl. In some embodiments, [0154] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B4, B5, or B6 comprises one or more N. In some embodiments, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B4, B5, or B6 comprises two or more N. [0155] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is C 1-6 alkyl, wherein the C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D10 ) 2 . In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -OH. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -O-C 1-6 alkyl. [0156] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is cycloalkyl, wherein the cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D10 ) 2 . In some embodiments, the cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D4, D5, D6, or D7 is . [0157] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is aryl, wherein the aryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D10 ) 2 . In some embodiments, the aryl is a monocyclic 6- membered aryl. In some embodiments, the aryl is substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments, the aryl is substituted with -CN. [0158] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is heteroaryl, wherein the heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0159] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is a monocyclic 5 or 6-membered heteroaryl comprising one or more N, wherein the monocyclic 5 or 6-membered heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, - OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, - C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CN. In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CF 3 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -OCH 3 . [0160] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is -O-C 1-6 alkyl, wherein the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D10 ) 2 . In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with halo. [0161] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O- heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1- 6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D10 ) 2 . In some embodiments, the -O-aryl or -O-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -O-aryl or -O-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0162] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is -C(O)-C 1-6 alkyl, wherein the -C(O)-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D10 ) 2 . In some embodiments, the - C(O)-C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the -C(O)-C 1-6 alkyl is unsubstituted or substituted with -CN. In some embodiments, D4, D5, D6, or D7 is or . [0163] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is -C(O)-cycloalkyl, wherein the -C(O)-cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D10 ) 2 . In some embodiments, the - C(O)-cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D4, D5, D6, or D7 is . [0164] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is -C(O)-heterocyclyl, wherein the -C(O)-heterocyclyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D10 ) 2 . In some embodiments, the - C(O)-heterocyclyl is unsubstituted or substituted with halo. In some embodiments, D4, D5, D6, or D7 is [0165] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is -C(O)-aryl or -C(O)-heteroaryl, wherein the -C(O)-aryl or - C(O)-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1- 6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D10 ) 2 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CF 3 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0166] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is -N(R 2D1 )(R 2D2 ). In some embodiments, D4, D5, D6, or D7 is [0167] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, R 2D1 is H. In some embodiments, R 2D1 is C 1-6 alkyl. In some embodiments, R 2D2 is aryl. In some embodiments, R 2D2 is heteroaryl. In some embodiments, the heteroaryl is a 6-membered heteroaryl. [0168] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is -C(O)N(R 2D3 )(R 2D4 ). In some embodiments, D4, D5, D6, or [0169] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, R 2D3 is H. In some embodiments, R 2D3 is C 1-6 alkyl. In some embodiments, R 2D4 is C 1-6 alkyl. In some embodiments, R 2D3 is H and R 2D4 is C 1-6 alkyl. [0170] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, D4, D5, D6, or D7 is -N(R 2D5 )C(O)R 2D6 . In some embodiments, D4, D5, D6, or [0171] In some embodiments of the compounds of Formula II and stereoisomers and pharmaceutically acceptable salts thereof, R 2D5 is H. In some embodiments, R 2D5 is C 1-6 alkyl. In some embodiments, R 2D6 is C 1-6 alkyl. In some embodiments, R 2D6 is cycloalkyl. In some embodiments, R 2D6 is heterocyclyl. In some embodiments, R 2D5 is H and R 2D6 is C 1-6 alkyl, cycloalkyl, or heterocyclyl. [0172] In some embodiments, the compound is of Formula III: and stereoisomers and pharmaceutically acceptable salts thereof, wherein: X 1 is -N- or -CR 3a2 -; X 2 is -N- or -CR 3a4 -; R 3a1 , R 3a2 , R 3a3 , and R 3a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 3L1 R 3L2 , or C 2-6 alkynyl-NR 3L3 R 3L4 ; wherein if X 1 is -CR 3a2 - and X 2 is -CR 3a4 -, then at least one of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 ; R 3L1 , R 3L2 , R 3L3 , and R 3L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 3L1 , R 3L2 , R 3L3 , and R 3L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 3b1 -, or -CR 3b2 R 3b3 -, wherein R 3b1 , R 3b2 , and R 3b3 are independently H or C 1-6 alkyl; R 3f1 is H or C1-6alkyl; X 4 is -O-, -NR 3f2 -, or -CR 3f3 R 3f4 -, wherein R 3f2 , R 3f3 , and R 3f4 are independently selected from H and C 1-6 alkyl; B3 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B3 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo; D3 is C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 3D1 )(R 3D2 ), -C(O)N(R 3D3 )(R 3D4 ), or -N(R 3D5 )C(O)R 3D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D3 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 3D10 ) 2 , wherein each R 3D10 is independently H or C 1- 6 alkyl; R 3D1 , R 3D3 , and R 3D5 are independently H or C 1-6 alkyl; R 3D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 3D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 3D11 ) 2 , wherein each R 3D11 is independently H or C 1-6 alkyl; and R 3D4 and R 3D6 are independently C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 3D4 and R 3D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 3D12 ) 2 , wherein each R 3D12 is independently H or C 1-6 alkyl. [0173] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, X 1 is -CR 3a2 -, In some embodiments, X 2 is -CR 3a4 -. In some embodiments, X 1 is - N-. In some embodiments, X 2 is -N-. In some embodiments, X 1 is -CR 3a2 - and X 2 is -CR 3a4 -. In some embodiments, X 1 is -CR 3a2 - and X 2 is -N-. In some embodiments, X 1 is -N- and X 2 is -CR 3a4 -. [0174] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is not H. In some embodiments, R 3a1 is not H. In some embodiments, R 3a2 is not H. In some embodiments, R 3a3 is not H. In some embodiments, R 3a4 is not H. [0175] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is halo. In some embodiments, R 3a1 is halo. In some embodiments, R 3a2 is halo. In some embodiments, R 3a3 is halo. In some embodiments, R 3a4 is halo. In some embodiments, the halo is F, Cl, or Br. In some embodiments, the halo is Cl. In some embodiments, the halo is F. In some embodiments, R 3a1 is F. In some embodiments, R 3a3 is F. In some embodiments, R 3a3 is F. [0176] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is -OH. In some embodiments, R 3a1 is - OH. In some embodiments, R 3a2 is -OH. In some embodiments, R 3a3 is -OH. In some embodiments, R 3a4 is -OH. [0177] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is C 1-6 alkyl. In some embodiments, R 3a1 is C 1-6 alkyl. In some embodiments, R 3a2 is C 1-6 alkyl. In some embodiments, R 3a3 is C 1-6 alkyl. In some embodiments, R 3a4 is C 1-6 alkyl. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, R 3a1 , R 3a2 , R 3a3 , or R 3a4 is -CH 3 , -CH 2 CH 3 , -CH 2 CF 3 , -CF 3 , - [0178] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is C 2-6 alkenyl. In some embodiments, R 3a1 is C 2-6 alkenyl. In some embodiments, R 3a2 is C 2-6 alkenyl. In some embodiments, R 3a3 is C 2-6 alkenyl. In some embodiments, R 3a4 is C 2-6 alkenyl. In some embodiments, the C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkenyl is substituted with halo. In some embodiments, R 3a1 , R 3a2 , R 3a3 , or R 3a4 is or . [0179] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is C 2-6 alkynyl. In some embodiments, R 3a1 is C 2-6 alkynyl. In some embodiments, R 3a2 is C 2-6 alkynyl. In some embodiments, R 3a3 is C 2-6 alkynyl. In some embodiments, R 3a4 is C 2-6 alkynyl. In some embodiments, the C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkynyl is substituted with halo. In some embodiments, the C 2-6 alkynyl is substituted with cycloalkyl. In some embodiments, R 3a1 , R 3a2 , R 3a3 , or R 3a4 is , [0180] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is cycloalkyl. In some embodiments, R 3a1 is cycloalkyl. In some embodiments, R 3a2 is cycloalkyl. In some embodiments, R 3a3 is cycloalkyl. In some embodiments, R 3a4 is cycloalkyl. In some embodiments, the cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the cycloalkyl is substituted with halo. In some embodiments, R 3a1 , R 3a2 , R 3a3 , or R 3a4 is . [0181] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is heterocyclyl. In some embodiments, R 3a1 is heterocyclyl. In some embodiments, R 3a2 is heterocyclyl. In some embodiments, R 3a3 is heterocyclyl. In some embodiments, R 3a4 is heterocyclyl. In some embodiments, the heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the heterocyclyl is substituted with halo. In some embodiments, the heterocyclyl is 3 substituted with -OH. In some embodiments, R a1 , R 3a2 , R 3a3 , or R 3a4 is , , [0182] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is aryl or heteroaryl. In some embodiments, R 3a1 is aryl or heteroaryl. In some embodiments, R 3a2 is aryl or heteroaryl. In some embodiments, R 3a3 is aryl or heteroaryl. In some embodiments, R 3a4 is aryl or heteroaryl. In some embodiments, the aryl or heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, R 3a1 , R 3a2 , R 3a3 , or R 3a4 is . [0183] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is -O-C 1-6 alkyl. In some embodiments, R 3a1 is -O-C 1-6 alkyl. In some embodiments, R 3a2 is -O-C 1-6 alkyl. In some embodiments, R 3a3 is -O-C 1- 6 alkyl. In some embodiments, R 3a4 is -O-C 1-6 alkyl. In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, - OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 1-6 alkyl is substituted with halo. In some embodiments, the -O-C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, R 3a1 , R 3a2 , R 3a3 , or R 3a4 is -OCH 3 , -OCHF 2 , -OCH 2 CF 3 , [0184] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is -O-C 2-6 alkenyl. In some embodiments, R 3a1 is -O-C 2-6 alkenyl. In some embodiments, R 3a2 is -O-C 2-6 alkenyl. In some embodiments, R 3a3 is -O-C 2- 6 alkenyl. In some embodiments, R 3a4 is -O-C 2-6 alkenyl. In some embodiments, the -O-C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 2-6 alkenyl is substituted with halo. [0185] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is -O-C 2-6 alkynyl. In some embodiments, R 3a1 is -O-C 2-6 alkynyl. In some embodiments, R 3a2 is -O-C 2-6 alkynyl. In some embodiments, R 3a3 is -O-C 2- 6 alkynyl. In some embodiments, R 3a4 is -O-C 2-6 alkynyl. In some embodiments, the -O-C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 2-6 alkynyl is substituted with halo. In some embodiments, R 3a1 , R 3a2 , R 3a3 , [0186] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is -O-cycloalkyl. In some embodiments, R 1a1 , R 2a1 , R 3a1 , R 4a1 , R 5a1 , R 6a1 , or R 7a1 is -O-cycloalkyl. In some embodiments, R 1a2 , R 2a2 , R 3a2 , R 4a2 , R 5a2 , R 6a2 , or R 7a2 is -O-cycloalkyl. In some embodiments, R 3a3 is -O-cycloalkyl. In some embodiments, R 3a4 is cycloalkyl. In some embodiments, the -O-cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-cycloalkyl is substituted with halo. In some embodiments, the -O-cycloalkyl is substituted with C 1-6 haloalkyl. In some embodiments, . [0187] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is -O-heterocyclyl. In some embodiments, R 3a1 is -O-heterocyclyl. In some embodiments, R 3a2 is -O-heterocyclyl. In some embodiments, R 3a3 is -O- heterocyclyl. In some embodiments, R 3a4 is -O-heterocyclyl. In some embodiments, the -O-heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heterocyclyl is substituted with halo. [0188] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is -O-aryl. In some embodiments, R 3a1 is - O-aryl. In some embodiments, R 3a2 is -O-aryl. In some embodiments, R 3a3 is -O-aryl. In some embodiments, R 3a4 is -O-aryl. In some embodiments, the -O-aryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-aryl is substituted with halo. In some embodiments, the -O-aryl is substituted with C 1-6 alkyl. In some embodiments, the -O-aryl is substituted with -C(O)NH 2 . In some embodiments, the aryl of is a 6 membered monocyclic aryl. In some embodiments, R 3a3 is -O-aryl wherein the aryl is an unsubstituted 6- membered monocyclic aryl. In some embodiments, R 3a , , , , [0189] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is -O-heteroaryl. In some embodiments, R 3a1 is -O-heteroaryl. In some embodiments, R 3a2 is -O-heteroaryl. In some embodiments, R 3a3 is -O- heteroaryl. In some embodiments, R 3a4 is -O-heteroaryl. In some embodiments, the -O-heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heteroaryl is substituted with halo. In some embodiments, R 3a1 , R 3a2 , R 3a3 , . [0190] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is -SO 2 -cycloalkyl. In some embodiments, R 3a1 is -SO 2 -cycloalkyl. In some embodiments, R 3a2 is -SO 2 -cycloalkyl. In some embodiments, R 3a3 is -SO 2 -cycloalkyl. In some embodiments, R 3a4 is -SO 2 -cycloalkyl. In some embodiments, the -SO 2 -cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -SO 2 -cycloalkyl is substituted with halo. In some embodiments, [0191] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is -NR 3L1 R 3L2 . In some embodiments, R 3a1 is -NR 3L1 R 3L2 . In some embodiments, R 3a2 is -NR 3L1 R 3L2 . In some embodiments, R 3a3 is -NR 3L1 R 3L2 . In some embodiments, R 3a4 is -NR 3L1 R 3L2 . In some embodiments, R 3a1 , R 3a2 , R 3a3 , or R 3a4 is , [0192] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, R 3L1 and R 3L2 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 3L1 is H. In some embodiments, R 3L1 is H and R 3L2 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 3L1 is -CH 3 . In some embodiments, R 3L1 is -CH 3 and R 3L2 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 3L1 is H or -CH 3 and R 3L2 is C 1-6 alkyl. In some embodiments, R 3L1 is H or -CH3 and R 3L2 is C2-6alkynyl. In some embodiments, R 3L1 is H or -CH3 and R 3L2 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C 1-6 alkyl is substituted with cycloalkyl. [0193] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is C2-6alkynyl-NR 3L3 R 3L4 . In some embodiments, R 3a1 is C 2-6 alkynyl-NR 3L3 R 3L4 . In some embodiments, R 3a2 is C 2-6 alkynyl-NR 3L3 R 3L4 . In some embodiments, R 3a3 is C 2-6 alkynyl-NR 3L3 R 3L4 . In some embodiments, R 3a4 is C 2-6 alkynyl-NR 3L3 R 3L4 . [0194] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, R 3L3 and R 3L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 3L3 is H. In some embodiments, R 3L3 is H and R 3L4 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 3L3 is -CH3. In some embodiments, R 3L3 is -CH3 and R 3L4 is C1-6alkyl, C2-6alkynyl, or cycloalkyl. In some embodiments, R 3L3 is C 1-6 alkyl and R 3L4 is C 1-6 alkyl. In some embodiments, R 3L3 is H or -CH 3 and R 3L4 is C 1-6 alkyl. In some embodiments, R 3L3 is H or -CH 3 and R 3L4 is C 2-6 alkynyl. In some embodiments, R 3L3 is H or -CH 3 and R 3L4 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, [0195] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, two or more of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is not H. In some embodiments, R 3a3 is not H and one of R 3a1 , R 3a2 , or R 3a4 is not H. In some embodiments, R 3a2 is F or Cl and R 3a3 is -O-C 1-6 alkyl or -O-cycloalkyl, wherein the -O-C 1-6 alkyl or -O-cycloalkyl is unsubstituted or substituted with halo or - CN. In some embodiments, R 3a1 is not H and R 3a2 is not H. In some embodiments, R 3a1 is F or Cl and R 3a2 is F or Cl. [0196] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, X 3 is -O-. In some embodiments, X 3 is -NR 3b1 -. In some embodiments, X 3 is -NH- . In some embodiments, X 3 is -N(CH 3 )-. In some embodiments, X 3 is -CR 3b2 R 3b3 . In some embodiments, X 3 is -CH(CH 3 )-. In some embodiments, X 3 is -CD 2 -. In some embodiments, X 3 is -CH 2 -. [0197] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, X 4 is -O-. In some embodiments, X 4 is -NR 3f2 -. In some embodiments, X 4 is -NH- . In some embodiments, X 4 is -N(CH 3 )-. In some embodiments, X 4 is -CR 3f3 R 3f4 -. In some embodiments, X 4 is -CH(CH 3 )-. In some embodiments, X 4 is -CH 2 -. [0198] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, B3 is a 3-membered monocyclic heterocyclediyl, a 4-membered monocyclic heterocyclediyl, a 5-membered monocyclic heterocyclediyl comprising 2 or more N, a 6-membered monocyclic heterocyclediyl comprising 2 or more N, a 7-membered monocyclic heterocyclediyl, an 8- membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18- membered spirocyclic heterocyclediyl, wherein the 3-membered monocyclic heterocyclediyl, 4-membered monocyclic heterocyclediyl, 5-membered monocyclic heterocyclediyl, 6-membered monocyclic heterocyclediyl, 7-membered monocyclic heterocyclediyl, 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B3 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18-membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered polycyclic heterocyclediyl. In some embodiments, the 7 to 18-membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered spirocyclic heterocyclediyl. [0199] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, B3 is a 3 to 8-membered monocyclic heterocyclediyl, wherein the 3 to 8- membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 3 to 8- membered monocyclic heterocyclediyl is a 3, 4, 5, 6, 7, or 8-membered monocyclic heterocyclediyl. In some embodiments, B3 is a 4-membered monocyclic heterocyclediyl, wherein the 4-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B3 is . In some embodiments, B3 is a 6-membered monocyclic heterocyclediyl, wherein the 6-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B3 is . In some embodiments, B3 is a 7-membered monocyclic heterocyclediyl, wherein the 7-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments . [0200] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, B3 is a 7 to 18-membered polycyclic heterocyclediyl, wherein the 7 to 18- membered polycyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18- membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered polycyclic heterocyclediyl. In some embodiments, B3 is . In some embodiments, B3 is [0201] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, B3 is a 7 to 18-membered spirocyclic heterocyclediyl, wherein the 7 to 18- membered spirocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18- membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered spirocyclic heterocyclediyl. In some embodiments, [0202] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B3 comprises one or more N. In some embodiments, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B3 comprises two or more N. [0203] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is C 1-6 alkyl, wherein the C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, - OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 3D10 ) 2 . In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -OH. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -O-C 1-6 alkyl. [0204] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is cycloalkyl, wherein the cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, - OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 3D10 ) 2 . In some embodiments, the cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D3 is . [0205] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is aryl, wherein the aryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O- C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 3D10 ) 2 . In some embodiments, the aryl is a monocyclic 6-membered aryl. In some embodiments, the aryl is substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments, the aryl is substituted with - CN. [0206] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is heteroaryl, wherein the heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, - OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, - C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0207] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is a monocyclic 5 or 6-membered heteroaryl comprising one or more N, wherein the monocyclic 5 or 6-membered heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O- C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CN. In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CF 3 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -OCH 3 . [0208] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is -O-C 1-6 alkyl, wherein the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 3D10 ) 2 . In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with halo. [0209] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 3D10 ) 2 . In some embodiments, the -O- aryl or -O-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -O-aryl or -O- heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0210] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is -C(O)-C 1-6 alkyl, wherein the -C(O)-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 3D10 ) 2 . In some embodiments, the -C(O)-C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the -C(O)-C 1-6 alkyl is unsubstituted or substituted with -CN. In some embodiments, D3 is or . [0211] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is -C(O)-cycloalkyl, wherein the -C(O)-cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 3D10 ) 2 . In some embodiments, the -C(O)-cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D3 is . [0212] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is -C(O)-heterocyclyl, wherein the -C(O)-heterocyclyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C1-6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 3D10 )2. In some embodiments, the -C(O)-heterocyclyl is unsubstituted or substituted with halo. In some embodiments, D3 is or . [0213] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is -C(O)-aryl or -C(O)-heteroaryl, wherein the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, - CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O- C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 3D10 ) 2 . In some embodiments, the - C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -C(O)- aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CF 3 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0214] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically [0215] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, R 3D1 is H. In some embodiments, R 3D1 is C 1-6 alkyl. In some embodiments, R 3D2 is aryl. In some embodiments, R 3D2 is heteroaryl. In some embodiments, the heteroaryl is a 6-membered heteroaryl. [0216] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically alts thereof, D3 is -C(O)N(R 3D3 )(R 3 acceptable s D4 ). In some embodiments, D3 is . [0217] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, R 3D3 is H. In some embodiments, R 3D3 is C 1-6 alkyl. In some embodiments, R 3D4 is C 1-6 alkyl. In some embodiments, R 3D3 is H and R 3D4 is C 1-6 alkyl. [0218] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, D3 is -N(R 3D5 )C(O)R 3D6 -. In some embodiments, D3 is , [0219] In some embodiments of the compounds of Formula III and stereoisomers and pharmaceutically acceptable salts thereof, R 3D5 is H. In some embodiments, R 3D5 is C 1-6 alkyl. In some embodiments, R 3D6 is C 1-6 alkyl. In some embodiments, R 3D6 is cycloalkyl. In some embodiments, R 3D6 is heterocyclyl. In some embodiments, R 3D5 is H and R 3D6 is C 1-6 alkyl, cycloalkyl, or heterocyclyl. [0220] In some embodiments, the compound is of Formula IV: and stereoisomers and pharmaceutically acceptable salts thereof, wherein: X 1 is -N- or -CR 4a2 -; X 2 is -N- or -CR 4a4 -; R 4a1 , R 4a2 , R 4a3 , and R 4a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 4L1 R 4L2 , or C 2-6 alkynyl-NR 4L3 R 4L4 ; wherein if X 1 is -CR 4a2 - and X 2 is -CR 4a4 -, then at least one of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2; R 4L1 , R 4L2 , R 4L3 , and R 4L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 4L1 , R 4L2 , R 4L3 , and R 4L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 4b1 -, or -CR 4b2 R 4b3 -, wherein R 4b1 , R 4b2 , and R 4b3 are independently H or C 1-6 alkyl; NH 2 , or -NHSO 2 CH 3 ; B4 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B4 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo; D4 is C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 4D1 )(R 4D2 ), -C(O)N(R 4D3 )(R 4D4 ), or -N(R 4D5 )C(O)R 4D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D4 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D10 ) 2 , wherein each R 4D10 is independently H or C 1- 6 alkyl; R 4D1 , R 4D3 , and R 4D5 are independently H or C 1-6 alkyl; R 4D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 4D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 4D11 ) 2 , wherein each R 4D11 is independently H or C 1-6 alkyl; and R 4D4 and R 4D6 are independently C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 4D4 and R 4D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D12 ) 2 , wherein each R 4D12 is independently H or C 1-6 alkyl; provided that 4a2 , then R is not F, if and R 4c2 is F, then R 4a2 is not F. [0221] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, X 1 is -CR 4a2 -. In some embodiments, X 2 is -CR 4a4 -. In some embodiments, X 1 is -N-. In some embodiments, X 2 is -N-. In some embodiments, X 1 is -CR 4a2 - and X 2 is - CR 4a4 -. In some embodiments, X 1 is -CR 4a2 -, and X 2 is -N-. In some embodiments, X 1 is -N- and X 2 is - CR 4a4 -. [0222] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, R 4a2 is H, Cl, Br, I, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 4L1 R 4L2 , or C 2-6 alkynyl-NR 4L3 R 4L4 , wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 -cycloalkyl of R 4a2 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . [0223] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, R 4a4 is H, F, Br, I, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 4L1 R 4L2 , or C 2-6 alkynyl-NR 4L3 R 4L4 , wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl -O-heteroaryl, or -SO 2 -cycloalkyl of R 4a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C1-6alkyl, C1-6haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2. [0224] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is not H. In some embodiments, R 4a1 is not H. In some embodiments, R 4a2 is not H. In some embodiments, R 4a3 is not H. In some embodiments, R 4a4 is not H. [0225] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is halo. In some embodiments, R 4a1 is halo. In some embodiments, R 4a2 is halo. In some embodiments, R 4a3 is halo. In some embodiments, R 4a4 is halo. In some embodiments, the halo is F, Cl, or Br. In some embodiments, the halo is Cl. In some embodiments, the halo is F. In some embodiments, R 4a1 is F. In some embodiments, R 4a3 is F. In some embodiments, R 4a3 is F. [0226] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is -OH. In some embodiments, R 4a1 is -OH. In some embodiments, R 4a2 is -OH. In some embodiments, R 4a3 is -OH. In some embodiments, R 4a4 is -OH. [0227] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is C 1-6 alkyl. In some embodiments, R 4a1 is C 1-6 alkyl. In some embodiments, R 4a2 is C 1-6 alkyl. In some embodiments, R 4a3 is C 1- 6 alkyl. In some embodiments, R 4a4 is C 1-6 alkyl. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, R 4a1 , R 4a2 , R 4a3 , or R 4a4 is -CH 3 , -CH 2 CH 3 , - . [0228] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is C 2-6 alkenyl. In some embodiments, R 4a1 is C 2-6 alkenyl. In some embodiments, R 4a2 is C 2-6 alkenyl. In some embodiments, R 4a3 is C 2-6 alkenyl. In some embodiments, R 4a4 is C 2-6 alkenyl. In some embodiments, the C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkenyl is substituted with halo. In some embodiments, R 4a1 , R 4a2 , R 4a3 , or [0229] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is C 2-6 alkynyl. In some embodiments, R 4a1 is C2-6alkynyl. In some embodiments, R 4a2 is C2-6alkynyl. In some embodiments, R 4a3 is C 2-6 alkynyl. In some embodiments, R 4a4 is C 2-6 alkynyl. In some embodiments, the C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkynyl is substituted with halo. In some embodiments, the C 2-6 alkynyl is substituted with cycloalkyl. In some embodiments, R 4a1 , R 4a2 , R 4a3 , or R 4a4 is , [0230] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is cycloalkyl. In some embodiments, R 4a1 is cycloalkyl. In some embodiments, R 4a2 is cycloalkyl. In some embodiments, R 4a3 is cycloalkyl. In some embodiments, R 4a4 is cycloalkyl. In some embodiments, the cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the cycloalkyl is substituted with halo. In some embodiments, R 4a1 , R 4a2 , R 4a3 , or [0231] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is heterocyclyl. In some embodiments, R 4a1 is heterocyclyl. In some embodiments, R 4a2 is heterocyclyl. In some embodiments, R 4a3 is heterocyclyl. In some embodiments, R 4a4 is heterocyclyl. In some embodiments, the heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the heterocyclyl is substituted with halo. In some embodiments, the heterocyclyl is substituted with -OH. In some embodiments, R 4a1 , R 4a2 , R 4a3 , or R 4a4 is , [0232] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 R 4a2 , R 4a3 , and R 4a4 is aryl or heteroaryl. In some embodiments, R 4a1 is aryl or heteroaryl. In some embodiments, R 4a2 is aryl or heteroaryl. In some embodiments, R 4a3 is aryl or heteroaryl. In some embodiments, R 4a4 is aryl or heteroaryl. In some embodiments, the aryl or heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, R 4a1 , R 4a2 , R 4a3 , or R 4a4 is . [0233] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is -O-C 1-6 alkyl. In some embodiments, R 4a1 is -O-C 1-6 alkyl. In some embodiments, R 4a2 is -O-C 1-6 alkyl. In some embodiments, R 4a3 is -O-C 1-6 alkyl. In some embodiments, R 4a4 is -O-C 1-6 alkyl. In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 1-6 alkyl is substituted with halo. In some embodiments, the -O-C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, R 4a1 , R 4a2 , R 4a3 , or R 4a4 is -OCH 3 , -OCHF 2 , -OCH 2 CF 3 , . [0234] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is -O-C 2-6 alkenyl. In some embodiments, R 4a1 is -O-C 2-6 alkenyl. In some embodiments, R 4a2 is -O-C 2-6 alkenyl. In some embodiments, R 4a3 is -O-C 2-6 alkenyl. In some embodiments, R 4a4 is -O-C 2-6 alkenyl. In some embodiments, the -O-C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 2-6 alkenyl is substituted with halo. [0235] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is -O-C 2-6 alkynyl. In some embodiments, R 4a1 is -O-C 2-6 alkynyl. In some embodiments, R 4a2 is -O-C 2-6 alkynyl. In some embodiments, R 4a3 is -O-C 2-6 alkynyl. In some embodiments, R 4a4 is -O-C 2-6 alkynyl. In some embodiments, the -O-C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH2. In some embodiments, the -O-C2-6alkynyl is substituted with halo. In some embodiments, [0236] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is -O-cycloalkyl. In some embodiments, R 4a1 is -O-cycloalkyl. In some embodiments, R 4a2 is -O-cycloalkyl. In some embodiments, R 4a3 is -O-cycloalkyl. In some embodiments, R 4a4 is cycloalkyl. In some embodiments, the - O-cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and - C(O)NH 2 . In some embodiments, the -O-cycloalkyl is substituted with halo. In some embodiments, the - O-cycloalkyl is substituted with C1-6haloalkyl. In some embodiments, R 4a1 , R 4a2 , R 4a3 , or R 4a4 is [0237] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is -O-heterocyclyl. In some embodiments, R 4a1 is -O-heterocyclyl. In some embodiments, R 4a2 is -O-heterocyclyl. In some embodiments, R 4a3 is -O-heterocyclyl. In some embodiments, R 4a4 is -O-heterocyclyl. In some embodiments, the -O-heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heterocyclyl is substituted with halo. [0238] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is -O-aryl. In some embodiments, R 4a1 is -O-aryl. In some embodiments, R 4a2 is -O-aryl. In some embodiments, R 4a3 is -O- aryl. In some embodiments, R 4a4 is -O-aryl. In some embodiments, the -O-aryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-aryl is substituted with halo. In some embodiments, the -O-aryl is substituted with C 1-6 alkyl. In some embodiments, the -O-aryl is substituted with -C(O)NH 2 . In some embodiments, the aryl of is a 6 membered monocyclic aryl. In some embodiments, R 4a3 is -O-aryl wherein the aryl is an unsubstituted 6- membered monocyclic aryl. In some embodiments, R 4a1 , R 4a2 , R 4a3 , or R 4a4 is , [0239] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is -O-heteroaryl. In some embodiments, R 4a1 is -O-heteroaryl. In some embodiments, R 4a2 is -O-heteroaryl. In some embodiments, R 4a3 is -O-heteroaryl. In some embodiments, R 4a4 is -O-heteroaryl. In some embodiments, the -O-heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heteroaryl is substituted with halo. In some embodiments, [0240] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is -SO 2 -cycloalkyl. In some embodiments, R 4a1 is -SO 2 -cycloalkyl. In some embodiments, R 4a2 is -SO 2 -cycloalkyl. In some embodiments, R 4a3 is -SO 2 -cycloalkyl. In some embodiments, R 4a4 is -SO 2 -cycloalkyl. In some embodiments, the -SO 2 -cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -SO 2 -cycloalkyl is substituted with halo. In some embodiments, [0241] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is -NR 4L1 R 4L2 . In some embodiments, R 4a1 is -NR 4L1 R 4L2 . In some embodiments, R 4a2 is -NR 4L1 R 4L2 . In some embodiments, R 4a3 is -NR 4L1 R 4L2 . In some embodiments, R 4a4 is -NR 4L1 R 4L2 . In some embodiments, R 4a1 , R 4a2 , R 4a3 , or R 4a4 is [0242] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, R 4L1 and R 4L2 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 4L1 is H. In some embodiments, R 4L1 is H and R 4L2 is C 1-6 alkyl, C 2- 6 alkynyl, or cycloalkyl. In some embodiments, R 4L1 is -CH 3 . In some embodiments, R 4L1 is -CH 3 and R 4L2 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 4L1 is H or -CH 3 and R 4L2 is C 1-6 alkyl. In some embodiments, R 4L1 is H or -CH 3 and R 4L2 is C 2-6 alkynyl. In some embodiments, R 4L1 is H or -CH 3 and R 4L2 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1- 6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C 1-6 alkyl is substituted with cycloalkyl. [0243] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is C 2-6 alkynyl- NR 4L3 R 4L4 . In some embodiments, R 4a1 is C 2-6 alkynyl-NR 4L3 R 4L4 . In some embodiments, R 4a2 is C 2- 6 alkynyl-NR 4L3 R 4L4 . In some embodiments, R 4a3 is C 2-6 alkynyl-NR 4L3 R 4L4 . In some embodiments, R 4a4 is C 2-6 alkynyl-NR 4L3 R 4L4 . [0244] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, R 4L3 and R 4L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 4L3 is H. In some embodiments, R 4L3 is H and R 4L4 is C 1-6 alkyl, C 2- 6 alkynyl, or cycloalkyl. In some embodiments, R 4L3 is -CH 3 . In some embodiments, R 4L3 is -CH 3 and R 4L4 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 4L3 is C 1-6 alkyl and R 4L4 is C 1-6 alkyl. In some embodiments, R 4L3 is H or -CH 3 and R 4L4 is C 1-6 alkyl. In some embodiments, R 4L3 is H or -CH 3 and R 4L4 is C 2-6 alkynyl. In some embodiments, R 4L3 is H or -CH 3 and R 4L4 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1-6alkyl, C1-6haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, R 4a1 , R 4a2 , R 4a3 , or R 4a4 is . [0245] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, two or more of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is not H. In some embodiments, R 4a3 is not H and one of R 4a1 , R 4a2 , or R 4a4 is not H. In some embodiments, R 4a2 is F or Cl and R 4a3 is -O-C 1-6 alkyl or -O-cycloalkyl, wherein the -O-C 1-6 alkyl or -O-cycloalkyl is unsubstituted or substituted with halo or -CN. In some embodiments, R 4a1 is not H and R 4a2 is not H. In some embodiments, R 4a1 is F or Cl and R 4a2 is F or Cl. [0246] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, X 3 is -O-. In some embodiments, X 3 is -NR 4b1 -. In some embodiments, X 3 is -NH-. In some embodiments, X 3 is -N(CH 3 )-. In some embodiments, X 3 is -CR 4b2 R 4b3 - . In some embodiments, X 3 is -CH(CH 3 )-. In some embodiments, X 3 is -CD 2 -. In some embodiments, X 3 is -CH 2 -. [0247] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, some embodiments, one or more of X 10 , X 11 , and X 12 is -N-. In some embodiments, X 10 is -N-. In some embodiments, X 11 is -N-. In some embodiments, X 12 is -N-. In some embodiments, X 10 is -CR 4c1 -. In some embodiments, X 11 is -CR 4c2 -. In some embodiments, X 12 is -CR 4c3 -. In some embodiments, X 10 is -CR 4c1 -, X 11 is -CR 4c2 -, and X 12 is -CR 4c3 - . [0248] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 4c1 , R 4c2 , R 4c3 , and R 4c4 is not H. In some embodiments, one or more of R 4c1 , R 4c2 , R 4c3 , and R 4c4 is -CN. In some embodiments, one or more of R 4c1 , R 4c2 , R 4c3 , and R 4c4 is independently Cl, F, or Br. In some embodiments, two or more of R 4c1 , R 4c2 , R 4c3 , and R 4c4 are independently Cl, F, or Br. In some embodiments, R 4c2 is F and R 1c1 , R 1c3 , and R 4c1 , R 4c3 , and R 4c4 are H. In some embodiments, R 4c1 , R 4c2 , R 4c3 , and R 4c4 are H. [0249] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, some embodiments, A4 is [0250] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, B4 is a 3-membered monocyclic heterocyclediyl, a 4-membered monocyclic heterocyclediyl, a 5-membered monocyclic heterocyclediyl comprising 2 or more N, a 6- membered monocyclic heterocyclediyl comprising 2 or more N, a 7-membered monocyclic heterocyclediyl, an 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl, wherein the 3-membered monocyclic heterocyclediyl, 4-membered monocyclic heterocyclediyl, 5-membered monocyclic heterocyclediyl, 6- membered monocyclic heterocyclediyl, 7-membered monocyclic heterocyclediyl, 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo C 1 6 alkyl, and oxo. In some embodiments, the 7 to 18-membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered polycyclic heterocyclediyl. In some embodiments, the 7 to 18-membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered spirocyclic heterocyclediyl. [0251] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, B4 is a 3 to 8-membered monocyclic heterocyclediyl, wherein the 3 to 8-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 3 to 8-membered monocyclic heterocyclediyl is a 3, 4, 5, 6, 7, or 8-membered monocyclic heterocyclediyl. In some embodiments, B4 is a 4-membered monocyclic heterocyclediyl, wherein the 4-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B4 is . In some embodiments, B4 is a 6-membered monocyclic heterocyclediyl, wherein the 6-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B4 is . In some embodiments, B4 is a 7-membered monocyclic heterocyclediyl, wherein the 7-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, . [0252] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, B4 is a 7 to 18-membered polycyclic heterocyclediyl, wherein the 7 to 18-membered polycyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18-membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered polycyclic heterocyclediyl. In some embodiments, B4 is . In some embodiments, B4 is

[0253] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, B4 is a 7 to 18-membered spirocyclic heterocyclediyl, wherein the 7 to 18-membered spirocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18-membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered [0254] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B4 comprises one or more N. In some embodiments, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B4 comprises two or more N. [0255] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is C 1-6 alkyl, wherein the C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D10 ) 2 . In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -OH. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -O-C 1-6 alkyl. [0256] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is cycloalkyl, wherein the cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C1-6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D10 )2. In some embodiments, the cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D4 is . [0257] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is aryl, wherein the aryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D10 ) 2 . In some embodiments, the aryl is a monocyclic 6- membered aryl. In some embodiments, the aryl is substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments, the aryl is substituted with -CN. [0258] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is heteroaryl, wherein the heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl,C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0259] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is a monocyclic 5 or 6-membered heteroaryl comprising one or more N, wherein the monocyclic 5 or 6-membered heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, - OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, - C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CN. In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CF 3 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -OCH 3 . [0260] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is -O-C 1-6 alkyl, wherein the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D10 ) 2 . In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with halo. [0261] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O- heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1- 6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D10 )2. In some embodiments, the -O-aryl or -O-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -O-aryl or -O-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0262] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is -C(O)-C 1-6 alkyl, wherein the -C(O)-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D10 ) 2 . In some embodiments, the - C(O)-C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the -C(O)-C 1-6 alkyl is unsubstituted or substituted with -CN. In some embodiments, D4 is or . [0263] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is -C(O)-cycloalkyl, wherein the -C(O)-cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D10 ) 2 . In some embodiments, the - C(O)-cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D4 is [0264] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is -C(O)-heterocyclyl, wherein the -C(O)-heterocyclyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D10 ) 2 . In some embodiments, the - C(O)-heterocyclyl is unsubstituted or substituted with halo. In some embodiments, D4 is or [0265] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is -C(O)-aryl or -C(O)-heteroaryl, wherein the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1- 6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D10 ) 2 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CF 3 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0266] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is -N(R 4D1 )(R 4D2 ). In some embodiments, D4 is [0267] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, R 4D1 is H. In some embodiments, R 4D1 is C 1-6 alkyl. In some embodiments, R 4D2 is aryl. In some embodiments, R 4D2 is heteroaryl. In some embodiments, the heteroaryl is a 6-membered heteroaryl. [0268] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is -C(O)N(R 4D3 )(R 4D4 ). In some embodiments, D4 is . [0269] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, R 4D3 is H. In some embodiments, R 4D3 is C 1-6 alkyl. In some embodiments, R 4D4 is C 1-6 alkyl. In some embodiments, R 4D3 is H and R 4D4 is C 1-6 alkyl. [0270] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, D4 is -N(R 4D5 )C(O)R 4D6 . In some embodiments, D4 is [0271] In some embodiments of the compounds of Formula IV and stereoisomers and pharmaceutically acceptable salts thereof, R 4D5 is H. In some embodiments, R 4D5 is C 1-6 alkyl. In some embodiments, R 4D6 is C 1-6 alkyl. In some embodiments, R 4D6 is cycloalkyl. In some embodiments, R 4D6 is heterocyclyl. In some embodiments, R 4D5 is H and R 4D6 is C 1-6 alkyl, cycloalkyl, or heterocyclyl. [0272] In some embodiments, the compound is of Formula V: and stereoisomers and pharmaceutically acceptable salts thereof, wherein: X 1 is -N- or -CR 5a2 -; X 2 is -N- or -CR 5a4 -; R 5a1 , R 5a2 , R 5a3 , and R 5a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 5L1 R 5L2 , or C 2-6 alkynyl-NR 5L3 R 5L4 ; wherein if X 1 is -CR 5a2 - and X 2 is -CR 5a4 -, then at least one of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 ; R 5L1 , R 5L2 , R 5L3 , and R 5L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 5L1 , R 5L2 , R 5L3 , and R 5L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 5b1 -, or -CR 5b2 R 5b3 -, wherein R 5b1 , R 5b2 , and R 5b3 are independently H or C 1-6 alkyl; A5 is R 5c1 , R 5c2 , R 5c3 , R 5c4 , R 5c5 , R 5c6 , R 5c7 , and R 5c8 are independently H, halo, -CN, -SO 2 CH 3 , -SO 2 NH 2 , or -NHSO 2 CH 3 ; R 5g1 is H or C 1-6 alkyl; B5 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B5 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo; D5 is C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 5D1 )(R 5D2 ), -C(O)N(R 5D3 )(R 5D4 ), or -N(R 5D5 )C(O)R 5D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D5 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 5D10 ) 2 , wherein each R 5D10 is independently H or C 1- 6alkyl; R 5D1 , R 5D3 , and R 5D5 are independently H or C 1-6 alkyl; R 5D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 5D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 5D11 ) 2 , wherein each R 5D11 is independently H or C 1-6 alkyl; and [0273] R 5D4 and R 5D6 are independently C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 5D4 and R 5D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 5D12 ) 2 , wherein each R 5D12 is independently H or C 1-6 alkyl.In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, X 1 is -CR 5a2 -. In some embodiments, X 2 is -CR 5a4 -. In some embodiments, X 1 is -N-. In some embodiments, X 2 is -N-. In some embodiments, X 1 is -CR 5a2 - and X 2 is -CR 5a4 -. In some embodiments, X 1 is -CR 5a2 - and X 2 is -N-. In some embodiments, X 1 is -N- and X 2 is -CR 5a4 -. [0274] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is not H. In some embodiments, R 5a1 is not H. In some embodiments, R 5a2 is not H. In some embodiments, R 5a3 is not H. In some embodiments, R 5a4 is not H. [0275] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is halo. In some embodiments, R 5a1 is halo. In some embodiments, R 5a2 is halo. In some embodiments, R 5a3 is halo. In some embodiments, R 5a4 is halo. In some embodiments, the halo is F, Cl, or Br. In some embodiments, the halo is Cl. In some embodiments, the halo is F. In some embodiments, R 5a1 is F. In some embodiments, R 5a3 is F. In some embodiments, R 5a3 is F. [0276] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is -OH. In some embodiments, R 5a1 is - OH. In some embodiments, R 5a2 is -OH. In some embodiments, R 5a3 is -OH. In some embodiments, R 5a4 is -OH. [0277] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is C 1-6 alkyl. In some embodiments, R 5a1 is C 1-6 alkyl. In some embodiments, R 5a2 is C 1-6 alkyl. In some embodiments, R 5a3 is C 1-6 alkyl. In some embodiments, R 5a4 is C 1-6 alkyl. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, R 5a1 , R 5a2 , R 5a3 , or R 5a4 is -CH 3 , -CH 2 CH 3 , -CH 2 CF 3 , -CF 3 , - [0278] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is C 2-6 alkenyl. In some embodiments, R 5a1 is C 2-6 alkenyl. In some embodiments, R 5a2 is C 2-6 alkenyl. In some embodiments, R 5a3 is C 2-6 alkenyl. In some embodiments, R 5a4 is C 2-6 alkenyl. In some embodiments, the C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkenyl is substituted with halo. In some embodiments, R 5a1 , R 5a2 , R 5a3 , or R 5a4 is or . [0279] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is C 2-6 alkynyl. In some embodiments, R 5a1 is C2-6alkynyl. In some embodiments, R 5a2 is C2-6alkynyl. In some embodiments, R 5a3 is C2-6alkynyl. In some embodiments, R 5a4 is C 2-6 alkynyl. In some embodiments, the C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkynyl is substituted with halo. In some embodiments, the C 2-6 alkynyl is substituted with cycloalkyl. In some embodiments, R 5a1 , R 5a2 , R 5a3 , or R 5a4 is , [0280] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is cycloalkyl. In some embodiments, R 5a1 is cycloalkyl. In some embodiments, R 5a2 is cycloalkyl. In some embodiments, R 5a3 is cycloalkyl. In some embodiments, R 5a4 is cycloalkyl. In some embodiments, the cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the cycloalkyl is substituted with halo. In some embodiments, R 5a1 , R 5a2 , R 5a3 , or R 5a4 is . [0281] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is heterocyclyl. In some embodiments, R 5a1 is heterocyclyl. In some embodiments, R 5a2 is heterocyclyl. In some embodiments, R 5a3 is heterocyclyl. In some embodiments, R 5a4 is heterocyclyl. In some embodiments, the heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the heterocyclyl is substituted with halo. In some embodiments, the heterocyclyl is substituted with -OH. In some embodiments, R 5a1 , R 5a2 , R 5a3 , or R 5a4 is , [0282] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is aryl or heteroaryl. In some embodiments, R 5a1 is aryl or heteroaryl. In some embodiments, R 5a2 is aryl or heteroaryl. In some embodiments, R 5a3 is aryl or heteroaryl. In some embodiments, R 5a4 is aryl or heteroaryl. In some embodiments, the aryl or heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, R 5a1 , R 5a2 , R 5a3 , or R 5a4 is . [0283] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is -O-C 1-6 alkyl. In some embodiments, R 5a1 is -O-C 1-6 alkyl. In some embodiments, R 5a2 is -O-C 1-6 alkyl. In some embodiments, R 5a3 is -O-C 1- 6 alkyl. In some embodiments, R 5a4 is -O-C 1-6 alkyl. In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, - OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 1-6 alkyl is substituted with halo. In some embodiments, the -O-C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, R 5a1 , R 5a2 , R 5a3 , or R 5a4 is -OCH 3 , -OCHF 2 , -OCH 2 CF 3 , . [0284] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is -O-C 2-6 alkenyl. In some embodiments, R 5a1 is -O-C 2-6 alkenyl. In some embodiments, R 5a2 is -O-C 2-6 alkenyl. In some embodiments, R 5a3 is -O-C 2- 6 alkenyl. In some embodiments, R 5a4 is -O-C 2-6 alkenyl. In some embodiments, the -O-C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 2-6 alkenyl is substituted with halo. [0285] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is -O-C 2-6 alkynyl. In some embodiments, R 5a1 is -O-C 2-6 alkynyl. In some embodiments, R 5a2 is -O-C 2-6 alkynyl. In some embodiments, R 5a3 is -O-C 2- 6 alkynyl. In some embodiments, R 5a4 is -O-C 2-6 alkynyl. In some embodiments, the -O-C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C2-6alkynyl is substituted with halo. In some embodiments, R 5a1 , R 5a2 , R 5a3 , [0286] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is -O-cycloalkyl. In some embodiments, R 5a1 is -O-cycloalkyl. In some embodiments, R 5a2 is -O-cycloalkyl. In some embodiments, R 5a3 is -O- cycloalkyl. In some embodiments, R 5a4 is cycloalkyl. In some embodiments, the -O-cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-cycloalkyl is substituted with halo. In some embodiments, the -O- cycloalkyl is substituted with C1-6haloalkyl. In some embodiments, R 5a1 , R 5a2 , R 5a3 , or R 5a4 is , [0287] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is -O-heterocyclyl. In some embodiments, R 5a1 is -O-heterocyclyl. In some embodiments, R 5a2 is -O-heterocyclyl. In some embodiments, R 5a3 is -O- heterocyclyl. In some embodiments, R 5a4 is -O-heterocyclyl. In some embodiments, the -O-heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heterocyclyl is substituted with halo. [0288] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is -O-aryl. In some embodiments, R 5a1 is - O-aryl. In some embodiments, R 5a2 is -O-aryl. In some embodiments, R 5a3 is -O-aryl. In some embodiments, R 5a4 is -O-aryl. In some embodiments, the -O-aryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-aryl is substituted with halo. In some embodiments, the -O-aryl is substituted with C 1-6 alkyl. In some embodiments, the -O-aryl is substituted with -C(O)NH 2 . In some embodiments, the aryl of is a 6 membered monocyclic aryl. In some embodiments, R 5a3 is -O-aryl wherein the aryl is an unsubstituted 6- membered monocyclic aryl. In some embodiments, R 5a1 , R 5a2 , R 5a3 , or R 5a4 is , [0289] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is -O-heteroaryl. In some embodiments, R 5a1 is -O-heteroaryl. In some embodiments, R 5a2 is -O-heteroaryl. In some embodiments, R 5a3 is -O- heteroaryl. In some embodiments, R 5a4 is -O-heteroaryl. In some embodiments, the -O-heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heteroaryl is substituted with halo. In some embodiments, R 5a1 , R 5a2 , R 5a3 , . [0290] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is -SO 2 -cycloalkyl. In some embodiments, R 5a1 is -SO 2 -cycloalkyl. In some embodiments, R 5a2 is -SO 2 -cycloalkyl. In some embodiments, R 5a3 is -SO 2 -cycloalkyl. In some embodiments, R 5a4 is -SO 2 -cycloalkyl. In some embodiments, the -SO 2 -cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -SO 2 -cycloalkyl is substituted with halo. In some embodiments, [0291] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is -NR 5L1 R 5L2 . In some embodiments, R 5a1 is -NR 5L1 R 5L2 . In some embodiments, R 5a2 is -NR 5L1 R 5L2 . In some embodiments, R 5a3 is -NR 5L1 R 5L2 . In 5 a4 5L1 5L2 In some embodiments, R 5a some embodiments, R is -NR R . 1 , R 5a2 , R 5a3 , or R 5a4 is , , [0292] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, R 5L1 and R 5L2 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 5L1 is H. In some embodiments, R 5L1 is H and R 5L2 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 5L1 is -CH 3 . In some embodiments, R 5L1 is -CH 3 and R 5L2 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 5L1 is H or -CH 3 and R 5L2 is C 1-6 alkyl. In some embodiments, R 5L1 is H or -CH 3 and R 5L2 is C 2-6 alkynyl. In some embodiments, R 5L1 is H or -CH 3 and R 5L2 is cycloalkyl. In some embodiments, the C1-6alkyl, C2-6alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C 1-6 alkyl is substituted with cycloalkyl. [0293] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is C 2-6 alkynyl-NR 5L3 R 5L4 . In some embodiments, R 5a1 is C 2-6 alkynyl-NR 5L3 R 5L4 . In some embodiments, R 5a2 is C 2-6 alkynyl-NR 5L3 R 5L4 . In some embodiments, R 5a3 is C 2-6 alkynyl-NR 5L3 R 5L4 . In some embodiments, R 5a4 is C 2-6 alkynyl-NR 5L3 R 5L4 . [0294] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, R 5L3 and R 5L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 5L3 is H. In some embodiments, R 5L3 is H and R 5L4 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 5L3 is -CH 3 . In some embodiments, R 5L3 is -CH 3 and R 5L4 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 5L3 is C 1-6 alkyl and R 5L4 is C 1-6 alkyl. In some embodiments, R 5L3 is H or -CH 3 and R 5L4 is C 1-6 alkyl. In some embodiments, R 5L3 is H or -CH 3 and R 5L4 is C 2-6 alkynyl. In some embodiments, R 5L3 is H or -CH 3 and R 5L4 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C1-6alkyl is substituted with halo. In some embodiments, the C1-6alkyl is substituted with cycloalkyl. In some embodiments, [0295] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, two or more of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is not H. In some embodiments, R 5a3 is not H and one of R 5a1 , R 5a2 , or R 5a4 is not H. In some embodiments, R 5a2 is F or Cl and R 5a3 is -O-C 1-6 alkyl or -O-cycloalkyl, wherein the -O-C 1-6 alkyl or -O-cycloalkyl is unsubstituted or substituted with halo or - CN. In some embodiments, R 5a1 is not H and R 5a2 is not H. In some embodiments, R 5a1 is F or Cl and R 5a2 is F or Cl. [0296] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, X 3 is -O-. In some embodiments, X 3 is -NR 5b1 . In some embodiments, X 3 is -NH-. In some embodiments, X 3 is -N(CH 3 )-. In some embodiments, X 3 is -CR 5b2 R 5b3 -. In some embodiments, X 3 is -CH(CH 3 )-. In some embodiments, X 3 is -CD 2 -. In some embodiments, X 3 is -CH 2 -. [0297] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, some embodiments, one or more of X 10 , X 11 , and X 12 is -N-. In some embodiments, X 10 is -N-. In some embodiments, X 11 is -N-. In some embodiments, X 12 is -N- . In some embodiments, X 10 is -CR 5c1 . In some embodiments, X 11 is -CR 5c2 -. In some embodiments, X 12 is -CR 5c3 -. In some embodiments, X 10 is -CR 5c1 -, X 11 is -CR 5c2 - and X 12 is -CR 5c3 . [0298] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 5c1 , R 5c2 , R 5c3 , and R 5c4 is not H. In some embodiments, one or more of R 5c1 , R 5c2 , R 5c3 , and R 5c4 is -CN. In some embodiments, one or more of R 5c1 , R 5c2 , R 5c3 , and R 5c4 is independently Cl, F, or Br. In some embodiments, two or more of R 5c1 , R 5c2 , R 5c3 , and R 5c4 are independently Cl, F, or Br. In some embodiments, R 5c2 is F and R 5c1 , R 5c3 , and R 5c4 are H. In some embodiments, R 5c1 , R 5c2 , R 5c3 , and R 5c4 are H. [0299] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically [0300] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, B5 is a 3-membered monocyclic heterocyclediyl, a 4-membered monocyclic heterocyclediyl, a 5-membered monocyclic heterocyclediyl comprising 2 or more N, a 6-membered monocyclic heterocyclediyl comprising 2 or more N, a 7-membered monocyclic heterocyclediyl, an 8- membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18- membered spirocyclic heterocyclediyl, wherein the 3-membered monocyclic heterocyclediyl, 4-membered monocyclic heterocyclediyl, 5-membered monocyclic heterocyclediyl, 6-membered monocyclic heterocyclediyl, 7-membered monocyclic heterocyclediyl, 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B5 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18-membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered polycyclic heterocyclediyl. In some embodiments, the 7 to 18-membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered spirocyclic heterocyclediyl. [0301] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, B5 is a 3 to 8-membered monocyclic heterocyclediyl, wherein the 3 to 8- membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 3 to 8- membered monocyclic heterocyclediyl is a 3, 4, 5, 6, 7, or 8-membered monocyclic heterocyclediyl. In some embodiments, B5 is a 4-membered monocyclic heterocyclediyl, wherein the 4-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the N 1 -6 . In some embodiments, B5 is 2B group consisting of halo, C alkyl, and oxo or . In some embodiments, B5 is a 6-membered monocyclic heterocyclediyl, wherein the 6-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B5 is . In some embodiments, B5 is a 7-membered monocyclic heterocyclediyl, wherein the 7-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, . [0302] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, B5 is a 7 to 18-membered polycyclic heterocyclediyl, wherein the 7 to 18- membered polycyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18- membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered . [0303] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, B5 is a 7 to 18-membered spirocyclic heterocyclediyl, wherein the 7 to 18- membered spirocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18- membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered spirocyclic heterocyclediyl. In some embodiments, [0304] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B5 comprises one or more N. In some embodiments, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B5 comprises two or more N. [0305] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, R 5g1 is H. In some embodiments, R 5g1 is C 1-6 alkyl. In some embodiments, R 5g1 is - CH 3 . [0306] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is C 1-6 alkyl, wherein the C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, - OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 5D10 ) 2 . In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -OH. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -O-C 1-6 alkyl. [0307] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is cycloalkyl, wherein the cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, - OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 5D10 ) 2 . In some embodiments, the cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D5 is . [0308] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is aryl, wherein the aryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O- C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 5D10 ) 2 . In some embodiments, the aryl is a monocyclic 6-membered aryl. In some embodiments, the aryl is substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments, the aryl is substituted with - CN. [0309] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is heteroaryl, wherein the heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, - OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, - C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0310] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is a monocyclic 5 or 6-membered heteroaryl comprising one or more N, wherein the monocyclic 5 or 6-membered heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O- C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CN. In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CF 3 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -OCH 3 . [0311] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is -O-C 1-6 alkyl, wherein the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 5D10 ) 2 . In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with halo. [0312] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 5D10 ) 2 . In some embodiments, the -O- aryl or -O-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -O-aryl or -O- heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0313] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is -C(O)-C 1-6 alkyl, wherein the -C(O)-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 5D10 ) 2 . In some embodiments, the -C(O)-C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the -C unsubstituted or substituted with -CN. In some embodiments, D5 is . [0314] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is -C(O)-cycloalkyl, wherein the -C(O)-cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 5D10 ) 2 . In some embodiments, the -C(O)-cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D5 is . [0315] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is -C(O)-heterocyclyl, wherein the -C(O)-heterocyclyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 5D10 ) 2 . In some embodiments, the -C(O)-heterocyclyl is unsubstituted or substituted with halo. In some embodiments, D5 is or . [0316] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is -C(O)-aryl or -C(O)-heteroaryl, wherein the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, - CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, C1-6alkyl-OH, -O-C1-6haloalkyl, C1-6alkyl-O- C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 5D10 ) 2 . In some embodiments, the - C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -C(O)- aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CF 3 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0317] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is -N(R 5D1 )(R 5D2 ). In some embodiments, D5 is , [0318] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, R 5D1 is H. In some embodiments, R 5D1 is C 1-6 alkyl. In some embodiments, R 5D2 is aryl. In some embodiments, R 5D2 is heteroaryl. In some embodiments, the heteroaryl is a 6-membered heteroaryl. [0319] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is -C(O)N(R 5D3 )(R 5D4 ). In some embodiments, D5 is . [0320] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, R 5D3 is H. In some embodiments, R 5D3 is C 1-6 alkyl. In some embodiments, R 5D4 is C 1-6 alkyl. In some embodiments, R 5D3 is H and R 5D4 is C 1-6 alkyl. [0321] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, D5 is -N(R 5D5 )C(O)R 5D6 -. In some embodiments, D5 is , [0322] In some embodiments of the compounds of Formula V and stereoisomers and pharmaceutically acceptable salts thereof, R 5D5 is H. In some embodiments, R 5D5 is C 1-6 alkyl. In some embodiments, R 5D6 is C 1-6 alkyl. In some embodiments, R 5D6 is cycloalkyl. In some embodiments, R 5D6 is heterocyclyl. In some embodiments, R 5D5 is H and R 5D6 is C 1-6 alkyl, cycloalkyl, or heterocyclyl. [0323] In some embodiments, the compound is of Formula VI: and stereoisomers and pharmaceutically acceptable salts thereof, wherein: X 1 is -N- or -CR 6a2 -; X 2 is -N- or -CR 6a4 -; R 6a1 , R 6a2 , R 6a3 , and R 6a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 6L1 R 6L2 , or C 2-6 alkynyl-NR 6L3 R 6L4 ; wherein if X 1 is -CR 6a2 - and X 2 is -CR 6a4 -, then at least one of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 ; R 6L1 , R 6L2 , R 6L3 , and R 6L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 6L1 , R 6L2 , R 6L3 , and R 6L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 6b1 -, or -CR 6b2 R 6b3 -, wherein R 6b1 , R 6b2 , and R 6b3 are independently H or C 1-6 alkyl; NH 2 , or -NHSO 2 CH 3 ; B6 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B6 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo; D6 is C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 6D1 )(R 6D2 ), -C(O)N(R 6D3 )(R 6D4 ), or -N(R 6D5 )C(O)R 6D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D6 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D10 ) 2 , wherein each R 6D10 is independently H or C 1- 6 alkyl; R 6D1 , R 6D3 , and R 6D5 are independently H or C 1-6 alkyl; R 6D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 6D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 6D11 ) 2 , wherein each R 6D11 is independently H or C 1-6 alkyl; and R 6D4 and R 6D6 are independently C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 6D4 and R 6D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D12 ) 2 , wherein each R 6D12 is independently H or C 1-6 alkyl. [0324] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, X 1 is -CR 6a2 -. In some embodiments, X 2 is -CR 6a4 -. In some embodiments, X 1 is -N-. In some embodiments, X 2 is -N-. In some embodiments, X 1 is -CR 6a2 - and X 2 is - CR 6a4 -. In some embodiments, X 1 is -CR 6a2 - and X 2 is -N-. In some embodiments, X 1 is -N- and X 2 is - CR 6a4 -. [0325] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is not H. In some embodiments, R 6a1 is not H. In some embodiments, R 6a2 is not H. In some embodiments, R 6a3 is not H. In some embodiments, R 6a4 is not H. [0326] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is halo. In some embodiments, R 6a1 is halo. In some embodiments, R 6a2 is halo. In some embodiments, R 6a3 is halo. In some embodiments, R 6a4 is halo. In some embodiments, the halo is F, Cl, or Br. In some embodiments, the halo is Cl. In some embodiments, the halo is F. In some embodiments, R 6a1 is F. In some embodiments, R 6a3 is F. In some embodiments, R 6a3 is F. [0327] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is -OH. In some embodiments, R 6a1 is -OH. In some embodiments, R 6a2 is -OH. In some embodiments, R 6a3 is -OH. In some embodiments, R 6a4 is -OH. [0328] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is C 1-6 alkyl. In some embodiments, R 6a1 is C 1-6 alkyl. In some embodiments, R 6a2 is C 1-6 alkyl. In some embodiments, R 6a3 is C 1- 6 alkyl. In some embodiments, R 6a4 is C 1-6 alkyl. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, R 6a1 , R 6a2 , R 6a3 , or R 6a4 is -CH 3 , -CH 2 CH 3 , - [0329] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is C 1-6 alkyl. In some embodiments, R 7a1 is C 1-6 alkyl. In some embodiments, R 7a2 is C 1-6 alkyl. In some embodiments, R 7a3 is C 1- 6 alkyl. In some embodiments, R 7a4 is C 1-6 alkyl. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, R 7a1 , R 7a2 , R 7a3 , or R 7a4 is -CH 3 , -CH 2 CH 3 , - [0330] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is C 2-6 alkenyl. In some embodiments, R 6a1 is C 2-6 alkenyl. In some embodiments, R 6a2 is C 2-6 alkenyl. In some embodiments, R 6a3 is C 2-6 alkenyl. In some embodiments, R 6a4 is C 2-6 alkenyl. In some embodiments, the C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkenyl is substituted with halo. In some embodiments, R 6a1 , R 6a2 , R 6a3 , or [0331] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is C 2-6 alkynyl. In some embodiments, R 6a1 is C 2-6 alkynyl. In some embodiments, R 6a2 is C 2-6 alkynyl. In some embodiments, R 6a3 is C 2-6 alkynyl. In some embodiments, R 6a4 is C 2-6 alkynyl. In some embodiments, the C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkynyl is substituted with halo. In some embodiments, the C 2-6 alkynyl is substituted with cycloalkyl. In some embodiments, R 6a1 , R 6a2 , R 6a3 , or R 6a4 is , [0332] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is cycloalkyl. In some embodiments, R 6a1 is cycloalkyl. In some embodiments, R 6a2 is cycloalkyl. In some embodiments, R 6a3 is cycloalkyl. In some embodiments, R 6a4 is cycloalkyl. In some embodiments, the cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the cycloalkyl is substituted with halo. In some embodiments, R 6a1 , R 6a2 , R 6a3 , or [0333] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is heterocyclyl. In some embodiments, R 6a1 is heterocyclyl. In some embodiments, R 6a2 is heterocyclyl. In some embodiments, R 6a3 is heterocyclyl. In some embodiments, R 6a4 is heterocyclyl. In some embodiments, the heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the heterocyclyl is substituted with halo. In some embodiments, the heterocyclyl is substituted with -OH. In some embodiments, R 6a1 , R 6a2 , R 6a3 , or R 6a4 is , [0334] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is aryl or heteroaryl. In some embodiments, R 6a1 is aryl or heteroaryl. In some embodiments, R 6a2 is aryl or heteroaryl. In some embodiments, R 6a3 is aryl or heteroaryl. In some embodiments, R 6a4 is aryl or heteroaryl. In some embodiments, the aryl or heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, R 6a1 , R 6a2 , R 6a3 , or R 6a4 is . [0335] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is -O-C 1-6 alkyl. In some embodiments, R 6a1 is -O-C 1-6 alkyl. In some embodiments, R 6a2 is -O-C 1-6 alkyl. In some embodiments, R 6a3 is -O-C 1-6 alkyl. In some embodiments, R 6a4 is -O-C 1-6 alkyl. In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 1-6 alkyl is substituted with halo. In some embodiments, the -O-C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, R 6a1 , R 6a2 , R 6a3 , or R 6a4 is -OCH 3 , -OCHF 2 , -OCH 2 CF 3 , [0336] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is -O-C 2-6 alkenyl. In some embodiments, R 6a1 is -O-C 2-6 alkenyl. In some embodiments, R 6a2 is -O-C 2-6 alkenyl. In some embodiments, R 6a3 is -O-C 2-6 alkenyl. In some embodiments, R 6a4 is -O-C 2-6 alkenyl. In some embodiments, the -O-C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 2-6 alkenyl is substituted with halo. [0337] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is -O-C 2-6 alkynyl. In some embodiments, R 6a1 is -O-C 2-6 alkynyl. In some embodiments, R 6a2 is -O-C 2-6 alkynyl. In some embodiments, R 6a3 is -O-C 2-6 alkynyl. In some embodiments, R 6a4 is -O-C 2-6 alkynyl. In some embodiments, the -O-C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH2. In some embodiments, the -O-C2-6alkynyl is substituted with halo. In some embodiments, [0338] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is -O-cycloalkyl. In some embodiments, R 6a1 is -O-cycloalkyl. In some embodiments, R 6a2 is -O-cycloalkyl. In some embodiments, R 6a3 is -O-cycloalkyl. In some embodiments, R 6a4 is cycloalkyl. In some embodiments, the - O-cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and - C(O)NH 2 . In some embodiments, the -O-cycloalkyl is substituted with halo. In some embodiments, the - O-cycloalkyl is substituted with C 1-6 haloalkyl. In some embodiments, R 6a1 , R 6a2 , R 6a3 , or R 6a4 is [0339] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is -O-heterocyclyl. In some embodiments, R 6a1 is -O-heterocyclyl. In some embodiments, R 6a2 is -O-heterocyclyl. In some embodiments, R 6a3 is -O-heterocyclyl. In some embodiments, R 6a4 is -O-heterocyclyl. In some embodiments, the -O-heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heterocyclyl is substituted with halo. [0340] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is -O-aryl. In some embodiments, R 6a1 is -O-aryl. In some embodiments, R 6a2 is -O-aryl. In some embodiments, R 6a3 is -O- aryl. In some embodiments, R 6a4 is -O-aryl. In some embodiments, the -O-aryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-aryl is substituted with halo. In some embodiments, the -O-aryl is substituted with C1-6alkyl. In some embodiments, the -O-aryl is substituted with -C(O)NH 2 . In some embodiments, the aryl of is a 6 membered monocyclic aryl. In some embodiments, R 6a3 is -O-aryl wherein the aryl is an unsubstituted 6- membered monocyclic aryl. In some embodiments, R 6a1 , R 6a2 , R 6a3 , or R 6a4 is , [0341] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is -O-heteroaryl. In some embodiments, R 6a1 is -O-heteroaryl. In some embodiments, R 6a2 is -O-heteroaryl. In some embodiments, R 6a3 is -O-heteroaryl. In some embodiments, R 6a4 is -O-heteroaryl. In some embodiments, the -O-heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heteroaryl is substituted with halo. In some embodiments, [0342] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is -SO 2 -cycloalkyl. In some embodiments, R 6a1 is -SO 2 -cycloalkyl. In some embodiments, R 6a2 is -SO 2 -cycloalkyl. In some embodiments, R 6a3 is -SO 2 -cycloalkyl. In some embodiments, R 6a4 is -SO 2 -cycloalkyl. In some embodiments, the -SO 2 -cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -SO 2 -cycloalkyl is substituted with halo. In some embodiments, [0343] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is -NR 6L1 R 6L2 . In some embodiments, R 6a1 is -NR 6L1 R 6L2 . In some embodiments, R 6a2 is -NR 6L1 R 6L2 . In some embodiments, R 6a3 is -NR 6L1 R 6L2 . In some embodiments, R 6a4 is -NR 6L1 R 6L2 . In some embodiments, R 6a1 , R 6a2 , R 6a3 , or R 6a4 is . [0344] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, R 6L1 and R 6L2 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 6L1 is H. In some embodiments, R 6L1 is H and R 6L2 is C 1-6 alkyl, C 2- 6 alkynyl, or cycloalkyl. In some embodiments, R 6L1 is -CH 3 . In some embodiments, R 6L1 is -CH 3 and R 6L2 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 6L1 is H or -CH 3 and R 6L2 is C 1-6 alkyl. In some embodiments, R 6L1 is H or -CH 3 and R 6L2 is C 2-6 alkynyl. In some embodiments, R 6L1 is H or -CH 3 and R 6L2 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1- 6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C1-6alkyl is substituted with cycloalkyl. [0345] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is C 2-6 alkynyl- NR 6L3 R 6L4 . In some embodiments, R 6a1 is C 2-6 alkynyl-NR 6L3 R 6L4 . In some embodiments, R 6a2 is C 2- 6 alkynyl-NR 6L3 R 6L4 . In some embodiments, R 6a3 is C 2-6 alkynyl-NR 6L3 R 6L4 . In some embodiments, R 6a4 is C 2-6 alkynyl-NR 6L3 R 6L4 . [0346] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, R 6L3 and R 6L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 6L3 is H. In some embodiments, R 6L3 is H and R 6L4 is C 1-6 alkyl, C 2- 6 alkynyl, or cycloalkyl. In some embodiments, R 6L3 is -CH 3 . In some embodiments, R 6L3 is -CH 3 and R 6L4 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 6L3 is C 1-6 alkyl and R 6L4 is C 1-6 alkyl. In some embodiments, R 6L3 is H or -CH 3 and R 6L4 is C 1-6 alkyl. In some embodiments, R 6L3 is H or -CH 3 and R 6L4 is C 2-6 alkynyl. In some embodiments, R 6L3 is H or -CH 3 and R 6L4 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, R 6a1 , R 6a2 , R 6a3 , or R 6a4 is . [0347] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, two or more of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is not H. In some embodiments, R 6a3 is not H and one of R 6a1 , R 6a2 , or R 6a4 is not H. In some embodiments, R 6a2 is F or Cl and R 6a3 is -O-C 1-6 alkyl or -O-cycloalkyl, wherein the -O-C 1-6 alkyl or -O-cycloalkyl is unsubstituted or substituted with halo or -CN. In some embodiments, R 6a1 is not H and R 6a2 is not H. In some embodiments, R 6a1 is F or Cl and R 6a2 is F or Cl. [0348] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, X 3 is -O-. In some embodiments, X 3 is -NR 6b1 -. In some embodiments, X 3 is -NH-. In some embodiments, X 3 is -N(CH 3 )-. In some embodiments, X 3 is -CR 6b2 R 6b3 - . In some embodiments, X 3 is -CH(CH 3 )-. In some embodiments, X 3 is -CD 2 -. In some embodiments, X 3 is -CH 2 -. [0349] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, some embodiments, one or more of X 10 , X 11 , and X 12 is -N-. In some embodiments, X 10 is -N-. In some embodiments, X 11 is -N-. In some embodiments, X 12 is -N-. In some embodiments, X 10 is -CR 6c1 -. In some embodiments, X 11 is -CR 6c2 -. In some embodiments, X 12 is -CR 6c3 -. In some embodiments, X 10 is -CR 6c1 -, X 11 is -CR 6c2 -, and X 12 is -CR 6c3 - . [0350] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 6c1 , R 6c2 , R 6c3 , and R 6c4 is not H. In some embodiments, one or more of R 6c1 , R 6c2 , R 6c3 , and R 6c4 is -CN. In some embodiments, one or more of R 6c1 , R 6c2 , R 6c3 , and R 6c4 is independently Cl, F, or Br. In some embodiments, two or more of R 6c1 , R 6c2 , R 6c3 , and R 6c4 are independently Cl, F, or Br. In some embodiments, R 6c2 is F and R 6c1 , R 6c3 , and R 6c4 are H. In some embodiments, R 6c1 , R 6c2 , R 6c3 , and R 6c4 are H. [0351] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, some embodiments, A6 is [0352] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, B6 is a 3-membered monocyclic heterocyclediyl, a 4-membered monocyclic heterocyclediyl, a 5-membered monocyclic heterocyclediyl comprising 2 or more N, a 6- membered monocyclic heterocyclediyl comprising 2 or more N, a 7-membered monocyclic heterocyclediyl, an 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl, wherein the 3-membered monocyclic heterocyclediyl, 4-membered monocyclic heterocyclediyl, 5-membered monocyclic heterocyclediyl, 6- membered monocyclic heterocyclediyl, 7-membered monocyclic heterocyclediyl, 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18-membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered polycyclic heterocyclediyl. In some embodiments, the 7 to 18-membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered spirocyclic heterocyclediyl. [0353] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, B6 is a 3 to 8-membered monocyclic heterocyclediyl, wherein the 3 to 8-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 3 to 8-membered monocyclic heterocyclediyl is a 3, 4, 5, 6, 7, or 8-membered monocyclic heterocyclediyl. In some embodiments, B6 is a 4-membered monocyclic heterocyclediyl, wherein the 4-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the N group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B6 is 2B or . In some embodiments, B6 is a 6-membered monocyclic heterocyclediyl, wherein the 6-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, B6 is . In some embodiments, B6 is a 7-membered monocyclic heterocyclediyl, wherein the 7-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, . [0354] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, B6 is a 7 to 18-membered polycyclic heterocyclediyl, wherein the 7 to 18-membered polycyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18-membered polycyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered polycyclic heterocyclediyl. In some embodiments, B6 is . In some embodiments, B6 is [0355] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, B6 is a 7 to 18-membered spirocyclic heterocyclediyl, wherein the 7 to 18-membered spirocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. In some embodiments, the 7 to 18-membered spirocyclic heterocyclediyl is a 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18-membered spirocyclic heterocyclediyl. In some embodiments, [0356] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B6 comprises one or more N. In some embodiments, the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B6 comprises two or more N. [0357] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is C 1-6 alkyl, wherein the C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, C1-6alkyl-OH, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D10 ) 2 . In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -OH. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -O-C 1-6 alkyl. [0358] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is cycloalkyl, wherein the cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D10 ) 2 . In some embodiments, the cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D6 is . [0359] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is aryl, wherein the aryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D10 ) 2 . In some embodiments, the aryl is a monocyclic 6- membered aryl. In some embodiments, the aryl is substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments, the aryl is substituted with -CN. [0360] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is heteroaryl, wherein the heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0361] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is a monocyclic 5 or 6-membered heteroaryl comprising one or more N, wherein the monocyclic 5 or 6-membered heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, - OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, - C(O)NH2, -C(O)NHCH3, and -C(O)N(CH3)2. In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CN. In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CF 3 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -OCH 3 . [0362] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is -O-C 1-6 alkyl, wherein the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D10 ) 2 . In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with halo. [0363] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O- heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1- 6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D10 ) 2 . In some embodiments, the -O-aryl or -O-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -O-aryl or -O-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0364] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is -C(O)-C 1-6 alkyl, wherein the -C(O)-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D10 ) 2 . In some embodiments, the - C(O)-C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the - unsubstituted or substituted with -CN. In some embodiments, D6 is [0365] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is -C(O)-cycloalkyl, wherein the -C(O)-cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D10 ) 2 . In some embodiments, the - C(O)-cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D6 is . [0366] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is -C(O)-heterocyclyl, wherein the -C(O)-heterocyclyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D10 ) 2 . In some embodiments, the - C(O)-heterocyclyl is unsubstituted or substituted with halo. In some embodiments, D6 is or . [0367] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is -C(O)-aryl or -C(O)-heteroaryl, wherein the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1- 6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D10 ) 2 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CF 3 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0368] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is -N(R 6D1 )(R 6D2 ). In some embodiments, D6 is [0369] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, R 6D1 is H. In some embodiments, R 6D1 is C 1-6 alkyl. In some embodiments, R 6D2 is aryl. In some embodiments, R 6D2 is heteroaryl. In some embodiments, the heteroaryl is a 6-membered heteroaryl. [0370] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is -C(O)N(R 6D3 )(R 6D4 ). In some embodiments, D6 is . [0371] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, R 6D3 is H. In some embodiments, R 6D3 is C 1-6 alkyl. In some embodiments, R 6D4 is C1-6alkyl. In some embodiments, R 6D3 is H and R 6D4 is C1-6alkyl. [0372] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, D6 is -N(R 6D5 )C(O)R 6D6 -. In some embodiments, D6 is [0373] In some embodiments of the compounds of Formula VI and stereoisomers and pharmaceutically acceptable salts thereof, R 6D5 is H. In some embodiments, R 6D5 is C 1-6 alkyl. In some embodiments, R 6D6 is C 1-6 alkyl. In some embodiments, R 6D6 is cycloalkyl. In some embodiments, R 6D6 is heterocyclyl. In some embodiments, R 6D5 is H and R 6D6 is C 1-6 alkyl, cycloalkyl, or heterocyclyl. [0374] In some embodiments, the compound is of Formula VII: and stereoisomers and pharmaceutically acceptable salts thereof, wherein: X 1 is -N- or -CR 7a2 -; X 2 is -N- or -CR 7a4 -; R 7a1 , R 7a2 , R 7a3 , and R 7a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 7L1 R 7L2 , or C 2-6 alkynyl-NR 7L3 R 7L4 ; wherein if X 1 is -CR 7a2 - and X 2 is -CR 7a4 -, then at least one of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO2- cycloalkyl of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 ; R 7L1 , R 7L2 , R 7L3 , and R 7L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 7L1 , R 7L2 , R 7L3 , and R 7L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 7b1 -, or -CR 7b2 R 7b3 -, wherein R 7b1 , R 7b2 , and R 7b3 are independently H or C 1-6 alkyl; 2 , or -NHSO 2 CH 3 ; X 5 is -CH(R 7g3 )- or -CH(R 7g4 )CH 2 N(R 7g5 )-; R 7g2 , R 7g3 , R 7g4 , and R 7g5 are independently H or C 1-6 alkyl; D7 is C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 7D1 )(R 7D2 ), -C(O)N(R 7D3 )(R 7D4 ), or -N(R 7D5 )C(O)R 7D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D7 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D10 ) 2 , wherein each R 7D10 is independently H or C 1- 6 alkyl; R 7D1 , R 7D3 , and R 7D5 are independently H or C1-6alkyl; R 7D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 7D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 7D11 )2, wherein each R 7D11 is independently H or C1-6alkyl; and R 7D4 and R 7D6 are independently C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 7D4 and R 7D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D12 ) 2 , wherein each R 7D12 is independently H or C 1-6 alkyl. [0375] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, X 1 is -CR 7a2 -. In some embodiments, X 2 is -CR 7a4 -. In some embodiments, X 1 is -N-. In some embodiments, X 2 is -N-. In some embodiments, X 1 is -CR 7a2 - and X 2 is - CR 7a4 -. In some embodiments, X 1 is -CR 7a2 - and X 2 is -N-. In some embodiments, X 1 is -N- and X 2 is - CR 7a4 -. [0376] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 is not H. In some embodiments, R 7a2 is not H. In some embodiments, R 7a3 is not H. In some embodiments, R 7a4 is not H. [0377] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is halo. In some embodiments, R 7a1 is halo. In some embodiments, R 7a2 is halo. In some embodiments, R 7a3 is halo. In some embodiments, R 7a4 is halo. In some embodiments, the halo is F, Cl, or Br. In some embodiments, the halo is Cl. In some embodiments, the halo is F. In some embodiments, R 7a1 is F. In some embodiments, R 7a3 is F. In some embodiments, R 7a3 is F. [0378] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -OH. In some embodiments, R 7a1 is -OH. In some embodiments, R 7a2 is -OH. In some embodiments, R 7a3 is -OH. In some embodiments, R 7a4 is -OH. [0379] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is C 2-6 alkenyl. In some embodiments, R 7a1 is C 2-6 alkenyl. In some embodiments, R 7a2 is C 2-6 alkenyl. In some embodiments, R 7a3 is C 2-6 alkenyl. In some embodiments, R 7a4 is C 2-6 alkenyl. In some embodiments, the C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkenyl is substituted with halo. In some embodiments, R 7a1 , R 7a2 , R 7a3 , or [0380] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is C 2-6 alkynyl. In some embodiments, R 7a1 is C2-6alkynyl. In some embodiments, R 7a2 is C2-6alkynyl. In some embodiments, R 7a3 is C 2-6 alkynyl. In some embodiments, R 7a4 is C 2-6 alkynyl. In some embodiments, the C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the C 2-6 alkynyl is substituted with halo. In some embodiments, the C 2-6 alkynyl is substituted with cycloalkyl. In some embodiments, R 7a1 , R 7a2 , R 7a3 , or R 7a4 is , [0381] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is cycloalkyl. In some embodiments, R 7a1 is cycloalkyl. In some embodiments, R 7a2 is cycloalkyl. In some embodiments, R 7a3 is cycloalkyl. In some embodiments, R 7a4 is cycloalkyl. In some embodiments, the cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the cycloalkyl is substituted with halo. In some embodiments, R 7a1 , R 7a2 , R 7a3 , or [0382] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is heterocyclyl. In some embodiments, R 7a1 is heterocyclyl. In some embodiments, R 7a2 is heterocyclyl. In some embodiments, R 7a3 is heterocyclyl. In some embodiments, R 7a4 is heterocyclyl. In some embodiments, the heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the heterocyclyl is substituted with halo. In some embodiments, the heterocyclyl is substituted with -OH. In some embodiments, R 7a1 , R 7a2 , R 7a3 , or R 7a4 is , [0383] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 R 7a2 , R 7a3 , and R 7a4 is aryl or heteroaryl. In some embodiments, R 7a1 is aryl or heteroaryl. In some embodiments, R 7a2 is aryl or heteroaryl. In some embodiments, R 7a3 is aryl or heteroaryl. In some embodiments, R 7a4 is aryl or heteroaryl. In some embodiments, the aryl or heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, R 7a1 , R 7a2 , R 7a3 , or R 7a4 is . [0384] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -O-C 1-6 alkyl. In some embodiments, R 7a1 is -O-C 1-6 alkyl. In some embodiments, R 7a2 is -O-C 1-6 alkyl. In some embodiments, R 7a3 is -O-C 1-6 alkyl. In some embodiments, R 7a4 is -O-C 1-6 alkyl. In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 1-6 alkyl is substituted with halo. In some embodiments, the -O-C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, R 7a1 , R 7a2 , R 7a3 , or R 7a4 is -OCH 3 , -OCHF 2 , -OCH 2 CF 3 , . [0385] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -O-C 2-6 alkenyl. In some embodiments, R 7a1 is -O-C 2-6 alkenyl. In some embodiments, R 7a2 is -O-C 2-6 alkenyl. In some embodiments, R 7a3 is -O-C 2-6 alkenyl. In some embodiments, R 7a4 is -O-C 2-6 alkenyl. In some embodiments, the -O-C 2-6 alkenyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-C 2-6 alkenyl is substituted with halo. [0386] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -O-C 2-6 alkynyl. In some embodiments, R 7a1 is -O-C 2-6 alkynyl. In some embodiments, R 7a2 is -O-C 2-6 alkynyl. In some embodiments, R 7a3 is -O-C 2-6 alkynyl. In some embodiments, R 7a4 is -O-C 2-6 alkynyl. In some embodiments, the -O-C 2-6 alkynyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH2. In some embodiments, the -O-C2-6alkynyl is substituted with halo. In some embodiments, [0387] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -O-cycloalkyl. In some embodiments, R 7a1 is -O-cycloalkyl. In some embodiments, R 7a2 is -O-cycloalkyl. In some embodiments, R 7a3 is -O-cycloalkyl. In some embodiments, R 7a4 is cycloalkyl. In some embodiments, the - O-cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and - C(O)NH 2 . In some embodiments, the -O-cycloalkyl is substituted with halo. In some embodiments, the - O-cycloalkyl is substituted with C1-6haloalkyl. In some embodiments, R 7a1 , R 7a2 , R 7a3 , or R 7a4 is [0388] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -O-heterocyclyl. In some embodiments, R 7a1 is -O-heterocyclyl. In some embodiments, R 7a2 is -O-heterocyclyl. In some embodiments, R 7a3 is -O-heterocyclyl. In some embodiments, R 7a4 is -O-heterocyclyl. In some embodiments, the -O-heterocyclyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heterocyclyl is substituted with halo. [0389] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -O-aryl. In some embodiments, R 7a1 is -O-aryl. In some embodiments, R 7a2 is -O-aryl. In some embodiments, R 7a3 is -O- aryl. In some embodiments, R 7a4 is -O-aryl. In some embodiments, the -O-aryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-aryl is substituted with halo. In some embodiments, the -O-aryl is substituted with C 1-6 alkyl. In some embodiments, the -O-aryl is substituted with -C(O)NH 2 . In some embodiments, the aryl of is a 6 membered monocyclic aryl. In some embodiments, R 7a3 is -O-aryl wherein the aryl is an unsubstituted 6- membered monocyclic aryl. In some embodiments, R 7a1 , R 7a2 , R 7a3 , or R 7a4 is , [0390] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -O-heteroaryl. In some embodiments, R 7a1 is -O-heteroaryl. In some embodiments, R 7a2 is -O-heteroaryl. In some embodiments, R 7a3 is -O-heteroaryl. In some embodiments, R 7a4 is -O-heteroaryl. In some embodiments, the -O-heteroaryl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -O-heteroaryl is substituted with halo. In some embodiments, [0391] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -SO 2 -cycloalkyl. In some embodiments, R 7a1 is -SO 2 -cycloalkyl. In some embodiments, R 7a2 is -SO 2 -cycloalkyl. In some embodiments, R 7a3 is -SO 2 -cycloalkyl. In some embodiments, R 7a4 is -SO 2 -cycloalkyl. In some embodiments, the -SO 2 -cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . In some embodiments, the -SO 2 -cycloalkyl is substituted with halo. In some embodiments, [0392] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -NR 7L1 R 7L2 . In some embodiments, R 7a1 is -NR 7L1 R 7L2 . In some embodiments, R 7a2 is -NR 7L1 R 7L2 . In some embodiments, R 7a3 is -NR 7L1 R 7L2 . In some embodiments, R 7a4 is -NR 7L1 R 7L2 . In some embodiments, R 7a1 , R 7a2 , R 7a3 , or R 7a4 is [0393] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, R 7L1 and R 7L2 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 7L1 is H. In some embodiments, R 7L1 is H and R 7L2 is C 1-6 alkyl, C 2- 6 alkynyl, or cycloalkyl. In some embodiments, R 7L1 is -CH 3 . In some embodiments, R 7L1 is -CH 3 and R 7L2 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 7L1 is H or -CH 3 and R 7L2 is C 1-6 alkyl. In some embodiments, R 7L1 is H or -CH 3 and R 7L2 is C 2-6 alkynyl. In some embodiments, R 7L1 is H or -CH 3 and R 7L2 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1- 6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C 1-6 alkyl is substituted with cycloalkyl. [0394] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is C 2-6 alkynyl- NR 7L3 R 7L4 . In some embodiments, R 7a1 is C 2-6 alkynyl-NR 7L3 R 7L4 . In some embodiments, R 7a2 is C 2- 6 alkynyl-NR 7L3 R 7L4 . In some embodiments, R 7a3 is C 2-6 alkynyl-NR 7L3 R 7L4 . In some embodiments, R 7a4 is C 2-6 alkynyl-NR 7L3 R 7L4 . [0395] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, R 7L3 and R 7L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 7L3 is H. In some embodiments, R 7L3 is H and R 7L4 is C 1-6 alkyl, C 2- 6 alkynyl, or cycloalkyl. In some embodiments, R 7L3 is -CH 3 . In some embodiments, R 7L3 is -CH 3 and R 7L4 is C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl. In some embodiments, R 7L3 is C 1-6 alkyl and R 7L4 is C 1-6 alkyl. In some embodiments, R 7L3 is H or -CH 3 and R 7L4 is C 1-6 alkyl. In some embodiments, R 7L3 is H or -CH 3 and R 7L4 is C 2-6 alkynyl. In some embodiments, R 7L3 is H or -CH 3 and R 7L4 is cycloalkyl. In some embodiments, the C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1-6alkyl, C1-6haloalkyl, cycloalkyl, and halocycloalkyl. In some embodiments, the C 1-6 alkyl is substituted with halo. In some embodiments, the C 1-6 alkyl is substituted with cycloalkyl. In some embodiments, R 7a1 , R 7a2 , R 7a3 , or R 7a4 is . [0396] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, two or more of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is not H. In some embodiments, R 7a3 is not H and one of R 7a1 , R 7a2 , or R 7a4 is not H. In some embodiments, R 7a2 is F or Cl and R 7a3 is -O-C 1-6 alkyl or -O-cycloalkyl, wherein the -O-C 1-6 alkyl or -O-cycloalkyl is unsubstituted or substituted with halo or -CN. In some embodiments, R 7a1 is not H and R 7a2 is not H. In some embodiments, R 7a1 is F or Cl and R 7a2 is F or Cl. [0397] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, X 3 is -O-. In some embodiments, X 3 is -NR 7b1 -. In some embodiments, X 3 is -NH-. In some embodiments, X 3 is -N(CH 3 )-. In some embodiments, X 3 is -CR 7b2 R 7b3 - . In some embodiments, X 3 is -CH(CH 3 )-. In some embodiments, X 3 is -CD 2 -. In some embodiments, X 3 is -CH 2 -. [0398] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, some embodiments, one or more of X 10 , X 11 , and X 12 is -N-. In some embodiments, X 10 is -N-. In some embodiments, X 11 is -N-. In some embodiments, X is N . In some embodiments, X is CR . In some embodiments, X is CR . In some embodiments, X 12 is -CR 7c3 -. In some embodiments, X 10 is -CR 7c1 -, X 11 is -CR 7c2 -, and X 12 is -CR 7c3 - . [0399] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, one or more of R 7c1 , R 7c2 , R 7c3 , and R 7c4 is not H. In some embodiments, one or more of R 7c1 , R 7c2 , R 7c3 , and R 7c4 is -CN. In some embodiments, one or more of R 7c1 , R 7c2 , R 7c3 , and R 7c4 is independently Cl, F, or Br. In some embodiments, two or more of R 7c1 , R 7c2 , R 7c3 , and R 7c4 are independently Cl, F, or Br. In some embodiments, R 7c2 is F and R 7c1 , R 7c3 , and R 7c4 are H. In some embodiments, R 7c1 , R 7c2 , R 7c3 , and R 7c4 are H. [0400] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, some embodiments, A7 is [0401] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, R 7g2 is H. In some embodiments, R 7g2 is C 1-6 alkyl. In some embodiments, R 7g2 is -CH 3 . In some embodiments, R 7g2 is -CH 2 CH 3 . [0402] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, X 5 is -CH(R 7g3 )-, wherein R 7g3 is H or C 1-6 alkyl. In some embodiments, X 5 is -CH 2 -. In some embodiments, X 5 is -CH(CH 3 )-. [0403] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, X 5 is -CH(R 7g4 )CH 2 N(R 7g5 )-, wherein R 7g4 and R 7g5 are independently H or C 1-6 alkyl. In some embodiments, R 7g4 is H and R 7g5 is C 1-6 alkyl. In some embodiments, R 7g5 is -CH 3 . In some embodiments, R 7g5 is -CH 2 CH 3 . In some embodiments, R 7g4 is H and R 7g5 is H. [0404] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is C 1-6 alkyl, wherein the C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D10 ) 2 . In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -OH. In some embodiments, the C 1-6 alkyl is unsubstituted or substituted with -O-C 1-6 alkyl. [0405] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is cycloalkyl, wherein the cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D10 ) 2 . In some embodiments, the cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D7 is . [0406] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is aryl, wherein the aryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D10 ) 2 . In some embodiments, the aryl is a monocyclic 6- membered aryl. In some embodiments, the aryl is substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments, the aryl is substituted with -CN. [0407] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is heteroaryl, wherein the heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0408] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is a monocyclic 5 or 6-membered heteroaryl comprising one or more N, wherein the monocyclic 5 or 6-membered heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, - OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, - C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CN. In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -CF 3 . In some embodiments, the monocyclic 5 or 6-membered heteroaryl is substituted with -OCH 3 . [0409] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is -O-C 1-6 alkyl, wherein the -O-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D10 ) 2 . In some embodiments, the -O-C 1-6 alkyl is unsubstituted or substituted with halo. [0410] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O- heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1- 6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D10 ) 2 . In some embodiments, the -O-aryl or -O-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -O-aryl or -O-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0411] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is -C(O)-C 1-6 alkyl, wherein the -C(O)-C 1-6 alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D10 ) 2 . In some embodiments, the - C(O)-C 1-6 alkyl is unsubstituted or substituted with halo. In some embodiments, the -C(O)-C 1-6 alkyl is unsubstituted or substituted with -CN. In some embodiments, D7 is or . [0412] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is -C(O)-cycloalkyl, wherein the -C(O)-cycloalkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D10 ) 2 . In some embodiments, the - C(O)-cycloalkyl is unsubstituted or substituted with halo. In some embodiments, D7 is . [0413] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is -C(O)-heterocyclyl, wherein the -C(O)-heterocyclyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1-6haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, C1-6alkyl-OH, -O-C1-6haloalkyl, C1-6alkyl-O-C1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D10 ) 2 . In some embodiments, the - C(O)-heterocyclyl is unsubstituted or substituted with halo. In some embodiments, D7 is or . [0414] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is -C(O)-aryl or -C(O)-heteroaryl, wherein the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1- 6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D10 ) 2 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with halo. In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CF 3 . In some embodiments, the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with -CN. In some embodiments, [0415] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is -N(R 7D1 )(R 7D2 ). In some embodiments, D7 is [0416] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, R 7D1 is H. In some embodiments, R 7D1 is C 1-6 alkyl. In some embodiments, R 7D2 is aryl. In some embodiments, R 7D2 is heteroaryl. In some embodiments, the heteroaryl is a 6-membered heteroaryl. [0417] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is -C(O)N(R 7D3 )(R 7D4 ). In some embodiments, D7 is . [0418] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, R 7D3 is H. In some embodiments, R 7D3 is C 1-6 alkyl. In some embodiments, R 7D4 is C 1-6 alkyl. In some embodiments, R 7D3 is H and R 7D4 is C 1-6 alkyl. [0419] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, D7 is -N(R 7D5 )C(O)R 7D6 -. In some embodiments, D7 is . [0420] In some embodiments of the compounds of Formula VII and stereoisomers and pharmaceutically acceptable salts thereof, R 7D5 is H. In some embodiments, R 7D5 is C 1-6 alkyl. In some embodiments, R 7D6 is C 1-6 alkyl. In some embodiments, R 7D6 is cycloalkyl. In some embodiments, R 7D6 is heterocyclyl. In some embodiments, R 7D5 is H and R 7D6 is C 1-6 alkyl, cycloalkyl, or heterocyclyl. [0421] In some embodiments, the compound, and stereoisomers and pharmaceutically acceptable salt thereof, is selected from the group consisting

[0422] In some embodiments, the compound, and stereoisomers and pharmaceutically acceptable salt thereof, is selected from the group consisting

O O O [0423] In some embodiments, the compound, and stereoisomers and pharmaceutically acceptable salt O [0424] In some embodiments, the compound, and stereoisomers and pharmaceutically acceptable salt

[0425] In some embodiments, the compound, and stereoisomers and pharmaceutically acceptable salt t . [0426] Unless otherwise stated, structures depicted herein are also meant to include salts (e.g. pharmaceutically acceptable salts), solvates, hydrates, and isomers (e.g. stereoisomers) thereof. Accordingly, the present disclosure is directed to compounds of Formulae I, II, III, IV, V, VI, and VII and salts, solvates, hydrates, and isomers thereof. Moreover, reference to compounds of Formulae I, II, III, IV, V, VI, and VII and stereoisomers and pharmaceutically acceptable salts thereof is considered to include reference to solvates, hydrates, and isomers (e.g. stereoisomers) of any thereof. [0427] In some embodiments, the compound is a solvate, hydrate, or isomer (e.g. stereoisomer) of a compound of Formulae I, II, III, IV, V, VI, and VII and stereoisomers and pharmaceutically acceptable salts thereof. [0428] Unless otherwise stated, structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of a hydrogen atom by deuterium or tritium, the replacement of a carbon atom by 13 C or 14 C, the replacement of a nitrogen atom by 15 N, or the replacement of an oxygen atom by 17 O or 18 O are within the scope of the present disclosure. Such isotopically labeled compounds are useful as research or diagnostic tools. Methods of Synthesizing the Compounds [0429] The compounds of Formulae I, II, III, IV, V, VI, and VII and stereoisomers and pharmaceutically acceptable salts thereof may be prepared by methods known in the art of organic synthesis. It is well understood that protecting groups for sensitive or reactive groups are employed where necessary in accordance with general principles or chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Greene and P. G. M. Wuts, “Protective Groups in Organic Synthesis”, Third edition, Wiley, New York 1999, which is hereby incorporated by reference in its entirety). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The selection processes, as well as the reaction conditions and order of their execution, shall be consistent with the preparation of the disclosed compounds (e.g., Formulae I, II, III, IV, V, VI, and VII and stereoisomers and pharmaceutically acceptable salts thereof). [0430] Those skilled in the art will recognize if a stereocenter exists in the compounds disclosed compounds (e.g., Formulae I, II, III, IV, V, VI, and VII and stereoisomers and pharmaceutically acceptable salts thereof). Accordingly, the present disclosure includes both possible stereoisomers (unless specified in the synthesis) and includes not only racemic compounds but the individual enantiomers and/or diastereomers as well. When a compound is desired as a single enantiomer or diastereomer, it may be obtained by stereospecific synthesis or by resolution of the final product or any convenient intermediate. Resolution of the final product, an intermediate, or a starting material may be affected by any suitable method known in the art. See, for example, “Stereochemistry of Organic Compounds” by E. L. Eliel, S. H. Wilen, and L. N. Mander (Wiley-Interscience, 1994), which is hereby incorporated by reference in its entirety. [0431] The compounds described herein may be made from commercially available starting materials or synthesized using known organic, inorganic, and/or enzymatic processes. [0432] Compounds of Formulae I, II, III, IV, V, VI, and VII can be prepared according to procedures outlined in Schemes and Examples herein. In the Examples section, compounds of the present disclosure are further exemplified by specific examples. Unless otherwise specified, all temperatures were expressed in °C and all reactions are conducted at room temperature. Pharmaceutical Compositions [0433] The compounds of Formulae I, II, III, IV, V, VI, and VII and stereoisomers and pharmaceutically acceptable salts thereof may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the disclosed compound and stereoisomers and pharmaceutically acceptable salts thereof is in association with a pharmaceutically acceptable adjuvant, diluent or carrier. Conventional procedures for the selection and preparation of suitable pharmaceutical formulations are described in, for example, “Pharmaceuticals - The Science of Dosage Form Designs”, M. E. Aulton, Churchill Livingstone, 1988, which is hereby incorporated by reference in its entirety. [0434] The present disclosure also provides a pharmaceutical composition comprising a compound of Formulae I, II, III, IV, V, VI, or VII, and stereoisomers and pharmaceutically acceptable salts thereof, and a pharmaceutically acceptable carrier. [0435] The present disclosure also provides a compound of Formulae I, II, III, IV, V, VI, or VII, and stereoisomers and pharmaceutically acceptable salts thereof, for use in medicine. [0436] The present disclosure also provides a pharmaceutical composition comprising a compound of Formulae I, II, III, IV, V, VI, or VII, and stereoisomers and pharmaceutically acceptable salts thereof, in association with a pharmaceutically acceptable adjuvant, diluent or carrier. [0437] The present disclosure further provides a process for the preparation of a pharmaceutical composition of the present disclosure which comprises mixing a compound of Formulae I, II, III, IV, V, VI, or VII and stereoisomers and pharmaceutically acceptable salts thereof with a pharmaceutically acceptable adjuvant, diluent or carrier. [0438] Depending on the mode of administration, the pharmaceutical composition will comprise from about 0.05 to about 99 %w (per cent by weight), more particularly from about 0.05 to about 80 %w, still more particularly from about 0.10 to about 70 %w, and even more particularly from about 0.10 to about 50 %w, of active ingredient, all percentages by weight being based on total composition. [0439] The pharmaceutical compositions may be administered topically (e.g. to the skin or to the lung and/or airways) in the form, e.g., of creams, solutions, suspensions, heptafluoroalkane (HFA) aerosols and dry powder formulations, for example, formulations in the inhaler device known as the Turbuhaler ® ; or systemically, e.g. by oral administration in the form of tablets, capsules, syrups, powders or granules; or by parenteral administration in the form of a sterile solution, suspension or emulsion for injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion); or by rectal administration in the form of suppositories. [0440] Dry powder formulations and pressurized HFA aerosols of the compounds of the present disclosure (including stereoisomers and pharmaceutically acceptable salts thereof) may be administered by oral or nasal inhalation. For inhalation, the compound is desirably finely divided. The finely divided compound preferably has a mass median diameter of less than 10 micrometres (μm), and may be suspended in a propellant mixture with the assistance of a dispersant, such as a C8-C20 fatty acid or salt thereof, (for example, oleic acid), a bile salt, a phospholipid, an alkyl saccharide, a perfluorinated or polyethoxylated surfactant, or other pharmaceutically acceptable dispersant. [0441] The compounds of the present disclosure may also be administered by means of a dry powder inhaler. The inhaler may be a single or a multi dose inhaler, and may be a breath actuated dry powder inhaler. [0442] One possibility is to mix the finely divided compound of the present disclosure with a carrier substance, for example, a mono-, di- or polysaccharide, a sugar alcohol, or another polyol. Suitable carriers are sugars, for example, lactose, glucose, raffinose, melezitose, lactitol, maltitol, trehalose, sucrose, mannitol; and starch. Alternatively the finely divided compound may be coated by another substance. The powder mixture may also be dispensed into hard gelatin capsules, each containing the desired dose of the active compound. [0443] Another possibility is to process the finely divided powder into spheres which break up during the inhalation procedure. This spheronized powder may be filled into the drug reservoir of a multidose inhaler, for example, that known as the Turbuhaler ® in which a dosing unit meters the desired dose which is then inhaled by the patient. With this system the active ingredient, with or without a carrier substance, is delivered to the patient. [0444] Another possibility is to process the compound as an amorphous dispersion in a polymer matrix such as hydroxypropyl methylcellulose (HPMC) or hydroxypropyl methylcellulose acetate succinate (HPMCAS). As the name suggests, spray-dried dispersions (SDDs) are obtained by dissolving drug and polymer in an organic solvent, atomizing the resulting solution into droplets, and evaporation to dried solid particles. SDDs are usually amenable for use a variety of final oral dosage forms, including capsules and tablets. [0445] For oral administration the compound of the present disclosure may be admixed with an adjuvant or a carrier, for example, lactose, saccharose, sorbitol, mannitol; a starch, for example, potato starch, corn starch or amylopectin; a cellulose derivative; a binder, for example, gelatin or polyvinylpyrrolidone; and/or a lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the like, and then compressed into tablets. If coated tablets are required, the cores, prepared as described above, may be coated with a concentrated sugar solution which may contain, for example, gum arabic, gelatin, talcum and titanium dioxide. Alternatively, the tablet may be coated with a suitable polymer dissolved in a readily volatile organic solvent. [0446] For the preparation of soft gelatin capsules, the compound of the present disclosure may be admixed with, for example, a vegetable oil or polyethylene glycol. Hard gelatin capsules may contain granules of the compound using either the above-mentioned excipients for tablets. Also liquid or semisolid formulations of the compound of the present disclosure may be filled into hard gelatin capsules. [0447] Liquid preparations for oral application may be in the form of syrups or suspensions, for example, solutions containing the compound of the present disclosure, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol. Optionally such liquid preparations may contain colouring agents, flavouring agents, saccharine and/or carboxymethylcellulose as a thickening agent or other excipients known to those skilled in art. Methods of Treatment [0448] The terms “treat,” “treating,” or “treatment”, as used herein, refer to any indicia of success in the amelioration of a disorder (such as injury, disease pathology, or condition), including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the disorder more tolerable to the subject; slowing or stopping the rate of degeneration, decline, or development; slowing the progression of disorder; making the final point of degeneration less debilitating; improving a subject’s physical or mental well-being; or relieving or causing regression of the disorder. The treatment of symptoms, including the amelioration of symptoms, can be based on objective or subjective parameters, which may include the results of a physical examination, a neuropsychiatric exam, and/or a psychiatric evaluation. Certain methods and uses disclosed herein may treat cancer by, for example, causing remission of cancer, slowing the rate of growth of cancer cells, slowing the rate of spread of cancer cells, reducing metastasis, or reducing the growth of metastatic tumors, reducing the size of one or more tumors, reducing the number of one or more tumors, or any combinations thereof. [0449] The terms “administered”, “administration”, or “administering”, as used herein, refers to either directly administering a disclosed compound (and stereoisomers and pharmaceutically acceptable salts thereof) or a composition to a subject, including an animal, in need of treatment by bringing such individual in contact with, or otherwise exposing such individual to, such compound. [0450] As used herein, the term “subject” encompasses mammals and non-mammals. Examples of mammals include, but are not limited to, any member of the class Mammalia: humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like. Examples of non-mammals include, but are not limited to, birds, fish and the like. In one embodiment of the present disclosure, the mammal is a human. [0451] A “patient” is a mammal, e.g., a human, mouse, rat, guinea pig, dog, cat, horse, cow, pig, or non-human primate, such as a monkey, chimpanzee, baboon or rhesus. “Patient” includes both humans and animals. [0452] “Inhibition”, as used herein, refers to reducing the activity of or complete inhibition of the target. activity may include reducing the activity of a component of a pathway to a level that is still detectable. Full inhibition may include stopping all activity of a component of a pathway (such as stopping the activity of PARP7), or reducing the activity of a component of a pathway to a level below detection. Inhibition of a component of a pathway may be measured directly or indirectly, using any methods known in the art. “Selective inhibition of PARP7”, as used herein, refers to wherein in vitro IC50 for PARP7 activity is less than about 10-fold compared to in vitro IC50 for other PARP family member activity in similar bio-assay formats, particularly PARP1 and PARP2. In some embodiments, the compound of Formulae I, II, III, IV, V, VI, and VII and stereoisomers and pharmaceutically acceptable salts thereof selectively inhibits PARP7. [0453] The term “disorder”, as used herein, refers to and is used interchangeably with the terms disease, condition, or illness, unless otherwise indicated. [0454] The term “disorder”, as used herein, refers to and is used interchangeably with the terms disease, condition, or illness, unless otherwise indicated. [0455] In some embodiments, the present disclosure provides compounds which are suitable for use in the treatment of one or more disorders which are linked to PARP7 (e.g. the overexpression of PARP). “Responsive to inhibition of PARP7”, as used herein, refers to disorders expected to improve with administration of inhibiting doses of the compound. Cancer is a disorder that may be responsive to treatment by administration of the compound. Some specific cancers responsive to treatment by administration of the compound can be identified by analysis of in vitro expression levels and abnormal activity of PARP7 in cancer cell lines. Additionally, in vitro cell viability assays can be used to identify specific cancer cell lines that are responsive to treatment by administration of the compound. Cancer cell lines responsive to PARP7 inhibitor compounds in vitro are predicted to be responsive to PARP7 inhibitor compounds in vivo, for example in xenograft models for cancer growth inhibition in mice. Also, some cancers may be responsive to PARP7 inhibitors indirectly by activating the immune system of the patient rather than by causing death of the cancer cells directly. Additional methods may be used to identify PARP7-responsive cancers such tumor-specific mutations in particular genes, tumor gene expression patterns as well as biomarkers in the blood for example circulating tumor DNA, RNA or proteins. [0456] The compounds of Formulae I, II, III, IV, V, VI, and VII and stereoisomers and pharmaceutically acceptable salts thereof have activity as pharmaceuticals, as discussed herein. [0457] The present disclosure provides a compound of Formulae I, II, III, IV, V, VI, or VII, and stereoisomers and pharmaceutically acceptable salts thereof, or a pharmaceutical composition thereof for use in the treatment of a disorder in a subject in need thereof. [0458] The present disclosure provides a compound of Formulae I, II, III, IV, V, VI, or VII, and stereoisomers and pharmaceutically acceptable salts thereof, or a pharmaceutical composition thereof for the treatment of a disorder in a subject in need thereof. [0459] The present disclosure provides a compound of Formulae I, II, III, IV, V, VI, or VII, and stereoisomers and pharmaceutically acceptable salts thereof, or a pharmaceutical composition thereof, for use in the treatment of a disorder that is responsive to inhibition of PARP7. [0460] The present disclosure provides a compound of Formulae I, II, III, IV, V, VI, or VII, and stereoisomers and pharmaceutically acceptable salts thereof, or a pharmaceutical composition thereof, in the treatment of a disorder that is responsive to inhibition of PARP7. [0461] The present disclosure provides a compound of Formulae I, II, III, IV, V, VI, or VII, and stereoisomers and pharmaceutically acceptable salts thereof, or a pharmaceutical composition thereof, for use in the manufacture of a medicament for the treatment of a disorder that is responsive to inhibition of PARP7. [0462] The present disclosure also provides a method of treating a disorder in a subject in need thereof, wherein the disorder is mediated by PARP7, comprising administering to the subject a compound of Formulae I, II, III, IV, V, VI, or VII, and stereoisomers and pharmaceutically acceptable salts thereof, or a pharmaceutical composition thereof. [0463] In some embodiments, the disorder is selected from the group consisting of cancer, cardiovascular disorder, neurological disorder, inflammatory disorder, autoimmune disorder, and infectious disease. [0464] In some embodiments, wherein the disorder is cancer. In some embodiments, the cancer is of solid organ origin or of hematopoietic origin. [0465] In some embodiments, the disorder is cancer, the cancer is of solid organ origin, and the solid organ is selected from the group consisting of the brain, breast, colon, endometrium, esophagus, head and neck, upper gastrointestinal tract, respiratory tract, lung, kidney, liver, lower gastrointestinal tract, small intestine, large intestine, ovary, pancreas, prostate, stomach, testes, and urinary tract. In some embodiments, the disorder is cancer and the cancer is adenocarcinoma. In some embodiments, the disorder is non-small cell lung cancer. In some embodiments, the cancer is squamous cell carcinoma of the lung (SCCL). [0466] In some embodiments, the disorder is cancer, and the cancer is leukemia or lymphoma. In some embodiments, the leukemia is acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), or chronic myelogenous leukemia (CML). In some embodiments, the lymphoma is Hodgkin's lymphoma , non-Hodgkin's lymphoma, multiple myeloma, B-cell lymphoma, diffuse large B-cell lymphoma (DLBCL), chronic lymphocyteic lymphoma (CLL), T-cell lymphoma, hairy cell lymphoma, or Burkett's lymphoma. [0467] In some embodiments, the disorder is cancer and the cancer is selected from the group consisting of cancer of the bladder, bone cancer, cancer of the cervix, cancer of the epithelium, cancer of the gallbladder, cancer of the rectum, skin cancer, thyroid cancer, and cancer of the uterus. [0468] In some embodiments, a compound of Formulae I, II, III, IV, V, VI, or VII, and stereoisomers and pharmaceutically acceptable salts thereof, is administered to a subject in need thereof to inhibit a component of the PARP7 pathway. In certain embodiments, the compound or stereoisomer or a pharmaceutically acceptable salt thereof, is administered as a pharmaceutical composition, as described herein. [0469] In some embodiments, inhibition of a component of the PARP7 pathway is measured directly, for example by measuring the product of a reaction catalyzed by a PARP7 pathway component. Inhibition of PARP7 activation may in some embodiments be demonstrated by western blotting and quantitatively assessing the levels of fulllength and cleaved PARP7 proteins from a cell line treated with compounds in vitro or in vivo. [0470] In some embodiments, inhibition of a component of the PARP7 pathway is measured indirectly, for example by measuring the level of expression of one or more genes that are regulated by PARP7. The inhibition of a component of the PARP7 pathway, such as inhibition of catalytic activity (MARylation), may modulate the expression of one or more genes that are regulated by PARP7, for example IFN-β. The transcription levels may be assessed, for example, by transcriptomic analysis, including but not limited to q-PCR. Modulation of one, two, three, four, five, or more genes may indicate inhibition of PARP activation. This evaluation of endogenous IFN-β gene expression may be assessed in cell lines (such as CT26 cell lines) or primary cells (such as fibroblasts of mouse, rat, or human origin). In some embodiments, the gene transcription levels of IFN-β are evaluated. Inhibition of PARP7 activation may in some embodiments be demonstrated by detection of IFN-β secreted by cells treated with compounds in vitro or in vivo. Example Embodiments [0471] Embodiment I-1. A compound of Formula I: and stereoisomers and pharmaceutically acceptable salts thereof, wherein: X 1 is -N- or -CR 1a2 -; X 2 is -N- or -CR 1a4 -; R 1a1 , R 1a2 , R 1a3 , and R 1a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1-6alkyl, -O-C2-6alkenyl, -O-C2-6alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 1L1 R 1L2 , or C 2-6 alkynyl-NR 1L3 R 1L4 ; wherein if X 1 is -CR 1a2 - and X 2 is -CR 1a4 -, then at least one of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 1a1 , R 1a2 , R 1a3 , and R 1a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2; R 1L1 , R 1L2 , R 1L3 , and R 1L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 1L1 , R 1L2 , R 1L3 , and R 1L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 1b1 -, or -CR 1b2 R 1b3 -, wherein R 1b1 , R 1b2 , and R 1b3 are independently H or C 1-6 alkyl; bond marked 2B is to D3, D4, D5, or D6; R 3f1 is H or C 1-6 alkyl; X 4 is -O-, -NR 3f2 -, or -CR 3f3 R 3f4 -, wherein R 3f2 , R 3f3 , and R 3f4 are independently selected from H and C1-6alkyl; wherein a bond marked 2B is to D3, D4, D5, or D6; X 5 is -CH(R 7g3 )- or -CH(R 7g4 )CH 2 N(R 7g5 )-; R 5g1 , R 7g2 , R 7g3 , R 7g4 , and R 7g5 are independently H or C1-6alkyl; B3, B4, B5, and B6 are independently a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18- membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B3, B4, B5, and B6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1- 6 alkyl, and oxo; D3, D4, D5, D6, and D7 are independently C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, - O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)- heteroaryl, -N(R 1D1 )(R 1D2 ), -C(O)N(R 1D3 )(R 1D4 ), or -N(R 1D5 )C(O)R 1D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D3, D4, D5, D6, and D7 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, - CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O- C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D10 ) 2 , wherein each R 1D10 is independently H or C 1-6 alkyl; R 1D1 , R 1D3 , and R 1D5 are independently H or C 1-6 alkyl; R 1D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 1D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 1D11 ) 2 , wherein each R 1D11 is independently H or C 1-6 alkyl; and R 1D4 and R 1D6 are independently C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 1D4 and R 1D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 1D12 ) 2 , wherein each R 1D12 is independently H or C 1-6 alkyl; provided that , then R 1a2 is not F, if and R 1c2 is F, then R 1a2 is not F. [0472] Embodiment I-2. A compound of Formula II: and stereoisomers and pharmaceutically acceptable salts thereof, wherein: X 1 is -N- or -CR 2a2 -; X 2 is -N- or -CR 2a4 -; R 2a1 , R 2a2 , R 2a3 , and R 2a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 2L1 R 2L2 , or C 2-6 alkynyl-NR 2L3 R 2L4 ; wherein if X 1 is -CR 2a2 - and X 2 is -CR 2a4 -, then at least one of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 2a1 , R 2a2 , R 2a3 , and R 2a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 ; R 2L1 , R 2L2 , R 2L3 , and R 2L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1- 6alkyl, C2-6alkynyl, or cycloalkyl of R 2L1 , R 2L2 , R 2L3 , and R 2L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 2b1 -, or -CR 2b2 R 2b3 -, wherein R 2b1 , R 2b2 , and R 2b3 are independently H or C 1-6 alkyl; A2 is

bond marked 2B is to D4, D5, or D6; X 5 is -CH(R 7g3 )- or -CH(R 7g4 )CH 2 N(R 7g5 )-; R 5g1 , R 7g2 , R 7g3 , R 7g4 , and R 7g5 are independently H or C 1-6 alkyl; B4, B5, and B6 are independently a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18- membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B4, B5, and B6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1- 6 alkyl, and oxo; provided that B4 is not D4, D5, D6, and D7 are independently C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O- aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 2D1 )(R 2D2 ), -C(O)N(R 2D3 )(R 2D4 ), or -N(R 2D5 )C(O)R 2D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D4, D5, D6, and D7 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D10 ) 2 , wherein each R 2D10 is independently H or C 1-6 alkyl; R 2D1 , R 2D3 , and R 2D5 are independently H or C 1-6 alkyl; R 2D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 2D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 2D11 ) 2 , wherein each R 2D11 is independently H or C 1-6 alkyl; and R 2D4 and R 2D6 are independently C1-6alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 2D4 and R 2D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 2D12 ) 2 , wherein each R 2D12 is independently H or C 1-6 alkyl; provided that if B4 is and D4 is -CHF 2 , -CF 2 CH 3 , or -CF 2 CF 3 , then R 2a2 is not F, if X 3 is -O-, B4 is and D4 is -C(O)-aryl, then R 2a4 is not Cl, and if D4 is and R 2c2 is F, then R 2a2 is not F. [0473] Embodiment I-3. A compound of Formula III: and stereoisomers and pharmaceutically acceptable salts thereof, wherein: X 1 is -N- or -CR 3a2 -; X 2 is -N- or -CR 3a4 -; R 3a1 , R 3a2 , R 3a3 , and R 3a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 3L1 R 3L2 , or C 2-6 alkynyl-NR 3L3 R 3L4 ; wherein if X 1 is -CR 3a2 - and X 2 is -CR 3a4 -, then at least one of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 3a1 , R 3a2 , R 3a3 , and R 3a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 ; R 3L1 , R 3L2 , R 3L3 , and R 3L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 3L1 , R 3L2 , R 3L3 , and R 3L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 3b1 -, or -CR 3b2 R 3b3 -, wherein R 3b1 , R 3b2 , and R 3b3 are independently H or C 1-6 alkyl; R 3f1 is H or C1-6alkyl; X 4 is -O-, -NR 3f2 -, or -CR 3f3 R 3f4 -, wherein R 3f2 , R 3f3 , and R 3f4 are independently selected from H and C 1-6 alkyl; B3 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B3 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C1-6alkyl, and oxo; D3 is C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 3D1 )(R 3D2 ), -C(O)N(R 3D3 )(R 3D4 ), or -N(R 3D5 )C(O)R 3D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D3 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 3D10 ) 2 , wherein each R 3D10 is independently H or C 1- 6 alkyl; R 3D1 , R 3D3 , and R 3D5 are independently H or C 1-6 alkyl; R 3D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 3D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 3D11 ) 2 , wherein each R 3D11 is independently H or C 1-6 alkyl; and R 3D4 and R 3D6 are independently C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 3D4 and R 3D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 3D12 ) 2 , wherein each R 3D12 is independently H or C 1-6 alkyl. [0474] Embodiment I-4. The compound of embodiment I-2, and stereoisomers and pharmaceutically acceptable salts thereof, wherein the compound is of Formula IV: X 1 is -N- or -CR 4a2 -; X 2 is -N- or -CR 4a4 -; R 4a1 , R 4a2 , R 4a3 , and R 4a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 4L1 R 4L2 , or C 2-6 alkynyl-NR 4L3 R 4L4 ; wherein if X 1 is -CR 4a2 - and X 2 is -CR 4a4 -, then at least one of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 4a1 , R 4a2 , R 4a3 , and R 4a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 ; R 4L1 , R 4L2 , R 4L3 , and R 4L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 4L1 , R 4L2 , R 4L3 , and R 4L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 4b1 -, or -CR 4b2 R 4b3 -, wherein R 4b1 , R 4b2 , and R 4b3 are independently H or C 1-6 alkyl; NH 2 , or -NHSO 2 CH 3 ; B4 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B4 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo; provided that B4 is not D4 is C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 4D1 )(R 4D2 ), -C(O)N(R 4D3 )(R 4D4 ), or -N(R 4D5 )C(O)R 4D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D4 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D10 ) 2 , wherein each R 4D10 is independently H or C 1- 6 alkyl; R 4D1 , R 4D3 , and R 4D5 are independently H or C 1-6 alkyl; R 4D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 4D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 4D11 ) 2 , wherein each R 4D11 is independently H or C 1-6 alkyl; and R 4D4 and R 4D6 are independently C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 4D4 and R 4D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 4D12 ) 2 , wherein each R 4D12 is independently H or C 1-6 alkyl; provided that , then R 4a2 is not F, if X 3 is -O-, B4 is , and D4 is -C(O)-aryl, then R 4a4 is not and R 4c2 is F, then R 4a2 is not F. [0475] Embodiment I-5. The compound of embodiment I-2, and stereoisomers and pharmaceutically acceptable salts thereof, wherein the compound is of Formula V: X 1 is -N- or -CR 5a2 -; X 2 is -N- or -CR 5a4 -; R 5a1 , R 5a2 , R 5a3 , and R 5a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 5L1 R 5L2 , or C 2-6 alkynyl-NR 5L3 R 5L4 ; wherein if X 1 is -CR 5a2 - and X 2 is -CR 5a4 -, then at least one of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 5a1 , R 5a2 , R 5a3 , and R 5a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 ; R 5L1 , R 5L2 , R 5L3 , and R 5L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 5L1 , R 5L2 , R 5L3 , and R 5L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 5b1 -, or -CR 5b2 R 5b3 -, wherein R 5b1 , R 5b2 , and R 5b3 are independently H or C 1-6 alkyl; , or -NHSO 2 CH 3 ; is H or C 1-6 alkyl; B5 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B5 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo; D5 is C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 5D1 )(R 5D2 ), -C(O)N(R 5D3 )(R 5D4 ), or -N(R 5D5 )C(O)R 5D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D5 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C1-6alkyl, C1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 5D10 ) 2 , wherein each R 5D10 is independently H or C 1- 6 alkyl; R 5D1 , R 5D3 , and R 5D5 are independently H or C 1-6 alkyl; R 5D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 5D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 5D11 ) 2 , wherein each R 5D11 is independently H or C 1-6 alkyl; and R 5D4 and R 5D6 are independently C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 5D4 and R 5D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 5D12 ) 2 , wherein each R 5D12 is independently H or C 1-6 alkyl. [0476] Embodiment I-6. The compound of embodiment I-2, and stereoisomers and pharmaceutically acceptable salts thereof, wherein the compound is of Formula VI: X 1 is -N- or -CR 6a2 -; X 2 is -N- or -CR 6a4 -; R 6a1 , R 6a2 , R 6a3 , and R 6a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 6L1 R 6L2 , or C 2-6 alkynyl-NR 6L3 R 6L4 ; wherein if X 1 is -CR 6a2 - and X 2 is -CR 6a4 -, then at least one of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is not H; wherein each C1-6alkyl, C2-6alkenyl, C2-6alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 6a1 , R 6a2 , R 6a3 , and R 6a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 ; R 6L1 , R 6L2 , R 6L3 , and R 6L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 6L1 , R 6L2 , R 6L3 , and R 6L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C1-6alkyl, C1-6haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 6b1 -, or -CR 6b2 R 6b3 -, wherein R 6b1 , R 6b2 , and R 6b3 are independently H or C 1-6 alkyl; NH 2 , or -NHSO 2 CH 3 ; B6 is a 3 to 8-membered monocyclic heterocyclediyl, a 7 to 18-membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3 to 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B6 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo; D6 is C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 6D1 )(R 6D2 ), -C(O)N(R 6D3 )(R 6D4 ), or -N(R 6D5 )C(O)R 6D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D6 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D10 ) 2 , wherein each R 6D10 is independently H or C 1- 6 alkyl; R 6D1 , R 6D3 , and R 6D5 are independently H or C1-6alkyl; R 6D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 6D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 6D11 ) 2 , wherein each R 6D11 is independently H or C 1-6 alkyl; and R 6D4 and R 6D6 are independently C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 6D4 and R 6D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C1-6alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 6D12 ) 2 , wherein each R 6D12 is independently H or C 1-6 alkyl. [0477] Embodiment I-7. The compound of embodiment I-2, and stereoisomers and pharmaceutically acceptable salts thereof, wherein the compound is of Formula VII: X 1 is -N- or -CR 7a2 -; X 2 is -N- or -CR 7a4 -; R 7a1 , R 7a2 , R 7a3 , and R 7a4 are independently H, halo, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O- heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 7L1 R 7L2 , or C 2-6 alkynyl-NR 7L3 R 7L4 ; wherein if X 1 is -CR 7a2 - and X 2 is -CR 7a4 -, then at least one of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is not H; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 7a1 , R 7a2 , R 7a3 , and R 7a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 ; R 7L1 , R 7L2 , R 7L3 , and R 7L4 are independently H, C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl, wherein each C 1-6 alkyl, C 2-6 alkynyl, or cycloalkyl of R 7L1 , R 7L2 , R 7L3 , and R 7L4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, and halocycloalkyl; X 3 is -O-, -NR 7b1 -, or -CR 7b2 R 7b3 -, wherein R 7b1 , R 7b2 , and R 7b3 are independently H or C 1-6 alkyl; 7 NH 2 , or -NHSO 2 CH 3 ; X 5 is -CH(R 7g3 )- or -CH(R 7g4 )CH 2 N(R 7g5 )-; R 7g2 , R 7g3 , R 7g4 , and R 7g5 are independently H or C1-6alkyl; D7 is C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1-6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, -C(O)-heteroaryl, -N(R 7D1 )(R 7D2 ), -C(O)N(R 7D3 )(R 7D4 ), or -N(R 7D5 )C(O)R 7D6 ; wherein the C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-aryl, -O-heteroaryl, -C(O)-C 1- 6 alkyl, -C(O)-cycloalkyl, -C(O)-heterocyclyl, -C(O)-aryl, or -C(O)-heteroaryl of D7 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, C 1-6 alkyl-OH, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D10 ) 2 , wherein each R 7D10 is independently H or C 1- 6 alkyl; R 7D1 , R 7D3 , and R 7D5 are independently H or C 1-6 alkyl; R 7D2 is aryl or heteroaryl, wherein the aryl or heteroaryl of R 7D2 is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C1-6alkyl, -O-C1-6haloalkyl, C1-6alkyl-O-C1-6alkyl, C1-6alkyl-O-C1-6haloalkyl, -C(O)- cycloalkyl, and -C(O)N(R 7D11 ) 2 , wherein each R 7D11 is independently H or C 1-6 alkyl; and R 7D4 and R 7D6 are independently C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, wherein each C 1-6 alkyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl of R 7D4 and R 7D6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1- 6 haloalkyl, -C(O)-cycloalkyl, and -C(O)N(R 7D12 ) 2 , wherein each R 7D12 is independently H or C 1-6 alkyl. [0478] Embodiment I-8. The compound of any one of embodiments I-1 to I-7, and stereoisomers and pharmaceutically acceptable salts thereof, wherein X 1 is -CR 1a2 -, -CR 2a2 -, -CR 3a2 -, -CR 4a2 -, -CR 5a2 -, -CR 6a2 - , or -CR 7a2 -. [0479] Embodiment I-9. The compound of any one of embodiments I-1 to I-8, and stereoisomers and pharmaceutically acceptable salts thereof, wherein X 2 is -CR 1a4 -, -CR 2a4 -, -CR 3a4 -, -CR 4a4 -, -CR 5a4 -, -CR 6a4 - , or -CR 7a4 -. [0480] Embodiment I-10. The compound of any one of embodiments I-1, I-2, I-4, I-8, and I-9, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1a2 , R 2a2 , or R 4a2 is H, Cl, Br, I, - OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2- 6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, - NR 1L1 R 1L2 , C 2-6 alkynyl-NR 1L3 R 1L4 , -NR 2L1 R 2L2 , C 2-6 alkynyl-NR 2L3 R 2L4 , -NR 4L1 R 4L2 , or C 2-6 alkynyl- NR 4L3 R 4L4 ; wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 1a2 , R 2a2 , or R 4a2 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C1-6alkyl, and -C(O)NH2. [0481] Embodiment I-11. The compound of any one of embodiments I-1, I-2, I-4, and I-8 to I-10, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1a4 , R 2a4 , or R 4a4 is H, F, Br, I, -OH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1-6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, -SO 2 -cycloalkyl, -NR 1L1 R 1L2 , C 2- wherein each C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, -O-C 1- 6 alkyl, -O-C 2-6 alkenyl, -O-C 2-6 alkynyl, -O-cycloalkyl, -O-heterocyclyl, -O-aryl, -O-heteroaryl, or -SO 2 - cycloalkyl of R 1a4 , R 2a4 , or R 4a4 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, -OH, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, halocycloalkyl, -O-C 1-6 alkyl, and -C(O)NH 2 . [0482] Embodiment I-12. The compound of any one of embodiments I-1 to I-11, and stereoisomers and pharmaceutically acceptable salts thereof, wherein one, two, three, or four of R 1a1 , R 1a2 , R 1a3 , and R 1a4 , R 2a1 , R 2a2 , R 2a3 , and R 2a4 , R 3a1 , R 3a2 , R 3a3 , and R 3a4 , R 4a1 , R 4a2 , R 4a3 , and R 4a4 , R 5a1 , R 5a2 , R 5a3 , and R 5a4 , R 6a1 , R 6a2 , R 6a3 , and R 6a4 , or R 7a1 , R 7a2 , R 7a3 , and R 7a4 is not H. [0483] Embodiment I-13. The compound of any one of embodiments I-1 to I-12, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1a3 , R 2a3 , R 3a3 , R 4a3 , R 5a3 , R 6a3 , or R 7a3 is not H. [0484] Embodiment I-14. The compound of any one of embodiments I-1 to I-13, and stereoisomers and pharmaceutically acceptable salts thereof, wherein one, two, three, or four of R 1a1 , R 1a2 , R 1a3 , and R 1a4 , [0485] Embodiment I-15. The compound of any one of embodiments I-1 to I-14, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1a1 , R 2a1 , R 3a1 , R 4a1 , R 5a1 , R 6a1 , or R 7a1 is F or Cl. [0486] Embodiment I-16. The compound of any one of embodiments I-1 to I-9 and I-11 to I-15, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1a2 , R 2a2 , R 3a2 , R 4a2 , R 5a2 , R 6a2 , or R 7a2 is F or Cl. [0487] Embodiment I-17. The compound of any one of embodiments I-1 to I-16, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1a3 , R 2a3 , R 3a3 , R 4a3 , R 5a3 , R 6a3 , or R 7a3 is F or Cl. [0488] Embodiment I-18. The compound of any one of embodiments I-1 to I-17, and stereoisomers and pharmaceutically acceptable salts thereof, wherein one, two, three, or four of R 1a1 , R 1a2 , R 1a3 , and R 1a4 , R 2a1 , R 2a2 , R 2a3 , and R 2a4 , R 3a1 , R 3a2 , R 3a3 , and R 3a4 , R 4a1 , R 4a2 , R 4a3 , and R 4a4 , R 5a1 , R 5a2 , R 5a3 , and R 5a4 , R 6a1 , R 6a2 , R 6a3 , and R 6a4 , or R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -OH. [0489] Embodiment I-19. The compound of any one of embodiments I-1 to I-16 and I-18, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1a3 , R 2a3 , R 3a3 , R 4a3 , R 5a3 , R 6a3 , or R 7a3 is -OH. [0490] Embodiment I-20. The compound of any one of embodiments I-1 to I-19, and stereoisomers and pharmaceutically acceptable salts thereof, wherein one, two, three, or four of R 1a1 , R 1a2 , R 1a3 , and R 1a4 , R 2a1 , R 2a2 , R 2a3 , and R 2a4 , R 3a1 , R 3a2 , R 3a3 , and R 3a4 , R 4a1 , R 4a2 , R 4a3 , and R 4a4 , R 5a1 , R 5a2 , R 5a3 , and R 5a4 , R 6a1 , R 6a2 , R 6a3 , and R 6a4 , or R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -O-C 1-6 alkyl or -O-cycloalkyl, wherein the -O-C 1-6 alkyl or -O-cycloalkyl is unsubstituted or substituted with halo or -CN. [0491] Embodiment I-21. The compound of any one of embodiments I-1 to I-16, I-18, and I-20, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1a3 , R 2a3 , R 3a3 , R 4a3 , R 5a3 , R 6a3 , or R 7a3 is -O-C 1-6 alkyl or -O-cycloalkyl, wherein the -O-C 1-6 alkyl or -O-cycloalkyl is unsubstituted or substituted with halo or -CN. [0492] Embodiment I-22. The compound of any one of embodiments I-1 to I-21, and stereoisomers and pharmaceutically acceptable salts thereof, wherein one, two, three, or four of R 1a1 , R 1a2 , R 1a3 , and R 1a4 , R 2a1 , R 2a2 , R 2a3 , and R 2a4 , R 3a1 , R 3a2 , R 3a3 , and R 3a4 , R 4a1 , R 4a2 , R 4a3 , and R 4a4 , R 5a1 , R 5a2 , R 5a3 , and R 5a4 , R 6a1 , R 6a2 , R 6a3 , and R 6a4 , or R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -O-C 2-6 alkynyl, wherein the -O-C 2-6 alkynyl is unsubstituted or substituted with halo. [0493] Embodiment I-23. The compound of any one of embodiments I-1 to I-16, I-18, I-20, and I-22, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1a3 , R 2a3 , R 3a3 , R 4a3 , R 5a3 , R 6a3 , or R 7a3 is -O-C 2-6 alkynyl, wherein the -O-C 2-6 alkynyl is unsubstituted or substituted with halo. [0494] Embodiment I-24. The compound of any one of embodiments I-1 to I-23, and stereoisomers and pharmaceutically acceptable salts thereof, wherein one, two, three, or four of R 1a1 , R 1a2 , R 1a3 , and R 1a4 , R 2a1 , R 2a2 , R 2a3 , and R 2a4 , R 3a1 , R 3a2 , R 3a3 , and R 3a4 , R 4a1 , R 4a2 , R 4a3 , and R 4a4 , R 5a1 , R 5a2 , R 5a3 , and R 5a4 , R 6a1 , R 6a2 , R 6a3 , and R 6a4 , or R 7a1 , R 7a2 , R 7a3 , and R 7a4 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O- heteroaryl is unsubstituted or substituted with halo. [0495] Embodiment I-25. The compound of any one of embodiments I-1 to I-16, I-18, I-20, I-22, and I-24, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1a3 , R 2a3 , R 3a3 , R 4a3 , R 5a3 , R 6a3 , or R 7a3 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O-heteroaryl is unsubstituted or substituted with halo. [0497] Embodiment I-27. The compound of any one of embodiments I-1 to I-26, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1a1 , R 2a1 , R 3a1 , R 4a1 , R 5a1 , R 6a1 , or R 7a1 is F or Cl and R 1a2 , R 2a2 , R 3a2 , R 4a2 , R 5a2 , R 6a2 , or R 7a2 is F or Cl. [0499] Embodiment I-29. The compound of any one of embodiments I-1 to I-28, and stereoisomers and pharmaceutically acceptable salts thereof, wherein X 3 is -O-. [0500] Embodiment I-30. The compound of any one of embodiments I-1 to I-29, and stereoisomers and pharmaceutically acceptable salts thereof, wherein X 3 is -CH 2 -. [0501] Embodiment I-31. The compound of any one of embodiments I-1, I-3, and I-8 to I-30, and stereoisomers and pharmaceutically acceptable salts thereof, wherein X 4 is -O-. [0502] Embodiment I-32. The compound of any one of embodiments I-1, I-3, and I-8 to I-30, and stereoisomers and pharmaceutically acceptable salts thereof, wherein X 4 is -NR 3f2 -. [0504] Embodiment I-34. The compound of any one of embodiments I-1, I-2 and I-4 to I-30, and stereoisomers and pharmaceutically acceptable salts thereof, wherein A1, A2, A4, A5, A6, or A7 is [0505] Embodiment I-35. The compound of any one of embodiments I-1, I-2, and I-4 to I-34, and stereoisomers and pharmaceutically acceptable salts thereof, wherein one or more of X 10 , X 11 , and X 12 is - N-. [0506] Embodiment I-36. The compound of any one of embodiments I-1, I-2, I-4 to I-30, and I-34, and stereoisomers and pharmaceutically acceptable salts thereof, wherein X 10 is -CR 1c1 -, -CR 2c1 -, -CR 4c1 -, -CR 5c1 -, -CR 6c1 -, or -CR 7c1 -, X 11 is -CR 1c2 -, -CR 2c2 -, -CR 4c2 -, -CR 5c2 -, -CR 6c2 -, or -CR 7c2 -, and X 12 is -CR 1c3 - , -CR 2c3 -, -CR 4c3 -, -CR 5c3 -, -CR 6c3 -, or -CR 7c3 -. [0507] Embodiment I-37. The compound of any one of embodiments I-1, I-2, I-4 to I-30, and I-34 to I-36, and stereoisomers and pharmaceutically acceptable salts thereof, wherein one, two, three, or four of 1 1 1 2 1 3 1 4 2 1 2 2 2 3 2 4 , R 4c1 , R 4c2 , R 4c3 , and R 4c4 , R 5c1 , R 5c2 , R 5c3 , and R 5c4 , R 6c1 , not H. [0508] Embodiment I-38. The compound of any one of embodiments I-1, I-2, I-4 to I-30, and I-34 to I-37 and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1c2 , R 2c2 , R 4c2 , R 5c2 , R 6c2 , 2 1 1 1 R 1c4 , R 2c1 , R 2c3 , and R 2c4 , R 4c1 , R 4c3 , and R 4c4 , R 5c1 , R 5c3 , and R 5c4 , R 6c1 , R 6c3 , 4 are H. [0509] Embodiment I-39. The compound of any one of embodiments I-1, I-2, I-4 to I-30, and I-34 to I-36, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 1c1 , R 1c2 , R 1c3 , and R 1c4 , R 2c1 , R 2c2 , R 2c3 , and R 2c4 , R 4c1 , R 4c2 , R 4c3 , and R 4c4 , R 5c1 , R 5c2 , R 5c3 , and R 5c4 , R 6c1 , R 6c2 , R 6c3 , and R 6c4 , or R 7c1 , R 7c2 , R 7c3 , and R 7c4 are H. [0510] Embodiment I-40. The compound of any one of embodiments I-1, I-2, and I-4 to I-30, and stereoisomers and pharmaceutically acceptable salts thereof, wherein A1, A2, A4, A5, A6, or A7 is [0511] Embodiment I-41. The compound of any one of embodiments I-1 to I-6 and I-8 to I-40, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is a 3-membered monocyclic heterocyclediyl, a 4-membered monocyclic heterocyclediyl, a 5-membered monocyclic heterocyclediyl comprising 2 or more N, a 6-membered monocyclic heterocyclediyl comprising 2 or more N, a 7-membered monocyclic heterocyclediyl, an 8-membered monocyclic heterocyclediyl, a 7 to 18- membered polycyclic heterocyclediyl, or a 7 to 18-membered spirocyclic heterocyclediyl; wherein the 3-membered monocyclic heterocyclediyl, 4-membered monocyclic heterocyclediyl, 5- membered monocyclic heterocyclediyl, 6-membered monocyclic heterocyclediyl, 7-membered monocyclic heterocyclediyl, 8-membered monocyclic heterocyclediyl, 7 to 18-membered polycyclic heterocyclediyl, or 7 to 18-membered spirocyclic heterocyclediyl of B3, B4, B5, or B6 is unsubstituted or substituted with one or more substituents independently selected from the group consisting of halo, C 1- 6 alkyl, and oxo. [0512] Embodiment I-42. The compound of any one of embodiments I-1 to I-6 and I-8 to I-40, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is a 3 to 8- membered monocyclic heterocyclediyl, wherein the 3 to 8 membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1- 6alkyl, and oxo. [0513] Embodiment I-43. The compound of any one of embodiments I-1 to I-6 and I-8 to I-42, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is a 4-membered monocyclic heterocyclediyl, wherein the 4-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1-6 alkyl, and oxo. [0514] Embodiment I-44. The compound of any one of embodiments I-1 to I-6 and I-8 to I-43, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is or . [0515] Embodiment I-45. The compound of any one of embodiments I-1 to I-6, I-8 to I-40 and I-42, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is a 6- membered monocyclic heterocyclediyl, wherein the 6-membered monocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1- 6 alkyl, and oxo. [0516] Embodiment I-46. The compound of any one of embodiments I-1 to I-6, I-8 to I-42 and I-45, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is . [0517] Embodiment I-47. The compound of any one of embodiments I-1 to I-6 and I-8 to I-42, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is [0518] Embodiment I-48. The compound of any one of embodiments I-1 to I-6 and I-8 to I-41, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is a 7 to 18- membered polycyclic heterocyclediyl, wherein the 7 to 18-membered polycyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1- 6 alkyl, and oxo. [0519] Embodiment I-49. The compound of any one of embodiments I-1 to I-6, I-8 to I-41, and I-48, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is . [0520] Embodiment I-50. The compound of any one of embodiments I-1 to I-6, I-8 to I-41, and I-48, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is [0521] Embodiment I-51. The compound of any one of embodiments I-1 to I-6, I-8 to I-41, and I-48, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is [0522] Embodiment I-52. The compound of any one of embodiments I-1 to I-6 and I-8 to I-41, and stereoisomers and pharmaceutically acceptable salts thereof, wherein B3, B4, B5, or B6 is a 7 to 18- membered spirocyclic heterocyclediyl, wherein the 7 to 18-membered spirocyclic heterocyclediyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, C 1- 6 alkyl, and oxo. [0523] Embodiment I-53. The compound of any one of embodiments I-1 to I-6 and I-8 to I-52, and stereoisomers and pharmaceutically acceptable salts thereof, wherein the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B3, B4, B5, or B6 comprises one or more N. [0524] Embodiment I-54. The compound of any one of embodiments I-1 to I-6, I-8 to I-43, and I-45 to I-54, and stereoisomers and pharmaceutically acceptable salts thereof, wherein the monocyclic heterocyclediyl, polycyclic heterocyclediyl, or spirocyclic heterocyclediyl of B3, B4, B5, or B6 comprises two or more N. [0525] Embodiment I-55. The compound of any one of embodiments I-1, I-5, I-8 to I-30, and I-34 to I-54, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 5g1 is H. [0526] Embodiment I-56. The compound of any one of embodiments I-1, I-5, I-8 to I-30, and I-34 to I-54, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 5g1 is -CH 3 . [0527] Embodiment I-57. The compound of any one of embodiments I-1, I-7, I-8 to I-30, and I-34 to I-54, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 7g2 is H. [0528] Embodiment I-58. The compound of any one of embodiments I-1, I-7, I-8 to I-30, and I-34 to I-54, and stereoisomers and pharmaceutically acceptable salts thereof, wherein R 7g2 is -CH 3 . [0529] Embodiment I-59. The compound of any one of embodiments I-1, I-7, I-8 to I-30, I-34 to I-54, I-57, and I-58 and stereoisomers and pharmaceutically acceptable salts thereof, wherein X 5 is -CH(R 7g3 )-, wherein R 7g3 is H or C1-6alkyl. [0530] Embodiment I-60. The compound of any one of embodiments I-1, I-7, I-8 to I-30, I-34 to I-54, I-57, and I-58, and stereoisomers and pharmaceutically acceptable salts thereof, wherein X 5 is - CH(R 7g4 )CH 2 N(R 7g5 )-, wherein R 7g4 and R 7g5 are independently H or C 1-6 alkyl. [0531] Embodiment I-61. The compound of any one of embodiments I-1 to I-60, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is C 1-6 alkyl, wherein the C 1- 6 alkyl is unsubstituted or substituted with halo. [0532] Embodiment I-62. The compound of any one of embodiments I-1 to I-60, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is cycloalkyl, wherein the cycloalkyl is unsubstituted or substituted with halo. [0533] Embodiment I-63. The compound of any one of embodiments I-1 to I-60, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is aryl, wherein the aryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0534] Embodiment I-64. The compound of any one of embodiments I-1 to I-60 and I-63, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is a monocyclic 6-membered aryl, wherein the monocyclic 6-membered aryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)- cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0535] Embodiment I-65. The compound of any one of embodiments I-1 to I-60, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is heteroaryl, wherein the heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1- 6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0536] Embodiment I-66. The compound of any one of embodiments I-1 to I-60 and I-65, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is a monocyclic 5 or 6-membered heteroaryl comprising one or more N, wherein the monocyclic 5 or 6- membered heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1- 6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and - C(O)N(CH 3 ) 2 . [0537] Embodiment I-67. The compound of any one of embodiments I-1 to I-60, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is -O-C 1-6 alkyl or C 1-6 alkyl- O-C1-6alkyl, wherein the -O-C1-6alkyl or C1-6alkyl-O-C1-6alkyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, - OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, - C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0538] Embodiment I-68. The compound of any one of embodiments I-1 to I-60, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is -O-aryl or -O-heteroaryl, wherein the -O-aryl or -O-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1- 6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0539] Embodiment I-69. The compound of any one of embodiments I-1 to I-60, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is -C(O)-C 1-6 alkyl, -C(O)- cycloalkyl, or -C(O)-heterocyclyl, wherein the -C(O)-C1-6alkyl, -C(O)-cycloalkyl, or -C(O)-heterocyclyl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O-C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl- O-C 1-6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0540] Embodiment I-70. The compound of any one of embodiments I-1 to I-60, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is -C(O)-aryl or -C(O)- heteroaryl, wherein the -C(O)-aryl or -C(O)-heteroaryl is unsubstituted or substituted with one or more substituents selected from the group consisting of halo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, cycloalkyl, -OH, -O- C 1-6 alkyl, -O-C 1-6 haloalkyl, C 1-6 alkyl-O-C 1-6 alkyl, C 1-6 alkyl-O-C 1-6 haloalkyl, -C(O)-cycloalkyl, -C(O)NH 2 , -C(O)NHCH 3 , and -C(O)N(CH 3 ) 2 . [0541] Embodiment I-71. The compound of any one of embodiments I-1 to I-60, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is -N(R 1D1 )(R 1D2 ), - N(R 2D1 )(R 2D2 ), -N(R 3D1 )(R 3D2 ), -N(R 4D1 )(R 4D2 ), -N(R 5D1 )(R 5D2 ), -N(R 6D1 )(R 6D2 ), or -N(R 7D1 )(R 7D2 ). [0542] Embodiment I-72. The compound of any one of embodiments I-1 to I-60, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is -C(O)N(R 1D3 )(R 1D4 ), - C(O)N(R 2D3 )(R 2D4 ), -C(O)N(R 3D3 )(R 3D4 ), -C(O)N(R 4D3 )(R 4D4 ), -C(O)N(R 5D3 )(R 5D4 ), -C(O)N(R 6D3 )(R 6D4 ), or -C(O)N(R 7D3 )(R 7D4 ). [0543] Embodiment I-73. The compound of any one of embodiments I-1 to I-60, and stereoisomers and pharmaceutically acceptable salts thereof, wherein D3, D4, D5, D6, or D7 is -N(R 1D5 )C(O)R 1D6 -, - N(R 2D5 )C(O)R 2D6 -, -N(R 3D5 )C(O)R 3D6 -, -N(R 4D5 )C(O)R 4D6 , -N(R 5D5 )C(O)R 5D6 -, -N(R 6D5 )C(O)R 6D6 -, or - N(R 7D5 )C(O)R 7D6 -. [0544] Embodiment I-74. A compound, and stereoisomers and pharmaceutically acceptable salts

[0545] Embodiment I-75. A compound, and stereoisomers and pharmaceutically acceptable salts thereof, selected from the group consisting O O [0546] Embodiment I-76. A compound, and stereoisomers and pharmaceutically acceptable salts t O [0548] Embodiment I-78. A compound, and stereoisomers and pharmaceutically acceptable salts thereof, selected from the group consisting . [0549] Embodiment I-79. The compound of any one of embodiments I-1 to I-78, wherein the compound selectively inhibits PARP7. [0550] Embodiment I-80. A pharmaceutical composition comprising a compound of any one of embodiments I-1 to I-79, and stereoisomers and pharmaceutically acceptable salts thereof, and a pharmaceutically acceptable carrier. [0551] Embodiment I-81. A compound of any one of embodiments I-1 to I-79, and stereoisomers and pharmaceutically acceptable salts thereof, for use in medicine. [0552] Embodiment I-82. A compound of any one of embodiments I-1 to I-79, and stereoisomers and pharmaceutically acceptable salts thereof, for use in the treatment of a disorder that is responsive to inhibition of PARP7. [0553] Embodiment I-83. Use of a compound of any one of embodiments I-1 to I-79, and stereoisomers and pharmaceutically acceptable salts thereof, in the treatment of a disorder that is responsive to inhibition of PARP7. [0554] Embodiment I-84. A compound of any one of embodiments I-1 to I-79, and stereoisomers and pharmaceutically acceptable salts thereof, for use in the manufacture of a medicament for the treatment of a disorder that is responsive to inhibition of PARP7. [0555] Embodiment I-85. A method of treating a disorder in a subject in need thereof, wherein the disorder is mediated by PARP7, comprising administering to the subject a compound of any one of embodiments I-1 to I-79, and stereoisomers and pharmaceutically acceptable salts thereof. [0556] Embodiment I-86. The compound of embodiment I-82 or I-84, the use of embodiment I-83, or the method of embodiment I-85, wherein the disorder is selected from the group consisting of cancer, cardiovascular disorder, neurological disorder, inflammatory disorder, autoimmune disorder, and infectious disease. [0557] Embodiment I-87. The compound of embodiment I-82, I-84, or I-86, the use of embodiment I- 83 or I-86, or the method of embodiment I-85 or I-86, wherein the disorder is cancer and the cancer is of solid organ origin or of hematopoietic origin. [0558] Embodiment I-88. The compound of embodiment I-82, I-84, I-86, or I-87 the use of embodiment I-83, I-86, or I-87 or the method of embodiment I-85, I-86, or I-87, wherein the disorder is cancer, the cancer is of solid organ origin, and the solid organ is selected from the group consisting of the brain, breast, colon, endometrium, esophagus, head and neck, upper gastrointestinal tract, respiratory tract, lung, kidney, liver, lower gastrointestinal tract, small intestine, large intestine, ovary, pancreas, prostate, stomach, testes, and urinary tract. [0559] Embodiment I-89. The compound of embodiment I-82, I-84, I-86, or I-87 the use of embodiment I-83, I-86, or I-87 or the method of embodiment I-85, I-86, or I-87, wherein the disorder is cancer and the cancer is leukemia or lymphoma. [0560] Embodiment I-90. The compound, use, or method of embodiment I-89, wherein the leukemia is acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), or chronic myelogenous leukemia (CML). [0561] Embodiment I-91. The compound, use, or method of embodiment I-89, wherein the lymphoma is Hodgkin's lymphoma , non-Hodgkin's lymphoma, multiple myeloma, B-cell lymphoma, diffuse large B-cell lymphoma (DLBCL), chronic lymphocytic lymphoma (CLL), T-cell lymphoma, hairy cell lymphoma, or Burkett's lymphoma. [0562] Embodiment I-92. The compound of embodiment I-82, I-84, or I-86, the use of embodiment I- 83 or I-86, or the method of embodiment I-85 or I-86, wherein the disorder is cancer and the cancer is selected from the group consisting of cancer of the bladder, bone cancer, cancer of the cervix, cancer of the epithelium, cancer of the gallbladder, cancer of the rectum, skin cancer, thyroid cancer, and cancer of the uterus. Examples General Synthetic Procedures [0563] In order that the invention described herein may be more fully understood, the following examples are set forth. The synthetic and biological examples described in this application are offered to illustrate the compounds, pharmaceutical compositions, and methods provided herein and are not to be construed in any way as limiting their scope. [0564] The compounds provided herein can be prepared from readily available starting materials using modifications to the specific synthesis protocols set forth below that would be well known to those of skill in the art. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvents used, but such conditions can be determined by those skilled in the art by routine optimization procedures. [0565] Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. The choice of a suitable protecting group for a particular functional group as well as suitable conditions for protection and deprotection are well known in the art. For example, numerous protecting groups, and their introduction and removal, are described in Greene et al., Protecting Groups in Organic Synthesis, Second Edition, Wiley, New York, 1991, and references cited therein. ABBREVIATIONS

[0566] Routine 1 H NMR spectra were recorded on 400, or 500 MHz spectrometers (Agilent, Oxford or Bruker) at ambient temperature. NMR solvents, d-chloroform (CDCl 3 ), d 6 -dimethylsulfoxide (DMSO-d 6 ), and deuterated methanol (CD 3 OD) were purchased from commercial suppliers and used without further purification. Spectra were processed using the automatic phasing and polynomial baseline correction features of the software. In cases where two adjacent peaks of equal or unequal height were observed, these two peaks may be labeled as either a multiplet or as a doublet. In the case of a doublet, a coupling constant using this software may be assigned. In any given example, one or more protons may not be observed due to obscurity by water and/or solvent peaks. Spectral data are reported as follows: chemical shift (multiplicity [singlet (s), broad singlet (bs), doublet (d), triplet (t), quartet (q), sextuplet (sex), multiplet (m), apparent (app), doublet of doublets (dd), doublet of doublet of doublets (ddd), doublet of triplets (dt)], coupling constant, integration). Chemical shifts are reported in ppm (δ), and coupling constants are reported in Hz. 1 H Resonances are referenced to solvent residual peaks for CDCl 3 (7.26 ppm), DMSO-d 6 (2.50 ppm), CD 3 OD (3.30 ppm). [0567] The HPLC-UV/MS instrumentation for product analysis consisted of a Waters Alliance 2695 with a column heater coupled with a ZQ 4000 mass spectrometer. The MS was equipped with an electrospray ionization (ESI) source and used in scan mode (100 - 1200 amu, source temperature: 150 °C) for both positive and negative ionization. The HPLC was equipped with a Waters photodiode array detector (PDA) 2998 (range used: 195-320nm). The analytical method was developed on a XBridge C18 column (3.5 µm particle size, 4.6 x 30 mm) with a 10 mM buffer (ammonium formate pH 3.8 or ammonium bicarbonate pH 10) - A% and acetonitrile - B% as the mobile phase. A flow rate of 3 mL/min at 25 °C was set and the following gradient was used: 1) 5% B isocratic for 0.2 min, 5% - 100% B in 1.8 min, 100% B for 1 min. or 2) 5% B isocratic for 0.2 min, 5% - 100% B in 5.8 min, 100% B for 1 min. [0568] In some other instances, HPLC-UV/MS instrumentation for product analysis consisted of a Agilent LCMS, Column: Waters X-Bridge C18 (50 mm x 4.6 mm x 3.5 μm); Column Temperature: 40 °C; Flow Rate: 1.5 mL/min; Mobile Phase: from 90% [water + 10 mM NH 4 HCO 3 ] and 5% [CH 3 CN] to 0% [water + 10 mM NH4HCO3] and 100% [CH3CN] in 6.0 min, then under this condition for 2.0 min, finally changed to 90% [water + 10 mM NH 4 HCO 3 ] and 5% [CH 3 CN] in 0.1 min and under this condition for 1 min. Prep-HPLC conditions [0569] An example of preparative HPLC condition employed to purify products is described below. Purifications were not limited to the gradient illustrated below and variations of the illustrated gradient were made according to polarity of the products obtained. 1.1 Chromatographic Equipment Gilson Prep-HPLC system: GX-281 sample manager, 306 pump, 806 Manometric module, 811D DYNAMIC Mixer, UV/VIS-156 1.2 Chromatographic condition Column: Waters X-Bridge TM Prep C185µm OBD TM , 19 × 250 mm Flowrate: 20 mL/min Gradient: Table 1: Chromatographic gradient conditions Wavelength: 214 nm and 254 nm. Examples [0570] The following synthetic examples are provided to illustrate the present disclosure, and should not be construed as limiting thereof. In these examples, all parts and percentages are by weight, unless otherwise noted. Synthesis of Example 1: 6-(4-(3-((7-Hydroxy-4-oxo-3,4-dihydrophthalazin- 1yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile tert-Butyl 4-(5-cyanopyridin-2-yl)piperazine-1-carboxylate [0571] A mixture of 6-chloronicotinonitrile (10.0 g, 72.2 mmol), tert-butyl piperazine-1-carboxylate (13.6 g, 72.9 mmol) and K 2 CO 3 (17.0 g, 122.7 mmol) in MeCN (10 mL) was stirred at 60 °C for 12 hours. The mixture was cooled to rt, diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 0 to 30%) to give tert- butyl 4-(5-cyanopyridin-2-yl)piperazine-1-carboxylate as a white solid (15.4 g, 73% yield). LCMS ESI m/z: 289 [M+H] + . Step 2: 6-(Piperazin-1-yl)nicotinonitrile hydrochloride [0572] A solution of tert-butyl 4-(5-cyanopyridin-2-yl)piperazine-1-carboxylate (15.4 g, 53.4 mmol) in HCl/dioxane (4M, 50 mL) was stirred at rt for 1 hour. The reaction mixture was concentrated in vacuo to give 6-(piperazin-1-yl)nicotinonitrile hydrochloride as a white solid (11.5 g, 96% yield). This was used without further purification. LCMS ESI m/z 189 [M+H] + . Step 3: 6-(4-(2-Fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e [0573] To a solution of 2-fluoro-5-formylbenzoic acid (10.0 g, 59.5 mmol) and 6-(piperazin-1- yl)nicotinonitrile (10.2 g, 54.1 mmol) in DMF (100 mL) was added EDCI (12.1 g, 81.1 mmol), HOBt (11.0 g, 81.1 mmol) and DIPEA (16.3 g, 162 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (100 mL) and extracted with EtOAc (100 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 0 to 30%) to give 6-(4-(2- fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitrile as a white solid (18 g, 94% yield). LCMS ESI m/z 339 [M+H] + . Step 4 3-Hydroxy-5-methoxyisobenzofuran-1(3H)-one [0574] To a stirred solution of 2-bromo-4-methoxybenzoic acid (5.00 g, 21.6 mmol) in anhydrous THF (5.17 mL) at -78 °C was added n-BuLi (2.77 g, 43.3 mmol) dropwise under argon. After stirred at this temperature for 0.5 hour, the mixture was treated with DMF (4.75 g, 64.9 mmol, 5.03 mL). The resulting solution was kept at -78 °C for 1 hour, then the reaction mixture was allowed to warm to ambient temperature. Water (10 mL) was added dropwise to the mixture, the aqueous layer was washed with diethyl ether (2 x 30 mL), shaken, and acidified with 2 M HCl until the solution reached pH 5~6. The organic layer was separated, dried and concentrated to the residue. The residue was purified by silica gel chromatography (petroleum ether : EtOAc = 10: 1 to 3: 1) to give 3-hydroxy-5-methoxy-3H- isobenzofuran-1-one (1.5 g, 38% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.18 (d, J = 6.8 Hz, 1H), 7.78 (d, J = 9.0 Hz, 1H), 7.22 (d, J = 5.9 Hz, 2H), 6.62 (d, J = 6.8 Hz, 1H), 3.94 (s, 3H). Step 5 Dimethyl 6-methoxy-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphonate [0575] To a solution of 3-hydroxy-5-methoxy-3H-isobenzofuran-1-one (1.50 g, 8.33 mmol) in dimethylphosphite (15 mL) was stirred at 100 °C for 2 hours then cooled to room temperature. The mixture was poured into water (30 mL), extracted with ethyl acetate (40 mL) for twice. The combined organic layers were washed with brine, dried and concentrated to the crude. The crude was purified by silica gel chromatography (petroleum ether : EtOAc = 30: 1 to 1: 1) to give dimethyl 6-methoxy-3-oxo- 1,3-dihydroisobenzofuran-1-ylphosphonate (1.70 g, 75% yield). LCMS ESI m/z: 273.0 [M + H] + . Step 6 6-(4-(3-((6-Methoxy-3-oxoisobenzofuran-1(3H)-ylidene)methyl) benzoyl) piperazin-1- yl)nicotinonitrile [0576] To a mixture of dimethyl 6-methoxy-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphonate (800 mg, 2.94 mmol) in THF (20 mL) was added 6-[4-(3-formylbenzoyl)piperazin-1-yl]pyridine-3- carbonitrile (1.13 g, 3.53 mmol) and Et 3 N (892 mg, 8.82 mmol, 1.23 mL). The resulting mixture was stirred at 70 °C for 10 hours. The mixture was quenched with sat. NaHSO3 solution (50 mL) and filtered. The filter cake was washed with H 2 O (20 mL x 2), dried and concentrated under reduced pressure to give 6-(4-(3-((6-methoxy-3-oxoisobenzofuran-1(3H)-ylidene)methyl) benzoyl)piperazin-1- yl)nicotinonitrile (900 mg, 66% yield) as a white solid. LCMS ESI 467.2 [M + H] + . Step 7

6-(4-(3-((7-Methoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl) benzoyl)piperazin-1-yl)nicotinonitrile [0577] To a mixture of 6-(4-(3-((6-methoxy-3-oxoisobenzofuran-1(3H)- ylidene)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (759 mg, 1.57 mmol) in THF (9.26 mL) was added hydrazine hydrate (784 mg, 15.7 mmol, 764 μL). The resulting mixture was stirred at 70 °C for 3 hours. The mixture was cooled to rt. and filtered. The filter cake was washed with a solution of petroleum ether/EtOAc =10/1 (20 mL x 2) and dried under reduced pressure to give 6-(4-(3-((7-methoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile (500 mg, 64% yield) as a white solid. LCMS ESI m/z 481.2 [M + H] + . Step 8 6-(4-(3-((7-Hydroxy-4-oxo-3,4-dihydrophthalazin-1yl)methyl)b enzoyl)piperazin-1-yl)nicotinonitrile [0578] To a solution of 6-(4-(3-((7-methoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (130 mg, 271 μmol) in DMF (2 mL) was added LiCl (34.4 mg, 812 μmol, 16.6 μL) at 25 °C. The reaction was stirred at 150 °C for 6 hours under microwave. The mixture was filtered and purified by prep-HPLC to give 6-(4-(3-((7-hydroxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile (51.6 mg, 41% yield) as a white solid. LCMS ESI 467.2 [M + H] + . 1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.32 (s, 1H), 8.51 (d, J = 2.0 Hz, 1H), 8.10 (d, J = 8.7 Hz, 1H), 7.89 (dd, J = 9.1, 2.4 Hz, 1H), 7.43 – 7.37 (m, 2H), 7.35 (s, 1H), 7.32 – 7.26 (m, 1H), 7.22 (dd, J = 8.7, 2.3 Hz, 1H), 7.11 (d, J = 2.2 Hz, 1H), 6.90 (d, J = 9.1 Hz, 1H), 4.25 (s, 2H), 3.68 (m, 8H). Synthesis of Example 2: 6-(4-(3-((4-Oxo-7-phenoxy-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

5-Phenoxyisobenzofuran-1(3H)-one [0579] To a mixture of 5-bromoisobenzofuran-1(3H)-one (5.0 g, 23 mmol) and phenol (4.3 g, 46 mmol) in dry DMF (100 mL) was successively added CuBr (0.33 g, 2.3 mmol), pentane-2,4-dione (0.46 g, 4.6 mmol) and K 2 CO 3 (6.4 g, 46 mmol) at room temperature. The resulting mixture was flushed with N 2 for 10 min. Then the reaction was allowed to heat to 90 °C and stirred at this temperature for overnight. The slurry was filtered over a pad of celite. The filtrate was concentrated. The filtrate was diluted with H 2 O (100 ml). The mixture was extracted with EtOAc (50 mL x 3). The combined organic layers were washed with 1 N NaOH (40 mL x 2). The organic layer was separated, dried, filtered and concentrated. The crude product was purified by silica gel chromatography (petroleum ether/EtOAc = 30/1 to 10/1, v/v) to give 5-phenoxyisobenzofuran-1(3H)-one (2.0 g, 38% yield) as a light yellow solid. 1 H NMR (400 MHz, CDCl 3 ) δ 7.85 (d, J = 8.0 Hz, 1H), 7.45-7.41 (m, 2H), 7.45-7.26 (m, 1H), 7.26-7.25 (m, 1H), 7.13-7.08 (m, 2H), 6.94 (d, J = 4.0 Hz, 1H), 5.33 (d, J = 4.0 Hz, 2H). Step 2 2-(Hydroxymethyl)-4-phenoxybenzoic acid [0580] To a solution of 5-phenoxyisobenzofuran-1(3H)-one (1.4 g, 6.2 mmol), MeOH (15 mL) and H 2 O (15 mL) was added KOH (0.69 g, 12 mmol) at room temperature. The resulting mixture was allowed to warm to 50 °C, and stirred at the same temperature for 3 hours. The volatile was removed. The residue was dissolved in H 2 O (40 mL). The solution was acidified with 1 N HCl until the solution reached pH 2-3. The suspension was filtered, and the solid was collected. The product was dried in vacuo to give 2- (hydroxymethyl)-4-phenoxybenzoic acid (1.3 g, 87%) as a white solid, which was directly used for the next step without further purification. 1 H NMR (400 MHz, CDCl3) δ 7.91 (d, J = 8.0 Hz, 1H), 7.47-7.45 (m, 2H), 7.30-7.29 (m, 1H), 7.25-7.23 (m, 1H), 7.12-7.10 (m, 2H), 6.90-6.87 (m, 1H), 4.82 (d, J = 8.0 Hz, 2H). Step 3 3-Hydroxy-5-phenoxyisobenzofuran-1(3H)-one [0581] To a slurry of 2-(hydroxymethyl)-4-phenoxybenzoic acid (1.1 g, 4.5 mmol) in dry DCM (30 mL) was added Dess-Martin periodinane (2.3 g, 5.4 mmol) in portionwise at 0 °C. Then the reaction was allowed to warm to room temperature, and stirred at the same temperature for 2 hours. The suspension was filtered, and the solid was rinsed with DCM (50 mL). The filtrate was washed by a sat. aq of Na 2 SO 3 (30 mL x 2). The organic layer was separated, dried, filtered and concentrated. The residue was purified by silica gel chromatography (DCM/MeOH = 20/1 to 10/1, v/v) to give 3-hydroxy-5- phenoxyisobenzofuran-1(3H)-one (0.6 g, 55% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 7.81 (d, J = 8.0 Hz, 1H), 7.44-7.43 (m, 2H), 7.28-7.23 (m, 2H), 7.17 (dd, J = 4.0 Hz, J = 8.0 Hz, 1H), 7.11-7.08 (m, 3H), 6.52 (s, 1H). Step 4 Dimethyl 3-oxo-6-phenoxy-1,3-dihydroisobenzofuran-1-ylphosphonate [0582] A flask were charged 3-hydroxy-5-phenoxyisobenzofuran-1(3H)-one (0.6 g, 2.5 mmol) and dimethyl phosphonate (10 mL) under N 2 . The mixture was heated to 100 °C, and stirred at the same temperature for overnight. The reaction was diluted with EtOAc (50 mL). The solution was washed by water (15 mL x 7). The organic layer was separated, dried, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 1/1, v/v) to give dimethyl 3-oxo-6- phenoxy-1,3-dihydroisobenzofuran-1-ylphosphonate (0.15 g, 18% yield) as a yellow solid. Step 5 6-(4-(3-((3-Oxo-6-phenoxyisobenzofuran-1(3H)-ylidene)methyl) benzoyl)piperazin-1- yl)nicotinonitrile [0583] To a mixture of dimethyl 3-oxo-6-phenoxy-1,3-dihydroisobenzofuran-1-ylphosphonate (0.15 g, 0.45 mmol) and 6-(4-(3-formylbenzoyl)piperazin-1-yl)nicotinonitrile (0.43 g, 1.3 mmol) in dry THF (5 mL) was added Et 3 N (91 mg, 0.9 mmol) dropwise at 0 °C under N 2 . The reaction mixture was allowed to warm to room temperature, and stirred at the same temperature for overnight. The mixture was poured into a mixture of DCM (50 mL) and a sat. aq of NaHSO 3 (50 mL). The solution was vigorously stirred at room temperature for 1 hour. The organic layer was separated, dried, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 5/1 to 1/1, v/v) to give 6-(4- (3-((3-oxo-6-phenoxyisobenzofuran-1(3H)-ylidene)methyl)benzo yl)piperazin-1-yl)nicotinonitrile (0.2 g, 83% yield) as a white solid. LCMS ESI m/z: 529.2 [M+H] + . Step 6 6-(4-(3-((4-Oxo-7-phenoxy-3,4-dihydrophthalazin-1-yl)methyl) benzoyl)piperazin-1-yl)nicotinonitrile [0584] A mixture of 6-(4-(3-((3-oxo-6-phenoxyisobenzofuran-1(3H)- ylidene)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (0.22 g, 0.44 mmol) and N 2 H 4 •H 2 O (0.8 mL) in THF (1.6 mL) was stirred at 70 °C for 2 hours in a sealed tube. The slurry was filtered, and the solid was collected. The product was triturated in MTBE (20 mL) for overnight. The suspension was filtered. The product was dried in vacuo to give 6-(4-(3-((4-oxo-7-phenoxy-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (0.11 g, 49% yield) as a white solid. LCMS ESI m/z: 543.2 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.56 (s, 1H), 8.49 (d, J = 4.0 Hz, 1H), 8.26 (d, J = 8.0 Hz, 1H), 7.87 (dd, J = 8.0, 4.0 Hz, 1H), 7.46 (t, J = 8.0 Hz, 2H), 7.38-7.37 (m, 3H), 7.30 - 7.28 (m, 4H), 7.09 (d, J = 8.0 Hz, 2H), 6.87 (d, J = 12.0 Hz, 1H), 4.25 (s, 2H), 3.76-3.62 (m, 6H), 3.41-3.40 (m, 2H). Synthesis of Example 3: 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-2-yn-1-yloxy)-3,4-dihydroph thalazin- 1yl)methyl)benzoyl) piperazin-1-yl)nicotinonitrile 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-2-yn-1-yloxy)-3,4-dihydroph thalazin-1yl)methyl)benzoyl) piperazin-1-yl)nicotinonitrile [0585] A mixture of 6-(4-(3-((7-hydroxy-4-oxo-3,4-dihydrophthalazin-1yl)methyl)b enzoyl)piperazin- 1-yl)nicotinonitrile (100 mg, 0.21 mmol), 3-bromoprop-1-yne (26 mg, 0.22 mmol) and K 2 CO 3 (86 mg, 0.63 mmol) in DMF (5 mL) was stirred at 50 °C for 1 hour. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was concentrated in vacuo and the residue was purified by prep-HPLC to give 6-(4-(2-fluoro-5-((4-oxo-7-(prop-2-yn-1- yloxy)-3,4-dihydrophthalazin-1-yl)methyl) benzoyl)piperazin-1-yl)nicotinonitrile as a white solid (40 mg, 37% yield). LCMS ESI m/z: 523 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 9.95 (s, 1H), 8.34 (dd, J = 10.0, 5.5 Hz, 2H), 7.59 (dd, J = 9.0, 2.3 Hz, 1H), 7.29 (dd, J = 12.8, 4.1 Hz, 3H), 7.14 (d, J = 2.4 Hz, 1H), 7.00 (t, J = 8.7 Hz, 1H), 6.55 (d, J = 9.0 Hz, 1H), 4.71 (d, J = 2.4 Hz, 2H), 4.18 (s, 2H), 3.82 (s, 2H), 3.71 (s, 2H), 3.62 (t, J = 5.1 Hz, 2H), 3.36 (s, 2H), 2.58 (t, J = 2.4 Hz, 1H). Synthesis of Example 4: 6-(4-(3-((7-Methoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0586] Following the procedure described in Example 1 and making non-critical variations as required to replace 2-(hydroxymethyl)-4-phenoxybenzoic acid with 2-(hydroxymethyl)-4-methoxybenzoic acid, 6- (4-(3-((7-methoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)be nzoyl)piperazin-1-yl)nicotinonitrile was obtained as a white solid. LCMS ESI m/z: 481.1 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.45 (s, 1H), 8.52 (d, J = 2.0 Hz, 1H), 8.19 (d, J = 8.8 Hz, 1H), 7.90 (dd, J = 9.1, 2.4 Hz, 1H), 7.41 (m, 4H), 7.30 (t, J = 5.4 Hz, 2H), 6.92 (d, J = 9.1 Hz, 1H), 4.35 (s, 2H), 3.88 (s, 3H), 3.69 (s, 8H). Synthesis of Example 5: 6-(4-(3-((7-Chloro-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

5-Chloro-3-hydroxyisobenzofuran-1(3H)-one [0587] To a mixture of 4-chlorobenzoic acid (1.0 g, 6.4 mmol) in THF (10 mL) was added TMEDA (1.6 g, 14.1 mmol). The mixture was stirred at -78 o C for 2 minutes. Then s-BuLi (14.1 mL, 14.1 mmol, 1 M in THF) was drop-wise into the mixture, which was stirred at -78 o C for another 30 min. DMF (2 mL) was added to the mixture, and the mixture was stirred at -78 o C for another 1 h. The mixture was quenched with 4 N aq. HCl, extracted with EtOAc (30 mL), dried over Na2SO4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 10/1 to 5/1, v/v) to give 5-chloro-3-hydroxyisobenzofuran-1(3H)-one (330 mg, 28% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 8.64 (d, J = 2.0 Hz, 1H), 8.24 (dd, J = 8.4, 2.1 Hz, 1H), 7.84 (s, 1H), 7.66 (s, 1H), 6.61 (s, 1H). Step 2 Dimethyl 6-chloro-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphonate [0588] Following the procedure of step 5 in Example 1, but starting with 5-chloro-3- hydroxyisobenzofuran-1(3H)-one gave the title compound as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ) δ 7.88 (d, J = 8.2 Hz, 1H), 7.75 (s, 1H), 7.57 (d, J = 8.2 Hz, 1H), 5.67 (m, 1H), 3.94 (m, 3H), 3.75-3.55 (m, 3H). Step 3

6-(4-(3-((6-Chloro-3-oxo-1,3-dihydroisobenzofuran-1yl)(metho xy)methyl)benzoyl) piperazin-1- yl)nicotinonitrile [0589] To a solution of dimethyl 6-chloro-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphonate (100 mg, 0.4 mmol) in THF (2 mL) was added 6-(4-(3-formylbenzoyl)piperazin-1-yl)nicotinonitrile (366 mg, 1.1 mmol). Then Et 3 N (116 mg, 1.1 mmol) was drop-wise into the mixture, which was stirred at 20 o C for 16 hrs. The solids were collected by filtration, washed with petroleum ether (5 mL x 2) and dried under vacuum to give 6-(4-(3-((6-chloro-3-oxo-1,3-dihydroisobenzofuran-1- yl)(methoxy)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (150 mg, 84% yield) as a yellow solid. LCMS ESI m/z: 503.1 [M+H] + . Step 4 6-(4-(3-((6-Chloro-3-oxoisobenzofuran-1(3H)-ylidene)methyl)b enzoyl)piperazin-1-yl)nicotinonitrile [0590] To a mixture of 6-(4-(3-((6-chloro-3-oxo-1,3-dihydroisobenzofuran-1- yl)(methoxy)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (130 mg, 0.3 mmol) in toluene (5 mL) was added PTSA (5 mg, 0.03 mmol). The resulting mixture was stirred at 100 o C for 1 hr. The mixture was concentrated to give the crude 6-(4-(3-((6-chloro-3-oxoisobenzofuran-1(3H)- ylidene)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (110 mg, 90% yield) as yellow gum. LCMS ESI m/z: 571.1 [M+H] + . Step 5

6-(4-(3-((7-Chloro-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)b enzoyl)piperazin-1-yl)nicotinonitrile [0591] Following the procedure of step 7 in Example 1, but starting with 6-(4-(3-((6-chloro-3- oxoisobenzofuran-1(3H)-ylidene)methyl)benzoyl)piperazin-1-yl )nicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 484.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.64 (s, 1H), 8.44 (d, J = 1.9 Hz, 1H), 8.18 (d, J = 8.5 Hz, 1H), 7.97 (d, J = 1.9 Hz, 1H), 7.85 – 7.77 (m, 2H), 7.37 – 7.31 (m, 3H), 7.26 – 7.20 (m, 1H), 6.84 (m, 1H), 4.30 (s, 2H), 3.61 (s, 6H), 3.32 (d, J = 6.8 Hz, 2H). Synthesis of Example 6: 6-(4-(3-((7-Fluoro-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile Step 1 5-Fluoro-3-hydroxyisobenzofuran-1(3H)-one [0592] Following the procedure of step 4 in Example 1, but starting with 4-fluorobenzoic acid gave the title compound as a yellow solid. LCMS ESI m/z: 169.1 [M+H] + . Step 2 Dimethyl 6-fluoro-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphonate [0593] Following the procedure of step 5 in Example 1, but starting with 5-fluoro-3- hydroxyisobenzofuran-1(3H)-one gave the title compound as a yellow oil. LCMS ESI m/z: 261.1 [M+H] + . Step 3 6-(4-(3-((6-Fluoro-3-oxoisobenzofuran-1(3H)-ylidene)methyl)b enzoyl)piperazin-1-yl)nicotinonitrile [0594] Following the procedure of step 6 in Example 1, but starting with dimethyl 6-fluoro-3-oxo-1,3- dihydroisobenzofuran-1-ylphosphonate gave the title compound as a yellow solid. LCMS ESI m/z: 454.1 [M+H] + . Step 4 2-(2-(3-(4-(5-Cyanopyridin-2-yl)piperazine-1-carbonyl)phenyl )acetyl)-4-fluorobenzohydrazide [0595] To a mixture of 6-(4-(3-((6-fluoro-3-oxoisobenzofuran-1(3H)- ylidene)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (0.14 g, 0.64 mmol) and N 2 H 4 •H 2 O (0.8 mL) in THF (1.6 mL) was stirred at 25 °C for 16 hours in a sealed tube. The reaction mixture was concentrated to give the crude product 2-(2-(3-(4-(5-cyanopyridin-2-yl)piperazine-1-carbonyl)phenyl )acetyl)-4- fluorobenzohydrazide as a yellow solid. LCMS ESI m/z: 487.1 [M+H] + . Step 5 6-(4-(3-((7-Fluoro-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)b enzoyl)piperazin-1-yl)nicotinonitrile [0596] A mixture of 2-(2-(3-(4-(5-cyanopyridin-2-yl)piperazine-1-carbonyl)phenyl )acetyl)-4- fluorobenzohydrazide (0.012 g, 0.02 mmol) in dioxane (5 mL) was stirred at 100 °C for 5 hours in a sealed tube. The reaction mixture was concentrated to the crude. The residue was purified by Prep-TLC (DCM/MeOH = 10/1) to give 6-(4-(3-((7-fluoro-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (8 mg, 70% yield) as a white solid. LCMS ESI m/z: 468.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.68 (s, 1H), 8.52 (m, 1H), 8.34 (m, 1H), 7.91 (m, 1H), 7.89 (m, 1H), 7.70 (m, 1H), 7.43 (m, 3H), 7.31 (m, 1H), 6.91 (m, 1H), 4.31 (s, 2H), 3.69 (m, 8H). Synthesis of Example 7: 6-(4-(5-((7-(Cyclopropylmethoxy)-4-oxo-3,4-dihydrophthalazin -1- yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile

Methyl 2-bromo-4-(cyclopropylmethoxy)benzoate [0597] A mixture of methyl 2-bromo-4-hydroxybenzoate (1.00 g, 4.33 mmol), (bromomethyl)cyclopropane (701 mg, 5.19 mmol) and K 2 CO 3 (1.79 g, 12.98 mmol) in DMF (10 mL) was stirred at 65 °C under Ar (g) for 12 hours. The mixture was cooled to rt, diluted with H 2 O (20 mL) and extracted with EtOAc (30 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 2-bromo-4- (cyclopropylmethoxy)benzoate as a yellow oil (1.23 g, 98% yield). This was used without further purification. LCMS ESI m/z : 285 [M+H] + . Step 2 2-Bromo-4-(cyclopropylmethoxy)benzoic acid [0598] A mixture of methyl 2-bromo-4-(cyclopropylmethoxy)benzoate (1.23 g, 4.31 mmol) and LiOH- H 2 O (362 mg, 8.62 mmol) in MeOH (12 mL) and H 2 O (4 mL) was stirred at 50 °C for 12 hours. The organic solvent was removed in vacuo and aq HCl (2N) was added until the solution reached pH 5~6, then extracted with EtOAc (30 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give 2-bromo-4-(cyclopropylmethoxy)benzoic acid as a white solid (1.1 g, 94%). This was used without further purification. LCMS ESI m/z : 271 [M+H] + . Step 3 5-(Cyclopropylmethoxy)-3-hydroxyisobenzofuran-1(3H)-one [0599] A solution of 2-bromo-4-(cyclopropylmethoxy)benzoic acid (900 mg, 3.32 mmol) in THF (10 mL) was cooled to -78°C, then n-BuLi (2.5 M in heptane, 2.66 mL, 6.64 mmol) was addded dropwise under Ar (g). The reaction was stirred at -78 °C for 15 min, then DMF (533 mg, 7.30 mmol) was added. The reaction was stirred at -78 °C for 20 min, then quenched with sat. NH 4 Cl soltion (10 mL), added aq HCl (1N) until the solution reached pH 5~6. The mixture was extracted with EtOAc (3 x 30 mL). The combined organic layer was washed with bine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 30 to 50%) to give 5- (cyclopropylmethoxy)-3-hydroxyisobenzofuran-1(3H)-one (110 mg, 14% yield). LCMS ESI m/z 221 (M+H) + . Step 4 Dimethyl (6-(cyclopropylmethoxy)-3-oxo-1,3-dihydroisobenzofuran-1-yl) phosphonate [0600] A solution of 5-(cyclopropylmethoxy)-3-hydroxyisobenzofuran-1(3H)-one (110 mg, 0.50 mmol) in dimethyl phosphonate (5 mL) was stirred at 100 °C for 3 hours. The mixture was cooled to rt, diluted with H2O (10 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 40 to 60 %) to give dimethyl (6- (cyclopropylmethoxy)-3-oxo-1,3-dihydroisobenzofuran-1-yl)pho sphonate as a yellow oil (135 mg, 87% yield). LCMS ESI m/z : 313 [M+H] + . Step 5

6-(4-(5-((6-(Cyclopropylmethoxy)-3-oxoisobenzofuran-1(3H)-yl idene)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0601] A mixture of dimethyl (6-(cyclopropylmethoxy)-3-oxo-1,3-dihydroisobenzofuran-1- yl)phosphonate (110 mg, 0.35 mmol), 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (143 mg, 0.42 mmol ) and Et 3 N (107 mg, 1.06 mmol) in THF (5 mL) was stirred at 60 °C for 3 hours. The mixture was cooled to rt, diluted with H 2 O (30 mL) and extracted with DCM (30 mL x 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give 6-(4-(5-((6- (cyclopropylmethoxy)-3-oxoisobenzofuran-1(3H)-ylidene)methyl )-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile as a white solid (180 mg, 97% yield). This was used without further purification. LCMS ESI m/z: 525 [M+H] + ). Step 6 6-(4-(5-((7-(Cyclopropylmethoxy)-4-oxo-3,4-dihydrophthalazin -1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0602] To a solution of 6-(4-(5-((6-(cyclopropylmethoxy)-3-oxoisobenzofuran-1(3H)-yl idene)methyl)- 2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile (180 mg, 0.34 mmol) in THF (5 mL) was added N 2 H 4 •H 2 O (136 mg, 6.8 mmol) and heated to 60 °C for 3 hours. The solids were collected by filtration, washed with water and dried under vacuum to give (4-(5-((7-(cyclopropylmethoxy)-4-oxo-3,4-dihydrophthalazin-1 - yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile as a white solid (24 mg, 13% yield). LCMS ESI m/z 539 [M+H] + ). 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.44 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.48 – 7.36 (m, 3H), 7.26 (dd, J = 14.9, 5.7 Hz, 2H), 6.93 (d, J = 9.1 Hz, 1H), 4.31 (s, 2H), 3.95 (d, J = 7.0 Hz, 2H), 3.82 – 3.68 (m, 4H), 3.67 – 3.57 (m, 2H), 3.32 – 3.29 (m, 2H), 1.29 – 1.14 (m, 1H), 0.63 – 0.49 (m, 2H), 0.41 – 0.27 (m, 2H). Synthesis of Example 8: 6-(4-(5-((7-(But-3-yn-2-yloxy)-4-oxo-3,4-dihydrophthalazin-1 -yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile 6-(4-(5-((7-(But-3-yn-2-yloxy)-4-oxo-3,4-dihydrophthalazin-1 -yl)methyl)-2-fluorobenzoyl)piperazin- 1-yl)nicotinonitrile [0603] A mixture of 6-(4-(2-fluoro-5-((7-hydroxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (40 mg, 0.08 mmol), but-3-yn-2-yl 4- methylbenzenesulfonate (27 mg, 0.12 mmol) and K 2 CO 3 (33 mg, 0.24 mmol) in DMF (5 mL) was stirred at 50°C for 1 hour. The solid were filtered off, and the filtrate was concentrated in vacuo. The residue was purified by prep-HPLC to give the desired product 6-(4-(5-((7-(but-3-yn-2-yloxy)-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)piperazin-1-y l)nicotinonitrile as a white solid (29 mg, 68% yield). LCMS ESI m/z 537 (M+H) + ). 1 H–NMR (400 MHz, DMSO–d 6 ) δ 12.49 (s, 1H), 8.20 (d, J = 8.8 Hz, 1H), 7.90 (dd, J = 9.2, 2.4 Hz, 1H), 7.49 – 7.37 (m, 4H), 7.26 (t, J = 9.2 Hz, 1H), 6.92 (d, J = 8.8 Hz, 1H), 5.37 – 5.35 (m, 1H), 4.38 – 4.21 (m, 2H), 3.77 – 3.73 (m, 4H), 3.62 – 3.54 (m, 3H), 3.32 – 3.29 (m, 2H), 1.58 (d, J = 6.8 Hz, 3H). Synthesis of Example 9: 6-(4-(2-Fluoro-5-((4-oxo-7-(2,2,2-trifluoroethoxy)-3,4-dihyd rophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile 3,5-Dibromoisobenzofuran-1(3H)-one [0604] A mixture of 5-bromoisobenzofuran-1(3H)-one (13.5 g, 63.4 mmol), NBS (13.5 g, 76.1 mmol) and AIBN (1.04 g, 6.34 mmol) in CCl 4 (100 mL) was heated to 80 °C and stirred for 16 hours. The mixture was cooled to rt, diluted with H 2 O (30 mL) and extracted with DCM (80 mL x 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petrole ether = 0 to 15%) to afford 3,5- dibromoisobenzofuran-1(3H)-one as a light yellow solid (8.8 g, 48% yield). LCMS ESI m/z 291 [M+H] + . Step 2 5-(Difluoromethoxy)-3-hydroxyisobenzofuran-1(3H)-one [0605] A mixture of 3,5-dibromoisobenzofuran-1(3H)-one (8.8 g, 16.6 mmol) and KOH (2.79 g, 49.7 mmol) in H2O (25 mL) was stirred at 80 °C for 3 hours. The reaction was cooled to rt and aq HCl (1N) was added until the solution reached pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 5-bromo-3-hydroxyisobenzofuran-1(3H)-one as a light yellow solid (6.4 g, 92% yield). This was used without further purification. LCMS ESI m/z 229 [M+H] + . Step 3 Dimethyl (6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate [0606] A solution of 5-bromo-3-hydroxyisobenzofuran-1(3H)-one (6.4 g, 22.35 mmol) in dimethyl phosphonate (50 mL) was heated to 100 °C and stirred for 2 hours. The mixture was cooled to rt, diluted with H2O (100 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30 to 50%) to afford dimethyl (6-bromo-3-oxo-1,3- dihydroisobenzofuran-1-yl)phosphonate as a white solid (5.68 g, 70 %). LCMS ESI m/z 321 [M+H] + . Step 4 Methyl-5-((6-bromo-3-oxoisobenzofuran-1(3H)-ylidene)methyl)- 2-fluorobenzoate [0607] A solution of dimethyl (6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (5.00 g,15.6 mmol), methyl 2-fluoro-5-formylbenzoate (3.12 g, 17.13 mmol) and Et 3 N (4.73 g, 46.72 mmol) in THF (50 mL) was stirred at rt under Ar (g) for 12 hours. The reaction mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over Na2SO4, filtered and concentrated in vacuo to afford methyl 5-((6-bromo-3-oxoisobenzofuran-1(3H)- ylidene)methyl)-2-fluorobenzoate as a white solid (5.01 g, 85% yield). This was used without further purification. LCMS ESI m/z 377 (M+H) + . Step 5 Methyl 5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoate [0608] A solution of methyl 5-((6-bromo-3-oxoisobenzofuran-1(3H)-ylidene)methyl)-2-fluor obenzoate (5.80 g, 13.3 mmol) and N 2 H 4 •H 2 O (1.33g, 26.5 mmol) in THF (50 mL) was stirred at 70 °C for 4 hours. The solids were filtered and washed with water (30 mL) to afford methyl 5-((7-bromo-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoate as a white solid (4.1 g, 79% yield). This was used without further purification. LCMS ESI m/z 391 (M+H) + . Step 6 (4-(4-Fluoro-3-(methoxycarbonyl)benzyl)-1-oxo-1,2-dihydropht halazin-6-yl)boronic acid [0609] A mixture of methyl 5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoate (529 mg, 1.11 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (1.41 g, 5.54 mmol), KOAc (653 mg, 6.65 mmol) and Pd(dppf)Cl 2 (49 mg, 0.06 mmol) in DMSO (10 mL) was stirred at 80 °C under Ar (g) for 2 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to afford (4-(4-fluoro-3-(methoxycarbonyl)benzyl)-1-oxo-1,2- dihydrophthalazin-6-yl)boronic acid as a light yellow solid (520 mg, 67% yield). This was used without further purification. LCMS ESI m/z 357 (M+H) + . Step 7 Methyl 2-fluoro-5-((4-oxo-7-(2,2,2-trifluoroethoxy)-3,4-dihydrophth alazin-1-yl)methyl)benzoate [0610] A mixture of (4-(4-fluoro-3-(methoxycarbonyl)benzyl)-1-oxo-1,2-dihydropht halazin-6- yl)boronic acid (60 mg, 0.17 mmol ), Cu(OAc) 2 ·H 2 O (4 mg, 0.02 mmol) and DMAP (41 mg, 0.33 mmol) in CF 3 CH 2 OH (1 mL) was stirred at 40 °C under O 2 (g) for 12 hours. The reaction was quenched with water (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 50 to 70%) to give methyl 2-fluoro-5-((4-oxo-7-(2,2,2- trifluoroethoxy)-3,4-dihydrophthalazin-1-yl)methyl)benzoate as a white solid (25 mg, 27% yield). LCMS ESI m/z 411[M+H] + . Step 8 2-Fluoro-5-((4-oxo-7-(2,2,2-trifluoroethoxy)-3,4-dihydrophth alazin-1-yl)methyl)benzoic acid [0611] A mixture of methyl 2-fluoro-5-((4-oxo-7-(2,2,2-trifluoroethoxy)-3,4-dihydrophth alazin-1- yl)methyl)benzoate (25 mg, 0.06 mmol) and NaOH (24 mg, 0.6 mmol) in MeOH (1 mL), THF (1 mL) and H 2 O (1 mL) was stirred at rt for 1 hour. The organic solvent was removed in vacuo and aq HCl (1N) was added to until the solution reached pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 2-fluoro-5-((4-oxo-7-(2,2,2-trifluoroethoxy)-3,4-dihydrophth alazin-1- yl)methyl)benzoic acid as a white solid (20 mg, 83% yield). LCMS ESI m/z 397 [M+H] + . Step 9 6-(4-(2-Fluoro-5-((4-oxo-7-(2,2,2-trifluoroethoxy)-3,4-dihyd rophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0612] To a solution of 6-(piperazin-1-yl)nicotinonitrile (12 mg, 0.06 mmol) and 2-fluoro-5-((4-oxo-7- (2,2,2-trifluoroethoxy)-3,4-dihydrophthalazin-1-yl)methyl)be nzoic acid (20 mg, 0.05 mmol) in DMF (1 mL) was added EDCI (10 mg, 0.05 mmol), HOBt (10 mg, 0.08 mmol) and DIPEA (20 mg, 0.15 mmol). The reaction mixture was stirred at rt for 2 hours. The reaction mixture was purified by prep-HPLC to afford 6-(4-(2-fluoro-5-((4-oxo-7-(2,2,2-trifluoroethoxy)-3,4-dihyd rophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile as a white solid (12 mg, 43% yield). LCMS ESI m/z 567 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.53 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 8.23 (d, J = 8.7 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.56 – 7.50 (m, 2H), 7.48 – 7.42 (m, 2H), 7.25 (t, J = 8.9 Hz, 1H), 6.92 (d, J = 9.2 Hz, 1H), 4.99 (q, J = 8.7 Hz, 2H), 4.33 (s, 2H), 3.80 – 3.70 (m, 4H), 3.65 – 3.59 (m, 2H), 3.17 (s, 2H). Synthesis of Example 10: 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile Step 1

6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1-yl)methyl)benzoyl)piperazin-1- yl)nicotinonitrile [0613] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (420 mg, 0.77 mmol), tributyl(prop-1-ynyl)stannane (760 mg, 2.30 mmol) and Pd(dppf)Cl 2 (58 mg, 0.08 mmol) in 1,4-dioxane (10 mL) were stirred at 100 °C under Ar (g) for 16 hours. The reaction was cooled to rt, diluted with water (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 50 to 80%) to give 6-(4-(2-fluoro-5- ((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazin-1-yl)methyl)be nzoyl)piperazin-1-yl)nicotinonitriletert- butyl as a white solid (145 mg, 37% yield). LCMS ESI m/z 508 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.64 (s, 1H), 8.52 (d, J = 8.2 Hz, 1H), 8.20 (d, J = 8.2 Hz, 1H), 7.98 – 7.87 (m, 2H), 7.78 (dd, J = 8.2, 1.2 Hz, 1H), 7.42 – 7.26 (m, 3H), 6.93 (d, J = 9.1 Hz, 1H), 4.34 (s, 2H), 3.75 (d, J = 10.2 Hz, 4H), 3.63 (s, 2H), 3.31 (s, 2H), 2.08 (s, 3H). Synthesis of Example 11: 6-(4-(2-Fluoro-5-((7-(3-methoxyazetidin-1-yl)-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile

6-(4-(5-((6-Bromo-3-oxoisobenzofuran-1(3H)-ylidene)methyl)-2 -fluorobenzoyl)piperazin-1- yl)nicotinonitrile [0614] A mixture of dimethyl (6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (3.5 g, 10.90 mmol), 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (4.43 g, 13.08 mmol) and Et 3 N (3.31 g, 32.70 mmol) in THF (70 mL) was heated to 60 °C and stirred for 3 hours. The mixture was cooled to rt, diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with water and brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 0 to 50%) to afford 6-(4-(5-((6- bromo-3-oxoisobenzofuran-1(3H)-ylidene)methyl)-2-fluorobenzo yl)piperazin-1-yl)nicotinonitrile as a yellow solid (3.6 g, 62% yield). LCMS ESI m/z 533 [M+H] + . Step 2 6-(4-(5-((7-Bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoyl)piperazin-1- yl)nicotinonitrile [0615] To a solution of 6-(4-(5-((6-bromo-3-oxoisobenzofuran-1(3H)-ylidene)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (3.60 g, 6.75 mmol) in THF (10 mL) was added N 2 H 4 •H 2 O (4.60 g, 67.5 mmol), then the mixture was stirred at 70 °C for 4 hours. The solids were collected by filtration, washed with water and dried under vacuum to give 6-(4-(5-((7-bromo-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)piperazin-1-y l)nicotinonitrile as a white solid (2.9 g, 79% yield). This was used without further purification. LCMS ESI m/z 547 [M+H] + . Step 3 6-(4-(2-Fluoro-5-((7-(3-methoxyazetidin-1-yl)-4-oxo-3,4-dihy drophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0616] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (100 mg, 0.18 mmol), Pd 2 (dba) 3 (10 mg, 0.01 mmol), XPhos (9.0 mg, 0.02 mmol), sodium tert-butoxide (53 mg, 0.55 mmol) and 3-methoxyazetidine (48 mg, 0.55 mmol) in DMA (2 mL) was stirred at 130 °C for 2 hours. The mixture was cooled to rt, filtered, and the filtrate was purified by prep-HPLC to give 6-(4-(2-fluoro-5-((7-(3-methoxyazetidin-1-yl)-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)benzoyl) piperazin-1-yl)nicotinonitrile as a white solid (11 mg, 18% yield). LCMS ESI m/z 554 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.17 (s, 1H), 8.52 (d, J = 1.9 Hz, 1H), 8.01 (d, J = 8.7 Hz, 1H), 7.91 (dd, J = 9.1, 2.3 Hz, 1H), 7.45 – 7.39 (m, 2H), 7.25 (t, J = 9.3 Hz, 1H), 6.92 (d, J = 9.1 Hz, 1H), 6.85 (dd, J = 8.7, 2.2 Hz, 1H), 6.58 (d, J = 2.1 Hz, 1H), 4.39 – 4.30 (m, 3H), 4.23 (s, 3H), 3.82 – 3.69 (m, 6H), 3.62 (t, J = 7.5 Hz, 2H), 3.24 (s, 4H). Synthesis of Example 12: 6-(4-(5-((7-(Cyclopropylamino)-4-oxo-3,4-dihydrophthalazin-1 -yl)methyl)- 2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile

6-(4-(5-((7-(Cyclopropylamino)-4-oxo-3,4-dihydrophthalazin-1 -yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0617] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (200 mg, 0.36 mmol), 3-methoxyazetidine (63 mg, 1.10 mmol), Pd 2 (dba) 3 (20 mg, 0.02 mmol), XPhos (42 mg, 0.09 mmol) and sodium tert-butoxide (70 mg, 0.73 mmol) in DMA (5 mL) was stirred at 130 °C under Ar (g) for 1.5 hours. The mixture was cooled to rt, filtered, and the filtrate was purified by prep-HPLC to give 6-(4-(5-((7-(cyclopropylamino)-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl) piperazin-1-yl)nicotinonitrile as a white solid (14 mg, 7% yield). LCMS ESI m/z 524 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.12 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 7.93 (d, J = 8.7 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.48 – 7.40 (m, 1H), 7.36 (dd, J = 6.4, 2.1 Hz, 1H), 7.26 (t, J = 9.0 Hz, 1H), 7.14 (s, 1H), 7.04 (dd, J = 8.7, 1.9 Hz, 1H), 6.95 – 6.88 (m, 2H), 4.21 (s, 2H), 3.82 – 3.66 (m, 6H), 3.30 (d, J = 3.7 Hz, 2H), 2.37 (s, 1H), 0.79 – 0.70 (m, 2H), 0.37 – 0.29 (m, 2H). Synthesis of Example 13: 6-(4-(5-((7-Cyclopropoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile Methyl 2-bromo-4-(vinyloxy)benzoate [0618] A mixture of methyl 2-bromo-4-hydroxybenzoate (2.00 g, 8.66 mmol), 2,4,6-trivinyl- 1,3,5,2,4,6-trioxatriborinane (2.00 g, 8.31 mmol), pyridine (6.85 g, 86.6 mmol) and Cu(OAc) 2 (1.57 g, 8.66 mmol) in DCM (20 mL) was stirred at rt under O 2 (g) for 48 hours. The reaction was concentrated in vacuo, then diluted with water (30 mL) and extracted with EtOAc (30 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 0 to 5%) to afford methyl 2-bromo- 4-(vinyloxy)benzoate as a yellow oil (1.6 g, 72% yield). LCMS ESI m/z 257 [M+H] + . Step 2 Methyl 2-bromo-4-cyclopropoxybenzoate [0619] To a solution of Et 2 Zn (1 M in hexanes, 35 mL, 35 mmol) in DCM (50 mL) was added TFA (3.99 g, 35 mmol) at 0 °C under N 2 (g). The mixture was stirred for 30 min at 0 °C, then diiodomethane (9.38 g, 35 mmol) was added and stirred for another 30 min. To the reaction mixture, a solution of methyl 2-bromo-4-vinyloxy-benzoate (1.5 g, 5.83 mmol) in DCM (10 mL) was added dropwise at 0 °C, then stirred at rt for 16 hours. The mixture was quenched with sat.NaHCO 3 (20 mL) and extracted with DCM (50 mL x 3). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 5%) to afford methyl 2-bromo-4-(cyclopropoxy)benzoate (780 mg, 49% yield) as a light yellow oil. LCMS ESI m/z 271 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 7.83 (d, J = 8.8 Hz, 1H), 7.35 (s, 1H), 7.00 (dd, J = 8.8, 2.8 Hz, 1H), 3.89 (s, 3H), 3.80 – 3.75 (m, 1H), 0.84 – 0.78 (m, 4H). Step 3 2-Bromo-4-cyclopropoxybenzoic acid [0620] A mixture of methyl 2-bromo-4-cyclopropoxybenzoate (780 mg, 2.88 mmol) and LiOH-H 2 O (605 mg, 14.4 mmol) in MeOH (8 mL) and H 2 O (4 mL) was stirred at rt for 3 hours. The organic solvent was removed in vacuo and aq HCl (1N) was added until the solution reached pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 2-bromo-4- cyclopropoxybenzoic acid as a white solid (640 mg, 87% yield). This was used without further purification. LCMS ESI m/z 257 [M+H] + . Step 4 5-Cyclopropoxy-3-hydroxyisobenzofuran-1(3H)-one [0621] A mixture of 2-bromo-4-cyclopropoxybenzoic acid (400 mg, 1.57 mmol) in THF (5 mL) was cooled to -78 °C, then n-BuLi (2.5 M in heptane, 1.25 mL, 3.11 mmol) was addded dropwise under Ar (g). The reaction was stirred at -78 °C for 15 min, then DMF (252 mg, 3.45 mmol) was added. The reaction was kept at -78 °C for 20 min, then quenched with sat. NH4Cl soltion (5 mL). To the resulting mixture, was added aq HCl (1N) until the solution reached pH 5~6, then extracted with EtOAc (20 x 3 mL). The combined organic layer was washed with bine, dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30 to 50%) to give 5-cyclopropoxy-3-hydroxyisobenzofuran-1(3H)-one as a colorless oil (90 mg, 28% yield). LCMS ESI m/z 207 (M+H) + . Step 5 Dimethyl (6-cyclopropoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphon ate [0622] A mixture of 5-cyclopropoxy-3-hydroxyisobenzofuran-1(3H)-one (90 mg, 0.44 mmol) in dimethyl phosphonate (617 mg, 5.6 mmol) was stirred at 100 °C for 3 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 50%) to give dimethyl (6- cyclopropoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate as a yellow oil (80 mg, 61% yield). LCMS ESI m/z 299 [M+H] + . Step 6 6-(4-(5-((6-Cyclopropoxy-3-oxoisobenzofuran-1(3H)-ylidene)me thyl)-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile [0623] A mixture of dimethyl(6-cyclopropoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl) phosphonate (80 mg, 0.27 mmol), 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (109 mg, 0.32 mmol ) and Et 3 N (81 mg, 0.80 mmol) in THF (5 mL) was heated to 60 °C and stirred for 3 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with DCM (10 mL x 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to afford 6-(4-(5-((6-cyclopropoxy- 3-oxoisobenzofuran-1(3H)-ylidene)methyl)-2-fluorobenzoyl) piperazin-1-yl)nicotinonitrile as a white solid (130 mg, 95% yield). This was used without further purification. LCMS ESI m/z 511 [M+H] + . Step 7 6-(4-(5-((7-Cyclopropoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile [0624] To a solution of 6-(4-(5-((6-cyclopropoxy-3-oxoisobenzofuran-1(3H)-ylidene) methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile (140 mg, 0.27 mmol) in THF (5 mL) was added N 2 H 4 •H 2 O (108 mg, 5.4 mmol), then stirred at 60 °C for 3 hours. The reaction was cooled to rt. The solids were collected by filtration, washed with water (5 mL) and dried under vacuo to give 6-(4-(5-((7-cyclopropoxy- 4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)pip erazin-1-yl)nicotinonitrile as a white solid (60 mg, 42% yield). LCMS ESI m/z 525 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.48 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 8.22 – 8.12 (m, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.52 – 7.37 (m, 4H), 7.27 (t, J = 9.0 Hz, 1H), 6.93 (d, J = 9.1 Hz, 1H), 4.31 (s, 2H), 4.04 – 3.94 (m, 1H), 3.74 (d, J = 12.0 Hz, 4H), 3.61 (d, J = 4.0 Hz, 2H), 3.31 (s, 2H), 0.85 (t, J = 6.2 Hz, 2H), 0.64 (s, 2H). Synthesis of Example 14: 6-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile

Methyl 2-bromo-4-hydroxybenzoate [0625] To a solution of 2-bromo-4-hydroxybenzoic (5.00 g, 23.04 mmol) in MeOH (75 mL) was added conc. H 2 SO 4 (4.50 g, 23.0 mmol, 2.50 mL) at rt, then stirred at 70 °C for 4 hours. The reaction was concentrated in vacuum, then diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to afford methyl 2-bromo-4-hydroxybenzoate as a white solid (5.20 g, 97% yield). This was used without further purification. LCMS ESI m/z 231 [M+H] + ). Step 2 methyl 2-bromo-4-cyclobutoxybenzoate [0626] A mixture of methyl 2-bromo-4-hydroxybenzoate (1.20 g, 5.19 mmol), K 2 CO 3 (2.15 g, 15.6 mmol) and bromocyclobutane (855 mg, 6.34 mmol) in DMF (24 mL) was stirred at 75 °C for 18 hours. The reaction was cooled to rt, diluted with H 2 O (5 mL) and extracted with EtOAc (30 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 7%) to give methyl 2-bromo-4-cyclobutoxybenzoate as a colorless oil (1.00 g, 81% yield). LCMS ESI m/z 285 [ Step 3 2-Bromo-4-cyclobutoxybenzoic acid [0627] A mixture of methyl 2-bromo-4-cyclobutoxybenzoate (1.20 g, 4.21 mmol) and LiOH-H 2 O (406 mg, 9.68 mmol) in MeOH (6 mL) and H 2 O (2 mL) was stirred at rt for 14 hours. The organic solvent was removed in vacuo and aq HCl (1N) was added until the solution reached pH 5~6. The solids were collected by filtration, washed with H 2 O and dried under vacuum to give 2-bromo-4-cyclobutoxybenzoic acid as a white solid (1.01 g, 88% yield). The product was used without further purification. LCMS ESI m/z 271 [M+H] + . Step 4 5-Cyclobutoxy-3-hydroxyisobenzofuran-1(3H)-one [0628] A solution of 2-bromo-4-cyclobutoxybenzoic acid (400 mg, 1.48 mmol) in THF (12 mL) was cooled to -78 °C, then n-BuLi (2.5 M in heptane, 1.18 mL, 2.96 mmol) was addded dropwise under Ar (g). The reaction was stirred at -78 °C for 15 min, then DMF (238 mg, 3.26 mmol) was added. The reaction was stirred at -78 °C for 20 min, then quenched with sat. NH 4 Cl soltion (10 mL). The mixture was neutralizated by aq HCl (1N) until the solution reached pH 5~6, then extracted with EtOAc (30 x 3 mL). The combined organic layer was washed with bine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30 to 50%) to give 5-cyclobutoxy-3-hydroxyisobenzofuran-1(3H)-one as a white solid (80 mg, 24% yield). LCMS ESI m/z 221 (M+H) + . Step 5 Dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te [0629] A solution of 5-cyclobutoxy-3-hydroxyisobenzofuran-1(3H)-one (80 mg, 1.80 mmol) in dimethylphosphite (6 mL) was stirred at 100 °C for 2 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 50 to 60 %) to give dimethyl (6-cyclobutoxy-3-oxo-1,3- dihydroisobenzofuran-1-yl)phosphonate as a colorless oil (62 mg, 54% yield). LCMS ESI m/z 313 [M+H] + . Step 6 6-(4-(2,3-Difluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophth alazin-1-yl)methyl)benzoyl)piperazin- 1-yl)nicotinonitrile [0630] A solution of dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (50 mg, 0.16 mmol), 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (65 mg, 0.19 mmol) and Et 3 N (49 mg, 0.48 mmol) in anhydrous THF (5 mL) was stirred at 25 °C under Ar (g) for 15 hours. Hydrazine hydrate (50 mg, 1 mmol) was added and the reaction was stirred at 70 °C for 1 hour. The reaction was cooled to rt, then THF was removed under vacuum. The formed solid was washed with water (10 mL) and petroleum ether/EtOAc (2:1, 10 mL) to give methyl 5-[(7-bromo-4-oxo-3H-phthalazin-1- yl)methyl]-2,3-difluoro-benzoate as a white solid (35 mg, 41% yield). LCMS ESI m/z 539 [M+H] + ). 1 H NMR (400 MHz, CDCl 3 ) δ 10.04 (s, 1H), 8.42 (d, J = 2.2 Hz, 1H), 8.35 (d, J = 8.8 Hz, 1H), 7.65 (dd, J = 9.0, 2.3 Hz, 1H), 7.38 – 7.30 (m, 2H), 7.20 (dd, J = 8.8, 2.4 Hz, 1H), 7.07 (t, J = 8.8 Hz, 1H), 6.91 (d, J = 2.3 Hz, 1H), 6.62 (d, J = 9.0 Hz, 1H), 4.67 (d, J = 7.0 Hz, 1H), 4.22 (s, 2H), 3.91 – 3.87 (m, 2H), 3.79 – 3.76 (m, 2H), 3.69 (m, 2H), 3.54 – 3.33 (m, 2H), 2.42 (m, 2H), 2.23 – 2.09 (m, 2H), 1.96 –1.84 (m, 1H), 1.84 – 1.70 (m, 1H). Synthesis of Example 15: 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-2-ynylamino)-3,4-dihydropht halazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

6-(4-(2-Fluoro-5-((4-oxo-7-(prop-2-ynylamino)-3,4-dihydropht halazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0631] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (200 mg, 0.37 mmol), prop-2-yn-1-amine (403 mg, 7.33 mmol), Pd(OAc) 2 (17 mg, 0.07 mmol), Brettphos (37 mg, 0.07 mmol) and sodium tert-butoxide (71 mg, 0.74 mmol) in 1,4-dioxane (10 mL) was stirred at 100°C for 8 hours. The reaction was cooled to rt, then the solids were filtered off. The filtrate was concentrated in vacuo and the residue was purified by prep- HPLC to give 6-(4-(2-fluoro-5-((4-oxo-7-(prop-2-ynylamino)-3,4-dihydropht halazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile as a white solid (35 mg, 18% yield). LCMS ESI m/z 522 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.66 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 8.23 (d, J = 8.2 Hz, 1H), 7.93 – 7.89 (m, 2H), 7.79 (dd, J = 8.2, 1.3 Hz, 1H), 7.44 – 7.37 (m, 2H), 7.26 (t, J = 9.0 Hz, 1H), 6.93 (d, J = 9.1 Hz, 1H), 4.35 (s, 2H), 3.79 – 3.70 (m, 4H), 3.65 – 3.57 (m, 4H), 3.31 – 3.27 (m, 2H), 2.76 (s, 1H). Synthesis of Example 16: 6-(4-(2-Fluoro-5-((7-(methylamino)-4-oxo-3,4-dihydrophthalaz in-1- yl)methyl)benzoyl) piperazin-1-yl)nicotinonitrile

6-(4-(2-Fluoro-5-((7-(methylamino)-4-oxo-3,4-dihydrophthalaz in-1-yl)methyl)benzoyl) piperazin-1- yl)nicotinonitrile [0632] To a solution of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (50.0 mg, 91.3 μmol) in DMA (3 mL) was added methanamine hydrochloride (47.49 mg, 456.72 μmol, HCl), t-BuONa (61.45 mg, 0.639 mmol) , Pd 2 (dba) 3 (8.4 mg, 9.1 μmol) , and XPhos (8.71 mg, 18.3 μmol). This reaction mixture was heated to 130 °C for 16 hours. The mixture was purified by Prep-HPLC to give 6-(4-(2-fluoro-5-((7- (methylamino)-4-oxo-34-dihydrophthalazin-1-yl)methyl)benzoyl )piperazin-1-yl)nicotinonitrile (10.0 mg, 20% yield) as a white solid. LCMS ESI m/z 497.7 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.07 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 7.95 – 7.86 (m, 2H), 7.47 – 7.35 (m, 2H), 7.25 (t, J = 9.0 Hz, 1H), 6.98 (dd, J = 8.8, 2.1 Hz, 1H), 6.92 (d, J = 9.1 Hz, 1H), 6.78 (d, J = 5.0 Hz, 1H), 6.63 (d, J = 2.0 Hz, 1H), 4.21 (s, 2H), 3.81 – 3.68 (m, 4H), 3.60 (s, 2H), 3.31 – 3.28 (m, 2H), 2.73 (d, J = 4.9 Hz, 3H). Synthesis of Example 17: 6-(4-(2-Fluoro-5-((7-(methyl(prop-2-ynyl)amino)-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile 6-(4-(2-Fluoro-5-((7-(methyl(prop-2-ynyl)amino)-4-oxo-3,4-di hydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0633] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (200 mg, 0.37 mmol), N-methylprop-2-yn-1-amine (505 mg, 7.33 mmol), Pd(OAc) 2 (17 mg, 0.07 mmol), Brettphos (37 mg, 0.07 mmol) and sodium tert-butoxide (71 mg, 0.74 mmol) in 1,4-dioxane (10 mL) was stirred at 100°C for 8 hours. The reaction was cooled to rt, then the solids were filtered off. The filtrate was concentrated in vacuo and the residue was purified by silica gel chromatography (EtOAc/petroleum ether = 80% to 100%) to give 6-(4-(2-fluoro-5-((7- (methyl(prop-2-ynyl)amino)-4-oxo-3,4-dihydrophthalazin-1-yl) methyl) benzoyl) piperazin-1- yl)nicotinonitrile as a white solid (45 mg, 23% Yield). LCMS ESI m/z 536 (M+H) + . 1 H NMR (400 MHz, MeOD) δ 8.42 (d, J = 1.9 Hz, 1H), 8.32 (d, J = 8.2 Hz, 1H), 7.95 (s, 1H), 7.76 (dd, J = 9.1, 2.3 Hz, 2H), 7.50 – 7.45 (m, 1H), 7.40 – 7.36 (m, 1H), 7.18 (t, J = 9.0 Hz, 1H), 6.87 (d, J = 9.1 Hz, 1H), 4.37 (s, 2H), 3.86 – 3.81 (m, 4H), 3.70 – 3.66 (m, 4H), 3.44 – 339 (m 2H) 251 (s, 3H). Synthesis of Example 18: 6-(4-(5-((7-(3,3-Difluoroazetidin-1-yl)-4-oxo-3,4-dihydropht halazin-1- yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile 6-(4-(5-((7-(3,3-Difluoroazetidin-1-yl)-4-oxo-3,4-dihydropht halazin-1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0634] To a solution of 6-[4-[5-[(7-bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro- benzoyl]piperazin-1-yl]pyridine-3-carbonitrile (99.6 mg, 182 μmol) in DMA (2 mL) was added 3,3- difluoroazetidine hydrochloride (118 mg, 910 μmol) , t-BuONa (122 mg, 1.27 mmol) , Pd 2 (dba) 3 (16.7 mg, 18.2 μmol), and XPhos (17.4 mg, 36.4 μmol). The resulting mixture was heated to 130 °C for 16 hours. The mixture was purified by Prep-HPLC to give 6-[4-[5-[[7-(3,3-difluoroazetidin-1-yl)-4-oxo-3H- phthalazin-1-yl]methyl]-2-fluoro-benzoyl]piperazin-1-yl]pyri dine-3-carbonitrile (18.03 mg, 16% yield) as a white solid. LCMS ESI m/z 559.7 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.27 (s, 1H), 8.52 (d, J = 2.0 Hz, 1H), 8.09 (d, J = 8.7 Hz, 1H), 7.90 (dd, J = 9.1, 2.4 Hz, 1H), 7.48 – 7.42 (m, 2H), 7.26 (t, J = 9.3 Hz, 1H), 7.01 (dd, J = 8.7, 2.2 Hz, 1H), 6.93 (d, J = 9.1 Hz, 1H), 6.82 (d, J = 2.2 Hz, 1H), 4.44 (t, J = 12.3 Hz, 4H), 4.26 (s, 2H), 3.76 (m, 4H), 3.63 (m, 2H), 3.36 (m, 2H). Synthesis of Example 19: 6-(4-(5-((7-(But-2-ynyloxy)-4-oxo-3,4-dihydrophthalazin-1-yl )methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile 4-(3-(4-(5-cyanopyridin-2-yl)piperazine-1-carbonyl)-4-fluoro benzyl)-1-oxo-1,2-dihydrophthalazin-6- ylboronic acid [0635] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (400 mg, 0.73 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'- bi(1,3,2-dioxaborolane) (556 mg, 2.19 mmol), Cs 2 CO 3 (714 mg, 2.19 mmol) and Pd(dppf)Cl 2 (58 mg, 0.08 mmol) in DMSO (10 mL) was stirred at 80°C under Ar (g) for 16 hours. The reaction was cooled to rt, diluted with water (20 mL) and extracted with EtOAc (3 × 20 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (DCM/MeOH = 0% to 10%) to give 4-(3-(4-(5-cyanopyridin-2-yl)piperazine-1- carbonyl)-4-fluorobenzyl)-1-oxo-1,2-dihydrophthalazin-6-ylbo ronic acid (240 mg, 64% yield) as a white solid. LCMS ESI m/z: 513 [M+H] + . Step 2 6-(4-(2-fluoro-5-((7-hydroxy-4-oxo-3,4-dihydrophthalazin-1-y l)methyl)benzoyl) piperazin-1- yl)nicotinonitrile [0636] A solution of 4-(3-(4-(5-cyanopyridin-2-yl)piperazine-1-carbonyl)-4-fluoro benzyl)-1-oxo-1,2- dihydrophthalazin-6-ylboronic acid (240 mg, 0.47 mmol) and H 2 O 2 (180 mg, 4.70 mmol) in dioxane (8 mL) were stirred at rt for 10 mins, then NaOH (56 mg, 1.41 mmol) in H 2 O (1 mL) was added. The reaction mixture was stirred rt for 16 hours, diluted with H2O (10 mL) and extracted with EtOAc (3 × 20 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (DCM/MeOH = 0% to 10%) to give 6-(4-(2-fluoro-5-((7- hydroxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)benzoyl)pipe razin-1-yl)nicotinonitrile (40 mg, 18% yield) as a white solid. LCMS ESI m/z: 485 [M+H] + . Step 3 6-(4-(5-((7-(but-2-ynyloxy)-4-oxo-3,4-dihydrophthalazin-1-yl )methyl)-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile [0637] A mixture of 6-(4-(2-fluoro-5-((7-hydroxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (40 mg 008 mmol), 1-bromobut-2-yne (16 mg, 0.12 mmol) and K 2 CO 3 (33 mg, 0.24 mmol) in DMF (5 mL) was stirred at 50°C for 1 hour. The solids were filtered off, and the filtrate was concentrated in vacuo. The residue was purified by pre-HPLC to give 2- chloro-4-isopropoxy-1,8-naphthyridine (25 mg, 58% yield) as a white solid. LCMS ESI m/z: 537 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.48 (s, 1H), 8.52 (d, J= 2.2 Hz, 1H), 8.19 (d, J= 8.8 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.48 –7.39 (m, 3H), 7.36 –7.33 (m, 1H), 7.26 (t, J= 9.0 Hz, 1H), 6.93 (d, J = 9.1 Hz, 1H), 4.91 (d, J= 2.2 Hz, 2H), 4.30 (s, 2H), 3.75 (m, 4H), 3.62 (m, 2H), 3.32 –3.30 (m, 2H), 1.78 (s, 3H). Synthesis of Example 20: 6-(4-(5-((7-(cyclopropylmethylamino)-4-oxo-3,4-dihydrophthal azin-1- yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile 6-(4-(5-((7-(Cyclopropylmethylamino)-4-oxo-3,4-dihydrophthal azin-1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0638] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (200 mg, 0.37 mmol), cyclopropylmethanamine (105 mg, 1.48 mmol), Pd 2 (dba) 3 (65 mg, 0.07 mmol), XPhos (33 mg, 0.07 mmol) and t-BuONa (107 mg, 1.11 mmol) in DMA (10 mL) was stirred at 130°C for 14 hours. The reaction was cooled to rt, then the solids were filtered off. The filtrate was concentrated in vacuo and the residue was purified by prep-HPLC to give 6-(4-(5-((7-(cyclopropylmethylamino)-4-oxo-3,4-dihydrophthal azin-1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile as a white solid (30 mg, 15% Yield). LCMS ESI m/z 538 (M+H) + . 1 H NMR (400 MHz, DMSO–d 6 ) δ 12.06 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 7.90 (dd, J = 9.0, 2.5 Hz, 2H), 7.46 – 7.40 (m, 1H), 7.39 – 7.34 (m, 1H), 7.25 (t, J = 9.0 Hz, 1H), 7.03 (dd, J = 8.9, 2.1 Hz, 1H), 6.92 (d, J = 9.2 Hz, 1H), 6.86 (t, J = 5.2 Hz, 1H), 6.69 (s, 1H), 4.20 (s, 2H), 3.76 (s, 2H), 3.73 (s, 2H), 3.60 (s, 2H), 3.31 – 3.27 (m, 2H), 2.95 (t, 2H), 1.05 – 0.90 (m, 1H), 0.48 – 0.40 (m, 2H), 0.24 – 0.15 (m, 2H). Synthesis of Example 21: 6-(4-(5-((7-(Difluoromethoxy)-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)- 2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile 5-(Difluoromethoxy)isobenzofuran-1(3H)-one [0639] To a solution of 5-hydroxy-3H-isobenzofuran-1-one (2.00 g, 13.3 mmol) and sodium 2-chloro- 2,2-difluoroacetate (6.09 g, 40.0 mmol) in DMF (20 mL) was added K 2 CO 3 (5.52 g, 40.0 mmol), then the mixture was stirred at 100 ℃ for 48 hours. The mixture was cooled to rt, poured into a saturated solution of NH 4 Cl (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over MgSO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 15%) to give 5-(difluoromethoxy)-3H-isobenzofuran-1- one as a white solid (1.83 g, 28% yield). LCMS ESI m/z 201 (M+H) + . 1 H NMR (400 MHz, CDCl 3 ) δ 7.94 (d, J = 8.4 Hz, 1H), 7.28 (dd, J = 8.5 Hz, 1H), 7.24 (s, 1H), 6.63 (t, J = 72.4 Hz, 1H), 5.32 (s, 2H). Step 2 3-Bromo-5-(difluoromethoxy)isobenzofuran-1(3H)-one [0640] A mixture of 5-(difluoromethoxy)-3H-isobenzofuran-1-one (750 mg, 3.75 mmol), NBS (801 mg, 4.5 mmol) and AIBN (62 mg, 3.75 mmol) in CCl 4 (10 mL) was heated to 80 °C and stirred for 16 hours. The mixture was cooled to rt, diluted with H 2 O (30 mL) and extracted with DCM (30 mL x 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 9%) to obtain 3-bromo-5- (difluoromethoxy)-3H-isobenzofuran-1-one as a white solid (580 mg, 55%). LCMS ESI m/z 279 (M+H) + . Step 3 5-(Difluoromethoxy)-3-hydroxyisobenzofuran-1(3H)-one [0641] A solution of 3-bromo-5-(difluoromethoxy)-3H-isobenzofuran-1-one (580 mg, 2.08 mmol) in H 2 O (25 mL) was stirred at 100 °C for 3 hours. The reaction was cooled to rt and extracted with DCM (50 mL x 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give 5-(difluoromethoxy)-3-hydroxy-3H-isobenzofuran-1-one as a light yellow oil (390 mg, 87% yield). LCMS ESI m/z 217 (M+H) + . Step 4 Dimethyl (6-(difluoromethoxy)-3-oxo-1,3-dihydroisobenzofuran-1-yl)pho sphonate [0642] A solution of 5-(difluoromethoxy)-3-hydroxy-3H-isobenzofuran-1-one (390 mg, 1.80 mmol) in dimethylphosphite (30 mL) was stirred at 100 °C for 2 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with water and brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give dimethyl (6- (difluoromethoxy)-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosph onate as a light yellow oil (230 mg, 41% yield). This was used without further purification. LCMS ESI m/z 309 (M+H) + . Step 5 6-(4-(5-((6-(Difluoromethoxy)-3-oxoisobenzofuran-1(3H)-ylide ne)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0643] To a solution of (6-(difluoromethoxy)-3-oxo-1,3-dihydroisobenzofuran-1-yl)pho sphonate (230 mg, 0.75 mmol) in THF (dry, 5 mL) under Ar (g), was added 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1- yl)nicotinonitrile (303 mg, 0.90 mmol) and triethylamine (197 mg, 1.95 mmol). The reaction mixture was stirred at rt for 12 hours, then poured into water (10 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to give 6-[4-[5-[6-(difluoromethoxy)-3-oxo-isobenzofuran-1-ylidene]m ethyl]-2-fluoro-benzoyl]piperazin-1- yl]pyridine-3-carbonitrile as a yellow solid (397 mg, 41% yield). LCMS ESI m/z 521 (M+H) + . Step 6 6-(4-(5-((7-(Difluoromethoxy)-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)-2-fluorobenzoyl)piperazin- 1-yl)nicotinonitrile [0644] To a solution of 6-[4-[5-[6-(difluoromethoxy)-3-oxo-isobenzofuran-1-ylidene]m ethyl]-2-fluoro- benzoyl]piperazin-1-yl]pyridine-3-carbonitrile (397 mg, 0.63 mmol) in THF (10 mL) was added N 2 H 4 •H 2 O (655 mg, 11.14 mmol), and the mixture was stirred at 70 °C for 4 hours. The solids were filtered, washed with water (5 mL x 2), and dried under vacuum to give 6-(4-(5-((7-(difluoromethoxy)-4- oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)piper azin-1-yl)nicotinonitrile as a white solid (260 mg, 44% yield). LCMS ESI m/z 535 (M+H) + . 1 H NMR (400 MHz, DMSO-d6) δ 12.66 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 8.32 (d, J = 8.6 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.69 – 7.60 (m, 2H), 7.51 – 7.21 (m, 4H), 6.92 (d, J = 9.1 Hz, 1H), 4.33 (s, 2H), 3.77-3.73 (m, 4H), 3.61 (m, 2H), 3.31 m, 2H). Synthesis of Example 22: 6-(4-(2-Fluoro-5-((4-oxo-7-((2,2,2-trifluoroethyl)amino)-3,4 - dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile 6-(4-(2-Fluoro-5-((4-oxo-7-((2,2,2-trifluoroethyl)amino)-3,4 -dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0645] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (200 mg, 0.36 mmol), 2,2,2-trifluoroethan-1-amine (108 mg, 1.10 mmol), Pd 2 (dba) 3 (20 mg, 0.02 mmol), XPhos (17 mg, 0.04 mmol) and sodium tert-butoxide (105 mg, 1.10 mmol) in DMA (5 mL) was stirred at 130 °C for 2 hours. The mixture was cooled to rt, filtered and the filtrate was purified by prep-HPLC to give 6-(4-(2-fluoro-5-((4-oxo-7-((2,2,2- trifluoroethyl)amino)-3,4-dihydrophthalazin-1-yl)methyl)benz oyl) piperazin-1-yl)nicotinonitrile as a white solid (19 mg, 9% yield). LCMS ESI m/z 566 (M+H) + . 1 H NMR (400 MHz, DMSO-d6) δ 12.17 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 7.97 (d, J = 8.8 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.50 – 7.42 (m, 1H), 7.36 (dd, J = 6.5, 2.0 Hz, 1H), 7.30 – 7.15 (m, 3H), 7.02 (d, J = 1.8 Hz, 1H), 6.91 (d, J = 9.1 Hz, 1H), 4.23 (s, 2H), 4.20 – 4.08 (m, 2H), 3.74 (m, 4H), 3.59 – 3.51 (m, 2H), 3.31 – 3.26 (m, 2H). Synthesis of Example 23: 6-(4-(2-Fluoro-5-((7-(6-hydroxy-2-azaspiro[3.3]heptan-2-yl)- 4-oxo-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile 2-Azaspiro[3.3]heptan-6-ol hydrochloride [0646] To a solution of tert-butyl 6-hydroxy-2-azaspiro[3.3]heptane-2-carboxylate (200 mg, 937 μmol) in HCl/MeOH (4 M, 4 mL) at 25 °C. The reaction mixture was stirred at 25 °C for 5 hours. The mixture was concentrated to give 2-azaspiro[3.3]heptan-6-ol hydrochloride (100 mg, 71% yield) as a white oil. Step 2

6-(4-(2-Fluoro-5-((7-(6-hydroxy-2-azaspiro[3.3]heptan-2-yl)- 4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0647] To a solution of 6-[4-[5-[(7-bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro- benzoyl]piperazin-1-yl]pyridine-3-carbonitrile (150 mg, 274 μmol) in DMF (4 mL) was added 2- azaspiro[3.3]heptan-6-ol hydrochloride (62.0 mg, 548 μmol), t-BuOK (185 mg, 1.64 mmol), X-Phos (26.1 mg, 54.8 μmol) and Pd 2 (dba) 3 (15.8 mg, 27.4 μmol) at 25 °C under N 2 (g). The reaction was stirred at 110 °C under microwave for 45 min. The mixture was filtered and purified by Pre-HPLC to give 6-(4- (2-fluoro-5-((7-(6-hydroxy-2-azaspiro[3.3]heptan-2-yl)-4-oxo -3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (13.5 mg, 9% yield) as a white solid. LCMS ESI m/z 580.3 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.14 (s, 1H), 8.52 (d, J = 2.3 Hz, 1H), 7.99 (d, J = 8.7 Hz, 1H), 7.90 (dd, J = 9.1, 2.4 Hz, 1H), 7.41 (t, J = 6.5 Hz, 2H), 7.25 (t, J = 9.2 Hz, 1H), 6.92 (d, J = 9.0 Hz, 1H), 6.79 (dd, J = 8.7, 2.2 Hz, 1H), 6.51 (d, J = 2.1 Hz, 1H), 5.07 (d, J = 6.1 Hz, 1H), 4.21 (s, 2H), 4.01 (dd, J = 13.4, 7.0 Hz, 1H), 3.89 (d, J = 17.3 Hz, 4H), 3.75 (m, 4H), 3.62 (m, 2H), 2.46 (dd, J = 7.6, 4.9 Hz, 4H), 2.07 – 1.97 (m, 2H). Synthesis of Example 24: 6-(4-(2-Fluoro-5-((4-oxo-7-(3,3,3-trifluoroprop-1-ynyl)-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile

6-(4-(2-Fluoro-5-((4-oxo-7-(3,3,3-trifluoroprop-1-ynyl)-3,4- dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0648] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (200 mg, 0.37 mmol), tributyl(3,3,3-trifluoroprop-1- ynyl)stannane (422 mg, 1.10 mmol) and Pd(dppf)Cl 2 (58 mg, 0.08 mmol) in 1,4-dioxane (10 mL) were stirred at 100 °C under Ar (g) for 14 hours. The mixture was cooled to rt, diluted with water (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 80 % to 100 %) to give 6-(4-(2-fluoro-5-((4-oxo-7-(3,3,3-trifluoroprop-1-ynyl)-3,4- dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile as a white solid (35 mg, 17% yield). LCMS ESI m/z 561 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.81 (s, 1H), 8.53 – 8.45 (m, 2H), 8.32 (d, J = 8.2 Hz, 1H), 8.12 (d, J = 9.4 Hz, 1H), 7.90 (dd, J = 9.1, 2.4 Hz, 1H), 7.46 – 7.27 (m, 3H), 6.92 (d, J = 9.1 Hz, 1H), 4.38 (s, 2H), 3.80-3.70 (m, 4H), 3.65 – 3.58 (m, 2H), 3.32 – 3.29 (m, 2H). Synthesis of Example 25: 6-(4-(2-Fluoro-5-((4-oxo-7-(3,3,3-trifluoropropyl)-3,4-dihyd rophthalazin- 1-yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

6-(4-(2-Fluoro-5-((4-oxo-7-(3,3,3-trifluoropropyl)-3,4-dihyd rophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0649] A mixture of 6-(4-(2-fluoro-5-((4-oxo-7-(3,3,3-trifluoroprop-1-ynyl)-3,4- dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (22 mg, 0.04 mmol) and Pd/C (10%, 4 mg) in MeOH (2 mL) were stirred at rt for 14 hours under H 2 (g). The mixture was filtered and concentrated in vacuo. The residue was purified by prep-HPLC to afford 6-(4-(2-fluoro-5-((4-oxo-7-(3,3,3-trifluoropropyl)-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile as a white solid (10 mg, 45% yield). LCMS ESI m/z 561 (M+H) + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.04 (s, 1H), 8.45 – 8.38 (m, 2H), 7.68 – 7.59 (m, 2H), 7.53 (s, 1H), 7.39 – 7.31 (m, 2H), 7.08 (t, J = 8.8 Hz, 1H), 6.61 (d, J = 8.9 Hz, 1H), 4.28 (s, 2H), 3.89 (m, 2H), 3.77 (m, 2H), 3.69 (t, J = 4.9 Hz, 2H), 3.43 (m, 2H), 3.03 (t, J = 7.6 Hz, 2H), 2.51 – 2.35 (m, 2H). Synthesis of Example 26: 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-2-yn-1-yloxy)-3,4-dihydroph thalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

6-(4-(2-Fluoro-5-((4-oxo-7-(prop-2-yn-1-yloxy)-3,4-dihydroph thalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0650] A mixture of 6-(4-(2-fluoro-5-((4-oxo-7-(prop-2-yn-1-yloxy)-3,4-dihydroph thalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (100 mg, 0.21 mmol), 3-bromoprop-1-yne (26 mg, 0.22 mmol) and K 2 CO 3 (86 mg, 0.63 mmol) in DMF (5 mL) was stirred at 50 °C for 1 hour. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was concentrated in vacuo and the residue was purified by prep-HPLC to give 6-(4-(2-fluoro-5-((4- oxo-7-(prop-2-yn-1-yloxy)-3,4-dihydrophthalazin-1-yl)methyl) benzoyl)piperazin-1-yl)nicotinonitrile as a white solid (40 mg, 37% yield). LCMS ESI m/z 523 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 9.95 (s, 1H), 8.34 (dd, J = 10.0, 5.5 Hz, 2H), 7.59 (dd, J = 9.0, 2.3 Hz, 1H), 7.29 (dd, J = 12.8, 4.1 Hz, 3H), 7.14 (d, J = 2.4 Hz, 1H), 7.00 (t, J = 8.7 Hz, 1H), 6.55 (d, J = 9.0 Hz, 1H), 4.71 (d, J = 2.4 Hz, 2H), 4.18 (s, 2H), 3.82 (m, 2H), 3.71 (m, 2H), 3.62 (m, 2H), 3.36 (s, 2H), 2.58 (t, J = 2.4 Hz, 1H). Synthesis of Example 27: (E)-6-(4-(2-Fluoro-5-((4-oxo-7-(3,3,3-trifluoroprop-1-enyl)- 3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile

(E)-5-(3,3,3-Trifluoroprop-1-en-1-yl)isobenzofuran-1(3H)-one [0651] To a solution of 5-bromoisobenzofuran-1(3H)-one (4.5 g, 21.12 mmol) in NMP (50 mL) was added 2-(trifluoromethyl)prop-2-enoic acid (4.14 g, 29.57 mmol), Pd(OAc) 2 (474 mg, 2.11 mmol) and CuO (1.70 g, 21.1 mmol) at 25 °C under N 2 (g). The reaction mixture was stirred at 130 °C for 15 hours. Water (20 mL) and EtOAc (30 mL) were added to the mixture. The organic layer was separated, washed with brine, dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 20/1 to 2/1) to give (E)-5-(3,3,3-trifluoroprop-1-en-1- yl)isobenzofuran-1(3H)-one (1.6 g, 33.2% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 7.95 (d, J = 8.0 Hz, 1H), 7.63 (d, J = 8.0 Hz, 1H), 7.57 (s, 1H), 7.29 – 7.22 (m, 1H), 6.36 (dq, J = 16.1, 6.3 Hz, 1H), 5.35 (s, 2H). Step 2 (E)-3-Bromo-5-(3,3,3-trifluoroprop-1-en-1-yl)isobenzofuran-1 (3H)-one [0652] To a solution of (E)-5-(3,3,3-trifluoroprop-1-en-1-yl)isobenzofuran-1(3H)-one (1.00 g, 4.38 mmol) in CCl 4 (10 mL) was added 1-bromopyrrolidine-2,5-dione (1.17 g, 6.57 mmol, 557.72 μL), AIBN (72.0 mg, 438 μmol) at 25 °C under N 2 (g). The reaction mixture was stirred at 80 °C for 15 hours. Water (20 mL) and EtOAc (30 mL) were added to the mixture. The organic layer was separated, washed with brine, dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleumether/EtOAc = 20/1 to 0/1) to give (E)-3-bromo-5-(3,3,3-trifluoroprop-1-en-1- yl)isobenzofuran-1(3H)-one (1.00 g, 74% yield) as a white solid. Step 3 (E)-3-Hydroxy-5-(3,3,3-trifluoroprop-1-en-1-yl)isobenzofuran -1(3H)-one [0653] A solution of (E)-3-bromo-5-(3,3,3-trifluoroprop-1-en-1-yl)isobenzofuran-1 (3H)-one (1.5 g, 4.88 mmol) in 1 mol/L KOH (15 mL) was stirred at 100 °C for 1 hour. The mixture was acidified with 1N HCl until the solution reached pH 2~3. The solids were collected by filtration, washed with water and dried under vacuum to give (E)-3-hydroxy-5-(3,3,3-trifluoroprop-1-en-1-yl)isobenzofuran -1(3H)-one (1 g, 84% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.27 (d, J = 8.5 Hz, 1H), 8.04 (s, 1H), 7.96 (d, J = 8.0 Hz, 1H), 7.87 (d, J = 7.9 Hz, 1H), 7.54 (dd, J = 16.2, 1.9 Hz, 1H), 7.15 – 6.99 (m, 1H), 6.70 (d, J = 8.3 Hz, 1H). Step 4 Dimethyl (E)-(3-oxo-6-(3,3,3-trifluoroprop-1-en-1-yl)-1,3-dihydroisob enzofuran-1-yl)phosphonate [0654] A solution of (E)-3-hydroxy-5-(3,3,3-trifluoroprop-1-en-1-yl)isobenzofuran -1(3H)-one (1.00 g, 4.10 mmol) in dimethylphosphite (4 mL) was stirred at 100 °C for 3 hours. Water (20 mL) and EtOAc (30 mL) were added to the mixture. The organic layer was separated, washed with brine, dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleumether/EtOAc = 20/1 to 1/2) to give dimethyl (E)-(3-oxo-6-(3,3,3-trifluoroprop- 1-en-1-yl)-1,3-dihydroisobenzofuran-1-yl)phosphonate (750 mg, 55% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.06 – 7.92 (m, 3H), 7.61 (dd, J = 16.3, 2.1 Hz, 1H), 7.07 (dq, J = 16.2, 6.9 Hz, 1H), 6.37 (d, J = 11.2 Hz, 1H), 3.82 (d, J = 10.9 Hz, 3H), 3.65 (d, J = 10.7 Hz, 3H). Step 5 6-(4-(2-Fluoro-5-(((Z)-3-oxo-6-((E)-3,3,3-trifluoroprop-1-en -1-yl)isobenzofuran-1(3H)- ylidene)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0655] To a solution of (E)-(3-oxo-6-(3,3,3-trifluoroprop-1-en-1-yl)-1,3-dihydroisob enzofuran-1- yl)phosphonate (300 mg, 892.33 μmol) in THF (4 mL) was added 6-[4-(2-fluoro-5-formyl- benzoyl)piperazin-1-yl]pyridine-3-carbonitrile (305 mg, 901 μmol), Et 3 N (271 mg, 2.68 mmol, 373 μL) at 25 °C. The reaction mixture was stirred at 25 °C for 8 hours. Water (10 mL) and EtOAc (10 mL) were added to the mixture, the organic layer was separated, washed with brine, dried over Na 2 SO 4 and concentrated to give 6-(4-(2-fluoro-5-(((Z)-3-oxo-6-((E)-3,3,3-trifluoroprop-1-en -1-yl)isobenzofuran- 1(3H)-ylidene)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (280 mg crude, 57% yield) as a white solid. Step 6 (E)-6-(4-(2-Fluoro-5-((4-oxo-7-(3,3,3-trifluoroprop-1-en-1-y l)-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0656] To a solution of 6-(4-(2-fluoro-5-(((Z)-3-oxo-6-((E)-3,3,3-trifluoroprop-1-en -1- yl)isobenzofuran-1(3H)-ylidene)methyl)benzoyl)piperazin-1-yl )nicotinonitrile (280 mg, 511 μmol) in THF (2 mL) was added hydrazine hydrate (2.05 g, 32.8 mmol, 2 mL, 80% purity) at 25 °C under N 2 (g). The reaction mixture was stirred at 70 °C for 2 hours. The mixture was filtered and purified by prep-HPLC to give (E)-6-(4-(2-fluoro-5-((4-oxo-7-(3,3,3-trifluoroprop-1-en-1-y l)-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile (49.8 mg, 17 % yield) as a white solid. LCMS ESI m/z 562.5 + 1 (400 MHz, DMSO-d 6 ) δ 12.66 (s, 1H), 8.51 (d, J = 1.9 Hz, 1H), 8.33 (s, 1H), 8.28 (d, J = 8.3 Hz, 1H), 8.16 (dd, J = 8.3, 1.2 Hz, 1H), 7.89 (dd, J = 9.1, 2.4 Hz, 1H), 7.56 (dd, J = 16.3, 2.1 Hz, 1H), 7.50 (dd, J = 10.5, 4.1 Hz, 2H), 7.27 (dd, J = 12.6, 6.0 Hz, 1H), 7.15 – 7.02 (m, 1H), 6.91 (d, J = 9.1 Hz, 1H), 4.36 (s, 2H), 3.75 (m, 4H), 3.59 (m, 2H), 3.30 (m, 2H). Synthesis of Example 28: 6-(4-(5-((7-(Dimethylamino)-4-oxo-3,4-dihydrophthalazin-1-yl )methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile 6-(4-(5-((7-(dimethylamino)-4-oxo-3,4-dihydrophthalazin-1-yl )methyl)-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile [0657] To a solution of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (100 mg, 183 μmol) in DMA (3 mL) was added dimethylamine (33.0 mg, 731 μmol), t-BuONa (44.0 mg, 457 μmol), Pd 2 (dba) 3 (1.67 mg, 1.83 μmol), and XPhos (1.74 mg, 3.65 μmol). The reaction mixture was heated to 110 °C for 16 hours. The mixture was filtered and concentrated to the crude. The crude was purified by prep-HPLC to give 6-(4-(5- ((7-(dimethylamino)-4-oxo-3,4-dihydrophthalazin-1-yl)methyl) -2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile (30 mg, 31% yield) as a white solid. LCMS ESI m/z 511.7 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.13 (s, 1H), 8.52 (d, J = 2.3 Hz, 1H), 8.01 (d, J = 9.0 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.44 (t, J = 5.0 Hz, 2H), 7.28 – 7.16 (m, 2H), 6.93 (d, J = 9.1 Hz, 1H), 6.76 (d, J = 2.3 Hz, 1H), 4.27 (s, 2H), 3.75 (m, 4H), 3.61 (s, 2H), 3.23 (m, 2H), 3.02 (s, 6H). Synthesis of Example 29: 6-(4-(5-((7-(Cyclobutylamino)-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)- 2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile 6-(4-(5-((7-(Cyclobutylamino)-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)-2-fluorobenzoyl)piperazin- 1-yl)nicotinonitrile [0658] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (200 mg 037 mmol) cyclobutanamine (130 mg, 1.83 mmol), Pd(OAc) 2 (15 mg, 0.07 mmol), Brettphos (39 mg, 0.07 mmol) and NaOtBu (70 mg, 0.73 mmol) in DMA (2 mL) was stirred at 130 °C under Ar (g) for 1 hour. The reaction was cooled to rt and purified by pre-HPLC to give 6-(4-(5-((7-(cyclobutylamino)-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile as a white solid (19.6 mg, 10% yield). LCMS ESI m/z 538 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 9.62 (s, 1H), 8.42 (d, J = 2.0 Hz, 1H), 8.18 (d, J = 8.7 Hz, 1H), 7.66 (dd, J = 9.0, 2.3 Hz, 1H), 7.37 – 7.34 (m, 1H), 7.33 – 7.29 (m, 1H), 7.06 (t, J = 8.8 Hz, 1H), 6.86 (dd, J = 8.7, 2.2 Hz, 1H), 6.62 (d, J = 9.0 Hz, 1H), 6.48 (d, J = 2.2 Hz, 1H), 4.62 (d, J = 6.2 Hz, 1H), 4.18 (s, 2H), 3.94 – 3.86 (m, 3H), 3.81 – 3.75 (m, 2H), 3.68 (m, 2H), 3.43 (m, 2H), 2.45 – 2.37 (m, 2H), 1.86 (m, 4H). Synthesis of Example 30: 6-(4-(2-fluoro-5-((4-oxo-7-(trifluoromethoxy)-3,4-dihydropht halazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile 3-Hydroxy-5-(trifluoromethoxy)isobenzofuran-1(3H)-one [0659] A solution of 2-bromo-4-(trifluoromethoxy)benzoic acid (200 mg, 0.70 mmol) in THF (6 mL) was cooled to -78°C, then n-BuLi (2.5M in heptane, 0.56 mL, 1.4 mmol) was addded dropwise under Ar (g). The reaction was stirred at -78°C for 15 min, then DMF (112 mg, 1.54 mmol) was added. The reaction was kept at -78 °C for 20 min, then quenched with sat. NH4Cl soltion (50 mL). The resulting mixture was added aq HCl (1N) until the solution reached pH 5~6, then extracted with EtOAc (30 x 3 mL). The combined organic layer was washed with bine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30 to 50%) to give 3-hydroxy-5-(trifluoromethoxy)isobenzofuran-1(3H)-one as a white solid (75 mg, 45% yield). LCMS ESI m/z 235 (M+H) + . Step 2 Dimethyl (3-oxo-6-(trifluoromethoxy)-1,3-dihydroisobenzofuran-1-yl)ph osphonate [0660] A solution of 3-hydroxy-5-(trifluoromethoxy)isobenzofuran-1(3H)-one (70 mg, 0.30 mmol) in dimethylphosphite (5 mL) was heated to 100 °C and stirred for 2 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give dimethyl (6- (difluoromethoxy)-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosph onate as a light yellow oil (90 mg, 83% yield). LCMS ESI m/z 327 (M+H) + . Step 3 6-(4-(2-Fluoro-5-((3-oxo-6-(trifluoromethoxy)isobenzofuran-1 (3H)- ylidene)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0661] A solution of dimethyl (3-oxo-6-(trifluoromethoxy)-1,3-dihydroisobenzofuran-1- yl)phosphonate (90 mg, 0.28 mmol), 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (112 mg, 0.33 mmol) and Et 3 N (84 mg, 0.83 mmol) in THF (3 mL) was stirred at rt under Ar (g) for 12 hours. The mixture was diluted with water (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to give 6-(4- (2-fluoro-5-((3-oxo-6-(trifluoromethoxy)isobenzofuran-1(3H)- ylidene)methyl)benzoyl)piperazin-1- yl)nicotinonitrile as a yellow solid (140 mg, 94% yield) LCMS ESI m/z 539 (M+H) + . Step 4 6-(4-(2-fluoro-5-((4-oxo-7-(trifluoromethoxy)-3,4-dihydropht halazin-1-yl)methyl)benzoyl)piperazin- 1-yl)nicotinonitrile [0662] A mixture of 6-(4-(2-fluoro-5-((3-oxo-6-(trifluoromethoxy)isobenzofuran-1 (3H)- ylidene)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (70 mg, 0.13 mmol) and N 2 H 4 •H 2 O (77 mg, 1.3 mmol) in THF (2 mL) was stirred at 70 °C for 3 hours. The solids were filtered and washed with water (5 mL), dried under vacuum to give 6-(4-(2-fluoro-5-((4-oxo-7-(trifluoromethoxy)-3,4-dihydropht halazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile as a white solid (26 mg, 36% yield). LCMS ESI m/z 553 (M+H) + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.02 (s, 1H), 8.52 (d, J = 8.8 Hz, 1H), 8.42 (d, J = 2.1 Hz,1H), 7.66 (dd, J = 9.0, 2.3 Hz, 1H), 7.57 (d, J = 8.8 Hz, 1H), 7.49 (s, 1H), 7.39 – 7.32 (m, 2H), 7.09 (t, J = 9.0 Hz, 1H), 6.62 (d, J = 9.0 Hz, 1H), 4.26 (s, 2H), 3.90 (m, 2H), 3.78 (m, 2H), 3.69 (t, J = 5.1 Hz, 2H), 3.42 (m, 2H). Synthesis of Example 31: 6-(4-(2-Fluoro-5-((4-oxo-7-(2-azaspiro[3.3]heptan-2-yl)-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile Step 1 6-(4-(2-Fluoro-5-((4-oxo-7-(2-azaspiro[3.3]heptan-2-yl)-3,4- dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0663] 6-[4-[5-[(7-Bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro- benzoyl]piperazin-1- yl]pyridine-3-carbonitrile (50.0 mg, 91.3 μmol), 2-azaspiro[3.3]heptane (44.4 mg, 457 μmol), Pd 2 (dba) 3 (5.3 mg, 9.1 μmol), t-BuONa (61.5 mg, 639 μmol), XPhos (8.7 mg, 18.3 μmol) and DMA (3 mL) were added to a 10 mL sealed tube. The resultant mixture was stirred at 120 °C for 0.5 hr under N 2 (g). The mixture was purified by prep-HPLC to afford 6-[4-[5-[[7-(2-azaspiro[3.3]heptan-2-yl)-4-oxo-3H- phthalazin-1-yl]methyl]-2-fluoro-benzoyl]piperazin-1-yl]pyri dine-3-carbonitrile (8.05 mg, 16% yield) as a white solid. LCMS ESI m/z 563.8 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.14 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 7.99 (d, J = 8.7 Hz, 1H), 7.90 (dd, J = 9.1, 2.4 Hz, 1H), 7.42 (dd, J = 8.8, 5.8 Hz, 2H), 7.25 (t, J = 8.9 Hz, 1H), 6.92 (d, J = 9.1 Hz, 1H), 6.81 (dd, J = 8.7, 2.1 Hz, 1H), 6.52 (d, J = 2.1 Hz, 1H), 4.22 (s, 2H), 3.90 (s, 4H), 3.75 (m, 4H), 3.62 (m, 2H), 3.37 (m, 2H), 2.17 (t, J = 7.5 Hz, 4H), 1.85 – 1.76 (m, 2H). Synthesis of Example 32: (E)-6-(4-(2-fluoro-5-((4-oxo-7-(4,4,4-trifluorobut-2-en-2-yl )-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile

5-(1-Ethoxyvinyl)isobenzofuran-1(3H)-one [0664] To a solution of 5-bromo-3H-isobenzofuran-1-one (3.00 g, 164 mmol) in toluene (101 mL) was added tributyl(1-ethoxyvinyl)stannane (89.0 g, 246 mmol, 83.3 mL), Pd(PPh 3 ) 4 (9.49 g, 8.21 mmol) at 25 °C under N 2 . The reaction mixture was stirred at 100 °C for 14 hrs. Water and EtOAc were added to the mixture. The organic layer was separated, washed with NaCl (aq), dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 20/1 to 0/1) to give 5-(1-ethoxyvinyl)-3H-isobenzofuran-1-one (30 g, 89% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-D6) δ 7.89 (s, 1H), 7.82 (s, 2H), 5.41 (s, 2H), 4.98 (d, J = 3.0 Hz, 1H), 4.49 (d, J = 3.0 Hz, 1H), 3.93 (t, J = 7.0 Hz, 2H), 1.37 (t, J = 7.0 Hz, 3H). Step 2 5-Acetylisobenzofuran-1(3H)-one [0665] A solution of 5-(1-ethoxyvinyl)-3H-isobenzofuran-1-one (30 g, 146.90 mmol) in 4M HCl/EtOAc (180 mL) was stirred at 25 °C for 2 hrs. The reaction mixture was concentrated to give 5- acetyl-3H-isobenzofuran-1-one (25 g, 96% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.23 (s, 1H), 8.12 (d, J = 8.0 Hz, 1H), 7.97 (d, J = 8.0 Hz, 1H), 5.49 (s, 2H), 2.67 (s, 3H). Step 3 (Z)- and (E)-tert-Butyl 3-(1-oxo-1,3-dihydroisobenzofuran-5-yl)but-2-enoate [0666] To a solution of 5-acetyl-3H-isobenzofuran-1-one (1.80 g, 10.2 mmol) in THF (20 mL) was added tert-butyl 2-diethoxyphosphorylacetate (3.35 g, 13.3 mmol, 3.12 mL) and t-BuOK (1.49 g, 13.3 mmol). The reaction mixture was stirred at 70 °C for 20 hrs. The reaction mixture was concentrated under vacuum. The residue was poured into water, extracted with EtOAc. The combined organic extracts were dried by Na 2 SO 4 , filtered and evaporated under reduced pressure to give viscous oils, which were purified by silica gel chromatography (petroleum ether/EtOAc = 10/1 to 2/1) to give the mixture of (E)-tert-butyl 3-(1-oxo-1,3-dihydroisobenzofuran-5-yl)but-2-enoate and (Z)-tert-butyl 3-(1-oxo-1,3- dihydroisobenzofuran-5-yl)but-2-enoate (1.2 g, 43% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6) δ 7.85 (d, J = 8.2 Hz, 2H), 7.73 (d, J = 8.7 Hz, 1H), 6.13 (d, J = 1.2 Hz, 1H), 5.41 (d, J = 6.7 Hz, 2H), 2.51 (d, J = 1.2 Hz, 3H), 1.48 (s, 9H). Step 4 (Z)- and (E)-3-(1-Oxo-1,3-dihydroisobenzofuran-5-yl)but-2-enoic acid [0667] The mixture product from above (8.00 g, 29.2 mmol) was added to 4M HCl/EtOAc (4 M, 100 mL). The reaction mixture was stirred at 25 °C for 3 hrs. The reaction mixture was concentrated to give a mixture of (E)-3-(1-oxo-1,3-dihydroisobenzofuran-5-yl)but-2-enoic acid and (Z)-3-(1-oxo-1,3- dihydroisobenzofuran-5-yl)but-2-enoic acid (5.78 g, 91% yield) was a yellow solid. LCMS ESI m/z: 219.1 [M + H] + . Step 5 (E)-5-(4,4,4-trifluorobut-2-en-2-yl)isobenzofuran-1(3H)-one [0668] To a solution of mixture (E)-3-(1-oxo-1,3-dihydroisobenzofuran-5-yl)but-2-enoic and (Z)-3-(1- oxo-1,3-dihydroisobenzofuran-5-yl)but-2-enoic acid (100 mg, 458 μmol), CuCl (45.4 mg, 458 μmol), Ag 2 CO 3 (75.8 mg, 275 μmol), and CF 3 SO 2 Na (215 mg, 1.37 mmol) in DCE (2 mL) at 0 °C was slowly added TBHP (295 mg, 2.29 mmol, 70% purity) with stirring. The reaction was allowed to warm to 70 °C and then stirred for 24 h under N 2 . Water and EtOAc were added to the mixture, which became two phases. The organic layer was separated, washed with NaCl (aq), dried over Na2SO4 and concentrated to give a residue. The residue was purified by prepared-TLC (petroleum ether/EtOAc = 2/1) to give (E)-5- (4,4,4-trifluorobut-2-en-2-yl)isobenzofuran-1(3H)-one (25 mg, 23% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 7.92 – 7.85 (m, 2H), 7.78 (d, J = 8.2 Hz, 1H), 6.34 (dd, J = 8.6, 1.3 Hz, 1H), 5.44 (s, 2H), 2.32 (dd, J = 2.1, 1.4 Hz, 3H). Step 6 (E)-2-(Hydroxymethyl)-4-(4,4,4-trifluorobut-2-en-2-yl)benzoi c acid [0669] To a solution of (E)-5-(4,4,4-trifluorobut-2-en-2-yl)isobenzofuran-1(3H)-one (630 mg, 2.60 mmol) in THF (3 mL) was added LiOH (186.90 mg, 7.80 mmol) at 25 °C. The reaction mixture was stirred at 25 °C for 3 hr. The mixture was adjusted to pH 6 with 1M HCl (aq). EtOAc was added to the mixture, which became two phases. The organic layer was separated, washed with NaCl (aq), dried over Na 2 SO 4 and concentrated to give (E)-2-(hydroxymethyl)-4-(4,4,4-trifluorobut-2-en-2-yl)benzoi c acid (400 mg, 59% yield) as a white solid. LCMS ESI m/z: 258.9 [M + H] + . Step 7 (E)-3-Hydroxy-5-(4,4,4-trifluorobut-2-en-2-yl)isobenzofuran- 1(3H)-one [0670] To a solution of (E)-2-(hydroxymethyl)-4-(4,4,4-trifluorobut-2-en-2-yl)benzoi c acid (800 mg, 3.07 mmol) in DCM (6 mL) was added Dess-Martin Periodinane (1.51 g, 3.38 mmol, 95% purity) at 25 °C under N 2 . The reaction mixture was stirred at 50 °C for 16 hrS. DCM was added to the mixture, after stirring the mixture for 30 min, the solids were collected, washed with DCM and filtrate dried under reduced pressure to give the crude product. The crude was purified by silica gel chromatography (petroleum ether/EtOAc = 20/1 to 0/1) to give (E)-3-hydroxy-5-(4,4,4-trifluorobut-2-en- 2-yl)isobenzofuran-1(3H)-one (450 mg, 57% yield) as a yellow oil. LCMS ESI m/z: 256.8 [M + H] + . Step 8, 9 and 10

(E)-6-(4-(2-Fluoro-5-((4-oxo-7-(4,4,4-trifluorobut-2-en-2-yl )-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0671] Following the three-step procedure in Example 1, but starting with (E)-3-hydroxy-5-(4,4,4- trifluorobut-2-en-2-yl)isobenzofuran-1(3H)-one gave the title compound as a white solid. LCMS ESI m/z: 576.7 [M + H] + . 1 H NMR (400 MHz, DMSO-d6) δ12.64 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 8.26 (d, J = 8.3 Hz, 1H), 8.09 (s,1H), 8.03 (d, J = 8.4 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.46 (t, J = 6.9 Hz, 2H), 7.26 (t, J = 8.9 Hz,1H), 6.92 (d, J = 9.1 Hz, 1H), 6.41 (dd, J = 16.8, 8.1 Hz, 1H), 4.42 (s, 2H), 3.74 (m, 4H), 3.60 (m, 2H), 3.30 – 3.26 (m, 2H), 2.32 (s, 3H). Synthesis of Example 33: 6-(4-(2-Fluoro-5-((4-oxo-7-((3,3,3-trifluoropropyl)amino)-3, 4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile

6-(4-(2-Fluoro-5-((4-oxo-7-((3,3,3-trifluoropropyl)amino)-3, 4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0672] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (200 mg, 0.37 mmol), 3,3-trifluoropropan-1-amine (124 mg, 1.10 mmol), Pd 2 (dba) 3 (33 mg, 0.04 mmol), BrettPhos (39.0 mg, 0.07 mmol) and t-BuONa (211 mg, 2.19 mmol) in DMA (2 mL) was heated at 160 °C by microwave for 0.5 hour. The reaction was cooled to rt and purified by prep-HPLC to give 6-(4-(2-fluoro-5-((4-oxo-7-((3,3,3-trifluoropropyl)amino)-3, 4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile as a white solid (51 mg, 24% yield). LCMS ESI m/z: 580 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 9.81 (s, 1H), 8.42 (d, J = 1.9 Hz, 1H), 8.23 (d, J = 8.7 Hz, 1H), 7.66 (dd, J = 9.0, 2.3 Hz, 1H), 7.38 – 7.35 (m, 1H), 7.34 – 7.30 (m, 1H), 7.06 (t, J = 8.8 Hz, 1H), 6.93 (dd, J = 8.8, 2.3 Hz, 1H), 6.62 (d, J = 9.0 Hz, 1H), 6.58 (d, J = 2.2 Hz, 1H), 4.65 (t, J = 6.0 Hz, 1H), 4.20 (s, 2H), 3.89 (m, 2H), 3.78 (m, 2H), 3.70 (t, J = 5.1 Hz, 2H), 3.52 – 3.41 (m, 4H), 2.45 – 2.33 (m, 2H). Synthesis of Example 34: 6-(4-(2-Fluoro-5-((4-oxo-7-(3-(trifluoromethyl)azetidin-1-yl )-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile

6-(4-(2-Fluoro-5-((4-oxo-7-(3-(trifluoromethyl)azetidin-1-yl )-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0673] To a solution of 6-[4-[5-[(7-bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro- benzoyl]piperazin-1-yl]pyridine-3-carbonitrile (50.0 mg, 91.3 μmol) in DMA (2 mL) was added 3- (trifluoromethyl)azetidine hydrochloride (73.8 mg, 456 μmol), t-BuONa (61.5 mg, 639 μmol), Pd 2 (dba) 3 (8.4 mg, 9.1 μmol) and XPhos (8.7 mg, 18.3 μmol). The reaction mixture was stirred at 110 °C under microwave for 0.5 hour. The mixture was purified by Prep-HPLC to give 6-[4-[2-fluoro-5-[[4-oxo-7-[3- (trifluoromethyl)azetidin-1-yl]-3H-phthalazin-1-yl]methyl]be nzoyl]piperazin-1-yl]pyridine-3-carbonitrile (15.6 mg, 29% yield) as a white solid. LCMS ESI m/z: 591.8 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.22 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 8.04 (d, J = 8.7 Hz, 1H), 7.90 (dd, J = 9.1, 2.4 Hz, 1H), 7.50 – 7.41 (m, 2H), 7.25 (t, J = 9.0 Hz, 1H), 6.92 (dd, J = 8.8, 2.2 Hz, 2H), 6.68 (d, J = 2.1 Hz, 1H), 4.25 (s, 2H), 4.18 (t, J = 8.7 Hz, 2H), 4.02 (dd, J = 8.8, 5.1 Hz, 2H), 3.75 (m, 5H), 3.62 (m, 2H), 3.32 – 3.29 (m, 2H). Synthesis of Example 35: 6-(4-(5-((7-(6,6-Difluoro-2-azaspiro[3.3]heptan-2-yl)-4-oxo- 3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)piperazin-1-y l)nicotinonitrile

6-(4-(5-((7-(6,6-Difluoro-2-azaspiro[3.3]heptan-2-yl)-4-oxo- 3,4-dihydrophthalazin-1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0674] A mixture of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (200 mg, 0.37 mmol), 6,6-difluoro-2-azaspiro[3.3]heptane (200 mg, 1.48 mmol), Pd 2 (dba) 3 (65.0 mg, 0.07 mmol), XPhos (33.0 mg, 0.07 mmol) and t-BuONa (107 mg, 1.11 mmol) in DMA (10 mL) was stirred at 130 °C for 16 hours. The reaction was cooled to rt, then the solids were filtered off. The filtrate was concentrated in vacuo and the residue was purified by prep- HPLC to give 6-(4-(5-((7-(6,6-difluoro-2-azaspiro[3.3]heptan-2-yl)-4-oxo- 3,4-dihydrophthalazin-1- yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile as a white solid (28 mg, 13% yield). LCMS ESI m/z: 600 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.18 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 8.01 (d, J = 8.7 Hz, 1H), 7.90 (dd, J = 9.1, 2.4 Hz, 1H), 7.47 – 7.38 (m, 2H), 7.25 (t, J = 8.9 Hz, 1H), 6.93 (d, J = 9.0 Hz, 1H), 6.85 (dd, J = 8.7, 2.1 Hz, 1H), 6.59 (d, J = 2.1 Hz, 1H), 4.23 (s, 2H), 4.05 (s, 4H), 3.76 (m, 4H), 3.62 (m, 2H), 3.36 (m, 2H), 2.87 (t, J = 12.4 Hz, 4H). Synthesis of Example 36: 6-(4-(5-((7-(1,1-Difluoroethyl)-4-oxo-3,4-dihydrophthalazin- 1-yl)methyl)- 2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile 5-(1,1-Difluoroethyl)isobenzofuran-1(3H)-one [0675] To a solution of 5-acetyl-3H-isobenzofuran-1-one (950 mg, 5.39 mmol) in DCM (20 mL) was added DAST (8.69 g, 53.9 mmol, 7.12 mL). The reaction mixture was stirred at 55 °C for 16 hours. The mixture was poured to sat. NaHCO 3 , extracted with DCM (20 mL x 3). The extracts were washed with sat. brine, dried over Na 2 SO 4 , filtered. The organic layer was concentrated to give the crude. The crude was purified by silica gel chromatography to give 5-(1,1-difluoroethyl)-3H-isobenzofuran-1-one (400 mg, 37 % yield) as a grey solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 7.97 (d, J = 8.0 Hz, 1H), 7.91 (d, J = 0.7 Hz, 1H), 7.78 (dd, J = 8.0, 0.6 Hz, 1H), 5.47 (s, 2H), 2.03 (t, J = 19.0 Hz, 3H). Step 2 5-(1,1-Difluoroethyl)isobenzofuran-1(3H)-one [0676] To a solution of 5-(1,1-difluoroethyl)-3H-isobenzofuran-1-one (350 mg, 1.77 mmol) in CCl 4 (20 mL) was added NBS (472 mg, 2.65 mmol, 225 μL) and AIBN (29.0 mg, 177 μmol). The mixture was stirred at 80 °C for 16 hours. The mixture was purified by silica gel chromatography (petroleum ether: EtOAc = 4:1) to give 3-bromo-5-(1,1-difluoroethyl)-3H-isobenzofuran-1-one (420 mg, 86% yield) as a light yellow oil. 1 H NMR (400 MHz, CDCl 3 ) δ 7.99 (d, J = 7.9 Hz, 1H), 7.76 (d, J = 8.8 Hz, 2H), 7.42 (s, 1H), 1.98 (t, J = 18.2 Hz, 3H). Step 3 5-(1,1-Difluoroethyl)-3-hydroxyisobenzofuran-1(3H)-one [0677] To a 50 mL round bottle flask was added 3-bromo-5-(1,1-difluoroethyl)-3H-isobenzofuran-1- one (420 mg, 1.52 mmol) and KOH (1 M, 20 mL). The mixture was stirred at 100 °C for 1 hour. The mixture was acidified to pH 5 with 1N HCl, extracted with DCM (20 mL x 3). The extracts were washed with brine, dried over Na 2 SO 4 , filtered and concentrated to give 5-(1,1-difluoroethyl)-3-hydroxy-3H- isobenzofuran-1-one as a light yellow solid (290 mg, 89% yield). LCMS ESI m/z: 212.9 [M + H] + . Step 4 Dimethyl 6-(1,1-difluoroethyl)-3-oxo-1,3-dihydroisobenzofuran-1-ylpho sphonate [0678] To a 50 mL round bottle flask was added 5-(1,1-difluoroethyl)-3-hydroxy-3H-isobenzofuran-1- one (290 mg, 1.35 mmol) and dimethylphosphite (149 mg, 1.35 mmol, 5 mL). The mixture was stirred at 100 °C for 16 hours. The mixture was poured into water (50 mL), extracted with DCM (30 mL x 3), dried over Na 2 SO 4 , filtered and concentrated to the residue. The residue was purified by silica gel chromatography (PE: EA= 10: 1) to give dimethyl 6-(1,1-difluoroethyl)-3-oxo-1,3-dihydroisobenzofuran- 1-ylphosphonate (810 mg, crude) as a light yellow oil. LCMS ESI m/z: 307.1 [M + H] + . Step 5 6-(4-(5-((6-(1,1-Difluoroethyl)-3-oxoisobenzofuran-1(3H)-yli dene)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0679] To a solution of dimethyl 6-(1,1-difluoroethyl)-3-oxo-1,3-dihydroisobenzofuran-1- ylphosphonate (442 mg, 1.31 mmol) in THF (15 mL) was added ET3N (397 mg, 3.92 mmol, 546 μL) and 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (800 mg, 1.31 mmol). The mixture was stirred at rt for 16 hours. The mixture was poured into water (30 mL), extracted with DCM (50 mL x 3). The organic layers were dried over Na 2 SO 4 , filtered and concentrated to the crude. The crude was purified by silica gel chromatography (PE: EA= 1: 1) to give 6-[4-[5-[[6-(1,1-difluoroethyl)-3-oxo-isobenzofuran- 1-ylidene]methyl]-2-fluoro-benzoyl]piperazin-1-yl]pyridine-3 -carbonitrile (290 mg, 43% yield) as a light yellow oil. LCMS ESI m/z: 519.2 [M + H] + . Step 6 6-(4-(5-((7-(1,1-Difluoroethyl)-4-oxo-3,4-dihydrophthalazin- 1-yl)methyl)-2-fluorobenzoyl)piperazin- 1-yl)nicotinonitrile [0680] To a solution of 6-[4-[5-[[6-(1,1-difluoroethyl)-3-oxo-isobenzofuran-1-yliden e]methyl]-2- fluoro-benzoyl]piperazin-1-yl]pyridine-3-carbonitrile (290 mg, 559 μmol) in THF (4 mL) was added N 2 H 4 ·H 2 O (2 mL). The mixture was stirred at 80 °C for 3 hours. The mixture was purified by Prep- HPLC to give 6-(4-(5-((7-(1,1-difluoroethyl)-4-oxo-3,4-dihydrophthalazin- 1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile (101 mg, 34% yield) as a white solid. LCMS ESI m/z: 532.8 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ12.73 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 8.37 (d, J = 8.3 Hz, 1H), 8.06 (s, 1H), 8.00 (d, J = 8.4 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.49 – 7.40 (m, 2H), 7.26 (t, J = 9.0 Hz, 1H), 6.91 (d, J = 9.1 Hz, 1H), 4.42 (s, 2H), 3.75 (m, 4H), 3.60 (m, 2H), 3.30 (m, 2H), 2.03 (t, J = 19.1 Hz, 3H). Synthesis of Example 37: 6-(4-(5-((7-(Cyclopropylethynyl)-4-oxo-3,4-dihydrophthalazin -1- yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile

6-(4-(5-((7-(Cyclopropylethynyl)-4-oxo-3,4-dihydrophthalazin -1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0681] To a mixture of 6-[4-[5-[(7-bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro- benzoyl]piperazin-1-yl]pyridine-3-carbonitrile (50.0 mg, 91.3 μmol) in THF (1.5 mL) was added ethynylcyclopropane (12.1 mg, 183 μmol, 15.46 μL) , CuI (8.70 mg, 45.7 μmol, 1.55 μL), Pd(PPh 3 ) 2 Cl 2 (6.41 mg, 9.13 μmol), Et 3 N (27.7 mg, 274 μmol, 38.2 μL). The reaction mixture was stirred under microwave at 110 °C for 1 hour. The mixture was purified by Prep-HPLC to give 6-(4-(5- ((7-(cyclopropylethynyl)-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile (8.43 mg, 17% yield) as a white solid. LCMS ESI m/z: 532.8 [M + H] + . 1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.61 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 8.18 (d, J = 8.2 Hz, 1H), 7.90 (dd, J = 9.3, 2.5 Hz, 2H), 7.77 – 7.72 (m, 1H), 7.41 (d, J = 5.7 Hz, 2H), 7.27 (d, J = 9.5 Hz, 1H), 6.92 (d, J = 9.1 Hz, 1H), 4.33 (s, 2H), 3.75 (m, 4H), 3.63 (m, 2H), 3.33 (m, 2H), 1.58 (s, 1H), 0.97 – 0.86 (m, 2H), 0.84 – 0.70 (m, 2H). Synthesis of Example 38: 6-(4-(2-Fluoro-5-((4-oxo-7-(pyridin-2-yloxy)-3,4-dihydrophth alazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile Methyl 2-bromo-4-(pyridin-2-yloxy)benzoate [0682] A mixture of methyl 2-bromo-4-fluorobenzoate (2.50 g, 10.82 mmol) and 2-fluoropyridine (1 mL) was stirred at 120 °C for 16 hours. The reaction was cooled to rt, diluted with H 2 O (100 mL) and extracted with EtOAc (100 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0% to 7%) to give methyl 2-bromo-4-(pyridin-2- yloxy)benzoate (2.83 g, 85% yield) as a colorless oil. LCMS ESI m/z: 308.0 [M+H] + . Step 2 2-Bromo-4-(pyridin-2-yloxy)benzoic acid [0683] A mixture of methyl 2-bromo-4-(pyridin-2-yloxy)benzoate (2.83 g, 9.18 mmol) and NaOH (1.84 g, 45.92 mmol) in MeOH (30 mL) and H 2 O (10 mL) was stirred at rt for 2 hours. The organic solvent was removed in vacuo and aq HCl (1N) was added until the solution reached pH 5~6. The formed solid was collected by filtration, washed with H 2 O, dried under vacuum to give 2-bromo-4-(pyridin-2- yloxy)benzoic acid (2.2 g, 81% yield) as a white solid. LCMS ESI m/z: 294.0 [M+H] + . Step 3 3-Hydroxy-5-(pyridin-2-yloxy)isobenzofuran-1(3H)-one [0684] A solution of 2-bromo-4-(pyridin-2-yloxy)benzoic acid (1.90 g, 6.46 mmol) in THF (30 mL) was cooled to -78 °C, then n-BuLi (2.5 M in heptane, 5.7 mL, 14.25 mmol) was addded dropwise under Ar (g). The reaction was stirred at -78 °C for 30 min, then added DMF (1.05 g, 14.33 mmol). The reaction was stirred at -78 °C for 20 min, then quenched with sat. NH 4 Cl soltion (10 mL), neutralizated by aq HCl (1N) until the solution reached pH 5~6, and extracted with EtOAc (30 x 3 mL). The combined organic layer was washed with bine, dried over Na 2 SO 4 , filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30% to 50%) to give 3-hydroxy-5- (pyridin-2-yloxy)isobenzofuran-1(3H)-one (680 mg, 43% yield) as a white solid. LCMS ESI m/z: 244.1 [M+H] + . Step 4 Dimethyl (3-oxo-6-(pyridin-2-yloxy)-1,3-dihydroisobenzofuran-1-yl)pho sphonate [0685] A solution of 3-hydroxy-5-(pyridin-2-yloxy)isobenzofuran-1(3H)-one (480 mg, 1.97 mmol) in dimethyl phosphonate (5 mL) was stirred at 100 °C for 16 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 50% to 60 %) to give dimethyl (3-oxo-6-(pyridin-2- yloxy)-1,3-dihydroisobenzofuran-1-yl)phosphonate (200 mg, 30% yield) as a white solid. LCMS ESI m/z: 334.1 [M+H] + . Step 5

6-(4-(2-Fluoro-5-((4-oxo-7-(pyridin-2-yloxy)-3,4-dihydrophth alazin-1-yl)methyl)benzoyl)piperazin- 1-yl)nicotinonitrile [0686] A solution of dimethyl (3-oxo-6-(pyridin-2-yloxy)-1,3-dihydroisobenzofuran-1-yl)pho sphonate (200 mg, 0.59 mmol), 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (212 mg, 0.63 mmol) and Et 3 N (181 mg, 1.79 mmol) in anhydrous THF (2 mL) was stirred at 60 °C under Ar (g) for 15 hours. Hydrazine hydrate (46 mg, 0.78 mmol) was added and the reaction was stirred at 70 °C for 1 hour. The reaction was cooled to rt, then THF was removed under vacuum. The formed solid was filtered, washed with water (10 mL) and petroleum ether/EtOAc (2:1) to give 6-(4-(2-fluoro-5-((4-oxo-7-(pyridin- 2-yloxy)-3,4-dihydrophthalazin-1-yl)methyl)benzoyl)piperazin -1-yl)nicotinonitrile (60 mg, 20% yield) as a white solid. LCMS ESI m/z: 562.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.60 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 8.28 (d, J = 8.7 Hz, 1H), 8.17 – 8.13 (m, 1H), 7.95 – 7.88 (m, 2H), 7.67 (d, J = 2.1 Hz, 1H), 7.58 – 7.53 (m, 1H), 7.44 – 7.34 (m, 2H), 7.27 – 7.14 (m, 3H), 6.89 (d, J = 9.1 Hz, 1H), 4.28 (s, 2H), 3.73 (m, , 4H), 3.56 (m, 2H), 3.26 (m, 2H). Synthesis of Example 39: 6-(4-(2-Fluoro-5-((7-(naphthalen-2-yloxy)-4-oxo-3,4-dihydrop hthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

Methyl 2-bromo-4-(naphthalen-2-yloxy)benzoate [0687] A mixture of ethyl methyl 2-bromo-4-fluorobenzoate (3.00 g,12.9 mmol), naphthalen-2-ol (2.78 g, 19.3 mmol) and K 2 CO 3 (3.56 g, 25.8 mmol) in DMF (30 mL) was stirred at 100 °C for 16 hours. The mixture was diluted with H 2 O (60 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0% to 50%) to give methyl 2-bromo-4-(naphthalen-2- yloxy)benzoate (4.00 g, 87% yield) as a white solid. LCMS ESI m/z: 357 [M+H]+. Step 2 2-Bromo-4-(naphthalen-2-yloxy)benzoic acid [0688] A mixture of methyl 2-bromo-4-(naphthalen-2-yloxy)benzoate (4.00 g, 11.2 mmol) and NaOH (2.24 g, 56.0 mmol) in MeOH (15 mL) and H 2 O (5 mL) was stirred at rt for 2 hours. The organic solvent was removed in vacuo and aq HCl (1N) was added until the solution reached pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 2-bromo-4-(naphthalen-2- yloxy)benzoic acid (3.5 g, 91% yield) as a white solid. LCMS ESI m/z: 343 [M+H] + . Step 3 3-Hydroxy-5-(naphthalen-2-yloxy)isobenzofuran-1(3H)-one [0689] A solution of 2-bromo-4-(naphthalen-2-yloxy)benzoic acid (1.00 g, 2.91 mmol) in THF (30 mL) was cooled to -78°C, then n-BuLi (2.5 M in heptane, 2.33 mL, 5.83 mmol) was addded dropwise under Ar (g). The reaction was stirred at -78 °C for 30 min, then added DMF (469 mg, 6.41 mmol). The reaction was stirred at -78 °C for 20 min, then quenched with sat. NH 4 Cl soltion (10 mL) and added aq HCl (1M) until the solution reached pH 5~6. The mixture was extracted with EtOAc (30 x 3 mL). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0% to 20%) to give 3- hydroxy-5-(naphthalen-2-yloxy)isobenzofuran-1(3H)-one (540 mg, 63% yield) as a white solid. LCMS ESI m/z: 293 [M+H] + . Step 4 Dimethyl (6-(naphthalen-2-yloxy)-3-oxo-1,3-dihydroisobenzofuran-1-yl) phosphonate [0690] A solution of 3-hydroxy-5-(naphthalen-2-yloxy)isobenzofuran-1(3H)-one (540 mg, 1.85 mmol) in dimethyl phosphonate (10 mL) was stirred at 100°C for 16 hours. The mixture was cooled to rt, diluted with H 2 O (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give dimethyl (6- (naphthalen-2-yloxy)-3-oxo-1,3-dihydroisobenzofuran-1-yl)pho sphonate (690 mg, 97% yield) as a light yellow oil. LCMS ESI m/z: 385 [M+H] + . Step 5 6-(4-(2-fluoro-5-((7-(naphthalen-2-yloxy)-4-oxo-3,4-dihydrop hthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0691] A solution of dimethyl (6-(naphthalen-2-yloxy)-3-oxo-1,3-dihydroisobenzofuran-1- yl)phosphonate (200 mg, 0.52 mmol), 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (176 mg, 0.52 mmol) and Et 3 N (158 mg, 1.56 mmol) in THF (5 mL) was stirred at 25 °C under Ar (g) for 15 hours. Hydrazine hydrate (46 mg, 0.78 mmol) was added and the reaction was stirred at 70 ℃ for 1 hour. The reaction was cooled to rt, and the solvent was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/EA (2:1, 10 mL) to give 6-(4-(2-fluoro-5-((7- (naphthalen-2-yloxy)-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )benzoyl)piperazin-1-yl)nicotinonitrile (119 mg, 37% yield) as a white solid. LCMS ESI m/z: 611 [M+H]+. 1 H NMR (400 MHz, DMSO–d 6 ) δ 12.58 (s, 1H), 8.49 – 8.44 (m, 1H), 8.27 (d, J = 8.7 Hz, 1H), 8.05 – 7.97 (m, 2H), 7.88 – 7.81 (m, 2H), 7.59 – 7.57 (m, 1H), 7.56 – 7.50 (m, 2H), 7.50 – 7.48 (m, 1H), 7.48 – 7.44 (m, 1H), 7.35 – 7.30 (m, 2H), 7.29 – 7.24 (m, 1H), 7.06 (t, J = 9.0 Hz, 1H), 6.84 (d, J = 9.1 Hz, 1H), 4.23 (s, 2H), 3.78 – 3.68 (m, 4H), 3.60 – 3.55 (m, 2H), 3.27 – 3.23 (m, 2H). Synthesis of Example 40: 6-(4-(5-((7-Cyclopropyl-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile 6-(4-(5-((7-Cyclopropyl-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile [0692] 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoyl)piperazin-1- yl)nicotinonitrile (27.7 mg, 0.05 mmol), cyclopropylboronic acid (13.0 mg , 0.15 mmol), Pd(OAc) 2 (3.0 mg, 0.013 mmol), PCy 3 (8.0 mg, 0.029 mmol), K 2 CO 3 (21.0 mg, 0.15 mmol), and toluene were added in a pressure vial under N 2 atmosphere. The mixture was heated at 100 o C overnight. The crude residue was purified by flash (0-100% EtOAc in hexanes) to afford the product as a white solid (4.3 mg, 16% yield). LCMS ESI m/z: 509.2 [M+H] + 1 H NMR (400 MHz, CDCl 3 ) δ 10.22 (s, 1H), 8.41 (d, J = 1.9 Hz, 1H), 8.32 (d, J = 8.2 Hz, 1H), 7.42 – 7.30 (m, 4H), 7.08 (d, J = 8.8 Hz, 1H), 6.61 (d, J = 8.9 Hz, 1H), 4.27 (s, 2H), 3.89 (s, 2H), 3.77 (s, 2H), 3.73 – 3.60 (m, 2H), 3.44 (s, 2H), 2.01 (m, 1H), 1.26 (m, 2H), 1.16 – 1.09 (m, 2H). Synthesis of Example 41: 6-(4-(2-Fluoro-5-((7-neopentyl-4-oxo-3,4-dihydrophthalazin-1 - yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile 6-(4-(2-Fluoro-5-((7-neopentyl-4-oxo-3,4-dihydrophthalazin-1 -yl)methyl)benzoyl)piperazin-1- yl)nicotinonitrile [0693] 6-(4-(5-((7-Bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoyl)piperazin-1- yl)nicotinonitrile (27.7 mg, 0.05 mmol), neopentylboronic acid (0.017 g, 0.15 mmol), Pd(OAc) 2 (.0 mg, 0.013 mmol), PCy 3 (8.0 mg, 0.029 mmol), K 2 CO 3 (21.0 mg, 0.15 mmol), and toluene were added in a pressure vial under N 2 atmosphere. The mixture was heated at 100 o C overnight. The reaction was cooled to rt and concentrated in vacuo. The crude residue was purified via a flash chromatography (0-100% EtOAc in hexanes) to afford the product as a white solid (5.4 mg, 19% yield). LC-MS ESI m/z 539.4 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 9.91 (s, 1H), 8.35 (d, J = 1.7 Hz, 1H), 8.28 (d, J = 8.1 Hz, 1H), 7.59 (dd, J = 9.0, 2.3 Hz, 1H), 7.46 (dd, J = 8.1, 1.5 Hz, 1H), 7.35 (s, 1H), 7.29 (m, 2H), 6.98 (t, J = 8.8 Hz, 1H), 6.55 (d, J = 9.0 Hz, 1H), 4.21 (s, 2H), 3.82 (s, 2H), 3.70 (s, 2H), 3.62 (s, 2H), 3.34 (s, 2H), 1.98 (s, 2H), 0.95 (s, 9H). Synthesis of Example 42: 4-(4-Fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1- carbonyl)benzyl)-6-(3,3,3-trifluoroprop-1-yn-1-yl)phthalazin -1(2H)-one 4-(4-Fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1-carbonyl)benzyl)-6-(3,3,3- trifluoroprop-1-yn-1-yl)phthalazin-1(2H)-one [0694] 6-Bromo-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one (24.0 mg, 0.04 mmol) was dissolved in anhydrous toluene (0.5 mL) under argon gas and the solution was degassed with argon for 5 min. Palladium tetrakis(triphenylphosphine) (7.0 mg, 0.00583 mmol.) was added quickly and the solution degassed with argon and stirred for 10 min. To this mixture tributyl(3,3,3-trifluoroprop-1-yn-1-yl)stannane (0.025 uL, 0.06 mmol) was added dropwise. The reaction mixture was heated at 125 °C for 4 h in a sealed tube. The reaction mixture was cooled to rt and concentrated in vacuo. The crude residue was purified via flash chromatography (0-80% EtOAc in hexanes) to afford the product as a white solid (6.6 mg, 27% yield). LCMS ESI m/z: 605.0 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) 9.78 (s, 1H), 8.33 (s, 2H), 8.13 (d, J = 8.9 Hz, 1H), 7.74 – 7.65 (m, 2H), 7.11-7.16 (m, 2H), 6.91 (t, J = 8.8 Hz, 1H), 4.06 (s, 2H), 3.84 (s, 2H), 3.74 – 3.67 (m, 4H), 3.22 (s, 2H). Synthesis of Example 43: 6-Ethyl-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one 6-Ethyl-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one [0695] 6-Bromo-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one (0.030 g, 0.05 mmol), ethylboronic acid (0.008 g, 3.0 equiv), Pd(OAc) 2 (2 mg), PCy 3 (4 mg), K 2 CO 3 (21 mg) and toluene were added in a pressure vial under N 2 atmosphere. The mixture was heated at 100 o C overnight. The reaction was cooled to rt and concentrated in vacuo. The crude residue was purified via flash chromatgraphy (0-100% EtOAc in hexanes) to afford the product as a white solid (8.4 mg, 31% yield). LCMS ESI m/z: 540.4 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.12 (s, 1H), 8.43 (s, 2H), 8.26 (d, J = 7.9 Hz, 1H), 7.34 – 7.27 (m, 4H), 7.00 (t, J = 8.8 Hz, 1H), 4.19 (s, 2H), 3.92 – 3.77 (m, 4H), 3.32 (s, 2H), 2.75 (q, J = 7.2 Hz, 1H), 1.23 (t, J = 7.2 Hz, 1H). Synthesis of Example 44: 4-(4-Fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1- carbonyl)benzyl)-6-isobutylphthalazin-1(2H)-one 4-(4-Fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1-carbonyl)benzyl)-6- isobutylphthalazin-1(2H)-one [0696] 6-Bromo-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one (0.030 g, 0.05 mmol) isobutylboronic acid (0.011 g, 3.0 equiv), Pd(OAc) 2 (2 mg), PCy 3 (4 mg), K 2 CO 3 (21 mg), and toluene were added in a pressure vial under N 2 atmosphere. The mixture was heated at 100 o C overnight. The reaction was cooled to rt and concentrated in vacuo. The crude residue was purified by flash chromatography (0-100% EtOAc in hexanes) to afford the product as a white solid (12.8 mg, 49% yield). LCMS ESI m/z: 568.4 [M+H] + . 1 H NMR (400 MHz, CDCl3) δ 10.25 (s, 1H), 8.43 (s, 2H), 8.31 (d, J = 8.0 Hz, 1H), 7.48 (d, J = 8.0 Hz, 1H), 7.38 (s, 1H), 7.28 (d, J = 17.5 Hz, 2H), 6.98 (t, J = 8.8 Hz, 1H), 4.22 (s, 2H), 3.94 (s, 2H), 3.84 – 3.77 (m, 4H), 3.31 (s, 2H), 2.54 (d, J = 7.1 Hz, 2H), 1.80 (m, 1H), 0.79 (d, J = 6.6 Hz, 6H). Synthesis of Example 45: 6-Cyclopropyl-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin -2- yl)piperazine-1-carbonyl)benzyl)phthalazin-1(2H)-one 6-Cyclopropyl-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin -2-yl)piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one [0697] 6-Bromo-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one (0.030 g, 0.05 mmol), cyclopropylboronic acid (0.0065 g, 3.0 equiv), Pd(OAc) 2 (2 mg), PCy 3 (4 mg), K 2 CO 3 (21 mg), and toluene were added in a pressure vial under N 2 atmosphere. The resulting mixture was heated at 100 o C overnight. The reaction was cooled to rt and concentrated in vacuo. The crude residue was purified via flash chromatpgraphy (0-100% EtOAc in hexanes) to afford the product as a white solid (9.8 mg, 37% yield). LCMS ESI m/z: 552.7 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.12 (s, 1H), 8.43 (s, 2H), 8.26 (d, J = 7.9 Hz, 1H), 7.34 – 7.27 (m, 4H), 7.00 (t, J = 8.8 Hz, 1H), 4.19 (s, 2H), 3.92 – 3.77 (m, 4H), 3.32 (s, 2H), 1.94 (m, 1H), 0.87 – 0.65 (m, 4H). Synthesis of Example 46: 6-(3-(Dimethylamino)prop-1-yn-1-yl)-4-(4-fluoro-3-(4-(5- (trifluoromethyl)pyrimidin-2-yl)piperazine-1-carbonyl)benzyl )phthalazin-1(2H)-one 6-(3-(Dimethylamino)prop-1-yn-1-yl)-4-(4-fluoro-3-(4-(5-(tri fluoromethyl)pyrimidin-2- yl)piperazine-1-carbonyl)benzyl)phthalazin-1(2H)-one [0698] 6-bromo-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one (0.030 g, 0.05 mmol) was added to a microwave vial and dissolved in anhydrous THF (1 mL) under argon. CuI (4 mg) and Pd(PPh 3 ) 2 Cl 2 (2 mg) was added to the stirred solution under argon and the mixture was stirred at rt for 15 mins. To this mixture as added triethylamine (60 uL), stirred for another 5 mins, and N,N-dimethylprop-2-yn-1-amine was added dropwise. The mixture was heated in a microwave reactor at 125 o C for 1 hr. The crude residue was purified via a flash chromatography (0-80% EtOAc in hexanes) to afford the product as a white solid (9.0 mg, 30% yield). LCMS ESI m/z: 593.9 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.46 (s, 1H), 8.44 (s, 2H), 8.33 (d, J = 8.2 Hz, 1H), 7.82 – 7.67 (m, 2H), 7.28 (ddd, J = 13.7, 5.6, 2.4 Hz, 2H), 7.01 (t, J = 8.8 Hz, 1H), 4.21 (s, 2H), 3.95 (s, 2H), 3.86 – 3.79 (m, 4H), 3.64 (s, 2H), 3.33 (s, 2H), 2.50 (s, 6H). Synthesis of Example 47: 6-(Cyclopropylethynyl)-4-(4-fluoro-3-(4-(5-(trifluoromethyl) pyrimidin-2- yl)piperazine-1-carbonyl)benzyl)phthalazin-1(2H)-one 6-(Cyclopropylethynyl)-4-(4-fluoro-3-(4-(5-(trifluoromethyl) pyrimidin-2-yl)piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one [0699] 6-Bromo-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one (0.030 g, 0.05 mmol) was added to a microwave vial and dissolved in anhydrous THF (1 mL) under argon gas. CuI (4 mg) and Pd(PPh 3 ) 2 Cl 2 (2 mg) was added to the stirred solution under argon and the mixture was stirred at rt for 15 mins. To this mixture was added triethylamine (60 uL), stirred for another 5 mins, and then the cyclopropylalkyne was added dropwise. The mixture was heated in a microwave reactor at 125 o C for 1 hr. The crude residue was purified via a flash chromatography (0-80% EtOAc in hexanes) to afford the product as a white solid (0.014 g, 49% yield). LCMS ESI m/z: 576.9 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.58 (s, 1H), 8.50 (s, 2H), 8.35 (d, J = 8.8 Hz, 1H), 7.72 – 7.62 (m, 2H), 7.34 - 7.42 (m, 2H), 7.07 (t, J = 8.8 Hz, 1H), 4.25 (s, 2H), 4.02 (s, 2H), 3.40 (s, 2H), 1.55 – 1.41 (m, 1H), 0.97 – 0.77 (m, 4H). Synthesis of Example 48: 4-(4-Fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1- carbonyl)benzyl)-6-(prop-1-yn-1-yl)phthalazin-1(2H)-one 4-(4-Fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1-carbonyl)benzyl)-6-(prop-1-yn-1- yl)phthalazin-1(2H)-one [0700] 6-Bromo-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one (0.029 g, 0.053 mmol) was dissolved in anhydrous toluene (1 mL) under argon gas and the solution degassed with argon for 5 min. Palladium tetrakis(triphenylphosphine) (7 mg, 0.00583 mmol) was added quickly to this mixture and the solution was degassed with argon for 10 min. Tributyl(prop-1-yn-1-yl)stannane (0.040 uL, 1.5 mmol) was added dropwise. The reaction mixture was heated at 125 °C for 4 h in a sealed tube. The reaction was cooled to rt and concentrated in vacuo. The crude residue was purified by flash chromatography (0-80% EtOAc in hexanes) to afford the product as a white solid (21.6 mg, 74% yield). LCMS ESI m/z: 550.2 [M+H] + . 1 H NMR (400 MHz, CD 3 OD) δ 8.58 – 8.42 (m, 2H), 8.21 (d, J = 8.3 Hz, 1H), 7.79 (s, 1H), 7.68 (dd, J = 8.3, 1.4 Hz, 1H), 7.33 (dd, J = 6.3, 2.2 Hz, 1H), 7.12 (t, J = 9.0 Hz, 1H), 4.30 (s, 2H), 4.00 – 3.93 (m, 4H), 3.81 – 3.75 (m, 4H), 2.00 (s, 3H). Synthesis of Example 49: 4-(4-Fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1- carbonyl)benzyl)-6-(3,3,3-trifluoroprop-1-yn-1-yl)phthalazin -1(2H)-one 4-(4-Fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1-carbonyl)benzyl)-6-(3,3,3- trifluoroprop-1-yn-1-yl)phthalazin-1(2H)-one [0701] 6-Bromo-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one (0.024 g, 0.04 mmol) was dissolved in anhydrous toluene (0.5 mL) under argon gas and the solution was degassed with argon for 5 min. Palladium tetrakis(triphenylphosphine) (7 mg, 0.00583 mmol) was added quickly and the solution degassed with argon and stirred for 10 min. To this mixture tributyl(3,3,3-trifluoroprop-1-yn-1-yl)stannane (0.025 uL, 0.06 mmol) was added dropwise. The reaction mixture was heated at 125 °C for 4 h in a sealed tube. The reaction mixture was cooled to rt and concentrated in vacuo. The crude residue was purified by flash chromatography (0-80% EtOAc in hexanes) to afford the product as a white solid (6.6 mg, 27% yield). LCMS ESI m/z: 605.0 [M+H] + . 1 H NMR (400 MHz, CDCl3) 9.78 (s, 1H), 8.33 (s, 2H), 8.13 (d, J = 8.9 Hz, 1H), 7.74 – 7.65 (m, 2H), 7.11-7.16 (m, 2H), 6.91 (t, J = 8.8 Hz, 1H), 4.06 (s, 2H), 3.84 (s, 2H), 3.74 – 3.67 (m, 4H), 3.22 (s, 2H). Synthesis of Example 51: 6-(4-(5-((7-(3,3-Difluorocyclobutoxy)-4-oxo-3,4-dihydrophtha lazin-1- yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile Dimethyl (6-(3,3-difluorocyclobutoxy)-3-oxo-1,3-dihydroisobenzofuran- 1-yl)phosphonate [0702] A mixture of dimethyl (6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (3H)-one (25 mg, 0.078 mmol), Pd 2 (dba) 3 (2.5 mg, 0.007 mmol), Rockphos (6.1 mg, 0.012 mmol), cesium carbonate (38 mg, 0.12 mmol) and 3,3-difluorocyclobutanol (43 mg, 0.39 mmol) in toluene (1.7 mL) was stirred at 90 °C for 5 hours. The mixture was cooled to rt and concentrated in vacuo. The resulting dimethyl (6-(3,3-difluorocyclobutoxy)-3-oxo-1,3-dihydroisobenzofuran- 1-yl)phosphonate was obtained as crude (brown oil) and carried forward without further purification. LCMS ESI m/z: 349.2 [M+H] + . Step 2 6-(4-(5-((7-(3,3-Difluorocyclobutoxy)-4-oxo-3,4-dihydrophtha lazin-1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0703] A mixture of dimethyl (6-(3,3-difluorocyclobutoxy)-3-oxo-1,3-dihydroisobenzofuran- 1- yl)phosphonate (25 mg, 0.078 mmol), 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (24 mg, 0.078 mmol) and Et3N (22 mg, 0.21 mmol) in THF (0.7 mL) was stirred at rt for 17 hours. To the reaction mixture was added N 2 H 4 •H 2 O (0.01 mL, 0.32 mmol), then stirred at 70 °C for 1 hour. The reaction mixture was concentrated in vacuo and the resulting crude was subjected to reverse phase column chromatography (MeCN: ammonium formate buffered water = 0 to 100%) to afford 6-(4-(5-((7-(3,3- difluorocyclobutoxy)-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile as a white solid (4 mg, 0.059 mmol, 12% yield). LCMS ESI m/z: 575.3 [M+H] + . 1 H- NMR (400 MHz, DMSO-d 6 ) δ 8.51 (d, J = 2.2 Hz, 1H), 8.19 (d, J = 8.8 Hz, 1H), 7.90 (dd, J = 9.2, 2.2 Hz, 1H), 7.47 – 7.42 (m, 2H), 7.39 (dd, J = 8.8, 2.3 Hz, 1H), 7.28 (d, J = 9.3 Hz, 1H), 7.18 (d, J = 1.7 Hz, 1H), 6.92 (d, J = 9.1 Hz, 1H), 4.99 – 4.91 (m, 1H), 4.32 (s, 2H), 3.78 – 3.74 (m, 2H), 3.73 – 3.69 (m, 2H), 3.63 – 3.59 (m, 2H), 3.33 – 3.28 (m, 2H), 3.26 – 3.18 (m, 2H), 2.76 – 2.67 (m, 2H). Synthesis of Example 52: 6-(4-(5-((7-(3-cyanocyclobutoxy)-4-oxo-3,4-dihydrophthalazin -1- yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile formate salt [0704] Following the procedure described in Example 51 and making non-critical variations as required to replace dimethyl (6-(3,3-difluorocyclobutoxy)-3-oxo-1,3-dihydroisobenzofuran- 1- yl)phosphonate with dimethyl (6-(3-cyanocyclobutoxy)-3-oxo-1,3-dihydroisobenzofuran-1- yl)phosphonate, 6-(4-(5-((7-(3-cyanocyclobutoxy)-4-oxo-3,4-dihydrophthalazin -1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile was obtained as a white solid (1 mg, 0.002 mmol, 2% yield). LCMS ESI m/z: 564.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.50 (s, 1H), 8.51 (d, J = 1.9 Hz, 1H), 8.48 (s, 1H), 8.17 (dd, J = 8.9, 2.1 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.45 – 7.41 (m, 2H), 7.33 (dd, J = 8.9, 2.3 Hz, 1H), 7.27 (t, J = 8.9 Hz, 1H), 7.12 (dd, J = 7.0, 3.0 Hz, 1H), 6.93 (d, J = 9.1 Hz, 1H), 4.90 – 4.82 (m, 1H), 4.30 (s, 2H), 3.79 – 3.76 (m, 2H), 3.74 – 3.71 (m, 2H), 3.62 (dd, J = 6.0, 3.3 Hz, 2H), 3.17 – 3.08 (m, 3H), 2.96 – 2.84 (m, 2H), 2.40 – 2.33 (m, 2H). Synthesis of Example 53: 6-(4-(5-((7-(cyclobutylsulfonyl)-4-oxo-3,4-dihydrophthalazin -1-yl)methyl)- 2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile 2-Ethylhexyl 3-((4-(3-(4-(5-cyanopyridin-2-yl)piperazine-1-carbonyl)-4-fl uorobenzyl)-1-oxo-1,2- dihydrophthalazin-6-yl)thio)propanoate [0705] To a solution of 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoyl)piperazin-1-yl)nicotinonitrile (200 mg, 0.365 mmol), 2-Ethylhexyl-3-Mercaptopropionate (80 mg, 0.365 mmol) and N,N-diisopropylethylamine (129 µL, 0.731 mmol) in 1,4-dioxane (3.65 mL) were added bis(dibenzylideneacetone)palladium(0) (4 mg, 0.007 mmol) and 4,4-bis(diphenylphosphino)- 9,9-dimethylxanthene (8 mg, 0.014 mmol) at room temperature. The resulting mixture was degassed with N 2 for 5 mins charged with a N 2 balloon. The reaction mixture was heated to 110 o C. After 180 mins, the reaction mixture diluted with EtOAc (50 mL) and washed with brine. The organic layer was dried over Na2SO4 and filtered. The solvent was removed to afford the crude product of 2-ethylhexyl 3-((4-(3-(4-(5- cyanopyridin-2-yl)piperazine-1-carbonyl)-4-fluorobenzyl)-1-o xo-1,2-dihydrophthalazin-6- yl)thio)propanoate as yellow oil (172 mg, 69 % yield). LCMS ESI m/z: 685.5 [M+H] + . Step 2 6-(4-(2-Fluoro-5-((7-mercapto-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1- yl)nicotinonitrile [0706] To a solution of 2-ethylhexyl 3-((4-(3-(4-(5-cyanopyridin-2-yl)piperazine-1-carbonyl)-4- fluorobenzyl)-1-oxo-1,2-dihydrophthalazin-6-yl)thio)propanoa te (142 mg, 0.207 mmol) in THF (2.07 mL) was added potassium tert-butoxide (58 uL, 0.415 mmol) at 0 o C. After 110 mins, the reaction mixture was concentrated under reduced pressure and purified with C-18 silica gel reverse-phase chromatography using ACN/AmF from 0% to 60% to afford 6-(4-(2-fluoro-5-((7-mercapto-4-oxo-3,4-dihydrophthalazin- 1-yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (78.0 mg, 71 % yield) as a white solid. LCMS ESI m/z: 501.3 [M+H] + . Step 3 6-(4-(5-((7-(cyclobutylthio)-4-oxo-3,4-dihydrophthalazin-1-y l)methyl)-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile [0707] To a solution of 6-(4-(2-fluoro-5-((7-mercapto-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (20 mg, 0.400 mmol) in MeCN (400 µL) were added bromocyclobutane (23 uL, 0.240 mmol) and cesium carbonate (13 mg, 0.400 mmol) . The reaction vessel was charged with N 2 and heated to 120 o C with a microwave reactor. After 20 mins, the reaction was concentrated under reduced pressure. The crude was used in the next step without further purification. LCMS ESI m/z: 555.3 [M+H] + . Step 4 6-(4-(5-((7-(cyclobutylsulfonyl)-4-oxo-3,4-dihydrophthalazin -1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0708] To a solution of the crude product of 6-(4-(5-((7-(cyclobutylthio)-4-oxo-3,4-dihydrophthalazin- 1-yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile (25 mg, 0.451 mmol) in dichloromethane (2.25 mL) was added 3-chloroperbenzoic acid (31 mg, 0.180 mmol). The resulting suspension was stirred at room temperature. After 2 hours, the reaction mixture was concentrated under reduced pressure. The crude was purified with C18 silica gel reverse-phase chromatography using MeCN/Ammonium formate from 0% to 70% to afford (17 mg, 63 % yield) as a white powder. LCMS ESI m/z: 587.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.89 (s, 1H), 8.49 (d, J = 2.1 Hz, 1H), 8.46 (d, J = 8.2 Hz, 1H), 8.24 – 8.16 (m, 2H), 7.89 (dd, J = 9.1, 2.3 Hz, 1H), 7.44 – 7.34 (m, 2H), 7.25 (t, J = 9.0 Hz, 1H), 6.89 (d, J = 9.1 Hz, 1H), 4.43 (s, 2H), 4.21 (p, J = 8.0 Hz, 1H), 3.72 (dd, J = 18.3, 4.1 Hz, 4H), 3.57 (s, 2H), 3.27 (d, J = 4.4 Hz, 2H), 2.30 – 2.16 (m, 2H), 2.08 – 1.96 (m, 2H), 1.95 – 1.72 (m, 2H). Synthesis of Example 54: 6-(4-(5-((7-((3,3-Difluorocyclobutyl)sulfonyl)-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)piperazin-1-y l)nicotinonitrile

Step 1 6-(4-(5-((7-((3,3-difluorocyclobutyl)thio)-4-oxo-3,4-dihydro phthalazin-1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0709] Following the procedure described in Example 53 and making non-critical variation as required to replace bromocyclobutane with 3-bromo-1,1-difluorocyclobutane, the crude product of 6-(4-(5-((7- ((3,3-difluorocyclobutyl)thio)-4-oxo-3,4-dihydrophthalazin-1 -yl)methyl)-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile was obtained as a yellow oil (crude). LCMS ESI m/z: 591.3 [M+H] + . Step 2 6-(4-(5-((7-((3,3-difluorocyclobutyl)sulfonyl)-4-oxo-3,4-dih ydrophthalazin-1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0710] Following the procedure described in Example 53 and making non-critical variation as required to replace 6-(4-(5-((7-(cyclobutylthio)-4-oxo-3,4-dihydrophthalazin-1-y l)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile with 6-(4-(5-((7-((3,3-difluorocyclobutyl)thio)-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)piperazin-1-y l)nicotinonitrile, 6-(4-(5-((7-((3,3- difluorocyclobutyl)thio)-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile was obtained as a white solid (16 mg, 22% yield). LCMS ESI m/z: 623.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.91 (s, 1H), 8.50 – 8.47 (m, 2H), 8.30 (d, J = 1.2 Hz, 1H), 8.27 (dd, J = 8.3, 1.6 Hz, 1H), 7.89 (dd, J = 9.1, 2.3 Hz, 1H), 7.46 – 7.39 (m, 1H), 7.36 (dd, J = 6.5, 2.1 Hz, 1H), 7.26 (t, J = 9.0 Hz, 1H), 6.89 (d, J = 9.1 Hz, 1H), 4.45 (s, 2H), 4.38 – 4.21 (m, 1H), 3.71 (dd, J = 18.0, 4.1 Hz, 4H), 3.57 (s, 2H), 3.30 (d, J = 20.9 Hz, 2H), 3.04 – 2.76 (m, 4H). Synthesis of Example 55: 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1-yl)methyl)benzoyl)piperazin-1- yl)pyrazine-2-carbonitrile

Methyl-5-((6-bromo-3-oxoisobenzofuran-1(3H)-ylidene)methyl)- 2-fluorobenzoate [0711] A solution of dimethyl (6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (5.00 g, 15.6 mmol), methyl 2-fluoro-5-formylbenzoate (3.12 g, 17.1 mmol) and Et 3 N (4.73 g, 46.72 mmol) in THF (50 mL) was stirred at rt under Ar (g) for 12 hours. The reaction mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to afford methyl 5-((6-bromo-3-oxoisobenzofuran-1(3H)- ylidene)methyl)-2-fluorobenzoate as a white solid (5.01 g, 85% yield). This was used without further purification. LCMS ESI m/z: 377 (M+H) + . Step 2 Methyl 5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoate [0712] A solution of methyl 5-((6-bromo-3-oxoisobenzofuran-1(3H)-ylidene)methyl)-2-fluor obenzoate (5.80 g, 13.3 mmol) and N 2 H 4 •H 2 O (1.33 g, 26.5 mmol) in THF (50 mL) was stirred at 70 °C for 4 h. The solids were filtered and washed with water (30 mL) to afford methyl 5-((7-bromo-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoate as a white solid (4.1 g, 79% yield). This was used without further purification. LCMS ESI m/z: 391 (M+H) + . Step 3 Methyl 2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1-yl)methyl)benzoate [0713] A mixture of tributyl(prop-1-ynyl)stannane (8.08 g, 24.5 mmol), methyl 5-[(7-bromo-4-oxo- 3H-phthalazin-1-yl)methyl]-2-fluoro-benzoate (4.00 g, 8.18 mmol) and Pd(dppf)Cl 2 (599 mg, 0.82 mmol) in 1,4-dioxane (40 mL) was stirred at 100 °C under Ar (g) for 5 hours. The reaction mixture was concentrated in vacuo and the residue was purified by silica gel chromatography (MeOH/DCM = 0 to 20%) to afford methyl 2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1-yl)methyl)benzoate as a white solid (3.2 g, 96% yield). LCMS ESI m/z: 351 [M+H] + . Step 4 2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1-yl)methyl)benzoic acid [0714] A mixture of methyl 2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1- yl)methyl)benzoate(3.20 g, 9.13 mmol) and NaOH (1.10 g, 27.40 mmol) in MeOH (30 mL) and H 2 O (10 mL) was stirred at rt for 2 hours. The organic solvent was removed in vacuo and aq HCl (1N) was added until the solution reached pH 5~6. The solids were collected by filtration, washed with H 2 O (5 mL) and dried under vacuum to give 2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1- yl)methyl)benzoic acid as a white solid (3.08 g, 90% yield). LCMS ESI m/z: 337 [M+H] + . Step 5 tert-Butyl 4-(6-cyanopyrazin-2-yl)piperazine-1-carboxylate [0715] To a solution of tert-butyl piperazine-1-carboxylate (265 mg, 1.07 mmol) in toluene (6 mL) was added 6-chloropyrazine-2-carbonitrile (150 mg, 1.07 mmol), triethylamine (326 mg, 3.22 mmol, 449 μL) at 25 °C . After stirring the reaction mixture at 100 °C for 3 hr. Water and EtOAc were added to the mixture, which became two phases. The organic layer was separated, washed with NaCl (aq), dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 20/1 to 2/1) to give tert-butyl 4-(6-cyanopyrazin-2- yl)piperazine-1-carboxylate (200 mg, crude) as a yellow solid. LCMS ESI m/z: 290.1 [M + H] + . Step 6 and 7 6-(4-(2-fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1-yl)methyl)benzoyl)piperazin-1- yl)pyrazine-2-carbonitrile [0716] Following the two-step procedure in Example 59, but starting with tert-butyl 4-(6- cyanopyrazin-2-yl)piperazine-1-carboxylate gave the title compound as a white solid. LCMS ESI m/z: 508.2 [M + H] + . 1 H NMR (400 MHz, DMSO-d6) δ12.62 (s, 1H), 8.64 (s, 1H), 8.35 (s, 1H), 8.20 (d, J = 8.2 Hz, 1H), 7.93 (s, 1H), 7.78 (dd, J = 8.2, 1.3 Hz, 1H), 7.45 – 7.38 (m, 2H), 7.29 – 7.23 (m, 1H), 4.34 (s, 2H), 3.75 (s, 4H), 3.59 (s, 2H), 3.34 (s, 2H), 2.09 (s, 3H). Synthesis of Example 56: 2-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1- yl)methyl)benzoyl)piperazin-1-yl)-N-methylisonicotinamide

2-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1-yl)methyl)benzoyl)piperazin-1- yl)-N-methylisonicotinamide [0717] Following the five-step procedure above in Example 55, but starting with 2-chloroisonicotinic acid gave the title compound as a white solid. LCMS ESI m/z: 539.2 [M + H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.63 (s, 1H), 8.55 (d, J = 4.6 Hz, 1H), 8.21 (dd, J = 7.7, 7.0 Hz, 2H), 7.93 (s, 1H), 7.78 (dd, J = 8.2, 1.3 Hz, 1H), 7.41 (dd, J = 6.2, 2.2 Hz, 2H), 7.25 (t, J = 9.3 Hz, 1H), 7.16 (s, 1H), 7.01 (d, J = 5.2 Hz, 1H), 4.34 (s, 2H), 3.75 (s, 2H), 3.63 (s, 2H), 3.50 (s, 2H), 3.32 (s, 2H), 2.78 (d, J = 4.5 Hz, 3H), 2.08 (s, 3H). Synthesis of Example 57: 4-(4-Fluoro-3-(4-(4,4,4-trifluorobutanoyl)piperazine-1-carbo nyl)benzyl)-6- (prop-1-ynyl)phthalazin-1(2H)-one

tert-Butyl 4-(4,4,4-trifluorobutanoyl)piperazine-1-carboxylate [0718] To a solution of 4,4,4-trifluorobutanoic acid (577 mg, 4.06 mmol) in DMF (10 mL) was added Et 3 N (1.64 g, 16.2 mmol) and HATU (2.32 g, 6.09 mmol). The reaction mixture was stirred at rt for 10 minutes, tert-butyl piperazine-1-carboxylate (1.0 g, 4.06 mmol) was added, and then stirred at rt for 3 hrs. The reaction mixture was poured into water (30 mL), extracted with Ethyl Acetate (50 mL x 3), washed with brine, dried and concentrated. The residue was purified by silica gel chromatography to give tert-butyl 4-(4,4,4-trifluorobutanoyl)piperazine-1-carboxylate (750 mg, 59% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ) δ 3.65 – 3.56 (m, 2H), 3.53 – 3.38 (m, 6H), 2.56 (m, 4H), 1.48 (s, 9H). Step 2 and 3 4-(4-Fluoro-3-(4-(4,4,4-trifluorobutanoyl)piperazine-1-carbo nyl)benzyl)-6-(prop-1-ynyl)phthalazin- 1(2H)-one [0719] Following the two-step procedure Example 59, but starting with tert-butyl 4-(4,4,4- trifluorobutanoyl)piperazine-1-carboxylate gave the title compound as a white solid. LCMS ESI m/z: 528.7 [M + H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.62 (s, 1H), 8.20 (d, J = 8.3 Hz, 1H), 7.92 (d, J = 13.2 Hz, 1H), 7.77 (d, J = 7.3 Hz, 1H), 7.46 – 7.34 (m, 2H), 7.25 (d, J = 7.2 Hz, 1H), 4.33 (s, 2H), 3.74 – 3.46 (m, 5H), 3.40 (m, 2H), 3.20 (m, 2H), 2.60 (m, 3H), 2.10 (s, 3H). Synthesis of Example 58: 3-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1- yl)methyl)benzoyl)piperazin-1-yl)-3-oxopropanenitrile 2-Cyanoacetyl chloride [0720] Oxalyl dichloride (2.24 g, 17.6 mmol, 1.54 mL) was added drop-wise to a solution of 2- cyanoacetic acid (1.00 g, 11.8 mmol) in DCM (15 mL) and DMF (0.1 mL) at 0 °C. The resultant mixture was stirred at rt for 4 hr. The mixture concentrated under vacuum to give 2-cyanoacetyl chloride (1.2 g, crude) as a black oil, which was used to next step without further purification. Step 2 tert-Butyl 4-(2-cyanoacetyl)piperazine-1-carboxylate [0721] 2-Cyanoacetyl chloride (1.22 g, 11.79 mmol) in DCM (10 mL) was added to a solution of tert- butyl piperazine-1-carboxylate (3.19 g, 12.97 mmol), Et 3 N (5.96 g, 58.93 mmol, 8.21 mL), DMAP (143.99 mg, 1.18 mmol) and DCM (10 mL) at 0 °C. The resultant mixture was stirred at rt for 12 hr. The mixture was concentrated to give a crude residue, which was purified by silica gel chromatography (petroleum ether: EtOAc = 10:1 to 1:1) to give tert-butyl 4-(2-cyanoacetyl)piperazine-1-carboxylate (1.00 g, 34% yield) as a yellow solid. 1 H NMR (400 MHz, CDCl3) δ 3.61 – 3.54 (m, 2H), 3.53 – 3.46 (m, 4H), 3.41 (dd, J = 9.9, 4.8 Hz, 4H), 1.44 (s, 9H). Step 3 and 4 3-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1-yl)methyl)benzoyl) piperazin-1- yl)-3-oxopropanenitrile [0722] Following the two-step procedure in Example 59, but starting with tert-butyl 4-(2- cyanoacetyl)piperazine-1-carboxylate gave the title compound as a light yellow solid. LCMS ESI m/z: 472.2 [M + H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.62 (s, 1H), 8.20 (d, J = 8.2 Hz, 1H), 7.92 (d, J = 12.6 Hz, 1H), 7.78 (d, J = 8.2 Hz, 1H), 7.44 – 7.35 (m, 2H), 7.24 (t, J = 8.8 Hz, 1H), 4.33 (s, 2H), 4.09 (s, 1H), 4.01 (s, 1H), 3.70– 3.52 (m, 4H), 3.42 (s, 2H), 3.20 (s, 2H), 2.11 (s, 3H). Synthesis of Example 59: 4-(3-(3-(2-Ethylphenyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[4 ,3-a]pyrazine- 7-carbonyl)-4-fluorobenzyl)-6-(prop-1-ynyl)phthalazin-1(2H)- one

Methyl 2-ethylbenzoate [0723] Into a 40 ml vial were added 2-ethylbenzoic acid (5.00 g, 33.3 mmol), DMF (20 mL), iodomethane (5.67 g, 40.0 mmol, 2.49 mL) and potassium carbonate (13.8 g, 99.9 mmol, 6.03 mL) at room temperature. The mixture was stirred for 3.5 hr at 75 °C. The mixture was extracted with Ethyl Acetate, washed with water. The organic phase was dried over Na 2 SO 4 , concentrated under reduced pressure to give methyl 2-ethylbenzoate (4.8 g, 88% yield) as a white solid. 1 H NMR (400 MHz, DMSO- d6) δ 7.76 (dd, J = 7.8, 1.3 Hz, 1H), 7.50 (td, J = 7.6, 1.4 Hz, 1H), 7.38 – 7.26 (m, 2H), 3.83 (s, 3H), 2.88 (q, J = 7.5 Hz, 2H), 1.16 (m, 3H). Step 2 2-Ethylbenzohydrazide [0724] A mixture of methyl 2-ethylbenzoate (4 g, 24.36 mmol) and Hydrazine monohydrate (9.76 g, 194.88 mmol, 9.50 mL) in EtOH (4 mL) was stirred at 100 °C under microwave for 3 hr. The reaction solution was concentrated under reduced pressure, diluted with DCM, washed with water. The organic phase was dried over Na 2 SO 4 and concentrated to give 2-ethylbenzohydrazide (2.56 g, 64% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ 9.40 (s, 1H), 7.30 (m, 4H), 4.43 (d, J = 4.0 Hz, 2H), 2.69 (q, J = 7.5 Hz, 2H), 1.14 (t, J = 7.5 Hz, 3H). Step 3 Benzyl 3-methoxy-5,6-dihydropyrazine-1(2H)-carboxylate [0725] To a solution of benzyl 3-oxopiperazine-1-carboxylate (5 g, 21.34 mmol) in DCM (200 mL) were added trimethyloxonium tetrafluoroborate (11.05 g, 74.71 mmol) and Na 2 CO 3 (45.25 g, 426.89 mmol) at room temperature. The mixture was stirred at room temperature for 6 hr. The mixture was poured into water. The organic layer was separated, dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by silica gel chromatography (eluent) to give benzyl 3-methoxy-5,6- dihydropyrazine-1(2H)-carboxylate (1 g, 19% yield) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ) δ 7.40 – 7.29 (m, 5H), 5.16 (s, 2H), 3.96 (s, 2H), 3.68 (s, 3H), 3.54 (s, 2H), 3.46 (m, 2H). Step 4 Benzyl 3-(2-ethylphenyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine -7(8H)-carboxylate [0726] Into a 40 mL sealed tube were added benzyl 6-methoxy-3,5-dihydro-2H-pyrazine-4-carboxylate (1.2 g, 4.83 mmol), toluene (70 mL) and 2-ethylbenzohydrazide (793.65 mg, 4.83 mmol) at room temperature. The solution was stirred overnight. The reaction system was concentrated under reduced pressure to remove the solvent. The residue was purified by silica gel chromatography (eluent) to give benzyl benzyl 3-(2-ethylphenyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine -7(8H)-carboxylate (1.5 g, 86% yield) as a yellow oil. LCMS ESI m/z: 363.2 [M+H] + . Step 5 3-(2-Ethylphenyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]p yrazine [0727] A mixture of benzyl 3-(2-ethylphenyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine -7(8H)- carboxylate (750 mg, 1.66 mmol) and Pd/C (500 mg, 4.12 mmol) in MeOH (5 mL) was stirred at room temperature for 2 h under hydrogen atmosphere. The mixture was filtered and concentrated to give 3-(2- ethylphenyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazi ne (320 mg, 85% yield) as a yellow oil. LCMS ESI m/z: 229.2 [M+H] + . Step 6

4-(3-(3-(2-Ethylphenyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[4 ,3-a]pyrazine-7-carbonyl)-4- fluorobenzyl)-6-(prop-1-ynyl)phthalazin-1(2H)-one [0728] A mixture of 2-fluoro-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)methyl]b enzoic acid (50 mg, 148.67 μmol), 3-(2-ethylphenyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]p yrazine (45 mg, 197.12 μmol), DIPEA (77 mg, 596.90 μmol), EDCI (43 mg, 225.13 μmol) and HOBt (30.2 mg, 223.51 μmol) in DMF (3 mL) was stirred at room temperature for 3 hr under nitrogen atmosphere. The mixture was extracted with Ethyl Acetate, washed with water. The organic phase was dried over Na 2 SO 4 , concentrated under reduced pressure. The residue was purified by prep-HPLC to give 4-(3-(3-(2-ethylphenyl)-5,6,7,8- tetrahydro-[1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl)-4-fluo robenzyl)-6-(prop-1-ynyl)phthalazin-1(2H)- one (5.0 mg, 6.1% yield) as a white solid. LCMS ESI m/z: 547.2 [M+H] + . 1 H NMR (400 MHz, DMSO- d 6 ) δ 12.59 (s, 1H), 8.17 (m, J = 27.4, 8.2 Hz, 1H), 7.88 (d, J = 50.9 Hz, 1H), 7.80 – 7.61 (m, 1H), 7.48 (dt, J = 17.5, 7.4 Hz, 3H), 7.43 – 7.23 (m, 4H), 5.01 (s, 1H), 4.69 (s, 1H), 4.39 – 4.28 (m, 2H), 3.95 (d, J = 91.4 Hz, 1H), 3.61 (s, 2H), 2.55 (q, J = 7.1 Hz, 2H), 2.13 – 2.00 (m, 3H), 1.10 – 0.99 (m, 3H). Synthesis of Example 60: 6-(6-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1- yl)methyl)benzoyl)-2,6-diazaspiro[3.3]heptan-2-yl)nicotinoni trile

tert-Butyl 6-(5-cyanopyridin-2-yl)-2,6-diazaspiro[3.3]heptane-2-carboxy late [0729] A mixture of 6-chloropyridine-3-carbonitrile (476 mg, 3.43 mmol), tert-butyl 2,6- diazaspiro[3.3]heptane-2-carboxylate (683 mg, 3.43 mmol) and K 2 CO 3 (1.19 g, 8.59 mmol) in MeCN (10 mL) was stirred at 60 °C for 16 hours. The reaction was diluted with H 2 O (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to afford tert-butyl 6-(5-cyanopyridin-2-yl)-2,6-diazaspiro[3.3]heptane-2- carboxylate as a white solid (1.03 g, 92% yield). This was used without further purification. LCMS ESI m/z: 301 [M+H] + . Step 2: 6-(2,6-Diazaspiro[3.3]heptan-2-yl)nicotinonitrile [0730] A solution of tert-butyl 6-(5-cyanopyridin-2-yl)-2,6-diazaspiro[3.3]heptane-2-carboxy late (400 mg, 1.33 mmol) in DCM (5 mL) and TFA (1 mL) was stirred at rt for 1 hour. The mixture was removed in vacuo to give 6-(2,6-diazaspiro[3.3]heptan-2-yl)nicotinonitrile as a yellow oil (266 mg, 99% yield). This was used without further purification. LCMS ESI m/z: 201 [M+H] + . Step 3

6-(6-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1-yl)methyl)benzoyl)-2,6- diazaspiro[3.3]heptan-2-yl)nicotinonitrile [0731] To a solution of 6-(2,6-diazaspiro[3.3]heptan-2-yl)nicotinonitrile (89 mg, 0.45 mmol) and 2- fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin-1- yl)methyl)benzoic acid (50 mg, 0.15 mmol) in DMF (1 mL) was added EDCI (43 mg, 0.22 mmol), HOBt (30mg, 0.22 mmol) and DIPEA (96 mg, 0.75 mmol). The reaction mixture was stirred at rt for 2 hours, then purified by prep-HPLC to give 6-(6- (2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin -1-yl)methyl)benzoyl)-2,6- diazaspiro[3.3]heptan-2-yl)nicotinonitrile as a white solid (45 mg, 58% yield). LCMS ESI m/z: 519 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 9.84 (s, 1H), 8.39 (d, J = 1.6 Hz, 1H), 8.36 (d, J = 8.2 Hz, 1H), 7.72 (d, J = 8.2 Hz, 1H), 7.70 (s, 1H), 7.61 (dd, J = 8.8, 2.2 Hz, 1H), 7.52 – 7.49 (m, 1H), 7.38 – 7.31 (m, 1H), 7.09 – 7.04 (m, 1H), 6.24 (d, J = 8.7 Hz, 1H), 4.38 (s, 2H), 4.31 – 4.21 (m, 8H), 2.10 (s, 3H). Synthesis of Example 61: 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1- yl)methyl)benzoyl)-3-methylpiperazin-1-yl)nicotinonitrile

tert-Butyl 4-(5-cyanopyridin-2-yl)-2-methylpiperazine-1-carboxylate [0732] A mixture of 6-chloronicotinonitrile (500 mg, 3.61 mmol), tert-butyl 2-methylpiperazine-1- carboxylate (730 mg, 3.64 mmol) and K 2 CO 3 (848 mg, 6.13 mmol) in MeCN (20 mL) was stirred at 60 °C for 12 hours. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to afford tert-butyl 4-(5-cyanopyridin-2-yl)-2-methylpiperazine-1-carboxylate as a white solid (700 mg, 63% yield). This was used without further purification. LCMS ESI m/z: 247 [M-Boc+H] + . Step 2 6-(3-Methylpiperazin-1-yl)nicotinonitrile hydrochloride [0733] To a solution of tert-butyl 4-(5-cyanopyridin-2-yl)-2-methylpiperazine-1-carboxylate (700 mg, 2.32 mmol) in 1,4-dioxane (10 mL) was added HCl/1,4-dioxane (4M, 10 mL). The mixture was stirred at rt for 1 hour. Solvent was removed in vacuo to afford 6-(3-methylpiperazin-1-yl)nicotinonitrile hydrochloride as a white solid (450 mg, 96% yield). This was used without further purification. LCMS ESI m/z: 203 [M+H] + . Step 3 6-(4-(2-fluoro-5-formylbenzoyl)-3-methylpiperazin-1-yl)nicot inonitrile [0734] To a solution of 6-(3-methylpiperazin-1-yl)nicotinonitrile (450 mg, 2.22 mmol) and 2-fluoro-5- formylbenzoic acid (549 mg, 3.26 mmol) in DMF (5 mL) was added EDCI (663 mg, 4.08 mmol), HOBt (467 mg, 3.46 mmol) and DIPEA (893 mg, 8.16 mmol). The reaction mixture was stirred at rt for 12 hours, then diluted with water (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to afford 6-(4-(2- fluoro-5-formylbenzoyl)-3-methylpiperazin-1-yl)nicotinonitri le as a white solid (636 mg, 81% yield). LCMS ESI m/z: 353 [M+H] + . Step 4 6-(4-(5-((6-Bromo-3-oxoisobenzofuran-1(3H)-ylidene)methyl)-2 -fluorobenzoyl)-3-methylpiperazin- 1-yl)nicotinonitrile [0735] A solution of 6-(4-(2-fluoro-5-formylbenzoyl)-3-methylpiperazin-1-yl)nicot inonitrile (336 mg, 0.99 mmol), dimethyl (6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (270 mg, 0.84 mmol) and Et 3 N (252 mg, 2.49 mmol) in THF (5 mL) was stirred at rt under Ar (g) for 12 hours. The reaction was diluted with water (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to afford 6-(4-(5-((6- bromo-3-oxoisobenzofuran-1(3H)-ylidene)methyl)-2-fluorobenzo yl)-3-methylpiperazin-1- yl)nicotinonitrile as a white solid (420 mg, 94% yield). This was used without further purification. LCMS ESI m/z: 539 (M+H) + . Step 5 6-(4-(5-((7-Bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoyl)-3-methylpiperazin-1- yl)nicotinonitrile [0736] A mixture of 6-(4-(5-((6-bromo-3-oxoisobenzofuran-1(3H)-ylidene)methyl)-2 -fluorobenzoyl)- 3-methylpiperazin-1-yl)nicotinonitrile (340 mg, 0.62 mmol) and N 2 H 4 •H 2 O (366 mg, 6.2 mmol) in THF (5 mL) was stirred at 70 °C for 3 hours. The solids were filtered and washed with water (10 mL) to afford 6-(4-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoyl)-3-methylpiperazin-1- yl)nicotinonitrile as a white solid (302 mg, 87% yield). LCMS ESI m/z: 561 (M+H) + . Step 6 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1-yl)methyl)benzoyl)-3- methylpiperazin-1-yl)nicotinonitrile [0737] A mixture of tributyl(prop-1-ynyl)stannane (176 mg, 0.53 mmol), 6-(4-(5-((7-bromo-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)-3-methylpipe razin-1-yl)nicotinonitrile (100 mg, 0.18 mmol) and Pd(dppf)Cl 2 (13 mg, 0.02 mmol) in 1,4-dioxane (5 mL) was stirred at 100 °C under Ar (g) for 16 hours. The reaction mixture was concentrated in vacuo and the residue was purified by prep-HPLC to afford 6-(4-(2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1-yl)methyl)benzoyl)-3- methylpiperazin-1-yl)nicotinonitrile as a white solid (32 mg, 35% yield). LCMS ESI m/z: 521 [M+H] + ; 1 H NMR (400 MHz, CDCl3) δ 10.48 (s, 1H), 8.46 – 8.31 (m, 2H), 7.71 (d, J = 7.3 Hz, 2H), 7.63 (dd, J = 9.0, 2.2 Hz, 1H), 7.32 (d, J = 8.5 Hz, 2H), 7.07 (t, J = 8.8 Hz, 1H), 6.59 (t, J = 7.9 Hz, 1H), 5.10 – 4.59 (m, 1H), 4.45 – 4.14 (m, 4H), 3.90 – 2.99 (m, 4H), 2.09 (s, 3H), 1.30 (d, J = 6.6 Hz, 3H). Synthesis of Example 62: 6-(8-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1- yl)methyl)benzoyl)-3,8-diazabicyclo[3.2.1]octan-3-yl)nicotin onitrile tert-Butyl 3-(5-cyanopyridin-2-yl)-3,8-diazabicyclo[3.2.1]octane-8-carb oxylate [0738] A mixture of 6-chloronicotinonitrile (500 mg, 3.61 mmol), tert-butyl-3,8- diazabicyclo[3.2.1]octane-8-carboxylate (766 mg, 3.61 mmol) and K 2 CO 3 (848 mg, 6.13 mmol) in MeCN (10 mL) was stirred at 60 °C for 12 hours. The mixture was diluted with water (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to afford tert-butyl 3-(5-cyanopyridin-2-yl)-3,8-diazabicyclo[3.2.1]octane-8- carboxylate as a white solid (1.02 g, 90% yield). This was used without further purification. LCMS ESI m/z : 247 [M+H] + . Step 2 6-(3,8-Diazabicyclo[3.2.1]octan-3-yl)nicotinonitrile [0739] To a solution of tert-butyl 3-(5-cyanopyridin-2-yl)-3,8-diazabicyclo[3.2.1]octane-8-carb oxylate (200 mg, 0.64 mmol) in 1,4-dioxane (1 mL) was added HCl/dioxane (4M, 1 mL). The mixture was stirred at rt for 1 hour. Solvent was removed in vacuo to afford 6-(3,8-diazabicyclo[3.2.1]octan-3- yl)nicotinonitrile as a white solid (130 mg, 96% yield). This was used without further purification. LCMS ESI m/z : 215 [M+H] + . Step 3 6-(8-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1-yl)methyl)benzoyl)-3,8- diazabicyclo[3.2.1]octan-3-yl)nicotinonitrile [0740] To a solution of 6-(3,8-diazabicyclo[3.2.1]octan-3-yl)nicotinonitrile (100 mg, 0.40 mmol) and 2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1-yl)methyl)benzoic acid (201 mg, 0.60 mmol) in DMF (2 mL) was added EDCI (114 mg, 0.60 mmol), HOBt (81 mg, 0.60 mmol) and DIPEA (155 mg, 1.20 mmol). The reaction was stirred at rt for 2 h. The reaction mixture was purified by prep- HPLC to afford 6-(8-(2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1-yl)methyl)benzoyl)- 3,8-diazabicyclo[3.2.1]octan-3-yl)nicotinonitrile as a white solid (55 mg, 26% yield). LCMS ESI m/z : 533 [M+H] + ); 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.63 (s, 1H), 8.51 (d, J = 2.3 Hz, 1H), 8.19 (d, J = 8.2 Hz, 1H), 7.94 (s, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.79 – 7.76 (m, 1H), 7.51 – 7.46 (m, 1H), 7.45 – 7.39 (m, 1H), 7.29 – 7.23 (m, 1H), 6.85 (d, J = 9.1 Hz, 1H), 4.83 – 4.77 (m, 1H), 4.34 (s, 2H), 4.26 (d, J = 11.5 Hz, 1H), 4.10 (d, J = 11.2 Hz, 1H), 3.87 – 3.81 (m, 1H), 3.14 (d, J = 11.3 Hz, 1H), 2.92 (d, J = 12.3 Hz, 1H), 2.07 (s, 3H), 1.93 – 1.80 (m, 2H), 1.71 – 1.58 (m, 2H). Synthesis of Example 63: 4-[[3-(3-cyclopropyl-6,8-dihydro-5H-[1,2,4]triazolo[4,3-a]py razine-7- carbonyl)-4-fluoro-phenyl]methyl]-6-prop-1-ynyl-2H-phthalazi n-1-one

N'-pyrazin-2-ylcyclopropanecarbohydrazide [0741] A mixture of pyrazin-2-ylhydrazine (1 g, 9.08 mmol) and Et 3 N (1.84 g, 18.16 mmol, 2.53 mL) in DCM (12 mL) was stirred at rt. Cyclopropanecarbonyl chloride (1.04 g, 9.99 mmol, 908.03 μL) was added dropwise to the reaction mixture at room temperature under nitrogen atmosphere. The solution was stirred at room temperature for 3 hours. The solution was concentrated under reduced pressure. The residue was purified by silica gel chromatography (DCM: MeOH= 12: 1) to give N'-pyrazin-2- ylcyclopropanecarbohydrazide (430 mg, 27% yield). LCMS ESI m/z : 179.1 [M + H] + . Step 2 3-Cyclopropyl-[1,2,4]triazolo[4,3-a]pyrazine [0742] A solution of N'-pyrazin-2-ylcyclopropanecarbohydrazide (200 mg, 841.79 μmol) in PPA (3 mL) was stirred at 150 °C for 2 hours and then for 36 hours at 100 °C under nitrogen atmosphere. The mixture was worked up with EtOAc and water. The mixture was added ammonium hydroxide until reached pH 1. The organic phase was dried over Na 2 SO 4 , concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM: MeOH = 10:1) to give 3-cyclopropyl-[1,2,4]triazolo[4,3- a]pyrazine (80 mg, 59% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6) δ 9.32 (d, J = 1.6 Hz, 1H), 8.61 (dd, J = 4.8, 1.6 Hz, 1H), 7.93 (d, J = 4.8 Hz, 1H), 2.48 – 2.43 (m, 1H), 1.18 (ddd, J = 11.6, 5.3, 3.2 Hz, 2H), 1.10 (qd, J = 5.0, 2.2 Hz, 2H). Step 3 3-Cyclopropyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyraz ine [0743] A mixture of 3-cyclopropyl-[1,2,4]triazolo[4,3-a]pyrazine (80 mg, 499.45 μmol) and Pd/C (60.66 mg, wet) in EtOH (5 mL) was stirred at room temperature under hydrogen atmosphere for 16 hours. The mixture was filtered. The filtrate was concentrated under reduced pressure to give 3- cyclopropyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin e (40 mg, 49% yield) as a colorless oil. LCMS ESI m/z : 165.2 [M + H] + . Step 4 4-[[3-(3-Cyclopropyl-6,8-dihydro-5H-[1,2,4]triazolo[4,3-a]py razine-7-carbonyl)-4-fluoro- phenyl]methyl]-6-prop-1-ynyl-2H-phthalazin-1-one [0744] A solution of 2-methyl-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)methyl]b enzoic acid (99.40 mg, 299.07 μmol), 3-cyclopropyl-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyraz ine(152-4) (60 mg, 365.39 μmol), DIPEA (141.41 mg, 1.10 mmol), EDCI (104.68 mg, 548.09 μmol) and HOBt (74.06 mg, 548.09 μmol) in DMF (8 mL) was stirred at room temperature under nitrogen atmosphere for 3 hours. The solution was dissolved in EtOAc (20 mL), washed with water (10 mL X 3). The organic phase was dried over Na 2 SO 4 , concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM: MeOH = 10: 1) to give 4-[[3-(3-cyclopropyl-6,8-dihydro-5H-[1,2,4]triazolo[4,3-a]py razine-7-carbonyl)-4-fluoro- phenyl]methyl]-6-prop-1-ynyl-2H-phthalazin-1-one (8.5 mg, 4.8% yield) as a white solid. LCMS ESI 483.2 [M + H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.52 (s, 1H), 8.35 (d, J = 8.2 Hz, 1H), 7.71 (d, J = 7.4 Hz, 2H), 7.47 – 7.31 (m, 2H), 7.11 (t, J = 8.9 Hz, 1H), 4.92 (d, J = 133.5 Hz, 2H), 4.32 – 3.70 (m, 6H), 2.10 (s, 3H), 1.70 – 1.65 (m, 1H), 1.13 (dd, J = 7.2, 3.3 Hz, 2H), 1.06 (d, J = 7.7 Hz, 2H). Synthesis of Example 64: 6-(3-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1- yl)methyl)benzoyl)-3,8-diazabicyclo[3.2.1]octan-8-yl)nicotin onitrile 6-(3-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1-yl)methyl)benzoyl)-3,8- diazabicyclo[3.2.1]octan-8-yl)nicotinonitrile [0745] Following the three-step procedure above in Example 55, but starting with tert-butyl 3,8- diazabicyclo[3.2.1]octane-3-carboxylate gave the title compound as a white solid. LCMS ESI m/z: 533.2 [M + H] + . 1 H NMR (400 MHz, DMSO-d6) δ12.61 (s, 1H), 8.52 (d, J = 2.1 Hz, 1H), 8.19 (d, J = 8.2 Hz, 1H), 7.90 (dd, J = 9.0, 2.3 Hz, 2H), 7.78 (dd, J = 8.2, 1.2 Hz, 1H), 7.50 – 7.29 (m, 2H), 7.24 (t, J = 8.9 Hz, 1H), 6.90 (d, J = 9.0 Hz, 1H), 4.80 (s, 1H), 4.60 (s, 1H), 4.33 (s, 3H), 3.27 (s, 1H), 3.11 (m, 1H), 2.99 (d, J = 12.6 Hz, 1H), 2.09 (m, 3H), 2.01 – 1.84 (m, 2H), 1.75 (m, 2H). Synthesis of Example 65: Preparation of 4-(4-fluoro-3-(3-(trifluoromethyl)-5,6,7,8-tetrahydro- [1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl)benzyl)-6-(prop-1- yn-1-yl)phthalazin-1(2H)-one 6-Bromo-4-(4-fluoro-3-(3-(trifluoromethyl)-5,6,7,8-tetrahydr o-[1,2,4]triazolo[4,3-a]pyrazine-7- carbonyl)benzyl)phthalazin-1(2H)-one [0746] To a solution of 5-[(7-bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro-benzoi c acid (450 mg, 1.19 mmol) in DMF (10 mL) was added HATU (544.39 mg, 1.43 mmol) and Et 3 N (482.92 mg, 4.77 mmol, 665.19 μL). The mixture was stirred at rt for 0.5 hour.3-(trifluoromethyl)-5,6,7,8-tetrahydro- [1,2,4]triazolo[4,3-a]pyrazine (272.75 mg, 1.19 mmol) was added, then stirred at rt for another 16 h. The reaction mixture was poured into water (50 mL), extracted with EtOAc (50 mL x 3). The extracts were washed with brine, dried and concentrated to give 6-bromo-4-[[4-fluoro-3-[3-(trifluoromethyl)-6,8- dihydro-5H-[1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl]phenyl] methyl]-2H-phthalazin-1-one (700 mg, crude) as a white solid. LCMS ESI 548.7 [M - H] -. Step 2 4-(4-Fluoro-3-(3-(trifluoromethyl)-5,6,7,8-tetrahydro-[1,2,4 ]triazolo[4,3-a]pyrazine-7- carbonyl)benzyl)-6-(prop-1-yn-1-yl)phthalazin-1(2H)-one [0747] To a solution of 6-bromo-4-[[4-fluoro-3-[3-(trifluoromethyl)-6,8-dihydro-5H- [1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl]phenyl]methyl]-2H- phthalazin-1-one (700 mg, 253.95 μmol) in PhCH 3 (4 mL) was added tributyl(prop-1-ynyl)stannane (835.77 mg, 2.54 mmol) and Pd(PPh 3 ) 4 (29.35 mg, 25.39 μmol) The mixture was stirred at 100 °C under microwave for 1 hour. CsF (1 g) and water (20 mL) were added to the reaction mixture. The mixture was stirred at rt for another 0.5 hour. The mixture was extracted with EtOAc (30 mL x 3), dried and concentrated to the residue. The residue was purified by Prep-HPLC to give 4-[[4-fluoro-3-[3-(trifluoromethyl)-6,8-dihydro-5H-[1,2,4]tr iazolo[4,3- a]pyrazine-7-carbonyl]phenyl]methyl]-6-prop-1-ynyl-2H-phthal azin-1-one (21.28 mg, 16 % yield) as a light yellow solid. LCMS ESI 511.2 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.61 (s, 1H), 8.20 (d, J = 8.2 Hz, 1H), 7.90 (d, J = 21.3 Hz, 1H), 7.77 (dd, J = 8.2, 1.3 Hz, 1H), 7.53 – 7.46 (m, 1H), 7.41 (d, J = 5.5 Hz, 1H), 7.31 (t, J = 8.8 Hz, 1H), 5.03 (m, 1H), 4.71 (m, 1H), 4.35 (s, 2H), 4.23 (m, 1H), 4.15 (m, 1H), 4.02 (m, 1H), 3.70 (m, 1H), 2.10 (s, 3H). Synthesis of Example 66: 6-((1R,4R)-5-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydro phthalazin- 1-yl)methyl)benzoyl)-2,5-diazabicyclo[2.2.1]heptan-2-yl)nico tinonitrile

6-((1R,4R)-2,5-Diazabicyclo[2.2.1]heptan-2-yl)nicotinonitril e [0748] Following the general procedure above in Example 55, but starting with (1R,4R)-tert-butyl 2,5- diazabicyclo[2.2.1]heptane-2-carboxylate and 6-chloronicotinonitrile gave the title compound as a colorless oil. LCMS ESI m/z: 201.1 [M+H] + . Step 3 6-((1R,4R)-5-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydro phthalazin-1-yl)methyl)benzoyl)-2,5- diazabicyclo[2.2.1]heptan-2-yl)nicotinonitrile [0749] Following the procedure in Example 55, but starting with 6-((1R,4R)-2,5- diazabicyclo[2.2.1]heptan-2-yl)nicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 518.7 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.61 (d, J = 18.5 Hz, 1H), 8.51 – 8.41 (m, 1H), 8.20 (dd, J = 12.8, 8.2 Hz, 1H), 7.96 – 7.73 (m, 3H), 7.53 – 7.11 (m, 3H), 6.59 (d, J = 38.1 Hz, 1H), 4.98 (s, 1H), 4.33 (m, 2H), 4.19 (s, 1H), 3.67 – 3.56 (m, 1H), 3.42 (m, 3H), 2.10 (t, J = 8.3 Hz, 3H), 1.97 (m, 2H). Synthesis of Example 67: 6-(4-Fluoro-1-(2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperidin-4-yl)nicotin onitrile tert-Butyl 4-(5-cyanopyridin-2-yl)-4-hydroxypiperidine-1-carboxylate [0750] A solution of 2,5-dibromopyridine (705 mg, 3 mmol) in THF (20 mL) was cooled to -78 °C, then n-BuLi (2.5M in heptane, 1.32 mL, 3.3 mmol) was addded dropwise under Ar (g). The reaction was stirred at -78 °C for 15 min, then tert-butyl 4-oxopiperidine-1-carboxylate (716 mg, 3.6 mmol) was added. The reaction was kept at -78°C for 20 min, then quenched with sat. NH 4 Cl solution (10 mL) and extracted with EtOAc (30 × 3 mL). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 20 to 40%) to give tert-butyl 4-(5-bromopyridin-2-yl)-4-hydroxypiperidine-1-carboxylate as a white solid (520 mg, 49% yield). LCMS ESI 357 (M+H) + . Step 2 tert-Butyl 4-(5-cyanopyridin-2-yl)-4-hydroxypiperidine-1-carboxylate [0751] A mixture of tert-butyl 4-oxopiperidine-1-carboxylate (520 mg, 1.46 mol), Zn(CN) 2 (347 mg, 2.92 mmol) and Pd(PPh 3 ) 4 in dry DMF (10 mL) was stirred at 100 ºC under Ar (g) for 2 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give tert-butyl 4-(5-cyanopyridin-2-yl)-4-hydroxypiperidine-1-carboxylate as light yellow oil (380 mg, 86% yield). LCMS ESI 304 (M+H) + . Step 3: tert-Butyl 4-(5-cyanopyridin-2-yl)-4-fluoropiperidine-1-carboxylate [0752] To a solution of tert-butyl 4-(5-cyanopyridin-2-yl)-4-hydroxypiperidine-1-carboxylate (380 mg, 1.25 mmol) in DCM (10 mL) was added BAST (966 mg, 4.37 mmol) in portions at 0 °C. The reaction was stirred at rt for 12 hours. The mixture was diluted with H 2 O (10 mL) and extracted with DCM (3 x 20 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 0 to 10%) to give tert-butyl 4-(5- cyanopyridin-2-yl)-4-fluoropiperidine-1-carboxylate as a yellow oil (170 mg, 45% yield). LCMS ESI m/z: 306 (M+H) + . 1 H NMR (400 MHz, CDCl 3 ) δ 8.20 (s, 1H), 8.02 (dd, J = 8.0, 2.0 Hz, 1H), 7.73 (t, J = 7.6 Hz, 1H), 4.15 – 4.05 (m, 4H), 3.18 – 3.03 (m, 2H), 2.38 – 2.15 (m, 2H), 1.49 (s, 9H). Step 4 6-(4-Fluoropiperidin-4-yl)nicotinonitrile [0753] To a solution of tert-butyl 4-(5-cyanopyridin-2-yl)-4-fluoropiperidine-1-carboxylate (115 mg, 0.37 mmol) in 1,4-dioxane (1 mL) was added HCl/dioxane (4 M , 1 mL). The mixture was stirred at rt for 1 hour, then concentrated in vacuo to afford 6-(4-fluoropiperidin-4-yl)nicotinonitrileas a white solid (75 mg, 98% yield). This was used without further purification. LCMS ESI m/z: 206 [M+H] + . Step 5 6-(4-Fluoro-1-(2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dih ydrophthalazin-1- yl)methyl)benzoyl)piperidin-4-yl)nicotinonitrile [0754] To a solution of 6-(4-fluoropiperidin-4-yl)nicotinonitrile (75 mg, 0.31 mmol) and 2-fluoro-5- ((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin-1-yl)methyl )benzoic acid (167 mg, 0.40 mmol) in DMF (2 mL) was added EDCI (77 mg, 0.50 mmol), HOBt (67 mg, 0.50 mmol) and DIEPA (214 mg, 1.66 mmol). The reaction mixture was stirred ar rt for 2 hours. The reaction was purified by prep-HPLC to give 6-(4-fluoro-1-(2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dih ydrophthalazin-1-yl)methyl) benzoyl)piperidin-4-yl)nicotinonitrile as a white solid (65 mg, 37% yield). LCMS ESI m/z: 524 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.13 (s, 1H), 8.85 (d, J = 1.0 Hz, 1H), 8.35 (d, 1H), 8.03 (dd, J = 8.3, 2.1 Hz, 1H), 7.77 – 7.66 (m, 3H), 7.37 (dd, J = 6.1 Hz, 1H), 7.33 – 7.27 (m, 1H), 7.07 (t, J = 8.8 Hz, 1H), 4.82 (dd, J = 13.6 Hz, 1H), 4.25 (s, 2H), 3.56 (t, 2H), 3.21 (t, J = 12.7 Hz, 1H), 2.56 – 2.16 (m, 2H), 2.09 (s, 3H), 2.03 – 1.93 (m, 1H), 1.86 – 1.73 (m, 1H). Synthesis of Example 68 : 6-((1S,4S)-5-(2-fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydro phthalazin-1- yl)methyl)benzoyl)-2,5-diazabicyclo[2.2.1]heptan-2-yl)nicoti nonitrile tert-Butyl(1S,4S)-5-(5-cyano-2-pyridyl)-2,5-diazabicyclo[2.2 .1]heptane-2-carboxylate [0755] A mixture of tert-butyl (1S,4S)-2,5-diazabicyclo[2.2.1]heptane-2-carboxylate (200.00 mg, 1.01 mmol), 6-chloropyridine-3-carbonitrile (181.70 mg, 1.31 mmol) and K 2 CO 3 (278.42 mg, 2.02 mmol) in NMP (8 mL) was stirred at room temperature. The mixture was stirred for 3 hours at 80°C under nitrogen atmosphere. The mixture was dissolved in EtOAc (20 mL), washed with water (20 mL x 3). The organic phase was dried over Na 2 SO 4 , concentrated under reduced pressure. The residue was purified by Prep- TLC (petroleum ether: EtOAc = 1: 1) to afford tert-butyl (1S,4S)-5-(5-cyano-2-pyridyl)-2,5- diazabicyclo[2.2.1]heptane-2-carboxylate (270 mg, 89% yield) as a colorless semi-solid. LCMS ESI m/z 245.1 [M - 56] + . Step 2 6-[(1S,4S)-2,5-Diazabicyclo[2.2.1]heptan-2-yl]pyridine-3-car bonitrile [0756] A mixture of tert-butyl (1S,4S)-5-(5-cyano-2-pyridyl)-2,5-diazabicyclo[2.2.1]heptane -2- carboxylate (270 mg, 898.94 μmol) and HCl in MeOH (4 mol/L 5 mL) was stirred at rt for 1 hour. The reaction system was concentrated under reduced pressure to give 6-[(1S,4S)-2,5- diazabicyclo[2.2.1]heptan-2-yl]pyridine-3-carbonitrile (200 mg, crude) as a colorless oil. LCMS ESI m/z 201.1 [M + H] + . Step 3 6-[(1S,4S)-5-[2-Fluoro-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin -1-yl)methyl]benzoyl]-2,5- diazabicyclo[2.2.1]heptan-2-yl]pyridine-3-carbonitrile [0757] A mixture of 2-fluoro-5-[(4-oxo-7-prop 1 ynyl 3H phthalazin-1-yl)methyl]benzoic acid (50 mg, 148.67 μmol), 6-[(1S,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl]pyridine-3-car bonitrile (35.72 mg, 178.40 μmol), EDCI (42.59 mg, 223.00 μmol), HOBt (30.13 mg, 223.00 μmol) and DIPEA (57.54 mg, 446.01 μmol) in DMF (5 mL) was stirred for 3 hours at room temperature under nitrogen atmosphere. The solution was poured into water (10 mL), extracted with EtOAc (10 mL X 3). The organic layers were dried over Na 2 SO 4 , concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM: MeOH = 10: 1) to give 6-[(1S,4S)-5-[2-fluoro-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin -1- yl)methyl]benzoyl]-2,5-diazabicyclo[2.2.1]heptan-2-yl]pyridi ne-3-carbonitrile (7.0 mg, 9% yield) as a white solid. LCMS ESI m/z 519.2 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.63 (s, 1H), 8.52 – 8.39 (m, 1H), 8.20 (dd, J = 12.7, 8.2 Hz, 1H), 7.94 – 7.74 (m, 3H), 7.49 – 7.31 (m, 2H), 7.31 – 7.14 (m, 1H), 6.63 (s, 1H), 4.98 (s, 1H), 4.32 (dd, J = 27.3, 4.2 Hz, 2H), 4.18 (s, 1H), 3.84 – 3.54 (m, 2H), 3.43 (dd, J = 14.7, 9.9 Hz, 2H), 2.11 (d, J = 5.4 Hz, 3H), 1.97 (dd, J = 27.2, 11.0 Hz, 2H). Synthesis of Example 69: 2-((1-(2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophth alazin-1- yl)methyl)benzoyl)azetidin-3-yl)(methyl)amino) isonicotinonitrile tert-Butyl 3-((4-cyanopyridin-2-yl)(methyl)amino)azetidine-1-carboxylat e [0758] A mixture of 2-chloroisonicotinonitrile (100 mg, 0.72 mmol), tert-butyl 3- (methylamino)azetidine-1-carboxylate (400 mg, 2.17 mmol) and Et 3 N (150 mg, 1.44 mmol) was stirred at 90 °C for 16 hours. The mixture was cooled to rt, diluted with H 2 O (5 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 40 to 60%) to afford tert-butyl 3-((4-cyanopyridin-2-yl)(methyl)amino)azetidine-1-carboxylat e as a colorless oil (110 mg, 53% yield). LCMS ESI m/z 289 [M+H] + . Step 2 2-(Azetidin-3-yl(methyl)amino)isonicotinonitrile [0759] To a solution of tert-butyl 3-((4-cyanopyridin-2-yl)(methyl)amino)azetidine-1-carboxylat e (0.11 g, 0.38 mmol) in DCM (5 mL) was added TFA (0.5 mL). The mixture was stirred at rt for 1 hour. The solution was concentrated in vacuo to give 2-(azetidin-3-yl(methyl)amino)isonicotinonitrile as a colorless oil (70 mg, 97% yield). This was used without further purification. LCMS ESI m/z 189 [M+H] + . Step 3 2-((1-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophth alazin-1-yl)methyl)benzoyl)azetidin-3- yl)(methyl)amino)isonicotinonitrile [0760] To a solution of 2-(azetidin-3-yl(methyl)amino)isonicotinonitrile (70 mg, 0.40 mmol) and 2- fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin-1- yl)methyl)benzoic acid (147 mg, 0.44 mmol) in DMF (5 mL) was added EDCI (115 mg, 0.60 mmol), HOBt (81 mg, 0.60 mmol) and DIPEA (154 mg, 1.20 mmol). The mixture was stirred at rt for 16 hours. The reaction was purified by prep-HPLC to give 2-((1-(2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophth alazin-1-yl)methyl)benzoyl)azetidin- 3-yl)(methyl)amino)isonicotinonitrile as a white solid (10 mg, 4% yield). LCMS ESI 507 (M+H) + ; 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.63 (s, 1H), 8.61 (t, J = 5.9 Hz, 1H), 8.43 (d, J = 6.7 Hz, 1H), 8.20 (d, J = 8.2 Hz, 1H), 7.95 (s, 1H), 7.90 (s, 1H), 7.77 (dd, J = 8.2, 1.4 Hz, 1H), 7.35 (dd, J = 6.7, 2.2 Hz, 1H), 7.25 (dd, J = 6.6, 1.4 Hz, 1H), 7.19 (dd, J = 10.1, 8.6 Hz, 1H), 4.82 (dd, J = 13.2, 11.4 Hz, 1H), 4.56 (dd, J = 13.5, 6.2 Hz, 1H), 4.51 – 4.42 (m, 1H), 3.80 – 3.69 (m, 2H), 3.68 – 3.60 (m, 2H), 3.16 (s, 3H), 2.10 (s, 3H). Synthesis of Example 70: 6-((1-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophth alazin-1- yl)methyl)benzoyl)azetidin-3-yl)(methyl)amino)nicotinonitril e 2,2,2-trifluoroacetate

tert-Butyl 3-((5-cyanopyridin-2-yl)(methyl)amino)azetidine-1-carboxylat e [0761] A mixture of 6-chloropyridine-3-carbonitrile (500 mg, 3.61 mmol), tert-butyl N-[2- (ethylamino)ethyl]carbamate (672 mg, 3.61 mmol) and K 2 CO 3 (1.25 g, 9.02 mmol) in MeCN (20 mL) was stirred at 60 °C for 16 hours. The reaction was concentrated in vacuo, diluted with water (30 mL) and extracted with EtOAc (30 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 10 to 20%) to give tert-butyl 3-((5-cyanopyridin-2- yl)(methyl)amino)azetidine-1-carboxylate as a white solide (1.2 g, 70% yield). LCMS ESI m/z 291 [M+H] + . Step 2 6-(Azetidin-3-yl(methyl)amino)nicotinonitrile [0762] A solution of tert-butyl 3-((5-cyanopyridin-2-yl)(methyl)amino)azetidine-1-carboxylat e (200 mg, 0.69 mmol) in DCM (5 mL) and TFA (0.5 mL) was stirred at rt for 1 hour. The solvent was removed in vacuo to give 6-(azetidin-3-yl(methyl)amino)nicotinonitrile as a yellow oil (129 mg, quant.). This was used without further purification. LCMS ESI m/z 189 [M+H] + . Step 3 6-((1-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophth alazin-1-yl)methyl)benzoyl)azetidin-3- yl)(methyl)amino)nicotinonitrile 2,2,2-trifluoroacetate [0763] To a solution of 6-(azetidin-3-yl(methyl)amino)nicotinonitrile (80 mg, 0.13 mmol) and 2- fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin-1- yl)methyl)benzoic acid (50 mg, 0.13mmol) in DMF (2 mL) was added EDCI (38 mg, 0.2 mmol), HOBt (27 mg, 0.2 mmol) and DIPEA (88 mg, 0.67 mmol). The reaction mixture was stirred at rt for 2 hours. The reaction was purified by prep-HPLC to give 6-((1-(2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophth alazin-1-yl)methyl)benzoyl)azetidin-3- yl)(methyl)amino)nicotinonitrile 2,2,2-trifluoroacetate as a white solid (17.5 mg, 21% yield). LCMS ESI m/z 507 (M+H) + ; 1 H NMR (400 MHz, DMSO–d 6 ) δ 12.63 (s, 1H), 9.03 (s, 1H), 8.62 (s, 1H), 8.30 (dd, J = 9.4, 1.7 Hz, 1H), 8.20 (d, J = 8.2 Hz, 1H), 7.92 (s, 1H), 7.78 (d, J = 8.3 Hz, 1H), 7.47 (s, 1H), 7.35 (d, J = 9.3 Hz, 2H), 7.26 – 7.15 (m, 1H), 4.79 (t, J = 11.4 Hz, 1H), 4.64 – 4.45 (m, 2H), 4.30 (s, 2H), 3.79 (d, J = 10.8 Hz, 1H), 3.59 (d, J = 14.2 Hz, 1H), 3.22 (s, 3H), 2.11 (s, 3H). Synthesis of Example 71: 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1- yl)methyl)benzoyl)-3,3-dimethyl-2-oxopiperazin-1-yl)nicotino nitrile

tert-Butyl 4-(5-cyanopyridin-2-yl)-2,2-dimethyl-3-oxopiperazine-1-carbo xylate [0764] A mixture of tert-butyl 2,2-dimethyl-3-oxopiperazine-1-carboxylate (600 mg, 2.63 mmol), 6- bromonicotinonitrile (481 mg, 2.63 mmol), K 2 CO 3 (726 mg, 5.26 mmol), CuI (5 mg, 0.03 mmol) and TMEDA (31 mg, 0.26 mmol) in 1,4-dioxane (3 mL) was stirred at 120 °C under Ar (g) for 16 h. The solids were removed by filtration, and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 9 %) to give tert-butyl 4-(5-cyanopyridin-2-yl)- 2,2-dimethyl-3-oxopiperazine-1-carboxylate (230 mg, 26% yield) as a white solid. LCMS ESI m/z 331 [M+H] + . Step 2 6-(3,3-Dimethyl-2-oxopiperazin-1-yl)nicotinonitrile [0765] A solution of tert-butyl 4-(5-cyanopyridin-2-yl)-2,2-dimethyl-3-oxopiperazine-1-carbo xylate (230 mg, 0.70 mmol) in HCl/dioxane (4M, 5 mL) was stirred at 25 °C for 1 hour. The reaction mixture was concentrated in vacuo to give 6-(3,3-dimethyl-2-oxopiperazin-1-yl)nicotinonitrile as a white solid (180 mg, 97% yield). This was used without further purification. LCMS ESI m/z 231 [M+H] + . Step 3

6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1-yl)methyl)benzoyl)-3,3- dimethyl-2-oxopiperazin-1-yl)nicotinonitrile [0766] A solution of 2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1-yl)methyl) benzoic acid (304 mg, 0.90 mmol) in SOCl 2 (1.95 g, 16.42 mmol) was stirred at 25 °C under Ar (g) for 3 hours. The mixture was concentrated in vacuo to obtain a white solid. The obtained solid was dissolved in DCM (20 mL), then a solution of 6-(3,3-dimethyl-2-oxopiperazin-1-yl)nicotinonitrile (180 mg, 0.82 mmol) and Et 3 N (249 mg, 2.46 mmol) in DCM (10 mL) was added. The mixture was stirred at rt under Ar (g) for 4 hours, then diluted with H 2 O (20 mL) and extracted with DCM (30 mL x 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 50%) to afford 6-(4-(2-fluoro-5-((4- oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin-1-yl)methyl)ben zoyl)-3,3-dimethyl-2-oxopiperazin-1- yl)nicotinonitrile as a white solid (174 mg, 39% yield). LCMS ESI 549 (M+H) + ; 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.64 (s, 1H), 8.93 (dd, J = 2.3, 0.8 Hz, 1H), 8.31 (dd, J = 8.9, 2.3 Hz, 1H), 8.22 – 8.20 (m, 1H), 8.20 – 8.18 (m, 1H), 7.95 (s, 1H), 7.78 (dd, J = 8.2, 1.4 Hz, 1H), 7.45 (dd, J = 6.5, 2.1 Hz, 1H), 7.42 – 7.37 (m, 1H), 7.29 – 7.22 (m, 1H), 4.34 (s, 2H), 4.06 – 4.00 (m, 2H), 3.58 (s, 2H), 2.08 (d, J = 2.1 Hz, 3H), 1.83 (s, 6H). Synthesis of Example 72: 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1- yl)methyl)benzoyl)-2-methylpiperazin-1-yl)nicotinonitrile

tert-Butyl 4-(5-cyanopyridin-2-yl)-3-methylpiperazine-1-carboxylate [0767] To a solution of 6-chloropyridine-3-carbonitrile (1.5 g, 10.83 mmol) in NMP (15 mL) was added tert-butyl 3-methylpiperazine-1-carboxylate (2.17 g, 10.83 mmol) and K 2 CO 3 (2.99 g, 21.65 mmol). The reaction mixture was stirred at 80 °C under N 2 (g) for 18 hours. The reaction mixture was worked up with water (20 mL) and EtOAc (30 mL). The organic layer was washed with brine, dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 20/1 to 4/1) to give tert-butyl 4-(5-cyano-2-pyridyl)-3-methyl-piperazine-1-carboxylate (1.8 g, 55% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 8.42 (d, J = 2.0 Hz, 1H), 7.63 (dd, J = 9.0, 2.3 Hz, 1H), 6.56 (d, J = 9.0 Hz, 1H), 4.53 (s, 1H), 4.12 (dd, J = 14.3, 7.1 Hz, 2H), 3.98 (d, J = 24.8 Hz, 1H), 3.31 – 3.12 (m, 2H), 3.02 (s, 1H), 1.49 (s, 9H), 1.20 (d, J = 6.7 Hz, 3H). Step 2 6-(2-Methyl-4-piperazin-1-yl)nicotinonitrile [0768] A solution of tert-butyl 4-(5-cyano-2-pyridyl)-3-methyl-piperazine-1-carboxylate (1.8 g, 5.95 mmol) in EtOAc (5 mL) was added HCl/EA (12 mL, 4 M). The reaction mixture was stirred at rt for 3 hours. The mixture was concentrated to give 6-(2-methylpiperazin-1-yl)pyridine-3-carbonitrile (1.4 g, crude). LCMS ESI m/z 203.2 [M + H] + . Step 3 6-(4-(2-Fluoro-5-formylbenzoyl)-2-methylpiperazin-1-yl)nicot inonitrile [0769] To a solution of 2-fluoro-5-formyl-benzoic acid (352.14 mg, 2.09 mmol) in DMF (8 mL) was added HATU (955.69 mg, 2.51 mmol) and Et 3 N (423.89 mg, 4.19 mmol, 583.87 μL). The reaction mixture was stirred at rt for 15 min.6-(2-Methylpiperazin-1-yl)pyridine-3-carbonitrile (500 mg, 2.09 mmol) was added to the mixture and the reaction mixture was stirred at rt for another 8 hours. The mixture was poured into water (20 mL), extracted with EtOAc (20 mL X 3). The organic layer was concentrated to the crude. The crude was purified by silica gel chromatography (petroleum ether/EtOAc = 2/1 ) to give 6-[4-(2-fluoro-5-formyl-benzoyl)-2-methyl-piperazin-1-yl]pyr idine-3-carbonitrile (550 mg, 75% yield) as a white solid. LCMS ESI m/z 353.1 [M + H] + . Step 4 6-(4-(5-((6-Bromo-3-oxoisobenzofuran-1(3H)-ylidene)methyl)-2 -fluorobenzoyl)-2-methylpiperazin- 1-yl)nicotinonitrile [0770] To a solution of 5-bromo-3-dimethoxyphosphoryl-3H-isobenzofuran-1-one (210 mg, 654.08 μmol) in THF (10 mL) was added 6-[4-(2-fluoro-5-formyl-benzoyl)-2-methyl-piperazin-1-yl]pyr idine-3- carbonitrile (276.57 mg, 784.90 μmol) and Et 3 N (198.56 mg, 1.96 mmol, 273.50 μL). The reaction mixture was stirred at rt for 16 hours. The mixture was poured into sat. NaHSO 3 , extracted with DCM (30 mL x 3), dried over Na 2 SO 4 . The organic layer was concentrated to give 6-[4-[5-[(6-bromo-3-oxo- isobenzofuran-1-ylidene)methyl]-2-fluoro-benzoyl]-2-methyl-p iperazin-1-yl]pyridine-3-carbonitrile (330 mg, crude) as a light yellow solid. LCMS ESI m/z 549.6 [M + H] + . Step 5

6-(4-(5-((7-Bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoyl)-2-methylpiperazin-1- yl)nicotinonitrile [0771] A solution of 6-[4-[5-[(Z)-(6-bromo-3-oxo-isobenzofuran-1-ylidene)methyl]- 2-fluoro-benzoyl]- 2-methyl-piperazin-1-yl]pyridine-3-carbonitrile (330 mg, 602.88 μmol) and N 2 H 4 •H 2 O (602.88 μmol, 2 mL, 80% purity) in THF (5 mL) was heated to 80 °C for 3 hours. The mixture was poured into water (20 mL), extracted with DCM (20 mL x 3). The extracts was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography to give 6-[4-[5-[(7-bromo- 4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro-benzoyl]-2-methyl- piperazin-1-yl]pyridine-3-carbonitrile (230 mg, 68% yield) as a light yellow gum. LCMS ESI m/z 561.1 [M + H] + . Step 6 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1-yl)methyl)benzoyl)-2- methylpiperazin-1-yl)nicotinonitrile [0772] To a mixture of 6-[4-[5-[(7-bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro- benzoyl]-2- methyl-piperazin-1-yl]pyridine-3-carbonitrile (100 mg, 178.12 μmol) in toluene (2 mL) was added Pd(PPh 3 ) 4 (20.58 mg, 17.81 μmol) and tributyl(prop-1-ynyl)stannane (117.24 mg, 356.25 μmol). The reaction mixture was stirred at 100 °C under microwave for 1 hr. The mixture was purified by Prep- HPLC to give 6-[4-[2-fluoro-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)me thyl]benzoyl]-2-methyl- piperazin-1-yl]pyridine-3-carbonitrile (24.57 mg, 27% yield) as a white solid; LCMS ESI 520.8 [M + H] + ; 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.63 (s, 1H), 8.52 (t, J = 2.5 Hz, 1H), 8.20 (dd, J = 8.2, 1.0 Hz, 1H), 7.90 (ddd, J = 15.0, 11.3, 7.4 Hz, 2H), 7.77 (d, J = 8.1 Hz, 1H), 7.41 (ddd, J = 23.5, 13.5, 5.2 Hz, 2H), 7.26 (t, J = 9.0 Hz, 1H), 6.88 (dd, J = 11.6, 9.3 Hz, 1H), 4.79 (s, 1H), 4.53 (s, 1H), 4.46 – 4.10 (m, 4H), 3.44 (d, J = 13.1 Hz, 1H), 3.29 – 2.99 (m, 3H), 2.08 (d, J = 4.8 Hz, 3H), 1.02 (dd, J = 99.0, 5.5 Hz, 3H). Synthesis of Example 73: 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1- yl)methyl-d2)benzoyl)piperazin-1-yl)nicotinonitrile 6-(4-(5-((7-Bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl-d2 )-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile [0773] A solution of 6-[4-[5-[(7-bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro- benzoyl]piperazin- 1-yl]pyridine-3-carbonitrile (100 mg, 0.18 mmol) and DBU (56 mg, 0.37 mmol) in dry DMSO-d 6 (15 mL) and D 2 O (5 mL) were stirred at 70 ºC under Ar (g) for 18 hours. The reaction was cooled to rt, diluted with H 2 O (20 mL) and filtered. The obtained solids were dried under vacuum to obtain 6-[4-[5- [(7-bromo-4-oxo-3H-phthalazin-1-yl)-dideuterio-methyl]-2-flu oro-benzoyl]piperazin-1-yl]pyridine-3- carbonitrile as a white solid (88 mg, 88% yield). This was used without further purification. LCMS ESI m/z: 549 [M+H] + . Step 2

6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1-yl)methyl- d2)benzoyl)piperazin-1-yl)nicotinonitrile [0774] A mixture of 6-[4-[5-[(7-bromo-4-oxo-3H-phthalazin-1-yl)-dideuterio-methy l]-2-fluoro- benzoyl]piperazin-1-yl]pyridine-3-carbonitrile (88 mg, 0.16 mmol), tributyl(prop-1-ynyl)stannane (160 mg, 0.48 mmol) and Pd(dppf)Cl 2 (15 mg, 0.02 mmol) in 1,4-dioxane (5 mL) was stirred at 100 °C under Ar (g) for 6 hours. The reaction mixture was concentrated in vacuo and the residue was purified by prep- HPLC to afford 6-(4-(2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1-yl)methyl- d2)benzoyl)piperazin-1-yl)nicotinonitrile (31 mg, 37% yield) as a white solid. LCMS ESI m/z 509 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.64 (s, 1H), 8.51 (d, J = 2.0 Hz, 1H), 8.19 (d, J = 8.2 Hz, 1H), 7.94 (s, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.78 (dd, J = 8.2, 1.4 Hz, 1H), 7.47 – 7.37 (m, 2H), 7.26 (t, J = 9.1 Hz, 1H), 6.93 (d, J = 9.1 Hz, 1H), 3.75 (dd, J = 13.7, 3.6 Hz, 4H), 3.62 (t, J = 4.7 Hz, 2H), 3.35 – 3.25 (m, 2H), 2.08 (s, 3H). Synthesis of Example 74: 6-(4-(2,3-Difluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophth alazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

Methyl 5-bromo-2,3-difluorobenzoate [0775] To a solution of 5-bromo-2,3-difluoro-benzoic acid (1.1 g, 4.64 mmol) in MeOH (25 mL) was added conc. H 2 SO 4 (4.05 g, 41.27 mmol, 2.20 mL) at rt, then stirred at 70 °C for 2 hours. The solution was concentrated in vacuo. This residue was diluted with water (30 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 4%) to afford methyl 5-bromo-2,3-difluoro-benzoate as a colorless oil (1.08 g, 93% yield). LCMS ESI m/z: 251 [M+H] + . Step 2 Methyl 2,3-difluoro-5-vinylbenzoate [0776] A mixture of methyl 5-bromo-2,3-difluoro-benzoate (0.98 g, 3.90 mmol), potassium vinyltrifluoroborate (1.05 g, 7.81 mmol), K 2 CO 3 (2.16 g, 15.62 mmol) and Pd(dppf)Cl 2 (286 mg, 0.39 mmol) in MeCN (16 mL) and H 2 O (4 mL) was stirred at 80 °C under Ar (g) for 7 hours. The mixture was cooled to rt, diluted with H2O (5 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 7%) to give methyl 2,3-difluoro-5- vinyl-benzoate as a colorless oil (0.7 g, 90% yield). LCMS ESI m/z: 199 [M+H] + . 1 HNMR (400 MHz, CDCl 3 ) δ 7.69 – 7.61 (m, 1H), 7.39 – 7.36 (m, 1H), 6.63 – 6.59 (m, 1H), 5.73 (d, J = 16 Hz, 1H), 5.35 (d, J = 12 Hz, 1H). Step 3 Methyl 2,3-difluoro-5-formylbenzoate [0777] To a mixture of methyl 2,3-difluoro-5-vinyl-benzoate (678 mg, 3.42 mmol) in THF (10 mL) and H 2 O (10 mL) was added K 2 OsO 4 -2H 2 O (13 mg, 0.34 mmol). The mixture was stirred at rt for 5 min, then NaIO 4 (2.20 g, 10.26 mmol) was added and the mixture was stirred at rt for 2 hours. The solids were removed by filtration. The filtrate was diluted with H 2 O (5 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 2,3-difluoro-5-formyl-benzoate as a colorless oil (0.68 g, 99% yield). This was used without further purification. LCMS ESI m/z: 201 [M+H] + . Step 4 [0778] Methyl 5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2,3-dif luorobenzoate A solution of 5-bromo-3-dimethoxyphosphoryl-3H-isobenzofuran-1-one (176 mg, 0.55 mmol), methyl 2,3- difluoro-5-formyl-benzoate (100 mg, 0.50 mmol) and Et 3 N (152 mg, 1.50 mmol) in anhydrous THF (5 mL) was stirred at 25 °C under Ar (g) for 15 hours. Hydrazine hydrate (50 mg, 1 mmol) was added and the reaction was stirred at 70 ℃ for 1 hour. The reaction was cooled to rt, then THF was removed under vacuum. The solids were washed with water (10 mL) and petroleum ether/EtOAc (2:1, 10 mL) to give methyl 5-[(7-bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2,3-difluoro-be nzoate as a white solid (130 mg, 64% yield). LCMS ESI m/z: 408.9 [M+H] + . Step 5 Methyl 2,3-difluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazin -1-yl)methyl)benzoate [0779] A mixture of tributyl(prop-1-ynyl)stannane (230 mg, 0.70 mmol), methyl 5-[(7-bromo-4-oxo- 3H-phthalazin-1-yl)methyl]-2,3-difluoro-benzoate (130 mg, 0.32 mmol) and Pd(dppf)Cl 2 (16 mg, 0.02 mmol) in 1,4-dioxane (5 mL) was stirred at 100 °C under Ar (g) for 5 hours. The reaction mixture was concentrated in vacuo and the residue was purified by silica gel chromatography (MeOH/DCM = 0 to 30%) to afford methyl 2,3-difluoro-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)meth yl]benzoate as a white solid (52 mg, 44% yield). LCMS ESI m/z: 369 [M+H] + . Step 6 2,3-Difluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthala zin-1-yl)methyl)benzoic acid [0780] A mixture of methyl 2,3-difluoro-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)meth yl]benzoate (52 mg, 0.14 mmol) and NaOH (17 mg, 0.42 mmol) in MeOH (2 mL), THF (2 mL) and H 2 O (2 mL) was stirred at rt for 17 hours. The organic solvent was removed in vacuo and aq HCl (1N) was added until the solution reached pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 2,3-difluoro-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)meth yl]benzoic acid as a white solid (34 mg, 68% yield). LCMS ESI m/z: 355 [M+H] + . Step 7

6-(4-(2,3-Difluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophth alazin-1-yl)methyl)benzoyl)piperazin- 1-yl)nicotinonitrile [0781] To a solution of 6-piperazin-1-ylpyridine-3-carbonitrile (36 mg, 0.16 mmol) and 2,3-difluoro-5- [(4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)methyl]benzoic acid (32 mg, 0.09 mmol) in DMF (5 mL) was added EDCI (26 mg, 0.13 mmol), HOBt (18 mg, 0.13 mmol) and DIPEA (58 mg, 0.45 mmol). The reaction mixture was stirred at rt for 2 hours. The reaction was purified by prep-HPLC to give 6-[4-[2,3- difluoro-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)methyl]b enzoyl]piperazin-1-yl]pyridine-3- carbonitrile as a white solid (31 mg, 65% yield). LCMS ESI m/z: 525.2 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.19 (s, 1H), 8.42 (d, J = 1.9 Hz, 1H), 8.37 (d, J = 8.2 Hz, 1H), 7.73 (dd, J = 8.2, 1.3 Hz, 1H), 7.69 – 7.64 (m, 2H), 7.22 – 7.14 (m, 1H), 7.12 (d, J = 4.8 Hz, 1H), 6.62 (d, J = 8.9 Hz, 1H), 4.23 (s, 2H), 3.89 (s, 2H), 3.78 (s, 2H), 3.70 (t, J = 5.1 Hz, 2H), 3.44 (s, 2H), 2.09 (s, 3H). Synthesis of Example 75: 6-(Ethyl(2-(1-oxo-6-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophtha lazin-1- yl)methyl)isoindolin-2-yl)ethyl)amino)nicotinonitrile

4-Methylisophthalic acid [0782] To a solution of 3-bromo-4-methyl-benzoic acid (10.0 g, 46.5 mmol) in THF (50 mL) was slowly added methylmagnesium bromide (1M in THF, 51 mL, 51 mmol) at 0°C under Ar (g). The mixture was stirred at 0°C for 2 hours, then cooled to -78 °C and added n-BuLi (2.5 M in Hexane, 36 mL, 90 mmol) dropwise. The reaction was warmed to -40 °C under CO 2 (g) for 30 min, then stirred at rt for 12 hours. The reaction mixture was quenched with sat. aq NH 4 Cl (10 mL) and extracted with EtOAc (50 mL × 3). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo to give 4- methylbenzene-1,3-dicarboxylic acid as a white solid (8.2 g, 43% yield). This was used without further purification. LCMS ESI m/z: 181 [M+H] + . Step 2 Dimethyl 4-methylisophthalate [0783] To a solution of 4-methylbenzene-1,3-dicarboxylic acid (7.2 g, 14.39 mmol) in MeOH (150 mL) was added conc. H 2 SO 4 (5 mL) in portions. The reaction mixture was stirred at 70°C for 8 hours, then cooled to rt, diluted with H2O (50 mL) and extracted with EtOAc (50 mL × 3). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 10 to 20%) to afford ethyl dimethyl 4-methylisophthalate as a yellow oil (7.1 g, 85% yield). LCMS ESI m/z: 209 [M+H] + . Step 3 Dimethyl 4-(bromomethyl)isophthalate [0784] A mixture of ethyl dimethyl 4-methylisophthalate (2.6 g, 12.49 mmol), NBS (2.44 g, 13.74 mmol) and AIBN (410 mg, 2.50 mmol) in CCl 4 (20 mL) was stirred at 70 °C for 15 hours. The mixture was cooled to rt, diluted with H 2 O (30 mL) and extracted with DCM (30 mL × 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 10%) to obtain dimethyl 4- (bromomethyl)isophthalate as a white solid (2.11 g, 59% yield). LCMS ESI m/z: 287 (M+H) + . Step 4 Methyl 2-(2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)-3-oxoisoindol ine-5-carboxylate [0785] A solution of dimethyl 4-(bromomethyl)isophthalate (542 mg, 1.89 mmol), 6-((2- aminoethyl)(ethyl)amino)nicotinonitrile (450 mg, 1.98 mmol) and DIPEA (732 mg, 5.66 mmol) in DMF (6 mL) was stirred at rt under Ar (g) for 12 hours. The mixture was diluted with H 2 O (5 mL) and extracted with EtOAc (15 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 50 to 70%) to give methyl 2-(2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)-3- oxoisoindoline-5-carboxylate as a colorless oil (495 mg, 79% yield). LCMS ESI m/z: 365 [M+H] + . Step 5 6-(Ethyl(2-(6-(hydroxymethyl)-1-oxoisoindolin-2-yl)ethyl)ami no)nicotinonitrile [0786] To a mixture of methyl 2-(2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)-3-oxoisoindol ine-5- carboxylate (445 mg, 1.22 mmol) in THF (10 mL) was added LiBH 4 (2M in THF, 1.22 mL, 2.44 mmol) in portions. The mixture was stirred at rt for 18 hours, then diluted with water (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was dried over Na 2 SO 4 , filtered, and concentrated in vacuum.The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 60 to 80%) to afford 6-(ethyl(2-(6-(hydroxymethyl)-1-oxoisoindolin-2-yl)ethyl)ami no)nicotinonitrile as a yellow oil (360 mg, 88% yield). LCMS ESI m/z: 337 [M+H] + . Step 6 6-(Ethyl(2-(6-formyl-1-oxoisoindolin-2-yl)ethyl)amino)nicoti nonitrile [0787] A mixture of 6-(ethyl(2-(6-(hydroxymethyl)-1-oxoisoindolin-2-yl)ethyl)ami no)nicotinonitrile (360 mg, 1.07 mmol) and MnO 2 (464 mg, 5.34 mmol) in DCM (15 mL) was stirred at 50°C for 12 hours. The reaction was filtered and the filtrate was concentrated in vacuo to obtain 6-(ethyl(2-(6-formyl-1- oxoisoindolin-2-yl)ethyl)amino)nicotinonitrile as a white solid (278 mg, 77% yield). This was used without further purification. LCMS ESI m/z: 335 [M+H] + . Step 7 6-((2-(6-((7-Bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)- 1-oxoisoindolin-2- yl)ethyl)(ethyl)amino)nicotinonitrile [0788] A solution of 6-(ethyl(2-(6-formyl-1-oxoisoindolin-2-yl)ethyl)amino)nicoti nonitrile (250 mg, 0.75 mmol), 5-bromo-3-dimethoxyphosphoryl-3H-isobenzofuran-1-one (240 mg, 0.75 mmol) and Et 3 N (228 mg, 2.25 mmol) in anhydrous THF (10 mL) was stirred at 25 °C under Ar (g) for 15 hours. To the reaction mixture, hydrazine hydrate (265 mg, 4.5 mmol) was added and the reaction was stirred at 70 ℃ for 1 hour. The reaction was cooled to rt, then THF was removed under vacuum. The solids were washed with H 2 O (10 mL) and petroleum ether/EA (2:1, 10 mL) to give 6-((2-(6-((7-bromo-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-1-oxoisoindolin-2-yl)ethyl)(e thyl)amino)nicotinonitrile as a white solid (290 mg, 84% yield). This was used without further purification. LCMS ESI m/z: 543 [M+H] + . Step 8 6-(Ethyl(2-(1-oxo-6-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophtha lazin-1-yl)methyl)isoindolin-2- yl)ethyl)amino)nicotinonitrile [0789] A mixture of tributyl(prop-1-ynyl)stannane (230. mg, 0.70 mmol), 6-((2-(6-((7-bromo-4-oxo- 3,4-dihydrophthalazin-1-yl)methyl)-1-oxoisoindolin-2-yl)ethy l)(ethyl)amino)nicotinonitrile (100 mg, 0.18 mmol) and Pd(dppf)Cl 2 (16 mg, 0.02 mmol) in 1,4-dioxane (5 mL) was stirred at 100 °C under Ar (g) for 5 hours. The reaction mixture was concentrated in vacuo and the residue was purified by prep-HPLC to afford methyl 2,3-difluoro-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)meth yl]benzoate as a white solid (26 mg, 29% yield). LCMS ESI m/z: 503 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 9.92 (s, 1H), 8.35 (d, J = 8.8 Hz, 1H), 8.28 (d, J = 2.1 Hz, 1H), 7.74 – 7.67 (m, 3H), 7.53 (dd, J = 9.0, 2.3 Hz, 1H), 7.46 (d, J = 7.8 Hz, 1H), 7.38 (d, J = 7.8 Hz, 1H), 6.53 (d, J = 9.1 Hz, 1H), 4.44 (s, 2H), 4.33 (s, 2H), 3.90 – 3.79 (m, 4H), 3.52 (q, J = 7.1 Hz, 2H), 2.09 (s, 3H), 1.19 (t, J = 7.1 Hz, 3H). Synthesis of Example 76: 2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1- yl)methyl)benzonitrile

[0790] 3,5-Dibromoisobenzofuran-1(3H)-one To a solution of 5-bromo-3H-isobenzofuran-1- one (10.0 g, 46.9 mmol) in CCl 4 (100 mL) was added AIBN (771 mg, 4.7 mmol) and 1- bromopyrrolidine-2,5-dione (10.0 g, 56.3 mmol). The resulting mixture was stirred at 80 °C for 16 hours. Water (90 mL) and EtOAc (120 mL) were added to the mixture, which became two phases. The organic layer was washed with brine, dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 10/1) to give 3,5-dibromo-3H- isobenzofuran-1-one (9 g, 66% yield) as a white solid. 1 H-NMR (400 MHz, CDCl 3 ) δ 7.80 – 7.78 (m, 1H), 7.78 (s, 1H), 7.76 (d, J = 1.3 Hz, 1H), 7.34 (s, 1H). Step 2 5-Bromo-3-hydroxyisobenzofuran-1(3H)-one [0791] To a mixture of 3,5-dibromo-3H-isobenzofuran-1-one (8.00 g, 27.4 mmol) in water (50 mL) was added KOH (3.08 g, 54.8 mmol). The resulting mixture was stirred at 100 °C for 2 hours. The mixture was cooled to rt, acidified with 1 M HCl until the solution reached pH 5~6. The mixture was filtered, the filter cake was washed with H 2 O (20 mL x 2) and dried under reduced pressure to give 5- bromo-3-hydroxy-3H-isobenzofuran-1-one (5.4 g, 86% yield) as a white solid. 1 H-NMR (400 MHz, DMSO-d 6 ) δ 8.28 (d, J = 8.2 Hz, 1H), 7.93 (s, 1H), 7.86 (dd, J = 8.1, 1.4 Hz, 1H), 7.77 (d, J = 8.1 Hz, 1H), 6.65 (d, J = 8.1 Hz, 1H). Step 3 Dimethyl 6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphonate [0792] 5-bromo-3-hydroxy-3H-isobenzofuran-1-one (5.00 g, 21.8 mmol) was added into dimethylphosphite (15 mL) and the resulting mixture was stirred at 100 °C for 3 hours. The mixture was quenched with H 2 O (50 mL) and extracted with EtOAc (60 mL x 2). The combined organic phase was washed with H 2 O (40 mL x 4), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give the crude product, which was purified by silica gel chromatography (petroleum ether/EtOAc = 30/1 to 1/1) to afford dimethyl 6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphonate (4.00 g, 57% yield) as a white solid. 1 H-NMR (400 MHz, DMSO-d 6 ) δ 7.89 (d, J = 0.8 Hz, 2H), 7.86 – 7.83 (m, 1H), 6.35 (dd, J = 11.6, 0.7 Hz, 1H), 3.82 (d, J = 10.9 Hz, 3H), 3.65 (d, J = 10.7 Hz, 3H). Step 4 5-[(Z)-(6-Bromo-3-oxo-isobenzofuran-1-ylidene)methyl]-2-fluo ro-benzonitrile [0793] To a mixture of 5-bromo-3-dimethoxyphosphoryl-3H-isobenzofuran-1-one (3.00 g, 9.34 mmol) in THF (40 mL) was added 2-fluoro-5-formyl-benzonitrile (2.79 g, 18.69 mmol) and N,N- diethylethanamine (2.84 g, 28.03 mmol, 3.91 mL). The resulting mixture was stirred at 25 °C for 12 hours. The mixture was quenched with sat. NaHSO 3 solution (30 mL) and filtered. The filter cake was washed with H 2 O (20 mL x 2) and dried under reduced pressure to give 5-[(Z)-(6-bromo-3-oxo- isobenzofuran-1-ylidene)methyl]-2-fluoro-benzonitrile (3.00 g, 93% yield) as a white solid. 1 H-NMR (400 MHz, DMSO-d 6 ) δ 8.42 (d, J = 1.0 Hz, 1H), 8.20 – 8.07 (m, 2H), 7.94 – 7.85 (m, 2H), 7.67 (t, J = 9.1 Hz, 1H), 7.08 (s, 1H). Step 5 5-[(7-Bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro-benzon itrile [0794] To a mixture of 5-[(Z)-(6-bromo-3-oxo-isobenzofuran-1-ylidene)methyl]-2-fluo ro-benzonitrile (3.00 g, 8.72 mmol) in THF (30 mL) was added hydrazine;hydrate (818 mg, 13.1 mmol, 797 μL). The resulting mixture was stirred at 70 °C for 4 hours. The mixture was cooled to rt. and filtered. The filter cake was washed with a solution of petrol ether/EtOAc =10/1 (20 mL x 2) and dried under reduced pressure to give 5-[(7-bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro-benzon itrile (3.00 g, 77 % yield) as a yellow solid. 1 H-NMR (400 MHz, DMSO-d 6 ) δ 8.22 (d, J = 1.7 Hz, 1H), 8.17 (d, J = 8.5 Hz, 1H), 8.02 (dd, J = 8.5, 1.8 Hz, 1H), 7.90 (dd, J = 6.2, 2.2 Hz, 1H), 7.76 – 7.68 (m, 1H), 7.48 (t, J = 9.1 Hz, 1H), 4.38 (s, 2H). Step 6 2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1-yl)methyl)benzonitrile [0795] 5-[(7-bromo-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro-benzon itrile (1.50 g, 4.19 mmol), tributyl(prop-1-ynyl)stannane (2.76 g, 8.38 mmol), Pd(PPh 3 ) 4 (484 mg, 419 μmol) and toluene (20 mL) were added to a 100 mL bottled flask. The mixture was purged with N 2 (g) for 3 times. The resultant mixture was stirred at 100 °C for 12 hours. The mixture was filtered and concentrated under reduced pressure to give the crude product, which was purified by prep-HPLC to afford 2-fluoro-5-((4-oxo-7- (prop-1-yn-1-yl)-3,4-dihydrophthalazin-1-yl)methyl)benzonitr ile (6.7 mg, 8% yield) as a white solid. LCMS ESI m/z: 317.9 [M + H] + . 1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.59 (s, 1H), 8.20 (d, J = 8.2Hz, 1H), 7.97 (s, 1H), 7.88 (dd, J = 6.3, 2.2 Hz, 1H), 7.79 (dd, J = 8.2, 1.4 Hz, 1H), 7.75 – 7.69 (m, 1H), 7.47 (t, J = 9.1 Hz, 1H), 4.37 (s, 2H), 2.12 (s, 3H). Synthesis of Example 77: 6-(4-(2,4-Difluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrop hthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

Methyl 5-bromo-2,4-difluorobenzoate [0796] To a solution of 5-bromo-2,4-difluoro-benzoic acid (2.00 g, 8.44 mmol) in MeOH (30 mL) was added conc. H 2 SO 4 (2 mL) in portions. The mixture was heated to 70 °C for 2 hours. The solution was concentrated in vacuo. The residue was diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 4%) to afford methyl 5-bromo-2,4-difluoro-benzoate as a colorless liquid (1.87 g, 86% yield). LCMS ESI m/z: 251 [M+H] + . Step 2 Methyl 2,4-difluoro-5-vinylbenzoate [0797] A mixture of methyl 5-bromo-2,4-difluoro-benzoate (1.87 g, 7.45 mmol), potassium vinyltrifluoroborate (1.99 g, 14.9 mmol), K 2 CO 3 (4.12 g, 29.8 mmol) and Pd(dppf)Cl 2 (545 mg, 0.75 mmol) in MeCN (30 mL) and H 2 O (8 mL) was stirred at 80 °C under Ar (g) for 2 hours. The mixture was cooled to rt, diluted with H 2 O (5 mL) and extracted with EtOAc (10 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromategraphy (EtOAc/petroleum ether = 0 to 7%) to give methyl 2,4-difluoro-5- vinyl-benzoate as a colorless oil (1.26 g, 85% yield). LCMS ESI m/z: 199.1 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 8.10 (t, J = 8.0 Hz, 1H), 6.88 – 6.73 (m, 2H), 5.84 (d, J = 16 Hz, 1H), 5.42 (d, J = 12 Hz, 1H), 3.93 (s, 3H). Step 3 Methyl 2,4-difluoro-5-formylbenzoate [0798] To a mixture of methyl 2,4-difluoro-5-vinyl-benzoate (1.26 g, 6.36 mmol) in THF (20 mL) and H 2 O (15 mL) was added K 2 OsO 4 -2H 2 O (23 mg, 0.06 mmol). The mixture was stirred at room temperature for 5 min, then NaIO 4 (3.98 g, 18.62 mmol) was added and the mixture was stirred at rt for 2 hours. The solids were removed by filtration. The filtrate was diluted with H 2 O (5 mL) and extracted with EtOAc (30 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 2,4-difluoro-5-formyl-benzoate (1.14 g, 92% yield). This was used without further purification. LCMS ESI m/z: 201[M+H] + . Step 4 Methyl 5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2,4-dif luorobenzoate [0799] A solution of 5-bromo-3-dimethoxyphosphoryl-3H-isobenzofuran-1-one (642 mg, 2.00 mmol), methyl 2,4-difluoro-5-formyl-benzoate (400 mg, 2.00 mmol) and Et 3 N (607 mg, 6.00 mmol) in anhydrous THF (16 mL) was stirred at 25 °C under Ar (g) for 3 hours. Hydrazine hydrate (220 mg, 3.73 mmol) was added and the reaction was stirred at 70 °C for 2 hour. The reaction was cooled to rt, then solvent was removed under vacuum. The solids were washed with water (10 mL) and petroleum ether/EtOAc (10:1, 10 mL) to give methyl 5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2,4-dif luorobenzoate as a yellow solid (664 mg, 82% yield). LCMS ESI m/z: 408.9 [M+H] + . Step 5 Methyl 2,4-Difluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazin -1-yl)methyl)benzoate [0800] A mixture of tributyl(prop-1-ynyl)stannane (354 mg, 1.08 mmol), methyl 5-[(7-bromo-4-oxo- 3H-phthalazin-1-yl)methyl]-2,3-difluoro-benzoate (200 mg, 0.49 mmol) and Pd(dppf)Cl 2 (25 mg, 0.04 mmol) in 1,4-dioxane (5 mL) were stirred at 100 °C under Ar (g) for 3 hours. The reaction mixture was concentrated in vacuo and the residue was purified by silica gel chromatography (MeOH/DCM 0-30%) to afford methyl 2,4-difluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazin -1-yl)methyl)benzoate as a yellow solid (97 mg, 54% yield). LCMS ESI m/z: 369 [M+H] + . Step 6 2,4-Difluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthala zin-1-yl)methyl)benzoic acid [0801] A mixture of methyl 2,4-difluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazin -1- yl)methyl)benzoate (97 mg, 0.26 mmol) and NaOH (32 mg, 0.79 mmol) in MeOH (2 mL), THF (2 mL) and H 2 O (2 mL) was stirred at rt for 1 hour. The organic solvent was removed in vacuum and aq HCl (1N) was added until the solution reached to pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 2,4-difluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthala zin-1- yl)methyl)benzoic acid as a white solid (60 mg, 67% yield). LCMS ESI m/z: 355 [M+H] + . Step 7 6-(4-(2,4-Difluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophth alazin-1-yl)methyl)benzoyl)piperazin- 1-yl)nicotinonitrile [0802] To a solution of 6-piperazin-1-ylpyridine-3-carbonitrile (69 mg, 0.30 mmol) and 2,4-difluoro-5- ((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin-1-yl)methyl )benzoic acid (60 mg, 0.17 mmol) in DMF (4 mL) was added EDCI (49 mg, 0.25 mmol), HOBT (34 mg, 0.25 mmol) and DIPEA (109 mg, 0.85 mmol). The reaction mixture was stirred at rt for 2 hours. The reaction was purified by prep-HPLC to give 6-(4-(2,4-difluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophth alazin-1-yl)methyl)benzoyl)piperazin-1- yl)nicotinonitrile as a white solid (35 mg, 42% yield). LCMS ESI m/z: 525 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.17 (s, 1H), 8.41 (d, J = 2.0 Hz, 1H), 8.35 (d, J = 8.2 Hz, 1H), 7.77 (s, 1H), 7.74 (d, J = 8.3 Hz, 1H), 7.65 (dd, J = 9.0, 2.2 Hz, 1H), 7.31 (t, J = 7.7 Hz, 1H), 6.94 (t, J = 9.3 Hz, 1H), 6.61 (d, J = 9.0 Hz, 1H), 4.24 (s, 2H), 3.86 (s, 2H), 3.72 (d, J = 28.1 Hz, 4H), 3.43 (s, 2H), 2.12 (s, 3H). Synthesis of Example 78: 6-(Ethyl(2-((2,2,2-trifluoro-1-(2-fluoro-5-((4-oxo-7-(prop-1 -yn-1-yl)-3,4- dihydrophthalazin-1-yl)methyl)phenyl)ethyl)amino)ethyl)amino )nicotinonitrile 2-(3-Bromo-4-fluorophenyl)-1,3-dioxolane [0803] To a mixture of 3-bromo-4-fluorobenzaldehyde (5.00 g, 24.6 mmol), ethylene glycol (22.9 g, 369 mmol) and TsOH (2.12 g, 12.3 mmol) in toluene (60 mL) was stirred under reflux for 2 hour. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , and filtered and concentrated in vacuo. The residue was purified by prep-HPLC to give 2-(3-bromo-4-fluorophenyl)-1,3-dioxolane (5.44 g, 89% yield) as a white solid. LCMS ESI m/z: 247/249 [M+H] + . Step 2 1-(5-(1,3-Dioxolan-2-yl)-2-fluorophenyl)-2,2,2-trifluoroetha n-1-one [0804] A solution of 2-(3-bromo-4-fluorophenyl)-1,3-dioxolane (2.00 g, 8.10 mmol) in THF (10 mL) was cooled to -78°C, then n-BuLi (2.5M in heptane, 3.88 mL, 9.71 mmol) was added dropwise under Ar (g). The reaction was stirred at -78°C for 15 min, then ethyl 2,2,2-trifluoroacetate (1.50 g, 10.52 mmol) was added dropwise. The reaction was kept at -78°C for 20 min, then quenched with sat. NH 4 Cl soltion (10 mL) and neutralized with aq HCl (1 mol/L) until the solution reached pH 5~6. The mixture was extracted with EtOAc (50 x 3 mL). The combined organic layer was washed with bine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 20% to 30%) to give 1-(5-(1,3-dioxolan-2-yl)-2-fluorophenyl)-2,2,2- trifluoroethan-1-one (950 mg, 44 % yield) as a white solid. LCMS ESI m/z: 265 [M+H] + . Step 3 6-((2-((1-(5-(1,3-Dioxolan-2-yl)-2-fluorophenyl)-2,2,2-trifl uoroethyl)amino)ethyl) (ethyl)amino)nicotinonitrile [0805] A solution of dimethyl 1-(5-(1,3-dioxolan-2-yl)-2-fluorophenyl)-2,2,2-trifluoroetha n-1-one (450 mg, 1.70 mmol), 6-((2-aminoethyl)(ethyl) amino)nicotinonitrile (463 mg, 2.04 mmol), TiCl 4 (323 mg, 1.70 mmol) and DIPEA (880 mg, 6.81 mmol) in anhydrous DCM (5 mL) was stirred at 25 °C under Ar (g) for 1 hours. NaBH 4 (322 mg, 8.52 mmol) was added and the reaction was stirred at 25 °C for 2 hour. The mixture was diluted with H 2 O (10 mL) and extracted with DCM (20 mL × 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 10% to 20%) to give 6-((2-((1-(5-(1,3- dioxolan-2-yl)-2-fluorophenyl)-2,2,2-trifluoroethyl)amino)et hyl) (ethyl) amino)nicotinonitrile (280 mg, 37% yield) as a yellow oil. LCMS ESI m/z: 439 [M+H] + . Step 4 tert-Butyl (1-(5-(1,3-dioxolan-2-yl)-2-fluorophenyl)-2,2,2-trifluoroeth yl)(2-((5-cyanopyridin-2- yl)(ethyl)amino)ethyl)carbamate [0806] A mixture of 6-((2-((1-(5-(1,3-dioxolan-2-yl)-2-fluorophenyl)-2,2,2-trifl uoroethyl)amino) ethyl)(ethyl)amino)nicotinonitrile (295 mg, 0.67 mmol), Boc 2 O (146 mg, 0.67 mmol), DIPEA (87 mg, 0.67 mmol) and DMAP (82 mg, 0.67 mmol) in DCM (6 mL) was stirred at 25 °C under Ar (g) for 12 hours. The mixture was diluted with H 2 O (5 mL) and extracted with DCM (10 mL × 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromategraphy (EtOAc/petroleum ether = 0% to 7%) to give tert-butyl (1-(5-(1,3- dioxolan-2-yl)-2-fluorophenyl)-2,2,2-trifluoroethyl)(2-((5-c yanopyridin-2- yl)(ethyl)amino)ethyl)carbamate (240 mg, 66 % yield) as a colorless oil. LCMS ESI m/z: 539 [M+H] + . Step 5 tert-Butyl (2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)(2,2,2-trifluoro -1-(2-fluoro-5- formylphenyl)ethyl)carbamate [0807] A solution of tert-butyl (1-(5-(1,3-dioxolan-2-yl)-2-fluorophenyl)-2,2,2-trifluoroeth yl)(2-((5- cyanopyridin-2-yl)(ethyl)amino)ethyl)carbamate (240 mg, 0.45 mmol) and PTSA (2 mg, 0.01 mmol) in actone (5 mL) and H 2 O (0.5 mL) was stirred at rt for 1 hour. The organic solvent was removed in vacuum and aq HCl (1N) was added until the solution reached to pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give tert-butyl (2-((5-cyanopyridin-2- yl)(ethyl)amino)ethyl)(2,2,2-trifluoro-1-(2-fluoro-5-formylp henyl)ethyl)carbamate (200 mg, 90% yield) as a white solid. LCMS ESI m/z: 495 [M+H] + . This was used without further purification. Step 6 tert-Butyl (1-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-f luorophenyl)-2,2,2- trifluoroethyl)(2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl) carbamate [0808] A solution of dimethyl (6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (130 mg, 0.41 mmol), tert-butyl (2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)(2,2,2-trifluoro -1-(2-fluoro-5- formylphenyl)ethyl)carbamate (200 mg, 0.41 mmol) and Et 3 N (123 mg, 1.21 mmol) in anhydrous THF (10 mL) was stirred at 50 °C under Ar (g) for 12 hours. Hydrazine hydrate (24 mg, 0.48 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/ EtOAc (10:1, 10 mL) to give tert-butyl (1-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-f luorophenyl)-2,2,2- trifluoroethyl)(2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)c arbamate (200 mg, 89% yield) as a yellow solid. LCMS ESI m/z: 703 [M+H] + . Step 7 tert-Butyl (2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)(2,2,2-trifluoro -1-(2-fluoro-5-((4-oxo-7-(prop- 1-yn-1-yl)-3,4-dihydrophthalazin-1-yl)methyl)phenyl)ethyl) carbamate [0809] A mixture of tert-butyl (1-(5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2- fluorophenyl)-2,2,2-trifluoroethyl)(2-((5-cyanopyridin-2-yl) (ethyl)amino)ethyl)carbamate (200 mg, 0.28 mmol), tributyl(prop-1-yn-1-yl)stannane (281 mg, 0.85 mmol), Pd(dppf)Cl 2 (21 mg, 0.03 mmol) in dioxane (20 mL) was stirred at 100 °C under Ar (g) for 16 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL x 3) The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30% to 60%) to give tert-butyl (2-((5-cyanopyridin-2- yl)(ethyl)amino)ethyl)(2,2,2-trifluoro-1-(2-fluoro-5-((4-oxo -7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin-1- yl)methyl)phenyl)ethyl)carbamate (160 mg, 84% yield) as a white solid. LCMS ESI m/z: 663 [M+H] + . Step 8 6-(Ethyl(2-((2,2,2-trifluoro-1-(2-fluoro-5-((4-oxo-7-(prop-1 -yn-1-yl)-3,4-dihydrophthalazin-1- yl)methyl)phenyl)ethyl)amino)ethyl)amino)nicotinonitrile [0810] To a solution of tert-butyl (2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)(2,2,2-trifluoro -1-(2- fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin-1- yl)methyl)phenyl)ethyl)carbamate (160 mg, 0.24 mmol) in DCM (5 mL) was added TFA (5 mL). The reaction mixture was stirred at rt for 1 hour. The reaction was concentrated in vacuo and the residue was purified by prep-HPLC to give the 6-(ethyl(2- ((2,2,2-trifluoro-1-(2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3 ,4-dihydrophthalazin-1- yl)methyl)phenyl)ethyl)amino)ethyl)amino)nicotinonitrile (90 mg, 66% yield) as a white solid. LCMS ESI m/z: 563 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.63 (s, 1H), 8.36 (s, 1H), 8.17 (d, J= 8.4Hz, 1H), 7.81 (s, 1H), 7.72–7.69 (m, 2H), 7.58–7.56 (m, 1H), 7.35–7.32 (m, 1H), 7.19 (t, J = 8.8Hz, 1H), 6.60 (d, J = 9.2Hz, 1H), 4.72–4.70 (m, 1H), 4.30–4.28 (m, 2H), 3.46–3.41 (m, 5H), 2.61 (t, J = 6.8Hz, 2H), 2.06 (s, 3H),1.00 (t, J = 6.8Hz, 3H). Synthesis of Example 79: 4-(3-(4-(1,5-Dimethyl-1H-imidazol-2-yl)piperazine-1-carbonyl )-4- fluorobenzyl)-6-(prop-1-ynyl)phthalazin-1(2H)-one

tert-Butyl 4-(1,5-dimethyl-1H-imidazol-2-yl)piperazine-1-carboxylate [0811] To a solution of 2-bromo-1,5-dimethyl-imidazole (100 mg, 571.34 μmol) in NMP (0.5 mL) was added tert-butyl piperazine-1-carboxylate (160 mg, 857 μmol), DIPEA (369 mg, 2.86 mmol, 498 μL). The reaction mixture stirred at 150 °C under microwave for 10 hr. Di-tert-butyl dicarbonate (312 mg, 1.43 mmol, 328 μL) was added to the reaction mixture and stirred at 25 °C for 2 hr. The reaction mixture was diluted in ethyl acetate (10 mL), washed with water (10 mL), brine, dried and concentrated to the residue. The residue was purified by prepare TLC(PE/EA=0/1) to give tert-butyl 4-(1,5-dimethylimidazol-2- yl)piperazine-1-carboxylate (54 mg, 34% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 6.54 (d, J = 0.7 Hz, 1H), 3.55 (dd, J = 6.2, 3.9 Hz, 4H), 3.37 (s, 3H), 3.07 – 2.94 (m, 4H), 2.14 (d, J = 1.0 Hz, 3H), 1.47 (s, 9H). Step 2 1-(1,5-Dimethyl-1H-imidazol-2-yl)piperazine [0812] To a solution of tert-butyl 4-(1,5-dimethylimidazol-2-yl)piperazine-1-carboxylate (54 mg, 193 μmol) in 4M EA/HCl (3 mL). The reaction mixture was stirred at rt. for 2 hours. The mixture was concentrated to give 1-(1,5-dimethylimidazol-2-yl)piperazine (40 mg, 96% yield, HCl) as a white solid. LCMS ESI m/z: 181.1 [M + H] + . The crude material was used as is in the next step. Step 3

4-(3-(4-(1,5-Dimethyl-1H-imidazol-2-yl)piperazine-1-carbonyl )-4-fluorobenzyl)-6-(prop-1- ynyl)phthalazin-1(2H)-one [0813] Following the general amide coupling procedure above in Example 55, but starting with 1-(1,5- dimethyl-1H-imidazol-2-yl)piperazine gave the title compound as a white solid. LCMS ESI m/z: 498.8 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ10.36 (s, 1H), 8.35 (d, J = 8.2 Hz, 1H), 7.75 (s, 1H), 7.70 (d, J = 8.2 Hz, 1H), 7.38 (d, J = 6.3 Hz, 1H), 7.32 (s, 1H), 7.07 (t, J = 8.8 Hz, 1H), 6.88 (s, 1H), 4.25 (s, 2H), 3.71 - 3.55 (m, 4H), 3.53 (s, 3H), 3.49 - 3.30 (m, 4H), 2.25 (s, 3H), 2.11 (s, 3H). Synthesis of Example 80: 6-(6,6-Difluoro-4-(2-fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)-1,4-diazepan-1-yl)nic otinonitrile

N,N'-(Ethane-1,2-diyl)bis(4-methylbenzenesulfonamide) [0814] To a solution of ethane-1,2-diamine (10.0 g, 166vmmol, 11.1 mL) taken under argon atmosphere and cooled to 0 °C, then NaOH (13.3 g, 333 mmol, 6.25 mL) in H 2 O (134 mL) was added slowly at 0 °C. The resulting mixture was dropwise added to the cooled solution of 4- methylbenzenesulfonyl chloride (63.45 g, 332.79 mmol) in THF (134 mL) at 0 °C and then stirred at 25 °C for 16 hrs. The solid suspension was filtered off and the resulting solid was filtered through sintered funnel and re-crystallized from MeOH to afford 4-methyl-N-[2-(p- tolylsulfonylamino)ethyl]benzenesulfonamide (31 g, 51% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ 7.61 (d, J = 8.2 Hz, 4H), 7.38 (d, J = 8.0 Hz, 4H), 2.72 (s, 4H), 2.39 (s, 6H). Step 2 1,4-Ditosyl-1,4-diazepan-6-ol [0815] To a stirred solution of 4-methyl-N-[2-(p-tolylsulfonylamino)ethyl]benzenesulfonamide (14.0 g, 38.0 mmol) in ethanol (140 mL), sodium ethanolate (4.15 g, 61.0 mmol) followed by 1,3- dibromopropan-2-ol (8.53 g, 39.1 mmol) were added. Then the reaction mixture was heated to reflux at 80 °C for 16 hours. The solvent was evaporated, diluted with H 2 O and neutralize the reaction mixture with citric acid and extracted with EtOAc. Combined organic layers were dried over Na 2 SO 4 and concentrated. The crude obtained was again suspended in EtOH-H 2 O (2:1) and basified with 2 N NaOH at 0 °C carefully (pH 10). The white suspension was decanted first and then filtered, the solid residue was washed with H2O followed by EtOH and finally dried to get 1,4-bis(p-tolylsulfonyl)-1,4-diazepan-6-ol (13.0 g, 81% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 7.71 (d, J = 8.2 Hz, 4H), 7.47 (d, J = 8.1 Hz, 4H), 5.32 (d, J = 4.7 Hz,1H), 3.85 – 3.73 (m, 1H), 3.52 (d, J = 1.8 Hz, 4H), 3.20 – 3.11 (m, 2H), 2.91 (dd, J = 13.9, 8.1 Hz, 2H), 2.45 (s, 6H). Step 3 1,4-Ditosyl-1,4-diazepan-6-one [0816] To a solution of 1,4-bis(p-tolylsulfonyl)-1,4-diazepan-6-ol (13 g, 30.62 mmol) in DCM (300 mL) was added DMP (26.0 g, 61.2 mmol) portion wise at 0 °C during 30 min. The reaction mixture was slowly allowed to 25 °C and stirring was continued for 15 hrs. The solids were filtered and washed with DCM. The organic layer was concentrated and purified by silica gel chromatography to afford 1,4-bis(p- tolylsulfonyl)-1,4-diazepan-6-one (12.0 g, 74% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 7.70 (d, J = 8.3 Hz, 4H), 7.43 (d, J = 8.0 Hz, 4H), 3.93 (s, 4H), 3.58 (s, 4H), 2.40 (s, 6H). Step 4 6,6-Difluoro-1,4-ditosyl-1,4-diazepane [0817] In a polystyrene reaction vessel containing a solution of 1,4-bis(p-tolylsulfonyl)-1,4-diazepan- 6-one (12.0 g, 28.4 mmol) in DCM (100 mL) was added N-ethyl-N-(trifluoro-sulfanyl)ethanamine (22.9 g, 142 mmol, 18.8 mL) at 0 °C under N 2 atmosphere. The reaction mixture was slowly allowed to warm to 25 °C and stirred for 16 h. The reaction mixture was diluted with DCM (40 ml) and carefully basified with NaHCO 3 adjust pH 7. The organic layer was separated, and aqueous layer was extracted with DCM (30 ml x 2). The combined organic layers were washed with brine (30 ml), dried over Na2SO4 and concentrated to get crude product. The crude was purified by silica gel chromatography to afford 6,6- difluoro-1,4-bis(p-tolylsulfonyl)-1,4-diazepane (8.0 g, 63% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 7.65 (d, J = 8.3 Hz, 4H), 7.33 (d, J = 8.0 Hz, 4H), 3.68 (t, J = 12.3 Hz, 4H), 3.41 (s, 4H), 2.44 (s, 6H). Step 5 6,6-Difluoro-1,4-diazepane [0818] In a polystyrA suspension of 6,6-difluoro-1,4-bis(p-tolylsulfonyl)-1,4-diazepane (1.2 g, 2.70 mmol) and phenol (1.02 g, 10.8 mmol, 949 μL) in HBr (728 mg, 2.70 mmol, 10 mL, 30% purity) was heated to 110 °C in a sealed tube. After 2 hrs, the mixture was cooled to 25 °C, the solvent removed in vacuum and the residue was triturated with MTBE and washed with EtOH and finally dried to give 6,6- difluoro-1,4-diazepane (550 mg, 68% yield) as an off-white solid. 1 H NMR (400 MHz, MeOD) δ 4.07 (t, J = 12.3 Hz, 4H), 3.78 (s, 4H). Step 6 tert-Butyl 6,6-difluoro-1,4-diazepane-1-carboxylate [0819] To a solution of 6,6-difluoro-1,4-diazepane (800 mg, 2.68 mmol, 2HBr) in H 2 O (24 mL) was added NaHCO 3 (677 mg, 8.05 mmol), which was dissolved in THF (45 mL) at 0 °C , and then Boc 2 O ( , 2.82 mmol) was added dropwise, and then stirred at rt. for 24 hrs. The mixture was concentrated and extracted with EtOAc (30 mL x 2), washed with brine, dried, concentrated and purified by silica gel chromatography to give tert-butyl 6,6-difluoro-1,4-diazepane-1-carboxylate (290 mg, 46% yield) as a white solid. 1 H NMR (400 MHz, MeOD) δ 3.84 (t, J = 12.5 Hz, 2H), 3.48 (s, 2H), 3.05 (t, J = 13.4 Hz, 2H), 2.94 – 2.85 (m, 2H), 1.46 (s, 9H). Step 7 6-(6,6-Difluoro-1,4-diazepan-1-yl)nicotinonitrile [0820] A solution of tert-butyl 6,6-difluoro-1,4-diazepane-1-carboxylate (100 mg, 423 μmol), 6- chloropyridine-3-carbonitrile (58.7 mg, 423 μmol) and DIPEA (164 mg, 1.27 mmol) in NMP (1.5 mL) was stirred at 150 °C under microwave for 40 min. The reaction mixture was diluted in EA, washed with water 3 times. The organic layer was concentrated to the crude and purified by Pre-TLC (EtOAc: Petroleum ether = 3: 1) to give 6-(6,6-difluoro-1,4-diazepan-1-yl)pyridine-3-carbonitrile (40 mg, 40% yield) as a yellow solid. LCMS ESI m/z: 239.2 [M + H] + . Step 8 6-(6,6-Difluoro-4-(2-fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-di hydrophthalazin-1-yl)methyl)benzoyl)- 1,4-diazepan-1-yl)nicotinonitrile [0821] Following the general amide coupling procedure in Example 55, but starting with 6-(6,6- difluoro-1,4-diazepan-1-yl)nicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 557.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.61 (d, J = 5.6 Hz, 1H), 8.51 (dd, J = 35.7, 2.1 Hz, 1H), 8.20 (dd, J = 8.2, 3.8 Hz, 1H), 7.92 (dt, J = 21.0, 12.1 Hz, 2H), 7.77 (dd, J = 11.3, 4.2 Hz, 1H), 7.43 – 7.34 (m, 1H), 7.30 – 7.18 (m, 2H), 7.00 (d, J = 9.1 Hz, 1H), 4.33 (m, 4H), 4.15 (s, 1H), 3.93 (m, 3H), 3.76 (s, 1H), 3.50 (t, J = 5.3 Hz, 1H), 2.09 (d, J = 4.0 Hz, 3H). Exampe 81 (SP-PAR7000076-NX-1) 6-(4-(2-Fluoro-5-((8-methoxy-4-oxo-7-(prop-1-yn-1-yl)-3,4-di hydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

5-Bromo-4-fluoroisobenzofuran-1(3H)-one [0822] A mixture of (3-bromo-2-fluorophenyl)methanol (4 g, 19.51 mmol), thallium(III) trifluoroacetate (10.71 g, 19.71 mmol) and TFA (20 mL) was stirred at room temperature for 1 day. The solution was concentrated to dryness under reduced pressure at room temperature and azeotroped with dichloromethane. The reaction mixture was then treated with PdCl 2 (345.96 mg, 1.95 mmol), LiCl (1.65 g, 39.02 mmol, 799.83 μL), MgO (1.61 g, 39.02 mmol) and MeOH (20 mL). The reaction mixture was then degassed and purged with CO several times and then stirred under CO for 1 day. EtOAc (80 mL) was added the reaction mixture to precipitate the salts. The black solution was filtered through a celite pad, washed with EtOAc (40 mL), adsorbed onto silica and purified by silica gel chromatography to afford 5- bromo-4-fluoro-3H-isobenzofuran-1-one (1 g, 22% yield) as a white solid. 1 H NMR (400 MHz, DMSO- d 6 ) δ 7.95 (dd, J = 8.0, 5.9 Hz, 1H), 7.66 (d, J = 8.1 Hz, 1H), 5.55 (s, 2H). Step 2 3,5-Dibromo-4-fluoroisobenzofuran-1(3H)-one [0823] To a solution of 5-bromo-4-fluoro-3H-isobenzofuran-1-one (1 g, 4.33 mmol) in CCl 4 (15 mL) was added 1-bromopyrrolidine-2,5-dione (924.52 mg, 5.19 mmol, 440.67 μL), AIBN (71.08 mg, 432.87 μmol). The reaction mixture was stirred at 80 °C for 18 hours under N 2 (g). Water (20 mL) and EtOAc (30 mL) were added to the mixture. The organic layer was separated, washed with brine, dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 20/1 to 0/1) to give 3,5-dibromo-4-fluoroisobenzofuran- 1(3H)-one (1.2 g, 90% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 7.99 (dd, J = 8.0, 5.9 Hz, 1H), 7.61 (d, J = 8.0 Hz, 1H), 6.82 (s, 1H). Step 3 5-Bromo-3,4-dihydroxyisobenzofuran-1(3H)-one [0824] A solution of 3,5-dibromo-4-fluoroisobenzofuran-1(3H)-one (1.2 g, 3.87 mmol) in 1 mol/L KOH (10 mL) was stirred at 100 °C for 3 hours. The mixture was acidified with 1N HCl until the solution reached pH 2~3, extracted with EtOAc (20 mL × 3). The organic layer was washed with water (20 mL), dried over Na 2 SO 4 , and concentrated to give 5-bromo-3,4-dihydroxyisobenzofuran-1(3H)-one (800 mg, 84% yield). The crude was taken to the next step without purification. Step 4 Dimethyl (6-bromo-7-hydroxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosp honate [0825] A solution of 5-bromo-3,4-dihydroxy-3H-isobenzofuran-1-one (800 mg, 3.26 mmol) in dimethyl phosphonate (1 mL) was stirred at 100 °C for 3 hours. Water (20 mL) and EtOAc (20 mL) were added to the mixture. The organic layer was separated, washed with brine, dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 20/1 to 0/1) to give dimethyl (6-bromo-7-hydroxy-3-oxo-1,3- dihydroisobenzofuran-1-yl)phosphonate (800 mg, 73% yield) as a white soild. LCMS ESI m/z 339.0 [M + H] + . Step 5 6-(4-(5-((6-Bromo-7-hydroxy-3-oxoisobenzofuran-1(3H)-ylidene )methyl)-2-fluorobenzoyl)piperazin- 1-yl)nicotinonitrile [0826] To a solution of dimethyl (6-bromo-7-hydroxy-3-oxo-1,3-dihydroisobenzofuran-1- yl)phosphonate (750 mg, 2.23 mmol) in THF (7 mL) was added 6-[4-(2-fluoro-5-formyl- benzoyl)piperazin-1-yl]pyridine-3-carbonitrile (790.48 mg, 2.34 mmol), Et 3 N (675.48 mg, 6.68 mmol, 930.41 μL) at 25 °C. The reaction mixture was stirred at 25 °C for 12 hours. The mixture was quenched with sat. Na 2 SO 3 solution (5 mL) and filtered. The filter cake was washed with H 2 O (5 mL * 2), dried under reduced pressure to give 6-(4-(5-((6-bromo-7-hydroxy-3-oxoisobenzofuran-1(3H)-ylidene )methyl)- 2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile (750 mg, 61% yield) as a yellow solid. LCMS ESI m/z 549.0 [M + H] + . Step 6 6-(4-(5-((6-Bromo-7-methoxy-3-oxoisobenzofuran-1(3H)-ylidene )methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0827] To a solution of 6-(4-(5-((6-bromo-7-hydroxy-3-oxoisobenzofuran-1(3H)-ylidene )methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile (80 mg, 145.63 μmol) in DMF (1 mL) was added K 2 CO 3 (60.38 mg, 436.88 μmol) and CH 3 I (31.01 mg, 218.44 μmol) at 25 °C . The reaction mixture was stirred at 25 °C for 4 hours. H 2 O (10 mL) was added to the mixture and filtered. The filter cake was washed with water (2 mL) and dried under reduced pressure to give 6-(4-(5-((6-bromo-7-methoxy-3-oxoisobenzofuran- 1(3H)-ylidene)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotin onitrile (58 mg, 71% yield). Step 7 6-(4-(5-((7-Bromo-8-methoxy-4-oxo-3,4-dihydrophthalazin-1-yl )methyl)-2-fluorobenzoyl)piperazin- 1-yl)nicotinonitrile [0828] To a solution of 6-(4-(5-((6-bromo-7-methoxy-3-oxoisobenzofuran-1(3H)-ylidene )methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile (58 mg, 102.95 μmol) in THF (2 mL) was added hydrazine hydrate (12.88 mg, 205.90 μmol, 12.55 μL, 80% purity) at 25 °C under N 2 (g). The reaction mixture was stirred at 75 °C for 4 hours. H 2 O (5 mL) was added to the mixture. The solids were collected by filtration, washed with water and dried under vacuum to give 6-(4-(5-((7-bromo-8-methoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)piperazin-1-y l)nicotinonitrile (55 mg, 93% yield). 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.76 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 8.09 (d, J = 8.5 Hz, 1H), 7.99 (d, J = 8.5 Hz, 1H), 7.90 (dd, J = 9.0, 2.3 Hz, 1H), 7.37 – 7.17 (m, 3H), 6.92 (d, J = 9.0 Hz, 1H), 4.39 (s, 2H), 3.86 (s, 3H), 3.63 (dd, J = 88.3, 44.8 Hz, 8H). Step 8 6-(4-(2-Fluoro-5-((8-methoxy-4-oxo-7-(prop-1-yn-1-yl)-3,4-di hydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0829] To a solution of 6-(4-(5-((7-bromo-8-methoxy-4-oxo-3,4-dihydrophthalazin-1-yl )methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile (45 mg, 77.94 μmol) in toluene (1 mL) was added tributyl(prop-1-ynyl)stannane (51.30 mg, 155.87 μmol), Pd(PPh 3 ) 4 (9.01 mg, 7.79 μmol) at 25 °C under N 2 (g). The reaction was stirred at 100 °C under microwave for 1 hour. The mixture was filtered and purified by prep-HPLC to give 6-[4-[2-fluoro-5-[(8-methoxy-4-oxo-7-prop-1-ynyl-3H- phthalazin-1-yl)methyl]benzoyl]piperazin-1-yl]pyridine-3-car bonitrile (7.65 mg, 18% yield) as a white solid. LCMS ESI m/z 537.2 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.68 (s, 1H), 8.51 (d, J = 2.0 Hz, 1H), 7.98 (d, J = 8.2 Hz, 1H), 7.90 (dd, J = 9.1, 2.4 Hz, 1H), 7.78 (d, J = 8.2 Hz, 1H), 7.32 – 7.27 (m, 1H), 7.22 (dt, J = 12.6, 5.9 Hz, 2H), 6.92 (d, J = 9.1 Hz, 1H), 4.35 (s, 2H), 3.91 (s, 3H), 3.79 – 3.69 (m, 4H), 3.62 (s, 2H), 3.29 (s, 2H), 2.15 (s, 3H). Synthesis of Example 82: 6-(4-(3-((6-Fluoro-7-methoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile 2-(2-Bromo-4-fluoro-5-methoxyphenyl)-1,3-dioxolane [0830] To a solution of 2-bromo-4-fluoro-5-methoxybenzaldehyde (2.0 g, 8.6 mmol) and (CH 2 OH) 2 (2.6 g, 43 mmol) in dry toluene (100 mL) was added TsOH (0.29 g, 0.17 mmol) at room temperature. The resulting mixture was allowed to heat to 120 ° C, and stirred at 120 ° C for 24 hours. The mixture was diluted with EtOAc (100 mL). The solution was washed by a sat. aq of NaHCO 3 (100 mL x 3). The organic layer was separated, dried, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 20/1 to 10/1, v/v) to give 2-(2-bromo-4-fluoro-5- methoxyphenyl)-1,3-dioxolane (2.2 g, 92% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 7.54 (d, J = 12.0 Hz, 1H), 7.21 (d, J = 8.0 Hz, 1H), 5.83 (s, 1H), 4.09-4.05 (m, 2H), 3.95-3.92 (m, 2H), 3.81 (s, 3H). Step 2 2-(1,3-Dioxolan-2-yl)-5-fluoro-4-methoxybenzonitrile [0831] A slurry of 2-(2-bromo-4-fluoro-5-methoxyphenyl)-1,3-dioxolane (1.0 g, 3.6 mmol) and CuCN (0.33 g, 3.6 mmol) in dry DMF (20 mL) was stirred at 165 °C (oil bath) for 1 hour. The reaction mixture was poured into H 2 O (50 mL), extracted with EA (30 mL x 3). The combined organic layers were washed with brine (20 mL x 3), dried, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 50/1 to 2/1, v/v) to give 2-(1,3-dioxolan-2-yl)-5-fluoro-4- methoxybenzonitrile (0.7 g, 88% yield) as an off-white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 7.54 (d, J = 12.0 Hz, 1H), 7.48 (d, J = 8.0 Hz,, 1H), 5.93 (s, 1H), 4.20-4.19 (m, 2H), 4.09-4.07 (m, 2H), 4.00 (m, 3H). Step 3 6-Fluoro-3-hydroxy-5-methoxyisobenzofuran-1(3H)-one [0832] A slurry of solution of 2-(1,3-dioxolan-2-yl)-5-fluoro-4-methoxybenzonitrile (0.6 g, 2.6 mmol) in a mixture of conc. H 2 SO 4 (8 mL) and H 2 O (8 mL) was stirred at 100 °C for 15 hours. The reaction solution was added dropwise to a solution of sat. NaHCO 3 (200 mL). The solution was extracted with EtOAc (30 mL x 4). The combined organic layers were dried, filtered, and concentrated. The residue was purified by silica gel chromatography (DCM/MeOH = 40/1 to 20/1, v/v) to give the desired compound of 6-fluoro-3-hydroxy-5-methoxyisobenzofuran-1(3H)-one (0.37 g, 60% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 7.57 (d, J = 12.0 Hz, 1H), 6.88 (d, J = 4.0 Hz, 1H), 6.54 (s, 1H), 3.89 (s, 3H). Step 4 Dimethyl 5-fluoro-6-methoxy-3-oxo-1,3-dihydroisobenzofuran-1-ylphosph onate [0833] Following the procedure of step 5 in Example 1 but starting with 6-fluoro-3-hydroxy-5- methoxyisobenzofuran-1(3H)-one gave the title compound as a yellow oil. 1 H NMR (400 MHz, DMSO- d 6 ) δ 7.59 (d, J = 12.0 Hz, 1H), 7.29-7.27 (m, 1H), 5.64 (d, J = 12.0 Hz,1H), 4.02 (s, 3H), 3.95 (d, J = 12.0 Hz,1H), 3.63 (d, J = 8.0 Hz,1H). Step 5 6-(4-(3-((5-Fluoro-6-methoxy-3-oxoisobenzofuran-1(3H)-yliden e)methyl)benzoyl)piperazin-1- yl)nicotinonitrile [0834] Following the procedure of step 6 in Example 1 but starting with dimethyl 5-fluoro-6-methoxy- 3-oxo-1,3-dihydroisobenzofuran-1-ylphosphonate gave the title compound as a light yellow solid. LCMS ESI m/z: 485.1 [M+H] + . Step 6 6-(4-(3-((6-Fluoro-7-methoxy-4-oxo-3,4-dihydrophthalazin-1-y l)methyl)benzoyl)piperazin-1- yl)nicotinonitrile [0835] Following the procedure of step 7 in Example 1, but starting with 6-(4-(3-((5-fluoro-6- methoxy-3-oxoisobenzofuran-1(3H)-ylidene)methyl)benzoyl)pipe razin-1-yl)nicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 498.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.59 (s, 1H), 8.51 (d, J = 4.0 Hz, 1H), 7.93 (d, J = 4.0 Hz, 1H), 7.89 (dd, J = 4.0 Hz, J = 8.0 Hz, 1H), 7.52 (d, J = 8.0 Hz, 1H), 7.46 - 7.38 (m, 3H), 7.30 (d, J = 8.0 Hz, 1H), 6.32 (d, J = 12.0 Hz, 1H), 4.38 (s, 2H), 3.97 (s, 3H), 3.68-3.59 (m, 6H), 3.41-3.40 (m, 2H). Synthesis of Example 83: 6-(4-(2-Fluoro-5-((6-methyl-4-oxo-7-(prop-1-yn-1-yl)-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile 5-Bromo-6-methyl-3H-isobenzofuran-1-one [0836] 4-bromo-3-methyl-benzoic acid (5 g, 23.25 mmol), dibromomethane (4.04 g, 23.25 mmol) and K 2 HPO 4 .3H 2 O (15.92 g, 69.75 mmol) were added to a 100 mL sealed tube. The mixture was purged with N 2 (g) for 2 times, then Pd(OAc) 2 (522.01 mg, 2.33 mmol) was added. The resultant mixture was stirred at 140 °C for 12 hours. After the reaction mixture was cooled to room temperature, the content was diluted with CH 2 Cl 2 (20 mL), filtered through a celite pad. The organic layer was washed with H 2 O (20 mL), brine (20 mL), dried over Na 2 SO 4 , concentrated in vacuum to afford the crude product, which was purified by silica gel chromatography (petroleum ether: EtOAc = 10: 1 to 5: 1) to afford 5-bromo-6- methyl-3H-isobenzofuran-1-one (3.7 g, 70% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 7.77 (s, 1H), 7.70 (s, 1H), 5.26 (s, 2H), 2.51 (s, 3H). Step 2 3,5-Dibromo-6-methyl-3H-isobenzofuran-1-one [0837] 5-Bromo-6-methyl-3H-isobenzofuran-1-one (3.7 g, 16.30 mmol), NBS (3.19 g, 17.93 mmol, 1.52 mL), AIBN (267.59 mg, 1.63 mmol) and CCl 4 (40 mL) were added to a 100 mL bottled flask. The mixture was stirred at 80 °C for 12 hours. The mixture was diluted with EtOAc (40 mL), and washed with H 2 O (20 mL), brine (30 mL), dried over Na 2 SO 4 , concentrated in vacuum to afford the crude product, which was purified by silica gel chromatography (petroleum ether: EtOAc = 10:1 to 5:1) to afford 3,5- dibromo-6-methyl-3H-isobenzofuran-1-one (2 g, 40% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 7.82 (s, 1H), 7.77 (s, 1H), 7.33 (s, 1H), 2.54 (s, 3H) Step 3 5-Bromo-3-hydroxy-6-methylisobenzofuran-1(3H)-one [0838] 3,5-Dibromo-6-methyl-3H-isobenzofuran-1-one (2 g, 6.54 mmol), KOH (733.53 mg, 13.07 mmol, 358.87 μL) and water (20 mL) were added to a 250 mL bottled flask. The mixture was stirred at 100 °C for 2 hours. The mixture was diluted in EtOAc (20 mL), the organic layer was washed with H 2 O (10 mL), brine (30 mL), dried over Na 2 SO 4 , concentrated in vacuum to afford the crude product, which was purified by silica gel chromatography (petroleum ether: EtOAc = 10: 1 to 5: 1) to afford 5-bromo-3- hydroxy-6-methylisobenzofuran-1(3H)-one (1.5 g, 94.4% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.30 (d, J = 7.6 Hz, 1H), 7.97 (s, 1H), 7.88 (s, 1H), 6.68 (d, J = 5.5 Hz, 1H), 2.52 (s, 3H). Step 4 Dimethyl (6-bromo-5-methyl-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosph onate [0839] 5-Bromo-3-hydroxy-6-methyl-3H-isobenzofuran-1-one (1.5 g, 6.17 mmol) was added into dimethylphosphite (679.16 mg, 6.17 mmol, 5 mL). The resulting mixture was stirred at 100 °C for 6 hours. The mixture was quenched with H2O (20 mL), extracted with EtOAc (20 mL x 2). The combined organic phase was washed with H 2 O (20 mL x 2), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to afford the crude product, which was purified by silica gel chromatography (petroleum ether: EtOAc = 20:1 to 1:1) to afford dimethyl (6-bromo-5-methyl-3-oxo-1,3-dihydroisobenzofuran-1- yl)phosphonate (850 mg, 41% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 7.94 (s, 1H), 7.79 (s, 1H), 5.64 (d, J = 10.9 Hz, 1H), 3.93 (d, J = 10.9 Hz, 3H), 3.68 (d, J = 10.7 Hz, 3H), 2.52 (s, 3H). Step 5 6-(4-(5-((6-bromo-5-methyl-3-oxoisobenzofuran-1(3H)-ylidene) methyl)-2-fluorobenzoyl)piperazin- 1-yl)nicotinonitrile [0840] To a mixture of dimethyl (6-bromo-5-methyl-3-oxo-1,3-dihydroisobenzofuran-1- yl)phosphonate (740 mg, 2.21 mmol) in THF (10 mL) was added 6-[4-(2-fluoro-5-formyl- benzoyl)piperazin-1-yl]pyridine-3-carbonitrile (1.34 g, 3.98 mmol) and Et3N (670.39 mg, 6.63 mmol, 923.41 μL). The resulting mixture was stirred at 25 °C for 12 hours. The mixture was quenched with sat. NaHSO 3 solution (20 mL) and filtered. The filter cake was washed with H 2 O (20 mL x 2) and dried under reduced pressure to give 6-(4-(5-((6-bromo-5-methyl-3-oxoisobenzofuran-1(3H)-ylidene) methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile (1.36 g, crude) as a yellow solid. Step 6 6-[4-[5-[(7-Bromo-6-methyl-4-oxo-3H-phthalazin-1-yl)methyl]- 2-fluoro-benzoyl]piperazin-1- yl]pyridine-3-carbonitrile [0841] To a mixture of 6-(4-(5-((6-bromo-5-methyl-3-oxoisobenzofuran-1(3H)-ylidene) methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile (1.36 g, 2.48 mmol) in THF (20 mL) was added hydrazine hydrate (310.95 mg, 4.97 mmol, 302.77 μL). The resulting mixture was stirred at 70 °C for 4 hours. The mixture was cooled to rt. and filtered. The filter cake was washed with a solution of petrol ether/EtOAc =10/1 (20 mL × 2). The solid was dried under reduced pressure to give 6-[4-[5-[(7-bromo-6- methyl-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluoro-benzoyl]pip erazin-1-yl]pyridine-3-carbonitrile (1.1 g, 79% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.68 (s, 1H), 8.57 (d, J = 2.2 Hz, 1H), 8.24 (d, J = 16.4 Hz, 2H), 7.96 (dd, J = 9.1, 2.3 Hz, 1H), 7.54 – 7.41 (m, 2H), 7.32 (t, J = 9.0 Hz, 1H), 6.98 (d, J = 9.1 Hz, 1H), 4.39 (s, 2H), 3.80 (d, J = 7.9 Hz, 4H), 3.66 (s, 2H), 3.37 (d, J = 2.4 Hz, 2H), 2.59 (s, 3H). Step 7 6-(4-(2-Fluoro-5-((6-methyl-4-oxo-7-(prop-1-yn-1-yl)-3,4-dih ydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0842] 6-[4-[5-[(7-Bromo-6-methyl-4-oxo-3H-phthalazin-1-yl)methyl]- 2-fluoro-benzoyl]piperazin-1- yl]pyridine-3-carbonitrile (100 mg, 178.12 μmol), tributyl(prop-1-ynyl)stannane (117.24 mg, 356.25 μmol), palladium;triphenylphosphane (20.58 mg, 17.81 μmol) and toluene (2 mL) were added to a 10 mL sealed tube. The mixture was purged with N 2 (g) for 3 times, the resultant mixture was stirred at 100 °C for 1 hr. The mixture was concentrated under reduced pressure to give the crude product, which was purified by prep-HPLC to afford 6-(4-(2-fluoro-5-((6-methyl-4-oxo-7-(prop-1-yn-1-yl)-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile (17.8 mg, 19% yield) as a white solid. LCMS ESI 520.8 [M + H] + . 1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.53 (s, 1H), 8.51 (d, J = 2.3Hz, 1H), 8.12 (s, 1H), 7.90 (dd, J = 9.0, 2.3 Hz, 2H), 7.43 –7.37 (m, 2H), 7.25 (t, J = 8.9 Hz, 1H), 6.92 (d, J = 9.1 Hz, 1H),4.31 (s, 2H), 3.74 (d, J = 9.4 Hz, 4H), 3.61 (s, 2H), 3.30 – 3.24 (m, 2H), 2.51 (d, J = 1.4 Hz, 3H), 2.13 (s, 3H). Synthesis of Example 84: 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-5-(trifluorometh yl)-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile

4-Bromo-2-methyl-6-(trifluoromethyl)aniline [0843] To a solution of 2-methyl-6-(trifluoromethyl)aniline (5.00 g, 28.60 mmol) in MeCN (20 mL) was added NBS (5.09 g, 28.6 mmol) in portions. The reaction mixture was stirred at rt for 3 hours, then diluted with EtOAc (100 mL) and water (100 mL). The organic phase was separated and the aqueous phase was extracted with EtOAc (50 mL × 2). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 10%) to afford 4-bromo-2-methyl-6-(trifluoromethyl)aniline as a yellow oil (6.90 g, 95% yield). LCMS ESI m/z 254 [M+H] + . Step 2 4-Bromo-2-methyl-6-(trifluoromethyl)benzonitrile [0844] A mixture of 4-bromo-2-methyl-6-(trifluoromethyl)aniline (6.90 g, 27.27 mmol), tert-Butyl nitrite (14.05 g, 136.36 mmol) and CuCN (3.83 g, 40.91 mmol) in DMSO (100 mL) were stirred at 70°C under Ar (g) for 1 hour. The reaction mixture was cooled to rt, diluted with water (50 mL) and extracted with EtOAc (3 × 50 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 10 to 20%) to afford 4-bromo-2-methyl-6-(trifluoromethyl)benzonitrile as a yellow oil (3.62 g, 50% yield). LCMSESI m/z 264 [M+H] + . Step 3 4-Bromo-2-methyl-6-(trifluoromethyl)benzamide [0845] A mixture of 4-bromo-2-methyl-6-(trifluoromethyl)benzonitrile (3.62 g, 13.76 mmol) and KOH (15.42 g, 275.29 mmol) in H 2 O (50 mL) were stirred at 100 °C for 16 hours. The mixture was cooled to rt, then extracted with EtOAc (3 × 50 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30 to 50%) to give 4-bromo-2-methyl-6-(trifluoromethyl)benzamide as a yellow solid (2.32 g, 60% yield). LC-MSESI m/z 282 [M+H] + . Step 4 4-Bromo-2-methyl-6-(trifluoromethyl)benzoic acid [0846] To a mixture of 4-bromo-2-methyl-6-(trifluoromethyl)benzamide (2.32 g, 8.26 mmol) in H 2 SO 4 (40 mL) was added a solution of NaNO 2 (9.68 g, 140.36 mmol) in H 2 O (15 mL) dropwise at 0°C. After stirred at 0°C for 30 mins, the reaction mixture was heated to 130 °C for 14 hours. The mixture was cooled to rt, diluted with H 2 O (40 mL) and extracted with EtOAc (3 × 40 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 50% to 80%) to give 4-bromo-2-methyl-6-(trifluoromethyl)benzoic acid as a yellow solid (2.11 g, 91% yield). LCMS ESI m/z 265, [M-H 2 O+H] + . Step 5 5-Bromo-7-(trifluoromethyl)isobenzofuran-1(3H)-one [0847] A mixture of 4-bromo-2-methyl-6-(trifluoromethyl)benzoic acid (2.11 g, 7.48 mmol), KBrO3 (7.49 g, 44.89 mmol) and NaHSO3 (4.67 g, 44.89 mmol) in H2O (50 mL) were stirred at rt for 5 days. The reaction mixture was extracted with EtOAc (3 × 50 mL). The combined organic layer was dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 0 to 10%) to give 5-bromo-7-(trifluoromethyl)isobenzofuran-1(3H)-one as a yellow solid (1.26 g, 60% yield). LCMS ESI m/z 281 [M+H]+.1H NMR (400 MHz, CDCl3) δ 7.97 (s, 1H), 7.68 (s, 1H), 5.33 (s, 2H). Step 6 3,5-Dibromo-7-(trifluoromethyl)isobenzofuran-1(3H)-one [0848] A mixture of 5-bromo-7-(trifluoromethyl)isobenzofuran-1(3H)-one (1.26 g, 4.50 mmol), AIBN (148 mg, 0.90 mmol) and NBS (960 mg, 5.40 mmol) in CCl4 (20 mL) were stirred at 80°C for 8 hours. The mixture was concentrated in vacuo, and the residue was purified by silica gel chromatography (EtOAc/pet ether = 0 to 10%) to give 3,5-dibromo-7-(trifluoromethyl)isobenzofuran-1(3H)-one as a yellow solid (880 mg, 55% yield). LCMS ESI m/z 359 [M+H]+. Step 7 5-Bromo-3-hydroxy-7-(trifluoromethyl)isobenzofuran-1(3H)-one [0849] A solution of 3,5-dibromo-7-(trifluoromethyl)isobenzofuran-1(3H)-one (880 mg, 2.46 mmol) in H2O (10 mL) and THF (10 mL) was stirred at 90 °C for 6 hours. The reaction mixture was cooled to rt, then extracted with EtOAc (3x 30 mL). The combined organic layer was dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30 to 40%) to give 5-bromo-3-hydroxy-7-(trifluoromethyl)isobenzofuran-1(3H)-one (470 mg, 65% yield). LCMS ESI m/z 297 [M+H]+. Step 8 Dimethyl 6-bromo-3-oxo-4-(trifluoromethyl)-1,3-dihydroisobenzofuran-1 -ylphosphonate [0850] A mixture of 5-bromo-3-hydroxy-7-(trifluoromethyl)isobenzofuran-1(3H)-one (470 mg, 1.59 mmol) in dimethyl phosphonate (5 mL) was stirred at 100 °C for 12 hours. The mixture was cooled to rt, diluted with H2O (20 mL) and extracted with EtOAc (3 × 20 mL). The combined organic layer was dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30 to 40%) to give dimethyl 6-bromo-3-oxo-4-(trifluoromethyl)-1,3- dihydroisobenzofuran-1-ylphosphonate as a yellow oil (380 mg, 63% yield). LCMS ESI m/z 389 [M+H]+. Step 9 6-(4-(5-((7-Bromo-4-oxo-5-(trifluoromethyl)-3,4-dihydrophtha lazin-1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0851] A mixture of 6-bromo-3-oxo-4-(trifluoromethyl)-1,3-dihydroisobenzofuran-1 -ylphosphonate (380 mg, 0.98 mmol), 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (330 mg, 0.98 mmmol) and Et3N (300 mg, 3 mmol) in THF (7 mL) was stirred at rt for 12 hours. To the reaction mixture was added N2H4•H2O (353 mg, 6 mmol), then stirred at 70 °C for 2 hours. The reaction mixture was concentrated in vacuo. The solids were diluted with MeOH (5 mL), filtered and dried under vacuum to give 6-(4-(5-((7-bromo-4-oxo-5-(trifluoromethyl)-3,4-dihydrophtha lazin-1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile as a white solid (150 mg, 25% yield). This crude product was used in the next step without further purification. LCMS ESI m/z 615 [M+H]+. Step 10 6-(4-(2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-5-(trifluoromethyl) -3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0852] A mixuture of 6-(4-(5-((7-bromo-4-oxo-5-(trifluoromethyl)-3,4-dihydrophtha lazin-1- yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile (150 mg, 0.25 mmol), tributyl(prop-1- ynyl)stannane (330 mg, 1.00 mmol) and Pd(dppf)Cl2 (22 mg, 0.03 mmol) in 1,4-dioxane (8 mL) was stirred at 100 °C under Ar (g) for 10 hours. The reaction mixuture was cooled to rt, diluted with water (20 mL) and extracted with EtOAc (3 × 20 mL). The combined organic layer was dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 80 to 100%) to give the desired product as a white solid (80 mg, 57% yield). LCMS ESI m/z 575 [M+H]+.1H NMR (400 MHz, DMSO–d6) δ 12.79 (s, 1H), 8.51 (d, J = 2.0 Hz, 1H), 8.10 (s, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.44 – 7.39 (m, 2H), 7.26 (t, J = 9.3 Hz, 1H), 6.92 (d, J = 9.2 Hz, 1H), 4.38 (s, 2H), 3.76 (d, J = 8.3 Hz, 4H), 3.61 (s, 2H), 3.32 – 7.29 (m, 2H), 2.11 (s, 3H). Synthesis of Example 85: 6-(4-(5-((5-chloro-4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophtha lazin-1- yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile 5-Bromo-7-chloroisobenzofuran-1(3H)-one [0853] A mixture of methyl 4-bromo-2-chloro-6-methylbenzoate (2.01 g, 7.59 mmol), NBS (6.75 g, 37.95 mmol) and AIBN (1.25 g, 7.59 mmol) in CCl 4 (100 mL) was stirred at 80 °C for 18 hours. The mixture was cooled to rt, diluted with H 2 O (50 mL), and extracted with DCM (50 mL × 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 10%) to obtain 5-bromo-7- chloroisobenzofuran-1(3H)-one as a white solid (1.41 g, 75% yield). LCMS ESI m/z 267 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 7.66 (s, 1H), 7.56 (s, 1H), 5.25 (s, 2H). Step 2 3,5-Dibromo-7-chloroisobenzofuran-1(3H)-one [0854] A mixture of methyl 5-bromo-7-chloroisobenzofuran-1(3H)-one (1.41 g, 5.70 mmol), NBS (1.12 g, 6.27 mmol) and AIBN (187 mg, 1.14 mmol) in CCl 4 (30 mL) was stirred at 80 °C for 15 hours. The mixture was cooled to rt, diluted with H 2 O (30 mL), and extracted with DCM (50 mL × 3). The combined organic layer dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 0 to 10%) to afford 3,5-dibromo-7- chloroisobenzofuran-1(3H)-one as a white solid (1.16 g, 62% yield). LCMS ESI m/z 326 [M+H] + . Step 3 5-Bromo-7-chloro-3-hydroxyisobenzofuran-1(3H)-one [0855] A mixture of 3,5-dibromo-7-chloroisobenzofuran-1(3H)-one (1.16 g, 3.55 mmol) in H 2 O (25 mL) was stirred at 100 °C for 4 hours. The reaction was cooled to rt and extracted with DCM (50 mL × 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give 5-bromo-7-chloro-3-hydroxyisobenzofuran-1(3H)-one as a light yellow oil (553 mg, 59% yield). LCMS ESI m/z 244 [M – H 2 O + H] + . Step 4 Dimethyl 6-bromo-4-chloro-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphon ate [0856] A solution of 5-bromo-7-chloro-3-hydroxyisobenzofuran-1(3H)-one (658 mg, 2.50 mmol) in dimethylphosphite (10 mL) was heated to 100 °C for 2 hours. The reaction mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give dimethyl 6- bromo-4-chloro-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphonat e as a light yellow oil (649 mg, 73% yield). LCMS ESI m/z 355 [M+H] + . Step 5

6-(4-(5-((7-Bromo-5-chloro-4-oxo-3,4-dihydrophthalazin-1-yl) methyl)-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile [0857] A solution of 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (649 mg, 1.92 mmol), dimethyl 6-bromo-4-chloro-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphon ate (649 mg, 1.83 mmol) and Et 3 N (554 mg, 5.48 mmol) in anhydrous THF (10 mL) was stirred at 25 °C under Ar (g) for 15 hours. To the reaction solution, hydrazine hydrate (914 mg, 18.26 mmol) was added and stirred at 70 °C for 1 hour. The reaction was cooled to rt, then THF was removed under vacuum. The solids were filtered, washed with water (10 mL) and petroleum ether/EtOAc (2:1, 10 mL) to give 6-(4-(5-((7-bromo-5-chloro- 4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)pip erazin-1-yl)nicotinonitrile as a white solid (135 mg, 12% yield). LCMS ESI m/z 581 [M+H] + . Step 6 6-(4-(5-((5-Chloro-4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalaz in-1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0858] A mixture of tributyl(prop-1-ynyl)stannane (183 mg, 0.56 mmol), 6-(4-(5-((7-bromo-5-chloro- 4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)pip erazin-1-yl)nicotinonitrile (100 mg, 0.19 mmol) and Pd(dppf)Cl 2 (15 mg, 0.02 mmol) in 1,4-dioxane (5 mL) was stirred at 100 °C under Ar (g) for 5 hours. The reaction mixture was concentrated in vacuo and the residue was purified by silica gel chromatography (MeOH/DCM = 0 to 5%) to afford 6-(4-(5-((5-chloro-4-oxo-7-(prop-1-ynyl)-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)piperazin-1-y l)nicotinonitrile as a white solid (26 mg, 28% yield). LCMS ESI m/z 541 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 9.75 (s, 1H), 8.42 (d, J = 1.9 Hz, 1H), 7.69 (d, J = 1.3 Hz, 1H), 7.66 (dd, J = 9.0, 2.3 Hz, 1H), 7.56 (d, J = 1.3 Hz, 1H), 7.35 – 7.27 (m, 2H), 7.08 (t, J = 8.8 Hz, 1H), 6.62 (d, J = 9.1 Hz, 1H), 4.20 (s, 2H), 3.90 (s, 2H), 3.78 (s, 2H), 3.69 (t, J = 5.2 Hz, 2H), 3.43 (s, 2H), 2.08 (s, 3H). Synthesis of Example 86: 6-(4-(2-Fluoro-5-((8-methyl-4-oxo-7-(prop-1-ynyl)-3,4-dihydr ophthalazin- 1-yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile 5-Bromo-4-methylisobenzofuran-1(3H)-one [0859] Into a single neck round bottom flask were added (3-bromo-2-methyl-phenyl)methanol (2.7 g, 13.43 mmol), TFA (30 mL) and LiCl (1.14 g, 26.86 mmol, 550.53 μL) at room temperature. The mixture was stirred overnight. The reaction was concentrated under reduced pressure. To the residue were added MeOH (70 mL), MgO (1.11 g, 26.86 mmol), PdCl 2 (237.69 mg, 1.34 mmol) and TTFA (7.29 g, 13.43 mmol) at room temperature. The mixture was stirred for 2 hr at room temperature under CO atmosphere. The mixture was diluted in EA, washed with water. The organic phase was dried over Na 2 SO 4 , concentrated under reduced pressure. The residue was purified by silica gel chromatography to give 5- bromo-4-methyl-3H-isobenzofuran-1-one (2.4 g, 57% yield) as a white solid. 1 H NMR (400 MHz, DMSO-D6) δ 7.87 (d, J = 8.1 Hz, 1H), 7.67 – 7.64 (m, 1H), 5.49 (s, 2H), 2.39 (s, 3H). Step 2 3,5-Dibromo-4-methylisobenzofuran-1(3H)-one [0860] Into a 100ml single round bottom flask were added 5-bromo-4-methyl-3H-isobenzofuran-1-one (900 mg, 3.96 mmol) CCl 4 (12 mL), AIBN (65.01 mg, 396.38 μmol) and NBS (917.12 mg, 5.15 mmol, 437.14 μL) at room temperature. The mixture was stirred overnight at 80°C under nitrogen atmosphere. The solvent was removed under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether: EtOAc = 15: 1) to give 3,5-dibromo-4-methyl-3H-isobenzofuran-1-one (880 mg, 73% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 7.83 (d, J = 8.1 Hz, 1H), 7.64 (d, J = 8.1 Hz, 1H), 7.30 (d, J = 5.2 Hz, 1H), 2.48 (s, 3H). Step 3 5-Bromo-3-hydroxy-4-methylisobenzofuran-1(3H)-one [0861] Into a 100 mL single round bottom flask were added 3,5-dibromo-4-methyl-3H-isobenzofuran- 1-one (850 mg, 2.78 mmol), H 2 O (14 mL) and KOH (779.43 mg, 13.89 mmol, 381.33 μL) at room temperature. The mixture was stirred for 40 min at 100 °C under nitrogen atmosphere. The reaction was allowed to cool to room temperature. The solution was adjusted to PH 1.0 with 1mol/L HCl aqueous solution. The precipitate was filtered. The filtrate was extracted with DCM. The organic phase was dried over Na2SO4. The precipitate was dissolved in the organic phase and concentrated under reduced pressure to give 5-bromo-3-hydroxy-4-methyl-3H-isobenzofuran-1-one (600 mg, 89% yield) as a white solid. LCMS ESI m/z 244.8 [M + H] + . Step 4 Dimethyl 6-bromo-7-methyl-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphon ate [0862] Into a 100 mL single round bottom flask were added 5-bromo-3-hydroxy-4-methyl-3H- isobenzofuran-1-one (600 mg, 2.47 mmol) and dimethyl phosphonate (8 mL) at room temperature. The solution was stirred for 16 hr at 100 °C under nitrogen atmosphere. The temperature was allowed to cool to room temperature. EA and water were added to the solution. The organic phase was dried over Na 2 SO 4 , concentrated under reduced pressure. The residue was purified by silica gel chromatography (EtOAc: Petroleum ether = 2: 1) to give dimethyl 6-bromo-7-methyl-3-oxo-1,3-dihydroisobenzofuran-1- ylphosphonate (380 mg, 46% yield) as a white solid. LCMS ESI m/z 337.0 [M + H] + . Step 5 5-((6-bromo-7-methyl-3-oxoisobenzofuran-1(3H)-ylidene)methyl )-2-fluorobenzonitrile [0863] Into a 100 mL single round bottom flask were added dimethyl 6-bromo-7-methyl-3-oxo-1,3- dihydroisobenzofuran-1-ylphosphonate (300 mg, 895.29 μmol) THF (20 mL) 2-fluoro-5-formyl- benzonitrile (226.96 mg, 1.52 mmol) and Et 3 N (271.78 mg, 2.69 mmol, 374.36 μL) at room temperature. The solution was stirred for 24 hr at room temperature under nitrogen atmosphere. The reaction was quenched with NaHSO 3 (aq) (30 mL), extracted with EA (30 mL), washed with water (20 mL). The organic phase was dried over Na 2 SO 4 , concentrated under reduced pressure. The residue was purified by triturated with DCM. The white solids were collected by filtration to give 5-((6-bromo-7-methyl-3- oxoisobenzofuran-1(3H)-ylidene)methyl)-2-fluorobenzonitrile (110 mg, 34% yield) as a white solid. 1 H NMR (400 MHz, DMSO-D6) δ 8.37 (dd, J = 6.4, 2.2 Hz, 1H), 8.34 – 8.27 (m, 1H), 7.97 (d, J = 8.1 Hz, 1H), 7.76 (d, J = 8.0 Hz, 1H), 7.68 (t, J = 9.1 Hz, 1H), 6.93 (s, 1H), 2.78 (s, 3H). Step 6 5-((7-bromo-8-methyl-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2-fluorobenzonitrile [0864] To a mixture of 5-((6-bromo-7-methyl-3-oxoisobenzofuran-1(3H)-ylidene)methyl )-2- fluorobenzonitrile (110 mg, 307.12 μmol) in THF (3 mL) was added hydrazine hydrate (38.44 mg, 614.25 μmol, 37.43 μL) at 25 °C . After stirring the reaction mixture at 70 °C for 3 hr. Water and EtOAc were added to the mixture, which became two phases. The organic layer was separated, washed with NaCl (aq), dried over Na2SO4 and concentrated to give 5-((7-bromo-8-methyl-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)-2-fluorobenzonitrile (110 mg, 96.2% yield) as a yellow solid. LCMS ESI m/z 372.0 [M+H] + . Step 7 5-((7-Bromo-8-methyl-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2-fluorobenzoic acid [0865] To a solution of 5-[(7-bromo-8-methyl-4-oxo-3H-phthalazin-1-yl)methyl]-2-fluo ro-benzonitrile (110 mg, 295.55 μmol) in water (4 mL) and EtOH (1 mL) was added KOH (165.83 mg, 2.96 mmol, 81.13 μL) at 25 °C. After stirring the reaction mixture at 100 °C for 2 hr .Ethanol was evaporated off and the aqueous was extracted with ethyl acetate . The aqueous was then acidified with conc HCl until the solution reached pH 1. The solids were collected by filtration and dried under vacuum to give 5-((7- bromo-8-methyl-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fl uorobenzoic acid (95 mg, crude) as a red solid. LCMS ESI m/z 390.7 [M+H] + . Step 8 6-(4-(5-((7-bromo-8-methyl-4-oxo-3,4-dihydrophthalazin-1-yl) methyl)-2-fluorobenzoyl)piperazin-1- yl)nicotinonitrile [0866] Following the amide coupling procedure in Example 9, but starting with 5-((7-Bromo-8- methyl-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenz oic acid and 6-(piperazin-1- yl)nicotinonitrile gave the title compound as a red solid. LCMS ESI m/z 560.7 [M + H] + . Step 9

6-(4-(2-Fluoro-5-((8-methyl-4-oxo-7-(prop-1-ynyl)-3,4-dihydr ophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0867] To a solution of 6-[4-[5-[(7-bromo-8-methyl-4-oxo-3H-phthalazin-1-yl)methyl]- 2-fluoro- benzoyl]piperazin-1-yl]pyridine-3-carbonitrile (110 mg, 195.94 μmol) in toluene (1 mL) was added tributyl(prop-1-ynyl)stannane (128.97 mg, 391.87 μmol), Pd(PPh 3 ) 4 (22.64 mg, 19.59 μmol) at 25 °C under N 2 (g). The reaction was stirred under microwave at 100 °C for 1 hr. The reaction mixture was filtered and concentrated. The residure was purified by prep-HPLC to give 6-[4-[2-fluoro-5-[(8- methyl-4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)methyl]benzoyl ]piperazin-1-yl]pyridine-3-carbonitrile (12.4 mg, 12% yield) as a white solid. LCMS ESI m/z 520.7 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.65 (s, 1H), 8.51 (d, J = 2.3 Hz, 1H), 8.13 (d, J = 8.2 Hz, 1H), 7.90 (dd, J = 9.1, 2.4 Hz, 1H), 7.78 (d, J = 8.2 Hz, 1H), 7.29 – 7.19 (m, 3H), 6.92 (d, J = 9.2 Hz, 1H), 4.51 (s, 2H), 3.73 (d, J = 13.0 Hz, 4H), 3.61 (s, 2H), 3.34 (d, J = 3.2 Hz, 2H), 2.79 (s, 3H), 2.14 (s, 3H). Synthesis of Example 87: 6-(4-(3-((5,6-Difluoro-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

6-(4-(3-Ethynylbenzoyl)piperazin-1-yl)nicotinonitrile [0868] To a solution of 3-ethynylbenzoic acid (4.0 g, 21.3 mmol) in THF (30 mL) was added 6- (piperazin-1-yl)nicotinonitrile (3.1 g, 21.3 mmol), HATU (9.7 g, 25.5 mmol) and DIPEA (8.2 g, 63.8 mmol). The resulting mixture was stirred at 20 o C for 2 h. Then water (30 mL) and EtOAc (40 mL) were added to the mixture, which was became two phases. The organic layer was separated, washed with water (10 mL x 2), aq. NH 4 Cl (10 mL x 2), brine (10 mL x 2), dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (PE/EA = 10/1 to 2/1, v/v) to give 6-(4-(3- ethynylbenzoyl)piperazin-1-yl)nicotinonitrile (6.0 g, 89% yield) as a white solid. LCMS ESI m/z 317.1 [M+H] + . Step 2 6-(4-(3-((3,4-Difluoro-2-formylphenyl)ethynyl)benzoyl)pipera zin-1-yl)nicotinonitrile [0869] A mixture of 6-(4-(3-ethynylbenzoyl)piperazin-1-yl)nicotinonitrile (3 g, 9.5 mmol) in DMF (15 mL) was added 6-bromo-2,3-difluorobenzaldehyde (2.1 g, 9.5 mmol), Pd(PPh 3 ) 2 Cl 2 (0.7 g, 1.0 mmol), CuI (0.2 g, 1.0 mmol) and ET3N (2.9 g, 28.5 mmol). The resulting mixture was stirred at rt for 1 h. The mixture was concentrated to give a residue. The residue was purified by triturated with petroleum ether/EtOAc (Petroleum ether/EtOAc = 5/1) (20 mL) to give 6-(4-(3-((3,4-difluoro-2- formylphenyl)ethynyl)benzoyl)piperazin-1-yl)nicotinonitrile (2.0 g, 46% yield) as a yellow solid. LCMS ESI m/z 457.1 [M+H] + . Step 3 6-(4-(3-((4,5-difluoro-3-oxoisobenzofuran-1(3H)-ylidene)meth yl)benzoyl)piperazin-1- yl)nicotinonitrile [0870] To a solution of 6-(4-(3-((3,4-difluoro-2-formylphenyl)ethynyl)benzoyl)pipera zin-1- yl)nicotinonitrile (1.0 g, 2.2 mmol) in MeCN (10 mL) was added 1,3-dimesitylimidazolidin-1-ium chloride (113 mg, 0.3 mmol) and DBU (100 mg, 0.7 mmol). The resulting mixture was stirred at 80 o C for 2 h under O 2 . Water (10 mL) and EtOAc (20 mL) were added to the mixture, which became two phases. The organic layer was separated, washed with brine (10 mL× 2), dried over Na 2 SO 4 and concentrated to a residue. The residue was purified by silica gel chromatography (PE/EA = 10/1 to 2/1, v/v) to give (Z)-6- (4-(3-((4,5-difluoro-3-oxoisobenzofuran-1(3H)-ylidene)methyl )benzoyl)piperazin-1-yl)nicotinonitrile (500 mg, 48% yield) as a yellow solid. LCMS ESI m/z 473.1 [M+H] + . Step 4 6-(2-(3-(4-(5-Cyanopyridin-2-yl)piperazine-1-carbonyl)phenyl )acetyl)-2,3-difluorobenzohydrazide [0871] Following the procedure of step 5 in Example 6, but starting with 6-(4-(3-((4,5-difluoro-3- oxoisobenzofuran-1(3H)-ylidene)methyl)benzoyl)piperazin-1-yl )nicotinonitrile gave the title compound as a yellow solid. LCMS ESI m/z 504.1 [M+H] + Step 5 6-(4-(3-((5,6-difluoro-4-oxo-3,4-dihydrophthalazin-1-yl)meth yl)benzoyl)piperazin-1- yl)nicotinonitrile [0872] Following the procedure of step 6 in Example 6 but starting with 6-(2-(3-(4-(5-cyanopyridin-2- yl)piperazine-1-carbonyl)phenyl)acetyl)-2,3-difluorobenzohyd razide gave the title compound as a yellow solid. LCMS ESI m/z 486.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.64 (s, 1H), 8.51 (m, 1H), 8.03 (m, 1H), 7.88 (m, 1H), 7.82 (m, 1H), 7.41 (m, 3H), 7.28 (m, 1H), 6.91 (m, 1H), 4.34 (s, 2H), 3.68 (m, 6H), 3.36 (m, 2H). Synthesis of Example 88: 6-(4-(3-((5-Fluoro-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

2-Fluorobenzoyl chloride [0873] 2-Fluorobenzoic acid (20.0 g, 142.74 mmol) was added into sulfurous dichloride (15 mL) slowly at 0 °C. Then N,N-dimethylformamide (1 mL) was added dropwise to the solution at 0 °C. The mixture was stirred at 80 °C for 3 hr. The reaction mixture was concentrated under reduced pressure to give 2-fluorobenzoyl chloride (22.6 g, 100% yield) as brown oil. Step 2 N,N-Diethyl-2-fluorobenzamide [0874] To a solution of 2-fluorobenzoyl chloride (22.6 g, 142.54 mmol) in dichloromethane (100 mL) was added diethylamine (22.1 mL, 213.80 mmol). The resulting mixture was stirred at 80 °C for 6 hr. The mixture was quenched with water (200 mL), extracted with dichloromethane (200 mL x 2). The combined organic phase was washed successively with hydrochloric acid (200 mL x 2, 1 M) and saturated sodium chloride solution (200 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give N,N-diethyl-2-fluorobenzamide (27.8 g, 100% yield) as a brown oil. Step 3 N,N-Diethyl-2-fluoro-6-formylbenzamide [0875] To a solution of N,N-diethyl-2-fluorobenzamide (10.0 g, 51.22 mmol) in tetrahydrofuran (100 mL) was added N,N,N',N'-Tetramethylethylenediamine (TMEDA, 16.9 mL, 112.68 mmol). sec- Butyllithium (s-BuLi, 88.7 mL, 112.68 mmol, 1.3 M in hexane) was added dropwise at -78 °C under nitrogen atmosphere and then the mixture was stirred at -78 °C for 0.5 hr under nitrogen atmosphere. N,N- dimethylformamide (19.8 mL, 256.10 mmol) was added slowly at -78 °C under nitrogen atmosphere. The resulting mixture was stirred at -78 °C for 1 h under nitrogen atmosphere. The mixture was quenched with hydrochloric acid (200 mL, 1 M), extracted with ethyl acetate (200 mL x 2). The combined organic phase was washed saturated sodium chloride solution (200 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether/ ethyl acetate = 10/1, v/v) to give N,N-diethyl-2-fluoro-6-formylbenzamide (4.75 g, 42% yield) as a yellow oil. LCMS ESI m/z: 223.9 [M+H] + . Step 4 7-Fluoro-3-hydroxyisobenzofuran-1(3H)-one [0876] A mixture of N,N-diethyl-2-fluoro-6-formylbenzamide (4.75 g, 21.28 mmol) was stirred at 100 °C for 2 hr. The mixture was diluted with water (100 mL), extracted with ethyl acetate (100 mL x 2). The combined organic phase was washed with saturated sodium chloride solution (100 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether/ ethyl acetate = 4/1, v/v) to give 7-fluoro-3-hydroxyisobenzofuran- 1(3H)-one (2.19 g, 61% yield) as a white solid. Step 5, 6 and 7 6-(4-(3-((5-Fluoro-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)b enzoyl)piperazin-1-yl)nicotinonitrile [0877] Following the three-step phthalazinone synthesis procedure in Example 1, but starting with 7- fluoro-3-hydroxyisobenzofuran-1(3H)-one gave the title compound as a yellow solid. LCMS ESI m/z 468.8 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.56 (s, 1H), 8.51 (d, J = 4.0 Hz, 1H), 7.91 - 7.88 (m, 2H), 7.77 (d, J = 8.0 Hz, 1H), 7.62 - 7.57 (m, 2H), 7.43 - 7.38 (m, 3H), 7.29 (d, J = 8.0 Hz, 1H), 6.92 (d, J = 8.0 Hz, 1H), 4.34 (s, 2H), 3.74 - 3.67 (m, 6H), 3.34 - 3.31 (m, 2H). Synthesis of Example 89: 6-(4-(2-Fluoro-5-((8-methoxy-4-oxo-7-phenoxy-3,4-dihydrophth alazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile methyl 3-methoxy-4-phenoxybenzoate [0878] A mixture of methyl 4-hydroxy-3-methoxybenzoate (5 g, 27.45 mmol), iodobenzene (11.20 g, 0.30 mmol) and Cu 2 O (9.43 g, 65.87 mmol) in 246 trimethylpyridine (10 mL) was stirred at 180 °C for 2 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na2SO4, and filtered and concentrated in vacuo. The residue was purified by prep-HPLC to give methyl 3-methoxy-4- phenoxybenzoate (4.90 g, 69% yield) as a white solid. LCMS ESI m/z: 259 [M+H] + . Step 2 3-methoxy-4-phenoxybenzoic acid [0879] A solution of methyl 3-methoxy-4-phenoxybenzoate (4.90 g, 18.97 mmol) and LiOH (3.41 g, 142.27 mmol) in MeOH (30 mL) and H 2 O (10 mL) was stirred at rt for 1 hour. The organic solvent was removed in vacuum and aq HCl (1N) was added until the solution reached to pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 3-methoxy-4-phenoxybenzoic acid (4.54 mg, 97% yield) as a white solid. LCMS ESI m/z 245 [M+H] + . This crude product was used in the next step without further purification. Step 3 N,N-diethyl-3-methoxy-4-phenoxybenzamide [0880] A mixture of 3-methoxy-4-phenoxybenzoic acid (4.24 g, 17.36 mmol) and thionyl chloride (41.31 g, 347.20 mmol) was stirred at 80 °C under Ar (g) for 3 hours. The reaction was concentrated under vacuum, the residue was dissolved in DCM (30 mL), then added a solution of Et 2 NH (1.90 g, 26.04 mmol) and Et 3 N (5.27 g, 52.08 mmol) in DCM (100 mL). The reaction was stirred at rt for 1 hour, then diluted with water (100 mL) and extracted with DCM (100 mL x 3). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 20 to 40%) to give N,N-diethyl-3-methoxy-4- phenoxybenzamide (5.28 g, 95% yield) as a yellow oil. LCMS ESI m/z 300 [M+H] + . Step 4 N,N-diethyl-2-formyl-3-methoxy-4-phenoxybenzamide [0881] A solution of N,N-diethyl-3-methoxy-4-phenoxybenzamide (600 mg, 2.00 mmol) and TMEDA (175 mg, 1.50 mmol) in THF (10 mL) was cooled to -78°C, then n-BuLi(2.5M in heptane, 0.60 mL, 1.50 mmol) was added dropwise under Argon. The reaction was stirred at -78°C for 30 min, then added DMF (123 mg, 1.70 mmol) dropwise. The reaction was stirred at -78°C for 20 min, then quenched with sat. NH 4 Cl soltion (10 mL) and neutralized aq HCl (1N) until the solution reached pH 5~6. The mixture was extracted with EtOAc (50 x 3 mL). The combined organic layer was washed with bine, dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 10% to 20%) to give N,N-diethyl-2-formyl-3-methoxy-4-phenoxybenzamide (160 mg, 27 % yield) as a white solid. LCMS ESI m/z: 328 [M+H] + . Step 5 3-Hydroxy-4-methoxy-5-phenoxyisobenzofuran-1(3H)-one [0882] A mixture of N,N-diethyl-2-formyl-3-methoxy-4-phenoxybenzamide (160 mg, 0.49 mmol) in 6 M HCI (5 mL) was stirred at 100 °C for 16 hours. The reaction was cooled to rt. The solid was collected by filteration, washed with water and dried under vacuum to give 3-hydroxy-4-methoxy-5- phenoxyisobenzofuran-1(3H)-one (150 mg, 95% yield) as a yellow solid. LCMS ESI m/z: 273 [M+H] + . This crude product was used in the next step without further purification. Step 6 7-Methoxy-3-oxo-6-phenoxy-1,3-dihydroisobenzofuran-1-yl dimethyl phosphate [0883] A solution of 3-hydroxy-4-methoxy-5-phenoxyisobenzofuran-1(3H)-one (150 mg, 0.55 mmol) in dimethyl phosphonate (1.21 g, 11.02 mmol) was stirred at 100°C for 2 hours. The mixture was cooled to rt, diluted with H 2 O (100 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 10% to 15%) to afford 7-methoxy- 3-oxo-6-phenoxy-1,3-dihydroisobenzofuran-1-yl dimethyl phosphate (95 mg, 20% yield) as a white solid. LCMS ESI m/z: 381 [M+H] + . Step 7

6-(4-(2-Fluoro-5-((8-methoxy-4-oxo-7-phenoxy-3,4-dihydrophth alazin-1-yl)methyl) benzoyl)piperazin-1-yl)nicotinonitrile [0884] A solution of 7-methoxy-3-oxo-6-phenoxy-1,3-dihydroisobenzofuran-1-yl dimethyl phosphate (95 mg, 0.25 mmol), 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (97.87 mg, 0.29 mmol) and Et 3 N (80 mg, 0.79 mmol) in anhydrous THF (5 mL) was stirred at 25 °C under Ar (g) for 3 hours. Hydrazine hydrate (20 mg, 0.39 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and pet ether/ EtOAc (10:1, 10 mL) to give 6-(4-(2-fluoro-5-((8-methoxy-4-oxo-7- phenoxy-3,4-dihydrophthalazin-1-yl)methyl)benzoyl)piperazin- 1-yl)nicotinonitrile (19 mg, 32% yield ) as a yellow. LCMS ESI m/z: 591 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.63 (s, 1H), 8.50 (d, J = 2.2 Hz, 1H), 8.05 (d, J = 8.7 Hz, 1H), 7.89 (dd, J = 9.1, 2.3 Hz, 1H), 7.46 – 7.35 (m, 3H), 7.34 – 7.28 (m, 1H), 7.22 (dt, J = 14.8, 6.2 Hz, 3H), 7.05 (d, J = 7.8 Hz, 2H), 6.90 (d, J = 9.2 Hz, 1H), 4.39 (s, 2H), 3.79 (s, 3H), 3.78 – 3.68 (m, 4H), 3.64 – 3.56 (m, 2H), 3.31 – 3.25 (m, 2H). Synthesis of Example 90: 6-(4-(2-fluoro-5-((4-oxo-7-(m-tolyloxy)-3,4-dihydrophthalazi n-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

Methyl 2-bromo-4-(m-tolyloxy)benzoate [0885] A mixture of methyl methyl 2-bromo-4-fluorobenzoate (2.00 g, 8.58 mmol), K 2 CO 3 (2.37 g, 17.16 mmol) and m-cresol (1.39 g, 12.87 mmol) in DMF (20 mL) was stirred at 100 °C for 16 hours. The reaction was cooled to rt, diluted with H 2 O (100 mL) and extracted with EtOAc (100 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0% to 7%) to give methyl 2-bromo-4-(m-tolyloxy)benzoate (2.45 g, 89% yield) as a colorless oil. LCMS ESI m/z 321.0 [M+H] + . Step 2 2-Bromo-4-(m-tolyloxy)benzoic acid [0886] A mixture of methyl methyl 2-bromo-4-(m-tolyloxy)benzoate (2.45 g, 7.63 mmol) and LiOH (1.83 g, 76.28 mmol) in MeOH (21 mL) and H 2 O (7 mL) was stirred at rt for 2 hours. The organic solvent was removed in vacuo and aq HCl (1N) was added until the solution reached pH 5~6. The formed solid was collected by filtration and dried under vacuum to give 2-bromo-4-(m-tolyloxy)benzoic (2.3 g, 98% yield) as a white solid. LCMS ESI m/z 307.0 [M+H] + . Step 3 O O O OH 3-Hydroxy-5-(m-tolyloxy)isobenzofuran-1(3H)-one [0887] A solution of 2-bromo-4-(m-tolyloxy)benzoic acid (2.0 g, 6.51 mmol) in THF (35 mL) was cooled to -78 °C, then n-BuLi (2.5M in heptane, 5.21 mL, 13.02 mmol) was addded dropwise under Ar (g). The reaction was stirred at -78 °C for 30 min, then added DMF (1.05 g, 14.33 mmol). The reaction was stirred at -78°C for 20 min, then quenched with sat. NH 4 Cl soltion (10 mL), neutralizated by aq HCl (1N) until the solution reached pH 5~6 and extracted with EtOAc (30 x 3 mL). The combined organic layer was washed with bine, dried over Na 2 SO 4 and filtered. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30% to 50%) to give 3-hydroxy-5-(m-tolyloxy)isobenzofuran- 1(3H)-one (900 mg, 54% yield) as a white solid. LCMS ESI m/z 257.1 [M+H] + . S Dimethyl (3-oxo-6-(m-tolyloxy)-1,3-dihydroisobenzofuran-1-yl)phosphon ate [0888] A solution of 3-hydroxy-5-(m-tolyloxy)isobenzofuran-1(3H)-one (500 mg, 1.95 mmol) in dimethyl phosphonate (5 mL) was stirred at 100 °C for 16 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 50% to 60 %) to give dimethyl (3-oxo-6-(m-tolyloxy)-1,3- dihydroisobenzofuran-1-yl)phosphonate (350 mg, 52% yield) as a off-white solid. LCMS ESI m/z 349.1 [M+H] + . Step 5 6-(4-(2-Fluoro-5-((4-oxo-7-(m-tolyloxy)-3,4-dihydrophthalazi n-1-yl)methyl)benzoyl)piperazin-1- yl)nicotinonitrile [0889] A solution of dimethyl (3-oxo-6-(m-tolyloxy)-1,3-dihydroisobenzofuran-1-yl)phosphon ate (250 mg, 0.72 mmol), 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (255 mg, 0.75 mmol) and Et 3 N (218 mg, 2.15 mmol) in anhydrous THF (5 mL) was stirred at 60 °C under Ar (g) for 15 hours. Hydrazine hydrate (55 mg, 0.93 mmol) was added and the reaction was stirred at 70 °C for 1 hour. The reaction was cooled to rt, then THF was removed under vacuum. The formed solid was washed with water (10 mL) and petroleum ether/EtOAc (2:1, 10 mL) to give 6-(4-(2-fluoro-5-((4-oxo-7-(m-tolyloxy)-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)piperazin-1-yl)nicotin onitrile (343 mg, 83% yield) as a white solid. LCMS ESI m/z 575.1 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.55 (s, 1H), 8.49 (d, J = 2.2 Hz, 1H), 8.24 (d, J = 8.7 Hz, 1H), 7.87 (dd, J = 9.1, 2.3 Hz, 1H), 7.38 – 7.30 (m, 5H), 7.21 (t, J = 9.3 Hz, 1H), 7.09 (d, J = 7.5 Hz, 1H), 6.93 – 6.86 (m, 3H), 4.23 (s, 2H), 3.79 – 3.69 (m, 4H), 3.59 (s, 2H), 3.28 (s, 2H), 2.30 (s, 3H). Synthesis of Example 91: 6-(4-(2-fluoro-5-((7-methoxy-4-oxo-3,4-dihydropyrido[4,3-d]p yridazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile 6-Methoxy-4-methylnicotinic acid [0890] To a solution of 5-bromo-2-methoxy-4-methyl-pyridine (10 g, 49.49 mmol) in THF (100 mL) was dropwised with n-BuLi (2.5 M, 23.76 mL) under -78 °C , and then stirred at -78 °C for 0.5 hr. The reaction mixture was charged with CO 2 balloon, and stirred at -78 °C for 2 hr, then 1 N HCl aq. was added to quench the reaction, warmed to rt. The solution was extracted with EA, washed with brine, dried and concentrated to give 6-methoxy-4-methyl-pyridine-3-carboxylic acid (15 g, crude) as a white solid. Step 2 Methyl 6-methoxy-4-methylnicotinate [0891] To a solution of 6-methoxy-4-methyl-pyridine-3-carboxylic acid (15 g, 89.73 mmol) in MeOH (100 mL) was dropwised with H 2 SO 4 (10 mL), and then heated to 68 °C for 16 hr. The reaction mixture was concentrated to give a residue, 100 mL H2O was added, and then the solution was basitified until the solution reached pH 9 by sat. NaHCO 3 , extracted with EtOAc (100 mL x 3), washed with brine, dried and concentrated to give methyl 6-methoxy-4-methylnicotinate (1.3 g, 8.0% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 8.75 (s, 1H), 6.58 (s, 1H), 3.97 (s, 3H), 3.88 (s, 3H), 2.57 (s, 3H). Step 3 Methyl 4-(dibromomethyl)-6-methoxynicotinate [0892] To a sealed tube was added methyl 6-methoxy-4-methyl-pyridine-3-carboxylate (600 mg, 3.31 mmol), AIBN (10 mg), NBS (1.30 g, 7.29 mmol) and CCl 4 (18 mL), and then heated to 80 °C for 16 h. The mixture was concentrated and purified by silica gel chromatography to give methyl 4- (dibromomethyl)-6-methoxy-pyridine-3-carboxylate (950 mg, 85% yield) as a light yellow solid. 1 H NMR (400 MHz, CDCl 3 ) δ 8.78 (s, 1H), 7.95 (s, 1H), 7.43 (s, 1H), 4.03 (s, 3H), 3.94 (s, 3H). Step 4 1-Hydroxy-6-methoxyfuro[3,4-c]pyridin-3(1H)-one [0893] To a 25 mL round bottle was added methyl 4-(dibromomethyl)-6-methoxy-pyridine-3- carboxylate (350 mg, 1.03 mmol), KOH (289.67 mg, 5.16 mmol, 141.72 μL), H 2 O (10 mL), and then heated to 100 °C for 3 hr, The mixture was acidified with 1 N HCl until the solution reached pH 5 and then extracted with EtOAc (15 mL x 3), washed with brine, dried and concentrated to give 1-hydroxy-6- methoxy-1H-furo[3,4-c]pyridin-3-one (80 mg, 42.77% yield) as a light yellow solid. Step 5, 6 and 7

6-(4-(2-Fluoro-5-((7-methoxy-4-oxo-3,4-dihydropyrido[4,3-d]p yridazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [0894] Following the three-step phthalazinone synthesis procedure above in Example 1, but starting with 1-hydroxy-6-methoxyfuro[3,4-c]pyridin-3(1H)-one gave the title compound as a white solid. LCMS ESI m/z: 499.7 [M + H] + . 1 H NMR (400 MHz, DMSO-d6) δ12.59 (s, 1H), 9.14 (s, 1H), 8.52 (d, J = 2.1 Hz, 1H), 7.91 (dd, J = 9.1, 2.1 Hz, 1H), 7.45 (t, J = 6.8 Hz, 2H), 7.26 (t, J = 8.9 Hz, 1H), 7.18 (s, 1H), 6.93 (d, J = 9.2 Hz, 1H), 4.27 (s, 2H), 4.00 (s, 3H), 3.75 (d, J = 12.2 Hz, 4H), 3.64 (s, 2H), 3.37 (s, 2H). Synthesis of Example 92: 6-(4-(2-Fluoro-5-((5-oxo-5,6-dihydropyrido[2,3-d]pyridazin-8 - yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

[0895] 1 (0.405 g, 3.00 mmol) and 1-bromopyrrolidine-2,5-dione (0.59 g, 3.3 mmol) were dissolved CCI4 and heated at reflux for 2 hours. The reaction was cooled and filtered, the filtrate was evaporated to afford 7-bromofuro[3,4-b]pyridin-5(7H)-one(2), as a yellow solid (quantitative yield), which was used directly in the next stage without further purification. [0896] 2 (3.0, 0.642 mmol) and triphenylphosphine (0.86 g, 3.3 mmol) were heated at reflux in acetonitrile (20 mL) for 4h. The reaction was cooled and concentrated and purified by using flash column (DCM/MeOH) to afford the desired material 3. [0897] 3 (0.605 g, 1.27 mmol) and 2-fluoro-5-formylbenzonitrile (0.23 g, 1.52 mmol) were dissolved DCM (7 mL), to this was added triethylamine (0.27 mL, 1.9 mmol) and the reaction was stirred overnight. The reaction mixture was quenched with water (50 mL), extracted with EA (2 x 15 mL), the organic layer was dried over MgSO4 , filtered and evaporated to afford an orange gum crude. This was passed through a plug of silica eluting with ethyl acetate to afford 4 as a pale yellow oil. (0.13g, 35%) [0898] To the above crude 4 (0.13 g) was added water (2 mL), EtOH (2 mL) and DMF (0.12 mL). Hydrazine hydrate (0.16mL) were added and the reaction was heated at reflux overnight. The reaction was cooled, and the precipitate was collected by filtration, washed with EtOH (25 mL) and air dried to afford the desired compound 5 as a white solid (78.2 mg, 56%) [0899] To 5 (78.2 mg) were added KOH (0.15 g, 10.0 mmol) and EtOH (1 mL) and water (3.5 mL) and heated at 1000C for 5 hours. The ethanol was evaporated off and the aqueous was extracted with ethyl acetate (2 x 5mL). The aqueous was then acidified to pH1 with cone HCI to afford a solid, which was filtered, washed with water and dried to the desired material 6 as a beige solid. (73 mg, 87%) [0900] 2-fluoro-5-((5-oxo-5,6-dihydropyrido[2,3-d]pyridazin-8-yl)me thyl)benzoic acid (0.028 g, 0.069 mmol) and 6-(piperazin-1-yl)nicotinonitrile (0.016 g, 0.083 mmol, 1.2 equiv.) were dissolved in anhydrous DMF (0.4 mL) and cooled to 0 o C. To this mixture was added diisopropylethylamine (35 uL, 3.0 equiv.) followed by addition of propylphosphonic anhydride (T3P) (90 uL, 0.13 mmol, 3.0 equiv.) (50% in EtOAc). The mixture was stirred for 30 min at rt. The mixture was diluted with ethyl acetate (5 mL) and sat. NaHCO 3 (5 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate. The organic extracts were washed with brine (5 mL), filtered through a silica plug, and dried over MgSO 4 . After filtration, the solvent was removed in vacuo. Flash chromatography (0-100% EtOAc in Heptanes) over silica gel afforded the product as a white solid (0.0277g). LCMS ESI m/z: 470.1 [M + H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.87 (s, 1H), 9.10 (dd, J = 4.6, 1.8 Hz, 1H), 8.69 (dd, J = 8.1, 1.8 Hz, 1H), 8.44 – 8.39 (m, 1H), 7.76 – 7.62 (m, 2H), 7.51 (ddd, J = 13.2, 5.0, 2.2 Hz, 2H), 7.03 (t, J = 8.8 Hz, 1H), 6.62 (d, J = 9.0 Hz, 1H), 4.45 (s, 2H), 3.89 (s, 2H), 3.84 – 3.72 (m, 2H), 3.44 (s, 2H). Synthesis of Example 93: 6-(4-(5-((7-Cyclobutoxy-4-oxo-5-(trifluoromethyl)-3,4-dihydr ophthalazin- 1-yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile

5-Bromo-7-(trifluoromethyl)isobenzofuran-1(3H)-one [0901] To a high pressure reaction vessel was added 4-bromo-2-(trifluoromethyl)benzoic acid (1.80 g, 6.49 mmol), Pd(OAc) 2 (729 mg, 3.25 mmol), potassium phosphate dibasic (3.46 g, 19.5 mmol), and finally methylene bromide (14 mL, 6.49 mmol). The tube was sealed and the reaction was heated to 140 ℃ for 24 hours. The crude reaction mixture was filtered through a 0.45 ^m syringe filter and the resulting filtrate was concentrated under vacuum. The resulting residue was dissolved in minimal DCM and subjected to normal phase chromotography (Heptanes-EtOAc, 5% to 100% over 10 minutes). The collected fractions were concentrated under vacuum to afford 5-bromo-7-(trifluoromethyl)isobenzofuran- 1(3H)-one (630 mg, 35 %) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.31 (s, 1H), 8.13 (s, 1H), 5.45 (s, 2H). Step 2 5-Cyclobutoxy-7-(trifluoromethyl)isobenzofuran-1(3H)-one [0902] A flame-dried vial with stir bar was charged with 5-bromo-7-(trifluoromethyl)isobenzofuran- 1(3H)-one (606 mg, 2.16 mmol), Pd(OAc) 2 (19.8 mg, 86.3 umol), Cs 2 CO 3 (1.42 g, 4.31 mmol) and 5- [di(1-adamantyl)phosphino]-12,32,52-triphenyl-12H-[1,42]bipy razole (114 mg, 173 umol). The vial was sealed with a septum and purged with nitrogen gas. A separate screw cap vial was charged with toluene (3.72 mL) and cyclobutanol (517 uL, 6.47 mmol), capped and degassed for a 5 minutes period using a nitrogen balloon and sonication. The mixture was then added to the vial under nitrogen and the reaction mixture was stirred at 100 °C for 24 hours. After 20 hours of stirring, the reaction mixture then cooled and filtered. The filtrate was then concentrated under vacuum and the resulting residue was suspended in minimal DCM and subjected to normal-phase chromotography (5%-95% Heptanes:EtOAc) to afford 5- cyclobutoxy-7-(trifluoromethyl)isobenzofuran-1(3H)-one (110 mg, 19 % yield) as a white solid. LCMS ESI m/z: 273.2 [M+H] + . Step 3 3-Bromo-5-cyclobutoxy-7-(trifluoromethyl)isobenzofuran-1(3H) -one [0903] To a solution of cyclobutoxy-7-(trifluoromethyl)isobenzofuran-1(3H)-one (110 mg, 0.404 mmol) in CCl 4 (1.99 mL) was added n-bromosuccinimide (72.6 mg, 0.404 mmol) and finally Luperox A98, benzoyl peroxide (4.31 uL, 0.0202 mmol). The solution was refluxed for 24 hours. After completion, the reaction was concentrated under vacuum and the resulting solution was dissolved in minimal DCM and subjected to normal phase chromotography (Heptanes:EtOAc, 5-95%) to afford 3-bromo-5- cyclobutoxy-7-(trifluoromethyl)isobenzofuran-1(3H)-one (142 mg, 100% yield) as an off-white solid which was carried forward to the next step. LCMS ESI m/z: 367 [M+OH]-. Step 4 5-Cyclobutoxy-3-hydroxy-7-(trifluoromethyl)isobenzofuran-1(3 H)-one [0904] A mixture of 3-bromo-5-cyclobutoxy-7-(trifluoromethyl)isobenzofuran-1(3H) -one (142 mg, 404 umol) in H 2 O (5 mL) was stirred at reflux for 16 hours. The reaction was cooled to room and extracted with DCM (10 mL × 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuum to afford 5-cyclobutoxy-3-hydroxy-7-(trifluoromethyl)isobenzofuran-1(3 H)- one (117 mg, 100% yield) as a light-yellow solid which was carried forward. LCMS ESI m/z: 287.1 [M- H] + . Step 5 Dimethyl (6-cyclobutoxy-3-oxo-4-(trifluoromethyl)-1,3-dihydroisobenzo furan-1-yl)phosphonate [0905] To a microwave vial was added 5-cyclobutoxy-3-hydroxy-7-(trifluoromethyl)isobenzofuran- 1(3H)-one (117 mg, 406 umol) and dimethyl phosphite (1.00 mL). The solution was sparged with nitrogen and the reaction was stirred at 110 °C for 45 minutes. The reaction was then diluted with water and EtOAc and extracted three times. The pooled organic layers were then washed with water and finally brine. The organic layer was then concentrated under vacuum to afford a yellow oil. The oil was left under high vacuum overnight to afford dimethyl (6-cyclobutoxy-3-oxo-4-(trifluoromethyl)-1,3- dihydroisobenzofuran-1-yl)phosphonate (154 mg, 100% yield) as an off-white solid. LCMS ESI m/z: 381.2 [M-H] + . Step 6 6-(4-(5-((7-cyclobutoxy-4-oxo-5-(trifluoromethyl)-3,4-dihydr ophthalazin-1-yl)methyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinonitrile [0906] To a glass vial containing (6-cyclobutoxy-3-oxo-4-(trifluoromethyl)-1,3-dihydroisobenzo furan- 1-yl)phosphonate (163 mg, 429 umol) in THF (21.4 mL) was added triethylamine (179 uL, 1.29 mmol) and 6-(4-(2-fluoro-5-formylbenzoyl)piperazin-1-yl)nicotinonitril e (160 mg, 472 umol). The reaction was heated to 70 °C for 48 hours. Hydrazine hydrate solution (54.4 uL, 1.71 mmol) was added and the reaction was left to stir for another 48 hours at 70 °C. After completion of the reaction, the solution was concentrated under vacuum. Methanol was then added to the resulting residue which induced the formation of a white solid. The solid was filtered off and the eluent was again concentrated. The residue was dissolved in minimal DMSO and subjected to reverse phase chromatography (10 mM aqueous ammonium formate : MeCN, 5-95%). The fractions containing product were pooled and lyophilized to afford 6-(4-(5-((7-cyclobutoxy-4-oxo-5-(trifluoromethyl)-3,4-dihydr ophthalazin-1- yl)methyl)-2-fluorobenzoyl)piperazin-1-yl)nicotinonitrile (8.20 mg, 3.2 % yield) as a white powder. LCMS ESI m/z: 607.4 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.64 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.62 (d, J = 2.0 Hz, 1H), 7.45 – 7.39 (m, 2H), 7.32 – 7.23 (m, 2H), 6.93 (d, J = 9.1 Hz, 1H), 4.93 (p, J = 7.1 Hz, 1H), 4.37 (s, 2H), 3.75 (s, 2H), 3.72 (s, 2H), 3.60 (s, 2H), 3.29 (s, 2H), 2.34 (d, J = 7.5 Hz, 2H), 2.05 – 1.93 (m, 2H), 1.78 (q, J = 10.2 Hz, 1H), 1.66 (dd, J = 18.6, 8.5 Hz, 1H). Synthesis of Example 94: 6-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-1,4-diazepan-1-yl)nicotinonitrile Methyl 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoate [0907] A solution of dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (1.6 g, 4.20 mmol), methyl 2-fluoro-5-formylbenzoate (762 mg, 4.20 mmol) and Et 3 N (700 mg, 6.9 mmol) in anhydrous THF (10 mL) was stirred at 60 °C under Ar (g) for 15 hours. Hydrazine hydrate (202 mg, 6.3 mmol) was added and the reaction was heated to 70°C for 1 hour. The reaction was concentrated under vacuum. The formed solids were washed with water (10 mL) and pet ether/EA (2:1, 10 mL) to afford methyl 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoate as a white solid (1.16 g, 59% yield). LCMS ESI m/z 383 [M+H] + . Step 2 5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic aciD [0908] A mixture solution of methyl 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoate (1.16 g , 3.03 mmol) and LiOH-H 2 O (636 mg, 15.15 mmol) in MeOH (15 mL) and H 2 O (5 mL) was stirred at rt for 3 hours. The organic solvent was removed under vacuo and aq HCl (1N) was added until the solution reached pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid as a white solid (1.05 g, 89% yield). This was used without further purification. LCMS ESI m/z 369 [M+H] + . Step 3 tert-Butyl 4-(5-cyanopyridin-2-yl)-1,4-diazepane-1-carboxylate [0909] A mixture of 6-chloronicotinonitrile (300 mg, 2.17 mmol), tert-butyl 1,4-diazepane-1- carboxylate (434 mg, 2.17 mmol) and K 2 CO 3 (599 mg, 4.34 mmol) in ACN (5 mL) was stirred at 60 °C for 16 hours. The reaction was concentrated in vacuo, diluted with water (20 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 10% to 50%) to give tert-butyl 4-(5-cyanopyridin-2-yl)-1,4-diazepane-1- carboxylate (620 mg, 95% yield) as a white solid. LCMS ESI m/z: 303 [M+H] + . Step 4 6-(1,4-Diazepan-1-yl)nicotinonitrile [0910] A solution of tert-butyl 4-(5-cyanopyridin-2-yl)-1,4-diazepane-1-carboxylate (620 mg, 2.05 mmol) in HCl/dioxane (4.0 M, 10 mL, 40.0 mmol) was stirred at rt for 1 hour. The solvent was removed in vacuo to give 6-(azetidin-3-yl(methyl)amino)nicotinonitrile (400 mg, 96% yield) as a white solid. This was used without further purification. LCMS ESI m/z: 203 [M+H] + . Step 5 6-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-1,4-diazepan-1- yl)nicotinonitrile [0911] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (80 mg, 0.22 mmol) and 6-(1,4-diazepan-1-yl)nicotinonitrile (62 mg, 0.26 mmol) in DMF (2 mL) was added EDCI (62 mg, 0.33 mmol), HOBt (44 mg, 0.33 mmol) and DIPEA (140 mg, 1.10 mmol). The reaction mixture was stirred at rt for 2 hours. The reaction was purified by prep-HPLC to give 6-(4-(5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoyl)-1,4-diazepan-1- yl)nicotinonitrile (52 mg, 43% yield) as a white solid. LCMS ESI m/z: 553 [M+H] + . 1 H NMR (400 MHz, DMSO–d 6 ) δ 12.44 (s, 1H), 8.50 – 8.30 (m, 1H), 8.19 – 8.12 (m, 1H), 7.87 – 7.80 (m, 1H), 7.43 – 7.28 (m, 2H), 7.26 – 7.11 (m, 1H), 7.11 – 7.06 (m, 1H), 7.05 – 6.74 (m, 2H), 4.87 – 4.75 (m, 1H), 4.25 (d, J = 24.2 Hz, 2H), 3.91 – 3.47 (m, 7H), 3.28 – 3.14 (m, 1H), 2.42 – 2.32 (m, 2H), 2.05 – 1.94 (m, 2H), 1.89 – 1.37 (m, 4H). Synthesis of Example 95: 2-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)isonicotinonitrile

tert-Butyl 4-(4-cyanopyridin-2-yl)piperazine-1-carboxylate [0912] A mixture of 2-chloroisonicotinonitrile (200 mg, 1.44 mmol), tert-butyl piperazine-1- carboxylate (323 mg, 1.73 mmol) and DIPEA (746 mg, 5.77 mmol) in DMF (5 mL) was stirred at 100 °C for 20 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 0 to 10%) to give tert-butyl 4-(4-cyanopyridin-2-yl)piperazine-1-carboxylate as a white solid (350 mg, 84% yield). LCMS ESI m/z 289 [M+H] + . Step 2 2-(Piperazin-1-yl)isonicotinonitrile [0913] A solution of tert-butyl 4-(4-cyanopyridin-2-yl)piperazine-1-carboxylate (350 mg, 1.21 mmol) in DCM (5 mL) and TFA (0.5 mL) was stirred at 25 °C for 2 hours. The reaction mixture was concentrated in vacuo to afford 2-(piperazin-1-yl)isonicotinonitrile as a yellow oil (366 mg, 99% yield). This was used without further purification. LCMS ESI m/z 189 [M+H] + . Step 3 2-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1- yl)isonicotinonitrile [0914] To a solution of 2-(piperazin-1-yl)isonicotinonitrile (43 mg, 0.23 mmol) and 5-((7-cyclobutoxy- 4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenzoic acid (70 mg, 0.19 mmol) in DMF (3 mL) was added EDCI (36 mg, 0.19 mmol), HOBt (39 mg, 0.29 mmol) and DIPEA (74 mg, 0.57 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was concentrated in vacuo and the residue was purified by prep-HPLC to give 2-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)isonicotinonitrile as a white solid (55 mg, 54% yield). LCMS ESI m/z 539 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.45 (s, 1H), 8.31 (d, J = 5.0 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.42 (t, J = 6.4 Hz, 2H), 7.34 – 7.22 (m, 3H), 7.11 (d, J = 2.3 Hz, 1H), 7.00 (dd, J = 5.0, 1.0 Hz, 1H), 4.91 – 4.76 (m, 1H), 4.31 (s, 2H), 3.77 – 3.62 (m, 4H), 3.52 (s, 2H), 3.29 (s, 2H), 2.44 – 2.34 (m, 2H), 2.06 – 1.95 (m, 2H), 1.83 – 1.58 (m, 2H). Synthesis of Example 96: 6-(8-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-3,8-diazabicyclo[3.2.1]octan-3-yl)nicotinonit rile

tert-Butyl 3-(5-cyanopyridin-2-yl)-3,8-diazabicyclo[3.2.1]octane-8-carb oxylate [0915] A mixture of 6-chloronicotinonitrile (300 mg, 2.17 mmol), tert-butyl 3,8- diazabicyclo[3.2.1]octane-8-carboxylate (460 mg, 2.17 mmol) and K 2 CO 3 (509 mg, 3.68 mmol) in MeCN (5 mL) was stirred at 60 °C for 16 hours. The reaction was concentrated in vacuo, diluted with water (20 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 10% to 50%) to give tert-butyl 3-(5-cyanopyridin-2-yl)-3,8- diazabicyclo[3.2.1]octane-8-carboxylate (580 mg, 85% yield) as a white solid. LCMS ESI m/z: 315 [M+H] + . Step 2 6-(3,8-Diazabicyclo[3.2.1]octan-3-yl)nicotinonitrile [0916] A solution of tert-butyl 3-(5-cyanopyridin-2-yl)-3,8-diazabicyclo[3.2.1]octane-8-carb oxylate (580 mg, 1.84 mmol) in HCl/dioxane (4.0M, 5 mL, 20.0 mmol) was stirred at rt for 1 hour. The solvent was removed in vacuo to give 6-(3,8-diazabicyclo[3.2.1]octan-3-yl)nicotinonitrile (370 mg, 94% yield) as a white solid. This was used without further purification. LCMS ESI m/z: 215 [M+H] + . Step 3

6-(8-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-3,8- diazabicyclo[3.2.1]octan-3-yl)nicotinonitrile [0917] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (80 mg, 0.22 mmol) and 6-(3,8-diazabicyclo[3.2.1]octan-3-yl)nicotinonitrile (163 mg, 0.66 mmol) in DMF (2 mL) was added EDCI (62 mg, 0.33 mmol), HOBt (44 mg, 0.33 mmol) and DIPEA (140 mg, 1.10 mmol). The reaction mixture was stirred at rt for 2 h. The reaction was purified by prep-HPLC to give 6-(8-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-3,8- diazabicyclo[3.2.1]octan-3-yl)nicotinonitrile (38 mg, 31% yield) as a white solid. LCMS ESI m/z: 565 [M+H] + . 1 H NMR (400 MHz, DMSO–d 6 ) δ 12.45 (s, 1H), 8.52 – 8.50 (m, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.92 – 7.88 (m, 1H), 7.51 – 7.47 (m, 1H), 7.46 – 7.41 (m, 1H), 7.33 – 7.24 (m, 2H), 7.13 – 7.10 (m, 1H), 6.85 (d, J = 9.2 Hz, 1H), 4.87 – 4.78 (m, 2H), 4.32 (s, 2H), 4.25 (d, J = 12.4 Hz, 1H), 4.08 (d, J = 12.1 Hz, 1H), 3.88 – 3.82 (m, 1H), 3.13 (d, J = 11.7 Hz, 1H), 2.90 (d, J = 12.1 Hz, 1H), 2.43 – 2.36 (m, 2H), 2.03 – 1.95 (m, 2H), 1.91 – 1.79 (m, 2H), 1.78 – 1.61 (m, 4H). Synthesis of Example 97: 6-cyclobutoxy-4-(4-fluoro-3-(3-(5-(trifluoromethyl)pyrimidin -2-yl)-3,8- diazabicyclo [3.2.1]octane-8-carbonyl)benzyl)phthalazin-1(2H)-one

tert-Butyl 3-(5-(trifluoromethyl)pyrimidin-2-yl)-3,8-diazabicyclo [3.2.1] octane-8-carboxylate [0918] A mixture of tert-butyl 3,8-diazabicyclo[3.2.1]octane-8-carboxylate (200 mg, 0.94 mmol), 2- chloro-5-(trifluoromethyl)pyrimidine (172 mg, 0.94 mmol) and K 2 CO 3 (221 mg, 0.02 mmol) in ACN (4 mL) was stirred at 60 °C for 12 hours. The solid was filtered off, and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 40% to 50%) to give the desired product tert-butyl 3-(5-(trifluoromethyl)pyrimidin-2-yl)-3,8-diazabicyclo[3.2.1 ]octane-8- carboxylate (302 mg, 89% yield) as a white solid. LCMS ESI m/z 359.1 [M+H] + . Step 2 3-(5-(trifluoromethyl)pyrimidin-2-yl)-3,8-diazabicyclo[3.2.1 ]octane [0919] To a solution of tert-butyl3-(5-(trifluoromethyl)pyrimidin-2-yl)-3,8- diazabicyclo[3.2.1] octane- 8-carboxylate (300 mg, 0.84 mmol) in dioxane (1 mL) was added HCl/dioxane (4.0M in dioxane, 2 mL, 8.0 mmol). The mixture was stirred at rt for 1 hour, then concentrated in vacuo to afford 3-(5- (trifluoromethyl)pyrimidin-2-yl)-3,8-diazabicyclo [3.2.1]octane (193 mg, 89 % yield) as a white solid. This was used without further purification. LCMS ESI m/z 259.1 [M+H] + . Step 3

6-cyclobutoxy-4-(4-fluoro-3-(3-(5-(trifluoromethyl)pyrimidin -2-yl)-3,8-diazabicyclo[3.2.1]octane-8- carbonyl)benzyl)phthalazin-1(2H)-one [0920] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 –fluorobenzoic acid (80 mg, 0.21 mmol) and 3-(5-(trifluoromethyl)pyrimidin-2-yl)-3,8-dia zabicyclo[3.2.1]octane (76.8 mg, 0.40 mmol) in DMF (2 mL) was added EDCI (63 mg, 0.32 mmol), HOBt (44 mg, 0.32 mmol) and DIPEA (140 mg, 1.09 mmol). The reaction mixture was stirred at rt for 2 hours. The reaction was purified by prep-HPLC to give 6-cyclobutoxy-4-(4-fluoro-3-(3-(5-(trifluoromethyl)pyrimidin -2-yl)-3,8- diazabicyclo [3.2.1]octane-8-carbonyl)benzyl)phthalazin-1(2H)-one (45 mg, 34% yield) as a white solid. LCMS ESI 609.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.46 (s, 1H), 8.73 (s, 1H), 8.26 – 8.06 (m, 1H), 7.50 (dd, J = 6.4, 2.0 Hz, 1H), 7.46 – 7.40 (m, 1H), 7.34 – 7.24 (m, 2H), 7.11 (d, J = 2.3 Hz, 1H), 4.84 (dd, J = 14.1, 6.9 Hz, 2H), 4.67 – 4.23 (m, 4H), 3.87 (d, J = 5.2 Hz, 1H), 3.10 (dd, J = 86.6, 12.5 Hz, 2H), 2.38 (m, J = 25.4, 12.7 Hz, 2H), 2.07 – 1.43 (m, 8H). Synthesis of Example 98: 6-Cyclobutoxy-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin -2-yl)-1,4- diazepane-1-carbonyl)benzyl)phthalazin-1(2H)-one

tert-Butyl 4-(5-(trifluoromethyl)pyrimidin-2-yl)-1,4-diazepane-1-carbox ylate [0921] A mixture of tert-butyl 1,4-diazepane-1-carboxylate (300 mg, 1.50 mmol), 2-chloro-5- (trifluoromethyl)pyrimidine (273 mg, 3.61 mmol) and K 2 CO 3 (352 mg, 2.55 mmol) in ACN (5 mL) was stirred at 60 °C for 16 hours. The reaction was concentrated in vacuo, diluted with water (30 mL) and extracted with EtOAc (30 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 10% to 20%) to give tert-butyl 4-(5- (trifluoromethyl)pyrimidin-2-yl)-1,4-diazepane-1-carboxylate (466 mg, 90% Yield) as a white solid. LCMS ESI m/z: 347 [M+H] + . Step 2 1-(5-(Trifluoromethyl)pyrimidin-2-yl)-1,4-diazepane [0922] To a solution of tert-butyl 4-(5-(trifluoromethyl)pyrimidin-2-yl)-1,4-diazepane-1-carb oxylate (466 mg, 1.35 mmol) ) in dioxane (1 mL) was added HCl/dioxane (4.0M in dioxane, 2 mL, 8.0 mmol). The mixture was stirred at rt for 1 hour. The solvent was removed in vacuo to give 1-(5- (trifluoromethyl)pyrimidin-2-yl)-1,4-diazepane (203 mg 61% yield) as a white solid. This was used without further purification. LCMS ESI m/z: 548 [M+H] + . Step 3 6-Cyclobutoxy-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin -2-yl)-1,4-diazepane-1- carbonyl)benzyl)phthalazin-1(2H)-one [0923] To a solution of 1-(5-(trifluoromethyl)pyrimidin-2-yl)-1,4-diazepane (74 mg, 0.26 mmol) and 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (80mg, 0.22mmol) in DMF (1 mL) was added EDCI (62 mg, 0.32 mmol), HOBt (44 mg, 0.32 mmol) and DIPEA (140 mg, 1.1 mmol). The reaction mixture was stirred at rt for 2 hours. The reaction was purified by prep-HPLC to give 6-cyclobutoxy-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin -2-yl)-1,4-diazepane-1- carbonyl)benzyl)phthalazin-1(2H)-one (45 mg, 61% yield) as a white solid. LCMS ESI m/z 597.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.44 (d, J = 4.5 Hz, 1H), 8.81 – 8.46 (m, 2H), 8.16 (dd, J = 8.8, 2.3 Hz, 1H), 7.38 – 7.02 (m, 5H), 4.87 – 4.76 (m, 1H), 4.25 (d, J = 23.3 Hz, 2H), 3.94 (d, J = 28.3 Hz, 1H), 3.90 – 3.77 (m, 4H), 3.65 (s, 1H), 3.43 (t, J = 5.6 Hz, 1H), 3.22 (t, J = 5.8 Hz, 1H), 2.38 (s, 2H), 2.04 – 1.43 (m, 6H). Synthesis of Example 99: 6-(3-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-3,8-diazabicyclo[3.2.1]octan-8-yl)nicotinonit rile

tert-Butyl 3-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2-fluorobenzoyl)-3,8- diazabicyclo[3.2.1]octane-8-carboxylate [0924] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (300 mg, 0.81 mmol) and tert-butyl 3,8-diazabicyclo[3.2.1]octane-8-carboxylate (173 mg, 0.81 mmol) in DMF (3 mL) was added EDCI (165 mg, 1.22 mmol), HOBt (234 mg, 1.22 mmol) and DIPEA (316 mg, 2.44 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (PE: EA = 9: 1 to 1: 1) to give tert-butyl methyl(3-oxo-3-(4-(5- (trifluoromethyl)pyrimidin-2-yl)piperazin-1-yl)propyl)carbam ate (426 mg, 0.75 mmol, 93% yield) as a white solid. LCMS ESI m/z: 563.1 [M+H] + . Step 2 4-(3-(3,8-Diazabicyclo[3.2.1]octane-3-carbonyl)-4-fluorobenz yl)-6-cyclobutoxyphthalazin-1(2H)-one [0925] To a solution of tert-butyl 3-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2- fluorobenzoyl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (395 mg, 0.70 mmol) in dioxane (5 mL) was added HCl/Dioxane (4M, 5 mL) and stirred at 25 °C for 1 hour. The mixture was concentrated in vacuo to afford 4-(3-(3,8-diazabicyclo[3.2.1]octane-3-carbonyl)-4-fluorobenz yl)-6-cyclobutoxyphthalazin-1(2H)- one as a white solid (320 mg, 99% yield). This was used without further purification. LCMS ESI m/z: 463.1 [M+H] + . Step 3 6-(3-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-3,8- diazabicyclo[3.2.1]octan-8-yl)nicotinonitrile [0926] To a solution of 4-(3-(3,8-diazabicyclo[3.2.1]octane-3-carbonyl)-4-fluorobenz yl)-6- cyclobutoxyphthalazin-1(2H)-one (80 mg, 0.17 mmol) and 6-chloronicotinonitrile (29 mg, 0.21 mmol) in DMF (1 mL) was added DIPEA (67 mg, 0.51 mmol). The reaction mixture was stirred at 90 °C for 16 hours. The reaction was purified by prep-HPLC to give 6-(3-(5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)-3,8-diazabic yclo[3.2.1]octan-8-yl)nicotinonitrile (56.9 mg, 58% yield) as a white solid. LCMS ESI m/z: 565.0 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.45 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.89 (dd, J = 9.0, 2.3 Hz, 1H), 7.39 (dd, J = 7.0, 4.6 Hz, 2H), 7.29 (dt, J = 8.9, 4.5 Hz, 1H), 7.23 (t, J = 8.8 Hz, 1H), 7.08 (s, 1H), 6.89 (d, J = 9.0 Hz, 1H), 4.86 – 4.73 (m, 2H), 4.58 (s, 1H), 4.29 (s, 3H), 3.30 – 3.18 (m, 1H), 3.11 (d, J = 12.3 Hz, 1H), 2.97 (d, J = 12.6 Hz, 1H), 2.37 (d, J = 5.4 Hz, 2H), 2.04 – 1.87 (m, 4H), 1.75 (t, J = 8.6 Hz, 2H), 1.63 (d, J = 8.6 Hz, 2H). Synthesis of Example 100: 6-Cyclobutoxy-4-(4-fluoro-3-(8-(5-(trifluoromethyl)pyrimidin -2-yl)-3,8- diazabicyclo[3.2.1]octane-3-carbonyl)benzyl)phthalazin-1(2H) -one

6-Cyclobutoxy-4-(4-fluoro-3-(8-(5-(trifluoromethyl)pyrimidin -2-yl)-3,8-diazabicyclo[3.2.1]octane-3- carbonyl)benzyl)phthalazin-1(2H)-one [0927] To a solution of 4-(3-(3,8-diazabicyclo[3.2.1]octane-3-carbonyl)-4-fluorobenz yl)-6- cyclobutoxyphthalazin-1(2H)-one (100 mg, 0.22 mmol) and 2-chloro-5-(trifluoromethyl)pyrimidine (47 mg, 0.26 mmol) in DMF (3 mL) was added DIPEA (84 mg, 0.65 mmol). The reaction mixture was stirred at 60 o C for 3 hours. The mixture was purified by prep-HPLC to give 6-cyclobutoxy-4-(4-fluoro-3-(8-(5- (trifluoromethyl)pyrimidin-2-yl)-3,8-diazabicyclo[3.2.1]octa ne-3-carbonyl)benzyl) phthalazin-1(2H)-one (39 mg, 30% yield) as a white solid. LCMS ESI m/z: 609.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.45 (s, 1H), 8.73 (s, 2H), 8.15 (d, J = 8.8 Hz, 1H), 7.40 (dd, J = 7.0, 4.6 Hz, 2H), 7.30 (dd, J = 8.8, 2.3 Hz, 1H), 7.24 (t, J = 8.9 Hz, 1H), 7.08 (s, 1H), 4.92 – 4.78 (m, 2H), 4.67 (d, J = 6.1 Hz, 1H), 4.43 – 4.24 (m, 3H), 3.31 – 3.25 (m, 1H), 3.18 (d, J = 14.8 Hz, 1H), 3.01 (d, J = 12.4 Hz, 1H), 2.38 (d, J = 6.6 Hz, 2H), 2.04 – 1.87 (m, 4H), 1.75 (t, J = 9.6 Hz, 2H), 1.70 – 1.48 (m, 2H). Synthesis of Example 101: 6-Cyclobutoxy-4-(3-(4-(cyclopropanecarbonyl)-1,4-diazepane-1 - carbonyl)-4-fluorobenzyl)phthalazin-1(2H)-one

tert-Butyl 4-(cyclopropanecarbonyl)-1,4-diazepane-1-carboxylate [0928] To a solution of tert-butyl 1,4-diazepane-1-carboxylate (300 mg, 1.50 mmol) and cyclopropanecarboxylic acid (155 mg, 1.80 mmol) in DMF (2 mL) was added EDCI (471 mg, 2.25 mmol), HOBt (304 mg, 2.25 mmol) and DIPEA (581 mg, 4.49 mmol). The resulting reaction mixture was stirred at rt for 16 hours, then the reaction was quenched with water (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic phase was washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (Petroleum ether/EtOAc = 4/1) to give tert-butyl 4-(cyclopropanecarbonyl)-1,4-diazepane-1-carboxylate (380 mg, 95% yield) as a colorless oil. LCMS ESI m/z: 269.1 [M+H] + . Step 2 Cyclopropyl(1,4-diazepan-1-yl)methanone [0929] To a solution of tert-butyl 4-(cyclopropanecarbonyl)-1,4-diazepane-1-carboxylate (380 mg, 1.42 mmol) in dioxane (5 mL) was added HCl/Dioxane (4M in dioxane, 5 mL) and stirred at 25 °C for 1 hour. The mixture was concentrated in vacuo to afford cyclopropyl(1,4-diazepan-1-yl)methanone as a white solid (238 mg, 82% yield). This was used without further purification. LCMS ESI m/z: 169.1 [M+H] + . Step 3 6-Cyclobutoxy-4-(3-(4-(cyclopropanecarbonyl)-1,4-diazepane-1 -carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [0930] To a solution of cyclopropyl(1,4-diazepan-1-yl)methanone (50 mg, 0.30 mmol) and 5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoic acid (109 mg, 0.30 mmol) in DMF (2 mL) was added EDCI (85 mg, 0.45 mmol), HOBt (60 mg, 0.45 mmol) and DIPEA (115 mg, 0.90 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was poured into water (10 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 and concentrated in vacuo. The residue was purified by prep-HPLC to give 6-cyclobutoxy-4-(3- (4-(cyclopropanecarbonyl)-1,4-diazepane-1-carbonyl)-4-fluoro benzyl)phthalazin-1(2H)-one (43 mg, 28% yield) as a white solid. LCMS ESI m/z: 519.1 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.48 – 12.40 (1 H, m), 8.15 (1 H, d, J 8.8), 7.45 – 7.35 (1 H, m), 7.33 – 7.15 (3 H, m), 7.13 – 7.03 (1 H, m), 4.83 (1 H, dt, J 10.8, 7.0), 4.29 (2 H, t, J 6.1), 3.81 (1 H, t, J 10.9), 3.69 (1 H, d, J 5.4), 3.65 – 3.55 (2 H, m), 3.41 (2 H, d, J 5.9), 3.27 – 3.15 (2 H, m), 2.43 – 2.30 (2 H, m), 2.05 – 1.95 (2 H, m), 1.92 – 1.82 (1 H, m), 1.82 – 1.57 (3 H, m), 1.34 (1 H, t, J 37.5), 0.59 (4 H, dt, J 86.1, 5.5). Synthesis of Example 102: 6-Cyclobutoxy-4-(4-fluoro-3-(3-((5-(trifluoromethyl)pyrimidi n-2- yl)amino)azetidine-1-carbonyl)benzyl)phthalazin-1(2H)-one Step 1 tert-Butyl 3-((5-(trifluoromethyl)pyrimidin-2-yl)amino)azetidine-1-carb oxylate [0931] To a solution of 2-chloro-5-(trifluoromethyl)pyrimidine (300 mg, 1.64 mmol) and tert-butyl 3- aminoazetidine-1-carboxylate (283 mg, 1.64 mmol) in MeCN (3 mL) was added K 2 CO 3 (681 mg, 4.93 mmol). The reaction mixture was stirred at 60 o C for 16 hours, then quenched with water (10 mL) and extracted with EtOAc (10 mL). The organic phase was washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (Petroleum ether/EtOAc= 5/1) to give tert-butyl 3-((5-(trifluoromethyl)pyrimidin-2-yl)amino)azetidine-1-carb oxylate (520 mg, 99% yield) as a white solid. LCMS ESI m/z: 319.1 [M+H] + . Step 2 N-(Azetidin-3-yl)-5-(trifluoromethyl)pyrimidin-2-amine [0932] To a solution of tert-butyl (1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methy l)-2- fluorobenzoyl)azetidin-3-yl)(methyl)carbamate (222 mg, 0.41 mmol) in DCM (10 mL) was added TFA (1 mL) and stirred at 25 °C for 2 hours. The reaction mixture was concentrated in vacuo to afford 6- cyclobutoxy-4-(4-fluoro-3-(3-(methylamino)azetidine-1-carbon yl)benzyl)phthalazin-1(2H)-one (346 mg, 97% yield) as a colorless oil. This was used without further purification. LCMS ESI m/z: 219.1 [M+H] + . Step 3 6-Cyclobutoxy-4-(4-fluoro-3-(3-((5-(trifluoromethyl)pyrimidi n-2-yl)amino)azetidine-1- carbonyl)benzyl)phthalazin-1(2H)-one [0933] To a solution of N-(azetidin-3-yl)-5-(trifluoromethyl)pyrimidin-2-amine (50 mg, 0.23 mmol) and 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (50 mg, 0.23 mmol) in DMF (2 mL) was added EDCI (66 mg, 0.34 mmol), HOBt (46 mg, 0.34 mmol) and DIPEA (89 mg, 0.69 mmol). The reaction mixture was stirred at rt for 16 h. The mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by prep-HPLC to give 6-cyclobutoxy-4-(4-fluoro-3-(3-((5-(trifluoromethyl)pyrimidi n-2-yl)amino) azetidine-1- carbonyl)benzyl)phthalazin-1(2H)-one (88.7 mg, 68% yield) as a white solid. LCMS ESI m/z: 569.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.45 (1 H, s), 8.71 (1 H, d, J 6.1), 8.67 (1 H, s), 8.14 (1 H, d, J 8.8), 7.46 (2 H, dd, J 12.0, 4.9), 7.33 – 7.17 (2 H, m), 7.08 (1 H, d, J 1.8), 4.81 (1 H, dd, J 14.0, 7.1), 4.66 (1 H, d, J 5.8), 4.41 – 4.17 (4 H, m), 3.94 (2 H, ddd, J 13.9, 9.4, 5.2), 2.41 – 2.26 (2 H, m), 1.96 (2 H, d, J 9.8), 1.82 – 1.53 (2 H, m). Synthesis of Example 103: 6-Cyclobutoxy-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin -2- yl)octahydropyrrolo[3,2-b]pyrrole-1-carbonyl)benzyl)phthalaz in-1(2H)-one tert-Butyl 4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2-fluoro benzoyl)hexahydropyrrolo[3,2-b]pyrrole-1(2H)-carboxylate [0934] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (250 mg, 0.68 mmol) and tert-butyl hexahydropyrrolo[3,2-b]pyrrole-1(2H)-carboxylate (173 mg, 0.81 mmol) in DMF (2 mL) was added EDCI (195 mg, 1.02 mmol), HOBt (138 mg, 1.02 mmol) and DIPEA (439 mg, 3.40 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 40% to 50%) to give tert-butyl 4-(5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)hexahydropyrr olo[3,2-b]pyrrole-1(2H)-carboxylate (360 mg 94% yield) as a white solid; LCMS ESI m/z 563.3 [M+H] + . Step 2 6-Cyclobutoxy-4-(4-fluoro-3-(octahydropyrrolo[3,2-b]pyrrole- 1-carbonyl)benzyl)ph thalazin-1(2H)- one [0935] To a solution of tert-butyl 4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2- fluorobenzoyl)hexahydropyrrolo[3,2-b]pyrrole-1(2H)-carboxyla te (360 mg, 0.64 mmol) in dioxane (1 mL) was added HCl/dioxane (4.0 M, 2 mL, 8.00 mmol). The mixture was stirred at rt for 1 hour. The solution was concentrated in vacuo to give 6-cyclobutoxy-4-(4-fluoro-3-(octahydropyrrolo[3,2-b]pyrrole- 1-carbonyl)benzyl) phthalazin-1(2H)-one (310 mg 97% yield) as a white solid. This was used without further purification. LCMS ESI m/z 463.1 [M+H] + . Step 3 6-Cyclobutoxy-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin -2-yl)octahydropyrrolo[3,2-b]pyrrole- 1-carbonyl)benzyl)phthalazin-1(2H)-one [0936] A solution of 6-cyclobutoxy-4-(4-fluoro-3-(octahydropyrrolo[3,2-b]pyrrole- 1-carbony l)benzyl)phthalazin-1(2H)-one (150 mg, 0.30 mmol), 2-chloro-5-(trifluorometh yl)pyrimidine (55 mg, 0.30 mmol) and DIPEA (155mg, 1.20 mmol) in DMF (2 mL) was stirred at rt under Ar (g) for 12 hours, then purified by pre-HPLC to afford 6-cyclobutoxy-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrimidin -2- yl)octahydropyrrolo[3,2-b]pyrrole-1-carbonyl)benzyl)phthalaz in-1(2H)-one (56 mg 31% yield) as a colorless oil. LCMS ESI m/z 609.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.46 (s, 1H), 8.75 (s, 2H), 8.15 (m, 1H), 7.60 – 7.06 (m, 5H), 4.94 – 4.68 (m, 3H), 4.38 – 4.26 (m, 2H), 4.17 – 3.94 (m, 1H), 3.46 (m, 1H), 3.31 – 3.10 (m, 2H), 2.40 – 2.27 (m, 3H), 2.21 – 2.12 (m, 2H), 2.08 – 1.87 (m, 3H), 1.67 (m, 2H). Synthesis of Example 104: 6-((1-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2- fluorobenzoyl) azetidin-3-yl)amino)nicotinonitrile tert-Butyl 3-((5-Cyanopyridin-2-yl)amino)azetidine-1-carboxylate [0937] A mixture of 6-chloropyridine-3-carbonitrile (300 mg, 2.17 mmol), tert-butyl 3-aminoazetidine- 1-carboxylate (746 mg, 4.33 mmol) and K 2 CO 3 (748 mg, 5.41 mmol) in MeCN (10 mL) was stirred under reflux for 16 hours. The reaction was concentrated in vacuo, diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (PE/EA= 5/1) to give tert-butyl 3-((5-cyanopyridin-2-yl)amino)azetidine-1-carboxylate (384 mg, 51% yield) as a white solid. LCMS ESI m/z: 525 [M+H] + . Step 2 6-(Azetidin-3-ylamino)nicotinonitrile [0938] A solution of tert-butyl 3-((5-cyanopyridin-2-yl)amino)azetidine-1-carboxylate (384 mg, 1.12 mmol) in DCM (5 mL) and TFA (0.5 mL) was stirred at rt for 1 hour. The solvent was removed in vacuo to give 6-(azetidin-3-ylamino)nicotinonitrile (322 mg, 100% yield) as a pale yellow oil. LCMS ESI m/z: 175.1 [M+H] + . This was used without further purification. Step 3 6-((1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2-fluorobenzoyl) azetidin-3- yl)amino)nicotinonitrile [0939] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (80 mg, 0.22 mmol) and 6-(azetidin-3-ylamino)nicotinonitrile (270 mg, 0.44mmol) in DMF (2 mL) was added EDCI (62 mg, 0.33 mmol), HOBt (44 mg, 0.33 mmol) and DIPEA (140 mg, 1.09 mmol). The reaction mixture was stirred at rt for 1 hour. The reaction was purified by prep-HPLC to give 6-((1-(5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoyl)azetidin-3- yl)amino)nicotinonitrile (54 mg, 47% yield) as a white solid. LCMS ESI m/z: 525.0 [M+H] + . 1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.45 (s, 1H), 8.40 (d, J = 2.0 Hz, 1H), 8.23 (d, J = 6.0 Hz, 1H), 8.14 (d, J = 8.8 Hz, 1H), 7.75 (dd, J = 8.8, 2.2 Hz, 1H), 7.51 – 7.39 (m, 2H), 7.30 – 7.18 (m, 2H), 7.07 (d, J = 2.3 Hz, 1H), 6.56 (d, J = 8.8 Hz, 1H), 4.81 (p, J = 7.2 Hz, 1H), 4.62 (s, 1H), 4.40 – 4.20 (m, 4H), 3.89 (dd, J = 10.2, 5.1 Hz, 1H), 3.79 (dd, J = 8.9, 5.0 Hz, 1H), 2.40 – 2.28 (m, 2H), 1.97 (dd, J = 10.0, 7.8 Hz, 2H), 1.80 – 1.56 (m, 2H). Synthesis of Example 105: 5-((1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2- fluorobenzoyl)azetidin-3-yl)(methyl)amino)nicotinonitrile

tert-Butyl 3-((5-cyanopyridin-3-yl)(methyl)amino)azetidine-1-carboxylat e [0940] A mixture of 5-bromonicotinonitrile (300 mg, 1.64 mmol), tert-butyl 3-(methylamino)azetidine- 1-carboxylate (458 mg, 2.46 mmol), Pd 2 (dba) 3 (150 mg, 0.16 mmol), Xantphos (190 mg, 0.33 mmol) and t-BuONa (236 mg, 2.46 mmol) in toluene (5 mL) was stirred at 115 °C for 3 hours. The mixture was cooled to rt, diluted with H 2 O (5 mL), and extracted with EtOAc (10 mL x 3). The combined organic layer was washed by brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 20 to 35%) to give tert-butyl 3-((5-cyanopyridin-3- yl)(methyl)amino)azetidine-1-carboxylate as a yellow oil (244 mg, 51% yield). LCMS ESI m/z 289 [M+H] + . Step 2 5-(Azetidin-3-yl(methyl)amino)nicotinonitrile [0941] A solution of tert-butyl 3-((5-cyanopyridin-3-yl)(methyl)amino)azetidine-1-carboxylat e (244 mg, 0.85 mmol) in DCM (3 mL) and TFA (0.3 mL) was stirred at rt for 1 hour. The reaction was concentrated under vacuum to afford 5-(azetidin-3-yl(methyl)amino)nicotinonitrile as a yellow oil (155 mg, 97% yield). This was used without further purification. LCMS ESI m/z 189 [M+H] + . Step 3 5-((1-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2-fluorobenzoyl)azetidin-3- yl)(methyl)amino)nicotinonitrile [0942] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (60 mg, 0.16 mmol) and 5-(azetidin-3-yl(methyl)amino)nicotinonitrile (67 mg, 0.36 mmol) in DMF (2 mL) was added EDCI (47 mg, 0.24 mmol), HOBt (33 mg, 0.24 mmol) and DIPEA (63 mg, 0.49 mmol). The reaction mixture was stirred at rt for 24 hours, then purified by prep-HPLC to give 5-((1-(5- ((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-f luorobenzoyl)azetidin-3-yl)(methyl)amino) nicotinonitrile as a white solid (36 mg, 41% yield). LCMS ESI m/z 539 [M+H] + . 1 H-NMR (400 MHz, DMSO–d 6 ) δ 12.47 (s, 1H), 8.42 (d, J = 3.0 Hz, 1H), 8.32 (d, J = 1.5 Hz, 1H), 8.14 (d, J = 8.8 Hz, 1H), 7.62 (dd, J = 2.9, 1.7 Hz, 1H), 7.51 (dd, J = 6.5, 2.1 Hz, 1H), 7.48 – 7.43 (m, 1H), 7.31 – 7.21 (m, 2H), 7.10 (d, J = 2.3 Hz, 1H), 4.89 – 4.72 (m, 2H), 4.49 – 4.19 (m, 4H), 4.05-3.98 (m, 2H), 2.98 (s, 3H), 2.43 – 2.30 (m, 2H), 2.06 – 1.90 (m, 2H), 1.83 – 1.56 (m, 2H). Synthesis of Example 106: 2-(1-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)azetidin-3-ylamino)isonicotinonitrile

tert-Butyl 3-(4-cyanopyridin-2-ylamino)azetidine-1-carboxylate [0943] tert-butyl 3-aminoazetidine-1-carboxylate (2.1 g, 12.19 mmol) , 2-chloropyridine-4-carbonitrile (2.53 g, 18.29 mmol) , potassium carbonate (3.37 g, 24.39 mmol, 1.47 mL) and DMF (20 mL) were added to a 100 mL bottled flask. The resultant mixture was stirred at 80 °C for 12 hr. The mixture was diluted with H 2 O (20 mL) and extracted with EA (40 mL X 2). The combined organic phases were washed with brine (30 mL), dried over Na 2 SO 4 and evaporated, and the residue was purified by silica gel chromatography to give tert-butyl 3-(4-cyanopyridin-2-ylamino)azetidine-1-carboxylate (360 mg, 10.8% yield) as a yellow solid. LCMS ESI m/z: 275.1 [M+H] + . Step 2 2-(Azetidin-3-ylamino)isonicotinonitrile [0944] tert-Butyl 3-[(4-cyano-2-pyridyl)amino]azetidine-1-carboxylate (360 mg, 1.31 mmol), TFA (2.96 g, 25.96 mmol, 2 mL) and DCM (4 mL) were added to a 50 mL bottled flask. The resultant mixture was stirred at rt for 2 hr. The solvent removed in vacuum to give 2-(azetidin-3-ylamino)pyridine-4- carbonitrile (600 mg, crude), which was used to next step without futher purification. LCMS ESI m/z: 175.2 [M+H] + . Step 3 2-(1-(2-Fluoro-5-formylbenzoyl)azetidin-3-ylamino)isonicotin onitrile [0945] 2-(azetidin-3-ylamino)pyridine-4-carbonitrile (540.03 mg, 1.87 mmol) was added to a solution of 2-fluoro-5-formyl-benzoic acid (210 mg, 1.25 mmol) , HATU (716.19 mg, 1.88 mmol), DIPEA (645.75 mg, 5.00 mmol, 870.28 μL) and DMF (10 mL) . The resultant mixture was stirred at rt for 12 hr . The mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL X 2). The combined organic phases were washed with brine (20 mL), dried over Na 2 SO 4 and concentrated. The residue was purified by Prep-HPLC to give 2-(1-(2-fluoro-5-formylbenzoyl)azetidin-3-ylamino)isonicotin onitrile (350 mg, 86% yield) as a yellow solid. LCMS ESI m/z: 325.1 [M+H] + . Step 4 2-(1-(5-((6-Cyclobutoxy-3-oxoisobenzofuran-1(3H)-ylidene)met hyl)-2-fluorobenzoyl)azetidin-3- ylamino)isonicotinonitrile [0946] To a mixture of 5-(cyclobutoxy)-3-dimethoxyphosphoryl-3H-isobenzofuran-1-one (70 mg, 224.18 μmol) in THF (10 mL) was added 2-[[1-(2-fluoro-5-formyl-benzoyl)azetidin-3- yl]amino]pyridine-4-carbonitrile (109.05 mg, 336.26 μmol) and ET 3 N (68.05 mg, 672.53 μmol, 93.74 μL). The resulting mixture was stirred at rt for 24 hr. The mixture was quenched with sat. NaHSO 3 solution (5 mL), and filtered. The filter cake was washed with H 2 O (10 mL) and dried under reduced pressure to give 2-(1-(5-((6-cyclobutoxy-3-oxoisobenzofuran-1(3H)-ylidene)met hyl)-2- fluorobenzoyl)azetidin-3-ylamino)isonicotinonitrile (70 mg, crude) as a yellow solid. LCMS ESI m/z: 510.7 [M+H] + . Step 5 2-(1-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)azetidin-3- ylamino)isonicotinonitrile [0947] To a mixture of 2-[[1-[5-[(E)-[6-(cyclobutoxy)-3-oxo-isobenzofuran-1-ylidene ]methyl]-2- fluoro-benzoyl]azetidin-3-yl]amino]pyridine-4-carbonitrile (70 mg, 137.12 μmol) in THF (5 mL) was added N O (8.58 mg, 137.12 μmol, 8.35 μL) . The resulting mixture was stirred at 70 °C for 4 hr. The mixture was concentrated under reduced pressure to give the crude product, which was purified by Pre-HPLC to afford 2-[[1-[5-[[7-(cyclobutoxy)-4-oxo-3H-phthalazin-1-yl]methyl]- 2-fluoro- benzoyl]azetidin-3-yl]amino]pyridine-4-carbonitrile (17.2 mg, 23.9% yield) as a white solid. LCMS ESI m/z: 525.2 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.44 (s, 1H), 8.15 (dd, J = 11.5, 7.0 Hz, 2H), 7.78 (d, J = 6.0 Hz, 1H), 7.49 – 7.41 (m, 2H), 7.26 (ddd, J = 18.4, 9.3, 5.4 Hz, 2H), 7.08 (d, J = 2.3 Hz, 1H), 6.89 (dd, J = 5.2, 1.3 Hz, 1H), 6.82 (s, 1H), 4.85 – 4.78 (m, 1H), 4.59 – 4.52 (m, 1H), 4.37 – 4.23 (m, 4H), 3.89 (dd, J = 10.2, 5.1 Hz, 1H), 3.76 (dd, J = 8.8, 5.0 Hz, 1H), 2.40 – 2.31 (m, 2H), 1.97 (dd, J = 9.7, 5.0 Hz, 2H), 1.79 – 1.60 (m, 2H). Synthesis of Example 107: 6-Cyclobutoxy-4-(3-(8-(cyclopropanecarbonyl)-3,8- diazabicyclo[3.2.1]octane-3-carbonyl)-4-fluorobenzyl)phthala zin-1(2H)-one 6-Cyclobutoxy-4-(3-(8-(cyclopropanecarbonyl)-3,8-diazabicycl o[3.2.1]octane-3-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [0948] To a solution of 4-(3-(3,8-diazabicyclo[3.2.1]octane-3-carbonyl)-4-fluorobenz yl)-6- cyclobutoxyphthalazin-1(2H)-one (80 mg, 0.17 mmol) and cyclopropanecarboxylic acid (22 mg, 0.26 mmol) in DMF (2 mL) was added EDCI (50 mg, 0.26 mmol), HOBt (35 mg, 0.26 mmol) and DIPEA (67 mg, 0.51 mmol). The reaction mixture was stirred at 50 °C for 1 hour. The reaction was purified by prep- HPLC to give 6-cyclobutoxy-4-(3-(8-(cyclopropanecarbonyl)-3,8-diazabicycl o[3.2.1]octane-3-carbonyl)- 4-fluorobenzyl) phthalazin-1(2H)-one (35 mg, 38% yield) as a white solid. LCMS ESI m/z: 531 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.45 (s, 1H), 8.18 – 8.10 (m, 1H), 7.46 – 7.34 (m, 2H), 7.32 – 7.28 (m, 1H), 7.26 – 7.19 (m, 1H), 7.13 – 7.03 (m, 1H), 4.96 – 4.64 (m, 2H), 4.59 – 4.46 (m, 1H), 4.35 – 4.26 (m, 3H), 3.22 – 3.04 (m, 2H), 3.00 – 2.81 (m, 1H), 2.39 (s, 2H), 2.09 – 1.90 (m, 4H), 1.84 – 1.74 (m, 2H), 1.71 – 1.62 (m, 2H), 0.82 – 0.60 (m, 5H). Synthesis of Example 108: 2-((1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2- fluorobenzoyl) azetidin-3-yl)(methyl)amino)-N-methylpyrimidine-4-carboxamid e 2-Chloro-N-methylpyrimidine-4-carboxamide [0949] A solution of 2-chloropyrimidine-4-carboxylic acid (500 mg, 3.15 mmol), DMF (23 mg, 0.32 mmol) in DCM (10 mL) was added oxalyl dichloride (400 mg, 3.15 mmol) dropwise at 0 °C. The reaction was stirred at rt for 1 hour, then concentrated in vacuo. The residue was dissolved in DCM (10 mL), then added a solution of methanamine hydrochloride (426 mg, 6.30 mmol) and DIPEA (1.22 g, 9.46 mmol) in DCM (10 mL). The mixture was stirred at rt for 1 hour, then diluted with H 2 O (30 mL) and extracted with DCM (30 mL × 3). The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromategraphy (EtOAc/petroleum ether = 0% to 30%) to give methyl 2,4-difluoro-5-vinyl-benzoate (310 mg, 57% yield) as a white solid. LCMS ESI m/z: 172 [M+H] + . Step 2 tert-Butyl (1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methy l)-2-fluorobenzoyl)azetidin- 3-yl)(methyl)carbamate [0950] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (120 mg, 0.33 mmol) and tert-butyl azetidin-3-yl(methyl)carbamate (73 mg, 0.39 mmol) in DMF (2 mL) was added EDCI (62 mg, 0.33 mmol), HOBt (66 mg, 0.49 mmol) and DIPEA (126 mg, 0.98 mmol). The reaction mixture was stirred at 45 °C for 1 hour. The mixture was diluted with H 2 O (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromategraphy (EtOAc/pet ether = 0% to 50%) to give tert-butyl (1-(5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)azetidin-3-yl )(methyl)carbamate (100 mg, 57% yield) as a white solid. LCMS ESI m/z: 537 [M+H] + . Step 3 6-Cyclobutoxy-4-(4-fluoro-3-(3-(methylamino)azetidine-1-carb onyl) benzyl)phthalazin-1(2H)-one [0951] A solution of tert-butyl (1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methy l)-2- fluorobenzoyl)azetidin-3-yl)(methyl)carbamate (100 mg, 0.19 mmol) in DCM (5 mL) and TFA (0.5 mL) was stirred at rt for 1 hour. The solvent was removed in vacuo to give 6-cyclobutoxy-4-(4-fluoro-3-(3- (methylamino)azetidine-1-carbonyl)benzyl)phthalazin-1(2H)-on e (80 mg, 78% yield) as a yellow oil. This was used without further purification. LCMS ESI m/z: 437 [M+H] + . Step 4 2-((1-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2-fluorobenzoyl) azetidin-3- yl)(methyl)amino)-N-methylpyrimidine-4-carboxamide [0952] A solution of 6-cyclobutoxy-4-(4-fluoro-3-(3-(methylamino)azetidine-1- carbonyl)benzyl)phthalazin-1(2H)-one (60 mg, 0.14 mmol), 2-chloro-N-methylpyrimidine-4-carboxamide (71 mg, 0.41 mmol) and DIPEA (53 mg, 0.41 mmol) in DMF (1 mL) was stirred at 90 °C for 2 hours. The mixture was concentrated in vacuo and the residue was purified by prep-HPLC to give 2-((1-(5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoyl)azetidin-3-yl)(methyl)amino)-N- methylpyrimidine-4-carboxamide (36 mg, 45% yield) as a white solid. LCMS ESI m/z: 572 [M+H] + . 1 H– NMR (400 MHz, DMSO-d 6 ) δ 12.47 (s, 1H), 8.76 – 8.70 (m, 1H), 8.58 (d, J = 4.8 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.54 – 7.50 (m, 1H), 7.48 – 7.43 (m, 1H), 7.30 – 7.23 (m, 2H), 7.16 (d, J = 4.8 Hz, 1H), 7.12 – 7.10 (m, 1H), 5.74 (s, 1H), 4.86 – 4.78 (m, 1H), 4.37 – 4.30 (m, 3H), 4.23 – 4.14 (m, 3H), 3.21 (s, 3H), 2.81 (d, J = 4.8 Hz, 3H), 2.39 – 2.32 (m, 2H), 2.02 – 1.92 (m, 2H), 1.78 – 1.59 (m, 2H). Synthesis of Example 109: 6-Cyclobutoxy-4-(3-(4-(cyclopropanecarbonyl)octahydropyrrolo [3,2- b]pyrrole-1-carbonyl)-4-fluorobenzyl)phthalazin-1(2H)-one 6-cyclobutoxy-4-(3-(4-(cyclopropanecarbonyl)octahydropyrrolo [3,2-b]pyrrole-1-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [0953] To a solution of 6-cyclobutoxy-4-(4-fluoro-3-(octahydropyrrolo[3,2-b]pyrrole- 1- carbonyl)benzyl)phthalazin-1(2H)-one (150 mg, 0.30 mmol) and cyclopropanecarboxylic acid (26 mg, 0.30 mmol) in DMF (5 mL) was added EDCI (86 mg, 0.45 mmol), HOBt (61 mg, 0.45 mmol) and DIPEA (155 mg, 1.2 mmol). The mixture was stirred at rt for 16 hours. The reaction was purified by prep- HPLC to give 6-cyclobutoxy-4-(3-(4-(cyclopropanecarbonyl)octahydropyrrolo [3,2-b]pyrrole-1-carbonyl)- 4-fluorobenzyl)phthalazin-1(2H)-one (56 mg 38% yield) as a white solid. LCMS ESI m/z 531.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.46 (s, 1H), 8.15 (m, 1H), 7.57 – 7.17 (m, 4H), 7.11 (m, 1H), 4.91 – 4.66 (m, 2H), 4.50 (m, 1H), 4.30 (s, 2H), 3.90 (m, 1H), 3.48 (m, 1H), 3.26 – 3.02 (m, 2H), 2.37 (t, J = 13.3 Hz, 2H), 2.26 – 2.07 (m, 2H), 2.00 (d, J = 5.5 Hz, 3H), 1.92 – 1.56 (m, 4H), 0.87 – 0.65 (m, 4H). Synthesis of Example 110: 6-Cyclobutoxy-4-(3-(3-((1,5-dimethyl-1H-imidazol-2- yl)(methyl)amino)azetidine-1-carbonyl)-4-fluorobenzyl)phthal azin-1(2H)-one Step 1 tert-Butyl 3-((1,5-dimethyl-1H-imidazol-2-yl)(methyl)amino)azetidine-1- carboxylate [0954] A mixture of 2-bromo-1,5-dimethyl-1H-imidazole (180 mg, 1.03 mmol), tert-butyl 3- (methylamino)azetidine-1-carboxylate (2.87 g, 15.43 mmol), brettphos Pd G3 (93 mg, 0.10 mmol), Xphos (196 mg, 0.41 mmol) and t-BuONa (197 mg, 2.06 mmol) in dioxane (3 mL) was was stirred at 110 °C under Ar (g) for 16 hours. The mixture was cooled to rt, diluted with H2O (10 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 30 to 50%) to give tert-butyl 3-((1,5-dimethyl-1H-imidazol-2- yl)(methyl)amino)azetidine-1-carboxylate as a white solid (80 mg, 27% yield). LCMS ESI m/z: 281 [M+H] + . Step 2 N-(Azetidin-3-yl)-N-1,5-trimethyl-1H-imidazol-2-amine [0955] To a solution of tert-butyl 3-((1,5-dimethyl-1H-imidazol-2-yl)(methyl)amino)azetidine-1- carboxylate (80 mg, 0.28 mmol) in DCM (1 mL) was added TFA (472 mg, 0.414 mmol). The mixture was stirred at rt for 1 hour. Solvent was removed in vacuo to afford N-(azetidin-3-yl)-N-1,5-trimethyl-1H- imidazol-2-amine as a white solid (72 mg, 96% yield). LCMS ESI m/z: 181 [M+H] + . This was used without further purification. Step 3 6-Cyclobutoxy-4-(3-(3-((1,5-dimethyl-1H-imidazol-2-yl)(methy l)amino)azetidine-1-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [0956] To a solution of N-(azetidin-3-yl)-N-1,5-trimethyl-1H-imidazol-2-amine (47 mg, 0.26 mmol) and 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (80 mg, 0.22 mmol) in DMF (2 mL) was added EDCI (63 mg, 0.33 mmol), HOBt (44 mg, 0.33 mmol) and DIPEA (84 mg, 0.65 mmol). The reaction mixture was stirred at rt for 1 hour. The reaction was purified by prep- HPLC to give 6-cyclobutoxy-4-(3-(3-((1,5-dimethyl-1H-imidazol-2-yl)(methy l)amino)azetidine-1- carbonyl)-4-fluorobenzyl) phthalazin-1(2H)-one (31 mg, 26% yield) as a white solid. LCMS ESI m/z: 531 [M+H] + . 1 H–NMR (400 MHz, DMSO-D6) δ 12.46 (s, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.48 – 7.38 (m, 2H), 7.30 (dd, J = 8.8, 2.3 Hz, 1H), 7.25 – 7.16 (m, 1H), 7.07 (d, J = 2.3 Hz, 1H), 6.33 (s, 1H), 4.90 – 4.77 (m, 1H), 4.37 – 4.24 (m, 2H), 4.17 – 3.95 (m, 3H), 3.82 – 3.63 (m, 2H), 3.32 (s, 3H), 2.52 (s, 3H), 2.42 – 2.30 (m, 2H), 2.08 (s, 3H), 2.05 – 1.90 (m, 2H), 1.85 – 1.55 (m, 2H). Synthesis of Example 111: 6-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)hexahydropyrrolo[3,2-b]pyrrol-1(2H)-yl)nicotin onitrile 6-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)hexahydropyrrolo[3,2-b]pyrrol-1(2H)-yl)nicotin onitrile [0957] A mixture of 6-cyclobutoxy-4-(4-fluoro-3-(octahydropyrrolo[3,2-b]pyrrole- 1- carbonyl)benzyl)phthalazin-1(2H)-one (150 mg, 0.30 mmol), 6-chloronicotinonitrile (42 mg, 0.30 mmol) and DIPEA (155 mg, 1.20 mmol) in DMF (2 mL) was stirred at 50 °C for 1 hour. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by prep-HPLC to give 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)hexahydropyrrolo[3,2-b]pyrrol-1(2H)-yl)nicotin onitrile (32 mg, 18% yield) as a white solid. LCMS ESI m/z: 565 [M+H] + . 1 H–NMR (400 MHz, DMSO-d 6 ) δ 12.46 (s, 1H), 8.52 (d, J = 2.0 Hz, 1H), 8.15 (d, J = 3.9 Hz, 1H), 7.87 (dd, J = 9.0, 2.3 Hz, 1H), 7.54 – 7.34 (m, 2H), 7.29 (dd, J = 8.8, 2.3 Hz, 1H), 7.22 (t, J = 9.1 Hz, 1H), 7.11 (d, J = 2.3 Hz, 1H), 6.69 (d, J = 9.0 Hz, 1H), 4.90 – 4.55 (m, 3H), 4.29 (s, 2H), 3.93 – 3.76 (m, 1H), 3.45 – 3.37 (m, 1H), 3.27 – 3.18 (m, 2H), 2.44 – 2.31 (m, 2H), 2.31 – 2.11 (m, 2H), 2.10 – 1.86 (m, 4H), 1.82 – 1.70 (m, 1H), 1.69 – 1.55 (m, 1H). Synthesis of Example 112: 6-Cyclobutoxy-4-(3-(3-(cyclopropanecarbonyl)-3,8- diazabicyclo[3.2.1]octane-8-carbonyl)-4-fluorobenzyl)phthala zin-1(2H)-one tert-Butyl 3-(cyclopropanecarbonyl)-3,8-diazabicyclo[3.2.1]octane-8-car boxylate [0958] A mixture of tert-butyl 3,8-diazabicyclo[3.2.1]octane-8-carboxylate (110 mg, 0.52 mmol) and cyclopropanecarboxylic acid (54 mg, 0.62 mmol) in DMF (2 mL) was added EDCI (149 mg, 0.78 mmol), HOBT (105 mg, 0.78 mmol) and DIPEA (201 mg, 1.55 mmol). The reaction mixture was stirred at rt for 16 hours, then quenched with water (10 mL) and extracted with DCM (50 mL). The organic phase was dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (PE/EA= 2/1) to give 6-(4-(3-formylbenzoyl)piperazin-1-yl)nicotinonitrile (141 mg, 0.45 mmol, 97% yield) as a colorless oil. LCMS ESI m/z: 281.0 [M+H] + . Step 2 (3,8-Diazabicyclo[3.2.1]octan-3-yl)(cyclopropyl)methanone [0959] To a solution of tert-butyl 3-(cyclopropanecarbonyl)-3,8-diazabicyclo[3.2.1]octane-8- carboxylate (141 mg, 0.50 mmol) in dioxane (5 mL) was added HCl/dioxane (5 mL) and stirred at 25 °C for 1 hour. The mixture was concentrated in vacuo to afford (3,8-diazabicyclo[3.2.1]octan-3- yl)(cyclopropyl)methanone (90 mg, 0.42 mmol, 83% yield) as a white solid. This was used without further purification. LCMS ESI m/z: 181.0 [M+H] + . Step 3 6-Cyclobutoxy-4-(3-(3-(cyclopropanecarbonyl)-3,8-diazabicycl o[3.2.1]octane-8-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [0960] To a solution of (3,8-diazabicyclo[3.2.1]octan-3-yl)(cyclopropyl)methanone (50 mg, 0.28 mmol) and 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (102 mg, 0.28 mmol) in DMF (2 mL) was added EDCI (80 mg, 0.42 mmol), HOBt (56 mg, 0.42 mmol) and DIPEA (108 mg, 0.83 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was poured into water (10 mL) and extracted with ethyl acetate (20 mL x3). The combined organic layer was washed with brine, dried over Na 2 SO 4 and concentrated in vacuo. The residue was purified by prep-HPLC to give 6- cyclobutoxy-4-(3-(3-(cyclopropanecarbonyl)-3,8-diazabicyclo[ 3.2.1]octane-8-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one (70 mg, 0.13 mmol, 48% yield) as a white solid. LCMS ESI m/z: 531.2 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.45 (s, 1H), 8.14 (d, J = 5.8 Hz, 1H), 7.43 (s, 2H), 7.28 (dd, J = 16.1, 8.6 Hz, 2H), 7.10 (s, 1H), 4.89 – 4.79 (m, 1H), 4.69 (s, 1H), 4.31 (s, 2H), 4.25 – 3.90 (m, 2H), 3.72 (s, 1H), 3.15 – 2.75 (m, 2H), 2.50 (dt, J = 3.5, 1.7 Hz, 4H), 2.39 (d, J = 6.5 Hz, 2H), 2.05 – 1.90 (m, 2H), 1.87 – 1.71 (m, 2H), 1.65 (dd, J = 18.3, 8.9 Hz, 1H), 1.56 – 1.42 (m, 1H), 0.74 (s, 4H). Synthesis of Example 113: N-(1-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)azetidin-3-yl) tert-butyl (1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methy l)-2-fluoro benzoyl)azetidin- 3-yl)carbamate [0961] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluo robenzoic acid (200 mg, 0.54 mmol) and tert-butyl azetidin-3-ylcarbamate (94 mg, 0.54 mmol) in DMF (2 mL) was added EDCI (109 mg, 0.54 mmol), HOBt (110 mg, 0.81 mmol) and DIPEA (351 mg, 2.70 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 40% to 45%) to give tert-butyl (1-(5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)azetidin-3-yl )carbamate (263 mg 93% yield) as a white solid. LCMS ESI 491.2 [M+H] + . Step 2 4-(3-(3-aminoazetidine-1-carbonyl)-4-fluorobenzyl)-6-cyclobu toxyphth alazin-1(2H)-one [0962] To a solution of tert-butyl (1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methy l)-2- fluorobenzoyl)azetidin-3-yl)carbamate (260 mg, 0.50 mmol) in DCM (5 mL) was added TFA (0.5 mL). The mixture was stirred at rt for 1 hour. The solution was concentrated in vacuo to give 4-(3-(3- aminoazetidine-1-carbonyl)-4-fluorobenzyl)-6-cyclo butoxyphthalazin-1(2H)-one (200 mg 95% yield) as a colorless oil. This was used without further purification. LCMS ESI 423 [M+H] + . Step 3 N-(1-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenz oyl)azetidin-3- yl)cyclopropanecarboxamide [0963] To a solution of 4-(3-(3-aminoazetidine-1-carbonyl)-4-fluorobenzyl)-6-cyclobu toxyphthalazin- 1(2H)-one (100 mg, 0.19 mmol) and cyclopropanecarboxylic acid (16 mg, 0.19 mmol) in DMF (5 mL) was added EDCI (54 mg, 0.28 mmol), HOBt (38 mg, 0.28 mmol) and DIPEA (96 mg, 0.75 mmol). The mixture was stirred at rt for 16 hours. The reaction was purified by prep-HPLC to give N-(1-(5-((7- cyclobutoxy-4-oxo-3,4-dihyd rophthalazin-1-yl)methyl)-2-fluorobenzoyl)azetidin-3- yl)cyclopropanecarboxamide (64 mg 95% yield) as a white solid. LCMS ESI 491.2 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.47 (s, 1H), 8.75 (m, 1H), 8.15 (m, 1H), 7.52 – 7.37 (m, 2H), 7.33 – 7.17 (m, 2H), 7.09 (m, 1H), 4.84 (m, 1H), 4.51 (m 1H), 4.34 – 4.10 (m, 4H), 3.89 – 3.73 (m, 2H), 2.37 (d, J = 3.4 Hz, 2H), 2.00 (m, 2H), 1.82 – 1.59 (m, 2H), 1.49 (m 1H), 0.73 – 0.63 (m, 4H). Synthesis of Example 114: N-(1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)azetidin-3-yl)-4,4,4-trifluoro-N-methylbutanam ide

N-(1-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)azetidin-3-yl)- 4,4,4-trifluoro-N-methylbutanamide [0964] To a solution of 6-cyclobutoxy-4-(4-fluoro-3-(3-(methylamino)azetidine-1- carbonyl)benzyl)phthalazin-1(2H)-one (75 mg, 0.17 mmol) and 4,4,4-trifluorobutanoic acid (49 mg, 0.34 mmol) in DMF (2 mL) was added EDCI (49 mg, 0.26 mmol), HOBt (35 mg, 0.26 mmol) and DIPEA (111 mg, 0.86 mmol). The reaction mixture was stirred at 45 °C for 2 hours. The mixture was concentrated in vacuo and the residue was purified by prep-HPLC to afford N-(1-(5-((7-cyclobutoxy-4- oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)azeti din-3-yl)-4,4,4-trifluoro-N- methylbutanamide as a white solid (50 mg, 52% yield). LCMS ESI m/z 561 (M+H) + . 1 H–NMR (400 MHz, DMSO-d 6 ) δ 12.46 (s, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.51 – 7.47 (m, 1H), 7.47 – 7.42 (m, 1H), 7.32 – 7.28 (m, 1H), 7.27 – 7.20 (m, 1H), 7.12 – 7.09 (m, 1H), 5.17 – 4.94 (m, 1H), 4.88 – 4.80 (m, 1H), 4.31 (s, 2H), 4.21 – 3.99 (m, 4H), 3.01 (s, 2H), 2.91 (s, 1H), 2.63 – 2.56 (m, 2H), 2.47 – 2.32 (m, 4H), 2.03 – 1.94 (m, 2H), 1.83 – 1.74 (m, 1H), 1.70 – 1.61 (m, 1H). Synthesis of Example 115: 6-(5-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl) hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)nicotinonitrile

tert-Butyl 5-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2-fluo robenzoyl)hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate [0965] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (300 mg, 0.81 mmol) and tert-butyl hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (207 mg, 0.98 mmol) in DMF (2 mL) was added EDCI (234 mg, 1.22 mmol), HOBt (165 mg, 1.22 mmol) and DIPEA (526 mg, 4.07 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 40% to 50%) to give tert-butyl 5-(5-((7-cyclobutoxy-4- oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)hexa hydropyrrolo[3,4-c]pyrrole-2(1H)- carboxylate (436 mg 95% yield) as a white solid. LCMS ESI 563 [M+H] + . Step 2 6-Cyclobutoxy-4-(4-fluoro-3-(octahydropyrrolo[3,4-c]pyrrole- 2-carbo nyl)benzyl)phthalazin-1(2H)- one [0966] To a solution of tert-butyl 5-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2- fluorobenzoyl)hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxyla te (436 mg, 0.75 mmol) ) in dioxane (2 mL) was added HCl/dioxane (4.0M, 4 mL, 16.00 mmol). The mixture was stirred at rt for 1 hour. The solvent was removed in vacuo to give 6-cyclobutoxy-4-(4-fluoro-3-(octahydropyrrolo[3,4-c]pyrrole- 2- carbonyl)be nzyl)phthalazin-1(2H)-one (322 mg 90% yield) as a white solid. This was used without further purification. LCMS ESI m/z 463 [M+H] + . Step 3 6-(5-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl) hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)nicotinonitrile [0967] To a solution of 6-cyclobutoxy-4-(4-fluoro-3-(octahydropyrrolo[3,4-c]pyrrole- 2- carbonyl)benzyl)phthalazin-1(2H)-one (60 mg, 0.13 mmol) and 6-chloronicotinonitrile (54 mg, 0.39 mmol) in DMF (1 mL) was added DIPEA (84 mg, 0.65 mmol). The reaction mixture was stirred at 90 °C for 2 hours. The mixture was concentrated in vacuo and the residue was purified by prep-HPLC to give 6- (5-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methy l)-2-fluorobenzoyl)hexahydropyrrolo[3,4- c]pyrrol-2(1H)-yl)nicotinonitrile (35 mg, 47% yield) as a white solid. LCMS ESI m/z: 565 [M+H] + . 1 H– NMR (400 MHz, DMSO-d 6 ) δ 12.45 (s, 1H), 8.49 – 8.45 (m, 1H), 8.14 (d, J = 8.8 Hz, 1H), 7.83 (dd, J = 8.9, 2.3 Hz, 1H), 7.41 – 7.36 (m, 2H), 7.31 – 7.28 (m, 1H), 7.25 – 7.19 (m, 1H), 7.08 – 7.06 (m, 1H), 6.53 (d, J = 9.0 Hz, 1H), 4.86 – 4.78 (m, 1H), 4.29 (s, 2H), 3.81 – 3.70 (m, 2H), 3.67 – 3.58 (m, 1H), 3.51 – 3.40 (m, 3H), 3.30 – 3.22 (m, 1H), 3.13 – 3.06 (m 2H) 304 – 297 (m, 1H), 2.41 – 2.34 (m, 2H), 2.03 – 1.95 (m, 2H), 1.82 – 1.73 (m, 1H), 1.69 – 1.61 (m, 1H). Synthesis of Example 116: 6-(3-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-3,8-diazabicyclo[3.2.1]octan-8-yl)nicotinonit rile tert-Butyl (1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methy l)-2-fluorobenzoyl)azetidin- 3-yl)(methyl)carbamate [0968] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (250 mg, 0.68 mmol) and tert-butyl azetidin-3-yl(methyl)carbamate (139 mg, 0.75 mmol) in DMF (5 mL) was added EDCI (195 mg, 1.02 mmol), HOBt (138 mg, 1.02 mmol) and DIPEA (263 mg, 2.04 mmol). The reaction mixture was stirred at rt for 16 hours, then the reaction was quenched with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The organic phase was washed with sat. NaCl solution, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The crude product was purified by silica gel chromatography (Petroleum ether/EtOAc = 2/1) to give 6-(4-(3-formylbenzoyl)piperazin-1- yl)nicotinonitrile (242 mg, 66% yield) as a white solid. LCMS ESI m/z: 537.1 [M+H] + . Step 2 6-Cyclobutoxy-4-(4-fluoro-3-(3-(methylamino)azetidine-1-carb onyl)benzyl)phthalazin-1(2H)-one [0969] To a solution of tert-butyl (1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methy l)-2- fluorobenzoyl)azetidin-3-yl)(methyl)carbamate (222 mg, 0.41 mmol) in DCM (10 mL) was added TFA (1 mL) and stirred at 25°C for 2 hours. The reaction mixture was concentrated in vacuo to afford 6- cyclobutoxy-4-(4-fluoro-3-(3-(methylamino)azetidine-1-carbon yl)benzyl)phthalazin-1(2H)-one (180 mg, 0.41 mmol, 99% yield) as a yellow oil. This was used without further purification. LCMS ESI m/z: 437.1 [M+H] + . Step 3 6-((1-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2-fluorobenzoyl) azetidin-3- yl)(methyl)amino)pyrazine-2-carbonitrile [0970] To a solution of 6-cyclobutoxy-4-(4-fluoro-3-(3-(methylamino)azetidine-1- carbonyl)benzyl)phthalazin-1(2H)-one (100 mg, 0.23 mmol) and 6-chloropyrazine-2-carbonitrile (96 mg, 0.69 mmol) in DMF (1 mL) was added DIPEA (81 mg, 0.63 mmol). The reaction mixture was stirred at 90 °C for 2 hours. The reaction was purified by prep-HPLC to give 6-((1-(5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)azetidin-3-yl )(methyl)amino)pyrazine-2-carbonitrile (54.5 mg, 54% yield) as a white solid. LCMS ESI m/z: 540.1 [M+H] + . 1 H–NMR (400 MHz, DMSO-d6) δ 12.46 (s, 1H), 8.53 (s, 1H), 8.35 (s, 1H), 8.14 (d, J = 8.8 Hz, 1H), 7.53 (dd, J = 6.4, 2.1 Hz, 1H), 7.47 – 7.41 (m, 1H), 7.32 – 7.20 (m, 2H), 7.09 (d, J = 2.2 Hz, 1H), 5.33 – 5.14 (m, 1H), 4.82 (p, J = 7.1 Hz, 1H), 4.37 – 4.27 (m, 3H), 4.24 – 4.06 (m, 3H), 3.13 (s, 3H), 2.43 – 2.27 (m, 2H), 2.04 – 1.90 (m, 2H), 1.74 (dt, J = 18.1, 9.1 Hz, 1H), 1.62 (dt, J = 18.4, 9.3 Hz, 1H). Synthesis of Example 117: N-(1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)azetidin-3-yl)-N-methylazetidine-2-carboxamide tert-butyl 2-((1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2- fluorobenzoyl)azetidin-3-yl)(methyl)carbamoyl)azetidine-1-ca rboxylate [0971] To a solution of 6-cyclobutoxy-4-(4-fluoro-3-(3-(methylamino)azetidine-1- carbonyl)benzyl)phthalazin-1(2H)-one (89 mg, 0.20 mmol) and 1-(tert-butoxycarbonyl)azetidine-2- carboxylic acid (50 mg, 0.25 mmol) in DMF (3 mL) was added EDCI (39 mg, 0.20 mmol), HOBt (42 mg, 0.30 mmol) and DIPEA (79 mg, 0.61 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na SO filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 30 to 50%) to give tert-butyl 2-((1- (5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)- 2-fluorobenzoyl)azetidin-3- yl)(methyl)carbamoyl)azetidine-1-carboxylate as a yellow solid (95 mg, 75% yield). LCMS ESI m/z 620 [M+H] + . Step 2 N-(1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)azetidin-3-yl)-N- methylazetidine-2-carboxamide [0972] A solution of tert-butyl 2-((1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2- fluorobenzoyl)azetidin-3-yl)(methyl)carbamoyl)azetidine-1-ca rboxylate (95 mg, 0.15 mmol) in DCM (5 mL) and TFA (0.5 mL) was stirred at rt for 2 hours. The mixture was concentrated in vacuo and the residue was purified by prep-HPLC to give N-(1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)-2-fluorobenzoyl)azetidin-3-yl)-N-methylazetidine- 2-carboxamide as a white solid (35 mg, 44% yield). LCMS ESI m/z 520 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.48 (s, 1H), 8.29 (s, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.55 – 7.41 (m, 2H), 7.31 (dd, J = 8.8, 2.2 Hz, 1H), 7.27 – 7.19 (m, 1H), 7.10 (t, J = 4.1 Hz, 1H), 5.36 – 5.04 (m, 1H), 4.84 (p, J = 7.2 Hz, 1H), 4.72 – 4.57 (m, 1H), 4.31 (s, 2H), 4.21 (d, J = 7.9 Hz, 1H), 4.08 (dd, J = 17.3, 10.9 Hz, 4H), 3.55 – 3.38 (m, 2H), 2.87 (dd, J = 36.6, 23.1 Hz, 3H), 2.72 – 2.57 (m, 1H), 2.38 (d, J = 4.3 Hz, 2H), 1.99 (t, J = 7.9 Hz, 2H), 1.83 – 1.60 (m, 2H), 1.23 (s, 1H). Synthesis of Example 118: 2-Cyano-N-(1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin- 1- yl)methyl)-2-fluorobenzoyl)azetidin-3-yl)-N-methylacetamide

2-Cyano-N-(1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin- 1-yl)methyl)-2- fluorobenzoyl)azetidin-3-yl)-N-methylacetamide [0973] To a solution of 2-cyanoacetic acid (75 mg, 0.17 mmol) and 6-cyclobutoxy-4-(4-fluoro-3-(3- (methylamino)azetidine-1-carbonyl)benzyl)phthalazin-1(2H)-on e (29 mg, 0.34mmol) in DMF (1 mL) was added EDCI (49 mg, 0.26 mmol), HOBt (35 mg, 0.26 mmol) and DIPEA (111 mg, 0.86 mmol). The reaction mixture was stirred at rt for 3 hours and purified by prep-HPLC to give 2-cyano-N-(1-(5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoyl)azetidin-3-yl)-N- methylacetamide as a white solid (25 mg, 29% yield). LCMS ESI 504 (M+H) + . 1 H NMR (400 MHz, CDCl 3 ) δ 9.90 (s, 1H), 8.35 (d, J = 8.8 Hz, 1H), 7.51 (s, 1H), 7.34 (s, 1H), 7.20 (d, J = 6.7 Hz, 1H), 7.05 (t, J = 9.1 Hz, 1H), 6.90 (s, 1H), 5.25 (s, 1H), 4.79 – 4.61 (m, 1H), 4.48 – 4.36 (m, 1H), 4.33 – 4.08 (m, 5H), 3.52 (s, 2H), 3.14 (d, J = 13.4 Hz, 3H), 2.41 (s, 2H), 2.22 – 2.08 (m, 2H), 1.90 (d, J = 10.9 Hz, 1H), 1.83 – 1.67 (m, 1H). Synthesis of Example 119: 6-(4-(5-((7-(cyclopropylmethoxy)-4-oxo-3,4-dihydrophthalazin -1- yl)methyl)-2-fluor obenzoyl)piperazin-1-yl)nicotinonitrile

6-Cyclobutoxy-4-(3-(5-(cyclopropanecarbonyl)octahydropyrrolo [3,4-c]pyrrole-2-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [0974] To a solution of cyclopropanecarboxylic acid (17.3 mg, 0.20 mmol) and 6-cyclobutoxy-4-(4- fluoro-3-(octahydropyrrolo[3,4-c]pyrrole-2-carbonyl)benzyl)p hthalazin-1(2H)-one (100 mg, 0.20 mmol) in DMF (2 mL) was added EDCI (58 mg, 0.30 mmol), HOBt (41 mg, 0.30 mmol) and DIPEA (104 mg, 0.80 mmol). The reaction mixture was stirred at rt for 16 hours, then purified by prep-HPLC to afford 6- cyclobutoxy-4-(3-(5-(cyclopro panecarbonyl)octahydropyrrolo[3,4-c]pyrrole-2-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one (52 mg, 49% yield) as a white solid. LCMS ESI m/z 531.2 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.45 (s, 1H), 8.15 (m, 1H), 7.40 (m, 2H), 7.30 m, 1H), 7.23 (m, 1H), 7.08 (s, 1H), 4.89 – 4.78 (m, 1H), 4.29 (s, 2H), 3.90 – 3.68 (m, 2H), 3.61 – 3.54 (m, 1H), 3.45 (m, 3H), 3.23 – 2.81 (m, 4H), 2.39 (s, 2H), 2.00 (d, J = 2.6 Hz, 2H), 1.82 – 1.63 (m, 3H), 0.75 – 0.67 (m, 4H). Synthesis of Example 120: 6-(4-(3-((7-Hydroxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

6-Cyclobutoxy-4-(4-fluoro-3-(3-(methyl(4-(trifluoromethyl)py rimidin-2-yl)amino) azetidine-1- carbonyl)benzyl)phthalazin-1(2H)-one [0975] A mixture of 6-cyclobutoxy-4-(4-fluoro-3-(3-(methylamino)azetidine-1-carb onyl)benzyl) phthalazin-1(2H)-one (55 mg, 0.13 mmol), 2-chloro-4-(trifluoromethyl) pyrimidine (46 mg, 0.26 mmol) in DMF (1 mL) was added DIPEA (49 mg, 0.39 mmol). The reaction was stirred at 90 o C for 1 hour. The reaction was purified by prep-HPLC to give 6-cyclobutoxy-4-(4-fluoro-3-(3-(methyl(4- (trifluoromethyl)pyrimidin-2-yl)amino) azetidine-1-carbonyl)benzyl) phthalazin-1(2H)-one (29 mg, 40% yield) as a white solid. LCMS ESI m/z: 583.0 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.47 (s, 1H), 8.70 (d, J = 4.8 Hz, 1H), 8.14 (d, J = 8.8 Hz, 1H), 7.53 (dd, J = 6.5, 2.2 Hz, 1H), 7.47 – 7.41 (m, 1H), 7.31 – 7.20 (m, 2H), 7.13 – 7.08 (m, 2H), 5.35 (s, 1H), 4.82 (p, J = 7.1 Hz, 1H), 4.41 – 4.26 (m, 3H), 4.18 (m, 3H), 3.17 (s, 3H), 2.41 – 2.29 (m, 2H), 2.03 – 1.89 (m, 2H), 1.82 – 1.53 (m, 2H). Synthesis of Example 121: N-(1-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluoroben zoyl)azetidin-3-yl)-N-methylcyclopropanecarboxamide

N-(1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobe nzoyl)azetidin-3-yl)- N-methylcyclopropanecarboxamide [0976] To a solution of cyclopropanecarboxylic acid (40 mg, 0.81 mmol) and 6-cyclobutoxy-4-(4- fluoro-3-(3-(methylamino)azetidine-1-carbonyl)benzyl)phthala zin-1(2H)-one (110 mg, 0.25 mmol) in DMF (2 mL) was added EDCI (62 mg, 0.48 mmol), HOBt (32 mg, 0.24 mmol) and DIPEA (62 mg, 0.48 mmol). The reaction mixture was stirred at rt for 16 hours, then purified by pre-HPLC to afford N-(1-(5- ((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-f luorobenzoyl)azetidin-3-yl)-N- methylcyclopropanec arboxamide (36 mg 28 % yield) as a white solid. LCMS ESI m/z: 505.1 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.46 (s, 1H), 8.15 (m, 1H), 7.55 – 7.48 (m, 1H), 7.43 (s, 1H), 7.30 (m, 1H), 7.25 (d, J = 9.0 Hz, 1H), 7.11 (s, 1H), 5.22 (m, 1H), 4.94 – 4.73 (m, 1H), 4.30 (s, 2H), 4.11 (m, 3H), 3.15 (s, 2H), 2.90 (s, 1H), 2.37 (s, 2H), 2.13 – 1.17 (m, 6H), 0.72 (s, 4H). Synthesis of Example 122: N-(1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)azetidin-3-yl)-N-methyloxetane-2-carboxamide N-(1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)azetidin-3-yl)-N- methyloxetane-2-carboxamide [0977] To a solution of 6-cyclobutoxy-4-(4-fluoro-3-(3-(methylamino)azetidine-1-carb onyl)benzyl) phthalazin-1(2H)-one (75 mg, 0.17 mmol) and oxetane-2-carboxylic acid (92 mg, 0.38 mmol) in DMF (2 mL) was added EDCI (49 mg, 0.26 mmol), HOBt (35 mg, 0.26 mmol) and DIPEA (67 mg, 0.51 mmol). The reaction was stirred at 45 °C for 4 hours and purified by pre-HPLC to give N-(1-(5-((7-cyclobutoxy- 4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)aze tidin-3-yl)-N-methyloxetane-2- carboxamide as a white solid (33 mg, 37% yield). LCMS ESI m/z 521 (M+H) + . 1 H–NMR (400 MHz, DMSO–d6) δ 12.46 (s, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.47 (d, J = 22.8 Hz, 2H), 7.33 – 7.18 (m, 2H), 7.10 (s, 1H), 5.44 – 5.30 (m, 1H), 5.15 – 4.45 (m, 3H), 4.33 (d, J = 19.1 Hz, 3H), 4.26 – 3.97 (m, 4H), 2.90 (d, J = 25.3 Hz, 3H), 2.81 – 2.65 (m, 2H), 2.46 – 2.30 (m, 2H), 2.08 – 1.92 (m, 2H), 1.87 – 1.57 (m, 2H). Synthesis of Example 123: 2-((1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2- fluorobenzoyl)azetidin-3-yl)(methyl)amino)-N-methylisonicoti namide

2-Fluoro-N-methylisonicotinamide [0978] A solution of 2-fluoroisonicotinic acid (500 mg, 3.54 mmol), oxalyl chloride (450 mg, 387.19 mmol) and DMF (15 mg, 0.2 mmol) in anhydrous DCM (10 mL) was stirred at rt under Ar (g) for 30 mins. The reaction was concentrated under vacuum. The residue was dissolved in DCM (5 mL). This solution was added slowly to a mixture of DIPEA (1.37 g, 10.63 mmol) and methylamine hydrochloride (478 mg, 7.08 mmol) in DCM (10 mL) at 0 °C. The reaction mixture was stirred at rt for 1 hour, then diluted with water (10 mL) and extracted with DCM (10 mL x3). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo to give 2-fluoro-N-methylisonicotinamide as a yellow solid (331 mg, 61% yield). This was used without further purification. LCMS ESI m/z 155 [M+H] + . Step 2 tert-Butyl 3-(methyl(4-(methylcarbamoyl)pyridin-2-yl)amino)azetidine-1- carboxylate [0979] A mixture of 2-fluoro-N-methylisonicotinamide (331 mg, 1.64 mmol), tert-butyl 3- (methylamino)azetidine-1-carboxylate (1.2 g, 6.42 mmol) and DIPEA (693 mg, 5.35 mmol) in DMF (3 mL) was stirred at 130 °C for 16 hours. The mixture was cooled to rt, diluted with H 2 O (5 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed by brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 30% to 45%) to obtain tert-butyl 3-(methyl(4-(methylcarbamoyl)pyridin-2- yl)amino)azetidine-1-carboxylate as a yellow oil (103 mg, 15% yield). LCMS ESI m/z 321 [M+H] + . Step 3: 2-(Azetidin-3-yl(methyl)amino)-N-methylisonicotinamide [0980] A solutuon of tert-butyl 3-(methyl(4-(methylcarbamoyl)pyridin-2-yl)amino)azetidine-1- carboxylate (103 mg, 0.31 mmol) in DCM (3 mL) and TFA (0.3 mL) was stirred at rt for 1 hour. The solvent was removed under vacuum to afford 2-(azetidin-3-yl(methyl)amino)-N-methylisonicotinamide as a yellow oil (68 mg, 99% yield). This was used without further purification. LCMS ESI m/z 221 [M+H] + . Step 4 2-((1-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2-fluorobenzoyl)azetidin-3- yl)(methyl)amino)-N-methylisonicotinamide [0981] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (60 mg, 0.16 mmol) and 2-(azetidin-3-yl(methyl)amino)-N-methylisonicotinamide (103 mg, 0.20 mmol) in DMF (2 mL) was added EDCI (47 mg, 0.24 mmol), HOBt (33 mg, 0.24 mmol) and DIPEA (63 mg, 0.49 mmol). The reaction was stirred at rt for 24 hours. The reaction was purified by prep-HPLC to give 2-((1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)-2-fluorobenzoyl) azetidin-3- yl)(methyl)amino)-N-methylisonicotinamide as a white solid (29 mg, 30% yield). LCMS ESI m/z 571 (M+H) + . 1 H-NMR (400 MHz, DMSO–d 6 ) δ 12.51 (s, 1H), 9.42 (d, J = 4.5 Hz, 1H), 8.78 (t, J = 5.7 Hz, 1H), 8.44 (s, 1H), 8.33 (d, J = 6.6 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.60 (s, 1H), 7.47 – 7.27 (m, 3H), 7.24 – 7.06 (m, 3H), 4.90 – 4.74 (m, 2H), 4.67 – 4.54 (m, 1H), 4.49 – 4.39 (m, 1H), 4.34 – 3.94 (m, 5H), 3.83 – 3.72 (m, 1H), 3.66 – 3.54 (m, 1H), 2.78 (d, J = 4.4 Hz, 3H), 2.46 – 2.34 (m, 2H), 2.09 – 1.94 (m, 2H), 1.87 – 1.58 (m, 2H). Synthesis of Example 124: 2-((1-(2-Fluoro-5-((7-(oxetan-3-yloxy)-4-oxo-3,4-dihydrophth alazin-1- yl)methyl)benzoyl)azetidin3yl)(methyl)amino)isonicotinonitri le

3-Hydroxy-5-methoxyisobenzofuran-1(3H)-one [0982] A solution of 2-bromo-4-methoxybenzoic acid (4.98 g, 21.64 mmol) in THF (50 mL) was cooled to -78 °C, then n-BuLi (2.5M in heptane, 17.31 mL, 43.28 mmol) was added dropwise under Ar (g). The reaction was stirred at -78°C for 15 mins, then DMF (3.48 g, 47.61 mmol) was added dropwise. The reaction was kept at -78°C for 20 mins, then quenched with sat. NH 4 Cl soltion (20 mL). The resulting mixture was added aq HCl (1N) until the solution reached pH 5~6, then extracted with EtOAc (50 x 3 mL). The combined organic layer was washed with bine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 30 to 50%) to give 3- hydroxy-5-methoxyisobenzofuran-1(3H)-one as a white solid (1.5 g, 38% yield). LCMS ESI m/z 181 [M+H] + ). Step 2 Dimethyl (6-methoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate [0983] A solution of 3-hydroxy-5-methoxyisobenzofuran-1(3H)-one (1.5 g, 8.30 mmol) in dimethylphosphite (30 mL) was heated to 100 °C and stirred for 2 hours. The mixture was cooled to rt, diluted with H2O (10 mL) and extracted with EtOAc (30 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give dimethyl (6- methoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate as a light yellow oil (1.29 g, 57% yield). This was used without further purification. LCMS ESI m/z 273 [M+H] + . Step 3 Methyl 2-fluoro-5-((7-methoxy-4-oxo-3,4-dihydrophthalazin-1-yl)meth yl)benzoate [0984] A solution of dimethyl (6-methoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (1.29 g, 4.74 mmol), methyl 2-fluoro-5-formylbenzoate (863 mg, 4.74 mmol) and Et 3 N (1.44 g, 14.22 mmol) in anhydrous THF (10 mL) was stirred at 60 °C under Ar(g) for 5 hours. Hydrazine hydrate (228 mg, 7.11 mmol) was added and the reaction was stirred at 70 °C for 1 hour. The reaction was concentrated under vacuum. The formed solids were washed with water (10 mL) and pet ether/EA (2:1, 10 mL) to give methyl 2-fluoro-5-((7-hydroxy-4-oxo-3,4-dihydrophthalazin-1-yl)meth yl)benzoate as a white solid (1.15 g, 71% yield). This was used without further purification. LCMS ESI m/z 343 [M+H] + . Step 4 Methyl 2-fluoro-5-((7-hydroxy-4-oxo-3,4-dihydrophthalazin-1-yl)meth yl)benzoate [0985] A solution of methyl 2-fluoro-5-((7-methoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoate (800 mg, 1.46 mmol) and BBr 3 (30% in DCM, 12.17 g, 14.6 mmol) in DCM (40 mL) was stirred at rt for 16 hours. The mixture was diluted with H 2 O (20 mL) and extracted with DCM (20 mL x 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 2-fluoro-5-((7-hydroxy-4-oxo-3,4-dihydrophthalazin-1-yl)meth yl)benzoate as a white solid (500 mg, 65% yield). This was used without further purification. LCMS ESI m/z 329 [M+H] + . Step 5 Methyl 2-fluoro-5-((7-(oxetan-3-yloxy)-4-oxo-3,4-dihydrophthalazin- 1-yl)methyl)benzoate [0986] A mixture of methyl 2-fluoro-5-((7-hydroxy-4-oxo-3,4-dihydrophthalazin-1-yl)meth yl)benzoate (300 mg, 0.91 mmol), 3-bromooxetane (371 mg, 2.73 mmol) and K 2 CO 3 (377 mg, 2.73 mmol) in DMF (3 mL) was stirred at 110 °C for 6 hours. The mixture was cooled to rt, poured into H 2 O (5 mL), then aq HCl (1N) was added until the solution reached pH 5~6. The mixture was extracted with EtOAc (20 mL x3). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 2-fluoro-5-((7-(oxetan-3-yloxy)-4-oxo-3,4-dihydrophthalazin- 1-yl)methyl)benzoate as a white solid (210 mg, 60% yield). This was used without further purification. LCMS ESI m/z 384 [M+H] + . Step 6 2-Fluoro-5-((7-(oxetan-3-yloxy)-4-oxo-3,4-dihydrophthalazin- 1-yl)methyl)benzoic acid [0987] A mixture of methyl 2-fluoro-5-((7-(oxetan-3-yloxy)-4-oxo-3,4-dihydrophthalazin- 1- yl)methyl)benzoate (200 mg, 0.52 mmol) and LiOH-H 2 O (168 mg, 4 mmol) in MeOH (4 mL) and H 2 O (2 mL) was stirred at rt for 3 hours. The organic solvent was removed in vacuo and aq HCl (1N) was added until the solution reached pH 5~6. The solids were collected by filtration, washed with water (5 mL) and dried under vacuum to give 2-fluoro-5-((7-(oxetan-3-yloxy)-4-oxo-3,4-dihydrophthalazin- 1- yl)methyl)benzoic acid as a white solid (123 mg.64% yield). This was used without further purification. LCMS ESI m/z 370 [M+H] + . Step 7 2-((1-(2-Fluoro-5-((7-(oxetan-3-yloxy)-4-oxo-3,4-dihydrophth alazin-1-yl)methyl)benzoyl)azetidin-3- yl)(methyl)amino)isonicotinonitrile [0988] To a solution of 2-(azetidin-3-yl(methyl)amino)isonicotinonitrile (70 mg, 0.37 mmol) and 2- fluoro-5-((7-(oxetan-3-yloxy)-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoic acid (123 mg, 0.33 mmol) in DMF (2 mL) was added EDCI (117 mg, 0.61 mmol), HOBt (82 mg, 0.60 mmol) and DIPEA (158 mg, 1.22 mmol). The mixture was stirred at rt for 2 hours, then purified by prep-HPLC to give 2-((1- (2-fluoro-5-((7-(oxetan-3-yloxy)-4-oxo-3,4-dihydrophthalazin -1-yl)methyl)benzoyl)azetidin-3- yl)(methyl)amino)isonicotinonitrile as a yellow solid (46 mg, 65% yield). LCMS ESI m/z 541 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.51 (s, 1H), 8.59 (s, 1H), 8.44 (d, J = 4 Hz, 1H), 8.19 (s, 1H), 7.96 (s, 1H), 7.44 (d, J = 8 Hz, 1H), 7.31 (m, 1H), 7.26 (m, 1H), 7.23 – 7.15 (m, 1H), 7.00 (m, 1H), 5.47 (m, 1H), 4.93 (m, 2H), 4.87 – 4.79 (m, 1H), 4.56 (m,, 1H), 4.49 (t, J = 5.9 Hz, 3H), 4.27 (s, 2H), 3.65 (s, 1H), 3.62 (s, 3H). Synthesis of Example 125: 6-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-6,6-difluoro-1,4-diazepan-1-yl)nicotinonitril e

6-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-6,6-difluoro- 1,4-diazepan-1-yl)nicotinonitrile [0989] Following the amide coupling procedure in Example 126, but starting with methyl 5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoic acid and 6-(6,6-difluoro-1,4- diazepan-1-yl)nicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 589.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ12.42 (d, J = 5.2 Hz, 1H), 8.50 (dd, J = 35.1, 2.0 Hz, 1H), 8.15 (dd, J = 8.8, 4.2 Hz, 1H), 7.92 (dd, J = 9.0, 2.3 Hz, 1H), 7.38 (dd, J = 13.7, 11.1 Hz, 1H), 7.33 – 7.14 (m, 3H), 7.11 – 6.95 (m, 2H), 4.81 (m, 1H), 4.39 – 4.21 (m, 4H), 4.15 (s, 1H), 3.90 (m, 3H), 3.73 (s, 1H), 3.49 (t, J = 5.3 Hz, 1H), 2.37 (s, 2H), 2.00 (t, J = 9.6 Hz, 2H), 1.77 (m, 1H), 1.71 – 1.59 (m, 1H). Synthesis of Example 126: 6-Cyclobutoxy-4-(3-(6,6-difluoro-4-(5-(trifluoromethyl)pyrim idin-2-yl)- 1,4-diazepane-1-carbonyl)-4-fluorobenzyl)phthalazin-1(2H)-on e

6,6-Difluoro-1-(5-(trifluoromethyl)pyrimidin-2-yl)-1,4-diaze pane [0990] A solution of 6,6-difluoro-1,4-diazepane hydrochloride (27 mg, 156.43 μmol), 2-chloro-5- (trifluoromethyl)pyrimidine (28.55 mg, 156.43 μmol) and DIPEA (101.09 mg, 782.14 μmol) in NMP (1 mL) was stirred at 100 °C under microwave for 20 min. The reaction mixture was diluted in EtOAc, washed with water 3 times. The organic layer was concentrated to the crude and purified by Prep-TLC (EtOAc: Petroleum ether = 3: 1) to give 6,6-difluoro-1-[5-(trifluoromethyl)pyrimidin-2-yl]-1,4-diaze pane (40.0 mg, 91% yield) as a white solid. LCMS ESI m/z: 282.9 [M+H] + . Step 2

6-Cyclobutoxy-4-(3-(6,6-difluoro-4-(5-(trifluoromethyl)pyrim idin-2-yl)-1,4-diazepane-1-carbonyl)- 4-fluorobenzyl)phthalazin-1(2H)-one [0991] Following the amide coupling procedure above in Example 94, but starting with 6,6-difluoro-1- (5-(trifluoromethyl)pyrimidin-2-yl)-1,4-diazepane and 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)-2-fluorobenzoic acid gave the title compound as a white solid. LCMS ESI m/z: 633.2 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.42 (d, J = 3.2 Hz, 1H), 8.75 (d, J = 51.0 Hz, 2H), 8.15 (dd, J = 8.8, 5.5 Hz, 1H), 7.45 – 7.34 (m, 1H), 7.34 – 7.12 (m, 3H), 7.06 (m, 1H), 4.80 (m, 1H), 4.42 (s, 2H), 4.32 – 4.04 (m, 4H), 3.94 (s, 2H), 3.77 (s, 1H), 3.51 (t, J = 5.3 Hz, 1H), 2.36 (m, 2H), 2.07 – 1.91 (m, 2H), 1.76 (m, 1H), 1.65 (m, 1H). Synthesis of Example 127: 6-Cyclobutoxy-4-(3-(4-(cyclopropanecarbonyl)-6,6-difluoro-1, 4- diazepane-1-carbonyl)-4-fluorobenzyl)phthalazin-1(2H)-one tert-butyl 4-(cyclopropanecarbonyl)-6,6-difluoro-1,4-diazepane-1-carbox ylate [0992] To a solution of tert-butyl 6,6-difluoro-1,4-diazepane-1-carboxylate (140 mg, 592.57 μmol) in THF (4 mL) was added Et 3 N (179.89 mg, 1.78 mmol, 247.78 μL) and cyclopropanecarbonyl chloride (74.33 mg, 711.09 μmol, 64.64 μL) at 25 °C . After stirring the reaction mixture 25 °C for 1 hr. NH 4 Cl(aq.) and EtOAc were added in the mixture, which became two phases. The organic layer was separated, washed with aq. Na 2 CO 3 , dried over Na 2 SO 4 and concentrated to give tert-butyl 4- (cyclopropanecarbonyl)-6,6-difluoro-1,4-diazepane-1-carboxyl ate (130 mg, 72% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3) δ 3.99 (t, J = 11.9 Hz, 2H), 3.88 – 3.51 (m, 6H), 1.48 (s, 9H), 1.20 – 1.14 (m, 1H), 1.05 – 0.99 (m, 2H), 0.83 (td, J = 6.8, 3.8 Hz, 2H). Step 2 cyclopropyl(6,6-difluoro-1,4-diazepan-1-yl)methanone hydrochloride [0993] To a mixture of tert-butyl 4-(cyclopropanecarbonyl)-6,6-difluoro-1,4-diazepane-1-carbox ylate (140 mg, 460.02 μmol) in 4M HCl/EA (5 mL). The reaction mixture was stirred at rt. for 2 hr. The mixture was concentrated to give cyclopropyl-(6,6-difluoro-1,4-diazepan-1-yl)methanone (110 mg, 457.04 μmol, 99.35% yield, HCl) as a white solid. LCMS ESI m/z: 205.2 [M+H] + . Step 3 6-cyclobutoxy-4-(3-(4-(cyclopropanecarbonyl)-6,6-difluoro-1, 4-diazepane-1-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [0994] Following the amide coupling procedure above in Example 94, but starting with cyclopropyl(6,6-difluoro-1,4-diazepan-1-yl)methanone hydrochloride gave the title compound as a white solid. LCMS ESI m/z: 555.2 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.42 (d, J = 2.4 Hz, 1H), 8.15 (dd, J = 8.8, 3.2 Hz, 1H), 7.49 – 7.19 (m, 4H), 7.15 – 7.01 (m, 1H), 4.89 – 4.73 (m, 1H), 4.29 (s, 2H), 4.26 – 4.10 (m, 2H), 3.97 (m, 2H), 3.76 (m, 2H), 3.64 – 3.34 (m, 2H), 2.38 (s, 2H), 2.00 (d, J = 8.0 Hz, 3H), 1.83 – 1.61 (m, 2H), 0.70 (m, 4H). Synthesis of Example 128: 6-cyclobutoxy-4-(4-fluoro-3-(4-(4-methoxypyridin-2-yl)pipera zine-1- carbonyl)benzyl)phthalazin-1(2H)-one tert-butyl 4-(4-methoxypyridin-2-yl)piperazine-1-carboxylate [0995] To a solution of 2-chloro-4-methoxy-pyridine (200 mg, 1.39 mmol) in PhCH3 (4 mL) was added tert-butyl piperazine-1-carboxylate (778.37 mg, 4.18 mmol), Pd 2 (dba) 3 (63.78 mg, 69.65 μmol), BINAP (86.74 mg, 139.30 μmol) and t-BuOK (468.95 mg, 4.18 mmol). The reaction mixture was stirred under microwave at 115 °C for 1 hr. The reaction mixture was filtered. The filtrate was poured into water (10 mL), extracted with EA (10 mL* 2). The organic layer was washed with brine, dried and concentrated to the residue. The residue was purified by silica gel chromatography to give tert-butyl 4-(4- methoxy-2-pyridyl)piperazine-1-carboxylate (130 mg, 31.8% yield) as a light green oil. LCMS ESI m/z: 294.3 [M+H] + . Step 2 and 3

6-cyclobutoxy-4-(4-fluoro-3-(4-(4-methoxypyridin-2-yl)pipera zine-1-carbonyl)benzyl)phthalazin- 1(2H)-one [0996] Following the amide coupling two-step procedure above in Example 94, but starting with tert- butyl 4-(4-methoxypyridin-2-yl)piperazine-1-carboxylate gave the title compound as a white solid. LCMS ESI m/z: 543.8 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.44 (s, 1H), 8.15 (t, J = 6.1 Hz, 1H), 7.94 (d, J = 5.7 Hz, 1H), 7.41 (dd, J = 6.3, 2.2 Hz, 2H), 7.34 – 7.19 (m, 2H), 7.11 (d, J = 2.3 Hz, 1H), 6.36 – 6.27 (m, 2H), 4.89 – 4.77 (m, 1H), 4.31 (s, 2H), 3.78 (s, 3H), 3.71 (s, 2H), 3.57 (d, J = 4.8 Hz, 2H), 3.42 (s, 2H), 3.27 (s, 2H), 2.45 – 2.34 (m, 2H), 2.08 – 1.94 (m, 2H), 1.72 (m, 2H). Synthesis of Example 129: 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)-5-(trifluoromethyl)nicotinonit rile tert-butyl 4-(5-bromo-3-(trifluoromethyl)pyridin-2-yl)piperazine-1-carb oxylate [0997] Following the general one-step procedure above in Example 23, but starting with 5-bromo-2- chloro-3-(trifluoromethyl)pyridine gave the title compound as yellow oil. LCMS ESI m/z: 356.0 [M+H- 56] + . Step 2, 6-(piperazin-1-yl)-5-(trifluoromethyl)nicotinonitrile [0998] A mixture of tert-butyl 4-[5-bromo-3-(trifluoromethyl)-2-pyridyl]piperazine-1-carbox ylate (100 mg, 243.77 μmol), Zinc Cyanide (17.17 mg, 146.26 μmol, 9.27 μL) and Pd 2 (dba) 3 (22.32 mg, 24.38 μmol) in DMF (1.5 mL) was stirred at 140 °C under N 2 for 60 min. The reaction was worked up with water and EtOAc, the organic layer was dried and concentrated. The crude product was used to the next step directly. LCMS ESI m/z: 257.2 [M+H] + . Step 3, 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1-yl)- 5-(trifluoromethyl)nicotinonitrile [0999] Following the amide coupling procedure above in Example 94, but starting with 6-(piperazin- 1-yl)-5-(trifluoromethyl)nicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 606.7 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.44 (s, 1H), 8.82 (d, J = 2.0 Hz, 1H), 8.57 (d, J = 2.0 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.47 – 7.37 (m, 2H), 7.34 – 7.21 (m, 2H), 7.09 (d, J = 2.3 Hz, 1H), 4.83 (p, J = 7.1 Hz, 1H), 4.30 (s, 2H), 3.76 (s, 2H), 3.61 (s, 2H), 3.46 (s, 2H), 3.33 (s, 2H), 2.45 – 2.31 (m, 2H), 2.06 – 1.92 (m, 2H), 1.82 – 1.58 (m, 2H). Synthesis of Example 130: 6-cyclobutoxy-4-(4-fluoro-3-(4-(5-(hydroxymethyl)pyridin-2- yl)piperazine-1-carbonyl)benzyl)phthalazin-1(2H)-one tert-butyl 4-(5-(methoxycarbonyl)pyridin-2-yl)piperazine-1-carboxylate [1000] To a solution of methyl 6-chloronicotinate (500 mg, 2.91 mmol) and tert-butyl piperazine-1- carboxylate (543 mg, 2.91 mmol) in DMF (5 mL) was added DIPEA (1.51 g, 11.66 mmol). The reaction mixture was stirred at 130 °C for 3 hours, then quenched with water (30 mL) and extracted with EtOAc (30 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 10% to 40%) to give tert-butyl 4-(5-(methoxycarbonyl)pyridin-2-yl)piperazine-1-carboxylate (780 mg, 83% yield) as a white solid. LCMS ESI m/z: 322 [M+H] + . Step 2 methyl 6-(piperazin-1-yl)nicotinate [1001] A solution of tert-butyl 4-(5-(methoxycarbonyl)pyridin-2-yl)piperazine-1-carboxylate (780 mg, 2.43 mmol) in HCl/Dioxane (4M, 10 mL) was stirred at rt for 1 hour. The solvent was removed in vacuo to give methyl 6-(piperazin-1-yl)nicotinate (510 mg, 95% yield) as a white solid. This was used without further purification. LCMS ESI m/z: 222 [M+H] + . Step 3 methyl 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinate [1002] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (100 mg, 0.27 mmol) and methyl 6-(piperazin-1-yl)nicotinate (96 mg, 0.30 mmol) in DMF (2 mL) was added EDCI (78 mg, 0.41 mmol), HOBt (55 mg, 0.41 mmol) and DIPEA (175 mg, 1.36 mmol). The reaction was stirred at rt for 4 hours, then quenched with water (20 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 10% to 70%) to give methyl 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinate (130 mg, 84% yield) as a white solid. LCMS ESI m/z: 572 [M+H] + . Step 4 6-cyclobutoxy-4-(4-fluoro-3-(4-(5-(hydroxymethyl)pyridin-2-y l)piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one [1003] To a solution of methyl 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)nicotinate (130 mg, 0.23 mmol) in THF (5 mL) was added LiBH 4 (15 mg, 0.68 mmol). The reaction mixture was stirred at rt for 72 hours. The reaction was purified by prep-HPLC to give 6-cyclobutoxy-4-(4-fluoro-3-(4-(5-(hydroxymethyl)pyridin-2-y l)piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one (36 mg, 29%) as a white solid. LCMS ESI m/z: 544 [M+H] + . 1 H– NMR (400 MHz, CDCl3) δ 10.83 (s, 1H), 8.35 (d, J = 8.8 Hz, 1H), 8.17 – 8.14 (m, 1H), 7.57 (dd, J = 8.7, 2.3 Hz, 1H), 7.37 – 7.34 (m, 1H), 7.34 – 7.29 (m, 1H), 7.20 (dd, J = 8.8, 2.3 Hz, 1H), 7.06 (t, J = 8.8 Hz, 1H), 6.91 (d, J = 2.3 Hz, 1H), 6.66 (d, J = 8.7 Hz, 1H), 4.71 – 4.63 (m, 1H), 4.57 (s, 2H), 4.23 (s, 2H), 3.93 – 3.84 (m, 2H), 3.65 – 3.60 (m, 2H), 3.56 – 3.51 (m, 2H), 3.45 – 3.36 (m, 2H), 2.45 – 2.38 (m, 2H), 2.20 – 2.11 (m, 2H), 1.93 – 1.85 (m, 1H), 1.81 – 1.71 (m, 1H). Synthesis of Example 131: 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)-5-methoxynicotinonitrile tert-butyl 4-(5-bromo-3-methoxypyridin-2-yl)piperazine-1-carboxylate [1004] Following the general one-step procedure above in Example 130, but starting with 5-bromo-2- chloro-3-methoxypyridine gave the title compound as yellow oil. LCMS ESI m/z: 374.1 [M+H] + . Step 2 tert-butyl 4-(5-cyano-3-methoxypyridin-2-yl)piperazine-1-carboxylate [1005] To a solution of tert-butyl 4-(5-bromo-3-methoxy-2-pyridyl)piperazine-1-carboxylate (100 mg, 268.63 μmol) in DMF (2 mL) was added Zn(CN) 2 (31.54 mg, 268.63 μmol), Pd 2 (dba) 3 (24.60 mg, 26.86 μmol), S-phos (11.03 mg, 26.86 μmol), and then the sealed tube was irradiated in microwave at 140 °C for 1.5 hr. The reaction was poured into H 2 O (10 mL), extracted with EA (10 mL). The organic layer was washed with brine, dried and concentrated. The residue was purified by Prep-TLC to give tert-butyl 4-(5-cyano-3-methoxy-2-pyridyl)piperazine-1-carboxylate (40 mg, 46.7% yield) as a yellow solid. LCMS ESI m/z: 262.9 [M+H-56] + . Step 3, and 4 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1-yl)- 5-methoxynicotinonitrile [1006] Following the deprotection and amide coupling two-step procedure above in Example 94, but starting with tert-butyl 4-(5-cyano-3-methoxypyridin-2-yl)piperazine-1-carboxylate gave the title compound as a white solid. LCMS ESI m/z: 568.8 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.44 (s, 1H), 8.21 (d, J = 1.7 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.61 (d, J = 1.7 Hz, 1H), 7.39 (d, J = 6.4 Hz, 2H), 7.33 – 7.21 (m, 2H), 7.08 (d, J = 2.2 Hz, 1H), 4.86 – 4.78 (m, 1H), 4.30 (s, 2H), 3.84 (s, 3H), 3.72 (s, 2H), 3.64 (s, 2H), 3.48 (s, 2H), 3.28 (s, 2H), 2.44 – 2.34 (m, 2H), 1.99 (t, J = 10.6 Hz, 2H), 1.77 (d, J = 10.1 Hz, 1H), 1.65 (dd, J = 19.0, 8.7 Hz, 1H). Synthesis of Example 132: 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)-4-(trifluoromethyl)nicotinonit rile

6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1-yl)- 4-(trifluoromethyl)nicotinonitrile [1007] Following the general three-step procedure above in Example 130, but starting with 6-chloro-4- (trifluoromethyl)nicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 606.6 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.44 (s, 1H), 8.73 (s, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.43 (t, J = 9.0 Hz, 2H), 7.35 – 7.22 (m, 3H), 7.10 (d, J = 2.2 Hz, 1H), 4.88 – 4.79 (m, 1H), 4.31 (s, 2H), 3.88 (s, 2H), 3.74 (s, 4H), 3.36 (s, 2H), 2.40 (m, 2H), 2.06 – 1.96 (m, 2H), 1.82 – 1.63 (m, 2H). Synthesis of Example 133: 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)-2-methylnicotinonitrile

6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1-yl)- 2-methylnicotinonitrile [1008] Following the general three-step procedure above in Example 130, but starting with 6-chloro-2- methylnicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 552.7 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.44 (s, 1H), 8.16 (d, J = 8.7 Hz, 1H), 7.82 (d, J = 8.9 Hz, 1H), 7.40 (d, J = 6.1 Hz, 2H), 7.35 – 7.19 (m, 2H), 7.09 (s, 1H), 6.75 (m, 1H), 4.86 – 4.79 (m, 1H), 4.31 (s, 2H), 3.73 (d, J = 12.6 Hz, 4H), 3.58 (s, 2H), 3.29 – 3.23 (m, 2H), 2.47 (s, 3H), 2.39 (m, 2H), 2.05 – 1.95 (m, 2H), 1.81 – 1.63 (m, 2H). Synthesis of Example 134: 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)-4-methylnicotinonitrile

6-hydroxy-4-methylnicotinonitrile [1009] To a solution of 6-methoxy-4-methyl-pyridine-3-carbonitrile (200 mg, 1.35 mmol) in CH 3 CN (3 mL) was added Chlorotrimethylsilane (293.30 mg, 2.70 mmol, 342.64 μL) and NaI (404.67 mg, 2.70 mmol, 110.35 μL). The reaction mixture was stirred at rt. for 18 hr. Water and EtOAc were added in the mixture, which became two phases. The organic layer was separated, washed with NaCl (aq), dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 20/1 to 1/1) to give 6-hydroxy-4-methyl-pyridine-3- carbonitrile (76 mg, 41.9% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-D6) δ 12.26 (s, 1H), 8.20 (s, 1H), 6.32 (s, 1H), 2.22 (d, J = 0.9 Hz, 3H). Step 2 6-chloro-4-methylnicotinonitrile [1010] To a solution of 6-hydroxy-4-methyl-pyridine-3-carbonitrile (76 mg, 566.59 μmol) in dioxane (3 mL) was added POCl 3 (130.32 mg, 849.89 μmol) at 25 °C under N 2 . The reaction mixture was stirred at 120 °C for 3 hr. The reaction mixture was poured into ice-cold water, extracted with EA. The organic layer was washed with NaCl (aq), dried over Na 2 SO 4 and concentrated to give a residue. The residue was purified by silica gel chromatography (petroleum ether/EtOAc = 20/1 to 1/1) to give 6-chloro-4- methylnicotinonitrile (54 mg, 62.4% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-D6) δ 8.82 (s, 1H), 7.75 (s, 1H), 2.51 (s, 3H). Step 3, 4 and 5 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1-yl)- 4-methylnicotinonitrile [1011] Following the general three-step procedure above in Example 130, but starting with 6-chloro-4- methylnicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 552.7 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.44 (s, 1H), 8.42 (s, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.42 (t, J = 8.0 Hz, 2H), 7.34 – 7.23 (m, 2H), 7.10 (d, J = 2.1 Hz, 1H), 6.86 (s, 1H), 4.83 (m, 1H), 4.30 (s, 2H), 3.73 (d, J = 6.8 Hz, 4H), 3.60 (s, 2H), 3.30 – 3.26 (m, 2H), 2.39 (s, 2H), 2.35 (s, 3H), 2.06 – 1.95 (m, 2H), 1.80 – 1.63 (m, 2H). Synthesis of Example 135: 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)-2-(trifluoromethyl)nicotinonit rile Step 1, 2 and 3 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1-yl)- 2-(trifluoromethyl)nicotinonitrile [1012] Following the general three-step procedure above in Example 130, but starting with 6-chloro-2- (trifluoromethyl)nicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 606.7 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.44 (s, 1H), 8.15 (dd, J = 9.0, 4.0 Hz, 2H), 7.46 – 7.38 (m, 2H), 7.34 –7.25 (m, 2H), 7.19 (d, J = 9.2 Hz, 1H), 7.09 (d, J = 2.2 Hz, 1H), 4.88 – 4.80 (m, 1H), 4.31 (s, 2H), 3.85 – 3.63 (m, 6H), 3.33 (s, 2H), 2.43 – 2.34 (m, 2H), 2.06 – 1.96 (m, 2H), 1.70 (m,2H). Synthesis of Example 136: 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)-2-methoxynicotinonitrile

6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1-yl)- 2-methoxynicotinonitrile [1013] Following the general three-step procedure above in Example 130, but starting with 6-chloro-2- methoxynicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 568.8 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.44 (s, 1H), 8.15 (t, J = 6.9 Hz, 1H), 7.81 (dd, J = 8.6, 4.5 Hz, 1H), 7.48 – 7.37 (m, 2H), 7.35 – 7.22 (m, 2H), 7.09 (d, J = 2.3 Hz, 1H), 6.46 (d, J = 8.7 Hz, 1H), 4.87 – 4.78 (m, 1H), 4.31 (s, 2H), 3.90 (s, 3H), 3.74 (d, J = 8.8 Hz, 4H), 3.59 (s, 2H), 3.30 – 3.25 (m, 2H), 2.43 – 2.33 (m, 2H), 2.06 – 1.94 (m, 2H), 1.83 – 1.60 (m, 2H). Synthesis of Example 137: 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)-4-methoxynicotinonitrile Step 1, 2 and 3

6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1-yl)- 4-methoxynicotinonitrile [1014] Following the general three-step procedure above in Example 130, but starting with 6-chloro-4- methoxynicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 568.8 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.44 (s, 1H), 8.32 (s, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.43 (dd, J = 10.6,5.9 Hz, 2H), 7.33 – 7.23 (m, 2H), 7.10 (d, J = 2.3 Hz, 1H), 6.42 (s, 1H), 4.87 – 4.79 (m, 1H), 4.31 (s, 2H),3.93 (s, 3H), 3.73 (m, 6H), 3.29 (s, 2H), 2.39 (m, 2H), 2.00 (m, 2H), 1.84 – 1.63 (m, 2H). Synthesis of Example 138: 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)-5-methylnicotinonitrile tert-butyl 4-(5-cyano-3-methylpyridin-2-yl)piperazine-1-carboxylate [1015] 6-chloro-5-methyl-pyridine-3-carbonitrile (100 mg, 655.39 μmol), tert-butyl piperazine-1- carboxylate (161.42 mg, 655.39 μmol, CH 3 COOH) DIPEA (42352 mg, 3.28 mmol, 570.79 μL) and NMP (0.5 mL) were added to a 10 mL sealed tube. The reaction mixture was stirred at 150 °C for 40 min under microwave. The mixture was diluted in EA (10 mL), washed with water (10 mL* 2), brine, dried and concentrated to the crude. The residue was purified by silica gel chromatography (PE: EA =10:1 to 1: 1) to give tert-butyl 4-(5-cyano-3-methyl-2-pyridyl)piperazine-1-carboxylate (150 mg, 75.6% yield) as a yellow solid. LCMS ESI m/z: 247.2 [M - 56] + . Step 2 5-methyl-6-(piperazin-1-yl)nicotinonitrile [1016] tert-butyl 4-(5-cyano-3-methyl-2-pyridyl)piperazine-1-carboxylate (150 mg, 496.08 μmol), TFA (1.48 g, 12.98 mmol, 1 mL) and DCM (2 mL) were added to a 100 mL bottled flask. The resultant mixture was stirred at 25 °C for 2 hr. The mixture was concentrated to give 5-methyl-6- piperazin-1-yl-pyridine-3-carbonitrile (150 mg, crude, TFA) as a yellow oil. LCMS ESI m/z: 203.2 [M + H] + . Step 3 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1-yl)- 5-methylnicotinonitrile [1017] Following the general procedure above in Example 55, but starting with 5-methyl-6-(piperazin- 1-yl)nicotinonitrile gave the title compound as a white solid. LCMS ESI m/z: 553.2 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.44 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.92 (d, J = 1.5 Hz, 1H), 7.46– 7.38 (m, 2H), 7.32 – 7.22 (m, 2H), 7.10 (d, J = 2.3 Hz, 1H),4.87 – 4.79 (m, 1H), 4.31 (s, 2H), 3.77 (s, 2H), 3.38 – 3.32 (m, 4H), 3.23 (s, 2H), 2.43 – 2.35 (m, 2H), 2.26 (s, 3H), 2.05 – 1.95(m, 2H), 1.82 – 1.63 (m, 2H). Synthesis of Example 139: 6-(7-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyra zin-3-yl)nicotinonitrile

methyl 5-cyanopicolinate [1018] A mixture of 6-chloropyridine-3-carbonitrile (500 mg, 3.61 mmol), ET3N (730.32 mg, 7.22 mmol, 1.01 mL), Pd(dppf)Cl 2 (264.05 mg, 360.87 μmol) and Methanol (10 mL) was stirred at 75 °C for 12 hr under CO (g) (15 psi). The mixture was concentrated to a crude residue, which was purified by silica gel chromatography (PE: EA = 10:1 to 1: 1) to give methyl 5-cyanopyridine-2-carboxylate (120 mg, 20.5% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-D6) δ 9.20 (m, 1H), 8.53 (dd, J = 8.2, 2.1 Hz, 1H), 8.20 (dd, J = 8.2, 0.6 Hz, 1H), 3.92 (s, 3H). Step 2, 4, 5 and 6 6-(7-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-5,6,7,8- tetrahydro-[1,2,4]triazolo[4,3-a]pyrazin-3-yl)nicotinonitril e [1019] Following the general four-step procedure in Example 1, but starting with methyl 5- cyanopicolinate gave the title compound as a white solid. LCMS ESI m/z: 577.2 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.43 (s, 1H), 9.15 (s, 1H), 8.47 (dd, J = 8.4, 2.1 Hz, 1H), 8.34 (d, J = 8.3 Hz, 1H), 8.15 (d, J = 8.7 Hz, 1H), 7.48 (t, J = 6.1 Hz, 2H), 7.31 (dd, J = 10.3, 7.1 Hz, 2H), 7.08 (s, 1H), 5.05 (s, 1H), 4.87 – 4.77 (m, 1H), 4.72 (s, 1H), 4.56 (s, 1H), 4.40 (s, 1H), 4.32 (s, 2H), 4.13 (s, 1H), 3.68 (s, 1H), 2.37 (s, 2H), 2.07 – 1.91 (m, 2H), 1.83 – 1.60 (m, 2H). Synthesis of Example 140: 6-cyclobutoxy-4-(4-fluoro-3-(4-(5-hydroxypyridin-2-yl)pipera zine-1- carbonyl)benzyl)phthalazin-1(2H)-one 5-(benzyloxy)-2-chloropyridine [1020] A mixture of 6-chloropyridin-3-ol (1.00 g, 7.75 mmol), (bromomethyl)benzene (1.58 g, 9.30 mmol) and K 2 CO 3 (1.60 g, 11.63 mmol) in DMF (20 mL) was stirred at 130 °C for 12 hours. The reaction was cooled to rt, diluted with H 2 O (50 mL) and extrated with EtOAc (3 x 30 mL). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to give 5-(benzyloxy)- 2-chloropyridine (1.25 g, 74% yield) as a yellow solid. LCMS ESI m/z: 220 [M+H] + . Step 2 tert-butyl 4-(5-(benzyloxy)pyridin-2-yl)piperazine-1-carboxylate [1021] A mixture of 5-(benzyloxy)-2-chloropyridine (1.25 g, 5.68 mmol), tert-butyl piperazine-1- carboxylate (1.06 g, 5.68 mmol), Pd 2 (dba) 3 (530 mg, 0.57 mmol), BINAP (706 mg, 1.13 mmol) and t- BuONa (1.64 g, 17.04 mmol) in toluene (50 mL) was stirred at 110 °C under Ar (g) for 2 hours. The reaction was cooled to rt, diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 10% to 30%) to give tert-butyl 4-(5-cyanopyridin-2-yl)piperazine-1-carboxylate (1.05 g, 50% yield) as a white solid. LCMS ESI m/z: 370 [M+H] + . Step 3 1-(5-(benzyloxy)pyridin-2-yl)piperazine [1022] A solution of tert-butyl 4-(5-(benzyloxy)pyridin-2-yl)piperazine-1-carboxylate (370 mg, 1 mmol) in HCl/dioxane (4M, 5 mL) was stirred at rt for 1 hour. The solvent was removed in vacuo to give 1-(5-(benzyloxy)pyridin-2-yl)piperazine (240 mg, 94% yield) as a white solid. This was used without further purification. (ESI 432 [M+H] + ). LCMS ESI m/z: 270 [M+H] + . Step 4

4-(3-(4-(5-(benzyloxy)pyridin-2-yl)piperazine-1-carbonyl)-4- fluorobenzyl)-6-cyclobutoxyphthalazin- 1(2H)-one [1023] To a solution of 1-(5-(benzyloxy)pyridin-2-yl)piperazine (73 mg, 0.27 mmol) and 5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoic acid (100 mg, 0.27 mmol) in DMF (2 mL) was added EDCI (52 mg, 0.27 mmol), HOBT (55 mg, 0.41 mmol) and DIPEA (105 mg, 0.81 mmol). The reaction mixture was stirred at rt for 2 hours. The residue was purified by prep-HPLC to afford 4-(3-(4-(5-(benzyloxy)pyridin-2-yl)piperazine-1-carbonyl)-4- fluorobenzyl)-6- cyclobutoxyphthalazin-1(2H)-one (90 mg, 54% yield) as a white solid. LCMS ESI m/z: 620 [M+H] + . Step 5 6-cyclobutoxy-4-(4-fluoro-3-(4-(5-hydroxypyridin-2-yl)pipera zine-1-carbonyl)benzyl)phthalazin- 1(2H)-one [1024] A mixture of 4-(3-(4-(5-(benzyloxy)pyridin-2-yl)piperazine-1-carbonyl)-4- fluorobenzyl)-6- cyclobutoxyphthalazin-1(2H)-one (90 mg, 0.15 mmol) and Pd/C (18 mg) in MeOH (5 mL) was stirred under H 2 at room temperature for 2 hours. The mixture was filtered and concentrated in vacuo. The residue was purified by prep-HPLC to give 6-cyclobutoxy-4-(4-fluoro-3-(4-(5-hydroxypyridin-2- yl)piperazine-1-carbonyl)benzyl)phthalazin-1(2H)-one (53 mg, 67% yield) as a white solid. LCMS ESI m/z: 530 [M+H] + . 1 H–NMR (400 MHz, DMSO-d 6 ) δ 12.45 (s, 1H), 9.08 (s, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.75 (d, J = 3.2 Hz, 1H), 7.42 (d, J = 3.2 Hz, 1H) 740 – 722 (m 4H), 7.10 – 7.07 (m, 2H), 4.84 – 4.81 (m, 1H), 4.30 (s, 2H), 3.72 (s, 2H), 3.36 – 3.22 (m, 6H), 2.42 – 2.38 (m, 2H), 2.07 – 1.75 (m, 4H). Synthesis of Example 141: 5-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl) piperazin-1-yl)nicotinonitrile tert-butyl 4-(5-cyanopyridin-3-yl)piperazine-1-carboxylate [1025] A mixture of 5-bromonicotinonitrile (300 mg, 1.64 mmol), tert-butyl piperazine-1-carboxylate (458 mg, 2.46 mmol), brettphos Pd G3 (143 mg, 0.16 mmol), Xphos (313 mg, 0.65 mmol) and t-BuONa (315 mg, 3.28 mmol) in dioxane (3mL) was stirred at 110 °C under Ar (g) for 16 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 20% to 30%) to give tert-butyl 4-(5- cyanopyridin-3-yl)piperazine-1-carboxylate (138 mg, 29% yield) as a white solid. LCMS ESI m/z: 289 [M+H] + . Step 2 5-(piperazin-1-yl)nicotinonitrile [1026] A solution of tert-butyl 4-(5-cyanopyridin-3-yl)piperazine-1-carboxylate (138 mg, 0.48 mmol) in HCl/dioxane (4M, 2 mL) was stirred at 25 °C for 1 hour. The reaction mixture was concentrated in vacuo to give 5-(piperazin-1-yl)nicotinonitrile as a white solid (90 mg, 99% yield). LCMS ESI m/z: 189 [M+H] + . This was used without further purification. Step 3 5-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl) piperazin-1- yl)nicotinonitrile [1027] To a solution of 5-(piperazin-1-yl)nicotinonitrile (79 mg, 0.26 mmol) and 5-((7-cyclobutoxy-4- oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenzoic acid (80 mg, 0.22 mmol) in DMF (2 mL) was added EDCI (51 mg, 0.33 mmol), HOBt (44 mg, 0.33 mmol) and DIPEA (140 mg, 0.65 mmol). The reaction mixture was stirred at 45 °C for 1 hour. The reaction was purified by prep-HPLC to give 5-(4-(5- ((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-f luorobenzoyl)piperazin-1-yl)nicotinonitrile (36 mg, 31% yield) as a white solid. LCMS ESI m/z: 539 [M+H] + . 1 H–NMR (400 MHz, DMSO-D6) δ 12.46 (s, 1H), 8.59 (d, J = 2.9 Hz, 1H), 8.38 (d, J = 1.6 Hz, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.79 (q, J = 2.8, 1.7 Hz, 1H), 7.47 – 7.38 (m, 2H), 7.34 – 7.23 (m, 2H), 7.11 (d, J = 2.3 Hz, 1H), 4.84 (p, J = 7.1 Hz, 1H), 4.31 (s, 2H), 3.76 (s, 2H), 3.42 – 3.38 (m, 2H), 3.32 – 3.29 (m, 2H), 3.25 (s, 2H), 2.45 – 2.30 (m, 2H), 2.07 – 1.92 (m, 2H), 1.83 – 1.55 (m, 2H). Synthesis of Example 142: 6-Cyclobutoxy-4-(3-(5-(cyclopropanecarbonyl)-2,5- diazabicyclo[2.2.2]octane-2-carbonyl)-4-fluorobenzyl)phthala zin-1(2H)-one

6-cyclobutoxy-4-(3-(5-(cyclopropanecarbonyl)-2,5-diazabicycl o[2.2.2]octane-2-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [1028] To a solution of 4-(3-(2,5-diazabicyclo[2.2.2]octane-2-carbonyl)-4-fluorobenz yl)-6- cyclobutoxyphthalazin-1(2H)-one (100 mg, 0.22 mmol) and cyclopropanecarboxylic acid (22 mg, 0.26 mmol) in DMF (3 mL) was added EDCI (62 mg, 0.32 mmol), HOBt (44 mg, 0.32 mmol) and DIPEA (84 mg, 0.65 mmol). The reaction mixture was stirred at 50 o C for 1 hour. The reaction was purified by prep- HPLC to give 6-cyclobutoxy-4-(3-(5-(cyclopropanecarbonyl)-2,5-diazabicycl o[2.2.2]octane-2-carbonyl)- 4-fluorobenzyl) phthalazin-1(2H)-one (52 mg, 0.10 mmol, 45% yield) as a white solid. LCMS ESI m/z: 531.2 [M+H] + . 1 H–NMR (400 MHz, DMSO-d6) δ 12.46 (d, J = 3.7 Hz, 1H), 8.14 (dd, J = 8.8, 1.7 Hz, 1H), 7.52 – 7.41 (m, 1H), 7.37 (dd, J = 6.7, 5.0 Hz, 1H), 7.32 – 7.19 (m, 2H), 7.13 – 7.01 (m, 1H), 4.89 – 4.77 (m, 1H), 4.73 – 4.62 (m, 1H), 4.35 – 4.25 (m, 2H), 3.73 – 3.50 (m, 4H), 3.40 (d, J = 11.8 Hz, 2H), 2.44 – 2.32 (m, 2H), 2.04 – 1.91 (m, 3H), 1.82 – 1.73 (m, 3H), 1.69 – 1.56 (m, 2H), 0.78 – 0.65 (m, 4H). Synthesis of Example 143: 6-(3-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-3,8-diazabicyclo[3.2.1]octan-8-yl)nicotinonit rile tert-butyl 5-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2-fluorobenzoyl)-2,5- diazabicyclo[2.2.2]octane-2-carboxylate [1029] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (300 mg, 0.81 mmol) and tert-butyl 2,5-diazabicyclo[2.2.2]octane-2-carboxylate (173 mg, 0.81 mmol) in DMF (3 mL) was added EDCI (165 mg, 1.22 mmol), HOBt (234 mg, 1.22 mmol) and DIPEA (316 mg, 2.44 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The crude was purified by silica gel chromatography (PE: EA = 9: 1 to 1: 1) to give tert-butyl methyl(3-oxo-3-(4-(5- (trifluoromethyl)pyrimidin-2-yl)piperazin-1-yl)propyl)carbam ate (408 mg, 0.73 mmol, 89% yield) as a white solid. LCMS ESI m/z: 563.2 [M+H] + . Step 2 4-(3-(2,5-diazabicyclo[2.2.2]octane-2-carbonyl)-4-fluorobenz yl)-6-cyclobutoxyphthalazin-1(2H)-one [1030] To a solution of tert-butyl 5-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2- fluorobenzoyl)-2,5-diazabicyclo[2.2.2]octane-2-carboxylate (408 mg, 0.73 mmol) in dioxane (5 mL) was added HCl/dioxane (4M, 5 mL) and stirred at 25 °C for 1 hour. The mixture was concentrated in vacuo to afford 4-(3-(2,5-diazabicyclo[2.2.2]octane-2-carbonyl)-4-fluorobenz yl)-6-cyclobutoxyphthalazin-1(2H)- one (361 mg, 0.72 mmol, 99% yield) as a brown oil. This was used without further purification. LCMS ESI m/z: 463.2 [M+H] + . Step 3 6-(5-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-2,5- diazabicyclo[2.2.2]octan-2-yl)nicotinonitrile [1031] To a solution of 4-(3-(2,5-diazabicyclo[2.2.2]octane-2-carbonyl)-4-fluorobenz yl)-6- cyclobutoxyphthalazin-1(2H)-one (80 mg, 0.17 mmol) and 6-chloronicotinonitrile (29 mg, 0.21 mmol) in DMF (1 mL) was added DIPEA (67 mg, 0.51 mmol). The reaction mixture was stirred at 90 °C for 16 hours. The reaction was purified by prep-HPLC to give 6-(5-(5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)-2,5-diazabic yclo[2.2.2]octan-2-yl)nicotinonitrile (31 mg, 0.05 mmol, 32% yield) as a white solid. LCMS ESI m/z: 565.0 [M+H] + . 1 H–NMR (400 MHz, DMSO-d6) δ 12.46 (d, J = 7.3 Hz, 1H), 8.48 (t, J = 4.9 Hz, 1H), 8.16 (dd, J = 12.7, 8.8 Hz, 1H), 7.87 (d, J = 7.2 Hz, 1H), 7.45 (dd, J = 5.8, 2.5 Hz, 1H), 7.36 (dd, J = 7.6, 5.5 Hz, 1H), 7.34 – 7.29 (m, 1H), 7.29 – 7.18 (m, 1H), 7.08 (d, J = 1.9 Hz, 1H), 6.62 (s, 1H), 4.83 (dd, J = 14.2, 7.0 Hz, 2H), 4.32 (t, J = 14.7 Hz, 2H), 3.65 (t, J = 17.5 Hz, 3H), 3.48 (d, J = 27.7 Hz, 2H), 2.43 – 2.33 (m, 2H), 1.99 (d, J = 8.1 Hz, 2H), 1.89 (s, 3H), 1.80 – 1.59 (m, 3H). Synthesis of Example 144: 6-cyclobutoxy-4-(4-fluoro-3-(5-(5-(trifluoromethyl)pyrimidin -2-yl)-2,5- diazabicyclo [2.2.2]octane-2-carbonyl)benzyl)phthalazin-1(2H)-one Step 1

6-cyclobutoxy-4-(4-fluoro-3-(5-(5-(trifluoromethyl)pyrimidin -2-yl)-2,5-diazabicyclo [2.2.2]octane-2- carbonyl)benzyl)phthalazin-1(2H)-one [1032] To a solution of 4-(3-(2,5-diazabicyclo[2.2.2]octane-2-carbonyl)-4-fluorobenz yl)-6- cyclobutoxyphthalazin-1(2H)-one (120 mg, 0.15 mmol), 2-chloro-5-(trifluoromethyl) pyrimidine (37 mg, 0.20 mmol) in DMF (1 mL) was added DIPEA (100 mg, 0.78 mmol). The reaction mixture was stirred at 90 o C for 1 hour. The mixture was purified by prep-HPLC to give 6-cyclobutoxy-4-(4-fluoro-3-(5-(5- (trifluoromethyl)pyrimidin-2-yl)-2,5-diazabicyclo[2.2.2]octa ne-2-carbonyl) benzyl) phthalazin-1(2H)-one (62 mg, 40% yield) as a white solid. LCMS ESI m/z: 609.2 [M+H] + . 1 H–NMR (400 MHz, CDCl3) δ 10.73 (s, 1H), 8.54 (d, J = 2.9 Hz, 1H), 8.45 (dd, J = 23.1, 2.4 Hz, 1H), 8.35 (t, J = 8.0 Hz, 1H), 7.37 – 7.27 (m, 2H), 7.19 (dd, J = 8.8, 2.3 Hz, 1H), 7.07 (t, J = 8.7 Hz, 1H), 6.92 – 6.87 (m, 1H), 5.21 (d, 1H), 4.71 – 4.61 (m, 1H), 4.22 (d, J = 8.2 Hz, 2H), 3.97 – 3.71 (m, 4H), 3.58 (d, J = 11.0 Hz, 1H), 2.47 – 2.35 (m, 2H), 2.18 – 1.85 (m, 6H), 1.83 – 1.73 (m, 2H). Synthesis of Example 145: 6-cyclobutoxy-4-(4-fluoro-3-(3-(trifluoromethyl)-5,6,7,8-tet rahydro- [1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl)benzyl)phthalazin- 1(2H)-one

6-cyclobutoxy-4-(4-fluoro-3-(3-(trifluoromethyl)-5,6,7,8-tet rahydro-[1,2,4]triazolo[4,3-a]pyrazine-7- carbonyl)benzyl)phthalazin-1(2H)-one [1033] Following the procedure above in Example 1, but starting with 5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoic acid gave the title compound as a white solid. LCMS ESI m/z: 542.7 [M + H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.43 (s, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.54 – 7.47 (m, 1H), 7.44 (d, J = 5.4 Hz, 1H), 7.36 – 7.24 (m, 2H), 7.06 (d, J = 13.4 Hz, 1H), 5.02 (s, 1H), 4.86 – 4.77 (m, 1H), 4.69 (s, 1H), 4.31 (s, 2H), 4.18 (m, 2H), 3.99 (s, 1H), 3.68 (s, 1H), 2.43 – 2.31 (m, 2H), 2.05 – 1.93 (m, 2H), 1.78 (m, 1H), 1.64 (m, 1H). Synthesis of Example 146: 6-cyclobutoxy-4-(3-(3-cyclopropyl-5,6,7,8-tetrahydro-[1,2,4] triazolo[4,3- a]pyrazine-7-carbonyl)-4-fluorobenzyl)phthalazin-1(2H)-one Step 1 6-cyclobutoxy-4-(3-(3-cyclopropyl-5,6,7,8-tetrahydro-[1,2,4] triazolo[4,3-a]pyrazine-7-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [1034] Following the procedure above in Example 8, but starting with 3-cyclopropyl-5,6,7,8- tetrahydro-[1,2,4]triazolo[4,3-a]pyrazine gave the title compound as a white solid. LCMS ESI m/z: 514.8 [M + H] + . 1 H NMR (400 MHz, DMSO-d6) δ12.43 (s, 1H), 8.15 (dd, J = 8.6, 4.9 Hz, 1H), 7.46 (dd, J = 10.7, 5.8 Hz, 2H), 7.31 (t, J = 8.7 Hz, 2H), 7.07 (d, J = 9.1 Hz, 1H), 4.83 (dd, J = 14.3, 7.1 Hz, 2H), 4.53 (s, 1H), 4.31 (s, 2H), 4.09 (s, 1H), 3.82 (s, 1H), 3.62 (s, 1H), 2.36 (s, 2H), 2.04 – 1.72 (m, 4H), 1.65 (m, 1H), 1.25 (s, 1H), 1.02 – 0.93 (m, 2H), 0.87 (d, J = 2.6 Hz, 2H). Synthesis of Example 147: 6-cyclobutoxy-4-(4-fluoro-3-(5-(5-(trifluoromethyl)pyrimidin -2- yl)octahydrop yrr olo[3,4-c]pyrrole-2-carbonyl)benzyl)phthalazin-1(2H)-one O NH N O F N O N N F N F F

tert-butyl 5-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2-fluo robenzoyl)hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate [1035] To a solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (300 mg, 0.81 mmol) and tert-butyl hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (207 mg, 0.98 mmol) in DMF (2 mL) was added EDCI (234 mg, 1.22 mmol), HOBt (165 mg, 1.22 mmol) and DIPEA (526 mg, 4.07 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/pet ether = 40% to 50%) to give tert-butyl 5-(5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)hexa hydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (436 mg 95% yield) as a white solid, LCMS ESI 563 [M+H] + . Step 2

6-cyclobutoxy-4-(4-fluoro-3-(octahydropyrrolo[3,4-c]pyrrole- 2-carbo nyl)benzyl)phthalazin-1(2H)- one [1036] To a solution of tert-butyl 5-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )-2- fluorobenzoyl)hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxyla te (436 mg, 0.75 mmol) ) in dioxane (2 mL) was added HCl/dioxane (4.0M, 4 mL, 16.00 mmol). The mixture was stirred at rt for 1 hour. The solvent was removed in vacuo to give 6-cyclobutoxy-4-(4-fluoro-3-(octahydropyrrolo[3,4-c]pyrrole- 2- carbonyl)be nzyl)phthalazin-1(2H)-one (322 mg 90% yield) as a white solid. This was used without further purification. LCMS ESI 463 [M+H] + . Step 3 6-cyclobutoxy-4-(4-fluoro-3-(5-(5-(trifluoromethyl)pyrimidin -2-yl)octahydropyrr olo[3,4-c]pyrrole- 2-carbonyl)benzyl)phthalazin-1(2H)-one [1037] A mixture of 6-cyclobutoxy-4-(4-fluoro-3-(octahydropyrrolo[3,4-c]pyrrole- 2- carbonyl)benzyl)phthalazin-1(2H)-one (200 mg, 0.40 mmol), 2-chloro-5-(triflu oromethyl)pyrimidine (81 mg, 0.44 mmol) and DIPEA (207 mg, 1.60 mmol) in MeCN (3 mL) was stirred at 50°C for 1 hour. The reaction was concentrated in vacuo. The residue was purified by prep-HPLC to give the desired product 6- cyclobutoxy-4-(4-fluoro-3-(5-(5-(trifluoromethyl)pyrimidin-2 -yl)octahydropyrrolo[3,4-c]pyrrole-2- carbonyl)benzyl)phthalazin-1(2H)-one (85 mg, 35% yield) as a white solid. LCMS ESI 609.2 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ 12.45 (s, 1H), 8.70 (d, J = 2.1 Hz, 2H), 8.14 (d, J = 8.8 Hz, 1H), 7.46 – 7.34 (m, 2H), 7.33 – 7.17 (m, 2H), 7.08 (d, J = 2.3 Hz, 1H), 4.82 (p, J = 7.1 Hz, 1H), 4.34 – 4.24 (m, 2H), 3.88 – 3.65 (m, 3H), 3.54 – 3.35 (m, 4H), 3.19 – 2.94 (m, 3H), 2.37 (m, 2H), 2.10 – 1.89 (m, 2H), 1.81 – 1.57 (m, 2H). Synthesis of Example 148: 6-(6-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-2,6-diazaspiro[3.3]heptan-2-yl)nicotinonitril e tert-butyl 6-(5-cyanopyridin-2-yl)-2,6-diazaspiro[3.3]heptane-2-carboxy late [1038] A mixture of 6-chloronicotinonitrile (241 mg, 1.74 mmol), tert-butyl 2,6- diazaspiro[3.3]heptane-2-carboxylate (328 mg, 1.65 mmol) and potassium carbonate (572 mg, 4.14 mmol) in MeCN (3.00 mL) was stirred at 60 °C overnight. The reaction was diluted with H 2 O (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduce pressure. The residue was purified with silica gel chromatography using EtOAc/Heptanes from 0% to 100% to provide tert-butyl 6-(5-cyanopyridin-2-yl)- 2,6-diazaspiro[3.3]heptane-2-carboxylate (250 mg, 50%) as a white solid. 1 H NMR (400 MHz, CDCl3) δ 8.35 (d, J = 1.7 Hz, 1H), 7.57 (dd, J = 8.8, 2.2 Hz, 1H), 6.22 (d, J = 8.8 Hz, 1H), 4.21 (s, 4H), 4.11 (s, 4H), 1.42 (s, 9H). Step 2 6-(2,6-diazaspiro[3.3]heptan-2-yl)nicotinonitrile [1039] To a solution of tert-butyl 6-(5-cyanopyridin-2-yl)-2,6-diazaspiro[3.3]heptane-2-carboxy late (250 mg, 0.832 mmol) in dichlorormethane (1.5 mL) was added trifluoroacetic acid (1.5 mL). The resulting mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure to give 6-(2,6-diazaspiro[3.3]heptan-2-yl)nicotinonitrile (165 mg, 99 %) as yellow oil. LCMS ESI m/z: 201.2, [M+H] + . Step 3 6-(6-(2-fluoro-5-formylbenzoyl)-2,6-diazaspiro[3.3]heptan-2- yl)nicotinonitrile [1040] To a solution of 6-(2,6-diazaspiro[3.3]heptan-2-yl)nicotinonitrile, 2-fluoro-5-formylbenzoic acid, N,N-diisopropylethylamine (321 µL, 1.83 mmol) in MeCN (3.00 mL) was added 1- propylphosphonic acid cyclic anhydride (546 µL, 0.917 mmol) . The reaction mixture was stirred at room temperature for 5 hours. The reaction was diluted with H 2 O (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography using EtOAc/heptanes from 0% to 100 % to provide 6-(6-(2-fluoro-5-formylbenzoyl)-2,6- diazaspiro[3.3]heptan-2-yl)nicotinonitrile (234 mg, 80 %) as a white solid. LCMS ESI m/z: 351.2, [M+H] + . Step 4

6-(6-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-2,6- diazaspiro[3.3]heptan-2-yl)nicotinonitrile [1041] Following the procedure described in Example 151, and making non-critical variation as required to replace a with 6-(6-(2-fluoro-5-formylbenzoyl)-2,6-diazaspiro[3.3]heptan-2- yl)nicotinonitrile, the product of 6-(6-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-2,6- diazaspiro[3.3]heptan-2-yl)nicotinonitrile was obtained as a white solid (59 mg, 23%). LCMS ESI m/z: 551.4 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.48 (s, 1H), 8.45 (dd, J = 2.2, 0.6 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.82 (dd, J = 8.8, 2.3 Hz, 1H), 7.51 (dd, J = 6.6, 2.1 Hz, 1H), 7.48 – 7.40 (m, 1H), 7.30 (dd, J = 8.8, 2.3 Hz, 1H), 7.24 (dd, J = 9.8, 8.7 Hz, 1H), 7.12 (d, J = 2.3 Hz, 1H), 6.43 (d, J = 8.8 Hz, 1H), 4.95 – 4.78 (m, 1H), 4.30 (s, 2H), 4.23 (t, J = 4.7 Hz, 4H), 4.18 (d, J = 7.7 Hz, 4H), 2.37 (dt, J = 9.2, 8.0 Hz, 2H), 2.07 – 1.92 (m, 2H), 1.84 – 1.57 (m, 2H). Synthesis of Example 149: 6-(2-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-2,5-diazaspiro[3.4]octan-5-yl)nicotinonitrile

tert-butyl 5-(5-cyanopyridin-2-yl)-2,5-diazaspiro[3.4]octane-2-carboxyl ate [1042] A solution of 6-chloronicotinonitrile (261 mg, 1.880 mmol), tert-butyl 2,5- diazaspiro[3.4]octane-2-carboxylate (400 mg, 1.880 mmol) and DIPEA (0.9 mL) in NMP (3.0 mL) was stirred at 120 ℃. The reaction was diluted with H 2 O (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography using EtOAc/heptanes from 0% to 100 % to afford tert-butyl 5-(5-cyanopyridin-2-yl)-2,5-diazaspiro[3.4]octane-2-carboxyl ate (382 mg, 57 %) as a white solid.1H NMR (400 MHz, CDCl 3 ) δ 8.39 (d, J = 1.8 Hz, 1H), 7.62 (dd, J = 8.9, 2.3 Hz, 1H), 6.46 (d, J = 8.7 Hz, 1H), 4.77 (d, J = 8.3 Hz, 2H), 3.69 (d, J = 8.5 Hz, 2H), 3.45 (t, J = 6.6 Hz, 2H), 2.31 (t, J = 6.8 Hz, 2H), 2.01 – 1.83 (m, 2H), 1.48 (s, 9H). Step 2 6-(2,5-diazaspiro[3.4]octan-5-yl)nicotinonitrile [1043] Following the procedure described in Example 148, and making non-critical variation as required to replace tert-butyl 6-(5-cyanopyridin-2-yl)-2,6-diazaspiro[3.3]heptane-2-carboxy late with tert- butyl 5-(5-cyanopyridin-2-yl)-2,5-diazaspiro[3.4]octane-2-carboxyl ate, the crude product of 6-(2,5- diazaspiro[3.4]octan-5-yl)nicotinonitrile was obtained as a yellow oil (172 mg, 99 %). LCMS ESI m/z: 215.3, [M+H] + . Step 3 6-(2-(2-fluoro-5-formylbenzoyl)-2,5-diazaspiro[3.4]octan-5-y l)nicotinonitrile [1044] Following the procedure described in Example 148, and making non-critical variation as required to replace 6-(2,6-diazaspiro[3.3]heptan-2-yl)nicotinonitrile with 6-(2,5-diazaspiro[3.4]octan-5- yl)nicotinonitrile, 6-(2-(2-fluoro-5-formylbenzoyl)-2,5-diazaspiro[3.4]octan-5-y l)nicotinonitrile was obtained as a white solid (172 mg, 99 %). LCMS ESI m/z: 365.3, [M+H] + . Step 4 6-(2-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-2,5- diazaspiro[3.4]octan-5-yl)nicotinonitrile [1045] Following the procedure described in Example 148, and making non-critical variation as required to replace 6-(6-(2-fluoro-5-formylbenzoyl)-2,6-diazaspiro[3.3]heptan-2- yl)nicotinonitrile with 6- (2-(2-fluoro-5-formylbenzoyl)-2,5-diazaspiro[3.4]octan-5-yl) nicotinonitrile, 6-(2-(5-((7-cyclobutoxy-4- oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)-2,5- diazaspiro[3.4]octan-5-yl)nicotinonitrile was obtained as a white solid (351 mg, 79 %). LCMS ESI m/z: 565.3, [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.70 – 12.19 (m, 1H), 8.77 – 8.38 (m, 1H), 8.28 – 8.05 (m, 1H), 8.02 – 7.80 (m, 1H), 7.55 – 7.33 (m, 2H), 7.33 – 7.16 (m, 2H), 7.16 – 6.98 (m, 1H), 6.98 – 6.70 (m, 1H), 4.95 – 4.59 (m, 1H), 4.41 – 4.12 (m, 2H), 3.85 (d, J = 48.3 Hz, 1H), 3.65 – 3.32 (m, 2H), 3.20 – 2.85 (m, 2H), 2.34 (d, J = 4.4 Hz, 1H), 2.00 – 1.87 (m, 2H), 1.77 – 1.56 (m, 2H), 1.35 – 1.17 (m, 2H), 0.77 (ddd, J = 26.3, 13.2, 6.5 Hz, 1H), 0.38 (ddd, J = 15.0, 10.2, 4.9 Hz, 1H). Synthesis of Example 150: 6-(5-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-2,5-diazabicyclo[4.1.0]heptan-2-yl)nicotinoni trile tert-butyl 5-(5-cyanopyridin-2-yl)-2,5-diazabicyclo[4.1.0]heptane-2-car boxylate [1046] Following the procedure described in Example 148, and making non-critical variation as required to replace tert-butyl 2,6-diazaspiro[3.3]heptane-2-carboxylate with, tert-butyl 5-(5-cyanopyridin- 2-yl)-2,5-diazabicyclo[4.1.0]heptane-2-carboxylate was obtained as a white solid (330 mg, 66%). LCMS ESI m/z: 301.3, [M+H] + . Step 2 6-(2,5-diazabicyclo[4.1.0]heptan-2-yl)nicotinonitrile [1047] Following the procedure described in Example 148, and making non-critical variation as required to replace tert-butyl 6-(5-cyanopyridin-2-yl)-2,6-diazaspiro[3.3]heptane-2-carboxy late with tert- butyl 5-(5-cyanopyridin-2-yl)-2,5-diazabicyclo[4.1.0]heptane-2-car boxylate, the crude product of 6-(2,5- diazabicyclo[4.1.0]heptan-2-yl)nicotinonitrile was obtained as a yellow oil (218 mg, 99%). LCMS ESI m/z: 301.3, [M+H] + . Step 3 6-(5-(2-fluoro-5-formylbenzoyl)-2,5-diazabicyclo[4.1.0]hepta n-2-yl)nicotinonitrile [1048] Following the procedure described in Example 148, and making non-critical variation as required to replace 6-(2,6-diazaspiro[3.3]heptan-2-yl)nicotinonitrile with 6-(2,5- diazabicyclo[4.1.0]heptan-2-yl)nicotinonitrile, 6-(5-(2-fluoro-5-formylbenzoyl)-2,5- diazabicyclo[4.1.0]heptan-2-yl)nicotinonitrile was obtained as a white solid (280 mg, 73 %). LCMS ESI m/z: 351.3, [M+H] + . Step 4 6-(5-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-2,5- diazabicyclo[4.1.0]heptan-2-yl)nicotinonitrile [1049] Following the procedure described in Example 148, and making non-critical variation as required to replace 6-(6-(2-fluoro-5-formylbenzoyl)-2,6-diazaspiro[3.3]heptan-2- yl)nicotinonitrile with 6- (5-(2-fluoro-5-formylbenzoyl)-2,5-diazabicyclo[4.1.0]heptan- 2-yl)nicotinonitrile, 6-(5-(5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoyl)-2,5-diazabicyclo[4.1.0]heptan- 2-yl)nicotinonitrile was obtained as a white solid (85 mg, 29 %). LCMS ESI m/z: 551.3, [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.70 – 12.19 (m, 1H), 8.77 – 8.38 (m, 1H), 8.28 – 8.05 (m, 1H), 8.02 – 7.80 (m, 1H), 7.55 – 7.33 (m, 2H), 7.33 – 7.16 (m, 2H), 7.16 – 6.98 (m, 1H), 6.98 – 6.70 (m, 1H), 4.95 – 4.59 (m, 1H), 4.41 – 4.12 (m, 2H), 3.85 (d, J = 48.3 Hz, 1H), 3.65 – 3.32 (m, 2H), 3.20 – 2.85 (m, 2H), 2.34 (d, J = 4.4 Hz, 1H), 2.00 – 1.87 (m, 2H), 1.77 – 1.56 (m, 2H), 1.35 – 1.17 (m, 2H), 0.77 (ddd, J = 26.3, 13.2, 6.5 Hz, 1H), 0.38 (ddd, J = 15.0, 10.2, 4.9 Hz, 1H). Synthesis of Example 151: 6-(7-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-2,7-diazaspiro[4.4]nonan-2-yl)nicotinonitrile formate salt tert-butyl 7-(5-cyanopyridin-2-yl)-2,7-diazaspiro[4.4]nonane-2-carboxyl ate [1050] A mixture of 6-chloronicotinonitrile (77 mg, 0.55 mmol), 6-(2,7-diazaspiro[4.4]nonan-2- yl)nicotinonitrile (130 mg, 0.55 mmol) and DIPEA (0.20 mL, 1.14 mmol) in NMP (2 mL) was stirred at 120 °C for 24 hours. The mixture was cooled to rt, diluted with H 2 O (50 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/Heptanes = 0 to 50%) to give tert-butyl 7-(5-cyanopyridin-2-yl)-2,7-diazaspiro[4.4]nonane-2-carboxyl ate (178 mg, 0.54 mmol, 99%) as a clear oil. LCMS ESI m/z: 329.3 [M+H] + . Step 2 6-(2,7-diazaspiro[4.4]nonan-2-yl)nicotinonitrile [1051] A mixture of tert-butyl 7-(5-cyanopyridin-2-yl)-2,7-diazaspiro[4.4]nonane-2-carboxyl ate (178 mg, 0.54 mmol), TFA (1.8 mL) in DCM (1.8 mL) was stirred at rt for 1 hour. The mixture concentrated in vacuo to give 6-(2,7-diazaspiro[4.4]nonan-2-yl)nicotinonitrile (124 mg, 0.54 mmol, 99%) as a clear oil that was carried forward without further purification. LCMS ESI m/z: 229.3 [M+H] + . Step 3 6-(7-(2-fluoro-5-formylbenzoyl)-2,7-diazaspiro[4.4]nonan-2-y l)nicotinonitrile [1052] A mixture of 6-(2,7-diazaspiro[4.4]nonan-2-yl)nicotinonitrile (124 mg, 0.54 mmol), 2-fluoro-5- formylbenzoic acid (91 mg, 0.54 mmol), T3P (0.44 mL, 0.60 mmol), and DIPEA (0.48 mL, 2.72 mmol) in MeCN (2.2 mL) was stirred at rt for 24 hours. The mixture was diluted with H 2 O (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/Heptanes = 0 to 100%) to give 6-(7-(2-fluoro-5-formylbenzoyl)-2,7-diazaspiro[4.4]nonan-2- yl)nicotinonitrile (21 mg, 0.06 mmol, 8%) as a clear oil. LCMS ESI m/z: 379.3 [M+H] + . Step 4 6-(7-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-2,7- diazaspiro[4.4]nonan-2-yl)nicotinonitrile formate salt [1053] A mixture of dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (17 mg, 0.056 mmol), 6-(7-(2-fluoro-5-formylbenzoyl)-2,7-diazaspiro[4.4]nonan-2-y l)nicotinonitrile (21 mg, 0.056 mmol) and Et 3 N (0.02 mL, 0.17 mmol) in THF (0.6 mL) was stirred at rt for 18 hours. To the reaction mixture was added N O (0.01 mL, 0.32 mmol), then stirred at 70 °C for 1 hour. The reaction mixture was concentrated in vacuo and the resulting crude was subjected to reverse phase column chromatography (MeCN: AMF = 0 to 100%) to afford 6-(7-(5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)-2,7-diazaspi ro[4.4]nonan-2-yl)nicotinonitrile formate salt as a white solid (2 mg, 0.004 mmol, 7%). LCMS ESI m/z: 579.5 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.46 (s, 1H), 8.46 (d, J = 8.2 Hz, 1H), 8.24 (s, 1H), 8.14 (dd, J = 8.7, 7.8 Hz, 1H), 7.82 (ddd, J = 11.3, 9.0, 2.1 Hz, 1H), 7.47 – 7.36 (m, 2H), 7.32 – 7.27 (m, 1H), 7.26 – 7.20 (m, 1H), 7.07 (dd, J = 27.6, 2.2 Hz, 1H), 6.57 – 6.49 (m, 1H), 4.86 – 4.77 (m, 1H), 4.29 (d, J = 10.2 Hz, 2H), 3.63 – 3.50 (m, 2H), 3.49 (d, J = 4.3 Hz, 1H), 3.30 – 3.22 (m, 3H), 3.11 (s, 1H), 2.41 – 2.32 (m, 4H), 2.14 – 1.77 (m, 6H), 1.77 – 1.59 (m, 1H). Synthesis of Example 152: 6-(7-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)-1,7-diazaspiro[3.5]nonan-1-yl)nicotinonitrile tert-Butyl 1-(5-cyanopyridin-2-yl)-1,7-diazaspiro[3.5]nonane-7-carboxyl ate [1054] The solution of 6-Chloronicotinonitrile (148 mg, 1.07 mmol) , tert-Butyl 1,7- diazaspiro[3.5]nonane-7-carboxylate (250 mg, 1.07 mmol) and DIPEA (0.3 mL) in NMP (3.0 mL) was stirred o/n at 120 ℃. The reaction was diluted with H 2 O (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/heptanes = 0 to 100 %) to give tert- Butyl 1-(5-cyanopyridin-2-yl)-1,7-diazaspiro[3.5]nonane-7-carboxyl ate (271 mg, 77 % yield). LCMS ESI 329.3 [M+H] + . Step 2 6-(1,7-diazaspiro[3.5]nonan-1-yl)nicotinonitrile [1055] The solution of tert-Butyl 1-(5-cyanopyridin-2-yl)-1,7-diazaspiro[3.5]nonane-7-carboxyl ate (271 mg, 824 umol), TFA (2 mL) and CH 2 Cl 2 (2 mL) was stirred at rt for 1h. The reaction mixture was concentrated in vacuo to obtain 6-(1,7-diazaspiro[3.5]nonan-1-yl)nicotinonitrile (172 mg, 99 % yield). LCMS ESI 229.3 [M+H] + .. Step 3 6-(7-(2-Fluoro-5-formylbenzoyl)-1,7-diazaspiro[3.5]nonan-1-y l)nicotinonitrile [1056] To a solution of 2-fluoro-5-formylbenzoic acid (127 mg, 753 umol) , N,N- Diisopropylethylamine (290 uL, 1.66 mmol) , 1-Propylphosphonic acid cyclic anhydride (493 uL, 829 umol) in MeCN (3.00 mL) was added 6-(1,7-diazaspiro[3.5]nonan-1-yl)nicotinonitrile (172 mg, 753 umol) . The reaction mixture was stirred o/n at rt. The reaction was diluted with H 2 O (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/heptanes = 0 to 100 %) to give 6-(7-(2-fluoro-5-formylbenzoyl)-1,7-diazaspiro[3.5]nonan-1- yl)nicotinonitrile (246 mg, 86 %). LCMS ESI 379.3 [M+H] + .. Step 4 6-(7-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)-1,7- diazaspiro[3.5]nonan-1-yl)nicotinonitrile [1057] Following the procedure described in Example 148, and making non-critical variation as required to replace 6-(6-(2-fluoro-5-formylbenzoyl)-2,6-diazaspiro[3.3]heptan-2- yl)nicotinonitrile with 6- (7-(2-fluoro-5-formylbenzoyl)-1,7-diazaspiro[3.5]nonan-1-yl) nicotinonitrile; 6-(7-(5-((7-cyclobutoxy-4- oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluorobenzoyl)-1,7- diazaspiro[3.5]nonan-1-yl)nicotinonitrile was obtained as a white solid (51.3 mg, 23 % yield). LCMS (ESI) m/z: 579.4 [M+H] + . 1 H NMR (400 MHz, 100 °C DMSO-d 6 ) δ 12.10 (s, 1H), 8.35 (d, J = 1.8 Hz, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.70 (dd, J = 8.8, 2.3 Hz, 1H), 7.45 – 7.36 (m, 1H), 7.31 (dd, J = 6.4, 2.0 Hz, 1H), 7.26 (dd, J = 8.8, 2.3 Hz, 1H), 7.19 (t, J = 9.0 Hz, 1H), 7.06 (d, J = 2.3 Hz, 1H), 6.30 (d, J = 8.8 Hz, 1H), 5.66 (s, 2H), 4.81 (p, J = 6.9 Hz, 1H), 4.30 (s, 2H), 3.94 (t, J = 7.5 Hz, 2H), 2.96 (s, 3H), 2.45 – 2.26 (m, 6H), 2.15 – 1.97 (m, 2H), 1.91 – 1.63 (m, 4H). Synthesis of Example 154: 2-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2- fluorobenzoyl)piperazin-1-yl)pyrimidine-5-carbonitrile

2-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2-fluorobenzoyl)piperazin-1- yl)pyrimidine-5-carbonitrile [1058] Following the general three-step procedure above in Example 130, but starting with 2- chloropyrimidine-5-carbonitrile gave the title compound as a white solid. LCMS ESI m/z: 539.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ12.44 (s, 1H), 8.79 (s, 2H), 8.16 (d, J = 8.8 Hz, 1H), 7.44 – 7.40 (m, 2H), 7.33 – 7.24 (m, 2H), 7.10 (d, J = 2.3 Hz, 1H), 4.85 (m, 1H), 4.31 (s, 2H), 3.93 (s, 2H),3.75 (m, 4H), 3.31 – 3.29 (m, 2H), 2.44 – 2.36 (m, 2H), 2.04 – 1.97 (m, 2H), 1.80 – 1.64 (m,2H). Synthesis of Example 155: 6-Cyclobutoxy-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyridin-2 - yl)piperazine-1-carbonyl)benzyl)phthalazin-1(2H)-one

tert-butyl 4-(5-(trifluoromethyl)pyridin-2-yl)piperazine-1-carboxylate [1059] 2-chloro-5-(trifluoromethyl)pyridine (200 mg, 1.10 mmol, 141.14 μL), tert-butyl piperazine-1- carboxylate (271.34 mg, 1.10 mmol), Cs 2 CO 3 (1.79 g, 5.51 mmol) and NMP (2 mL) were added to a 10 mL sealed tube. The resultant mixture was stirred at 150 °C for 1 hr under microwave. The mixture was poured into water (10 mL), extracted with EA (10 mL x 2). The organic layer was washed with brine, dried and concentrated to a crude residue. The residue was purified by silica gel chromatography (Petroleum ether: EtOAc = 10: 1 to 1: 1) to give tert-butyl 4-[5-(trifluoromethyl)-2-pyridyl]piperazine-1- carboxylate (180 mg, 34.0% yield) as a yellow oil. LCMS ESI m/z: 276.1 [M+H-56] + . Step 2 and 3 6-cyclobutoxy-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyridin-2 -yl)piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one [1060] Following the general two-step procedure above in Example 130, but starting with tert-butyl 4- (5-(trifluoromethyl)pyridin-2-yl)piperazine-1-carboxylate gave the title compound as a white solid. LCMS ESI m/z: 581.7 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.44 (s, 1H), 8.43 (s, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.84 (dd, J = 9.1,2.4 Hz, 1H), 7.43 (t, J = 66 Hz 2H) 734 – 7.23 (m, 2H), 7.10 (d, J = 2.3 Hz, 1H), 6.95 (m,1H), 4.88 – 4.78 (m, 1H), 4.31 (s, 2H), 3.73 (s, 4H), 3.58 (s, 2H), 3.31 – 3.27 (m, 2H), 2.39 (m, 2H), 2.00 (m, 2H), 1.70 (m, 2H). Synthesis of Example 156: 6-Cyclobutoxy-4-(4-fluoro-3-(4-(4-(trifluoromethyl)pyridin-2 - yl)piperazine-1-carbonyl)benzyl)phthalazin-1(2H)-one Step 1 tert-butyl 4-(4-(trifluoromethyl)pyridin-2-yl)piperazine-1-carboxylate [1061] 2-chloro-4-(trifluoromethyl)pyridine (100 mg, 550.83 μmol, 70.87 μL), tert-butyl piperazine-1- carboxylate (135.67 mg, 550.83 μmol), Cs 2 CO 3 , (538.42 mg, 1.65 mmol) and NMP (1 mL) were added to a 10 mL sealed tube. The reaction mixture was stirred under microwave at 130 °C for 2 hr .The mixture was poured into H 2 O (10 mL), extracted with EA (10 mL *2). The combined organic layers were washed with brine, dried and concentrated to give a crude product tert-butyl 4-(4-(trifluoromethyl)pyridin-2- yl)piperazine-1-carboxylate as a yellow solid. LCMS ESI m/z: 332.2 [M+H] + . Step 2 and 3

6-cyclobutoxy-4-(4-fluoro-3-(4-(4-(trifluoromethyl)pyridin-2 -yl)piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one [1062] Following the general two-step procedure above in Example 130, but starting with tert-butyl 4- (4-(trifluoromethyl)pyridin-2-yl)piperazine-1-carboxylate gave the title compound as a white solid. LCMS ESI m/z: 582.2 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.45 (s, 1H), 8.35 (d, J = 5.1 Hz, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.46 –7.39 (m, 2H), 7.29 (m, 2H), 7.13 – 7.09 (m, 2H), 6.94 (d, J = 5.2 Hz, 1H), 4.89 –4.81 (m, 1H), 4.31 (s, 2H), 3.72 (d, J = 7.5 Hz, 4H), 3.55 (s, 2H), 3.30 (d, J = 4.9 Hz, 2H), 2.40 (m, 2H), 2.08 – 1.97 (m, 2H), 1.83 – 1.65 (m, 2H). Synthesis of Example 157: 6-cyclobutoxy-4-(3-(4-(5-(difluoromethyl)pyrimidin-2-yl)pipe razine-1- carbonyl)-4-fluorobenzyl)phthalazin-1(2H)-one Step 1, 2-Chloro-5-(difluoromethyl)pyrimidine [1063] To a solution of 2-chloropyrimidine-5-carbaldehyde (200 mg, 1.40 mmol) in DCM (5 mL) was added DAST (2.26 g, 14.03 mmol, 1.85 mL), and then the mixture was stirred at rt for 5 hr. The reaction mixture was poured into sat. NaHCO 3 (20 mL) , extracted with DCM (10 mL). The organic layer was washed with brine, dried and concentrated to give 2-chloro-5-(difluoromethyl)pyrimidine (235 mg, crude) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ) δ 8.79 (s, 2H), 6.78 (t, J = 55.1 Hz, 1H). Step 2, 3 and 4 6-Cyclobutoxy-4-(3-(4-(5-(difluoromethyl)pyrimidin-2-yl)pipe razine-1-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [1064] Following the general three-step procedure above in Example 130, but starting with 2-chloro-5- (difluoromethyl)pyrimidine gave the title compound as a white solid. LCMS ESI m/z: 564.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.45 (s, 1H), 8.58 (s, 2H), 8.16 (d, J = 8.8 Hz, 1H), 7.45 – 7.38 (m, 2H), 7.34 – 7.22 (m, 2H), 7.13 – 6.83 (m, 2H), 4.90 – 4.76 (m, 1H), 4.31 (s, 2H), 3.89 (s, 2H), 3.73 (s, 4H), 3.30 (s, 2H), 2.45 – 2.35 (m, 2H), 2.07 – 1.95 (m, 2H), 1.84 – 1.61 (m, 2H). Synthesis of Example 158: 6-cyclobutoxy-4-(4-fluoro-3-(4-(6-(trifluoromethyl)pyridazin -3- yl)piperazine-1-carbonyl)benzyl)phthalazin-1(2H)-one

6-Cyclobutoxy-4-(4-fluoro-3-(4-(6-(trifluoromethyl)pyridazin -3-yl)piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one [1065] Following the general three-step procedure above in Example 130, but starting with 3-chloro-6- (trifluoromethyl)pyridazine gave the title compound as a white solid. LCMS ESI m/z: 582.7 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 8.37 (d, J = 8.8 Hz, 1H), 7.54 (d, J = 14.2 Hz, 1H), 7.35 (dd, J = 11.7, 5.4 Hz, 2H), 7.26 (s, 1H),7.24 (s, 1H), 7.09 (t, J = 8.7 Hz, 1H), 7.01 – 6.93 (m, 2H), 4.74– 4.67 (m, 1H), 4.27 (s, 2H), 3.95 (s, 2H), 3.84 (t, J = 4.8 Hz, 4H), 3.51 (s, 2H), 2.44 (s, 2H), 2.23 – 2.12 (m, 2H), 1.91 (m, 1H), 1.82 – 1.73 (m, 1H). Synthesis of Example 159: 6-Cyclobutoxy-4-(4-fluoro-3-(4-(6-(trifluoromethyl)pyridin-3 - yl)piperazine-1-carbonyl)benzyl)phthalazin-1(2H)-one

6-cyclobutoxy-4-(4-fluoro-3-(4-(6-(trifluoromethyl)pyridin-3 -yl)piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one [1066] Following the general three-step procedure above in Example 130, but starting with 5-chloro-2- (trifluoromethyl)pyridine gave the title compound as a white solid. LCMS ESI m/z: 581.7 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.45 (s, 1H), 8.43 (d, J = 2.8 Hz, 1H), 8.17 (d, J = 8.8 Hz, 1H), 7.68 (d, J = 8.8 Hz, 1H), 7.48 – 7.39 (m, 3H), 7.35 – 7.23 (m, 2H), 7.12 (d, J = 2.3 Hz, 1H), 4.89 – 4.79 (m, 1H), 4.32 (s, 2H), 3.79 (s, 2H), 3.47 (s, 2H), 3.34 (s, 2H), 3.31 – 3.26 (m, 2H), 2.39 (m, 2H), 2.00 (m, 2H), 1.72 (m, 2H). Synthesis of Example 160: 6-cyclobutoxy-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrazin-2 - yl)piperazine-1-carbonyl)benzyl)phthalazin-1(2H)-one

2-(Piperazin-1-yl)-5-(trifluoromethyl)pyrazine [1067] To a solution of 2-chloro-5-(trifluoromethyl)pyrazine (100 mg, 547.85 μmol) in NMP (3 mL) was added tert-butyl piperazine-1-carboxylate (102.04 mg, 547.85 μmol), DIPEA (354.02 mg, 2.74 mmol, 477.12 μL), and then the sealed tube was irradiated microwave at 150 °C for 2 hr. The reaction mixture was diluted in EA (10 mL), washed with water (10 mL* 2), brine, dried and concentrated. The residue was purified by silica gel chromatography to give 2-piperazin-1-yl-5-(trifluoromethyl)pyrazine (65 mg, 51.0% yield) as a grey solid. LCMS ESI m/z: 233.1 [M + H] + . Step 2, 6-cyclobutoxy-4-(4-fluoro-3-(4-(5-(trifluoromethyl)pyrazin-2 -yl)piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one [1068] Following the general one-step procedure above in Example 130, but starting with 2- (piperazin-1-yl)-5-(trifluoromethyl)pyrazine gave the title compound as a white solid. LCMS ESI m/z: 582.7 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.44 (s, 1H), 8.51 (s, 1H), 8.42 (s, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.43 (dd, J = 9.5, 6.0 Hz, 2H), 7.35 – 7.23 (m, 2H), 7.11 (d, J = 2.3 Hz, 1H), 4.90 – 4.78 (m, 1H), 4.31 (s, 2H), 3.82 (d, J = 5.3 Hz, 2H), 3.76 (s, 2H), 3.67 (s, 2H), 3.33 (s, 2H), 2.45 – 2.35 (m, 2H), 2.07 – 1.95 (m, 2H), 1.84 – 1.61 (m, 2H). Synthesis of Example 161: 6-cyclobutoxy-4-(3-(4-(5-(1,1-difluoroethyl)pyrimidin-2-yl)p iperazine-1- carbonyl)-4-fluorobenzyl)phthalazin-1(2H)-one 2-Chloro-5-(1-ethoxyvinyl)pyrimidine [1069] To a solution of tributyl(1-ethoxyvinyl)stannane (3.73 g, 10.34 mmol, 3.49 mL) in DMF (15 mL) was added 5-bromo-2-chloro-pyrimidine (2.0 g, 10.34 mmol), Pd(PPh 3 ) 2 Cl 2 (362.87 mg, 516.99 μmol). The reaction mixture was heated to 100 °C for 3 hr. Water (50 mL) and KF (1 g) was added to the reaction mixture, the mixture was stirred at rt for another 10 minutes, extracted with EtOAc (50 mL). The organic layer was washed with brine, dried and concentrated to give 2-chloro-5-(1- ethoxyvinyl)pyrimidine (2.2 g, crude) as a brown solid. Step 2, 1-(2-Chloropyrimidin-5-yl)ethanone [1070] To a mixture of 2-chloro-5-(1-ethoxyvinyl)pyrimidine (2.2 g, 11.92 mmol) in H 2 O (40 mL) was added HCl/EtOAc (4 M, 10 mL), and then stirred at rt for 1 hr. The mixture was extracted with EtOAc (40 mL), the organic layer was washed with brine, dried, and concentrated to the residue. The residue was purified by silica gel chromatography to give 1-(2-chloropyrimidin-5-yl)ethanone (900 mg, 48% yield) as a yellow solid. LCMS ESI m/z: 157.1 [M+H] + . Step 3, 2-chloro-5-(1,1-difluoroethyl)pyrimidine [1071] To a solution of 1-(2-chloropyrimidin-5-yl)ethanone (500 mg, 3.19 mmol) in DCM (10 mL) was added DAST (5.15 g, 31.93 mmol, 4.22 mL). The reaction mixture was stirred at rt for 48 hr. The mixture was cooled to 0°C, and sat. NaHCO 3 was added. The organic phase was partitioned, washed with brine, dried and concentrated to a residue. The residue was purified by silica gel chromatography to give 2-chloro-5-(1,1-difluoroethyl)pyrimidine (400 mg, 70.1% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 8.77 (s, 2H), 2.00 (t, J = 18.4 Hz, 3H). Step 4, 5 and 6 6-Cyclobutoxy-4-(3-(4-(5-(1,1-difluoroethyl)pyrimidin-2-yl)p iperazine-1-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [1072] Following the general three-step procedure above in Example 130, but starting with 2-chloro-5- (1,1-difluoroethyl)pyrimidine gave the title compound as a white solid. LCMS ESI m/z: 578.7 [M+H] + . 1 tert-Butyl 4-(5-cyclopropylpyrimidin-2-yl)piperazine-1-carboxylate [1073] A mixture of tert-butyl 4-(5-bromopyrimidin-2-yl)piperazine-1-carboxylate (500 mg, 1.46 mmol), cyclopropylboronic acid (150.16 mg, 1.75 mmol), dicesium carbonate (949.30 mg, 2.91 mmol), cyclopentyl(diphenyl)phosphane dichloromethane dichloropalladium iron (1.43 g, 1.75 mmol) and water (0.5 mL) in Dioxane (2 mL) was stirred under N 2 at 100 °C for 16 hr . The reaction mixture was concentrated to a residue. The residue was purified by silica gel chromatography (Petroleum ether: EtOAc = 10:1) to give tert-butyl 4-(5-cyclopropylpyrimidin-2-yl)piperazine-1-carboxylate (200 mg, 45% yield) as a white solid. LCMS ESI m/z: 305.2 [M + H] + . Step 2 and 3 6-Cyclobutoxy-4-(3-(4-(5-cyclopropylpyrimidin-2-yl)piperazin e-1-carbonyl)-4- fluorobenzyl)phthalazin-1(2H)-one [1074] Following the general two-step procedure above in Example 130, but starting with tert-butyl 4- (5-cyclopropylpyrimidin-2-yl)piperazine-1-carboxylate gave the title compound as a white solid. LCMS ESI m/z: 555.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.44 (s, 1H), 8.19 (s, 2H), 8.15 (d, J = 8.8 Hz, 1H), 7.41 (dd, J = 6.1, 2.0 Hz, 2H), 7.31 (dd, J =8.8, 2.1 Hz, 1H), 7.25 (t, J = 9.3 Hz, 1H), 7.10 (d, J = 2.1 Hz,1H), 4.88 – 4.77 (m, 1H), 4.31 (s, 2H), 3.78 – 3.58 (m, 6H), 3.25 (s, 2H), 2.39 (m, 2H), 2.00 (m, 2H), 1.81 – 1.72 (m, 2H), 1.66 (dd, J = 19.0, 9.1 Hz, 1H), 0.91 – 0.84 (m, 2H), 0.67 – 0.60 (m, 2H). Synthesis of Example 163: 6-Cyclobutoxy-4-(3-(3-(difluoromethyl)-5,6,7,8-tetrahydro- [1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl)-4-fluorobenzyl)ph thalazin-1(2H)-one

tert-Butyl 3-formyl-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine-7(8H)-ca rboxylate [1075] A solution of tert-butyl 3-(hydroxymethyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine -7(8H)- carboxylate (0.2 g, 1.30 mmol) and MnO 2 (1.13 g, 12.97 mmol) in DCM (5 mL) was stirred for 16 hr at 25 °C. The reaction mixture was filtered through a pad of celite, and the filtrate was concentrated to give tert-butyl 3-formyl-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine-7(8H)-ca rboxylate (0.2 g, 91% yield). LCMS ESI m/z: 252.9 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 9.97 (s, 1H), 4.80 (br s, 2H), 4.04 – 4.02 (m, 2H), 3.78 – 3.74 (m, 2H), 1.44 (s, 9H). Step 2, 3 and 4 6-Cyclobutoxy-4-(3-(3-(difluoromethyl)-5,6,7,8-tetrahydro-[1 ,2,4]triazolo[4,3-a]pyrazine-7- carbonyl)-4-fluorobenzyl)phthalazin-1(2H)-one [1076] Following the general three-step procedure above in Example 130, but starting with tert-butyl 3-formyl-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine-7(8H)-ca rboxylate gave the title compound as a white solid. LCMS ESI m/z: 525.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.43 (s, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.54 – 7.43 (m, 2H), 7.40 – 7.15 (m, 3H), 7.07 (s, 1H), 5.00 (s, 1H), 4.89 – 4.79 (m, 1H), 4.66 (s, 1H), 4.31 (s, 2H), 4.25 – 3.94 (m, 3H), 3.69 (s, 1H), 2.37 (s, 2H), 1.99 (m, 2H), 1.84 – 1.70 (m, 1H), 1.69 – 1.58 (m, 1H). Synthesis of Example 164: 6-Cyclobutoxy-4-(4-fluoro-3-(3-(fluoromethyl)-5,6,7,8-tetrah ydro- [1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl)benzyl)phthalazin- 1(2H)-one Step 1 tert-Butyl 3-(fluoromethyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine- 7(8H)-carboxylate [1077] To a solution of tert-butyl 3-(hydroxymethyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine -7(8H)- carboxylate (100 mg, 393.26 μmol) in dry DCM (4.0 mL) was added dropwise DAST (633.90 mg, 3.93 mmol, 519.59 μL). The reaction mixture was stirred at 30 °C for 2 hr. The mixture was poured into sat. NaHCO 3 , extracted with DCM (10 mL x 3). The extracts were washed with brine, dried over Na 2 SO 4 , filtered and concentrated. The crude was purified by silica gel chromatography to give tert-butyl 3- (fluoromethyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine-7( 8H)-carboxylate (70 mg, 65.9% yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 5.64 (d, J = 48 Hz, 1H), 4.71 (br, 2H), 4.09 –4.06 (m, 2H), 3.81 –3.78 (m, 2H), 1.49 (s, 9H). Step 2 and 3

6-Cyclobutoxy-4-(4-fluoro-3-(3-(fluoromethyl)-5,6,7,8-tetrah ydro-[1,2,4]triazolo[4,3-a]pyrazine-7- carbonyl)benzyl)phthalazin-1(2H)-one [1078] Following the general two-step procedure above in Example 130, but starting with tert-butyl 3- (fluoromethyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine-7( 8H)-carboxylate gave the title compound as a white solid. LCMS ESI m/z: 506.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ12.43 (s, 1H), 8.16 (d, J = 8.7 Hz, 1H), 7.52 – 7.42 (m, 2H), 7.34 – 7.26 (m, 2H), 7.08 (s, 1H), 5.59 (m, 2H), 5.00 – 4.59 (m, 3H), 4.32 (s, 2H), 4.21 – 3.86 (m, 3H), 3.67 (s, 1H), 2.37 (s, 2H), 2.06 – 1.93 (m, 2H), 1.70 (m, 2H). Synthesis of Example 165: 6-Cyclobutoxy-4-(4-fluoro-3-(3-methoxy-5,6,7,8-tetrahydro- [1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl)benzyl)phthalazin- 1(2H)-one 6-Cyclobutoxy-4-(4-fluoro-3-(3-methoxy-5,6,7,8-tetrahydro-[1 ,2,4]triazolo[4,3-a]pyrazine-7- carbonyl)benzyl)phthalazin-1(2H)-one [1079] Following the general two-step procedure above in Example 130, but starting with tert-butyl 3- methoxy-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine-7(8H)-car boxylate gave the title compound as a white solid. LCMS ESI m/z: 504.8 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.43 (s, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.46 (d, J = 5.9 Hz, 2H), 7.34 – 7.25 (m, 2H), 7.08 (d, J = 2.2 Hz, 1H), 4.83 (m, 2H), 4.46 (s, 1H), 4.30 (s, 2H), 4.03 (m, 4H), 3.79 (s, 1H), 3.61 (s, 2H), 2.43 – 2.33 (m, 2H), 1.99 (m, 2H), 1.83 – 1.61 (m, 2H). Synthesis of Example 166: 6-Cyclobutoxy-4-(4-fluoro-3-(3-(hydroxymethyl)-5,6,7,8-tetra hydro- [1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl)benzyl)phthalazin- 1(2H)-one tert-butyl 3-(hydroxymethyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine -7(8H)-carboxylate [1080] To a solution of 7-tert-butyl 3-ethyl 5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine-3,7(8H)- dicarboxylate (2.0 g, 6.75 mmol) in MeOH (60 mL) was added NaBH 4 (2.05 g, 54.00 mmol). The mixture was stirred at 30°C for 2 hr. Water (50 mL) was added and stirred for another 10 min. The reaction mixture was concentrated to give a crude solid. The crude product was purified by silica gel chromatography (DCM: MeOH = 10: 1) to give tert-butyl 3-(hydroxymethyl)-5,6-dihydro- [1,2,4]triazolo[4,3-a]pyrazine-7(8H)-carboxylate (1.2 g, 66% yield) as a white solid. LCMS ESI m/z: 255.2 [M + H] + . Step 2 and 3 6-Cyclobutoxy-4-(4-fluoro-3-(3-(hydroxymethyl)-5,6,7,8-tetra hydro-[1,2,4]triazolo[4,3-a]pyrazine-7- carbonyl)benzyl)phthalazin-1(2H)-one [1081] Following the general two-step procedure above in Example 130, but starting with tert-butyl 3- (hydroxymethyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine-7 (8H)-carboxylate gave the title compound as a white solid. LCMS ESI m/z: 504.7 [M+H] + . 1 H NMR (400 MHz, DMSO-D6) δ12.43 (s, 1H), 8.16 (dd, J = 8.7, 4.9 Hz, 1H), 7.50 – 7.43 (m, 2H), 7.30 (dd, J = 12.6, 5.1 Hz, 2H), 7.08 (s, 1H), 5.49 (dd, J = 12.5, 6.9 Hz, 1H), 4.92 (s, 1H), 4.83 (p, J = 7.1 Hz, 1H), 4.69 – 4.46 (m, 3H), 4.32 (s, 2H), 4.19 – 3.84 (m, 3H), 3.65 (s, 1H), 2.38 (s, 2H), 2.06 – 1.94 (m, 2H), 1.84 – 1.73 (m, 1H), 1.71 – 1.59 (m, 1H). Synthesis of Example 167: 6-Cyclobutoxy-4-(4-fluoro-3-(3-(methoxymethyl)-5,6,7,8-tetra hydro- [1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl)benzyl)phthalazin- 1(2H)-one

tert-Butyl 3-(methoxymethyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine -7(8H)-carboxylate [1082] A mixture of tert-butyl 3-(hydroxymethyl)-6,8-dihydro-5H-[1,2,4]triazolo[4,3-a]pyraz ine-7- carboxylate (150 mg, 589.89 μmol) and NaH (54.25 mg, 2.36 mmol) in dry THF (10 mL) was stirred at 0°C for 15 min. Then CH 3 I (0.47 g, 3.3 mmol) was added dropwise to the reaction mixture. The reaction mixture was allowed to warm to room temperature and stirred for another 2 hr. Sat. NH 4 C1 solution was added and the resulting mixture was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous Na 2 S0 4 and concentrated to give tert-butyl 3-(methoxymethyl)- 5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine-7(8H)-carboxylate (120 mg, 76% yield) as a white solid. The residue was used for next step directly. LCMS ESI m/z: 269.2 [M + H] + . Step 2 and 3 6-Cyclobutoxy-4-(4-fluoro-3-(3-(methoxymethyl)-5,6,7,8-tetra hydro-[1,2,4]triazolo[4,3-a]pyrazine- 7-carbonyl)benzyl)phthalazin-1(2H)-one [1083] Following the general two-step procedure above in Example 130, but starting with tert-butyl 3- (methoxymethyl)-5,6-dihydro-[1,2,4]triazolo[4,3-a]pyrazine-7 (8H)-carboxylate gave the title compound as a white solid. LCMS ESI m/z: 519.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.45 (s, 1H), 8.17 (t, J = 7.3 Hz, 1H), 7.46 (d, J = 6.6 Hz, 2H), 7.36 –7.26 (m, 2H), 7.09 (d, J = 6.2 Hz, 1H), 4.94(s, 1H), 4.89– 4.81 (m, 1H), 4.65– 4.48 (m, 3H), 4.33 (s, 2H), 4.09 (s, 2H),3.87 (s, 1H), 3.65 (s, 1H), 3.29 (d, J = 2.8 Hz, 3H), 2.38 (s, 2H), 2.00 (s, 2H), 1.83–1.75 (m, 1H), 1.67 (m, 1H). Synthesis of Example 168: 6-Cyclobutoxy-4-(4-fluoro-3-(2-(hydroxymethyl)-4,5,6,7- tetrahydropyrazolo[1,5-a]pyrazine-5-carbonyl)benzyl)phthalaz in-1(2H)-one (4,5,6,7-Tetrahydropyrazolo[1,5-a]pyrazin-2-yl)methanol [1084] To a solution of ethyl 4-oxo-6,7-dihydro-5H-pyrazolo[1,5-a]pyrazine-2-carboxylate (200 mg, 956.02 μmol) in THF (10 mL) was added LiAlH 4 (145.14 mg, 3.82 mmol) at 0 °C, and then the mixture was heated to 70 °C for 4 hr. The reaction solution was sequentially quenched with H 2 O (200 mg), 15% NaOH (400 mg) and H 2 O (200 mg). The mixture was stirred at rt for 10 minutes. The solids were removed by filtration, and the filtrate was concentrated to give (4,5,6,7-tetrahydropyrazolo[1,5-a]pyrazin- 2-yl)methanol (110 mg, crude) as a light yellow oil, and it will be used in the next step directly. LCMS ESI m/z: 154.0 [M + H] + . Step 2 6-Cyclobutoxy-4-(4-fluoro-3-(2-(hydroxymethyl)-4,5,6,7-tetra hydropyrazolo[1,5-a]pyrazine-5- carbonyl)benzyl)phthalazin-1(2H)-one [1085] Following the general one-step procedure above in Example 130, but starting with (4,5,6,7- tetrahydropyrazolo[1,5-a]pyrazin-2-yl)methanol gave the title compound as a white solid. LCMS ESI m/z: 503.8 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ12.43 (s, 1H), 8.15 (dd, J = 8.7, 3.9 Hz, 1H), 7.45 (dd, J = 7.0, 3.8 Hz, 2H), 7.29 (td, J = 8.9, 4.7 Hz, 2H), 7.14 – 7.05 (m, 1H), 6.02 (m, 1H), 5.01 – 4.91 (m, 1H), 4.82 (m, 2H), 4.50 – 4.27 (m, 4H), 4.10 (s, 2H), 3.97 (d, J = 5.2 Hz, 1H), 3.66 (s, 1H), 2.36 (m, 2H), 1.99 (m, 2H), 1.86 – 1.56 (m, 2H). Synthesis of Example 169: 4-(4-chloro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1- carbonyl)benzyl)-6-cyclobutoxyphthalazin-1(2H)-one Methyl 2-chloro-5-methylbenzoate [1086] To a solution of 2-chloro-5-methylbenzoic acid (5.00 g, 29.31 mmol) in MeOH (20 mL) was added conc. H 2 SO 4 (5 mL) in portions. The mixture was stirred at 70 °C for 2 hours. The solution was concentrated in vacuo, the residue was diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 2-chloro-5-methylbenzoate (5.0 g, 91% yield) as a white oil. LCMS ESI m/z: 185.0 [M+H] + . Step 2 Methyl 2-chloro-5-(dibromomethyl)benzoate [1087] A mixture of methyl methyl 2-chloro-5-methylbenzoate (500 mg, 2.71 mmol), NBS (1.59 g, 8.94 mmol) and AIBN (133 mg, 0.81 mmol ) in ACN (10 mL) was stirred under reflux for 16 hours. The mixture was cooled to rt, diluted with H 2 O (30 mL), and extracted with DCM (30 mL x 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0% to 15%) to obtain methyl methyl 2- chloro-5-(dibromomethyl)benzoate (615 mg, 62% yield) as a yellow oil. LCMS ESI m/z: 341 [M+H] + . Step 3 Methyl 2-chloro-5-formylbenzoate [1088] To a solution of methyl methyl 2-chloro-5-(dibromomethyl)benzoate (615 mg, 1.80 mmol) in EtOH (10 mL), was added AgNO 3 (3.05 g, 17.96 mmol) and H 2 O (20 mL). The mixture was stirred at 80 °C for 30 minutes. The solution was concentrated in vacuo. This residue was diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 2-chloro-5-formylbenzoate (370 mg, 88% yield) as a yellow solid. LCMS ESI m/z: 199.0 [M+H] + . Step 4 methyl 2-chloro-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl) benzoate [1089] A solution of dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (200 mg, 0.64 mmol), methyl 2-chloro-5-formylbenzoate (134 mg, 0.67 mmol) and Et 3 N (194 mg, 1.92 mmol) in anhydrous THF (6 mL) was stirred at 25 °C under Ar (g) for 3 hours. Hydrazine hydrate (49 mg, 0.83 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/ EtOAc (10:1, 10 mL) to give methyl 2-chloro-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoate (125 mg, 49% yield) as a white solid. LCMS ESI m/z: 399.1 [M+H] + . Step 5 2-chloro-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl)benzoic acid [1090] A solution of methyl 2-chloro-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoate (125 mg, 0.31 mmol) and LiOH (75 mg, 3.13 mmol) in MeOH (3 mL) and H 2 O (1 mL) was stirred at rt for 30 minutes. The organic solvent was removed in vacuum and aq HCl (1N) was added until the solution reached to pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 2-chloro-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl)benzoic acid (120 mg, 99% yield) as a white solid. LCMS ESI m/z: 385.1 [M+H] + . Step 6 4-(4-chloro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1-carbonyl)benzyl)-6- cyclobutoxyphthalazin-1(2H)-one [1091] To a solution of 2-(piperazin-1-yl)-5-(trifluoromethyl)pyrimidine (92 mg, 0.34 mmol) and 2- chloro-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)me thyl)benzoic acid (120 mg, 0.31 mmol) in DMF (3 mL) was added EDCI (90 mg, 0.47 mmol), HOBt (63 mg, 0.47 mmol) and DIPEA (201 mg, 1.56 mmol). The reaction mixture was stirred at 45 °C for 1 hour. The reaction was purified by prep- HPLC to give 4-(4-chloro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1-carbonyl)benzyl)-6- cyclobutoxyphthalazin-1(2H)-one (98 mg, 52% yield) as a white solid. LCMS ESI m/z: 599.1 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.54 (s, 1H), 8.49 (s, 2H), 8.36 (d, J = 8.8 Hz, 1H), 7.37 (d, J = 8.1 Hz, 1H), 7.28 (d, J = 2.1 Hz, 1H), 7.25 (s, 1H), 7.19 (dd, J = 8.8, 2.3 Hz, 1H), 6.89 (d, J = 2.3 Hz, 1H), 4.66 (p, J = 7.1 Hz, 1H), 4.23 (s, 2H), 4.09 – 3.77 (m, 6H), 3.39 – 3.19 (m, 2H), 2.48 – 2.36 (m, 2H), 2.22 – 2.09 (m, 2H), 1.94 – 1.70 (m, 2H). Synthesis of Example 170: 6-cyclobutoxy-4-(3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pip erazine-1- carbonyl)benzyl)phthalazin-1(2H)-one Methyl 3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)be nzoate [1092] A solution of dimethyl (6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (200 mg, 0.64 mmol), methyl 3-formylbenzoate (105 mg, 0.64 mmol) and Et 3 N (194 mg, 1.92 mmol) in anhydrous THF (5 mL) was stirred at 25 °C under Ar (g) for 15 hours. Hydrazine hydrate (43 mg, 0.86 mmol) was added and the reaction was stirred at 70 ℃ for 1 hour. The reaction was cooled to rt, then THF was removed under vacuum. The formed solid was washed with water (10 mL) and petroleum ether/EA (2:1, 10 mL) to give methyl 3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)be nzoate (100 mg, 48% yield) as a white solid. LCMS ESI m/z: 365 [M+H] + . Step 2 3-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)be nzoic acid [1093] A solution of methyl 3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)be nzoate (100 mg, 0.27 mmol) and LiOH (66 mg, 2.70 mmol) in MeOH (3 mL) and H 2 O (1 mL) was stirred at rt for 2 hours. The organic solvent was removed in vacuo and aq HCl (1 mol/L) was added until the solution reached pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)be nzoic acid (80 mg, 83% yield) as a white solid. LCMS ESI m/z: 351 [M+H] + . Step 3 6-Cyclobutoxy-4-(3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pip erazine-1-carbonyl)benzyl)phthalazin- 1(2H)-one [1094] To a solution of 3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)be nzoic acid (60 mg, 0.17 mmol) and 2-(piperazin-1-yl)-5-(trifluoromethyl)pyrimidine (55 mg, 0.21 mmol) in DMF (2 mL) was added EDCI (49 mg, 0.26 mmol), HOBt (35 mg, 0.26 mmol) and DIPEA (111 mg, 0.86 mmol). The reaction mixture was stirred at 45 °C for 1 hour. The reaction was purified by prep-HPLC to give 6- cyclobutoxy-4-(3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piper azine-1-carbonyl)benzyl)phthalazin-1(2H)- one (34 mg, 35% yield) as a white solid (34 mg). LCMS ESI m/z: 565 [M+H] + . 1 H NMR (400 MHz, DMSO–d6) δ 12.48 (s, 1H), 8.74 (s, 2H), 8.16 (d, J = 8.8 Hz, 1H), 7.44 – 7.40 (m, 3H), 7.33 – 7.29 (m, 2H), 7.12 – 7.08 (m, 1H), 4.86 – 4.78 (m, 1H), 4.34 (s, 2H), 3.98 – 3.65 (m, 6H), 3.46 – 3.38 (m, 2H), 2.42 – 2.32 (m, 2H), 2.05 – 1.93 (m, 2H), 1.81 – 1.72 (m, 1H), 1.69 – 1.60 (m, 1H). Synthesis of Example 171: 4-(2-chloro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1- carbonyl)benzyl)-6-cyclobutoxyphthalazin-1(2H)-one Methyl 2-chloro-3-methylbenzoate [1095] To a solution of 2-chloro-3-methylbenzoic acid (5.00 g, 29.31mmol) in MeOH (15 mL) was added conc. H 2 SO 4 (6.25 mL) in portions. The mixture was stirred at 25 °C for 20 hours. The solution was concentrated in vacuo. This residue was diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 2-chloro-3-methylbenzoate (5.20 g, 96% yield) as a white solid. LCMS ESI m/z: 185 [M+H] + . This was used without further purification. Step 2 Methyl 2-chloro-3-(dibromomethyl)benzoate [1096] A mixture of methyl 2-chloro-3-methylbenzoate (500 mg, 2.71 mmol), NBS (1.59 g, 8.94 mmol) and AIBN (133 mg, 0.81 mmol ) in ACN (10 mL) was stirred under reflux for 16 hours. The mixture was cooled to rt, diluted with H2O (30 mL), and extracted with DCM (30 mL x 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0% to 15%) to obtain methyl 2-chloro-3- (dibromomethyl)benzoate (797 mg, 86% yield) as a white solid. LCMS ESI m/z: 342/344 [M+H] + . Step 3 Methyl 2-chloro-3-formylbenzoate [1097] To a solution of methyl 2-chloro-3-(dibromomethyl)benzoate (700 mg, 2.04 mmol) in EtOH (10 mL), was added AgNO 3 (3.47 g, 9.58 mmol) and H 2 O (5 mL). The mixture was stirred at 80 °C for 30 minutes. The solution was concentrated in vacuo, diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 2-chloro-3-formylbenzoate (370 mg, 91% yield) as a yellow solid. LCMS ESI m/z: 199 [M+H] + . This was used without further purification. Step 4 Methyl 2-chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl) benzoate [1098] A solution of dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (200 mg, 0.64 mmol), methyl 2-chloro-3-formylbenzoate (134 mg, 0.67 mmol) and Et 3 N (194 mg, 1.92 mmol) in anhydrous THF (6 mL) was stirred at 25 °C under Ar (g) for 3 hours. Hydrazine hydrate (49 mg, 0.83 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/EtOAc (10:1, 10 mL) to give methyl 2-chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoate (96 mg, 38 % yield) as a white solid. LCMS ESI m/z: 399 [M+H] + . Step 5 2-chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl)benzoic acid [1099] A solution of methyl 2-chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoate (96 mg, 0.24 mmol) and LiOH (60 mg, 2.51 mmol) in MeOH (3 mL) and H 2 O (1 mL) was stirred at rt for 1 hour. The organic solvent was removed in vacuum and aq HCl (1N) was added until the solution reached to pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 2-chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl)benzoic acid (90 mg, 93% yield) as a white solid. LCMS ESI m/z: 384 [M+H] + . This was used without further purification. Step 6 4-(2-Chloro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1-carbonyl)benzyl)-6- cyclobutoxyphthalazin-1(2H)-one [1100] To a solution of 2-(piperazin-1-yl)-5-(trifluoromethyl)pyrimidine (92 mg, 0.34 mmol) and 5- ((7-2-chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1 -yl)methyl)benzoic acid (120 mg, 0.31 mmol) in DMF (3 mL) was added EDCI (90 mg, 0.47 mmol), HOBt (63 mg, 0.47 mmol) and DIPEA (201 mg, 1.56 mmol). The reaction mixture was stirred at 45 °C for 1 hour. The reaction was purified by prep-HPLC to give the 4-(2-chloro-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1- carbonyl)benzyl)-6-cyclobutoxyphthalazin-1(2H)-one (37 mg, 20% yield) as a white solid. LCMS ESI m/z: 599 [M+H] + . 1 H–NMR (400 MHz, DMSO-d 6 ) δ 12.42 (s, 1H), 8.74 (s, 2H), 8.18 (d, J = 8.8 Hz, 1H), 7.39 – 7.28 (m, 4H), 7.16 (d, J = 2.3 Hz, 1H), 4.89 (p, J = 7.1 Hz, 1H), 4.50 – 4.36 (m, 2H), 4.04 – 3.71 (m, 6H), 3.27 (t, J = 5.1 Hz, 2H), 2.49 – 2.41 (m, 2H), 2.14 – 1.98 (m, 2H), 1.88 – 1.59 (m, 2H). Synthesis of Example 172: 6-Cyclobutoxy-4-(3-fluoro-4-(4-(5-(trifluoromethyl)pyrimidin -2- yl)piperazine-1-carbonyl)benzyl)phthalazin-1(2H)-one Methyl 4-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoate [1101] A solution of methyl 2-fluoro-4-formylbenzoate (150 mg, 0.82 mmol), dimethyl (6-bromo-3- oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (257 mg, 0.82 mmol) and Et 3 N (250 mg, 2.47 mmol) in anhydrous THF (10 mL) was stirred at 25 °C under Ar (g) for 3 hours. Hydrazine hydrate (83 mg, 1.66 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and concentrated under reduced pressure to give 6-(4-(3-formylbenzoyl)piperazin-1-yl)nicotinonitrile (209 mg, 66% yield) as a white solid. LCMS ESI m/z: 383.0 [M+H] + . This was used without further purification. Step 2 4-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid [1102] A mixture of methyl 4-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 - fluorobenzoate (209 mg, 0.55 mmol) and LiOH (131 mg, 5.47 mmol) in MeOH (5 mL) and H 2 O (5 mL) was stirred at rt for 16 hours. The organic solvent was removed in vacuo, aq 2N HCl was added until the solution reached pH 5~6, then extracted with EtOAc (30 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give 4-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoic acid (200 mg, 99% yield) as a white solid. LCMS ESI m/z: 369.0 [M+H] + . This was used without further purification. Step 3 6-Cyclobutoxy-4-(3-fluoro-4-(4-(5-(trifluoromethyl)pyrimidin -2-yl)piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one [1103] To a solution of 4-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (100 mg, 0.27 mmol) and 2-(piperazin-1-yl)-5-(trifluoromethyl)pyrimidine (54 mg, 0.27 mmol) in DMF (3 mL) was added HOBt (55 mg, 0.41 mmol), EDCI (78 mg, 0.41 mmol) and DIPEA (105 mg, 0.81 mmol). The reaction mixture was stirred at rt for 16 hours. The reaction was purified by prep-HPLC to give 6-cyclobutoxy-4-(3-fluoro-4-(4-(5-(trifluoromethyl)pyrimidin -2-yl)piperazine-1- carbonyl)benzyl)phthalazin-1(2H)-one (41.1 mg, 26 % yield) as a white solid. LCMS ESI m/z: 583.2 [M+H] + . 1 H–NMR (400 MHz, DMSO-d 6 ) δ 12.48 (s, 1H), 8.74 (s, 2H), 8.17 (d, J = 8.8 Hz, 1H), 7.39 (t, J = 7.5 Hz, 1H), 7.33 (d, J = 2.0 Hz, 1H), 7.31 (d, J = 2.3 Hz, 1H), 7.23 (d, J = 7.8 Hz, 1H), 7.11 (d, J = 2.3 Hz, 1H), 4.86 (dd, J = 14.2, 7.1 Hz, 1H), 4.35 (s, 2H), 3.92 (d, J = 5.4 Hz, 2H), 3.80 (d, J = 5.2 Hz, 2H), 3.74 (d, J = 5.0 Hz, 2H), 3.31 (s, 2H), 2.43 – 2.30 (m, 2H), 2.07 – 1.95 (m, 2H), 1.80 (dd, J = 20.2, 10.1 Hz, 1H), 1.66 (dd, J = 18.5, 8.4 Hz, 1H). Synthesis of Example 173: 4-(2-Chloro-5-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1- carbonyl)benzyl)-6-cyclobutoxyphthalazin-1(2H)-one methyl 4-chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl) benzoate [1104] A solution of dimethyl (6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (200 mg, 0.64 mmol), methyl 4-chloro-3-formylbenzoate (127 mg, 0.64 mmol) and Et 3 N (194 mg, 1.92 mmol) in anhydrous THF (5 mL) was stirred at 25 °C under Ar (g) for 15 hours. Hydrazine hydrate (49 mg, 0.83 mmol) was added and the reaction was stirred at 70 ℃ for 1 hour. The reaction was cooled to rt, then THF was removed under vacuum. The formed solid was washed with water (10 mL) and petroleum ether/EA (2:1, 10 mL) to give methyl 4-chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoate (140 mg, 55% yield) as a white solid. LCMS ESI m/z: 399 [M+H] + . Step 2 4-Chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl)benzoic acid [1105] A solution of methyl 4-chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoate (140 mg, 0.35 mmol) and LiOH (84 mg, 3.5 mmol) in MeOH (3 mL) and H 2 O (1 mL) was stirred at rt for 2 hours. The organic solvent was removed in vacuo and aq HCl (1 M) was added until the solution reached pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 4-chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl)benzoic acid (116 mg, 86% yield) as a white solid. LCMS ESI m/z: 385 [M+H] + . Step 3 4-(2-chloro-5-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1-carbonyl)benzyl)-6- cyclobutoxyphthalazin-1(2H)-one [1106] To a solution of 4-chloro-3-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl)benzoic acid (116 mg, 0.30 mmol) and 2-(piperazin-1-yl)-5-(trifluoromethyl)pyrimidine (89 mg, 0.33 mmol) in DMF (2 mL) was added EDCI (87 mg, 0.45 mmol), HOBt (61 mg, 0.45 mmol) and DIPEA (195 mg, 1.50 mmol). The reaction mixture was stirred at 45 °C for 1 hour. The reaction was purified by prep- HPLC to give 4-(2-chloro-5-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1-carbonyl) benzyl)-6- cyclobutoxyphthalazin-1(2H)-one (75 mg, 41% yield) as a white solid. LCMS ESI m/z: 599 [M+H] + . 1 H NMR (400 MHz, DMSO–d 6 ) δ 12.39 (s, 1H), 8.74 (s, 2H), 8.18 (d, J = 8.8 Hz, 1H), 7.61 – 7.58 (m, 1H), 7.40 – 7.35 (m, 3H), 7.18 – 7.15 (m, 1H), 4.93 – 4.86 (m, 1H), 4.43 (s, 2H), 3.95 – 3.63 (m, 6H), 2.48 – 2.35 (m, 3H), 2.20 – 1.91 (m, 3H), 1.84 – 1.76 (m, 1H), 1.71 – 1.64 (m, 1H). Synthesis of Example 174: 4-(3-chloro-5-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1- carbonyl)benzyl)-6-cyclobutoxyphthalazin-1(2H)-one Methyl 3-chloro-5-(hydroxymethyl)benzoate [1107] To a solution of dimethyl 5-chloroisophthalate (500 mg, 2.19 mmol) in THF (10 mL) was added DIBAL-H (1M in THF, 6.6 ML, 6.6 mmol) at 0 °C, then the mixture was stirred at rt for 30 minutes. The reaction was quenched with saturated aqueous solution of potassium sodium tartrate tetrahydrate (20 mL), then extracted with EtOAc (20 mL x 3). The combined organic phase was washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (Petroleum ether/EtOAc = 3/1) to give methyl 3-chloro-5- (hydroxymethyl)benzoate (213 mg, 49% yield) as a colorless oil. LCMS ESI m/z: 201.1 [M+H] + . Step 2 Methyl 3-chloro-5-formylbenzoate [1108] To a solution of methyl 3-chloro-5-(hydroxymethyl)benzoate (213 mg, 1.06 mmol) in DCM (5 mL) was added MnO 2 (1.38 g, 15.93 mmol), then the mixture was stirred at rt for 16 hours. The solid was removed by filtration and the filtrate was concentrated in vacuo to afford methyl 3-chloro-5- formylbenzoate (142 mg, 67% yield) as a colorless oil. LCMS ESI m/z: 199.0 [M+H] + . Step 3 Methyl 3-chloro-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl) benzoate [1109] A solution of methyl 3-chloro-5-formylbenzoate (142 mg, 0.71 mmol), dimethyl (6- cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (223 mg, 0.71 mmol) and Et 3 N (217 mg, 2.14 mmol) in anhydrous THF (5 mL) was stirred at 45 °C under Ar (g) for 16 hours. N 2 H 4 -H 2 O (72 mg, 1.43 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and concentrated under reduced pressure to give methyl 3-chloro-5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)benzoate (155 mg, 54% yield) as a white solid. LCMS ESI m/z: 399.1 [M+H] + . This was used without further purification. Step 4 3-Chloro-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl)benzoic acid [1110] A mixture of methyl 3-chloro-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoate (155 mg, 0.39 mmol) and LiOH (93 mg, 3.89 mmol) in MeOH (3 mL) and H 2 O (3 mL) was stirred at rt for 3 hours. The organic solvent was removed in vacuo and aq HCl (2N) was added until the solution reached pH 5~6, then extracted with EtOAc (30 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give 3-chloro- 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)be nzoic acid (137 mg, 92% yield) as a white solid. LCMS ESI m/z: 385.1 [M+H] + . This was used without further purification. Step 5 4-(3-Chloro-5-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1-carbonyl)benzyl)-6- cyclobutoxyphthalazin-1(2H)-one [1111] To a solution of 3-chloro-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl) methyl)benzoic acid (137 mg, 0.36 mmol) and 2-(piperazin-1-yl)-5-(trifluoromethyl)pyrimidine (83 mg, 0.36 mmol) in DMF (3 mL) was added EDCI (102 mg, 0.53 mmol), HOBt (72 mg, 0.53 mmol) and DIPEA (138 mg, 1.07 mmol). The reaction mixture was stirred at rt for 16 hours. The reaction was purified by prep-HPLC to give 4-(3-chloro-5-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi ne-1-carbonyl)benzyl)-6- cyclobutoxyphthalazin-1(2H)-one (93 mg, 44% yield) as a white solid. LCMS ESI m/z: 599.0 [M+H] + . 1 H–NMR (400 MHz, CDCl 3 ) δ 12.47 (s, 1H), 8.74 (s, 2H), 8.16 (d, J = 8.8 Hz, 1H), 7.54 (s, 1H), 7.38 (d, J = 1.7 Hz, 1H), 7.36 – 7.30 (m, 2H), 7.13 (d, J = 2.3 Hz, 1H), 4.87 (p, J = 7.0 Hz, 1H), 4.36 (s, 2H), 4.03 – 3.61 (m, 6H), 3.36 (s, 1H), 3.31 (s, 1H), 2.44 – 2.35 (m, 2H), 2.08 – 1.94 (m, 2H), 1.79 (dd, J = 20.1, 10.2 Hz, 1H), 1.66 (dd, J = 18.5, 8.3 Hz, 1H). Synthesis of Example 175: 2-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2,3- dihydrobenzofuran-7-carbonyl)piperazin-1-yl)isonicotinonitri le

Methyl 5-bromo-2,3-dihydrobenzofuran-7-carboxylate [1112] To a solution of 5-bromo-2,3-dihydrobenzofuran-7-carboxylic acid (1.00 g, 4.11 mmol) in MeOH (5 mL) was added conc. H 2 SO 4 (0.87 mL) in portions. The mixture was stirred at 25 °C for 20 hours. The solution was concentrated in vacuo. This residue was diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 5-bromo-2,3-dihydrobenzofuran-7-carboxylate (1.20 g, 99% yield) as a colorless oil. LCMS ESI m/z: 257 [M+H] + . This was used without further purification. Step 2 Methyl 5-vinyl-2,3-dihydrobenzofuran-7-carboxylate [1113] A mixture of 5-bromo-2,3-dihydrobenzofuran-7-carboxylate (1.2 g, 4.48 mmol), potassium vinyltrifluoroborate (603 mg, 4.50 mmol), K2CO3 (1.54g, 11.20 mmol) and Pd(PPh3)4 (259 mg, 0.224 mmol) in DMF (20 mL) was stirred at 80 °C under Ar (g) for 4 hours. The mixture was cooled to rt, diluted with H 2 O (20 mL) and extracted with EtOAc (30 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromategraphy (EtOAc/petroleum ether = 10 to 15%) to give methyl 5-vinyl-2,3- dihydrobenzofuran-7-carboxylate (807 mg, 89% yield) as a colorless oil. LCMS ESI m/z: 205 [M+H] + . Step 3 Methyl 5-formyl-2,3-dihydrobenzofuran-7-carboxylate [1114] To a mixture of methyl 5-vinyl-2,3-dihydrobenzofuran-7-carboxylate (607 mg, 2.76 mmol) in THF (8 mL) and H 2 O (5 mL) was added K 2 OsO 4 -2H 2 O (10 mg, 0.03 mmol). The mixture was stirred at room temperature for 5 min, then NaIO 4 (1.77 g, 8.29 mmol) was added and the mixture was stirred at 80 ℃ for 2 hours. The solids were removed by filtration. The filtrate was diluted with H 2 O (5 mL) and extracted with EtOAc (30 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 5-formyl-2,3-dihydrobenzofuran-7- carboxylate (284 mg, 49% yield). LCMS ESI m/z: 207 [M+H] + . This was used without further purification. Step 4 Methyl 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 ,3-dihydrobenzofuran-7- carboxylate [1115] A solution of dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (200 mg, 0.97 mmol), methyl 5-formyl-2,3-dihydrobenzofuran-7-carboxylate (504 mg, 0.97 mmol) and Et 3 N (294 mg, 2.91 mmol) in anhydrous THF (3 mL) was stirred at 25 °C under Ar (g) for 3 hours. Hydrazine hydrate (72 mg, 1.44 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/ EtOAc (10:1, 10 mL) to give methyl 5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2,3-dihydrobenzofuran-7-carbo xylate (130 mg, 37 % yield) as a yellow solid. LCMS ESI m/z: 407 [M+H] + . Step 5 5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 ,3-dihydrobenzofuran -7-carboxylic acid [1116] A solution of methyl 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 ,3- dihydrobenzofuran-7-carboxylate (130 mg, 0.37 mmol) and NaOH (147 mg, 3.69 mmol) in MeOH (2 mL), THF (2 mL) and H 2 O (2 mL) was stirred at 45 °C for 1 hour. The organic solvent was removed in vacuum and aq HCl (1N) was added until the solution reached to pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 5-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)-2,3-dihydrobenzofuran-7-carbo xylic acid (90 mg, 72% yield) as white solid. LCMS ESI m/z: 393 [M+H] + . Step 6 tert-Butyl 4-(4-cyanopyridin-2-yl)piperazine-1-carboxylate [1117] A mixture of 2-chloroisonicotinonitrile (300 mg, 2.17 mmol), tert-butyl piperazine-1- carboxylate (403 mg, 2.17 mmol) and DIPEA (1.12 g, 8.66 mmol) in DMF (10 mL) was stirred at 100 °C for 20 hours. The reaction was diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0% to 10%) to give tert-butyl 4-(4-cyanopyridin-2-yl)piperazine-1-carboxylate (340 mg, 45% yield ) as a white solid. LCMS ESI m/z: 289 [M+H] + . Step 7 2-(Piperazin-1-yl)isonicotinonitrile [1118] A solution of tert-butyl 4-(4-cyanopyridin-2-yl)piperazine-1-carboxylate (340 mg, 4.14 mmol) in HCl/dioxane (4M, 10 mL) was stirred at rt for 1 hour. The solvent was removed in vacuo to give 2- (piperazin-1-yl)isonicotinonitrile (609 mg, 85% yield ) as a white solid. LCMS ESI m/z: 189 [M+H] + . This was used without further purification. Step 8 2-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2,3-dihydrobenzofuran-7- carbonyl)piperazin-1-yl)isonicotinonitrile [1119] To a solution of 2-(piperazin-1-yl)isonicotinonitrile (172 mg, 0.28 mmol) and 5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2,3-dih ydrobenzofuran-7-carboxylic acid (90 mg, 0.23 mmol) in DMF (2 mL) was added EDCI (66 mg, 0.34 mmol), HOBt (47 mg, 0.34 mmol) and DIPEA (89 mg, 0.67 mmol). The reaction mixture was stirred at rt for 1 hour. The reaction was purified by prep- HPLC to give 2-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)-2,3-dihydrobenzofuran- 7-carbonyl)piperazin-1-yl)isonicotinonitrile (25 mg, 19% yield) as a white solid. LCMS ESI m/z: 562 [M+H] + . 1 H–NMR (400 MHz, DMSO-d 6 ) δ 12.44 (s, 1H), 8.31 (d, J = 5.0 Hz, 1H), 8.14 (d, J = 8.8 Hz, 1H), 7.34 – 7.27 (m, 2H), 7.23 (s, 1H), 7.14 (dd, 2H), 6.99 (dd, J = 5.0 Hz, 1H), 4.84 (p, J = 7.1 Hz, 1H), 4.53 (t, J = 8.7 Hz, 2H), 4.21 (s, 2H), 3.73 – 3.49 (m, 6H), 3.30 (s, 2H), 3.15 (t, J = 8.5 Hz, 2H), 2.47 – 2.34 (m, 2H), 2.06 – 1.93 (m, 2H), 1.86 – 1.59 (m, 2H). Synthesis of Example 176: 6-(4-(6-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)picolinoyl)piperazin-1-yl)nicotinonitrile

Methyl 6-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)pi colinate [1120] A solution of dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (189 mg, 0.606 mmol), methyl 6-formyl-2-pyridinecarboxylate (100 mg, 0.606 mmol) and Et 3 N (285 mg, 1.82 mmol) in anhydrous THF (3 mL) was stirred at 25 °C under Ar (g) for 15 hours. Hydrazine hydrate (72 mg, 1.44 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/ EtOAc (2:1, 10 mL) to give methyl 6-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)picolinate (221 mg, 85 % yield) as a as a white solid. LCMS ESI m/z: 366.2 [M+H] + . Step 2 6-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)pi colinic acid [1121] A solution of methyl 6-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)pi colinate (221 mg, 606 umol), 1 M LiOH in H 2 O (606 uL, 1.21 mmol) and THF (3.03 mL) was stirred at 25 °C under N 2 (g) for 15 hours. The reaction was stopped and the product was precipitated out of solution by addition of 10% citric acid in water until pH was found to be acidic (pH = 2-3). Product precipitated as a bright yellow solid. Filtration over a Buchner and the solid was washed with water (2 x 20 mL), dried in vacuo. Used as is for the next step (150 mg, 65 %). LCMS ESI m/z 352.1 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 13.23 (s, 1H), 12.46 (s, 1H), 8.11 (d, J = 8.8 Hz, 1H), 7.92 – 7.83 (m, 2H), 7.49 (dd, J = 6.5, 2.3 Hz, 1H), 7.26 (dd, J = 8.8, 2.4 Hz, 1H), 7.20 (d, J = 2.3 Hz, 1H), 4.78 (p, J = 7.1 Hz, 1H), 4.45 (s, 2H), 2.43 – 2.32 (m, 2H), 1.97 (ddd, J = 12.2, 8.9, 2.5 Hz, 2H), 1.86 – 1.54 (m, 3H). Step 3 6-(4-(6-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)picolinoyl)piperazin-1- yl)nicotinonitrile [1122] A solution of 6-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)pi colinic acid (30.0 mg, 85.4 umol), 6-(piperazin-1-yl)nicotinonitrile (16.1 mg, 85.4 umol) and HATU (40.2 mg, 102 umol) in anhydrous DMF (854 uL) was stirred at 25 °C under N 2 (g) for 2 hours. The crude solution was purified by reverse phase chromatography (100% aqueous ammonium formate (AMF) solution (pH = 3.8) to 70% MeCN in AMF). Fractions containing pure product were lyophilized overnight providing 6-(4-(6- ((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)pico linoyl)piperazin-1-yl)nicotinonitrile as a white solid (31.0 mg, 69 % yield). LCMS ESI m/z 522.3 (M+H) + .1H NMR (400 MHz, DMSO-d6) δ 12.45 (s, 1H), 8.50 (d, J = 2.2 Hz, 1H), 8.14 (d, J = 8.8 Hz, 1H), 7.90 (dd, J = 9.4, 6.6 Hz, 2H), 7.52 (d, J = 7.7 Hz, 1H), 7.48 (d, J = 7.7 Hz, 1H), 7.29 (dd, J = 8.8, 2.2 Hz, 1H), 7.08 (d, J = 2.3 Hz, 1H), 6.84 (d, J = 9.1 Hz, 1H), 4.78 – 4.67 (m, 1H), 4.46 (s, 2H), 3.72-3.64 (m, 4H), 3.29 – 3.21 (m, 2H), 2.35-2.27 (m, 2H), 1.99-1.86 (m, 2H), 1.78-1.52 (m, 2H). Synthesis of Example 177: 6-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)nicotinoyl)piperazin-1-yl)nicotinonitrile 5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)ni cotinonitrile [1123] A solution of dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (355 mg, 1.14 mmol), 5-formylnicotinonitrile (150 mg, 1.14 mmol) and Et 3 N (346 mg, 3.41 mmol) in anhydrous THF (5.68 mL) was stirred at 25 °C under Ar (g) for 15 hours. Hydrazine hydrate (72 mg, 2.27 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/ EtOAc (2:1, 10 mL) to give 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)nicotinonitrile (210 mg, 56% yield) as a as a white solid. 1 H NMR (400 MHz, cdcl3) δ 9.01 (d, J = 2.1 Hz, 1H), 8.76 (d, J = 1.9 Hz, 1H), 8.60 (t, J = 2.1 Hz, 1H), 7.85 (dd, J = 8.9, 6.6 Hz, 1H), 7.09 – 6.99 (m, 2H), 6.28 (s, 1H), 4.90 – 4.69 (m, 1H), 2.60 – 2.50 (m, 2H), 2.36 – 2.19 (m, 2H), 1.96 (d, J = 10.4 Hz, 1H), 1.86 – 1.68 (m, 1H). Step 2 5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)ni cotinic acid [1124] A solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)ni cotinonitrile (200 mg, 0.602 mmol), an 8.0 M aqueous solution of sodium hydroxide (1.5 mL, 20 mmol), and Ethanol (3.02 mL) was stirred at 25 °C then heated to 100 °C for 1 hour. The reaction was cooled to rt, then the reaction was made acidic by addition of citric acid 10% in water (30 mL) until pH was found to be 2-3. The formed solid was triturated and filtered and dried in vacuo. The solid was then triturated again in in 40% EtOAc in 60% Heptanes (30 mL) for 30 minutes before being filtered on a Buchner.5-((7-Cyclobutoxy-4- oxo-3,4-dihydrophthalazin-1-yl)methyl)nicotinic acid (170 mg, 80 %) isolated as a white solid. LCMS ESI m/z: 352.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.41 (s, 1H), 8.90 (d, J = 2.0 Hz, 1H), 8.77 (d, J = 2.1 Hz, 1H), 8.22 – 8.10 (m, 2H), 7.30 (dd, J = 8.8, 2.3 Hz, 1H), 7.17 (d, J = 2.3 Hz, 1H), 4.96 – 4.81 (m, 1H), 4.40 (s, 2H), 2.57 – 2.34 (m, 2H), 2.13 – 1.93 (m, 2H), 1.93 – 1.55 (m, 2H). Step 3 6-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)nicotinoyl)piperazin-1- yl)nicotinonitrile [1125] A solution of 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)ni cotinic acid (30.0 mg, 85.4 umol), 6-(piperazin-1-yl)nicotinonitrile (16.1 mg, 85.4 umol) and HATU (40.2 mg, 102 umol) in anhydrous DMF (854 uL) was stirred at 25 °C under N 2 (g) for 2 hours. The crude solution was purified by reverse phase chromatography (100% aqueous ammonium formate (AMF) solution (pH = 3.8) to 70% MeCN in AMF). Fractions containing pure product were lyophilized overnight providing 6-(4-(5- ((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)nico tinoyl)piperazin-1-yl)nicotinonitrile as a white solid (30.5 mg, 68 % yield). LCMS ESI m/z 522.3 (M+H) + .1H NMR (400 MHz, DMSO-d 6 ) δ 12.43 (s, 1H), 8.64 (d, J = 2.1 Hz, 1H), 8.50 (dd, J = 4.4, 2.0 Hz, 2H), 8.14 (d, J = 8.8 Hz, 1H), 7.88 (dd, J = 9.1, 2.4 Hz, 1H), 7.76 (t, J = 1.9 Hz, 1H), 7.32 (dd, J = 8.8, 2.3 Hz, 1H), 7.15 (d, J = 2.4 Hz, 1H), 6.90 (d, J = 9.2 Hz, 1H), 4.87 (p, J = 7.7 Hz, 1H), 4.37 (s, 2H), 3.80-3.58 (m, 6H), 3.42-3.34 (m, 2H), 2.41- 2.34 (m, 2H), 2.03-1.95 (m, 2H), 1.82 – 1.55 (m, 2H). Synthesis of Example 178: 6-(4-(3-Bromo-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin- 1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile Methyl 3-bromo-5-(hydroxymethyl)benzoate [1126] To a stirred solution of dimethyl 5-bromoisophthalate (5.4 g, 20 mmol) in methanol (300 mL) at 0 0 C was slowly added sodium tetrahydroborate (10 g, 0.28 mol). The mixture was stirred at rt for 3 h, and then treated with water (300 mL). The volatiles were removed in vacuo and the aqueous phase was extracted with CH 2 Cl 2 (100 niL x 3). The combined organic layers were washed with brine, dried over anhydrous MgSO 4 , and concentrated in vacuo. The resulting oil was purified by flash chromatography (100% Heptanes to 50% EtOAc in Heptanes) to afford the title compound. methyl 3-bromo-5- (hydroxymethyl)benzoate (1.50 g, 56 %). 1 H NMR (400 MHz, CDCl3) δ 8.08 (s, 1H), 7.94 (s, 1H), 7.73 (s, 1H), 4.74 (s, 2H), 3.92 (s, 3H). Step 2 Methyl 3-bromo-5-formylbenzoate [1127] To a stirred solution of methyl 3-bromo-5-(hydroxymethyl)benzoate (1.35 g, 5.49 mmol) in DCM (27.5 mL) at rt was added manganese (IV) oxide activated (2.65 g, 27.5 mmol). The mixture was stirred at rt for 12 h. The crude was filtrated over Celite cake and the filtrate was evaoprated to dryness. The resulting solid was purified by flash chromatography (100% Heptanes to 20% EtOAc in hepatnes) to afford the title compound. Methyl 3-bromo-5-formylbenzoate (854 mg, 64 %) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 10.01 (s, 1H), 8.44 (d, J = 1.3 Hz, 1H), 8.42 (d, J = 1.7 Hz, 1H), 8.23 – 8.16 (m, 1H), 3.97 (s, 3H). Step 3 Methyl 3-bromo-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)m ethyl)benzoate [1128] A solution of methyl 3-bromo-5-formylbenzoate (625 mg, 2.06 mmol), dimethyl (6- cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (355 mg, 1.14 mmol) and Triethylamine (865 uL, 6.17 mmol) in anhydrous THF (10.3 mL) was stirred at 25 °C under N 2 (g) for 15 hours. Dilution with EtOAc (60 mL) and transfer to a 125 mL extraction funnel. Wash the solution with NaHCO 3 (sat. aq.) and brine (1 x). Dry organic layer over Na 2 SO 4 , filter and evaporate to dryness. The oil was redissolved in anhydrous THF (10.3 mL) and then hydrazine hydrate solution (196 uL, 6.17 mmol) was added and the reaction was stirred at 70 °C for 1 hour. The reaction was cooled to rt, then THF was removed under vacuum. The crude was resuspended in a 75% EtOAc in heptane mix and triturated for 1 hour. Filtration of the solid provided a white solid which was dried for 30 minutes. Methyl 3-bromo-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)m ethyl)benzoate (740 mg, 81 %) recuperated. LCMS ESI m/z: 444.2 [M+H] + . Step 4 3-Bromo-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)m ethyl)benzoic acid [1129] A solution of methyl 3-bromo-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoate (450 mg, 1.02 mmol), 1 M LiOH in H 2 O (2.03 mL, 2.03 mmol) and THF (5.08 mL) was stirred at 25 °C under N 2 (g) for 15 hours. The reaction was stopped and the product was precipitated out of solution by addition of 10% citric acid in water until pH was found to be acidic (pH = 2-3). Product precipitated as a bright yellow solid. Filtration over a Buchner and the solid was washed with water (2 x 20 mL), dried in vacuo.3-Bromo-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin- 1- yl)methyl)benzoic acid used as is for the next step (410 mg, 94 %). LCMS ESI m/z 429.2 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 13.31 (s, 1H), 12.44 (s, 1H), 8.13 (d, J = 8.8 Hz, 1H), 7.86 (dd, J = 5.5, 1.5 Hz, 3H), 7.28 (dd, J = 8.8, 2.4 Hz, 1H), 7.11 (d, J = 2.3 Hz, 1H), 4.85 (p, J = 7.0 Hz, 1H), 4.35 (s, 2H), 2.41 – 2.26 (m, 2H), 2.07 – 1.88 (m, 2H), 1.88 – 1.49 (m, 2H). Step 5 6-(4-(3-Bromo-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin- 1-yl)methyl)benzoyl)piperazin-1- yl)nicotinonitrile [1130] A solution of 3-bromo-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)m ethyl)benzoic acid (36.7 mg, 85.4 umol), 6-(piperazin-1-yl)nicotinonitrile (16.1 mg, 85.4 umol) and HATU (40.2 mg, 102 umol) in anhydrous DMF (854 uL) was stirred at 25 °C under N 2 (g) for 2 hours. The crude solution was purified by reverse phase chromatography (100% aqueous ammonium formate (AMF) solution (pH = 3.8) to 90% MeCN in AMF). Fractions containing pure product were lyophilized overnight providing 6- (4-(3-bromo-5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1- yl)nicotinonitrile as a white solid (51.2 mg, 71 % yield). LCMS ESI m/z 599.1 (M+H) + .1H NMR (400 MHz, DMSO-d 6 ) δ 12.45 (s, 1H), 8.49 (d, J = 2.5 Hz, 1H), 8.13 (d, J = 8.8 Hz, 1H), 7.88 (dd, J = 9.0, 2.4 Hz, 1H), 7.68 (s, 1H), 7.51 – 7.45 (m, 1H), 7.34 (s, 1H), 7.30 (dd, J = 8.8, 2.3 Hz, 1H), 7.10 (d, J = 2.4 Hz, 1H), 6.89 (d, J = 9.0 Hz, 1H), 4.87 – 4.76 (m, 1H), 4.32 (s, 2H), 3.80-3.51 (m, 6H), 2.35-2.26 (m, 2H), 2.05-1.94 (m, 2H), 1.81 – 1.54 (m, 2H). Synthesis of Example 179: 6-(4-(4-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)picolinoyl)piperazin-1-yl)nicotinonitrile 4-(Hydroxymethyl)picolinonitrile [1131] To a flame-dried, nitrogen-flushed 100 mL round-bottom flask was added 2-cyanoisonicotinic acid (1.00 g, 6.75 mmol) and anhydrous DCM (10.0 mL). The solution was cooled to 0 °C in a water/ice bath. DMF (2-3 drops) was added along with oxalyl chloride (1.75 mL, 20.3 mmol) dropwise. The reaction was stirred at 0 °C to rt over 2 hours. Then, the solution was evaoprated to dryness to provide an orange oil. It was directly redissolved in THF (10.0 mL) and cooled to 0 °C. Methanol (5.00 mL) was added and 1 M Lithium borohydride solution in THF (3.38 mL, 6.75 mmol) was added dropwise. The reaction was stirred from 0 °C to rt overnight. Additionnal amount of 1 M lithium borohydride (3.38 mL, 6.75 mmol) was added dropwise at rt. After 2 more hours, the solution was evaporated to dryness. It resulted in a gum/solid which was redissolved in EtOAc (60 mL). The organic layer was washed with HCl 1N (2 x 50 mL), and brine (2 x 50 mL). The organic layer was dried over Na 2 SO 4 , filtered and evaporated to dryness. It resulted in a yellow solid. Purification of the crude over flash chromatography (100% heptanes to 80% EtOAc in heptanes). Evaporation of the fraction provided 4- (hydroxymethyl)picolinonitrile (450 mg, 50 %) as a white solid. LCMS ESI m/z 135.1 (M+H) + . 1 H NMR (400 MHz, CDCl 3 ) δ 8.67 (d, J = 5.1 Hz, 1H), 7.73 (s, 1H), 7.51 (d, J = 4.5 Hz, 1H), 4.82 (s, 2H), 1.75 (s, 1H). Step 2 4-Formylpicolinonitrile [1132] To a 100 mL round-bottom flask was added 4-(hydroxymethyl)picolinonitrile (450 mg, 3.35 mmol), and it was dissolved in DCM (16.8 mL). The reaction was initiated by addition of Manganese (IV) oxide activated (1.94 g, 20.1 mmol) which created a black suspension. The suspension was vigorously stirred at rt and monited after 24 hours. The reaction was stopped and filtered over Celite. The celite cake was washed with DCM (2 x 30 mL) and filtrate was evaporated to dryness. It resulted in a white foam. The crude was then purified by flash chromatography (100% Heptanes to 60% EtOAc in heptanes). Evaporation of clean fraction provided 4-formylpicolinonitrile (110 mg, 25% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 10.12 (s, 1H), 9.02 (d, J = 4.9 Hz, 1H), 8.11 (s, 1H), 7.94 (dd, J = 4.9, 1.5 Hz, 1H). Step 3 4-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)pi colinonitrile [1133] A solution of dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (208 mg, 0.667 mmol), 4-formylpicolinonitrile (80 mg, 0.606 mmol) and Et 3 N (285 mg, 1.82 mmol) in anhydrous THF (6.1 mL) was stirred at 25 °C under Ar (g) for 15 hours. Hydrazine hydrate (38 mg, 1.21 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and Heptanes/ EtOAc (2:1, 30 mL) to give 4-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)pi colinonitrile (201 mg, 99 % yield) as a as a white solid. LCMS ESI m/z: 319.2 [M+H] + . Step 4 4-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)pi colinic acid [1134] A solution of 4-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)pi colinonitrile (201 mg, 605 umol), an 8.0 M aqueous solution of Sodium hydroxide (3.03 mL), and Ethanol (3.02 mL) was stirred at 25 °C then heated to 100 °C for 1 hour. The reaction was cooled to rt, then the reaction was made acidic by addition of citric acid 10% in water (30 mL) until pH was found to be 2-3. The formed solid was triturated and filtered and dried in vacuo. The solid was then triturated again in in 40% EtOAc in 60% Heptanes (30 mL) for 30 minutes before being filtered on a Buchner.4-((7-Cyclobutoxy-4-oxo- 3,4-dihydrophthalazin-1-yl)methyl)picolinic acid (170 mg, 80 %) isolated as a white solid. LCMS ESI m/z: 352.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.48 (s, 1H), 8.56 (d, J = 4.9 Hz, 1H), 8.14 (d, J = 8.8 Hz, 1H), 8.01 (s, 1H), 7.49 (d, J = 4.4 Hz, 1H), 7.30 (dd, J = 8.8, 1.9 Hz, 1H), 7.07 (d, J = 1.8 Hz, 1H), 4.90 – 4.74 (m, 1H), 4.39 (s, 2H), 2.48-2.29 (m, 2H), 2.01-1.92 (m, 2H), 1.76 (dd, J = 19.0, 9.3 Hz, 1H), 1.62 (dd, J = 19.1, 9.4 Hz, 1H). Step 5 6-(4-(4-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)picolinoyl)piperazin-1- yl)nicotinonitrile [1135] A solution of 4-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)pi colinic acid (40.0 mg, 0.114 mmol), 6-(piperazin-1-yl)nicotinonitrile (21.4 mg, 0.114 mmol) and HATU (53.6 mg, 0.137 mmol) in anhydrous DMF (1.14 mL) was stirred at 25 °C under N 2 (g) for 2 hours. The crude solution was purified by reverse phase chromatography (100% aqueous ammonium formate (AMF) solution (pH = 3.8) to 60% MeCN in AMF). Fractions containing pure product were lyophilized overnight providing 6- (4-(4-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methy l)picolinoyl)piperazin-1-yl)nicotinonitrile as a white solid (23.0 mg, 39 % yield). LCMS ESI m/z 522.3 (M+H) + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.49 (s, 1H), 8.56 – 8.44 (m, 2H), 8.14 (d, J = 8.8 Hz, 1H), 7.87 (dd, J = 9.1, 2.4 Hz, 1H), 7.60 (s, 1H), 7.40 (dd, J = 5.1, 1.5 Hz, 1H), 7.30 (dd, J = 8.8, 2.3 Hz, 1H), 7.06 (d, J = 2.3 Hz, 1H), 6.90 (d, J = 9.0 Hz, 1H), 4.83 (p, J = 6.6 Hz, 1H), 4.37 (s, 2H), 3.82 – 3.48 (m, 8H), 2.34 (ddd, J = 12.7, 9.7, 5.2 Hz, 2H), 2.03 – 1.90 (m, 2H), 1.82 – 1.54 (m, 2H). Synthesis of Example 180: 6-(4-(2-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)isonicotinoyl)piperazin-1-yl)nicotinonitrile 2-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)is onicotinonitrile [1136] A solution of dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (189 mg, 0.606 mmol), 2-formylisonicotinonitrile (80 mg, 0.606 mmol) and Et 3 N (185 mg, 1.82 mmol) in anhydrous THF (3.03 mL) was stirred at 25 °C under Ar (g) for 15 hours. Hydrazine hydrate (38.8 mg, 1.21 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/ EtOAc (2:1, 10 mL) to give 2-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)isonicotinonitrile (192 mg, 95% yield) as a as a white solid. LCMS ESI m/z 333.1 (M+H) + . Step 2 2-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)is onicotinic acid [1137] A solution of 2-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)is onicotinonitrile (180 mg, 0.542 mmol), an 8.0 M aqueous solution of sodium hydroxide (4.16 mL, 33.3 mmol), and Ethanol (2.71 mL) was stirred at 25 °C then heated to 100 °C for 1 hour. The reaction was cooled to rt, then the reaction was made acidic by addition of citric acid 10% in water (30 mL) until pH was found to be 2-3. The formed solid was triturated and filtered and dried in vacuo. The solid was then triturated again in in 40% EtOAc in 60% Heptanes (30 mL) for 30 minutes before being filtered on a Buchner.2- ((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)ison icotinic acid (170 mg, 89 % yield) isolated as a white solid. LCMS ESI m/z: 352.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.43 (s, 1H), 8.58 (d, J = 5.0 Hz, 1H), 8.10 (d, J = 8.7 Hz, 1H), 7.79 (s, 1H), 7.60 (d, J = 3.9 Hz, 1H), 7.25 (dd, J = 8.8, 2.3 Hz, 1H), 7.11 (d, J = 2.2 Hz, 1H), 4.79 – 4.70 (m, 1H), 4.46 (s, 2H), 2.40-2.27 (m, 2H), 2.04 – 1.90 (m, 2H), 1.80 – 1.53 (m, 2H). Step 3 6-(4-(2-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)isonicotinoyl)piperazin-1- yl)nicotinonitrile [1138] A solution of 2-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)is onicotinic acid (40.0 mg, 0.114 mmol), 6-(piperazin-1-yl)nicotinonitrile (21.4 mg, 0.114 mmol) and HATU (53.6 mg, 137 mmol) in anhydrous DMF (1.14 mL) was stirred at 25 °C under N 2 (g) for 2 hours. The crude solution was purified by reverse phase chromatography (100% aqueous ammonium formate (AMF) solution (pH = 3.8) to 70% MeCN in AMF). Fractions containing pure product were lyophilized overnight providing 6-(4-(2-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)isonicotinoyl)piperazin-1- yl)nicotinonitrile as a white solid (18 mg, 30 % yield). LCMS ESI m/z 522.3 (M+H) + . 1 H NMR (400 MHz, DMSO-d6) δ 12.45 (s, 1H), 8.50 (d, J = 2.2 Hz, 1H), 8.14 (d, J = 8.8 Hz, 1H), 7.90 (dd, J = 9.4, 6.6 Hz, 2H), 7.52 (d, J = 7.7 Hz, 1H), 7.48 (d, J = 7.7 Hz, 1H), 7.29 (dd, J = 8.8, 2.2 Hz, 1H), 7.08 (d, J = 2.3 Hz, 1H), 6.84 (d, J = 9.1 Hz, 1H), 4.78 – 4.67 (m, 1H), 4.46 (s, 2H), 3.72-3.64 (m, 4H), 3.29 – 3.21 (m, 2H), 2.35-2.27 (m, 2H), 1.99-1.86 (m, 2H), 1.78-1.52 (m, 2H). Synthesis of Example 181: 4-(4-(4-Fluoro-3-(4-methylpiperazine-1-carbonyl)benzyl)-1-ox o-1,2- dihydrophthalazin-6-yloxy)benzamide 4-(1-oxo-1,3-dihydroisobenzofuran-5-yloxy)benzamide [1139] To a mixture of 5-bromoisobenzofuran-1(3H)-one (5.0 g, 24 mmol) and 4-hydroxybenzamide (4.5 g, 33 mmoL) in dry DMF (100 mL) was successively added CuBr (0.65 g, 4.5 mmol), pentane-2,4- dione (0.45 g, 4.5 mmol) and K 2 CO 3 (4.5 g, 33 mmol) at room temperature. The resulting mixture was flushed with N 2 for 10 min. Then the reaction was allowed to heat to 90 °C, stirred at this temperature for overnight. The slurry was filtered over a pad of celite. The filtrate was diluted with H 2 O (100 mL). The mixture was extracted with EA (50 mL x 3). The combined organic layers were washed with 1 N NaOH (40 mL x 2). The organic layer was separated, dried, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether/ ethyl acetate = 10/1 to 1/1, v/v) to give 4-(1-oxo-1,3- dihydroisobenzofuran-5-yloxy)benzamide (1.7 g, 27% yield) as a light yellow solid. LCMS ESI m/z: 270.0 [M + H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 7.98 (d, J = 8.0 Hz, 2H), 7.88-7.86 (m, 1H), 7.74-7.72 (m, 1H), 7.71 (s, 1H), 7.37 (s, 1H), 7.20 (d, J = 8.0 Hz, 2H), 6.79-6.75 (m, 1H), 5.34 (s, 2H). Step 2 4-(4-Carbamoylphenoxy)-2-(hydroxymethyl)benzoic acid [1140] To a solution of 4-(1-oxo-1,3-dihydroisobenzofuran-5-yloxy)benzamide (1.5 g, 5.6 mmol) in MeOH (15 mL) and H 2 O (15 mL) was added NaOH (0.60 g, 17 mmol) at room temperature. The resulting mixture was stirred at the same temperature for overnight. The volatile was removed. The residue was dissolved in H 2 O (40 mL). The solution was acidified with 1 N HCl until the solution reached PH 4 ~ 5. The solids were collected by filtration and dried under vacuum to give 4-(4- carbamoylphenoxy)-2-(hydroxymethyl)benzoic acid (1.3 g, 80% yield) as a white solid, which was directly used for the next step without further purification. 1 H NMR (400 MHz, CDCl 3 ) δ 7.96-7.94 (m, 4H), 7.36 (m, 2H), 7.15-7.12 (m, 2H), 6.99 (m, 1H), 4.84 (d, J = 8.0 Hz, 2H). Step 3, 4, 5 and 6 4-(4-(4-Fluoro-3-(4-methylpiperazine-1-carbonyl)benzyl)-1-ox o-1,2-dihydrophthalazin-6- yloxy)benzamide [1141] Following the four-step procedure in Example 1, but starting with 4-(4-carbamoylphenoxy)-2- (hydroxymethyl)benzoic acid gave the title compound as a white solid. LCMS ESI m/z: 515.8 [M + H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.58 (s, 1H), 8.28 (d, J = 8.0 Hz, 1H), 7.98 (s, 1H), 7.96-7.93 (m, 2H), 7.49 (d, J = 4.0 Hz, 1H), 7.45 (dd, J = 4.0 Hz, J = 8.0 Hz, 1H), 7.37 (s, 1H), 7.32-7.28 (m, 1H), 7.25 (dd, J = 4.0 Hz, J = 8.0 Hz, 1H), 7.18 (t, J = 12.0 Hz, 1H), 7.12-7.10 (m, 2H), 4.24 (s, 2H), 3.60 (m, 2H), 3.11 (m, 2H), 2.33-2.30 (m, 2H), 2.14 (m, 5H). Synthesis of Example 182: 6-(4-(3-((7-hydroxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile

[1142] To a solution of 5-bromo-2-fluorobenzoic acid (2.0 g, 9.13 mmol) in MeOH (5 mL) was added conc. H 2 SO 4 (2 mL) in portions. The mixture was stirred at 25 °C for 20 hours. The solution was concentrated in vacuo. This residue was diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl methyl 5-bromo-2-fluorobenzoate (2.03 g, 95% yield) as a colorless oil. LCMS ESI m/z: 233 [M+H] + . Step 2 Methyl 5-bromo-2-(methylthio)benzoate [1143] To a solution of methyl 5-bromo-2-fluorobenzoate (2.03 g, 8.71 mmol) in DMAC (20 mL) was added sodium thiomethoxide (0.69 g, 9.58 mmol) in portions. The mixture was stirred at 50 °C for 3 hours, then diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 5-bromo-2-(methylthio)benzoate (1.6 g, 70.34% yield) as a colorless oil. LCMS ESI m/z: 261/263 [M+H] + . This was used without further purification. Step 3 Methyl 5-bromo-2-(methylsulfonyl)benzoate [1144] To a solution of methyl 5-bromo-2-(methylthio)benzoate (1.6 g, 6.13 mmol) in MeOH (20 mL), was added a solution of potassium peroxymonosulfate (0.69g, 9.58 mmol) in H 2 O (5 mL) dropwise. The mixture was stirred at 60 °C for 2 hours. The solution was concentrated in vacuo. This residue was diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 5- bromo-2-(methylsulfonyl)benzoate (1.55 g, 86% yield) as a white solid. LCMS ESI m/z: 293/295 [M+H] + . This was used without further purification. Step 4 Methyl 2,4-difluoro-5-vinylbenzoate [1145] A mixture of bromo-2-(methylsulfonyl)benzoate (560 mg, 1.91 mmol), potassium vinyltrifluoroborate (264 mg, 1.97 mmol), K 2 CO 3 (660 mg, 4.78 mmol) and Pd(PPh 3 ) 4 (110 mg, 0.95 mmol) in DMF (10 mL) was stirred at 80 °C under Ar (g) for 4 hours. The mixture was cooled to rt, diluted with H 2 O (5 mL) and extracted with EtOAc (10 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromategraphy (EtOAc/petroleum ether = 0% to 7%) to give methyl 2-(methylsulfonyl)-5- vinylbenzoate (453 mg, 98 % yield) as a colorless oil. LCMS ESI m/z: 241 [M+H] + . Step 5 5-Formyl-2-(methylsulfonyl)benzoic acid [1146] To a mixture of methyl 2,4-difluoro-5-vinyl-benzoate (453 mg, 1.89 mmol) in THF (24 mL) and H 2 O (18 mL) was added K 2 OsO 4 -2H 2 O (7 mg, 0.02 mmol). The mixture was stirred at room temperature for 5 min, then NaIO 4 (1.28 g, 6 mmol) was added and the mixture was stirred at 80 °C for 2 hours. The solids were removed by filtration. The filtrate was diluted with H 2 O (5 mL) and extracted with EtOAc (30 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give methyl 5-formyl-2-(methylsulfonyl)benzoate (210 mg, 43% yield) as a colorless oil. LCMS ESI m/z: 243 [M+H] + . This was used without further purification. Step 6 Methyl 5-((7-methoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-(me thylsulfonyl) benzoate [1147] A solution of dimethyl (6-methoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (211 mg, 0.78 mmol), methyl 5-formyl-2-(methylsulfonyl)benzoate (237 mg, 0.78 mmol) and Et 3 N (238 mg, 2.35 mmol) in anhydrous THF (16 mL) was stirred at 25 °C under Ar (g) for 3 hours. Hydrazine hydrate (39 mg, 1.17 mmol) was added and the reaction was stirred at 70 °C for 2 hour. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/ EtOAc (10:1, 10 mL) to give methyl 5-((7-methoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)-2-(methylsulfonyl)benzoate (154 mg, 48% yield) as a yellow solid. LCMS ESI m/z: 403 [M+H] + . Step 7

5-((7-Methoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-(me thylsulfonyl)benzoic acid [1148] A solution of methyl 5-((7-methoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2- (methylsulfonyl)benzoate (97 mg, 0.26 mmol) and NaOH (32 mg, 0.79 mmol) in MeOH (2 mL), THF (2 mL) and H 2 O (2 mL) was stirred at rt for 1 hour. The organic solvent was removed in vacuum and aq HCl (1N) was added until the solution reached to pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 5-((7-methoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2- (methylsulfonyl) benzoic acid (102 mg, 68% yield) as a white solid. LCMS ESI m/z: 389 [M+H] + . This was used without further purification. Step 8 6-(4-(5-((7-Methoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl) -2-(methylsulfonyl) benzoyl)piperazin-1-yl)nicotinonitrile [1149] To a solution of 6-piperazin-1-ylpyridine-3-carbonitrile (130 mg, 0.57 mmol) and 5-((7- methoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-(methylsu lfonyl)benzoic acid (102 mg, 0.26 mmol) in DMF (4 mL) was added EDCI (75 mg, 0.39 mmol), HOBt (53 mg, 0.39 mmol) and DIPEA (102 mg, 0.79 mmol). The reaction mixture was stirred 45 °C for 1 hour. The reaction was purified by prep-HPLC to give the 6-(4-(5-((7-methoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl) -2- (methylsulfonyl)benzoyl)piperazin-1-yl) nicotinonitrile (55 mg, 37% yield) as a white solid. LCMS ESI m/z: 559 [M+H] + . 1 H–NMR (400 MHz, DMSO-D6) δ 12.46 (s, 1H), 8.52 (d, J = 2.1 Hz, 1H), 8.19 (d, J = 8.8 Hz, 1H), 7.95 – 7.88 (m, 2H), 7.63 (dd, J = 8.2, 1.5 Hz, 1H), 7.57 (d, J = 1.3 Hz, 1H), 7.43 (dd, J = 8.8, 2.4 Hz, 1H), 7.33 (d, J = 2.3 Hz, 1H), 6.92 (d, J = 9.1 Hz, 1H), 4.46 (d, J = 2.4 Hz, 2H), 3.90 (s, 3H), 3.86 – 3.59 (m, 6H), 3.27 (s, 3H), 3.25 – 3.12 (m, 2H). Synthesis of Example 183: 6-(4-(2-Fluoro-5-((4-oxo-7-(222-trifluoroethyl)-3,4-dihydrop hthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile (4-(4-Fluoro-3-(methoxycarbonyl)benzyl)-1-oxo-1,2-dihydropht halazin-6-yl)boronic acid [1150] A mixture of methyl 5-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoate (529 mg, 1.11 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (1.41 g, 5.54 mmol), KOAc (653 mg, 6.65 mmol) and Pd(dppf)Cl 2 (49 mg, 0.06 mmol) in DMSO (10 mL) was stirred at 80 °C under Ar (g) for 2 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to afford (4-(4-fluoro-3-(methoxycarbonyl)benzyl)-1-oxo-1,2- dihydrophthalazin-6-yl)boronic acid as a light yellow solid (520 mg, 67% yield). This was used without further purification. LCMS ESI 357 (M+H) + . Step 2 tert-Butyl 4-(5-cyanopyridin-2-yl)-4-fluoropiperidine-1-carboxylate [1151] To a mixture of (4-(4-fluoro-3-(methoxycarbonyl)benzyl)-1-oxo-1,2-dihydropht halazin-6- yl)boronic acid (520 mg, 0.74 mmol), 1,1,1-trifluoro-2-iodoethane (781 mg, 3.72 mmol) and Cs 2 CO 3 (970 mg, 2.98 mmol) in DMSO (10 mL) was added Pd 2 (dba) 3 (41 mg, 0.05 mmol) and XantPhos (73 mg, 0.13 mmol) at rt under Ar (g). The reaction was stirred at 65 °C for 16 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was concentrated in vacuo and the residue was purified by prep-HPLC to give dimethyl (6-(difluoromethoxy)- 3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate as a light yellow oil (60 mg, 20% yield). LCMS ESI 357 (M+H) + . Step 3 2-Fluoro-5-((4-oxo-7-(2,2,2-trifluoroethyl)-3,4-dihydrophtha lazin-1-yl)methyl)benzoic acid [1152] A mixture of tert-butyl 4-(5-cyanopyridin-2-yl)-4-fluoropiperidine-1-carboxylate (60 mg, 0.15 mmol) and NaOH (18 mg, 0.46 mmol) in THF (3 mL) and H 2 O (1 mL) was stirred at rt for 1 hour. The organic solvent was removed under vacuum and aq HCl (1N) was added until the solution reached pH 5~6. The solids were collected by filtration, washed with water and dried under vacuum to give 2-fluoro- 5-((4-oxo-7-(2,2,2-trifluoroethyl)-3,4-dihydrophthalazin-1-y l)methyl)benzoic acid as a white solid (57 mg, 99% yield). This was used without further purification. LCMS ESI 381 (M+H) + . Step 4

6-(4-(2-Fluoro-5-((4-oxo-7-(2,2,2-trifluoroethyl)-3,4-dihydr ophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile [1153] To a solution of 2-fluoro-5-((4-oxo-7-(2,2,2-trifluoroethyl)-3,4-dihydrophtha lazin-1- yl)methyl)benzoic acid (57 mg, 0.15 mmol) and 6-(piperazin-1-yl)nicotinonitrile (44 mg, 0.18 mmol) in DMF (2 mL) was added EDCI (35 mg, 0.23 mmol), HOBt (31 mg, 0.23 mmol) and DIPEA (98 mg, 0.76 mmol). The reaction mixture was stirred at rt for 14 hours. The reaction was purified by prep-HPLC to give 6-(4-(2-fluoro-5-((4-oxo-7-(2,2,2-trifluoroethyl)-3,4-dihydr ophthalazin-1- yl)methyl)benzoyl)piperazin-1-yl)nicotinonitrile as a white solid (28 mg, 32% yield). LCMS ESI 551 (M+H) + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.27 (s, 1H), 8.46 (d, J = 8.1 Hz, 1H), 8.42 (d, J = 1.8 Hz, 1H), 7.71 – 7.61 (m, 3H), 7.36 (d, J = 5.5 Hz, 2H), 7.07 (t, J = 11.2, 6.6 Hz, 1H), 6.61 (d, J = 9.0 Hz, 1H), 4.30 (s, 2H), 3.89 (s, 4H), 3.78 (t, 2H), 3.66 (t, J = 5.1 Hz, 2H), 3.53 (dd, J = 21.5, 11.0 Hz, 2H). Synthesis of Example 184: 6-(5-(3-((4-Oxo-7-(pyridin-3-yl)-3,4-dihydrophthalazin-1- yl)methyl)benzoyl)-2,5-diazabicyclo[2.2.1]heptan-2-yl)nicoti nonitrile

6-(5-(3-((7-Bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)be nzoyl)-2,5-diazabicyclo[2.2.1]heptan- 2-yl)nicotinonitrile [1154] Following the general procedure in Example 1, but starting with 6-fluoronicotinonitrile and tert-butyl 2,5-diazabicyclo[2.2.1]heptane-2-carboxylate gave the title compound as a yellow solid. LCMS ESI m/z: 541.1 [M + H] + . Step 6 6-(5-(3-((4-Oxo-7-(pyridin-3-yl)-3,4-dihydrophthalazin-1-yl) methyl)benzoyl)-2,5- diazabicyclo[2.2.1]heptan-2-yl)nicotinonitrile [1155] To a solution of 6-(5-(3-((7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)be nzoyl)-2,5- diazabicyclo[2.2.1]heptan-2-yl)nicotinonitrile (90 mg 016 mmol) and pyridin-3-ylboronic acid (23 mg, 0.18 mmol) in DMF (2 ml) was added Pd(dppf)Cl 2 (12 mg, 0.016 mmol) and K 2 CO 3 (69 mg, 0.50 mmol) at 25 ℃. The reaction mixture was stirred at 140℃ for 4 hr under N2. The mixture was diluted with water and extracted with EtOAc, the organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated to the crude, which was purified by prep-HPLC to give 6-(5-(3-((4-oxo-7-(pyridin-3-yl)-3,4- dihydrophthalazin-1-yl)methyl)benzoyl)-2,5-diazabicyclo[2.2. 1]heptan-2-yl)nicotinonitrile (22 mg, 25% yield) as a white solid. LCMS ESI m/z: 539.7 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.66 (d, J = 20.4 Hz, 1H), 8.98 (d, J = 9.3 Hz, 1H), 8.67 (s, 1H), 8.30 (ddd, J = 45.5, 32.0, 23.2 Hz, 5H), 7.80 (dd, J = 38.9, 9.0 Hz, 1H), 7.61 – 7.27 (m, 5H), 5.54 – 4.67 (m, 2H), 4.58 – 4.19 (m, 3H), 3.61 (d, J = 10.3 Hz, 2H), 3.18 – 3.03 (m, 1H), 1.98 (dd, J = 18.3, 10.3 Hz, 2H). Synthesis of Example 185: 4-(4-Fluoro-3-(3-(trifluoromethyl)-5,6,7,8-tetrahydro-[1,2,4 ]triazolo[4,3- a]pyrazine-7-carbonyl)benzyl)-6-(prop-2-yn-1-ylamino)phthala zin-1(2H)-one Step 1

6-Bromo-4-(4-fluoro-3-(3-(trifluoromethyl)-5,6,7,8-tetrahydr o-[1,2,4]triazolo[4,3-a]pyrazine-7- carbonyl)benzyl)phthalazin-1(2H)-one [1156] To a solution of 5-bromo-3-[[4-fluoro-3-[3-(trifluoromethyl)-6,8-dihydro-5H- [1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl]phenyl]methylene]i sobenzofuran-1-one (500 mg, 930.65 μmol) in THF (10 mL) was added N 2 H 4 ·H 2 O (174.71 mg, 2.79 mmol, 80% purity). The mixture was stirred at 70 °C for 3 hours. The reaction mixture was poured into water (10 mL), extracted with EtOAc (20 mL x 3). The extracts were washed with brine, dried and concentrated to give 6-bromo-4-[[4-fluoro-3- [3-(trifluoromethyl)-6,8-dihydro-5H-[1,2,4]triazolo[4,3-a]py razine-7-carbonyl]phenyl]methyl]-2H- phthalazin-1-one (400 mg, crude) as a white solid. LCMS ESI m/z 553.0 [M + H] + . Step 2: 4-(4-Fluoro-3-(3-(trifluoromethyl)-5,6,7,8-tetrahydro-[1,2,4 ]triazolo[4,3-a]pyrazine-7- carbonyl)benzyl)-6-(prop-2-yn-1-ylamino)phthalazin-1(2H)-one [1157] To a solution of 6-bromo-4-[[4-fluoro-3-[3-(trifluoromethyl)-6,8-dihydro-5H- [1,2,4]triazolo[4,3-a]pyrazine-7-carbonyl]phenyl]methyl]-2H- phthalazin-1-one (100 mg, 181.39 μmol) in dioxane (3 mL) was added prop-2-yn-1-amine (59.94 mg, 1.09 mmol, 69.70 μL), t-BuONa (34.86 mg, 362.79 μmol), Pd(OAc) 2 (8.14 mg, 36.28 μmol) and Brettphos (19.47 mg, 36.28 μmol). The mixture was stirred at 100 °C under microwave for 1 hour. The mixture was purified by Prep-HPLC and Prep-TLC to give 4-[[4-fluoro-3-[3-(trifluoromethyl)-6,8-dihydro-5H-[1,2,4]tr iazolo[4,3-a]pyrazine-7- carbonyl]phenyl]methyl]-6-(prop-2-ynylamino)-2H-phthalazin-1 -one (3.67 mg, 3.8% yield) as a white solid. LCMS ESI 527.7 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.21 (s, 1H), 8.05 (d, J = 8.7 Hz, 1H), 7.60 (s, 1H), 7.48 (d, J = 5.2 Hz, 1H), 7.38 (t, J = 9.0 Hz, 1H), 7.17 (td, J = 8.7, 3.9 Hz, 2H), 6.91 (d, J = 18.2 Hz, 1H), 5.10 (s, 1H), 4.80 (s, 1H), 4.30 (s, 3H), 4.07 (d, J = 5.6 Hz, 3H), 3.79 (s, 1H), 3.15 (s, 1H), 2.16 – 2.01 (m, 1H). Synthesis of Example 186: 8-Fluoro-4-(4-(4-methylpiperazine-1-carbonyl)benzyl)phthalaz in-1(2H)- one 4-Formylbenzoyl chloride [1158] 4-Formylbenzoic acid (1.0 g, 6.66 mmol) was added slowly into sulfurous dichloride (3 mL) at 0 °C. The mixture was stirred at 80°C for 5 hr. The mixture was concentrated under reduced pressure to give 4-formylbenzoyl chloride (1.12 g, 100% yield) as a brown oil. LCMS ESI m/z: 541.1 [M + H] + . Step 2 4-(4-methylpiperazine-1-carbonyl)benzaldehyde [1159] To a mixture of 4-formylbenzoyl chloride (1.12 g, 6.64 mmol) in dichloromethane (13 mL) was added triethylamine (807 mg, 7.97 mmol).1-methylpiperazine (800 mg, 7.97 mmol) was added slowly. The resulting mixture was stirred at room temperature overnight. The mixture was quenched with methanol (50 mL) and concentrated under reduced pressure. The residue was purified by silica gel chromatography (methanol / dichloromethane = 1/30, v/v) to give 4-(4-methylpiperazine-1- carbonyl)benzaldehyde (482 mg, 30.8% yield) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ 10.08 (s, 1H), 8.01 (d, J = 8.1 Hz, 2H), 7.68 (d, J = 8.0 Hz, 2H), 3.59 (s, 2H), 3.30 – 3.09 (m, 6H), 2.76 (s, 3H). Step 3, 4 and 5 8-Fluoro-4-(4-(4-methylpiperazine-1-carbonyl)benzyl)phthalaz in-1(2H)-one [1160] Following the three-step procedure above in Example 6, but starting with 4-(4- methylpiperazine-1-carbonyl)benzaldehyde and dimethyl 4-fluoro-3-oxo-1,3-dihydroisobenzofuran-1- ylphosphonate gave the title compound as a white solid. LCMS ESI m/z: 380.9 [M + H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.57 (s, 1H), 7.89 (td, J = 8.1, 4.9 Hz, 1H), 7.77 (d, J = 8.1 Hz, 1H), 7.59 (dd, J = 11.3, 8.2 Hz, 1H), 7.37 (d, J = 8.1 Hz, 2H), 7.31 (d, J = 8.1 Hz, 2H), 4.32 (s, 2H), 3.48 (m, 4H), 2.41 – 2.22 (m, 4H), 2.19 (s, 3H). Synthesis of Example 187 and 188: 6-(4-(2-fluoro-5-(4-oxo-6-(prop-1-ynyl)-3,4-dihydrophthalazi n-1- yloxy)benzoyl)piperazin-1-yl)nicotinonitrile and 6-(4-(2-fluoro-5-(4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazi n-1-yloxy)benzoyl)piperazin-1- yl)nicotinonitrile

methyl 5-(7-bromo-4-chlorophthalazin-1-yloxy)-2-fluorobenzoate and methyl 5-(6-bromo-4- chlorophthalazin-1-yloxy)-2-fluorobenzoate [1161] To a solution of methyl 2-fluoro-5-hydroxy-benzoate (800 mg, 4.70 mmol) in DMF (15 mL) was added NaH (197.49 mg, 4.94 mmol, 60% purity) at 0 °C, then the mixture was added dropwise to a solution of 6-bromo-1,4-dichloro-phthalazine (1.44 g, 5.17 mmol) in THF (60 mL) during 1 hr, then stirred at rt. for 3 hr. TLC (PE:EA=10:1) showed new spots. The reaction mixture was poured into H 2 O (30 mL), extracted with EA (50 mL* 3), washed with brine, dried over Na 2 SO 4 and concentrated. The residue was purified by silica gel chromatography to give the mixture of methyl 5-(7-bromo-4- chlorophthalazin-1-yloxy)-2-fluorobenzoate and methyl 5-(6-bromo-4-chlorophthalazin-1-yloxy)-2- fluorobenzoate (1.1 g, 56.8% yield) as a yellow solid and inseperable mixture, it will be used in the next step directly. For major isomer: 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.70 (d, J = 1.8 Hz, 1H), 8.44 (dd, J = 8.8, 1.9 Hz, 1H), 8.27 (d, J = 8.8 Hz, 1H), 7.97 (dd, J = 6.0, 3.0 Hz, 1H), 7.80 (dd, J = 5.2, 3.2 Hz, 1H), 7.58 (dd, J = 10.3, 9.1 Hz, 1H), 3.93 (s, 3H). Step 2 Methyl 5-(7-bromo-4-oxo-3,4-dihydrophthalazin-1-yloxy)-2-fluorobenz oate and methyl 5-(6-bromo- 4-oxo-3,4-dihydrophthalazin-1-yloxy)-2-fluorobenzoate [1162] To a 100 mL round bottle was added the mixture of methyl 5-(7-bromo-4-chloro-phthalazin-1- yl)oxy-2-fluoro-benzoate and methyl 5-(6-bromo-4-chloro-phthalazin-1-yl)oxy-2-fluoro-benzoate (800 mg, 1.94 mmol), then CH 3 COOH (20 mL) was added. The reaction mixture was heated to 120 °C for 2 hr. The reaction mixture was concentrated to give a residue, poured into H 2 O (20 mL), stirred at rt. for 2 minutes. The solids were collected by filtration and dried under vacuum to give the mixture of methyl 5- (7-bromo-4-oxo-3,4-dihydrophthalazin-1-yloxy)-2-fluorobenzoa te and methyl 5-(6-bromo-4-oxo-3,4- dihydrophthalazin-1-yloxy)-2-fluorobenzoate as an inseparable mixture (760 mg, 99% yield) as a white solid. LCMS ESI m/z: 390.7 [M - H] -. Step 3 5-(7-Bromo-4-oxo-3,4-dihydrophthalazin-1-yloxy)-2-fluorobenz oic acid and 5-(6-bromo-4-oxo-3,4- dihydrophthalazin-1-yloxy)-2-fluorobenzoic acid [1163] To a solution of the mixture of methyl 5-(7-bromo-4-oxo-3,4-dihydrophthalazin-1-yloxy)-2- fluorobenzoate and methyl 5-(6-bromo-4-oxo-3,4-dihydrophthalazin-1-yloxy)-2-fluorobenz oate (760 mg, 1.93 mmol) in H 2 O (10 mL) and THF (10 mL) was added LiOH (138.89 mg, 5.80 mmol), then stirred at rt. for 2 hr. The mixture was washed with DCM, the aqueous layer was collected and acidified with 1 M HCl aq. until the solution reached pH 5. The solids were collected by filtration and dried under vacuum to give 5-(7-bromo-4-oxo-3,4-dihydrophthalazin-1-yloxy)-2-fluorobenz oic acid and 5-(6-bromo-4-oxo-3,4- dihydrophthalazin-1-yloxy)-2-fluorobenzoic acid as an inseparable mixture (580 mg, 79% yield) as a white solid. LCMS ESI m/z: 390.7 [M - H] -. Step 4 and 5

6-(4-(2-fluoro-5-(4-oxo-6-(prop-1-ynyl)-3,4-dihydrophthalazi n-1-yloxy)benzoyl)piperazin-1- yl)nicotinonitrile and 6-(4-(2-fluoro-5-(4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazi n-1- yloxy)benzoyl)piperazin-1-yl)nicotinonitrile [1164] Following the general two-step procedure above in Example 1, but starting with the mixture of 5-(7-bromo-4-oxo-3,4-dihydrophthalazin-1-yloxy)-2-fluorobenz oic acid and 5-(6-bromo-4-oxo-3,4- dihydrophthalazin-1-yloxy)-2-fluorobenzoic acid gave the mixture of title compound (80 mg) as a white solid. The mixture was purified by prepare TLC (toluene/DCM/THF = 1/6/1, Petroleum ether/toluene/DCM/THF= 1/ 2/ 3/ 2 to petroleum ether/toluene/DCM/THF =1/ 2/ 2/ 2) to give 6-(4-(2- fluoro-5-(4-oxo-6-(prop-1-ynyl)-3,4-dihydrophthalazin-1-ylox y)benzoyl)piperazin-1-yl)nicotinonitrile (13 mg) and 6-(4-(2-fluoro-5-(4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazi n-1-yloxy)benzoyl)piperazin-1- yl)nicotinonitrile (10.8 mg) as white solids. LCMS ESI m/z: 509.2 [M+H] + . [1165] 6-(4-(2-fluoro-5-(4-oxo-6-(prop-1-ynyl)-3,4-dihydrophthalazi n-1-yloxy)benzoyl)piperazin-1- yl)nicotinonitrile 1 H NMR (400 MHz, DMSO-d 6 ) δ12.06 (s, 1H) , 8.51 (d, J = 2.2 Hz, 1H), 8.14 (d, J = 1.2 Hz, 1H), 8.07 (d, J = 8.3 Hz, 1H), 7.95 (dd, J = 8.3, 1.6 Hz, 1H), 7.90 (dd, J = 9.1, 2.3 Hz, 1H), 7.49 – 7.38 (m, 3H), 6.94 (d, J = 9.1 Hz, 1H), 3.81 – 3.66 (m, 6H), 3.38 (s, 2H), 2.14 (s, 3H). [1166] 6-(4-(2-fluoro-5-(4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazi n-1-yloxy)benzoyl)piperazin-1- yl)nicotinonitrile 1 H NMR (400 MHz, DMSO-d 6 ) δ12.03 (s, 1H), 8.51 (d, J = 2.1 Hz, 1H), 8.21 (d, J = 8.2 Hz, 1H), 8.03 (s, 1H), 7.94 – 7.87 (m, 2H), 7.52 – 7.38 (m, 3H), 6.94 (d, J = 9.1 Hz, 1H), 3.81 – 3.67 (m, 6H), 3.39 (s, 2H), 2.13 (s, 3H). Synthesis of Example 189: 2-((1-((5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)m ethyl)-2- fluorophenyl) sulfonyl)azetidin-3-yl)(methyl)amino)isonicotinonitrile

Step 1 3-((3-((tert-butoxycarbonyl)(methyl)amino)azetidin-1-yl)sulf onyl)-4-fluorobenzoic acid [1167] A solution of 3-(chlorosulfonyl)-4-fluorobenzoic acid (1 g, 4.19 mmol), tert-butyl azetidin-3- yl(methyl)carbamate (780 mg, 4.19 mmol) and Et 3 N (1.7 g, 16.76 mmol) in THF (10 mL) was stirred ar rt for 2 hours. The solution was concentrated in vacuo, then diluted with water (50 mL) and extracted with EtOAc (30 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (DCM/MEOH= 10/1) to give 3-((3-((tert-butoxycarbonyl)(methyl)amino)azetidin-1-yl)sulf onyl)-4-fluorobenzoic acid (1.40 g, 88% yield) as a white solid. LCMS ESI m/z: 389.1 [M+H] + . Step 2 Methyl 3-((3-((tert-butoxycarbonyl)(methyl)amino)azetidin-1-yl)sulf onyl)-4-fluorobenzoate [1168] To a mixture of 3-((3-((tert-butoxycarbonyl)(methyl)amino)azetidin-1-yl)sulf onyl)-4- fluorobenzoic acid (1.4 g, 3.71 mmol) and K 2 CO 3 (768 mg, 5.56 mmol) in DMF (15 mL) was added MeI (631 mg,4.45 mmol). The reaction was stirred at rt for 2 hours, then diluted with water (50 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (PE/EA= 1/1) to afford methyl 3-((3-((tert-butoxycarbonyl) (methyl) amino) azetidin-1- yl)sulfonyl)-4-fluorobenzoate(1.10 g, 65% yield) as a colorless oil. LCMS ESI m/z: 403.1 [M+H] + . Step 3 tert-butyl (1-((2-fluoro-5-(hydroxymethyl)phenyl)sulfonyl)azetidin-3-yl )(methyl) carbamate [1169] To a solution of methyl 3-((3-((tert-butoxycarbonyl)(methyl)amino)azetidin-1-yl)sulf onyl)-4- fluorobenzoate (1.1 g, 2.73 mmol) in THF (10 mL) was added NaBH 4 ( 1.5 g, 40 mmol). The mixture was stirred at rt for 2 hours, then diluted with water (50 mL) and extracted with EtOAc (30 mL × 3). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (PE/EA= 1/1) to afford tert-butyl (1-((2-fluoro-5-(hydroxymethyl) phenyl) sulfonyl)azetidin-3-yl)(methyl)carbamate (780 mg, 65% yield) as a yellow oil. LCMS ESI m/z: 375.0 [M+H] + . Step 4 tert-butyl (1-((2-fluoro-5-formylphenyl)sulfonyl)azetidin-3-yl)(methyl) carbamate [1170] A mixture of tert-butyl (1-((2-fluoro-5-(hydroxymethyl)phenyl)sulfonyl)azetidin-3- yl)(methyl)carbamate (780 mg, 2.08 mmol) and MnO 2 (2.71 g, 27.94 mmol) in DCM (20 mL) was stirred under reflux for 2 hours. The reaction mixture was cooled to rt and filtered. The filtrate was concentrated in vacuo to give tert-butyl (1-((2-fluoro-5-formylphenyl)sulfonyl)azetidin-3-yl)(methyl) carbamate (650 mg, 83% yield) as a white solid. LCMS ESI m/z: 373.1 [M+H] + . This was used without further purification. Step 5 tert-butyl (1-((5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)meth yl)-2- fluorophenyl)sulfonyl)azetidin-3-yl)(methyl)carbamate [1171] A solution of tert-butyl (1-((2-fluoro-5-formylphenyl)sulfonyl)azetidin-3-yl)(methyl) carbamate (650 mg, 1.75 mmol) and methyl dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1- yl)phosphonate (546 mg, 1.75 mmol) in anhydrous THF (6 mL) was stirred at 25 °C under Ar (g) for 3 hours. Hydrazine hydrate (130 mg, 2.62 mmol) was added and the reaction was stirred at 70 °C for 2 hours. The reaction was cooled to rt, then THF was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/EA (10:1, 10 mL) to give tert-butyl (1-((5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor ophenyl)sulfonyl)azetidin-3- yl)(methyl)carbamate (640 mg, 64% yield) as a yellow solid. LCMS ESI m/z: 573.2 [M+H] + . Step 6 6-cyclobutoxy-4-(4-fluoro-3-((3-(methylamino)azetidin-1-yl)s ulfonyl)benzyl) phthalazin-1(2H)-one [1172] To a solution of tert-butyl (1-((5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)meth yl)-2- fluorophenyl)sulfonyl)azetidin-3-yl)(methyl)carbamate (640 mg, 1.12 mmol) in dioxane (5 mL) was added HCl/dioxane (4M, 10 mL) and stirred at 25 °C for 1 hour. The mixture was concentrated in vacuo to afford 6-cyclobutoxy-4-(4-fluoro-3-((3-(methylamino)azetidin-1-yl)s ulfonyl)benzyl)phthalazin-1(2H)- one (528 mg, 100% yield) as a white solid. LCMS ESI m/z: 473.1 [M+H] + . This was used without further purification. Step 7

2-((1-((5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)m ethyl)-2-fluorophenyl) sulfonyl)azetidin-3-yl)(methyl)amino)isonicotinonitrile [1173] A solution of 6-cyclobutoxy-4-(4-fluoro-3-((3-(methylamino)azetidin-1-yl)s ulfonyl) benzyl)phthalazin-1(2H)-one (140 mg, 0.28 mmol), 2-fluoroisonicotinonitrile (140 mg, 11.38 mmol) in DMSO (2 mL) was stirred at 80 o C for 16 hours. The reaction was purified by prep-HPLC to give 2-((1- ((5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl) -2-fluorophenyl)sulfonyl)azetidin-3- yl)(methyl)amino)isonicotinonitrile (57 mg, 33% yield) as a white solid. LCMS ESI m/z: 575.1 [M+H] + . 1H-NMR (400 MHz, DMSO-d 6 ) δ 12.47 (s, 1H), 8.41 (d, J = 3.0 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.83 (s, 1H), 7.60 (d, J = 5.1 Hz, 1H), 7.47 – 7.37 (m, 1H), 7.30 (dd, J = 8.8, 2.3 Hz, 1H), 7.21 (d, J = 4.1 Hz, 1H), 7.18 – 7.08 (m, 2H), 4.88 – 4.78 (m, 1H), 4.75 – 4.64 (m, 1H), 4.61 – 4.49 (m, 1H), 4.42 – 4.16 (m, 4H), 3.25 – 3.17 (m, 1H), 2.96 (s, 3H), 2.45 – 2.35 (m, 2H), 2.08 – 1.93 (m, 2H), 1.84 – 1.73 (m, 1H), 1.72 – 1.61 (m, 1H). Synthesis of Example 190: 6-(4-(2-fluoro-5-((4-oxo-3,4-dihydrophthalazin-1- yl)oxy)benzoyl)piperazin-1-yl)nicotinonitrile [1174] To a solution of 2-fluoro-5-((4-oxo-3,4-dihydrophthalazin-1-yl)oxy)benzoic acid (0.062 g, 0.2 mmol) synthesized following Example 185 and 186 procedures and 6-(piperazin-1-yl)nicotinonitrile (0.040 g, 0.2 mmol) in anhydrous DMF (1.0 mL) at 0 o C was added diisopropylethylamine (0.10 mL, 0.6 mmol), followed by addition of propylphosphonic anhydride (T3P) (0.25 mL, 0.4 mmol) (50% in EtOAc). The mixture was stirred for 30 min at rt. Upon completion of the reaction, it was dissolved in ethyl acetate (5 mL) and added with sat. NaHCO 3 (5 mL). The layers were separated and the aqueous layer was extracted with ethyl acetate. The organic extracts were washed with brine (5 mL), filtered through a silica plug and dried over MgSO 4 . After filtration, the solvent was removed in vacuo. Flash chromatography over silica gel (0 to 100% EtOAc in Heptanes) afforded the product as a white solid (0.0274 g, 28% yield). LCMS ESI m/z 471.2 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 10.20 (s, 1H), 8.40 – 8.32 (m, 2H), 8.11 – 8.04 (m, 1H), 7.83 (dtd, J = 23.1, 7.4, 1.3 Hz, 2H), 7.57 (dd, J = 9.0, 2.3 Hz, 1H), 7.31 – 7.22 (m, 2H), 7.17 – 7.07 (m, 1H), 6.60 – 6.55 (m, 1H), 3.84 (s, 2H), 3.71 (dq, J = 12.0, 6.8, 6.0 Hz, 4H), 3.63 – 3.45 (m, 2H). Synthesis of Example 191: 4-(((4-Oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin -1-yl)butan- 2-yl)amino)methyl)-6-(prop-1-yn-1-yl)phthalazin-1(2H)-one Step 1 6-Bromo-4-(hydroxymethyl)phthalazin-1(2H)-one [1175] To a solution of methyl 7-bromo-4-oxo-3,4-dihydrophthalazine-1-carboxylate (600 mg, 2.12 mmol) in MeOH (5 mL) was added NaBH 4 (802 mg, 21.20 mmol), the mixture was stirred at rt for 16 hours. The solvent was removed in vacuo, the residue was diluted with H2O (10 mL), aq 6N HCl was added until the solution reached pH 4. The solid was colleted by filtration and dried under vacuum to give 6-bromo-4-(hydroxymethyl)-2H-phthalazin-1-one (358 mg, 1.40 mmol, 66% yield) as a white solid. LCMS ESI m/z: 255.1 [M+H] + . Step 2 6-Bromo-4-(bromomethyl)phthalazin-1(2H)-one [1176] To a solution of 6-bromo-4-(hydroxymethyl)-2H-phthalazin-1-one (258 mg, 1.01 mmol) in THF (10 mL) was added PBr 3 (411 mg, 1.52 mmol) slowly, the mixture was stirred at rt for 0.5 h. The reaction was quenched with water (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic phase was washed with sat. NaCl solution, dried over Na 2 SO 4 , filtered and concentrated in vacuo to give 6-bromo-4-(bromomethyl)phthalazin-1(2H)-one (226 mg, 70% yield) as a white solid. LCMS ESI m/z: 317.1 [M+H] + . Step 3 6-Bromo-4-(((4-oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl)p iperazin-1-yl)butan-2- yl)amino)methyl)phthalazin-1(2H)-one [1177] A solution of 6-bromo-4-(bromomethyl)phthalazin-1(2H)-one (276 mg, 0.89 mmol), 3-amino- 1-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1-yl)butan -1-one (307 mg, 0.88 mmol) and DIPEA (337 mg, 2.60 mmol) in DMF (3 mL) was stirred at rt for 1 hour. The mixture was purified by prep-HPLC to give 6-bromo-4-(((4-oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl)p iperazin-1-yl)butan-2- yl)amino)methyl)phthalazin-1(2H)-one (130 mg, 27% yield) as a white solid. LCMS ESI m/z: 554.1 [M+H] + . Step 4

4-(((4-Oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin -1-yl)butan-2-yl)amino)methyl)-6- (prop-1-yn-1-yl)phthalazin-1(2H)-one [1178] To a solution of 6-bromo-4-[[[1-methyl-3-oxo-3-[4-[5-(trifluoromethyl)pyrimid in-2- yl]piperazin-1-yl]propyl]amino]methyl]-2H-phthalazin-1-one (110 mg, 0.20 mmol) in 1,4-dioxane (10 mL) was added Pd(dppf)Cl 2 (15 mg, 0.20 mmol) and tributyl(prop-1-ynyl)stannane (196 mg, 0.60 mmol), then the mixture was stirred at 120 °C under Ar atmosphere for 2 hours. The mixture was purified by prep-HPLC to afford 4-(((4-oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin -1-yl)butan-2- yl)amino)methyl)-6-(prop-1-yn-1-yl)phthalazin-1(2H)-one (75 mg, 0.13 mmol, 68% yield) as a white solid. LCMS ESI m/z: 514.2 [M+H] + . 1 H NMR (400 MHz, CDCl 3 ) δ 12.61 (s, 1H), 8.73 (s, 2H), 8.17 (d, J = 7.8 Hz, 1H), 8.14 (s, 1H), 8.11 (s, 1H), 7.75 (dd, J = 8.2, 1.3 Hz, 1H), 4.13 (d, J = 13.8 Hz, 1H), 3.98 (d, J = 13.8 Hz, 1H), 3.85 – 3.70 (m, 6H), 3.51 (d, J = 4.5 Hz, 2H), 3.15 (dd, J = 12.5, 6.3 Hz, 2H), 2.63 (dd, J = 15.2, 7.0 Hz, 1H), 2.44 – 2.34 (m, 1H), 2.10 (s, 3H), 1.15 (d, J = 6.3 Hz, 3H). Synthesis of Example 192: 4-(1-(3-Oxo-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi n-1- yl)propoxy)ethyl)-6-(prop-1-ynyl)phthalazin-1(2H)-one

6-Bromo-4-ethylphthalazin-1(2H)-one [1179] A mixture of dimethyl 6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-ylphosphonate (1.00 g, 3.13 mmol), acetaldehyde (275 mg, 6.26 mmmol) and Et 3 N (947 mg, 9.39 mmol) in THF (10mL) was stirred at rt for 12 hours. To the reaction mixture, N 2 H 4 •H 2 O (368 mg, 6.26 mmol) was added and stirred at 70°C for 2 hours. The reaction was cooled to rt, concentrated in vacuo and the residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 20%) to give 6-bromo-4-ethylphthalazin-1(2H)-one as a yellow solid (790 mg, 99% yield). LCMS ESI m/z: 253 [M+H] + . 1 HNMR (400 MHz, DMSO–d 6 ) δ 12.58 (s, 1H), 8.18 (s, 1H), 8.16 (d, J = 8.8 Hz, 1H), 8.01 (dd, J = 8.4, 1.6 Hz, 1H), 2.99 (q, J = 14.8, 7.2 Hz, 2H), 1.23 (t, J = 7.2 Hz, 3H). Step 2 6-Bromo-4-(1-bromoethyl)phthalazin-1(2H)-one [1180] A mixture of 5-bromo-7-(trifluoromethyl)isobenzofuran-1(3H)-one (790 mg, 3.12 mmol), NBS (667 mg, 3.74 mmol) and AIBN (102 mg, 0.62 mmol) in CCl 4 (10 mL) was stirred at 80°C for 8 hours. The mixture was concentrated in vacuo, and the residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 20%) to give 6-bromo-4-(1-bromoethyl)phthalazin-1(2H)-one as a yellow solid (460 mg, 45% yield). LCMS ESI m/z: 331 [M+H] + . 1 H NMR (400 MHz, DMSO–d 6 ) δ 12.68 (s, 1H), 8.36 (s, 1H), 8.19 (d, J = 8.8 Hz, 1H), 8.04 (dd, J = 8.4, 1.6 Hz, 1H), 6.10 (q, J = 6.4 Hz, 1H), 2.05 (d, J = 6.4 Hz, 3H). Step 3 3-(1-(7-Bromo-4-oxo-3,4-dihydrophthalazin-1-yl)ethoxy)propan oic acid [1181] A solution of 6-bromo-4-(1-bromoethyl)phthalazin-1(2H)-one (460 mg, 1.39 mmol) in methyl 3-hydroxypropanoate (0.6 mL) were stirred at 100 °C for 5 hours. The reaction was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (3x 20 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was dissolved in MeOH (3 mL), then added a solution of LiOH (580 mg, 13.9 mmol) in H 2 O (1 mL). The reaction mixture was stirred at rt for 6 hours, then aq HCl (1N) was added until the solution reached pH 5~6. The mixture was extracted with EtOAc (3 × 20 mL). The combined organic layer was dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 60 to 80%) to give 3-(1-(7- bromo-4-oxo-3,4-dihydrophthalazin-1-yl)ethoxy)propanoic acid as a yellow solid (210 mg, 38% yield). LCMS ESI m/z: 341 [M+H] + . Step 4 6-Bromo-4-(1-(3-oxo-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazin-1- yl)propoxy)ethyl)phthalazin-1(2H)-one [1182] A solution of 3-(1-(7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)ethoxy)propan oic acid (210 mg, 0.62 mmol), EDCI (118 mg, 0.62 mmol), 2-(piperazin-1-yl)-5-(trifluoromethyl)pyrimidine (145 mg, 0.62 mmol), HOBT(126 mg, 0.93 mmol) and DIPEA (320 mg, 2.48 mmol) in DMF (5 mL) were stirred at rt for 6 hours. The mixture was diluted with H 2 O (5 mL) and extracted with EtOAc (3 × 20 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 80 to 100%) to give 6-bromo-4-(1-(3- oxo-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1-yl)p ropoxy)ethyl)phthalazin-1(2H)-one as a white solid (260 mg, 76% yield). LCMS ESI m/z: 555 [M+H] + . Step 5 4-(1-(3-Oxo-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazi n-1-yl)propoxy)ethyl)-6-(prop-1- ynyl)phthalazin-1(2H)-one [1183] A mixture of 6-bromo-4-(1-(3-oxo-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl) piperazin-1- yl)propoxy)ethyl)phthalazin-1(2H)-one (260 mg, 0.47 mmol), tributyl(prop-1-ynyl)stannane (618 mg, 1.87 mmol) and Pd(dppf)Cl 2 (37 mg, 0.05 mmol) in 1,4-dioxane (8 mL) were stirred at 100 °C for 5 hours. The mixture was cooled to rt, diluted with water (20 mL) and extracted with EtOAc (3 × 20 mL). The combined organic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (MeOH/DCM = 0 to 5%) to give 4-(1-(3-oxo-3-(4-(5- (trifluoromethyl)pyrimidin-2-yl)piperazin-1-yl)propoxy)ethyl )-6-(prop-1-ynyl)phthalazin-1(2H)-one as a white solid (75 mg, 30% yield). LCMS ESI m/z: 515.3 [M+H] + . 1 H NMR (400 MHz, MeOD) δ 8.58 (s, 2H), 8.28 – 8.23 (m, 2H), 7.74 (dd, J = 8.3, 1.4 Hz, 1H), 4.90 – 4.87 (m, 1H), 3.88 – 3.81 (m, 5H), 3.77 – 3.70 (m, 1H), 3.67 – 3.59 (m, 4H), 2.79 – 2.60 (m, 2H), 2.08 (s, 3H), 1.62 (d, J = 6.7 Hz, 3H). Synthesis of Example 193: 4-(1-(Methyl(3-oxo-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)p iperazin-1- yl)propyl) amino)ethyl)-6-(prop-1-yn-1-yl)phthalazin-1(2H)-one

tert-Butyl (4-oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1-y l)butan-2-yl)carbamate [1184] To a solution of 3-((tert-butoxycarbonyl)(methyl)amino)propanoic acid (1.00 g, 4.92 mmol) and 2-(piperazin-1-yl)-5-(trifluoromethyl)pyrimidine (1.32 g, 4.92 mmol) in DMF (10 mL) was added EDCI (1.41 g, 7.38 mmol), HOBt (1.00 g, 7.38 mmol) and DIPEA (1.59 g, 12.30 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0% to 33%) to give tert-butyl methyl(3-oxo-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperaz in-1- yl)propyl)carbamate (1.95 g, 91% yield) as a white solid. LCMS ESI m/z: 418 [M+H] + . Step 2 3-(Methylamino)-1-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pipe razin-1-yl)propan-1-one [1185] A solution of tert-butyl 4-(5-cyanopyridin-2-yl)piperazine-1-carboxylate (1.95 g, 9.58 mmol) in HCl/Dioxane (4M, 5 mL) was stirred at 25 °C for 1 hour. The reaction mixture was concentrated in vacuo to give 3-(methylamino)-1-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pipe razin-1-yl)propan-1-one (1.48 g, 99% yield) as a white solid. LCMS ESI m/z: 318 [M+H] + . This was used without further purification. Step 3

6-Bromo-4-(1-(methyl(3-oxo-3-(4-(5-(trifluoromethyl)pyrimidi n-2-yl)piperazin-1- yl)propyl)amino)ethyl)phthalazin-1(2H)-one [1186] A solution of 6-bromo-4-(1-bromoethyl)phthalazin-1(2H)-one (113 mg, 0.34 mmol), 3- (methylamino)-1-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pipera zin-1-yl)propan-1-one (140 mg, 0.44 mmol) and DIPEA (175 mg, 1.36 mmol) in MeCN (5 mL) was stirred at rt for 2 hours. The reaction was purified by prep-HPLC to give 6-bromo-4-(1-(methyl(3-oxo-3-(4-(5-(trifluoromethyl)pyrimidi n-2-yl)piperazin-1- yl)propyl)amino)ethyl)phthalazin-1(2H)-one (183 mg, 95% yield) as a white solid. LCMS ESI m/z: 568 [M+H] + . Step 4 4-(1-(methyl(3-oxo-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)p iperazin-1-yl)propyl)amino)ethyl)-6- (prop-1-yn-1-yl)phthalazin-1(2H)-one [1187] A mixture of tributyl(3,3,3-trifluoroprop-1-ynyl)stannane (260 mg, 0.68 mmol), 6-bromo-4-[1- [methyl-[3-oxo-3-[4-[5-(trifluoromethyl)pyrimidin-2-yl]piper azin-1-yl]propyl]amino]ethyl]-2H- phthalazin-1-one (129 mg, 0.23 mmol) and Pd(dppf)Cl 2 (16 mg, 0.04 mmol) in DMF (4 mL) was stirred at 100 °C under Ar (g) for 19 hours. The reaction mixture was concentrated in vacuo and the residue was purified by prep-HPLC to afford 4-(1-(methyl(3-oxo-3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)p iperazin-1- yl)propyl) amino)ethyl)-6-(prop-1-yn-1-yl)phthalazin-1(2H)-one (42 mg, 35 % yield) as a white solid. LCMS ESI m/z: 528 [M+H] + . 1 H-NMR (400 MHz, DMSO-d 6 ) δ 12.58 (s, 1H), 8.73 (s, 2H), 8.18 – 8.09 (m, 2H), 7.70 (d, J = 8.2 Hz, 1H), 4.40 – 4.37 (m, 1H), 3.82 – 3.74 (m, 2H), 3.65 (t, J = 4.9 Hz, 2H), 3.47 (t, J = 4.9 Hz, 2H), 3.39 – 3.35 (m, 1H), 3.32 – 3.25 (m, 1H), 2.75 – 2.66 (m, 2H), 2.43 (t, J = 6.7 Hz, 2H), 2.25 (s, 3H), 2.07 (s, 3H), 1.28 (d, J = 6.6 Hz, 3H). Synthesis of Example 194: 4-(((4-Oxo-4-(4-(5(trifluoromethyl)pyrimidin-2-yl)piperazin- 1-

tert-Butyl (4-oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1-y l)butan-2-yl)carbamate [1188] To a solution of 3-((tert-butoxycarbonyl)amino)butanoic acid (2.5 g, 12.30 mmol) and 2- (piperazin-1-yl)-5-(trifluoromethyl)pyrimidine (2.86 g, 10.63 mmol) in DMF (30 mL) was added EDCI (3.54 g, 18.45 mmol), HOBt (2.49 g, 18.45 mmol) and DIPEA (4.77 g, 36.90 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 40 to 50%) to give tert-butyl (4-oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1-y l)butan-2- yl)carbamate as a white solid (4.01 g, 78% yield). LCMS ESI m/z: 418 [M+H] + . Step 2 3-Amino-1-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1- yl)butan-1-one [1189] To a solution of tert-butyl 4-(5-cyanopyridin-2-yl)piperazine-1-carboxylate (4.00 g, 9.58 mmol) in 1,4-dioxane (20 mL) was added HCl/dioxane (4 M, 5 mL), then stirred at 25 °C for 1 hour. The reaction mixture was concentrated in vacuo to give 3-amino-1-(4-(5-(trifluoromethyl)pyrimidin-2- yl)piperazin-1-yl)butan-1-one as a white solid (3.73 g, 99% yield). This was used without further purification. LCMS ESI m/z: 318 [M+H] + . Step 3 6-Bromo-4-(1-((4-oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl )piperazin-1-yl)butan-2- yl)amino)ethyl)phthalazin-1(2H)-one [1190] To a solution of 6-bromo-4-(1-bromoethyl)phthalazin-1(2H)-one (160 mg, 0.48 mmol) and 3- amino-1-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1-yl )butan-1-one (199 mg, 0.63 mmol) in MeCN (5 mL) was added DIPEA (249 mg, 1.93 mmol). The reaction mixture was stirred at rt for 2 hours. The mixture was concentrated in vacuo and the residue was purified by prep-HPLC to give 6-bromo-4-(1-((4- oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1-yl)b utan-2-yl)amino)ethyl)phthalazin-1(2H)-one as a white solid (60 mg, 22% yield). LCMS ESI m/z: 568 [M+H] + . Step 4 4-(((4-Oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin -1-yl)butan-2-yl)amino)methyl)-6- (prop-1-yn-1-yl)phthalazin-1(2H)-one [1191] A mixture of tributyl(prop-1-ynyl)stannane (107 mg, 0.33 mmol), 6-bromo-4-(1-((4-oxo-4-(4- (5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1-yl)butan2yl)a mino)ethyl) phthalazin-1-(2H)-one (60 mg, 0.11 mmol) and Pd(dppf)Cl 2 (6 mg, 0.08 mmol) in 1,4-dioxane (6 mL) was stirred at 100 °C under Ar (g) for 2 hours. The mixture was concentrated in vacuo and the residue was purified by prep-HPLC to give 4- (((4-oxo-4-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1 -yl)butan-2-yl)amino)methyl)-6-(prop-1-yn- 1-yl)phthalazin-1(2H)-one as a white solid (35 mg, 63% yield). LCMS ESI m/z: 528.1 [M+H] + . 1 HNMR (400 MHz, DMSO-d6) δ 12.62 (d, J = 4.5 Hz, 1H), 8.73 (s, 2H), 8.40 (d, J = 13.0 Hz, 1H), 8.17 (d, J = 9.2 Hz, 1H), 7.80 – 7.73 (m, 1H), 4.45 (dd, J = 21.8, 6.5 Hz, 1H), 3.84 – 3.67 (m, 4H), 3.53 – 3.44 (m, 5H), 3.08 (d, J = 6.2 Hz, 1H), 2.96 (d, J = 6.4 Hz, 1H), 2.37 – 2.26 (m, 1H), 2.10 (d, J = 11.6 Hz, 3H), 1.42 – 1.34 (m, 3H), 1.06 (dd, J = 6.1, 2.6 Hz, 3H). Synthesis of Example 195: 4-(6-Oxo-6-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1 -yl)hexan-2- yl)-6-(prop-1-yn-1-yl)phthalazin-1(2H)-one Ethyl-5-(6-bromo-3-oxoisobenzofuran-1(3H)-ylidene)hexanoate [1192] To a solution of dimethyl (6-bromo-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphonate (2.0 g, 6.23 mmol) in THF (18 mL) was added LiHMDS (1.0 M in THF, 6.23 mL, 6.23 mmol) at -78 °C, then stirred at -78 °C for 1 hour. Ethyl 5-oxohexanoate (1.18 g, 7.48 mmol) was added to the mixture and the reaction was stirred at rt for 18 hours. The mixture was diluted with H 2 O (50 mL) and extracted with DCM (100 mL x 3). The combined organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0 to 10%) to afford ethyl 5-(6-bromo-3-oxoisobenzofuran-1(3H)-ylidene)hexanoate as a colorless oil (590 mg, 27% yield). LCMS ESI m/z: 353 [M+H] + . Step 2 Ethyl 5-(7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)hexanoate [1193] To a solution of ethyl 5-(6-bromo-3-oxoisobenzofuran-1(3H)-ylidene)hexanoate (590 mg, 1.67 mmol) in THF (10 mL) was added N 2 H 4 •H 2 O (340 mg, 16.70 mmol). The reaction mixture was stirred at 60 °C for 3 hours, then cooled to rt, diluted with H 2 O (5 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 10 to 28%) to give ethyl 5-(7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)hexanoate as a white solid (350 mg, 57% yield). LCMS ESI m/z: 367 [M+H] + . 5-(7-Bromo-4-oxo-3,4-dihydrophthalazin-1-yl)hexanoic acid [1194] To a solution of methyl 2-bromo-4-cyclopropoxybenzoate (350 mg, 0.95 mmol) in 1,4-dioxane (4 mL) and H 2 O (2 mL) was added LiOH-H 2 O (400 mg, 9.53 mmol). The mixture was stirred at 25 °C for 3 hours. The organic solvent was removed in vacuo and aq HCl (1N) was added until the solution reached pH 5~6, then extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to afford 2-bromo-4- cyclopropoxybenzoic acid as a white solid (320 mg, 86% yield). This was used without further purification. LCMS ESI m/z: 339 [M+H] + . Step 4 6-Bromo-4-(6-oxo-6-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pip erazin-1-yl)hexan-2-yl)phthalazin- 1(2H)-one [1195] To a solution of 5-(7-bromo-4-oxo-3,4-dihydrophthalazin-1-yl)hexanoic acid (100 mg, 0.29 mmol) and 2-(piperazin-1-yl)-5-(trifluoromethyl)pyrimidine (75 mg, 0.32 mmol) in DMF (5 mL) was added EDCI (85 mg, 0.44 mmol), HOBt (60 mg, 0.44 mmol) and DIPEA (114 mg, 0.88 mmol). The mixture was stirred ar rt for 16 hours. The reaction was purified by prep-HPLC to afford 6-bromo-4-(6- oxo-6-(4-(5-(trifluoromethyl) pyrimidin-2-yl)piperazin-1-yl)hexan-2-yl)phthalazin-1(2H)-on e as a white solid (80 mg, 49% yield). LCMS ESI m/z: 553 [M+H] + . Step 5 4-(6-Oxo-6-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1 -yl)hexan-2-yl)-6-(prop-1-yn-1- yl)phthalazin-1(2H)-one [1196] A mixture of 6-bromo-4-(6-oxo-6-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pip erazin-1-yl)hexan- 2-yl)phthalazin-1(2H)-one (80 mg, 0.14 mmol), tributyl(prop-1-yn-1-yl)stannane (143 mg, 0.42 mmol) and Pd(dppf)Cl 2 (11 mg, 0.01 mmol) in 1,4-dioxane (3 mL) was stirred at 100 °C under Ar (g) for 3 hours. The reaction mixture was concentrated in vacuo. The residue was purified by silica gel chromatography (MeOH/DCM 0-10%) to afford 4-(6-oxo-6-(4-(5-(trifluoromethyl)pyrimidin-2-yl)piperazin-1 -yl)hexan-2- yl)-6-(prop-1-yn-1-yl)phthalazin-1(2H)-one (27 mg, 36% yield) as a white solid. LCMS ESI m/z: 513.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.56 (s, 1H), 8.72 (s, 2H), 8.20 (d, J = 8.2 Hz, 1H), 8.03 (s, 1H), 7.78 (d, J = 9.2 Hz, 1H), 3.79 (d, J = 19.9 Hz, 4H), 3.55 – 3.50 (m, 4H), 2.37 (t, J = 6.7 Hz, 2H), 2.12 (s, 3H), 2.02 – 1.96 (m, 1H), 1.80 (d, J = 6.5 Hz, 1H), 1.54 (d, J = 3.3 Hz, 3H), 1.22 (d, J = 6.4 Hz, 3H). Synthesis of Example 196: 6-(4-(3-(((7-Cyclopropoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)amino)butanoyl)piperazin-1-yl)nicotinonitrile Step 1 : 6-Cyclopropoxy-4-methylphthalazin-1(2H)-one [1197] A mixture of dimethyl (6-cyclopropoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphon ate (420 mg, 1.41 mmol), paraformaldehyde (423 mg, 14.08 mmol) and K 2 CO 3 (389 mg, 2.84 mmol) in anhydrous THF (5 mL) was stirred at 25 °C under Ar (g) for 15 hours. Hydrazine hydrate (1.25 g, 20.03 mmol) was added and the reaction was stirred at 60 °C for 3 hours. THF was removed under vacuum. The formed solids were collected by filtration, washed with water (10 mL) and petroleum ether/EA (2:1, 10 mL) to give 6-cyclopropoxy-4-methylphthalazin-1(2H)-one as a white solid (270 mg, 83% yield). This was used without further purification. LCMS ESI m/z 203 [M+H] + . Step 2 : 4-(Bromomethyl)-6-cyclopropoxyphthalazin-1(2H)-one [1198] A mixture of 6-cyclopropoxy-4-methylphthalazin-1(2H)-one (225 mg, 1.04 mmol), NBS (445 mg, 2.50 mmol) and AIBN (513 mg, 3.12 mmol) in CCl 4 (8 mL) was stirred at 80 °C for 19 hours. The reaction was quenched with H 2 O (10 mL) and extracted with DCM (20 mL × 3). The combined oragnic layer was dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30 to 50%) to obtain 4-(bromomethyl)-6- cyclopropoxyphthalazin-1(2H)-one as a yellow oil (145 mg, 32% yield). LCMS ESI m/z 295 [M+H] + . Step 3: 6-(4-(3-(((7-Cyclopropoxy-4-oxo-3,4-dihydrophthalazin-1-yl)m ethyl)amino) butanoyl)piperazin-1-yl)nicotinonitrile [1199] A mixture of 4-(bromomethyl)-6-cyclopropoxyphthalazin-1(2H)-one (130 mg, 0.30 mmol), 6- (4-(3-aminobutanoyl)piperazin-1-yl)nicotinonitrile (129 mg, 0.47 mmol) and DIPEA (153 mg, 1.18 mmol) in MeCN (3 mL) was stirred at rt for 16 hours. The mixture was diluted with H2O (5 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was concentrated in vacuo and the residue was purified by prep-HPLC to give 6-(4-(3-(((7-cyclopropoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)amino) butanoyl)piperazin-1-yl)nicotinonitrile as a white solid (15.6 mg, 11% yield). LCMS ESI m/z 488 (M+H) + . 1 H–NMR (400 MHz, DMSO–d 6 ) δ 12.64 (s, 1H), 8.51 (d, J = 2.2 Hz, 1H), 8.21 (d, J = 8.8 Hz, 1H), 7.89 (dd, J = 9.1, 2.3 Hz, 1H), 7.65 – 7.53 (m, 2H), 6.93 (d, J = 9.2 Hz, 1H), 4.36 (d, 2H), 4.15 – 4.07 (m, 1H), 3.79 – 3.52 (m, 10H), 2.72 (d, J = 37.6 Hz, 2H), 1.28 (d, J = 6.1 Hz, 3H), 0.94 – 0.86 (m, 2H), 0.78 – 0.71 (m, 2H). Synthesis of Example 197: Preparation of N-(2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)-2-fluoro- 5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin-1-yl)meth yl)benzamide tert-Butyl (2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)carbamate [1200] A mixture of 6-chloropyridine-3-carbonitrile (810 mg, 5.84 mmol), tert-butyl N-[2- (ethylamino)ethyl]carbamate (1 g, 5.31 mmol) and K 2 CO 3 (1.84 g, 13.28 mmol) in MeCN (10 mL) was stirred at 60 °C for 16 hours. The reaction was concentrated in vacuo, diluted with water (30 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 10 to 20%) to give tert-butyl (2-((5-cyanopyridin-2- yl)(ethyl)amino)ethyl)carbamate as a white solid (1.2 g, 70% yield). LCMS ESI m/z: 291 [M+H] + . Step 2 6-((2-Aminoethyl)(ethyl)amino)nicotinonitrile [1201] A solution of tert-butyl (2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)carbamate (1.2 g, 4.14 mmol) in HCl/dioxane (4 M, 10 mL) was stirred at rt for 1 hour. The solvent was removed in vacuo to give 6-((2-aminoethyl)(ethyl)amino)nicotinonitrile as a white solid (1.12 g, 97% yield). This was used without further purification. LCMS ESI m/z: 191 [M+H] + . Step 3 N-(2-((5-Cyanopyridin-2-yl)(ethyl)amino)ethyl)-2-fluoro-5-(( 4-oxo-7-(prop-1-yn-1-yl)-3,4- dihydrophthalazin-1-yl)methyl)benzamide [1202] To a solution of 2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1- yl)methyl)benzoic acid (100 mg, 0.3 mmol) and 2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4- dihydrophthalazin-1-yl)methyl)benzoic acid (90 mg, 0.3mmol) in DMF (2 mL) was added EDCI (86 mg, 0.45 mmol), HOBt (60 mg, 0.45 mmol) and DIPEA (192 mg, 1.49 mmol). The reaction mixture was stirred at rt for 1 hour. The reaction was purified by prep-HPLC to give N-(2-((5-cyanopyridin-2- yl)(ethyl)amino)ethyl)-2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl) -3,4-dihydrophthalazin-1- yl)methyl)benzamide as a white solid (74 mg, 49%). LCMS ESI m/z : 509 [M+H] + ); 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.65 (s, 1H), 8.46 (d, J = 2.1 Hz, 1H), 8.42 (d, J = 2.5 Hz, 1H), 8.20 (d, J = 8.2 Hz, 1H), 7.92 (s, 1H), 7.84 – 7.75 (m, 2H), 7.53 – 7.44 (m, 2H), 7.23 (dd, J = 10.4, 8.5 Hz, 1H), 6.81 (d, J = 9.1 Hz, 1H), 4.32 (s, 2H), 3.63 (d, J = 5.9 Hz, 2H), 3.55 (d, J = 6.9 Hz, 2H), 3.44 (dd, J = 12.5, 6.3 Hz, 2H), 2.11 (s, 3H), 1.09 (t, J = 7.0 Hz, 3H). Synthesis of Example 198: N-(2-((5-Cyanopyridin-2-yl)(ethyl)amino)ethyl)-2-fluoro-N-me thyl-5-((4- oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin-1-yl)methyl)ben zamide

tert-Butyl (2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)(methyl)carbamat e [1203] To a solution of tert-butyl (2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)carbamate (300 mg, 1.03 mmol) in DMF (5 mL) was added NaH (60%, 41 mg, 1.03 mmol) at 0 °C under N 2 (g) . The mixture was stirred at rt for 1 hour, then CH 3 I (170 mg, 1.20 mmol) was added and stirred at rt for 12 hours. The reaction was quenched with water (10 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by prep-HPLC to give tert-butyl (2-((5-cyanopyridin-2- yl)(ethyl)amino)ethyl)(methyl) carbamate as a white solid (93 mg, 30% yield). LCMS ESI m/z: 247 [M+H] + . Step 2 6-(Ethyl(2-(methylamino)ethyl)amino)nicotinonitrile [1204] To a solution of tert-butyl (2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)(methyl) carbamate (90 mg, 0.30 mmol) in 1,4-dioxane (1 mL) was added HCl/dioxane (4M, 1 mL). The mixture was stirred at rt for 1 hour, then concentrated in vacuo to afford 6-(ethyl(2-(methylamino)ethyl)amino)nicotinonitrile as a white solid (60 mg, 99% yield). LCMS ESI m/z: 205 [M+H] + . Step 3 N-(2-((5-Cyanopyridin-2-yl)(ethyl)amino)ethyl)-2-fluoro-N-me thyl-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4- dihydrophthalazin-1-yl)methyl)benzamide [1205] To a solution of 6-(ethyl(2-(methylamino)ethyl)amino)nicotinonitrile (60 mg, 0.30 mmol) and 2-fluoro-5-((4-oxo-7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1-yl)methyl)benzoic acid (99 mg, 0.30 mmol) in DMF (2 mL) was added EDCI (84 mg, 0.44 mmol), HOBt (60 mg, 0.44 mmol) and DIPEA (114 mg, 0.88 mmol). The reaction mixture was stirred at rt for 2 hours. The reaction was purified by prep-HPLC to afford N-(2-((5-cyanopyridin-2-yl)(ethyl)amino)ethyl)-2-fluoro-N-me thyl-5-((4-oxo-7- (prop-1-yn-1-yl)-3,4-dihydrophthalazin-1-yl)methyl)benzamide as a white solid (48 mg, 31% yield). LCMS ESI m/z 523 (M+H) + ; 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.48 (s, 1H), 8.45 (s, 0.5H), 8.22 – 8.14 (m, 1H), 8.03 (s, 0.5H), 7.89 – 7.84 (m, 1H), 7.82 – 7.77 (m, 0.5H), 7.74 (d, J = 7.9 Hz, 0.5H), 7.69 (d, J = 8.2 Hz, 0.5H), 7.54 (d, J = 8.8 Hz, 0.5H), 7.38 – 7.30 (m, 1H), 7.21 – 7.05 (m, 2H), 6.81 (d, J = 9.1 Hz, 0.5H), 6.29 (d, J = 8.2 Hz, 0.5H), 4.25 (m, 2H), 3.82 – 3.74 (m, 1H), 3.69 – 3.63 (m, 1H), 3.61 – 3.53 (m, 1H), 3.53 – 3.46 (m, 1H), 3.40 – 3.34 (m, 1H), 3.13 – 3.10 (m, 1H), 3.04 (s, 2H), 2.83 (s, 1H), 2.11 – 2.06 (m, 3H), 1.14 (t, J = 7.0 Hz, 1H), 0.90 (t, J = 7.0 Hz, 2H). Synthesis of Example 199: 2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazin-1-y l)methyl)- N-((5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl)methyl)benzamid e

tert-Butyl (2-oxo-2-(2-(2,2,2-trifluoroacetyl)hydrazineyl)ethyl)carbama te [1206] To a solution of tert-butyl (2-hydrazineyl-2-oxoethyl)carbamate (4.8 g, 23.62 mmol) in CH 3 CN (100 mL) was added DIPEA (3.66 g, 28.34 mmol, 4.94 mL), cooled to -45 °C, 2,2,2-trifluoroacetic anhydride (5.46 g, 25.98 mmol, 3.67 mL) was added dropwise. The mixture was warmed to rt and stirred for 16 hours. The reaction mixture was concentrated to give a residue. The residue was dissolved in water (100 mL), extracted with EtOAc (100 mL x 3). The extracts were washed with brine, dried and concentrated to the crude. The crude was purified by silica gel chromatography (petroleum ether/EtOAc = 10/1 to 1/1 ) to give tert-butyl (2-oxo-2-(2-(2,2,2-trifluoroacetyl)hydrazineyl)ethyl)carbama te (6.4 g, 95% yield) as a light yellow gum. 1 H NMR (400 MHz, DMSO-d 6 ) δ 11.45 (s, 1H), 10.24 (s, 1H), 7.10 (t, J = 6.1 Hz, 1H), 3.63 (d, J = 6.2 Hz, 2H), 1.38 (s, 9H). Step 2 tert-Butyl ((5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl)methyl)carbamate [1207] To a solution of tert-butyl N-[2-oxo-2-[2-(2,2,2-trifluoroacetyl)hydrazino]ethyl]carbama te (500 mg, 1.75 mmol) in CH 3 CN (5 mL) was added DIPEA (1.31 g, 10.17 mmol, 1.77 mL) and PPh 3 (1.89 g, 7.19 mmol). The mixture was stirred at 25 °C for 5 minutes then perchloroethane (954.53 mg, 4.03 mmol, 456.49 μL) was added. The reaction mixture was stirred at rt. for another 20 hours. The mixture was concentrated to the residue. The residue was was worked up with water (100 mL) and EtOAc (350 mL). The organic layer was concentrated to the crude. The crude was purified by silica gel chromatography (EtOAc: petroleum ether = 5-15%) to give tert-butyl N-[[5-(trifluoromethyl)-1,3,4- oxadiazol-2-yl]methyl]carbamate (300 mg, 64% yield) as a yellow solid. 1 H- NMR (400 MHz, CDCl 3 ) δ 5.27 (s, 1H), 4.65 (d, J = 5.6 Hz, 2H), 1.46 (s, 9H). Step 3: (5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl)methanamine [1208] To a solution of tert-butyl N-[[5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl]methyl]carbamat e (150 mg, 561.37 μmol) in DCM (2 mL) was added CF 3 COOH (1 mL). The solution was stirred at rt. for 4 hours. The mixture was concentrated to give 5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl]methanamine (160 mg, crude) as a yellow oil. Step 4 2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazin-1-y l)methyl)-N-((5-(trifluoromethyl)- 1,3,4-oxadiazol-2-yl)methyl)benzamide [1209] To a solution of 2-fluoro-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)methyl]b enzoic acid (57.43 mg, 170.75 μmol) in DMF (2 mL) was added DIPEA (294.24 mg, 2.28 mmol, 396.55 μL), EDCI (163.66 mg, 853.75 μmol) and HOBt (115.36 mg, 853.75 μmol). The reaction mixture was stirred at rt for 5 minutes, 5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl]methanamine (160 mg, 569.17 μmol) was added to the mixture. The mixture was stirred at rt for another 16 hours. The mixture was purified by prep-HPLC to give 2-fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazin-1-y l)methyl)-N-((5-(trifluoromethyl)- 1,3,4-oxadiazol-2-yl)methyl)benzamide (3.22 mg, 1.0% yield) as a white solid. LCMS ESI m/z 485.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ12.62 (s, 1H), 9.09 (s, 1H), 8.20 (d, J = 8.2 Hz, 1H), 7.95 (s, 1H), 7.78 (d, J = 8.2 Hz, 1H), 7.62 (dd, J = 6.8, 2.0 Hz, 1H), 7.57 – 7.48 (m, 1H), 7.29 (dd, J = 10.4, 8.6 Hz, 1H), 4.81 (d, J = 5.5 Hz, 2H), 4.35 (s, 2H), 2.11 (s, 3H). Synthesis of Example 200: 2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazin-1-y l)methyl)- N-(1-(5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl)ethyl)benzami de

tert-Butyl 1-hydrazinyl-1-oxopropan-2-ylcarbamate [1210] To a solution of methyl (2S)-2-(tert-butoxycarbonylamino)propanoate (5 g, 24.60 mmol) in Ethanol (140 mL) was added hydrazine hydrate (19.00 g, 379.63 mmol, 18.50 mL), The reaction mixture was stirred at 25 °C for 12 hrs. The reaction mixture was concentrated under reduced pressure. The residue was diluted with H 2 O (30 mL) and extracted with DCM (50 mL X 2). The combined organic layers were washed with brine (30 ml), dried over Na 2 SO 4 and concentrated to afford tert-butyl 1- hydrazinyl-1-oxopropan-2-ylcarbamate (3.7 g, 74.0% yield) as a white solid. 1 H NMR (400MHz, DMSO- d 6 ) δ 8.97 (s, 1H), 6.82 (d, J = 7.6 Hz, 1H), 4.17 (s, 2H), 3.97 – 3.85 (m, 1H), 1.37 (s, 9H), 1.14 (d, J = 7.1 Hz, 3H) Step 2 tert-Butyl 1-oxo-1-(2-(2,2,2-trifluoroacetyl)hydrazinyl)propan-2-ylcarb amate [1211] To a solution of tert-butyl N-[(1S)-2-hydrazino-1-methyl-2-oxo-ethyl]carbamate (3.7 g, 18.21 mmol) and DIPEA (2.82 g, 21.85 mmol, 3.81 mL) in CH 3 CN (80 mL) at -45 °C under N 2 was added TFAA (4.21 g, 20.03 mmol, 2.83 mL) . The reaction was gradually warmed to RT and further stirred for 12 hours. The solvent was removed by evaporation and the residue partitioned between H 2 O (25 mL) and EtOAc (25 mL). The organic phase was separated and the aqueous phase was re-extracted with EtOAc (25 mL). The combined organic phases were washed with brine (30 mL), dried over MgSO4 and evaporated, and the residue was purified by silica gel chromatography to afford tert-butyl 1-oxo-1-(2- (2,2,2-trifluoroacetyl)hydrazinyl)propan-2-ylcarbamate (4 g, 73% yield) as a yellow oil. LCMS ESI m/z: 200.1 [M+H] + . Step 3 tert-butyl 1-(5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl)ethylcarbamate [1212] To a mixture of tert-butyl N-[1-methyl-2-oxo-2-[2-(2,2,2- trifluoroacetyl)hydrazino]ethyl]carbamate (4 g, 13.37 mmol) in CH 3 CN (50 mL) was added PPh 3 (14.37 g, 54.80 mmol) , DIPEA (10.02 g, 77.53 mmol, 13.50 mL). The reaction mixture was stirred at rt for 5 minutes, then 1,1,1,2,2,2-hexachloroethane (7.28 g, 30.74 mmol, 3.48 mL) was added, and stirred at rt for 16 hr , The mixture was concentrated to the crude. The crude was diluted into EtOAc (100 mL), washed with water, brine, dried and concentrated. The residue was purified by silica gel chromatography (0-100% EtOAc in heptanes) to give tert-butyl N-[1-[5-(trifluoromethyl)-1,3,4-oxadiazol-2- yl]ethyl]carbamate (2.1 g, 56% yield) as a light yellow oil. 1 H NMR (400MHz, CDCl 3 ) δ 5.13 (d, J = 14.3 Hz, 2H), 1.64 (d, J = 7.0 Hz, 3H), 1.44 (s, 9H) Step 4 1-(5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl)ethanamine [1213] tert-Butyl N-[1-[5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl]ethyl]carbama te (1 g, 3.56 mmol), HCl/dioxane (5 mL) and 1.4-dioxane (10 mL) were added to a 50 mL bottled flask. The resultant mixture was stirred at rt for 12 hr. The solvent removed in vacuo to give 1-(5-(trifluoromethyl)-1,3,4- oxadiazol-2-yl)ethanamine (1 g, crude) , which was used to next step without further purification. 1 HNMR (400MHz, DMSO-d 6 ) δ 9.18 (s, 2H), 4.95 (d, J = 7.0 Hz, 1H), 1.68 (d, J = 6.9 Hz, 3H). Step 5 2-Fluoro-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophthalazin-1-y l)methyl)-N-(1-(5-(trifluoromethyl)- 1,3,4-oxadiazol-2-yl)ethyl)benzamide [1214] Following the general amide coupling procedure in Example 1, but starting with (S)-1-(5- (trifluoromethyl)-1,3,4-oxadiazol-2-yl)ethanamine gave the title compound as a white solid. LCMS ESI m/z: 499.7 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.61 (s, 1H), 9.09 (d, J = 7.5 Hz, 1H), 8.20 (d, J = 8.2 Hz, 1H), 7.94 (s, 1H), 7.77 (dd, J = 8.2, 1.3 Hz, 1H), 7.56 (dd, J = 6.8, 2.3 Hz, 1H), 7.51 – 7.46 (m, 1H), 7.26 (dd, J = 10.2, 8.6 Hz, 1H), 5.47 – 5.41 (m, 1H), 4.34 (s, 2H), 2.11 (s, 3H), 1.62 (d, J = 7.0 Hz, 3H). Synthesis of Example 201: N-(1-(5-Cyanopyridin-2-yl)azetidin-3-yl)-2-fluoro-N-methyl-5 -((4-oxo-7- (prop-1-yn-1-yl)-3,4-dihydrophthalazin-1-yl)methyl)benzamide tert-Butyl (1-(5-cyanopyridin-2-yl)azetidin-3-yl)(methyl)carbamate [1215] A mixture of 6-chloropyridine-3-carbonitrile (372 mg, 2.68 mmol), tert-butyl N-(azetidin-3-yl)- N-methyl-carbamate (500 mg, 2.68 mmol) and K 2 CO 3 (1.11 g, 8.05 mmol) in MeCN (20 mL) was stirred at 60 °C under Ar (g) for 16 hours. The mixture was cooled to rt, diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give tert-butyl (1-(5-cyanopyridin-2-yl)azetidin-3- yl)(methyl)carbamate as a white solid (0.5 g, 65% yield). This was used without further purification. LCMS ESI m/z 289 [M+H] + . Step 2 6-(3-(Methylamino)azetidin-1-yl)nicotinonitrile [1216] A mixture of tert-butyl N-[1-(5-cyano-2-pyridyl)azetidin-3-yl]-N-methyl-carbamate (500 mg, 1.73 mmol) in DCM (5 mL) and TFA (1 mL) was stirred at 25 °C for 1 hour. The reaction was concentrated in vacuo to give 6-(3-(methylamino)azetidin-1-yl)nicotinonitrile as a brown oil (305 mg, 92% yield). This was used without further purification. LCMS ESI m/z 189 [M+H] + . Step 3 N-(1-(5-Cyanopyridin-2-yl)azetidin-3-yl)-2-fluoro-N-methyl-5 -((4-oxo-7-(prop-1-yn-1-yl)-3,4- dihydrophthalazin-1-yl)methyl)benzamide [1217] To a solution of 2-fluoro-5-[(4-oxo-7-prop-1-ynyl-3H-phthalazin-1-yl)methyl]b enzoic acid (50 mg, 0.15 mmol) and 6-[3-(methylamino)azetidin-1-yl]pyridine-3-carbonitrile (37 mg, 0.16 mmol) in DMF (2 mL) was added HOBt (30 mg, 0.23 mmol), EDCI (43 mg, 0.23 mmol) and DIPEA (58 mg, 0.45 mmol). The reaction mixture was stirred at rt for 2 hours and purified by prep-HPLC to give N-(1-(5- cyanopyridin-2-yl)azetidin-3-yl)-2-fluoro-N-methyl-5-((4-oxo -7-(prop-1-yn-1-yl)-3,4-dihydrophthalazin- 1-yl)methyl)benzamide as a white solid (38 mg, 50% yield). Yield: 50% LCMS ESI m/z 507 (M+H) + ; 1 H NMR (400 MHz, DMSO–d 6 ) δ 12.48 (s, 1H), 8.43 (s, 1H), 8.20 (d, J = 8.2 Hz, 1H), 7.90 (s, 1H), 7.80 (d, J = 6.7 Hz, 1H), 7.74 (dd, J = 8.2, 1.4 Hz, 1H), 7.44 – 7.26 (m, 2H), 7.22 (t, J = 9.1 Hz, 1H), 6.43 (s, 1H), 5.28 (s, 0.5H), 4.56 (s, 0.5H), 4.32 (s, 3H), 4.22 – 4.16 (m, 2H), 4.15 – 4.05 (m, 1H), 2.90 (s, 3H), 2.08 (s, 3H). Synthesis of Example 202: 2-Fluoro-N-methyl-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophtha lazin-1- yl)methyl)-N-((5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl)meth yl)benzamide

tert-Butyl 2-oxo-2-(2-(2,2,2-trifluoroacetyl)hydrazinyl)ethylcarbamate [1218] To a solution of tert-butyl N-(2-hydrazino-2-oxo-ethyl)carbamate (5.00 g, 26.4 mmol) in CH3CN (100 mL) was added DIPEA (4.10 g, 31.7 mmol, 5.52 mL), and then cooled to -45 °C, then trifluoroacetic anhydride (6.11 g, 29.1 mmol, 4.11 mL) was added dropwise, after the addition, the mixture warmed to rt. slowly, and then stirred at rt. for 16 hrs. The reaction mixture was concentrated. The residue was diluted in H 2 O (100 mL), extracted with EA (100 mL x 3), washed with brine, dried, concentrated and purified by silica gel chromatography to give tert-butyl N-[2-oxo-2-[2-(2,2,2- trifluoroacetyl)hydrazino]ethyl]carbamate (5.5 g, 73% yield) as a colorless oil. 1 H NMR (400 MHz, DMSO-d 6 ) δ 11.45 (s, 1H), 10.23 (s, 1H), 7.10 (t, J = 6.1 Hz, 1H), 3.62 (t, J = 9.7 Hz, 2H), 1.38 (s, 9H). Step 2 tert-Butyl (5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl)methylcarbamate [1219] To a solution of tert-butyl N-[2-oxo-2-[2-(2,2,2-trifluoroacetyl)hydrazino]ethyl]carbama te (5.50 g, 19.3 mmol) in CH 3 CN (100 mL) was added DIPEA (14.5 g, 112 mmol, 19.5 mL) and PPh 3 (20.7 g, 79.1 mmol), and stirred at rt. for 5 minutes, and then perchloroethane (10.5 g, 44.4 mmol, 5.02 mL) was added, and stirred at rt. for 20 hrs. The mixture was concentrated and 50 mL H 2 O was added, extracted with EtOAc (100 mL x 3), washed with brine, dried, concentrated and purified by silica gel chromatography (EtOAc: petroleum ether = 1: 10) to give tert-butyl N-[[5-(trifluoromethyl)-1,3,4- oxadiazol-2-yl]methyl]carbamate (3.5 g, 68% yield) as a yellow solid. 1 H NMR (400 MHz, CDCl 3 ) δ 5.20 (s, 1H), 4.65 (d, J = 5.6 Hz, 2H), 1.46 (s, 9H). Step 3 (5-(Trifluoromethyl)-1,3,4-oxadiazol-2-yl)methanamine [1220] To a solu tion of tert-butyl N-[[5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl]methyl]carbamat e (220 mg, 823μmol) in DCM (2 mL) was added CF 3 COOH (1 mL), and then stirred at rt. for 4 hrs. The reaction mixture was concentrated to give [5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl]methanamine (240 mg, crude) as a light yellow solid. Step 4 N-Methyl-1-(5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl)methana mine [1221] A mixture of [5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl]methanamine (150 mg, 898 μmol) and formaldehyde (72.9 mg, 898 μmol, 67 μL) in DCE (9.93 mL) was stirred at 25°C for 1 hr, sodium triacetoxyborahydride (285 mg, 1.35 mmol) was added to the reaction mixture. Then the reaction mixture was stirred for another 2 hrs. The reaction mixture was acidified with NaHCO 3 (aq.), extracted with EtOAc, the organic layer was concentrated to give N-methyl-1-[5-(trifluoromethyl)-1,3,4- oxadiazol-2-yl]methanamine (100 mg, crude), the crude was used to the next step without further purification. LCMS ESI m/z: 181.9 [M + H] + . Step 5 2-Fluoro-N-methyl-5-((4-oxo-7-(prop-1-ynyl)-3,4-dihydrophtha lazin-1-yl)methyl)-N-((5- (trifluoromethyl)-1,3,4-oxadiazol-2-yl)methyl)benzamide [1222] Following the general amide coupling procedure in step 2 in Example 1, but starting with N- methyl-1-(5-(trifluoromethyl)-1,3,4-oxadiazol-2-yl)methanami ne gave the title compound as a white solid. LCMS ESI m/z: 500.1 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.61 (s, 1H), 8.19 (d, J = 8.2 Hz, 1H), 7.92-7.91 (m, 1H), 7.76 (dd, J = 8.2, 1.3 Hz, 1H), 7.44-7.36 (m, 2H), 7.29-7.22 (m, 1H), 5.06 (s, 2H), 4.35 (s, 2H), 2.97 (s, 3H), 2.10 (s, 3H). Synthesis of Example 203: 5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-N -(1- (cyclopropanecarbonyl)azetidin-3-yl)-2-fluorobenzamide tert-Butyl (1-(cyclopropanecarbonyl)azetidin-3-yl)carbamate [1223] A mixture of cyclopropanecarboxylic acid (300 mg, 3.48 mmol) and tert-butyl azetidin-3- ylcarbamate (600 mg, 3.48 mmol) in DMF (10 mL) was added EDCI (1 g, 5.23 mmol), HOBt (706 mg, 5.23 mmol) and DIPEA (1.35 g, 10.45 mmol). The resulting reaction mixture was stirred at 50 o C for 1 hour, then the reaction was quenched with water (10 mL) and extracted with DCM (30 mL). The organic phase was dried over Na 2 SO 4 , filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (petroleum ether/EtOAc= 2/1) to give 6-(4-(3-formylbenzoyl)piperazin-1- yl)nicotinonitrile (560 mg, 67% yield) as a white solid. LCMS ESI m/z: 241.1 [M+H] + . Step 2 (3-Aminoazetidin-1-yl)(cyclopropyl)methanone [1224] To a solution of tert-butyl (1-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methy l)-2- fluorobenzoyl)azetidin-3-yl)(methyl)carbamate (560 mg, 2.33 mmol) in DCM (10 mL) was added TFA (1 mL) and stirred at 25 °C for 2 hours. The reaction mixture was concentrated in vacuo to afford (3- aminoazetidin-1-yl)(cyclopropyl)methanone (326 mg, 99% yield) as a colorless oil. This was used without further purification. LCMS ESI m/z: 141.1 [M+H] + . Step 3 5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-N -(1-(cyclopropanecarbonyl)azetidin- 3-yl)-2-fluorobenzamide [1225] To a solution of (3-aminoazetidin-1-yl)(cyclopropyl)methanone (30 mg, 0.21 mmol) and 5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoic acid (79 mg, 0.21 mmol) in DMF (3 mL) was added EDCI (62 mg, 0.32 mmol), HOBt (43 mg, 0.32 mmol) and DIPEA (83 mg, 0.64 mmol). The reaction mixture was stirred at rt for 16 hours. The mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (10 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by prep-HPLC to give 5- ((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-N-( 1-(cyclopropanecarbonyl) azetidin-3-yl)-2- fluorobenzamide (88.7 mg, 68% yield) as a white solid. LCMS ESI m/z: 491.2 [M+H] + . 1 H–NMR (400 MHz, DMSO-d 6 ) δ 12.46 (s, 1H), 8.98 (d, J = 6.7 Hz, 1H), 8.14 (d, J = 8.8 Hz, 1H), 7.63 (dd, J = 6.7, 2.0 Hz, 1H), 7.53 – 7.40 (m, 1H), 7.34 – 7.19 (m, 2H), 7.11 (d, J = 2.2 Hz, 1H), 4.84 (p, J = 7.0 Hz, 1H), 4.74 – 4.62 (m, 1H), 4.53 (t, J = 8.3 Hz, 1H), 4.30 (s, 2H), 4.12 (dd, J = 15.7, 7.2 Hz, 2H), 3.80 (dd, J = 9.7, 5.3 Hz, 1H), 2.39 (dt, J = 9.1, 8.1 Hz, 2H), 2.08 – 1.90 (m, 2H), 1.76 (dt, J = 24.5, 12.4 Hz, 1H), 1.63 (dt, J = 18.6, 9.4 Hz, 1H), 1.56 – 1.47 (m, 1H), 0.69 (d, J = 7.7 Hz, 4H). Synthesis of Example 204: N-(1-(5-Cyanopyridin-2-yl)azetidin-3-yl)-5-((7-cyclobutoxy-4 -oxo-3,4- dihydrophthalazin-1-yl)methyl)-2-fluorobenzamide

tert-Butyl (1-(5-cyanopyridin-2-yl)azetidin-3-yl)carbamate [1226] A mixture of 6-chloronicotinonitrile (241 mg, 1.74 mmol), tert-butyl azetidin-3-ylcarbamate (315 mg, 1.83 mmol) and DIPEA (675 mg, 5.23 mmol) in DMF (6 mL) was stirred at 60 °C for 12 hours. The reaction was diluted with water (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to afford tert-butyl (1-(5-cyanopyridin-2-yl)azetidin-3-yl)carbamate (477 mg, 99% yield) as a off-white solid. LCMS ESI m/z: 275.1 [M+H] + . Step 2 6-(3-Aminoazetidin-1-yl)nicotinonitrile [1227] To a solution of tert-butyl (1-(5-cyanopyridin-2-yl)azetidin-3-yl)carbamate (477 mg, 1.74 mmol) in DCM (10 mL) was added TFA (991 mg, 8.70 mmol). The mixture was stirred at rt for 4 hours. Solvent was removed in vacuo to afford 6-(3-aminoazetidin-1-yl)nicotinonitrile (303 mg, 85% yield) as a white solid. LCMS ESI m/z: 175.1 [M+H] + . Step 3 5-N-(1-(5-Cyanopyridin-2-yl)azetidin-3-yl)-5-((7-cyclobutoxy -4-oxo-3,4-dihydrophthalazin-1- yl)methyl)-2-fluorobenzamide [1228] To a solution of 6-(3-aminoazetidin-1-yl)nicotinonitrile (40 mg, 0.23 mmol) and 5-((7- cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2-fluor obenzoic acid (84 mg, 0.23 mmol) in DMF (6 mL) was added EDCI (66 mg, 0.34 mmol), HOBt (46 mg, 0.34 mmol) and DIPEA (118 mg, 0.92 mmol). The reaction mixture was stirred at rt for 4 hours. The reaction was purified by prep-HPLC to give N-(1-(5-cyanopyridin-2-yl)azetidin-3-yl)-5-((7-cyclobutoxy-4 -oxo-3,4-dihydrophthalazin-1-yl)methyl)-2- fluorobenzamide (77 mg, 63% yield) as a off-white solid. LCMS ESI m/z: 525.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) δ 12.46 (s, 1H), 9.02 (d, J = 6.9 Hz, 1H), 8.47 (d, J = 1.9 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.84 (dd, J = 8.8, 2.2 Hz, 1H), 7.66 – 7.61 (m, 1H), 7.50 – 7.44 (m, 1H), 7.32 – 7.22 (m, 2H), 7.11 (d, J = 2.1 Hz, 1H), 6.48 (d, J = 8.8 Hz, 1H), 4.89 – 4.79 (m, 2H), 4.38 (t, J = 8.5 Hz, 2H), 4.30 (s, 2H), 4.00 (dd, J = 9.3, 5.4 Hz, 2H), 2.44 – 2.35 (m, 2H), 2.04 – 1.95 (m, 2H), 1.83 – 1.60 (m, 2H). Synthesis of Example 205: 5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluoro-N- (1-(5-(trifluoromethyl)pyrimidin-2-yl)azetidin-3-yl)benzamid e

tert-Butyl (1-(5-(trifluoromethyl)pyrimidin-2-yl)azetidin-3-yl)carbamat e [1229] A mixture of 2-chloro-5-(trifluoromethyl)pyrimidine (183 mg, 1.74 mmol), tert-butyl azetidin- 3-ylcarbamate (300 mg, 1.74 mmol) and DIPEA (675 mg, 5.23 mmol) in DMF (10 mL) was stirred at rt for 2 hours. The reaction was diluted with water (20 mL) and extracted with EtOAc (20 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to afford tert-butyl (1-(5-(trifluoromethyl)pyrimidin-2-yl)azetidin-3-yl)carbamat e (545 mg, 99% yield) as a white solid. LCMS ESI m/z: 319.1 [M+H] + . Step 2 1-(5-(Trifluoromethyl)pyrimidin-2-yl)azetidin-3-amine [1230] To a solution of tert-butyl (1-(5-(trifluoromethyl)pyrimidin-2-yl)azetidin-3-yl)carbamat e (545 mg, 1.71 mmol) in DCM (10 mL) was added TFA (991 mg, 8.70mmol). The mixture was stirred at rt for 4 hours. The solvent was removed in vacuo to afford 1-(5-(trifluoromethyl)pyrimidin-2-yl)azetidin-3- amine (350 mg, 94% yield) as a white solid. LCMS ESI m/z: 219.1 [M+H] + . Step 3 5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluoro-N-(1-(5- (trifluoromethyl)pyrimidin-2-yl)azetidin-3-yl)benzamide [1231] To a solution of 1-(5-(trifluoromethyl)pyrimidin-2-yl)azetidin-3-amine (50 mg, 0.23 mmol) and 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluorobenzoic acid (84 mg, 0.23 mmol) in DMF (4 mL) was added EDCI (66 mg, 0.34 mmol), HOBt (46 mg, 0.34 mmol) and DIPEA (118 mg, 0.92 mmol). The reaction mixture was stirred at rt for 4 hours. The reaction was purified by prep-HPLC to give 5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl)-2 -fluoro-N-(1-(5- (trifluoromethyl)pyrimidin-2-yl)azetidin-3-yl)benzamide (56 mg, 43% yield) as a white solid. LCMS ESI m/z: 569.2 [M+H] + . 1 H–NMR (400 MHz, DMSO-d 6 ) δ 12.46 (s, 1H), 9.02 (d, J = 6.9 Hz, 1H), 8.70 (s, 2H), 8.15 (d, J = 8.8 Hz, 1H), 7.69 – 7.63 (m, 1H), 7.46 (s, 1H), 7.32 – 7.20 (m, 2H), 7.12 (d, J = 2.2 Hz, 1H), 4.89 – 4.78 (m, 2H), 4.44 (t, J = 8.7 Hz, 2H), 4.30 (s, 2H), 4.12 – 4.05 (m, 2H), 2.45 – 2.36 (m, 2H), 2.06 – 1.95 (m, 2H), 1.82 – 1.59 (m, 2H). Synthesis of Example 206: 6-Cyclobutoxy-4-((3-(4-(cyclopropanecarbonyl)piperazin-1- yl)benzo[d]isoxazol-7-yl)methyl)phthalazin-1(2H)-one 6-Cyclobutoxy-4-((3-(4-(cyclopropanecarbonyl)piperazin-1-yl) benzo[d]isoxazol-7- yl)methyl)phthalazin-1(2H)-one [1232] To a solution of 6-cyclobutoxy-4-((3-(piperazin-1-yl)benzo[d]isoxazol-7-yl)me thyl)phthalazin- 1(2H)-one (108 mg, 0.23 mmol) and cyclopropanecarboxylic acid (20 mg, 0.23 mmol) in DMF (1.5 mL) was added EDCI (66 mg, 0.35 mmol), HOBt (47 mg, 0.35 mmol) and DIPEA (149 mg, 1.15 mmol). The reaction mixture was stirred at 45℃ for 1 hour, then purified by prep-HPLC to afford 6-cyclobutoxy-4- ((3-(4-(cyclopropanecarbonyl)piperazin-1-yl)benzo[d]isoxazol -7-yl)methyl)phthalazin-1(2H)-one (40 mg, 35% yield) as a off-white solid. LCMS ESI m/z: 500.1 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.43 (s, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.89 (d, J = 8.0 Hz, 1H), 7.42 (d, J = 7.2 Hz, 1H), 7.32 – 7.22 (m, 2H), 7.12 (d, J = 2.1 Hz, 1H), 4.75 – 4.66 (m, 1H), 4.50 (s, 2H), 3.88 (s, 2H), 3.67 (s, 2H), 3.56 – 3.42 (m, 4H), 2.37 – 2.28 (m, 2H), 2.06 – 1.92 (m, 3H), 1.83 – 1.57 (m, 2H), 0.81 – 0.69 (m, 4H). Synthesis of Example 207: 6-Cyclobutoxy-4-((3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pi perazin-1- yl)benzo[d]isoxazol-7-yl)methyl)phthalazin-1(2H)-one 6-Cyclobutoxy-4-((3-(4-(5-(trifluoromethyl)pyrimidin 2 yl)piperazin-1-yl)benzo[d] isoxazol-7- yl)methyl)phthalazin-1(2H)-one [1233] To a solution of 6-cyclobutoxy-4-((3-(piperazin-1-yl)benzo[d]isoxazol-7-yl)me thyl)phthalazin- 1(2H)-one (100 mg, 0.23 mmol) and 2-chloro-5-(trifluoromethyl) pyrimidine (46 mg, 0.25 mmol) in DMF (2 mL) was added DIPEA (89 mg, 0.69 mmol). The reaction mixture was stirred at 60 °C for 1 hour. The reaction mixture was purified by prep-HPLC to give 6-(4-(7-((7-cyclobutoxy-4-oxo-3,4- dihydrophthalazin-1-yl)methyl)benzo[d]isoxazol-3-yl)piperazi n-1-yl)nicotinonitrile (84 mg, 63% yield) as a white solid. LCMS ESI m/z: 578 (M+H) + . 1 H–NMR (400 MHz, DMSO-d 6 ) δ 12.43 (s, 1H), 8.77 (s, 2H), 8.15 (d, J = 8.8 Hz, 1H), 7.93 (d, J = 8 Hz, 1H), 7.43 (d, J = 7.2 Hz, 1H), 7.32 – 7.25 (m, 2H), 7.13 (s, 1H), 4.73 (t, J = 7.2 Hz, 1H), 4.51 (s, 2H), 4.07 – 4.04 (m, 4H), 3.62 – 3.58 (m, 4H), 2.34 – 2.31 (m, 2H), 2.01 – 1.61 (m, 4H). Synthesis of Example 208: 6-(4-(7-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzo[d]isoxazol-3-yl)piperazin-1-yl)nicotinonitri le

7-bromobenzo[d]isoxazol-3(2H)-one [1234] To a solution of N-hydroxyacetamide (5 g, 66.7 mmol) in DMF (50 mL) was added t-BuONa (7.47 g, 66.7 mmol) and stirred at rt for 30 min. Then ethyl 3-bromo-2-fluorobenzoate (7.73 g, 33.34 mmol) in DMF (20 mL) was added and the reaction was stirred at 90°C for 4 hours. The mixture was cooled to rt, diluted with H 2 O (50 mL) and extracted with EtOAc (30 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30 to 50%) to give 7- bromobenzo[d]isoxazol-3(2H)-one (2.55 g, 36% yield) as a white solid. LCMS ESI m/z: 214 [M+H] + . Step 2 7-Bromo-3-chlorobenzo[d]isoxazole [1235] To a mixture of 7-bromobenzo[d]isoxazol-3(2H)-one (2.55 g, 11.92 mmol) in phosphoryl trichloride (20 mL) was added Et 3 N (2.41 g, 23.84 mmol). The mixture was stirred at 130 °C for 12 hours. The reaction was carefully diluted with H 2 O (50 mL) and extrated with EtOAc (3 x 30 mL). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to give 7-bromo-3-chlorobenzo[d]isoxazole (1.95 g, 71% yield) as a yellow solid. LCMS ESI m/z: 232 [M+H] + . Step 3 tert-butyl 4-(7-bromobenzo[d]isoxazol-3-yl)piperazine-1-carboxylate [1236] A solution of 7-bromo-3-chlorobenzo[d]isoxazole (1.95 g, 8.41 mmol), tert-butyl piperazine-1- carboxylate (4.69 g, 25.23 mmol) and 2,3,4,6,7,8,9,10-octahydropyrimido[1,2-a]azepine (1.92 g, 12.61 mmol) in pyridine (20 mL) was stirred at 110 °C for 12 hours. The reaction was cooled to rt, diluted with water (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30 to 50 %) to give tert-butyl 4-(7-bromobenzo[d]isoxazol-3- yl)piperazine-1-carboxylate (1.88 g, 59% yield) as a white solid. LCMS ESI m/z: 382 [M+H] + . Step 4 tert-Butyl 4-(7-vinylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate [1237] A mixture of tert-butyl 4-(7-bromobenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (1.88 g, 4.92 mmol), potassium vinyltrifluoroborate (0.99 g, 7.38 mmol), Cs 2 CO 3 (4.01 g, 12.3 mmol) and Pd(dppf)Cl 2 (366 mg, 0.5 mmol) in dioxane (20 mL) was stirred at 80 °C under Ar (g) for 2 hours. The mixture was cooled to rt, diluted with H 2 O (20 mL) and extracted with EtOAc (20 mL × 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromategraphy (EtOAc/petroleum ether = 10 to 20%) to give tert- butyl 4-(7-vinylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (1.26 g, 78% yield) as a white solid. LCMS ESI m/z: 330 [M+H] + . Step 5 tert-Butyl 4-(7-formylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate [1238] To a mixture of tert-butyl 4-(7-vinylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (1.10 g, 3.33 mmol) in THF (40 mL) and H 2 O (25 mL) was added K 2 OsO 4 -2H 2 O (33 mg, 0.09 mmol). The mixture was stirred at room temperature for 5 min, then NaIO 4 (2.20 g, 10.19 mmol) was added and the mixture was stirred at rt for 2 hours. The solids were removed by filtration. The filtrate was diluted with H 2 O (10 mL) and extracted with EtOAc (30 mL x 3). The combined organic layer was washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to give tert-butyl 4-(7- formylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (0.89 g, 81% yield). This was used without further purification. LCMS ESI m/z: 332 [M+H] + . Step 6 tert-Butyl 4-(6-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )benzo [d]isoxazol-3- yl)piperazine-1-carboxylate [1239] A solution of tert-butyl 4-(7-formylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (450 mg, 1.36 mmol), dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (636 mg, 2.04 mmol) and Et 3 N (412 mg, 4.07 mmol) in THF (5 mL) was stirred at 60 °C under Ar (g) for 12 hours. Hydrazine hydrate (102 mg, 2.04 mmol) was added and the reaction was heated to 70 ℃ for 2 hours. The reaction was cooled to rt, and the solvent was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/EtOAc (2:1, 10 mL) to give tert-butyl 4-(7-((7-cyclobutoxy-4- oxo-3,4-dihydrophthalazin-1-yl)methyl)benzo[d]isoxazol-3-yl) piperazine-1-carboxylate (440 mg, 61% yield) as a white solid. LCMS ESI m/z: 532 [M+H] + . Step 7 6-Cyclobutoxy-4-((3-(piperazin-1-yl)benzo[d]isoxazol-7-yl)me thyl)phthalazin-1(2H)-one [1240] A solution of tert-butyl 4-(7-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzo[d]isoxazol-3-yl)piperazine-1-carboxylate (440 mg, 0.83 mmol) in HCl/dioxane (4 M, 5 mL) was stirred at rt for 1 hour. The solvent was removed in vacuo to give 6-cyclobutoxy-4-((3- (piperazin-1-yl)benzo[d]isoxazol-7-yl)methyl)phthalazin-1(2H )-one (350 mg, 98% yield) as a white solid. This was used without further purification. LCMS ESI 432 [M+H] + ). Step 8 6-(4-(7-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)benzo[d]isoxazol-3-yl)piperazin- 1-yl)nicotinonitrile [1241] To a solution of 6-cyclobutoxy-4-((3-(piperazin-1-yl)benzo[d]isoxazol-7-yl)me thyl)phthalazin- 1(2H)-one (100 mg, 0.23 mmol) and 6-chloronicotinonitrile (35 mg, 0.25 mmol) in DMF (2 mL) was added DIPEA (89 mg, 0.69 mmol). The reaction mixture was stirred at 60 °C for 6 hours. The mixture was purified by prep-HPLC to give 6-(4-(7-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzo[d]isoxazol-3-yl)piperazin-1-yl)nicotinonitri le (38 mg, 31% yield) as a white solid. LCMS ESI m/z: 534 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.43 (s, 1H), 8.53 (d, J = 1.6 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.93 – 7.89 (m, 2H), 7.42 (d, J = 7.2 Hz, 1H), 7.31 – 7.24 (m, 2H), 7.12 (d, J = 1.2 Hz, 1H), 7.01 (d, J = 9.2 Hz, 1H), 4.73 (t, J = 7.2 Hz, 1H), 4.50 (s, 2H), 3.90 – 3.87 (m, 4H), 3.61 – 3.58 (m, 4H), 2.36 – 2.30 (m, 2H), 2.00 – 1.61 (m, 4H). Synthesis of Example 209: 6-Cyclobutoxy-4-((3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pi perazin-1- yl)be nzo[d]isoxazol-5-yl)methyl)phthalazin-1(2H)-on 5-Bromobenzo[d]isoxazol-3(2H)-one [1242] To a solution of N-hydroxyacetamide (3.40 g, 45.30 mmol) in DMF (20 mL) was addded t- BuONa (8.70 g, 90.60 mmol) under Ar (g). The reaction was stirred at rt for 40 min, then added methyl 5- bromo-2-fluorobenzoate (5.20 g, 22 mmol). The reaction was stirred at 90 °C for 40 min, then cooled to rt and quenched with sat. NH 4 Cl soltion (10 mL). The mixture was extracted with EtOAc (30 × 3 mL). The combined organic layer was washed with bine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 20% to 40%) to give 5- bromobenzo[d]isoxazol-3(2H)-one (2 g 42% yield) as a white solid. LCMS ESI 214 [M+H] + . Step 2 6-Bromo-3-chlorobenzo[d]isoxazole [1243] To a solution of 5-bromobenzo[d]isoxazol-3(2H)-one (2.00 g, 9.38 mmol) in pyridine (1.66 g , 21 mmol) was added POCl 3 (6.45 g , 42 mmol) and Et 3 N (1.51 g, 14.95 mmol). The mixture was stirred at 110 °C for 12 hours. The reaction was carefully added to water (10 mL), then extrated with EtOAc (3 x 30 mL). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to give 5-bromo-3-chlorobenzo[d]isoxazole (1.56 g 72% yield) as a white solid. LCMS ESI 232 [M+H] + . This was used without further purification. Step 3 tert-Butyl 4-(5-bromobenzo[d]isoxazol-3-yl)piperazine-1-carboxylate [1244] A mixture of 5-bromo-3-chlorobenzo[d]isoxazole (1.56 g, 6.80 mmol), tert-butyl piperazine-1- carboxylate (1.25 g, 6.70 mmol) and 2,3,4,6,7,8,9,10-octahydropyri mido[1,2 -a]azepine (786 mg, 5.16 mmol) in pyridine (20 mL) was stirred at 110 °C for 12 hours. The reaction was diluted with water (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography (EtOAc/petroleum ether = 30% to 50%) to give tert-butyl 4-(5-bromobenzo[d]isoxazol-3-yl)piperazine-1- carboxylate (1.20 g 78% yield) as a white solid. LCMS ESI 382 [M+H] + . Step 4 tert-Butyl 4-(5-vinylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate [1245] A mixture of tert-butyl 4-(5-bromobenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (2.00 g, 5.20 mmol), potassium vinyltrifluoroborate (505 mg,3.77 mmol), Pd(dppf)Cl2 (230 mg, 0.31 mmol) and K2CO3 (1.53 g, 11.10 mmol) in dioxane (10 mL) was stirred at 80 ℃ for 5 hours. The reaction mixture was diluted with H2O (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layer was washed with brine, dried over Na 2 SO 4 and filtered. The solvent was removed in vacuo and the residue was purified by silica gel chromatography (EtOAc/petroleum ether = 10% to 30%) to give tert-butyl 4-(5- vinylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (850 mg, 49% yield) as a white solid. LCMS ESI 330 [M+H] + . Step 5 tert-Butyl 4-(5-formylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate [1246] To a mixture of tert-butyl 4-(5-vinylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (850 mg, 2.6 mmol) in THF (36 mL) and H 2 O (24 mL) was added K 2 OsO 4 -2H 2 O (27 mg, 0.10 mmol). The mixture was stirred at rt for 5 min, then added NaIO 4 (1.8 g, 4.80 mmol) and stirred at rt for 4 hours. The reaction was diluted with H 2 O (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layer was washed with brine, dried over Na 2 SO 4 and filtered. The solvent was removed in vacuo to give tert-butyl 4- (5-formylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (835 mg 97% yield) as a white solid. This was used without further purification. LCMS ESI 330 [M+H] + . Step 6 tert-Butyl 4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )benzo[d]is oxazol-3- yl)piperazine-1-carboxylate [1247] A solution of tert-butyl 4-(5-formylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (420 mg, 1.27 mmol), dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosp honate (396 mg, 1.27 mmol) and Et 3 N (385 mg, 3.80 mmol) in THF (5 mL) was stirred at 50 °C under Ar (g) for 15 hours. Hydrazine hydrate (102 mg, 2.04 mmol) was added and the reaction was stirred at 70 ℃ for 1 hour. The reaction was cooled to rt, and the solvent was removed under vacuum. The formed solids were filterated, washed with water (10 mL) and petroleum ether/EA (2:1, 10 mL) to give tert-butyl 4-(5-((7-cyclobutoxy- 4-oxo-3,4-dihydrophthalazin-1-yl)methyl)benzo[d]isoxazol-3-y l)piperazine-1-carboxylate (490 mg 80% yield) as a white solid. LCMS ESI 532 [M+H] + . Step 7 6-cyclobutoxy-4-((3-(piperazin-1-yl)benzo[d]isoxazol-5-yl)me thyl)phth alazin-1(2H)-one [1248] A solution of tert-butyl 4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzo[d]isoxazol-3-yl)piperazine-1-carboxylate (490 mg, 0.94 mmol) in HCl/dioxane (4M, 5 mL) was stirred at rt for 1 hour. The solvent was removed in vacuo to give 6-cyclobutoxy-4-((3- (piperazin-1-yl)benzo[d]isoxazol-5-yl)methyl)phthalazin-1(2H )-one (395 mg 99% yield) as a white solid. This was used without further purification. LCMS ESI 432[M+H] + . Step 8 6-Cyclobutoxy-4-((3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pi perazin-1-yl)be nzo[d]isoxazol-5- yl)methyl)phthalazin-1(2H)-one [1249] To a solution of 6-cyclobutoxy-4-((3-(piperazin-1-yl)benzo[d]isoxazol-5-yl)me thyl)phthalazin- 1(2H)-one (120 mg, 0.24 mmol) and 2-chloro-5-(trifluoromethyl) pyrimidine (50 mg, 0.28 mmol) in DMF (2 mL) was added DIPEA (112 mg, 0.85 mmol). The reaction mixture was stirred at 60 °C for 2 hours. The mixture was purified by prep-HPLC to give 6-cyclobutoxy-4-((3-(4-(5-(trifluoromethyl)pyrimidin-2- yl)piperazin-1-yl)benzo[d]isoxazol-5-yl)methyl)phthalazin-1( 2H)-one (77 mg 60% yield) as a white solid. LCMS ESI 578 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.44 (s, 1H), 8.77 (s, 2H), 8.27 – 8.06 (m, 2H), 7.57 – 7.42 (m, 2H), 7.33 – 7.15 (m, 2H), 4.85 (p, J = 7.1 Hz, 1H), 4.42 (s, 2H), 4.14 – 3.99 (m, 4H), 3.68 – 3.50 (m, 4H), 2.44 – 2.29 (m, 2H), 2.08 – 1.91 (m, 2H), 1.84 – 1.62 (m, 2H). Synthesis of Example 210: 6-cyclobutoxy-4-((3-(4-(cyclopropanecarbonyl)piperazin-1- yl)benzo[d]isoxazol-5-yl)methyl)phthalazin-1(2H)-one Step 1 6-Cyclobutoxy-4-((3-(4-(cyclopropanecarbonyl)piperazin-1-yl) benzo[d]isoxazol-5- yl)methyl)phthalazin-1(2H)-one [1250] To a solution of cyclopropanecarboxylic acid (24 mg, 0.28 mmol) and 6-cyclobutoxy-4-((3- (piperazin-1-yl)benzo[d]isoxazol-5-yl)methyl)phthalazin-1(2H )-one (120 mg, 0.26 mmol) in DMF (2 mL) was added EDCI (74 mg, 0.38 mmol), HOBt (52 mg, 0.38mmol) and DIPEA (133 mg, 1.03 mmol). The reaction mixture was stirred at rt for 16 hours, then purified by prep-HPLC to afford 6-cyclobutoxy-4-((3- (4-(cyclopropanecarbonyl)piperazin-1-yl)benzo[d]isoxazol-5-y l)methyl)phthalazin-1(2H)-one (53 mg 41% yield) as a white solid. LCMS ESI 500 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.44 (s, 1H), 8.20 – 8.10 (m, 2H), 7.50 (d, J = 8.7 Hz, 1H), 7.44 (d, J = 8.9 Hz, 1H), 7.30 (dd, J = 8.8, 2.2 Hz, 1H), 7.19 (d, J = 2.1 Hz, 1H), 4.85 (p, J = 7.0 Hz, 1H), 4.40 (s, 2H), 3.89 (s, 2H), 3.69 (s, 2H), 3.50 (s, 4H), 2.41 – 2.32 (m, 2H), 2.12 – 1.95 (m, 3H), 1.78 (q, J = 10.0 Hz, 1H), 1.64 (m, 1H), 0.81 – 0.67 (m, 4H). Synthesis of Example 211: 6-(4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzo[d]isoxazol-3-yl)piperazin-1-yl)nicotinonitri le

6-(4-(5-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)benzo [d]isoxazol-3-yl)piperazin- 1-yl)nicotinonitrile [1251] A solution of 6-cyclobutoxy-4-((3-(piperazin-1-yl)benzo[d]isoxazol-5-yl)me thy l)phthalazin- 1(2H)-one (120 mg, 0.26 mmol), 6-chloronicotinonitrile (39 mg, 0.30 mmol) and DIPEA (133 mg, 1.00 mmol) in DMF (2 mL) was stirred at rt under Ar (g) for 12 hours, then purified by prep-HPLC to afford 6- (4-(5-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methy l)benzo[d]isoxazol-3-yl)piperazin-1- yl)nicotinonitrile (75 mg 55% yield) as a white solid. LCMS ESI 534 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.44 (s, 1H), 8.54 (d, J = 2.2 Hz, 1H), 8.15 (d, J = 8.7 Hz, 2H), 7.92 (dd, J = 9.1, 2.3 Hz, 1H), 7.57 – 7.39 (m, 2H), 7.30 (dd, J = 8.8, 2.3 Hz, 1H), 7.19 (d, J = 2.3 Hz, 1H), 7.03 (d, J = 9.1 Hz, 1H), 4.85 (p, J = 7.1 Hz, 1H), 4.42 (s, 2H), 3.97 – 3.84 (m, 4H), 3.68 – 3.52 (m, 4H), 2.37 (m, 2H), 2.04 – 1.92 (m, 2H), 1.69 (m, 2H). Synthesis of Example 212: 6-Cyclobutoxy-4-((3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pi perazin-1- yl)benzo[d]isoxazol-6-yl)methyl)phthalazin-1(2H)-one

6-Bromo-3-chlorobenzo[d]isoxazole [1252] To a mixture of 6-bromobenzo[d]isoxazol-3(2H)-one (1.60 g, 7.48 mmol) in phosphoryl trichloride (20 mL) was added Et 3 N (1.51 g, 14.95 mmol). The mixture was stirred at 130 °C for 12 hours.The reaction was carefully added to water (10 mL), then extrated with EtOAc (3 x 30 mL). The combined organic layer was washed with brine, dried over Na 2 SO 4 and filtered. The solvent was removed in vacuo to give 6-bromo-3-chlorobenzo[d]isoxazole (1.16 g, 67% yield) as a white solid. LCMS ESI m/z: 232 [M+H] + . Step 2 tert-Butyl 4-(6-bromobenzo[d]isoxazol-3-yl)piperazine-1-carboxylate [1253] A mixture of 6-bromo-3-chlorobenzo[d]isoxazole (1.00 g, 4.30 mmol), tert-butyl piperazine-1- carboxylate (801 mg, 4.30 mmol) and 2,3,4,6,7,8,9,10-octahydropyrimido[1,2-a]azepine (786 mg, 5.16 mmol) in pyridine (20 mL) was stirred at 110 °C for 12 hours. The reaction was diluted with water (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic layer was washed with brine, dried over Na 2 SO 4 and filtered. The solvent was removed in vacuo and the residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0% to 50%) to give tert-butyl 4-(6-bromobenzo[d]isoxazol-3- yl)piperazine-1-carboxylate (1.20 g, 73% yield) as a white solid. LCMS ESI m/z: 382 [M+H] + . Step 3 tert-butyl 4-(6-vinylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate [1254] A mixture of tert-butyl 4-(6-bromobenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (1.20 g, 3.14 mmol), potassium vinyltrifluoroborate (505 mg,3.77 mmol), Pd(dppf)Cl 2 (230 mg, 0.31 mmol) and K 2 CO 3 (1.08 g, 7.85 mmol) in dioxane (10 mL) was stirred at 80 °C for 5 hours. The mixture was cooled to rt, diluted with H 2 O (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layer was washed with brine, dried over Na 2 SO 4 and filtered. The solvent was removed in vacuo and the residue was purified by silica gel chromatography (EtOAc/petroleum ether = 0% to 30%) to give tert-butyl 4-(6- vinylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (710 mg, 69% yield) as a white solid. LCMS ESI m/z: 330 [M+H] + . Step 4 tert-butyl 4-(6-formylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate [1255] To a mixture of tert-butyl 4-(6-formylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (700 mg, 1.99 mmol) in THF (9 mL) and H 2 O (6 mL) was added K 2 OsO 4 -2H 2 O (37 mg, 0.10 mmol). The mixture was stirred at rt for 5 min, added NaIO 4 (1.28 g, 5.96 mmol) and stirred at rt for 4 hours. Then THF was removed under reduced pressure, diluted with H 2 O (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layer was washed with brine, dried over Na 2 SO 4 and filtered. The solvent was removed in vacuo to give tert-butyl 4-(6-vinylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (710 mg, 69% yield) as a white solid. This was used without further purification. LCMS ESI m/z: 332 [M+H] + . Step 5 tert-butyl 4-(6-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)methyl )benzo[d]isoxazol-3- yl)piperazine-1-carboxylate [1256] A solution of tert-butyl 4-(6-formylbenzo[d]isoxazol-3-yl)piperazine-1-carboxylate (450 mg, 1.36 mmol), dimethyl (6-cyclobutoxy-3-oxo-1,3-dihydroisobenzofuran-1-yl)phosphona te (636 mg, 2.04 mmol) and Et3N (412 mg, 4.07 mmol) in THF (5 mL) was stirred at 25 °C under Ar (g) for 15 hours. Hydrazine hydrate (102 mg, 2.04 mmol) was added and the reaction was stirred at 70 ℃ for 1 hour. The reaction was cooled to rt, and the solvent was removed under vacuum. The formed solids were washed with water (10 mL) and petroleum ether/EtOAc (1:1, 10 mL) to give tert-butyl 4-(6-((7-cyclobutoxy-4- oxo-3,4-dihydrophthalazin-1-yl)methyl)benzo[d]isoxazol-3-yl) piperazine-1-carboxylate (430 mg, 60% yield) as a white solid. LCMS ESI m/z: 532 [M+H]+. Step 6 6-cyclobutoxy-4-((3-(piperazin-1-yl)benzo[d]isoxazol-6-yl)me thyl)phthalazin-1(2H)-one [1257] A solution of tert-butyl 4-(6-((7-cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzo[d]isoxazol-3-yl)piperazine-1-carboxylate (90 mg, 0.17 mmol) in HCl/dioxane (4M, 5 mL) was stirred at rt for 1 hour. The solvent was removed in vacuo to give 6-cyclobutoxy-4-((3- (piperazin-1-yl)benzo[d]isoxazol-6-yl)methyl)phthalazin-1(2H )-one (70 mg, 94% yield) as a white solid. This was used without further purification. LCMS ESI m/z: 532 [M+H] + . Step 7 6-cyclobutoxy-4-((3-(4-(5-(trifluoromethyl)pyrimidin-2-yl)pi perazin-1-yl)benzo[d]isoxazol-6- yl)methyl)phthalazin-1(2H)-one [1258] To a solution of 6-cyclobutoxy-4-((3-(piperazin-1-yl)benzo[d]isoxazol-6-yl)me thyl)phthalazin- 1(2H)-one (60 mg, 0.14 mmol) and 2-chloro-5-(trifluoromethyl)pyrimidine (25 mg, 0.14 mmol) in DMF (1 mL) was added DIPEA (72 mg, 0.56 mmol). The reaction mixture was stirred at 60 °C for 2 hours. The mixture was purified by prep-HPLC to give 6-cyclobutoxy-4-((3-(4-(5-(trifluoromethyl)pyrimidin-2- yl)piperazin-1-yl)benzo[d]isoxazol-6-yl)methyl) phthalazin-1(2H)-one (60 mg, 75% yield) as a white solid. LCMS ESI m/z: 532 [M+H] + . 1 H–NMR (400 MHz, DMSO-d 6 ) δ 12.46 (s, 1H), 8.75 (s, 2H), 8.15 (d, J = 8.8 Hz, 1H), 7.98 – 7.94 (m, 1H), 7.58 (s, 1H), 7.32 – 7.28 (m, 1H), 7.27 – 7.24 (m, 1H), 7.15 – 7.12 (m, 1H), 4.87 – 4.79 (m, 1H), 4.43 (s, 2H), 4.06 – 3.99 (m, 4H), 3.59 – 3.54 (m, 4H), 2.41 – 2.35 (m, 2H), 2.03 – 1.93 (m, 2H), 1.83 – 1.73 (m, 1H), 1.69 – 1.59 (m, 1H). Synthesis of Example 213: 6-cyclobutoxy-4-((3-(4-(cyclopropanecarbonyl)piperazin-1- yl)benzo[d]isoxazol-6-yl)methyl)phthalazin-1(2H)-one 6-cyclobutoxy-4-((3-(4-(cyclopropanecarbonyl)piperazin-1-yl) benzo[d]isoxazol-6-y l)methyl)phthalazin-1(2H)-one [1259] To a solution of 6-cyclobutoxy-4-((3-(piperazin-1-yl)benzo[d]isoxazol-6-yl)me th yl)phthalazin- 1(2H)-one (110 mg, 0.34 mmol) and cyclopropanecarboxylic acid (20 mg, 0.23 mmol) in DMF (2 mL) was added EDCI (68 mg, 0.35 mmol), HOBt (48 mg, 0.35 mmol) and DIPEA (122 mg, 0.94 mmol). The reaction mixture was stirred at rt for 12 hours, then purified by prep-HPLC to afford 6-cyclobutoxy-4-((3- (4-(cyclopropanec arbonyl)piperazin-1-yl)benzo[d]isoxazol-6-yl)methyl)phthalaz in-1(2H)-one (80 mg 68% yield) as a white solid. LCMS ESI 500.3 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.47 (s, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.93 (d, J = 8.3 Hz, 1H), 7.58 (s, 1H), 7.30 (dd, J = 8.8, 2.3 Hz, 1H), 7.25 (d, J = 8.3 Hz, 1H), 7.14 (d, J = 2.3 Hz, 1H), 4.84 (dd, J = 14.1, 7.1 Hz, 1H), 4.43 (s, 2H), 3.86 (s, 2H), 3.65 (s, 2H), 3.46 (d, J = 22.6 Hz, 4H), 2.36 (dd, J = 10.1, 7.3 Hz, 2H), 2.04 – 1.95 (m, 3H), 1.83 – 1.73 (m, 1H), 1.64 (dd, J = 18.8, 8.5 Hz, 1H), 0.77 – 0.72 (m, 4H). Synthesis of Example 214: 6-(4-(6-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1- yl)methyl)benzo[d]isoxazol-3-yl)piperazin-1-yl)nicotinonitri le 6-(4-(6-((7-Cyclobutoxy-4-oxo-3,4-dihydrophthalazin-1-yl)met hyl)benzo[d]isoazol-3-yl)piperazin-1- yl)nicotinonitrile [1260] A solution of 6-cyclobutoxy-4-((3-(piperazin-1-yl)benzo[d]isoxazol-6-yl)me thyl)ph thalazin- 1(2H)-one (120 mg, 0.30 mmol), 6-chloronicotinonitrile (71 mg, 0.50 mmol) and DIPEA (132 mg, 1.20 mmol) in DMF (2 mL) was stirred at rt under Ar (g) for 12 hours, then purified by prep-HPLC to afford 6- (4-(6-((7-cyclobutoxy-4-oxo-3,4-dihydroph thalazin-1-yl)methyl)benzo[d]isoxazol-3-yl)piperazin-1- yl)nicotinonitrile diazepane (60 mg 41% yield) as a white solid. LCMS ESI 534.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.47 (s, 1H), 8.53 (d, J = 2.3 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 7.98 – 7.87 (m, 2H), 7.59 (s, 1H), 7.31 – 7.24 (m, 2H), 7.14 (d, J = 2.3 Hz, 1H), 7.01 (d, J = 9.1 Hz, 1H), 4.91 – 4.75 (m, 1H), 4.43 (s, 2H), 3.96 – 3.82 (m, 4H), 3.61 – 3.49 (m, 4H), 2.40 – 2.32 (m, 2H), 2.04 – 1.91 (m, 2H), 1.70 (m, 2H). Results Table 2: Example Compounds

Biological Testing Methods Biological Example 1: Protocol for the PARP7 Biochemical Assay: [1261] 25 µL of 100nM His-GST-PARP7 in PARP buffer for reaction (PBR) (50mM HEPES pH 7.5, 100mM NaCl, 4mM MgCl 2 , 0.2mM TCEP) was added to each well of XpressBio GSH-coated 384 well plates and incubated for 1h at RT. The plate was then washed for 3 times in 50µL PBST (1X PBS, 0.02% Tween-20), followed by one wash in 1X PBS and one wash PBR. Each wash was incubated for 3 min. After washing, varying concentrations of each inhibitor was pre-incubated with 20 µM (2X) 6-a-NAD + . The solution was added to the plate, diluted to 1X in PBR, and incubated at 30˚C for 90 min. Following the enzymatic reaction, the plate was washed 3 times in PBST, once in PBS for 3 min each wash. The wells were then subjected to 25µL of click reaction buffer consisting of 100µM TBTA, 1mM CuSO 4 , 100µM Biotin-N 3 , 2mM TCEP made up in 1X PBS. The wells were incubated in click reaction buffer for 30min at 30˚C. The click buffer was removed, and the plate was washed 3 times with PBST and once with PBS as before. The plate was then blocked with 5% BSA in PBST. The plate was then washed 3 times with PBST and once with PBS. To develop, the plate was incubated with 25µL of 0.05ng/µL Step-HRP with 0.02% BSA in PBS. The plate was washed 3 times with PBS before developing with QuantaRed per manufacturer directions. Fluorescence (Excitation 550 nm, Emission 620 nm) was immediately read using a Paradigm Plate Reader. Biological Example 2: Protocol for the PARP1 and PARP2 Biochemical Assays: [1262] N-Terminal His-tagged PARPs (PARP1, PARP2) and SRPK2 were expressed and purified as previously described (Carter-O’Connell et al., J. Am. Chem. Soc.2014, 136, 14, 5201–5204.) PARP1 was purified to greater than 90% and SRPK2 to 70% or greater by an in-gel standard curve of Bovine Serum Albumin (Bio-Rad). PARP1 and PARP2 plate assays were performed as described in (Kirby et al, STAR Protocols, 2021, 2(1), 100344.) 50µL of 1µM SRPK2 in PARP Buffer for reaction (PBR) (50mM HEPES pH 7.5, 100mM NaCl, 4mM MgCl 2 , 0.2mM TCEP) was added to wells of a Nickel-NTA coated 96-well plate (Thermo Scientific) and incubated at RT for 1h. The solution was removed, and the plate was washed 3 times with PBST (1X PBS, 0.1% Tween-20), once with 1X PBS, and once with PBR. Each wash was incubated for 3 min. After washing, varying concentrations of each inhibitor was pre-incubated with 20 µM (2X) 6-a-NAD + .25µL of the 2X 6-a-NAD + /inhibitor mixture was added to the plate with 25µL of 20nM of PARP1 or PARP2 in PBR with 0.1mg/mL Activated DNA (Sigma). PARP final concentration 10nM. The reaction mixture was incubated at 30˚C for 1h. The plate was then washed 3 time with PBST and once with PBS. The wells were subjected to a 50µL click reaction consisting of 100µM TBTA, 1mM CuSO 4 , 100µM Biotin-N 3 , 2mM TCEP made up in 1X PBS at RT for 30 min. The plate was then washed 3 times with PBST before blocking in 100µL of 1% Milk (Carnation) in PBST at RT for 30 min. The plate was again washed 3 times with PBST and once with PBS before incubation with Strep-HRP (0.05ng/µL Strep-HRP (Fisher Scientific), 300ng/µL BSA, 1X PBS) for 30 min at RT. The plate was washed 3 time in PBST and once with PBS before development with QuantaRed (Pierce) per manufacturer directions. The plate was immediately read for fluorescence (Excitation 550 nm, Emission 620 nm) on Spectra Max i3 (Molecular Devices) following development. Three parameter nonlinear regression was performed with GraphPad Prism 9 to obtain IC 50 values. Biological Example 3: Protocol for the Cell Proliferation Assays Using NCI-H1373 (human lung adenocarcinoma), HARA (human lung squamous cell carcinoma) and CT26 (mouse colon adenocarcinoma) cell lines [1263] An assay to measure inhibition of NCI-H1373, HARA or CT26 cell growth was performed using CellTiter-Glo (CTG) One Solution Assay reagent (for detection of ATP by luminescence; Promega, G8461) and an EnVision microplate reader (Perkin Elmer, 2104-0010) in a 384-well format. On the day prior to treatment, NCI-H1373, HARA or CT26 cells were dissociated using trypsin-EDTA 0.25% and seeded in white 384-well plates at a density of 500 cells per well in medium (RPMI 1640 with L- glutamine + 10% FBS). Outermost wells were filled with only medium to mitigate edge effects. On the day of treatment, one row of wells in one plate was treated with CTG reagent. The plate was then covered with a clear plate seal and a white adhesive backing and read on the EnVision plate reader 30 minutes after the addition of CTG reagent in order to provide a Day 0 measurement of cell viability. Serial dilutions of compounds to be tested were prepared in DMSO, then all further diluted by the same factor in medium to minimize potential precipitation. Compounds diluted in medium were then transferred to the 384-well NCI-H1373, HARA or CT26 plates (0.5% DMSO final). Plates were cultured for 6 days. On Day 6, viability was measured using CTG reagent as described above. Growth curves and GI 50 values were calculated using Excel (Microsoft Office Professional Plus 2013) with XLFit add-in (ID Business Solutions Limited, version 5.5.0.5). The average Day 0 reading was subtracted from all Day 6 readings, and cell viability for each inhibitor at each concentration was calculated as a percentage of average vehicle-only reading. Fit Model (205) Dose Response One Site (a 4 parameter logistic model) was applied (Fit = (A+((B=A)/(1+((C/x)^D)))); Inv = (C/((((B-A)/(y-A))-1)^(1/D))); Res = (y-fit)). This model was used to calculate absolute GI 50 values (compound concentration at which cell viability is 50% of untreated control). Results [1264] Results of certain compounds are shown in Table 3. Table legend: [1265] IC 50 Range Values for PARP7, PARP1 and PARP2 biochemical assays: **** is [IC 50 ] < 50 nM; *** is 50 nM ≤ [IC 50 ] < 100 nM; ** is 100 nM ≤ [IC 50 ] < 500 nM; * is 500 nM ≤ [IC 50 ] < 2000 nM; - is 2000 nM ≤ [IC 50 ] where [IC 50 ] is the IC 50 concentration value in nanomolar units. [1266] GI 50 Range Values for NCI-H1373, HARA and CT26 cell proliferation assays: +++ is [GI 50 ] < 100 nM; ++ is 100 nM ≤ [GI 50 ] < 1000 nM; + is 1000 nM ≤ [GI 50 ] < 5000 nM; ^ is 5000 nM ≤ [GI 50 ] where [GI 50 ] is the GI 50 concentration value in nanomolar units. Table 3

Biological Example 4: Structurally distinct PARP7 inhibitors reveal new insights into the regulation of cytosolic nucleic acid-sensing by PARP7 SUMMARY [1267] The mono-ADP-ribosyltransferase PARP7 is an aryl hydrocarbon receptor (AHR)-stimulated gene that has emerged as a negative regulator of cytosolic nucleic acid (NA)-sensors of the innate immune system. Herein, we apply a rational design strategy for parlaying a pan-PARP inhibitor into a selective inhibitor of PARP7 (Example 10). Example 10 potently inhibits PARP7-mediated MARylation in vitro and in cells, and displays PARP family-wide selectivity. Consistent with recent studies using structurally distinct PARP7 inhibitor RBN-2397, co-treatment of mouse embryonic fibroblasts (MEFs) with Example 10 and an AHR agonist synergistically induced the expression of an AHR-target gene. Likewise, co- treatment of MEFs with Example 10 and cytoplasmic NA-sensor ligands synergistically induced the expression of the type I interferon, IFN-β. In mouse colon carcinoma (CT-26) cells, Example 10 treatment alone induced IFN-β signaling in a STING-dependent manner. Both Example 10 and RBN-2397 increased endogenous PARP7 protein levels in the nucleus of CT-26 cells, demonstrating that the catalytic activity of PARP7 regulates its protein levels in cells. Curiously, saturating doses of Example 10 and RBN-2397 achieved different maximum levels of PARP7 protein, which correlated with the magnitude of type I interferon gene expression. INTRODUCTION [1268] Cytosolic nucleic acid (NA)-sensing serves as the first line of defense against pathogens. When viruses and bacteria enter cells, they release their NAs into the cytoplasm, which are promptly detected by cytosolic NA sensors. DNA derived from viruses and bacteria is recognized by cytosolic NA sensors, namely cyclic cGMP-AMP (cGAMP) synthase (cGAS). Binding of pathogen-derived DNA to cGAS stimulates the production of cGAMP, which binds to and activates stimulator of interferon genes protein (STING). RNA derived from viruses is recognized by a different set of cytosolic NA sensors, the most well-studied being retinoic acid-inducible gene I (RIG-I). While the activation mechanisms of STING and RIG-I are distinct, they both converge on common signal transducers, such as TANK binding kinase 1 (TBK1) and its target interferon regulatory factor 3 (IRF3), the activation of which leads to type I interferon (IFN-I) production. IFN-I functions as both an autocrine and paracrine factor to activate the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway and subsequently induce interferon stimulated genes or (ISGs), which enhance the innate response and orchestrate an adaptive immune response (e.g., activation of cytotoxic T cells to kill infected cells). While involved in host defense against pathogens, overactivation of cytosolic NA-sensing can lead to autoimmune and inflammatory disorders. Thus, repressors of cytosolic NA-sensing are potentially useful for attenuating innate immunity. [1269] PARP7 (also known as TiPARP) has emerged as a potential repressor of cytosolic NA-sensing. PARP7 is a member of a family of 17 enzymes in humans called PARPs. PARP7, like most other PARP family members, catalyzes the transfer of ADP-ribose (ADPr) from nicotinamide adenine dinucleotide (NAD + ) to amino acids on protein targets, a post-translational modification (PTM) known as mono-ADP- ribosylation (MARylation). PARP7 was initially identified as a gene that is strongly upregulated by synthetic agonists (e.g., the carcinogen 2,3,7,8-tetrachlorodibenzo-p-dioxin or TCDD) of the aryl hydrocarbon receptor (AHR). Subsequent studies demonstrated that PARP7 is a negative regulator of AHR-ligand induced transcription. The repressor function of PARP7 is dependent on its catalytic activity, and it is postulated that this is due to MARylation of AHR, although the consequences of AHR MARylation on its function are not understood. [1270] Recently, PARP7 was identified as an AHR regulated gene that represses IFN-I signaling in response to cytosolic NA-sensor ligands and several types of RNA viruses. In mouse embryonic fibroblasts (MEFs), knockout of PARP7 in the presence of cytosolic NA-sensor ligands (i.e. the synthetic RIG-I ligand 3pRNA and the STING agonist cGAMP) synergistically induced I β expression. Expression of wild-type (WT), but not a catalytically inactive PARP7 mutant in PARP7 knockout MEFs rescued these effects, suggesting that the catalytic activity of PARP7 is potentially important for repressing cytosolic NA sensing and downstream IFN-β signaling. [1271] Beyond host defense against pathogens, cytosolic NA-sensing—in particular the cGAS/STING pathway—is involved in preventing an inappropriate immune response to cytosolic self-DNA. Cancer cells contain cytosolic self-DNA (e.g., due to defects in DNA repair) and must repress cGAS/STING to evade detection and elimination by adaptive immune responses. Therefore repressors of cGAS/STING, like PARP7, are potential therapeutic targets in the burgeoning area of immune-oncology. Indeed, a pyridazinone-based PARP7 inhibitor (RBN-2397) treatment induced STING-dependent, IFN-I signaling in certain cancer cells, resulting in immune-cell dependent killing in vivo. This study not only validates the role of PARP7 as a repressor of cGAS/STING signaling, but also demonstrates the therapeutic potential of PARP7 inhibitors as immunomodulatory agents. [1272] Intriguingly, stable knockout of PARP7 in mouse colorectal cancer cells (CT-26 cells) did not affect tumor growth in vivo and only weakly activated IFN-I signaling in vitro. Moreover, a CRISPR/Cas9 screen identified PARP7 itself as a hit for conferring RBN-2397 resistance in NIH-1373 cells. Together, these results suggest that the immunomodulatory effects of RBN-2397 are not simply due to catalytic inhibition, but may involve other mechanisms dependent on PARP7. [1273] Here, we used rational drug design to convert a PARP1/2/7 inhibitor into a selective PARP7 inhibitor (Example 10). Example 10 achieves selectivity for PARP7 over PARP1/2 by exploiting a hydrophobic sub-pocket adjacent to the NAD + binding site found in PARP7 but not PARP1/2. Cell-based studies using Example 10 validate the requirement of PARP7’s catalytic activity for its repressor functions in both AHR and IFN-I signaling. Example 10, as well as RBN-2397 increase PARP7 protein levels in the nucleus of CT-26 cells, suggesting that PARP7 stability is regulated by its activity. Yet despite being equipotent against PARP7 catalytic activity in vitro, RBN-2397 leads to a two-fold higher increase in PARP7 protein levels at saturating doses. Intriguingly, this difference correlated with a two-fold higher induction of an IFN-I-responsive luciferase reporter, potentially suggesting that, in addition to serving as derepressor of IFN-I-mediated transcription, PARP7 inhibitors may induce cGAS/STING signaling via a PARP7-inhibitor complex. Parlaying a PARP1/2/7 inhibitor into a selective PARP7 inhibitor [1274] MARylating PARPs—like PARP7—are the largest sub-group of the PARP family, yet are the least understood. This is because unlike PARylating PARPs, namely PARP1 and PARP2, there is a dearth of highly selective MARylating PARP inhibitors. An obstacle toward the development of a selective inhibitor of PARP7, or any PARP, is the high structural conservation among PARP catalytic domains; while the sequence homology is only ~50%, their structures are highly conserved, especially in the NAD + binding site, which is the major binding site for the majority of PARP inhibitors. To address this challenge, we identified previously a unique, hydrophobic cavity abutting the nicotinamide sub-pocket of the NAD + binding site in MARylating that can be exploited for the development of selective inhibitors of MARylating PARPs. Crystal structure analysis of one of our MARylating PARP inhibitors, ITK6, bound to PARP14 (PDB: 6FZM) shows that Leu1782 (human PARP14) acts as a “gatekeeper” (a term we borrow from the protein kinase field), allowing hydrophobic substituents emanating from the inhibitor scaffold to access the hydrophobic cavity. A hydrophobic amino acid at the gatekeeper position is conserved across the MARylating PARP sub-group, and indeed a growing number of MARylating PARPs exploit the hydrophobic cavity. PARylating PARPs have a conserved glutamate at the gatekeeper position, and analysis of multiple structures of PARP1 and PARP2 shows that this glutamate occludes access to the hydrophobic cavity, consistent with previous studies showing that MARylating PARP inhibitors containing substituents that access the hydrophobic cavity show selectivity over PARylating PARPs. [1275] A step in our rational design strategy is the identification of a NAD + -competitive inhibitor scaffold that contains a position that can be easily derivatized with a substituent that will interact favorably with the unique hydrophobic cavity in MARylating PARPs. In recent work, we found that the NAD + -competitive inhibitor Phthal01 (FIG.1A), which is based on a phthalazinone scaffold, is a potent, but non-specific inhibitor of PARP7. Using our PARP inhibitor screening platform, PARP activity screening and inhibitor testing assay (PASTA), we found that Pthal01 exhibited double-digit nanomolar potency against three PARPs: PARP1, 2, and PARP7, but was at least 12-fold more potent against PARP7 compared to all other PARP family members (FIG.1B). To parlay Phthal01 into a more selective inhibitor of PARP7, we designed and synthesized a Phthal01 analogue (Example 10) that contains a propynyl at the C-6 position of the phthalazinone scaffold (FIG.1A and FIG.12). We hypothesized that the C-6 propynyl would occupy the hydrophobic cavity in PARP7, but would clash with the glutamate gatekeeper in PARylating PARPs. We chose a propynyl because our previous studies suggested that this was a privileged substituent for generating selective MARylating PARP inhibitors. [1276] We used PASTA to profile the selectivity of Example 10, as well as the recently described PARP7 inhibitor RBN-20397 (FIG.1A), across the PARP family. Similar to Phthal01, we found that Example 10 potently inhibits PARP7 with an IC 50 = 13.7 nM (FIG.1B, FIG.6, and Table 4); however unlike Pthal01, Example 10 displays ~75-fold selectivity for PARP7 over PARP2 and it does not inhibit PARP1 up to 3 µM. Consistent with the previous study that first described RBN-2397, we found that RBN-2397 potently inhibits PARP2 (IC 50 = 30.3 nM), but does not inhibit most other PARP family members up to 3 µM. However, we did identify two discrepancies: using PASTA, we found that RBN- 2397 weakly inhibits PARP1 (IC 50 = 2639 nM) and PARP12 (IC 50 = 716 nM) whereas the previous study found that RBN-2397 was more potent (i.e. double digit nanomolar inhibition) against these PARPs. While the PARP inhibitor screening assay used in the previous study is similar to PASTA, there are a few differences that could account for this variance. Our in vitro IC 50 values for RBN-2397 against PARP1 and PARP12 are congruent with previously obtained cellular IC50 values for RBN-2397 against PARP1 and PARP12, suggesting PASTA accurately predicates the cellular efficacy of PARP inhibitors. Example 10 is >50-fold selective for PARP7 over most PARP family members beyond PARP1 and PARP2; two exceptions are PARP10 and PARP11, however, Example 10 still exhibits ~10-fold selectivity for PARP7 over these MARylating PARPs. The excellent family-wide selectivity of Example 10 for PARP7 over other PARP family members makes it a useful chemical tool for probing PARP7-specific functions in physiological and pathophysiological contexts. Table 4 [1277] PASTA showed that the addition of the propynyl group at the C-6 position of Example 10 dramatically improved its selectivity for PARP7 over PARP1 and PARP2 compared to Phthal01. To gain insight into this improved selectivity, we used an induced-fit docking (IFD) approach to predict the binding mode of Example 10 to a homology model of PARP7 (see Methods). IFD showed that the propynyl group at the C-6 position of Example 10 occupies the hydrophobic cavity, the entrance to which is controlled by the isoleucine (Ile631, human PARP7 numbering) gatekeeper in PARP7 (FIG.1C). Overlay of the crystal structure of PARP2 bound to olaparib (PDB: 4TVJ) shows that the glutamate (Glu558, human PARP2 numbering) gatekeeper prevents access to the hydrophobic cavity, and thus we predict that it would clash with the C-6 propynyl group of Example 10. Taken together these modeling studies support the notion that Example 10 achieves selectivity for PARP7 over PARP2 (and by extension all PARylating PARPs) by exploiting the hydrophobic cavity in PARP7. [1278] We next sought to determine if Example 10 inhibits the catalytic activity of PARP7 in cells. In previous studies we showed that that overexpression of GFP-PARP7 in HEK 293T cells leads to MARylation of PARP7, which can be assessed by western blot using an antibody that recognizes MARylation. We found that treatment of GFP-PARP7 expressing HEK 293T cells with Example 10 lead to a dose-dependent decrease in PARP7 MARylation (EC 50 = 8 nM) (FIG.1D, FIG.1E, and FIG.8). These results not only show that Example 10 is active in cells, but also provide evidence that the MAR signal detected on PARP7 is due to auto-MARylation and not trans-MARylation by another PARP. We also observed that Example 10 increases GFP-PARP7 protein levels in a dose dependent manner. This is consistent with our previous studies using Phthal01, as well as studies using a catalytically inactive PARP7 mutant. Example 10 validates the necessity of the catalytic activity of PARP7 for its repressor function on AHR activity [1279] Having established that Example 10 inhibits PARP7 catalytic activity in cells, we next sought to determine the effects of Example 10 on endogenous PARP7 function. For these initial studies, we focused on the role of PARP7 in ligand-mediated AHR transcriptional regulation in mouse embryonic fibroblasts (MEFs). We treated wild type (WT) MEFs with increasing concentrations of Example 10 in the presence or absence of the synthetic AHR agonist TCDD, and subsequently measured the expression of a major AHR target gene, CYP1A1, by quantitative reverse transcription PCR (RT-qPCR). Treatment of WT MEFs with Example 10 in the presence of 1 nM TCDD dramatically increased CYP1A1 mRNA levels in a dose-dependent manner (saturating effects observed at 100 nM Example 10) (FIG.2A). In the absence of TCDD, Example 10 had no effect on CYP1A1 mRNA levels (FIG.2A). Likewise, TCDD treatment alone did not induce CYP1A1. In contrast to WT MEFs, PARP7 -/- MEFs treated with 1 nM showed a robust induction of CYP1A1 (FIG.2B). Example 10 treatment did not further enhance the effects of TCDD treatment on CYP1A1 mRNA levels in PARP7 -/- MEFs (FIG.2B). Treatment of WT MEFs with increasing concentrations of TCDD in the presence of 100 nM Example 10 (saturating dose) lead to a strong, dose-dependent induction of CYP1A1 mRNA (FIG.2C). In PARP7 -/- MEFs TCDD treatment alone increased the levels of CYP1A1 in a dose-dependent manner (FIG.2D). Treatment of PARP7 -/- MEFs with 100 nM Example 10 did not further increase TCDD-mediated CYP1A1 induction (FIG.2D). Our results with Example 10 are congruent with recent studies showing that treatment with RBN-2397, or replacement of endogenous PARP7 with a catalytically inactive variant (H532A PARP7), profoundly enhanced TCDD-mediated AHR transcription. Taken together, these results strongly support the hypothesis that PARP7-mediated MARylation represses AHR activity in a TCDD-dependent manner. Example 10 synergistically induces IFN-β expression in the presence of cytosolic NA-sensor ligands [1280] Replacement of WT PARP7 with a catalytically inactive mutant (H532A PARP7) induces the IFN-I, IFN-β. Consistent with these studies, treatment of MEFs with 100 nM Example 10 alone induced a 3-fold increase in the IFN-I IFN-β mRNA as determined by RT-qPCR (FIG.3A). Strikingly, when MEFs were stimulated with the RIG-I agonist 3pRNA we observed a synergistic induction of IFN-β mRNA (FIG.3B). Similar results were obtained with the STING agonist cGAMP (FIG.3C). Our results support the hypothesis that the repressor function of PARP7 on IFN-I expression is dependent on its catalytic activity. Structurally distinct PARP7 inhibitors with non-intersecting PARP family-wide selectivity profiles induce IFN-I signaling in cancer cells [1281] Treatment of cancer cells (e.g., CT-26 and NCI-H1373) with RBN-2397 induced IFN-β expression and downstream signaling, suggesting that inhibition of PARP7 catalytic activity is responsible for this effect. Given that other PARP family members have been implicated in IFN-I signaling (e.g., PARP1/2 and PARP11), we reasoned that the non-overlapping selectivities of Example 10 and RBN-2397 across the PARP family would prove advantageous in studies aimed at validating PARP7 inhibition as the sole driver of IFN-β signaling in cancer cells. To evaluate the effects of these PARP7 inhibitors on IFN-β signaling in CT-26 cells, we monitored changes in STAT1 and STAT1 Tyr701 phosphorylation (pSTAT1) by western blot. Treatment of CT-26 cells with increasing concentrations of either Example 10 or RBN-2397 for 16 h dose-dependently increased STAT1 and pSTAT1 levels (FIG.4A). Quantification of the STAT1 and pSTAT1 western blot data showed a nearly indistinguishable response between RBN- 2397 and Example 10 (FIG.4B). Moreover, the increase in STAT1 and pSTAT1 by 300 nM (saturating dose) Example 10 and RBN-2397 showed the same time dependence; noticeable increases in pSTAT1 and STAT1 occurred at 4 h and 16 h, respectively (FIG.10A). Together, these results provide evidence the induction of IFN-β signaling in CT-26 cells by Example 10 and RBN-2397 is due to PARP7 inhibition, and not inhibition of other PARP family members. [1282] Previous studies showed that RBN-2397-mediated induction of IFN-β is dependent on a functional STING pathway; however, whether PARP7 acts upstream or downstream of STING was not clear. STING activation leads to auto-phosphorylation (Ser172) and activation of TBK1, followed by TBK1-mediated phosphorylation of IRF3 on Ser396. This leads to IRF3 dimerization and nuclear translocation where it regulates the transcription of IFN-I and other cytokines. Activation of STING by various ligands promotes its ubiquitylation and subsequent degradation by autophagy, attenuating the IFN-I response. Indeed, we found that the mouse-specific STING agonist DMXAA decreased substantially STING levels and induced the appearance of a higher molecular weight band (FIG.4C). In contrast, both PARP7 inhibitors only decreased slightly STING levels in a dose-dependent (FIG.4C) and time-dependent (FIG.10B) manner. Although this is inconsistent with canonical STING activation, it is possible that PARP7 inhibition activates STING in a non-canonical manner. We therefore looked at pTBK1 and pIRF3. In contrast to DMXAA, which induced robustly both pTBK1 and pIRF3, PARP7 inhibition did not induce pTBK1 and only increased slightly pIRF3 (FIG.4C). Taken together, these results suggest that PARP7 inhibition regulates IFN-β signaling downstream of TBK1/IRF3. [1283] Activation of the IFN-I-mediated STAT1 signaling pathway in certain cancer cells can lead to a loss of viability in a cell-autonomous manner. Indeed, RBN-2397 was shown to decrease the viability of NCI-H1373, but not CT-26 cells. Consistent with this finding, Example 10 dose-dependently decreased the viability of NCI-H1373 cells (FIG.9), but did not decrease viability in CT-26 cells. The EC 50 for Example 10 on NCI-H1373 is 104 nM whereas we found that the EC 50 for RBN-2397 is 17.8 nM. Why Example 10 is ~6-fold less potent than RBN-2397 is not clear considering their similar potencies against PARP7 in vitro and their similar efficacies in inducing IFN-I in CT-26 cells. Nevertheless, these results strengthen the notion that the loss of PARP7 catalytic activity decreases viability in NCI-H1373. PARP7 inhibitors increase endogenous PARP7 levels, and the extent of this increase correlates with the magnitude of the IFN-I transcriptional response [1284] Since we observed an increase in the protein levels of ectopically expressed GFP-PARP7 upon treatment with Example 10, we wondered if Example 10 and RBN-2397 increased the levels of endogenous PARP7 in CT-26 cells. In the absence of inhibitor, the levels of endogenous PARP7 were nearly undetectable; however, increasing concentrations of Example 10 and RBN-2397 increased PARP7 levels (FIG.4D). Quantification of the western blot data revealed that RBN-2397 caused an unexpected 2- fold increase in PARP7 protein abundance compared to Example 10 at saturating doses (FIG.4E). Given this difference, we asked if the increase in PARP7 protein levels occurred on similar timescales. The time- dependent increase in PARP7 protein levels was similar for both inhibitors, with maximal PARP7 protein levels occurring ~16 h (FIG.10C). [1285] Although both PARP7 inhibitors induced similar levels of pSTAT1/STAT1 (FIG.4B), we wondered if the levels of PARP7 inhibitor-induced secreted IFN-β were different between Example 10 and RBN-2397. Using ELISA, we found that RBN-2397 increased significantly the levels of secreted IFN-β to a greater extent than Example 10 at saturating doses (FIG.4F). As expected, the PARP7 inhibitor-induced induction of IFN-β was completely blocked by the covalent STING inhibitor H-151 (FIG.4F). Since PARP7 induces IFN-β and given that many PARPs are interferon stimulated genes (ISGs), we wondered if IFN-β alone increased PARP7 levels. In contrast to PARP7 inhibitors, recombinant IFN-β did not increase PARP7 levels despite strong induction of pSTAT1/STAT1 (FIG. 10D); hence, the PARP7-inhibitor induced increase in PARP7 levels is independent of IFN-β. [1286] To determine if the higher levels of RBN-2397-induced IFN-β compared to Example 10 correlated with higher levels of IFN-β-dependent transcription, we generated an interferon-sensitive response element (ISRE)-firefly luciferase (Luc)—CT-26 cell line using lentiviral transduction. ISRE-Luc have been used in various cell lines as a convenient reporter of IFN-I-dependent transcription. We treated ISRE-Luc)—CT-26 cells with increasing concentrations of either Example 10 or RBN-2397 for 16 h, and then measured Luc-generated luminesce in lysates. Intriguingly, while the EC 50 for the two inhibitors was similar, the magnitude of the response was greater (~2-fold) for RBN-2397 compared to Example 10 at saturating doses (FIG.4G). The extent of the ISRE increases is well correlated with the extent of the PARP7 protein level increases in the presence of saturating inhibitors. Taken together, these results possibly hint at additional mechanisms—beyond catalytic inhibition—by which PARP7 inhibitors regulate IFN-I signaling. PARP7 inhibition increases PARP7 abundance in the nucleus [1287] To gain insight into how PARP7 inhibitor-mediated increases in PARP7 protein levels could influence IFN-I transcriptional output, we next determined (i) the endogenous PARP7 localization, which is currently unknown, and (ii) if PARP7 localization changes upon inhibitor treatment, as we have observed for other PARPs. Unfortunately, there is only one validated PARP7 antibody, which in our hands did not work for immunofluorescence (IF). We therefore sought a strategy in which endogenous PARP7 could be replaced with an epitope-tagged variant. We envisioned that the epitope tag would be small enough so as to not perturb endogenous PARP7 function or localization. [1288] Recently, a versatile, small epitope tag, termed HiBiT, was developed. Importantly, an antibody against HiBiT has been used in IF applications. Another feature of the HiBiT is that it binds with high affinity to LgBiT, the binding of which reconstitutes a luminescent protein akin to Nano luciferase. The small size of the HiBiT allows for efficient knock-in using CRISPR/Cas9. We therefore generated CT-26 cells in which endogenous PARP7 was replaced with HiBiT-PARP7. HiBiT-PARP7—CT-26 cells were treated with 300 nM Example 10 or RBN-2397, or vehicle control (DMSO) for 18 h. HiBiT-PARP7 localization was assessed by IF using an anti-HiBiT antibody. In the absence of PARP7 inhibitors, we could not detect HiBiT-PARP7, preventing us for asking if PARP7 localization changes upon inhibitor treatment (FIG.5A). In the presence of Example 10 and RBN-2397, a nuclear HiBiT signal that colocalized with DAPI could be detected (FIGS.5B, 5C, and 11). The HiBiT-PARP7 signal was stronger for RBN-2397 compared to Example 10, consistent with western blot analysis of endogenous, untagged PARP7. Importantly, no signal was detected in WT CT-26 cells treated with 300 nM RBN-2397, demonstrating the specificity of the HiBiT-PARP7 signal in HiBiT-PARP7—CT-26 cells (FIG.5A). To confirm our localization studies, we biochemically fractionated lysates from WT CT-26 cells treated with or without PARP7 inhibitors (300 nM). Using western blot with a validated PARP7 antibody, PARP7 was not detected in either the cytoplasmic or nuclear fractions; in contrast, PARP7 was detected robustly in the nuclear fraction (FIG.5C). A very faint signal could be detected in the cytoplasmic fraction (FIG.5C). Together, these results show that in CT-26 cells, the PARP7—inhibitor complex is localized predominately to the nucleus. [1289] Because we could not detect HiBiT-PARP7 in the absence of PARP7 inhibitors, we could not determine if PARP7 inhibitors altered the localization of PARP7. We therefore turned to live-cell imaging experiments using ectopically expressed GFP-PARP7 in HeLa cells, which are commonly used for these types of studies. HeLa cells were transfected with GFP-PARP7 as well as mRuby2-Nup90, which was used as a nuclear marker. These transfected cells were treated with 300 nM RBN-2397, Example 10, or DMSO and localization was monitored via live-cell imaging over a period of 2 h. In the absence of PARP7 inhibitors, GFP-PARP7 localized to the nucleus, consistent with our IF studies using HiBiT- PARP7. In some cells, GFP-PARP7 was found in large puncta in the nucleus (FIG.5D).Within 30 min of PARP7 inhibitor treatment, these nuclear puncta disappeared, and GFP-PARP7 was instead localized in a diffuse, pan-nuclear pattern (FIG.5D). The pan-nuclear localization of GFP-PARP7 in the presence of inhibitors is similar to the localization of GFP-tagged, catalytically inactive PARP7. Taken together, these results suggest that PARP7 catalytic activity controls its localization in cells. DISCUSSION [1290] Much attention—both in terms of fundamental research and therapeutic development—has been given to PARylating PARPs (i.e., PARP1, PARP2, and PARP5a/b (Tankyrase1/2) in large part because multiple classes of potent and selective inhibitors based on distinct chemotypes are available. While genetic tools, such as RNAi and most recently CRISPR/Cas9, have been powerful means to prosecute the biology of the lesser known MARylating PARPs, selective small molecule inhibitors of MARylating PARP offer unique advantages over genetic strategies, chiefly among them is their ability to directly assess the functional role of PARP catalytic activity in cells. Compared to PARylating PARPs, there is a dearth of inhibitors selective for individual MARylating PARPs, which has hampered the studies focused on elucidating the physiological and pathophysiological roles of their catalytic activity. Previously we uncovered a hydrophobic cavity in MARylating PARPs, the accessibility of which is controlled by a hydrophobic gatekeeper amino acid. The hydrophobic cavity is not accessible in PARylating PARPs because a glutamate in the gatekeeper position blocks the entrance to the hydrophobic cavity. In this study, we exploited differences in the gatekeeper position between PARylating PARPs and MARylating PARPs to rationally convert a pan-PARP inhibitor into a selective PARP7 inhibitor, Example 10. [1291] Example 10 is structurally distinct from the previously described PARP7 inhibitor RBN-2397. Example 10 is based on a phthalazinone scaffold whereas RBN-2397 is based on a pyridazinone scaffold. When profiled in our PARP inhibitor screening platform, PASTA, Example 10 and RBN-2397 inhibited PARP7 with similar efficacy, but showed different selectivity across the family. Of particular note, Example 10 showed better selectivity for PARP7 compared to the PARylating PARP, PARP2. Modeling suggests that the improved PARP7 to PARP2 selectivity is due to a greater engagement of the hydrophobic cavity by Example 10 versus RBN-2397. [1292] Example 10 was used to investigate the role of PARP7 catalytic activity in regulating AHR transcription and IFN-I signaling driven by RIG-I and STING in MEFs. Example 10 synergized with an AHR agonist to enhance AHR-mediated transcription. Similarly, Example 10 synergized with RIG- I/STING ligands to induce the expression the IFN-I, IFN-β. These results described here, together with previous studies using RBN-2397 and a catalytically inactive PARP7 mutant, support the hypothesis that the catalytic activity of PARP7 acts as a repressor AHR and RIG-I/STING-mediated IFN-I signaling. In the case of AHR, it was shown that PARP7 can MARylate AHR in vitro and in cells; however, whether AHR MARylation is the means by which PARP7 catalytic activity regulates AHR-mediated transcription remains to be determined. In the case of RIG-I/STING-mediated IFN-I signaling, PARP7 was shown to MARylate TBK1 in vitro, but studies showing directly MARylation of TBK1 are lacking. Using a combined chemical genetics and proximity labeling approach, we identified several direct targets of PARP7, several of which have known roles in IFN-I signaling. We did identify TBK1 as a PARP7 interacting partner, but not as a MARylation target. One possible reason for this is that MARylation of TBK1 by PARP7 is context dependent (for example, during cytoplasmic NA-sensor stimulation). [1293] Curiously, we found that the RBN-2397 and Example 10 increased endogenous PARP7 protein levels in CT-26 cells. Previous studies found that a catalytically inactive GFP-PARP7 mutant is expressed at a higher level than WT GFP-PARP7. Additionally, the expression levels of WT GFP-PARP7 could be increased using MG132. Our results presented here, together with the aforementioned studies, support the hypothesis that PARP7 stability is regulated by its catalytic activity in a ubiquitin-dependent manner. How catalytic activity of PARP7 regulates its stability is not clear; however, we and others have identified several ubiquitin E3 ligases that interact with PARP7. [1294] Finally, we found that the magnitude of IFN-I-dependent transcription correlated with the levels of PARP7 protein levels in the presence of PARP7 inhibitors. While this is corelative at the moment, one intriguing possibility is that the PARP7 inhibitor—PARP7 complex drives IFN-I signaling. Biological Example 5: Methods for the Figures and for Biological Example 4 Cell Culture [1295] All cells were cultured at 37˚C with 5% CO 2 . HEK293T cells were cultured in DMEM + 1X Glutamax + 10% FBS. CT-26 and NCI-H1373 cells were cultured in RPMI-1640 + 10% FBS. ISRE-Luc expressing CT-26 cells were cultured in RPMI-1640 + 10% FBS + 20 µg/ml puromycin (Sigma). Stable Cell Line Production [1296] ISRE-Luc CT-26 cells were produced by lentiviral transduction. Briefly, 400,000 CT-26 cells in 800 µl of media were incubated with 200µl of ISRE Luciferase Reporter Lentivirus (BPS Biosciences, #79824) with 8 µg/ml polybrene. The mixture was centrifuged at 800g for 30 min at 25˚C. Media was removed and cells were plated in 2 ml of fresh media.48 h post transduction, cells were selected for with 20 µg/ml puromycin. Following selection, cells were continuously cultured win 20 µg/ml puromycin as described above. [1297] HiBiT-PARP7 KICT-26 cells were generated by Synthego via CRISPR/Cas9 Knock-in (KI) of GTGAGCGGCTGGCGGCTGTTCAAGAAGATTAGC to the PARP7 N-Terminus. The Knock-in cell pool was cultured as previously described for CT-26 cells. PASTA Biochemical PARP screening Assay [1298] N-Terminal His-tagged PARPs and SRPK2 were expressed and purified as previously described (Carter-O’Connell et al., J. Am. Chem. Soc.2014, 136, 14, 5201–5204.) Proteins were quantified against an in-gel standard curve of Bovine Serum Albumin (Bio-Rad) using ImageLab (BioRad). [1299] PARPs 1, 2, 5cat, 6, 8, 10, 11, 12cat, 14wwe, 15cat plate assays were performed as described (Kirby et al, STAR Protocols, 2021, 2(1), 100344.). Briefly, 50 µl of 1 µM SRPK2 in PARP Buffer for reaction (PBR) (50 mM HEPES pH 7.5, 100 mM NaCl, 4 mM MgCl 2 , 0.2 mM TCEP) was added to wells of a Nickel-NTA coated 96-well plate (Thermo Scientific) and incubated at RT for 1 h. The solution was removed, and the plate was washed 3 times with PBST (1X PBS, 0.1% Tween-20), once with 1X PBS, and once with PBR. Each wash was incubated for 3 min. After washing, varying concentrations of each inhibitor was pre-incubated with 20 µM (2X) 6-a-NAD + (prepared as described) (Carter-O’Connell et al., J. Am. Chem. Soc.2014, 136, 14, 5201–5204.) 25 µl of the 2X 6-a-NAD + /inhibitor mixture was added to the plate with 25 µl of 2X PARP. For PARP1 and PARP2, 0.1 mg/ml Activated DNA (Sigma) was supplemented. The reaction mixture was incubated at 30˚C for 1 h. The plate was then washed 3 time with PBST and once with PBS. The wells were subjected to a 50µL click reaction consisting of 100 µM biotin- PEG3-Azide (Click Chemistry Tools), 100 µM Tris[(1-benzyl-1H-1,2,3-triazol-4-yl)methyl] amine (TBTA) (Click Chemistry Tools), 1 mM CuSO4 and 1 mM TCEP in 1X PBS at RT for 30 min. The plate was then washed 3 times with PBST before blocking in 100 µl of 1% Milk (Carnation) in PBST at RT for 30 min. The plate was again washed 3 times with PBST and once with PBS before incubation with Strep- HRP (0.05 ng/µl Strep-HRP (Fisher Scientific), 300 ng/µl BSA, 1X PBS) for 30 min at RT. The plate was washed 3 time in PBST and once with PBS before development with QuantaRed (Pierce) per manufacturer directions. The plate was immediately read for fluorescence (Excitation 550 nm, Emission 620 nm) on Spectra Max i3 (Molecular Devices) following development. Three parameter nonlinear regression was performed with GraphPad Prism 9 to obtain IC 50 values. [1300] For PARP3, PARP4bc, and PARP16, 200-350ng of PARP was directly adhered to nickel coated 96-well plates for 60 min at 25˚C in PBR. PARP3 was activated by Dnick 5’P as previously described (Langelier, Marie-France et al. Nucleic acids research vol.42,12 (2014): 7762-75. ). The plate was washed three times with PBST, once with PBS, and once with PBR. Varying concentrations of inhibitor were pre-incubated with NAD + and added to the plate (final NAD + concentration: 100 µM for PARP3, PARP4bc; 400 µM for PARP16). The plate was incubated at 30˚C for 60 min, then washed three times with PBST before blocking with 5% Milk in PBST. The plate was washed three times with PBST and then incubated with 1:5000 (PARP3) or 1:25000 (PARP4bc, PARP16) Poly/ Mono-ADP Ribose rabbit monoclonal antibody (Cell Signaling Technology) in PBST with 2% BSA and 0.05% NaN 3 for 30 min at 25˚C. The plate was washed three times with PBST before incubation with 1:5000 (PARP3) or 1:25000 (PARP4bc, PARP16) anti-Rabbit HRP (Jackson ImmunoResearch) in 5% Milk in PBST for 30 min at 25˚C. The plate was then washed three times with PBST and once with PBS before developing as described previously. [1301] GST-tagged PARP7 was expressed and purified as previously described (Hutin et al., 2018). Proteins were quantified by an in-gel standard curve of Bovine Serum Albumin (Bio-Rad). [1302] For PARP7, 250ng of PARP7 was adhered to glutathione coated 96-well plates (Thermo Scientific) in PBR for 60 min at RT. The plate was washed three times with PBST (1X PBS + 0.1% Tween-20), once with PBS, and once with PBR (50 mM HEPES pH 7.5, 100 mM NaCl, 4 mM MgCl 2 , 0.2 mM TCEP). After washing, varying concentrations of each inhibitor was pre-incubated with 20 µM (2X) 6-a-NAD + . The solution was added to the plate, diluted to 1X in PBR, and incubated at 30˚C for 60 min. The plate was washed three times with PBST and once with PBS before performing click conjugation (100 µM biotin-PEG3-Azide, 100 µM Tris[(1-benzyl-1H-1,2,3-triazol-4-yl)methyl] amine (TBTA), 1 mM CuSO4, 1 mM TCEP, 1X PBS) for 30 min at RT. The plate was then washed three times with PBST and blocked with 1% Milk (Carnation) in PBST. The plate was subsequently washed three times in PBST and once in PBS before incubation with Strep-HRP (300 ng/µl BSA, 0.05 ng/µl Strep-HRP (Jackson ImmunoResearch), 1X PBS) for 30 min at RT. The plate was washed three time in PBST and once in PBS before development with QuantaRed Enhanced Chemifluorescent HRP Substrate as described previously. Molecular Modeling [1303] Due to the lack of experimental structure of PARP7, a homology model was built based on the sequence alignment.6V3W is the PDB code of PARP12 complex with a small molecule RBN-2397 and used as the template to develop the homology model of the PARP7 catalytic domain. A pharmacophore model was developed and used to place the RBN-2397 and Example 10 efficiently in the pocket followed by binding affinity assessment in the process of docking. Both the pharmacophore development and docking were carried out by MOE software package from CCG (https://www.chemcomp.com/). Western Blotting [1304] CT-26 cells at 75% confluency were treated with PARP7 inhibitors from 0-1 µM for 16 h for the dose-response or at 300 nM for the time-course (0-24 h). As a positive control, cells were treated with DMXAA (10 µg/ml) for one hour. At the end of treatment, cells were washed once with cold PBS and the plate was frozen in -80˚C prior to lysis. For lysis, the plate was thawed on ice, and lysed using a cytosolic lysis buffer (50 mM HEPES pH 7.4, 150 mM NaCl, 1 mM MgCl2, 1% triton X-100 supplemented with fresh 100 µM TCEP, 1X protease inhibitor (Roche), 1X phosphatase inhibitors (Sigma cocktail 2&3), and 30 µM Pan-PARP inhibitor Pthtal01. Lysates were clarified by centrifugation at 14,000 rpm at 4˚C for 10 minutes. Supernatants were transferred into a new tube and protein concentration was determined using the Bradford assay (Bio-Rad).4X sample buffer (40% glycerol, 200mM Tris-Cl (pH 6.8), 8% SDS, 4% β- mercaptoethanol, 0.02% bromophenol blue) was added to normalized lysates and samples were boiled at 95˚C for 5 min. Total protein was separated on a 10% SDS-PAGE gel, transferred to nitrocellulose membrane using Trans-Blot Turbo Transfer System (BioRad), followed by blocking with 5% milk (Carnation) in TBST. The blots were probed overnight at 4˚C against the following antibodies (Cell Signaling Technology): STING (1:000, D2P2F), Rig-I (1:1000, D14G6), phospho-STAT1 (Tyr701) (1:000, D4A7), STAT1 (1:000, 9172), phospho-IRF3 (Ser396) (1:1000, 4D4G), IRF3 (1:1000, D6I4C), Phospho-TBK1 (Ser172) (1:1000, D52C2), TBK1 (1:1000, D1B4), and tubulin (1:2000, DM1A). PARP7 was detected using an in-house mouse anti-PARP7 antibody (Rasmussen 2021). After PBST washes, blots were incubated with a goat anti-rabbit (1:10000, Jackson Immuno Reseach Labs) or goat anti-mouse (1:5000, Invitrogen) HRP-conjugated secondary antibody in 5% milk in TBST for 1 hour at room temperature. Blots were washed with TBST and developed using SuperSignal™ West Pico or Femto chemiluminescent substrate (Thermo Scientific) and imaged on a ChemiDoc Gel Imaging System (BioRad). [1305] MEFs isolated from C57BL/6 mice were plated at a density of 1.0 x 10 5 cells per ml in a 6-well plate. The following day, cells were treated with DMSO, 100 nM RBN-2397 or 100 nM KMR for 6 hours. Two wells were pooled, and pellets were resuspended in 20 ml of PBS-T [phosphate buffered saline (PSB) containing 0.1%Tween20]. The cells were lysed in 200 µl of SDS lysis buffer (1% SDS, 100 mM Tris, pH 7.5, 200 mM NaCl, 10% glycerol) supplemented with 1x protease inhibitors cocktail (Merck) and 2 mM DTT. Samples were sonicated using a Bioruptor at low intensity for two 30 seconds on/off cycles then centrifuge at 20,000xg for 10 min at 4 o C. Protein concentration was determined, and sample diluted with Laemmli buffer and incubated at 95 o C for 5 min.25 µg of protein were separated by SDS- PAGE and transferred to a PVDF membrane. Membranes were blocked for 1 h at room temperature in 5% non-fat milk dissolved in TBS containing 0.1% Tween20 (TBS-T). Membranes were then incubated overnight at 4 o C with an in-house generated anti-PARP7 (Rasmussen, 2021), 1:8000 dilution of anti-AHR (Enzo Life Sciences SA210; lot# 03011639) followed by incubation with the appropriate secondary antibodies. PVDF membranes were stripped and incubated with anti-β-actin antibody 1:4000 (Sigma- Aldrich; A-2228). RNA Extraction and Gene Expression Analysis [1306] CT26 cells, MEFs isolated Parp7 +/+ or Parp7 -/- mice (MacPherson, 2013 #2606) were plated at a density of 1.0 x 10 5 cells per ml in a 12-well plate. The following day, cells were treated for 24 hours with DMSO, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (Accustandard; New Haven, CT, USA), KMR or TCDD + KMR at the doses indicated in the figure captions. For the 3pRNA and cGAMP studies, Parp7 +/+ MEFs were plated at a density of 1.0 x 10 5 cells per ml in a 12-well plate. The following day, the cells were transfected with 100 ng/ml 3p-hpRNA (Invivogen) or 5 mg/ml 2’3’-cGAMP (Invivogen) using Lipofectamine 2000 and immediately treated with 100 nM KMR for 4 h. Total RNA was isolated using Aurum™ Total RNA isolation kit (BioRad), and 1 mg of RNA was reverse transcribed using High Capacity cDNA Reverse Transcription Kit according to the manufacturer’s instructions (Applied Biosystems). Each RT-qPCR reaction consisted of 0.1 µl forward primer, 0.1 µl reverse primer, 5 µl 2X SsoAdvanced Universal SYBR Green Supermix (BioRad), 1 µl of cDNA and dH 2 O to a total volume of 10 µl. The primers used for TATA-binding protein (TBP) were forward 5’- TTGTACCGCAGCTGCAAAAT-3’ and reverse 5’-TATATTCGGCGTTTCGGGCA-3’, for PARP7 antisense (AS) were forward 5’-CTATGCGCGTGTTTTGCAG-3’ and reverse 5’- TCTGGGTGATGAAGGTCGTAG-3’, for Cytochrome P450 family 1 subfamily A member 1 (CYP1A1) were forward 5’-TGGTCTCCCTTCTCTACACTCTTGT-3’ and reverse 5’- ATTTTCCCTATTACATTAAATCAATGGTTCT-3’, and for IFNb were forward 5’- TGGGAGATGTCCTCAACTGC-3’ and reverse 5’-CCAGGAGTAGCTGTTGTACT-3’. All target transcripts were normalized to TBP and analyzed using the comparative cycle threshold (CT) (ΔΔCT) method. ISRE Luciferase Assay [1307] 10,000 ISRE-Luc CT-26 or WT CT-26 cells were seeded into wells of a white 96-well plate and grown overnight. Compounds were serial diluted at 1000X concentration in DMSO. For dosing, compound dose response curves were dilute 1:100 in culture media, then added at 1:10 dilution to cell culture. Final DMSO concentration was 0.1%. Cells were incubated in treatment for 18 hours. Following treatment, the plates were developed using ONE-Glo Luciferase Assay system (Promega) per manufacturer directions. The luminescence signal was read on a Spectra Max i3 (Molecular Devices) plate reader with a 2 second integration time. WT CT-26 signal was background subtracted and curves were fit using a four-parameter linear regression in GraphPad Prism 9. Interferon-β ELISA [1308] CT-26 cells were seeded into a 6-well plate. After 24 h, the growth media was replaced, and cells were treated with inhibitors. After treatment, growth media was harvested and assayed with PBL Verikine Mouse IFN-β ELISA kit following the manufacturer protocol. HiBiT Staining [1309] 50,000 HiBiT-PARP7 KI CT-26 or WT CT-26 cells were plated onto coverslips. After 24 hours, the cells were treated with PARP7 inhibitors or DMSO for 18 hours. The cells were then washed once with cold PBS and fixed with 4% PFA in PBS at RT for 10 min. The coverslips were washed three times in PBS over a 5-minute period. The HiBiT antibody (Promega) was then added at 1:1000 dilution in antibody incubation buffer (3% Horse Serum, 0.5% Triton X-100, 0.025% NaN 3 , PBS) for 2 hours at RT. The coverslips were washed three times in PBST (PBS, 0.1% Tween-20) for 5 min per wash. Subsequently, the coverslips were incubated with 1:1000 AlexaFluor657 anti-Mouse in antibody incubation buffer for 1 hour at RT protected from light. Finally, the coverslips were counterstained with 1:5000 DAPI in PBS for 10 min before washing three times in PBST. The coverslips were then mounted onto slides with Citifluor CFMR2 Antifade (Electron Microscopy Sciences) and sealed with CoverGrip (Biotium). Imaging was performed on a Ziess LSM880 confocal laser scanning microscope with Airyscan with a 63x oil objective at 2048x2048 resolution DAPI excitation was performed with a 405 nm laser at low (≤ 1%) laser power. Red fluorophores were excited with a 559 nm laser at 7.5% laser power. Cellular Fractionation [1310] CT26 cells were culture as described above. Cells were treated with RBN-2397 or Example 10 for 18h before harvest. Nuclear–cytoplasmic fractionation was carried out using the NE-PER™ Nuclear and Cytoplasmic Extraction Reagents kit (Thermo Fisher Scientific) according to the manufacturer’s protocol. Western blot was performed as described above. Live Cell Imaging [1311] Hela cells were seeded and grown in 8 well-chambered glass slides (Lab-Tek, Nunc, Cat No. 177402). Cells were subsequently transfected with 100ng DNA of each of the plasmid constructs (GFP- PARP7 and mRuby2-Nup50-N-10 (Addgene plasmid 55908)) using jetOptimus DNA transfection reagent (Polyplus, Cat No.117-07), according to manufacturer protocol.10h after transfection, media containing transfection mixture was removed. Live cell imaging was performed on the Olympus FV1200 microscope equipped with an environmental box. Cells were kept at 37C and 5% CO2 during the whole course of imaging. All images were taken with the 63x oil objective NA=1,35, sequential acquisition. Acquired images were analyzed using ImageJ software. Cell Titer-Glo Viability [1312] NCI-H1373 cells were seeded in a white 96-well plate (Corning) at a density of 1,000 cells per well. After 24 hours, cells were treated with PARP7 inhibitors and incubated at 37˚C for 6 days. Cell viability was assessed using CellTiter-Glo (CTG) 2.0 (Promega). After addition of the CTG reagent (following manufacturer protocol), the plate was incubated on an orbital shaker for 15 min (protected from light) at room temperature. Chemiluminescence was measured on a Spectra Max i3 (Molecular Devices), normalized to DMSO-treated wells, and IC 50 values were calculated by non-linear regression (three-parameter fit) analysis using Prism 9 (GraphPad). Chemistry Methods Synthesis of Compound 1B [1313] 5-bromo-phthalide (2g, 9.44mmol) was heated to reflux in carbon tetrachloride (CCl4) with Azobisisobutyronitrile (160mg, 0.944 mmol) and N-Bromosuccinimide, (1.9g, 10.7 mmol) for 4 hours until reaction was completed. The reaction was then filtered and the filtrate was concentrated. The crude mixture was then purified via column purification using combiflash ISCO purification system using a mobile phase gradient from 0 to 30% ethyl acetate in hexanes.2.35g of compound 1B was obtained (86% yield). 1 H NMR (400 MHz, CDCl3) δ 7.84 – 7.71 (m, 3H), 7.34 (s, 1H). Synthesis of Compound 2B [1314] Under dry conditions, in a clean, oven dried flask, compound 1B (white solid) (2g, 6.9mmol) was heated slightly (50˚C) under vacuum pressure with dry triphenyl phosphine (1.81g, 6.9mmol) for 30 mins. Triphenyl phosphine and compound 1 were both dried over P2O5 overnight prior to starting the reaction. To the dry starting materials, dry THF was added and the reaction was allowed to stir under reflux overnight. A white precipitate formed quickly after reaction started. The white precipitate, compound 2B, was filtered out and dried under vacuum. (2.6g, 80% yield) 1 H NMR (400 MHz, CDCl 3 ) δ 10.29 (s, 1H), 7.88 (ddd, J = 19.4, 15.0, 9.1 Hz, 8H), 7.70 (td, J = 7.9, 3.7 Hz, 7H), 7.60 (d, J = 8.2 Hz, 2H), 7.25 (s, 1H). Synthesis of Compound 3B

[1315] Compound 2B (0.475 g, 1.0mmol) and 2-fluoro-5-formylbenzonitrile (.180 g, 1.2 mmol) were dissolved DCM (4.0 mL), to this was added triethylamine (0.18 mL, 1.3 mmol) and the reaction was stirred overnight. The reaction mixture was quenched with water (5 mL), extracted with DCM (2 x 10 mL), the organic layer was dried over MgSO4, filtered and evaporated the solvent to yield a pale white solid (0.335 g, 98%). 1 H NMR (400 MHz, DMSO) δ 8.22 (d, J = 1.7 Hz, 1H), 8.17 (d, J = 8.5 Hz, 1H), 8.02 (dd, J = 8.5, 1.8 Hz, 1H), 7.90 (dd, J = 6.2, 2.2 Hz, 1H), 7.73 (ddd, J = 7.9, 5.3, 2.3 Hz, 1H), 7.48 (t, J = 9.1 Hz, 1H), 4.38 (s, 2H). [1316] To the crude were added water (2.5 mL), EtOH (2.5 mL) and DMF (0.25 mL). Refluxed the mixture and added Hydrazine hydrate (0.3 mL, 9.7 mmol) and left to reflux overnight. The reaction was cooled and the precipitate was collected by filtration, washed with EtOH (5 mL) and dried under high vacuum to afford compound 3B as a white solid (0.2 g, 60%) 1 H NMR (400 MHz, DMSO) δ 8.22 (d, J = 1.7 Hz, 1H), 8.17 (d, J = 8.5 Hz, 1H), 8.02 (dd, J = 8.5, 1.8 Hz, 1H), 7.90 (dd, J = 6.2, 2.2 Hz, 1H), 7.73 (ddd, J = 7.9, 5.3, 2.3 Hz, 1H), 7.48 (t, J = 9.1 Hz, 1H), 4.38 (s, 2H). Synthesis of Compound 4B 4B [1317] Compound 3B (0.20 g , 0.57mmol) and potassium hydroxide (0.323g, 5.75 mmol) were added to ethanol (2 mL) and water (7 mL) and heated at 100 o C for 4.5 hours. The ethanol was evaporated off and the aqueous was extracted with ethyl acetate (2 x 5mL). The aqueous was then acidified to pH=1 with conc. HCI to form a precipitate which was sonicated, filtered, washed with water and dried under high vacuum to afford a beige solid (0.2 g, 94 % yield). 1 H NMR (400 MHz, DMSO) δ 12.68 (s, 1H), 8.27 – 8.07 (m, 2H), 7.99 (d, J = 8.5 Hz, 1H), 7.86 – 7.71 (m, 1H), 7.62 – 7.48 (m, 1H), 7.24 (t, J = 9.6 Hz, 1H), 4.35 (d, J = 8.2 Hz, 2H). Synthesis of Compound Compound 4B (100 mg, 0.266mmol) and 6-(piperizino)pyridine-3-carbonitrile (50mg, 0.266mmol) were dissolved in 700uL DMF in a small scintillation vile.1-propane phosphoric acid cyclic anhydride (338uL, 0.532mmol) and DIPEA (140uL, 0.798mmol) were added via syringe and the reaction was stirred until complete conversion of the starting material was observed (2.5hours). The reaction was monitored via TLC using 5% MeOH in DCM with 1-2drops glacial acetic acid (to separate the starting material acid from product). The reaction was then dissolved in ethyl acetate (5mL) and washed with saturated NaCO3, ddH2O, and brine. The organic layer was then concentrated to yield 102mg of compound 5B (70% yield) 1 H NMR (400 MHz, DMSO) δ 12.69 (s, 1H), 8.50 (d, J = 2.4 Hz, 1H), 8.16 (dd, J = 5.2, 3.3 Hz, 2H), 7.94 (ddd, J = 45.0, 8.8, 2.1 Hz, 2H), 7.41 (dd, J = 8.5, 4.4 Hz, 2H), 7.26 (t, J = 8.9 Hz, 1H), 6.91 (d, J = 9.1 Hz, 1H), 4.34 (s, 2H), 3.74 (d, J = 10.3 Hz, 4H), 3.61 (s, 2H). Example 10 In a clean, oven dried 15mL round bottom flask, Compound 5B (50mg, 0.092mmol) was co-evaporated with dry toluene (5mL) and allowed to sit under high vacuum for 20 mins. Dry toluene (1mL) was added and argon was bubbled through the solution for 10 mins. Under argon pressure, Palladium Tetrakis (10.4mg, 0.009mmol) and Tributyl (1-propynyl) tin (30.4uL, 0.1mmol) were added and the reaction was refluxed under argon pressure for 3 hours until reaction had reached completion. The crude reaction was purified via dry loading method (1g silica) on a 4g silica manual column. The column was initially washed with 100% ethyl acetate to elute excess triphenyl phosphine, followed by 5%MeOH in ethyl acetate to separate impurities and elute desired compound. Fractions containing desired compound were pooled and concentrated to yield 22mg of Example 10 (47% yield).1H NMR (400 MHz, DMSO) δ 12.61 (s, 1H), 8.49 (s, 1H), 8.18 (d, J = 8.2 Hz, 1H), 7.89 (d, J = 18.6 Hz, 2H), 7.82 – 7.63 (m, 3H), 7.40 (s, 2H), 7.25 (d, J = 8.5 Hz, 1H), 6.90 (d, J = 9.2 Hz, 1H), 4.32 (s, 2H), 4.12 (s, 2H), 3.84 – 3.55 (m, 8H). Equivalents [1318] While the present disclosure has been described in conjunction with the specific embodiments set forth above, many alternatives, modifications and other variations thereof will be apparent to those of ordinary skill in the art. All such alternatives, modifications and variations are intended to fall within the spirit and scope of the present disclosure.