Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HSD17B13 INHIBITORS AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2023/043836
Kind Code:
A1
Abstract:
Described herein are compounds that are HSD17B13 inhibitors, methods of making such compounds, pharmaceutical compositions and medicaments comprising such compounds, and methods of using such compounds in the treatment of conditions, diseases, or disorders associated with HSD17B13 activity.

Inventors:
NAGASAWA JOHNNY Y (US)
BOTROUS IRINY (US)
LAI ANDILIY G (US)
HUDSON ANDREW R (US)
SMITH NICHOLAS D (US)
GOVEK STEVEN P (US)
Application Number:
PCT/US2022/043527
Publication Date:
March 23, 2023
Filing Date:
September 14, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
METACRINE INC (US)
International Classes:
C07D401/10; A61K31/4192; A61K31/444; A61K31/496; A61K31/519; A61K31/5377; A61P1/16; C07D401/14; C07D403/04; C07D403/10; C07D413/04; C07D413/10; C07D471/04; C07D487/04
Domestic Patent References:
WO2019133629A12019-07-04
WO2021003295A12021-01-07
WO2022072512A12022-04-07
WO2022072517A12022-04-07
Attorney, Agent or Firm:
MALECHA, James (US)
Download PDF:
Claims:
CLAIMS WHAT IS CLAIMED IS: 1. A compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof: wherein: X1, X2, and X3 are each independently CR3 or N; Y1 is CR4 or N; Y2 is N(R9), O, S, or C(R4)2; Y3 is CR4 or N; Y4 is C(O), C(R4)2, N(R15), O, or S; Y5 is C(O), C(R4)2, or N(R15), wherein at least one of Y4 and Y5 is C(O); Z1, Z2, Z3, and Z4 are each independently CR5 or N; L1 is selected from a bond, -O-, -N(R10)- -C(O)-, -S-, -S(O)-, -S(O)2-, -C(O)N(R10)-, - N(R10)C(O)-, -C(R10)(R11)N(R10)-, and -N(R10)C(R10)(R11)-; L2 is C1-6alkylene optionally substituted with -OH; R1 is selected from: a) C3-10cycloalkyl and C2-9heterocycloalkyl, wherein C3-10cycloalkyl and C2- 9heterocycloalkyl are optionally substituted with one, two, or three R6; and b) C6-10aryl and C1-9heteroaryl, wherein C6-10aryl and C1-9heteroaryl are optionally substituted with one, two, or three R7; R2 is selected from H, halogen, -CN, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, -SR10, -N(R10)(R11), -C(O)OR10, - OC(O)N(R10)(R11), -N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, - C(O)R13, -S(O)R13, -OC(O)R13, -C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), - N(R12)C(O)R13, -S(O)2R13, -S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), - CH2C(O)N(R10)(R11), -CH2N(R12)C(O)R13, -CH2S(O)2R13, and - CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2- 9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, and - N(R10)(R11); each R3 and each R4 are each independently selected from H, halogen, -CN, C1-6alkyl, C1-6haloalkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6- 10aryl, C1-9heteroaryl, -OR10, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), - N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, -S(O)R13, - OC(O)R13, -C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, - S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), -CH2C(O)N(R10)(R11), - CH2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, and -N(R10)(R11); each R5 is independently selected from H, halogen, hydroxy, -CN, C1-6alkyl, C1- 6haloalkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), - N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, -S(O)R13, - OC(O)R13, -C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, - S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), -CH2C(O)N(R10)(R11), - CH2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, and -N(R10)(R11); each R6 is independently selected from halogen, oxo, -CN, C1-6alkyl, C1-6haloalkyl, C2- 6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, - OR10, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), -N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, -S(O)R13, -OC(O)R13, - C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, - S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), -CH2C(O)N(R10)(R11), - CH2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, -N(R10)(R11), and -C(O)OR10; each R7 are each independently selected from halogen, -CN, C1-6alkyl, C1-6haloalkyl, C2- 6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, - OR10, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), -N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, -S(O)R13, -OC(O)R13, - C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, - S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), -CH2C(O)N(R10)(R11), - CH2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, -N(R10)(R11), and -C(O)OR10; R8 is selected from C2-6alkyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl, wherein C2-6alkyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three R14; R9 is selected from H, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2- 9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, -C(O)R13, -C(O)OR10, -C(O)N(R10)(R11), and -S(O)2R13, wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2- 9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, and - N(R10)(R11); each R10 is independently selected from hydrogen, C1-6alkyl, C1-6 haloalkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl, wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6- 10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C1-6alkyl, C1-6haloalkyl, C1-6alkoxy, C3- 6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl; each R11 is independently selected from hydrogen, C1-6alkyl, and C1-6haloalkyl; each R12 is independently selected from hydrogen, C1-6alkyl, and C1-6haloalkyl; each R13 is independently selected C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2- 9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl, wherein C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, -N(R10)(R11), -C(O)OR10, -N(R12)C(O)R16, C1-6alkyl, C1-6haloalkyl, C1- 6alkoxy, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl; each R14 is independently selected from halogen, oxo, -CN, C1-6alkyl, C1-6haloalkyl, C2- 6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, - OR10, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), -N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, -S(O)R13, -OC(O)R13, - C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, - S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), -CH2C(O)N(R10)(R11), - CH2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C1-6alkyl, C1-6haloalkyl, -OR10, -N(R10)(R11), and -C(O)OR10; R15 is selected from hydrogen, C1-6alkyl, C1-6 haloalkyl, C2-6alkenyl, C2-6alkynyl, C3- 6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, -C(O)R13, -S(O)R13, - S(O)2R13, -C(O)OR10, -S(O)2OR10, -C(O)N(R10)(R11), -S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C1-6alkyl, C1-6haloalkyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6- 10aryl, C1-9heteroaryl, -OR10, -SR10, -N(R10)(R11), -C(O)OR10, -C(O)N(R10)(R11), - S(O)R13, -S(O)2R13, -S(O)2N(R10)(R11), -OC(O)N(R10)(R11), -N(R12)C(O)R13, - N(R12)C(O)OR13, and -N(R12)C(O)N(R10)(R11); and each R16 is independently selected C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2- 9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl. 2. A compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof: wherein: X1, X2, and X3 are each independently CR3 or N; Y1 is CR4 or N; Y2 is N(R9), O, S, or C(R4)2; Y3 is CR4 or N; Y4 is C(O), C(R4)2, N(R15), O, or S; Y5 is C(O), C(R4)2, or N(R15), wherein at least one of Y4 and Y5 is C(O); Z1, Z2, Z3, and Z4 are each independently CR5 or N; L1 is selected from a bond, -O-, -N(R10)- -C(O)-, -S-, -S(O)-, -S(O)2-, -C(O)N(R10)-, - N(R10)C(O)-, -C(R10)(R11)N(R10)-, and -N(R10)C(R10)(R11)-; L2 is C1-6alkylene optionally substituted with -OH; R1 is selected from: a) C3-10cycloalkyl and C2-9heterocycloalkyl, wherein C3-10cycloalkyl and C2- 9heterocycloalkyl are optionally substituted with one, two, or three R6; and b) C6-10aryl and C1-9heteroaryl, wherein C6-10aryl and C1-9heteroaryl are optionally substituted with one, two, or three R7; R2 is selected from H, halogen, -CN, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, -SR10, -N(R10)(R11), -C(O)OR10, - OC(O)N(R10)(R11), -N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, - C(O)R13, -S(O)R13, -OC(O)R13, -C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), - N(R12)C(O)R13, -S(O)2R13, -S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), - CH2C(O)N(R10)(R11), -CH2N(R12)C(O)R13, -CH2S(O)2R13, and - CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2- 9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, and - N(R10)(R11); each R3 and each R4 are each independently selected from H, halogen, -CN, C1-6alkyl, C1-6haloalkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6- 10aryl, C1-9heteroaryl, -OR10, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), - N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, -S(O)R13, - OC(O)R13, -C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, - S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), -CH2C(O)N(R10)(R11), - CH2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, and -N(R10)(R11); each R5 is independently selected from H, halogen, hydroxy, -CN, C1-6alkyl, C1- 6haloalkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), - N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, -S(O)R13, - OC(O)R13, -C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, - S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), -CH2C(O)N(R10)(R11), - CH2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, and -N(R10)(R11); each R6 is independently selected from halogen, oxo, -CN, C1-6alkyl, C1-6haloalkyl, C2- 6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, - OR10, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), -N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, -S(O)R13, -OC(O)R13, - C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, - S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), -CH2C(O)N(R10)(R11), - CH2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, -N(R10)(R11), and -C(O)OR10; each R7 are each independently selected from halogen, -CN, C1-6alkyl, C1-6haloalkyl, C2- 6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, - OR10, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), -N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, -S(O)R13, -OC(O)R13, - C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, - S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), -CH2C(O)N(R10)(R11), - CH2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, -N(R10)(R11), and -C(O)OR10; R8 is selected from C2-6alkyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl, wherein C2-6alkyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three R14; R9 is selected from H, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2- 9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, -C(O)R13, -C(O)OR10, -C(O)N(R10)(R11), and -S(O)2R13, wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2- 9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, and - N(R10)(R11); each R10 is independently selected from hydrogen, C1-6alkyl, C1-6 haloalkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl, wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6- 10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C1-6alkyl, C1-6haloalkyl, C1-6alkoxy, C3- 6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl; each R11 is independently selected from hydrogen, C1-6alkyl, and C1-6haloalkyl; each R12 is independently selected from hydrogen, C1-6alkyl, and C1-6haloalkyl; each R13 is independently selected C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2- 9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl, wherein C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C1-6alkyl, C1-6haloalkyl, C1-6alkoxy, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl; each R14 is independently selected from halogen, oxo, -CN, C1-6alkyl, C1-6haloalkyl, C2- 6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, - OR10, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), -N(R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, -S(O)R13, -OC(O)R13, - C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, - S(O)2N(R10)(R11)-, S(=O)(=NH)N(R10)(R11), -CH2C(O)N(R10)(R11), - CH2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C1-6alkyl, C1-6haloalkyl, -OR10, -N(R10)(R11), and -C(O)OR10; and R15 is selected from hydrogen, C1-6alkyl, C1-6 haloalkyl, C2-6alkenyl, C2-6alkynyl, C3- 6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, C1-9heteroaryl, -C(O)R13, -S(O)R13, - S(O)2R13, -C(O)OR10, -S(O)2OR10, -C(O)N(R10)(R11), -S(O)2N(R10)(R11), wherein C1-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C1-6alkyl, C1-6haloalkyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6- 10aryl, C1-9heteroaryl, -OR10, -SR10, -N(R10)(R11), -C(O)OR10, -C(O)N(R10)(R11), - S(O)R13, -S(O)2R13, -S(O)2N(R10)(R11), -OC(O)N(R10)(R11), -N(R12)C(O)R13, - N(R12)C(O)OR13, and -N(R12)C(O)N(R10)(R11).

3. The compound of claim 1 or claim 2, or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (II’): Formula (II’). 4. The compound of any one of claims 1-3, or a pharmaceutically acceptable salt or solvate thereof, wherein Y2 is N(R9). 5. The compound of claim 4, or a pharmaceutically acceptable salt or solvate thereof, wherein R9 is selected from H and C1-6alkyl. 6. The compound of claim 5, or a pharmaceutically acceptable salt or solvate thereof, wherein R9 is H. 7. The compound of claim 5, or a pharmaceutically acceptable salt or solvate thereof, wherein R9 is C1-6alkyl. 8. The compound of claim 1 or claim 2, or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (III’): Formula (III’). 9. The compound of claim 1 or claim 2, or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IV’): Formula (IV’). 10. The compound of claim 8 or claim 9, or a pharmaceutically acceptable salt or solvate thereof, wherein Y3 is N.

11. The compound of claim 8 or claim 9, or a pharmaceutically acceptable salt or solvate thereof, wherein Y3 is CR4. 12. The compound of claim 11, or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is H, halogen, C1-6alkyl, C1-6haloalkyl, -N(R10)(R11), or -C(O)R13. 13. The compound of claim 12, or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is H, halogen, C1-6alkyl, or -N(R10)(R11). 14. The compound of any one of claims 1-13, or a pharmaceutically acceptable salt or solvate thereof, wherein Y1 is N. 15. The compound of any one of claims 1-14, or a pharmaceutically acceptable salt or solvate thereof, wherein X1, X2, and X3 are CR3. 16. The compound of claim 1-7, or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IIa’): Formula (IIa’). 17. The compound of any one of claims 8, 12, and 13, or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IIIa’): Formula (IIIa’). 18. The compound of claim 9, or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IVa’): Formula (IVa’). 19. The compound of any one of claims 1-18, or a pharmaceutically acceptable salt or solvate thereof, wherein Z2 is N. 20. The compound of any one of claims 1-18, or a pharmaceutically acceptable salt or solvate thereof, wherein Z2 is CR5. 21. The compound of any one of claims 1-20, or a pharmaceutically acceptable salt or solvate thereof, wherein Z1 and Z3 are CR5. 22. The compound of any one of claims 1-20, or a pharmaceutically acceptable salt or solvate thereof, wherein Z1 is N; and Z3 are CR5. 23. The compound of any one of claims 1-20, or a pharmaceutically acceptable salt or solvate thereof, wherein Z3 is N; and Z1 is CR5. 24. The compound of any one of claims 1-23, or a pharmaceutically acceptable salt or solvate thereof, wherein Z4 is N. 25. The compound of any one of claims 1-23, or a pharmaceutically acceptable salt or solvate thereof, wherein Z4 is CR5. 26. The compound of any one of claims 1-25, or a pharmaceutically acceptable salt or solvate thereof, wherein each R5 is independently selected from H, halogen, -CN, C1- 6alkyl, and -OR10. 27. The compound of any one of claims 1-26, or a pharmaceutically acceptable salt or solvate thereof, wherein each R5 is H. 28. The compound of any one of claims 1-27, or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is unsubstituted C1-6alkylene. 29. The compound of any one of claims 1-28, or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is -CH2-. 30. The compound of any one of claims 1-29, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is phenyl optionally substituted with one, two, or three R14. 31. The compound of any one of claims 1-30, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is unsubstituted phenyl. 32. The compound of any one of claims 1-29, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is C1-9heteroaryl optionally substituted with one, two, or three R14. 33. The compound of claim 32, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is unsubstituted C1-9heteroaryl.

34. The compound of any one of claims 1-29, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is C2-9heterocycloalkyl optionally substituted with one, two, or three R14. 35. The compound of claim 34, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is unsubstituted C2-9heterocycloalkyl. 36. The compound of any one of claims 1-29, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is C3-6cycloalkyl optionally substituted with one, two, or three R14. 37. The compound of claim 36, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is unsubstituted C3-6cycloalkyl. 38. The compound of any one of claims 1-29, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is C2-6alkyl optionally substituted with one, two, or three R14. 39. The compound of claim 38, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is unsubstituted C2-6alkyl. 40. The compound of any one of claims 1-39, or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is a bond. 41. The compound of any one of claims 1-39, or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is - N(R10)-. 42. The compound of any one of claims 1-41, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C2-9heterocycloalkyl optionally substituted with one, two, or three R6. 43. The compound of any one of claims 1-42, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C2-9heterocycloalkyl selected from piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, and diazepanyl, wherein piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, and diazepanyl are optionally substituted with one, two, or three R6. 44. The compound of any one of claims 1-43, or a pharmaceutically acceptable salt or solvate thereof, wherein -R1-L2-R8 is 45. The compound of any one of claims 1-44, or a pharmaceutically acceptable salt or solvate thereof, wherein each R6 is independently selected from oxo, C1-6alkyl, -OR10, - C(O)OR10, -N(R12)C(O)R13, -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, - C(O)N(R10)(R11), -S(O)2R13, and -S(O)2N(R10)(R11). 46. The compound of any one of claims 1-45, or a pharmaceutically acceptable salt or 47. The compound of any one of claims 1-41, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C3-8cycloalkyl optionally substituted with one, two, or three R6. 48. The compound of any one of claims 1-41, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C6-10aryl substituted with one, two, or three R7. 49. The compound of any one of claims 1-41, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C1-9heteroaryl substituted with one, two, or three R7. 50. The compound of any one of claims 1-49, or a pharmaceutically acceptable salt or solvate thereof, wherein each R3 is independently selected from H, halogen, -CN, C1- 6alkyl, C1-6haloalkyl, and -OR10. 51. The compound of any one of claims 1-50, or a pharmaceutically acceptable salt or solvate thereof, wherein each R3 is independently selected from H, halogen, and C1- 6haloalkyl. 52. The compound of any one of claims 1-51, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is H. 53. The compound of any one of claims 1-51, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is halogen.

54. A compound selected from:

-245-

and or a pharmaceutically acceptable salt or solvate thereof.

55. A compound selected from: or a pharmaceutically acceptable salt or solvate thereof. 56. A pharmaceutical composition comprising a compound of any one of claims 1-55, or a pharmaceutically acceptable salt or solvate thereof, and at least one pharmaceutically acceptable excipient. 57. The pharmaceutical composition of claim 56, wherein the pharmaceutical composition is formulated for administration to a mammal by intravenous administration, subcutaneous administration, oral administration, inhalation, nasal administration, dermal administration, or ophthalmic administration. 58. The pharmaceutical composition of claim 56, wherein the pharmaceutical composition is in the form of a tablet, a pill, a capsule, a liquid, a suspension, a gel, a dispersion, a solution, an emulsion, an ointment, or a lotion. 59. A method of treating or preventing a liver disease or condition in a mammal, comprising administering to the mammal a compound of any one of claims 1-55, or a pharmaceutically acceptable salt or solvate thereof. 60. The method of claim 59, wherein the liver disease or condition is an alcoholic liver disease or condition. 61. The method of claim 59, wherein the liver disease or condition is a nonalcoholic liver disease or condition.

62. The method of claim 59, wherein the liver disease or condition is liver inflammation, fatty liver (steatosis), liver fibrosis, hepatitis, cirrhosis, hepatocellular carcinoma, or combinations thereof. 63. The method of claim 59, wherein the liver disease or condition is primary biliary cirrhosis, primary sclerosing cholangitis, cholestasis, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), or combinations thereof. 64. A method of treating or preventing a disease or condition in a mammal that would benefit from treatment with an HSD17B13 inhibitor, comprising administering to the mammal a compound of any one of claims 1-55, or a pharmaceutically acceptable salt or solvate thereof. 65. The method of claim 64, wherein the disease or condition in the mammal that would benefit from treatment with an HSD17B13 inhibitor mammal is a liver disease or condition as described in claim 62 or claim 63. 66. A method of modulating hydroxysteroid 17β-dehydrogenase 13 (HSD17B13) activity in a mammal comprising administering to the mammal a compound of any one of claims 1- 55, or a pharmaceutically acceptable salt or solvate thereof. 67. The method of claim 66, wherein modulating comprises inhibiting HSD17B13 activity. 68. The method of claim 66 or claim 67, wherein the mammal has a liver disease or condition as described in claim 62 or claim 63.

Description:
HSD17B13 INHIBITORS AND USES THEREOF CROSS-REFERENCE [0001] This application claims the benefit of U.S. Provisional Application Nos.63/244,675 filed on September 15, 2021 and 63/325,045 filed on March 29, 2022, each of which is incorporated by reference in its entirety. FIELD OF THE INVENTION [0002] Described herein are compounds that are hydroxysteroid 17β-dehydrogenase 13 (HSD17B13) inhibitors, methods of making such compounds, pharmaceutical compositions and medicaments comprising such compounds, and methods of using such compounds in the treatment of conditions, diseases, or disorders associated with HSD17B13 activity. BACKGROUND OF THE INVENTION [0003] Hydroxysteroid dehydrogenase 17 β13 (HSD17b13) is a member of the short-chain dehydrogenase/reductase enzymes highly expressed in the liver on lipid droplets. It has been shown to oxidize retinol, steroids such as estradiol, and bio-active lipids like leukotriene B4. Loss of HSD17b13 expression and enzymatic activity is associated with decreased incidence of liver disease. Inhibition of HSD17b13 enzymatic activity can be used for the treatment of liver diseases that result in hepatic inflammation, fibrosis, cirrhosis, and development of hepatocellular carcinoma. SUMMARY OF THE INVENTION [0004] In one aspect, described herein is a compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof: wherein:

X 1 , X 2 , and X 3 are each independently CR 3 or N; Y 1 is CR 4 or N; Y 2 is N(R 9 ), O, S, or C(R 4 ) 2 ; Y 3 is CR 4 or N; Y 4 is C(O), C(R 4 ) 2 , N(R 15 ), O, or S; Y 5 is C(O), C(R 4 ) 2 , or N(R 15 ), wherein at least one of Y 4 and Y 5 is C(O); Z 1 , Z 2 , Z 3 , and Z 4 are each independently CR 5 or N; L 1 is selected from a bond, -O-, -N(R 10 )- -C(O)-, -S-, -S(O)-, -S(O) 2 -, -C(O)N(R 10 )-, - N(R 10 )C(O)-, -C(R 10 )(R 11 )N(R 10 )-, and -N(R 10 )C(R 10 )(R 11 )-; L 2 is C 1-6 alkylene optionally substituted with -OH; R 1 is selected from: a) C 3-10 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-10 cycloalkyl and C 2- 9 heterocycloalkyl are optionally substituted with one, two, or three R 6 ; and b) C 6-10 aryl and C 1-9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 ; R 2 is selected from H, halogen, -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , - OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , - C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), - CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and - CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 3 and each R 4 are each independently selected from H, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 1 0 aryl, C 1-9 heteroaryl , - OR10, -SR10 , -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 5 is independently selected from H, halogen, hydroxy, -CN, C 1-6 alkyl, C 1- 6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 6 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , - C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; each R 7 are each independently selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , - C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - C H2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 8 is selected from C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl, wherein C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three R 14 ; R 9 is selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -C(O)R 13 , -C(O)OR 10 , -C(O)N(R 10 )(R 11 ), and -S(O) 2 R 13 , wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 10 is independently selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 10aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3- 6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 11 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 12 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 13 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, -N(R 10 )(R 11 ), -C(O)OR 10 , -N(R 12 )C(O)R 16 , C 1-6 alkyl, C 1-6 haloalkyl, C 1- 6alkoxy, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 14 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , - C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 15 is selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3- 6cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -C(O)R 13 , -S(O)R 13 , - S(O) 2 R 13 , -C(O)OR 10 , -S(O) 2 OR 10 , -C(O)N(R 10 )(R 11 ), -S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 10aryl, C 1-9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -C(O)N(R 10 )(R 11 ), - S(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 ), -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , - N(R 12 )C(O)OR 13 , and -N(R 12 )C(O)N(R 10 )(R 11 ); and each R 16 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl. [0005] In another aspect, described herein is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof: wherein:

X 1 , X 2 , and X 3 are each independently CR 3 or N; Y 1 is CR 4 or N; Y 2 is N(R 9 ), O, S, or C(R 4 ) 2 ; Y 3 is CR 4 or N; Y 4 is C(O), C(R 4 ) 2 , N(R 15 ), O, or S; Y 5 is C(O), C(R 4 ) 2 , or N(R 15 ), wherein at least one of Y 4 and Y 5 is C(O); Z 1 , Z 2 , Z 3 , and Z 4 are each independently CR 5 or N; L 1 is selected from a bond, -O-, -N(R 10 )- -C(O)-, -S-, -S(O)-, -S(O) 2 -, -C(O)N(R 10 )-, - N(R 10 )C(O)-, -C(R 10 )(R 11 )N(R 10 )-, and -N(R 10 )C(R 10 )(R 11 )-; L 2 is C 1-6 alkylene optionally substituted with -OH; R 1 is selected from: a) C 3-10 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-10 cycloalkyl and C 2- 9 heterocycloalkyl are optionally substituted with one, two, or three R 6 ; and b) C 6-10 aryl and C 1-9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 ; R 2 is selected from H, halogen, -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , - OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , - C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), - CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and - CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 3 and each R 4 are each independently selected from H, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 1 0aryl, C1-9heteroaryl, -OR10, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 5 is independently selected from H, halogen, hydroxy, -CN, C 1-6 alkyl, C 1- 6haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 6 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , - C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; each R 7 are each independently selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , - C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - C H2N(R12)C(O)R13, -CH2S(O)2R13, and -CH2S(O)2N(R10)(R11), wherein C1-6alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 8 is selected from C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl, wherein C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three R 14 ; R 9 is selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -C(O)R 13 , -C(O)OR 10 , -C(O)N(R 10 )(R 11 ), and -S(O) 2 R 13 , wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 10 is independently selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 10aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3- 6cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 11 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 12 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 13 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 14 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , - C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6alkenyl, C2-6alkynyl, C3-6cycloalkyl, C2-9heterocycloalkyl, C6-10aryl, and C1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; and R 15 is selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3- 6cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -C(O)R 13 , -S(O)R 13 , - S(O) 2 R 13 , -C(O)OR 10 , -S(O) 2 OR 10 , -C(O)N(R 10 )(R 11 ), -S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 10aryl, C 1-9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -C(O)N(R 10 )(R 11 ), - S(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 ), -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , - N(R 12 )C(O)OR 13 , and -N(R 12 )C(O)N(R 10 )(R 11 ). [0006] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (II’): [0007] In some embodiments is a compound of Formula (I’), (I), or (II’), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is N(R 9 ). In some embodiments is a compound of Formula (I’), (I), or (II’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 9 is selected from H and C 1-6 alkyl. In some embodiments is a compound of Formula (I’), (I), or (II’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 9 is H. In some embodiments is a compound of Formula (I’), (I), or (II’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 9 is C 1-6 alkyl. [0008] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (III’): [0009] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IV’): [0010] In some embodiments is a compound of Formula (I’), (I), (III’), or (IV’), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is N. In some embodiments is a compound of Formula (I’), (I), (III’), or (IV’), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is CR 4 . In some embodiments is a compound of Formula (I’), (I), (III’), or (IV’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 4 is H, halogen, C 1- 6 alkyl, C 1-6 haloalkyl, -N(R 10 )(R 11 ), or -C(O)R 13 . In some embodiments is a compound of Formula (I’), (I), (III’), or (IV’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 4 is H, halogen, C 1-6 alkyl, or -N(R 10 )(R 11 ). In some embodiments is a compound of Formula (I’), (I), (II’), (III’), or (IV’), a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is N. In some embodiments is a compound of Formula (I’), (I), (II’), (III’), or (IV’), a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are CR 3 . [0011] In some embodiments is a compound of Formula (I’), (I), or (II’), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IIa’): Formula (IIa’). [0012] In some embodiments is a compound of Formula (I’), (I), or (III’), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IIIa’): Formula (IIIa’). [0013] In some embodiments is a compound of Formula (I’), (I), or (IV’), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IVa’): Formula (IVa’). [0014] In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is N. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is CR 5 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 and Z 3 are CR 5 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is N; and Z 3 are CR 5 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is N; and Z 1 is CR 5 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 4 is N. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 4 is CR 5 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 5 is independently selected from H, halogen, -CN, C 1-6 alkyl, and -OR 10 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 5 is H. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein L 2 is unsubstituted C 1- 6 alkylene. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein L 2 is -CH 2 -. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is phenyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted phenyl. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 1-9 heteroaryl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 1- 9 heteroaryl. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 2- 9 heterocycloalkyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 2-9 heterocycloalkyl. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 3-6 cycloalkyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 3-6 cycloalkyl. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 2-6 alkyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 2-6 alkyl. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is a bond. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is - N(R 10 )-. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl selected from piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, and diazepanyl, wherein piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, and diazepanyl are optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from oxo, C 1-6 alkyl, -OR 10 , -C(O)OR 10 , -N(R 12 )C(O)R 13 , - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -C(O)N(R 10 )(R 11 ), -S(O) 2 R 13 , and - S(O) 2 N(R 10 )(R 11 )-. In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 - In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 3-8 cycloalkyl optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 6-10 aryl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 3 is independently selected from H, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, and -OR 10 . In some embodiments is a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 3 is independently selected from H, halogen, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (I), (II), (IIa), (III), or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is H. In some embodiments is a compound of Formula (I), (II), (IIa), (III), or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is halogen. [0015] Any combination of the groups described above for the various variables is contemplated herein. Throughout the specification, groups and substituents thereof are chosen by one skilled in the field to provide stable moieties and compounds. [0016] In one aspect, described herein is a pharmaceutical composition comprising a compound described herein, or a pharmaceutically acceptable salt or solvate thereof, and at least one pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition is formulated for administration to a mammal by intravenous administration, subcutaneous administration, oral administration, inhalation, nasal administration, dermal administration, or ophthalmic administration. In some embodiments, the pharmaceutical composition is formulated for administration to a mammal by intravenous administration, subcutaneous administration, or oral administration. In some embodiments, the pharmaceutical composition is formulated for administration to a mammal by oral administration. In some embodiments, the pharmaceutical composition is in the form of a tablet, a pill, a capsule, a liquid, a suspension, a gel, a dispersion, a solution, an emulsion, an ointment, or a lotion. In some embodiments, the pharmaceutical composition is in the form of a tablet, a pill, or a capsule. [0017] In another aspect, described herein is a method of treating or preventing a liver disease or condition in a mammal, comprising administering to the mammal a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments, the liver disease or condition is an alcoholic liver disease or condition. In some embodiments, the liver disease or condition is a nonalcoholic liver disease or condition. In some embodiments, the liver disease or condition is liver inflammation, fatty liver (steatosis), liver fibrosis, hepatitis, cirrhosis, hepatocellular carcinoma, or combinations thereof. In some embodiments, the liver disease or condition is primary biliary cirrhosis, primary sclerosing cholangitis, cholestasis, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), or combinations thereof. [0018] In another aspect, described herein is a method of treating a disease or condition in a mammal that would benefit from hydroxysteroid 17β-dehydrogenase 13 (HSD17B13) inhibition comprising administering a compound as described herein, or pharmaceutically acceptable salt or solvate thereof, to the mammal in need thereof. In some embodiments, the disease or condition in a mammal that would benefit from HSD17B13 inhibition is liver inflammation, fatty liver (steatosis), liver fibrosis, hepatitis, cirrhosis, hepatocellular carcinoma, or combinations thereof. In some embodiments, the disease or condition in a mammal that would benefit from HSD17B13 inhibition is primary biliary cirrhosis, primary sclerosing cholangitis, cholestasis, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), or combinations thereof. [0019] In another aspect, described herein is a method of modulating hydroxysteroid 17β- dehydrogenase 13 (HSD17B13) activity in a mammal, comprising administering to the mammal a compound of Formula (I’), (I), (II’), (IIa’), (III’), (IIIa’), (IV’), or (IVa’), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments, modulating comprises inhibiting HSD17B13 activity. In some embodiments of a method of modulating HSD17B13 activity in a mammal, the mammal has a liver disease or condition selected from liver inflammation, fatty liver (steatosis), liver fibrosis, hepatitis, cirrhosis, hepatocellular carcinoma, and combinations thereof. In some embodiments of a method of modulating HSD17B13 activity in a mammal, the mammal has a liver disease or condition selected from primary biliary cirrhosis, primary sclerosing cholangitis, cholestasis, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), and combinations thereof. [0020] In any of the aforementioned aspects are further embodiments in which the effective amount of the compound described herein, or a pharmaceutically acceptable salt thereof, is: (a) systemically administered to the mammal; and/or (b) administered orally to the mammal; and/or (c) intravenously administered to the mammal; and/or (d) administered by inhalation; and/or (e) administered by nasal administration; or and/or (f) administered by injection to the mammal; and/or (g) administered topically to the mammal; and/or (h) administered by ophthalmic administration; and/or (i) administered rectally to the mammal; and/or (j) administered non- systemically or locally to the mammal. [0021] In any of the embodiments disclosed herein, the mammal or subject is a human. [0022] In some embodiments, compounds provided herein are administered to a human. [0023] In some embodiments, compounds provided herein are orally administered. [0024] Articles of manufacture, which include packaging material, a compound described herein, or a pharmaceutically acceptable salt thereof, within the packaging material, and a label that indicates that the compound or composition, or pharmaceutically acceptable salt, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, is used for the treatment, prevention or amelioration of one or more symptoms of a disease or condition that would benefit from HSD17B13 inhibition, are provided. [0025] Other objects, features and advantages of the compounds, methods and compositions described herein will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments, are given by way of illustration only, since various changes and modifications within the spirit and scope of the instant disclosure will become apparent to those skilled in the art from this detailed description. DETAILED DESCRIPTION OF THE INVENTION [0026] Hydroxysteroid dehydrogenase 17 β13 (HSD17b13) is a member of the short-chain dehydrogenase/reductase enzymes highly expressed in the liver on lipid droplets (Horiguchi et al Biochem Biophysl Res Comm, 2008, 370, 235). It has been shown to oxidize retinol, steroids such as estradiol, and bio-active lipids like leukotriene B4 (Abul-Husn et al NEJM, 2018, 378, 1096 and Ma et al Hepatology, 2019, 691504). Exosome sequencing analysis of a large patient population identified a minor allele of HSD17b13 (rs72613567:TA) that was associated with reduced odds of developing liver disease (Abul-Husn et al NEJM, 2018, 378, 1096). Relative to subjects with the common HSD17b13 allele (rs72613567:T), subjects with the TA variant have lower serum ALT and AST and lower odds of alcoholic liver disease with or without cirrhosis, nonalcoholic liver disease with or without cirrhosis, and lower odds of hepatocellular carcinoma. Liver pathology analysis reveals that the subjects with the rs72613567:TA allele have decreased odds of having liver pathology analysis classified as NASH vs normal, NASH vs simple steatosis or NASH with fibrosis vs simple steatosis. Liver injury associated with the PNPLA3 rs738409 (p.I148M) is mitigated by the presence of the rs72613567:TA allele of HSD17b13. Additionally hepatic PNPLA3 mRNA expression is decreased in subjects with the rs72613567:TA allele. The rs72613567:TA allele was found to produce a truncated protein which is unable to metabolize substrates such as estradiol, suggesting the hepatic protective effects of the rs72613567:TA allele is due to loss of enzymatic activity. [0027] Patients with NASH have shown elevated expression of hepatic of HSD17b13 mRNA relative to control subject. Further exploration of the role of HSD17b13 in NASH development identified a minor allele rs62305723 that encodes a P260S mutation of HSD17b13 that leads to loss of retinol metabolism and is associated with decreased hepatic ballooning and inflammation (Ma et al Hepatology, 2019, 691504). [0028] HSD17b13 rs72613567:TA minor allele is associated with loss of HSD17b13 protein expression in the liver and protection from nonalcoholic steatohepatitis, ballooning degeneration, lobular inflammation and fibrosis. Transcription analysis shows changes in immune-responsive pathways in subjects with rs72613567:TA relative to the major allele (Pirolat et al JLR, 2019, 60, 176). [0029] Subjects with the rs72613567:TA allele of HSD17b13 are not only found to have lower histological evidence of fibrosis, but decreased hepatic expression of fibrotic genes like TGFb2 and Col3a1. In addition loss of HSD17b13 due to the rs72613567:TA allele has been shown to significantly change the expression of inflammatory gene ALOX5 and decreased plasma IL1b, IL6 and IL-10 (Luukkonen et al, JCI, 2020, 5 e132158). HSD17b13 rs72613567:TA carriers also show increased hepatic phospholipids PC(p16:0/16:0), PE(p16:0/18:1), PC(44:5e), PC(36:2e), PE(34:0), PE(36:3) and PC(34:3) possibly due to decreased phospholipid degradation from a decreased hepatic expression of PLD4. [0030] The HSD17b13 rs72613567:TA allele, that has been shown to lack HSD17b13 enzymatic activity, is associated with decreased odds of developing severe fibrosis in patients with chronic HCV infection (About & Abel, NEJM, 2018, 379, 1875). Conversely the major allele rs72613567:T is associated with increasing the risk of development of fibrosis, cirrhosis and HCC in HCV infected patients with the PNPLA3 rs738409:G allele (De Benedittis et al. Gastroenterol Res Pract, 2020, 2020, 4216451). [0031] The loss of function minor allele HSD17b13 rs72613567:TA reduces the risk of developing cirrhosis and hepatocellular carcinoma, is associated with a lower risk of liver- related mortality in the general population and further in patients with cirrhosis (Gellbert- Kristensen et al, Hepatology, 2020, 71, 56). Loss of HSD17b13 function also protects against development of HCC in subjects with alcoholic liver disease (Yang et al, Hepatology, 2019, 70, 231 and Stickel et al, Hepatology, 2020, 72, 88). [0032] PNPLA3 rs738409:G is associated with increased fibrosis in patients with NAFLD. The minor HSD17b13 rs72613567:TA allele has been shown to counteract the PNPLA3 rs738409:G allele and decrease the prevalence of severe inflammation, ballooning and fibrosis (Seko et al, Liver Int, 2020, 40, 1686). [0033] Loss of HSD17b13 enzymatic activity due to carrying the rs72613567:TA allele may delay the onset of autoimmune hepatitis (Mederacke et al, Aliment Pharmacol Ther, 2020, 00, 1). [0034] HSD17b13 rs72613567:TA allele is associated with decreased fibrosis and cirrhosis in patents with copper induced liver injury from Wilson’s disease (Ferenci et al, 2019, JHEP, 1, 2). Compounds [0035] Compounds described herein, including pharmaceutically acceptable salts, prodrugs, active metabolites and pharmaceutically acceptable solvates thereof, are HSD17B13 inhibitors. [0036] In some embodiments is a compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof: wherein:

X 1 , X 2 , and X 3 are each independently CR 3 or N; Y 1 is CR 4 or N; Y 2 is N(R 9 ), O, S, or C(R 4 ) 2 ; Y 3 is CR 4 or N; Y 4 is C(O), C(R 4 ) 2 , N(R 15 ), O, or S; Y 5 is C(O), C(R 4 ) 2 , or N(R 15 ), wherein at least one of Y 4 and Y 5 is C(O); Z 1 , Z 2 , Z 3 , and Z 4 are each independently CR 5 or N; L 1 is selected from a bond, -O-, -N(R 10 )- -C(O)-, -S-, -S(O)-, -S(O) 2 -, -C(O)N(R 10 )-, - N(R 10 )C(O)-, -C(R 10 )(R 11 )N(R 10 )-, and -N(R 10 )C(R 10 )(R 11 )-; L 2 is C 1-6 alkylene optionally substituted with -OH; R 1 is selected from: a) C 3-10 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-10 cycloalkyl and C 2- 9 heterocycloalkyl are optionally substituted with one, two, or three R 6 ; and b) C 6-10 aryl and C 1-9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 ; R 2 is selected from H, halogen, -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , - OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , - C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), - CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and - CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 3 and each R 4 are each independently selected from H, halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 1 0 aryl, C 1-9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 5 is independently selected from H, halogen, hydroxy, -CN, C 1-6 alkyl, C 1- 6haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9heteroaryl, -SR10, -N(R10)(R11), -C(O)OR10, -OC(O)N(R10)(R11), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 6 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , - C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; each R 7 are each independently selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , - C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 8 is selected from C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl, wherein C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three R 14 ; R 9 is selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -C(O)R 13 , -C(O)OR 10 , -C(O)N(R 10 )(R 11 ), and -S(O) 2 R 13 , wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C6-10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 10 is independently selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 10aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3- 6cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 11 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 12 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 13 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, -N(R 10 )(R 11 ), -C(O)OR 10 , -N(R 12 )C(O)R 16 , C 1-6 alkyl, C 1-6 haloalkyl, C 1- 6alkoxy, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 14 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , - C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 15 is selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3- 6cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -C(O)R 13 , -S(O)R 13 , - S(O) 2 R 13 , -C(O)OR 10 , -S(O) 2 OR 10 , -C(O)N(R 10 )(R 11 ), -S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 10aryl, C 1-9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -C(O)N(R 10 )(R 11 ), - S (O)R13, -S(O)2R13, -S(O)2N(R10)(R11), -OC(O)N(R10)(R11), -N(R12)C(O)R13, - N(R 12 )C(O)OR 13 , and -N(R 12 )C(O)N(R 10 )(R 11 ); and each R 16 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl. [0037] In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof: wherein:

X 1 , X 2 , and X 3 are each independently CR 3 or N; Y 1 is CR 4 or N; Y 2 is N(R 9 ), O, S, or C(R 4 ) 2 ; Y 3 is CR 4 or N; Y 4 is C(O), C(R 4 ) 2 , N(R 15 ), O, or S; Y 5 is C(O), C(R 4 ) 2 , or N(R 15 ), wherein at least one of Y 4 and Y 5 is C(O); Z 1 , Z 2 , Z 3 , and Z 4 are each independently CR 5 or N; L 1 is selected from a bond, -O-, -N(R 10 )- -C(O)-, -S-, -S(O)-, -S(O) 2 -, -C(O)N(R 10 )-, - N(R 10 )C(O)-, -C(R 10 )(R 11 )N(R 10 )-, and -N(R 10 )C(R 10 )(R 11 )-; L 2 is C 1-6 alkylene optionally substituted with -OH; R 1 is selected from: a) C 3-10 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-10 cycloalkyl and C 2- 9 heterocycloalkyl are optionally substituted with one, two, or three R 6 ; and b) C 6-10 aryl and C 1-9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 ; R 2 is selected from H, halogen, -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , - OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , - C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), - CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 1 0 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 3 and each R 4 are each independently selected from H, halogen, -CN, C 1-6 alkyl, C 1- 6haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1- 9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 5 is independently selected from H, halogen, hydroxy, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C (O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, -S(O)2N(R10)(R11)-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 6 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; each R 7 are each independently selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 8 is selected from C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl, wherein C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three R 14 ; R 9 is selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -C(O)R 13 , -C(O)OR 10 , -C(O)N(R 10 )(R 11 ), and -S(O) 2 R 13 , wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R10 is independently selected from hydrogen, C1-6alkyl, C1-6 haloalkyl, C2-6alkenyl, C2- 6alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 11 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 12 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 13 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1- 6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl; each R 14 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , - N(R 10 )(R 11 ), and -C(O)OR 10 ; and R 15 is selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3- 6cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -C(O)R 13 , -S(O)R 13 , - S(O) 2 R 13 , -C(O)OR 10 , -S(O) 2 OR 10 , -C(O)N(R 10 )(R 11 ), -S(O) 2 N(R 10 )(R 11 ), wherein C 1- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -C(O)N(R 10 )(R 11 ), -S(O)R 13 , - S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 ), -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -N(R 12 )C(O)OR 13 , and -N(R 12 )C(O)N(R 10 )(R 11 ). [0038] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein J is In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein J is In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein J is . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein J is . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein J is . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein J is . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein J is . [0039] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is N(R 9 ). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is N(R 9 ) and R 9 is selected from H and C 1-6 alkyl. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is N(R 9 ) and R 9 is H. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is N(R 9 ) and R 9 is C 1-6 alkyl. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is C(R 4 ) 2 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is C(R 4 ) 2 and each R 4 is independently selected from H, halogen, C 1-6 alkyl, and -N(R 10 )(R 11 ). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is C(R 4 ) 2 and each R 4 is independently selected from H and C 1-6 alkyl. [0040] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is N. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is CR 4 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is CR 4 and R 4 is H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, - N(R 10 )(R 11 ), or -C(O)R 13 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is CR 4 and R 4 is H, halogen, C 1- 6 alkyl, or -N(R 10 )(R 11 ). [0041] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is N. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 and R 4 is H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, - N(R 10 )(R 11 ), or -C(O)R 13 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 and R 4 is H, halogen, or C 1-6 alkyl. [0042] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X1, X2, and X3 are each CR3 and each R3 is independently selected from H, halogen, - CN, C 1-6 alkyl, C 1-6 haloalkyl, and -OR 10 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 and each R 3 is independently selected from H, halogen, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 and each R 3 is independently selected from H, halogen, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(F), and X 3 is C(H). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(H). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(F). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(Cl). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(CH 3 ). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(Cl), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(Cl), X 2 is C(H), and X 3 is C(H). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(H), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(H), X 2 is C(H), and X 3 is C(F). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(H), X 2 is C(H), and X 3 is C(Cl). [0043] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is selected from H, halogen, C 1-6 alkyl, C 1- 6haloalkyl, and -OR 10 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is H. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is halogen. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is F. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is Cl. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is C 1-6 alkyl. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is C 1-6 haloalkyl. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is -OR 10 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is -OH. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is -OCH 3 . [0044] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 , Z 2 , and Z 3 are CR 5 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is N; and Z 2 and Z 3 are CR 5 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is N; and Z 1 and Z 3 are CR 5 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is N; and Z 1 and Z 2 are CR 5 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is CR 5 ; and Z 2 and Z 3 are N. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is CR 5 ; and Z 1 and Z 3 are N. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is CR 5 ; and Z 1 and Z 2 are N. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 5 is independently selected from H, halogen, -CN, C 1-6 alkyl, and -OR 10 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 5 is H. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 , Z 2 , and Z 3 are C(H). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is N; and Z 2 and Z 3 are C(H). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is N; and Z 1 and Z 3 are C(H). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is N; and Z 1 and Z 2 are C(H). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is C(H); and Z 2 and Z 3 are N. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z2 is C(H); and Z1 and Z3 are N. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is C(H); and Z 1 and Z 2 are N. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 4 is CR 5 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 4 is CR 5 and R 5 is selected H, halogen, -CN, C 1-6 alkyl, and -OR 10 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 4 is C(H). In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 4 is N. [0045] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is a bond. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -O-. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(R 10 )-. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(H)-. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -C(R 10 )(R 11 )N(R 10 )-. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is - CH 2 N(H)-. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(R 10 )C(R 10 )(R 11 )-. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(H)CH 2 -. [0046] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is selected from C 3-8 cycloalkyl and C 2- 9 heterocycloalkyl, wherein C 3-8 cycloalkyl and C 2-9 heterocycloalkyl are optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2- 9 heterocycloalkyl substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2- 9 heterocycloalkyl unsubstituted with R 6 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2- 9 heterocycloalkyl selected from piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, diazepanyl, 6- azaspiro[2.5]octanyl, 4,7-diazaspiro[2.5]octanyl, 7-oxa-4-azaspiro[2.5]octanyl, 5,8- diazaspiro[3.5]nonanyl, 8-oxa-5-azaspiro[3.5]nonanyl, or 2,6-diazaspiro[3.3]heptanyl, wherein piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, diazepanyl, 6-azaspiro[2.5]octanyl, 4,7- diazaspiro[2.5]octanyl, 7-oxa-4-azaspiro[2.5]octanyl, 5,8-diazaspiro[3.5]nonanyl, 8-oxa-5- azaspiro[3.5]nonanyl, or 2,6-diazaspiro[3.3]heptanyl are optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from C 1-6 alkyl, -OR 10 , -C(O)OR 10 , - N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -C(O)N(R 10 )(R 11 ), -S(O) 2 R 13 , and -S(O) 2 N(R 10 )(R 11 )-. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from -C(O)R 13 and -S(O) 2 R 13 . In some embodiments is a compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is , , In some embodiments is a compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (I’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . [0047] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 3-8 cycloalkyl optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 3-8 cycloalkyl substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 3-8 cycloalkyl unsubstituted with R 6 . [0048] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is selected from C 6-10 aryl and C 1-9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl unsubstituted with R 7 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl selected from pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, and thiadiazolyl, wherein pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, and thiadiazolyl are substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 6-10 aryl optionally substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl optionally substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl unsubstituted with R 7 . [0049] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is , , , , , , In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is [0050] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from oxo, C 1-6 alkyl, -OR 10 , -C(O)OR 10 , -N(R 12 )C(O)R 13 , -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , - C(O)N(R 10 )(R 11 ), -S(O) 2 R 13 , and -S(O) 2 N(R 10 )(R 11 )-. [0051] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is , , In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is [0052] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L 2 is unsubstituted C 1-6 alkylene. In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L 2 is -CH 2 -. [0053] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is phenyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted phenyl. [0054] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 1-9 heteroaryl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 1-9 heteroaryl. [0055] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 3-6 cycloalkyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 3-6 cycloalkyl. [0056] In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 2-6 alkyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (I’) or (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 2-6 alkyl. [0057] In some embodiments is a compound of Formula (II’), or a pharmaceutically acceptable salt or solvate thereof: wherein: X 1 , X 2 , and X 3 are each independently CR 3 or N; Y 1 is CR 4 or N; Y 2 is N(R 9 ), O, S, or C(R 4 ) 2 ; Z 1 , Z 2 , and Z 3 are each independently CR 5 or N; L 1 is selected from a bond, -O-, -N(R 10 )- -C(O)-, -S-, -S(O)-, -S(O) 2 -, -C(O)N(R 10 )-, - N(R 10 )C(O)-, -C(R 10 )(R 11 )N(R 10 )-, and -N(R 10 )C(R 10 )(R 11 )-; L 2 is C 1-6 alkylene optionally substituted with -OH; R 1 is selected from: a) C 3-10 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-10 cycloalkyl and C 2- 9 heterocycloalkyl are optionally substituted with one, two, or three R 6 ; and b) C 6-10 aryl and C 1-9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 ; R 2 is selected from H, halogen, -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , - OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , - C (O)R13, -S(O)R13, -OC(O)R13, -C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), - N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), - CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 10aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 3 and each R 4 are each independently selected from H, halogen, -CN, C 1-6 alkyl, C 1- 6haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1- 9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 5 is independently selected from H, halogen, hydroxy, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 6 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, t wo, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, -N(R10)(R11), and -C(O)OR 10 ; each R 7 are each independently selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 8 is selected from C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl, wherein C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three R 14 ; R 9 is selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -C(O)R 13 , -C(O)OR 10 , -C(O)N(R 10 )(R 11 ), and -S(O) 2 R 13 , wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 10 is independently selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2- 9heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 11 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 12 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 13 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, - N(R 10 )(R 11 ), -C(O)OR 10 , -N(R 12 )C(O)R 16 , C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3- 6cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; e ach R14 is independently selected from halogen, oxo, -CN, C1-6alkyl, C1-6haloalkyl, C2- 6alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , - N(R 10 )(R 11 ), and -C(O)OR 10 ; and each R 16 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl. [0058] In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof: wherein: X 1 , X 2 , and X 3 are each independently CR 3 or N; Y 1 is CR 4 or N; Y 2 is N(R 9 ), O, S, or C(R 4 ) 2 ; Z 1 , Z 2 , and Z 3 are each independently CR 5 or N; L 1 is selected from a bond, -O-, -N(R 10 )- -C(O)-, -S-, -S(O)-, -S(O) 2 -, -C(O)N(R 10 )-, - N(R 10 )C(O)-, -C(R 10 )(R 11 )N(R 10 )-, and -N(R 10 )C(R 10 )(R 11 )-; L 2 is C 1-6 alkylene optionally substituted with -OH; R 1 is selected from: a) C 3-10 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-10 cycloalkyl and C 2- 9 heterocycloalkyl are optionally substituted with one, two, or three R 6 ; and b) C 6-10 aryl and C 1-9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 ; R 2 is selected from H, halogen, -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , - OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , - C (O)R13, -S(O)R13, -OC(O)R13, -C(O)N(R10)(R11), -C(O)C(O)N(R10)(R11), - N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), - CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 10aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 3 and each R 4 are each independently selected from H, halogen, -CN, C 1-6 alkyl, C 1- 6haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1- 9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 5 is independently selected from H, halogen, hydroxy, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 6 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, t wo, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, -N(R10)(R11), and -C(O)OR 10 ; each R 7 are each independently selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 8 is selected from C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl, wherein C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three R 14 ; R 9 is selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -C(O)R 13 , -C(O)OR 10 , -C(O)N(R 10 )(R 11 ), and -S(O) 2 R 13 , wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 10 is independently selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2- 9heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 11 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 12 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 13 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1- 6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl; and e ach R14 is independently selected from halogen, oxo, -CN, C1-6alkyl, C1-6haloalkyl, C2- 6alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , - N(R 10 )(R 11 ), and -C(O)OR 10 . [0059] In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is N(R 9 ). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is N(R 9 ) and R 9 is selected from H and C 1-6 alkyl. In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is N(R 9 ) and R 9 is H. In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is N(R 9 ) and R 9 is C 1-6 alkyl. In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is C(R 4 ) 2 . In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is C(R 4 ) 2 and each R 4 is independently selected from H, halogen, C 1-6 alkyl, and -N(R 10 )(R 11 ). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 2 is C(R 4 ) 2 and each R 4 is independently selected from H and C 1-6 alkyl. [0060] In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is N. In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 . In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 and R 4 is H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, - N(R 10 )(R 11 ), or -C(O)R 13 . In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 and R 4 is H, halogen, or C 1-6 alkyl. [0061] In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 . In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X1, X2, and X3 are each CR3 and each R3 is independently selected from H, halogen, - CN, C 1-6 alkyl, C 1-6 haloalkyl, and -OR 10 . In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 and each R 3 is independently selected from H, halogen, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 and each R 3 is independently selected from H, halogen, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(F), and X 3 is C(H). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(H). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(F). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(Cl). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(CH 3 ). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(Cl), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(Cl), X 2 is C(H), and X 3 is C(H). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(H), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(H), X 2 is C(H), and X 3 is C(F). In some embodiments is a compound of Formula (II’) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(H), X 2 is C(H), and X 3 is C(Cl). [0062] In some embodiments is a compound of Formula (II’), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IIa’): [0063] In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IIa): ( ) [0064] In some embodiments is a compound of Formula (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 9 is selected from H and C 1-6 alkyl. In some embodiments is a compound of Formula (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 9 is C 1-6 alkyl. In some embodiments is a compound of Formula (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 9 is -CH 3 . In some embodiments is a compound of Formula (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 9 is H. [0065] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is selected from H, halogen, C 1- 6 alkyl, C 1-6 haloalkyl, and -OR 10 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is H. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is halogen. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is F. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is Cl. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is C 1-6 alkyl. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is C 1- 6haloalkyl. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is -OR 10 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is -OH. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is -OCH 3 . [0066] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 , Z 2 , and Z 3 are CR 5 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is N; and Z 2 and Z 3 are CR 5 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is N; and Z 1 and Z 3 are CR 5 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is N; and Z 1 and Z 2 are CR 5 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is CR 5 ; and Z 2 and Z 3 are N. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is CR 5 ; and Z 1 and Z 3 are N. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is CR 5 ; and Z 1 and Z 2 are N. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 5 is independently selected from H, halogen, - CN, C 1-6 alkyl, and -OR 10 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 5 is H. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 , Z 2 , and Z 3 are C(H). In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is N; and Z 2 and Z 3 are C(H). In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is N; and Z 1 and Z 3 are C(H). In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is N; and Z 1 and Z 2 are C(H). In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is C(H); and Z 2 and Z 3 are N. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is C(H); and Z 1 and Z 3 are N. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is C(H); and Z 1 and Z 2 are N. [0067] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is a bond. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -O-. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(R 10 )-. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(H)-. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -C(R 10 )(R 11 )N(R 10 )-. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -CH 2 N(H)-. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(R 10 )C(R 10 )(R 11 )-. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(H)CH 2 -. [0068] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is selected from C 3-8 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-8 cycloalkyl and C 2-9 heterocycloalkyl are optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2- 9 heterocycloalkyl optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl unsubstituted with R 6 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl selected from piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, diazepanyl, 6-azaspiro[2.5]octanyl, 4,7-diazaspiro[2.5]octanyl, 7-oxa-4-azaspiro[2.5]octanyl, 5,8-diazaspiro[3.5]nonanyl, 8-oxa-5-azaspiro[3.5]nonanyl, or 2,6- diazaspiro[3.3]heptanyl, wherein piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, diazepanyl, 6- azaspiro[2.5]octanyl, 4,7-diazaspiro[2.5]octanyl, 7-oxa-4-azaspiro[2.5]octanyl, 5,8- diazaspiro[3.5]nonanyl, 8-oxa-5-azaspiro[3.5]nonanyl, or 2,6-diazaspiro[3.3]heptanyl are optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, , , , , , , . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from C 1-6 alkyl, -OR 10 , -C(O)OR 10 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , - C(O)N(R 10 )(R 11 ), -S(O) 2 R 13 , and -S(O) 2 N(R 10 )(R 11 )-. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is , , , or . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (II’) or (IIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from -C(O)R 13 and -S(O) 2 R 13 . In some embodiments is a compound of Formula (II’) or (IIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is , , , , , In some embodiments is a compound of Formula (II’) or (IIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (II’) or (IIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (II’) or (IIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (II’) or (IIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (II’) or (IIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (II’) or (IIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (II’) or (IIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is [0069] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 3-8 cycloalkyl optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 3- 8cycloalkyl substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 3-8 cycloalkyl unsubstituted with R 6 . [0070] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is selected from C 6-10 aryl and C 1- 9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl unsubstituted with R 7 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl selected from pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, and thiadiazolyl, wherein pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, and thiadiazolyl are substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 6-10 aryl optionally substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl optionally substituted w ith one, two, or three R7. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl unsubstituted with R 7 . [0071] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is , In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is . [0072] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from oxo, C 1-6 alkyl, -OR 10 , -C(O)OR 10 , -N(R 12 )C(O)R 13 , -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , - C(O)R 13 , -C(O)N(R 10 )(R 11 ), -S(O) 2 R 13 , and -S(O) 2 N(R 10 )(R 11 )-. [0073] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is , , . [0074] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 2 is unsubstituted C 1-6 alkylene. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 2 is -CH 2 -. [0075] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is phenyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted phenyl. [0076] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 1-9 heteroaryl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 1-9 heteroaryl. [0077] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 3-6 cycloalkyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 3-6 cycloalkyl. [0078] In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 2-6 alkyl optionally substituted with one, two, or three R14. In some embodiments is a compound of Formula (II’), (II), (IIa’) or (IIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 2-6 alkyl. [0079] In some embodiments is a compound of Formula (III’), or a pharmaceutically acceptable salt or solvate thereof: wherein: X 1 , X 2 , and X 3 are each independently CR 3 or N; Y 1 is CR 4 or N; Y 3 is CR 4 or N; Z 1 , Z 2 , and Z 3 are each independently CR 5 or N; L 1 is selected from a bond, -O-, -N(R 10 )- -C(O)-, -S-, -S(O)-, -S(O) 2 -, -C(O)N(R 10 )-, - N(R 10 )C(O)-, -C(R 10 )(R 11 )N(R 10 )-, and -N(R 10 )C(R 10 )(R 11 )-; L 2 is C 1-6 alkylene optionally substituted with -OH; R 1 is selected from: a) C 3-10 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-10 cycloalkyl and C 2- 9 heterocycloalkyl are optionally substituted with one, two, or three R 6 ; and b) C 6-10 aryl and C 1-9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 ; R 2 is selected from H, halogen, -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , - OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , - C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), - CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 3 and each R 4 are each independently selected from H, halogen, -CN, C 1-6 alkyl, C 1- 6haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1- 9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), - N (R12)C(O)N(R10)(R11), -N(R12)C(O)OR13, -N(R12)S(O)2R13, -C(O)R13, -S(O)R13, - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 5 is independently selected from H, halogen, hydroxy, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 6 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; each R 7 are each independently selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9heterocycloalkyl, C6-10aryl, and C1-9heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 8 is selected from C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl, wherein C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three R 14 ; each R 10 is independently selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 11 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 12 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 13 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2- 6alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, - N(R 10 )(R 11 ), -C(O)OR 10 , -N(R 12 )C(O)R 16 , C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3- 6cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 14 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , - N(R 10 )(R 11 ), and -C(O)OR 10 ; and each R 16 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl. [0080] In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof: wherein: X 1 , X 2 , and X 3 are each independently CR 3 or N; Y 1 is CR 4 or N; Y 3 is CR 4 or N; Z 1 , Z 2 , and Z 3 are each independently CR 5 or N; L 1 is selected from a bond, -O-, -N(R 10 )- -C(O)-, -S-, -S(O)-, -S(O) 2 -, -C(O)N(R 10 )-, - N(R 10 )C(O)-, -C(R 10 )(R 11 )N(R 10 )-, and -N(R 10 )C(R 10 )(R 11 )-; L 2 is C 1-6 alkylene optionally substituted with -OH; R 1 is selected from: a) C 3-10 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-10 cycloalkyl and C 2- 9 heterocycloalkyl are optionally substituted with one, two, or three R 6 ; and b) C 6-10 aryl and C 1-9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 ; R 2 is selected from H, halogen, -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , - OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , - C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), - CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6- 1 0 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 3 and each R 4 are each independently selected from H, halogen, -CN, C 1-6 alkyl, C 1- 6haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1- 9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl a re optionally substituted with one, two, or three groups selected from halogen, C1-6alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 5 is independently selected from H, halogen, hydroxy, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 6 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; each R 7 are each independently selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 8 is selected from C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl, wherein C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three R14; each R 10 is independently selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 11 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 12 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 13 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1- 6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl; and each R 14 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , - N(R 10 )(R 11 ), and -C(O)OR 10 . [0081] In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is N. In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 . In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 and R 4 is H, halogen, C 1-6 alkyl, C 1- 6haloalkyl, -N(R 10 )(R 11 ), or -C(O)R 13 . In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 and R 4 is H, halogen, or C 1-6 alkyl. [0082] In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y3 is N. In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is CR 4 . In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is CR 4 and R 4 is H, halogen, C 1-6 alkyl, C 1- 6haloalkyl, -N(R 10 )(R 11 ), or -C(O)R 13 . In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is CR 4 and R 4 is H, halogen, C 1-6 alkyl, or -N(R 10 )(R 11 ). [0083] In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 . In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 and each R 3 is independently selected from H, halogen, - CN, C 1-6 alkyl, C 1-6 haloalkyl, and -OR 10 . In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 and each R 3 is independently selected from H, halogen, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 and each R 3 is independently selected from H, halogen, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(F), and X 3 is C(H). In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(H). In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(F). In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(Cl). In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(CH 3 ). In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(Cl), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(Cl), X 2 is C(H), and X 3 is C(H). In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(H), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is C(H), X2 is C(H), and X3 is C(F). In some embodiments is a compound of Formula (III’) or (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(H), X 2 is C(H), and X 3 is C(Cl). [0084] In some embodiments is a compound of Formula (III’), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IIIa’): ( ) [0085] In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IIIa): ( ) [0086] In some embodiments is a compound of Formula (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 3 is independently selected from H, halogen, - CN, C 1-6 alkyl, C 1-6 haloalkyl, and -OR 10 . In some embodiments is a compound of Formula (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 3 is independently selected from H, halogen, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 3 is independently selected from H, halogen, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 4 is H, halogen, C 1-6 alkyl, C 1-6 haloalkyl, -N(R 10 )(R 11 ), or -C(O)R 13 . In some embodiments is a compound of Formula (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 4 is H, halogen, C 1-6 alkyl, or -N(R 10 )(R 11 ). In some embodiments is a compound of Formula (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 4 is H. In some embodiments is a compound of Formula (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 4 is halogen. In some embodiments is a compound of Formula (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 4 is C 1-6 alkyl. In some embodiments is a compound of Formula (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is -CH3. In some embodiments is a compound of Formula (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 4 is -N(H)CH 3 . [0087] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is selected from H, halogen, C 1- 6 alkyl, C 1-6 haloalkyl, and -OR 10 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is H. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is halogen. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is F. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is Cl. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is C 1-6 alkyl. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is C 1- 6haloalkyl. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is -OR 10 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is -OH. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is - OCH 3 . [0088] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 , Z 2 , and Z 3 are CR 5 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is N; and Z 2 and Z 3 are CR 5 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is N; and Z 1 and Z 3 are CR 5 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is N; and Z 1 and Z 2 are CR 5 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is CR 5 ; and Z 2 and Z 3 are N. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is CR 5 ; and Z 1 and Z 3 are N. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is CR 5 ; and Z 1 and Z 2 are N. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 5 is independently selected from H, halogen, - CN, C 1-6 alkyl, and -OR 10 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 5 is H. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 , Z 2 , and Z 3 are C(H). In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is N; and Z 2 and Z 3 are C(H). In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is N; and Z 1 and Z 3 are C(H). In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is N; and Z 1 and Z 2 are C(H). In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is C(H); and Z 2 and Z 3 are N. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is C(H); and Z 1 and Z 3 are N. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is C(H); and Z 1 and Z 2 are N. [0089] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is a bond. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -O-. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(R 10 )-. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(H)-. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -C(R 10 )(R 11 )N(R 10 )-. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -CH 2 N(H)-. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is - N(R 10 )C(R 10 )(R 11 )-. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(H)CH 2 -. [0090] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is selected from C 3-8 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-8 cycloalkyl and C 2-9 heterocycloalkyl are optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2- 9 heterocycloalkyl optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl unsubstituted with R 6 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl selected from piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, diazepanyl, 6-azaspiro[2.5]octanyl, 4,7-diazaspiro[2.5]octanyl, 7-oxa-4-azaspiro[2.5]octanyl, 5,8-diazaspiro[3.5]nonanyl, 8-oxa-5-azaspiro[3.5]nonanyl, or 2,6- diazaspiro[3.3]heptanyl, wherein piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, diazepanyl, 6- azaspiro[2.5]octanyl, 4,7-diazaspiro[2.5]octanyl, 7-oxa-4-azaspiro[2.5]octanyl, 5,8- diazaspiro[3.5]nonanyl, 8-oxa-5-azaspiro[3.5]nonanyl, or 2,6-diazaspiro[3.3]heptanyl are optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, , , , , , , In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from C 1-6 alkyl, -OR 10 , -C(O)OR 10 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , - C(O)N(R 10 )(R 11 ), -S(O) 2 R 13 , and -S(O) 2 N(R 10 )(R 11 )-. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (III’) or (IIIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from -C(O)R 13 and -S(O) 2 R 13 . In some embodiments is a compound of Formula (III’) or (IIIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (III’) or (IIIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (III’) or (IIIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (III’) or (IIIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (III’) or (IIIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (III’) or (IIIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (III’) or (IIIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (III’) or (IIIa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is [0091] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 3-8 cycloalkyl optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C 3- 8 cycloalkyl substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 3-8 cycloalkyl unsubstituted with R 6 . [0092] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is selected from C 6-10 aryl and C 1- 9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1- 9 heteroaryl unsubstituted with R 7 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1- 9 heteroaryl selected from pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, and thiadiazolyl, wherein pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, and thiadiazolyl are substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 6-10 aryl optionally substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl optionally substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl unsubstituted with R 7 . [0093] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is , , , In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is , , , , , . [0094] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from oxo, C 1-6 alkyl, -OR 10 , -C(O)OR 10 , -N(R 12 )C(O)R 13 , -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , - C(O)R 13 , -C(O)N(R 10 )(R 11 ), -S(O) 2 R 13 , and -S(O) 2 N(R 10 )(R 11 )-. [0095] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is , , [0096] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 2 is unsubstituted C 1-6 alkylene. In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 2 is -CH 2 -. [0097] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is phenyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted phenyl. [0098] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 1-9 heteroaryl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 1-9 heteroaryl. [0099] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 3-6 cycloalkyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 3-6 cycloalkyl. [00100] In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 2-6 alkyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (III’), (III), (IIIa’) or (IIIa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 2-6 alkyl. [00101] In some embodiments is a compound of Formula (IV’), or a pharmaceutically acceptable salt or solvate thereof: wherein: X 1 , X 2 , and X 3 are each independently CR 3 or N; Y 1 is CR 4 or N; Y 3 is CR 4 or N; Z 1 , Z 2 , Z 3 , and Z 4 are each independently CR 5 or N; L 1 is selected from a bond, -O-, -N(R 10 )- -C(O)-, -S-, -S(O)-, -S(O) 2 -, -C(O)N(R 10 )-, - N(R 10 )C(O)-, -C(R 10 )(R 11 )N(R 10 )-, and -N(R 10 )C(R 10 )(R 11 )-; L 2 is C 1-6 alkylene optionally substituted with -OH; R 1 is selected from: a) C 3-10 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-10 cycloalkyl and C 2- 9 heterocycloalkyl are optionally substituted with one, two, or three R 6 ; and b) C 6-10 aryl and C 1-9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 ; each R 3 and each R 4 are each independently selected from H, halogen, -CN, C 1-6 alkyl, C 1- 6haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1- 9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 5 is independently selected from H, halogen, hydroxy, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, t wo, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, and - N(R 10 )(R 11 ); each R 6 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; each R 7 are each independently selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 8 is selected from C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl, wherein C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9heteroaryl are optionally substituted with one, two, or three R 14 ; each R 10 is independently selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 11 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 12 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 13 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C6-10aryl, and C1-9heteroaryl, wherein C1-6alkyl, C2-6alkenyl, C2- 6alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, - N(R 10 )(R 11 ), -C(O)OR 10 , -N(R 12 )C(O)R 16 , C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3- 6cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 14 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , - N(R 10 )(R 11 ), and -C(O)OR 10 ; and each R 16 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl. [00102] In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof: wherein: X 1 , X 2 , and X 3 are each independently CR 3 or N; Y 1 is CR 4 or N; Y 3 is CR 4 or N; Z 1 , Z 2 , Z 3 , and Z 4 are each independently CR 5 or N; L 1 is selected from a bond, -O-, -N(R 10 )- -C(O)-, -S-, -S(O)-, -S(O) 2 -, -C(O)N(R 10 )-, - N(R 10 )C(O)-, -C(R 10 )(R 11 )N(R 10 )-, and -N(R 10 )C(R 10 )(R 11 )-; L 2 is C 1-6 alkylene optionally substituted with -OH; R 1 is selected from: a) C 3-10 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-10 cycloalkyl and C 2- 9 heterocycloalkyl are optionally substituted with one, two, or three R 6 ; and b) C 6-10 aryl and C 1-9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 ; each R 3 and each R 4 are each independently selected from H, halogen, -CN, C 1-6 alkyl, C 1- 6haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1- 9 heteroaryl, -OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), - N(R 12 )C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , - OC(O)R 13 , -C(O)N(R 10 )(R 11 ), -C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , - S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), - CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and -N(R 10 )(R 11 ); each R 5 is independently selected from H, halogen, hydroxy, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , and - N(R 10 )(R 11 ); each R 6 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, t wo, or three groups selected from halogen, C1-6alkyl, C1-6haloalkyl, -OR10, -N(R10)(R11), and -C(O)OR 10 ; each R 7 are each independently selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C(O)C(O)N(R 10 )(R 11 ), -N(R 12 )C(O)R 13 , -S(O) 2 R 13 , -S(O) 2 N(R 10 )(R 11 )-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , -N(R 10 )(R 11 ), and -C(O)OR 10 ; R 8 is selected from C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl, wherein C 2-6 alkyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three R 14 ; each R 10 is independently selected from hydrogen, C 1-6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2- 9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl; each R 11 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 12 is independently selected from hydrogen, C 1-6 alkyl, and C 1-6 haloalkyl; each R 13 is independently selected C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2- 9heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl, wherein C 1-6 alkyl, C 2-6 alkenyl, C 2- 6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, hydroxy, C 1- 6 alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1- 9 heteroaryl; and each R 14 is independently selected from halogen, oxo, -CN, C 1-6 alkyl, C 1-6 haloalkyl, C 2- 6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, C 1-9 heteroaryl, - OR 10 , -SR 10 , -N(R 10 )(R 11 ), -C(O)OR 10 , -OC(O)N(R 10 )(R 11 ), -N(R 12 )C(O)N(R 10 )(R 11 ), - N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , -S(O)R 13 , -OC(O)R 13 , -C(O)N(R 10 )(R 11 ), - C (O)C(O)N(R10)(R11), -N(R12)C(O)R13, -S(O)2R13, -S(O)2N(R10)(R11)-, S(=O)(=NH)N(R 10 )(R 11 ), -CH 2 C(O)N(R 10 )(R 11 ), -CH 2 N(R 12 )C(O)R 13 , -CH 2 S(O) 2 R 13 , and -CH 2 S(O) 2 N(R 10 )(R 11 ), wherein C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, C 2-9 heterocycloalkyl, C 6-10 aryl, and C 1-9 heteroaryl are optionally substituted with one, two, or three groups selected from halogen, -CN, C 1-6 alkyl, C 1-6 haloalkyl, -OR 10 , - N(R 10 )(R 11 ), and -C(O)OR 10 . [00103] In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is N. In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is CR 4 . In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is CR 4 and R 4 is H, halogen, C 1-6 alkyl, C 1- 6haloalkyl, -N(R 10 )(R 11 ), or -C(O)R 13 . In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 3 is CR 4 and R 4 is H, halogen, C 1-6 alkyl, or -N(R 10 )(R 11 ). [00104] In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is N. In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 . In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 and R 4 is H, halogen, C 1-6 alkyl, C 1- 6haloalkyl, -N(R 10 )(R 11 ), or -C(O)R 13 . In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y 1 is CR 4 and R 4 is H, halogen, or C 1-6 alkyl. [00105] In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 . In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 and each R 3 is independently selected from H, halogen, - CN, C 1-6 alkyl, C 1-6 haloalkyl, and -OR 10 . In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 and each R 3 is independently selected from H, halogen, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 , X 2 , and X 3 are each CR 3 and each R 3 is independently selected from H, halogen, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is C(F), X2 is C(F), and X3 is C(H). In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(H). In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(F). In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(Cl). In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(F), X 2 is C(H), and X 3 is C(CH 3 ). In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(Cl), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(Cl), X 2 is C(H), and X 3 is C(H). In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(H), X 2 is C(H), and X 3 is C(CF 3 ). In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(H), X 2 is C(H), and X 3 is C(F). In some embodiments is a compound of Formula (IV’) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X 1 is C(H), X 2 is C(H), and X 3 is C(Cl). [00106] In some embodiments is a compound of Formula (IV’), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IVa’):

[00107] In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (IVa): [00108] In some embodiments is a compound of Formula (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 3 is independently selected from H, halogen, - CN, C 1-6 alkyl, C 1-6 haloalkyl, and -OR 10 . In some embodiments is a compound of Formula (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 3 is independently selected from H, halogen, C 1-6 alkyl, and C 1-6 haloalkyl. In some embodiments is a compound of Formula (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 3 is independently selected from H, halogen, and C 1-6 haloalkyl. [00109] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 , Z 2 , and Z 3 are CR 5 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is N; and Z 2 and Z 3 are CR 5 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is N; and Z 1 and Z 3 are CR 5 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is N; and Z 1 and Z 2 are CR 5 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is CR 5 ; and Z 2 and Z 3 are N. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is CR 5 ; and Z 1 and Z 3 are N. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is CR 5 ; and Z 1 and Z 2 are N. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 5 is independently selected from H, halogen, - CN, C 1-6 alkyl, and -OR 10 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 5 is H. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 , Z 2 , and Z 3 are C(H). In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is N; and Z 2 and Z 3 are C(H). In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is N; and Z 1 and Z 3 are C(H). In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is N; and Z 1 and Z 2 are C(H). In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 1 is C(H); and Z 2 and Z 3 are N. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 2 is C(H); and Z 1 and Z 3 are N. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 3 is C(H); and Z 1 and Z 2 are N. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 4 is CR 5 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 4 is CR 5 and R 5 is selected H, halogen, -CN, C 1-6 alkyl, and -OR 10 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 4 is C(H). In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein Z 4 is N. [00110] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is a bond. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -O-. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(R 10 )-. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(H)-. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -C(R 10 )(R 11 )N(R 10 )-. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -CH 2 N(H)-. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is - N(R 10 )C(R 10 )(R 11 )-. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 1 is -N(H)CH 2 -. [00111] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is selected from C 3-8 cycloalkyl and C 2-9 heterocycloalkyl, wherein C 3-8 cycloalkyl and C 2-9 heterocycloalkyl are optionally substituted with one, two, or three R6. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2- 9 heterocycloalkyl optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl unsubstituted with R 6 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 2-9 heterocycloalkyl selected from piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, diazepanyl, 6-azaspiro[2.5]octanyl, 4,7-diazaspiro[2.5]octanyl, 7-oxa-4-azaspiro[2.5]octanyl, 5,8-diazaspiro[3.5]nonanyl, 8-oxa-5-azaspiro[3.5]nonanyl, or 2,6- diazaspiro[3.3]heptanyl, wherein piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, tetrahydrofuranyl, pyrrolidinyl, oxetanyl, azetidinyl, aziridinyl, azepanyl, diazepanyl, 6- azaspiro[2.5]octanyl, 4,7-diazaspiro[2.5]octanyl, 7-oxa-4-azaspiro[2.5]octanyl, 5,8- diazaspiro[3.5]nonanyl, 8-oxa-5-azaspiro[3.5]nonanyl, or 2,6-diazaspiro[3.3]heptanyl are optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, , In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from C 1-6 alkyl, -OR 10 , -C(O)OR 10 , -N(R 12 )S(O) 2 R 13 , -C(O)R 13 , - C(O)N(R 10 )(R 11 ), -S(O) 2 R 13 , and -S(O) 2 N(R 10 )(R 11 )-. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is , , In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (IV’) or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from -C(O)R 13 and -S(O) 2 R 13 . In some embodiments is a compound of Formula (IV’) or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is , , In some embodiments is a compound of Formula (IV’) or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (IV’) or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is . In some embodiments is a compound of Formula (IV’) or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (IV’) or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (IV’) or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (IV’) or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is In some embodiments is a compound of Formula (IV’) or (IVa’), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is [00112] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 3-8 cycloalkyl optionally substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (IV’), ( IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C3- 8cycloalkyl substituted with one, two, or three R 6 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 3-8 cycloalkyl unsubstituted with R 6 . [00113] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is selected from C 6-10 aryl and C 1- 9 heteroaryl, wherein C 6-10 aryl and C 1-9 heteroaryl are optionally substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1-9 heteroaryl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1- 9 heteroaryl unsubstituted with R 7 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 1- 9 heteroaryl selected from pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, and thiadiazolyl, wherein pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, tetrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, and thiadiazolyl are substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is C 6-10 aryl optionally substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl optionally substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl substituted with one, two, or three R 7 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is phenyl unsubstituted with R 7 . [00114] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is , , In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is , , , , , . [00115] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein each R 6 is independently selected from oxo, C 1-6 alkyl, -OR 10 , -C(O)OR 10 , -N(R 12 )C(O)R 13 , -N(R 12 )C(O)OR 13 , -N(R 12 )S(O) 2 R 13 , - C(O)R 13 , -C(O)N(R 10 )(R 11 ), -S(O) 2 R 13 , and -S(O) 2 N(R 10 )(R 11 )-. [00116] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is , , , , In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein -R 1 -L 2 -R 8 is , , . [00117] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 2 is unsubstituted C 1-6 alkylene. In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein L 2 is -CH 2 -. [00118] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is phenyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted phenyl. [00119] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 1-9 heteroaryl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 1-9 heteroaryl. [00120] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 3-6 cycloalkyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 3-6 cycloalkyl. [00121] In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is C 2-6 alkyl optionally substituted with one, two, or three R 14 . In some embodiments is a compound of Formula (IV’), (IV), (IVa’) or (IVa), or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 is unsubstituted C 2-6 alkyl. [00122] Any combination of the groups described above for the various variables is contemplated herein. Throughout the specification, groups and substituents thereof are chosen by one skilled in the field to provide stable moieties and compounds. [00123] In some embodiments, compounds described herein include, but are not limited to, those described in Table 1. TABLE 1

[00124] In some embodiments, provided herein is a pharmaceutically acceptable salt or solvate of a compound that is described in Table 1. [00125] In some embodiments, compounds described herein include, but are not limited to, those described in Table 2. TABLE 2

[00126] In some embodiments, provided herein is a pharmaceutically acceptable salt or solvate of a compound that is described in Table 2. [00127] In another aspect, compounds described herein are in the form of pharmaceutically acceptable salts. As well, active metabolites of these compounds having the same type of activity are included in the scope of the present disclosure. In addition, the compounds described herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein. [00128] “Pharmaceutically acceptable,” as used herein, refers a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively nontoxic, i.e., the material is administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained. [00129] The term “pharmaceutically acceptable salt” refers to a form of a therapeutically active agent that consists of a cationic form of the therapeutically active agent in combination with a suitable anion, or in alternative embodiments, an anionic form of the therapeutically active agent in combination with a suitable cation. Handbook of Pharmaceutical Salts: Properties, Selection and Use. International Union of Pure and Applied Chemistry, Wiley-VCH 2002. S.M. Berge, L.D. Bighley, D.C. Monkhouse, J. Pharm. Sci.1977, 66, 1-19. P. H. Stahl and C. G. Wermuth, editors, Handbook of Pharmaceutical Salts: Properties, Selection and Use, Weinheim/Zürich: Wiley-VCH/VHCA, 2002. Pharmaceutical salts typically are more soluble and more rapidly soluble in stomach and intestinal fluids than non-ionic species and so are useful in solid dosage forms. Furthermore, because their solubility often is a function of pH, selective dissolution in one or another part of the digestive tract is possible, and this capability can be manipulated as one aspect of delayed and sustained release behaviors. Also, because the salt-forming molecule can be in equilibrium with a neutral form, passage through biological membranes can be adjusted. [00130] In some embodiments, pharmaceutically acceptable salts are obtained by reacting a compound described herein with an acid to provide a "pharmaceutically acceptable acid addition salt." In some embodiments, the compound described herein (i.e. free base form) is basic and is reacted with an organic acid or an inorganic acid. Inorganic acids include, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and metaphosphoric acid. Organic acids include, but are not limited to, 1-hydroxy-2-naphthoic acid; 2,2-dichloroacetic acid; 2-hydroxyethanesulfonic acid; 2-oxoglutaric acid; 4-acetamidobenzoic acid; 4-aminosalicylic acid; acetic acid; adipic acid; ascorbic acid (L); aspartic acid (L); benzenesulfonic acid; benzoic acid; camphoric acid (+); camphor-10-sulfonic acid (+); capric acid (decanoic acid); caproic acid (hexanoic acid); caprylic acid (octanoic acid); carbonic acid; cinnamic acid; citric acid; cyclamic acid; dodecylsulfuric acid; ethane-1,2-disulfonic acid; ethanesulfonic acid; formic acid; fumaric acid; galactaric acid; gentisic acid; glucoheptonic acid (D); gluconic acid (D); glucuronic acid (D); glutamic acid; glutaric acid; glycerophosphoric acid; glycolic acid; hippuric acid; isobutyric acid; lactic acid (DL); lactobionic acid; lauric acid; maleic acid; malic acid (- L); malonic acid; mandelic acid (DL); methanesulfonic acid; monomethyl fumarate, naphthalene-1,5-disulfonic acid; naphthalene-2-sulfonic acid; nicotinic acid; oleic acid; oxalic acid; palmitic acid; pamoic acid; phosphoric acid; proprionic acid; pyroglutamic acid (- L); salicylic acid; sebacic acid; stearic acid; succinic acid; sulfuric acid; tartaric acid (+ L); thiocyanic acid; toluenesulfonic acid (p); and undecylenic acid. [00131] In some embodiments, a compound described herein is prepared as a chloride salt, sulfate salt, bromide salt, mesylate salt, maleate salt, citrate salt or phosphate salt. [00132] In some embodiments, pharmaceutically acceptable salts are obtained by reacting a compound described herein with a base to provide a "pharmaceutically acceptable base addition salt." [00133] In some embodiments, the compound described herein is acidic and is reacted with a base. In such situations, an acidic proton of the compound described herein is replaced by a metal ion, e.g., lithium, sodium, potassium, magnesium, calcium, or an aluminum ion. In some cases, compounds described herein coordinate with an organic base, such as, but not limited to, ethanolamine, diethanolamine, triethanolamine, tromethamine, meglumine, N-methylglucamine, dicyclohexylamine, tris(hydroxymethyl)methylamine. In other cases, compounds described herein form salts with amino acids such as, but not limited to, arginine, lysine, and the like. Acceptable inorganic bases used to form salts with compounds that include an acidic proton, include, but are not limited to, aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, potassium carbonate, sodium hydroxide, lithium hydroxide, and the like. In some embodiments, the compounds provided herein are prepared as a sodium salt, calcium salt, potassium salt, magnesium salt, meglumine salt, N-methylglucamine salt or ammonium salt. [00134] It should be understood that a reference to a pharmaceutically acceptable salt includes the solvent addition forms. In some embodiments, solvates contain either stoichiometric or non- stoichiometric amounts of a solvent, and are formed during the process of isolating or purifying the compound with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Solvates of compounds described herein are conveniently prepared or formed during the processes described herein. In addition, the compounds provided herein optionally exist in unsolvated as well as solvated forms. [00135] The methods and formulations described herein include the use of N-oxides (if appropriate), crystalline forms (also known as polymorphs), or pharmaceutically acceptable salts of compounds described herein, as well as active metabolites of these compounds having the same type of activity. [00136] In some embodiments, sites on the organic groups (e.g., alkyl groups, aromatic rings) of compounds described herein are susceptible to various metabolic reactions. Incorporation of appropriate substituents on the organic groups will reduce, minimize or eliminate this metabolic pathway. In specific embodiments, the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a halogen, deuterium, an alkyl group, a haloalkyl group, or a deuteroalkyl group. [00137] In another embodiment, the compounds described herein are labeled isotopically (e.g., with a radioisotope) or by another other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels. [00138] Compounds described herein include isotopically-labeled compounds, which are identical to those recited in the various formulae and structures presented 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 the present compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine and chlorine, such as, for example, 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 35 S, 18 F, 36 Cl. In one aspect, isotopically-labeled compounds described herein, for example those into which radioactive isotopes such as 3 H and 14 C are incorporated, are useful in drug and/or substrate tissue distribution assays. In one aspect, substitution with isotopes such as deuterium affords certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements. In some embodiments, one or more hydrogen atoms of the compounds described herein is replaced with deuterium. [00139] In some embodiments, the compounds described herein possess one or more stereocenters and each stereocenter exists independently in either the R or S configuration. The compounds presented herein include all diastereomeric, enantiomeric, atropisomers, and epimeric forms as well as the appropriate mixtures thereof. The compounds and methods provided herein include all cis, trans, syn, anti, entgegen (E), and zusammen (Z) isomers as well as the appropriate mixtures thereof. [00140] Individual stereoisomers are obtained, if desired, by methods such as, stereoselective synthesis and/or the separation of stereoisomers by chiral chromatographic columns. In certain embodiments, compounds described herein are prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds/salts, separating the diastereomers and recovering the optically pure enantiomers. In some embodiments, resolution of enantiomers is carried out using covalent diastereomeric derivatives of the compounds described herein. In another embodiment, diastereomers are separated by separation/resolution techniques based upon differences in solubility. In other embodiments, separation of stereoisomers is performed by chromatography or by the forming diastereomeric salts and separation by recrystallization, or chromatography, or any combination thereof. Jean Jacques, Andre Collet, Samuel H. Wilen, “Enantiomers, Racemates and Resolutions”, John Wiley and Sons, Inc., 1981. In some embodiments, stereoisomers are obtained by stereoselective synthesis. [00141] In some embodiments, compounds described herein are prepared as prodrugs. A “prodrug” refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they are easier to administer than the parent drug. They are, for instance, bioavailable by oral administration whereas the parent is not. The prodrug may be a substrate for a transporter. Further or alternatively, the prodrug also has improved solubility in pharmaceutical compositions over the parent drug. In some embodiments, the design of a prodrug increases the effective water solubility. An example, without limitation, of a prodrug is a compound described herein, which is administered as an ester (the “prodrug”) but then is metabolically hydrolyzed to provide the active entity. A further example of a prodrug is a short peptide (polyaminoacid) bonded to an acid group where the peptide is metabolized to reveal the active moiety. In certain embodiments, upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically, or therapeutically active form of the compound. In certain embodiments, a prodrug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound. [00142] Prodrugs of the compounds described herein include, but are not limited to, esters, ethers, carbonates, thiocarbonates, N-acyl derivatives, N-acyloxyalkyl derivatives, quaternary derivatives of tertiary amines, N-Mannich bases, Schiff bases, amino acid conjugates, phosphate esters, and sulfonate esters. See for example Design of Prodrugs, Bundgaard, A. Ed., Elseview, 1985 and Method in Enzymology, Widder, K. et al., Ed.; Academic, 1985, vol.42, p.309-396; Bundgaard, H. “Design and Application of Prodrugs” in A Textbook of Drug Design and Development, Krosgaard-Larsen and H. Bundgaard, Ed., 1991, Chapter 5, p.113-191; and Bundgaard, H., Advanced Drug Delivery Review, 1992, 8, 1-38, each of which is incorporated herein by reference. In some embodiments, a hydroxyl group in the compounds disclosed herein is used to form a prodrug, wherein the hydroxyl group is incorporated into an acyloxyalkyl ester, alkoxycarbonyloxyalkyl ester, alkyl ester, aryl ester, phosphate ester, sugar ester, ether, and the like. In some embodiments, a hydroxyl group in the compounds disclosed herein is a prodrug wherein the hydroxyl is then metabolized in vivo to provide a carboxylic acid group. In some embodiments, a carboxyl group is used to provide an ester or amide (i.e. the prodrug), which is then metabolized in vivo to provide a carboxylic acid group. In some embodiments, compounds described herein are prepared as alkyl ester prodrugs. [00143] Prodrug forms of the herein described compounds, wherein the prodrug is metabolized in vivo to produce a compound described herein as set forth herein are included within the scope of the claims. In some cases, some of the herein-described compounds is a prodrug for another derivative or active compound. In some embodiments, a prodrug of the compound disclosed herein permits targeted delivery of the compound to a particular region of the gastrointestinal tract. Formation of a pharmacologically active metabolite by the colonic metabolism of drugs is a commonly used “prodrug” approach for the colon-specific drug delivery systems. [00144] In some embodiments, a prodrug is formed by the formation of a covalent linkage between drug and a carrier in such a manner that upon oral administration the moiety remains intact in the stomach and small intestine. This approach involves the formation of a prodrug, which is a pharmacologically inactive derivative of a parent drug molecule that requires spontaneous or enzymatic transformation in the biological environment to release the active drug. Formation of prodrugs has improved delivery properties over the parent drug molecule. The problem of stability of certain drugs from the adverse environment of the upper gastrointestinal tract can be eliminated by prodrug formation, which is converted into the parent drug molecule once it reaches the colon. Site specific drug delivery through site specific prodrug activation may be accomplished by the utilization of some specific property at the target site, such as altered pH or high activity of certain enzymes relative to the non-target tissues for the prodrug-drug conversion. [00145] In some embodiments, covalent linkage of the drug with a carrier forms a conjugate. Such conjugates include, but are not limited to, azo bond conjugates, glycoside conjugates, glucuronide conjugates, cyclodextrin conjugates, dextran conjugates or amino-acid conjugates. [00146] In additional or further embodiments, the compounds described herein are metabolized upon administration to an organism in need to produce a metabolite that is then used to produce a desired effect, including a desired therapeutic effect. [00147] A “metabolite” of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized. The term “active metabolite” refers to a biologically active derivative of a compound that is formed when the compound is metabolized. The term “metabolized,” as used herein, refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes) by which a particular substance is changed by an organism. Thus, enzymes may produce specific structural alterations to a compound. For example, cytochrome P450 catalyzes a variety of oxidative and reductive reactions while uridine diphosphate glucuronyltransferases catalyze the transfer of an activated glucuronic-acid molecule to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulphydryl groups. Metabolites of the compounds disclosed herein are optionally identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds. [00148] In additional or further embodiments, the compounds are rapidly metabolized in plasma. [00149] In additional or further embodiments, the compounds are rapidly metabolized by the intestines. [00150] In additional or further embodiments, the compounds are rapidly metabolized by the liver. Synthesis of Compounds [00151] Compounds described herein are synthesized using standard synthetic techniques or using methods known in the art in combination with methods described herein. [00152] Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology are employed. [00153] Compounds are prepared using standard organic chemistry techniques such as those described in, for example, March’s Advanced Organic Chemistry, 6 th Edition, John Wiley and Sons, Inc. Alternative reaction conditions for the synthetic transformations described herein may be employed such as variation of solvent, reaction temperature, reaction time, as well as different chemical reagents and other reaction conditions. The starting materials are available from commercial sources or are readily prepared. [00154] Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation, include for example, "Synthetic Organic Chemistry", John Wiley & Sons, Inc., New York; S. R. Sandler et al., "Organic Functional Group Preparations," 2nd Ed., Academic Press, New York, 1983; H. O. House, "Modern Synthetic Reactions", 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif.1972; T. L. Gilchrist, "Heterocyclic Chemistry", 2nd Ed., John Wiley & Sons, New York, 1992; J. March, "Advanced Organic Chemistry: Reactions, Mechanisms and Structure", 4th Ed., Wiley-Interscience, New York, 1992. Additional suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation, include for example, Fuhrhop, J. and Penzlin G. "Organic Synthesis: Concepts, Methods, Starting Materials", Second, Revised and Enlarged Edition (1994) John Wiley & Sons ISBN: 3-527-29074-5; Hoffman, R.V. "Organic Chemistry, An Intermediate Text" (1996) Oxford University Press, ISBN 0-19-509618-5; Larock, R. C. "Comprehensive Organic Transformations: A Guide to Functional Group Preparations" 2nd Edition (1999) Wiley-VCH, ISBN: 0-471-19031-4; March, J. "Advanced Organic Chemistry: Reactions, Mechanisms, and Structure" 4th Edition (1992) John Wiley & Sons, ISBN: 0-471-60180-2; Otera, J. (editor) "Modern Carbonyl Chemistry" (2000) Wiley- VCH, ISBN: 3-527-29871-1; Patai, S. "Patai's 1992 Guide to the Chemistry of Functional Groups" (1992) Interscience ISBN: 0-471-93022-9; Solomons, T. W. G. "Organic Chemistry" 7th Edition (2000) John Wiley & Sons, ISBN: 0-471-19095-0; Stowell, J.C., "Intermediate Organic Chemistry" 2nd Edition (1993) Wiley-Interscience, ISBN: 0-471-57456-2; "Industrial Organic Chemicals: Starting Materials and Intermediates: An Ullmann's Encyclopedia" (1999) John Wiley & Sons, ISBN: 3-527-29645-X, in 8 volumes; "Organic Reactions" (1942-2000) John Wiley & Sons, in over 55 volumes; and "Chemistry of Functional Groups" John Wiley & Sons, in 73 volumes. [00155] In some embodiments, compounds are prepared as described in the Examples. Certain Terminology [00156] Unless otherwise stated, the following terms used in this application have the definitions given below. The use of the term “including” as well as other forms, such as “include”, “includes,” and “included,” is not limiting. The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. [00157] As used herein, C 1- C x includes C 1- C 2 , C 1- C 3 ... C 1- C x . By way of example only, a group designated as "C 1- C 4 " indicates that there are one to four carbon atoms in the moiety, i.e. groups containing 1 carbon atom, 2 carbon atoms, 3 carbon atoms or 4 carbon atoms. Thus, by way of example only, "C 1- C 4 alkyl" indicates that there are one to four carbon atoms in the alkyl group, i.e., the alkyl group is selected from among methyl, ethyl, propyl, iso-propyl, n-butyl, iso- butyl, sec-butyl, and t-butyl. [00158] An “alkyl” group refers to an aliphatic hydrocarbon group. The alkyl group is branched or straight chain. In some embodiments, the “alkyl” group has 1 to 10 carbon atoms, i.e. a C 1- C 10 alkyl. Whenever it appears herein, a numerical range such as “1 to 10” refers to each integer in the given range; e.g., “1 to 10 carbon atoms” means that the alkyl group consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms,6 carbon atoms, etc., up to and including 10 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated. In some embodiments, an alkyl is a C 1- C 6 alkyl. In one aspect the alkyl is methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, or t-butyl. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tertiary butyl, pentyl, neopentyl, or hexyl. [00159] An “alkylene” group refers to a divalent alkyl group. Any of the above mentioned monovalent alkyl groups may be an alkylene by abstraction of a second hydrogen atom from the alkyl. In some embodiments, an alkylene is a C 1- C 6 alkylene. In other embodiments, an alkylene is a C 1- C 4 alkylene. In certain embodiments, an alkylene comprises one to four carbon atoms (e.g., C 1- C 4 alkylene). In other embodiments, an alkylene comprises one to three carbon atoms (e.g., C 1- C 3 alkylene). In other embodiments, an alkylene comprises one to two carbon atoms (e.g., C1-C2 alkylene). In other embodiments, an alkylene comprises one carbon atom (e.g., C 1 alkylene). In other embodiments, an alkylene comprises two carbon atoms (e.g.,C 2 alkylene). In other embodiments, an alkylene comprises two to four carbon atoms (e.g., C 2- C 4 alkylene). Typical alkylene groups include, but are not limited to, -CH 2 -, -CH(CH 3 )-, -C(CH 3 ) 2 -, -CH 2 CH 2 - , -CH 2 CH(CH 3 )-, -CH 2 C(CH 3 ) 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH 2 -, and the like. [00160] “Deuteroalkyl” refers to an alkyl group where 1 or more hydrogen atoms of an alkyl are replaced with deuterium. [00161] The term “alkenyl” refers to a type of alkyl group in which at least one carbon-carbon double bond is present. In one embodiment, an alkenyl group has the formula –C(R)=CR 2 , wherein R refers to the remaining portions of the alkenyl group, which may be the same or different. In some embodiments, R is H or an alkyl. In some embodiments, an alkenyl is selected from ethenyl (i.e., vinyl), propenyl (i.e., allyl), butenyl, pentenyl, pentadienyl, and the like. Non-limiting examples of an alkenyl group include -CH=CH 2 , -C(CH 3 )=CH 2 , - CH=CHCH 3 , -C(CH 3 )=CHCH 3 , and –CH 2 CH=CH 2 . [00162] The term “alkynyl” refers to a type of alkyl group in which at least one carbon-carbon triple bond is present. In one embodiment, an alkenyl group has the formula -C≡C-R, wherein R refers to the remaining portions of the alkynyl group. In some embodiments, R is H or an alkyl. In some embodiments, an alkynyl is selected from ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Non-limiting examples of an alkynyl group include -C≡CH, -C≡CCH 3 - C≡CCH 2 CH 3 , -CH 2 C≡CH. [00163] An “alkoxy” group refers to a (alkyl)O- group, where alkyl is as defined herein. [00164] The term “alkylamine” refers to the –N(alkyl)xHy group, where x is 0 and y is 2, or where x is 1 and y is 1, or where x is 2 and y is 0. [00165] The term “aromatic” refers to a planar ring having a delocalized ^-electron system containing 4n+2 π electrons, where n is an integer. The term “aromatic” includes both carbocyclic aryl (“aryl”, e.g., phenyl) and heterocyclic aryl (or “heteroaryl” or “heteroaromatic”) groups (e.g., pyridine). The term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon or nitrogen atoms) groups. [00166] The term “carbocyclic” or “carbocycle” refers to a ring or ring system where the atoms forming the backbone of the ring are all carbon atoms. The term thus distinguishes carbocyclic from “heterocyclic” rings or “heterocycles” in which the ring backbone contains at least one atom which is different from carbon. In some embodiments, at least one of the two rings of a bicyclic carbocycle is aromatic. In some embodiments, both rings of a bicyclic carbocycle are aromatic. Carbocycle includes cycloalkyl and aryl. [00167] As used herein, the term “aryl” refers to an aromatic ring wherein each of the atoms forming the ring is a carbon atom. In one aspect, aryl is phenyl or a naphthyl. In some embodiments, an aryl is a phenyl. In some embodiments, an aryl is a C 6 -C 10 aryl. Depending on the structure, an aryl group is a monoradical or a diradical (i.e., an arylene group). [00168] The term “cycloalkyl” refers to a monocyclic or polycyclic aliphatic, non-aromatic group, wherein each of the atoms forming the ring (i.e. skeletal atoms) is a carbon atom. In some embodiments, cycloalkyls are spirocyclic or bridged compounds. In some embodiments, cycloalkyls are fully saturated. In some embodiments, cycloalkyls are partially unsaturated. In some embodiments, cycloalkyls are optionally fused with an aromatic ring, and the point of attachment is at a carbon that is not an aromatic ring carbon atom. Cycloalkyl groups include groups having from 3 to 10 ring atoms. In some embodiments, cycloalkyl groups are selected from among cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctyl, spiro[2.2]pentyl, norbornyl and bicyclo[1.1.1]pentyl. In some embodiments, a cycloalkyl is a C 3- C 6 cycloalkyl. In some embodiments, a cycloalkyl is a monocyclic cycloalkyl. Monocyclic cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Polycyclic cycloalkyls include, for example, adamantyl, norbornyl (i.e., bicyclo[2.2.1]heptanyl), norbornenyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like [00169] The term “halo” or, alternatively, “halogen” or “halide” means fluoro, chloro, bromo or iodo. In some embodiments, halo is fluoro, chloro, or bromo. [00170] The term “haloalkyl” refers to an alkyl in which one or more hydrogen atoms are replaced by a halogen atom. In one aspect, a fluoroalkyl is a C 1- C 6 fluoroalkyl. [00171] The term “fluoroalkyl” refers to an alkyl in which one or more hydrogen atoms are replaced by a fluorine atom. In one aspect, a fluoroalkyl is a C 1- C 6 fluoroalkyl. In some embodiments, a fluoroalkyl is selected from trifluoromethyl, difluoromethyl, fluoromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, and the like. [00172] The term “heteroalkyl” refers to an alkyl group in which one or more skeletal atoms of the alkyl are selected from an atom other than carbon, e.g., oxygen, nitrogen (e.g., -NH-, - N(alkyl)-, sulfur, or combinations thereof. A heteroalkyl is attached to the rest of the molecule at a carbon atom of the heteroalkyl. In one aspect, a heteroalkyl is a C 1- C 6 heteroalkyl. [00173] The term “heteroalkylene” refers to a divalent heteroalkyl group. [00174] The term "heterocycle" or “heterocyclic” refers to heteroaromatic rings (also known as heteroaryls) and heterocycloalkyl rings (also known as heteroalicyclic groups) containing one to four heteroatoms in the ring(s), where each heteroatom in the ring(s) is selected from O, S and N, wherein each heterocyclic group has from 3 to 10 atoms in its ring system, and with the proviso that any ring does not contain two adjacent O or S atoms. In some embodiments, heterocycles are monocyclic, bicyclic, polycyclic, spirocyclic or bridged compounds. Non- aromatic heterocyclic groups (also known as heterocycloalkyls) include rings having 3 to 10 atoms in its ring system and aromatic heterocyclic groups include rings having 5 to 10 atoms in its ring system. The heterocyclic groups include benzo-fused ring systems. Examples of non- aromatic heterocyclic groups are pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, oxazolidinonyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, thioxanyl, piperazinyl, aziridinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1,2,3,6-tetrahydropyridinyl, pyrrolin-2-yl, pyrrolin-3-yl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3-azabicyclo[3.1.0]hexanyl, 3- azabicyclo[4.1.0]heptanyl, 3H-indolyl, indolin-2-onyl, isoindolin-1-onyl, isoindoline-1,3-dionyl, 3,4-dihydroisoquinolin-1(2H)-onyl, 3,4-dihydroquinolin-2(1H)-onyl, isoindoline-1,3-dithionyl, benzo[d]oxazol-2(3H)-onyl, 1H-benzo[d]imidazol-2(3H)-onyl, benzo[d]thiazol-2(3H)-onyl, and quinolizinyl. Examples of aromatic heterocyclic groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl. The foregoing groups are either C-attached (or C-linked) or N-attached where such is possible. For instance, a group derived from pyrrole includes both pyrrol-1-yl (N-attached) or pyrrol-3-yl (C-attached). Further, a group derived from imidazole includes imidazol-1-yl or imidazol-3-yl (both N- attached) or imidazol-2-yl, imidazol-4-yl or imidazol-5-yl (all C-attached). The heterocyclic groups include benzo-fused ring systems. Non-aromatic heterocycles are optionally substituted with one or two oxo (=O) moieties, such as pyrrolidin-2-one. In some embodiments, at least one of the two rings of a bicyclic heterocycle is aromatic. In some embodiments, both rings of a bicyclic heterocycle are aromatic. [00175] The terms “heteroaryl” or, alternatively, “heteroaromatic” refers to an aryl group that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur. Illustrative examples of heteroaryl groups include monocyclic heteroaryls and bicyclic heteroaryls. Monocyclic heteroaryls include pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, pyridazinyl, triazinyl, oxadiazolyl, thiadiazolyl, and furazanyl. Bicyclic heteroaryls include indolizine, indole, benzofuran, benzothiophene, indazole, benzimidazole, benzotriazole, purine, quinolizine, quinoline, isoquinoline, cinnoline, phthalazine, quinazoline, quinoxaline, 1,8- naphthyridine, and pteridine. In some embodiments, a heteroaryl contains 0-4 N atoms in the ring. In some embodiments, a heteroaryl contains 1-4 N atoms in the ring. In some embodiments, a heteroaryl contains 0-4 N atoms, 0-1 O atoms, and 0-1 S atoms in the ring. In some embodiments, a heteroaryl contains 1-4 N atoms, 0-1 O atoms, and 0-1 S atoms in the ring. In some embodiments, heteroaryl is a C 1- C 9 heteroaryl. In some embodiments, monocyclic heteroaryl is a C 1- C 5 heteroaryl. In some embodiments, monocyclic heteroaryl is a 5-membered or 6-membered heteroaryl. In some embodiments, bicyclic heteroaryl is a C 6- C 9 heteroaryl. [00176] A “heterocycloalkyl” or “heteroalicyclic” group refers to a cycloalkyl group that includes at least one heteroatom selected from nitrogen, oxygen and sulfur. In some embodiments, heterocycloalkyls are spirocyclic or bridged compounds. In some embodiments, heterocycloalkyls are fully saturated. In some embodiments, heterocycloalkyls are partially unsaturated. In some embodiments, a heterocycloalkyl is fused with an aryl or heteroaryl. In some embodiments, the heterocycloalkyl is oxazolidinonyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, piperidin-2-onyl, pyrrolidine-2,5-dithionyl, pyrrolidine-2,5-dionyl, pyrrolidinonyl, imidazolidinyl, imidazolidin-2-onyl, or thiazolidin-2-onyl. The term heteroalicyclic also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides. In one aspect, a heterocycloalkyl is a C 2- C 10 heterocycloalkyl. In another aspect, a heterocycloalkyl is a C 4 -C 10 heterocycloalkyl. In some embodiments, a heterocycloalkyl contains 0-2 N atoms in the ring. In some embodiments, a heterocycloalkyl contains 0-2 N atoms, 0-2 O atoms and 0-1 S atoms in the ring. [00177] The term “bond” or “single bond” refers to a chemical bond between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure. In one aspect, when a group described herein is a bond, the referenced group is absent thereby allowing a bond to be formed between the remaining identified groups. [00178] The term “moiety” refers to a specific segment or functional group of a molecule. Chemical moieties are often recognized chemical entities embedded in or appended to a molecule. [00179] The term “optionally substituted” or “substituted” means that the referenced group is optionally substituted with one or more additional group(s). In some other embodiments, optional substituents are individually and independently selected from D, halogen, -CN, -NH2, - NH(alkyl), -N(alkyl) 2 , -OH, -CO 2 H, -CO 2 alkyl, -C(=O)NH 2 , -C(=O)NH(alkyl), - C(=O)N(alkyl) 2 , -S(=O) 2 NH 2 , -S(=O) 2 NH(alkyl), -S(=O) 2 N(alkyl) 2 , -CH 2 CO 2 H, -CH 2 CO 2 alkyl, -CH 2 C(=O)NH 2 , -CH 2 C(=O)NH(alkyl), -CH 2 C(=O)N(alkyl) 2 , -CH 2 S(=O) 2 NH 2 , - CH 2 S(=O) 2 NH(alkyl), - CH 2 S(=O) 2 N(alkyl) 2 , alkyl, alkenyl, alkynyl, cycloalkyl, fluoroalkyl, heteroalkyl, alkoxy, fluoroalkoxy, heterocycloalkyl, aryl, heteroaryl, aryloxy, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulfone, and arylsulfone. The term “optionally substituted” or “substituted” means that the referenced group is optionally substituted with one or more additional group(s) individually and independently selected from D, halogen, -CN, -NH 2 , - NH(alkyl), -N(alkyl) 2 , -OH, -CO 2 H, -CO 2 alkyl, -C(=O)NH 2 , -C(=O)NH(alkyl), - C(=O)N(alkyl) 2 , -S(=O) 2 NH 2 , -S(=O) 2 NH(alkyl), -S(=O) 2 N(alkyl) 2 , alkyl, cycloalkyl, fluoroalkyl, heteroalkyl, alkoxy, fluoroalkoxy, heterocycloalkyl, aryl, heteroaryl, aryloxy, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulfone, and arylsulfone. In some other embodiments, optional substituents are independently selected from D, halogen, -CN, -NH 2 , - NH(CH 3 ), -N(CH 3 ) 2 , -OH, -CO 2 H, -CO 2 (C 1- C 4 alkyl), -C(=O)NH 2 , -C(=O)NH(C 1- C 4 alkyl), - C(=O)N(C 1- C 4 alkyl) 2 , -S(=O) 2 NH 2 , -S(=O) 2 NH(C 1- C 4 alkyl), -S(=O) 2 N(C 1- C 4 alkyl) 2 , C 1- C 4 alkyl, C 3- C 6 cycloalkyl, C 1- C 4 fluoroalkyl, C 1- C 4 heteroalkyl, C 1- C 4 alkoxy, C 1- C 4 fluoroalkoxy, -SC 1- C 4 alkyl, -S(=O)C 1- C 4 alkyl, and -S(=O) 2 C 1- C 4 alkyl. In some embodiments, optional substituents are independently selected from D, halogen, -CN, -NH 2 , -OH, -NH(CH 3 ), -N(CH 3 ) 2 , -CH 3 , -CH 2 CH 3 , -CF 3 , -OCH 3 , and -OCF 3 . In some embodiments, substituted groups are substituted with one or two of the preceding groups. In some embodiments, substituted groups are substituted with one of the preceding groups. In some embodiments, an optional substituent on an aliphatic carbon atom (acyclic or cyclic) includes oxo (=O). [00180] The term “acceptable” with respect to a formulation, composition or ingredient, as used herein, means having no persistent detrimental effect on the general health of the subject being treated. [00181] The term “modulate” as used herein, means to interact with a target either directly or indirectly so as to alter the activity of the target, including, by way of example only, to enhance the activity of the target, to inhibit the activity of the target, to limit the activity of the target, or to extend the activity of the target. [00182] The term “modulator” as used herein, refers to a molecule that interacts with a target either directly or indirectly. The interactions include, but are not limited to, the interactions of an agonist, partial agonist, an inverse agonist, antagonist, degrader, or combinations thereof. In some embodiments, a modulator is an agonist. [00183] The terms "administer," "administering", "administration," and the like, as used herein, refer to the methods that may be used to enable delivery of compounds or compositions to the desired site of biological action. These methods include, but are not limited to oral routes, intraduodenal routes, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intravascular or infusion), topical and rectal administration. Those of skill in the art are familiar with administration techniques that can be employed with the compounds and methods described herein. In some embodiments, the compounds and compositions described herein are administered orally. [00184] The terms “co-administration” or the like, as used herein, are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time. [00185] The terms “effective amount” or “therapeutically effective amount,” as used herein, refer to a sufficient amount of an agent or a compound being administered, which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms. An appropriate “effective” amount in any individual case is optionally determined using techniques, such as a dose escalation study. [00186] The terms “enhance” or “enhancing,” as used herein, means to increase or prolong either in potency or duration a desired effect. Thus, in regard to enhancing the effect of therapeutic agents, the term “enhancing” refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system. An “enhancing-effective amount,” as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system. [00187] The terms “kit” and “article of manufacture” are used as synonyms. [00188] The term “subject” or “patient” encompasses mammals. Examples of mammals include, but are not limited to, any member of the Mammalian class: 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. In one aspect, the mammal is a human. [00189] The terms “treat,” “treating” or “treatment,” as used herein, include alleviating, abating or ameliorating at least one symptom of a disease or condition, preventing additional symptoms, inhibiting the disease or condition, e.g., arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactically and/or therapeutically. Pharmaceutical compositions [00190] In some embodiments, the compounds described herein are formulated into pharmaceutical compositions. Pharmaceutical compositions are formulated in a conventional manner using one or more pharmaceutically acceptable inactive ingredients that facilitate processing of the active compounds into preparations that are used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. A summary of pharmaceutical compositions described herein is found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington’s Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins1999), herein incorporated by reference for such disclosure. [00191] In some embodiments, the compounds described herein are administered either alone or in combination with pharmaceutically acceptable carriers, excipients or diluents, in a pharmaceutical composition. Administration of the compounds and compositions described herein can be affected by any method that enables delivery of the compounds to the site of action. These methods include, though are not limited to delivery via enteral routes (including oral, gastric or duodenal feeding tube, rectal suppository and rectal enema), parenteral routes (injection or infusion, including intraarterial, intracardiac, intradermal, intraduodenal, intramedullary, intramuscular, intraosseous, intraperitoneal, intrathecal, intravascular, intravenous, intravitreal, epidural and subcutaneous), inhalational, transdermal, transmucosal, sublingual, buccal and topical (including epicutaneous, dermal, enema, eye drops, ear drops, intranasal, vaginal) administration, although the most suitable route may depend upon for example the condition and disorder of the recipient. By way of example only, compounds described herein can be administered locally to the area in need of treatment, by for example, local infusion during surgery, topical application such as creams or ointments, injection, catheter, or implant. The administration can also be by direct injection at the site of a diseased tissue or organ. [00192] In some embodiments, pharmaceutical compositions suitable for oral administration are presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. In some embodiments, the active ingredient is presented as a bolus, electuary or paste. [00193] Pharmaceutical compositions which can be used orally include tablets, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Tablets may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with binders, inert diluents, or lubricating, surface active or dispersing agents. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. In some embodiments, the tablets are coated or scored and are formulated so as to provide slow or controlled release of the active ingredient therein. All formulations for oral administration should be in dosages suitable for such administration. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In some embodiments, stabilizers are added. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or Dragee coatings for identification or to characterize different combinations of active compound doses. [00194] In some embodiments, pharmaceutical compositions are formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. The compositions may be presented in unit-dose or multi- dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or sterile pyrogen-free water, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described. [00195] Pharmaceutical compositions for parenteral administration include aqueous and non- aqueous (oily) sterile injection solutions of the active compounds which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. [00196] Pharmaceutical compositions may also be formulated as a depot preparation. Such long-acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. [00197] For buccal or sublingual administration, the compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner. Such compositions may comprise the active ingredient in a flavored basis such as sucrose and acacia or tragacanth. [00198] Pharmaceutical compositions may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter, polyethylene glycol, or other glycerides. [00199] Pharmaceutical compositions may be administered topically, that is by non-systemic administration. This includes the application of a compound of the present invention externally to the epidermis or the buccal cavity and the instillation of such a compound into the ear, eye and nose, such that the compound does not significantly enter the blood stream. In contrast, systemic administration refers to oral, intravenous, intraperitoneal and intramuscular administration. [00200] Pharmaceutical compositions suitable for topical administration include liquid or semi- liquid preparations suitable for penetration through the skin to the site of inflammation such as gels, liniments, lotions, creams, ointments or pastes, and drops suitable for administration to the eye, ear or nose. The active ingredient may comprise, for topical administration, from 0.001% to 10% w/w, for instance from 1% to 2% by weight of the formulation. [00201] Pharmaceutical compositions for administration by inhalation are conveniently delivered from an insufflator, nebulizer pressurized packs or other convenient means of delivering an aerosol spray. Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Alternatively, for administration by inhalation or insufflation, pharmaceutical preparations may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch. The powder composition may be presented in unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator. [00202] In some embodiments, a compound disclosed herein is formulated to provide a controlled release of the compound. Controlled release refers to the release of the compound described herein from a dosage form in which it is incorporated according to a desired profile over an extended period of time. Controlled release profiles include, for example, sustained release, prolonged release, pulsatile release, and delayed release profiles. In contrast to immediate release compositions, controlled release compositions allow delivery of an agent to a subject over an extended period of time according to a predetermined profile. Such release rates can provide therapeutically effective levels of agent for an extended period of time and thereby provide a longer period of pharmacologic response while minimizing side effects as compared to conventional rapid release dosage forms. Such longer periods of response provide for many inherent benefits that are not achieved with the corresponding short acting, immediate release preparations. [00203] Approaches to deliver the intact therapeutic compound to the particular regions of the gastrointestinal tract (e.g., such as the colon), include: [00204] (i) Coating with polymers: The intact molecule can be delivered to the colon without absorbing at the upper part of the intestine by coating of the drug molecule with the suitable polymers, which degrade only in the colon. [00205] (ii) Coating with pH-sensitive polymers: The majority of enteric and colon targeted delivery systems are based on the coating of tablets or pellets, which are filled into conventional hard gelatin capsules. Most commonly used pH-dependent coating polymers are methacrylic acid copolymers, commonly known as Eudragit® S, more specifically Eudragit® L and Eudragit® S. Eudragit® L100 and S 100 are copolymers of methacrylic acid and methyl methacrylate. Additional pH-dependent coating polymers include cellulose acetate phthalate (CAP), hydroxypropyl methylcellulose phthalate (HPMCP), polyvinyl acetate phthalate (PVAP) and cellulose acetate trimelliate. [00206] (iii) Coating with biodegradable polymers; [00207] (iv) Embedding in matrices; [00208] (v) Embedding in biodegradable matrices and hydrogels; [00209] (vi) Embedding in pH-sensitive matrices; [00210] (vii) Timed release systems; [00211] (viii)Redox-sensitive polymers; [00212] (ix) Bioadhesive systems; [00213] (x) Coating with microparticles; [00214] (xi) Osmotic controlled drug delivery. [00215] Another approach towards colon-targeted drug delivery or controlled-release systems includes embedding the drug in polymer matrices to trap it and release it in the colon. These matrices can be pH-sensitive or biodegradable. Matrix-Based Systems, such as multi-matrix (MMX)-based delayed-release tablets, ensure the drug release in the colon. [00216] Additional pharmaceutical approaches to targeted delivery of therapeutics to particular regions of the gastrointestinal tract are known. Chourasia MK, Jain SK, Pharmaceutical approaches to colon targeted drug delivery systems., J Pharm Sci.2003 Jan-Apr; 6(1):33-66. Patel M, Shah T, Amin A. Therapeutic opportunities in colon-specific drug-delivery systems Crit Rev Ther Drug Carrier Syst.2007; 24(2):147-202. Kumar P, Mishra B. Colon targeted drug delivery systems-an overview. Curr Drug Deliv.2008 Jul; 5(3):186-98. Van den Mooter G. Colon drug delivery. Expert Opin Drug Deliv.2006 Jan; 3(1):111-25. Seth Amidon, Jack E. Brown, and Vivek S. Dave, Colon-Targeted Oral Drug Delivery Systems: Design Trends and Approaches, AAPS PharmSciTech.2015 Aug; 16(4): 731–741. [00217] It should be understood that in addition to the ingredients particularly mentioned above, the compounds and compositions described herein may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents. Methods of Dosing and Treatment Regimens [00218] In one embodiment, the compounds described herein, or a pharmaceutically acceptable salt thereof, are used in the preparation of medicaments for the treatment of diseases or conditions in a mammal that would benefit from administration of an HSD17B13 inhibitor. Methods for treating any of the diseases or conditions described herein in a mammal in need of such treatment, involves administration of pharmaceutical compositions that include at least one compound described herein or a pharmaceutically acceptable salt, active metabolite, prodrug, or pharmaceutically acceptable solvate thereof, in therapeutically effective amounts to said mammal. [00219] In some embodiments, described herein is a method of treating or preventing a liver disease or condition in a mammal, comprising administering to the mammal a compound of Formula (I’), (I), (II’), (II), (IIa’), (IIa), (III’), (III), (IIIa’), (IIIa), (IV’), (IV), (IVa’), or (IVa), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments, described herein is a method of treating or preventing an alcoholic or nonalcoholic liver disease or condition in a mammal, comprising administering to the mammal a compound of Formula (I’), (I), (II’), (II), (IIa’), (IIa), (III’), (III), (IIIa’), (IIIa), (IV’), (IV), (IVa’), or (IVa), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments, the liver disease or condition is an alcoholic liver disease or condition. In some embodiments, the liver disease or condition is a nonalcoholic liver disease or condition. In some embodiments, the liver disease or condition is liver inflammation, fatty liver (steatosis), liver fibrosis, hepatitis, cirrhosis, hepatocellular carcinoma, or combinations thereof. In some embodiments, the liver disease or condition is primary biliary cirrhosis, primary sclerosing cholangitis, cholestasis, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), or combinations thereof. In some embodiments, the liver disease or condition is a chronic liver disease or condition. [00220] In some embodiments, described herein is a method of modulating HSD17B13 activity in a mammal, comprising administering to the mammal a compound of Formula (I’), (I), (II’), (II), (IIa’), (IIa), (III’), (III), (IIIa’), (IIIa), (IV’), (IV), (IVa’), or (IVa), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments, modulating comprises inhibiting HSD17B13 activity. In some embodiments of a method of modulating HSD17B13 activity in a mammal, the mammal has a liver disease or condition selected from liver inflammation, fatty liver (steatosis), liver fibrosis, hepatitis, cirrhosis, hepatocellular carcinoma, and combinations thereof. In some embodiments of a method of modulating HSD17B13 activity in a mammal, the mammal has a liver disease or condition selected from primary biliary cirrhosis, primary sclerosing cholangitis, cholestasis, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), and combinations thereof. [00221] In certain embodiments, the compositions containing the compound(s) described herein are administered for prophylactic and/or therapeutic treatments. In certain therapeutic applications, the compositions are administered to a patient already suffering from a disease or condition, in an amount sufficient to cure or at least partially arrest at least one of the symptoms of the disease or condition. Amounts effective for this use depend on the severity and course of the disease or condition, previous therapy, the patient's health status, weight, and response to the drugs, and the judgment of the treating physician. Therapeutically effective amounts are optionally determined by methods including, but not limited to, a dose escalation and/or dose ranging clinical trial. [00222] In prophylactic applications, compositions containing the compounds described herein are administered to a patient susceptible to or otherwise at risk of a particular disease, disorder, or condition. Such an amount is defined to be a "prophylactically effective amount or dose." In this use, the precise amounts also depend on the patient's state of health, weight, and the like. When used in patients, effective amounts for this use will depend on the severity and course of the disease, disorder, or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician. In one aspect, prophylactic treatments include administering to a mammal, who previously experienced at least one symptom of the disease being treated and is currently in remission, a pharmaceutical composition comprising a compound described herein, or a pharmaceutically acceptable salt thereof, in order to prevent a return of the symptoms of the disease or condition. [00223] In certain embodiments wherein the patient’s condition does not improve, upon the doctor’s discretion, the compounds are administered chronically, that is, for an extended period of time, including throughout the duration of the patient’s life in order to ameliorate or otherwise control or limit the symptoms of the patient’s disease or condition. [00224] In certain embodiments wherein a patient’s status does improve, the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time (i.e., a “drug holiday”). In specific embodiments, the length of the drug holiday is between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, or more than 28 days. The dose reduction during a drug holiday is, by way of example only, by 10%-100%, including by way of example only 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, and 100%. [00225] Once improvement of the patient's conditions has occurred, a maintenance dose is administered if necessary. Subsequently, in specific embodiments, the dosage or the frequency of administration, or both, is reduced, as a function of the symptoms, to a level at which the improved disease, disorder, or condition is retained. In certain embodiments, however, the patient requires intermittent treatment on a long-term basis upon any recurrence of symptoms. [00226] The amount of a given agent that corresponds to such an amount varies depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight, sex) of the subject or host in need of treatment, but nevertheless is determined according to the particular circumstances surrounding the case, including, e.g., the specific agent being administered, the route of administration, the condition being treated, and the subject or host being treated. [00227] In general, however, doses employed for adult human treatment are typically in the range of 0.01 mg-5000 mg per day. In one aspect, doses employed for adult human treatment are from about 1 mg to about 1000 mg per day. In one embodiment, the desired dose is conveniently presented in a single dose or in divided doses administered simultaneously or at appropriate intervals, for example as two, three, four or more sub-doses per day. [00228] In one embodiment, the daily dosages appropriate for the compound described herein, or a pharmaceutically acceptable salt thereof, are from about 0.01 to about 50 mg/kg per body weight. In some embodiments, the daily dosage or the amount of active in the dosage form are lower or higher than the ranges indicated herein, based on a number of variables in regard to an individual treatment regime. In various embodiments, the daily and unit dosages are altered depending on a number of variables including, but not limited to, the activity of the compound used, the disease or condition to be treated, the mode of administration, the requirements of the individual subject, the severity of the disease or condition being treated, and the judgment of the practitioner. [00229] Toxicity and therapeutic efficacy of such therapeutic regimens are determined by standard pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, the determination of the LD50 and the ED50. The dose ratio between the toxic and therapeutic effects is the therapeutic index and it is expressed as the ratio between LD50 and ED50. In certain embodiments, the data obtained from cell culture assays and animal studies are used in formulating the therapeutically effective daily dosage range and/or the therapeutically effective unit dosage amount for use in mammals, including humans. In some embodiments, the daily dosage amount of the compounds described herein lies within a range of circulating concentrations that include the ED50 with minimal toxicity. In certain embodiments, the daily dosage range and/or the unit dosage amount varies within this range depending upon the dosage form employed and the route of administration utilized. [00230] In any of the aforementioned aspects are further embodiments in which the effective amount of the compound described herein, or a pharmaceutically acceptable salt thereof, is: (a) systemically administered to the mammal; and/or (b) administered orally to the mammal; and/or (c) intravenously administered to the mammal; and/or (d) administered by injection to the mammal; and/or (e) administered topically to the mammal; and/or (f) administered non- systemically or locally to the mammal. [00231] In any of the aforementioned aspects are further embodiments comprising single administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered once a day; or (ii) the compound is administered to the mammal multiple times over the span of one day. [00232] In any of the aforementioned aspects are further embodiments comprising multiple administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered continuously or intermittently: as in a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the compound is administered to the mammal every 8 hours; (iv) the compound is administered to the mammal every 12 hours; (v) the compound is administered to the mammal every 24 hours. In further or alternative embodiments, the method comprises a drug holiday, wherein the administration of the compound is temporarily suspended or the dose of the compound being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound is resumed. In one embodiment, the length of the drug holiday varies from 2 days to 1 year. [00233] It is understood that the dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought, is modified in accordance with a variety of factors (e.g., the disease, disorder, or condition from which the subject suffers; the age, weight, sex, diet, and medical condition of the subject). Thus, in some instances, the dosage regimen actually employed varies and, in some embodiments, deviates from the dosage regimens set forth herein. [00234] The compounds described herein, or a pharmaceutically acceptable salt thereof, as well as combination therapies, are administered before, during or after the occurrence of a disease or condition, and the timing of administering the composition containing a compound varies. Thus, in one embodiment, the compounds described herein are used as a prophylactic and are administered continuously to subjects with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease or condition. In another embodiment, the compounds and compositions are administered to a subject during or as soon as possible after the onset of the symptoms. In specific embodiments, a compound described herein is administered as soon as is practicable after the onset of a disease or condition is detected or suspected, and for a length of time necessary for the treatment of the disease. In some embodiments, the length required for treatment varies, and the treatment length is adjusted to suit the specific needs of each subject. For example, in specific embodiments, a compound described herein or a formulation containing the compound is administered for at least 2 weeks, about 1 month to about 5 years. EXAMPLES [00235] The following examples are provided for illustrative purposes only and not to limit the scope of the claims provided herein. [00236] As used above, and throughout the description of the invention, the following abbreviations, unless otherwise indicated, shall be understood to have the following meanings: acac acetylacetone ACN or MeCN acetonitrile AcOH acetic acid Ac acetyl BINAP 2,2′-bis(diphenylphosphino)-1,1′-binaphthalene Bn benzyl BOC or Boc tert-butyl carbamate i-Bu iso-butyl t-Bu tert-butyl Cy cyclohexyl CDI 1,1-carbonyldiimidazole CPME cyclopentyl methyl ether DAST (diethylamino)sulfur trifluoride DBA or dba dibenzylideneacetone DCE dichloroethane (ClCH 2 CH 2 Cl) DCM dichloromethane (CH 2 Cl 2 ) DHP 3,4-dihydropyran DIBAL-H diisobutylaluminum hydride DIPEA or DIEA diisopropylethylamine DMAP 4-(N,N-dimethylamino)pyridine DME 1,2-dimethoxyethane DMF N,N-dimethylformamide DMA N,N-dimethylacetamide DMPU N,N′-dimethylpropyleneurea DMSO dimethylsulfoxide Dppf or dppf 1,1'-bis(diphenylphosphino)ferrocene EDC or EDCI N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride EEDQ 2-Ethoxy-1-ethoxycarbonyl-1,2-dihydroquinoline eq equivalent(s) Et ethyl Et2O diethyl ether EtOH ethanol EtOAc ethyl acetate HATU 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5- b]pyridinium 3-oxid hexafluorophosphate HMPA hexamethylphosphoramide HOBt 1-hydroxybenzotriazole HPLC high performance liquid chromatography IBX 2-iodoxybenzoic acid KOAc potassium acetate KHMDS potassium bis(trimethylsilyl)amide NaHMDS sodium bis(trimethylsilyl)amide LiHMDS lithium bis(trimethylsilyl)amide LAH lithium aluminum anhydride LCMS liquid chromatography mass spectrometry 2-MeTHF 2-methyltetrahydrofuran Me methyl MeOH methanol MOM methoxymethyl ether MS mass spectroscopy Ms mesyl MTBE methyl tert-butyl ether NBS N-bromosuccinimide NCS N-chlorosuccinimide NIS N-iodosuccinimide NMI 1-methylimidazole NMM N-methyl-morpholine NMP N-methyl-pyrrolidin-2-one NMR nuclear magnetic resonance OTf trifluoromethanesulfonate PCC pyridinium chlorochromate PE petroleum ether PG protecting group Ph phenyl PPTS pyridium p-toluenesulfonate iPr/i-Pr iso-propyl RP-HPLC reverse-phase high-pressure liquid chromatography rt room temperature SEM 2-(trimethylsilyl)ethoxymethyl TBS tert-butyldimethylsilyl TBAF tetra-n-butylammonium fluoride TBAI tetra-n-butylammonium iodide TCFH chloro-N,N,N’,N’-tetramethylformamidinium hexafluorophosphate TEA triethylamine TFA trifluoroacetic acid THF tetrahydrofuran TLC thin layer chromatography TMEDA N,N,N′,N′-tetramethylethylenediamine TMS trimethylsilyl TsOH/p-TsOH p-toluenesulfonic acid Intermediate 1 6-(Benzyloxy)-1-(4-bromophenyl)-5,7-difluoro-1H-benzo[d][1,2 ,3]triazole Step 1: 3-(Benzyloxy)-2,4-difluoro-6-nitroaniline [00237] Sodium hydride (3.12 g, 78.1 mmol, 60% in mineral oil) was added to a solution of 2,3,4-trifluoro-6-nitroaniline (5.00 g, 26.03 mmol) in THF (100 mL) at 0 °C under N 2 . The mixture was stirred for 0.5 h. Benzyl alcohol (4.06 mL, 39.0 mmol) in THF (10 mL) was added dropwise at 0 °C. The reaction mixture was stirred at rt for 1 h, poured into ice-water (100 mL) carefully, and then extracted with EtOAc (3×100 mL). The combined organic phases were washed with brine (100 mL), dried over Na 2 SO 4 , filtered, concentrated, and then triturated with EtOH (20 mL) for 30 min to obtain 3-(benzyloxy)-2,4-difluoro-6-nitroaniline (5 g) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 7.78 (d, 1H), 7.45-7.36 (m, 5H), 7.29 (s, 2H), 5.35 (s, 2H). Step 2: 3-(Benzyloxy)-N-(4-bromophenyl)-2,4-difluoro-6-nitroaniline [00238] Tris(dibenzylideneacetone)dipalladium(0) (1.63 g, 1.78 mmol) was added to a solution of 3-(benzyloxy)-2,4-difluoro-6-nitroaniline (5 g, crude), 1-bromo-4-iodobenzene (6.06 g, 21.4 mmol), XantPhos (2.06 g, 3.57 mmol), and t-BuONa (3.43 g, 35.7 mmol) in toluene (80 mL) under N 2 . The mixture was degassed and purged with N 2 3 times, stirred at 100 °C overnight, allowed to cool to rt, poured into H 2 O (80 mL), and then extracted with EtOAc (3×80 mL). The combined organic layers were washed with brine (2×50 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by column chromatography (petroleum ether/ethyl acetate=20/1 to 1/1) to obtain 3-(benzyloxy)-N-(4-bromophenyl)-2,4-difluoro-6- nitroaniline (4.06 g, 35% over two steps; contains 3.2 g ~80% 1 H NMR purity and 3.0 g ~50% 1 H NMR purity) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 8.53 (s, 1H), 8.01 (d, 1H), 7.43-7.38 (m, 5H), 7.34 (d, 2H), 6.70 (d, 2H), 5.35 (s, 2H); LCMS: 435.0 [M+H] + . Step 3: 5-(Benzyloxy)-N1-(4-bromophenyl)-4,6-difluorobenzene-1,2-dia mine [00239] Iron powder (2.46 g, 44.12 mmol) was added to a solution of 3-(benzyloxy)-N-(4- bromophenyl)-2,4-difluoro-6-nitroaniline (3.20 g, 7.35 mmol), NH 4 Cl (2.36 g, 44.12 mmol) in EtOH (40 mL), and H 2 O (10 mL). The mixture was stirred at 80 °C for 1 h, allowed to cool to rt, and then filtered. The filtrate was concentrated under vacuum to obtain 5-(benzyloxy)- N1-(4-bromophenyl)-4,6-difluorobenzene-1,2-diamine (3 g) as a red oil. 1 H NMR (400 MHz, DMSO-d6): δ 7.37-7.36 (m, 5H), 7.24-7.20 (m, 3H), 6.41-6.35 (m, 3H), 5.15 (s, 2H), 4.95 (s, 2H); LCMS: 405.0 [M+H] + . Step 4: 6-(Benzyloxy)-1-(4-bromophenyl)-5,7-difluoro-1H-benzo[d][1,2 ,3]triazole [00240] Sulfuric acid (7.50 mL, 141 mmol) in H 2 O (55 mL) and NaNO2 (715 mg, 10.4 mmol) in H 2 O (5 mL) were added to a solution of 5-(benzyloxy)-N1-(4-bromophenyl)-4,6- difluorobenzene-1,2-diamine (3 g, crude) in THF (30 mL) at 0 °C. The mixture was stirred for 1 h, allowed to warm slowly to rt, poured into H 2 O (20 mL), and then extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (2×20 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by column chromatography (petroleum ether/ethyl acetate=20/1 to 5/1) to obtain 6-(benzyloxy)-1-(4-bromophenyl)-5,7-difluoro-1H- benzo[d][1,2,3]triazole (2.20 g, 89% over two steps) as a red solid. 1 H NMR (400 MHz, DMSO- d6): δ 8.08 (d, 1H), 7.88 (d, 2H), 7.77 (d, 2H), 7.46-7.44 (m, 2H), 7.41-7.36 (m, 3H), 5.26 (s, 2H); LCMS: 416.0 [M+H] + . Intermediate 2 (R)-3-Benzyl-1-(methylsulfonyl)piperazine TFA salt Step 1: tert-Butyl (R)-2-benzyl-4-(methylsulfonyl)piperazine-1-carboxylate [00241] Methanesulfonyl chloride (0.42 mL, 5.43 mmol) was added to a solution of tert-butyl (R)-2-benzylpiperazine-1-carboxylate (1.00 g, 3.62 mmol) and Et3N (1.51 mL, 10.8 mmol) in CH 2 Cl 2 (15 mL) at 0 °C. The mixture was allowed to warm to rt, stirred overnight, poured into H 2 O (20 mL) slowly, and then extracted with CH 2 Cl 2 (3×30 mL). The combined organic layers were washed with brine (70 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate=20/1 to 3/1)) to give tert-butyl (R)-2- benzyl-4-(methylsulfonyl)piperazine-1-carboxylate (1.05 g, 82%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 7.30-7.26 (m, 2H), 7.22-7.18 (m, 3H), 4.30 (s, 1H), 3.96 (d, 1H), 3.53 (d, 1H), 3.40 (d, 1H), 3.17 (t, 1H), 2.88-2.66 (m, 7H), 1.24 (s, 9H); LCMS: 255.2 [(M- Boc)+H] + . Step 2: (R)-3-Benzyl-1-(methylsulfonyl)piperazine TFA salt [00242] Trifluoroacetic acid (5.25 mL, 68.6 mmol) was added to a solution of tert-butyl (R)-2- benzyl-4-(methylsulfonyl)piperazine-1-carboxylate (1.05 g, 2.96 mmol) in CH 2 Cl 2 (15 mL) at rt. The mixture was stirred for 2 h and then concentrated to obtain (R)-3-benzyl-1- (methylsulfonyl)piperazine TFA salt (1.20 g) as a yellow oil. LCMS: 255.2 [M+H] + . [00243] The Intermediates below were synthesized from the appropriate Boc-protected piperazine following the procedures described for Intermediate 2. Alternate conditions used: Step 1: 2 h-ON. 1 Step 2: 4 M HCl in dioxane, dioxane or 1:1 dioxane/DCM, rt, 15-31 h. 2 Prepared from Intermediate 2.01 by partitioning between DCM and sat. aq NaHCO 3 . Intermediate 3 (R)-3-(4-Methoxybenzyl)morpholine Hydrochloride Step 1: tert-Butyl (R,Z)-3-((2-tosylhydrazineylidene)methyl)morpholine-4-carbox ylate [00244] A mixture of tert-butyl (S)-3-formylmorpholine-4-carboxylate (950 mg, 4.41 mmol), 4- methylbenzenesulfonohydrazide (870 mg, 4.67 mmol), and methanol (20 mL) was stirred at rt overnight, concentrated, and then purified by silica gel chromatography (10-60% ethyl acetate in heptane) to give tert-butyl (R,Z)-3-((2-tosylhydrazineylidene)methyl)morpholine-4-carbox ylate (1.62 g, 96%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.16 (s, 1H), 7.68 (d, J = 7.8 Hz, 2H), 7.38 (d, J = 7.9 Hz, 2H), 7.21 (br s, 1H), 4.39 (br s, 1H), 3.89 (br d, J = 11.5 Hz, 1H), 3.65 (br d, J = 11.0 Hz, 1H), 3.54-3.38 (m, 2H), 3.33-3.23 (m, 1H), 2.90-2.68 (m, 1H), 2.38 (s, 3H), 1.55-1.22 (m, 9H); LCMS: 284.3 [(M-Boc)+H] + . Step 2: tert-Butyl (R)-3-(4-methoxybenzyl)morpholine-4-carboxylate [00245] A mixture of tert-butyl (R,Z)-3-((2-tosylhydrazineylidene)methyl)morpholine-4- carboxylate (200 mg, 0.52 mmol), 4-methoxyphenylboronic acid (107 mg, 0.70 mmol), dioxane (1 mL), and potassium carbonate (208 mg, 1.51 mmol) was heated at 110 °C for 2 h, allowed to cool to rt, and then diluted with 20 mL EtOAc. The organic layer was washed with 15 mL water, washed with 15 mL brine, dried (Na 2 SO 4 ), concentrated, and then purified by silica gel chromatography (0-20% ethyl acetate in heptanes) to give tert-butyl (R)-3-(4- methoxybenzyl)morpholine-4-carboxylate (65 mg, 40%). Step 3: (R)-3-(4-Methoxybenzyl)morpholine Hydrochloride Hydrochloric acid (4 M in dioxane, 0.60 mL, 2.40 mmol) was added to a solution of tert-butyl (R)-3-(4-methoxybenzyl)morpholine-4-carboxylate (65 mg, 0.21 mmol) and CH 2 Cl 2 (1 mL) at rt. The mixture was stirred for 1.5 h, concentrated, and then dried on high vacuum overnight to give (R)-3-(4-methoxybenzyl)morpholine hydrochloride (45 mg) as an off-white solid. LCMS: 208.4 [M+H] + . [00246] The Intermediates below were synthesized from Intermediate 3, Step 1 and the appropriate boronic acid following the procedures described for Intermediate 3, Steps 2-3. Alternate conditions used: 1 Step 2: 80 °C, 30 min then 110 °C, 90 min. Step 2: 1-2 h. Step 3: 1-2 h. Intermediate 4 2,6-Difluoro-3-iodo-5-(trifluoromethyl)phenol Step 1: (2,6-Difluoro-3-(trifluoromethyl)phenyl)boronic Acid [00247] n-Butyllithium (2.5 M in hexanes, 171 mL, 428 mmol) was added dropwise to a mixture of 2,4-difluoro-1-(trifluoromethyl)benzene (60.0 g, 329 mmol) in Et2O (~400 mL) at - 78 °C under N 2 . The reaction was stirred for 1 h. Trimethyl borate (44.7 mL, 395 mmol) in Et2O (200 mL) was added dropwise at -78 °C. The reaction was stirred for 1 h, allowed to warm to rt, stirred for 10 h, and then quenched slowly with aq. HCl (1 M, 500 mL) under ice cooling. The organic layer was separated and washed with brine (300 mL) to give (2,6-difluoro-3- (trifluoromethyl)phenyl)boronic acid as a solution in Et2O (~600 mL). LCMS: 225.1 [M-H]-. Step 2: 2,6-Difluoro-3-(trifluoromethyl)phenol [00248] Hydrogen peroxide (166 mL, 1.72 mol, 30% purity in H 2 O) was added to a solution of (2,6-difluoro-3-(trifluoromethyl)phenyl)boronic acid (74.4 g, 329 mmol) in Et2O (~600 mL) at 0 °C. The mixture was heated to 40 °C, stirred for 4 h, and then allowed to cool to rt. The aqueous layer was separated. The organic layer was cooled to 0 °C and then quenched with aqueous Na 2 O3 (20% in H 2 O, ~500 mL) keeping the temperature <20 °C. The organic layer was separated. The aqueous layer was extracted with EtOAc (2×300 ml). The organic layers were combined, washed with water (2×300 ml), washed with brine (300 ml), dried (Na 2 SO 4 ), filtered, concentrated and then purified by silica gel chromatography (petroleum ether/ethyl acetate=50:1 to 5:1) to give 2,6-difluoro-3-(trifluoromethyl)phenol (41.3 g, 63%) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6): δ 11.01 (s, 1H), 7.27-7.19 (m, 2H); LCMS: 196.9 [M-H]-. Step 3: 2-(Benzyloxy)-1,3-difluoro-4-(trifluoromethyl)benzene [00249] Benzyl bromide (43.2 mL, 363 mmol) was added to a mixture of 2,6-difluoro-3- (trifluoromethyl)phenol (60.3 g, 303 mmol), K 2 CO 3 (126 g, 909 mmol), and DMF (600 mL) at rt. The mixture was stirred at 50 °C for 12 h, cooled to rt, poured into H 2 O (500 mL) slowly, and then extracted with EtOAc (3×300 mL). The organic layers were combined, washed with brine (300 mL), dried (Na 2 SO 4 ), filtered, concentrated and then purified by silica gel chromatography (petroleum ether/ethyl acetate=100:1 to 10:1) to give 2-(benzyloxy)-1,3-difluoro-4- (trifluoromethyl)benzene (54.5 g, 62%) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6): δ 7.56- 7.50 (m, 1H), 7.43-7.34 (m, 6H), 5.24 (s, 2H). Step 4: 3-(Benzyloxy)-2,4-difluoro-1-iodo-5-(trifluoromethyl)benzene [00250] n-Butyllithium (2.5 M in hexanes, 104 mL, 260 mmol) was added dropwise to a mixture of 2-(benzyloxy)-1,3-difluoro-4-(trifluoromethyl)benzene (50.1 g, 173 mmol) in THF (300 mL) at -78 °C under N2. The mixture was stirred for 1 h. Iodine (88.1 g, 347 mmol) in THF (200 mL) was added dropwise into the mixture at -78 °C. The mixture was allowed to warm to rt, stirred for 12 h, diluted with sat. aq. Na 2 O3 (500 mL), and then extracted with EtOAc (3×300 mL). The combined organic layers were washed with brine (300 mL), dried (Na 2 SO 4 ), filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/EtOAc=1/0) to give 3-(benzyloxy)-2,4-difluoro-1-iodo-5-(trifluoromethyl)benzene (64.3 g, 89%) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6): δ 7.93 (t, 1H), 7.44-7.35 (m, 5H), 5.25 (s, 2H). Step 5: 2,6-Difluoro-3-iodo-5-(trifluoromethyl)phenol [00251] Boron tribromide (58.2 mL, 604 mmol) was added dropwise to a mixture of 3- (benzyloxy)-2,4-difluoro-1-iodo-5-(trifluoromethyl)benzene (50.2 g, 121 mmol) in CH 2 Cl 2 (500 mL) at -78 °C under N 2 . The mixture was stirred at rt for 4 h, quenched slowly with CH 3 OH (~200 mL) at 0 °C, concentrated, and then purified by silica gel chromatography (petroleum ether/EtOAc =50:1 to 5:1) to give 2,6-difluoro-3-iodo-5-(trifluoromethyl)phenol (34.6 g, 86%) as a red solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.39 (s, 1H), 7.59 (t, 1H); LCMS: 322.9 [M- H]-. [00252] The Intermediate below was synthesized from 2-chloro-4-fluoro-1- (trifluoromethyl)benzene following the procedures described for Intermediate 4. Alternate conditions used: Step 5: TFA, 70 °C, 2 h. Intermediate 5 3-Bromo-6-chloro-2-fluoro-5-(trifluoromethyl)phenol [00253] 1,3-Dichloro-5,5-dimethylhydantoin (5.52 g, 19.31 mmol) was added to the mixture of 3-bromo-2-fluoro-5-(trifluoromethyl)phenol (5.00 g, 19.3 mmol) and diisopropylamine HCl (0.03 g, 0.19 mmol) in toluene at 0 °C. The yellow suspension was stirred at 0 °C in the absence of light for 2 h, diluted with water, and then extracted with ethyl acetate. The organic layer was dried (MgSO 4 ), concentrated, and then purified by silica gel chromatography (0-50% CH 2 Cl 2 in heptane). The crude material was purified further by prep-HPLC (40-100% CH 3 CN in water with 0.1% TFA). The fractions were combined, concentrated, diluted with ethyl acetate, and then washed with NaHCO 3 . The aqueous layer was back extracted with ethyl acetate. The combined organics were washed with brine, dried (MgSO 4 ), filtered, and then concentrated to give 3-bromo-6-chloro-2-fluoro-5-(trifluoromethyl)phenol (3.3 g, 55%) as a white semi-solid. 1 H NMR (400 MHz, DMSO-d6): δ 6.49-6.42 (m, 1H). Intermediate 6 2-(2,4-Difluoro-3-(methoxymethoxy)-5-(trifluoromethyl)phenyl )-4,4,5,5-tetramethyl-1,3,2- dioxaborolane Step 1: 2-(3-Bromo-2,6-difluoro-5-(trifluoromethyl)phenyl)-4,4,5,5-t etramethyl-1,3,2- dioxaborolane [00254] A mixture of (1,5-cyclooctadiene)(methoxy)iridium(I) dimer (1.13 g, 1.71 mmol), 4,4'- di-tert-butyl-2,2'-bipyridine (0.46 g, 1.71 mmol), and bis(pinacolato)diboron (23.9 g, 94 mmol) was evacuated and refilled with nitrogen 3 times. Cyclopentyl methyl ether (90 mL) was added, and the mixture was evacuated and purged with nitrogen an additional 3 times.4-Bromo-1,5- difluoro-2-(trifluoromethyl)benzene (22.3 g, 85 mmol) was added. The reaction was heated at 100 ℃ overnight, cooled to rt, concentrated under reduced pressure, and then purified by silica gel chromatography (0-20% EtOAc/heptane) to give 2-(3-bromo-2,6-difluoro-5- (trifluoromethyl)phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaboro lane (29.3 g, 84%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 8.32 (t, J = 7.4 Hz, 1H), 1.32 (s, 12H). Step 2: 3-Bromo-2,6-difluoro-5-(trifluoromethyl)phenol [00255] Hydrogen peroxide (30 w/w in H 2 O, 69 mL) was slowly added to a solution of 2-(3- bromo-2,6-difluoro-5-(trifluoromethyl)phenyl)-4,4,5,5-tetram ethyl-1,3,2-dioxaborolane (23.6 g, 61 mmol) in methanol (240 mL). The clear solution was stirred at rt for 5 h, quenched by the slow addition of saturated aqueous Na 2 S 2 O 3 solution over ~1 h, stirred for 30 min, and then extracted twice with EtOAc. The combined organic layers were washed with brine, dried (MgSO 4 ), filtered, concentrated under reduced pressure, and then purified by silica gel chromatography (0-20% EtOAc/heptane) to give 3-bromo-2,6-difluoro-5- (trifluoromethyl)phenol (16.9 g, 73%) as a semi-solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.62 (s, 1H), 7.56 (t, J = 6.8 Hz, 1H). Step 3: 1-Bromo-2,4-difluoro-3-(methoxymethoxy)-5-(trifluoromethyl)b enzene [00256] Chloromethyl methyl ether (0.51 mL, 6.77 mmol) and DIEA (1.57 mL, 9.0 mmol) were added to a solution of 3-bromo-2,6-difluoro-5-(trifluoromethyl)phenol (1.25 g, 4.51 mmol) in CH 2 Cl 2 (10 mL) at 0 ℃. The reaction was stirred at rt overnight, diluted with water, and then extracted with CH 2 Cl 2 . The aqueous layer was extracted with CH 2 Cl 2 . The combined organics were dried (MgSO 4 ), filtered, concentrated under reduced pressure, and then purified by silica gel chromatography (0-20% EtOAc/heptane) to give 1-bromo-2,4-difluoro-3- (methoxymethoxy)-5-(trifluoromethyl)benzene (0.89 g, 58%) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6): δ 7.99 (t, J = 7.0 Hz, 1H), 5.26 (s, 2H), 3.51 (s, 3H). Step 4: 2-(2,4-Difluoro-3-(methoxymethoxy)-5-(trifluoromethyl)phenyl )-4,4,5,5- tetramethyl-1,3,2-dioxaborolane [00257] A mixture of 1-bromo-2,4-difluoro-3-(methoxymethoxy)-5-(trifluoromethyl)b enzene (0.47 g, 1.46 mmol), bis(pinacolato)diboron (558 mg, 2.2 mmol), and KOAc (287 mg, 2.92 mmol) in dioxane (5 mL) was evacuated and filled with nitrogen 3 times. Pd(dppf)Cl 2 (54 mg, 0.07 mmol) was added. The mixture was degassed and filled with nitrogen 3 more times, heated at 80 ℃ for 3 days, cooled to rt, concentrated under reduced pressure, and then purified by silica gel chromatography (0-20% EtOAc/heptanes) to give 2-(2,4-difluoro-3-(methoxymethoxy)-5- (trifluoromethyl)phenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaboro lane (0.36 g, 63%) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6): δ 7.62 (br t, J = 6.7 Hz, 1H), 5.21 (s, 2H), 3.50 (s, 3H), 1.44-1.25 (m, 12H). [00258] The Intermediates below were synthesized from the appropriate Intermediate or starting material following the procedures described for Intermediate 6.

Alternate conditions used: Step 1: 16-22 h. Step 2: 5-49 h. Step 3: 1 h-ON. Step 4: 85 °C, 0.5-1 h. 1 Synthesized from Intermediate 5 following the procedure described for Intermediate 6, Steps 3-4. 2 Isolated during the purification of Intermediate 6.01. 3 Synthesized from Intermediate 4.01 following the procedure described for Intermediate 6, Step 4 (80 °C for 16 h in sealed reactor). 4 Steps 1-2 only from 4- bromo-2-chloro-1,5-difluorobenzene. Intermediate 7 2-(3-(Benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-4,4 ,5,5-tetramethyl-1,3,2- dioxaborolane [00259] (1,5-Cyclooctadiene)(methoxy)iridium(I) dimer (92.1 mg, 0.139 mmol) and 4,4'-di- tert-butyl-2,2'-bipyridine (37.2 mg, 0.139 mmol) were added to a solution of Intermediate 4, Step 3 (4.01 g, 13.88 mmol) and 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (2.93 g, 11.52 mmol) in THF (40 mL) under N 2 . The mixture was stirred at 80 °C for 4 h, cooled to rt, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate =100/1 to 10/1) to give 2-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-4,4 ,5,5- tetramethyl-1,3,2-dioxaborolane (5.3 g, 92%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 7.45 (t, 1H), 7.43-7.38 (m, 5H), 5.22 (s, 2H), 1.31 (s, 12H). [00260] The Intermediate below was synthesized from 2,6-difluoro-3-(trifluoromethyl)phenol (Intermediate 4, Step 2) using the following sequence: Intermediate 6 (Step 3) and then Intermediate 7. Intermediate 7.01 2,4-Difluoro-3-methoxy-5-(tributylstannyl)benzonitrile Step 1: 2,4-Difluoro-3-methoxybenzonitrile [00261] CuCN (460 mg, 5.13 mmol) was added to a solution of 1,3-difluoro-4-iodo-2-methoxy- benzene (0.99 g, 3.67 mmol) and DMF (15 mL). The mixture was stirred at 120 °C for 22 h, cooled to rt, diluted with H 2 O (120 mL), and then extracted with EtOAc (120 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 99/1 to 82/18) to give 2,4-difluoro-3- methoxy-benzonitrile (293 mg, 47%) as a yellow solid. 1 H NMR (400 MHz, CD 3 OD): δ 7.40- 7.60 (m, 1H), 7.10-7.26 (m, 1H), 4.00-4.10 (s, 3H). Step 2: 2,4-Difluoro-3-methoxy-5-(tributylstannyl)benzonitrile [00262] n-BuLi (2 M in n-hexane, 950 µL) was added dropwise to a solution of i-Pr2NH (293 µL, 2.07 mmol) and THF (4.9 mL) maintaining the temperature below -65 °C. The reaction was stirred for 10 min and then a solution of 2,4-difluoro-3-methoxy-benzonitrile (292 mg, 1.73 mmol) and THF (10 mL) was added over 20 min at -65 °C. The reaction was stirred at -65 °C for 30 min, and then tributyl(chloro)stannane (1.03 mL, 3.82 mmol) was added. The mixture was stirred at -65 ℃ for 1 h, warmed to rt, poured into sat. aq. KF (15 mL), and then extracted with EtOAc (15 mL × 3). The combined organic layers were washed with brine (15 mL × 2), dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 1/0 to 97/3) to give 2,4-difluoro-3-methoxy-5-tributylstannyl- benzonitrile (522 mg, 66%) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ): δ 7.10-7.22 (m, 1H), 3.92-3.98 (m, 3H), 1.39-1.51 (m, 6H), 1.21-1.28 (m, 6H), 1.01-1.12 (m, 6H), 0.80-0.84 ppm (m, 9H). [00263] The Intermediate below was synthesized from 2-(benzyloxy)-1-chloro-3,5- difluorobenzene in a similar manner to that described in Intermediate 7.01, Step 2. Intermediate 8 5-Chloro-3-methyl-1H-pyrazolo[4,3-d]pyrimidine Step 1: 5-Chloro-3-iodo-1H-pyrazolo[4,3-d]pyrimidine [00264] A mixture of 5-chloro-1H-pyrazolo[4,3-d]pyrimidine (8.83 g, 57.1 mmol), N- iodosuccinimide (3.86 g 17.1 mmol), and DMF (90 mL) was stirred at rt overnight. Additional N-iodosuccinimide (3.86 g 17.1 mmol) was added. The reaction was stirred overnight. Additional N-iodosuccinimide (3.86 g 17.1 mmol) was added. The reaction was stirred for 1.5 h. Water was added. The reaction was extracted with 10% CH 3 OH in CH 2 Cl 2 (× 3). The combined organics were dried (MgSO 4 ), filtered, and concentrated. Toluene was added, and then the solution was concentrated to remove DMF. Water (500 mL) was added. Solids formed. The mixture was filtered, and the filter cake was washed with water. The solids were dried on a lyophilizer to give 5-chloro-3-iodo-1H-pyrazolo[4,3-d]pyrimidine (12.3 g, 73%) as a beige solid. 1 H NMR (400 MHz, DMSO-d6): δ 14.57 (br s, 1H), 9.31 (s, 1H); LCMS: 280.7 [M+H] + . Step 2: 5-Chloro-3-methyl-1H-pyrazolo[4,3-d]pyrimidine [00265] A mixture of 5-chloro-3-iodo-1H-pyrazolo[4,3-d]pyrimidine (1.00 g, 3.57 mmol), 2,4,6-trimethyl-1,3,5,2,4,6-trioxatriborinane (5.98 mL, 21.4 mmol, 50% w/w soln. in THF), Pd(dppf)Cl 2 (0.39 g, 0.53 mmol), K3PO4 (2.27 g, 10.7 mmol), and dioxane (6 mL) was degassed/backfilled with N 2 three times, heated in a microwave at 120 °C for 90 min, allowed to cool to rt, and then diluted with ethyl acetate. The organic layer was washed with water. The aqueous layer was extracted with ethyl acetate. The combined organic layers were dried (MgSO 4 ), filtered, and then concentrated. The residue was dissolved in CH 2 Cl 2 and 1 mL CH 3 OH and purified via silica gel chromatography (0-50% ethyl acetate in heptane) to give 5- chloro-3-methyl-1H-pyrazolo[4,3-d]pyrimidine (0.16 g, 25%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 13.76 (br s, 1H), 9.24 (s, 1H), 2.52 (s, 3H); LCMS: 168.8 [M+H] + . Intermediate 8.01 5-Bromo-6-fluoro-3-methyl-1H-pyrazolo[4,3-b]pyridine Step 1: 1-(3-Amino-5-fluoropyridin-2-yl)ethanone Bromo(methyl)magnesium (14.1 mL, 3 M in THF) was added dropwise over 0.5 h to a solution of 3-amino-5-fluoropicolinonitrile (2.90 g, 21.2 mmol) and THF (30 mL) at -15 °C under N 2 . The mixture was stirred at -15 °C for 0.5 h, stirred at rt for 5 h, poured into sat. aq. NH4Cl (100 mL), and then extracted with ethyl acetate (50 mL × 3). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 10/1 to 4/1) to give 1-(3-amino-5- fluoropyridin-2-yl)ethanone (2.10 g, 64%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 7.84 (d, 1H), 7.55-7.24 (m, 2H), 7.00 (d, 1H), 2.53 (s, 3H); LCMS: 155.2 [M+H] + . Step 2: 1-(3-Amino-6-bromo-5-fluoropyridin-2-yl)ethenone NBS (2.54 g, 14.3 mmol) was added to a solution of 1-(3-amino-5-fluoropyridin-2-yl)ethanone (2.00 g, 13.0 mmol) and MeCN (40 mL). The mixture was stirred at rt for 3 h, poured into sat. aq. Na 2 S 2 O3 (80 mL), and then extracted with ethyl acetate (40 mL × 3). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and concentrated to give 1- (3-amino-6-bromo-5-fluoropyridin-2-yl)ethanone (3.40 g) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 7.75-7.39 (m, 2H), 7.17 (d, 1H), 2.57 (s, 3H). LCMS: 233.0 [M+H] + . Step 3: 5-Bromo-6-fluoro-3-methyl-1H-pyrazolo[4,3-b]pyridine [00266] A solution of NaNO 2 (1.07 g, 15.6 mmol) and H 2 O (10 mL) was added dropwise over 0.5 h to a solution of 1-(3-amino-6-bromo-5-fluoropyridin-2-yl)ethanone (3.30 g, 14.2 mmol) and HCl (50 mL, 12 M) at 0 °C. The mixture was stirred at 0 °C for 0.5 h, and then a solution of SnCl 2 •2H 2 O (7.35 g, 32.6 mmol) in HCl (20 mL, 12 M) was added dropwise. The reaction was stirred at 0 °C for 1 h, poured into sat. aq. NaHCO 3 (1 L) slowly, and then extracted with ethyl acetate (100 mL × 3). The combined organic layers were washed with brine (100 mL), dried over Na 2 SO 4 , filtered, and concentrated to give 5-bromo-6-fluoro-3-methyl-1H-pyrazolo[4,3- b]pyridine (2.20 g) as a light yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 13.23 (s, 1H), 8.08 (d, 1H), 2.53-2.51 (m, 3H); LCMS: 230.0 [M+H] + . Intermediate 9 (R)-5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-methyl-1 H-pyrazolo[4,3-d]pyrimidine Step 1: 5-Chloro-3-methyl-2-((2-(trimethylsilyl)ethoxy)methyl)-2H-py razolo[4,3- d]pyrimidine [00267] (2-(Chloromethoxy)ethyl)trimethylsilane (18.3 mL, 103 mmol) was added dropwise to a mixture of Intermediate 8 (11.6 g, 68.8 mmol) and C s2 CO 3 (56.1 g, 172 mmol) in DMF (200 mL) at 0 °C. The mixture was stirred at rt for 2 h, poured into H 2 O (200 mL) slowly, and then filtered. The filtrate was extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (100 mL), dried (Na 2 SO 4 ), filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/EtOAc=30:1 to 3:1) to give 5-chloro-3-methyl-2-((2- (trimethylsilyl)ethoxy)methyl)-2H-pyrazolo[4,3-d]pyrimidine (5.32 g, 24%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 9.51 (s, 1H), 5.87 (s, 2H), 3.58 (t, 2H), 2.67 (s, 3H), 0.84 (s, 2H), -0.08 (s, 9H); LCMS: 299.0 [M+H] + . Step 2: (R)-5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-methyl-1 H-pyrazolo[4,3- d]pyrimidine [00268] A mixture of 5-chloro-3-methyl-2-((2-(trimethylsilyl)ethoxy)methyl)-2H-py razolo[4,3- d]pyrimidine (650 mg, 2.18 mmol), Intermediate 2.01 (696 mg, 2.39 mmol), potassium fluoride (379 mg, 6.53 mmol), and water (10 mL) was stirred at 100 °C overnight, allowed to cool to rt, and then diluted with EtOAc. The layers were separated, and the organic layer was dried (MgSO 4 ), filtered, and then concentrated. The residue was dissolved in CH 2 Cl 2 /TFA (2:1, 15 mL). The reaction was stirred at rt for 2 h and concentrated to give a mixture of (R)-5-(2-benzyl- 4-(methylsulfonyl)piperazin-1-yl)-3-methyl-1H-pyrazolo[4,3-d ]pyrimidine and (R)-(5-(2- benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-methyl-1H-pyrazol o[4,3-d]pyrimidin-1- yl)methanol. The mixture was dissolved in 10 mL CH 2 Cl 2 . Ethylenediamine (1 mL) was added, and the reaction was stirred over the weekend. Additional ethylendiamine (1 mL) was added. The reaction was stirred for 6 h and then diluted with water. The layers were separated, and the aqueous layer was extracted with EtOAc. The combined organic layers were dried (MgSO 4 ), filtered, concentrated, and then purified by silica gel chromatography (0-100% EtOAc in heptane) to give (R)-5-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-methyl-1 H-pyrazolo[4,3- d]pyrimidine (110 mg, 12%) as an off-white solid. 1 H NMR (400 MHz, DMSO-d6): δ 12.97 (s, 1H), 8.99 (s, 1H), 7.39-7.25 (m, 4H), 7.23-7.16 (m, 1H), 5.04-4.89 (m, 1H), 4.69 (br d, J = 13.1 Hz, 1H), 3.70 (br d, J = 10.6 Hz, 1H), 3.47 (br d, J = 11.5 Hz, 1H), 3.30-3.20 (m, 1H), 3.07 (br t, J = 11.2 Hz, 1H), 2.95-2.75 (m, 5H), 2.67 (br d, J = 10.5 Hz, 1H), 2.46 (s, 3H); LCMS: 387.5 [M+H] + . Intermediate 10 (R)-5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-chloro-1 H-pyrazolo[4,3-d]pyrimidine Step 1: 3,5-Dichloro-1H-pyrazolo[4,3-d]pyrimidine [00269] N-Chlorosuccinimide (1.04 g, 7.76 mmol) was added to a solution of 5-chloro-1H- pyrazolo[4,3-d]pyrimidine (1.00 g, 6.47 mmol) in DMF (20 mL). The reaction was stirred at rt for 18.5 h, diluted with ethyl acetate and water. The organic layer was dried (MgSO 4 ), filtered, concentrated, and then purified by silica gel chromatography (0-50% ethyl acetate in heptane) to give 3,5-dichloro-1H-pyrazolo[4,3-d]pyrimidine (1.01 g, 78%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 14.55-14.34 (m, 1H), 9.39 (s, 1H); LMCS: 189.2 [M+H] + . Step 2: 3,5-Dichloro-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol o[4,3-d]pyrimidine [00270] Sodium hydride (158 mg, 4.11 mmol) was added slowly to a mixture of 3,5-dichloro- 1H-pyrazolo[4,3-d]pyrimidine (432 mg, 2.29 mmol) and SEM-Cl (0.73 mL, 4.11 mmol) in THF (6 mL) at 0 °C. The reaction was stirred at rt for 30 min, quenched with water, and then extracted with ethyl acetate (twice). The combined organic layers were washed with brine, dried (MgSO 4 ), filtered, concentrated, and then purified by silica gel chromatography (0-50% ethyl acetate in heptane) to give 3,5-dichloro-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol o[4,3- d]pyrimidine (328 mg, 45%) as an off-white semi-solid. 1 H NMR (400 MHz, DMSO-d 6 ): δ 9.63 (s, 1H), 5.86 (s, 2H), 3.55-3.50 (m, 2H), 0.84-0.80 (m, 2H), -0.08 (s, 9H); LMCS: 319.4 [M+H] + . Step 3: (R)-5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-chloro-1 H-pyrazolo[4,3- d]pyrimidine [00271] A mixture of 3,5-dichloro-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol o[4,3- d]pyrimidine (100 mg, 0.313 mmol), Intermediate 2.01 (119 mg, 0.41 mmol), DIEA (0.21 mL, 1.25 mmol), and NMP (1 mL) was stirred at 140 °C overnight, allowed to cool to rt, and then diluted with EtOAc. The organic layer was washed with water, and the aqueous layer was extracted with EtOAc. The combined organic layers were dried (MgSO 4 ), filtered, concentrated, and then purified by silica gel chromatography (0-100% EtOAc in heptane). The residue was dissolved in 2 mL CH 2 Cl 2 and 1 mL TFA and then stirred at rt for 4 h. Additional trifluoroacetic acid (1 mL) was added. The reaction was stirred overnight, concentrated, dissolved in a mixture of EtOAc/heptane, and then concentrated to give crude (R)-5-(2-benzyl-4- (methylsulfonyl)piperazin-1-yl)-3-chloro-1H-pyrazolo[4,3-d]p yrimidine (17 mg, 13%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 12.73-11.49 (m, 1H), 9.08 (s, 1H), 7.41-7.32 (m, 2H), 7.32-7.24 (m, 2H), 7.22-7.14 (m, 1H), 4.97 (br d, J = 4.4 Hz, 1H), 4.68 (br d, J = 13.3 Hz, 1H), 3.71 (br d, J = 9.8 Hz, 1H), 3.48 (br d, J = 11.6 Hz, 1H), 3.33-3.25 (m, 1H), 3.12-3.01 (m, 1H), 2.89 (s, 3H), 2.88-2.80 (m, 2H), 2.76-2.68 (m, 1H); LCMS: 407.5 [M+H] + . [00272] The Intermediate below was synthesized from 5-chloro-1H-pyrazolo[4,3-d]pyrimidine following the procedure described for Intermediate 10, Step 2-3. Alternate conditions used: 1 Step 3: DIEA, NMP, 130 °C for 3 h, 140 °C for ON, and then 150 °C, 3 h; CH 2 Cl 2 /TFA (2:1), rt, overnight. Intermediate 11 3-Benzyl-4-(3-methyl-1H-pyrazolo[3,4-c]pyridin-5-yl)morpholi ne Step 1: 5-Bromo-3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[3, 4-c]pyridine [00273] A suspension of 5-bromo-3-methyl-1H-pyrazolo[3,4-c]pyridine (5.14 g, 24.2 mmol), 2H-3,4-dihydropyran (6.6 mL, 72.7 mmol), p-toluenesulfonic acid (0.55 g, 2.91 mmol), and CH 2 Cl 2 (50 mL) was stirred at rt for overnight. Additional 2H-3,4-dihydropyran (4.42 mL, 48.5 mmol) was added. The reaction was stirred at rt overnight and then diluted with NaHCO 3 . The organic layer was dried (MgSO 4 ), filtered, concentrated, and then purified by silica gel chromatography (0-100% ethyl acetate in heptane) to give 5-bromo-3-methyl-1-(tetrahydro-2H- pyran-2-yl)-1H-pyrazolo[3,4-c]pyridine (6 g, 83%) as an off-white semi-solid. 1 H NMR (400 MHz, DMSO-d6): δ 8.99 (d, J = 1.1 Hz, 1H), 8.08 (d, J = 1.1 Hz, 1H), 5.90 (dd, J = 2.4, 9.7 Hz, 1H), 3.94-3.81 (m, 1H), 3.79-3.67 (m, 1H), 2.50 (s, 3H), 2.07-1.93 (m, 2H), 1.79-1.54 (m, 4H); LCMS: 298.1 [M+H] + . Step 2: 3-Benzyl-4-(3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol o[3,4-c]pyridin-5- yl)morpholine [00274] A mixture of 5-bromo-3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[3, 4- c]pyridine (70 mg, 0.24 mmol), 3-benzylmorpholine (63 mg, 0.35 mmol), sodium tert-butoxide (68 mg, 0.71 mmol), Pd2(dba)3, and RuPhos (11 mg, 0.024 mmol) was degassed/backfilled with N 2 three times. Dioxane (2 mL) was added. The reaction was degassed/backfilled with N 2 for 3 min, stirred at 80 °C for 3 h, allowed to cool to rt, and then diluted with CH 2 Cl 2 and water. The organic layer was dried (MgSO 4 ), filtered, concentrated, and then purified by silica gel chromatography (0-50% ethyl acetate in heptane) to give 3-benzyl-4-(3-methyl-1-(tetrahydro- 2H-pyran-2-yl)-1H-pyrazolo[3,4-c]pyridin-5-yl)morpholine (56 mg, 60%) as a yellow oil. LCMS: 393.4 [M+H] + . Step 3: 3-Benzyl-4-(3-methyl-1H-pyrazolo[3,4-c]pyridin-5-yl)morpholi ne [00275] 3-Benzyl-4-(3-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol o[3,4-c]pyridin-5- yl)morpholine (56 mg, 0.24 mmol) was dissolved in 4 mL CH 2 Cl 2 and 2 mL TFA. The solution was stirred at rt overnight and then concentrated to give 3-benzyl-4-(3-methyl-1H-pyrazolo[3,4- c]pyridin-5-yl)morpholine (38 mg, 49%) as a yellow oil. LCMS: 309.3 [M+H] + . [00276] The Intermediates below were synthesized from the appropriate Intermediate or starting material following the procedures described for Intermediate 11. Alternate conditions used: Step 1: reaction time was 1-48 h. Step 2: reaction time was 3 h-overnight. Step 3: reaction time was 2 h-overnight. In some instances, additional t-BuONa, Pd2(dba)3, and RuPhos was needed. 1 Synthesized from 3-acetyl-5-bromo-1H-indazole using the following sequence: Step 1, fluorination (DAST, CH 2 Cl 2 , 50 °C, 3 days then rt, 10 days), Step 2, and then Step 3. 2 Isolated as a byproduct of Intermediate 11.09 (Step 2) and then deprotected following the procedure for Step 3. 3 Step 2: Xantphos, Xantphos Pd G4, Cs 2 CO 3 , toluene, 80 °C, 2 h. Intermediate 11.13 (R)-3-Benzyl-4-(3-methyl-1H-pyrazolo[4,3-d]pyrimidin-5-yl)mo rpholine (R)-3-Benzyl-4-(3-methyl-1H-pyrazolo[4,3-d]pyrimidin-5-yl)mo rpholine was synthesized from Intermediate 8 (Step 1) using the following sequence: Intermediate 10 (Step 2), Intermediate 8 (Step 2), Buchwald coupling with (R)-3-benzylmorpholine (Pd(OAc) 2 , BINAP, Cs2CO 3 , 100 °C, 1 4 h), and then deprotection (2:1 CH2Cl2/TFA, rt, 2 h). 1H NMR (400 MHz, DMSO-d6): δ 12.92 (s, 1H), 8.95 (s, 1H), 7.33-7.16 (m, 5H), 4.66-4.64 (m, 1H), 4.37 (d, 1H), 4.03-3.99 (m, 1H), 3.64 (d, 1H), 3.55-3.45 (m, 2H), 3.31-3.24 (m, 1H), 3.08-3.03 (m, 1H), 2.71 (dd, 1H), 2.45 (s, 3H); LCMS: 310.2 [M+H] + . Intermediate 11.14 (R)-5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-6-chloro-3 -methyl-1H-pyrazolo[4,3- b]pyridine (R)-5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-6-chloro-3 -methyl-1H-pyrazolo[4,3- b]pyridine was synthesized from 5-chloro-1H-pyrazolo[4,3-b]pyridine using the procedures described for Intermediate 8 (Step 1), Intermediate 11 (Step 1), Intermediate 8 (Step 2; 90 °C, overnight), Intermediate 11 (Step 2), Intermediate 10 (Step 1; CH 3 CN, 80 °C, 2 h), and then Intermediate 11 (Step 3; 1:1 TFA/CH 2 Cl 2 ). 1 H NMR (400 MHz, DMSO-d6): δ 12.86 (s, 1H), 8.09 (s, 1H), 7.26-7.14 (m, 5H), 4.03 (q, 3H), 3.59-3.45 (m, 2H), 3.22-3.06 (m, 2H), 2.97-2.91 (m, 4H), 2.86-2.80 (m, 1H), 1.99 (s, 3H); LCMS: 420.1 [M+H] + . Intermediate 12 6-Chloro-3-iodo-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[4,3 -c]pyridine [00277] 6-Chloro-3-iodo-1H-pyrazolo[4,3-c]pyridine (2.50 g, 8.91 mmol) was weighed into a 20 mL microwave vial. Tetrahydrofuran (5 mL), 2H-3,4-dihydropyran (1.47 mL, 16.1 mmol), and methanesulfonic acid (70 µL, 1.07 mmol) were added. The reaction was heated in a microwave at 90 °C for 1 h, allowed to cool to rt, diluted with 10 mL CH 2 Cl 2 , washed with saturated NaHCO 3 , concentrated, and then purified by silica gel chromatography (0-50% EtOAc in hexanes to give 6-chloro-3-iodo-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[4,3 -c]pyridine (1.69 g, 87%) as an off-white solid. 1 H NMR (400MHz, DMSO-d6): δ 8.65 (s, 1H), 8.00 (s, 1H), 5.90 (br d, J = 8.1 Hz, 1H), 3.89 (br d, J = 11.4 Hz, 1H), 3.81-3.70 (m, 1H), 2.37-2.26 (m, 1H), 2.04-1.94 (m, 2H), 1.76-1.64 (m, 1H), 1.64-1.53 (m, 2H); LCMS: 364.1 [M+H] + . [00278] The Intermediate below was synthesized from 6-chloro-3-iodo-1H-pyrazolo[3,4- d]pyrimidine following the procedure described for Intermediate 12. Alternate conditions used: 1 DHP, p-toluenesulfonic acid monohydrate, CH 2 Cl 2 , rt, 15 h. Intermediate 13 6-Chloro-3-(2,4-difluoro-3-(methoxymethoxy)-5-(trifluorometh yl)phenyl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidine Step 1: 6-Chloro-3-iodo-1-methyl-1H-pyrazolo[3,4-d]pyrimidine [00279] Iodomethane (7.99 mL, 128 mmol) was added to a mixture of 6-chloro-3-iodo-1H- pyrazolo[3,4-d]pyrimidine (20.1 g, 71.3 mmol) and K 2 CO 3 (29.6 g, 214 mmol) in DMF (200 mL) at 0 °C. The mixture was stirred at rt for 12 h, poured into H 2 O (300 mL) slowly, and then extracted with EtOAc (3×300 mL). The combined organic layers were washed with brine (300 mL), dried (Na 2 SO 4 ), filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate=30:1 to 3:1) to give 6-chloro-3-iodo-1-methyl-1H-pyrazolo[3,4- d]pyrimidine (15.2 g, 72%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 9.01 (s, 1H), 4.00 (s, 3H); LCMS: 294.9 [M+H] + . Step 2: 6-Chloro-3-(2,4-difluoro-3-(methoxymethoxy)-5-(trifluorometh yl)phenyl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidine [00280] Pd(dppf)Cl 2 .CH 2 Cl 2 (2.77 g, 3.40 mmol) was added to a mixture of 6-chloro-3-iodo-1- methyl-1H-pyrazolo[3,4-d]pyrimidine (10.0 g, 34.0 mmol), Intermediate 6 (15.0 g, 40.8 mmol), Na 2 CO 3 (2 M in H 2 O, 42.5 mL, 84.9 mmol), and dioxane (120 mL) under N 2 . The mixture was degassed and purged with N 2 3 times, stirred at 80 °C for 1 h, allowed to cool to rt, diluted with H 2O (500 mL), and then extracted with EtOAc (2×300 mL). The combined organic layers were washed with brine (300 mL), dried (Na 2 SO 4 ), filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/EtOAc=30:1 to 3:1) to give 6-chloro-3-(2,4-difluoro-3- (methoxymethoxy)-5-(trifluoromethyl)phenyl)-1-methyl-1H-pyra zolo[3,4-d]pyrimidine (13.8 g, 90%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 9.39 (d, 1H), 7.99 (t, 1H), 5.35 (s, 2H), 4.12 (s, 3H), 3.56 (s, 3H); LCMS: 409.0 [M+H] + . [00281] The Intermediates below were synthesized from the appropriate Intermediate and the appropriate starting material following the procedures described for Intermediate 13. Alternate conditions used: 1 Step 1: THF instead of DMF, rt, 2-15 h. 2 Step 1: CH 3 I, Cs 2 CO 3 , DMF, 50 °C, 2 h. 3 Step 1: 80 °C, 15 h. 4 Step 2 only. 5 Step 2: Pd(dppf)Cl 2 , KF, dioxane, water, 90 °C, 30-45 min. 6 Step 2: Intermediate 6.02, Pd(dppf)Cl 2 , K 2 CO 3 , CH 3 CN, microwave, 90 °C, 15 min. Intermediate 14 (R)-6-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-iodo-1-m ethyl-1H-pyrazolo[3,4- d]pyrimidine [00282] Intermediate 13, Step 1 (800 mg, 2.72 mmol) and Intermediate 2.01 (1.05 g, 4.13 mmol) were combined in a 20 mL microwave vial.1-Methyl-2-pyrrolidinone (8 mL) and then DIEA (1.35 mL, 7.75 mmol) were added. The reaction was heated in a microwave at 150 °C for 1 h, allowed to cool to rt, and then diluted with 30 mL ethyl acetate. The organic layer was washed with 30 mL water, washed with 20 mL brine, dried (Na 2 SO 4 ), filtered, and then concentrated. The residue was suspended in CH 2 Cl 2 :EtOAc (5:2, 10 mL), warmed with a heat gun, cooled to rt, and then filtered. The solids were washed with 10 mL EtOAc to give (R)-6-(2- benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-iodo-1-methyl-1H- pyrazolo[3,4-d]pyrimidine (664 mg, 48%) as a beige solid. The filtrate was purified by silica gel chromatography (0-15% EtOAc in hexanes). The fractions were concentrated and then suspended in CH 2 Cl 2 :EtOAc (5:2, 7 mL). The mixture was warmed with a heat gun, cooled to rt, and then filtered. The solids were washed with 5 mL EtOAc to give additional (R)-6-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-iodo- 1-methyl-1H-pyrazolo[3,4-d]pyrimidine (212 mg, 15%). 1 H NMR (400 MHz, DMSO-d6): δ 8.62 (s, 1H), 7.36-7.24 (m, 4H), 7.22-7.13 (m, 1H), 5.14 (br s, 1H), 4.82 (br d, J = 11.7 Hz, 1H), 3.87 (s, 3H), 3.70 (br d, J = 10.6 Hz, 1H), 3.49 (br d, J = 11.7 Hz, 1H), 3.41-3.34 (m, 1H), 3.10 (dd, J = 9.4, 12.8 Hz, 1H), 2.92-2.89 (m, 3H), 2.88-2.75 (m, 3H); LCMS: 513.4 [M+H] + . [00283] The Intermediates below were synthesized from the appropriate Intermediate and the appropriate amine following the procedure described for Intermediate 14. Alternate conditions used: Reaction time was 1-5 h. Reaction temp was 120 °C or 150 °C. 1 Synthesized from Intermediate 14.01 by methylation: MeI, NaH, DMF, 0 °C-rt, 1-3 h. 2 Used Intermediate 2.06 and Intermediate 13.10 (Step 1). 3 KF, NMP, 100 °C, overnight. Intermediate 15 6-(2-(Cyclopropylmethyl)-4-(methylsulfonyl)piperazin-1-yl)-3 -iodo-1-methyl-1H- pyrazolo[3,4-d]pyrimidine Step 1: tert-Butyl 3-(cyclopropylmethyl)-4-(3-iodo-1-methyl-1H-pyrazolo[3,4-d]p yrimidin- 6-yl)piperazine-1-carboxylate [00284] Intermediate 13, Step 1 (150 mg, 0.509 mmol) and tert-butyl 3- (cyclopropylmethyl)piperazine-1-carboxylate (147 mg, 0.611 mmol) were weighed into a 2 mL microwave vial.1-Methyl-2-pyrrolidinone (1 mL) and N,N-diisopropylethylamine (275 µL, 1.58 mmol) were added. The reaction was heated in a microwave at 100 °C for 1.5 h, allowed to cool to rt, and then diluted with 10 mL EtOAc. The organic layer was washed with 10 mL water, washed with 10 mL brine, dried (Na 2 SO 4 ), filtered, concentrated, and then purified by silica gel chromatography (0-20% EtOAc in hexanes) to give tert-butyl 3-(cyclopropylmethyl)-4-(3-iodo- 1-methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)piperazine-1-carbo xylate (129 mg, 51%) as a white foam. 1 H NMR (400 MHz, DMSO-d6): δ 8.49 (s, 1H), 4.91-4.73 (m, 1H), 4.46 (br dd, J = 1.4, 13.9 Hz, 1H), 4.01 (br d, J = 13.6 Hz, 1H), 3.95-3.86 (m, 1H), 3.72 (s, 3H), 3.00 (br d, J = 9.0 Hz, 2H), 2.82-2.67 (m, 1H), 1.63-1.45 (m, 1H), 1.34 (s, 9H), 1.15 (br s, 1H), 0.65-0.48 (m, 1H), 0.39-0.23 (m, 2H), 0.00 (br s, 2H); LCMS: 499.5 [M+H] + . Step 2: 6-(2-(Cyclopropylmethyl)piperazin-1-yl)-3-iodo-1-methyl-1H-p yrazolo[3,4- d]pyrimidine [00285] tert-Butyl 3-(cyclopropylmethyl)-4-(3-iodo-1-methyl-1H-pyrazolo[3,4-d]p yrimidin-6- yl)piperazine-1-carboxylate (125 mg, 0.251 mmol) was weighed into an 8 mL vial. Dichloromethane (1 mL) and TFA (200 µL) were added. The reaction was stirred at rt for 5 h, concentrated, diluted with CH 2 Cl 2 (1 mL), concentrated, diluted with CH 2 Cl 2 (1 mL), and then concentrated to give 6-(2-(cyclopropylmethyl)piperazin-1-yl)-3-iodo-1-methyl-1H-p yrazolo[3,4- d]pyrimidine, which was used in the next step without purification. Step 3: 6-(2-(Cyclopropylmethyl)-4-(methylsulfonyl)piperazin-1-yl)-3 -iodo-1-methyl-1H- pyrazolo[3,4-d]pyrimidine [00286] Triethylamine (100 µL, 0.717 mmol) was added to 6-(2-(cyclopropylmethyl)piperazin- 1-yl)-3-iodo-1-methyl-1H-pyrazolo[3,4-d]pyrimidine (from the previous step) in CH 2 Cl 2 (2 mL). The mixture was cooled in an ice/water bath. Methanesulfonyl chloride (22 µL, 0.28 mmol) was added at 0 °C. The reaction was stirred at rt for 17 h, diluted with 10 mL CH 2 Cl 2 , washed with 10 mL water, dried (Na 2 SO 4 ), filtered, concentrated, and then purified by silica gel chromatography (0-40% EtOAc in hexanes) to give 6-(2-(cyclopropylmethyl)-4- (methylsulfonyl)piperazin-1-yl)-3-iodo-1-methyl-1H-pyrazolo[ 3,4-d]pyrimidine (49 mg, 41%) as a white foam. 1 H NMR (400 MHz, DMSO-d6): δ 8.61 (s, 1H), 5.12 (br s, 1H), 4.76 (br d, J = 13.9 Hz, 1H), 3.83 (s, 3H), 3.71 (br d, J = 11.6 Hz, 1H), 3.60 (br d, J = 11.4 Hz, 1H), 3.17 (br t, J = 11.4 Hz, 1H), 2.96-2.88 (m, 4H), 2.84-2.75 (m, 1H), 1.80-1.68 (m, 1H), 1.47-1.36 (m, 1H), 0.62 (br dd, J = 1.5, 3.9 Hz, 1H), 0.42-0.27 (m, 2H), 0.20-0.00 (m, 2H); LCMS: 477.5 [M+H] + . [00287] The Intermediate below was isolated during the purification of Intermediate 15, Step 3. Alternate conditions used: Step 2: 2 h. Step 3: 0 °C-rt, 30 min. 1 Isolated during the purification of Intermediate 15, Step 3. 2 Synthesized from Intermediate 15 (Step 2) by acylation (dimethylcarbamyl chloride, TEA, CH 2 Cl 2 , 0 °C-rt, 1 h). 3 Synthesized from Intermediate 15 (Step 2) by amine addition (isocyanatotrimethylsilane, TEA, THF, 0 °C-rt, overnight). Intermediate 16 3-(Cyclohexylmethyl)morpholine Step 1: 2-Amino-3-cyclohexylpropan-1-ol [00288] BH3•Me2S (10 M, 5.26 mL, 52.6 mmol) was added dropwise to a mixture of 2-amino- 3-cyclohexylpropanoic acid (3.0 g, 17.5 mmol) and THF (50 mL) at 0 °C. The mixture was heated at 50 °C overnight, allowed to cool to rt, poured into MeOH (50 mL) slowly, stirred for 0.5 h, and then concentrated to give 2-amino-3-cyclohexylpropan-1-ol (2.87 g) as a colorless oil. Step 2: 2-(2-Chloroacetamido)-3-cyclohexylpropyl 2-chloroacetate [00289] 2-Chloroacetyl chloride (4.12 g, 36.5 mmol) was added dropwise to a mixture of 2- amino-3-cyclohexylpropan-1-ol (2.87 g, 18.3 mmol), K 2 CO 3 (5.04 g, 36.5 mmol), and MeCN (50 mL) at 0 °C. The mixture was stirred at rt for 2 h, poured into H 2 O (50 mL), and then extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and concentrated to give 2-(2-chloroacetamido)-3- cyclohexylpropyl 2-chloroacetate (5.1 g) as a yellow oil. Step 3: 2-Chloro-N-(1-cyclohexyl-3-hydroxypropan-2-yl)acetamide [00290] LiOH•H 2 O (3.45 g, 82.2 mmol) was added to a mixture of 2-(2-chloroacetamido)-3- cyclohexylpropyl 2-chloroacetate (5.1 g, 16.4 mmol) and THF (50 mL). The reaction was stirred at rt for 2 h, diluted with H 2 O (30 mL), and then extracted with EtOAc (30 mL × 3). The combined organic layers were washed with brine (100 mL), dried over Na 2 SO 4 , filtered, and concentrated to give 2-chloro-N-(1-cyclohexyl-3-hydroxypropan-2-yl)acetamide (3.75g) as a yellow oil. LCMS: 234.2 [M+H] + . Step 4: 5-(Cyclohexylmethyl)morpholin-3-one [00291] t-BuOK (9.00 g, 80.2 mmol) was added to a solution of 2-chloro-N-(1-cyclohexyl-3- hydroxypropan-2-yl)acetamide (3.75 g, 16.0 mmol) and i-PrOH (50 mL). The mixture was stirred at rt for 1 h, heated at 40 ℃ overnight, allowed to cool to rt, adjusted to pH=2 with 1 M HCl, and then concentrated. The residue was diluted with H 2 O (50 mL) and extracted with DCM (20 mL × 3). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and concentrated to give 5-(cyclohexylmethyl)morpholin-3-one (1.9 g) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6): δ 8.04 (s, 1H), 3.93-3.92 (d, 2H), 3.79-3.76 (m, 1H), 3.43-3.38 (m, 2H), 1.65-1.62 (d, 6H), 1.36-1.33 (m, 2H), 1.24-1.15 (m, 3H), 0.85-0.80 (m, 2H); LCMS: 198.2 [M+H] + . Step 5: 3-(Cyclohexylmethyl)morpholine [00292] BH 3 •Me 2 S (10 M, 2.40 mL, 24.0 mmol) was added dropwise to a mixture of 5- (cyclohexylmethyl)morpholin-3-one (1.58 g, 8.01 mmol) and THF (20 mL) at 0 °C. The mixture was stirred at rt overnight, poured into MeOH (50 mL) slowly, stirred for 0.5 h, and then concentrated to give 3-(cyclohexylmethyl)morpholine (1.4 g) as a colorless oil. [00293] The Intermediates below were synthesized from the appropriate 2-amino-3- (pyridinyl)propanoic acid using the following sequence: Intermediate 16 (Step 1: rt, overnight; Step 2: THF/water as solvent to provide free OH directly), cyclization (NaH, THF, 0 °C, 0.5 h), amide reduction (2.4 M LiAlH4 in THF, THF, rt, overnight). 1After amide reduction: Boc protection (Boc2O, CH 2 Cl 2 , TEA, rt, 2 h), silica gel purification, Boc deprotection (4 M HCl in EtOAc, rt, 1 h); 2 Step 1: 2.4 M LiAH 4 in THF, THF, 0 °C-rt, overnight. Intermediate 17 3-(Cyclohexylmethyl)-1-(methylsulfonyl)piperazine Step 1: Ethyl 2-(benzylamino)-3-cyclohexylpropanoate [00294] NaBH(OAc)3 (4.17 g, 19.67 mmol) was added to a mixture of ethyl 2-amino-3- cyclohexylpropanoate (1.96 g, 9.83 mmol), benzaldehyde (0.90 mL, 8.85 mmol), and DCE (30 mL) at 0 °C. The mixture was stirred at rt for 2 h, poured into H 2 O (30 mL), adjusted to pH=8 with sat. aq. NaHCO 3 , and then extracted with DCM (20 mL × 3). The organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 50/1 to 5/1) to give ethyl 2-(benzylamino)-3- cyclohexylpropanoate (1.76 g, 22%) as a yellow oil. LCMS: 290.2 [M+H] + . Step 2: Ethyl 2-(benzyl(2-((tert-butoxycarbonyl)amino)ethyl)amino)-3- cyclohexylpropanoate [00295] NaBH(OAc)3 (2.81 g, 13.3 mmol) was added to a mixture of ethyl 2-(benzylamino)-3- cyclohexylpropanoate (1.92 g, 6.63 mmol), tert-butyl (2-oxoethyl)carbamate (950 mg, 5.97 mmol), and DCE (20 mL) at 0 °C. The mixture was stirred at rt for 2 h, poured into H 2 O (20 mL), adjusted to pH=8 with sat. aq. NaHCO 3 , and then extracted with DCM (10 mL × 3). The organic layers were washed with brine (30 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 50/1 to 5/1) to give ethyl 2-(benzyl(2-((tert-butoxycarbonyl)amino)ethyl)amino)-3-cyclo hexylpropanoate (2.17 g, 76%) as a yellow oil. LCMS: 433.3 [M+H] + . Step 3: Ethyl 2-((2-aminoethyl)(benzyl)amino)-3-cyclohexylpropanoate hydrochloride [00296] A mixture of ethyl 2-(benzyl(2-((tert-butoxycarbonyl)amino)ethyl)amino)-3- cyclohexylpropanoate (2.04 g, 4.72 mmol) and HCl/EtOAc (4M, 30 mL) was stirred at rt for 0.5 h and then concentrated to give ethyl 2-((2-aminoethyl)(benzyl)amino)-3-cyclohexylpropanoate hydrochloride (1.6 g) as a yellow solid: LCMS: 333.3 [M+H] + . Step 4: 4-Benzyl-3-(cyclohexylmethyl)piperazin-2-one [00297] Sodium methoxide (1.15 g, 21.3 mmol) was added to a solution of ethyl 2-((2- aminoethyl)(benzyl)amino)-3-cyclohexylpropanoate hydrochloride (1.57 g, 4.26 mmol) and MeOH (20 mL). The mixture was stirred at rt for 2 h, poured into H 2 O (20 mL), and then filtered. The filter cake was washed with H 2 O (50 mL) and then dried under vacuum to give 4- benzyl-3-(cyclohexylmethyl)piperazin-2-one (1.4 g) as a white solid. 1 H NMR (400 MHz, CDCl 3 ): δ 7.34-7.28 (m, 5H), 5.94 (s, 1H), 3.89-3.86 (m, 1H), 3.60-3.57 (m, 1H), 3.52-3.48 (m, 1H), 3.28-3.24 (m, 1H), 3.19-3.16 (m, 1H) , 3.12-3.06 (m, 1H), 2.63-2.59 (m, 1H), 1.72-1.48 (m, 8H), 1.28-1.13 (m, 3H), 0.96-0.82 (m, 2H); LCMS: 287.3 [M+H] + . Step 5: 1-Benzyl-2-(cyclohexylmethyl)piperazine [00298] LiAlH4 (726 mg, 19.1 mmol) was added in 2 portions to a mixture of 4-benzyl-3- (cyclohexylmethyl)piperazin-2-one (1.37 g, 4.78 mmol) in THF (30 mL) at 0 °C under N 2 . The mixture was heated at 65 °C for 2 h, allowed to cool to rt, poured into sat. aq. NaK tartrate (30 mL), and then extracted with EtOAc (20 mL × 3). The organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, and concentrated to give 1-benzyl-2- (cyclohexylmethyl)piperazine (1.32 g) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ): δ 7.34-7.21 (m, 5H), 4.02 (d, 1H), 3.17 (d, 1H), 3.04-3.00 (m, 1H), 2.84-2.75 (m, 2H), 2.71-2.60 (m, 2H), 2.45-2.36 (m, 1H), 2.18-2.09 (m, 1H), 1.78-1.58 (m, 8H), 1.33-1.24 (m, 4H), 0.98-0.85 (m, 2H); LCMS: 273.2 [M+H] + . Step 6: 1-Benzyl-2-(cyclohexylmethyl)-4-(methylsulfonyl)piperazine [00299] MsCl (1.16 mL, 15.0 mmol) was added to a solution of 1-benzyl-2- (cyclohexylmethyl)piperazine (1.26 g, 4.63 mmol), Et3N (3.22 mL, 23.1 mmol), and DCM (20 mL) at 0 °C. The mixture was stirred at rt for 2 h, poured into water (20 mL), and then extracted with DCM (10 mL × 3). The organic layers were washed with brine (30 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 20/1 to 2/1) to give 1-benzyl-2-(cyclohexylmethyl)-4- (methylsulfonyl)piperazine (1.24 g, 82% over 4 steps) as a yellow solid. 1 H NMR (400 MHz, CDCl 3 ): δ 7.32-7.24 (m, 5H), 3.90 (d, 1H), 3.37-3.30 (m, 2H), 3.22-3.10 (m, 2H), 3.02-2.99 (m, 1H), 2.83-2.79 (m, 1H), 2.77 (s, 3H), 2.71-2.67 (m, 1H), 2.40-2.34 (m, 1H), 1.70-1.53 (m, 6H), 1.40-1.20 (m, 5H), 1.00-0.85 (m, 2H); LCMS: 351.2 [M+H]+. Step 7: 3-(Cyclohexylmethyl)-1-(methylsulfonyl)piperazine [00300] 1-Benzyl-2-(cyclohexylmethyl)-4-(methylsulfonyl)piperazine (553 mg, 1.58 mmol) and AcOH (0.24 mL, 3.94 mmol) were added to a mixture of 20% Pd(OH) 2 /C (50% water, 700 mg) and THF (20 mL) under N 2 . The mixture was degassed with 3 vacuum/H 2 cycles, stirred under H 2 (15 Psi) at rt for 2 h, and then filtered through Celite with MeOH (600 mL) washing. The filtrate was concentrated to give 3-(cyclohexylmethyl)-1-(methylsulfonyl)piperazine (530 mg) as a yellow oil. LCMS: 261.2 [M+H] + . Intermediate 18 tert-Butyl 4-amino-4-benzylpiperidine-1-carboxylate Step 1: 4-Benzyl-1-(tert-butoxycarbonyl)piperidine-4-carboxylic (isobutyl carbonic) anhydride [00301] Isobutyl carbonochloridate (1.54 mL, 11.7 mmol) was added dropwise to a solution of 4-benzyl-1-(tert-butoxycarbonyl)piperidine-4-carboxylic acid (2.50 g, 7.83 mmol), 4- methylmorpholine (1.2 mL, 10.9 mmol), and THF (50 mL) at -15 °C under N 2 . The reaction was stirred at -15 °C for 2 h to give 4-benzyl-1-(tert-butoxycarbonyl)piperidine-4-carboxylic (isobutyl carbonic) anhydride as a colorless solution. LCMS: 442.3 [M+Na] + . Step 2: tert-Butyl 4-(azidocarbonyl)-4-benzylpiperidine-1-carboxylate [00302] A solution of NaN3 (1.55 g, 23.8 mmol) an water (5 mL) was added dropwise to the solution of 4-benzyl-1-(tert-butoxycarbonyl)piperidine-4-carboxylic (isobutyl carbonic) anhydride (~5.96 mmol; estimated by LCMS purity) in THF (50 mL) at -15 °C under N 2 . The reaction mixture was allowed to warm to rt, stirred overnight, poured into ice/water (100 mL), and extracted with toluene (30 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, and then concentrated to ~90 mL to give a yellow solution of tert-butyl 4- (azidocarbonyl)-4-benzylpiperidine-1-carboxylate in toluene. Step 3: tert-Butyl 4-amino-4-benzylpiperidine-1-carboxylate [00303] The solution of tert-butyl 4-(azidocarbonyl)-4-benzylpiperidine-1-carboxylate (~4.65 mmol; estimated by TLC purity) in toluene (90 mL) was added dropwise over ~10 min to refluxing toluene (30 mL). The reaction was refluxed for 2 h, allowed to cool to rt, concentrated, and then diluted with THF (25 mL) and water (25 mL). A solution of KOH (1.06 g, 18.9 mmol) and water (5 mL) was added dropwise at 0 °C. The reaction was allowed to warm to rt, stirred overnight, concentrated to remove THF, diluted with water (25 mL), adjusted to pH~9 with aq. HCl (6M, ~3 mL), and then extracted with ethyl acetate (30 mL × 3). The combined organic layers were washed with brine (30 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 3/1) to give tert-butyl 4- amino-4-benzylpiperidine-1-carboxylate (900 mg, 40% over 3 steps) as a colorless oil. 1 H NMR (400 MHz, DMSO-d6): δ 7.32-7.16 (m, 5H), 3.66-3.51 (m, 2H), 3.20-3.05 (m, 2H), 2.64-2.57 (m, 2H), 1.43-1.28 (m, 13H), 1.25-1.11 (m, 2H); LCMS: 291.3 [M+H] + . [00304] The Intermediate below was synthesized from 3-benzyl-1-(tert- butoxycarbonyl)piperidine-3-carboxylic acid following the procedures described for Intermediate 18. Intermediate 19 Step 1: (R)-4-Benzylazetidin-2-one [00305] MsCl (0.24 mL, 2.76 mmol) was added to a mixture of (R)-3-amino-4-phenylbutanoic acid (451 mg, 2.52 mmol), NaHCO 3 (1.27 g, 15.1 mmol), and MeCN (15 mL) at 80 °C. The reaction was stirred at 80 °C for 2 h, allowed to cool to rt, filtered, and then concentrated to give (R)-4-benzylazetidin-2-one (621 mg; 5 batches combined during filtration) as white solid. Step 2: (R)-4-Benzyl-1-(tert-butyldimethylsilyl)azetidin-2-one [00306] A mixture of (R)-4-benzylazetidin-2-one (621 mg, 3.85 mmol), tert- butylchlorodimethylsilane (638 mg, 4.24 mmol), DIEA (1.0 mL, 5.78 mmol), and DCM (8 mL) was stirred at rt for 2 h under N 2 , concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 100/1 to 0/1) to give (R)-4-benzyl-1-(tert- butyldimethylsilyl)azetidin-2-one (623 mg, 58%) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ): δ 7.40-7.28 (m, 3H), 7.16 (d, 2H), 3.79-3.70 (m, 1H), 3.28-3.23 (m, 1H), 3.04-2.98 (m, 1H), 2.73- 2.68 (m, 1H), 2.63-2.57 (m, 1H), 1.02 (s, 9H), 0.32 (s, 3H), 0.29 (s, 3H); LCMS: 276.2 [M+H] + . Step 3: (3R,4R)-4-Benzyl-1-(tert-butyldimethylsilyl)-3-(methylthio)a zetidin-2-one [00307] LDA (2 M in THF, 0.85 mL) was added dropwise to a solution of (R)-4-benzyl-1-(tert- butyldimethylsilyl)azetidin-2-one (391 mg, 1.42 mmol) and THF (15 mL) at -78 °C. The reaction was stirred for 15 min, and then a solution of 1,2-dimethyldisulfane (69.2 mg, 0.732 mmol) in THF (0.5 mL) was added dropwise at -78 °C. The reaction was stirred at -78°C for 10 min, poured (while still cold) into water (30 mL), and then extracted with ethyl acetate (25 mL × 3). The combined organic phase was washed with brine (70 mL), dried with anhydrous Na 2 SO 4 , filtered, concentrated, and purified by silica gel chromatography (petroleum ether/ethyl acetate: 100/1 to 0/1) to give (3R,4R)-4-benzyl-1-(tert-butyldimethylsilyl)-3-(methylthio)a zetidin-2-one (301 mg, 65%) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ): δ 7.41-7.35 (m, 3H), 7.25-7.16 (m, 2H), 3.78-3.74 (m, 1H), 3.71-3.63 (m, 1H), 3.34-3.26 (m, 1H), 2.73-2.62 (m, 1H), 1.86-1.79 (m, 3H), 1.03 (s, 9H), 0.34 (s, 3H), 0.31 (s, 3H); LCMS: 322.2 [M+H] + . Step 4: (2R,3S)-2-Benzyl-3-(methylthio)azetidine [00308] BH3•THF (1 M in THF, 3.74 mL) was added to a solution of (3R,4R)-4-benzyl-1-(tert- butyldimethylsilyl)-3-(methylthio)azetidin-2-one (301 mg, 0.936 mmol) and THF (10 mL) at 0 °C. The reaction was stirred at rt for 12 h, and then MeOH (10 mL) was added. The reaction was heated at 75 °C for 2 h, allowed to cool to rt, and then concentrated to give (2R,3S)-2-benzyl-3- (methylthio)azetidine (165 mg) as colorless oil. LCMS: 194.2 [M+H] + . Intermediate 20 (R)-3-Benzyl-1-ethylpiperazine Step 1: (R)-tert-Butyl 2-benzyl-4-ethylpiperazine-1-carboxylate [00309] A mixture of (R)-tert-butyl 2-benzylpiperazine-1-carboxylate (1.0 g, 3.62 mmol), iodoethane (0.43 mL, 5.43 mmol), DIEA (1.89 mL, 10.85 mmol), and MeCN (10 mL) was stirred at rt for 2 h under N 2 , concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 5/1→2/1) to give (R)-tert-butyl 2-benzyl-4-ethylpiperazine-1- carboxylate (3.85 g, 87%) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6): δ 7.33-7.24 (m, 2H), 7.23-7.11 (m, 3H), 4.11 (s, 1H), 3.77 (d, 1H), 3.09 (t, 1H), 2.98-2.76 (m, 3H), 2.68 (d, 1H), 2.43- 2.28 (m, 1H), 2.27-2.15 (m, 1H), 1.82 (td, 2H), 1.41-1.11 (m, 9H), 0.99 (t, 3H); LCMS: 305.3 [M+H] + Step 2: (R)-3-Benzyl-1-ethylpiperazine hydrochloride [00310] A mixture of (R)-tert-butyl 2-benzyl-4-ethylpiperazine-1-carboxylate (3.85 g, 12.65 mmol) and HCl/EtOAc (4 M, 3.16 mL) was stirred at rt for 2 h and then concentrated to give (R)-3-benzyl-1-ethylpiperazine hydrochloride (3.0 g) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 12.11-11.75 (m, 1H), 10.67-10.30 (m, 1H), 10.04-9.67 (m, 1H), 7.48-7.14 (m, 5H), 3.99-3.83 (m, 1H), 3.80-3.62 (m, 1H), 3.60-3.37 (m, 5H), 3.35-3.03 (m, 2H), 2.93 (dd, 1H), 1.29-1.11 (m, 3H); LCMS: 205.2 [M+H] + . Step 3: (R)-3-Benzyl-1-ethylpiperazine [00311] A mixture of (R)-3-benzyl-1-ethylpiperazine hydrochloride (3.0 g, 12.46 mmol), Ambersep 900 OH anion exchange resin (250 mg), and MeOH (20 mL) was stirred at rt for 30 min. The reaction was filtered, and the filtrate was concentrated to give (R)-3-benzyl-1- ethylpiperazine (2.1 g) as a white solid. Intermediate 21 tert-Butyl 6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydrop yridine-1(2H)- carboxylate Step 1: tert-Butyl 6-((diphenoxyphosphoryl)oxy)-3,4-dihydropyridine-1(2H)-carbo xylate [00312] LiHMDS (37.6 mL, 1 M in THF) was added to a solution of tert-butyl 2- oxopiperidine-1-carboxylate (7.40 g, 27.6 mmol) in THF (50 mL) at -78 °C. The reaction was stirred at -78 °C for 1 h, and then a solution of diphenyl phosphorochloridate (5.0 g, 25.1 mmol) and THF (50 mL) was added at -78 °C. The mixture was allowed to warm to rt, stirred for 1 h, quenched with H 2 O (100 mL), and then extracted with EtOAc (50 mL × 3). The combined organic layers were washed with brine (50 mL × 2), dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 100/1 to 5/1) to give tert-butyl 6-((diphenoxyphosphoryl)oxy)-3,4-dihydropyridine-1(2H)-carbo xylate (9.62 g, 88%) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ): δ 8.56-7.96 (m, 1H), 7.30-7.20 (m, 4H), 7.20-7.15 (m, 2H), 7.12 (t, 4H), 3.58 (t, 1H), 3.54-3.47 (m, 1H), 2.50-2.38 (m, 1H), 2.16-2.03 (m, 1H), 1.79-1.71 (m, 2H), 1.50-1.41 (m, 6H), 1.36 (s, 3H); LCMS: 432.2 [M+H] + . Step 2: tert-Butyl 6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydrop yridine- 1(2H)-carboxylate [00313] Pd(PPh3) 2 Cl 2 (292.9 mg, 0.417 mmol) was added to a mixture of tert-butyl 6- ((diphenoxyphosphoryl)oxy)-3,4-dihydropyridine-1(2H)-carboxy late (6.0 g, 13.9 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborola n-2-yl)-1,3,2-dioxaborolane (5.30 g, 20.8 mmol), PPh3 (219 mg, 0.83 mmol), K 2 CO 3 (2.88 g, 20.9 mmol), and dioxane (500 mL). The mixture was degassed with 3 vacuum/N 2 cycles, heated at 90 °C for 7 h, allowed to cool to rt, quenched with H 2 O (100 mL), and then extracted with EtOAc (30 mL × 3). The combined organic layers were washed with brine (50 mL × 2), dried over Na 2 SO 4 , filtered, concentrated, and purified by silica gel chromatography (petroleum ether/ethyl acetate: 1/0 to 8/1) to give tert-butyl 6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydrop yridine- 1(2H)-carboxylate (562 mg, 13%) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ): δ 5.21 (d, 1H), 3.48-3.39 (m, 2H), 2.09-1.99 (m, 2H), 1.77 (d, 2H), 1.58-1.47 (m, 9H), 1.30-1.27 (m, 12H); LCMS: 310.3 [M+H] + . Intermediate 22 (R)-5-(2-Benzyl-4-ethylpiperazin-1-yl)-3-methyl-2H-pyrazolo[ 4,3-d]pyrimidine [00314] (R)-5-(2-Benzyl-4-ethylpiperazin-1-yl)-3-methyl-2H-pyrazolo[ 4,3-d]pyrimidine was synthesized from Intermediate 9 (Step 1) and Intermediate 20 following the procedures described for Intermediate 10 (Step 3; KF instead of DIEA) and then Intermediate 11 (Step 3). 1 H NMR (400 MHz, DMSO-d6): δ 9.57 (s, 1H), 9.27 (s, 1H), 7.71-7.43 (m, 5H), 5.22-4.99 (m, 1H), 4.80 (d, 1H), 2.86-2.81 (m, 9H), 2.77 (s, 3H), 1.36 (t, 3H); LCMS: 337.3 [M+H] + . Intermediate 23 (R)-3-Benzylthiomorpholine Step 1: (R)-2-((tert-Butoxycarbonyl)amino)-3-phenylpropyl methanesulfonate [00315] MsCl (1.39 mL, 17.9 mmol) was added to a mixture of tert-butyl (R)-(1-hydroxy-3- phenylpropan-2-yl)carbamate (3.01 g, 12.0 mmol), Et 3 N (2.50 mL, 17.9 mmol), and DCM (30 mL) at 0 °C. The reaction was stirred at 25 °C for 1 h, diluted with DCM (30 mL), washed with 1 M HCl (30 mL), washed with sat.aq. NaHCO 3 (30 mL), washed with brine (30 mL), dried (Na 2 SO 4 ), filtered, and concentrated to give (R)-2-((tert-butoxycarbonyl)amino)-3-phenylpropyl methanesulfonate (3.55 g) as a white solid. 1 H NMR (400 MHz, CDCl 3 ): δ 7.36-7.25 (m, 5H), 5.34 (s, 1H), 4.83-4.81 (m, 1H), 4.18-4.16 (m, 2H), 3.06 (s, 3H), 2.98– 2.96 (m, 2H), 1.46 (s, 9H); LCMS: 274.1 [(M-tBu+H)+H] + . Step 2: (R)-Methyl 2-((2-((tert-butoxycarbonyl)amino)-3-phenylpropyl)thio)aceta te [00316] Methyl 2-sulfanylacetate (1.51 mL, 16.7 mmol) was added to a mixture of (R)-2-((tert- butoxycarbonyl)amino)-3-phenylpropyl methanesulfonate (2.5 g, 7.59 mmol), K 2 CO 3 (2.1 g, 15.2 mmol), and MeCN (30 mL). The mixture was stirred at 80 °C for 1 h, cooled to rt, concentrated, diluted with EtOAc (50 mL), washed with 1 M HCl (20 mL), washed with sat.aq. NaHCO 3 (20 mL), washed with brine (20 mL), dried (Na 2 SO 4 ), filtered, and concentrated to give crude (R)-methyl 2-((2-((tert-butoxycarbonyl)amino)-3-phenylpropyl)thio)aceta te (2.55 g) as a yellow oil. LCMS: 240.1 [(M-Boc+H)+H)] + . Step 3: (R)-5-Benzylthiomorpholin-3-one [00317] A mixture of (R)-methyl 2-((2-((tert-butoxycarbonyl)amino)-3- phenylpropyl)thio)acetate (2.55 g, 7.51 mmol) and HCl/EtOAc (4 M, 1 mL) was stirred at rt for 1 h and then concentrated. A mixture of this residue, MeOH (1 mL), and Et3N (3.1 mL, 22.5 mmol) was stirred at 25 °C for 24 h, concentrated, diluted with EtOAc (30 mL), washed with 1 M HCl (10 mL), washed with sat.aq. NaHCO 3 (10 mL), washed with brine (10 mL), dried (Na 2 SO 4 ), filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/EtOAc: 20/1 to 2/1) to give (R)-5-benzylthiomorpholin-3-one (925 mg, 59%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 7.80 (s, 1H), 7.34-7.22 (m, 5H), 4.34-4.21 (m, 1H), 3.25-3.21 (m, 1H), 3.10-3.06 (m, 1H), 2.83-2.82 (m, 1H), 2.80-2.76 (m, 2H), 2.50-2.49 (m, 1H); LCMS: 208.1 [M+H] + . Step 4: (R)-3-Benzylthiomorpholine [00318] BH3•THF (1 M in THF, 7.4 mL) was slowly added to a mixture of (R)-5- benzylthiomorpholin-3-one (0.51 g, 2.46 mmol) and THF (2 mL) at 25 °C. The reaction was stirred at 70 °C for 2 h, cooled to rt, quenched with MeOH (~50 mL), and then concentrated. The residue, EtOAc (10 mL), and 1 M NaOH (20 mL) was stirred for 0.5 h and then extracted with EtOAc (10 mL × 2). The combined organic layers were washed with brine (20 mL), dried (Na2SO4), filtered, and concentrated to give (R)-3-benzylthiomorpholine (402 mg) as a yellow oil. LCMS: 194.1 [M+H] + . Compound 1 (R)-1-(4-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)phenyl)- 5,7-difluoro-1H- benzo[d][1,2,3]triazol-6-ol Step 1: (R)-1-(4-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)phenyl)- 6-(benzyloxy)-5,7- difluoro-1H-benzo[d][1,2,3]triazole [00319] Tris(dibenzylideneacetone)dipalladium(0) (66 mg, 0.07 mmol) was added to a solution of Intermediate 1 (300 mg, 0.72 mmol), Intermediate 2 (319 mg, 0.86 mmol), RuPhos (67.2 mg, 0.14 mmol), and t-BuONa (346 mg, 3.60 mmol) in dioxane (6 mL) under N 2 . The mixture was degassed and purged with N 2 3 times, stirred at 100 °C for 3 h, allowed to cool to rt, poured into H 2 O (10 mL), and then extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by column chromatography (petroleum ether/ethyl acetate=20/1 to 2/1) to obtain (R)-1-(4-(2- benzyl-4-(methylsulfonyl)piperazin-1-yl)phenyl)-6-(benzyloxy )-5,7-difluoro-1H- benzo[d][1,2,3]triazole (205 mg, 48%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 8.03 (d, 1H), 7.64-7.62 (m, 2H), 7.46-7.44 (m, 2H), 7.41-7.35 (m, 3H), 7.33-7.21 (m, 7H), 5.25 (s, 2H), 4.31 (d, 1H), 3.76-3.67 (m, 2H), 3.45 (d, 1H), 3.06 (d, 1H), 3.00-2.89 (m, 6H), 2.59 (d, 1H); LCMS: 590.2 [M+H] + Step 2: (R)-1-(4-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)phenyl)- 5,7-difluoro-1H- benzo[d][1,2,3]triazol-6-ol [00320] A mixture of (R)-1-(4-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)phenyl)- 6- (benzyloxy)-5,7-difluoro-1H-benzo[d][1,2,3]triazole (195 mg, 0.33 mmol) and TFA (3.0 mL, 40.5 mmol) was stirred at 50 °C for 3 h, allowed to cool to rt, quenched with sat. aq. NaHCO 3 until pH=~8, and then extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×10 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by prep-HPLC [water(FA)-CH 3 CN] to obtain (R)-1-(4-(2-benzyl-4-(methylsulfonyl)piperazin-1- yl)phenyl)-5,7-difluoro-1H-benzo[d][1,2,3]triazol-6-ol (75 mg, 45%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 10.96 (s, 1H), 7.90 (d, 1H), 7.63-7.60 (m, 2H), 7.32-7.19 (m, 7H), 4.30 (d, 1H), 3.71 (d, 2H), 3.44 (d, 1H), 3.25 (t, 1H), 3.05 (d, 1H), 2.99-2.88 (m, 5H), 2.57 (d, 1H); LCMS: 500.0 [M+H] + [00321] The Compounds below were synthesized from Intermediate 1 and the appropriate amine following the procedures described for Compound 1.

Compound 2 (R)-3-(5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-methy l-1H-pyrazolo[4,3- d]pyrimidin-1-yl)-2,6-difluoro-5-(trifluoromethyl)phenol [00322] A mixture of Intermediate 9 (88 mg, 0.23 mmol), Intermediate 4 (111 mg, 0.342 mmol), trans-N,N'-dimethylcyclohexane-1,2-diamine (28.7 µL, 0.182 mmol), CuI (17 mg, 0.091 mmol), potassium phosphate (193 mg, 0.911 mmol), and DMSO (3 mL) were degassed/backfilled with N 2 , stirred at 100 °C overnight, allowed to cool to rt, diluted with methanol, and then purified by prep-HPLC (20-100% CH 3 CN in water with 0.1% TFA) to give (R)-3-(5-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-methy l-1H-pyrazolo[4,3-d]pyrimidin-1- yl)-2,6-difluoro-5-(trifluoromethyl)phenol (68 mg, 49%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.84-11.58 (m, 1H), 9.01 (d, J = 2.3 Hz, 1H), 7.50 (t, J = 7.0 Hz, 1H), 7.41-7.23 (m, 4H), 7.22-7.13 (m, 1H), 5.07-4.97 (m, 1H), 4.72 (br d, J = 13.1 Hz, 1H), 3.72 (br d, J = 11.2 Hz, 1H), 3.49 (br d, J = 11.9 Hz, 1H), 3.39-3.26 (m, 1H), 3.14-3.03 (m, 1H), 2.94-2.88 (m, 3H), 2.88-2.79 (m, 2H), 2.73 (br dd, J = 3.4, 12.7 Hz, 1H), 2.60-2.52 (m, 3H); LCMS: 583.5 [M+H] + . [00323] The Compounds below were synthesized from the appropriate Intermediate and the appropriate aryl halide following the procedure described for Compound 2.

Alternate conditions used: Reaction time varied: 2-48 h. Reaction temp varied: 100-110 °C. In some instances, additional aryl iodide, bromide or chloride, CuI, trans-N,N'-dimethylcyclohexane-1,2- diamine, and K 3 PO 4 were needed for full conversion to desired product. 1Aryl bromide or chloride was used; 2 From Intermediate 11.06 (difluoro hydrolysis to ketone during coupling); 3 Synthesized from Compound 2.06 or 2.22 or 2.18: 0.4 M ammonia solution in dioxane or methylamine HCl, AdBrettPhos, AdBrettPhos Palladacycle Gen. 3, NaOtBu, dioxane, 80 °C, 2 h-overnight. 4 Synthesized from Compound 2.22: methylamine HCl, AdBrettPhos, AdBrettPhos Palladacycle Gen.3, NaOtBu, dioxane, 80 °C, 4.5 h, additional NaOtBu, AdBrettPhos, AdBrettPhos Palladacycle Gen 3 added, 80 °C, ON, and then 2 M methylamine in THF, AdBrettPhos, AdBrettPhos Palladacycle Gen.3, NaOtBu, 80 °C, 45 min. 5 Isolated in the synthesis of Compound 2.27. 6 Isolated in the synthesis of Compound 2.29. 7Bis[(tetrabutylammonium iodide)copper(I) iodide] instead of CuI and 110 °C instead of 100 °C. 8Isolated in the synthesis of Compound 2.36. Compound 3 (R)-3-(6-(3-Benzylmorpholino)-1-methyl-1H-pyrazolo[4,3-c]pyr idin-3-yl)-2,6-difluoro-5- (trifluoromethyl)phenol [00324] A mixture of Intermediate 13.01 (75 mg, 0.18 mmol), (R)-3-benzyl-morpholine (63 mg, 0.36 mmol), Pd2(dba)3 (19 mg, 0.021 mmol), RuPhos (20 mg, 0.043 mmol), NaO t Bu (40 mg, 0.42 mmol), and dioxane (2 mL) was purged with nitrogen for 5 min, heated in a microwave at 90 ℃ for 90 min, allowed to cool to rt, and then diluted with 20 mL EtOAc. The organic layer was washed with 15 mL saturated NH4Cl, washed with 15 mL brine, dried (Na 2 SO 4 ), filtered, concentrated, and then purified by silica gel chromatography (0-30% EtOAc in heptane) [Note: no purification was needed prior to deprotection for some compounds in the table below]. The residue was dissolved in CH 2 Cl 2 (2 mL) and TFA (1 mL), stirred at rt for 15 min, concentrated, and then purified by prep-HPLC to give (R)-3-(6-(3-benzylmorpholino)-1-methyl-1H- pyrazolo[4,3-c]pyridin-3-yl)-2,6-difluoro-5-(trifluoromethyl )phenol (38 mg, 41%) as an off- white solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.38 (br s, 1H), 8.81 (d, J = 3.1 Hz, 1H), 7.54 (t, J = 7.0 Hz, 1H), 7.35-7.26 (m, 4H), 7.24-7.15 (m, 1H), 6.81 (s, 1H), 4.63 (br d, J = 9.2 Hz, 1H), 4.10-3.99 (m, 4H), 3.86 (br d, J = 12.5 Hz, 1H), 3.71-3.65 (m, 1H), 3.64-3.55 (m, 1H), 3.52 (br d, J = 10.3 Hz, 1H), 3.30 (dt, J = 2.5, 12.4 Hz, 1H), 3.06 (br t, J = 11.7 Hz, 1H), 2.63 (br dd, J = 3.7, 12.5 Hz, 1H); LCMS 505.5 [M+H] + . [00325] The Compounds below were synthesized from the appropriate Intermediate and the appropriate amine following the procedure described for Compound 3. Alternate condition used: Reaction times varied: 60-90 min. 1 Demethylation: 1 M boron tribromide in CH 2 Cl 2 , CH 2 Cl 2 , 0 °C-rt, 3 h. 2 Debenzylation: Palladium hydroxide on carbon, THF, H 2 , rt, 15 h. 3 Boc protected amine was used. Compound 4 (R)-3-(6-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-1H-pyra zolo[4,3-c]pyridin-3-yl)-2,6- difluoro-5-(trifluoromethyl)phenol [00326] A mixture of Intermediate 13.05 (75 mg, 0.16 mmol), Intermediate 2.01 (59 mg, 0.20 mmol), tris(dibenzylideneacetone)dipalladium(0) (7.19 mg, 0.01 mmol), RuPhos (7.32 mg, 0.02 mmol), and sodium tert-butoxide (45 mg, 0.47 mmol) in dioxane (3 mL) was purged with nitrogen for 5 minutes, heated in a microwave at 90 °C for 75 min, and then partitioned between CH 2 Cl 2 and saturated sodium hydrogen carbonate solution. The aqueous layer was separated and extracted with CH 2 Cl 2 . The combined organics were washed with saturated sodium hydrogen carbonate solution, dried (Na 2 SO 4 ), and then concentrated under reduced pressure. The residue was taken up in CH 3 OH. Hydrogen chloride solution (4.0 M in dioxane, 0.20 mL, 0.78 mmol) was added. The reaction was stirred at rt for 15 h and then concentrated under reduced pressure. The residue was taken up in CH 2 Cl 2 (5 mL):TFA (2 mL), stirred at rt for 1 h, concentrated under reduced pressure, and then purified by prep-HPLC (5-95% CH 3 CN:H 2 O) to give the desired product (R)-3-(6-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-1H-pyra zolo[4,3-c]pyridin-3-yl)- 2,6-difluoro-5-(trifluoromethyl)phenol as a white powder (12 mg, 13%). 1 H NMR (400 MHz, DMSO-d6): δ 13.40 (br s, 1H), 11.83-11.00 (m, 1H), 8.86 (d, J = 3.2 Hz, 1H), 7.57 (t, J = 7.1 Hz, 1H), 7.30 (d, J = 4.3 Hz, 4H), 7.24-7.17 (m, 1H), 6.74 (s, 1H), 4.87-4.77 (m, 1H), 4.16 (br d, J = 13.0 Hz, 1H), 3.71 (br d, J = 11.2, 1H), 3.48 (br d, J = 11.6 Hz, 1H), 3.28 (dt, J = 3.4, 12.4 Hz, 1H), 3.06 (dd, J = 10.3, 12.9 Hz, 1H), 2.97-2.82 (m, 5H), 2.60 (dd, J = 3.4, 12.7 Hz, 1H); LCMS: 568.5 [M+H] + . [00327] The Compounds below were synthesized from Intermediate 13.05 and the appropriate amine following the procedure described for Compound 4. Alternate conditions used: 1 THP/MOM deprotection: 1.25 M HCl in CH 3 OH, rt, 50 min then TFA/CH 2 Cl 2 (1:2), rt, overnight. 2 Isolated during the purification of Compound 4.01. 3 MOM deprotection: 4 M HCl in dioxane, CH 3 OH, 17 h, concentrated, then THP deprotection: TFA/CH 2 Cl 2 (1:5), rt, 2 h, 40 °C, 1 h, concentrated, purified, and then 4 M HCl in dioxane, CH 3 OH, 50 °C, 24 h Compound 5 (R)-3-(6-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-1-methy l-1H-pyrazolo[3,4- d]pyrimidin-3-yl)-2,6-difluoro-5-(trifluoromethyl)phenol [00328] A mixture of Intermediate 13 (100 mg, 0.24 mmol), Intermediate 2.01 (93 mg, 0.32 mmol), DIEA (0.21 mL, 1.22 mmol) and NMP (1 mL) was heated in a microwave reactor at 140 ℃ for 15 h and then partitioned between saturated NaHCO 3 and EtOAc. The aqueous layer was extracted with EtOAc. The combined organics were dried (Na 2 SO 4 ) and concentrated (note: aqueous work up was omitted for some compounds in the table below). The residue was purified by silica gel chromatography (0-100% EtOAc in hexanes) and further purified by prep-HPLC (CH 3 CN:water with 0.1% TFA) to give (R)-3-(6-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-2,6-difluoro-5-(tri fluoromethyl)phenol (11 mg, 8%) as a white powder. 1 H NMR (400 MHz, DMSO-d6): δ 11.43 (br s, 1H), 9.00 (d, J = 2.7 Hz, 1H), 7.59 (t, J = 7.1 Hz, 1H), 7.40-7.32 (m, 2H), 7.29 (br t, J = 7.3 Hz, 2H), 7.22-7.14 (m, 1H), 5.22- 5.10 (m, 1H), 4.90-4.82 (m, 1H), 3.97 (s, 3H), 3.72 (br d, J = 11.5 Hz, 1H), 3.51 (br d, J = 11.9 Hz, 1H), 3.43-3.33 (m, 1H), 3.12 (br dd, J = 9.2, 12.9 Hz, 1H), 2.93-2.79 (m, 6H); LCMS: 583.5 [M+H] + . [00329] The Compounds below were synthesized from the appropriate Intermediate and the appropriate amine following the procedure described for Compound 5.

Alternate conditions used: In some instances, Intermediate 2.06 was used instead of Intermediate 2.01. Reaction temp varied: 120-150 °C in a microwave. Reaction time varied: 0.5-15 h. 1 DMA instead of NMP. 2 TFA/CH 2 Cl 2 (1:2), rt, 15 h was needed to remove THP PG. 3 TFA/CH 2 Cl 2 (1:5), rt, 6 h then 4 M HCl in dioxane/CH 3 OH (1:5), rt, 17 h and 50 °C, 2 h to remove THP PG. 4 70 °C, microwave, 1 h then TFA/CH 2 Cl 2 (1:5), rt, 30 min to remove MOM PG. 5 Synthesized from tert-butyl (R)-3-benzylpiperazine- 1-carboxylate following the procedure described for Compound 5 then 4 M HCl/EtOAc, rt, 2 h to remove Boc and benzyl PGs (benzyl PG was partially cleaved during thermal coupling prior to deprotection). Compound 6 Methyl (R)-3-benzyl-4-(3-(2,4-difluoro-3-hydroxy-5-(trifluoromethyl )phenyl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)piperazine-1-carboxylate Step 1: tert-Butyl (R)-3-benzyl-4-(3-(3-(benzyloxy)-2,4-difluoro-5-(trifluorome thyl)phenyl)- 1-methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)piperazine-1-carbo xylate [00330] A mixture of Intermediate 13.06 (1.03 g, 2.26 mmol), (R)-tert-butyl 3- benzylpiperazine-1-carboxylate (814 mg, 2.94 mmol), DIPEA (1.2 mL, 6.79 mmol), and NMP (20 mL) was stirred at 120 °C for 10 h. The reaction mixture was allowed to cool to rt, poured into H 2 O (30 mL) slowly, and then extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (20 mL), dried (Na 2 SO 4 ), filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate=30:1 to 3:1) to give (R)-tert-butyl 3-benzyl-4-(3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl )phenyl)-1-methyl- 1H-pyrazolo[3,4-d]pyrimidin-6-yl)piperazine-1-carboxylate (705 mg, 44%) as a yellow oil and (R)-tert-butyl 3-benzyl-4-(3-(2,4-difluoro-3-hydroxy-5-(trifluoromethyl)phe nyl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)piperazine-1-carboxylate (490 mg, 35%) as a yellow oil. (R)-tert-butyl 3-benzyl-4-(3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl )phenyl)-1-methyl- 1H-pyrazolo[3,4-d]pyrimidin-6-yl)piperazine-1-carboxylate: 1 H NMR (400 MHz, DMSO-d6): δ 8.99 (s, 1H), 7.87 (t, 1H), 7.51-7.14 (m, 10H), 5.36 (s, 2H) 4.97 (s, 1H), 4.73 (d, 1H), 3.96 (s, 3H), 3.66-3.51 (m, 1H), 3.12-2.91 (m, 6H), 1.36 (s, 9H); LCMS: 695.1 [M+H] + . (R)-tert-butyl 3-benzyl-4-(3-(2,4-difluoro-3-hydroxy-5-(trifluoromethyl)phe nyl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)piperazine-1-carboxylate: 1 H NMR (400 MHz, DMSO-d6): δ 8.97 (s, 1H), 7.52 (t, 1H), 7.34-7.22 (m, 5H), 4.96 (s, 1H), 4.64 (d, 1H), 3.96 (s, 3H), 3.78-3.66 (m, 1H), 3.09-2.86 (m, 6H), 1.46 (s, 9H); LCMS: 605.1 [M+H] + . Step 2: (R)-3-(3-(Benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl) -6-(2-benzylpiperazin- 1-yl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidine hydrochloride [00331] A mixture of (R)-tert-butyl 3-benzyl-4-(3-(3-(benzyloxy)-2,4-difluoro-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)piperazine-1-carboxylate (677 mg, 0.975 mmol) and 4 M HCl in EtOAc (20 mL, 80 mmol) was stirred at rt for 2 h. The reaction mixture was concentrated to dryness to give (R)-3-(3-(benzyloxy)-2,4-difluoro-5- (trifluoromethyl)phenyl)-6-(2-benzylpiperazin-1-yl)-1-methyl -1H-pyrazolo[3,4-d]pyrimidine hydrochloride (702 mg) as a yellow solid which was used directly in the next step without further purification. LCMS: 595.1 [M+H] + . Step 3: Methyl (R)-3-benzyl-4-(3-(3-(benzyloxy)-2,4-difluoro-5-(trifluorome thyl)phenyl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)piperazine-1-carboxy late [00332] Methyl carbonochloridate (0.075 mL, 0.975 mmol) was added to a mixture of (R)-3-(3- (benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-6-(2-ben zylpiperazin-1-yl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidine hydrochloride (0.41 g, 0.649 mmol), Et3N (0.45 mL, 3.25 mmol), and CH 2 Cl 2 (5 mL) at 0 °C. The mixture was stirred at rt for 2 h, poured into water (20 mL) slowly, and then extracted with CH 2 Cl 2 (2×10 ml). The combined organic layers were washed with brine (20 mL), dried (Na 2 SO 4 ), filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate=30:1 to 3:1) to give (R)-methyl 3-benzyl-4-(3-(3-(benzyloxy)-2,4- difluoro-5-(trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4 -d]pyrimidin-6-yl)piperazine-1- carboxylate (203 mg, 47%) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6): δ 8.96 (d, 1H), 7.86 (t, 1H), 7.40-7.20 (m, 10H), 5.35 (s, 2H), 5.02 (s, 1H), 4.66 (d, 1H), 4.09-4.03 (m, 1H), 3.95 (s, 3H), 3.69 (s, 3H), 3.16-2.76 (m, 6H); LCMS: 653.1 [M+H] + . Step 4: Methyl (R)-3-benzyl-4-(3-(2,4-difluoro-3-hydroxy-5-(trifluoromethyl )phenyl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)piperazine-1-carboxy late [00333] A mixture of (R)-methyl 3-benzyl-4-(3-(3-(benzyloxy)-2,4-difluoro-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)piperazine-1-carboxylate (188 mg, 0.288 mmol) and TFA (5 mL) was stirred at 70 °C for 2 h, allowed to cool to rt, concentrated, diluted with EtOAc (20 mL), adjusted to pH=~7 with sat. aq. NaHCO 3 , and then extracted with EtOAc (2×20 mL). The combined organic layers were washed with brine (20 mL), dried (Na 2 SO 4 ), filtered, concentrated, and then purified by reverse-phase HPLC [water (0.05% HCl)/CH3CN] to give (R)-methyl 3-benzyl-4-(3-(2,4-difluoro-3-hydroxy-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)piperazine-1-carboxylate (145 mg, 89%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.41 (s, 1H), 8.96 (d, 1H), 7.57 (t, 1H), 7.32-7.26 (m, 4H), 7.19-7.15 (m, 1H), 5.02 (s, 1H), 4.65 (d, 1H), 4.08 (s, 1H), 3.95 (s, 4H), 3.69 (s, 3H), 3.31-3.30 (m, 1H), 3.05 (s, 2H), 2.91-2.76 (m, 2H); LCMS: 563.1 [M+H] + . [00334] The Compounds below were synthesized from Intermediate 13.06, the appropriate amine, and the appropriate acylating agent following the procedures described for Compound 6.

Alternate conditions used: Step 1: KF, NMP, 90 °C, overnight. 1 Step 1: 110 °C, 1.5-2 h. Step 2: 2:1 or 3:1 CH 2 Cl 2 /TFA, rt, 1.5-2.5 h. 2 Step 3: 0-50 °C, ON. 3 From Compound 6, Step 2: Compound 6 (Step 3; DIEA instead of TEA, rt, 10 min-5 h) then deprotection [ 4 (10% palladium on carbon, THF, H 2 , rt, 25-30 min) or 5 (2:1 CH 2 Cl 2 /TFA or TFA, 70 °C, 20-60 min)]. 6 Acylation from residual TFA in sample during Step 3 conditions. 7 From Compound 6, Step 2: HATU coupling (appropriate acid, HATU, DIEA, DMF, rt, 1 h) then deprotection (10% palladium on carbon, THF, H 2 , rt, 100 min). 8 Hydrolysis step following Step 4 (1 N NaOH, 1:1 CH 3 OH/THF, rt, 1 h). [00335] The Compounds below were synthesized from Compound 6 (Step 2) or Compound 6.04 (Step 2) using the following sequence: acylation (isocyanatotrimethylsilane, TEA, THF, rt, 2 h) and then deprotection (TFA, 70 °C, 2 h). [00336] The Compound below was synthesized from Compound 6 (Step 2) using the following sequence: acylation (appropriate acid, HATU, TEA, CH 2 Cl 2 , rt, 2 h-ON), deprotection (TFA, 70 °C, 2 h), and then hydrolysis (LiOH·H 2 O, THF, H 2 O, rt, 2 h). [00337] The Compound below was synthesized from Compound 6 (Step 2) using the following sequence: acylation (TCFH, NMI, CH 3 CN, rt, 2 h) and then deprotection (TFA, 70 °C, 2 h). Compound 10 (R)-3-Benzyl-4-(3-(2,4-difluoro-3-hydroxy-5-(trifluoromethyl )phenyl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)-N-methylpiperazine-1-carboxam ide Step 1: (R)-3-(6-(2-Benzylpiperazin-1-yl)-1-methyl-1H-pyrazolo[3,4-d ]pyrimidin-3-yl)-2,6- difluoro-5-(trifluoromethyl)phenol hydrochloride [00338] A mixture of (R)-tert-butyl 3-benzyl-4-(3-(2,4-difluoro-3-hydroxy-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)piperazine-1-carboxylate (Compound 6, Step 1: 950 mg, 1.57 mmol) and 4 M HCl in EtOAc (20 mL, 80 mmol) was stirred at rt for 2 h. The reaction mixture was concentrated to dryness to give (R)-3-(6-(2- benzylpiperazin-1-yl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidin-3 -yl)-2,6-difluoro-5- (trifluoromethyl)phenol hydrochloride (562 mg) as a yellow solid and used directly in the next step without further purification. 1 H NMR (400 MHz, DMSO-d6): δ 9.66 (s, 1H), 9.51 (s, 1H), 8.99 (d, 1H), 7.45-7.27 (m, 5H), 7.17 (t, 1H), 5.25 (s, 1H), 4.90 (d, 1H), 3.96 (s, 3H), 3.49-3.36 (m, 4H), 3.13-2.98 (m, 4H); LCMS: 505.2 [M+H] + . Step 2: (R)-3-Benzyl-4-(3-(2,4-difluoro-3-hydroxy-5-(trifluoromethyl )phenyl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)-N-methylpiperazine-1-carboxam ide [00339] (R)-3-Benzyl-4-(3-(2,4-difluoro-3-hydroxy-5-(trifluoromethyl )phenyl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)-N-methylpiperazine-1-carboxam ide was synthesized from (R)-3- (6-(2-benzylpiperazin-1-yl)-1-methyl-1H-pyrazolo[3,4-d]pyrim idin-3-yl)-2,6-difluoro-5- (trifluoromethyl)phenol hydrochloride and methylcarbamic chloride following the procedure described for Compound 6, Step 3. LCMS: 562.1 [M+H] + . Compound 11 (R)-3-(6-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-1-methy l-1H-pyrazolo[3,4- d]pyrimidin-3-yl)-6-chloro-2-fluorophenol [00340] Intermediate 14 (40 mg, 0.078 mmol), 4-chloro-2-fluoro-3-hydroxyphenylboronic acid (22 mg, 0.12 mmol), Pd(dppf)Cl 2 (6 mg, 0.008 mmol), DME (1 mL), water (200 µL), and potassium carbonate (35 mg, 0.25 mmol) were combined in a microwave vial. The reaction was heated in a microwave at 90 °C for 20 min, allowed to cool to rt, and then diluted with 10 mL ethyl acetate. The organic layer was washed with 10 mL water, washed with 10 mL brine, dried (Na 2 SO 4 ), filtered, concentrated, and then purified by reverse-phase HPLC [40-90% CH 3 CN in water (with 0.1% TFA)] to give (R)-3-(6-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-1-methy l- 1H-pyrazolo[3,4-d]pyrimidin-3-yl)-6-chloro-2-fluorophenol (28 mg, 68%) as a pale yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 10.71-10.60 (m, 1H), 8.98 (d, J = 2.4 Hz, 1H), 7.39- 7.27 (m, 6H), 7.22-7.15 (m, 1H), 5.23-5.11 (m, 1H), 4.92-4.80 (m, 1H), 3.96 (s, 3H), 3.72 (br d, J = 12.1 Hz, 1H), 3.51 (br d, J = 11.7 Hz, 1H), 3.41-3.30 (m, 1H), 3.12 (dd, J = 9.7, 12.7 Hz, 1H), 2.93-2.77 (m, 6H); LCMS: 531.6 [M+H] + . [00341] The Compounds below were synthesized from the appropriate Intermediate and the appropriate boronic ester or boronic acid following the procedure described for Compound 11.

Alternate conditions used: Reaction time varied: 10 min-2 h. In some instances, conventional heating was used instead of the microwave. In some instances, 2:1 or 5:1 CH 2 Cl 2 /TFA was needed to remove protecting group. 1 Used appropriate methoxy boronic acid or boronic ester and then demethylated (1 M BBr3 in CH 2 Cl 2 , CH 2 Cl 2 , -78 °C-rt, 1-41 h). 2 K 2 CO 3 , Pd(dppf)Cl 2 , CH 3 CN or dioxane, microwave, 90- 100 °C, 10-30 min. 3 K 2 CO 3 , Pd(dppf)Cl 2 , dioxane/H 2 O (2:1), 80 °C or 100 °C, 2 h. 4 Synthesized from Intermediate 14 and the appropriate methoxy boronic acid using the following sequence: Suzuki coupling (Pd(PPh3)4, 2 M Na 2 CO 3 , dioxane, 100 °C, 12 h), nitrile hydrolyzed during coupling so amide dehydration (TFAA, CH 2 Cl 2 , rt, 1 h), and then demethylation (1 M BBr 3 in CH 2 Cl 2 , CH 2 Cl 2 , -78 °C, 30 min, rt, 2 h). 5 Pd(PPh 3 ) 4 , 2 M Na 2 CO 3 , dioxane/H 2 O (2:1), 80 °C, 2 h. 6 Used appropriate benzyl protected boronic acid or boronic ester and then debenzylated (TFA, 70 °C, 30 min-2 h or 10% palladium on carbon, THF, H 2 , rt, 2 h). 7 Synthesized from Intermediate 14.06 and Intermediate 7 using the following sequence: Compound 11 coupling, hydrolysis (1 N NaOH, 2:1 THF/CH 3 OH, rt, 3 h), HATU coupling (appropriate amine, HATU, DMF, rt, 15-20 min), and then deprotection (10% palladium on carbon, THF, H 2 , rt, 30-100 min). Compound 12 (S)-3-(6-(2-Benzylmorpholino)-1-methyl-1H-pyrazolo[3,4-d]pyr imidin-3-yl)-2,6-difluoro-5- (trifluoromethyl)phenol Step 1: (R)-(4-(3-(2,4-Difluoro-3-(methoxymethoxy)-5-(trifluoromethy l)phenyl)-1-methyl- 1H-pyrazolo[3,4-d]pyrimidin-6-yl)morpholin-2-yl)methanol [00342] A mixture of Intermediate 13 (504 mg, 1.23 mmol), (R)-morpholin-2-ylmethanol hydrochloride (381 mg, 2.48 mmol), NMP (1 mL), and DIEA (0.85 mL, 4.93 mmol) was stirred at 60 °C for 1 h, allowed to cool to rt, and then diluted with 20 mL EtOAc. The organic layer was washed with water (2×20 mL), washed with 15 mL brine, dried (Na 2 SO 4 ), concentrated, and then purified by silica gel chromatography (10-60% EtOAc in heptane) to give (R)-(4-(3-(2,4- difluoro-3-(methoxymethoxy)-5-(trifluoromethyl)phenyl)-1-met hyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)morpholin-2-yl)methanol (350 mg, 58%). 1 H NMR (400 MHz, DMSO-d6): δ 9.01 (d, J = 3.1 Hz, 1H), 7.93 (t, J = 7.2 Hz, 1H), 5.33 (s, 2H), 5.02-4.79 (m, 1H), 4.75 (br d, J = 12.8 Hz, 1H), 4.59 (br d, J = 13.2 Hz, 1H), 4.04-3.95 (m, 1H), 3.93 (s, 3H), 3.61-3.49 (m, 5H), 3.49-3.40 (m, 2H), 3.14-3.02 (m, 1H), 2.91-2.77 (m, 1H); LCMS: 490.6 [M+H] + . Step 2: (R)-4-(3-(2,4-Difluoro-3-(methoxymethoxy)-5-(trifluoromethyl )phenyl)-1-methyl- 1H-pyrazolo[3,4-d]pyrimidin-6-yl)morpholine-2-carbaldehyde [00343] Dess-martin periodinane (28 mg, 0.07 mmol) was added to a solution of (R)-(4-(3-(2,4- difluoro-3-(methoxymethoxy)-5-(trifluoromethyl)phenyl)-1-met hyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)morpholin-2-yl)methanol (30 mg, 0.06 mmol) and CH 2 Cl 2 (1 mL) at rt. The mixture was stirred for 50 min, diluted with 20 mL EtOAc, washed (15 mL 1:1 saturated Na 2 S 2 O 3 :saturated NaHCO 3 and then 15 mL brine), dried (Na 2 SO 4 ), and then concentrated to give (R)-4-(3-(2,4-difluoro-3-(methoxymethoxy)-5-(trifluoromethyl )phenyl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)morpholine-2-carbaldehyde. 1 H NMR (400 MHz, CDCl 3 ): δ 9.69 (s, 1H), 8.84 (d, J = 3.2 Hz, 1H), 7.91 (t, J = 7.2 Hz, 1H), 5.19 (s, 2H), 4.75 (dd, J = 2.9, 13.5 Hz, 1H), 4.48 (br d, J = 13.7 Hz, 1H), 4.12-3.98 (m, 2H), 3.93 (s, 3H), 3.72 (br t, J = 10.2 Hz, 1H), 3.58 (s, 3H), 3.41-3.30 (m, 1H), 3.25 (dd, J = 9.8, 13.2 Hz, 1H). Step 3: (R,E)-N'-((4-(3-(2,4-Difluoro-3-(methoxymethoxy)-5-(trifluor omethyl)phenyl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)morpholin-2-yl)methy lene)-4- methylbenzenesulfonohydrazide [00344] (R)-4-(3-(2,4-Difluoro-3-(methoxymethoxy)-5-(trifluoromethyl )phenyl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)morpholine-2-carbaldehyde was dissolved in methanol (2 mL). Hydrazino(4-methylphenyl)sulfone (12 mg, 0.06 mmol) was added to the solution. The mixture was stirred at rt over the weekend, concentrated, and then purified by silica gel chromatography (0-40% EtOAc in heptane) to give (R,E)-N'-((4-(3-(2,4-difluoro-3-(methoxymethoxy)-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)morpholin-2- yl)methylene)-4-methylbenzenesulfonohydrazide (50 mg). 1 H NMR (400 MHz, DMSO-d6): δ 11.42 (s, 1H), 9.05 (d, J = 2.7 Hz, 1H), 7.95 (br t, J = 7.0 Hz, 1H), 7.70 (d, J = 8.1 Hz, 2H), 7.42 (br d, J = 7.8 Hz, 2H), 7.25 (d, J = 3.8 Hz, 1H), 5.34 (s, 2H), 4.62-4.53 (m, 1H), 4.50 (br d, J = 12.8 Hz, 1H), 4.11-4.03 (m, 1H), 3.98-3.86 (m, 4H), 3.63-3.51 (m, 4H), 3.24-3.14 (m, 1H), 3.07 (dd, J = 10.6, 13.1 Hz, 1H), 2.39 (s, 3H); LCMS: 656.6 [M+H] + . Step 4: (S)-2-Benzyl-4-(3-(2,4-difluoro-3-(methoxymethoxy)-5-(triflu oromethyl)phenyl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)morpholine [00345] A mixture of (R,E)-N'-((4-(3-(2,4-difluoro-3-(methoxymethoxy)-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)morpholin-2- yl)methylene)-4-methylbenzenesulfonohydrazide (50 mg, 0.08 mmol), potassium carbonate (35 mg, 0.25 mmol), phenylboronic acid (15 mg, 0.12 mmol), and dioxane (1.00 mL) was stirred at 100 °C for 20 min, cooled to rt, and then diluted with 20 mL EtOAc. The organic layer was washed (15 mL water and then 15 mL brine), dried (Na 2 SO 4 ), concentrated, and then purified by silica gel chromatography (0-20% EtOAc in heptane) to give (S)-2-benzyl-4-(3-(2,4-difluoro-3- (methoxymethoxy)-5-(trifluoromethyl)phenyl)-1-methyl-1H-pyra zolo[3,4-d]pyrimidin-6- yl)morpholine. LCMS: 550.6 [M+H] + . Step 5: (S)-3-(6-(2-Benzylmorpholino)-1-methyl-1H-pyrazolo[3,4-d]pyr imidin-3-yl)-2,6- difluoro-5-(trifluoromethyl)phenol [00346] (S)-2-Benzyl-4-(3-(2,4-difluoro-3-(methoxymethoxy)-5-(triflu oromethyl)phenyl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)morpholine was stirred in 2 mL CH 2 Cl 2 and 1 mL TFA at rt for 20 min, concentrated, and then purified by RP-HPLC (20-80% CH 3 CN in water with 0.1% TFA) to give (S)-3-(6-(2-benzylmorpholino)-1-methyl-1H-pyrazolo[3,4-d]pyr imidin- 3-yl)-2,6-difluoro-5-(trifluoromethyl)phenol (10 mg, 26%) as an off-white solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.41 (s, 1H), 8.94 (d, J = 2.3 Hz, 1H), 7.57 (t, J = 7.2 Hz, 1H), 7.37-7.28 (m, 4H), 7.24 (br d, J = 6.7 Hz, 1H), 4.65 (br d, J = 13.0 Hz, 1H), 4.57 (br d, J = 13.2 Hz, 1H), 3.97 (br d, J = 10.4 Hz, 1H), 3.89 (s, 3H), 3.73-3.64 (m, 1H), 3.55-3.45 (m, 1H), 3.17 - 3.04 (m, 1H), 2.93-2.81 (m, 3H); LCMS: 506.6 [M+H] + . [00347] The Compounds below were synthesized from Compound 12, Step 3 and the appropriate boronic acid following the procedure described for Compound 12, Steps 4-5.

Alternate conditions used: Step 4: 100-110 °C, 15 min-3 h. [00348] The Compound below was synthesized from Compound 12, Step 2 using the following sequence: Grignard addition (THF, 1 M tosylmagnesium bromide, 0 °C, 20 min, rt, 3.5 h) and then deprotection (2:1 CH 2 Cl 2 /TFA, rt, 25 min). Compound 14 3-(6-(1-Benzylpiperidin-2-yl)-1-methyl-1H-pyrazolo[3,4-d]pyr imidin-3-yl)-2,6-difluoro-5- (trifluoromethyl)phenol hydrochloride Step 1: tert-Butyl 6-(3-(3-(Benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)- 1-methyl- 1H-pyrazolo[3,4-d]pyrimidin-6-yl)-3,4-dihydropyridine-1(2H)- carboxylate [00349] Pd(dppf)Cl 2 (321.8 mg, 0.44 mmol) was added to a mixture of Intermediate 21 (1.63 g, 5.28mmol), Intermediate 13.06 (2.00 g, 4.40mmol), K 3 PO 4 (2.33 g, 11.0 mmol), H 2 O (2 mL), and dioxane (10 mL). The mixture was degassed with 3 vacuum/N 2 cycles, heated at 80 °C overnight. allowed to cool to rt, quenched with H 2 O (10 mL), and then extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (20 mL × 2), dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 100/1 to 5/1) to give tert-butyl 6-(3-(3-(benzyloxy)-2,4-difluoro-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)-3,4-dihydropyridine- 1(2H)-carboxylate (610 mg, 23% ) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6) δ 9.41-9.37 (m, 1H), 8.02 (t, 1H), 7.58 (d, 2H), 7.53-7.44 (m, 3H), 6.28-6.15 (m, 1H), 5.45 (s, 2H), 4.24 (s, 1H), 4.18 (s, 2H), 3.76-3.67 (m, 1H), 3.38-3.36 (m, 1H), 2.40-2.46 (m, 2H), 1.96-1.84 (m, 2H), 1.14 (s, 3H), 1.11-0.86 (m, 6H); LCMS: 602.2 [M+H] + . Step 2: tert-Butyl 2-(3-(2,4-difluoro-3-hydroxy-5-(trifluoromethyl)phenyl)-1-me thyl-1H- pyrazolo[3,4-d]pyrimidin-6-yl)piperidine-1-carboxylate [00350] A mixture of tert-butyl 6-(3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)- 1- methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)-3,4-dihydropyridine -1(2H)-carboxylate (425.3 mg, 0.71 mmol), 10% Pd/C (291 mg), and THF (5 mL) was degassed 3 vacuum/H 2 cycles and then stirred at rt for 2 h under H 2 (15 psi). The reaction mixture was diluted with THF (50 mL) and then filtered. The filtrate was concentrated and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 100/1 to 3/1) to give tert-butyl 2-(3-(2,4-difluoro-3-hydroxy-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)piperidine-1-carboxylate (210 mg, 58%) as a colorless oil. LCMS: 514.3 [M+H] + . Step 3: 2,6-Difluoro-3-(1-methyl-6-(piperidin-2-yl)-1H-pyrazolo[3,4- d]pyrimidin-3-yl)-5- (trifluoromethyl)phenol [00351] A mixture of tert-butyl 2-(3-(2,4-difluoro-3-hydroxy-5-(trifluoromethyl)phenyl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)piperidine-1-carboxy late (210.0 mg, 0.41 mol) and HCl/EtOAc (10 mL, 4 M) was stirred at rt for 0.5 h and then concentrated to give 2,6-difluoro-3- (1-methyl-6-(piperidin-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3-y l)-5-(trifluoromethyl)phenol hydrochloride (180 mg) as a white solid. LCMS: 414.1 [M+H] + . Step 4: 3-(3-(Benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-6-( 1-benzylpiperidin-2- yl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidine hydrochloride [00352] BnBr (91 µL, 0.77 mmol) was added to a mixture of 2,6-difluoro-3-(1-methyl-6- (piperidin-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-5-(triflu oromethyl)phenol hydrochloride (115 mg, 0.26 mmol), K 2 CO 3 (177 mg, 1.28 mmol), and DMF (2 mL) at rt. The reaction was heated at 70 °C for 2 h under N 2 , allowed to cool to rt, quenched with H 2 O (30 mL), and then extracted with EtOAc (20 mL × 3). The combined organic layers were washed with brine (20 mL × 2), dried over Na 2 SO 4 , filtered, concentrated, and then purified by prep-TLC (PE/EA; 1:1) to give 3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-6-( 1-benzylpiperidin-2-yl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidine (70 mg, 46%) as a colorless oil. 1 H NMR (400 MHz, CDCl 3 ): δ 9.19 (d, 1H), 7.89-7.81 (m, 1H), 7.44-7.37 (m, 2H), 7.37-7.26 (m, 6H), 7.25-7.22 (m, 2H), 5.24 (s, 2H), 4.14 (s, 3H), 3.68-3.59 (m, 1H), 3.58-3.51 (m, 1H), 3.11 (d, 1H), 3.00 (d, 1H), 2.09-2.01 (m, 1H), 1.73-1.53 (m, 4H), 1.46-1.29 (m, 2H); LCMS: 594.2 [M+H] + . Step 5: 3-(6-(1-Benzylpiperidin-2-yl)-1-methyl-1H-pyrazolo[3,4-d]pyr imidin-3-yl)-2,6- difluoro-5-(trifluoromethyl)phenol hydrochloride [00353] A mixture of 3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-6-( 1- benzylpiperidin-2-yl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidine (120.0 mg, 0.20 mmol) and TFA (10 mL) was stirred at 50 °C for 2 h. The reaction mixture was cooled to rt, concentrated, and then purified by prep-HPLC (water(HCl)-ACN) to give 3-(6-(1-benzylpiperidin-2-yl)-1-methyl- 1H-pyrazolo[3,4-d]pyrimidin-3-yl)-2,6-difluoro-5-(trifluorom ethyl)phenol hydrochloride (85 mg, 77.9%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.58 (s, 1H), 10.35 (s, 1H), 9.54 (d, 1H), 7.65 (t, 1H), 7.47-7.36 (m, 5H), 4.70 (t, 1H), 4.34 (d, 1H), 4.23 (s, 3H), 4.16 (d, 1H), 3.48-3.40 (m, 1H), 3.32-3.11 (m, 1H), 2.20 (d, 1H), 1.99-1.78 (m, 4H), 1.78-1.60 (m, 1H); LCMS: 504.1 [M+H] + . [00354] The Compound below was synthesized from Intermediate 13.06 and tert-butyl 3- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5,6-dihydropyr idine-1(2H)-carboxylate following the procedures described for Compound 14, Steps 1-3 and then reductive amination (benzaldehyde, NaBH(OAc)3, CH 2 Cl 2 , rt, 2 h). Alternate conditions used: Step 1: 3 h. Step 2: EtOAc instead of THF as solvent. Step 2: rt, 0.5 h. Step 3: 2 h. Compound 15 3-(6-(2-(Cyclohexylmethyl)-4-(methylsulfonyl)piperazin-1-yl) -1-methyl-1H-pyrazolo[3,4- d]pyrimidin-3-yl)-2,6-difluoro-5-(trifluoromethyl)phenol Step 1: 3-(3-(Benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-6-( 2-(cyclohexylmethyl)- 4-(methylsulfonyl)piperazin-1-yl)-1-methyl-1H-pyrazolo[3,4-d ]pyrimidine [00355] A mixture of Intermediate 13.06 (507 mg, 1.11 mmol), Intermediate 17 (581 mg, 2.23 mmol), KF (324 mg, 5.57 mmol), and NMP (10 mL) was stirred at 120 °C overnight. The reaction mixture was cooled to rt, poured into H 2 O (10 mL), and then extracted with EtOAc (5 mL × 3). The organic layers were washed with brine (10 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 50/1 to 5/1) to give 3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-6-( 2- (cyclohexylmethyl)-4-(methylsulfonyl)piperazin-1-yl)-1-methy l-1H-pyrazolo[3,4-d]pyrimidine (276 mg, 36%) as a yellow oil. LCMS: 679.3 [M+H] + . Step 10: 3-(6-(2-(Cyclohexylmethyl)-4-(methylsulfonyl)piperazin-1-yl) -1-methyl-1H- pyrazolo[3,4-d]pyrimidin-3-yl)-2,6-difluoro-5-(trifluorometh yl)phenol [00356] 3-(3-(Benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-6-( 2-(cyclohexylmethyl)-4- (methylsulfonyl)piperazin-1-yl)-1-methyl-1H-pyrazolo[3,4-d]p yrimidine (268 mg, 0.395 mmol) was added to a mixture of 10% Pd/C (100 mg) and THF (5 mL) under N 2 . The mixture was degassed with 3 vacuum/H 2 cycles, stirred under H 2 (15 Psi) at rt for 2 h, and then filtered through Celite with THF (500 mL) washing. The filtrate was concentrated and then purified by reverse-phase HPLC (water(0.05%HCl)-MeCN) to give 3-(6-(2-(cyclohexylmethyl)-4- (methylsulfonyl)piperazin-1-yl)-1-methyl-1H-pyrazolo[3,4-d]p yrimidin-3-yl)-2,6-difluoro-5- (trifluoromethyl)phenol (96 mg, 41%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.38 (s, 1H), 8.96 (s, 1H), 7.57 (t, 1H), 5.26-5.15 (m, 1H), 4.85-4.82 (m, 1H), 3.92 (s, 3H), 3.62-3.59 (m, 1H), 3.52-3.49 (m, 1H), 3.26-3.20 (m, 1H), 2.94-2.90 (m, 1H), 2.87 (s, 3H), 2.82-2.76 (m, 1H), 1.91-1.77 (m, 2H), 1.66-1.55 (m, 5H), 1.17-0.92 (m, 6H); LCMS: 589.1 [M+H] + . [00357] The Compounds below were synthesized from Intermediate 13.06 or Intermediate 13.10 and the appropriate Intermediate or starting material following the procedures described for Compound 15.

Alternate conditions used: Step 1: 100-120 °C, 2 h-overnight. Step 2: 1-2 h. 1 Separated by SFC from Compound 15 (DAICEL CHIRALPAK IC: 250 mm x 30 mm, 10 µM; 0.1% NH H 2 O in i-PrOH); Compound 15.01 was the first eluting enantiomer. 2 Separated by SFC from Compound 15.03 (DAICEL CHIRALPAK AD: 250 mm x 30 mm, 10 µM; 0.1% NH3·H 2 O in EtOH); Compound 15.04 was the first eluting enantiomer. 3 Separated enantiomers by SFC (DAICEL CHIRALCEL OJ: 250 mm x 30 mm, 10 µm; 0.1% NH H 2 O in MeOH; B%: 5-30%, 13 min); Compound 15.10 was the first eluting isomer. 4 Step 2: TFA, 70 °C, 30 min. [00358] The Compound below was synthesized from Intermediate 13.06 and Intermediate 19 using the following sequence: Compound 15 (Step 1), oxidation (oxone, 1:1 THF/H 2 O, rt, 2 h), and then Compound 15 (Step 2). Compound 17 3-(6-((4-Benzyl-1-(methylsulfonyl)piperidin-4-yl)(methyl)ami no)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-3-yl)-2,6-difluoro-5-(trifluorometh yl)phenol Step 1: tert-Butyl 4-benzyl-4-((3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethy l)phenyl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)(methyl)amino)piperi dine-1-carboxylate [00359] A mixture of Intermediate 13.06 (1.01 g, 2.20 mmol), Intermediate 18 (830 mg, 2.86 mmol), KF (383 mg, 6.60 mmol), and NMP (50 mL) was stirred at 120 °C for 2 h, allowed to cool to rt, poured into H 2 O (100 mL), and then extracted with ethyl acetate (40 mL × 3). The combined organic phase was washed with brine (100 mL × 3), dried with anhydrous Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 100/1 to 0/1) to give tert-butyl 4-benzyl-4-((3-(3-(benzyloxy)-2,4-difluoro-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)amino)piperidine-1- carboxylate (211 mg, 13%) as a yellow oil. LCMS: 709.4 [M+H] + . Step 2: tert-Butyl 4-benzyl-4-((3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethy l)phenyl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)(methyl)amino)piperi dine-1-carboxylate [00360] NaH (23.1 mg, 0.578 mmol, 60% purity) was added to a solution of tert-butyl 4- benzyl-4-((3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl) phenyl)-1-methyl-1H-pyrazolo[3,4- d]pyrimidin-6-yl)amino)piperidine-1-carboxylate (205 mg, 289 umol) and DMF (5 mL) at 0 °C under N 2 . The reaction was stirred at rt for 0.5 h, and then MeI (27 uL, 0.434 mmol) was added. The reaction was stirred at rt for an additional hour, poured into H 2 O (40 mL), and then extracted with ethyl acetate (20 mL × 3).The combined organic phase was washed with brine (50 mL × 3), dried with anhydrous Na 2 SO 4 , filtered, concentrated, and then purified by prep- TLC (petroleum ether/ethyl acetate: 5/1) to give tert-butyl 4-benzyl-4-((3-(3-(benzyloxy)-2,4- difluoro-5-(trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4 -d]pyrimidin-6- yl)(methyl)amino)piperidine-1-carboxylate (105 mg, 50%) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ): δ 8.92 (d, 1H), 7.94 (t, 1H), 7.53-7.46 (m, 2H), 7.43-7.37 (m, 3H), 7.26-7.16 (m, 5H), 5.30 (s, 2H), 4.01 (s, 3H), 3.97-3.87 (m, 2H), 3.83-3.71 (m, 2H), 3.65 (s, 2H), 2.92 (s, 3H), 2.16- 2.03 (m, 2H), 1.95-1.86 (m, 2H), 1.56 (s, 9H); LCMS: 723.4 [M+H] + . Step 3: 3-(3-(Benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-N-( 4-benzylpiperidin-4- yl)-N,1-dimethyl-1H-pyrazolo[3,4-d]pyrimidin-6-amine [00361] A solution of tert-butyl 4-benzyl-4-((3-(3-(benzyloxy)-2,4-difluoro-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)(methyl)amino)piperidine- 1-carboxylate (105 mg, 0.145 mmol) and HCl/EtOAc (4 M, 23 mL) was stirred at rt for 1 h and then concentrated to give 3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-N-( 4- benzylpiperidin-4-yl)-N,1-dimethyl-1H-pyrazolo[3,4-d]pyrimid in-6-amine (90 mg) as a yellow oil. Step 4: N-(4-benzyl-1-(methylsulfonyl)piperidin-4-yl)-3-(3-(benzylox y)-2,4-difluoro-5- (trifluoromethyl)phenyl)-N,1-dimethyl-1H-pyrazolo[3,4-d]pyri midin-6-amine [00362] Methanesulfonyl chloride (0.02 mL, 0.244 mmol) was added to a solution of 3-(3- (benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl)-N-(4-ben zylpiperidin-4-yl)-N,1-dimethyl- 1H-pyrazolo[3,4-d]pyrimidin-6-amine (91 mg, 0.146 mmol), TEA (0.06 mL, 0.438 mmol), and DCM (4 mL). The reaction was stirred at rt for 2 h, poured into H 2 O (15 mL), and extracted with DCM (10 mL × 3). The combined organic phase was washed with brine (30 mL), dried with anhydrous Na 2 SO 4 , filtered, concentrated, and then purified by prep-TLC (petroleum ether/ethyl acetate: 2/1) to give N-(4-benzyl-1-(methylsulfonyl)piperidin-4-yl)-3-(3-(benzylox y)-2,4- difluoro-5-(trifluoromethyl)phenyl)-N,1-dimethyl-1H-pyrazolo [3,4-d]pyrimidin-6-amine (55 mg, 53%) as a white solid. 1 H NMR (400 MHz, CDCl 3 ): δ 8.93 (d, 1H), 7.94 (t, 1H), 7.55-7.45 (m, 2H), 7.44-7.37 (m, 3H), 7.24-7.20 (m, 3H), 7.05-6.99 (m, 2H), 5.30 (s, 2H), 4.02 (s, 3H), 3.65-3.55 (m, 2H), 2.94 (s, 3H), 2.91-2.84 (m, 3H), 2.75-2.70 (m, 4H), 2.04-1.93 (m, 2H), 1.28- 1.25 (m, 2H); LCMS: 701.4 [M+H] + . Step 5: 3-(6-((4-Benzyl-1-(methylsulfonyl)piperidin-4-yl)(methyl)ami no)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-3-yl)-2,6-difluoro-5-(trifluorometh yl)phenol [00363] BCl 3 (1 M in DCM, 0.46 mL) was added to a solution of N-(4-benzyl-1- (methylsulfonyl)piperidin-4-yl)-3-(3-(benzyloxy)-2,4-difluor o-5-(trifluoromethyl)phenyl)-N,1- dimethyl-1H-pyrazolo[3,4-d]pyrimidin-6-amine (106 mg, 0.151 mmol) and DCM (10 mL) at - 70 °C. The mixture was stirred at -70 °C for 1 h, poured (while still cold) into a stirring mixture of MeOH (5 mL) and sat. aq. NaHCO 3 (20 mL), stirred for 5 min, and then extracted with DCM (15 mL × 3). The combined organic phase was washed with brine (50 mL), dried with anhydrous Na 2 SO 4 , filtered, concentrated, and then purified by reverse-phase HPLC (Phenomenex Luna C18; water (HCl)-ACN) to give 3-(6-((4-benzyl-1- (methylsulfonyl)piperidin-4-yl)(methyl)amino)-1-methyl-1H-py razolo[3,4-d]pyrimidin-3-yl)- 2,6-difluoro-5-(trifluoromethyl)phenol (24.4 mg, 26%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.40 (s, 1H), 9.01 (d, 1H), 7.61 (t, 1H), 7.26-7.14 (m, 3H), 7.05-7.69 (m, 2H), 3.95 (s, 3H), 3.54-3.25 (m, 4H), 3.11-2.76 (m, 10H), 2.07-1.94 (m, 2H); LCMS: 611.1 [M+H] + . [00364] The Compound below was synthesized from Intermediate 13.06 and Intermediate 18.01 following the procedures described for Compound 17. [00365] The Compounds below were synthesized from Intermediate 13.06 and the appropriate piperazine starting material following the procedures described for Compound 17 (Steps 1,3-4) and then deprotection (TFA, 50 °C or 70 °C, 2 h). Alternate conditions used: 1 Separated by SFC from Compound 18.04 (DAICEL CHIRALPAK IC: 250 mm x 30 mm, 10 µM; [0.1% NH H 2 O in i-PrOH] B%: 28%-28%, 10 min); Compound 18.05 was the first eluting enantiomer. Compound 19 (R)-3-(6-(2-(Cyclopropylmethyl)-4-(methylsulfonyl)piperazin- 1-yl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-3-yl)-2,6-difluoro-5-(trifluorometh yl)phenol [00366] (R)-3-(6-(2-(Cyclopropylmethyl)-4-(methylsulfonyl)piperazin- 1-yl)-1-methyl-1H- pyrazolo[3,4-d]pyrimidin-3-yl)-2,6-difluoro-5-(trifluorometh yl)phenol was synthesized from Intermediate 13.06 and tert-butyl 3-(cyclopropylmethyl)piperazine-1-carboxylate using the following sequence: Compound 17 [Steps 1 (110 °C, overnight), 3, & 4 (overnight)], deprotection (10% Pd/C, THF, H 2 , rt, 2 h), and then SFC separation (2 nd eluting enantiomer from DIACEL CHIRALPAK IG: 250 mm x 30 mm, 10 µM; 0.1% NH3·H 2 O in CH 3 OH). 1 H NMR (400 MHz, DMSO-d6): δ 11.66-11.19(m, 1H), 8.97 (d, 1H), 7.55 (t, 1H), 3.93 (s, 3H), 3.73 (d, 1H), 3.62 (d, 1H), 3.24-3.15 (m, 1H), 2.98-2.92(m, 1H), 2.92-2.88 (m, 3H), 2.82 (d, J = 11.9, 1H), 2.37-2.28 (m, 1H), 1.82-1.71 (m, 1H), 1.51-1.38 (m, 1H), 0.66 (d, 1H), 0.46-0.28 (m, 2H), 0.13-0.22 (m, 1H), 0.22-0.13 (m, 1H), 0.13-0.2 (m, 1H); LCMS: 547.1[M+H] + . [00367] The Compound below was the first eluting enantiomer from the SFC separation of Compound 19. [00368] The Compounds below were synthesized from Compound 19 (Step 2) using the following sequence: acylation (TFAA, TEA, CH 2 Cl 2 , rt, 1 h), SFC separation (DIACEL CHIRALPAK IG: 250 mm x 30 mm, 10 µM; [0.1% NH3·H 2 O in CH 3 OH] B%: 25%-25%, 11 min), and then each isomer was deprotected separately (TFA, 50 °C, 2 h). Compound 21 (R)-5-(6-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-1-methy l-1H-pyrazolo[3,4- d]pyrimidin-3-yl)-2,4-difluoro-3-hydroxybenzonitrile Step 1: (R)-5-(6-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-1-methy l-1H- pyrazolo[3,4-d]pyrimidin-3-yl)-2,4-difluoro-3-methoxybenzoni trile [00369] Pd(PPh3) 2 Cl 2 (13.7 mg, 19.5 µmol) and CuI (7.4 mg, 39 µmol) was added to a solution of Intermediate 14 (0.1 g, 195 µmol), Intermediate 7.01 (89.4 mg, 195 µmol), and dioxane (5 mL) under N 2 . The mixture was stirred at 100 °C for 16 h, cooled to rt, poured into H 2 O (30 mL), and then extracted with EtOAc (30 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 9/1 to 3/1) to give (R)-5-(6-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-2,4-difluoro-3-meth oxybenzonitrile (246 mg, 54%) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ): δ 8.96 (d, 1H), 7.96 (t, 1H), 7.41-7.46 (m, 2H), 7.33 (t, 2H), 7.18-7.25 (m, 1H), 5.19-5.30 (m, 1H), 4.92-5.04 (m, 1H), 4.13-4.18 (m, 3H), 4.04 (s, 3H), 3.91-3.99 (m, 1H), 3.76 (d, 1H), 3.40-3.52 (m, 1H), 3.23-3.33 (m, 1H), 2.74-2.90 (m, 6H); LCMS: 554.2 [M+H] + . Step 2: (R)-5-(6-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-1-methy l-1H- pyrazolo[3,4-d]pyrimidin-3-yl)-2,4-difluoro-3-hydroxybenzoni trile [00370] BBr3 (429 µL, 4.45 mmol) was added to a solution of (R)-5-(6-(2-benzyl-4- (methylsulfonyl)piperazin-1-yl)-1-methyl-1H-pyrazolo[3,4-d]p yrimidin-3-yl)-2,4-difluoro-3- methoxybenzonitrile (246 mg, 445 µmol) and DCM (5 mL) at 0 ℃. The mixture was stirred at rt for 16 h, quenched with dropwise addition of MeOH (10 mL), stirred for 10 min at rt, poured into H 2 O (20 mL), and then extracted with EtOAc (20 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, concentrated, and then purified by prep-HPLC (Phenomenex Luna C18; water(HCl)-ACN) to give (R)-5-(6-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-2,4-difluoro-3-hydr oxybenzonitrile (52 mg, 22%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.50 (s, 1H), 9.03 (d, 1H), 7.80 (t, 1H), 7.23- 7.42 (m, 4H), 7.11-7.22 (m, 1H), 5.07-5.33 (m, 1H), 4.69-4.97 (m, 1H), 3.97 (s, 3H), 3.67-3.79 (m, 1H), 3.46-3.57 (m, 2H), 3.04-3.18 (m, 1H), 2.75-2.94 (m, 6H); LCMS: 540.2 [M+H] + . [00371] The Compound below was synthesized from Intermediate 14 and Intermediate 7.02 following the procedures described for Compound 21 (Step 1) and then Compound 6 (Step 4). Alternate conditions used: Step 1: 2 h. Compound 22 (R)-3-(6-(2-Benzyl-4-ethylpiperazin-1-yl)-1-methyl-1H-pyrazo lo[3,4-d]pyrimidin-3-yl)-2,6- difluoro-5-(trifluoromethyl)phenol hydrochloride Step 1: (R)-tert-Butyl 3-benzyl-4-(3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl )phenyl)- 1-methyl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)piperazine-1-carbo xylate [00372] Pd(dppf)Cl 2 •CH 2 Cl 2 (321 mg, 0.392 mmol) was added to a mixture of Intermediate 14.03 (2.1 g, 3.93 mmol), Intermediate 7 (1.95 g, 4.72 mmol), Na 2 CO 3 (2 M in H 2 O, 4.9 mL), and dioxane (40 mL) under N 2 . The mixture was degassed with 3 vacuum/N 2 cycles, stirred at 80 °C for 2 h under N 2 , allowed to cool to rt, poured into H 2 O (100 mL), and then extracted with ethyl acetate (50 mL × 3). The combined organic layers were washed with brine (50 mL), dried (Na 2 SO 4 ), filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 5/1) to give (R)-tert-butyl 3-benzyl-4-(3-(3-(benzyloxy)-2,4-difluoro-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)piperazine-1-carboxylate (2.2 g, 80%) as yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 8.98 (s, 1H), 7.86 (t, 1H), 7.55 - 7.46 (m, 2H), 7.46 - 7.24 (m, 7H), 7.21 (d, 1H), 5.35 (s, 2H), 5.09 - 4.82 (m, 1H), 4.63 (d, 1H), 4.16 - 4.06 (m, 1H), 4.00 - 3.91 (m, 3H), 3.90 - 3.77 (m, 1H), 3.13 - 2.83 (m, 3H), 2.73 (d, 1H), 1.46 (s, 9H), 1.32 - 1.25 (m, 1H); LCMS: 695.7 [M+H] + . Step 2: (R)-3-(3-(Benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl) -6-(2-benzylpiperazin- 1-yl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidine hydrochloride [00373] A mixture of (R)-tert-butyl 3-benzyl-4-(3-(3-(benzyloxy)-2,4-difluoro-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi n-6-yl)piperazine-1-carboxylate (1.8 g, 2.59 mmol) and HCl/EtOAc (4 M, 50 mL) was stirred at rt for 2 h and then concentrated to give (R)-3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl) -6-(2-benzylpiperazin-1- yl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidine hydrochloride (1.9 g) as yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 9.75 (d, 1H), 9.57 (d, 1H), 9.00 (d, 1H), 7.87 (t, 1H), 7.53 - 7.47 (m, 2H), 7.46 - 7.36 (m, 5H), 7.30 (t, 2H), 7.21 - 7.15 (m, 1H), 5.36 (s, 2H), 5.25 (d, 1H), 4.92 (d, 1H), 3.96 (s, 3H), 3.56 - 3.31 (m, 3H), 3.21 - 2.96 (m, 4H); LCMS: 595.2 [M+H] + . Step 3: (R)-6-(2-Benzyl-4-ethylpiperazin-1-yl)-3-(3-(benzyloxy)-2,4- difluoro-5- (trifluoromethyl)phenyl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidi ne [00374] Iodoethane (0.03 mL, 0.48 mmol) was added to a mixture of (R)-3-(3-(benzyloxy)-2,4- difluoro-5-(trifluoromethyl)phenyl)-6-(2-benzylpiperazin-1-y l)-1-methyl-1H-pyrazolo[3,4- d]pyrimidine hydrochloride (150 mg, 0.237 mmol), K 2 CO 3 (98.6 mg, 0.713 mmol), and acetone (5 mL). The mixture was stirred at 50 °C overnight, allowed to cool to rt, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 1/1) to give (R)-6-(2- benzyl-4-ethylpiperazin-1-yl)-3-(3-(benzyloxy)-2,4-difluoro- 5-(trifluoromethyl)phenyl)-1- methyl-1H-pyrazolo[3,4-d]pyrimidine (92 mg, 62%) as a yellow oil. 1 H NMR (400 MHz, DMSO-d6): δ 8.96 (d, 1H), 7.87 (t, 1H), 7.61-7.08 (m, 9H), 7.63-7.02 (m, 1H), 5.36 (s, 2H), 5.05-4.90 (m, 1H), 4.73-4.58 (m, 1H), 4.07-3.87 (m, 3H), 3.18 (dd, 1H), 3.02 (d, 1H), 2.79 (d, 2H), 2.36-2.20 (m, 1H), 2.09-1.94 (m, 1H), 1.93-1.85 (m, 1H), 1.37-1.14 (m, 2H), 1.04 (t, 3H); LCMS: 623.3 [M+H] + . Step 4: (R)-3-(6-(2-Benzyl-4-ethylpiperazin-1-yl)-1-methyl-1H-pyrazo lo[3,4-d]pyrimidin-3- yl)-2,6-difluoro-5-(trifluoromethyl)phenol hydrochloride [00375] A mixture of (R)-3-(3-(benzyloxy)-2,4-difluoro-5-(trifluoromethyl)phenyl) -6-(2- benzylpiperazin-1-yl)-1-methyl-1H-pyrazolo[3,4-d]pyrimidine hydrochloride (70 mg, 0.106 mmol) and TFA (3 mL) was stirred at 70 °C for 2 h under N 2 , allowed to cool to rt, concentrated, and then purified by prep-HPLC (water (0.05% HCl)/CH 3 CN) to give (R)-3-(6-(2- benzyl-4-ethylpiperazin-1-yl)-1-methyl-1H-pyrazolo[3,4-d]pyr imidin-3-yl)-2,6-difluoro-5- (trifluoromethyl)phenol hydrochloride (45 mg, 73%) as a white solid. 1 H NMR (400 MHz, DMSO-d 6 ,): δ 11.42 (s, 1H), 11.02-10.36 (m, 1H), 9.00 (d, 1H), 7.58 (t, 1H), 7.45 (s, 2H), 7.35- 7.26 (m, 1H), 7.30 (t, 1H), 7.23-7.15 (m, 1H), 5.54-5.19 (m, 1H), 5.13-4.84 (m, 1H), 3.97 (s, 3H), 3.73-3.45 (m, 3H), 3.26-2.94 (m, 6H), 1.40-1.23 (m, 3H). LCMS: 533.3 [M+H] + . [00376] The Compounds below were synthesized from the appropriate Intermediates following the procedures described for Compound 22. Alternate conditions used: Step 3: CH 3 CN instead of acetone as solvent. Step 3: 80 °C. 1 Step 1 then Step 4. 2 Synthesized from Compound 22.03 (Step 1) using the following sequence: Boc deprotection (4 M HCl in EtOAC, rt, 2 h) and then methylation (formaldehyde in formic acid, 100 °C, overnight). 3 Synthesized from Compound 22.03 (Step 1) using the following sequence: Boc deprotection (4 M HCl in EtOAc, rt, 2 h) and then Compound 22 (Steps 3-4). 4 Synthesized from Compound 22.03 (Step 1) using the following sequence: Boc deprotection (4 M HCl in EtOAc, rt, 2 h), alkylation (appropriate alkylating agent, K 2 CO 3 , CH 3 CN, 80 °C, overnight), and then Compound 22 (Step 4). [00377] The Compounds below were synthesized from Compound 22.08 (Step 1) by oxidation (oxone, 1:1 THF, H 2 O, 0 °C, 1 h), prep-TLC separation, and then deprotection (Compound 22, Step 4). [00378] The Compound below was synthesized from Compound 22 (Step 2) using the following sequence: acylation ((4-nitrophenyl) carbonochloridate, TEA, CH 2 Cl 2 , 0 °C-rt, 2 h), alkylation (azetidine, K 2 CO 3 , DMF, rt, overnight), and then deprotection (10% palladium on carbon, THF, H 2 , rt, 2 h). [00379] The Compound below was synthesized from Compound 22.03 (Step 1) using the following sequence: Boc deprotection (4 M HCl in EtOAc, rt, 2 h), acylation ((4-nitrophenyl) carbonochloridate, TEA, CH 2 Cl 2 , 0 °C-rt, 2 h), alkylation (azetidine, K 2 CO 3 , DMF, 80 °C, overnight), and then deprotection (10% palladium on carbon, THF, H 2 , rt, 2 h). Compound 26 (R)-3-(5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-(meth ylamino)-1H-pyrazolo[4,3- d]pyrimidin-1-yl)-6-chloro-2-fluoro-5-(trifluoromethyl)pheno l Step 1: 5-Chloro-3-iodo-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyra zolo[4,3-d]pyrimidine [00380] SEM-Cl (6.6 mL, 37.4 mmol) was added dropwise to a solution of Intermediate 8, Step 1 (7.0 g, 25.0 mmol), DIEA (13.1 mL, 74.9 mmol), and DMF (50 mL) at 0 °C. The reaction was stirred at 25 °C for 2 h, poured into H 2 O (200 mL), and then extracted with EtOAc (50 mL × 3). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 30/1→3/1) to give 5-chloro-3-iodo-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyra zolo[4,3- d]pyrimidine (5.5 g, 54%) as a white solid. 1 H NMR (400 MHz, DMSO-d6): δ 9.52 (s, 1H), 5.87 (s, 2H), 3.57-3.55 (t, 2H), 0.83-0.80 (t, 2H), 0.10 (s, 9H); LCMS: 411.0 [M+H] + . Step 2: (R)-5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-iodo-1-( (2- (trimethylsilyl)ethoxy)methyl)-1H-pyrazolo[4,3-d]pyrimidine [00381] KF (2.84 g, 48.9 mmol) was added to a solution of 5-chloro-3-iodo-1-((2- (trimethylsilyl)ethoxy)methyl)-1H-pyrazolo[4,3-d]pyrimidine (2.0 g, 4.9 mmol) and Intermediate 2.06 (2.48 g, 9.74 mmol) in water (60 mL). The mixture was stirred at 100 °C for 16 h, additional KF (2.83 g, 48.7 mmol) was added, and the mixture was stirred at 100 °C for an additional 24 h. The reaction was cooled to rt and extracted with EtOAc (50 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 20/1→1/1) to give (R)-5-(2-benzyl-4- (methylsulfonyl)piperazin-1-yl)-3-iodo-1-((2-(trimethylsilyl )ethoxy)methyl)-1H-pyrazolo[4,3- d]pyrimidine (1.1 g, 36%) as yellow oil. 1 H NMR (400 MHz, CDCl 3 ): δ 8.92 (s, 1H), 7.56-7.55 (m, 2H), 7.38-7.34 (m, 2H), 7.29-7.23 (m, 1H), 5.72 (s, 2H), 5.19-5.09 (m, 1H), 4.97-4.88 (m, 1H), 3.96-3.94 (m, 1H), 3.79-3.76 (m, 1H), 3.66-3.61 (m, 2H), 3.59-3.57 (m, 1H), 3.30-3.24 (m, 1H), 2.88-2.75 (m, 6H), 0.93 (t, 2H), 0.01 (s, 9H); LCMS: 629.2 [M+H] + . Step 3: (R)-5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-iodo-1H- pyrazolo[4,3- d]pyrimidine [00382] TFA (20 mL, 270 mmol) was added to a solution of (R)-5-(2-benzyl-4- (methylsulfonyl)piperazin-1-yl)-3-iodo-1-((2-(trimethylsilyl )ethoxy)methyl)-1H-pyrazolo[4,3- d]pyrimidine (3.7 g, 5.9 mmol) and DCM (40 mL). The reaction was stirred at 20 °C for 16.5 h, concentrated, and then diluted with THF (50 mL) and 1 M NaOH (20 mL). This mixture was stirred at 20 °C for 0.5 h, diluted with water (30 mL), and then extracted with EtOAc (30 mL × 3). The combined organic extracts were dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 10/1→0/1) to give (R)-5- (2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-iodo-1H-pyrazo lo[4,3-d]pyrimidine (1.8 g, 48%) as a yellow solid. 1 H NMR (400 MHz, CDCl 3 ): δ 8.88 (s, 1H), 7.54-7.52 (m, 2H), 7.35-7.31 (m, 2H), 7.23-7.20 (m, 1H), 5.11-5.09 (m, 1H), 4.88-4.85 (d, 1H), 3.93 (d, 1H), 3.75 (d, 1H), 3.42 (t, 1H), 3.23-3.18 (m, 2H), 2.87-2.73 (m, 6H); LCMS: 499.0 [M+H]+. Step 4: (R)-3-(5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-iodo- 1H-pyrazolo[4,3- d]pyrimidin-1-yl)-6-chloro-2-fluoro-5-(trifluoromethyl)pheno l [00383] TEA (10.0 mL, 71.9 mmol), Cu(OAc) 2 (1.82 g, 10.0 mmol), and pyridine (10.0 mL, 124 mmol) were added to a solution of (R)-5-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-3- iodo-1H-pyrazolo[4,3-d]pyrimidine (1.0 g, 2.0 mmol), Intermediate 6.02 (1.02 g, 3.01 mmol), and DCM (10 mL). The mixture was degassed with 3 vacuum/O2 cycles, heated at 40 °C for 16 h under O2, cooled to rt, concentrated, diluted with 0.5 M HCl (50 mL), and then extracted with EtOAc (10 mL × 3). The combined organic layers were dried over Na 2 SO 4 , filtered, concentrated, and then purified by reverse-phase HPLC (Phenomenex Luna; water (HCl)-ACN) to give (R)-3-(5-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-iodo- 1H-pyrazolo[4,3- d]pyrimidin-1-yl)-6-chloro-2-fluoro-5-(trifluoromethyl)pheno l (0.6 g, 40%) as a yellow solid. 1 H NMR (400 MHz, CD 3 OD): δ 8.82 (d, 1H), 7.58 (d, 1H), 7.49 (d, 2H), 7.26 (t, 2H), 7.15 (t, 1H), 5.10-5.08 (m, 1H), 4.87-4.84 (m, 1H), 3.85 (d, 1H), 3.67 (d, 1H), 3.43-3.37 (m, 1H), 3.15-3.14 (m, 1H), 2.91-2.81 (m, 6H); LCMS: 710.9 [M+H] + . Step 5: (R)-3-(5-(2-Benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-(meth ylamino)-1H- pyrazolo[4,3-d]pyrimidin-1-yl)-6-chloro-2-fluoro-5-(trifluor omethyl)phenol [00384] Xantphos (40 mg, 69 umol), Cs 2 CO 3 (140 mg, 430 µmol), and Xantphos Pd G4 (40 mg, 42 µmol) were added to a solution of (R)-3-(5-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)- 3-iodo-1H-pyrazolo[4,3-d]pyrimidin-1-yl)-6-chloro-2-fluoro-5 -(trifluoromethyl)phenol (100 mg, 141 µmol), methyl amine (2 M in THF, 1 mL), and amyl alcohol (3 mL). The reaction was degassed with 3 vacuum/N 2 cycles, heated at 80 °C for 16 h in a sealed reactor, cooled to rt, diluted with 0.5 M HCl (50 mL), and then extracted with EtOAc (30 mL × 3; the reaction was run in 6 parallel batches and combined for work up). The combined organic layers were dried over Na 2 SO 4 , filtered, concentrated, and then purified by silica gel chromatography (petroleum ether/ethyl acetate: 5/1→0/1) followed by reverse-phase HPLC (Phenomenex Luna; water (HCl)-ACN) to give (R)-3-(5-(2-benzyl-4-(methylsulfonyl)piperazin-1-yl)-3-(meth ylamino)-1H- pyrazolo[4,3-d]pyrimidin-1-yl)-6-chloro-2-fluoro-5-(trifluor omethyl)phenol (120 mg, 23%) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): δ 11.59 (s, 1H), 8.81 (d, 1H), 7.49 (d, 1H), 7.34- 7.32 (m, 2H), 7.30-7.26 (m, 2H), 7.20-7.16 (m, 1H), 6.44 (s, 1H), 5.08-5.06 (m, 1H), 4.71 (d, 1H), 3.70 (d, 1H), 3.46 (d, 1H), 3.36-3.29 (m, 1H), 3.09 (dd, 1H), 2.95 (s, 3H), 2.89 (s, 3H), 2.86-2.82 (m, 2H), 2.74 (dd, 1H); LCMS: 614.0 [M+H] + . [00385] The Compounds below were synthesized in a similar manner to the procedures described herein and in PCT/US2021/052679. Example A-1: Parenteral Pharmaceutical Composition [00386] To prepare a parenteral pharmaceutical composition suitable for administration by injection (subcutaneous, intravenous), 1-1000 mg of a compound described herein, or a pharmaceutically acceptable salt or solvate thereof, is dissolved in sterile water and then mixed with 10 mL of 0.9% sterile saline. A suitable buffer is optionally added as well as optional acid or base to adjust the pH. The mixture is incorporated into a dosage unit form suitable for administration by injection. Example A-2: Oral Solution [00387] To prepare a pharmaceutical composition for oral delivery, a sufficient amount of a compound described herein, or a pharmaceutically acceptable salt thereof, is added to water (with optional solubilizer(s), optional buffer(s), and taste masking excipients) to provide a 20 mg/mL solution. Example A-3: Oral Tablet [00388] A tablet is prepared by mixing 20-50% by weight of a compound described herein, or a pharmaceutically acceptable salt thereof, 20-50% by weight of microcrystalline cellulose, 1-10% by weight of low-substituted hydroxypropyl cellulose, and 1-10% by weight of magnesium stearate or other appropriate excipients. Tablets are prepared by direct compression. The total weight of the compressed tablets is maintained at 100 -500 mg. Example A-4: Oral Capsule [00389] To prepare a pharmaceutical composition for oral delivery, 10-500 mg of a compound described herein, or a pharmaceutically acceptable salt thereof, is mixed with starch or other suitable powder blend. The mixture is incorporated into an oral dosage unit such as a hard gelatin capsule, which is suitable for oral administration. [00390] In another embodiment, 10-500 mg of a compound described herein, or a pharmaceutically acceptable salt thereof, is placed into size 4 capsule, or size 1 capsule (hypromellose or hard gelatin) and the capsule is closed. Example A-5: Topical Gel Composition [00391] To prepare a pharmaceutical topical gel composition, a compound described herein, or a pharmaceutically acceptable salt thereof, is mixed with hydroxypropyl cellulose, propylene glycol, isopropyl myristate and purified alcohol USP. The resulting gel mixture is then incorporated into containers, such as tubes, which are suitable for topical administration. Example B-1: HSD17b13 NAD(P)H-Glo Biochemical Assay Materials [00392] Recombinant human HSD17B13 enzyme. Substrate: estradiol (Sigma β-Estradiol E8875), 100 mM in DMSO. Cofactor: NAD+ Grade I free acid (Sigma 10127965001), 20 mM in H 2 O. Assay buffer final concentration: 20 mM Tris pH7.4 with 0.002% Tween-20 and 0.02% BSA. Assay performed in 384 well solid bottom plate (Corning 3570). Enzymatic activity detected by NAD(P)H-Glo™ Detection System (Promega G9062). Compounds [00393] Inhibitor compounds were serially diluted in DMSO and then further diluted in assay buffer to a 10X concentration consisting of 1% DMSO. Procedure [00394] HSD17b13 enzyme was diluted in 1X assay buffer to the desired enzyme concentration based on the specific activity of the enzyme lot.20 uL of diluted enzyme was added to each well along with 2.5 uL of 10X inhibitor solution. Assay plate was incubated at RT for 20 minutes, and then 2.5 uL of a 10X substrate/cofactor mix was added to each well for a final concentration of 50 uM estradiol and 1 mM NAD+. Assay plate was incubated at 37 °C for 3 hours. NAD(P)H-Glo™ Detection System reagents were prepared according to manufacturer’s specifications, and 25 uL was added to each well. After incubating for 1 hour at RT, luminescence was measured. [00395] Representative data for exemplary compounds disclosed herein is presented in Table 3. TABLE 3 Example B-2: HSD17b1 NAD(P)H-Glo Biochemical Assay Materials [00396] Recombinant human HSD17B1 enzyme. Substrate: testosterone (Sigma T1500), 100 mM in DMSO. Cofactor: NADP disodium salt (Sigma 10128031001), 20 mM in H 2 O. Assay buffer final concentration: 20 mM Tris pH 7.4 with 0.002% Tween-20 and 0.02% BSA. Assay performed in 384 well solid bottom plate (Corning 3570). Enzymatic activity detected by NAD(P)H-Glo™ Detection System (Promega G9062). Compounds [00397] Inhibitor compounds were serially diluted in DMSO and then further diluted in assay buffer to a 10X concentration consisting of 1% DMSO. Procedure [00398] HSD17b1 enzyme was diluted in 1X assay buffer to the desired enzyme concentration based on the specific activity of the enzyme lot.20 uL of diluted enzyme was added to each well along with 2.5 uL of the 10X inhibitor solution. Assay plate was incubated at RT for 20 minutes, and then 2.5 uL of a 10X substrate/cofactor mix was added to each well for a final concentration of 55 uM testosterone and 1 mM NADP. Assay plate was incubated at 37 °C for 1 hour. NAD(P)H-Glo™ Detection System reagents were prepared according to manufacturer’s specifications, and 25uL was added to each well. After incubating for 1 hour at RT, luminescence was measured. Example B-3: HSD17b2 NAD(P)H-Glo Biochemical Assay Materials and Setup [00399] Recombinant human HSD17B2 enzyme. Substrate: estradiol (Sigma β-Estradiol E8875) 2mM in DMSO. Cofactor: NAD+ Grade I free acid (Sigma 10127965001), 20mM in H 2 O. Assay buffer final concentration: 20mM Tris pH7.4 with 0.002% Tween-20 and 0.02% BSA. Assay performed in 384 well solid bottom plate (Corning 3570). Enzymatic activity detected by NAD(P)H-Glo™ Detection System (Promega G9062). Compounds [00400] Inhibitor compounds were serially diluted in DMSO and then further diluted in assay buffer to a 10X concentration consisting of 1% DMSO. Procedure [00401] HSD17b2 enzyme was diluted in 1X assay buffer to the desired enzyme concentration based on the specific activity of the enzyme lot. 20uL of diluted enzyme was added to each well along with 2.5 uL of 10X inhibitor solution. Assay plate was incubated at RT for 20 minutes, and then 2.5 uL of 10X substrate/cofactor mix was added to each well for a final assay concentration of 1 uM estradiol and 500 uM NAD+. Assay plate was incubated at RT for 1 hour. NAD(P)H-Glo™ Detection System reagents were prepared according to manufacturer’s specifications and 25 uL was added to each well. After incubating for 1 hour at RT, luminescence was measured. Example B-4: In Vitro HSD17b13 Cell Based Assay Seeding [00402] HEK293 cells were plated at 4,000,000 cells per T75 flask with EMEM (ATCC Cat # 30-2003) and 10% FBS (Sigma Cat # F2442) and then incubated at 37 °C in 5% CO 2 for 18 hours. Transfection and plate [00403] After the 18 h incubation, media was replaced with 15 mL of fresh media: EMEM without Phenol Red (Quality Biological Cat # 112-212-101), 10% CSS (Sigma Cat # F6765) and GlutaMax (Gibco Cat # 35050-061). In a polypropylene tube, 20 ug pCMV6 HSD17B13 (Origene Cat # RC213132) was diluted in OptiMEM (Life Technologies, Cat # 31985-062) to 2 mL. 60 uL of transfection reagent (X-tremeGENE HP Roche, Cat # 06366236001) was added, and the tube was vortexed and incubated at room temperature for 20 minutes. The transfection reagent/DNA mixture was added to the cells in the T75 flask, and the cells were incubated at 37 °C in 5% CO 2 for 18 hours. The next day, the cells were resuspended in EMEM media with 10% CSS and plated in a 96 well plate at 80,000 cells/well, 100 uL/well. Cells were incubated at 37 °C in 5% CO 2 for 18 hours. Test Compounds [00404] Compounds were serially diluted in DMSO (1000X final concentration) and then further diluted in EMEM media with 10% CSS to a 20X final concentration.10 uL of the 20X compound mix was added to each well of transfected cells, and the cells were incubated at 37 °C in 5% CO2 for 30 minutes. 100 uL of EMEM media with 100 uM estradiol (Sigma cat# E8875) was added to each well, and the cells were incubated for 4 hours at 37 °C in 5% CO 2 . The cell media was collected and examined for estradiol and estrone concentrations by LCMS. Example B-5: In Vitro HSD17b11 Cell Based Assay Seeding [00405] HEK293 cells were plated at 4,000,000 cells per T75 flask with EMEM (ATCC Cat # 30-2003) and 10% FBS (Sigma Cat # F2442) and then incubated at 37 °C in 5% CO 2 for 18 hours. Transfection and plate [00406] After the 18 h incubation, the media was replaced with 15 mL of fresh media: EMEM without Phenol Red (Quality Biological Cat # 112-212-101), 10% CSS (Sigma Cat # F6765) and GlutaMax (Gibco Cat # 35050-061). In a polypropylene tube, 20 ug pCMV6 HSD17B11 (Origene Cat # RC205941) was diluted in OptiMEM (Life Technologies, Cat # 31985-062) to 2 mL.60 uL of transfection reagent (X-tremeGENE HP Roche, Cat # 06366236001) was added, and the tube was vortexed and incubated at room temperature for 20 minutes. The transfection reagent/DNA mixture was added to the cells in the T75 flask, and the cells were incubated at 37 °C in 5% CO 2 for 18 hours. The next day, the transfected cells were resuspended in EMEM media with 10% CSS and plated in a 96 well plate at 80,000 cells/well, 100 uL/well. Cells were incubated at 37 °C in 5% CO 2 for 18 hours. Test Compounds [00407] Compounds were serially diluted in DMSO (1000X final concentration) and then further diluted in EMEM media with 10% CSS to a 20X final concentration.10 uL of the 20X compound mix was added to each well of the transfected cells, and the cells were incubated at 37 °C in 5% CO 2 for 30 minutes.100 uL of EMEM media with 60 uM of estradiol (Sigma cat# E8875) was added, and the cells were incubated for 4 hours at 37 °C in 5% CO 2 . The cell media was examined for estradiol and estrone concentrations by LCMS. Example B-6: NASH Activity Study (AMLN model) [00408] NASH is induced in male C57BL/6 mice by diet-induction with AMLN diet (DIO- NASH) (D09100301, Research Diet, USA) (40% fat (18% trans-fat), 40% carbohydrates (20% fructose) and 2% cholesterol). The animals are kept on the diet for 29 weeks. After 26 weeks of diet induction, liver biopsies are performed for base line histological assessment of disease progression (hepatosteatosis and fibrosis), stratified and randomized into treatment groups according to liver fibrosis stage, steatosis score, and body weight. Three weeks after biopsy the mice are stratified into treatment groups and dosed daily by oral gavage with an HSD17B13 inhibitor for 8 weeks. At the end of the study liver biopsies are performed to assess hepatic steatosis and fibrosis by examining tissue sections stained with H&E and Sirius Red, respectively. Total collagen content in the liver is measured by colorimetric determination of hydroxyproline residues by acid hydrolysis of collagen. Triglycerides and total cholesterol content in liver homogenates are measured in single determinations using autoanalyzer Cobas C- 111 with commercial kit (Roche Diagnostics, Germany) according to manufacturer`s instructions. Example B-7: CCl 4 Fibrosis Model [00409] Fibrosis is induced in C57BL/6 male mice by bi-weekly oral administration of CCl 4 . CCl 4 is formulated 1:4 in oil and is oral dosed at a final concentration of 0.5ul/g mouse. After 2- 4 weeks of fibrosis induction the compounds is administered daily by oral gavage for 2-8 weeks of treatment while continuing CCl 4 administration. At study termination livers are formalin fixed and stained with H&E or Sirius Red stain for histopathological evaluation of inflammation and fibrosis. Total collagen content is measured by colorimetric determination of hydroxyproline residues by acid hydrolysis of collagen. Collagen gene induction is measured by qPCR analysis of Col1a1 and Col3a1 mRNA. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) are measured by a clinical chemistry analyzer. Example B-8: Mouse PK Study [00410] The plasma pharmacokinetics of any one of the compounds disclosed herein as a test article is measured following a single bolus intravenous and oral administration to mice (CD-1, C57BL, and diet induced obesity mice). Test article is formulated for intravenous administration in a vehicle solution of DMSO, PEG400, hydroxypropyl-β-cyclodextrin (HPβCD) and is administered (for example at a dose volume of 3 mL/kg) at selected dose levels. An oral dosing formulation is prepared in appropriate oral dosing vehicles (vegetable oils, PEG400, Solutol, citrate buffer, or carboxymethyl cellulose) and is administered at a dose volume of 5~10 mL/kg at selected dose levels. Blood samples (approximately 0.15 mL) are collected by cheek pouch method at pre-determined time intervals post intravenous or oral doses into tubes containing EDTA. Plasma is isolated by centrifugation of blood at 10,000 g for 5 minutes, and aliquots are transferred into a 96-well plate and stored at -60 ^C or below until analysis. [00411] Calibration standards of test article are prepared by diluting DMSO stock solution with DMSO in a concentration range. Aliquots of calibration standards in DMSO are combined with plasma from naïve mouse so that the final concentrations of calibration standards in plasma are 10-fold lower than the calibration standards in DMSO. PK plasma samples are combined with blank DMSO to match the matrix. The calibration standards and PK samples are combined with ice-cold acetonitrile containing an analytical internal standard and centrifuged at 1850 g for 30 minutes at 4°C. The supernatant fractions are analyzed by LC/MS/MS and quantitated against the calibration curve. Pharmacokinetic parameters (area under the curve (AUC), Cmax, Tmax, elimination half-life (T1/2), clearance (CL), steady state volume of distribution (Vdss), and mean residence time (MRT)) are calculated via non-compartmental analysis using Microsoft Excel (version 2013). Example B-9: Mouse CDA-HFD NASH Model [00412] A NASH phenotype with mild fibrosis can be induced in C57BL/6 mice by feeding a choline-deficient diet with 0.1% methionine and 60% kcal fat (Research Diet A06071302) for 4- 12 weeks. After 4-6 weeks of diet induction compounds can be administered daily by oral gavage for 4-8 weeks of treatment while continuing CDA-HFD feeding. At study termination livers can be formalin fixed and stained with H&E and Sirius Red stain histopathological evaluation of steatosis, inflammation, and fibrosis. Total collagen content can be measured by colorimetric determination of hydroxyproline residues by acid hydrolysis of collagen. Collagen gene induction can be measured by qPCR analysis of Col1a1 or Col3a1. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) can be measured by a clinical chemistry analyzer.