TYK2 PSEUDOKINASE LIGANDS

Described herein are TYK2 pseudokinase ligands and methods of utilizing TYK2 pseudokinase ligands in the treatment of diseases, disorders or conditions. Also described herein are pharmaceutical compositions containing such compounds.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE

This application is a continuation application of U.S. patent application Ser. No. 17/090,805, filed on Nov. 5, 2020, which claims benefit of U.S. Provisional Application No. 62/933,179, filed on Nov. 8, 2019, and U.S. Provisional Application No. 63/046,514, filed on Jun. 30, 2020, each of which are herein incorporated by reference in their entirety.

BACKGROUND OF THE INVENTION

Janus kinase (JAK) is a family of intracellular, non-receptor tyrosine kinases that transduce cytokine-mediated signals via the JAK-STAT pathway. The four JAK family members are Janus kinase 1 (JAK1), Janus kinase 2 (JAK2), Janus kinase 3 (JAK3), and Tyrosine kinase 2 (TYK2) and have been shown to be key components of cytokine-mediated effects. Unlike JAK1 deficient mice, TYK2 deficient mice are viable and the TYK2 deficiency has been shown to be protective in various models of autoimmunity.

SUMMARY OF THE INVENTION

In one aspect, provided herein are compounds of Formula (I′):

  • wherein:
    • X is N or C(R6);
    • Y is C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, or

wherein C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, or C2-C9heteroaryl are optionally substituted by 1, 2, 3, or 4 R7;

    • L1 is a bond, —O—, —C(O)—, —N(R9)—, —N(R9)CH2—, or —CH2N(R9)—;
    • R1 is C3-C6cycloalkyl substituted by 1, 2, or 3 R13;
    • R2, R3, and R4 are independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • R5 is hydrogen or C1-C6alkyl;
    • each R6 is independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • each R7 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; or two R7 are combined to form a 5-, 6-, or 7-membered heterocycloalkyl ring or a 4-, 5-, or 6-membered cycloalkyl ring;
    • R8 is hydrogen, deuterium, halogen, C1-C6alkyl, or C1-C6deuteroalkyl;
    • R9 is hydrogen or C1-C6alkyl;
    • each R10 is independently selected from hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6heteroalkyl, and phenyl, wherein phenyl is optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R2)2;
    • each R11 is independently selected from C1-C6alkyl and C1-C6heteroalkyl;
    • each R12 is independently selected from hydrogen, C1-C6alkyl, and C1-C6haloalkyl;
    • each R13 is independently selected from halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; and
    • n is 0, 1, 2, 3, or 4;
    • or a pharmaceutically acceptable salt or solvate thereof.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C6-C10aryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is phenyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein n is 0, 1, or 2. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein each R7 is independently selected from halogen, C1-C6alkyl, C1-C6haloalkyl, and C1-C6alkoxy. In some embodiments is a compound of Formula I′) or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (IP), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein each R13 is independently selected from halogen, —OH, —CN, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, and oxo. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein each R13 is independently selected from halogen, —OH, and —CN. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl optionally substituted by 1 or 2 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is a bond. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)—. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are independently selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6alkyl. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6deuteroalkyl. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is hydrogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is hydrogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is hydrogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is hydrogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein X is N. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(R6). In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(H).

In another aspect, provided herein are compounds of Formula (I) or Formula (II):

  • wherein:
    • X is N or C(R6);
    • Y is C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, or

wherein C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, or C2-C9heteroaryl are optionally substituted by 1, 2, 3, or 4 R7;

    • L1 is a bond, —O—, —C(O)—, —N(R9)—, —N(R9)CH2—, or —CH2N(R9)—;
    • R1 is selected from hydrogen, deuterium, C1-C6alkyl, C1-C6deuteroalkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, and C2-C9heteroaryl, wherein C1-C6alkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl are optionally substituted by 1, 2, or 3 R7;
    • R2, R3, and R4 are independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • R5 is hydrogen or C1-C6alkyl;
    • each R6 is independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • each R7 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; or two R7 are combined to form a 5-, 6-, or 7-membered heterocycloalkyl ring or a 4-, 5-, or 6-membered cycloalkyl ring;
    • R8 is hydrogen, deuterium, halogen, C1-C6alkyl, or C1-C6deuteroalkyl;
    • R9 is hydrogen or C1-C6alkyl;
    • each R10 is independently selected from hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6heteroalkyl, and phenyl, wherein phenyl is optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R2)2;
    • each R11 is independently selected from C1-C6alkyl and C1-C6heteroalkyl;
    • each R12 is independently selected from hydrogen, C1-C6alkyl, and C1-C6haloalkyl; and
    • n is 0, 1, 2, 3, or 4;
    • or a pharmaceutically acceptable salt or solvate thereof.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof:

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof:

In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C6-C10aryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is phenyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein n is 0, 1, or 2. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein each R7 is independently selected from halogen, C1-C6alkyl, C1-C6haloalkyl, and C1-C6alkoxy. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is hydrogen, C1-C6alkyl, C1-C6deuteroalkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, and C2-C9heteroaryl, wherein C1-C6alkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl are optionally substituted by 1, 2, or 3 R7. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is hydrogen. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C1-C6alkyl optionally substituted by 1, 2, or 3 R7. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is unsubstituted C1-C6alkyl. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C3-C6cycloalkyl optionally substituted by 1, 2, or 3 R7. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is unsubstituted C3-C6cycloalkyl. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C2-C9heterocycloalkyl optionally substituted by 1, 2, or 3 R7. In some embodiments is a compound of Formula (I) or (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C2-C9heteroaryl optionally substituted by 1, 2, or 3 R7.

In another aspect, provided herein are compounds of Formula (III) or Formula (IV):

  • wherein:
    • X is N or C(R6);
    • Y is C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl, wherein C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl are optionally substituted by 1, 2, 3, or 4 R7;
    • L1 is a bond, —O—, —C(O)—, —N(R9)—, —N(R9)CH2—, or —CH2N(R9)—;
    • L2 is a bond, —N(R9)—, —O—, C1-C6alkyl, or C3-C6cycloalkyl;
    • each R1 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2; or two R1 are combined to form a 3-, 4-, 5-, or 6-membered cycloalkyl ring or a 3-, 4-, 5-, or 6-membered heterocycloalkyl ring;
    • R2, R3, and R4 are independently selected from hydrogen, deuterium, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6heteroalkyl, C3-C6cycloalkyl, and C2-C9heterocycloalkyl;
    • R5 is hydrogen or C1-C6alkyl;
    • each R6 is independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • each R7 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; or two R7 are combined to form a 5-, 6-, or 7-membered heterocycloalkyl ring or a 4-, 5-, or 6-membered cycloalkyl ring;
    • R8 is hydrogen, deuterium, halogen, C1-C6alkyl, or C1-C6deuteroalkyl;
    • R9 is hydrogen or C1-C6alkyl;
    • each R10 is independently selected from hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6heteroalkyl, and phenyl, wherein phenyl is optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2;
    • each R11 is independently selected from C1-C6alkyl and C1-C6heteroalkyl;
    • each R12 is independently selected from hydrogen, C1-C6alkyl, and C1-C6haloalkyl; and
    • p is 0, 1, 2, 3, or 4;
    • or a pharmaceutically acceptable salt or solvate thereof.

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof:

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof:

In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C6-C10aryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is phenyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, and C1-C6alkoxy. In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is selected from

In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein each R1 is hydrogen. In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein p is 1. In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein p is 2. In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein p is 0. In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is a bond. In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is —O—. In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is C1-C6alkyl. In some embodiments is a compound of Formula (III) or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is C3-C6cycloalkyl.

In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is a bond. In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)—. In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are independently selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6alkyl. In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6deuteroalkyl. In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is hydrogen. In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is hydrogen. In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is hydrogen. In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is hydrogen. In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X is N. In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(R6). In some embodiments is a compound of Formula (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(H).

In another aspect described herein is a pharmaceutical composition comprising a compound of Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient.

In another aspect described herein is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease is selected from rheumatoid arthritis, multiple sclerosis, psoriasis, lupus, intestinal bowel disease, Crohn's disease, ulcerative colitis, ankylosing spondylitis, vitiligo, and atopic dermatitis.

INCORPORATION BY REFERENCE

All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.

DETAILED DESCRIPTION OF THE INVENTION Definitions

In the context of this disclosure, a number of terms shall be utilized.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood to which the claimed subject matter belongs. In the event that there are a plurality of definitions for terms herein, those in this section prevail. All patents, patent applications, publications and published nucleotide and amino acid sequences (e.g., sequences available in GenBank or other databases) referred to herein are incorporated by reference. Where reference is made to a URL or other such identifier or address, it is understood that such identifiers can change and particular information on the internet can come and go, but equivalent information can be found by searching the internet. Reference thereto evidences the availability and public dissemination of such information.

It is to be understood that the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of any subject matter claimed. In this application, the use of the singular includes the plural unless specifically stated otherwise. It must be noted that, as used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. In this application, the use of “or” means “and/or” unless stated otherwise. Furthermore, 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.

Definition of standard chemistry terms may be found in reference works, including but not limited to, Carey and Sundberg “Advanced Organic Chemistry 4th Ed.” Vols. A (2000) and B (2001), Plenum Press, New York. Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques and pharmacology.

Unless specific definitions are provided, the nomenclature employed in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those recognized in the field. Standard techniques can be used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients. Standard techniques can be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Reactions and purification techniques can be performed e.g., using kits of manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures can be generally performed of conventional methods and as described in various general and more specific references that are cited and discussed throughout the present specification.

It is to be understood that the methods and compositions described herein are not limited to the particular methodology, protocols, cell lines, constructs, and reagents described herein and as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the methods, compounds, compositions described herein.

As used herein, C1-Cx includes C1-C2, C1-C3 . . . C1-Cx. C1-Cx refers to the number of carbon atoms that make up the moiety to which it designates (excluding optional substituents).

An “alkyl” group refers to an aliphatic hydrocarbon group. The alkyl groups may or may not include units of unsaturation. The alkyl moiety may be a “saturated alkyl” group, which means that it does not contain any units of unsaturation (i.e. a carbon-carbon double bond or a carbon-carbon triple bond). The alkyl group may also be an “unsaturated alkyl” moiety, which means that it contains at least one unit of unsaturation. The alkyl moiety, whether saturated or unsaturated, may be branched or straight chain.

The “alkyl” group may have 1 to 6 carbon atoms (whenever it appears herein, a numerical range such as “1 to 6” refers to each integer in the given range; e.g., “1 to 6 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 6 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated). The alkyl group of the compounds described herein may be designated as “C1-C6alkyl” or similar designations. By way of example only, “C1-C6alkyl” indicates that there are one to six carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from the group consisting of methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, t-butyl, n-pentyl, iso-pentyl, neo-pentyl, and hexyl. Alkyl groups can be substituted or unsubstituted. Depending on the structure, an alkyl group can be a monoradical or a diradical (i.e., an alkylene group).

An “alkoxy” refers to a “—O-alkyl” group, where alkyl is as defined herein.

The term “alkenyl” refers to a type of alkyl group in which the first two atoms of the alkyl group form a double bond that is not part of an aromatic group. That is, an alkenyl group begins with the atoms —C(R)═CR2, wherein R refers to the remaining portions of the alkenyl group, which may be the same or different. Non-limiting examples of an alkenyl group include —CH═CH2, —C(CH3)═CH2, —CH═CHCH3, —CH═C(CH3)2 and —C(CH3)═CHCH3. The alkenyl moiety may be branched, straight chain, or cyclic (in which case, it would also be known as a “cycloalkenyl” group). Alkenyl groups may have 2 to 6 carbons. Alkenyl groups can be substituted or unsubstituted. Depending on the structure, an alkenyl group can be a monoradical or a diradical (i.e., an alkenylene group).

The term “alkynyl” refers to a type of alkyl group in which the first two atoms of the alkyl group form a triple bond. That is, an alkynyl group begins with the atoms —C≡C—R, wherein R refers to the remaining portions of the alkynyl group. Non-limiting examples of an alkynyl group include —C≡CH, —C≡CCH3, —C≡CCH2CH3 and —C≡CCH2CH2CH3. The “R” portion of the alkynyl moiety may be branched, straight chain, or cyclic. An alkynyl group can have 2 to 6 carbons. Alkynyl groups can be substituted or unsubstituted. Depending on the structure, an alkynyl group can be a monoradical or a diradical (i.e., an alkynylene group).

“Amino” refers to a —NH2 group.

The term “alkylamine” or “alkylamino” refers to the —N(alkyl)xHy group, where alkyl is as defined herein and x and y are selected from the group x=1, y=1 and x=2, y=0. When x=2, the alkyl groups, taken together with the nitrogen to which they are attached, can optionally form a cyclic ring system. “Dialkylamino” refers to a —N(alkyl)2 group, where alkyl is as defined herein.

The term “aromatic” refers to a planar ring having a delocalized π-electron system containing 4n+2π electrons, where n is an integer. Aromatic rings can be formed from five, six, seven, eight, nine, or more than nine atoms. Aromatics can be optionally substituted. The term “aromatic” includes both aryl groups (e.g., phenyl, naphthalenyl) and heteroaryl groups (e.g., pyridinyl, quinolinyl).

As used herein, the term “aryl” refers to an aromatic ring wherein each of the atoms forming the ring is a carbon atom. Aryl rings can be formed by five, six, seven, eight, nine, or more than nine carbon atoms. Aryl groups can be optionally substituted. Examples of aryl groups include, but are not limited to phenyl, and naphthalenyl. Depending on the structure, an aryl group can be a monoradical or a diradical (i.e., an arylene group).

“Carboxy” refers to —CO2H. In some embodiments, carboxy moieties may be replaced with a “carboxylic acid bioisostere”, which refers to a functional group or moiety that exhibits similar physical and/or chemical properties as a carboxylic acid moiety. A carboxylic acid bioisostere has similar biological properties to that of a carboxylic acid group. A compound with a carboxylic acid moiety can have the carboxylic acid moiety exchanged with a carboxylic acid bioisostere and have similar physical and/or biological properties when compared to the carboxylic acid-containing compound. For example, in one embodiment, a carboxylic acid bioisostere would ionize at physiological pH to roughly the same extent as a carboxylic acid group. Examples of bioisosteres of a carboxylic acid include, but are not limited to,

and the like.

The term “cycloalkyl” refers to a monocyclic or polycyclic non-aromatic radical, wherein each of the atoms forming the ring (i.e. skeletal atoms) is a carbon atom. Cycloalkyls may be saturated, or partially unsaturated. Cycloalkyls may be fused with an aromatic ring (in which case the cycloalkyl is bonded through a non-aromatic ring carbon atom). Cycloalkyl groups include groups having from 3 to 10 ring atoms.

The terms “heteroaryl” or, alternatively, “heteroaromatic” refers to an aryl group that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur. An N-containing “heteroaromatic” or “heteroaryl” moiety refers to an aromatic group in which at least one of the skeletal atoms of the ring is a nitrogen atom.

A “heterocycloalkyl” group or “heteroalicyclic” group refers to a cycloalkyl group, wherein at least one skeletal ring atom is a heteroatom selected from nitrogen, oxygen and sulfur. The radicals may be fused with an aryl or heteroaryl. The term heteroalicyclic also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides. Unless otherwise noted, heterocycloalkyls have from 2 to 10 carbons in the ring. It is understood that when referring to the number of carbon atoms in a heterocycloalkyl, the number of carbon atoms in the heterocycloalkyl is not the same as the total number of atoms (including the heteroatoms) that make up the heterocycloalkyl (i.e. skeletal atoms of the heterocycloalkyl ring).

The term “halo” or, alternatively, “halogen” means fluoro, chloro, bromo and iodo.

The term “haloalkyl” refers to an alkyl group that is substituted with one or more halogens. The halogens may the same or they may be different. Non-limiting examples of haloalkyls include —CH2Cl, —CF3, —CHF2, —CH2CF3, —CF2CF3, and the like.

The terms “fluoroalkyl” and “fluoroalkoxy” include alkyl and alkoxy groups, respectively, that are substituted with one or more fluorine atoms. Non-limiting examples of fluoroalkyls include —CF3, —CHF2, —CH2F, —CH2CF3, —CF2CF3, —CF2CF2CF3, —CF(CH3)3, and the like. Non-limiting examples of fluoroalkoxy groups, include —OCF3, —OCHF2, —OCH2F, —OCH2CF3, —OCF2CF3, —OCF2CF2CF3, —OCF(CH3)2, and the like.

The term “deuteroalkyl” refers to an alkyl group that is substituted with one or more deuteriums.

The term “heteroalkyl” refers to an alkyl radical where one or more skeletal chain atoms is selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus, silicon, or combinations thereof. The heteroatom(s) may be placed at any interior position of the heteroalkyl group. Examples include, but are not limited to, —CH2—O—CH3, —CH2—CH2—O—CH3, —CH2—NH—CH3, —CH2—CH2—NH—CH3, —CH2—N(CH3)—CH3, —CH2—CH2—NH—CH3, —CH2—CH2—N(CH3)—CH3, —CH2—S—CH2—CH3, —CH2—CH2—S(O)—CH3, —CH2—CH2—S(O)2—CH3, —CH2—NH—OCH3, —CH2—O—Si(CH3)3, —CH2—CH═N—OCH3, and —CH═CH—N(CH3)—CH3. In addition, up to two heteroatoms may be consecutive, such as, by way of example, —CH2—NH—OCH3 and —CH2—O—Si(CH3)3. Excluding the number of heteroatoms, a “heteroalkyl” may have from 1 to 6 carbon atoms.

The term “bond” or “single bond” refers to a chemical bond between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure.

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.

As used herein, the substituent “R” appearing by itself and without a number designation refers to a substituent selected from among from alkyl, haloalkyl, heteroalkyl, alkenyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon), and heterocycloalkyl.

“Optional” or “optionally” means that a subsequently described event or circumstance may or may not occur and that the description includes instances when the event or circumstance occurs and instances in which it does not.

The term “optionally substituted” or “substituted” means that the referenced group may be substituted with one or more additional group(s) individually and independently selected from alkyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, —OH, alkoxy, aryloxy, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, alkylsulfone, arylsulfone, —CN, alkyne, C1-C6alkylalkyne, halo, acyl, acyloxy, —CO2H, —CO2-alkyl, nitro, haloalkyl, fluoroalkyl, and amino, including mono- and di-substituted amino groups (e.g. —NH2, —NHR, —N(R)2), and the protected derivatives thereof. By way of example, an optional substituents may be LsRs, wherein each Ls is independently selected from a bond, —O—, —C(═O)—, —S—, —S(═O)—, —S(═O)2—, —NH—, —NHC(O)—, —C(O)NH—, S(═O)2NH—, —NHS(═O)2, —OC(O)NH—, —NHC(O)O—, —(C1-C6alkyl)-, or —(C2-C6alkenyl)-; and each Rs is independently selected from among H, (C1-C6alkyl), (C3-C8cycloalkyl), aryl, heteroaryl, heterocycloalkyl, and C1-C6heteroalkyl. The protecting groups that may form the protective derivatives of the above substituents are found in sources such as Greene and Wuts, above.

As used herein, the term “about” or “approximately” means within 20%, preferably within 10%, and more preferably within 5% of a given value or range.

The term a “therapeutically effective amount” as used herein refers to the amount of a TYK2 pseudokinase ligand that, when administered to a mammal in need, is effective to at least partially ameliorate or to at least partially prevent conditions related to skin aging.

As used herein, the term “expression” includes the process by which polynucleotides are transcribed into mRNA and translated into peptides, polypeptides, or proteins.

The term “modulate” encompasses either a decrease or an increase in activity or expression depending on the target molecule.

The term “activator” is used in this specification to denote any molecular species that results in activation of the indicated receptor, regardless of whether the species itself binds to the receptor or a metabolite of the species binds to the receptor when the species is administered topically. Thus, the activator can be a ligand of the receptor or it can be an activator that is metabolized to the ligand of the receptor, i.e., a metabolite that is formed in tissue and is the actual ligand.

The term “patient” or “mammal” refers to a human, a non-human primate, canine, feline, bovine, ovine, porcine, murine, or other veterinary or laboratory mammal. Those skilled in the art recognize that a therapy which reduces the severity of a pathology in one species of mammal is predictive of the effect of the therapy on another species of mammal.

The term “soft-drug” as used herein, refers to drug substance and/or a chemical compound that is biologically active in the desired target tissue and that is metabolized, after exerting its effect in the target tissue, to a compound that is inactive against the biological target. In some embodiments, the soft-drug has no target biological activity in systemic circulation.

“Pharmaceutically acceptable salt” includes both acid and base addition salts. A pharmaceutically acceptable salt of any one of the compounds described herein is intended to encompass any and all pharmaceutically suitable salt forms. Preferred pharmaceutically acceptable salts of the compounds described herein are pharmaceutically acceptable acid addition salts, and pharmaceutically acceptable base addition salts.

“Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, hydroiodic acid, hydrofluoric acid, phosphorous acid, and the like. Also included are salts that are formed with organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc. and include, for example, acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. Exemplary salts thus include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, nitrates, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, trifluoroacetates, propionates, caprylates, isobutyrates, oxalates, malonates, succinate suberates, sebacates, fumarates, maleates, mandelates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, phthalates, benzenesulfonates, toluenesulfonates, phenylacetates, citrates, lactates, malates, tartrates, methanesulfonates, and the like. Also contemplated are salts of amino acids, such as arginates, gluconates, and galacturonates (see, for example, Berge S. M. et al., “Pharmaceutical Salts,”Journal of Pharmaceutical Science, 66:1-19 (1997)). Acid addition salts of basic compounds are prepared by contacting the free base forms with a sufficient amount of the desired acid to produce the salt.

“Pharmaceutically acceptable base addition salt” refers to those salts that retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. In some embodiments, pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts, and the like. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, for example, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, N,N-dibenzylethylenediamine, chloroprocaine, hydrabamine, choline, betaine, ethylenediamine, ethylenedianiline, N-methylglucamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like. See Berge et al., supra.

As used herein, “treatment” or “treating” or “palliating” or “ameliorating” are used interchangeably herein. These terms refer to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit. By “therapeutic benefit” is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient is still afflicted with the underlying disorder. For prophylactic benefit, the compositions are administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease has not been made.

TYK2 Pseudokinase Ligands

As a member of the JAK family of tyrosine kinases, TYK2 mediates the signaling of pro-inflammatory cytokines and therefore represents a target for treating various inflammatory and autoimmune diseases. The hallmark structural feature of the JAK family is the pseudokinase (JH2) domain immediately N-terminal to the catalytic domain (JH1). Although the JH2 domain shares the overall fold of a typical catalytic domain, a series of individual residue and conformational differences between the TYK2 JH1 and JH2 domains points to the lack of catalytic activity of the JH2 domain. The JH2 domains of the JAK family have been shown to regulate the function of the JH1 domains. The overall body of evidence is consistent with the TYK2 pseudokinase domain being auto-inhibitory, stabilizing the inactivated state of the kinase domain and that small molecule ligands can stabilize this auto-inhibitory conformation thereby preventing protein function in an allosteric manner (Moslin et al., Med. Chem. Commun., 2017, 700-712).

The compounds of Formula (I′) described herein are TYK2 pseudokinase ligands. The compounds of Formula (I′) described herein, and compositions comprising these compounds, are useful for the treatment of an inflammatory or autoimmune disease.

The urea compounds of Formula (I′) described herein have superior metabolic stability and accordingly superior in vivo exposure than the corresponding amide compounds. The improved pharmacokinetic properties of the urea compounds are attributed to the proteolytic stability of the urea functionality when compared to a simple amide. In addition, the replacement of amide with a urea changes the nature of the binding interaction with the pseudo-kinase domain of TYK2, enforcing a syn, anti-urea conformation, which in turn presents both N—H and C═O for direct, critical interactions with the back pocket of the protein. This change in binding mode leads to a different SAR of TYK2 activity and selectivity, as well as an improved pharmacokinetic and pharmacologic profile of the urea compounds versus the amide compounds.

In some embodiments, provided herein is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof:

  • wherein:
    • X is N or C(R6);
    • Y is C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, or

wherein C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, or C2-C9heteroaryl are optionally substituted by 1, 2, 3, or 4 R7;

    • L1 is a bond, —O—, —C(O)—, —N(R9)—, —N(R9)CH2—, or —CH2N(R9)—;
    • R1 is C3-C6cycloalkyl substituted by 1, 2, or 3 R13;
    • R2, R3, and R4 are independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • R5 is hydrogen or C1-C6alkyl;
    • each R6 is independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • each R7 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; or two R7 are combined to form a 5-, 6-, or 7-membered heterocycloalkyl ring or a 4-, 5-, or 6-membered cycloalkyl ring;
    • R8 is hydrogen, deuterium, halogen, C1-C6alkyl, or C1-C6deuteroalkyl;
    • R9 is hydrogen or C1-C6alkyl;
    • each R10 is independently selected from hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6heteroalkyl, and phenyl, wherein phenyl is optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2;
    • each R11 is independently selected from C1-C6alkyl and C1-C6heteroalkyl;
    • each R12 is independently selected from hydrogen, C1-C6alkyl, and C1-C6haloalkyl;
    • each R13 is independently selected from halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; and
    • n is 0, 1, 2, 3, or 4.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C6-C10aryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is phenyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, and —CN. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, and C1-C6alkoxy. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein two R7 are combined to form a 5- or 6-membered heterocycloalkyl ring. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are unsubstituted.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heterocycloalkyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C3-C6cycloalkyl optionally substituted with 1, 2, 3, or 4 R7. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, and oxo. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein two R7 are combined to form a 5- or 6-membered heterocycloalkyl ring.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(R9)—. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)—. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(R9)CH2—. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)CH2—. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —CH2N(R9)—.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —CH2N(H)—. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is a bond. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —O—. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —C(O)—.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein X is N. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(R6). In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(H).

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is C1-C6alkyl.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is hydrogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is C1-C6alkyl.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6alkyl. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are hydrogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are C1-C6alkyl.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is hydrogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is C1-C6alkyl.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is hydrogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is C1-C6alkyl.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is hydrogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is C1-C6alkyl.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is hydrogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is C1-C6alkyl.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C3-C6cycloalkyl substituted by 1, 2, or 3 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C3-C6cycloalkyl substituted by 1 or 2 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C3-C6cycloalkyl substituted by 1 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1, 2, or 3 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 or 2 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1, 2, or 3 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1 or 2 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopentyl substituted by 1, 2, or 3 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclohexyl substituted by 1, 2, or 3 R13. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein each R13 is independently selected from halogen, —OH, —CN, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, and oxo. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein each R13 is independently selected from halogen, —OH, —CN, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6alkoxy, and oxo. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein each R13 is independently selected from halogen, —OH, —CN, C1-C6alkyl, and C1-C6alkoxy. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein each R13 is independently selected from halogen, —OH, and —CN. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein each R13 is independently selected from halogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein each R13 is —OH. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein each R13 is —CN.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1, 2, or 3 R13, and each R13 is independently selected from halogen, —OH, —CN, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, and oxo. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 or 2 R13, and each R13 is independently selected from halogen, —OH, —CN, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6alkoxy, and oxo. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 R13, and R13 is selected from halogen, —OH, —CN, C1-C6alkyl, and C1-C6alkoxy. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 R13, and R13 is halogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 R13, and R13 is fluoro. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 R13, and R13 is —OH. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 R13, and R13 is —CN. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 R13, and R13 is C1-C6alkyl. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 R13, and R13 is C1-C6alkoxy. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1, 2, or 3 R13, and each R13 is independently selected from halogen, —OH, —CN, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, and oxo. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1 or 2 R13, and each R13 is independently selected from halogen, —OH, —CN, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6alkoxy, and oxo. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1 R13, and R13 is selected from halogen, —OH, —CN, C1-C6alkyl, and C1-C6alkoxy. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1 R13, and R13 is halogen. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1 R13, and R13 is fluoro. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1 R13, and R13 is —OH. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1 R13, and R13 is —CN. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1 R13, and R13 is C1-C6alkyl. In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1 R13, and R13 is C1-C6alkoxy.

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments is a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

In some embodiments, provided herein is a compound selected from:

or a pharmaceutically acceptable salt or solvate thereof.

In some embodiments, provided herein is a compound selected from:

or a pharmaceutically acceptable salt or solvate thereof.

Any combination of the groups described above for the various variables is contemplated herein. Throughout the specification, groups and substituents thereof can be chosen by one skilled in the field to provide stable moieties and compounds.

In some embodiments, the therapeutic agent(s) (e.g. compound of Formula (I′)) is present in the pharmaceutical composition as a pharmaceutically acceptable salt. In some embodiments, any compound described above is suitable for any method or composition described herein.

The compounds of Formula (I), (II), (III), or (IV) described herein are TYK2 pseudokinase ligands. The compounds of Formula (I), (II), (III), or (IV) described herein, and compositions comprising these compounds, are useful for the treatment of an inflammatory or autoimmune disease.

The urea compounds of Formula (I) or (II) described herein have superior metabolic stability and accordingly superior in vivo exposure than the corresponding amide compounds. The improved pharmacokinetic properties of the urea compounds are attributed to the proteolytic stability of the urea functionality when compared to a simple amide. In addition, the replacement of amide with a urea changes the nature of the binding interaction with the pseudo-kinase domain of TYK2, enforcing a syn, anti-urea conformation, which in turn presents both N—H and C═O for direct, critical interactions with the back pocket of the protein. This change in binding mode leads to a different SAR of TYK2 activity and selectivity, as well as an improved pharmacokinetic and pharmacologic profile of the urea compounds versus the amide compounds.

In some embodiments, provided herein is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof:

  • wherein:
    • X is N or C(R6);
    • Y is C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, or

wherein C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, or C2-C9heteroaryl are optionally substituted by 1, 2, 3, or 4 R7;

    • L1 is a bond, —O—, —C(O)—, —N(R9)—, —N(R9)CH2—, or —CH2N(R9)—;
    • R1 is selected from hydrogen, deuterium, C1-C6alkyl, C1-C6deuteroalkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, and C2-C9heteroaryl, wherein C1-C6alkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl are optionally substituted by 1, 2, or 3 R7;
    • R2, R3, and R4 are independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • R5 is hydrogen or C1-C6alkyl;
    • each R6 is independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • each R7 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; or two R7 are combined to form a 5-, 6-, or 7-membered heterocycloalkyl ring or a 4-, 5-, or 6-membered cycloalkyl ring;
    • R8 is hydrogen, deuterium, halogen, C1-C6alkyl, or C1-C6deuteroalkyl;
    • R9 is hydrogen or C1-C6alkyl;
    • each R10 is independently selected from hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6heteroalkyl, and phenyl, wherein phenyl is optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2;
    • each R11 is independently selected from C1-C6alkyl and C1-C6heteroalkyl;
    • each R12 is independently selected from hydrogen, C1-C6alkyl, and C1-C6haloalkyl; and
    • n is 0, 1, 2, 3, or 4.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C6-C10aryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is phenyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, and —CN. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, and C1-C6alkoxy. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein two R7 are combined to form a 5- or 6-membered heterocycloalkyl ring. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are unsubstituted.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heterocycloalkyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C3-C6cycloalkyl optionally substituted with 1, 2, 3, or 4 R7. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, and oxo. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein two R7 are combined to form a 5- or 6-membered heterocycloalkyl ring.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(R9)—. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)—. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(R9)CH2—. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)CH2—. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —CH2N(R9)—. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —CH2N(H)—. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is a bond. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —O—. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —C(O)—.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X is N. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(R6). In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(H).

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is C1-C6alkyl.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is hydrogen. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is C1-C6alkyl.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6alkyl. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are hydrogen. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are C1-C6alkyl.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is hydrogen. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is C1-C6alkyl.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is hydrogen. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is C1-C6alkyl.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is hydrogen. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is C1-C6alkyl.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is hydrogen. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is C1-C6alkyl.

In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is hydrogen. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C1-C6alkyl optionally substituted by 1, 2, or 3 R7. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is unsubstituted C1-C6alkyl. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C1-C6deuteroalkyl. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C3-C6cycloalkyl optionally substituted by 1, 2, or 3 R7. In some embodiments is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is unsubstituted C3-C6cycloalkyl.

In some embodiments, provided herein is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof:

  • wherein:
    • X is N or C(R6);
    • Y is C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, or

wherein C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, or C2-C9heteroaryl are optionally substituted by 1, 2, 3, or 4 R7;

    • L1 is a bond, —O—, —C(O)—, —N(R9)—, —N(R9)CH2—, or —CH2N(R9)—;
    • R1 is selected from hydrogen, deuterium, C1-C6alkyl, C1-C6deuteroalkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, and C2-C9heteroaryl, wherein C1-C6alkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl are optionally substituted by 1, 2, or 3 R7;
    • R2, R3, and R4 are independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • R5 is hydrogen or C1-C6alkyl;
    • each R6 is independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • each R7 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; or two R7 are combined to form a 5-, 6-, or 7-membered heterocycloalkyl ring or a 4-, 5-, or 6-membered cycloalkyl ring;
    • R8 is hydrogen, deuterium, halogen, C1-C6alkyl, or C1-C6deuteroalkyl;
    • R9 is hydrogen or C1-C6alkyl;
    • each R10 is independently selected from hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6heteroalkyl, and phenyl, wherein phenyl is optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2;
    • each R11 is independently selected from C1-C6alkyl and C1-C6heteroalkyl;
    • each R12 is independently selected from hydrogen, C1-C6alkyl, and C1-C6haloalkyl; and
    • n is 0, 1, 2, 3, or 4.

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C6-C10aryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is phenyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, and —CN. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, and C1-C6alkoxy. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein two R7 are combined to form a 5- or 6-membered heterocycloalkyl ring. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are unsubstituted.

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heterocycloalkyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C3-C6cycloalkyl optionally substituted with 1, 2, 3, or 4 R7. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, and oxo. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein two R7 are combined to form a 5- or 6-membered heterocycloalkyl ring.

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(R9)—. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)—. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(R9)CH2—. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)CH2—. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —CH2N(R9)—. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —CH2N(H)—. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is a bond. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —O—. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —C(O)—.

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X is N. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(R6). In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(H).

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is C1-C6alkyl.

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is hydrogen. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is C1-C6alkyl.

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6alkyl. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are hydrogen. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are C1-C6alkyl.

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is hydrogen. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is C1-C6alkyl.

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is hydrogen. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is C1-C6alkyl.

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is hydrogen. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is C1-C6alkyl.

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is hydrogen. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is C1-C6alkyl.

In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is hydrogen. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C1-C6alkyl optionally substituted by 1, 2, or 3 R7. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is unsubstituted C1-C6alkyl. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C1-C6deuteroalkyl. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C3-C6cycloalkyl optionally substituted by 1, 2, or 3 R7. In some embodiments is a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is unsubstituted C3-C6cycloalkyl.

In some embodiments, provided herein is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof:

  • wherein:
    • X is N or C(R6);
    • Y is C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl, wherein C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl are optionally substituted by 1, 2, 3, or 4 R7;
    • L1 is a bond, —O—, —C(O)—, —N(R9)—, —N(R9)CH2—, or —CH2N(R9)—;
    • L2 is a bond, —N(R9)—, —O—, C1-C6alkyl, or C3-C6cycloalkyl;
    • each R1 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)Rn, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2; or two R1 are combined to form a 3-, 4-, 5-, or 6-membered cycloalkyl ring or a 3-, 4-, 5-, or 6-membered heterocycloalkyl ring;
    • R2, R3, and R4 are independently selected from hydrogen, deuterium, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6heteroalkyl, C3-C6cycloalkyl, and C2-C9heterocycloalkyl;
    • R5 is hydrogen or C1-C6alkyl;
    • each R6 is independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • each R7 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; or two R7 are combined to form a 5-, 6-, or 7-membered heterocycloalkyl ring or a 4-, 5-, or 6-membered cycloalkyl ring;
    • R8 is hydrogen, deuterium, halogen, C1-C6alkyl, or C1-C6deuteroalkyl;
    • R9 is hydrogen or C1-C6alkyl;
    • each R10 is independently selected from hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6heteroalkyl, and phenyl, wherein phenyl is optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2;
    • each R11 is independently selected from C1-C6alkyl and C1-C6heteroalkyl;
    • each R12 is independently selected from hydrogen, C1-C6alkyl, and C1-C6haloalkyl; and
    • p is 0, 1, 2, 3, or 4.

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C6-C10aryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is phenyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is unsubstituted phenyl. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, and —CN. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from:

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heterocycloalkyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C3-C6cycloalkyl optionally substituted with 1, 2, 3, or 4 R7. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, and oxo. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heterocycloalkyl selected from:

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(R9)—. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)—. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(R9)CH2—. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)CH2—. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —CH2N(R9)—. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —CH2N(H)—. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is a bond. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —O—. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —C(O)—.

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is —N(R9)—. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is —N(H)—. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is a bond. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is —O—. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is C1-C6alkyl. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is C3-C6cycloalkyl.

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X is N. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(R6). In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(H).

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is C1-C6alkyl.

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is hydrogen. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is C1-C6alkyl.

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6alkyl. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are hydrogen. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are C1-C6alkyl.

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is hydrogen. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is C1-C6alkyl.

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is hydrogen. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is C1-C6alkyl.

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is hydrogen. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is C1-C6alkyl.

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is hydrogen. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is C1-C6alkyl.

In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein each R1 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein each R1 is hydrogen. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein two R1 are combined to form a 3-, 4-, 5-, or 6-membered cycloalkyl ring or a 3-, 4-, 5-, or 6-membered heterocycloalkyl ring. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein p is 0. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein p is 1. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein p is 2. In some embodiments is a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof, wherein p is 3.

In some embodiments, provided herein is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof:

  • wherein:
    • X is N or C(R6);
    • Y is C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl, wherein C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl are optionally substituted by 1, 2, 3, or 4 R7;
    • L1 is a bond, —O—, —C(O)—, —N(R9)—, —N(R9)CH2—, or —CH2N(R9)—;
    • L2 is a bond, —N(R9)—, —O—, C1-C6alkyl, or C3-C6cycloalkyl;
    • each R1 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2; or two R1 are combined to form a 3-, 4-, 5-, or 6-membered cycloalkyl ring or a 3-, 4-, 5-, or 6-membered heterocycloalkyl ring;
    • R2, R3, and R4 are independently selected from hydrogen, deuterium, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6heteroalkyl, C3-C6cycloalkyl, and C2-C9heterocycloalkyl;
    • R5 is hydrogen or C1-C6alkyl;
    • each R6 is independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
    • each R7 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; or two R7 are combined to form a 5-, 6-, or 7-membered heterocycloalkyl ring or a 4-, 5-, or 6-membered cycloalkyl ring;
    • R8 is hydrogen, deuterium, halogen, C1-C6alkyl, or C1-C6deuteroalkyl;
    • R9 is hydrogen or C1-C6alkyl;
    • each R10 is independently selected from hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6heteroalkyl, and phenyl, wherein phenyl is optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2;
    • each Ru is independently selected from C1-C6alkyl and C1-C6heteroalkyl;
    • each R12 is independently selected from hydrogen, C1-C6alkyl, and C1-C6haloalkyl; and
    • p is 0, 1, 2, 3, or 4.

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C6-C10aryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is phenyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is unsubstituted phenyl. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, and —CN. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from:

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heterocycloalkyl optionally substituted with 1, 2, or 3 R7. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C3-C6cycloalkyl optionally substituted with 1, 2, 3, or 4 R7. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, and oxo. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heterocycloalkyl selected from:

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(R9)—. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)—. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(R9)CH2—. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)CH2—. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —CH2N(R9)—. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —CH2N(H)—. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is a bond. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —O—. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —C(O)—.

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is —N(R9)—. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is —N(H)—. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is a bond. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is —O—. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is C1-C6alkyl. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein L2 is C3-C6cycloalkyl.

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X is N. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(R6). In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(H).

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is C1-C6alkyl.

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is hydrogen. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R4 is C1-C6alkyl.

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6alkyl. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are hydrogen. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are C1-C6alkyl.

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is hydrogen. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is C1-C6alkyl.

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is hydrogen. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is C1-C6alkyl.

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is hydrogen. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is C1-C6alkyl.

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is hydrogen. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is C1-C6alkyl.

In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein each R1 is selected from hydrogen and C1-C6alkyl. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein each R1 is hydrogen. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein two R1 are combined to form a 3-, 4-, 5-, or 6-membered cycloalkyl ring or a 3-, 4-, 5-, or 6-membered heterocycloalkyl ring. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein p is 0. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein p is 1. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein p is 2. In some embodiments is a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein p is 3.

In some embodiments, provided herein is a compound selected from:

or a pharmaceutically acceptable salt or solvate thereof.

In some embodiments, provided herein is a compound selected from:

or a pharmaceutically acceptable salt or solvate thereof.

Any combination of the groups described above for the various variables is contemplated herein. Throughout the specification, groups and substituents thereof can be chosen by one skilled in the field to provide stable moieties and compounds.

In some embodiments, the therapeutic agent(s) (e.g. compound of Formula (I), (II), (III), or (IV)) is present in the pharmaceutical composition as a pharmaceutically acceptable salt. In some embodiments, any compound described above is suitable for any method or composition described herein.

Further Forms of Compounds Disclosed Herein Isomers

Furthermore, in some embodiments, the compounds described herein exist as geometric isomers. In some embodiments, the compounds described herein possess one or more double bonds. The compounds presented herein include all cis, trans, syn, anti, entgegen (E), and zusammen (Z) isomers as well as the corresponding mixtures thereof. In some situations, compounds exist as tautomers. The compounds described herein include all possible tautomers within the formulas described herein. In some situations, the compounds described herein possess one or more chiral centers and each center exists in the R configuration or S configuration. The compounds described herein include all diastereomeric, enantiomeric, and epimeric forms as well as the corresponding mixtures thereof. In additional embodiments of the compounds and methods provided herein, mixtures of enantiomers and/or diastereoisomers, resulting from a single preparative step, combination, or interconversion, are useful for the applications described herein. In some embodiments, the compounds described herein are prepared as optically pure enantiomers by chiral chromatographic resolution of the racemic mixture. In some embodiments, the compounds described herein are prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers, and recovering the optically pure enantiomers. In some embodiments, dissociable complexes are preferred (e.g., crystalline diastereomeric salts). In some embodiments, the diastereomers have distinct physical properties (e.g., melting points, boiling points, solubilities, reactivity, etc.) and are separated by taking advantage of these dissimilarities. In some embodiments, the diastereomers are separated by chiral chromatography, or preferably, by separation/resolution techniques based upon differences in solubility. In some embodiments, the optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that does not result in racemization.

Labeled Compounds

In some embodiments, the compounds described herein exist in their isotopically-labeled forms. In some embodiments, the methods disclosed herein include methods of treating diseases by administering such isotopically-labeled compounds. In some embodiments, the methods disclosed herein include methods of treating diseases by administering such isotopically-labeled compounds as pharmaceutical compositions. Thus, in some embodiments, the compounds disclosed herein include isotopically-labeled compounds, which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that are incorporated into compounds described herein include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine, and chloride, such as 2H, 3H, 13C, 14C, 15N, 17O, 18O, 31P, 32P 35S, 18F, and 36Cl, respectively. Compounds described herein, and pharmaceutically acceptable salts, esters, solvate, hydrates, or derivatives thereof which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically-labeled compounds, for example those into which radioactive isotopes such as 3H and 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i. e., 3H and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavy isotopes such as deuterium, i.e., 2H, produces certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements. In some embodiments, the isotopically labeled compounds, pharmaceutically acceptable salt, ester, solvate, hydrate, or derivative thereof is prepared by any suitable method.

In some embodiments, the compounds described herein are labeled by other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.

Pharmaceutically Acceptable Salts

In some embodiments, the compounds described herein exist as their pharmaceutically acceptable salts. In some embodiments, the methods disclosed herein include methods of treating diseases by administering such pharmaceutically acceptable salts. In some embodiments, the methods disclosed herein include methods of treating diseases by administering such pharmaceutically acceptable salts as pharmaceutical compositions.

In some embodiments, the compounds described herein possess acidic or basic groups and therefore react with any of a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt. In some embodiments, these salts are prepared in situ during the final isolation and purification of the compounds described herein, or by separately reacting a purified compound in its free form with a suitable acid or base, and isolating the salt thus formed.

Solvates

In some embodiments, the compounds described herein exist as solvates. In some embodiments are methods of treating diseases by administering such solvates. Further described herein are methods of treating diseases by administering such solvates as pharmaceutical compositions.

Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and, in some embodiments, 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 the compounds described herein are conveniently prepared or formed during the processes described herein. By way of example only, hydrates of the compounds described herein are conveniently prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents including, but not limited to, dioxane, tetrahydrofuran, or MeOH. In addition, the compounds provided herein exist in unsolvated as well as solvated forms. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the compounds and methods provided herein.

Synthesis of Compounds

In some embodiments, the synthesis of compounds described herein are accomplished using means described in the chemical literature, using the methods described herein, or by a combination thereof. In addition, solvents, temperatures and other reaction conditions presented herein may vary.

In other embodiments, the starting materials and reagents used for the synthesis of the compounds described herein are synthesized or are obtained from commercial sources, such as, but not limited to, Sigma-Aldrich, FischerScientific (Fischer Chemicals), and AcrosOrganics.

In further embodiments, the compounds described herein, and other related compounds having different substituents are synthesized using techniques and materials described herein as well as those that are recognized in the field, such as described, for example, in Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989), March, Advanced Organic Chemistry 4th Ed., (Wiley 1992); Carey and Sundberg, Advanced Organic Chemistry 4th Ed., Vols. A and B (Plenum 2000, 2001), and Green and Wuts, Protective Groups in Organic Synthesis 3rd Ed., (Wiley 1999) (all of which are incorporated by reference for such disclosure). General methods for the preparation of compound as disclosed herein may be derived from reactions and the reactions may be modified by the use of appropriate reagents and conditions, for the introduction of the various moieties found in the formulae as provided herein. As a guide the following synthetic methods may be utilized.

Use of Protecting Groups

In the reactions described, it may be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, in order to avoid their unwanted participation in reactions. Protecting groups are used to block some or all of the reactive moieties and prevent such groups from participating in chemical reactions until the protective group is removed. It is preferred that each protective group be removable by a different means. Protective groups that are cleaved under totally disparate reaction conditions fulfill the requirement of differential removal.

Protective groups can be removed by acid, base, reducing conditions (such as, for example, hydrogenolysis), and/or oxidative conditions. Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and may be used to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis, and Fmoc groups, which are base labile. Carboxylic acid and hydroxy reactive moieties may be blocked with base labile groups such as, but not limited to, methyl, ethyl, and acetyl in the presence of amines blocked with acid labile groups such as t-butyl carbamate or with carbamates that are both acid and base stable but hydrolytically removable.

Carboxylic acid and hydroxy reactive moieties may also be blocked with hydrolytically removable protective groups such as the benzyl group, while amine groups capable of hydrogen bonding with acids may be blocked with base labile groups such as Fmoc. Carboxylic acid reactive moieties may be protected by conversion to simple ester compounds as exemplified herein, which include conversion to alkyl esters, or they may be blocked with oxidatively-removable protective groups such as 2,4-dimethoxybenzyl, while co-existing amino groups may be blocked with fluoride labile silyl carbamates.

Allyl blocking groups are useful in the presence of acid- and base-protecting groups since the former are stable and can be subsequently removed by metal or pi-acid catalysts. For example, an allyl-blocked carboxylic acid can be deprotected with a Pd0-catalyzed reaction in the presence of acid labile t-butyl carbamate or base-labile acetate amine protecting groups. Yet another form of protecting group is a resin to which a compound or intermediate may be attached. As long as the residue is attached to the resin, that functional group is blocked and cannot react. Once released from the resin, the functional group is available to react.

Typically blocking/protecting groups may be selected from:

Other protecting groups, plus a detailed description of techniques applicable to the creation of protecting groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999, and Kocienski, Protective Groups, Thieme Verlag, New York, NY, 1994, which are incorporated herein by reference for such disclosure).

Methods of Treatment and Prevention

In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a method of treating an inflammatory disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a method of treating an autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease, disorder, or condition is selected from rheumatoid arthritis, multiple sclerosis, psoriasis, lupus, intestinal bowel disease, Crohn's disease, ulcerative colitis, ankylosing spondylitis, vitiligo, and atopic dermatitis. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease, disorder, or condition is rheumatoid arthritis. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease, disorder, or condition is multiple sclerosis. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease, disorder, or condition is psoriasis. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease, disorder, or condition is lupus. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease, disorder, or condition is intestinal bowel disease. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease, disorder, or condition is Crohn's disease. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease, disorder, or condition is ulcerative colitis. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease, disorder, or condition is ankylosing spondylitis. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease, disorder, or condition is vitiligo. In some embodiments is a method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof, wherein the disease, disorder, or condition is atopic dermatitis.

Pharmaceutical Compositions and Methods of Administration

TYK2 pseudokinase ligands described herein are administered to subjects in a biologically compatible form suitable for administration to treat or prevent diseases, disorders or conditions. Administration of TYK2 pseudokinase ligands as described herein can be in any pharmacological form including a therapeutically effective amount of a TYK2 pseudokinase ligand alone or in combination with a pharmaceutically acceptable carrier.

In certain embodiments, the compounds described herein are administered as a pure chemical. In other embodiments, the compounds described herein are combined with a pharmaceutically suitable or acceptable carrier (also referred to herein as a pharmaceutically suitable (or acceptable) excipient, physiologically suitable (or acceptable) excipient, or physiologically suitable (or acceptable) carrier) selected on the basis of a chosen route of administration and standard pharmaceutical practice as described, for example, in Remington: The Science and Practice of Pharmacy (Gennaro, 21st Ed. Mack Pub. Co., Easton, PA (2005)).

Accordingly, provided herein is a pharmaceutical composition comprising at least one compound described herein, or a pharmaceutically acceptable salt, together with one or more pharmaceutically acceptable carriers. The carrier(s) (or excipient(s)) is acceptable or suitable if the carrier is compatible with the other ingredients of the composition and not deleterious to the recipient (i.e., the subject) of the composition.

In some embodiments is a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (I′), (I), (II), (III), or (IV), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof.

Another embodiment provides a pharmaceutical composition consisting essentially of a pharmaceutically acceptable carrier and a compound of Formula (I′), (I), (II), (III), or (IV) or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a pharmaceutical composition consisting essentially of a pharmaceutically acceptable carrier and a compound of Formula (I′), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a pharmaceutical composition consisting essentially of a pharmaceutically acceptable carrier and a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a pharmaceutical composition consisting essentially of a pharmaceutically acceptable carrier and a compound of Formula (II), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a pharmaceutical composition consisting essentially of a pharmaceutically acceptable carrier and a compound of Formula (III), or a pharmaceutically acceptable salt or solvate thereof. In some embodiments is a pharmaceutical composition consisting essentially of a pharmaceutically acceptable carrier and a compound of Formula (IV), or a pharmaceutically acceptable salt or solvate thereof.

In certain embodiments, the compound as described herein is substantially pure, in that it contains less than about 5%, or less than about 1%, or less than about 0.1%, of other organic small molecules, such as contaminating intermediates or by-products that are created, for example, in one or more of the steps of a synthesis method.

These formulations include those suitable for oral, topical, buccal, parenteral (e.g., subcutaneous, intramuscular, intradermal, or intravenous), or aerosol administration.

Exemplary pharmaceutical compositions are used in the form of a pharmaceutical preparation, for example, in solid, semisolid or liquid form, which includes one or more of a disclosed compound, as an active ingredient, in a mixture with an organic or inorganic carrier or excipient suitable for external, enteral or parenteral applications. In some embodiments, the active ingredient is compounded, for example, with the usual non-toxic, pharmaceutically acceptable carriers for tablets, pellets, capsules, suppositories, solutions, emulsions, suspensions, and any other form suitable for use. The active object compound is included in the pharmaceutical composition in an amount sufficient to produce the desired effect upon the process or condition of the disease.

In some embodiments, TYK2 pseudokinase ligands described herein are administered to subjects in a biologically compatible form suitable for topical administration to treat or prevent dermal diseases, disorders or conditions. By “biologically compatible form suitable for topical administration” is meant a form of the TYK2 pseudokinase ligand to be administered in which any toxic effects are outweighed by the therapeutic effects of the inhibitor. Administration of TYK2 pseudokinase ligands as described herein can be in any pharmacological form including a therapeutically effective amount of a TYK2 pseudokinase ligand alone or in combination with a pharmaceutically acceptable carrier.

Topical administration of a TYK2 pseudokinase ligand may be presented in the form of an aerosol, a semi-solid pharmaceutical composition, a powder, or a solution. By the term “a semi-solid composition” is meant an ointment, cream, salve, jelly, or other pharmaceutical composition of substantially similar consistency suitable for application to the skin. Examples of semi-solid compositions are given in Chapter 17 of The Theory and Practice of Industrial Pharmacy, Lachman, Lieberman and Kanig, published by Lea and Febiger (1970) and in Chapter 67 of Remington's Pharmaceutical Sciences, 15th Edition (1975) published by Mack Publishing Company.

Dermal or skin patches are another method for transdermal delivery of the therapeutic or pharmaceutical compositions described herein. Patches can provide an absorption enhancer such as DMSO to increase the absorption of the compounds. Patches can include those that control the rate of drug delivery to the skin. Patches may provide a variety of dosing systems including a reservoir system or a monolithic system, respectively. The reservoir design may, for example, have four layers: the adhesive layer that directly contacts the skin, the control membrane, which controls the diffusion of drug molecules, the reservoir of drug molecules, and a water-resistant backing. Such a design delivers uniform amounts of the drug over a specified time period, the rate of delivery has to be less than the saturation limit of different types of skin. The monolithic design, for example, typically has only three layers: the adhesive layer, a polymer matrix containing the compound, and a water-proof backing. This design brings a saturating amount of drug to the skin. Thereby, delivery is controlled by the skin. As the drug amount decreases in the patch to below the saturating level, the delivery rate falls.

In one embodiment, the topical composition may, for example, take the form of hydrogel based on polyacrylic acid or polyacrylamide; as an ointment, for example with polyethyleneglycol (PEG) as the carrier, like the standard ointment DAB 8 (50% PEG 300, 50% PEG 1500); or as an emulsion, especially a microemulsion based on water-in-oil or oil-in-water, optionally with added liposomes. Suitable permeation accelerators (entraining agents) include sulphoxide derivatives such as dimethylsulfoxide (DMSO) or decylmethylsulfoxide (decyl-MSO) and transcutol (diethyleneglycolmonoethylether) or cyclodextrin; as well as pyrrolidones, for example 2-pyrrolidone, N-methyl-2-pyrrolidone, 2-pyrrolidone-5-carboxylic acid, or the biodegradable N-(2-hydroxyethyl)-2-pyrrolidone and the fatty acid esters thereof; urea derivatives such as dodecylurea, 1,3-didodecylurea, and 1,3-diphenylurea; terpenes, for example D-limonene, menthone, a-terpinol, carvol, limonene oxide, or 1,8-cineol.

Ointments, pastes, creams and gels also can contain excipients, such as starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, and talc, or mixtures thereof. Powders and sprays also can contain excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Solutions of nanocrystalline antimicrobial metals can be converted into aerosols or sprays by any of the known means routinely used for making aerosol pharmaceuticals. In general, such methods comprise pressurizing or providing a means for pressurizing a container of the solution, usually with an inert carrier gas, and passing the pressurized gas through a small orifice. Sprays can additionally contain customary propellants, such a chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.

The carrier can also contain other pharmaceutically-acceptable excipients for modifying or maintaining the pH, osmolarity, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor of the formulation. The anti-skin aging compositions can also further comprise antioxidants, sun screens, natural retinoids (e.g., retinol), and other additives commonly found in skin treatment compositions.

In some embodiments for preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical carrier, e.g., conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g., water, to form a solid preformulation composition containing a homogeneous mixture of a disclosed compound or a non-toxic pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition is readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.

In solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the subject composition is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, cellulose, microcrystalline cellulose, silicified microcrystalline cellulose, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, hypromellose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as crospovidone, croscarmellose sodium, sodium starch glycolate, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, docusate sodium, cetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, in some embodiments, the compositions comprise buffering agents. In some embodiments, solid compositions of a similar type are also employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.

In some embodiments, a tablet is made by compression or molding, optionally with one or more accessory ingredients. In some embodiments, compressed tablets are prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. In some embodiments, molded tablets are made by molding in a suitable machine a mixture of the subject composition moistened with an inert liquid diluent. In some embodiments, tablets, and other solid dosage forms, such as dragees, capsules, pills and granules, are scored or prepared with coatings and shells, such as enteric coatings and other coatings.

Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the subject composition, in some embodiments, the liquid dosage forms contain inert diluents, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, cyclodextrins and mixtures thereof.

In some embodiments, suspensions, in addition to the subject composition, contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.

In some embodiments, powders and sprays contain, in addition to a subject composition, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. In some embodiments, sprays additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.

Compositions and compounds disclosed herein alternatively are administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation or solid particles containing the compound. In some embodiments, a non-aqueous (e.g., fluorocarbon propellant) suspension is used. In some embodiments, sonic nebulizers are used because they minimize exposing the agent to shear, which results in degradation of the compounds contained in the subject compositions. Ordinarily, an aqueous aerosol is made by formulating an aqueous solution or suspension of a subject composition together with conventional pharmaceutically acceptable carriers and stabilizers. The carriers and stabilizers vary with the requirements of the particular subject composition, but typically include non-ionic surfactants (Tweens, Pluronics, or polyethylene glycol), innocuous proteins like serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars or sugar alcohols. Aerosols generally are prepared from isotonic solutions.

Pharmaceutical compositions suitable for parenteral administration comprise a subject composition in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which are reconstituted into sterile injectable solutions or dispersions just prior to use, which, in some embodiments, contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.

Examples of suitable aqueous and non-aqueous carriers which are employed in the pharmaceutical compositions include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate and cyclodextrins. Proper fluidity is maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.

The dose of the composition comprising at least one compound described herein differs, depending upon the patient's (e.g., human) condition, that is, stage of the disease, general health status, age, and other factors.

Pharmaceutical compositions are administered in a manner appropriate to the disease to be treated (or prevented). An appropriate dose and a suitable duration and frequency of administration will be determined by such factors as the condition of the patient, the type and severity of the patient's disease, the particular form of the active ingredient, and the method of administration. In general, an appropriate dose and treatment regimen provides the composition(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit (e.g., an improved clinical outcome, such as more frequent complete or partial remissions, or longer disease-free and/or overall survival, or a lessening of symptom severity). Optimal doses are generally determined using experimental models and/or clinical trials. In some embodiments, the optimal dose depends upon the body mass, weight, or blood volume of the patient.

Oral doses typically range from about 1.0 mg to about 1000 mg, one to four times, or more, per day.

Dose administration can be repeated depending upon the pharmacokinetic parameters of the dosage formulation and the route of administration used.

It is especially advantageous to formulate compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms are dictated by and directly dependent on (a) the unique characteristics of the TYK2 pseudokinase ligand and the particular therapeutic effect to be achieved and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals. The specific dose can be readily calculated by one of ordinary skill in the art, e.g., according to the approximate body weight or body surface area of the patient or the volume of body space to be occupied. The dose will also be calculated dependent upon the particular route of administration selected. Further refinement of the calculations necessary to determine the appropriate dosage for treatment is routinely made by those of ordinary skill in the art. Such calculations can be made without undue experimentation by one skilled in the art in light of the TYK2 pseudokinase ligand activities disclosed herein in assay preparations of target cells. Exact dosages are determined in conjunction with standard dose-response studies. It will be understood that the amount of the composition actually administered will be determined by a practitioner, in the light of the relevant circumstances including the condition or conditions to be treated, the choice of composition to be administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the chosen route of administration.

Toxicity and therapeutic efficacy of such TYK2 pseudokinase ligands can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, for example, for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. TYK2 pseudokinase ligands that exhibit large therapeutic indices are preferred. While TYK2 pseudokinase ligands that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such inhibitors to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.

The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such TYK2 pseudokinase ligands lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any TYK2 pseudokinase ligand used in a method described herein, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of TYK2 pseudokinase ligand that achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.

EXAMPLES

The following examples are offered for purposes of illustration and are not intended to limit the scope of the claims provided herein. All literature citations in these examples and throughout this specification are incorporated herein by references for all legal purposes to be served thereby. The starting materials and reagents used for the synthesis of the compounds described herein may be synthesized or can be obtained from commercial sources, such as, but not limited to, Sigma-Aldrich, Acros Organics, Fluka, and Fischer Scientific.

Standard abbreviations and acronyms as defined in J. Org. Chem. 2007 72(1): 23A-24A are used herein. Other abbreviations and acronyms used herein are as follows:

AcOH acetic acid DMF dimethylformamide DMP Dess-Martin periodinane dppf (diphenylphosphino)ferrocene EtOAc ethyl acetate EtOH ethanol eq equivalent HBTU N,N,N′,N′-tetramethyl-O-(1H-benzotriazol-1- yl)uronium hexafluorophosphate LC-MS liquid chromatography-mass spectrometry MeOH methanol TEA triethylamine RT room temperature

Example 1: Synthesis of 5-chloro-N-(4-methoxybenzyl)-N-methyl-3-nitropyrazolo[1,5-a]pyrimidin-7-amine (Intermediate 1E)

Step 1: To a solution of 1A (100 g, 1.17 mol) and diethyl malonate (214 g, 1.34 mol) in EtOH (500 mL) was added EtONa (20% w/w in EtOH, 819 g, 2.41 mol). The reaction mixture was stirred at 80° C. overnight under N2. The solvent was removed under vacuum. The residue was diluted with water (2000 mL), acidified with 1N HCl to pH ˜2. Precipitates were formed and collected by filtration to afford 1B (125 g, 69%) as a brown solid.

Step 2: To a solution of 1B (100 g, 1.7 mmol.) in POCl3 (500 mL) was added Et2NPh (167.8 g, 1.12 mol, 1.7 eq.) below 30° C. The mixture was stirred at 90° C. for 5.0 hours. The reaction mixture was poured into ice-water (3000 mL), extracted with EtOAc (3*1000 mL). The combined organic layers were washed by saturated NaHCO3 and brine, dried over Na2SO4, filtrated and concentrated. The residue was purified by silica gel column to afford 1C (90 g, 73%) as a white solid.

Step 3: To sulfuric acid (600 mL, concentrated) in a 2000 mL flask at −10° C. was added nitric acid (300 mL, 70%) dropwise over 30 mins. Solid 1C (100 g) was added in portions while keeping the temperature below 0° C. The mixture was stirred at 0° C. for another 2.0 hours. The mixture was poured into ice-water (3.0 L) and extracted with DCM (1L*3). The combined organic layers were washed by saturated NaHCO3 and brine, dried over Na2SO4, filtrated and concentrated. The residue was triturated with Petroleum Ether/EtOAc (300 mL, 5/1, v/v). The solid was collected by filtrated and dried under vacuum to afford compound 1D (96 g, 77%) as a yellow solid.

Step 4: To a solution of compound 1D (100 g, 430 mmol) in dioxane (1500 mL) was added PMBNHCH3 (65 g, 430 mmol) and TEA (87.0 g, 860 mol). The mixture was stirred at 90° C. for 1.0 hour. The mixture was diluted with DCM (3000 mL) and washed by brine. The organic phase was dried over Na2SO4 and concentrated under vacuum. The residue was triturated with Petroleum Ether/EtOAc (1500 mL, 10/1, v/v). The solid was collected by filtrated and dried under vacuum to afford 5-chloro-N-(4-methoxybenzyl)-N-methyl-3-nitropyrazolo[1,5-a]pyrimidin-7-amine (1E) (130 g, 87%) as a yellow solid. LCMS: 248.0 [M+H]+.

Example 2: Synthesis of 7-fluoro-2,3-dihydrobenzo[b][1,4]dioxin-5-amine (Intermediate 2E)

Step 1: To a solution of 2A (100.0 g, 0.769 mol) in AcOH (800 mL) was added dropwise HNO3/AcOH (200 mL, v/v=1/1). The mixture was stirred at 10° C. for 90 min and then poured into ice-water (4 L). The solid was collected by filtration and washed by small amount of water to afford 2B (125 g, 93%), which was used in the next step without further purification.

Step 2: To a solution of 2B (125.0 g, 0.714 mol) in DMF (800 mL) at 25° C. was added 2-bromoethanol (220.0 g, 1.785 mol) and DIPEA (454.0 g, 3.570 mol). The mixture was stirred at 120° C. for 2 h. The reaction solution was diluted with water (8 L), extracted by EtOAc (4 L), dried over Na2SO4. The crude product was purified by column chromatography to afford 2C (116 g, 73%) as a yellow oil.

Step 3: To a solution of 2C (116 g, 0.530 mol) in DMF (600 mL) was added Cs2CO3 (460.0 g, 1.589 mol), then the mixture was heated to 80° C. for 1 h. The mixture was poured into ice-water (5 L), and extracted with EtOAc (3 L). The solvent was removed under reduced pressure, the residue was purified by column chromatography to afford 2D (85 g, 85%) as a light yellow solid.

Step 4: To a solution of 2D (70.0 g, 0.352 mol) in EtOAc (500 mL) at 25° C. was added Pd/C (7.0 g). The resulting mixture was stirred at r.t. under H2 atmosphere for 14 h. The mixture was filtered, and washed by EtOAc. The organic phase was collected and the solvent was removed under reduced pressure to afford 7-fluoro-2,3-dihydrobenzo[b][1,4]dioxin-5-amine (2E) (45 g, 75%) as a brown oil.

Example 3: Synthesis of phenyl ((1R,2S)-2-fluorocyclopropyl)carbamate (Intermediate 3C)

To a solution of compound 3A (100 g, 405 mmol) in DCM (1000 mL) at −20° C. was added TEA (122 g, 1.22 mol). Compound 3B (70 g, 445 mmol) was added into the mixture at −20° C. over 30 mins. The mixture was stirred at −20° C. for 2.0 hours. The reaction mixture was washed by saturated NaHCO3 and brine, the organic phase was dried over Na2SO4 and concentrated under vacuum to afford phenyl ((1R,2S)-2-fluorocyclopropyl)carbamate (3C) (70 g, 91%) as a brown solid. LCMS: 196.0 [M+H]+.

Example 4: Synthesis of 1-(5-((7-fluoro-2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-(methylamino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-((1R,2S)-2-fluorocyclopropyl)urea (5)

Step 1: To a solution of 1E (100 g, 288 mmol) and 2E (57 g, 345 mmol) in dry 1,4-dioxane (3000 mL) under N2 atmosphere was added Cs2CO3 (141 g, 432 mmol), Pd(OAc)2 (5.2 g, 23.3 mmol) and BINAP (28.6 g, 46.6 mmol). After stirring at 115° C. overnight, the reaction mixture was cooled to rt. and diluted with hexane (3000 mL). The solid was collected by filtration and washed with 2×1500 mL (50% hexane in DCM). The solid was suspended into 5000 mL water and stirred for 1 h. The solid was collected by filtration and dried under vacuum to afford compound 2 (90 g, 65%) as a brown solid.

Step 2: To a solution of compound 2 (70 g, 145 mmol) in NMP (2000 mL) was added saturated NH4Cl (aq., 1000 mL) and Fe (92.8 g, 1.45 mol). After stirring at 80° C. for 5.0 hours, the reaction mixture was cooled to rt. and filtrated. The filtrate was poured into water (20 L) and the solid was collected by filtration. The solid was dissolved with DCM (˜1500 mL) and flashed through a short silica gel column, the column was washed with another 3000 mL of DCM/MeOH=30/1. The organic phases were concentrated under vacuum. The residue was triturated by EtOH/MeCN (2000 mL, 5/1, v/v). The solid was collected by filtration and dried under vacuum to afford compound 3 (46 g, 71%) as a brown solid.

Step 3: To a solution of compound 3 (86 g, 0.19 mol, 1.0 eq.) in DMF (800 mL) was added 3C (44.7 g, 0.23 mol, 1.3 eq.) and TEA (38.6 g, 0.38 mol, 2 eq.). After stirring at 80° C. for 2 hours, the reaction mixture was cooled to RT and poured into water (8000 mL). The solid was collected by filtration and dried under vacuum. The residue was triturated by EtOH/MeCN (1200 mL, 5/1, v/v). The solid was collected by filtration and dried under vacuum to afford compound 4 (75 g, 71%) as a brown solid.

Step 4: To a solution of compound 4 (118 g, 0.21 mol, 1.0 eq.) in DCM (1200 mL) was added triethylsilane (37.3 g, 0.32 mol, 1.5 eq.). After stirring for 10 mins, TFA (240 mL) was added into the mixture and the reaction solution was stirred at rt. for 0.5 hour. TfOH (60 mL) was added and the reaction solution was stirred at rt. for another 2 hour. The reaction mixture was poured into saturated NaHCO3 (30 mL). The solid was collected by filtration and dried under vacuum. The solid was triturated with EtOH (500 mL) and collected by filtration. The solid was triturated with EtOAc (2×500 mL). The solid was collected by filtration and dried under vacuum to afford 1-(5-((7-fluoro-2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-(methylamino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-((1R,2S)-2-fluorocyclopropyl)urea (5) (80 g, 86%) as a yellow solid. LCMS: 432.1 [M+H]+.

Example 5: Synthesis of 1-(5-((2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-(methylamino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (7)

Step 1: To sulfuric acid (9 mL, concentrated) in a 100 mL flask at −10° C. was added nitric acid (4.5 mL, 70%) dropwise over 2 mins. Solid 5,7-dichloropyrazolo[1,5-a]pyrimidine (1AA) (1.0 g) was added in portions. The resulting mixture was continuously stirred at 0° C. for 6 hrs. LC/MS showed no starting material remaining. To the reaction was added 80 mL ice-water, and stirred at 0° C. for 20 mins. The mixture was extracted by DCM (3×60 mL) and washed with saturated NaHCO3 and brine. The solvent was removed in vacuo to afford 5,7-dichloro-3-nitropyrazolo[1,5-a]pyrimidine (1BB) (1.15 g) as a yellow solid which was used without further purification.

Step 2: 5,7-dichloro-3-nitropyrazolo[1,5-a]pyrimidine (1BB) (1.2 g, 1.0 eq) and TEA (0.624 g, 1.2 eq) was dissolved in DMF (12 mL). The solution was cooled at −30° C. (hexane/dry ice), then a solution of 1-(4-methoxyphenyl)-N-methylmethanamine (0.682 g, 0.98 eq) in DMF (2 mL) was added dropwise. The resulting mixture was stirred at −20° C. for 15 mins. The reaction mixture was poured into ice-water (200 mL) and stirred for 5 mins. The precipitate was collected by filtration, washed with water (2×20 mL), and dried in vacuo to afford 5-chloro-N-(4-methoxybenzyl)-N-methyl-3-nitropyrazolo[1,5-a]pyrimidin-7-amine (1E) (1.68 g, 94%) which was used without further purification.

Step 3: 5-chloro-N-(4-methoxybenzyl)-N-methyl-3-nitropyrazolo[1,5-a]pyrimidin-7-amine (1E) (1.68 g, 1.0 eq) was suspended into EtOH (100 mL). To the solution was added iron powder (4.0 g, 15 eq) and saturated aqueous NH4Cl (30 mL). The resulting mixture was heated at 90° C. for 35 mins. The excess iron was removed by filtration through celite-pad. The solvent was concentrated in vacuo. The residue was dissolved in ethyl acetate (70 mL), and washed with NaHCO3 (sat.) and brine. The crude mixture was purified on silica gel column to afford a yellow oil 5-chloro-N7-(4-methoxybenzyl)-N7-methylpyrazolo[1,5-a]pyrimidine-3,7-diamine (1F) (1.4 g, 91%) and it solidified overnight.

Step 4: 5-chloro-N7-(4-methoxybenzyl)-N7-methylpyrazolo[1,5-a]pyrimidine-3,7-diamine (1F) (120 mg, 1.0 eq) was dissolved in dry THE (8 mL) and cooled at 0° C. under N2. To the solution was added triphosgene (37 mg, 0.33 eq) and DIEA (54 mg, 1.1 eq). The resulting mixture was stirred at RT for 15 mins. The reaction mixture was cooled over ice-water bath, and added CH3NH2 (2M in THF, 0.6 mL, 3.0 eq) at 0° C. The resulting mixture was stirred at RT for 5 mins. The reaction mixture was diluted with DCM (50 mL), washed with NaHCO3 (sat.) and brine. The crude product was purified on ISCO to afford 1-(5-chloro-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (1G) (114 mg, 80%).

Step 5: 1-(5-chloro-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (1G) (100 mg, 1.0 eq), 2,3-dihydrobenzo[b][1,4]dioxin-5-amine (61 mg, 1.5 eq), Pd(OAc)2 (15 mg, 0.25 eq), BINAP (67 mg, 0.4 eq) and Cs2CO3 (156 mg, 1.8 eq) were combined in dry dioxane (8 mL). The mixture was bubbled N2 for 5 mins, then the reaction was heated at 115° C. in a sealed-tube for 8 hrs. The reaction was diluted with DCM (50 mL), washed with water and brine. The crude was purified on ISCO silica-gel column to afford 1-(5-((2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (6) (45 mg).

Step 6: To a solution of 1-(5-((2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (6) (45 mg) in DCM (2 mL) was added anisole (0.1 mL) and TFA (0.5 mL). The resulting mixture was stirred at RT for 1 hr. The solvent was removed in vacuo. The residue was dissolved in DCM (50 mL), washed with NaHCO3 (sat.) and brine. The crude mixture was purified on ISCO silica-gel column to afford 1-(5-((2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-(methylamino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (7) (22 mg, 64%) as a beige solid. LCMS: 370.4 (M+H)+.

Example 6: Synthesis of 1-(5-((2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-(methylamino)pyrazolo[1,5-a]pyrimidin-3-yl)urea (10)

Step 1: To a solution of 5-chloro-N7-(4-methoxybenzyl)-N7-methylpyrazolo[1,5-a]pyrimidine-3,7-diamine (1F) (150 mg, 1.0 eq) in dry THE (2 mL) was added TMSNCO (82 mg, 1.5 eq). The resulting mixture was heated at 60° C. for 1 hr. The solvent was removed in vacuo. The residue was suspended in MeOH (5 mL), and the mixture was heated at 60° C. for 1 hr. Upon cooling to room temperature, a solid was precipitated out and collected by filtration to afford 1-(5-chloro-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)urea (8) (120 mg, 70%) which was used without further purification.

Step 2: 1-(5-chloro-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)urea (120 mg, 1.0 eq), 2,3-dihydrobenzo[b][1,4]dioxin-5-amine (8) (76 mg, 1.5 eq), Pd(OAc)2 (12 mg, 0.15 eq), BINAP (42 mg, 0.2 eq) and Cs2CO3 (162 mg, 1.5 eq) were combined in dry dioxane (8 mL). The mixture was bubbled N2 for 5 mins, then the reaction was heated at 115° C. in a sealed-tube for 8 hrs. The reaction was diluted with DCM (50 mL), washed with water and brine. The crude was purified on ISCO silica-gel column to afford 1-(5-((2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)urea (9) (39 mg, 25%).

Step 3: To a solution of 1-(5-((2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)urea (9) (39 mg) in DCM (2 mL) was added anisole (0.1 mL) and TFA (0.5 mL). The resulting mixture was stirred at RT for 1 hr. The solvent was removed in vacuo. The residue was dissolved in DCM (50 mL), washed with NaHCO3 (sat.) and brine. The crude mixture was purified on ISCO silica-gel column to afford 1-(5-((2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-(methylamino)pyrazolo[1,5-a]pyrimidin-3-yl)urea (10) (20 mg, 69%) as a beige solid. LCMS: 356.5 (M+H)+.

Example 7: Synthesis of 1-(5-((2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-((methyl-d3 amino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (13)

Step 1: 5,7-dichloro-3-nitropyrazolo[1,5-a]pyrimidine (1BB) (500 mg, 1.0 eq) and TEA (0.26 g, 1.2 eq) was dissolved in DMF (8 mL). The solution was cooled at −20° C. (hexane dry ice), then CD3NH2·HCl (148 mg, 0.98 eq) was added. The resulting mixture was stirred at −20° C. for 1 hr. The reaction mixture was diluted with ethyl acetate (80 mL), washed with water (3×50 mL) and brine. The organic phase was dried over Na2SO4 and concentrated in vacuo to afford 5-chloro-N-(methyl-d3)-3-nitropyrazolo[1,5-a]pyrimidin-7-amine (11) (400 mg) as a yellow solid which was used without purification.

Step 2: To a solution of 5-chloro-N-(methyl-d3)-3-nitropyrazolo[1,5-a]pyrimidin-7-amine (11) (400 mg, 1.0 eq) in DMF (15 mL) was added K2CO3 (760 mg, 3.0 eq) and PMBCl (410 mg, 1.5 eq). The resulting mixture was stirred at RT for 2 days. The reaction mixture was diluted with ethyl acetate (100 mL), washed with water (3×60 mL) and brine. The crude mixture was purified on silica-gel column to afford 5-chloro-N-(4-methoxybenzyl)-N-(methyl-d3)-3-nitropyrazolo[1,5-a]pyrimidin-7-amine (12) (410 mg, 67%).

1-(5-((2,3-dihydrobenzo[b][1,4]dioxin-5-yl)amino)-7-((methyl-d3)amino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (13) was prepared from 5-chloro-N-(4-methoxybenzyl)-N-(methyl-d3)-3-nitropyrazolo[1,5-a]pyrimidin-7-amine (12) following the synthetic procedures outlined in Example 1, steps 3-6. LCMS: 373.5 (M+H)+.

Example 8: Synthesis of 1-(5-((1-(6-methoxypyridazin-3-yl)-2-oxo-1,2-dihydropyridin-3-yl)amino)-7-(methylamino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (17)

Step 1: To a suspension of 3,6-dibromopyridazine (1.0 g, 1.0 eq) in MeOH (50 mL) was added K2CO3 (1.2 g, 2.0 eq). The resulting mixture was heated at 90° C. in a sealed-tube for 15 hrs. The solvent was removed in vacuo. The residue was dissolved in ethyl acetate, washed with water and brine to afford 3-bromo-6-methoxypyridazine (14) (725 mg, 92%) which was used without further purification.

Step 2: Benzyl (2-oxo-1,2-dihydropyridin-3-yl)carbamate (900 mg, 1.0 eq), 3-bromo-6-methoxypyridazine (14) (700 mg, 1.0 eq), N1,N1,N2,N2-tetramethylethane-1,2-diamine (0.13 g, 0.3 eq), CuI (71 mg, 0.1 eq), and K2CO3 (1.1 g, 2.0 eq) were combined in dry dioxane (25 mL). The mixture was bubbled N2 gas for 5 mins. The resulting mixture was heated at 100° C. in a sealed-tube for 14 hrs. The reaction was diluted with ethyl acetate (100 mL) and washed with water (2×50 mL). The crude was purified on silica-gel column to afford benzyl (1-(6-methoxypyridazin-3-yl)-2-oxo-1,2-dihydropyridin-3-yl)carbamate (15) (362 mg, 27%).

Step 3: Benzyl (1-(6-methoxypyridazin-3-yl)-2-oxo-1,2-dihydropyridin-3-yl)carbamate (15) (360 mg) was dissolved in MeOH (20 mL) and ethyl acetate (20 mL). To the solution was added Pd/C (10%, 72 mg). The mixture was hydrogenated at H2 (50 psi) for 5 hrs. The catalyst was removed by filtration. The solvent was removed in vacuo to afford a beige solid. The solid was suspended in ethyl acetate (5 mL) and heated at reflux for 5 mins. Upon cooling to room temperature, a white solid precipitated out. The solid was collected by filtration and dried in vacuo to afford 3-amino-1-(6-methoxypyridazin-3-yl) pyridin-2(1H)-one (16) (149 mg, 67%).

1-(5-((1-(6-methoxypyridazin-3-yl)-2-oxo-1,2-dihydropyridin-3-yl)amino)-7-(methylamino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (17) was prepared from 3-amino-1-(6-methoxypyridazin-3-yl) pyridin-2(1H)-one (16) following the synthetic procedures outlined in Example 1, steps 4-6. LCMS: 437.5 (M+H)+.

Example 9: Synthesis of 1-(5-((2-methoxy-3-(1-methyl-1H-pyrazol-4-yl)phenyl)amino)-7-(methylamino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (19)

Step 1: 3-bromo-2-methoxyaniline(1.0 g, 1.0 eq), 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole(1.23 g, 1.2 eq) and PdCl2(dppf) (0.38 g, 0.1 eq) and K2CO3 (1.3 g, 2 eq) were combined in dioxane (30 mL) and water (10 mL). The mixture was degassed with bubbling N2 gas for 5 mins. The resulting mixture was heated at 90° C. under N2 for 14 hrs. The mixture was diluted with ethyl acetate (150 mL), washed with water (100 mL) and brine. The crude mixture was purified on silica-gel column to afford 2-methoxy-3-(1-methyl-1H-pyrazol-4-yl) aniline (18) (0.64 g, 63%) as a brown solid.

1-(5-((2-methoxy-3-(1-methyl-1H-pyrazol-4-yl)phenyl)amino)-7-(methylamino)pyrazolo[1,5-a]pyrimidin-3-yl)-3-methylurea (19) was prepared from 2-methoxy-3-(1-methyl-1H-pyrazol-4-yl) aniline (18) following the synthetic procedures outlined in Example 1, steps 4-6. LCMS: 422.5 (M+H)+.

Example 10: Synthesis of (13E,14E)-17-(methylamino)-4-oxa-2,7,9-triaza-1(5,3)-pyrazolo[1,5-a]pyrimidina-3(1,3)-benzenacyclononaphan-8-one (22)

5-chloro-N7-(4-methoxybenzyl)-N7-methylpyrazolo[1,5-a]pyrimidine-3,7-diamine (1F) (250 mg, 1.0 eq) was dissolved in dry THE (15 mL) and cooled at 0° C. under N2. To the solution was added triphosgene (77 mg, 0.33 eq) and DIEA (112 mg, 1.1 eq). The mixture was stirred at RT for 15 mins then was cooled over ice-water bath. 3-(2-aminoethoxy) aniline (132 mg, 1.1 eq) was added to the reaction mixture at 0° C. The resulting mixture was stirred at RT for 15 mins. The reaction mixture was diluted with DCM (60 mL), washed with NaHCO3 (sat.) and brine. The crude was purified on ISCO to afford 1-(2-(3-aminophenoxy)ethyl)-3-(5-chloro-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)urea (20) (258 mg, 66%).

1-(2-(3-aminophenoxy)ethyl)-3-(5-chloro-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)urea (20) (200 mg, 1.0 eq), Pd(OAc)2 (20 mg, 0.2 eq), BINAP (110 mg, 0.4 eq) and Cs2CO3 (215 mg, 1.5 eq) were combined in dry dioxane (25 mL). The mixture was bubbled with N2 gas for 5 mins. The resulting mixture was heated at 115° C. in a sealed-tube for 8 hrs. The reaction was diluted with DCM (100 mL) and ethyl acetate (100 mL). The insoluble solids were removed by filtration. The filtrate was concentrated in vacuo to afford a brown solid. The solid was suspended into DCM (20 mL) and the suspension was heated at reflux for 10 mins. Upon cooled to room temperature, the remaining solid was collected by filtration, washed with DCM (2×5 mL), and dried in vacuo to afford (1E,1E)-1-((4-methoxybenzyl)(methyl)amino)-4-oxa-2,7,9-triaza-1(5,3)-pyrazolo[1,5-a]pyrimidina-3(1,3)-benzenacyclononaphan-8-one (21) (75 mg, 40%) as a yellow solid.

(1E,1E)-1-((4-methoxybenzyl)(methyl)amino)-4-oxa-2,7,9-triaza-1(5,3)-pyrazolo[1,5-a]pyrimidina-3(1,3)-benzenacyclononaphan-8-one (21) (75 mg) was suspended in DCM (4 mL), added anisole (0.2 mL) and TFA (1 mL). The resulting mixture was stirred at RT for 30 min. The mixture was concentrated to afford a brown oil. The brown oil was dissolved in DCM (100 mL), washed with NaHCO3 (sat.) and brine. The organic phase was dried over Na2SO4 and concentrated in vacuo to afford a solid that was washed with DCM (2×5 mL) to provide (13E,14E)-17-(methylamino)-4-oxa-2,7,9-triaza-1(5,3)-pyrazolo[1,5-a]pyrimidina-3(1,3)-benzenacyclononaphan-8-one (22) (38 mg, 69%). LCMS: 340.3 (M+H)+.

Example 11: Synthesis of (13E,14E)-17-(methylamino)-2,6,8-triaza-1(5,3)-pyrazolo[1,5-a]pyrimidina-3(1,3)-benzenacyclooctaphan-7-one (25)

5-chloro-N7-(4-methoxybenzyl)-N7-methylpyrazolo[1,5-a]pyrimidine-3,7-diamine (1F) (180 mg, 1.0 eq) was dissolved in dry THE (12 mL) and cooled at 0° C. under N2. To the solution was added triphosgene (55 mg, 0.33 eq) and DIEA (80 mg, 1.1 eq). The resulting mixture was stirred at RT for 15 mins and then cooled over an ice-water bath again. 3-(2-aminoethyl)aniline (85 mg, 1.1 eq) was added to the reaction mixture at 0° C. The resulting mixture was stirred at RT for 15 mins. The reaction mixture was diluted with DCM (40 mL), washed with NaHCO3 (sat.) and brine. The crude was purified on ISCO to afford 1-(3-aminophenethyl)-3-(5-chloro-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)urea (23) (165 mg, 61%).

1-(3-aminophenethyl)-3-(5-chloro-7-((4-methoxybenzyl)(methyl)amino)pyrazolo[1,5-a]pyrimidin-3-yl)urea (23) (160 mg, 1.0 eq), Pd(OAc)2 (15 mg, 0.2 eq), BINAP (83 mg, 0.4 eq) and Cs2CO3 (165 mg, 1.5 eq) were combined in dry dioxane (25 mL). The mixture was bubbled with N2 gas for 5 mins. The resulting mixture was heated at 115° C. in a sealed-tube for 8 hrs. The reaction was diluted with DCM (100 mL) and ethyl acetate (100 mL). The insoluble solids were removed by filtration. The filtrate was concentrated in vacuo to afford a brown solid. The solid was suspended into DCM (20 mL) and the suspension was heated at reflux for 10 mins. Upon cooling to room temperature, the remaining solid was collected by filtration, washed with DCM (2×5 mL), and dried in vacuo to afford (1E,1E)-1-((4-methoxybenzyl)(methyl)amino)-2,6,8-triaza-1(5,3)-pyrazolo[1,5-a]pyrimidina-3(1,3)-benzenacyclooctaphan-7-one (24) (56 mg, 38%) as a yellow solid.

(1E,1E)-1-((4-methoxybenzyl)(methyl)amino)-2,6,8-triaza-1(5,3)-pyrazolo[1,5-a]pyrimidina-3(1,3)-benzenacyclooctaphan-7-one (24) (56 mg) was suspended in DCM (4 mL), added anisole (0.2 mL) and TFA (1 mL). The resulting mixture was stirred at RT for 30 min. The mixture was concentrated to afford a brown oil. The brown oil was dissolved in DCM (100 mL), washed with NaHCO3 (sat.) and brine. The organic phase was dried over Na2SO4 and concentrated in vacuo to afford a solid that was washed with DCM (2×5 mL) to provide (13E,14E)-17-(methylamino)-2,6,8-triaza-1(5,3)-pyrazolo[1,5-a]pyrimidina-3(1,3)-benzenacyclooctaphan-7-one (25) (30 mg, 73%). LCMS: 324.6 (M+H)+.

Compounds 26-187 were prepared by similar procedures as described in the preceding Examples.

MS Compound Structure Name [M + H]+ 26 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 414.2 27 1-(3-cyanocyclobutyl)-3-(5- ((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 435.2 28 1-(1-cyanocyclopropyl)-3-(5- ((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 421.1 29 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(3- hydroxycylcobutyl)urea 426.2 30 1-(2-hydroxycyclopropyl)-3-(7- (methylamino)-5-(quinoxalin-5- ylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 406.2 31 1-(2-hydroxycyclopropyl)-3-(7- (methylamino)-5-(quinolin-8- ylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 405.2 32 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(2- hydroxycyclopropyl)urea 412.2 33 1-(5-((7-cyano-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 439.5 34 1-(3,3-difluorocyclobutyl)-3-(5- ((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 446.1 35 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(3- fluorocyclobutyl)urea 428.1 36 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(3- oxocyclobutyl)urea 424.2 37 1-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(2- fluorocyclopropyl)urea 433.1 38 1-((1R,2S)-2- fluorocyclopropyl)-3-(7- (methylamino)-5-((2,3,5,6- tetrafluorophenyl)amino)pyrazolo [1,5-a]pyrimidin-3-yl)urea 428.2 39 1-((1R,2S)-2- fluorocyclopropyl)-3-(7- (methylamino)-5-(quinolin-8- ylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 407.1 40 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7-((methyl- d3)amino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 417.3 41 1-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1S,2S)-2- fluorocyclopropyl)urea 432.3 42 1-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1S,2R)-2- fluorocyclopropyl)urea 432.2 43 1-(2-cyanocyclobutyl)-3-(5- ((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 435.1 44 1-((1R,2S)-2- fluorocyclopropyl)-3-(7- (methylamino)-5-(quinoxalin-5- ylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 408.1 45 1-((1R,2S)-2-cyanocyclopropyl)- 3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 421.1 46 1-(2-cyanocyclobutyl)-3-(5-((7- fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 453.1 47 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(3- (hydroxymethyl)cyclobutyl)urea 440.1 48 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(3-hydroxy- 3-methylcyclobutyl)urea 440.2 49 1-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7-((methyl- d3)amino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 435.3 50 1-(5-(benzofuran-4-ylamino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 396.1 51 1-((1R,2S)-2-cyanocyclopropyl)- 3-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 439.0 52 1-(5-((2- (difluoromethoxy)pyridin-3- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 423.1 53 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-2-methyl-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 428.1 54 1-(5-((6-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 432.3 55 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-6-fluoro-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 432.1 56 1-((1S,2R)-2- hydroxycyclobutyl)-3-(7- (methylamino)-5-(1-(tetrahydro- 2H-pyran-4-yl)-1H-pyrrolo[2,3- b]pyridin-3-yl)pyrazolo[1,5- a]pyrimidin-3-yl)urea 477.3 57 1-((1S,2R)-2- hydroxycyclobutyl)-3-(5-(1- isopropyl-1H-pyrrolo[2,3- b]pyridin-3-yl)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 435.3 58 1-(2-hydroxycyclopropyl)-3-(5- (1-isopropyl-1H-pyrrolo[2,3- b]pyridin-3-yl)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 421.3 59 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1S,2R)-2- hydroxycyclobutyl)urea 426.4 60 1-(5-((3,5- dimethylphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 384.3 61 1-(5-((6,7-difluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 450.2 62 1-methyl-3-(5-((4-methyl-3,4- dihydro-2H- benzo[b][1,4]oxazin-8- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 383.5 63 1-(5-((3-chloro-2- methoxyphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 376.5 64 1-methyl-3-(7-(methylamino)-5- (quinolin-8- ylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 363.6 65 1-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 388.5 66 1-(5-((2-methoxy-6- methylphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 356.5 67 1-(5-((2,5- dimethoxyphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 372.4 68 1-(5-((2-methoxy-5- methylphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 356.5 69 1-cyclopropyl-3-(5-((7-fluoro- 2,3-dihydrobenzo[b][1,4]dioxin- 5-yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 415.2 70 1-(5-((3,5- dimethoxyphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 372.3 71 1-methyl-3-(7-(methylamino)-5- ((2-(oxazol-2- yl)phenyl)amino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 379.5 72 1-(5-(1-isopropyl-1H- pyrrolo[2,3-b]pyridin-3-yl)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 379.1 73 1-methyl-3-(7-((methyl- d3)amino)-5-(1-(tetrahydro-2H- pyran-4-yl)-1H-pyrrolo[2,3- b]pyridin-3-yl)pyrazolo[1,5- a]pyrimidin-3-yl)urea 424.4 74 1-methyl-3-(5-(1-methyl-1H- pyrrolo[2,3-b]pyridin-3-yl)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 351.2 75 1-methyl-3-(7-((methyl- d3)amino)-5-(1- (tetrahydrofuran-3-yl)-1H- pyrrolo[2,3-b]pyridin-3- yl)pyrazolo[1,5-a]pyrimidin-3- yl)urea 410.2 76 1-methyl-3-(7-(methylamino)-5- ((7-(trifluoromethyl)-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 439.5 77 1-(5-((7-chloro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 404.6 78 1-methyl-3-(5-((7-methyl-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 384.4 79 1-cyclopropyl-3-(5-((7-methyl- 2,3-dihydrobenzo[b][1,4]dioxin- 5-yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 410.4 80 1-methyl-3-(7-(methylamino)-5- (1-(tetrahydro-2H-pyran-4-yl)- 1H-pyrrolo[2,3-b]pyridin-3- yl)pyrazolo[1,5-a]pyrimidin-3- yl)urea 421.2 81 1-(5-((7-methoxy-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 400.3 82 1-cyclopropyl-3-(7- (methylamino)-5-((7- (trifluoromethyl)-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 464.5 83 1-(5-((7-cyano-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 395.2 84 1-cyclopropyl-3-(5-((7-methoxy- 2,3-dihydrobenzo[b][1,4]dioxin- 5-yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 426.3 85 1-(5-((7-chloro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3- cyclopropylurea 430.5 86 1-methyl-3-(7-(methylamino)-5- (1-(oxetan-3-yl)-1H-pyrrolo[2,3- b]pyridin-3-yl)pyrazolo[1,5- a]pyrimidin-3-yl)urea 393.1 87 1-(5-(benzo[d][1,3]dioxol-4- ylamino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 356.5 88 1-methyl-3-(7-(methylamino)-5- (1H-pyrrolo[2,3-b]pyridin-3- yl)pyrazolo[1,5-a]pyrimidin-3- yl)urea 337.2 89 1-(5-((7-cyano-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3- cyclopropylurea 412.4 90 1-(5-((2,6- difluorophenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 348.4 91 1-(5-(isoquinolin-4-ylamino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 363.6 92 1-(5-((2,3-dihydro- [1,4]dioxino[2,3-b]pyridin-8- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 371.4 93 1-cyclobutyl-3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 410.2 94 1-methyl-3-(7-(methylamino)-5- ((2-((tetrahydro-2H-pyran-4- yl)oxy)pyridin-3- yl)amino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 413.1 95 1-methyl-3-(7-(methylamino)-5- ((3-oxo-3,4-dihydro-2H- benzo[b][1,4]oxazin-5- yl)amino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 383.5 96 1-(5-((7-cyclopropyl-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 410.4 97 1-(5-((2,3- dimethoxyphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 372.3 98 1-methyl-3-(7-(methylamino)-5- ((3-oxo-3,4-dihydro-2H- benzo[b][1,4]oxazin-8- yl)amino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 383.5 99 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(2,2,2- trifluoroethyl)urea 438.2 100 1-(5-((2,3- dihydrobenzo[b][1,4]dixoin-5- y)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(3- hydroxypropyl)urea 414.2 101 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(prop-2-yn- 1-yl)urea 394.1 102 1-(cyanomethyl)-3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 395.2 103 1-(cyclopropylmethyl)-3-(5- ((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 410.2 104 1-(5-(benzofuran-4-ylamino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 352.1 105 1-cyclobutyl-3-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 428.5 106 1-(5-((7-cyano-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3- cyclobutylurea 435.5 107 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-6- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 370.3 108 1-cyclopentyl-3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 424.5 109 1-(5-((7-ethynyl-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 394.4 110 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-2-fluoro-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 388.2 111 1-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(prop-2-yn- 1-yl)urea 412.3 112 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(2- methoxyethyl)urea 414.5 113 1-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(2- methoxyethyl)urea 432.5 114 1-(cyanomethyl)-3-(5-((7-fluoro- 2,3-dihydrobenzo[b][1,4]dioxin- 5-yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 413.5 115 1-(5-((2,2- difluorobenzo[d][1,3]dioxol-4- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 392.2 116 1-(5-((2,2- dimethylbenzo[d][1,3]dioxol-4- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 384.2 117 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(oxetan-3- yl)urea 412.2 118 2-(3-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3- yl)ureido)acetamide 431.6 119 1-(1-cyanoethyl)-3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 409.5 120 1-(1-cyanoethyl)-3-(5-((7-fluoro- 2,3-dihydrobenzo[b][1,4]dioxin- 5-yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 427.4 121 (S)-1-(1-cyanoethyl)-3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 409.5 122 (R)-1-(1-cyanoethyl)-3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 409.5 123 (S)-1-(but-3-yn-2-yl)-3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 408.5 124 1-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 374.4 125 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(1,1- dioxidothietan-3-yl)urea 460.2 126 1-(1,1-dioxidothietan-3-yl)-3-(5- ((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 478.2 127 1-(cyanomethyl)-3-(7- (methylamino)-5-(quinolin-8- ylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 388.1 128 1-(cyanomethyl)-3-(5-((3,5- dimethylphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 365.2 129 1-(cyanomethyl)-3-(5-((2- methoxy-5- methylphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 381.3 130 1-(cyanomethyl)-3-(5-(1- isopropyl-1H-pyrrolo[2,3- b]pyridin-3-yl)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 404.1 131 1-(5-(benzofuran-4-ylamino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3- (cyanomethyl)urea 377.1 132 1-(cyanomethyl)-3-(7- (methylamino)-5-(quinoxalin-5- ylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 389.1 133 1-(2,2-difluoroethyl)-3-(5-((7- fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 438.2 134 1-(2,2-difluoroethyl)-3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 420.2 135 1-(5-((6-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(prop-2-yn- 1-yl)urea 412.2 136 1-(cyanomethyl)-3-(5-((6-fluoro- 2,3-dihydrobenzo[b][1,4]dioxin- 5-yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 413.2 137 1-(cyanomethyl)-3-(7- (methylamino)-5-(1-(tetrahydro- 2H-pyran-4-yl)-1H-pyrrolo[2,3- b]pyridin-3-yl)pyrazolo[1,5- a]pyrimidin-3-yl)urea 446.2 138 1-((1-cyanocyclopropyl)methyl)- 3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 435.2 139 1-cyclopropyl-3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 396.3 140 3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-1,1- dimethylurea 384.4 141 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(3-hydroxy- 2,2-dimethylpropyl)urea 442.7 142 1-(5-((8-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 388.5 143 1-(5-((3,5-difluoro-2- methoxyphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 378.6 144 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(methyl- d3)urea 373.5 145 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(2- hydroxyethyl)urea 400.2 146 1-methyl-3-(7-(methylamino)-5- (phenylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 312.2 147 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-ethylurea 384.4 148 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3- isopropylurea 398.4 149 1-(5-((2-methoxyphenyl)amino)- 7-(methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 342.4 150 1-(5-((3-methoxyphenyl)amino)- 7-(methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 342.4 151 1-(5-((4-methoxyphenyl)amino)- 7-(methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 342.4 152 1-(5-((3,5- dimethylphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 340.5 153 1-(5-(benzo[d][1,3]dioxol-4- ylamino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 356.5 154 1-(5-(chroman-5-ylamino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 368.4 155 1-(5-(cyclopentylamino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 304.5 156 1-methyl-3-(7-(methylamino)-5- ((2- (methylsulfonyl)phenyl)amino)py- razolo[1,5-a]pyrimidin-3- yl)urea 390.5 157 1-(5-(chroman-8-ylamino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 368.4 158 1-(5-((6-fluorochroman-8- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 386.4 159 1-methyl-3-(5- (methyl(phenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 326.6 160 1-methyl-3-(7-(methylamino)-5- (naphthalen-1- ylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 362.2 161 1-methyl-3-(7-(methylamino)-5- ((2- phenoxyphenyl)amino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 404.6 162 1-(5-((2- isopropoxyphenyl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 370.5 163 1-methyl-3-(5-((4-methyl-3,4- dihydro-2H- benzo[b][1,4]oxazin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 383.5 164 1-(5-((2-methoxypyridin-3- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 343.3 165 1-(5-([1,1′-biphenyl]-2- ylamino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 388.5 166 1-methyl-3-(7-(methylamino)-5- (quinoxalin-5- ylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 364.6 167 N-(7-(methylamino)-3-(3- methylureido)pyrazolo[1,5- a]pyrimidin-5-yl)benzamide 340.4 168 1-methyl-3-(7-(methylamino)-5- ((2- (trifluoromethoxy)phenyl)amino) pyrazolo[1,5-a]pyrimidin-3- yl)urea 396.4 169 1-(5-((3,4-dihydro-2H- benzo[b][1,4]dioxepin-6- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-methylurea 384.3 170 1-methyl-3-(7-(methylamino)-5- ((5,6,7,8-tetrahydronaphthalen- 1-yl)amino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 366.4 171 1-(5-((2- (difluoromethoxy)pyridin-3- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 423.1 172 1-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-2-methyl-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 428.1 173 1-(5-((6-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-(prop-2-yn- 1-yl)urea 412.2 174 1-(cyanomethyl)-3-(5-((6-fluoro- 2,3-dihydrobenzo[b][1,4]dioxin- 5-yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 413.2 175 1-(5-((6-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 432.3 176 1-(cyanomethyl)-3-(7- (methylamino)-5-(1-(tetrahydro- 2H-pyran-4-yl)-1H-pyrrolo[2,3- b]pyridin-3-yl)pyrazolo[1,5- a]pyrimidin-3-yl)urea 446.2 177 1-((1-cyanocyclopropyl)methyl)- 3-(5-((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 435.2 178 1-(3,3-difluoropropyl)-3-(5-((7- fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 452.1 179 1-(3,3-difluoropropyl)-3-(5- ((2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 434.3 180 1-((1-cyanocyclopropyl)methyl)- 3-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 453.3 181 1-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl-2,2,3,3-d4)amino)-7-((methyl- d3)amino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 439.6 182 1-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl-2,2,3,3-d4)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 436.4 183 1-((1R,2S)-2- fluorocyclopropyl)-3-(7- (methylamino)-5-((4- oxochroman-8- yl)amino)pyrazolo[1,5- a]pyrimidin-3-yl)urea 426.1 184 1-(5-((4,4-difluorochroman-8- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 448.1 185 1-(5-(((S)-7-fluoro-2- (hydroxymethyl)-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 461.4 186 1-(5-(((R)-7-fluoro-2- (hydroxymethyl)-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2S)-2- fluorocyclopropyl)urea 461.4 187 1-(5-((7-fluoro-2,3- dihydrobenzo[b][1,4]dioxin-5- yl)amino)-7- (methylamino)pyrazolo[1,5- a]pyrimidin-3-yl)-3-((1R,2R)-2- fluorocyclopropyl)urea 431.9

Example 12: Co-Stimulation Assay in Lysed Whole Blood; JAK2: GM-CSF Stimulated STAT5 Phosphorylation and JAK1/TYK2 Stimulated STAT1 Phosphorylation Assay Human Blood Lysis Using Abcam's RBC Lysis Buffer

Dilute RBC lysis buffer to 1× in distilled water. Add 2 mL blood to 38 mL of 1×RBC-lysis buffer. Incubate for 15 mins at RT, in dark. Spin at 300 g, 5 mins, to collect the pellet. Re-lyse if necessary. Re-suspend pellet in 5 mL of cRPMI.

Compound and Cytokine Treatment

Aliquot 80 μL of lysed human blood in to wells of 96 deep-well plate. Add 10 μl of (10×conc.) of different concentrations of compounds to all wells except controls (unstained and unstimulated) and mix it with the help of 100 uL multichannel. Add 10 uL of RPMI media in controls. For dilution of compounds and dilution range please refer Appendix. Incubate on water bath or CO2 incubator for 1 hour at 37° C. Add 10 l of (10×conc.) of cytokine mix (GM-CSF and IFNa) (final conc. 10 ng/mL of GM-CSF and 100 ng/mL of IFNa) to each well except unstimulated and unstained controls and incubate further for 20 minutes on water bath at 37° C.

RBC Lysis and Fixation

Add 900 μL of prewarmed 1×Fix/Lyse solution (Appendix) and mix it properly using 1000 μl multichannel, incubate further on water bath at 37° C. for 10 minutes (which includes time of addition). Centrifuge at 800×g for 5 minutes at 40° C.; remove 900 uL of supernatant and add 900 L of 1×PBS. Centrifuge at 800×g for 5 minutes at 40° C., remove 900 μL of supernatant. Wash one more time with 900 μL of PBS (optional) and resuspend pellets in 100 uL of PBS.

Permeabilization

Disrupt the pellet by gentle tapping and resuspend in 1000 μL of BD Phosflow Perm Buffer III and incubate plate on ice for 30 minutes. Centrifuge plate at 800×g for 5 minutes at 40° C. Wash two more times with 1000 μL of BD Pharmingen Stain Buffer.

Antibody Treatment

Disrupt the pellet by gentle tapping. Resuspend pellets in 100 uL of Stain Buffer and add 5 μL of pSTAT5_AF488 Ab and 5 uL of pSTAT1_PE in all wells except unstained control and mix properly using 200 μl multichannel, incubate overnight at 40° C. Add 900 μL of wash buffer and centrifuge at 1800 rpm for 3 minutes at 40° C. Wash one more time with 1000 μL of BD Pharmingen Stain Buffer. Finally resuspend the pellet in 300 uL of BD Pharmingen Stain Buffer. Transfer the cells to 96-well v-bottom plate and acquire the cells in Beckman Coulter CytExpert. Acquiring cells in Flow Cytometer: Keep the threshold value to 250 and cell concentration should not exceed 100-500 cells/μL. Acquire at least 5,000-10,000 cells.

APPENDIX Preparation of Reagents

RPMI 1640 Complete Medium: RPMI 1640 media+10% FBS.

Cytokine dilution: 1) GM-CSF Stock at 100 ug/mL. Prepare an intermediate dilution of 1 ug/mL by adding 2 uL of stock into 198 uL of cRPMI. Further dilute to 100 ng/mL by adding 100 uL of the intermediate stock to 900 uL of cRPMI. 2) IFNa Stock at 200 ug/mL. Dilute IFNa stock 1:200 by adding 5 uL of stock into the 1000 uL of 100 ng/mL GM-CSF working stock as above to give a combined working stock of 1000 ng/mL of IFNa and 100 ng/mL GM-CSF (10×). Keep it on ice until used.

Lyse/Fix buffer preparation: Dilute 5×Lyse/Fix buffer to 1×using MQ water and keep at 37° C. until used.

BD Phosflow perm buffer III: Keep on ice/fridge.

Compound Dilution

Final 10X concentration, concentration, Sample nM nM Dilution 1 10,000 100,000 2 μL of 10 mM compound + 198 μL of cRPMI media 2 3333.3 33,333 60 μL of A + 120 μL of cRPMI media 3 1111.1 11,111 60 μL of B + 120 μL of cRPMI media 4 370.4 3,704 60 μL of C + 120 μL of cRPMI media 5 123.5 1,235 60 μL of D + 120 μL of cRPMI media 6 41.2 412 60 μL of E + 120 μL of cRPMI media 7 13.7 137 60 μL of F + 120 μL of cRPMI media 8 4.6 46 60 μL of G + 120 μL of cRPMI media 9 0 0 2 μL of DMSO + 198 μL of cRPMI media

IC50 values are shown in the table below.

IFN-a/Jak1 Tyk2 Compound IC50 5 A 7 A 10 A 13 A 17 B 19 B 22 B 25 A 26 A 27 A 28 C 29 B 30 C 31 C 32 C 33 A 34 B 35 A 36 A 37 A 38 B 39 A 40 A 41 A 42 A 43 B 44 A 45 A 46 B 47 A 48 C 49 A 50 A 51 A 52 B 53 C 54 C 55 C 56 C 57 B 58 C 59 B 60 C 61 C 62 B 63 B 64 A 65 A 66 B 67 B 68 A 69 A 70 A 71 A 72 A 73 A 74 B 75 A 76 A 77 A 78 A 79 B 80 A 81 A 82 B 83 A 84 B 85 A 86 B 87 A 88 B 89 A 90 B 91 B 92 B 93 A 94 B 95 B 96 A 97 B 98 B 99 A 100 B 101 A 102 A 103 B 104 A 105 A 106 B 107 B 108 A 109 A 110 C 111 A 112 A 113 A 114 A 115 B 116 B 117 A 118 A 119 A 120 A 121 A 122 A 123 A 124 A 125 B 126 B 127 A 128 A 129 A 130 A 131 A 132 A 133 B 134 B 135 B 136 B 137 A 138 A 139 A 140 C 141 B 142 A 143 A 144 A 145 B 146 B 147 A 148 A 149 A 150 A 151 B 152 A 153 A 154 A 155 B 156 C 157 A 158 A 159 C 160 A 161 B 162 B 163 B 164 A 165 C 166 A 167 B 168 B 169 A 170 B 171 B 172 C 173 B 174 C 175 C 176 A 177 A 178 A 179 A 180 C 181 A 182 A 183 A 184 B 185 B 186 B 187 B A: IC50 < 1 uM; B: IC50 ≥ 1 uM and < 10 uM; C: IC50 ≥ 10 uM

Example 13: Human Liver Microsome Stability Assay

The microsome stability assay was carried out in 96-well microtiter plates at 37° C. Reaction mixtures (25 μL) contained a final concentration of 1 μM test compound, 0.5 mg/mL human liver microsomes protein, and 1 mM NADPH and/or 1 mM UDPGA (with alamethicin) in 100 mM potassium phosphate, pH 7.4 buffer with 3 mM MgCl2. At each of the time points (for example, 0, 15, 30, and 60 minutes), 150 μL of quench solution (100% acetonitrile with 0.1% formic acid) with internal standard was transferred to each well. Besides the zero minute controls, mixtures containing the same components except the NADPH were also prepared as the negative control. Verapamil was included as a positive control to verify assay performance. Plates were sealed, vortexed, and centrifuged at 4° C. for 15 minutes at 4000 rpm. The supernatant was transferred to fresh plates for LC/MS/MS analysis. Comparison data for microsome stability of urea compounds described herein versus the corresponding amide compounds are shown in Table 1. The ureas showed unexpected increased microsomal stability compared to the amides.

TABLE 1 Urea Corresponding Amide Microsome Stability, half >120 minutes 19 minutes life Microsome Stability, half >120 minutes 60 minutes life Microsome Stability, half >120 minutes 18 minutes life Microsome Stability, half >120 minutes <50 minutes life

Example 14: Rat Pharmacokinetics (PK)

Rat PK experiments were performed with an IV group and PO group included, with 3 rats per group. The formulation was solution for IV group and either solution or suspension formulation for the oral group(s). Plasma samples were harvested with K2EDTA as anticoagulant at 0.08, 0.25, 0.5, 1, 2, 4, 8, 12 and 24 hours for the IV group and at 0.25, 0.5, 1, 2, 4, 6, 8, 12 and 24 hours for the PO group, and analyzed by fit for purpose LC-MS/MS method. Non-compartmental analysis module in Phoenix WinNonlin® (Version 8.0) was used to assess the pharmacokinetic parameters. Maximum concentration (Cmax) were taken from the observed values. The ureas showed unexpected increased Cmax compared to the amides.

TABLE 2 Urea Corresponding Amide Cmax 1,688 ng/mL 400 ng/mL Cmax 5,000 ng/mL 1,900 ng/mL Cmax 1,800 ng/mL 400 ng/mL Cmax 915 ng/mL 200 ng/mL

Example 15: Hepatocyte Stability

Metabolic stability of compounds can be evaluated using human, rat, mouse, or other animal hepatocytes to predict intrinsic clearance. Human LiverPool™ 20-donor and animal cryopreserved hepatocytes were obtained from BioreclamationIVT.

Cryopreserved hepatocytes were removed from a liquid nitrogen tank and thawed in a 37° C. water bath. As soon as the cells pull away from the vial wall, they were decanted into 48 mL of warm HT medium. Cells were centrifuged for four minutes at 420 rpm (50 g). After removing the supernatant, the pellet was re-suspended in warm DMEM medium. Cell density was counted by a hemacytometer.

The assay was carried out in 96-well microtiter plates. Compound 5 was incubated for 0, 60, 120, and 180 minutes at 37° C. with hepatocytes. Reaction mixtures (50 μL) contain a final concentration of 1 μM test compound, 0.5 million cells/mL hepatocytes in the DMEM medium. At each of the time points (for example, 0, 1, 2, and 3 hours), 200 μL of quench solution (100% acetonitrile with 0.1% formic acid) with internal standard was transferred to each well. Midazolam was included as a positive control to verify assay performance. Plates were sealed and centrifuged at 4° C. for 15 minutes at 4000 rpm. The supernatant was transferred to fresh plates for LC/MS/MS analysis.

All samples were analyzed on LC/MS/MS using an AB Sciex API 4000 instrument, coupled to a Shimadzu LC-20AD LC Pump system. Analytical samples were separated using a Waters Atlantis T3 dC18 reverse phase HPLC column (20 mm×2.1 mm) at a flow rate of 0.5 mL/min. The mobile phase consists of 0.1% formic acid in water (solvent A) and 0.1% formic acid in 100% acetonitrile (solvent B).

The extent of metabolism was calculated as the disappearance of the test compound, compared to the 0-min control reaction incubations. Initial rates are calculated for the compound concentration and used to determine t1/2 values and subsequently, the intrinsic clearance, CLint=(0.693)(1/t1/2 (min))(mL incubation/million cells). As shown in Table 3, the urea compound (Compound 5) had an unexpected high metabolic stability compared to the corresponding amide compound.

TABLE 3 Urea Corresponding Amide Human Microsomal Stability, >360 minutes <100 minutes half life

The examples and embodiments described herein are for illustrative purposes only and in some embodiments, various modifications or changes are to be included within the purview of disclosure and scope of the appended claims.

Claims

1. A compound having the structure of Formula (I′):

wherein: X is N or C(R6); Y is C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, or
 R7 wherein C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, or C2-C9heteroaryl are optionally substituted by 1, 2, 3, or 4 R7;
L1 is a bond, —O—, —C(O)—, —N(R9)—, —N(R9)CH2—, or —CH2N(R9)—;
R1 is C3-C6cycloalkyl substituted by 1, 2, or 3 R13;
R2, R3, and R4 are independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
R5 is hydrogen or C1-C6alkyl;
each R6 is independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
each R7 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; or two R7 are combined to form a 5-, 6-, or 7-membered heterocycloalkyl ring or a 4-, 5-, or 6-membered cycloalkyl ring;
R8 is hydrogen, deuterium, halogen, C1-C6alkyl, or C1-C6deuteroalkyl;
R9 is hydrogen or C1-C6alkyl;
each R10 is independently selected from hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6heteroalkyl, and phenyl, wherein phenyl is optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R2)2;
each R1 is independently selected from C1-C6alkyl and C1-C6heteroalkyl;
each R12 is independently selected from hydrogen, C1-C6alkyl, and C1-C6haloalkyl;
each R13 is independently selected from halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; and
n is 0, 1, 2, 3, or 4;
or a pharmaceutically acceptable salt or solvate thereof.

2. The compound of claim 1, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C6-C10aryl optionally substituted with 1, 2, or 3 R7.

3. The compound of claim 1 or claim 2, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is phenyl optionally substituted with 1, 2, or 3 R7.

4. The compound of claim 1, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl optionally substituted with 1, 2, or 3 R7.

5. The compound of claim 4, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are optionally substituted with 1, 2, or 3 R7.

6. The compound of claim 1, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

7. The compound of claim 6, or a pharmaceutically acceptable salt or solvate thereof, wherein n is 0, 1, or 2.

8. The compound of any one of claims 1-7, or a pharmaceutically acceptable salt or solvate thereof, wherein each R7 is independently selected from halogen, C1-C6alkyl, C1-C6haloalkyl, and C1-C6alkoxy.

9. The compound of claim 1, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

10. The compound of claim 1, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

11. The compound of any one of claims 1-10, or a pharmaceutically acceptable salt or solvate thereof, wherein each R13 is independently selected from halogen, —OH, —CN, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, and oxo.

12. The compound of any one of claims 1-11, or a pharmaceutically acceptable salt or solvate thereof, wherein each R13 is independently selected from halogen, —OH, and —CN.

13. The compound of any one of claims 1-12, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclopropyl substituted by 1 R13.

14. The compound of any one of claims 1-12, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is cyclobutyl substituted by 1 or 2 R13.

15. The compound of any one of claims 1-11, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is

16. The compound of any one of claims 1-15, or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is a bond.

17. The compound of any one of claims 1-15, or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)—.

18. The compound of any one of claims 1-17, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are independently selected from hydrogen and C1-C6alkyl.

19. The compound of any one of claims 1-18, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6alkyl.

20. The compound of any one of claims 1-18, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6deuteroalkyl.

21. The compound of any one of claims 1-20, or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is hydrogen.

22. The compound of any one of claims 1-21, or a pharmaceutically acceptable salt or solvate thereof, wherein each R8 is hydrogen.

23. The compound of any one of claims 1-22, or a pharmaceutically acceptable salt or solvate thereof, wherein each R2 is hydrogen.

24. The compound of any one of claims 1-23, or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is hydrogen.

25. The compound of any one of claims 1-24, or a pharmaceutically acceptable salt or solvate thereof, wherein X is N.

26. The compound of any one of claims 1-24, or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(R6).

27. The compound of any one of claims 1-24, or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(H).

28. A compound selected from:

 or a pharmaceutically acceptable salt or solvate thereof.

29. A compound selected from:

 or a pharmaceutically acceptable salt or solvate thereof.

30. A compound having the structure of Formula (I) or Formula (II):

wherein: X is N or C(R6); Y is C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, or
w herein C1-C6alkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, or C2-C9heteroaryl are optionally substituted by 1, 2, 3, or 4 R7;
L1 is a bond, —O—, —C(O)—, —N(R9)—, —N(R9)CH2—, or —CH2N(R9)—;
R1 is selected from hydrogen, deuterium, C1-C6alkyl, C1-C6deuteroalkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, and C2-C9heteroaryl, wherein C1-C6alkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl are optionally substituted by 1, 2, or 3 R7;
R2, R3, and R4 are independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
R5 is hydrogen or C1-C6alkyl;
each R6 is independently selected from hydrogen, deuterium, C1-C6alkyl, and C1-C6deuteroalkyl;
each R7 is independently selected from deuterium, halogen, C1-C6alkyl, C1-C6deuteroalkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C1-C6heteroalkyl, C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl, oxo, —OR10, —N(R10)2, —CN, —C(═O)R11, —C(═O)OR10, —C(═O)N(R10)2, —NR10C(═O)R11, —NR10S(═O)2R11, —S(═O)2R11, and —S(═O)2N(R10)2, wherein C3-C6cycloalkyl, C2-C9heterocycloalkyl, C6-C10aryl, C2-C9heteroaryl are optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R12)2; or two R7 are combined to form a 5-, 6-, or 7-membered heterocycloalkyl ring or a 4-, 5-, or 6-membered cycloalkyl ring;
R8 is hydrogen, deuterium, halogen, C1-C6alkyl, or C1-C6deuteroalkyl;
R9 is hydrogen or C1-C6alkyl;
each R10 is independently selected from hydrogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6heteroalkyl, and phenyl, wherein phenyl is optionally substituted with 1, 2, or 3 groups selected from halogen, C1-C6alkyl, C1-C6haloalkyl, C1-C6alkoxy, C1-C6haloalkoxy, C2-C9heterocycloalkyl, C2-C9heteroaryl, —OR12, —N(R12)2, —C(═O)OR12, and —C(═O)N(R2)2;
each R11 is independently selected from C1-C6alkyl and C1-C6heteroalkyl;
each R12 is independently selected from hydrogen, C1-C6alkyl, and C1-C6haloalkyl; and
n is 0, 1, 2, 3, or 4;
or a pharmaceutically acceptable salt or solvate thereof.

31. The compound of claim 30, or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (I):

32. The compound of claim 30, or a pharmaceutically acceptable salt or solvate thereof, having the structure of Formula (II):

33. The compound of any one of claims 30-32, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C6-C10aryl optionally substituted with 1, 2, or 3 R7.

34. The compound of claim 33, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is phenyl optionally substituted with 1, 2, or 3 R7.

35. The compound of any one of claims 30-34, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl optionally substituted with 1, 2, or 3 R7.

36. The compound of claim 35, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is C2-C9heteroaryl selected from oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl, wherein oxazolyl, thiazolyl, pyrazolyl, furanyl, thienyl, pyrrolyl, imidazolyl, triazolyl, isoxazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and pyridazinyl are optionally substituted with 1, 2, or 3 R7.

37. The compound of an one of claims 30-32, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

38. The compound of claim 37, or a pharmaceutically acceptable salt or solvate thereof, wherein n is 0, 1, or 2.

39. The compound of any one of claims 30-38, or a pharmaceutically acceptable salt or solvate thereof, wherein each R7 is independently selected from halogen, C1-C6alkyl, C1-C6haloalkyl, and C1-C6alkoxy.

40. The compound of any-one of claims 30-32, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

41. The compound of any one of claims 30-32, or a pharmaceutically acceptable salt or solvate thereof, wherein Y is

42. The compound of any one of claims 30-41, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is hydrogen, C1-C6alkyl, C1-C6deuteroalkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, and C2-C9heteroaryl, wherein C1-C6alkyl, C3-C6cycloalkyl, —C1-C6alkyl-C3-C6cycloalkyl, C2-C9heterocycloalkyl, or C2-C9heteroaryl are optionally substituted by 1, 2, or 3 R7.

43. The compound of any one of claims 30-42, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is hydrogen.

44. The compound of any one of claims 30-42, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C1-C6alkyl optionally substituted by 1, 2, or 3 R7.

45. The compound of claim 44, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is unsubstituted C1-C6alkyl.

46. The compound of any one of claims 30-42, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C3-C6cycloalkyl optionally substituted by 1, 2, or 3 R7.

47. The compound of claim 46, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is unsubstituted C3-C6cycloalkyl.

48. The compound of any one of claims 30-42, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C2-C9heterocycloalkyl optionally substituted by 1, 2, or 3 R7.

49. The compound of any one of claims 30-42, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is C2-C9heteroaryl optionally substituted by 1, 2, or 3 R7.

50. The compound of any one of claims 30-49, or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is a bond.

51. The compound of any one of claims 30-49, or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is —N(H)—.

52. The compound of any one of claims 30-51, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are independently selected from hydrogen and C1-C6alkyl.

53. The compound of any one of claims 30-52, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6alkyl.

54. The compound of any one of claims 30-52, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is hydrogen and R4 is C1-C6deuteroalkyl.

55. The compound of any one of claims 30-54, or a pharmaceutically acceptable salt or solvate thereof, wherein R6 is hydrogen.

56. The compound of any one of claims 30-55, or a pharmaceutically acceptable salt or solvate thereof, wherein each R8 is hydrogen.

57. The compound of any one of claims 30-56, or a pharmaceutically acceptable salt or solvate thereof, wherein each R2 is hydrogen.

58. The compound of any one of claims 30-57, or a pharmaceutically acceptable salt or solvate thereof, wherein R5 is hydrogen.

59. The compound of any one of claims 30-58, or a pharmaceutically acceptable salt or solvate thereof, wherein X is N.

60. The compound of any one of claims 30-58, or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(R6).

61. The compound of any one of claims 30-58, or a pharmaceutically acceptable salt or solvate thereof, wherein X is C(H).

62. A compound selected from:

 or a pharmaceutically acceptable salt or solvate thereof.

63. A compound selected from:

 or a pharmaceutically acceptable salt or solvate thereof.

64. A pharmaceutical composition comprising a compound of any one of claims 1-63, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient.

65. A method of treating an inflammatory or autoimmune disease in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of any one of claims 1-63, or a pharmaceutically acceptable salt or solvate thereof.

66. The method of claim 65, wherein the disease is selected from rheumatoid arthritis, multiple sclerosis, psoriasis, lupus, intestinal bowel disease, Crohn's disease, ulcerative colitis, ankylosing spondylitis, vitiligo, and atopic dermatitis.

Patent History
Publication number: 20230348478
Type: Application
Filed: Jul 6, 2023
Publication Date: Nov 2, 2023
Inventors: Raju MOHAN (Encinitas, CA), John NUSS (Encinitas, CA), Jason HARRIS (Encinitas, CA), Shendong YUAN (Encinitas, CA)
Application Number: 18/348,296
Classifications
International Classification: C07D 487/04 (20060101); C07D 519/00 (20060101);