Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TYK2 INHIBITORS AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2024/042361
Kind Code:
A1
Abstract:
Described herein are compounds that are TYK2 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 that would benefit from modulation of TYK2 activity.

Inventors:
PANDEY ANJALI (US)
MANOJVEER SEETHARAMAN (IN)
THAKKAR MAHESH (IN)
DURAISWAMY ATHISAYAMANI (IN)
Application Number:
PCT/IB2023/000502
Publication Date:
February 29, 2024
Filing Date:
August 22, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SUDO BIOSCIENCES LTD (GB)
International Classes:
C07D519/00; A61K31/437; A61P19/02; C07D487/04
Domestic Patent References:
WO2022175745A12022-08-25
WO2022175746A12022-08-25
WO2022175747A12022-08-25
WO2022175752A12022-08-25
WO2020061101A12020-03-26
WO2020061103A12020-03-26
WO2004018482A22004-03-04
WO2015069310A12015-05-14
Foreign References:
US20150005297A12015-01-01
Other References:
"Handbook of Pharmaceutical Salts: Properties, Selection and Use", 2002, WILEY-VCH/VHCA, article "International Union of Pure and Applied Chemistry"
S.M. BERGEL.D. BIGHLEYD.C. MONKHOUSE, J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
JEAN JACQUESANDRE COLLETSAMUEL H. WILEN: "Enantiomers, Racemates and Resolutions", 1981, JOHN WILEY AND SONS, INC.
"Method in Enzymology", vol. 42, 1985, ACADEMIC, pages: 309 - 396
BUNDGAARD, H.: "A Textbook of Drug Design and Development", vol. 5, 1991, article "Design and Application of Prodrugs", pages: 113 - 191
BUNDGAARD, H., ADVANCED DRUG DELIVERY REVIEW, vol. 8, 1992, pages 1 - 38
"Remington: The Science and Practice of Pharmacy", 1995, JOHN WILEY AND SONS, INC.
HOOVER, JOHN E.: "Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING CO.
"Pharmaceutical Dosage Forms", 1980, MARCEL DECKER
"Pharmaceutical Dosage Forms and Drug Delivery Systems", 1999, LIPPINCOTT WILLIAMS & WILKINS
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A compound of Formula (I):

Formula (I), or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

Y is a ligand that binds the hinge-binding region in the allosteric binding site of TYK2; L is a linker that covalently connects Y and the rest of the molecule;

Ring A is an unsubstituted or substituted carbocyclic ring wherein A1 and A2 are both C, or an unsubstituted or substituted 5- or 6-membered heterocyclic ring wherein A1 and A2 are independently N or C, wherein if Ring A is substituted then Ring A is substituted with p instances of R8; each R8 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR17, -C(=O)R16, -CO2R16, - C(=O)N(R16)2, -N(R16)2, -NR16C(=O)R17, -SR16, -S(=O)R17, -SO2R17, or - SO2N(R16)2; wherein if R8 is attached to a nitrogen atom, then R8 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -C(=O)R16, -CO2R16, - C(=O)N(R16)2, -S(=O)R17, -SO2R17, or -SO2N(R16)2; or two R8 attached to the same carbon atom are taken together to form =0, =S, or =NH;

Z is -NR10-, -0-, -S-, -S(=0)-, or -SO2-;

R10 is hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, or monocyclic heterocycle; X1, X2, and X3 are each independently CR11 or N; each R11 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2- Ce alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR17, -C(=O)R16, -CO2R16, - C(=O)N(R16)2, -N(R16)2, -NR16C(=O)R17, -SR16, -S(=O)R17, -SO2R17, or - SO2N(R16)2; each R6 and R7 is independently hydrogen, deuterium, halogen, Ci-Ce alkyl, Ci-Ce deuteroalkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, monocyclic heterocycle, -CN, - OH, -OR17, -C(=O)R16, -CO2R16, -C(=O)N(R16)2, -N(R16)2, -NR16C(=O)R17, -SR16, - S(=O)R17, -SO2R17, or -SO2N(R16)2; or one R6 and one R7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C=O or a C3-C6 cycloalkane; each R16 is independently hydrogen, substituted or unsubstituted Ci-Ce alkyl, substituted or unsubstituted Ci-Ce fluoroalkyl, substituted or unsubstituted Ci-Ce heteroalkyl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted monocyclic 3- to 8-membered heterocycloalkyl, substituted or unsubstituted phenyl, or substituted or unsubstituted monocyclic heteroaryl; or two R16 on the same N atom are taken together with the N atom to which they are attached to form a substituted or unsubstituted N-containing heterocycle; and each R17 is independently substituted or unsubstituted Ci-Ce alkyl, substituted or unsubstituted Ci-Ce fluoroalkyl, substituted or unsubstituted Ci-Ce heteroalkyl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted monocyclic 3- to 8-membered heterocycloalkyl, substituted or unsubstituted phenyl, or substituted or unsubstituted monocyclic heteroaryl; wherein each substituted alkyl, substituted alkenyl, substituted alkynyl, substituted fluoroalkyl, substituted deuteroalkyl, substituted alkoxy, substituted fluoroalkoxy, substituted heteroalkyl, substituted carbocycle, and substituted heterocycle is substituted with one or more Rs groups independently selected from the group consisting of deuterium, halogen, Ci-Ce alkyl, monocyclic carbocycle, monocyclic heterocycle, -CN, -CH2CN, -OR18, -CH2OR18, -CO2R18, -CH2CO2R18, - C(=O)N(R18)2, -CH2C(=O)N(R18)2, -N(R18)2, -CH2N(R18)2, -NR18C(=O)R18, - CH2NR18C(=O)R18, -NR18SO2R19, -CH2NR18SO2R19, -SR18, -CH2SR18, -S(=O)R19, - CH2S(=O)R19, -SO2R19, -CH2SO2R19, -SO2N(R18)2, and -CH2SO2N(R18)2; each R18 is independently selected from hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C2-C6 heterocycloalkyl, phenyl, benzyl, 5- membered heteroaryl and 6-membered heteroaryl; or two R18 groups are taken together with the N atom to which they are attached to form a N-containing heterocycle; each R19 is independently selected from Ci-Ce alkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C2-C6 heterocycloalkyl, phenyl, benzyl, 5-membered heteroaryl, and 6-membered heteroaryl; n is 1, 2, or 3; and p is 0, 1, 2, or 3; provided the compound is not a compound listed in Table 2. The compound of claim 1, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, having a structure of Formula (II):

Formula (II) wherein:

L is a bond, -O-, -S-, or -N(R5)-, wherein:

R5 is hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, or monocyclic heterocycle;

Ring C is selected from unsubstituted or substituted carbocycle and unsubstituted or substituted heterocycle, wherein if Ring C is substituted carbocycle or substituted heterocycle, then Ring C is substituted with s instances of Rc, further wherein: s is 0, 1, 2, 3, 4, or 5; and each Rc is independently selected from selected from -N(R')(R2), -C(=0)-W-R4, and R12, wherein:

R1 is hydrogen, Ci-Ce alkyl, or Ci-Ce fluoroalkyl;

R2 is a Ring B that is an unsubstituted or substituted heterocycle or unsubstituted or substituted carbocycle, wherein if Ring B is substituted then Ring B is substituted with q instances of R13, further wherein: each R13 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR17, -C(=O)R16, -CO2R16, -C(=O)N(R16)2, - N(R16)2, -NR16C(=O)R17, -SR16, -S(=O)R17, -SO2R17, -SO2N(R16)2, or -P(=O)(R16)2; or two R13 groups on adjacent atoms of R2 are taken together with the intervening atoms to which they are attached to form an unsubstituted or substituted 5- or 6-membered monocyclic carbocycle or an unsubstituted or substituted 5- or 6-membered monocyclic heterocycle; and q is 0, 1, 2, 3, or 4; or R2 is -C(=O)R14, -C(=O)NR14R15, or -C(=O)OR14;

R14 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted monocyclic carbocycle, unsubstituted or substituted bicyclic carbocycle, unsubstituted or substituted monocyclic heterocycle, or unsubstituted or substituted bicyclic heterocycle;

R15 is hydrogen, Ci-Ce alkyl, or Ci-Ce fluoroalkyl; or R14 and R15 are taken together with the intervening atoms to which they are attached to form an unsubstituted or substituted 4- to 6-membered monocyclic heterocycle; or R1 and R15 are taken together with the intervening atoms to which they are attached to form an unsubstituted or substituted 5- or 6-membered monocyclic heterocycle;

W is -NR3- or -O-;

R3 is hydrogen, Ci-Ce alkyl, Ci-Ce deuteroalkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, or monocyclic heterocycle;

R4 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce alkoxy, unsubstituted or substituted C3-C6 cycloalkyl, or unsubstituted or substituted monocyclic heterocycle; or R3 and R4 are taken together with the N atom to which they are attached to form a substituted or unsubstituted N-containing heterocycle; or R3 and one R12 are taken together with the intervening atoms to which they are attached to form a substituted or unsubstituted 5- or 6-membered heterocycle; and each R12 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, - OH, -OR17, -C(=O)R16, -CO2R16, -C(=O)N(R16)2, -N(R16)2, - NR16C(=O)R17, -SR16, -S(=O)R17, -SO2R17, or -SO2N(R16)2; or two R12 attached to the same aliphatic carbon atom are taken together to form oxo; or two R12 attached to two adjacent carbon atoms are taken together to form a group selected from unsubstituted or substituted carbocycle and unsubstituted or substituted heterocycle. The compound of claim 2, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, having a structure of Formula (III- A):

Formula (III- A) or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

A1 and A2 are each independently N or C; and

A3 is S, O, N, NR8, CR8, or C=O; and

A4 and A5 are each independently S, O, N, NR8, or CR8; wherein at least one of A1 and A2 is C, or at least one of A3, A4, and A5 is CR8. The compound of claim 2, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, having a structure of Formula (III-B):

Formula (III-B), or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein: A6 is N or CR8. The compound according to any one of claims 2-4, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is selected from: unsubstituted or substituted carbocycle, unsubstituted or substituted monocyclic heterocycloalkyl, unsubstituted or substituted 5-membered heteroaryl, and unsubstituted or substituted bicyclic heterocycle; and , wherein:

X4, X5, and X6 are each independently CH, CRc, or N; and t is 0, 1, or 2. The compound of claim 5, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein when X4 is CH, X5 is N, and X6 is CH, then t is 1 or 2.

The compound according to any one of claims 2-6, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein when Ring then

Ring C is selected from:

RD is -N(R1)(R2), -C(=O)-W-R4, or R20, wherein:

R20 is chloro, bromo, iodo, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR17, -C(=O)R16, -CO2R16, - C(=O)N(R16)2, -N(R16)2, -NR16C(=O)R17, -SR16, -S(=O)R17, - SO2R17, or -SO2N(R16)2.

The compound according to any one of claims 2-7, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein when Ring then R4 is hydrogen, Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce alkoxy, unsubstituted or substituted C3-C6 cycloalkyl, or unsubstituted or substituted monocyclic heterocycle.

The compound according to any one of claims 2-6, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring The compound according to any one of claims 2-6 and 9, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring The compound according to any one of claims 5, 6, 9, and 10, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

X4 is CH; X5 is N; and X6 is CH;

X4 is CH; X5 is N; and X6 is CRc;

X4 is CH; X5 is CRc; and X6 is N;

X4 is N; X5 is N; and X6 is CH;

X4 is N; X5 is N; and X6 is N; or

X4 is CH; X5 is N; and X6 is N. The compound according to any one of claims 5, 6, and 9-11, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

X4 is CH; X5 is N; and X6 is CRc;

X4 is CH; X5 is CRc; and X6 is N; or

X4 is CH; X5 is N; and X6 is N. The compound according to any one of claims 2-12, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is selected from: The compound according to any one of claims 2-13, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is selected from: The compound according to any one of claims 2-5, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is unsubstituted or substituted bicyclic heterocycle. The compound according to any one of claims 2-5, and 15, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is substituted bicyclic heterocycle. The compound according to any one of claims 2-5, 15, and 16, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is selected from: The compound according to any one of claims 2-5 and 15-17, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is selected from:

The compound according to any one of claims 2-18, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each Rc is independently selected from -N(R4)(R2), -C(=O)-W-R4, and R12. The compound according to any one of claims 2-19, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein at least one Rc is -N(R1)(R2). The compound according to any one of claims 2-20, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein at least one Rc is -C(=0)-W-R4. The compound according to any one of claims 2-21, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein at least one Rc is R12. The compound according to any one of claims 2-15 and 19-21, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is selected from:

24. The compound according to any one of claims 2-22, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is selected from:

The compound according to any one of claims 2-24, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R1 is hydrogen or C1-C4 alkyl.

The compound according to any one of claims 2-25, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R1 is hydrogen.

The compound according to any one of claims 2-26, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein W is -NR3-.

The compound according to any one of claims 2-27, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R3 is hydrogen, C1-C4 alkyl, or C1-C4 deuteroalkyl.

The compound according to any one of claims 2-28, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R3 is hydrogen.

The compound according to any one of claims 2-29, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R4 is hydrogen, unsubstituted or substituted C1-C4 alkyl, unsubstituted or substituted C1-C4 deuteroalkyl, unsubstituted or substituted C1-C4 alkoxy, or unsubstituted or substituted C3-C6 cycloalkyl.

The compound according to any one of claims 2-30, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R4 is hydrogen, C1-C4 alkyl, C1-C4 deuteroalkyl, or C1-C4 alkoxy.

The compound according to any one of claims 2-31, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R4 is hydrogen, methyl, trideuteromethyl, methoxy, or ethoxy.

The compound according to any one of claims 2-30, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R4 is substituted C3-C6 cycloalkyl. The compound according to any one of claims 2-30 and 33, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R4 is substituted cyclopropyl or cyclobutyl. The compound according to any one of claims 2-30, 33, and 34, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R4 is 2-methoxycyclobutyl or 2- fluorocyclopropyl . The compound according to any one of claims 2-35, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R12 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, or - N(R16)2; or two R12 attached to the same aliphatic carbon atom are taken together to form oxo; or two R12 attached to two adjacent carbon atoms are taken together to form an unsubstituted or substituted heterocycle. The compound according to any one of claims 2-36, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R12 is independently substituted Ci-Ce alkyl, substituted carbocycle, substituted heterocycle; or two R12 attached to two adjacent carbon atoms are taken together to form a substituted heterocycle. The compound according to any one of claims 2-37, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R12 is independently substituted with 1, 2, or 3 Rs groups independently selected from deuterium, halogen, monocyclic carbocycle, and -CH2OH. The compound according to any one of claims 2-38, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R12 is independently substituted with 1, 2, or 3 Rs groups independently selected from deuterium, fluoro, cyclopropyl, and -CH2OH. The compound according to any one of claims 2-39, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R12 is independently fluoro, methyl, ethyl, trideuteromethyl, difluoromethyl, phenyl, 2,6-difluorophenyl, 2-hydroxymethyl- l,3,4-oxadiazol-5-yl, amino, or methylamino; or two R12 attached to the same aliphatic carbon atom are taken together to form oxo; or two R12 attached to two adjacent carbon atoms are taken together to form 2- cyclopropyl-4,5-dihydro-lH-imidazole. The compound according to any one of claims 2-40, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R12 is independently methyl, 2- hydroxymethyl-l,3,4-oxadiazol-5-yl, or amino; or two R12 attached to the same aliphatic carbon atom are taken together to form oxo. The compound of any one of claims 2-41, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R2 is a Ring B that is an unsubstituted or substituted heterocycle or unsubstituted or substituted carbocycle, wherein if Ring B is substituted then Ring B is substituted with q instances of R13. The compound of any one of claims 2-42, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

R2 is a Ring B that is an unsubstituted or substituted monocyclic heterocycle, wherein if Ring B is substituted then Ring B is substituted with q instances of R13. The compound of any one of claims 2-43, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

R2 is a Ring B that is an unsubstituted or substituted monocyclic 6-membered heteroaryl, wherein if Ring B is substituted then Ring B is substituted with q instances of R13. The compound of any one of claims 2-44, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

R2 is a Ring B that is an unsubstituted or substituted pyridinyl or unsubstituted or substituted pyrimidinyl, wherein if Ring B is substituted then Ring B is substituted with q instances of R13. The compound of any one of claims 2-45, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

Ring The compound of any one of claims 2-46, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein q is 1 or 2. The compound of any one of claims 2-47, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R13 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR17, - C(=O)R16, -CO2R16, -C(=O)N(R16)2, -N(R16)2, -NR16C(=O)R17, -SO2R17, -SO2N(R16)2, or

-P(=O)(R16)2. The compound of any one of claims 2-48, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R13 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, or -CN. The compound of any one of claims 2-49, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R13 is independently -F, -CH3, -CH2N(CH3)2, or -CN. The compound of any one of claims 2-50, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring B is 5-fluoropyridin-2-yl, 6-cyanopyridin-2- yl, 4-aminopyrimidin-6-yl, 5-((dimethylamino)methyl)pyridin-2-yl, or 2,6- dimethylpyrimidin-4-yl. The compound of any one of claims 2-51, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring B is 2,6-dimethylpyrimidin-4-yl. The compound of any one of claims 2-52, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R2 is -C(=O)R14, -C(=O)NR14R15, or -C(=O)OR14. The compound according to any one of claims 2-53, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R2 is -C(=O)R14. The compound of any one of claims 2-41 and 54, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R14 is unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C3-C4 cycloalkyl, or unsubstituted or substituted 4-membered heterocycloalkyl; wherein the substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, or substituted heterocycloalkyl is substituted with one or more Rs groups independently selected from the group consisting of deuterium, halogen, -CN, -NH2, - OH, -NH(CH3), -N(CH3)2, -CH3, -CH2CH3, -CHF2, -CF3, -0CH3, -0CHF2, and -OCF3. The compound of any one of claims 2-41, 54, and 55, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R14 is unsubstituted or substituted C3-C4 cycloalkyl. The compound of any one of claims 2-41 and 54-56, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R14 is cyclopropyl.

58. The compound according to any one of claims 2-57, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is selected from:

59. The compound according to any one of claims 2-58, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is selected from:

The compound according to any one of claims 2-59, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is selected from: The compound according to any one of claims 2-60, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring C is selected from: The compound according to any one of claims 1-61, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein L is a bond or -N(R5)-. The compound according to any one of claims 1-62, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein L is -N(R5)-. The compound according to any one of claims 1-63, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R5 is hydrogen. The compound according to any one of claims 1-62, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein L is a bond. The compound of any one of claims 1-65, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Ring A is an unsubstituted or substituted pyrazole or an unsubstituted or substituted triazole, wherein if Ring A is substituted then Ring A is substituted with p instances of R8. The compound of any one of claims 3-66, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

A1 is C; A2 is C; A3 is N; A4 is NR8; and A5 is CR8; or A1 is C; A2 is N; A3 is N; A4 is CR8; and A5 is N; or A1 is N; A2 is C; A3 is N; A4 is N; and A5 is CR8; The compound of any one of claims 1-67, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R8 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, or unsubstituted or substituted carbocycle; wherein if R8 is attached to a nitrogen atom, then R8 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, or unsubstituted or substituted carbocycle; or two R8 attached to the same carbon atom are taken together to form =0. The compound of any one of claims 1-68, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R8 is independently hydrogen or unsubstituted or substituted Ci-Ce alkyl. The compound of any one of claims 1-69, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R8 is independently hydrogen or methyl. The compound of any one of claims 1-70, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein p is 0 or 1. The compound of any one of claims 1-71, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein p is 1. The compound of any one of claims 1-72, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R6 and R7 is independently hydrogen, deuterium, Ci-Ce alkyl, or Ci-Ce deuteroalkyl; or one R6 and one R7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form a C3-C6 cycloalkane. The compound of any one of claims 1-73, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R6 and R7 is independently hydrogen or methyl. The compound of any one of claims 1-74, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

X1 is CR11, X2 is CR11, and X3 is CR11; or X1 is CR11, X2 is CR11, and X3 is N; or X1 is CR11, X2 is N, and X3 is CR11; or X1 is CR11, X2 is N, and X3 is N; or X1 is N, X2 is CR11, and X3 is CR11; or X1 is N, X2 is CR11, and X3 is N; or X1 is N, X2 is N, and X3 is CR11. The compound of any one of claims 1-75, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

X1 is CR11, X2 is CR11, and X3 is CR11; or X1 is CR11, X2 is CR11, and X3 is N. The compound of any one of claims 1-76, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R11 is independently hydrogen, halogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, -CN, -OH, -OR17, or -N(R16)2. The compound of any one of claims 1-77, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein each R11 is independently hydrogen. The compound of any one of claims 1-78, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

X1 is CH, X2 is CH, and X3 is CH; or X1 is CH, X2 is CH, and X3 is N. The compound of any one of claims 1-79, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein Z is -NR10-, -O-, or -S-. The compound of any one of claims 1-80, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R10 is hydrogen, Ci-Ce alkyl, or Ci-Ce fluoroalkyl. The compound of any one of claims 1-81, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R10 is Ci-Ce alkyl. The compound of any one of claims 1-82, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein R10 is methyl. The compound of any one of claims 1-83, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein n is 1 or 2.

The compound of any one of claims 1-84, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein selected from:

86. The compound according to any one of claims 1-85, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein the compound is selected from:

87. The compound according to any one of claims 1-86, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein the compound is selected from:

88. The compound according to any one of claims 1-87, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein the compound is selected from:

The compound according to any one of claims 1-88, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein the compound is selected from: A pharmaceutical composition comprising the compound of any one of claims 1-89, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, and a pharmaceutically acceptable excipient. A method of treating a disease or condition in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of any one of claims 1-89, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, or a pharmaceutical composition of claim 90. The method of claim 91, wherein the disease or condition is a TYK2 -mediated disease or condition. The method of claim 91 or claim 92, wherein the disease or condition is an inflammatory disease or condition or an autoimmune disease or condition. The method according to any one of claims 91-93, wherein the disease or condition is an inflammatory disease or condition. The method of either claim 93 or claim 94, wherein the inflammatory disease or condition is a neuroinflammatory disease or condition. The method of any one of claims 91-95, wherein the disease or condition is a neurodegenerative disease or condition. The method of any one of claims 91-96, wherein the disease or condition is selected from multiple sclerosis, stroke, epilepsy, encephalomyelitis, polyneuropathy, encephalitis, and a neuromyelitis optica spectrum disorder. The method of claim 97, wherein the disease or condition is multiple sclerosis. The method of claim 98, wherein the multiple sclerosis is relapsing or relapsingremitting. The method of claim 97, wherein the disease or condition is a neuromyelitis optica spectrum disorder. The method of claim 100, wherein the disease or condition is neuromyelitis optica. The method of claim 97, wherein the disease or condition is encephalomyelitis. The method of claim 102, wherein the disease or condition is acute disseminated encephalomy eliti s . The method of claim 97, wherein the disease or condition is polyneuropathy. The method of claim 104, wherein the disease or condition is chronic inflammatory demyelinating polyneuropathy. The method of claim 97, wherein the disease or condition is encephalitis. The method of claim 106, wherein the disease or condition is autoimmune encephalitis. The method of any one of claims 91-96, wherein the disease or condition is selected from rheumatoid arthritis, multiple sclerosis, psoriasis, psoriatic arthritis, lupus, systemic lupus erythematosus, Sjogren’s syndrome, ankylosing spondylitis, vitiligo, atopic dermatitis, scleroderma, alopecia, hidradenitis suppurativa, uveitis, dry eye, intestinal bowel disease, Crohn’s disease, ulcerative colitis, celiac disease, Bechet’s disease, type 1 diabetes, systemic sclerosis, and idiopathic pulmonary fibrosis.

Description:
TYK2 INHIBITORS AND USES THEREOF

CROSS-REFERENCE TO RELATED APPLICATION

[0001] This application claims benefit of U.S. Provisional Patent Application No. 63/400,689, filed on August 24, 2022, which is incorporated herein by reference in its entirety.

BACKGROUND OF THE INVENTION

[0002] The present disclosure relates to compounds that bind to the pseudokinase domain (JH2) of the non-receptor tyrosine-protein kinase 2 (TYK2). Compounds of the present disclosure may inhibit certain cytokine signaling, for example IL-12, IL-23, and IFN-a signaling. Additional aspects of the disclosure include pharmaceutical compositions comprising the compounds described herein, methods of using the compounds to treat certain diseases, and intermediates and processes useful in the synthesis of the compounds.

[0003] TYK2 is a non-receptor tyrosine kinase member of the Janus kinase (JAKs) family of protein kinases. The mammalian JAK family consists of four members, TYK2, JAK1, JAK2, and JAK3. JAK proteins, including TYK2, are integral to cytokine signaling. TYK2 associates with the cytoplasmic domain of type I and type II cytokine receptors, as well as interferon types I and III receptors, and is activated by those receptors upon cytokine binding. Cytokines implicated in TYK2 activation include interferons (e.g. IFN-a, IFN-P, IFN-K, IFN-5, IFN-S, IFN-T, IFN-CO, and IFN-^ (also known as limitin), and interleukins (e.g. IL-6, IL-10, IL-12, IL- 23, oncostatin M, ciliary neurotrophic factor, cardiotrophin 1, cardiotrophin-like cytokine, and LIF). The activated TYK2 then goes on to phosphorylate further signaling proteins such as members of the STAT family, including STAT1, STAT2, STAT4, and STAT6.

SUMMARY OF THE INVENTION

[0004] Compounds described herein are modulators of the JAK family of kinases. More specifically, the compounds of the present disclosure are inhibitors of TYK2. In some embodiments, compounds are selective for TYK2 over other JAKs. For example, compounds may bind specifically to the pseudokinase domain (JH2) of TYK2 thereby enhancing selectivity over JAK family members. In some embodiments, a compound of the present disclosure may be an allosteric modulator or noncompetitive inhibitor of TYK2. In additional embodiments, a compound described herein may be useful in the treatment of TYK2 mediated diseases or disorders. [0005] In one aspect, described herein is a compound of Formula (I):

Formula (I), or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

Y is a ligand that binds the hinge-binding region in the allosteric binding site of TYK2; L is a linker that covalently connects Y and the rest of the molecule;

Ring A is an unsubstituted or substituted carbocyclic ring wherein A 1 and A 2 are both C, or an unsubstituted or substituted 5- or 6-membered heterocyclic ring wherein A 1 and A 2 are independently N or C, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 ; each R 8 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , - C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , -NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , - SO2R 17 , or -SO2N(R 16 )2; wherein if R 8 is attached to a nitrogen atom, then R 8 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci- Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 ) 2 , -S(=O)R 17 , -SO2R 17 , or -SO2N(R 16 )2; or two R 8 attached to the same carbon atom are taken together to form =0, =S, or =NH;

Z is -NR 10 -, -0-, -S-, -S(=0)-, or -SO2-;

R 10 is hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, or monocyclic heterocycle;

X 1 , X 2 , and X 3 are each independently CR 11 or N; each R 11 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , -C(=O)R 16 , -CO 2 R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , - NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , -SO 2 R 17 , or -SO 2 N(R 16 ) 2 ; each R 6 and R 7 is independently hydrogen, deuterium, halogen, Ci-Ce alkyl, Ci-Ce deuteroalkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, monocyclic heterocycle, -CN, -OH, -OR 17 , -C(=O)R 16 , - CO 2 R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , -NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , -SO 2 R 17 , or - SO 2 N(R 16 ) 2 ; or one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C=O or a C3-C6 cycloalkane; each R 16 is independently hydrogen, substituted or unsubstituted Ci-Ce alkyl, substituted or unsubstituted Ci-Ce fluoroalkyl, substituted or unsubstituted Ci-Ce heteroalkyl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted monocyclic 3- to 8-membered heterocycloalkyl, substituted or unsubstituted phenyl, or substituted or unsubstituted monocyclic heteroaryl; or two R 16 on the same N atom are taken together with the N atom to which they are attached to form a substituted or unsubstituted N-containing heterocycle; and each R 17 is independently substituted or unsubstituted Ci-Ce alkyl, substituted or unsubstituted Ci-Ce fluoroalkyl, substituted or unsubstituted Ci-Ce heteroalkyl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted monocyclic 3- to 8-membered heterocycloalkyl, substituted or unsubstituted phenyl, or substituted or unsubstituted monocyclic heteroaryl; wherein each substituted alkyl, substituted alkenyl, substituted alkynyl, substituted fluoroalkyl, substituted deuteroalkyl, substituted alkoxy, substituted fluoroalkoxy, substituted heteroalkyl, substituted carbocycle, and substituted heterocycle is substituted with one or more R s groups independently selected from the group consisting of deuterium, halogen, Ci- Ce alkyl, monocyclic carbocycle, monocyclic heterocycle, -CN, -CH 2 CN, -OR 18 , -CH 2 OR 18 , -CO 2 R 18 , -CH 2 CO 2 R 18 , -C(=O)N(R 18 ) 2 , -CH 2 C(=O)N(R 18 ) 2 , -N(R 18 ) 2 , -CH 2 N(R 18 ) 2 , - NR 18 C(=O)R 18 , -CH 2 NR 18 C(=O)R 18 , -NR 18 SO 2 R 19 , -CH 2 NR 18 SO 2 R 19 , -SR 18 , -CH 2 SR 18 , - S(=O)R 19 , -CH 2 S(=O)R 19 , -SO 2 R 19 , -CH 2 SO 2 R 19 , -SO 2 N(R 18 ) 2 , and -CH 2 SO 2 N(R 18 ) 2 ; each R 18 is independently selected from hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C 2 -Ce heterocycloalkyl, phenyl, benzyl, 5-membered heteroaryl and 6-membered heteroaryl; or two R 18 groups are taken together with the N atom to which they are attached to form a N- containing heterocycle; each R 19 is independently selected from Ci-Ce alkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C2-C6 heterocycloalkyl, phenyl, benzyl, 5-membered heteroaryl, and 6-membered heteroaryl; n is 1, 2, or 3; and p is 0, 1, 2, or 3; provided the compound is not a compound listed in Table 2.

[0006] In some embodiments, the compound has a structure of Formula (II):

Formula (II) or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

L is a bond, -O-, -S-, or -N(R 5 )-, wherein:

R 5 is hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, or monocyclic heterocycle; Ring C is selected from unsubstituted or substituted carbocycle and unsubstituted or substituted heterocycle, wherein if Ring C is substituted carbocycle or substituted heterocycle, then Ring C is substituted with s instances of R c , further wherein: s is 0, 1, 2, 3, 4, or 5; and each R c is independently selected from selected from -N(R')(R 2 ), -C(=0)-W-R 4 , and R 12 , wherein:

R 1 is hydrogen, Ci-Ce alkyl, or Ci-Ce fluoroalkyl;

R 2 is a Ring B that is an unsubstituted or substituted heterocycle or unsubstituted or substituted carbocycle, wherein if Ring B is substituted then Ring B is substituted with q instances of R 13 , further wherein: each R 13 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , -C(=O)R 16 , - CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , -NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , - SO2R 17 , -SO 2 N(R 16 )2, or -P(=O)(R 16 ) 2 ; or two R 13 groups on adjacent atoms of R 2 are taken together with the intervening atoms to which they are attached to form an unsubstituted or substituted 5- or 6-membered monocyclic carbocycle or an unsubstituted or substituted 5- or 6-membered monocyclic heterocycle; and q is 0, 1, 2, 3, or 4; or R 2 is -C(=O)R 14 , -C(=O)NR 14 R 15 , or -C(=O)OR 14 ;

R 14 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted monocyclic carbocycle, unsubstituted or substituted bicyclic carbocycle, unsubstituted or substituted monocyclic heterocycle, or unsubstituted or substituted bicyclic heterocycle;

R 15 is hydrogen, Ci-Ce alkyl, or Ci-Ce fluoroalkyl; or R 14 and R 15 are taken together with the intervening atoms to which they are attached to form an unsubstituted or substituted 4- to 6-membered monocyclic heterocycle; or R 1 and R 15 are taken together with the intervening atoms to which they are attached to form an unsubstituted or substituted 5- or 6-membered monocyclic heterocycle;

W is -NR 3 - or -O-;

R 3 is hydrogen, Ci-Ce alkyl, Ci-Ce deuteroalkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, or monocyclic heterocycle;

R 4 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce alkoxy, unsubstituted or substituted C3-C6 cycloalkyl, or unsubstituted or substituted monocyclic heterocycle; or R 3 and R 4 are taken together with the N atom to which they are attached to form a substituted or unsubstituted N-containing heterocycle; or R 3 and one R 12 are taken together with the intervening atoms to which they are attached to form a substituted or unsubstituted 5- or 6-membered heterocycle; and each R 12 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , -C(=O)R 16 , -CO2R 16 , - C(=O)N(R 16 ) 2 , -N(R 16 )2, -NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , -SO2R 17 , or - SO 2 N(R 16 ) 2 ; or two R 12 attached to the same aliphatic carbon atom are taken together to form oxo; or two R 12 attached to two adjacent carbon atoms are taken together to form a group selected from unsubstituted or substituted carbocycle and unsubstituted or substituted heterocycle.

[0007] In some embodiments, the compound has a structure of Formula (III- A):

Formula (III- A) or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

A 1 and A 2 are each independently N or C; and

A 3 is S, O, N, NR 8 , CR 8 , or C=O; and

A 4 and A 5 are each independently S, O, N, NR 8 , or CR 8 ; wherein at least one of A 1 and A 2 is C, or at least one of A 3 , A 4 , and A 5 is CR 8 .

[0008] In some embodiments, the compound has a structure of Formula (III-B):

Formula (III-B), or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein: A 6 is N or CR 8 .

[0009] In other aspects, the present disclosure provides pharmaceutical compositions comprising a compound of the present disclosure, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, and a pharmaceutically acceptable excipient. [0010] In still other aspects, the present disclosure provides methods of treating a disease or condition in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, or a pharmaceutical composition of the present disclosure. In some embodiments, the disease or condition is a TYK2 -mediated disease or condition.

[0011] In still other aspects, the present disclosure provides 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, tautomers, pharmaceutically acceptable N- oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, is used for modulating TYK2, or for the treatment, prevention or amelioration of one or more symptoms of a disease or condition that would benefit from modulating TYK2, are provided.

[0012] 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

[0013] TYK2 activation has been linked to many diseases and disorders, including inflammatory diseases and disorders, autoimmune diseases and disorders, respiratory diseases and disorders, and cancer.

[0014] In particular, IL-23 activation of TYK2 is associated with inflammatory diseases such as inflammatory bowel disease (IBD), Crohn’s disease, celiac disease, and ulcerative colitis. As the downstream effector of IL-23, TYK2 also plays a role in psoriasis, ankylosing spondylitis, and Behcet’s disease. TYK2 has also been associated with diseases and conditions of the skin, such as psoriasis, vitiligo, atopic dermatitis, scleroderma; or diseases and conditions of the eye, such as Sjogren’s syndrome, uveitis, and dry eye.

[0015] TYK2 is associated with respiratory diseases and conditions such as asthma, chronic obstructive pulmonary disease (COPD), lung cancer, and cystic fibrosis. Goblet cell hyperplasia (GCH) and mucous hypersecretion is mediated by IL-13-induced activation of the TYK2/STAT6 pathway. [0016] TYK2 is also associated with autoimmune diseases and conditions, such as multiple sclerosis (MS), lupus, and systemic lupus erythematosus (SLE). Loss of function mutation in TYK2, leads to decreased demyelination and increased remyelination of neurons, further suggesting a role for TYK2 inhibitors in the treatment of MS and other CNS demyelination disorders. Various type I IFN signaling pathways dependent on TYK2 signaling have implicated TYK2 in SLE and other autoimmune diseases and conditions.

[0017] TYK2 is associated with arthritis, including psoriatic arthritis and rheumatoid arthritis. Decreased TYK2 activity leads to protection of joints from collagen antibody-induced arthritis, a model of human rheumatoid arthritis.

[0018] TYK2 has also been shown to play an important role in maintaining tumor surveillance and TYK2 knockout mice showed compromised cytotoxic T cell response, and accelerated tumor development. These effects are largely due to the efficient suppression of natural killer (NK) and cytotoxic T lymphocytes, suggesting that TYK2 inhibitors are highly suitable for the treatment of autoimmune disorders or transplant rejection. Although other JAK family members such as JAK3 have similar roles in the immune system, TYK2 is a superior target because of its involvement in fewer and more closely related signaling pathways, leading to fewer off-target effects. However, studies in T-cell acute lymphoblastic leukemia (T-ALL) indicate that T-ALL is highly dependent on IL-10 via TYK2/STAT1 signaling to maintain cancer cell survival through upregulation of anti-apoptotic protein BCL2. Knockdown of TYK2, but not other JAK family members, reduced cell growth. Thus, selective inhibition of TYK2 has been suggested as a suitable target for patients with IL-10 and/or BCL2-addicted tumors, such as 70% of adult T- cell leukemia cases.

[0019] TYK2-mediated STAT3 signaling has also been shown to mediate neuronal cell death caused by amyloid-P (AP) peptide. Decreased TYK2 phosphorylation of STAT3 following Ap administration lead to decreased neuronal cell death, and increased phosphorylation of STAT3 has been observed in postmortem brains of Alzheimer’s patients.

[0020] Inhibition of JAK-STAT signaling pathways is also implicated in hair growth, and the reversal of the hair loss associated with alopecia areata.

[0021] There is a continuing need to provide novel inhibitors having more effective or advantageous pharmaceutically relevant properties. In some embodiments, the TYK2 inhibitors show selectivity over JAK1, JAK2, and/or JAK3. In some embodiments, compounds with this selectivity (particularly over JAK2) deliver a pharmacological response that favorably treats one or more of the diseases or conditions described herein without the side-effects associated with the inhibition of JAK2. For example, compounds with increased activity or increased selectivity over other JAK kinases (especially JAK2). The present disclosure relates to compounds that bind to the pseudokinase domain (JH2) of the non-receptor tyrosine-protein kinase 2 (TYK2) and inhibit certain cytokine signaling, in particular IL-23 and IFNa signaling, to pharmaceutical compositions comprising the compounds, to methods of using the compounds to treat certain autoimmune diseases, multiple sclerosis (MS), lupus, and systemic lupus erythematosus (SLE), and other CNS demyelination disorders, and to intermediates and processes useful in the synthesis of the compounds.

[0022] In some embodiments, the TYK2 inhibitors described herein are used in the treatment of a disease or condition in a mammal, such as a human.

Compounds of the Present Disclosure

[0023] Compounds described herein, including pharmaceutically acceptable salts, tautomers, and solvates thereof, are inhibitors of TYK2. In some embodiments, compounds described herein are selective for TYK2 over other JAKs. In some embodiments, compounds described herein bind selectively/specifically to the pseudokinase domain (JH2) of TYK2. In some embodiments, a compound described herein binds to an allosteric site of TYK2. In additional embodiments, a compound described herein may be useful in the treatment of TYK2 mediated diseases or disorders.

[0024] In one aspect, described herein is a compound of Formula (I):

Formula (I), or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

Y is a ligand that binds the hinge-binding region in the allosteric binding site of TYK2;

L is a linker that covalently connects Y and the rest of the molecule;

Ring A is an unsubstituted or substituted carbocyclic ring wherein A 1 and A 2 are both C, or an unsubstituted or substituted 5- or 6-membered heterocyclic ring wherein A 1 and A 2 are independently N or C, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 ; each R 8 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , - C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , -NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , - SO2R 17 , or -SO2N(R 16 )2; wherein if R 8 is attached to a nitrogen atom, then R 8 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci- C , deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 ) 2 , -S(=O)R 17 , -SO2R 17 , or -SO2N(R 16 )2; or two R 8 attached to the same carbon atom are taken together to form =0, =S, or =NH;

Z is -NR 10 -, -O-, -S-, -S(=O)-, or -SO2-;

R 10 is hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, or monocyclic heterocycle;

X 1 , X 2 , and X 3 are each independently CR 11 or N; each R 11 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , -C(=0)R 16 , -CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , - NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , -SO2R 17 , or -SO 2 N(R 16 )2; each R 6 and R 7 is independently hydrogen, deuterium, halogen, Ci-Ce alkyl, Ci-Ce deuteroalkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, monocyclic heterocycle, -CN, -OH, -OR 17 , -C(=0)R 16 , - CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 )2, -NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , -SO2R 17 , or - SO 2 N(R 16 ) 2 ; or one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C=O or a C3-C6 cycloalkane; each R 16 is independently hydrogen, substituted or unsubstituted Ci-Ce alkyl, substituted or unsubstituted Ci-Ce fluoroalkyl, substituted or unsubstituted Ci-Ce heteroalkyl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted monocyclic 3- to 8-membered heterocycloalkyl, substituted or unsubstituted phenyl, or substituted or unsubstituted monocyclic heteroaryl; or two R 16 on the same N atom are taken together with the N atom to which they are attached to form a substituted or unsubstituted N-containing heterocycle; and each R 17 is independently substituted or unsubstituted Ci-Ce alkyl, substituted or unsubstituted Ci-Ce fluoroalkyl, substituted or unsubstituted Ci-Ce heteroalkyl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted monocyclic 3- to 8-membered heterocycloalkyl, substituted or unsubstituted phenyl, or substituted or unsubstituted monocyclic heteroaryl; wherein each substituted alkyl, substituted alkenyl, substituted alkynyl, substituted fluoroalkyl, substituted deuteroalkyl, substituted alkoxy, substituted fluoroalkoxy, substituted heteroalkyl, substituted carbocycle, and substituted heterocycle is substituted with one or more R s groups independently selected from the group consisting of deuterium, halogen, Ci- C , alkyl, monocyclic carbocycle, monocyclic heterocycle, -CN, -CH2CN, -OR 18 , -CH2OR 18 , -CO2R 18 , -CH2CO2R 18 , -C(=O)N(R 18 ) 2 , -CH 2 C(=O)N(R 18 ) 2 , -N(R 18 )2, -CH 2 N(R 18 ) 2 , - NR 18 C(=O)R 18 , -CH 2 NR 18 C(=O)R 18 , -NR 18 SO 2 R 19 , -CH 2 NR 18 SO 2 R 19 , -SR 18 , -CH2SR 18 , - S(=O)R 19 , -CH 2 S(=O)R 19 , -SO2R 19 , -CH2SO2R 19 , -SO 2 N(R 18 )2, and -CH 2 SO 2 N(R 18 )2; each R 18 is independently selected from hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C2-C6 heterocycloalkyl, phenyl, benzyl, 5-membered heteroaryl and 6-membered heteroaryl; or two R 18 groups are taken together with the N atom to which they are attached to form a N- containing heterocycle; each R 19 is independently selected from Ci-Ce alkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C2-C6 heterocycloalkyl, phenyl, benzyl, 5-membered heteroaryl, and 6-membered heteroaryl; n is 1, 2, or 3; and p is 0, 1, 2, or 3; provided the compound is not a compound listed in Table 2.

[0025] In some embodiments, the compound has a structure of Formula (I):

Formula (I), or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

Y is a ligand that binds the hinge-binding region in the allosteric binding site of TYK2;

L is a linker that covalently connects Y and the rest of the molecule;

Ring A is an unsubstituted or substituted carbocyclic ring wherein A 1 and A 2 are both C, or an unsubstituted or substituted 5- or 6-membered heterocyclic ring wherein A 1 and A 2 are independently N or C, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 ; each R 8 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , - C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , -NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , - SO2R 17 , or -SO2N(R 16 )2; wherein if R 8 is attached to a nitrogen atom, then R 8 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci- Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 ) 2 , -S(=O)R 17 , -SO2R 17 , or -SO2N(R 16 )2; or two R 8 attached to the same carbon atom are taken together to form =0, =S, or =NH;

Z is -NR 10 -, -0-, -S-, -S(=0)-, or -SO2-;

R 10 is hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, or monocyclic heterocycle;

X 1 , X 2 , and X 3 are each independently CR 11 or N; each R 11 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , -C(=0)R 16 , -CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , - NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , -SO2R 17 , or -SO 2 N(R 16 )2; each R 6 and R 7 is independently hydrogen, deuterium, halogen, Ci-Ce alkyl, Ci-Ce deuteroalkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, monocyclic heterocycle, -CN, -OH, -OR 17 , -C(=0)R 16 , - CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 )2, -NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , -SO2R 17 , or - SO 2 N(R 16 ) 2 ; or one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C=0 or a C3-C6 cycloalkane; each R 16 is independently hydrogen, substituted or unsubstituted Ci-Ce alkyl, substituted or unsubstituted Ci-Ce fluoroalkyl, substituted or unsubstituted Ci-Ce heteroalkyl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted monocyclic 3- to 8-membered heterocycloalkyl, substituted or unsubstituted phenyl, or substituted or unsubstituted monocyclic heteroaryl; or two R 16 on the same N atom are taken together with the N atom to which they are attached to form a substituted or unsubstituted N-containing heterocycle; and each R 17 is independently substituted or unsubstituted Ci-Ce alkyl, substituted or unsubstituted Ci-Ce fluoroalkyl, substituted or unsubstituted Ci-Ce heteroalkyl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted monocyclic 3- to 8-membered heterocycloalkyl, substituted or unsubstituted phenyl, or substituted or unsubstituted monocyclic heteroaryl; wherein each substituted alkyl, substituted fluoroalkyl, substituted deuteroalkyl, substituted alkoxy, substituted fluoroalkoxy, substituted heteroalkyl, substituted carbocycle, and substituted heterocycle is substituted with one or more R s groups independently selected from the group consisting of deuterium, halogen, Ci-Ce alkyl, monocyclic carbocycle, monocyclic heterocycle, -CN, -CH2CN, -OR 18 , -CH2OR 18 , -CO2R 18 , -CH2CO2R 18 , - C(=O)N(R 18 ) 2 , -CH 2 C(=O)N(R 18 ) 2 , -N(R 18 ) 2 , -CH 2 N(R 18 ) 2 , -NR 18 C(=O)R 18 , - CH 2 NR 18 C(=O)R 18 , -NR 18 SO 2 R 19 , -CH 2 NR 18 SO 2 R 19 , -SR 18 , -CH2SR 18 , -S(=O)R 19 , - CH 2 S(=O)R 19 , -SO2R 19 , -CH2SO2R 19 , -SO 2 N(R 18 )2, and -CH 2 SO 2 N(R 18 )2; each R 18 is independently selected from hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C2-C6 heterocycloalkyl, phenyl, benzyl, 5-membered heteroaryl and 6-membered heteroaryl; or two R 18 groups are taken together with the N atom to which they are attached to form a N- containing heterocycle; each R 19 is independently selected from Ci-Ce alkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C2-C6 heterocycloalkyl, phenyl, benzyl, 5-membered heteroaryl, and 6-membered heteroaryl; n is 1, 2, or 3; and p is 0, 1, 2, or 3; provided the compound is not a compound listed in Table 2. [0026] In some embodiments, the compound has a structure of Formula (II):

Formula (II) or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

L is a bond, -O-, -S-, or -N(R 5 )-, wherein:

R 5 is hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, or monocyclic heterocycle; Ring C is selected from unsubstituted or substituted carbocycle and unsubstituted or substituted heterocycle, wherein if Ring C is substituted carbocycle or substituted heterocycle, then Ring C is substituted with s instances of R c , further wherein: s is 0, 1, 2, 3, 4, or 5; and each R c is independently selected from selected from -N(R 1 )(R 2 ), -C(=0)-W-R 4 , and R 12 , wherein:

R 1 is hydrogen, Ci-Ce alkyl, or Ci-Ce fluoroalkyl;

R 2 is a Ring B that is an unsubstituted or substituted heterocycle or unsubstituted or substituted carbocycle, wherein if Ring B is substituted then Ring B is substituted with q instances of R 13 , further wherein: each R 13 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , -C(=O)R 16 , - CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , -NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , - SO2R 17 , -SO 2 N(R 16 )2, or -P(=O)(R 16 ) 2 ; or two R 13 groups on adjacent atoms of R 2 are taken together with the intervening atoms to which they are attached to form an unsubstituted or substituted 5- or 6-membered monocyclic carbocycle or an unsubstituted or substituted 5- or 6-membered monocyclic heterocycle; and q is 0, 1, 2, 3, or 4; or R 2 is -C(=O)R 14 , -C(=O)NR 14 R 15 , or -C(=O)OR 14 ; R 14 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted monocyclic carbocycle, unsubstituted or substituted bicyclic carbocycle, unsubstituted or substituted monocyclic heterocycle, or unsubstituted or substituted bicyclic heterocycle;

R 15 is hydrogen, Ci-Ce alkyl, or Ci-Ce fluoroalkyl; or R 14 and R 15 are taken together with the intervening atoms to which they are attached to form an unsubstituted or substituted 4- to 6-membered monocyclic heterocycle; or R 1 and R 15 are taken together with the intervening atoms to which they are attached to form an unsubstituted or substituted 5- or 6-membered monocyclic heterocycle;

W is -NR 3 - or -O-;

R 3 is hydrogen, Ci-Ce alkyl, Ci-Ce deuteroalkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, or monocyclic heterocycle;

R 4 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce alkoxy, unsubstituted or substituted C3-C6 cycloalkyl, or unsubstituted or substituted monocyclic heterocycle; or R 3 and R 4 are taken together with the N atom to which they are attached to form a substituted or unsubstituted N-containing heterocycle; or R 3 and one R 12 are taken together with the intervening atoms to which they are attached to form a substituted or unsubstituted 5- or 6-membered heterocycle; and each R 12 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , -C(=O)R 16 , -CO2R 16 , - C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , -NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , -SO2R 17 , or - SO 2 N(R 16 ) 2 ; or two R 12 attached to the same aliphatic carbon atom are taken together to form oxo; or two R 12 attached to two adjacent carbon atoms are taken together to form a group selected from unsubstituted or substituted carbocycle and unsubstituted or substituted heterocycle. [0027] In some embodiments, the compound has a structure of Formula (III- A):

Formula (III- A) or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein:

A 1 and A 2 are each independently N or C; and

A 3 is S, O, N, NR 8 , CR 8 , or C=O; and

A 4 and A 5 are each independently S, O, N, NR 8 , or CR 8 ; wherein at least one of A 1 and A 2 is C, or at least one of A 3 , A 4 , and A 5 is CR 8 .

[0028] In some embodiments, the compound has a structure of Formula (III-B):

Formula (III-B), or a pharmaceutically acceptable salt, tautomer, or solvate thereof, wherein: A 6 is N or CR 8 .

[0029] In some embodiments, Ring C is selected from: unsubstituted or substituted carbocycle, unsubstituted or substituted monocyclic heterocycloalkyl, unsubstituted or substituted 5-membered heteroaryl, and unsubstituted or substituted bicyclic heterocycle; and , wherein:

X 4 , X 5 , and X 6 are each independently CH, CR c , or N; and t is 0, 1, or 2. [0030] In some embodiments, when X 4 is CH, X 5 is N, and X 6 is CH, then t is 1 or 2. In some wherein:

R D is -N(R 1 )(R 2 ), -C(=O)-W-R 4 , or R 20 , wherein:

R 20 is chloro, bromo, iodo, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , -C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , - NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , -SO2R 17 , or -SO 2 N(R 16 )2.

[0031] In some embodiments, when Ring , then R 4 is hydrogen,

Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce alkoxy, unsubstituted or substituted C3-C6 cycloalkyl, or unsubstituted or substituted monocyclic heterocycle.

[0033] In some embodiments:

X 4 is CH; X 5 is N; and X 6 is CH; or X 4 is CH; X 5 is N; and X 6 is CR c ; or X 4 is CH; X 5 is CR c ; and X 6 is N; or X 4 is N; X 5 is N; and X 6 is CH; or X 4 is N; X 5 is N; and X 6 is N; or X 4 is CR c ; X 5 is N; and X 6 is CR c ; or X 4 is CR c ; X 5 is CR c ; and X 6 is CR c ; or X 4 is CH; X 5 is CH; and X 6 is CH; or X 4 is N; X 5 is N; and X 6 is CR c

[0034] In some embodiments:

X 4 is CH; X 5 is N; and X 6 is CH;

X 4 is CH; X 5 is N; and X 6 is CR c ;

X 4 is CH; X 5 is CR c ; and X 6 is N;

X 4 is N; X 5 is N; and X 6 is CH;

X 4 is N; X 5 is N; and X 6 is N; or X 4 is CH; X 5 is N; and X 6 is N.

[0035] In some embodiments:

X 4 is CH; X 5 is N; and X 6 is CH;

X 4 is CH; X 5 is N; and X 6 is CR c ;

X 4 is CH; X 5 is CR c ; and X 6 is N;

X 4 is N; X 5 is N; and X 6 is CH; or X 4 is N; X 5 is N; and X 6 is N.

[0036] In some embodiments:

X 4 is CH; X 5 is N; and X 6 is CR c ;

X 4 is CH; X 5 is CR c ; and X 6 is N;

X 4 is CH; X 5 is N; and X 6 is N.

[0037] In some embodiments:

X 4 is CH; X 5 is N; and X 6 is CR c ; or

X 4 is CH; X 5 is CR c ; and X 6 is N.

[0038] In some embodiments, X 4 is CH; X 5 is N; and X 6 is CH. In some embodiments, X 4 is CH; X 5 is N; and X 6 is CR c . In some embodiments, X 4 is CH; X 5 is CR c ; and X 6 is N. In some embodiments, X 4 is N; X 5 is N; and X 6 is CH. In some embodiments, X 4 is N; X 5 is N; and X 6 is N. In some embodiments, X 4 is CR c ; X 5 is N; and X 6 is CR c . In some embodiments, X 4 is CR c ; X 5 is CR c ; and X 6 is CR c . In some embodiments, X 4 is CH; X 5 is CH; and X 6 is CH. In some embodiments, X 4 is N; X 5 is N; and X 6 is CR c .

[0039] In some embodiments, Ring C is selected from:

[0040] In some embodiments, Ring C is selected from:

[0041] In some embodiments, Ring C is selected from:

[0042] In some embodiments, Ring C is selected from:

[0043] In some embodiments, Ring C is unsubstituted or substituted bicyclic heterocycle. In some embodiments, Ring C is substituted bicyclic heterocycle. In some embodiments, Ring C is selected from:

[0044] In some embodiments, Ring C is selected from:

[0045] In some embodiments, each R c is independently selected from -N(R 1 )(R 2 ), -C(=O)-W- R 4 , and R 12 . In some embodiments, at least one R c is -N(R 1 )(R 2 ). In some embodiments, at least one R c is -C(=O)-W-R 4 . In some embodiments, at least one R c is R 12 .

[0046] In some embodiments, Ring C is selected from:

[0048] In some embodiments, Ring C is selected from:

[0049] In some embodiments, R 1 is hydrogen or C1-C4 alkyl. In some embodiments, R 1 is hydrogen. In some embodiments, R 1 is methyl. In some embodiments, W is -NR 3 -.

[0050] In some embodiments, R 3 is hydrogen, C1-C4 alkyl, or C1-C4 deuteroalkyl. In some embodiments, R 3 is hydrogen or C1-C4 alkyl. In some embodiments, R 3 is hydrogen or C1-C4 deuteroalkyl. In some embodiments, R 3 is C1-C4 alkyl or C1-C4 deuteroalkyl. In some embodiments, R 3 is hydrogen, methyl, or trideuteromethyl. In some embodiments, R 3 is hydrogen. In some embodiments, R 3 is C1-C4 alkyl, such as methyl. In some embodiments, R 3 is C1-C4 deuteroalkyl, such as trideuteromethyl.

[0051] In some embodiments, R 4 is hydrogen, Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce alkoxy, unsubstituted or substituted C3-C6 cycloalkyl, or unsubstituted or substituted monocyclic heterocycle. In some embodiments, R 4 is hydrogen, Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce alkoxy, or unsubstituted or substituted C3-C6 cycloalkyl. In some embodiments, R 4 is hydrogen, Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce alkoxy, or unsubstituted or substituted monocyclic heterocycle. In some embodiments, R 4 is hydrogen, Ci-Ce alkyl, unsubstituted or substituted Ci- Ce fluoroalkyl, or unsubstituted or substituted Ci-Ce alkoxy. In some embodiments, R 4 is hydrogen, C1-C4 alkyl, unsubstituted or substituted C1-C4 fluoroalkyl, or unsubstituted or substituted C1-C4 alkoxy. In some embodiments, R 4 is hydrogen, Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl. In some embodiments, R 4 is hydrogen, Ci-Ce alkyl, or unsubstituted or substituted Ci-Ce alkoxy. In some embodiments, R 4 is hydrogen, C1-C4 alkyl, unsubstituted or substituted C1-C4 fluoroalkyl. In some embodiments, R 4 is hydrogen, C1-C4 alkyl, or unsubstituted or substituted C1-C4 alkoxy. In some embodiments, R 4 is hydrogen or Ci- C , alkyl. In some embodiments, R 4 is hydrogen or C1-C4 alkyl. In some embodiments, R 4 is hydrogen, methyl, ethyl, propyl, isopropyl, or butyl. In some embodiments, R 4 is hydrogen. In some embodiments, R 4 is methyl.

[0052] In some embodiments, R 4 is hydrogen, unsubstituted or substituted C1-C4 alkyl, unsubstituted or substituted C1-C4 deuteroalkyl, unsubstituted or substituted C1-C4 alkoxy, or unsubstituted or substituted C3-C6 cycloalkyl. In some embodiments, R 4 is hydrogen, C1-C4 alkyl, C1-C4 deuteroalkyl, or C1-C4 alkoxy. In some embodiments, R 4 is hydrogen, methyl, trideuteromethyl, methoxy, or ethoxy. In some embodiments, R 4 is substituted C3-C6 cycloalkyl. In some embodiments, R 4 is substituted cyclopropyl or cyclobutyl. In some embodiments, R 4 is 2-methoxy cyclobutyl or 2-fluorocyclopropyl.

[0053] In some embodiments, each R 12 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, or -N(R 16 )2; or two R 12 attached to the same aliphatic carbon atom are taken together to form oxo; or two R 12 attached to two adjacent carbon atoms are taken together to form an unsubstituted or substituted heterocycle. In some embodiments, each R 12 is independently substituted Ci-Ce alkyl, substituted carbocycle, substituted heterocycle; or two R 12 attached to two adjacent carbon atoms are taken together to form a substituted heterocycle. In some embodiments, each R 12 is independently substituted with 1, 2, or 3 R s groups independently selected from deuterium, halogen, monocyclic carbocycle, and -CH2OH. In some embodiments, each R 12 is independently substituted with 1, 2, or 3 R s groups independently selected from deuterium, fluoro, cyclopropyl, and -CH2OH. In some embodiments, each R 12 is independently fluoro, methyl, ethyl, trideuteromethyl, difluoromethyl, phenyl, 2,6-difluorophenyl, 2-hydroxymethyl-l,3,4-oxadiazol-5-yl, amino, or methylamino; or two R 12 attached to the same aliphatic carbon atom are taken together to form oxo; or two R 12 attached to two adjacent carbon atoms are taken together to form 2-cyclopropyl-4,5-dihydro-lH- imidazole. In some embodiments, each R 12 is independently fluoro, methyl, ethyl, trideuteromethyl, difluoromethyl, phenyl, 2,6-difluorophenyl, 2 -hydroxymethyl- 1,3, 4-oxadiazol- 5-yl, amino, or methylamino. In some embodiments, each R 12 is independently fluoro, methyl, ethyl, trideuteromethyl, or difluoromethyl. In some embodiments, each R 12 is independently fluoro, methyl, or trideuteromethyl. In some embodiments, each R 12 is independently methyl, 2- hydroxymethyl-l,3,4-oxadiazol-5-yl, or amino; or two R 12 attached to the same aliphatic carbon atom are taken together to form oxo. [0054] In some embodiments, each R 12 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, or -N(R 16 )2; or two R 12 attached to the same aliphatic carbon atom are taken together to form oxo; or two R 12 attached to two adjacent carbon atoms are taken together to form an unsubstituted or substituted heterocycle.

[0055] In some embodiments, each R 12 is independently substituted Ci-Ce alkyl, substituted carbocycle, or substituted heterocycle; or two R 12 attached to two adjacent carbon atoms are taken together to form a substituted heterocycle.

[0056] In some embodiments, each R 12 is independently substituted with 1, 2, or 3 R s groups independently selected from deuterium, halogen, monocyclic carbocycle, and -CH2OH. In some embodiments, each R 12 is independently substituted with 1, 2, or 3 R s groups independently selected from deuterium, fluoro, cyclopropyl, and -CH2OH.

[0057] In some embodiments, each R 12 is independently fluoro, methyl, ethyl, trideuteromethyl, difluoromethyl, phenyl, 2,6-difluorophenyl, 2-hydroxymethyl-l,3,4-oxadiazol-5-yl, amino, or methylamino; or two R 12 attached to the same aliphatic carbon atom are taken together to form oxo; or two R 12 attached to two adjacent carbon atoms are taken together to form 2-cyclopropyl-4,5- dihydro-lH-imidazole.

[0058] In some embodiments, each R 12 is independently methyl, 2-hydroxymethyl- 1,3,4- oxadiazol-5-yl, or amino; or two R 12 attached to the same aliphatic carbon atom are taken together to form oxo.

[0059] In some embodiments, R 2 is a Ring B that is an unsubstituted or substituted heterocycle or unsubstituted or substituted carbocycle, wherein if Ring B is substituted then Ring B is substituted with q instances of R 13 . In some embodiments, R 2 is a Ring B that is an unsubstituted or substituted monocyclic heterocycle, wherein if Ring B is substituted then Ring B is substituted with q instances of R 13 . In some embodiments, R 2 is a Ring B that is an unsubstituted or substituted monocyclic 6-membered heteroaryl, wherein if Ring B is substituted then Ring B is substituted with q instances of R 13 . In some embodiments, R 2 is a Ring B that is an unsubstituted or substituted pyridinyl or unsubstituted or substituted pyrimidinyl, wherein if Ring B is substituted then Ring B is substituted with q instances of R 13 . [0060] In some embodiments, Ring

4. In some embodiments, q is 0, 1, or 2. In some embodiments, q is 1 or 2. In some embodiments, q is 1. In some embodiments, q is 2.

[0061] In some embodiments, each R 13 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , -C(=O)R 16 , -CO2R 16 , - C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , -NR 16 C(=O)R 17 , -SO2R 17 , -SO 2 N(R 16 )2, or -P(=O)(R 16 ) 2 . In some embodiments, each R 13 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, -CN. In some embodiments, each R 13 is independently -F, -CH3, -CH2N(CH3)2, or -CN. In some embodiments, each R 13 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , -C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 )2, -N(R 16 ) 2 , -NR 16 C(=O)R 17 , -SO2R 17 , -SO 2 N(R 16 )2, or -P(=O)(R 16 )2. In some embodiments, each R 13 is independently halogen, unsubstituted or substituted Ci-Ce alkyl, or -CN. In some embodiments, each R 13 is independently -F, -CH3, -CH2N(CH3)2, or -CN. In some embodiments, at least one R 13 is -F. In some embodiments, at least one R 13 is -CH3. In some embodiments, at least one R 13 is -CH2N(CH3)2. In some embodiments, at least one R 13 is -CN.

[0062] In some embodiments, Ring B is 5-fluoropyridin-2-yl, 6-cyanopyridin-2-yl, 4- aminopyrimidin-6-yl, 5-((dimethylamino)methyl)pyridin-2-yl, or 2,6-dimethylpyrimidin-4-yl. In some embodiments, Ring B is 2,6-dimethylpyrimidin-6-yl. In some embodiments, Ring B is 5- fluoropyridin-2-yl, 6-cyanopyridin-2-yl, 4-aminopyrimidin-6-yl, 5- ((dimethylamino)methyl)pyridin-2-yl, or 2,6-dimethylpyrimidin-4-yl. In some embodiments, Ring B is 5-fluoropyridin-2-yl. In some embodiments, Ring B is 6-cyanopyridin-2-yl. In some embodiments, Ring B is 4-aminopyrimidin-6-yl. In some embodiments, Ring B is 5- ((dimethylamino)methyl)pyridin-2-yl. In some embodiments, Ring B is 2,6-dimethylpyrimidin- 4-yl. In some embodiments, Ring B is 2,6-dimethylpyrimidin-4-yl.

[0063] In some embodiments, R 2 is -C(=O)R 14 , -C(=O)NR 14 R 15 , or -C(=O)OR 14 . In some embodiments, R 2 is -C(=O)R 14 . In some embodiments, R 2 is -C(=O)R 14 , -C(=O)NR 14 R 15 , or - C(=O)OR 14 . In some embodiments, R 2 is -C(=O)R 14 or -C(=O)NR 14 R 15 . In some embodiments, R 2 is -C(=O)R 14 or -C(=O)OR 14 . In some embodiments, R 2 is -C(=O)NR 14 R 15 , or -C(=O)OR 14 . In some embodiments, R 2 is -C(=O)R 14 . In some embodiments, R 2 is -C(=O)NR 14 R 15 . In some embodiments, R 2 is -C(=O)OR 14 . In some embodiments, R 14 is unsubstituted or substituted Ci- Ce alkyl, unsubstituted or substituted C3-C4 cycloalkyl, or unsubstituted or substituted 4- membered heterocycloalkyl; wherein the substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, or substituted heterocycloalkyl is substituted with one or more R s groups independently selected from the group consisting of deuterium, halogen, -CN, -NH 2 , -OH, - NH(CH 3 ), -N(CH 3 ) 2 , -CH 3 , -CH 2 CH 3 , -CHF 2 , -CF 3 , -OCH 3 , -OCHF 2 , and -OCF 3 . In some embodiments, R 14 is unsubstituted Ci-Ce alkyl unsubstituted C -C4 cycloalkyl, or unsubstituted 4-membered heterocycloalkyl. In some embodiments, R 14 is unsubstituted or substituted C 3 -C4 cycloalkyl. In some embodiments, Ring B is cyclopropyl, cyclobutyl, oxetanyl, or azetidinyl. In some embodiments, Ring B is cyclopropyl or cyclobutyl. In some embodiments, R 14 is cyclopropyl. In some embodiments, R 14 is unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C -C4 cycloalkyl, or unsubstituted or substituted 4-membered heterocycloalkyl; wherein the substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, or substituted heterocycloalkyl is substituted with one or more R s groups independently selected from the group consisting of deuterium, halogen, -CN, -NH 2 , -OH, -NH(CH 3 ), -N(CH 3 ) 2 , -CH 3 , - CH 2 CH 3 , -CHF 2 , -CF 3 , -OCH 3 , -OCHF 2 , and -OCF 3 . In some embodiments, R 14 is unsubstituted or substituted C 3 -C4 cycloalkyl. In some embodiments, R 14 is cyclopropyl. In some embodiments, R 15 is hydrogen, Ci-Ce alkyl, or Ci-Ce fluoroalkyl. In some embodiments, R 15 is hydrogen, C1-C4 alkyl, or C1-C4 fluoroalkyl. In some embodiments, R 15 is hydrogen, methyl, ethyl, propyl, isopropyl, butyl, or trifluoromethyl. In some embodiments, R 15 is hydrogen or methyl. In some embodiments, R 15 is hydrogen. In some embodiments, R 15 is methyl.

[0064] In some embodiments, Ring C is selected from:

[0066] In some embodiments, Ring C is selected from: [0069] In some embodiments, Ring C is selected from: [0071] In some embodiments, Ring C is selected from:

[0073] In some embodiments, Ring C is selected from: [0074] In some embodiments, L is a bond, -O-, -S-, or -N(R 5 )-. In some embodiments, L is -O- , -S-, or -N(R 5 )-. In some embodiments, L is -O- or -N(R 5 )-. In some embodiments, L is -O-. In some embodiments, L is a bond or -N(R 5 )-. In some embodiments, L is a bond. In some embodiments, L is -N(R 5 )-. In some embodiments, R 5 is hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, or monocyclic heterocycle. In some embodiments, R 5 is hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, or C3-C6 cycloalkyl. In some embodiments, R 5 is hydrogen, C1-C4 alkyl, C1-C4 fluoroalkyl, or C3-C6 cycloalkyl. In some embodiments, R 5 is hydrogen, C1-C4 alkyl, or C1-C4 fluoroalkyl. In some embodiments, R 5 is hydrogen or C1-C4 alkyl. In some embodiments, R 5 is hydrogen or methyl. In some embodiments, R 5 is hydrogen. In some embodiments, R 5 is methyl.

[0075] In some embodiments, Ring A is an unsubstituted or substituted carbocyclic ring wherein A 1 and A 2 are both C, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In further embodiments, Ring A is an unsubstituted or substituted phenyl, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 .

[0076] In some embodiments, Ring A is an unsubstituted or substituted 5- or 6-membered heterocycloalkyl ring wherein A 1 and A 2 are independently N or C, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted 5-membered heterocycloalkyl ring wherein A 1 and A 2 are independently N or C, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted 6-membered heterocycloalkyl ring wherein A 1 and A 2 are independently N or C, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 .

[0077] In some embodiments, Ring A is an unsubstituted or substituted triazolone, unsubstituted or substituted pyridone, unsubstituted or substituted imidazolidinone, or unsubstituted or substituted oxazolidinone, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted triazolone or unsubstituted or substituted pyridone, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted triazolone, wherein if Ring A is substituted then Ring A is substituted with 1 R 8 .

[0079] In some embodiments, Ring A is an unsubstituted or substituted 5- or 6-membered heteroaryl ring wherein A 1 and A 2 are independently N or C, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 .

[0080] In some embodiments, Ring A is an unsubstituted or substituted 5-membered heteroaryl ring wherein A 1 and A 2 are independently N or C, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted pyrrole, unsubstituted or substituted furan, unsubstituted or substituted thiophene, unsubstituted or substituted pyrazole, unsubstituted or substituted imidazole, unsubstituted or substituted oxazole, unsubstituted or substituted isoxazole, unsubstituted or substituted thiazole, unsubstituted or substituted isothiazole, unsubstituted or substituted triazole, unsubstituted or substituted oxadiazole, unsubstituted or substituted thiadiazole, unsubstituted or substituted tetrazole, or unsubstituted or substituted triazolone, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted pyrazole, unsubstituted or substituted imidazole, unsubstituted or substituted oxazole, unsubstituted or substituted triazole, unsubstituted or substituted tetrazole, or unsubstituted or substituted triazolone, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted pyrazole or unsubstituted or substituted triazole, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 .

[0081] In some embodiments, Ring A is an unsubstituted or substituted pyrazole, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted imidazole, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted oxazole, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted triazole, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted tetrazole, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted triazol one, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . , some embodiments, Ring A is

[0083] In some embodiments, are each independently N or C; and A 3 , A 4 , and A 5 are each independently S, O, N, NR 8 , or CR 8 ; wherein at least one of A 1 and A 2 is not C, or at least one of A 3 , A 4 , and A 5 is not -CR 8 -.

[0084] In further embodiments:

A 1 is C; A 2 is C; A 3 is N; A 4 is NR 8 , O, or S; and A 5 is CR 8 ; or A 1 is C; A 2 is C; A 3 is NR 8 , O, or S; A 4 is N; and A 5 is CR 8 ; or A 1 is N; A 2 is C; A 3 is N; A 4 is N; and A 5 is CR 8 ; or A 1 is C; A 2 is N; A 3 is N; A 4 is CR 8 ; and A 5 is N; or A 1 is C; A 2 is N; A 3 is N; A 4 is N; and A 5 is N; or A 1 is N; A 2 is C; A 3 is N; A 4 is N; and A 5 is N; or A 1 is C; A 2 is N; A 3 is N; A 4 is CR 8 ; and A 5 is CR 8 ; or A 1 is C; A 2 is N; A 3 is N; A 4 is N; and A 5 is CR 8 ; or A 1 is C; A 2 is N; A 3 is CR 8 ; A 4 is N; and A 5 is CR 8 ; or A 1 is C; A 2 is N; A 3 is CR 8 ; A 4 is CR 8 ; and A 5 is N; or A 1 is N; A 2 is C; A 3 is N; A 4 is CR 8 ; and A 5 is CR 8 ; or A 1 is C; A 2 is C; A 3 is N; A 4 is CR 8 ; and A 5 is NR 8 , O, or S; or A 1 is C; A 2 is C; A 3 is NR 8 , O, or S; A 4 is CR 8 ; and A 5 is N; or A 1 is C; A 2 is N; A 3 is N or C=O; A 4 is NR 8 ; and A 5 is N; or A 1 is C; A 2 is C; A 3 is N or C=O; A 4 is NR 8 ; and A 5 is N.

[0085] In still further embodiments:

A 1 is C; A 2 is C; A 3 is N; A 4 is NR 8 , O, or S; and A 5 is CR 8 ; or A 1 is C; A 2 is N; A 3 is N; A 4 is CR 8 ; and A 5 is N; or A 1 is C; A 2 is N; A 3 is N; A 4 is CR 8 ; and A 5 is CR 8 ; or A 1 is C; A 2 is C; A 3 is N; A 4 is CR 8 ; and A 5 is NR 8 , O, or S; or A 1 is C; A 2 is N; A 3 is N or C=O; A 4 is NR 8 ; and A 5 is N; or A 1 is C; A 2 is C; A 3 is N or C=O; A 4 is NR 8 ; and A 5 is N.

[0086] In some embodiments, Ring A is an unsubstituted or substituted 6-membered heteroaryl ring wherein A 1 and A 2 are independently N or C, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted pyridine, unsubstituted or substituted pyridazine, unsubstituted or substituted pyrimidine, unsubstituted or substituted pyrazine, unsubstituted or substituted triazine, or unsubstituted or substituted pyridone, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted pyridine, unsubstituted or substituted pyrimidine, unsubstituted or substituted pyrazine, or unsubstituted or substituted pyridone, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 .

[0087] In some embodiments, Ring A is an unsubstituted or substituted pyridine, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted pyridazine, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted pyrimidine, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted pyrazine, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted triazine, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . In some embodiments, Ring A is an unsubstituted or substituted pyridone, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 .

[0088] In some embodiments, Ring some embodiments,

Ring some embodiments, Ring some embodiments, Ring

[0089] In some embodiments, wherein A 6 , A 7 , A 8 and A 9 are each independently CR 8 or N; wherein at least one of A 6 , A 7 , A 8 and A 9 is N.

[0090] In some embodiments, A 6 is N. In some embodiments, A 6 is CR 8 . In some embodiments, A 7 is CR 8 . In some embodiments, A 7 is N. In some embodiments, A 8 is CR 8 . In some embodiments, A 8 is N. In some embodiments, A 9 is CR 8 . In some embodiments, A 9 is N. In some embodiments, A 6 is N; A 7 is CR 8 or N; A 8 is CR 8 or N; and A 9 is CR 8 or N.

[0091] In some embodiments, wherein A 6 is N or CR 8 .

[0092] In some embodiments, Ring A is an unsubstituted or substituted pyrazole or an unsubstituted or substituted triazole, wherein if Ring A is substituted then Ring A is substituted with p instances of R 8 . [0093] In some embodiments A 1 and A 2 are each N. In some embodiments, A 1 and A 2 are each C. In some embodiments A 1 is C and A 2 is N. In some embodiments A 1 is N and A 2 is C. In some embodiments A 3 is S. In some embodiments A 3 is O. In some embodiments A 3 is CR 8 . In some embodiments A 3 is N. In some embodiments A 3 is NR 8 . In some embodiments A 3 is C=O.

In some embodiments A 4 and A 5 are each independently S, O, N, NR 8 , or CR 8 . In some embodiments A 4 and A 5 are each N or NR 8 . In some embodiments, A 4 is N and A 5 is NR 8 . In some embodiments A 4 is NR 8 and A 5 is N. In some embodiments, A 4 is CR 8 and A 5 is S. In some embodiments A 4 is CR 8 and A 5 is O. In some embodiments A 4 is N and A 5 is CR 8 . In some embodiments A 4 is O and A 5 is CR 8 . In some embodiments A 4 is O and A 5 is NR 8 .

[0094] In some embodiments, A 1 is C; A 2 is N or C; A 3 is N or C=O; A 4 is NR 8 or CR 8 ; and A 5 is N, S, or CR 8 .

[0095] In some embodiments:

A 1 is C; A 2 is C; A 3 is N; A 4 is NR 8 , O, or S; and A 5 is CR 8 ; or A 1 is C; A 2 is C; A 3 is NR 8 , O, or S; A 4 is N; and A 5 is CR 8 ; or A 1 is N; A 2 is C; A 3 is N; A 4 is N; and A 5 is CR 8 ; or A 1 is C; A 2 is N; A 3 is N; A 4 is CR 8 ; and A 5 is N; or A 1 is C; A 2 is N; A 3 is N; A 4 is N; and A 5 is N; or A 1 is N; A 2 is C; A 3 is N; A 4 is N; and A 5 is N; or A 1 is C; A 2 is N; A 3 is N; A 4 is CR 8 ; and A 5 is CR 8 ; or A 1 is C; A 2 is N; A 3 is N; A 4 is N; and A 5 is CR 8 ; or A 1 is C; A 2 is N; A 3 is CR 8 ; A 4 is N; and A 5 is CR 8 ; or A 1 is C; A 2 is N; A 3 is CR 8 ; A 4 is CR 8 ; and A 5 is N; or A 1 is N; A 2 is C; A 3 is N; A 4 is CR 8 ; and A 5 is CR 8 ; or A 1 is C; A 2 is C; A 3 is N; A 4 is CR 8 ; and A 5 is NR 8 , O, or S; or A 1 is C; A 2 is C; A 3 is NR 8 , O, or S; A 4 is CR 8 ; and A 5 is N; or A 1 is C; A 2 is N; A 3 is N or C=O; A 4 is NR 8 ; and A 5 is N; or A 1 is C; A 2 is C; A 3 is N or C=O; A 4 is NR 8 ; and A 5 is N.

[0096] In some embodiments:

A 1 is C; A 2 is C; A 3 is N; A 4 is NR 8 , O, or S; and A 5 is CR 8 ; or A 1 is C; A 2 is N; A 3 is N; A 4 is CR 8 ; and A 5 is N; or A 1 is C; A 2 is N; A 3 is N; A 4 is CR 8 ; and A 5 is CR 8 ; or A 1 is C; A 2 is C; A 3 is N; A 4 is CR 8 ; and A 5 is NR 8 , O, or S; or A 1 is C; A 2 is N; A 3 is N or C=O; A 4 is NR 8 ; and A 5 is N; or A 1 is C; A 2 is C; A 3 is N or C=O; A 4 is NR 8 ; and A 5 is N. [0097] In some embodiments:

A 1 is C; A 2 is C; A 3 is N; A 4 is NR 8 ; and A 5 is CR 8 ; or A 1 is C; A 2 is N; A 3 is N; A 4 is CR 8 ; and A 5 is N; or A 1 is N; A 2 is C; A 3 is N; A 4 is N; and A 5 is CR 8 .

[0098] In some embodiments, A 1 is C; A 2 is C; A 3 is N; A 4 is NR 8 , O, or S; and A 5 is CR 8 . In some embodiments, A 1 is C; A 2 is C; A 3 is NR 8 , O, or S; A 4 is N; and A 5 is CR 8 . In some embodiments, A 1 is N; A 2 is C; A 3 is N; A 4 is N; and A 5 is CR 8 . In some embodiments, A 1 is C;

A 2 is N; A 3 is N; A 4 is CR 8 ; and A 5 is N. In some embodiments, A 1 is C; A 2 is N; A 3 is N; A 4 is N; and A 5 is N. In some embodiments, A 1 is N; A 2 is C; A 3 is N; A 4 is N; and A 5 is N. In some embodiments, A 1 is C; A 2 is N; A 3 is N; A 4 is CR 8 ; and A 5 is CR 8 . In some embodiments, A 1 is C; A 2 is N; A 3 is N; A 4 is N; and A 5 is CR 8 . In some embodiments, A 1 is C; A 2 is N; A 3 is CR 8 ; A 4 is N; and A 5 is CR 8 . In some embodiments, A 1 is C; A 2 is N; A 3 is CR 8 ; A 4 is CR 8 ; and A 5 is N. In some embodiments, A 1 is N; A 2 is C; A 3 is N; A 4 is CR 8 ; and A 5 is CR 8 . In some embodiments, A 1 is C; A 2 is C; A 3 is N; A 4 is CR 8 ; and A 5 is NR 8 , O, or S. In some embodiments, A 1 is C; A 2 is C; A 3 is NR 8 , O, or S; A 4 is CR 8 ; and A 5 is N. In some embodiments, A 1 is C; A 2 is N; A 3 is N or C=O; A 4 is NR 8 ; and A 5 is N. In some embodiments, A 1 is C; A 2 is C; A 3 is N or C=O; A 4 is NR 8 ; and A 5 is N.

[0099] In some embodiments, each R 8 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci- C , fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, -OR 17 , -C(=O)R 16 , -CO2R 16 , - C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , -NR 16 C(=O)R 17 , -SR 16 , -S(=O)R 17 , -SO2R 17 , or -SO 2 N(R 16 )2; wherein if R 8 is attached to a nitrogen atom, then R 8 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 )2, -S(=O)R 17 , - SO2R 17 , or -SO2N(R 16 )2; or two R 8 attached to the same carbon atom are taken together to form =0, =S, or =NH. In some embodiments, each R 8 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -OH, - OR 17 , -C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , -NR 16 C(=O)R 17 ; wherein if R 8 is attached to a nitrogen atom, then R 8 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted C2-C6 alkenyl, unsubstituted or substituted C2-C6 alkynyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 )2; or two R 8 attached to the same carbon atom are taken together to form =0. In some embodiments, each R 8 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, - CN, -CO2R 16 ; wherein if R 8 is attached to a nitrogen atom, then R 8 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, unsubstituted or substituted Ci-Ce heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CO2R 16 ; or two R 8 attached to the same carbon atom are taken together to form =0. In some embodiments, each R 8 is independently hydrogen, halogen, unsubstituted or substituted C1-C4 alkyl, unsubstituted or substituted C1-C4 deuteroalkyl, unsubstituted or substituted C1-C4 fluoroalkyl, unsubstituted or substituted C1-C4 heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, -CN, -CO2R 16 ; wherein if R 8 is attached to a nitrogen atom, then R 8 is hydrogen, unsubstituted or substituted C1-C4 alkyl, unsubstituted or substituted C1-C4 deuteroalkyl, unsubstituted or substituted C1-C4 fluoroalkyl, unsubstituted or substituted C1-C4 heteroalkyl, unsubstituted or substituted carbocycle, unsubstituted or substituted heterocycle, - CO2R 16 ; or two R 8 attached to the same carbon atom are taken together to form =0. In some embodiments, each R 8 is independently hydrogen, fluorine, methyl, hydroxymethyl, ethyl, trideuteromethyl, trifluoromethyl, cyclopropyl, oxetane, -CN, or -CO2CH3; wherein if R 8 is attached to a nitrogen atom, then R 8 is hydrogen, methyl, hydroxymethyl, ethyl, trideuteromethyl, trifluoromethyl, cyclopropyl, oxetane, -CN, -CO2CH3, or 2-(dimethylamino)- eth-l-yl; or two R 8 attached to the same carbon atom are taken together to form =0. In some embodiments, each R 8 is independently hydrogen, halogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, or unsubstituted or substituted carbocycle; wherein if R 8 is attached to a nitrogen atom, then R 8 is hydrogen, unsubstituted or substituted Ci-Ce alkyl, unsubstituted or substituted Ci-Ce deuteroalkyl, unsubstituted or substituted Ci-Ce fluoroalkyl, or unsubstituted or substituted carbocycle; or two R 8 attached to the same carbon atom are taken together to form =0. In some embodiments, each R 8 is independently hydrogen or unsubstituted or substituted Ci-Ce alkyl. In some embodiments, each R 8 is independently hydrogen or methyl. [0100] In some embodiments p is 0, 1, 2, or 3. In some embodiments p is 0, 1, or 2. In some embodiments p is 0 or 1. In some embodiments p is 1 or 2. In some embodiments p is 0. In some embodiments p is 1. In some embodiments p is 2.

[0101] In some embodiments each R 6 and R 7 is independently hydrogen, deuterium, halogen, Ci-Ce alkyl, Ci-Ce deuteroalkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, monocyclic heterocycle, - CN, -OH, -OR 17 , -C(=O)R 16 , -CO2R 16 , -C(=O)N(R 16 ) 2 , -N(R 16 ) 2 , -NR 16 C(=O)R 17 , -SR 16 , - S(=O)R 17 , -SO2R 17 , or -SO2N(R 16 )2; or one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C=O or a C3-C6 cycloalkane. In some embodiments, each R 6 and R 7 is independently hydrogen, deuterium, halogen, Ci-Ce alkyl, Ci-Ce deuteroalkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, monocyclic heterocycle, -CN, -OH; or one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C=O or a C3-C6 cycloalkane. In some embodiments, each R 6 and R 7 is independently hydrogen, deuterium, halogen, Ci-Ce alkyl, Ci-Ce deuteroalkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl, monocyclic heterocycle; or one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C=O or a C3-C6 cycloalkane. In some embodiments, each R 6 and R 7 is independently hydrogen, deuterium, halogen, Ci-Ce alkyl, Ci-Ce deuteroalkyl, Ci-Ce fluoroalkyl, C3-C6 cycloalkyl; or one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C=O or a C3-C6 cycloalkane. In some embodiments, each R 6 and R 7 is independently hydrogen, deuterium, C1-C4 alkyl, C1-C4 deuteroalkyl, C1-C4 fluoroalkyl, C3-C6 cycloalkyl; or one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form C=O or a C3-C6 cycloalkane. In some embodiments, each R 6 and R 7 is independently hydrogen, deuterium, C1-C4 alkyl, C1-C4 deuteroalkyl; or one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form a C3-C6 cycloalkane. In some embodiments, each R 6 and R 7 is independently hydrogen, deuterium, Ci- Ce alkyl, or Ci-Ce deuteroalkyl; or one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form a C3-C6 cycloalkane. In some embodiments, each R 6 and R 7 is independently hydrogen, deuterium, C1-C4 alkyl, or C1-C4 deuteroalkyl. In some embodiments, each R 6 and R 7 is independently hydrogen, deuterium, methyl, ethyl, propyl, butyl, isopropyl, isobutyl, tert-butyl, trideuteromethyl, -CH2CD3, or - CD2CD3. In some embodiments, each R 6 and R 7 is independently hydrogen, deuterium, methyl, or trideuteromethyl. In some embodiments, each R 6 and R 7 is independently hydrogen or methyl. In some embodiments, each R 6 and R 7 is hydrogen. In some embodiments, each R 6 and R 7 is deuterium. In some embodiments, one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form a C3-C6 cycloalkane. In some embodiments, one R 6 and one R 7 attached to the same carbon atom are taken together with the carbon atom to which they are attached to form a cyclopropane.

[0102] In some embodiments:

X 1 is CR 11 , X 2 is CR 11 , and X 3 is CR 11 ; or X 1 is CR 11 , X 2 is CR 11 , and X 3 is N; or X 1 is CR 11 , X 2 is N, and X 3 is CR 11 ; or X 1 is CR 11 , X 2 is N, and X 3 is N; or X 1 is N, X 2 is CR 11 , and X 3 is CR 11 ; or X 1 is N, X 2 is CR 11 , and X 3 is N; or X 1 is N, X 2 is N, and X 3 is CR 11 .

[0103] In some embodiments:

X 1 is CR 11 , X 2 is CR 11 , and X 3 is CR 11 ; or X 1 is CR 11 , X 2 is CR 11 , and X 3 is N; or X 1 is CR 11 , X 2 is N, and X 3 is CR 11 ; or X 1 is N, X 2 is CR 11 , and X 3 is CR 11 .

[0104] In some embodiments:

X 1 is CR 11 , X 2 is CR 11 , and X 3 is CR 11 ; or or X 1 is CR 11 , X 2 is CR 11 , and X 3 is N.

[0105] In some embodiments, X 1 is CR 11 , X 2 is CR 11 , and X 3 is CR 11 . In some embodiments, X 1 is CR 11 , X 2 is CR 11 , and X 3 is N. In some embodiments, X 1 is CR 11 , X 2 is N, and X 3 is CR 11 . In some embodiments, X 1 is N, X 2 is CR 11 , and X 3 is CR 11 .

[0106] In some embodiments, each R 11 is independently hydrogen, halogen, Ci-Ce alkyl, Ci- C , fluoroalkyl, -CN, -OH, -OR 17 , -N(R 16 )2. In some embodiments, each R 11 is independently hydrogen, halogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, or -CN. In some embodiments, each R 11 is independently hydrogen, halogen, C1-C4 alkyl, or C1-C4 fluoroalkyl. In some embodiments, each R 11 is independently hydrogen, fluorine, methyl, or trifluoromethyl. In some embodiments, each R 11 is independently hydrogen or fluorine. In some embodiments, each R 11 is hydrogen.

[0107] In some embodiments:

X 1 is CH, X 2 is CH, and X 3 is CH; or X 1 is CF, X 2 is CH, and X 3 is CH; or X 1 is CH, X 2 is CF, and X 3 is CH; or X 1 is CH, X 2 is CH, and X 3 is CF; or X 1 is CH, X 2 is CH, and X 3 is N; or X 1 is CF, X 2 is CH, and X 3 is N; or X 1 is CH, X 2 is CF, and X 3 is N; or X 1 is CH, X 2 is N, and X 3 is CH; or X 1 is CF, X 2 is N, and X 3 is CH; or X 1 is CH, X 2 is N, and X 3 is CF; or X 1 is N, X 2 is CH, and X 3 is CH; or X 1 is N, X 2 is CF, and X 3 is CH; or X 1 is N, X 2 is CH, and X 3 is CF. [0108] In some embodiments: X 1 is CH, X 2 is CH, and X 3 is CH; or X 1 is CH, X 2 is CH, and X 3 is N. [0109] In some embodiments, X 1 is CH, X 2 is CH, and X 3 is CH. In some embodiments, In some embodiments, X 1 is CF, X 2 is CH, and X 3 is CH. In some embodiments, X 1 is CH, X 2 is CF, and X 3 is CH. In some embodiments, is CH, X 2 is CH, and X 3 is CF. In some embodiments, X 1 is CH, X 2 is CH, and X 3 is N. In some embodiments, X 1 is CF, X 2 is CH, and X 3 is N. In some embodiments, X 1 is CH, X 2 is CF, and X 3 is N. In some embodiments, X 1 is CH, X 2 is N, and X 3 is CH. In some embodiments, X 1 is CF, X 2 is N, and X 3 is CH. In some embodiments, X 1 is CH, X 2 is N, and X 3 is CF. In some embodiments, X 1 is N, X 2 is CH, and X 3 is CH. In some embodiments, X 1 is N, X 2 is CF, and X 3 is CH. In some embodiments, X 1 is N, X 2 is CH, and X 3 is CF. In some embodiments, Z is -NR 10 -, -O-, or -S-. In some embodiments, Z is -NR 10 - or -O-. In some embodiments Z is -O-. In some embodiments, Z is -NR 10 -.

[0110] In some embodiments, R 10 is hydrogen, Ci-Ce alkyl, or Ci-Ce fluoroalkyl. In some embodiments, R 10 is hydrogen, C1-C4 alkyl, or C1-C4 fluoroalkyl. In some embodiments, R 10 is hydrogen or Ci-Ce alkyl. In some embodiments, R 10 is Ci-Ce alkyl. In some embodiments, R 10 is C1-C4 alkyl. In some embodiments, R 10 is methyl, ethyl, propyl, isopropyl or butyl. In some embodiments, R 10 is methyl or ethyl. In some embodiments, R 10 is methyl. In some embodiments, R 10 is ethyl.

[0111] In some embodiments, n is 1 or 2. In some embodiments, n is 1. In some embodiments, n is 2. [0112] In some embodiments, selected from:

[0113] In some embodiments, the compound is selected from: [0114] In some embodiments, the compound is selected from:

or a pharmaceutically acceptable salt, tautomer, or solvate thereof.

[0115] In some embodiments, the compound is selected from:

or a pharmaceutically acceptable salt, tautomer, or solvate thereof.

[0116] In some embodiments, the compound is selected from: or a pharmaceutically acceptable salt, tautomer, or solvate thereof.

[0117] In some embodiments, the compound is selected from: or a pharmaceutically acceptable salt, tautomer, or solvate thereof.

or a pharmaceutically acceptable salt, tautomer, or solvate thereof.

[0119] In some embodiments, the compound is selected from: or a pharmaceutically acceptable salt, tautomer, or solvate thereof.

[0120] In other aspects, the present disclosure provides pharmaceutical compositions comprising a compound of the present disclosure, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, and a pharmaceutically acceptable excipient.

[0121] In still other aspects, the present disclosure provides methods of treating a disease or condition in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, or a pharmaceutical composition of the present disclosure. In some embodiments, the disease or condition is a TYK2 -mediated disease or condition. In some embodiments, the disease or condition is an inflammatory disease or condition or an autoimmune disease or condition. In some embodiments, the disease or condition is an inflammatory disease or condition. In some embodiments, the inflammatory disease or condition is a neuroinflammatory disease or condition. In some embodiments, the disease or condition is a neurodegenerative disease or condition. In some embodiments, the disease or condition is selected from multiple sclerosis, stroke, epilepsy, encephalomyelitis, polyneuropathy, encephalitis, or a neuromyelitis optica spectrum disorder. In some embodiments, the disease or condition is multiple sclerosis. In some embodiments, the multiple sclerosis is relapsing or relapsing-remitting. In some embodiments, the disease or condition is a neuromyelitis optica spectrum disorder. In some embodiments, the disease or condition is neuromyelitis optica. In some embodiments, the disease or condition is encephalomyelitis. In some embodiments, the disease or condition is acute disseminated encephalomyelitis. In some embodiments, the disease or condition is polyneuropathy. In some embodiments, the disease or condition is chronic inflammatory demyelinating polyneuropathy. In some embodiments, the disease or condition is encephalitis. In some embodiments, the disease or condition is autoimmune encephalitis. In some embodiments, the disease or condition is selected from rheumatoid arthritis, multiple sclerosis, psoriasis, psoriatic arthritis, lupus, systemic lupus erythematosus, Sjogren’s syndrome, ankylosing spondylitis, vitiligo, atopic dermatitis, scleroderma, alopecia, hidradenitis suppurativa, uveitis, dry eye, intestinal bowel disease, Crohn’s disease, ulcerative colitis, celiac disease, Bechet’s disease, type 1 diabetes, systemic sclerosis, and idiopathic pulmonary fibrosis. [0122] Also described herein are pharmaceutical compositions comprising a compound described herein, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, and a 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 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.

[0123] Described herein are compounds of Formula (I), or a pharmaceutically acceptable salt, tautomer, or solvate thereof useful in the treatment of TYK2 -mediated disorders. Described herein are compounds of Formula (I), or a pharmaceutically acceptable salt, tautomer, or solvate thereof, useful in the treatment of an inflammatory or autoimmune disease. In some embodiments, the disease is selected from: multiple sclerosis, such as relapsing or relapsingremitting multiple sclerosis; stroke; epilepsy; encephalomyelitis, such as acute disseminated encephalomyelitis; polyneuropathy, such as chronic inflammatory demyelinating polyneuropathy; encephalitis, such as autoimmune encephalitis; and a neuromyelitis optica spectrum disorder, such as neuromyelitis optica.

[0124] In any of the aforementioned aspects are further embodiments in which the effective amount of the compound of Formula (I), or a pharmaceutically acceptable salt, or solvate 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.

[0125] In any of the aforementioned aspects are further embodiments comprising single administrations of the effective amount of the compound, including further embodiments in which the compound is administered once a day to the mammal or the compound is administered to the mammal multiple times over the span of one day. In some embodiments, the compound is administered on a continuous dosing schedule. In some embodiments, the compound is administered on a continuous daily dosing schedule.

[0126] In any of the embodiments disclosed herein, the patient is a mammal. In any of the embodiments disclosed herein, the mammal is a human. In some embodiments, compounds provided herein are orally administered to a human.

[0127] 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, tautomers, pharmaceutically acceptable N-oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, is used for modulating TYK2, or for the treatment, prevention or amelioration of one or more symptoms of a disease or condition that would benefit from modulating TYK2, are provided.

[0128] 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. [0129] Exemplary compounds described herein include the compounds described in Table 1 :

Table 1:

1: N-(2-methyl-4-((2-methyl-2,4-dihydrochromeno[4,3-c]pyrazol-6 -yl)amino)-3-oxo-2,3- dihydro-lH-pyrazolo[3,4-b]pyridin-6-yl)cyclopropanecarboxami de;

2: N-(4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6 -yl)amino)-2-methyl-3-oxo-

2,3-dihydro-lH-pyrazolo[3,4-b]pyridin-6-yl)cyclopropaneca rboxamide;

3: (S)-N-(2-methyl-3-oxo-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4 ]triazolo[l,5-a]quinoxalin-6- yl)amino)-2,3-dihydro-lH-pyrazolo[3,4-b]pyridin-6-yl)cyclopr opanecarboxamide;

4: (S)-2-amino-6-(cyclopropanecarboxamido)-N-(methyl-d3)-4-((2, 4,5-trimethyl-4,5-dihydro- [ 1 , 2, 4]tri azolof 1 , 5-a]quinoxalin-6-yl)amino)nicotinamide;

5: (5-(4-((2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quino xalin-6-yl)amino)-6-((2,6- dimethylpyrimidin-4-yl)amino)pyridin-3-yl)-l,3,4-oxadiazol-2 -yl)methanol;

6: N-methyl-4-((7-methyl-6,7-dihydro-5H-benzo[f][l,2,4]triazolo [4,3-d][l,4]diazepin-8- yl)amino)-lH-pyrrolo[2,3-b]pyridine-5-carboxamide;

7: 4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6-yl )amino)-N-methyl-lH- pyrrolo[2,3-b]pyridine-5-carboxamide;

8: (S)-6-(cyclopropanecarboxamido)-N-methoxy-4-((2,4,5-trimethy l-4,5-dihydro-

[ 1 , 2, 4]tri azolof 1 , 5-a]quinoxalin-6-yl)amino)nicotinamide;

9: (S)-6-(cyclopropanecarboxamido)-N-ethoxy-4-((2,4,5-trimethyl -4,5-dihydro-

[ 1 , 2, 4]tri azolof 1 , 5-a]quinoxalin-6-yl)amino)nicotinamide;

10: (S)-6-(cyclopropanecarboxamido)-N-methoxy-4-((2,4,5-trimethy l-4,5-dihydro-

[ 1 , 2, 4]tri azolof 1 , 5-a]quinoxalin-6-yl)amino)pyridazine-3 -carboxamide;

11: (S)-6-((5-fluoropyridin-2-yl)amino)-N-methoxy-4-((2,4,5-trim ethyl-4,5-dihydro-

[ 1 , 2, 4]tri azolof 1 , 5-a]quinoxalin-6-yl)amino)pyridazine-3 -carboxamide;

12: (S)-6-((2,6-dimethylpyrimidin-4-yl)amino)-N-methoxy-4-((2,4, 5-trimethyl-4,5-dihydro-

[ 1 , 2, 4]tri azolof 1 , 5-a]quinoxalin-6-yl)amino)pyridazine-3 -carboxamide;

13: (S)-6-(cyclopropanecarboxamido)-N-ethoxy-4-((2,4,5-trimethyl -4,5-dihydro-

[ 1 , 2, 4]tri azolof 1 , 5-a]quinoxalin-6-yl)amino)pyridazine-3 -carboxamide;

14: (S)-6-((2,6-dimethylpyrimidin-4-yl)amino)-N-ethoxy-4-((2,4,5 -trimethyl-4,5-dihydro-

[ 1 , 2, 4]tri azolof 1 , 5-a]quinoxalin-6-yl)amino)pyridazine-3 -carboxamide;

15: (S)-N-ethoxy-6-((5-fluoropyridin-2-yl)amino)-4-((2,4,5-trime thyl-4,5-dihydro-

[ 1 , 2, 4]tri azolof 1 , 5-a]quinoxalin-6-yl)amino)pyridazine-3 -carboxamide; and 16: (S)-6-(cyclopropanecarboxamido)-N-ethoxy-4-((2,4,5-trimethyl -4,5-dihydro-2H- [l,2,3]triazolo[4,5-c][l,7]naphthyridin-6-yl)amino)pyridazin e-3-carboxamide.

[0131] In some embodiments, provided herein is a pharmaceutically acceptable salt of a compound that is described in Table 1. Table 2 provides other TYK2 inhibitors.

Table 2. Other TYK2 inhibitors.

[0132] In one aspect, compounds described herein are in the form of pharmaceutically acceptable salts. 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.

[0133] “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 at the concentration or amount used, 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.

[0134] 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/Zurich:Wiley-VCH/VHCA, 2002. Pharmaceutical salts typically are more soluble and more rapidly soluble in stomach and intestinal juices 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.

[0135] In some embodiments, pharmaceutically acceptable salts are obtained by reacting a compound of Formula (I) with an acid. In some embodiments, the compound of Formula (I) (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-hydroxy ethanesulfonic 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; naphthal ene-l,5-disulfonic acid; naphthal ene-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 (/?); and undecylenic acid.

[0136] In some embodiments, a compound of Formula (I) is prepared as a chloride salt, sulfate salt, bromide salt, mesylate salt, maleate salt, citrate salt or phosphate salt.

[0137] In some embodiments, pharmaceutically acceptable salts are obtained by reacting a compound of Formula (I) with a base. In some embodiments, the compound of Formula (I) is acidic and is reacted with a base. In such situations, an acidic proton of the compound of Formula (I) 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.

[0138] 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 crystallization 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.

[0139] The methods and formulations described herein include the use of A-oxides (if appropriate), or pharmaceutically acceptable salts of compounds having the structure of Formula (I), as well as active metabolites of these compounds having the same type of activity.

[0140] In some embodiments, sites on the organic radicals (e.g. alkyl groups, aromatic rings) of compounds of Formula (I) are susceptible to various metabolic reactions. Incorporation of appropriate substituents on the organic radicals 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.

[0141] 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.

[0142] 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, sulfur, fluorine chlorine, iodine, phosphorus, such as, for example, 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 0, 35 S, 18 F, 36 C1, 123 I, 124 I, 125 I, 134 1, 32 P and 33 P. 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.

[0143] In some embodiments, the compounds of Formula (I) possess one or more stereocenters and each stereocenter exists independently in either the R or S configuration. In some embodiments, the compound of Formula (I) exists in the R configuration. In some embodiments, the compound of Formula (I) exists in the S configuration. The compounds presented herein include all diastereomeric, individual enantiomers, 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.

[0144] Individual stereoisomers are obtained, if desired, by methods such as, stereoselective synthesis and/or the separation of stereoisomers by chiral chromatographic columns or the separation of diastereomers by either non-chiral or chiral chromatographic columns or crystallization and recrystallization in a proper solvent or a mixture of solvents. In certain embodiments, compounds of Formula (I) 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 individual enantiomers. In some embodiments, resolution of individual 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.

[0145] 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. 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.

[0146] Prodrugs of the compounds described herein include, but are not limited to, esters, ethers, carbonates, thiocarbonates, N-acyl derivatives, N-acyloxyalkyl derivatives, N- alkyloxyacyl 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. [0147] Prodrug forms of the herein described compounds, wherein the prodrug is metabolized in vivo to produce a compound of Formula (I) 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.

[0148] In some embodiments, any one of the hydroxyl group(s), amino group(s), and/or carboxylic acid group(s) are functionalized in a suitable manner to provide a prodrug moiety. In some embodiments, the prodrug moiety is as described above.

[0149] 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.

[0150] 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 sulfhydryl 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. [0151] In some instances, heterocyclic rings may exist in tautomeric forms. In such situations, it is understood that the structures of said compounds are illustrated or named in one tautomeric form but could be illustrated or named in the alternative tautomeric form. The alternative tautomeric forms are expressly included in this disclosure, such as, for example, the structures illustrated below. For example, pyridones could exist in the following tautomeric forms: are encapsulated within the group, “substituted pyridines.” Similarly, triazolones could exist in the following tautomeric forms, which include zwitterionic forms: all of which are encapsulated within the group, “substituted 5-membered heteroaryl.” Similarly, pyrazoles, triazoles, pyrimidines, and the like are known to tautomerize; for the purpose of this disclosure, all tautomeric forms (including charged and zwitterionic tautomers) are considered within the scope of the present disclosure.

General Synthesis of the Compounds of the Present Disclosure

[0152] Compounds of Formula (I) described herein are synthesized using standard synthetic techniques or using methods known in the art in combination with methods described herein.

[0153] Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC are employed.

[0154] 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.

[0155] In some embodiments, compounds are prepared as described in the Examples.

Certain Terminology

[0156] 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. [0157] As used herein, Ci-C x includes C1-C2, C1-C3 . . . Ci-C x . By way of example only, a group designated as “Ci-Ce” indicates that there are one to six 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, “C1-C4 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, Ao-propyl, //-butyl, iso- butyl, ec-butyl, and /-butyl.

[0158] 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 Ci- Cioalkyl. 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, 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 Ci-Ce 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.

[0159] An “alkylene” group refers to a divalent alkyl radical. 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 Ci-Ce alkylene. In other embodiments, an alkylene is a Ci-C4alkylene. Typical alkylene groups include, but are not limited to, -CH2-, -CH2CH2-, - CH2CH2CH2-, -CH2CH2CH2CH2-, and the like. In some embodiments, an alkylene is -CH2-. [0160] An “alkoxy” group refers to a (alkyl)O- group, where alkyl is as defined herein.

[0161] An “alkylthio” group refers to a (alkyl)S- group, where alkyl is as defined herein. [0162] An “alkylsulfoxide” group refers to a (alkyl)S(O)- group, where alkyl is as defined herein.

[0163] An “alkylsulfone” group refers to a (alkyl)S(O)2- group, where alkyl is as defined herein.

[0164] The term “alkylamine” refers to the -N(alkyl) x H y 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.

[0165] An “hydroxyalkyl” refers to an alkyl in which one hydrogen atom is replaced by a hydroxyl. In some embodiments, a hydroxyalkyl is a Ci-C4hydroxyalkyl. Typical hydroxyalkyl groups include, but are not limited to, -CH2OH, -CH2CH2OH, -CH2CH2CH2OH, - CH2CH2CH2CH2OH, and the like.

[0166] An “aminoalkyl” refers to an alkyl in which one hydrogen atom is replaced by an amino. In some embodiments, aminoalkyl is a Ci-C4aminoalkyl. Typical aminoalkyl groups include, but are not limited to, -CH2NH2, -CH 2 CH 2 NH 2 , -CH 2 CH 2 CH 2 NH 2 , - CH 2 CH 2 CH 2 CH 2 NH 2 , and the like.

[0167] 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 (z.e., vinyl), propenyl (z.e., allyl), butenyl, pentenyl, pentadienyl, and the like. Nonlimiting examples of an alkenyl group include -CH=CH 2 , -C(CH3)=CH 2 , -CH=CHCH 3 , - C(CH 3 )=CHCH 3 , and -CH 2 CH=CH 2 .

[0168] 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.

[0169] 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 Ci-Ceheteroalkyl.

[0170] The term “aromatic” refers to a planar ring having a delocalized > -electron system containing 4n+2 > 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 (z.e., rings which share adjacent pairs of carbon atoms) groups.

[0171] 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. Carbocycles include aryls and cycloalkyls.

[0172] 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 phenyl, naphthyl, indanyl, indenyl, or tetrahydronaphthyl. In some embodiments, an aryl is a Ce-Cioaryl. Depending on the structure, an aryl group is a monoradical or a diradical (i.e., an arylene group). [0173] An “aryloxy” group refers to a (aryl)O- group, where alkyl is as defined herein. [0174] An “arylthio” group refers to a (aryl)S- group, where alkyl is as defined herein. [0175] An “arylsulfoxide” group refers to a (aryl)S(O)- group, where alkyl is as defined herein.

[0176] An “arylsulfone” group refers to a (aryl)S(O)2- group, where alkyl is as defined herein. [0177] The term “cycloalkyl” refers to a monocyclic or polycyclic aliphatic, non-aromatic radical, 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 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, norbomyl, and bicyclo[l.l. l]pentyl. In some embodiments, a cycloalkyl is a Cs-Cecycloalkyl. In some embodiments, a cycloalkyl is a C3-C4cycloalkyl.

[0178] The term “halo” or, alternatively, “halogen” or “halide” means fluoro, chloro, bromo, or iodo. In some embodiments, halo is fluoro, chloro, or bromo.

[0179] 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 Ci-Cefluoroalkyl.

[0180] The term “deuteroalkyl” refers to an alkyl in which one or more hydrogen atoms are replaced by a deuterium atom. In one aspect, a deuteroalkyl is a Ci-Cedeuteroalkyl.

[0181] The term “heterocycle” or “heterocyclic” refers to heteroaromatic rings (also known as heteroaryls) and heterocycloalkyl rings 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. 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-l(2H)-onyl, 3,4-dihydroquinolin-2(lH)- onyl, isoindoline-1, 3-dithionyl, benzo[d]oxazol-2(3H)-onyl, lH-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 TV-attached where such is possible. For instance, a group derived from pyrrole includes both pyrrol-l-yl (TV- attached) or pyrrol-3-yl (C-attached). Further, a group derived from imidazole includes imidazol-l-yl or imidazol-3-yl (both TV-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 (=0) 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.

[0182] 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. Monocyclic heteroaryls include indolizine, indole, benzofuran, benzothiophene, indazole, benzimidazole, 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 0 atoms, and 0-1 S atoms in the ring. In some embodiments, a heteroaryl contains 1-4 N atoms, 0-1 0 atoms, and 0-1 S atoms in the ring. In some embodiments, heteroaryl is a C1-C9 heteroaryl. In some embodiments, monocyclic heteroaryl is a Ci-Csheteroaryl. In some embodiments, monocyclic heteroaryl is a 5-membered or 6-membered heteroaryl. In some embodiments, bicyclic heteroaryl is a C6-C9 heteroaryl.

[0183] A “heterocycloalkyl” group refers to a cycloalkyl group that includes at least one heteroatom selected from nitrogen, oxygen and sulfur. 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. In one aspect, a heterocycloalkyl is a C2-Cioheterocycloalkyl. In another aspect, a heterocycloalkyl is a C4-Cioheterocycloalkyl. In some embodiments, a heterocycloalkyl is monocyclic or bicyclic. In some embodiments, a heterocycloalkyl is monocyclic and is a 3, 4, 5, 6, 7, or 8-membered ring. In some embodiments, a heterocycloalkyl is monocyclic and is a 3, 4, 5, or 6-membered ring. In some embodiments, a heterocycloalkyl is monocyclic and is a 3 or 4-membered ring. 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.

[0184] The term “bond” or “single bond” refers to a chemical bond between two atoms, or two moi eties 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.

[0185] 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.

[0186] 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 halogen, -CN, -NH2, -NH(alkyl), -N(alkyl)2, -OH, -CO2H, -CO2alkyl, -C(=O)NH2, -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 halogen, -CN, -NH2, - NH(CH 3 ), -N(CH 3 ) 2 , -OH, -CO2H, -CO 2 (Ci-C 4 alkyl), -C(=O)NH 2 , -C(=O)NH(Ci-C 4 alkyl), - C(=O)N(Ci-C 4 alkyl) 2 , -S(=O) 2 NH 2 , -S(=O) 2 NH(Ci-C 4 alkyl), -S(=O) 2 N(Ci-C 4 alkyl) 2 , Ci- C 4 alkyl, C 3 -Cecycloalkyl, Ci-C 4 fluoroalkyl, Ci-C 4 heteroalkyl, Ci-C 4 alkoxy, Ci-C 4 fluoroalkoxy, -SCi-C 4 alkyl, -S(=O)Ci-C 4 alkyl, and -S(=O)2Ci-C 4 alkyl. In some embodiments, optional substituents are independently selected from halogen, -CN, -NH2, -OH, -NH(CH 3 ), -N(CH 3 )2, - CH 3 , -CH2CH 3 , -CHF2, -CF 3 , -OCH 3 , -OCHF2, and -OCF 3 . In some embodiments, substituted groups are substituted with one or two of the preceding groups. In some embodiments, an optional substituent on an aliphatic carbon atom (acyclic or cyclic) includes oxo (=0).

[0187] In some embodiments, each substituted alkyl, substituted alkenyl, substituted alkynyl, substituted deuteroalkyl, substituted alkoxy, substituted fluoroalkyl, substituted heteroalkyl, substituted carbocycle, and substituted heterocycle is substituted with one or more R s groups independently selected from the group consisting of deuterium, halogen, Ci-Ce alkyl, monocyclic carbocycle, monocyclic heterocycle, -CN, -OR 18 , -CO2R 18 , -C(=O)N(R 18 )2, - N(R 18 ) 2 , -NR 18 C(=O)R 19 , -SR 18 , -S(=O)R 19 , -SO2R 19 , and -SO 2 N(R 18 )2; each R 18 is independently selected from hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C2-C6 heterocycloalkyl, phenyl, benzyl, 5-membered heteroaryl and 6-membered heteroaryl; or two R 18 groups are taken together with the N atom to which they are attached to form a N-containing heterocycle; each R 19 is independently selected from Ci-Ce alkyl, Ci-Ce fluoroalkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C2-C6 heterocycloalkyl, phenyl, benzyl, 5- membered heteroaryl and 6-membered heteroaryl.

[0188] In some embodiments, each substituted alkyl, substituted fluoroalkyl, substituted heteroalkyl, substituted carbocycle, and substituted heterocycle is substituted with one or more R s groups independently selected from the group consisting of deuterium, halogen, Ci-Ce alkyl, monocyclic carbocycle, monocyclic heterocycle, -CN, -OR 18 , -CO2R 18 , -C(=O)N(R 18 )2, - N(R 18 ) 2 , -NR 18 C(=O)R 19 , -SR 18 , -S(=O)R 19 , -SO2R 19 , and -SO 2 N(R 18 )2; each R 18 is independently selected from hydrogen, Ci-Ce alkyl, Ci-Ce fluoroalkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C2-C6 heterocycloalkyl, phenyl, benzyl, 5-membered heteroaryl and 6-membered heteroaryl; or two R 18 groups are taken together with the N atom to which they are attached to form a N-containing heterocycle; each R 19 is independently selected from Ci-Ce alkyl, Ci-Ce fluoroalkyl, Ci-Ce heteroalkyl, C3-C6 cycloalkyl, C2-C6 heterocycloalkyl, phenyl, benzyl, 5- membered heteroaryl and 6-membered heteroaryl.

[0189] 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.

[0190] 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.

[0191] 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 antagonist. In some embodiments, a modulator is an inhibitor.

[0192] 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.

[0193] 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.

[0194] 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.

[0195] 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.

[0196] The term “pharmaceutical combination” as used herein, means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term “fixed combination” means that the active ingredients, e.g. a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term “non-fixed combination” means that the active ingredients, e.g. a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and a co-agent, are administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific intervening time limits, wherein such administration provides effective levels of the two compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more active ingredients. [0197] The terms “article of manufacture” and “kit” are used as synonyms.

[0198] 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.

[0199] 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 or progression of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a secondary condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactically and/or therapeutically.

Pharmaceutical Compositions

[0200] 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 & Wilkins 1999), herein incorporated by reference for such disclosure. [0201] A compound or a pharmaceutical composition of the present disclosure is, in some embodiments, useful for the treatment of a TYK2 mediated disease or disorder. In some embodiments, the pharmaceutical composition is effective at treating a disease or disorder wherein TYK2 is overexpressed or hyperactive. In some embodiments, the pharmaceutical composition is effective at treating a disease or disorder which would benefit from a reduction in TYK2 activity or expression.

[0202] In some embodiments, the pharmaceutical composition is useful in the treatment of disease or disorder associated with high levels of cytokines driven by TYK2, such as interferons (e.g. IFN-a, IFN-P, IFN-K, IFN-5, IFN-S, IFN-T, IFN-CO, and IFN-^ (also known as limitin), and interleukins (e.g. IL-6, IL-10, IL-12, IL-23, oncostatin M, ciliary neurotrophic factor, cardiotrophin 1, cardiotrophin-like cytokine, and LIF. In some embodiments, the disease or disorder is an inflammatory disease or disorder, an autoimmune disease or disorder, a respiratory disease or disorder, type 1 diabetes, and interferonopathies such as Alcardi-Goutieres syndrome, or combinations thereof.

[0203] In some embodiments, the pharmaceutical composition is useful in the treatment of an inflammatory disease or disorder. In some embodiments, the inflammatory disease or disorder is an auto-inflammatory disease or disorder, a host-mediated inflammatory disease or disorder, an injury-related inflammatory disease or disorder, an infection-related inflammatory disease or disorder, a hyperproliferative (e.g., cancer, fibrosis) mediated inflammatory disease or disorder. In some embodiments, the inflammatory disease or disorder or infection-related inflammatory disease or disorder is a respiratory disease or disorder. In some embodiments, the respiratory disease or disorder is associated with a viral in microbial infection. In some embodiments, the respiratory disease or disorder is a problematic immune response to a viral or microbial infection. In some embodiments, the respiratory disease or disorder is associated with a coronavirus such as MERS-CoV, SARS-CoV-1, or SARS-CoV-2. In some embodiments, the pharmaceutical composition is effective in decreasing symptoms associated with COVID-19, or an immune response associated therewith.

[0204] In some embodiments, the pharmaceutical composition is useful in the treatment of an autoimmune disease or disorders. In some embodiments, an autoimmune disease or disorder is rheumatoid arthritis, multiple sclerosis, psoriasis, psoriatic arthritis, lupus, systemic lupus erythematosus, Sjogren’s syndrome, ankylosing spondylitis, vitiligo, atopic dermatitis, scleroderma, alopecia, hidradenitis suppurativa, uveitis, dry eye, intestinal bowel disease, Crohn’s disease, ulcerative colitis, celiac disease, Bechet’s disease, type 1 diabetes, systemic sclerosis, and idiopathic pulmonary fibrosis. In some embodiments, an autoimmune disease or disorder is lupus or systemic lupus erythematosus. In some embodiments, an autoimmune disease or disorder is psoriasis. In some embodiments, an autoimmune disease or disorder is irritable bowel disease (IBS) or irritable bowel disease with diarrhea (IBS-D). In some embodiments, an autoimmune disease or disorder is dry eye or uveitis. In some embodiments, an autoimmune disease or disorder is Crohn’s disease. In some embodiments, an autoimmune disease or disorder is atopic dermatitis.

[0205] 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 effected 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, topical application such as creams or ointments. Additional examples of local administration of the present compounds include eye drops, ocular creams, gels or hydrogels, implants, transdermal patches, or drug depots. In some embodiments, a pharmaceutical composition is administered orally (e.g., in a liquid formulation, tablet, capsule, nebulized liquid, aerosolized liquid, dry powder spray).

[0206] 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.

[0207] 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.

[0208] 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 multidose 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.

[0209] Pharmaceutical compositions may also be formulated as a depot preparation. Such long-acting formulations may be administered by implantation (for example subcutaneously). 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.

[0210] Pharmaceutical compositions may be administered topically, that is by non-systemic administration. This includes the application of a compound of the present disclosure externally to the epidermis or the buccal cavity and the installation 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.

[0211] Pharmaceutical compositions suitable for topical administration include liquid or semiliquid 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.

[0212] 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.

[0213] 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

[0214] In one embodiment, the compound described herein, or a pharmaceutically acceptable salt, tautomer, or solvate thereof, are used in the preparation of medicaments for the treatment of diseases or conditions in a mammal that would benefit from modulation of TYK2 activity. 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.

[0215] 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.

[0216] 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.

[0217] In certain embodiments wherein the patient’s condition does not improve, upon the doctor’s discretion the administration of 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. [0218] 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. [0219] 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.

[0220] In general, however, doses employed for adult human treatment are typically in the range of 0.01 mg-2000 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.

[0221] In one embodiment, the daily dosages appropriate for the compound described herein, or a pharmaceutically acceptable salt thereof, described herein 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.

[0222] 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.

[0223] 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.

[0224] 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.

[0225] 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.

Combination Treatments

[0226] In certain instances, it is appropriate to administer at least one compound described herein, or a pharmaceutically acceptable salt thereof, in combination with one or more other therapeutic agents.

[0227] In one embodiment, the therapeutic effectiveness of one of the compounds described herein is enhanced by administration of an adjuvant (z.e., by itself the adjuvant has minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced). Or, in some embodiments, the benefit experienced by a patient is increased by administering one of the compounds described herein with another agent (which also includes a therapeutic regimen) that also has therapeutic benefit.

[0228] In one specific embodiment, a compound described herein, or a pharmaceutically acceptable salt thereof, is co-administered with a second therapeutic agent, wherein the compound described herein, or a pharmaceutically acceptable salt thereof, and the second therapeutic agent modulate different aspects of the disease, disorder or condition being treated, thereby providing a greater overall benefit than administration of either therapeutic agent alone. [0229] In any case, regardless of the disease, disorder or condition being treated, the overall benefit experienced by the patient may simply be additive of the two therapeutic agents or the patient may experience a synergistic benefit.

[0230] For combination therapies described herein, dosages of the co-administered compounds vary depending on the type of co-drug employed, on the specific drug employed, on the disease or condition being treated and so forth. In additional embodiments, when coadministered with one or more other therapeutic agents, the compound provided herein is administered either simultaneously with the one or more other therapeutic agents, or sequentially.

[0231] In combination therapies, the multiple therapeutic agents (one of which is one of the compounds described herein) are administered in any order or even simultaneously. If administration is simultaneous, the multiple therapeutic agents are, by way of example only, provided in a single, unified form, or in multiple forms (e.g., as a single pill or as two separate pills).

[0232] 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. EXAMPLES

[0233] As used above, and throughout the description of the disclosure, the following abbreviations, unless otherwise indicated, shall be understood to have the following meanings:

Abbreviations:

ACN acetonitrile

CAN ceric ammonium nitrate

DCM dichloromethane

DIBAL diisobutylaluminum hydride

DIPEA M -diisopropylethylamine

DMA dimethylacetamide

DMF 7V,7V-di methyl form am ide

DMSO dimethylsulfoxide

EtOAc ethyl acetate

EGTA ethylene glycol-bis(P-aminoethyl ether)-A,A,7V’,7V’ -tetraacetic acid

ES electrospray

FBS fetal bovine serum

GST glutathione S-transferase

HEK human embryonic kidney

HEPES 4-(2-hy droxy ethyl)- 1 -piperazineethanesulfonic acid

HMDS bis(trimethylsilyl)amide

HPLC high pressure liquid chromatography

HTRF homogenous time resolved fluorescence

IC50 half maximal inhibitory concentration

IFN interferon

IL interleukin

IPA isopropyl alcohol

JAK Janus kinase

LCMS liquid chromatography-mass spectrometry

MDI metered drug inhalant

MW micro wave

NMR nuclear magnetic resonance

SEAP secreted embryonic alkaline phosphatase

STAT signal transducer and activator of transcription

T3P propanephosphonic acid anhydride TBAF tetra-n-butylammonium fluoride

TBDMS tert-butyldimethylsilyl

TBDPS tert-butyldiphenylsilyl

TEA triethylamine

TFA trifluoroacetic acid

THF tetrahydrofuran

TLC thin-layer chromatography

TYK non-receptor tyrosine-protein kinase

[0234] The following examples are provided for illustrative purposes only and not to limit the scope of the claims provided herein.

I. Synthesis of Compounds

Example 1: Preparation of 2-methyl-2,4-dihydrochromeno[4,3-c]pyrazol-6-amine (1-1):

[0235] Step-1: 3-bromo-2-(prop-2-yn-l-yloxy)benzaldehyde (I-lb): To a stirred solution of 3-bromo-2-hydroxybenzaldehyde I-la (10.0 g, 49.7 mmol) in DMF (50.0 mL) was added anhydrous K2CO3 (20.6 g, 149 mmol) at 0 °C and stirred for 10 min. To this was then added propargyl bromide (7.54 mL, 99.5 mmol) and the reaction mixture was allowed to stir at room temperature for 16 h. The progress of the reaction was monitored by TLC. After complete consumption of starting material, water (250 mL) was added to it and extraction was carried out using EtOAc (100 mL x 3). The combined organic extracts were washed with water (100 mL x 3), brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford 3-bromo-2-(prop-2-yn-l-yloxy)benzaldehyde I-lb (11.3 g) as a yellow solid. LCMS (ES) m/z: 239.1 [M+H] + .

[0236] Step-2: 6-bromo-2,4-dihydrochromeno[4,3-c]pyrazole (I-lc): To a stirred solution of I-lb (11.3 g, 47.3 mmol) in MeOH (25.0 mL) and water (25.0 mL) was added KOBu tert (8.03 g, 70.9 mmol) and /?-toluenesulfonylhydrazide (17.6 g, 93.8 mmol) at room temperature. The reaction mixture was then stirred at 70 °C for 16 h. The progress of the reaction was monitored by TLC. After complete consumption of starting material, the reaction mixture was cooled to room temperature and water (250 mL) was added to it. Extraction was carried out using EtOAc (100 mL x 3); the combined organic extracts were washed with brine (100 mL x 2), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting crude was purified by Combi-Flash (using gradient elution of 0-20% EtOAc in hexane) to afford 6-bromo- 2,4-dihydrochromeno[4,3-c]pyrazole I-lc (10 g) as an off-white solid. LCMS (ES) m/z 251.0 [M+H] + .

[0237] Step-3: 6-bromo-2-methyl-2,4-dihydrochromeno[4,3-c] pyrazole (I-ld) and 6- bromo-l-methyl-l,4-dihydrochromeno[4,3-c]pyrazole (1-le): To a solution of I-lc (4.00 g, 15.9 mmol) in DMF (60.0 mL) was added CS2CO3 (10.4 g, 31.9 mmol) under nitrogen atmosphere and stirred for 30 min. To this was then added Mel (1.20 mL, 19.1 mmol) and the reaction mixture was stirred at 80 °C for 16 h in a sealed tube. The progress of the reaction was monitored by TLC. After completion, it was cooled to room temperature and water (50 mL) was added to it. Extraction was carried out using EtOAc (3 x 50 mL); the combined organic extracts were washed with water (50 mL x 3), brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting crude was purified by Combi-Flash (using gradient elution of 0-20% EtOAc in hexane) to afford 6-bromo-2-methyl-2,4- dihydrochromeno[4,3-c] pyrazole I-ld (3.4 g) as an off-white solid {LCMS (ES) m/z'. 265.0 [M+H] + . X H NMR (400 MHz, DMSO-tL) 3 7.61 (s, 1H); 7.58 (d, J= 7.2 Hz, 1H); 7.46 (d, J= 7.6 Hz, 1H); 6.93 (t, J= 7.6 Hz, 1H); 5.36 (s, 2H); 3.89 (s, 3H)} and 6-bromo- 1 -methyl- 1,4- dihydrochromeno[4,3-c]pyrazole 1-le (0.8 g) as an off-white solid {LCMS (ES) m/z 265.0 [M+H] + . X H NMR (400 MHz, DMSO-tL) 3 7.72 (d, J= 8.0 Hz, 1H); 7.55 (d, J= 8.0 Hz, 1H); 7.34 (s, 1H); 6.99 (t, J= 8.0 Hz, 1H); 5.30 (s, 2H); 3.87 (s, 3H)}.

[0238] Step-4: tert-butyl (2-methyl-2,4-dihydrochromeno[4,3-c] pyrazol-6-yl)carbamate (I-lf): Argon gas was purged through a stirred suspension of I-ld (3.40 g, 12.8 mmol), tertbutyl carbamate (2.25 g, 19.2 mmol) and CS2CO3 (8.36 g, 25.6 mmol) in 1,4-dioxane (20.00 mL) for 15 min. To this was then added [5-(diphenylphosphanyl)-9,9-dimethyl-9H-xanthen-4- yl]diphenylphosphane (1.48 g, 2.56 mmol) and Pd(OAc)2 (576 mg, 2.56 mmol). The reaction mixture was then stirred at 100 °C for 16 h in a sealed tube. It was then cooled to room temperature, filtered through Celite bed and washed with EtOAc (50 mL x 2). The combined filtrate was concentrated under reduced pressure and the residue was purified by Combi-Flash (using gradient elution of 0-50% EtOAc in hexane) to afford tert-butyl (2-methyl-2,4- dihydrochromeno[4,3-c] pyrazol-6-yl)carbamate I-lf (2.30 g) as a pale yellow solid. LCMS (ES) m/z: 301.9 [M+H] + . [0239] Step-5: 2-methyl-2,4-dihydrochromeno[4,3-c] pyrazol-6-amine (1-1): To a stirred solution of I-lf (2.30 g, 7.63 mmol) in DCM (40.0 mL) was added trifluoroacetic acid (10.0 mL) at 0 °C under nitrogen atmosphere and the reaction mixture was allowed to warm to room temperature over 2 h. The progress of the reaction was monitored by TLC. After completion, volatiles were removed under reduced pressure and saturated NaHCOs solution (50 mL) was added to the residue. Extraction was carried out using EtOAc (3 x 30 mL); the combined organic extracts were washed with water (30 mL), brine (30 mL), dried over anhydrous ISfeSCU, filtered and evaporated under reduced pressure to afford 2-methyl-2,4-dihydrochromeno[4,3-c] pyrazol- 6-amine 1-1 (1.20 g) as a pale yellow solid. LCMS (ES) m/z 202.1 [M+H] + . 'H NMR (400 MHz, DMSO ) 3 7.53 (s, 1H); 6.81 (d, J= 7.2 Hz, 1H); 6.70 (t, J= 7.2 Hz, 1H); 6.57 (d, J = 7.6 Hz, 1H); 5.19 (s, 2H); 4.75 (br s, 2H); 3.86 (s, 3H).

Example 2: Preparation of 2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6-amine

[0240] Step-1: Methyl 3-bromo-2-(methylamino)benzoate (I-2b): To a stirred solution of methyl 3-bromo-2-fluorobenzoate I-2a (5.0 g, 21.5 mmol) in 1,4-dioxane (20 mL) was added potassium carbonate (5.93 g, 42.9 mmol) and a solution of 2M MeNEL in THF (21.5 mL, 42.9 mmol) at room temperature. The reaction mixture was then stirred at 40 °C in a sealed tube for 16 h, while monitoring progress by LCMS. After completion, volatiles were removed under reduced pressure and the residue was partitioned between DCM (100 mL) and water (50 mL). The organic layer was separated and washed with brine (50 mL), dried over anhydrous ISfeSCL, filtered and concentrated under reduced pressure to provide desired compound methyl 3-bromo- 2-(methylamino) benzoate I-2b (5.0 g) as a yellow oil. LCMS (ES) m/z 244.0 [M+H] + ; 246.0. [0241] Step-2: (3-bromo-2-(methylamino)phenyl)methanol (I-2c): To a stirred solution of I-2b (9.50 g, 38.9 mmol) in anhydrous DCM (100 mL) was added a IM solution of DIBAL-H (117 mL, 117 mmol) drop wise at 0 °C. The reaction mixture was then allowed to warm to room temperature over 4 h, while monitoring reaction progress by TLC. After completion, it was cooled to 0 °C and quenched slowly with addition of 10% aqueous solution of citric acid (30 mL). Extraction was carried out using DCM (100 mL x 2); the combined organic extracts were washed with brine (100 mL), dried over anhydrous ISfeSCU, filtered and concentrated under reduced pressure. The resulting crude was purified by Combi-Flash (using gradient elution of 0- 60% EtOAc in hexane) to afford desired compound (3-bromo-2-(methylamino)phenyl)methanol

1-2c (6.5 g) as a colorless oil. LCMS (ES) m/z 216.0 [M+H] + , 217.8.

[0242] Step-3: 2-bromo-6-(((tert-butyldimethylsilyl)oxy)methyl)-N-methylani line (I-2d): To a stirred solution of I-2c (6.50 g, 30.1 mmol) in anhydrous DMF (60 mL) was added 1H- imidazole (3.07 g, 45.1 mmol) and TBDMSC1 (5.67 g, 37.6 mmol) at room temperature. The reaction mixture was then stirred at room temperature for 16 h. Saturated NaHCOs solution (50 mL) was added to it and extraction was carried out using diethyl ether (70 mL x 3). The combined organic extracts were washed with water (50 mL x 3), brine (50 mL), dried over anhydrous MgSCU, filtered and concentrated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-50% EtOAc in hexane) to afford desired compound

2-bromo-6-(((tert-butyldimethylsilyl)oxy)methyl)-N-methyl aniline I-2d (6.5 g) as a pale yellow oil. 'H NMR (400 MHz, CDCh) 3 7.41 (d, J= 8.0 Hz, 1H); 7.30 (d, J= 8.0 Hz, 1H); 6.82 (t, J = 8.0 Hz, 1H); 4.74 (s, 2H); 3.95 (s, 1H); 2.82 (s, 3H); 0.92 (s, 9H); 0.08 (s, 6H).

[0243] Step-4: 2-bromo-6-(((tert-butyldimethylsilyl)oxy)methyl)-N-methyl-N- (prop-2-yn- l-yl)aniline (I-2e): To a stirred solution of I-2d (6.50 g, 19.7 mmol) in ACN (65 mL) was added anhydrous K2CO3 (8.16 g, 59.0 mmol) and propargyl bromide (4.47 mL, 59.0 mmol) at room temperature. The reaction mixture was then stirred at 80 °C for 24 h, while monitoring reaction progress by TLC. After completion, it was cooled to room temperature, the base was filtered off through Celite and washed with ACN (10 mL x 2). The filtrate was concentrated under reduced pressure and the residue was diluted with EtOAc (100 mL). The organic layer was washed with water (50 mL), brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-50% EtOAc in hexane) to afford desired compound 2-bromo-6-{[(tert- butyldimethylsilyl)oxy]methyl}-N-methyl-N-(prop-2-yn-l-yl)an iline I-2e (7.0 g) as a brown solid. X H NMR (400 MHz, CDCh) d 7.50 (d, J= 7.6 Hz, 1H); 7.43 (d, J= 8.0 Hz, 1H); 7.09 (t, J = 8.0 Hz, 1H); 4.95-4.77 (m, 2H); 4.00-3.82 (m, 2H); 2.90 (s, 3H); 2.19 (t, J= 2.4 Hz, 1H); 0.92 (s, 9H); 0.08 (s, 6H).

Ill [0244] Step-5: (3-bromo-2-(methyl(prop-2-yn-l-yl)amino)phenyl)methanol (I-2f): IM solution of TBAF in THF (22.8 mL, 22.8 mmol) was added to a stirred solution of I-2e (7.00 g, 19.0 mmol) in THF (70 mL) at 0 °C and the reaction mixture was stirred for 30 min. After complete consumption of starting material (as indicated by TLC), saturated NH4CI solution (50 mL) was added to it and extraction was carried out using EtOAc (70 mL x 2). The combined organic extracts were washed with brine (70 mL), dried over MgSCU, filtered and concentrated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0- 100% EtOAc in hexane) to afford desired compound (3-bromo-2-[methyl(prop-2-yn-l- yl)amino]phenyl)methanol I-2f (4.5 g) as a colorless oil. LCMS (ES) m/z'. 254.0 [M+H] + , 256.0.

[0245] Step-6: 3-bromo-2-(methyl(prop-2-yn-l-yl)amino)benzaldehyde (I-2g): To a stirred solution of I-2f (4.50 g, 17.7 mmol) in DCM (50.0 mL) was added DMP (11.3 g, 26.6 mmol) at 0 °C and the reaction mixture was allowed to warm to room temperature over 1 h. The reaction progress was monitored by LCMS. After completion, it was filtered through Celite bed and washed with DCM (20 mL x 2). The resulting filtrate was washed with saturated NaHCOs solution (30 mL), water (30 mL), brined (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-50% EtOAc in hexane) to afford desired compound 3-bromo-2-[methyl(prop-2-yn- l-yl)amino]benzaldehyde I-2g (3.60 g) as a yellow oil. LCMS (ES) m z 252.0 [M+H] + , 254.0.

[0246] Step-7: 6-bromo-5-methyl-4,5-dihydro-2H-pyrazolo [4, 3-c] quinolone (I-2h): To a stirred solution of I-2g (3.60 g, 14.3 mmol) in MeOH (20.0 mL) and water (20.0 mL) was added KOtBu (2.75 g, 24.3 mmol) and /?-toluenesulfonylhydrazide (3.99 g, 21.4 mmol) sequentially at room temperature. The reaction mixture was stirred at 70 °C for 16 h, while monitoring reaction progress by TLC. After complete consumption of starting material, it was cooled to room temperature and MeOH was removed under reduced pressure. Water (30 mL) was added to the residue and extraction was carried out using EtOAc (3 x 50 mL). The combined organic extracts were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was then stirred in pentane (30 mL), filtered and dried to afford desired compound 6-bromo-5-methyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolone I-2h (3.5 g) as an off-white solid. LCMS (ES) m z 264.0 [M+H] + , 266.0.

[0247] Step-8: 6-bromo-2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolone (I-2i) & 6-bromo-l,5-dimethyl-4,5-dihydro-lH-pyrazolo[4,3-c]quinolone (I-2j): To a stirred solution of I-2h (5.00 g, 18.9 mmol) in DMF (60.0 mL) was added CS2CO3 (12.3 g, 37.9 mmol) and iodomethane (1.42 mL, 22.7 mmol). The reaction mixture was then stirred at 80 °C for 16 h in a sealed tube. After complete consumption of starting material, it was cooled to room temperature and water (60 mL) was added to it. Extraction was carried out using EtOAc (50 mL x 3); the combined organic extracts were washed with water (50 mL x 3), brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford 4.16 g of mixture of 6-bromo-2,5-dimethyl-2H,4H,5H-pyrazolo[4,3-c]quinolone (I-2i) and 6-bromo-l,5-dimethyl- 4,5-dihydro-lH-pyrazolo[4,3-c]quinolone (I-2j). LCMS (ES) m/z 278.1 [M+H] + , 280.1.

[0248] Step-9: tert-butyl (2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6- yl)carbamate (I-2k) and tert-butyl (l,5-dimethyl-4,5-dihydro-lH-pyrazolo[4,3-c]quinolin- 6-yl)carbamate (1-21): Argon gas was purged through a stirred suspension of (I-2i + I-2j) (4.00 g, 14.4 mmol), tert-butyl carbamate (2.53 g, 21.6 mmol) and CS2CO3 (9.37 g, 28.8 mmol) in 1,4- dioxane (50.0 mL) for 15 min. To this was then added [5-(diphenylphosphanyl)-9,9-dimethyl- 9H-xanthen-4-yl]diphenylphosphane (1.66 g, 2.88 mmol) and Pd(OAc)2 (646 mg, 2.88 mmol). The reaction mixture was then stirred at 100 °C for 16 h in a sealed tube. It was then cooled to room temperature, filtered through a pad of Celite and washed with EtOAc (50 mL x 2). The filtrate was concentrated under reduced pressure and the residue was purified by Combi-Flash (using gradient elution of 0-50% EtOAc in hexane) to afford tert-butyl N-{2,5-dimethyl- 2H,4H,5H-pyrazolo[4,3-c]quinolin-6-yl}carbamate I-2k (2.0 g) (as an off-white solid) and tertbutyl (l,5-dimethyl-4,5-dihydro-lH-pyrazolo[4,3-c]quinolin-6-yl)ca rbamate 1-21 (0.8 g). LCMS (ES) TWZZ: 314.9 [M+H] + .

[0249] Step-10: 2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6-amine (1-2): To a stirred solution of I-2k (1.00 g, 3.18 mmol) in DCM (10.0 mL) was added a 4M solution HC1 in 1,4-di oxane (5.00 mL) at 0 °C. The reaction mixture was stirred at room temperature for 16 h. After completion, volatiles were removed under reduced pressure and saturated sodium bicarbonate solution (30 mL) was added to the residue. Extraction was carried out using EtOAc (3 x 30 mL); the combined organic extracts were washed with water (30 mL), brine (30 mL), dried over anhydrous Na2SO4, filtered and evaporated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-80% EtOAc in hexane) to afford desired compound 2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6-amine 1-2 (0.5 g) as semisolid. LCMS (ES) m/z 215.2 [M+H] + . 'H NMR (400 MHz, DMSO-tL) 3 7.49 (s, 1H); 6.94 - 6.80 (m, 2H); 6.57 (dd, Ji = 1.6 Hz, J 2 = 7.6 Hz, 1H); 4.87 (s, 2H); 3.93 (s, 2H); 3.83 (s, 3H); 2.34 (s, 3H). Example 3: Preparation of (S)-2,4,5-trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinox alin-

6-amine (1-3):

[0250] Step-1: tert-butyl (S)-(l-amino-l-oxopropan-2-yl)(methyl)carbamate (I-3b): To a stirred solution of I-3a (15.0 g, 73.8 mmol) in THF (250 mL) was added TEA (13.5 mL, 95.9 mmol) and ethyl chloroformate (7.38 mL, 77.5 mmol) at 0 °C. It was then stirred at 0 °C for 1 h (Part A). 100 mL of THF in a separate round bottom flask was purged with NH3 gas at 0 °C for 15 min (Part B). NH3 in THF solution (Part B) was poured into the previous reaction mixture (Part A) at 0 °C. It was then allowed to stir at room temperature for 16 h. After completion, the reaction mixture was diluted with water (200 mL) and extracted with EtOAc (3 x 100 mL). The combined organic extracts were washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting residue was purified by Combi- Flash (using gradient elution of 0-30% EtOAc in hexane) to afford desired compound tert-butyl (S)-(l-amino-l-oxopropan-2-yl)(methyl)carbamate I-3b (10.0 g, 67%) as an off-white solid. LCMS (ES) m/r. 203.2 [M+H] + .

[0251] Step-2: tert-butyl (S,E)-(l-((l-(dimethylamino)ethylidene)amino)-l-oxopropan-2- yl)(methyl)carbamate (I-3c): To a stirred solution of I-3b (10.0 g, 49.4 mmol) in 1,4-dioxane (100 mL) was added 1,1 -dimethoxy -N,N-dimethylethan-l -amine (21.7 mL, 148 mmol) at room temperature. It was then stirred at 60 °C for 2 h. After complete consumption of starting material (as indicated by TLC), the reaction mixture was diluted with water (100 mL) and extracted with EtOAc (2 x 100 mL). The combined organic extracts were washed with brine (70 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford the desired compound tert-butyl (S,E)-( 1 -(( 1 -(dimethylamino)ethylidene)amino)- 1 -oxopropan-2- yl)(methyl)carbamate I-3c (12.8 g, 95%) as a yellow oil. LCMS (ES) m/z 272.0 [M+H] + . [0252] Step-3: tert-butyl (S)-(l-(l-(3-bromo-2-fluorophenyl)-3-methyl-lH-l,2,4-triazol -5- yl)ethyl)(methyl)carbamate (I-3d): To a stirred solution of I-3c (12.6 g, 46.3 mmol) and (3- bromo-2-fluorophenyl)hydrazine (free base) (9.5 g, 46.3 mmol) in 1,4-dioxane (60 mL) was added acetic acid (70 mL) slowly at room temperature. It was then allowed to stir at 80 °C for 2 h. After complete consumption of starting material, volatiles were removed under reduced pressure and saturated NaHCOs solution (100 mL) was added to the residue. Extraction was carried out using EtOAc (2 x 100 mL); the combined organic extracts were washed with water (100 mL), brine (100 mL), dried over anhydrous ISfeSCU, filtered and evaporated under reduced pressure. The resulting crude was purified by Combi-Flash (using gradient elution of 0-30% EtOAc in hexane) to afford the desired compound tert-butyl (S)-(l-(l-(3-bromo-2- fluorophenyl)-3-methyl-lH-l,2,4-triazol-5-yl)ethyl)(methyl)c arbamate I-3d (14.5 g, 75%) as a yellow oil. LCMS (ES) m/z 413.0 [M+H] + .

[0253] Step-4: (S)-l-(l-(3-bromo-2-fluorophenyl)-3-methyl-lH-l,2,4-triazol- 5-yl)-N- methylethan-l-amine (I-3e): A 4M solution of HC1 in 1,4-di oxane (70 mL) was added to I-3d (14.5 g, 35.1 mmol) at 0 °C. Then, the reaction mixture was stirred at room temperature for 16 h. After completion, volatiles were removed under reduced pressure and dried (co-evaporation with 1,4-di oxane) to afford the desired compound (S)-l-(l-(3-bromo-2-fluorophenyl)-3-methyl- lH-l,2,4-triazol-5-yl)-N-methylethan-l-amine I-3e (12.1 g crude HC1 salt, 98%) as a pale yellow solid. LCMS (ES) m/z 313.0 [M+H] + .

[0254] Step-5: (S)-6-bromo-2,4,5-trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5- a]quinoxaline (I-3f): To a stirred solution of I-3e (12.1 g, 38.6 mmol) in 1,4-dioxane (80 mL) was added DIPEA (33.7 mL, 193 mmol) slowly at 0 °C. The reaction mixture was then allowed to stir at 80 °C for 5 h. After completion (as indicated by TLC), volatiles were removed under reduced pressure and saturated NaHCOs solution (100 mL) was added to the residue. Extraction was carried out using EtOAc (3 x 100 mL). The combined organic extracts were washed with water (100 mL) and brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-30% EtOAc in hexane) to afford the desired compound (S)-6-bromo-2,4,5-trimethyl-4,5-dihydro- [l,2,4]triazolo[l,5-a]quinoxaline I-3f (9.5 g, 83%) as a yellow oil. LCMS (ES) m/z 293.1 [M+H] + .

[0255] Step-6: tert-butyl (S)-(2,4,5-trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5- a]quinoxalin-6-yl)carbamate (I-3g): Argon gas was purged through a stirred suspension of I-3f (5.0 g, 17.1 mmol), tert-butyl carbamate (3.0 g, 25.6 mmol) and CS2CO3 (11.1 g, 34.1 mmol) in 1,4-dioxane (30 mL) for 15 min. To this was then added [5-(diphenylphosphanyl)-9,9-dimethyl- 9H-xanthen-4-yl]diphenylphosphane (0.98 g, 1.71 mmol) and Pd(OAc)2 (0.38 g, 1.71 mmol). The reaction mixture was then stirred at 100 °C for 16 h in a sealed tube. It was then cooled to room temperature, filtered through celite bed and washed with EtOAc (150 mL x 2). The filtrate was concentrated under reduced pressure and the residue was purified by Combi-Flash (using gradient elution of 0-5% MeOH in DCM) to afford tert-butyl (S)-(2,4,5-trimethyl-4,5-dihydro- [l,2,4]triazolo[l,5-a]quinoxalin-6-yl)carbamate I-3g (5.1 g, 90%) as a yellow oil. LCMS (ES) m/r. 330.2 [M+H] + .

[0256] Step-7: (S)-2,4,5-trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinox alin-6-amine (1-3): To a stirred solution of I-3g (5.1 g, 15.5 mmol) in DCM (50.0 mL) was added a 4M solution HCI in 1,4-dioxane (40.0 mL) at 0 °C. The reaction mixture was stirred at room temperature for 4 h. After completion, volatiles were removed under reduced pressure and saturated sodium bicarbonate solution (50 mL) was added to the residue. Extraction was carried out using EtOAc (3 x 50 mL); the combined organic extracts were washed with water (50 mL), brine (70 mL), dried over anhydrous Na2SO4, filtered and evaporated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-60% EtOAc in hexane) to afford desired compound (S)-2,4,5-trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinox alin-6- amine 1-3 (2.6 g, 73%) as a yellow solid. LCMS (ES) m/z 230.2 [M+H] + . 'H NMR (400 MHz, CDCh) 3 7.15 (dd, Ji = 1.2 Hz, J 2 = 8.0 Hz, 1H); 7.07 (t, J= 8.0 Hz, 1H); 6.65 (dd, Ji = 1.2 Hz, J 2 = 8.0 Hz, 1H); 4.33 (q, J= 7.2 Hz, 1H); 3.79 (br s, 2H); 2.53 (s, 3H); 2.48 (s, 3H); 1.31 (d, J = 7.2 Hz, 3H).

Example 4: Preparation of 2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6- amine (1-4):

[0257] Step-1: tert-butyl (2-amino-2-oxoethyl)(methyl)carbamate (I-4b): To a stirred solution of I-4a (15.0 g, 79.3 mmol) in THF (150 mL) was added TEA (14.5 mL, 103 mmol) and ethyl chloroformate (9.03 g, 83.2 mmol) at 0 °C. It was then stirred at 0 °C for 1 h (Part A). 150 mL of THF in a separate round bottom flask was purged with NH3 gas at 0 °C for 15 min (Part B). NH3 in THF solution was poured into the previous reaction mixture (part A) at 0 °C. It was then allowed to stir at room temperature for 16 h. After completion, the reaction mixture was diluted with water (200 mL) and extracted with EtOAc (2 x 300 mL). The combined organic extracts were washed with brine (50 mL), dried over anhydrous Na2 SO4. filtered and concentrated under reduced pressure. The resulting residue was purified by Combi-Flash (using gradient elution of 0-30% EtOAc in hexane) to afford desired compound tert-butyl (2-amino-2- oxoethyl)(methyl)carbamate I-4b (10.0 g) as an off-white solid. LCMS (ES) m/z 189.2 [M+H] + . [0258] Step-2: tert-butyl (E)-(2-((l-(dimethylamino)ethylidene)amino)-2- oxoethyl)(methyl)carbamate (I-4c): To a stirred solution of I-4b (16.0 g, 85.0 mmol) in 1,4- dioxane (160 mL) was added 1,1 -dimethoxy -N,N-dimethylethan-l -amine (37.3 mL, 255 mmol) at room temperature. It was then stirred at 60 °C for 2 h. After completion, the reaction mixture was diluted with water (200 mL) and extracted with EtOAc (2 x 200 mL). The combined organic extracts were washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford the desired compound tert-butyl (E)-(2-((l- (dimethylamino)ethylidene)amino)-2-oxoethyl)(methyl)carbamat e I-4c (20.0 g) as a yellow oil. LCMS (ES) m/r. 258.2 [M+H] + .

[0259] Step-3: tert-butyl ((l-(3-bromo-2-fluorophenyl)-3-methyl-lH-l,2,4-triazol-5- yl)methyl)(methyl)carbamate (I-4d): To a stirred solution of I-4c (11.2 g, 43.9 mmol) and (3- bromo-2-fluorophenyl)hydrazine (9.00 g, 43.9 mmol) in 1,4-di oxane (100 mL) was added acetic acid (100 mL) slowly at room temperature. It was then allowed to stir at 80 °C for 1 h. After complete consumption of starting material, volatiles were removed under reduced pressure and saturated NaHCCL solution (300 mL) was added to the residue. Extraction was carried out using EtOAc (3 x 100 mL); the combined organic extracts were washed with water (50 mL), brine (30 mL), dried over anhydrous Na2SO4, filtered and evaporated under reduced pressure. The resulting crude was purified by Combi-Flash (using gradient elution of 0-30% EtOAc in hexane) to afford the desired compound tert-butyl ((l-(3-bromo-2-fluorophenyl)-3-methyl-lH-l,2,4- triazol-5-yl)methyl)(methyl)carbamate I-4d (15.0 g) as a yellow oil. LCMS (ES) m/z 399.1 [M+H] + .

[0260] Step-4: l-(l-(3-bromo-2-fluorophenyl)-3-methyl-lH-l,2,4-triazol-5-yl )-N- methylmethanamine (I-4e): A 4M solution of HC1 in 1,4-di oxane (100 mL, 400 mmol) was added to I-4d (15.0 g, 37.6 mmol) at 0 °C and the reaction mixture was allowed to warm to room temperature over 2 h. After completion, volatiles were removed under reduced pressure and dried (co-evaporation with 1,4-di oxane) to afford the desired compound l-(l-(3-bromo-2- fluorophenyl)-3-methyl-lH-l,2,4-triazol-5-yl)-N-methylmethan amine I-4e (15.0 g) as a pale yellow solid. LCMS (ES) m/z 299.1 [M+H] + .

[0261] Step-5: 6-bromo-2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quino xaline (1-41): To a stirred solution of I-4e (11.0 g, 36.8 mmol) in 1,4-dioxane (200 mL) was added DIPEA (150 mL) slowly at 0 °C. The reaction mixture was then allowed to stir at 80 °C for 1 h. After completion, dioxane was removed under reduced pressure and saturated NaHCOs solution (100 mL) was added to the residue. Extraction was carried out using EtOAc (3 x 70 mL). The combined organic extracts were washed with water (100 mL) and brine (50 mL), dried over anhydrous Na2 SO4. filtered and concentrated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-30% EtOAc in hexane) to afford the desired compound 6-bromo-2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quino xaline I-4f (7.0 g) as a yellow oil. LCMS (ES) m/z 279.0 [M+H] + .

[0262] Step-6: tert-butyl (2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6 - yl)carbamate (I-4g): Argon gas was purged through a stirred suspension of I-4f (3.0 g, 10.7 mmol), tert-butyl carbamate (1.89 g, 16.1 mmol) and CS2CO3 (7.0 g, 21.5 mmol) in 1,4-dioxane (30 mL) for 15 min. To this was then added [5-(diphenylphosphanyl)-9,9-dimethyl-9H-xanthen- 4-yl]diphenylphosphane (0.62 g, 1.07 mmol) and Pd(OAc)2 (0.24 g, 1.07 mmol). The reaction mixture was then stirred at 100 °C for 16 h in a sealed tube. It was then cooled to room temperature, filtered through celite bed and washed with EtOAc (150 mL x 2). The filtrate was concentrated under reduced pressure and the residue was purified by Combi-Flash (using gradient elution of 0-5% MeOH in DCM) to afford tert-butyl (2,5-dimethyl-4,5-dihydro- [l,2,4]triazolo[l,5-a]quinoxalin-6-yl)carbamate I-4g (3.2 g) as a yellow oil. LCMS (ES) m/z 316.0 [M+H] + .

[0263] Step-7: 2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6- amine (1-4): To a stirred solution of I-4g (3.0 g, 9.51 mmol) in DCM (30.0 mL) was added a 4M solution of HC1 in 1,4-dioxane (60.0 mL) at 0 °C. The reaction mixture was then stirred at room temperature for 2 h. After completion, volatiles were removed under reduced pressure and saturated sodium bicarbonate solution (30 mL) was added to the residue. Extraction was carried out using EtOAc (3 x 50 mL); the combined organic extracts were washed with water (50 mL), brine (75 mL), dried over anhydrous Na2SO4, filtered and evaporated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-60% EtOAc in hexane) to afford desired compound 2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6- amine 1-4 (1.6 g) as a yellow solid. LCMS (ES) m/z 216.2 [M+H] + . 'H NMR (400 MHz, DMSO-tL) 3 6.98 (t, J= 8.0 Hz, 1H); 6.86 (dd, Ji = 1.2 Hz, J 2 = 7.6 Hz, 1H); 6.62 (dd, Ji = 1.2 Hz, J 2 = 8.0 Hz, 1H); 5.23 (s, 2H); 4.25 (s, 2H); 2.44 (s, 3H); 2.35 (s, 3H). Example 5: Preparation of 7-methyl-6,7-dihydro-5H-benzo[f| [l,2,4]triazolo[4,3- d][l,4]diazepin-8-amine (1-5):

[0264] Step-1: ethyl 3-bromo-2-fluorobenzoate (I-5b): To a stirred solution of I-5a (25.2 g, 115 mmol) in EtOH (250 mL) was added concentrated sulfuric acid (20.0 mL) slowly at room temperature and the reaction mixture was stirred at reflux temperature for 16 h. The progress of the reaction was monitored by TLC. After completion, it was cooled to room temperature and volatiles were removed under reduced pressure. Saturated NaHCOs solution (100 mL) was then slowly added to the residue and extraction was carried out using DCM (100 mL x 3). The combined organic extracts were washed with brine (100 mL), dried over anhydrous ISfeSCU, filtered and concentrated under reduced pressure to afford ethyl 3-bromo-2-fluorobenzoate I-5b (25.4 g) as a thick oil. LCMS (ES) m/r. 247.0 [M+H] + .

[0265] Step-2: 3-bromo-2-fluorobenzohydrazide (I-5c): To a stirred solution of I-5b (15.1 g, 61.1 mmol) in MeOH (40.0 mL) was added hydrazine hydrate (9.18 g, 183 mmol) at room temperature and the reaction mixture was stirred at 50 °C for 16 h. After complete consumption of starting material, it was cooled to room temperature. The resulting solid was filtered, washed with EtOH (20 mL) and dried to afford desired compound 3-bromo-2-fluorobenzohydrazide I-5c (13.1 g) as an off-white solid. LCMS (ES) m/z 233.0 [M+H] + .

[0266] Step-3: N'-(3-bromo-2-fluorobenzoyl)-N,N-dimethylformohydrazonamide (I-5d): To a stirred solution of I-5c (12.2 g, 52.4 mmol) in ACN (200 mL) was added DMF-DMA (27.8 mL, 209 mmol) at room temperature and the reaction mixture was stirred at 90 °C for 6 h. The reaction progress was monitored by TLC. After complete consumption of starting material, it was cooled to room temperature and volatiles were removed under reduced pressure. The resulting solid was stirred in Et2O (30 mL), filtered and dried to afford desired compound N'-(3- bromo-2-fluorobenzoyl)-N,N-dimethylformohydrazonarnide I-5d (10.8 g) as an off-white solid. LCMS (ES) m/r. 288.0 [M+H] + .

[0267] Step-4: tert-butyl N-{2-[3-(3-bromo-2-fluorophenyl)-4H-l,2,4-triazol-4-yl]ethyl }- N-methylcarbamate (I-5e): To a stirred solution of I-5d (3.5 g, 12.1 mmol) in ACN (50.0 mL) was added AcOH (3.7 mL, 60.7 mmol) and tert-butyl (2-aminoethyl)(methyl)carbamate (6.3 mL, 36.4 mmol) at 0 °C. The reaction mixture was then stirred at 100 °C for 16 h, while monitoring reaction progress by TLC and LCMS. After completion, it was cooled to room temperature and volatiles were removed under reduced pressure. Saturated NaHCOs solution (50 mL) was added to it and extraction was carried out using EtOAc (3 x 50 mL). The combined organic extracts were washed with water (50 mL), brine (50 mL), dried over anhydrous ISfeSCU, filtered and concentrated under reduced pressure. The resulting residue was purified by Combi- Flash (using gradient elution of 0-5% MeOH in DCM) to afford desired compound tert-butyl N- {2-[3-(3-bromo-2-fluorophenyl)-4H-l,2,4-triazol-4-yl]ethyl}- N-methylcarbamate I-5e (3.2 g) as a thick oil. LCMS (ES) m/r. 399 (M+H) + .

[0268] Step-5: {2-[3-(3-bromo-2-fluorophenyl)-4H-l,2,4-triazol-4-yl]ethyl}( methyl)amine hydrochloride (I-5f): A 4M solution of HC1 in 1,4-dioxane (20.0 mL) was added to a stirred solution of I-5e (3.2 g, 8.01 mmol) in DCM (10.0 mL) at 0 °C and the reaction mixture was allowed to warm to room temperature over 2 h. After completion (as indicated by TLC), volatiles were removed under reduced pressure and the residue was stirred in pentane (30 mL). It was then filtered and dried to afford desired compound {2-[3-(3-bromo-2-fluorophenyl)-4H- l,2,4-triazol-4-yl]ethyl}(methyl)amine hydrochloride I-5f (2.2 g, HC1 salt) as an off-white solid. LCMS (ES) m/z: 299.1 [M+H] + .

[0269] Step-6: 8-bromo-7-methyl-6,7-dihydro-5H-benzo[f] [l,2,4]triazolo[4,3- d][l,4]diazepine (I-5g): To a stirred solution of I-5f (2.2 g, 6.56 mmol) in DMF (25.0 mL) was added DIPEA (3.5 mL, 19.7 mmol) and stirred for 15 min. To this was then added K2CO3 (2.72 g, 19.7 mmol) and the reaction mixture was stirred at 80 °C for 24 h. The progress of the reaction was monitored by LCMS and TLC. After completion, it was cooled to room temperature and water (50 mL) was added to it. Extraction was carried out using EtOAc (50 mL x 3); the combined organic extracts were washed with water (30 mL x 3), brine (30 mL), dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-10% MeOH in DCM) to afford desired compound 8-bromo-7-methyl-6,7-dihydro-5H-benzo[f][l,2,4]triazolo[4,3- d][l,4]diazepine I-5g (0.6 g) as a pale yellow thick oil. LCMS (ES) m/z 279.1 [M+2H] + . 1 H NMR (400 MHz, DMSO- dd) d 8.67 (s, 1H); 8.54 (dd, Ji = 1.6 Hz, J 2 = 8.0 Hz, 1H); 7.72 (dd, Ji = 1.6 Hz, J 2 = 8.0 Hz, 1H); 7.14 (t, J= 8.0 Hz, 1H); 4.45-4.43 (m, 2H); 3.49-3.45 (m, 2H); 2.86 (s, 3H).

[0270] Step-7: tert-butyl (7-methyl-6,7-dihydro-5H-benzo[f][l,2,4]triazolo[4,3- d][l,4]diazepin-8-yl)carbamate (I-5h): I-5h (0.52 g) was synthesized by following procedure as described for the synthesis of 1-1 (step-4) using I-5g (0.75 g, 2.69 mmol) as the starting material. LCMS (ES) m/z 316.0 [M+H] + .

[0271] Step-8: 7-methyl-6,7-dihydro-5H-benzo[f][l,2,4]triazolo[4,3-d][l,4]d iazepin-8- amine (1-5): 1-5 (0.34 g) was synthesized by following procedure as described for the synthesis of 1-3 (step-7) using I-5h (0.5 g, 1.59 mmol) as the starting material. LCMS (ES) m/z 216.2 [M+H] + . X H NMR (400 MHz, DMSO-d 6 ) 3 8.56 (s, 1H); 7.69 (dd, Ji = 0.8 Hz, J 2 = 8.0 Hz, 1H); 6.91 (t, J= 8.0 Hz, 1H); 6.74 (dd, Ji = 1.6 Hz, J 2 = 8.0 Hz, 1H); 5.02 (br s, 2H); 4.40-4.37 (m, 2H); 3.43-3.38 (m, 2H); 2.59 (s, 3H).

Example 6: Preparation of 4,6-dichloro-2-methyl-l,2-dihydro-3H-pyrazolo[3,4-b]pyridin- 3- one (A-1): i) SOCI 2 , DMF

80 °C, 16 h ii) methylhydrazine 0 Cl NaOH, DCM H N Na 2 CO 3 , 1 -butanol 50°C, 2 h N 120 °C, 16 h Step-1 Cl Step-2

A-1 a A-1 b A-1

[0272] Step-1: 2,4,6-trichloro-N-methylnicotinohydrazide (A-lb): To a solution of A-la (10.0 g, 44.2 mmol) in thionyl chloride (5.0 mL) was added DMF (0.4 mL, 0.44 mmol) drop wise at 0 °C. The reaction mixture was then allowed to stir at 80 °C for 16 h. After completion, volatiles were removed under reduced pressure and the residue was dried. It was then dissolved in anhydrous DCM (50 mL) and was added to a stirred solution of methylhydrazine (2.14 mL, 40.8 mmol) and aqueous NaOH (6.53 g in 30.0 mL water, 163 mmol) in anhydrous DCM (50 mL) under nitrogen atmosphere. The reaction mixture was then stirred at 50 °C for 2 h. After completion, water (50 mL) was then added to it and the organic layer was separated. It was then washed with saturated NaHCOs solution (30 mL), brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was then purified by Combi-Flash (using gradient elution of 0-40% EtOAc in hexane) to afford desired compound 2,4,6-trichloro- N-methylnicotinohydrazide A-1 (6.5 g) as a colorless liquid. LCMS (ES) m/z 254.1 [M+H] + .

[0273] Step-2: 4,6-dichloro-2-methyl-l,2-dihydro-3H-pyrazolo[3,4-b]pyridin- 3-one (A-1): To a stirred solution of A-lb (6.5 g, 25.5 mmol) in 1-butanol (50.0 mL) was added Na2CC>3 (2.98 g, 28.1 mmol) at room temperature. The reaction mixture was then stirred at 120 °C for 16 h. After completion, volatiles were removed under reduced pressure. An aqueous solution of 2N HC1 (50 mL) was then added to it. Extraction was then carried out using 10% MeOH in DCM (25 mL x 2). The combined organic extracts were washed with water (50 mL), brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was then purified by Combi-Flash (using gradient elution of 0-10% MeOH in DCM) to afford desired compound 4,6-dichloro-2-methyl-l,2-dihydro-3H-pyrazolo[3,4-b]pyridin- 3-one A-l (2.4 g) as an off-white solid. LCMS (ES) m/z 218.0 [M+H] + .

Example 7: Preparation of N-(2-methyl-4-((2-methyl-2,4-dihydrochromeno[4,3-c]pyrazol-6 - yl)amino)-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-b]pyridin-6-yl)c yclopropanecarboxamide

(Compound 1):

[0274] Step-1: 6-chloro-2-methyl-4-((2-methyl-2,4-dihydrochromeno[4,3-c]pyr azol-6- yl)amino)-l,2-dihydro-3H-pyrazolo[3,4-b]pyridin-3-one (la): To a stirred solution of 1-1 (0.25 g, 1.24 mmol) and A-l (0.298 g, 1.37 mmol) in anhydrous THF (6.00 mL) was added a IM solution of LiHMDS (in THF) (4.97 mL, 4.97 mmol) drop wise at 0 °C. The reaction mixture was allowed to stir at room temperature for 16 h, while monitoring reaction progress by TLC. After completion, it was quenched with addition of water (30 mL) and extraction was carried out using 5% MeOH in DCM (30 mL x 3). The combined organic extracts were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting crude was purified by Combi-Flash (using gradient elution 0-3% MeOH in DCM) to afford desired compound 6-chloro-2-methyl-4-((2-methyl-2,4- dihydrochromeno[4,3-c]pyrazol-6-yl)amino)-l,2-dihydro-3H-pyr azolo[3,4-b]pyridin-3-one la (0.16 g) as an off-white solid. LCMS (ES) m/z 383.1 [M+H] + .

[0275] Step-2: N-(2-methyl-4-((2-methyl-2,4-dihydrochromeno[4,3-c]pyrazol-6 - yl)amino)-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-b]pyridin-6-yl)c yclopropanecarboxamide (Compound 1): Argon gas was purged through a stirred suspension of la (0.12 g, 0.313 mmol), cyclopropanecarboxamide (0.032 g., 0.376 mmol) and CS2CO3 (0.204 g, 0.627 mmol) in DMA (2.00 mL) for 15 min. To this was then added [5-(diphenylphosphanyl)-9,9-dimethyl-9H- xanthen-4-yl]diphenylphosphane (0.018 g, 0.031 mmol) and tris(l,5-diphenylpenta-l,4-dien-3- one)dipalladium (0.028 g, 0.031 mmol). The reaction mixture was then irradiated at 130 °C for 1 h in a MW reactor. After completion, it was cooled to room temperature and filtered through Celite bed. It was washed with EtOAc (10 mL x 2) and the filtrate was concentrated under reduced pressure. The residue was purified by preparative HPLC to afford desired compound N- (2-methyl-4-((2-methyl-2,4-dihydrochromeno[4,3-c]pyrazol-6-y l)amino)-3-oxo-2,3-dihydro-lH- pyrazolo[3,4-b]pyridin-6-yl)cyclopropanecarboxamide 1 (40 mg) as an off-white solid. . LCMS (ES) m/z 432.3 [M+H] + . 'H NMR (400 MHz, DMSO-PL) 3 10.71 (s, 1H); 8.53 (s, 1H); 7.62- 7.54 (m, 2H); 7.38-7.30 (m, 2H); 7.02 (apparent t, J= 8.0 Hz, 1H); 5.31 (s, 2H); 3.87 (s, 3H); 3.28 (s, 3H); 2.01-1.92 (m, 1H); 0.82-0.72 (m, 4H).

Example 8: Preparation of N-(4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6 - yl)amino)-2-methyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-b]pyrid in-6- yl)cyclo Compound 2

[0276] Step-1: 4,6-dichloro-2-methyl-l-((2-(trimethylsilyl)ethoxy)methyl)-l ,2-dihydro- 3H-pyrazolo[3,4-b]pyridin-3-one (2a): To a stirred solution of A-l (1.0 g, 4.59 mmol) in anhydrous DMF (20.0 mL) was added NaH (60% suspension) (0.275 g, 6.88 mmol) at 0 °C and stirred for 15 min. To this was then added [2-(chloromethoxy)ethyl]trimethylsilane (0.918 g, 5.5 mmol) dropwise at 0 °C and the reaction mixture was allowed to warm to room temperature over 2 h. After completion, ice cold water (50 mL) was added to it and extraction was carried out using EtOAc (50 mL x 2). The combined organic extracts were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-40% EtOAc in hexane) to afford desired compound 4, 6-dichl oro-2 -methyl- l-((2-(trimethylsilyl)ethoxy)methyl)-l,2-dihydro-3H- pyrazolo[3,4-b]pyri din-3 -one 2a (0.63 g) as a yellow liquid. LCMS (ES) m/z 348.1 [M+H] + . [0277] Step-2: 6-chloro-4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quin olin-6- yl)amino)-2-methyl-l-((2-(trimethylsilyl)ethoxy)methyl)-l,2- dihydro-3H-pyrazolo[3,4- b]pyridin-3-one (2b): To a stirred solution of 1-2 (0.3 g, 1.4 mmol) and 2a (0.634 g, 1.82 mmol) in anhydrous DMA (8.0 mL) was added a IM solution of LiHMDS (in THF) (5.6 mL, 5.6 mmol) drop wise at 0 °C. The reaction mixture was allowed to stir at the same temperature for 1 h, while monitoring reaction progress by TLC. After completion, it was quenched by addition of saturated NH4CI solution (25 mL) and extraction was carried out using EtOAc (25 mL x 2). The combined organic extracts were washed with brine (25 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting crude was purified by Combi-Flash (using gradient elution 0-70% EtOAc in hexane) to afford desired compound 6- chloro-4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinol in-6-yl)amino)-2-methyl-l-((2- (trimethylsilyl)ethoxy)methyl)-l,2-dihydro-3H-pyrazolo[3,4-b ]pyridin-3-one 2b (0.65 g) as a yellow viscous liquid. LCMS (ES) m/z 526.2 [M+H] + .

[0278] Step-3: N-(4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6 -yl)amino)- 2-methyl-3-oxo-l-((2-(trimethylsilyl)ethoxy)methyl)-2,3-dihy dro-lH-pyrazolo[3,4- b]pyridin-6-yl)cyclopropanecarboxamide (2c): Argon gas was purged through a stirred suspension of 2b (0.65 g, 1.24 mmol), cyclopropanecarboxamide (0.21 g, 2.47 mmol) and CS2CO3 (1.21 g, 3.71 mmol) in 1,4-dioxane (12.0 mL) for 15 min. To this was then added [5- (diphenylphosphanyl)-9,9-dimethyl-9H-xanthen-4-yl]diphenylph osphane (0.071 g, 0.124 mmol) and Pd2(dba)3 (0.113 g, 0.124 mmol). The reaction mixture was then stirred at 130 °C for 3 h in a sealed tube. The reaction progress was monitored by TLC. After complete consumption of starting material, it was cooled to room temperature and filtered through celite bed. It was washed with MeOH (50 mL x 2) and the filtrate was concentrated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-80% EtOAc in hexane) to afford desired compound N-(4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6 - yl)amino)-2-methyl-3-oxo-l-((2-(trimethylsilyl)ethoxy)methyl )-2,3-dihydro-lH-pyrazolo[3,4- b]pyridin-6-yl)cyclopropanecarboxamide 2c (0.43 g) as a yellow liquid. LCMS (ES) m/z 575.3 [M+H] + .

[0279] Step-4: N-(4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6 -yl)amino)- 2-methyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-b]pyridin-6-yl)cy clopropanecarboxamide (Compound 2): To a stirred solution of 2c (0.43 g, 0.748 mmol) in DCM (5.0 mL) was added TFA (5.00 mL) at 0 °C and the reaction was allowed to warm to room temperature over 2 h. After complete consumption of starting material, volatiles were removed under reduced pressure. To this was then added 1,4-dioxane (5.0 mL) and aqueous ammonia solution (5.0 mL). The reaction mixture was stirred at room temperature for another 1 h and then volatiles were removed under reduced pressure. The residue was purified by preparative HPLC to afford desired compound N-(4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6 -yl)amino)-2- methyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-b]pyridin-6-yl)cycl opropanecarboxamide 2 (10 mg) as an off white solid. LCMS (ES) m/z 445.2 [M+H] + . 'H NMR (400 MHz, DMSO-cL) 3 10.62 (s, 1H); 10.57 (bs, 1H); 8.97 (s, 1H); 7.80 (s, 1H); 7.52 (s, 1H); 7.32 (t, J= 7.6 Hz, 2H); 7.13 (t, J= 8.0 Hz, 1H); 4.01 (s, 2H); 3.83 (s, 3H); 2.50 (s, 3H); 2.36 (s, 3H); 1.98-1.90 (m, 1H); 0.78- 0.70 (m, 4H).

Example 9: Preparation of (S)-N-(2-methyl-3-oxo-4-((2,4,5-trimethyl-4,5-dihydro-

[l,2,4]triazolo[l,5-a]quinoxalin-6-yl)amino)-2,3-dihydro- lH-pyrazolo[3,4-b]pyridin-6- yl)cyclopropanecarboxamide (Compound 3):

Compound 3

[0280] Step-1: (S)-6-chloro-2-methyl-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4 ]triazolo[l,5- a]quinoxalin-6-yl)amino)-l-((2-(trimethylsilyl)ethoxy)methyl )-l,2-dihydro-3H- pyrazolo[3,4-b]pyridin-3-one (3a): 3a (0.6 g) was synthesized by following procedure as described for the synthesis of compound 2 (step-2) using 2a (0.53 g, 1.53 mmol) and 1-3 (0.35 g, 1.53 mmol) as the starting materials. LCMS (ES) m/z 541.3 [M+H] + .

[0281] Step-2: (S)-N-(2-methyl-3-oxo-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4 ]triazolo[l,5- a]quinoxalin-6-yl)amino)-l-((2-(trimethylsilyl)ethoxy)methyl )-2,3-dihydro-lH- pyrazolo[3,4-b]pyridin-6-yl)cyclopropanecarboxamide (3b): 3b (0.46 g) was synthesized by following procedure as described for the synthesis of compound 2 (step-3) using 3a (0.6 g, 1.11 mmol) as the starting material. LCMS (ES) m/z 590.1 [M+H] + .

[0282] Step-3: (S)-N-(2-methyl-3-oxo-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4 ]triazolo[l,5- a] quinoxalin-6-yl)amino)-2,3-dihydro-lH-pyrazolo [3,4-b] pyridin-6- yl)cyclopropanecarboxamide (Compound 3): Compound 3 (43 mg) was synthesized by following procedure as described for the synthesis of compound 2 (step-4) using 3b (0.46 g, 0.78 mmol) as the starting material. LCMS (ES) m/z 460.4 [M+H] + . 'H NMR (400 MHz, DMSO ) 3 10.75 (s, 2H); 9.06 (s, 1H); 7.85 (s, 1H); 7.44-7.40 (m, 2H); 7.35 (t, J= 8.0 Hz, 1H); 4.59 (q, J= 7.2 Hz, 1H); 2.52 (s, 6H); 2.38 (s, 3H); 2.04-1.98 (m, 1H); 1.20 (d, J= 7.2 Hz, 3H); 0.84-0.78 (m, 4H).

Example 10: Preparation of (S)-2-amino-6-(cyclopropanecarboxamido)-N-(methyl-d3)-4-

((2,4,5-trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quino xalin-6-yl)amino)nicotinamide

(Compound 4):

[0283] Step-1: methyl (S)-2-amino-6-chloro-4-((2,4,5-trimethyl-4,5-dihydro- [l,2,4]triazolo[l,5-a]quinoxalin-6-yl)amino)nicotinate (4b): To a stirred solution of 4a (2.17 g, 9.81 mmol) and 1-3 (1.5 g, 6.54 mmol) in EtOH (45 mL) was added concentrated hydrochloric acid (7 mL) at room temperature. The reaction mixture was then refluxed for 16 h. After complete consumption of starting material, volatiles were evaporated under reduced pressure and saturated NaHCOs (30 mL) was added to it. Extraction was the carried out using EtOAc (75 ml x 2); the combined organic extracts were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting crude was purified by Combi-Flash (using gradient elution 0-50% EtOAc in hexane) to afford desired compound methyl (S)-2-amino-6-chloro-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4] triazolo[l,5- a]quinoxalin-6-yl)amino)nicotinate 4b (1.1 g). LCMS (ES) m/z 414.2 [M+H] + .

[0284] Step-2: (S)-2-amino-6-chloro-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4] triazolo[l,5- a]quinoxalin-6-yl)amino)nicotinic acid (4c): To a stirred solution of 4b (1.1 g, 2.66 mmol) in THF (20 mL) was added an aqueous solution of LiOHEbO (0.22 g, 5.32 mmol, in 5 mL water) and the reaction mixture was allowed to stir at room for 16 h. After complete consumption of starting material, volatiles were removed under reduced pressure and the aqueous layer was washed with diethyl ether (30 mL). The aqueous layer was then diluted with water (20 mL) and pH was adjusted to 4 by using IN aqueous HC1 solution. The resulting precipitate was collected by filtration, washed with water (5 mL x 2) and dried to afford desired compound S)-2-amino-6- chloro-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a ]quinoxalin-6-yl)amino)nicotinic acid 4c (0.7 g) as an off white solid. LCMS (ES) m/z 400.5 [M+H] + .

[0285] Step-3: (S)-2-amino-6-chloro-N-(methyl-</3)-4-((2,4,5-trimethyl-4 ,5-dihydro- [l,2,4]triazolo[l,5-a]quinoxalin-6-yl)amino)nicotinamide (4d): To a stirred solution of 4c (0.7 g, 1.75 mmol) and methan-t/j -amine hydrochloride (0.123 g, 1.75 mmol) in DMF (14.0 mL) was added DIPEA (0.97 mL, 5.25 mmol) and HATU (1.66 g, 4.38 mmol). The reaction mixture was stirred at room temperature for 2 h. After complete consumption of starting material, water (20 mL) was added to it and extraction was carried out using EtOAc (2 x 50 mL). The combined organic extracts were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was then purified by Combi-Flash (using gradient elution 0-5% MeOH in DCM) to afford desired (S)-2-amino-6-chloro-N-(methyl-t )-4- ((2,4,5-trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinoxal in-6-yl)amino)nicotinamide 4d (0.65 g) as a brown solid. LCMS (ES) m/z 416.2 [M+H] + .

[0286] Step-4: (S)-2-amino-6-(cyclopropanecarboxamido)-N-(methyl-</3)-4- ((2,4,5- trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6-yl) amino)nicotinamide (Compound 4): Argon gas was purged through a stirred suspension of 4d (0.3 g, 0.72 mmol), cyclopropanecarboxamide (0.092 g, 1.08 mmol) and CS2CO3 (0.588 g, 1.88 mmol) in 1,4- dioxane (5.0 mL) for 15 min. To this was then added Xphos Pd-G2 (0.052 g, 0.072 mmol). The reaction mixture was then stirred at 130 °C for 16 h in a sealed tube. After complete consumption of starting material, it was cooled to room temperature and filtered through Celite bed. It was washed with MeOH (25 mL x 2) and the filtrate was concentrated under reduced pressure. The residue was purified by prep-HPLC to afford desired compound (S)-2-amino-6- (cyclopropanecarboxamido)-N-(methyl-t )-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5- a]quinoxalin-6-yl)amino)nicotinamide 4 (5 mg) as an off-white solid. LCMS (ES) m/z 465.4 [M+H] + . X H NMR (400 MHz, DMSO-tL) 3 10.20 (s, 1H); 8.91 (s, 1H); 8.09 (s, 1H); 7.48 (s, 1H); 7.26-7.17 (m, 3H); 5.67 (s, 2H); 4.52 (q, J= 7.2 Hz, 1H); 2.47 (s, 3H); 2.34 (s, 3H); 2.00- 1.94 (m, 1H), 1.14 (d, J= 6.8 Hz, 3H); 0.79-0.71 (m, 4H).

Example 11: Preparation of (5-(4-((2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5- a]quinoxalin-6-yl)amino)-6-((2,6-dimethylpyrimidin-4-yl)amin o)pyridin-3-yl)-l,3,4- oxadiazol-2-yl)methanol (Compound 5):

Compound 5

[0287] Step-1: ethyl 4,6-dichloronicotinate (5b): To a stirred solution of 5a (15 g, 78.1 mmol) in DCM (100 mL) was added oxalyl chloride (8.2 mL, 90.5 mmol) drop wise at 0 °C followed by addition of catalytic amount of DMF (0.4 mL). The reaction mixture was then allowed to warm to room temperature over 2 h. To this was then added EtOH (36 mL, 617.0 mmol) and the reaction mixture was allowed to stir at room temperature for another 2 h. After complete consumption of starting material, water (50 mL) was added to it and the organic layer was separated. It was then washed with saturated NaHCCh solution (50 mL), brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was then purified by Combi-Flash (using gradient elution of 0-7% EtOAc in hexane) to afford desired compound ethyl 4,6-dichloronicotinate 5b (8.0 g) as a pale yellow liquid. LCMS (ES) m/r. QQ [M+H] + .

[0288] Step-2: ethyl 6-chloro-4-((2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5- a]quinoxalin-6-yl)amino)nicotinate (5c): To a stirred solution of 5b (1.72 g, 7.8 mmol) and 1-4 (1.0 g, 6.5 mmol) in EtOH (25 mL) was added concentrated HC1 (0.6 mL) at room temperature. The reaction mixture was then refluxed for 16 h. After completion, volatiles were evaporated under reduced pressure and saturated NaHCCh (30 mL) was added to it. Extraction was the carried out using DCM (50 ml x 2); the combined organic extracts were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting crude was purified by Combi-Flash (using gradient elution 0-50% EtOAc in hexane) to afford desired compound ethyl 6-chloro-4-((2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5- a]quinoxalin-6-yl)amino)nicotinate 5c (0.67 g) as a pale yellow solid. LCMS (ES) m/z 399 [M+H] + .

[0289] Step-3: 6-chloro-4-((2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a] quinoxalin-6- yl)amino)nicotinohydrazide (5d): To a stirred solution of 5c (0.66 g, 1.65 mmol) in MeOH (7 mL) was added hydrazine hydrate (0.25 mL, 4.96 mmol) at room temperature. The reaction mixture was then stirred at 80 °C for 5 h. After complete consumption of starting material, the resulting solid was filtered, washed with water (10 mL) and dried. It was then stirred in Et2O (10 mL), filtered and dried to afford desired compound 6-chloro-4-((2,5-dimethyl-4,5-dihydro- [l,2,4]triazolo[l,5-a]quinoxalin-6-yl)amino)nicotinohydrazid e 5d (0.55 g) as a yellow solid. LCMS (ES) m/z: 385.5 [M+H] + .

[0290] Step-4: methyl 5-(6-chloro-4-((2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5 - a]quinoxalin-6-yl)amino)pyridin-3-yl)-l,3,4-oxadiazole-2-car boxylate (5e): To a stirred solution of 5d (0.65 g, 1.69 mmol) and triethylamine (0.715 mL, 5.07 mmol) in DCM (6 mL) was added methyl chloroglyoxylate (0.177 mL, 0.312 mmol) at 0 °C. The reaction mixture was then allowed to stir at room temperature for 4 h. To this was then added 4-toluenesulfonyl chloride (0.642 g, 3.38 mmol) and the reaction mixture was stirred at room temperature for 16 h. Volatiles were then removed under reduced pressure and water (20 mL) was added to it. Extraction was carried out using EtOAc (30 mL x 2); the combined organic extracts were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting crude was purified by Combi-Flash (using gradient elution 0-7% MeOH in DCM) to afford desired compound methyl 5-(6-chloro-4-((2,5-dimethyl-4,5-dihydro- [ 1 ,2,4]triazolo[ 1 , 5-a]quinoxalin-6-yl)amino)pyri din-3 -yl)- 1 ,3 ,4-oxadiazole-2-carboxylate 5e (0.3 g) as a yellow solid. LCMS (ES) m/z 453.0 [M+H] + .

[0291] Step-5: (5-(6-chloro-4-((2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l, 5-a]quinoxalin- 6-yl)amino)pyridin-3-yl)-l,3,4-oxadiazol-2-yl)methanol (51): To a stirred solution of 5e (0.3 g, 0.664 mmol) in THF (1.0 mL) and MeOH (1.0 mL) was added 2M solution of LiBHj in THF (1.32 mL, 2.65 mmol) drop-wise at 0 °C. The reaction mixture was then allowed to warm to room temperature over 1 h. After complete consumption of starting material, saturated aqueous NH4CI solution (20 mL) was added to it and extraction was carried out using EtOAc (50 mL x 3). The combined organic extracts were washed with brine (50 mL x 2), dried over Na2SO4, filtered and concentrated under reduced pressure. The resulting residue was purified by Combi- Flash (using gradient elution of 0-5% MeOH in DCM) to afford (5-(6-chloro-4-((2,5-dimethyl- 4,5-dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6-yl)amino)pyri din-3-yl)-l,3,4-oxadiazol-2- yl)methanol 5f (0.115 g) as an-off white solid. LCMS (ES) m/z 425.2 [M+H] + .

[0292] Step-6: (5-(4-((2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quino xalin-6- yl)amino)-6-((2,6-dimethylpyrimidin-4-yl)amino)pyridin-3-yl) -l,3,4-oxadiazol-2- yl)methanol (Compound 5): Argon gas was purged through a stirred suspension of 5f (0.115 g, 0.271 mmol), 2,6-dimethylpyrimidin-4-amine (0.033 g, 0.271 mmol) and K2CO3 (0.112 g, 0.812 mmol) in 1,4-dioxane (5.0 mL) for 15 min. To this was then added [5-(diphenylphosphanyl)-9,9- dimethyl-9H-xanthen-4-yl]diphenylphosphane (0.031 g, 0.054 mmol) and Pd2(dba)3 (0.025 g, 0.027 mmol). The reaction mixture was then irradiated at 130 °C for 2 h in a MW reactor. After completion, it was cooled to room temperature and filtered through Celite bed. It was washed with MeOH (50 mL x 2). The filtrate was concentrated under reduced pressure and the residue was purified by prep-HPLC to afford (5-(4-((2,5-dimethyl-4,5-dihydro-[l,2,4]triazolo[l,5- a]quinoxalin-6-yl)amino)-6-((2,6-dimethylpyrimidin-4-yl)amin o)pyridin-3-yl)-l,3,4-oxadiazol- 2-yl)methanol 5 (11 mg) as an off-white solid. LCMS (ES) m/z 512.4 [M+H] + . 'H NMR (400 MHz, DMSO ) 3 10.24 (s, 1H); 9.76 (s, 1H); 8.68 (s, 1H); 8.26 (s, 1H); 7.54 (dd, Ji = 6.8 Hz, J2 = 1.2 Hz, 2H); 7.36 (t, J= 8.0 Hz, 1H); 7.06 (s, 1H); 6.01 (br s, 1H); 4.76 (s, 2H); 4.45 (s 2H); 2.59 (s, 3H); 2.39 (s, 3H); 2.30 (s, 3H); 2.29 (s, 3H).

Example 12: Preparation of N-methyl-4-((7-methyl-6,7-dihydro-5H- benzo[f][l,2,4]triazolo[4,3-d][l,4]diazepin-8-yl)amino)-lH-p yrrolo[2,3-b]pyridine-5- carboxamide (Compound 6):

[0293] Step-1: ethyl 4-chloro-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-pyrrolo[2,3 - b]pyridine-5-carboxylate (6b): To a stirred solution of 6a (3.00 g, 13.4 mmol) in DMF (30.0 mL) was added sodium hydride (60% suspension) (0.481 g, 20.0 mmol) at 0 °C and stirred for 15 min. To this was then added [2-(chloromethoxy)ethyl]trimethylsilane (3.34 g, 20.0 mmol) dropwise at 0 °C and the reaction mixture was allowed to warm to room temperature over 2 h. After completion (as indicated by TLC), ice cold water (50 mL) was added to it and extraction was carried out using DCM (60 mL x 2). The combined organic extracts were washed with brine (50 mL), dried over anhydrous Na2 SO4. filtered and concentrated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-5% MeOH in DCM) to afford desired compound ethyl 4-chloro-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-pyrrolo[2,3 - b]pyridine-5-carboxylate 6b (4.5 g) as a yellow liquid. LCMS (ES) m/z 355.1 [M+H] + .

[0294] Step-2: 4-chloro-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-pyrrolo[2,3 -b]pyridine- 5-carboxylic acid (6c): To a stirred solution of 6b (5.00 g, 14.1 mmol) in EtOH (50.0 mL) was added an aqueous solution of sodium hydroxide (5.63 g, 141 mmol, in 50 mL water). The reaction mixture was stirred at room temperature for 15 min, while monitoring reaction progress by TLC. After complete consumption of starting material, EtOH was removed under reduced pressure and the aqueous layer was acidified using IN aqueous HC1 solution. The resulting solid was filtered, washed with water (5 mL x 2) and dried. It was then stirred in pentane (20 mL), filtered and dried under vacuum to afford desired compound 4-chloro-l-((2- (trimethylsilyl)ethoxy)methyl)-lH-pyrrolo[2,3-b]pyridine-5-c arboxylic acid 6c (3.2 g) as an off- white solid. LCMS (ES) m/z 327.1 [M+H] + .

[0295] Step-3: 4-chloro-N-methyl-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-py rrolo[2,3- b]pyridine-5-carboxamide (6d): To a stirred solution of 6c (0.5 g, 1.53 mmol) in anhydrous DMF (5 mL) was added CDI (1.24 g, 7.65 mmol) at room temperature and the reaction mixture was stirred for 1 h. To this was then added a 2M solution of MeNH2 in THF (0.803 mL, 1.61 mmol). The reaction mixture was stirred at room temperature for another 1 h. After complete consumption of starting material, volatiles were removed under reduced pressure and water (30 mL) was added to the residue. Extraction was carried out using EtOAc (30 mL x 2); the combined organic extracts were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-50% EtOAc in hexane) to afford desired compound 4-chloro-N- methyl-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-pyrrolo[2,3-b ]pyridine-5-carboxamide 6d (0.4 g) as an off-white solid. LCMS (ES) m/z 340.2 [M+H] + .

[0296] Step-4: N-methyl-4-((7-methyl-6,7-dihydro-5H-benzo[f] [l,2,4]triazolo[4,3-d] [1,4] diazepin-8-yl)amino)-l-((2-(trimethylsilyl)ethoxy)methyl)-lH -pyrrolo[2,3-b]pyridine-5- carboxamide (6e): Argon gas was purged through a stirred suspension of 6d (0.474 g, 1.39 mmol), 1-5 (0.250 g, 1.16 mmol) and CS2CO3 (1.14 g, 3.48 mmol) in 1,4-dioxane (5.0 mL) for 15 min. To this was then added [5-(diphenylphosphanyl)-9,9-dimethyl-9H-xanthen-4- yl]diphenylphosphane (67.2 mg, 0.116 mmol) and Pd2(dba)3 (0.106 g, 0.116 mmol). The reaction mixture was then stirred at 130 °C for 2 h in a sealed tube. It was then cooled to room temperature, filtered through cellite bed and washed with EtOAc (30 mL x 2). The combined filtrate was concentrated under reduced pressure and the residue was purified by Combi-Flash (using gradient elution of 0-70% EtOAc in hexane) to afford desired compound N-methyl-4-((7- methyl-6,7-dihydro-5H-benzo[f] [ 1 ,2,4]triazolo[4,3 -d] [ 1 ,4]diazepin-8-yl)amino)- 1 -((2- (trimethylsilyl)ethoxy)methyl)-lH-pyrrolo[2,3-b]pyridine-5-c arboxamide 6e (0.4 g) as a yellow solid. LCMS (ES) m/z 519.3 [M+H] + .

[0297] Step-5: N-methyl-4-((7-methyl-6,7-dihydro-5H-benzo[f] [l,2,4]triazolo[4,3- d][l,4]diazepin-8-yl)amino)-lH-pyrrolo[2,3-b]pyridine-5-carb oxamide (Compound 6): To a stirred solution of 6e (0.425 g, 0.819 mmol) in DCM (8 mL) was added TFA (5.00 mL) at 0 °C and the reaction was allowed to warm to room temperature over 2 h. After complete consumption of starting material (as indicated by LCMS), volatiles were removed under reduced pressure. To this was added 1,4-dioxane (8 mL) and aqueous ammonia solution (4.00 mL). The reaction mixture was stirred at room temperature for another 2 h and then volatiles were removed under reduced pressure. The residue was purified by preparative HPLC to afford desired compound N-methyl-4-((7-methyl-6,7-dihydro-5H-benzo[f][l,2,4]triazolo [4,3- d][l,4]diazepin-8-yl)amino)-lH-pyrrolo[2,3-b]pyridine-5-carb oxamide 6 (13 mg) as an off white solid. LCMS (ES) m/z 389.2 [M+H] + . 'H NMR (400 MHz, DMSO-tL) 3 11.49 (s, 1H); 10.53 (s, 1H); 8.57 (s, 1H); 8.41 (d, J= 4.4 Hz, 1H); 8.38 (s, 1H); 8.22 (dd, Ji = 1.6 Hz, J 2 = 7.6 Hz, 1H); 7.10-7.00 (m, 2H); 7.00-6.98 (m, 1H); 5.50-5.49 (m, 1H); 4.33-4.28 (m, 2H); 3.34-3.30 (m, 2H); 2.74 (s, 3H); 2.73 (s, 3H).

Example 13: Preparation of 4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6- yl)amino)-N-methyl-lH-pyrrolo[2,3-b]pyridine-5-carboxamide (Compound 7):

[0298] Step-1: 4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6-yl )amino)-N- methyl-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-pyrrolo[2,3-b ]pyridine-5-carboxamide (7a): 7a (0.35 g) was synthesized by following procedure as described for the synthesis of compound 6 (step-4) using 6d (0.3 g, 0.88 mmol) and 1-2 (0.23 g, 1.06 mmol) as the starting materials. LCMS (ES) m/z: 518.3 [M+H] + .

[0299] Step-2: 4-((2,5-dimethyl-4,5-dihydro-2H-pyrazolo[4,3-c]quinolin-6-yl )amino)-N- methyl-lH-pyrrolo[2,3-b]pyridine-5-carboxamide (7): Compound 7 (14 mg) was synthesized by following procedure as described for the synthesis of compound 6 (step-5) using 7a (0.35 g, 0.676 mmol) as the starting material. LCMS (ES) m/z 388.2 [M+H] + . 'H NMR (400 MHz, DMSO-cLh) 11.54 (s, 1H); 10.50 (s, 1H); 8.45 (d, = 4.4 Hz, 1H); 8.43 (s, 1H); 7.57 (s, 1H); 7.42 (dd, Ji = 1.2 Hz, J 2 = 7.6 Hz, 1H); 7.10-7.00 (m, 2H); 6.97-6.95 (m, 1H); 5.68-5.66 (m, 1H); 4.07 (s, 2H); 3.90 (s, 3H); 2.80 (d, J= 4.4 Hz, 3H); 2.55 (s, 3H).

Example 14: Preparation of (S)-6-(cyclopropanecarboxamido)-N-methoxy-4-((2,4,5- trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6-yl) amino)nicotinamide

(Compound 8):

[0300] Step-1: 4,6-dichloro-N-methoxynicotinamide (8a): To a stirred solution of 5a (3.0 g, 15.6 mmol) in anhydrous DCM (50 mL) was added oxalyl chloride (1.74 mL, 20.3 mmol) drop wise at 0 °C followed by addition catalytic amount of DMF (1.0 mL). The reaction mixture was then allowed to warm to room temperature over 2 h. Volatiles were then removed under reduced pressure and the residue was dried. It was then dissolved in anhydrous DCM (40 mL) and this solution was added to a stirred solution of O-methylhydroxylamine hydrochloride (1.7 g, 20.3 mmol) and DIPEA (8.19 mL, 46.9 mmol) in anhydrous DCM (25 mL) at 0 °C under nitrogen atmosphere. The reaction mixture was then stirred at room temperature for 12 h. Water (50 mL x 2) was then added to it and the organic layer was separated. It was then washed with saturated NaHCCh solution (30 mL), brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was then purified by Combi-Flash (using gradient elution of 0-4% MeOH in DCM) to afford desired compound 4,6-dichloro-N- methoxynicotinamide 8a (2.2 g) as an off-white solid. LCMS (ES) m/z 221.0 [M+H] + .

[0301] Step-2: ((S)-6-chloro-N-methoxy-4-((2,4,5-trimethyl-4,5-dihydro- [l,2,4]triazolo[l,5-a]quinoxalin-6-yl)amino)nicotinamide (8b): 8b (0.09 g) was synthesized by following procedure as described for the synthesis of compound 5 (step-2) using 8a (0.48 g, 2.18 mmol) and 1-3 (0.5 g, 2.18 mmol) as the starting materials. LCMS (ES) m/z 414.1 [M+H] + . [0302] Step-3: (S)-6-(cyclopropanecarboxamido)-N-methoxy-4-((2,4,5-trimethy l-4,5- dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6-yl)amino)nicotina mide (Compound 8): Argon gas was purged through a stirred suspension of 8b (0.09 g, 0.217 mmol), cyclopropanecarboxamide (0.022 g, 0.261 mmol) and CS2CO3 (0.142 g, 0.435 mmol) in 1,4- dioxane (5.0 mL) for 15 min. To this was then added [5-(diphenylphosphanyl)-9,9-dimethyl-9H- xanthen-4-yl]diphenylphosphane (0.012 g, 0.022 mmol) and Pd2(dba)3 (0.02 g, 0.022 mmol). The reaction mixture was then stirred at 130 °C for 3 h in a sealed tube. The reaction progress was monitored by TLC. After complete consumption of starting material, it was cooled to room temperature and filtered through celite bed. It was washed with MeOH (50 mL x 2) and the filtrate was concentrated under reduced pressure. The residue was purified by prep-HPLC to afford desired compound (S)-6-(cyclopropanecarboxamido)-N-methoxy-4-((2,4,5-trimethy l-4,5- dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6-yl)amino)nicotina mide 8 (6 mg) as an off-white solid. LCMS (ES) m/z 463.4 [M+H] + . X H NMR (400 MHz, DMSO-tL) 3 11.83 (s, 1H); 10.82 (s, 1H); 10.16 (s, 1H); 8.37 (s, 1H); 8.11 (s, 1H); 7.41-7.33 (m, 2H); 7.27 (t, J= 8.0 Hz, 1H); 4.58 (q, J= 7.2 Hz, 1H); 3.72 (s, 3H); 2.52 (s, 3H); 2.36 (s, 3H); 2.00-1.92 (m, 1H); 1.16 (d, J= 8.0 Hz, 3H); 0.80-0.70 (m, 4H).

Example 15: Preparation of 2,4,5-trimethyl-4,5-dihydro-2H-[l,2,3]ti'iazolo[4,5- c][l,7]naphthyridin-6-amine (1-6):

Step-1: l-(5-bromo-2-methyl-2H-l,2,3-triazol-4-yl)ethan-l-one (I-6b): To a stirred solution of I-6a (40 g, 166 mmol) in THF (300 mL) was added 2M solution of isopropylmagnesium chloride in THF (91.3 mL, 183 mmol) at -30 °C and stirred for 1 h at the same temperature. To this was then added DMA (77 mL, 830 mmol) at -30 °C. The reaction mixture was slowly allowed to warm to room temperature over 1 h. After completion, it was quenched with addition of saturated NH4CI solution (100 mL) and extraction was carried out using EtOAc (75 mL x 3). The combined organic extracts were washed with water (50 mL), brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting crude was purified by Combi-Flash (using gradient elution 0-10% EtOAc in heptane) to afford desired compound l-(5-bromo-2-methyl-2H-l,2,3-triazol-4-yl)ethan-l-one I-6b (19 g) as a white solid. 'H NMR (400 MHz, DMSO-d6) 8 4.23 (s, 3H), 2.59 (s, 3H).

Step-2: l-(5-bromo-2-methyl-2H-l,2,3-triazol-4-yl)-N-methylethan-l-a mine (I-6c): To a stirred solution of I-6b (17 g, 83.3 mmol) in MeOH (80 mL) was added TEA (23.2 mL, 167 mmol) and methylamine hydrochloride (11.3 g, 167 mmol) at 0 °C. The reaction mixture was stirred for 16 h at room temperature. It was then cooled to 0 °C and NaBHi (6.3 g, 167 mmol) was added to it portion-wise. The reaction mixture was allowed to warm to room temperature over 2 h. After completion (as indicated by LCMS), saturated NaHCOs solution (30 mL) was added to it and washed with EtOAc (20 mL x 2). The aqueous NaHCOs solution containing 1- (5-bromo-2-methyl-2H-l,2,3-triazol-4-yl)-N-methylethan-l-ami ne I-6c was used in the next step without further purification. LCMS (ES) m/z 219.0 [M+l] + . Step-3: tert-butyl (l-(5-(2-chloro-3-fluoropyridin-4-yl)-2-methyl-2H-l,2,3-tria zol-4- yl)ethyl)(methyl)carbamate (I-6d): A solution of (Boc)2O (35.7 mL, 155 mmol) in THF (100 mL) was added to the aqueous NaHCOs solution containing I-6c and the reaction mixture was stirred at room temperature for 16 h. After completion, volatiles were removed under reduced pressure and water (50 mL) was added to it. Extraction was carried out using EtOAc (50 mL x 2). The combined organic extracts were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by Combi- Flash (using gradient elution of 0-50% EtOAc in hexane) to afford tert-butyl (l-(5-(2-chl oro-3 - fluoropyridin-4-yl)-2-methyl-2H-l,2,3-triazol-4-yl)ethyl)(me thyl)carbamate I-6d (16.0 g) as a colorless thick oil. LCMS (ES) m/z 219.1 [(M-Boc) + 1] + .

Step-4: tert-butyl (l-(5-(2-chloro-3-fluoropyridin-4-yl)-2-methyl-2H-l,2,3-tria zol-4- yl)ethyl)(methyl)carbamate (I-6e): Argon gas was purged through a solution of I-6d (18.0 g, 56.4 mmol), (2-chloro-3-fluoropyridin-4-yl)boronic acid (24.7 g, 141.0 mmol) and CsF (25.7 g, 169 mmol) in THF (50 mL) for 15 min. To this was added tri-tert-butylphosphonium tetrafluoroborate (0.98 g, 3.38 mmol) and Pd2(dba)s (2.58 g, 2.82 mmol). The reaction mixture was then stirred at 70 °C for 16 h in a sealed tube. After completion, the reaction mixture was filtered through celite bed and washed with EtOAc (50 mL x 2). The combined filtrate was concentrated under reduced pressure and the residue was purified by Combi-Flash (using gradient elution of 0-30% EtOAc in heptane) to afford tert-butyl (l-(5-(2-chloro-3- fhjoropyridin-4-yl)-2-methyl-2H-l,2,3-triazol-4-yl)ethyl)(me thyl)carbamate I-6e (14 g) as a pale yellow gum. LCMS (ES) m/z 370.0 [M+H] + .

Step-5: 6-chloro-2,4,5-trimethyl-4,5-dihydro-2H-[l,2,3]triazolo[4,5- c][l,7]naphthyridine (I- 6f): A 4M solution of HC1 in 1,4-dioxane (140 mL) was added to I-6e (14 g, 37.9 mmol) at 0 °C and the reaction mixture was stirred at room temperature for 16 h. After completion, volatiles were removed under reduced pressure and dried (co-evaporation with 1,4-dioxane). To this was added 1,4-dioxane (60 mL) and DIPEA (31.3 mL, 180.0 mmol) at room temperature. The reaction mixture was then stirred at 80 °C for 12 h. After completion, volatiles were removed under reduced pressure and the residue was purified by Combi-Flash (using gradient elution of 0-35% EtOAc in hexane) to afford the desired compound 6-chloro-2,4,5-trimethyl-4,5-dihydro- 2H-[l,2,3]triazolo[4,5-c][l,7]naphthyridine I-6f (8 g) as an off-white solid. LCMS (ES) m/z 250.0 [M+H] + .

Step-6: N-(2,4,5-trimethyl-4,5-dihydro-2H-[l,2,3]triazolo[4,5-c][l,7 ]naphthyridin-6- yl)cyclopropanecarboxamide (I-6g): Argon gas was purged through a stirred suspension of I- 6f (8 g, 32.0 mmol), cyclopropanecarboxamide (5.45 g, 64.1 mmol) and CS2CO3 (31.3 g, 96.1 mmol) in 1,4-dioxane (80 mL) for 15 min. To this was then added [5-(diphenylphosphanyl)-9,9- dimethyl-9H-xanthen-4-yl]diphenylphosphane (1.85 g, 3.2 mmol) and Pd2(dba)3 (2.93 g, 3.2 mmol). The reaction mixture was then stirred at 130 °C for 16 h in a sealed tube. It was then cooled to room temperature, filtered through celite bed and washed with EtOAc (50 mL x 2). The filtrate was concentrated under reduced pressure and the residue was purified by Combi- Flash (using gradient elution of 0-80% EtOAc in hexane) to afford N-(2,4,5-trimethyl-4,5- dihydro-2H-[l,2,3]triazolo[4,5-c][l,7]naphthyridin-6-yl)cycl opropanecarboxamide I-6g (8 g) as a pale yellow solid. LCMS (ES) m/z 299.2 [M+H] + .

Step-7: 2,4,5-trimethyl-4,5-dihydro-2H-[l,2,3]triazolo[4,5-c][l,7]na phthyridin-6-amine (I- 6h): To a stirred solution of I-6g (9.0 g, 30.2 mmol) in THF (300 mL) was added an aqueous solution of LiOH (3.61 g, 17.6 mmol, in 100 mL water) at room temperature. It was then stirred at 50 °C for 5 h. After completion, it was cooled to room temperature and water (20 mL) was added to it. Extraction was carried out using 10% MeOH in DCM (100 mL x 2); the combined organic extracts were washed with brine (50 mL), dried over anhydrous TsfeSCU, filtered and evaporated under reduced pressure. The residue was purified by Combi-Flash (using gradient elution of 0-10% MeOH in DCM) to afford desired compound 2,4,5-trimethyl-4,5-dihydro-2H- [l,2,3]triazolo[4,5-c][l,7]naphthyridin-6-amine I-6h (4.4 g) as an off-white solid. LCMS (ES) m/z; 231.2 [M+H] + .

Note: Racemate I-6h (4.4 g) was resolved by chiral HPLC separation [Column: CHIRALCEL OJ H (250 mm x 30 mm x 5 pm); Mobile phase: n-Hexane: Ethanol with 0.1% DEA (80:20); Flow rate: 40 mL/min] to afford two enantiomers {I-6i (1.8 g): peak-1; R t ; 6.8 min and 1-6 (1.8 g): peak-

2; R t ; 9.44 min}, the desired enantiomer 1-6 was used further without confirming its absolute configuration.

Example 16: Preparation of (S)-6-(cyclopropanecarboxamido)-N-ethoxy-4-((2,4,5- trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6-yl) amino)nicotinamide

(Compound 9):

Compound 9

Step-1: 4,6-dichloro-N-ethoxynicotinamide (9a): 9a (3.6 g) was synthesized by following procedure as described for the synthesis of compound 8 (step-1) using 5a (3.0 g, 15.6 mmol) as the starting material. LCMS (ES) m/z 235.0 [M+H] + .

Step-2: (S)-6-chloro-N-ethoxy-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4 ]triazolo[l,5- a]quinoxalin-6-yl)amino)nicotinamide (9b): 9b (0.19 g) was synthesized by following procedure as described for the synthesis of compound 5 (step-2) using 9a (0.74 g, 3.14 mmol) and 1-3 (0.6 g, 2.62 mmol) as the starting materials. LCMS (ES) m/z 428.2 [M+H] + .

Step-3: (S)-6-(cyclopropanecarboxamido)-N-ethoxy-4-((2,4,5-trimethyl -4,5-dihydro- [l,2,4]triazolo[l,5-a]quinoxalin-6-yl)amino)nicotinamide (Compound 9): Compound 9

(0.065 g) was synthesized by following procedure as described for the synthesis of compound 8 (step-3) using 9b (0.18 g, 0.43 mmol) as the starting material. LCMS (ES) m/z 477.4 [M+H] + . 1 H NMR (400 MHz, DMSO-cL) 3 11.71 (s, 1H), 10.81 (s, 1H), 10.11 (s, 1H), 8.38 (s, 1H), 8.10 (s, 1H), 7.42-7.25 (m, 3H), 4.59 (q, J= 7.2 Hz, 1H), 3.96 (q, J= 7.2 Hz, 2H), 2.53 (s, 3H), 2.37

(s, 3H), 1.99-1.96 (m, 1H), 1.23 (t, J = 7.2 Hz, 3H), 1.17 (d, J = 7.2 Hz, 3H), 0.78-0.76 (m, 4H). Example 17: Preparation of (S)-6-(cyclopropanecarboxamido)-N-methoxy-4-((2,4,5- trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6-yl) amino)pyridazine-3- carboxamide (Compound 10): cyclopropanecarboxamide, Cs 2 CO 3 , Xantphos Pd 2 (dba) 3 , 1 ,4-dioxane 130 °C, 3 h

Step-3

Compound 10

Step-1: (S)-6-chloro-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4]triazolo [l,5-a]quinoxalin-6- yl)amino)pyridazine-3-carboxylic acid (10b): To a stirred solution of 1-3 (1.0 g, 4.36 mmol) and 10a (1.31g, 6.54 mmol) in anhydrous THF (20.0 mL) was added a IM solution of LiHMDS (in THF) (17.4 mL, 17.4 mmol) drop wise at 0 °C. The reaction mixture was allowed to stir at same temperature for 1 h, while monitoring reaction progress by TLC. After completion, it was quenched with addition of 10% solution of citric acid (30 mL) and extraction was carried out using 5% MeOH in DCM (30 mL x 3). The combined organic extracts were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford desired compound (S)-6-chloro-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4]triazolo [l,5- a]quinoxalin-6-yl)amino)pyridazine-3-carboxylic acid 10b (1.5 g) as an off-white solid. LCMS (ES) TWZZ: 384.1 [M-H] + .

Step-2: (S)-6-chloro-N-methoxy-4-((2,4,5-trimethyl-4,5-dihydro-[l,2, 4]triazolo[l,5- a]quinoxalin-6-yl)amino)pyridazine-3-carboxamide (10c): To a stirred solution of 10b (1.5 g, 3.89 mmol) and O-methylhydroxylamine hydrochloride (0.812 g, 9.72 mmol) in DCE (30.0 mL) at 0 °C was added DIPEA (3.6 mL, 19.4 mmol) and 50% solution of T3P (in ethyl acetate) (10.0 mL, 19.4 mmol). The reaction mixture was stirred at room temperature for 16 h. After complete consumption of starting material, water (20 mL) was added to it and extraction was carried out using 10% MeOH in DCM (2 x 50 mL). The combined organic extracts were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was then purified by Combi-Flash (using gradient elution 0-5% MeOH in DCM) to afford desired (S)-6-chloro-N-methoxy-4-((2,4,5-trimethyl-4,5-dihydro-[l,2, 4]triazolo[l,5- a]quinoxalin-6-yl)amino)pyridazine-3-carboxamide 10c (1.4 g) as a pale yellow solid. LCMS (ES) TWZZ: 415.2 [M+H] + .

Step-3: (S)-6-(cyclopropanecarboxamido)-N-methoxy-4-((2,4,5-trimethy l-4,5-dihydro- [l,2,4]triazolo[l,5-a]quinoxalin-6-yl)amino)pyridazine-3-car boxamide (Compound 10):

Compound 10 (0.072 g) was synthesized by following procedure as described for the synthesis of compound 8 (step-3) using 10c (0.4 g, 0.96 mmol) as the starting material. LCMS (ES) m/z 464.4 [M+H] + . X H NMR (400 MHz, DMSO-tL) 3 12.33 (s, 1H); 11.33 (s, 1H); 10.55 (s, 1H); 8.18 (s, 1H); 7.50-7.48 (m, 1H); 7.37-7.28 (m, 2H); 4.61 (q, = 7.2 Hz, 1H); 3.75 (s, 3H); 2.56 (s, 3H); 2.37 (s, 3H); 2.09-2.06 (m, 1H); 1.17 (d, J= 7.2 Hz, 3H); 0.83-0.80 (m, 4H).

Below compounds were synthesized by following procedures described above using appropriate intermediates and amine/amide coupling partners for Buchwald reactions.

Example 18: Preparation of (S)-6-(cyclopropanecarboxamido)-N-ethoxy-4-((2,4,5- trimethyl-4,5-dihydro-[l,2,4]triazolo[l,5-a]quinoxalin-6-yl) amino)pyridazine-3- carboxamide (Compound 13):

Step-1: (S)-6-chloro-N-ethoxy-4-((2,4,5-trimethyl-4,5-dihydro-[l,2,4 ]triazolo[l,5- a]quinoxalin-6-yl)amino)pyridazine-3-carboxamide (13a): 13a (1.5 g) was synthesized by following procedure as described for the synthesis of compound 10 (step-2) using 10b (1.6 g,

4.15 mmol) as the starting material. LCMS (ES) m/z 429.2 [M+H] + .

Step-2: (S)-6-(cyclopropanecarboxamido)-N-ethoxy-4-((2,4,5-trimethyl -4,5-dihydro- [l,2,4]triazolo[l,5-a]quinoxalin-6-yl)amino)pyridazine-3-car boxamide (Compound 13):

Compound 13 (0.065 g) was synthesized by following procedure as described for the synthesis of compound 8 (step-3) using 13a (0.35 g, 0.82 mmol) as the starting material. LCMS (ES) m/z 478.4 [M+H] + . X H NMR (400 MHz, DMSO-cL) 3 12.21 (s, 1H); 11.31 (s, 1H); 10.53 (s, 1H);

8.16 (s, 1H); 7.50-7.48 (m, 1H); 7.36-7.28 (m, 2H); 4.60 (q, J= 6.8 Hz, 1H); 3.99 (q, J= 6.8 Hz, 2H); 2.56 (s, 3H); 2.37 (s, 3H); 2.09-2.06 (m, 1H); 1.23 (t, J= 6.8 Hz, 3H); 1.17 (d, J= 7.2 Hz, 3H); 0.82-0.79 (m, 4H).

Below compounds were synthesized by following procedures described above using appropriate intermediates and amine/amide coupling partners for Buchwald reactions. Example 19: Preparation of (S)-6-(cyclopropanecarboxamido)-N-ethoxy-4-((2,4,5- trimethyl-4,5-dihydro-2H-[l,2,3]triazolo[4,5-c][l,7]naphthyr idin-6-yl)amino)pyridazine-3- carboxamide (Compound 16):

Compound 16

Step-1: (S)-6-chloro-4-((2,4,5-trimethyl-4,5-dihydro-2H-[l,2,3]triaz olo[4,5- c][l,7]naphthyridin-6-yl)amino)pyridazine-3-carboxylic acid (16a): To a stirred solution of 1-6 (1.0 g, 4.34 mmol) and 10a (1.4 g, 6.95 mmol) in anhydrous THF (30.0 mL) was added a IM solution of LiHMDS (in THF) (17.4 mL, 17.4 mmol) drop wise at 0 °C. The reaction mixture was allowed to stir at 70 °C for 2 h, while monitoring reaction progress by TLC. After completion, it was quenched with addition of 10% solution of citric acid (30 mL) and extraction was carried out using 5% MeOH in DCM (30 mL x 3). The combined organic extracts were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford desired compound (S)-6-chloro-4-((2,4,5-trimethyl-4,5-dihydro-2H-

[1.2.3]triazolo[4,5-c][l,7]naphthyridin-6-yl)amino)pyrida zine-3-carboxylic acid 16a (1.9 g) as an brown solid. LCMS (ES) m/z 385.1 [M-H] + .

Step-2: (S)-6-chloro-N-ethoxy-4-((2,4,5-trimethyl-4,5-dihydro-2H-[l, 2,3]triazolo[4,5- c][l,7]naphthyridin-6-yl)amino)pyridazine-3-carboxamide (16b): 16b (0.35 g) was synthesized by following procedure as described for the synthesis of compound 10 (step-2) using 16a (1.9 g, 4.91 mmol) as the starting material LCMS (ES) m/z 430.2 [M+H] + .

Step-3: (S)-6-(cyclopropanecarboxamido)-N-ethoxy-4-((2,4,5-trimethyl -4,5-dihydro-2H-

[1.2.3]triazolo[4,5-c][l,7]naphthyridin-6-yl)amino)pyrida zine-3-carboxamide (Compound 16): Compound 16 (0.025 g) was synthesized by following procedure as described for the synthesis of compound 8 (step-3) using 16b (0.3 g, 0.698 mmol) as the starting material. LCMS (ES) m/z 479.4 [M+H] + . 'H NMR (400 MHz, DMSO-t/ 6 ) b 12.21 (s, 1H); 11.80 (s, 1H); 11.30 (s, 1H); 9.83 (s, 1H); 8.17 (d, J= 5.2 Hz, 1H); 7.30 (m, d, J= 5.2 Hz, 1H); 4.57 (q, J = 6.8 Hz, 1H); 4.24 (s, 3H); 4.01 (q, J= 7.2 Hz, 2H); 2.66 (s, 3H); 2.16-2.10 (m, 1H); 1.24 (t, J= 7.2 Hz, 3H); 1.16 (d, J = 7.2 Hz, 3H); 0.88-0.86 (m, 4H).

II. Biological Evaluation

Example B-l: HEK-Blue™ IL-23 and IFNa/ Reporter Assays for Profiling TYK2 Pseudokinase (JH2) Inhibition

[0303] HEK-Blue™ IL-23 and IFNa/p cells with a stably-integrated cytokine receptor and STAT3 or STAT1 express STAT-inducible secreted embryonic alkaline phosphatase (SEAP) reporter gene upon cytokine stimulation. These cells are plated in DMEM (Gibco) containing 10% heat-inactivated FBS (Gibco) and 100 U/mL PenStrep (Gibco) at 37° C under 5% CO2 conditions for 20-22 hours. The cells are then pretreated with serially diluted test compounds for 60 min prior to stimulation with either 10 ng/mL human recombinant IL-23 (Miltenyl Biotech) or 1 ng/mL human recombinant IFNa (InvivoGen) for 22-24 hours for IL-23 or 16-18 h for IFNa. SEAP induction is measured using the QUANTI Blue™ Solution (InvivoGen) according to the manufacturer's instructions. Inhibition data are calculated by comparison to no inhibitor control wells for 0% inhibition and non-stimulated control wells for 100% inhibition. Dose response curves are generated to determine the concentration required to inhibit 50% of cellular response (IC50) as derived by non-linear regression analysis.

Table B-l provides TYK2 inhibitory activity of illustrative compounds, where A means IC50 < 30 nM; B means IC50 is between 30 and 300 nM; C means IC50 is between 300 and 1000 nM; D means IC50 > 1000 nM; n/a means no observed activity at 1000 nM; and n.d. means not determined.

Table B-l: Representative TYK2 Inhibitory Activity

Example B-2: HTRF-Based Selectivity Assay:

[0304] The ability of compounds to inhibit the activity of JAK1, JAK2, JAK3 and TYK2 is measured using a recombinant purified His or GST-tagged catalytic domain for each enzyme (JAK1, JAK2 and TYK2 are generated in-house; JAK3 was purchased from Cama biosciences, Cat# 08-046) in an HTRF format biochemical assay. The reactions employs a commercial peptide substrate from Cisbio (Cat# 62TK0PEC). The basic assay protocol is as follows: first, 2.5 pL of diluted compounds (4x) in DMSO are dispensed into a 384-well Optiplate. Next, 2.5 pL of enzyme (final concentrations for enzymes are: TYK2- 700 ng/mL, JAK1- 80.6 ng/mL, JAK2- 2.1 ng/mL and JAK3- 171.8 ng/mL) is added and incubated at RT for 5-20 min. Finally, 5 pl of mixture of 2X ATP [Final concentration 20 pM for TYK2, 21.43 pM for JAK1, 14.7 pM for JAK2 and 2.12 pM for JAK3] + 2X Substrate [Final concentration 217 nM for TYK2, 454.7 nM for JAK1, 200 nM for JAK2 and 257.4 nM for JAK3] is added to 384 well Optiplate. Composition of Kinase assay buffer used in the assay is as follows: HEPES 50mM, EGTA ImM, MgCh lOmM, DTT 2mM, Tween-20 0.01% and water. Then the plates are shaken and then incubated at 26.5°C for 60 min. At the end of the incubation, 10 pL of mixture of 2X detection mix [(EU3+Cryptate(lX) + Streptavidin-XL665(final concentration: 62.5 nM) (HTRF KinEASE-TK kit Cat#62TK0PEC)] is added to the assay plate, shaken and incubated at 26.5°C for 60 min. Plates are then read on a Perkin Elmer Envision for HTRF signal (665 nm reading / 615 nm reading). After normalization to untreated controls, the percent inhibition of the HTRF signal at each compound concentration is calculated. The plot of percent inhibition versus the log of compound concentration is fit with a 4-parameter dose response equation to calculate IC50 values. Table B-2 provides selectivity data of illustrative compounds across the JAK family (TYK2, JAK1, JAK2, and JAK3) at the kinase domain (JH1), where A means IC50 < 30 nM; B means IC50 is between 30 and 300 nM; C means IC50 is between 300 and 1000 nM; D means IC50 > 1000 nM; n/a means no observed activity at 1000 nM; and n.d. means not determined.

Table B-2: HTRF-Based TYK2 Selectivity Data

Example B-3: HEK-Blue™ IL-2 and IFNy Reporter Assays for determining selectivity [0305] HEK-Blue™ IL-2 and IFNy reporter cells with a stably-integrated cytokine receptor and STAT5 or STAT1 express STAT -inducible secreted embryonic alkaline phosphatase (SEAP) reporter gene upon cytokine stimulation. These cells were plated in DMEM (Gibco) containing 10% heat-inactivated FBS (Gibco) and 100 U/mL PenStrep (Gibco) at 37°C under 5% CO2 conditions for 20-22 hours. The cells were then pretreated with serially diluted test compounds for 60 min prior to stimulation with either 4 ng/mL human recombinant IL-2 (Miltenyl Biotech) or 50 ng/mL human recombinant IFNy (InvivoGen) for 24 hours. SEAP induction was measured using the QUANTI-Blue™ Solution (InvivoGen) according to the manufacturer's instructions. Inhibition data were calculated by comparison to no inhibitor control wells for 0% inhibition and non-stimulated control wells for 100% inhibition. Dose response curves were generated to determine the concentration required to inhibit 50% of cellular response (IC50) as derived by non-linear regression analysis.

[0306] Table B-3 provides selectivity data (SEAP) of illustrative compounds for IL-2 and IFN- y, where A means IC50 < 30 nM; B means IC50 is between 30 and 300 nM; C means IC50 is between 300 and 1000 nM; D means IC50 > 1000 nM; n/a means no observed activity at 1000 nM; and n.d. means not determined.

Table B-3: SEAP Selectivity Assay Data at IL-2 and IFN-y [0307] The examples and embodiments described herein are for illustrative purposes only and various modifications or changes suggested to persons skilled in the art are to be included within the spirit and purview of this application and scope of the appended claims.