Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS AND COMPOSITIONS FOR TARGETING PD-L1
Document Type and Number:
WIPO Patent Application WO/2023/114365
Kind Code:
A1
Abstract:
The present disclosure related to compounds that can be useful as inhibitors of PD-1, PD-L1 or the PD-1/PD-L1 interaction. Also disclosed herein are pharmaceutical compositions of that can include a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and uses of or methods of using a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for the treatment of PD-L1 related diseases including but not limited to liver diseases, cancer, hepatocellular carcinoma, viral diseases, or hepatitis B.

Inventors:
WU TONGFEI (US)
RABOISSON PIERRE JEAN-MARIE BERNARD (US)
GONZALVEZ FRANCOIS (US)
Application Number:
PCT/US2022/052956
Publication Date:
June 22, 2023
Filing Date:
December 15, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ALIGOS THERAPEUTICS INC (US)
International Classes:
C07D401/14; A61K31/501; A61K31/513; A61P31/20; A61P35/00; C07D401/12; C07D403/14; C07D405/14; C07D413/14; C07D453/06; C07D487/10; C07D498/04
Domestic Patent References:
WO2020086556A12020-04-30
WO2020011209A12020-01-16
WO2019204609A12019-10-24
WO2019160882A12019-08-22
WO2020232256A12020-11-19
WO2020257549A22020-12-24
Attorney, Agent or Firm:
MILLER, Kimberly J. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A compound of Formula (I), or a pharmaceutically acceptable salt thereof, having the structure: wherein:

R13 is selected from the group consisting of -CM alkyl, -C1-4 haloalkyl, -CEbfCs-e monocyclic cycloalkyl), -CH2(4-6 membered monocyclic heterocyclyl) and -CH2(5-6 membered monocyclic heteroaryl);

Rlb is selected from the group consisting of hydrogen, -CM alkyl, -CM haloalkyl;

R3C is selected from the group consisting of-N(Rrai)Rni and -RXI;

R2a, R2b, R2C, R2e, R22g, R2h are jndependently selected from the group consisting of hydrogen and halogen; R2u and R21 is independently selected from hydrogen, halogen, cyano, CH3 CH2CH3. -CH2OH, -OCH3 and SCI k.

R'3 is selected from the group consisting of hydrogen, -CH3, -CF3 and -CHF2;

R3b is selected from the group consisting of hydrogen, halogen, cyano, -CH3, -CF3, -CHF2, -CH2CH3, CH2CH3, OCH3 and -SCH3;

Yla is selected from the group consisting of N (nitrogen) and -CH;

Ylb is selected from the group consisting of N (nitrogen) and C(R4);

Y2a is selected from the group consisting of N (nitrogen) and C(R5);

Y2b is selected from the group consisting of N (nitrogen) and C(R‘a), wherein Rld is selected from the group consisting of hydrogen and halogen;

R4 is selected from the group consisting of hydrogen, halogen and -CH3;

R’ is selected from the group consisting of hydrogen, halogen and -CH3 RZ3 is selected from the group consisting of hydrogen, -CM alkyl and -CM haloalkyl; and

RZ4 is selected from the group consisting of hydrogen, -CM alkyl and 5- to 6-membered monocyclic heterocyclyllCH?)- optionally substituted with -CM alkyl.

2. The compound of Claim 1, wherein Y2a is N.

3. The compound of Claim 1, wherein Y?a is C(R5).

4. The compound of Claim 3, wherein R5 is hydrogen.

5. The compound of Claim 3, wherein R? is halogen.

6. The compound of Claim 3, wherein R5 is -CHs.

7. The compound of any one of Claims 1-6, wherein R,a is hydrogen.

8. The compound of any one of Claims 1-6, wherein R ';! is -CHs.

9. The compound of any one of Claims 1-6, wherein R,a is -CF3.

10. The compound of any one of Claims 1-6, wherein R ';! is -CHF;?.

11. The compound of any one of Claims 1-10, wherein R3b is hydrogen.

12. The compound of any one of Claims 1-10, wherein Rjb is halogen.

13. The compound of any one of Claims 1-10, wherein R3b is cyano.

14. The compound of any one of Claims 1-10, wherein Rjb is -CH3 or -CFbCEh-

15. The compound of any one of Claims 1 - 10, wherein R,b is -CF3 or -CHF2.

16. The compound of any one of Claims 1-10, wherein R3b is -CH2OH, -OH, OCI h or SCI h

17. The compound of any one of Claims 1-16, wherein Yia is N.

18. The compound of any one of Claims 1-16, wherein Y 13 is -CH.

19. The compound of any one of Claims 1-18, wherein Ylb is N.

20. The compound of any one of Claims 1-18, wherein Y:° is C(R4).

21. The compound of Claim 20, wherein R4 is hydrogen.

22. The compound of Claim 20, wherein R4 is halogen.

23. The compound of Claim 20, wherein R4 is -CH3.

24. The compound of any one of Claims 1-23, wherein R,a is -CM alkyl.

25. The compound of any one of Claims 1-23, wherein Rla is -CM haloalkyl.

26. The compound of any one of Claims 1-23, wherein R,a is selected from the group consisting of -CHz(C3-6 monocyclic cycloalkyl), ~CHz(4-6 membered monocyclic heterocyclyl) and ---CH2.(5-6 membered monocyclic heteroaryl).

27. The compound of any one of Claims 1-26, wherein Rld is hydrogen.

28. The compound of any one of Claims 1-26, wherein R,d is halogen.

29. The compound of any one of Claims 1-28, wherein Rlc is ---N(Rm,)Rni.

30. The compound of Claim 29, wherein Rnl is hydrogen.

31. The compound of Claim 29, wherein Rn1 is -C1-4 alkyl.

32. The compound of Claim 29, wherein Rni is -C1 -4 haloalkyl, C3-6 monocyclic cycloalkyl(CH2)- or -C(=O)ORZ4

33. The compound of any one of Claims 29-32, wherein Rml is tetrahydrofuran or tetrahydro-2H-pyran, each optionally substituted with hydroxy.

41. The compound of any one of Claims 1-40, wherein R2a, R2b, R2c, R2e, R2g and R2h are each hydrogen.

42. The compound of any one of Claims 1 -40, wherein R2a, R2e, R2e, R2g and R2b are each hydrogen; and R2b halogen.

43. The compound of any one of Claims 1-40, wherein R'T R2c, R2e, R2g and R2h are each hydrogen; and R2a halogen.

44. The compound of any one of Claims 1-43, wherein R2d and R2f are each halogen.

45. The compound of any one of Claims 1-43, wherein R2d and R2f are each -CH3.

46. The compound of any one of Claims 1-43, wherein R2d is halogen; and R2f is - CHs.

47. The compound of any one of Claims 1 -43, wherein R2d is -CHs; and R2! is halogen,

48. The compound of any one of Claims 1-43, wherein R2d is cyano; and R2f is halogen,

49. The compound of any one of Claims 1-43, wherein R2d is -OCRs; and R2f is halogen,

50. The compound of any one of Claims 42-44 or 46-49, wherein the halogen is chloro or fluoro.  

the foregoing.

54. A pharmaceutical composition comprising an effective amount of a compound of any one of Claims 1-53, or a pharmaceutically acceptable salt thereof, and excipient.

55. A method for treating hepatitis B in a subject comprising administering to the subject in need thereof an effective amount of a compound of any one of Claims 1-53, or a pharmaceutically acceptable salt thereof

56. A method for treating hepatocellular carcinoma (HCC) in a subject comprising administering to the subject in need thereof an effective amount of a compound of any one of Claims 1-53, or a pharmaceutically acceptable salt thereof.

57. The method of any one of Claims 55-56, further comprising administering surgery, radiation therapy, chemotherapy, targeted therapy, immunotherapy, hormonal therapy, or antiviral therapy.

58. A compound of any one of Claims 1-53, or a pharmaceutically acceptable salt thereof, for use in treating hepatitis B.

59. A compound of any one of Claims 1-53, or a pharmaceutically acceptable salt thereof, for use in treating hepatocellular carcinoma (HCC).

60. The compound of any one of Claims 58-59, further comprising administering surgery, radiation therapy, chemotherapy, targeted therapy, immunotherapy, hormonal therapy, or antiviral therapy.

61. Use of a compound of any one of Claims 1-53, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for use in treating hepatitis B.

62. Use of a compound of any one of Claims 1-53, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for use in treating hepatocellular carcinoma (HCC).

63. The use of any one of Claims 61 -62, further comprising administering surgery, radiation therapy, chemotherapy, targeted therapy, immunotherapy, hormonal therapy, or antiviral therapy.

Description:
METHODS AND COMPOSITIONS FOR TARGETING PD-Ll

INCORPORATION BY REFERENCE TO ANY PRIORITY APPLICATIONS

[0001] Any and all applications for which a foreign or domestic priority claim is identified, for example, in the Application Data Sheet or Request as filed with the present application, are hereby incorporated by reference under 37 CFR 1.57, and Rules 4.18 and 20.6, including U.S. Provisional Application No. 63/265,510, filed December 16, 2021.

FIELD

[0002] The present application relates to the fields of chemistry, biochemistry, molecular biology and medicine. The present disclosure related to compounds that can be useful as inhibitors of PD-1, PD-L1 or the PD-1 /PD-Ll interaction. Also disclosed herein are pharmaceutical compositions of compounds described herein and uses of or methods of using the compounds for the treatment of PD-Ll related diseases including but not limited to liver diseases, cancer, hepatocellular carcinoma, viral diseases, or hepatitis B.

BACKGROUND

[0003] The programmed cell death 1 (PD-1) immune checkpoint expressed on the surface of activated CD4‘ f and CD8 + T cells controls an inhibitory mechanism to prevent autoimmunity. Engagement of PD-1 by programmed death-ligand 1 (PD-Ll) expressed on the multitude of cell types, including macrophages, dendritic cells, mast cells as well as cancer cells induces T cell exhaustion resulting in reduction or loss of effector cytokine production (e.g, 11,-2, TNF-a, IFN-y) and upregulation of other inhibitory receptors and immune checkpoints (e.g. CTLA-4, LAG-3, and BTLA), or T cell apoptosis. High expression of PD- Ll is exhibited by many types of cancers to escape tumor immune surveillance and has been associated with poorer prognosis. PD-1 -mediated immunosuppression is also linked to some viral infections, such as hepatitis B. There is an ongoing need for PD-1/PD-L1 therapies and improvements thereof for the treatment of disease. SUMMARY

[0004] Some embodiments disclosed herein relate to a compound of Formula (I), or a pharmaceutically acceptable salt thereof.

[0005] Some embodiments disclosed herein relate to a pharmaceutical composition that can contain an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof.

[0006] Some embodiments described herein relate to a method of treating a HBV and/or HDV infection that can include administering to a subject identified as suffering from the HBV and/or HDV infection an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein, or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein. Other embodiments described herein relate to a compound, or a pharmaceutically acceptable salt thereof, as described herein, or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein for the use of treating a HBV’ and/or HDV infection.

[0007] Some embodiments disclosed herein relate to a method of inhibiting replication of HBV and/or HDV that can include contacting a cell infected with the HBV and/or HDV with an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein, or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein. Other embodiments described herein relate to a compound, or a pharmaceutically acceptable salt thereof, as described herein, or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein for the use of inhibiting the replication HBV and/or HDV.

[0008] These are other embodiments are described in greater detail below.

DETAILED DESCRIPTION

[0009] Hepatocellular carcinoma (HCC) is the most common form of liver cancer. HCC can be caused by a variety of conditions, such as alcohol consumption, cirrhosis, and viral infections that cause hepatitis, such as hepatitis B virus, hepatitis C virus, and hepatitis D virus. The inflammation, fibrosis, and cirrhosis linked with these conditions can induce malignancies in affected liver cells. HCC has relatively poor prognosis, with a five-year survival rate of about 30%, depending on if full surgical resection of the tumor is possible.

[0010] For early disease, surgical resection is used. However, most HCC are identified at later stages because of difficulties in diagnosing. Upon late-stage diagnosis, the tumors are unresectable, and most patients are given systemic therapies. The current standard of care in front line are multi-kinase inhibitors (including, for example, sorafenib and/or lenvatinib). Most patients are refractory or relapse from these treatments and undergo second line therapies that have anti-angiogenic agents (including, for example, Regorafmib, Cabozantinib, and/or Ramicirumab) or immune checkpoint inhibitors (including, for example, nivolumab and/or pembrolizumab). However, most patients do not respond to first and second therapies, and the clinical benefit is poor, with overall survival not exceeding one year. In addition, biomarker driven therapies are lacking. Thus, there is a need to develop more tolerable and efficacious therapies for the treatment of HCC and related liver disorders.

[0011] HBV is a partially double-stranded circular DNA of about 3.2 kilobase (kb) pairs, and is classified into eight genotypes, A to H. The HBV replication pathway has been studied in great detail. One part of replication includes the formation of the covalently closed circular DNA (cccDNA) form. The presence of the cccDNA gives rise to the risk of viral reemergence throughout the life of the host organism. HBV carriers can transmit the disease for many years. An estimated 300 million people are living with hepatitis B virus infection, and it is estimated that over 750,000 people worldwide die of hepatitis B each year. In addition, immunosuppressed individuals or individuals undergoing chemotherapy are especially at. risk for reactivation of an HBV infection. HBV can be acute and/or chronic. Acute HBV infection can be either asymptomatic or present with symptomatic acute hepatitis.

[0012] HBV can be transmitted by blood, semen, and/or another body fluid. This can occur through direct blood-to-blood contact, unprotected sex, sharing of needles, and from an infected mother to her baby during the delivery process. The HBV surface antigen (HBsAg) is most frequently used to screen for the presence of this infection. Currently available medications do not cure HBV and/or HDV infection. Rather, the medications suppress replication of the virus.

[0013] The hepatitis D virus (HDV) is a DNA virus, also in the Hepadnaviridae family of viruses. HDV can propagate only in the presence of HBV. The routes of transmission of HDV are similar to those for HBV. Transmission of HDV can occur either via simultaneous infection with HBV (coinfection) or in addition to chronic hepatitis B or hepatitis B carrier state (superinfection). Both superinfection and coinfection with HDV results in more severe complications compared to infection with HB V alone. These complications include a greater likelihood of experiencing liver failure in acute infections and a rapid progression to liver cirrhosis, with an increased risk of developing liver cancer in chronic infections. In combination with hepatitis B, hepatitis D has the highest fatality rate of all the hepatitis infections, at 20%. There is currently no cure or vaccine for hepatitis D.

[0014] Programmed cell death 1, or programmed death 1 (PD-1) is a 268 amino acid long type I transmembrane protein found as a surface marker on T cells and other immune cells. As an immune checkpoint, PD-1 selves to negatively regulate immune responses to prevent autoimmune disorder. PD-1 protein (NCBI accession number NP 005009.2) is expressed from the cluster of differentiation 279 (CD279) gene (NCBI accession number NG-012110.1) or mRNA transcript (NCBI accession number NM_005018.3). In some preferred embodiments, PD-1 is the human PD-1 protein, and CD279 is the human CD279 transcript or gene on chromosome 2. It should be understood that a person with ordinary skill in the art would view' the terms PD-1 and CD279 as often nominally interchangeable when considering the nucleic acid (DNA or RNA) or corresponding translated protein, or the sequences thereof.

[0015] Programmed cell death-ligand 1, or programmed death-ligand 1 (PD-L1), also known as B7 homolog 1 (B7-H1) is 272 amino acid long type I transmembrane protein found as a surface marker on many different cell types. PD-L1 is a major ligand of PD-1 and results in inhibition of T cell cytotoxicity and cytokine production. Cancer cells such as HCC cells take advantage of this immune checkpoint by upregulating PD-L1 expression, resulting in dysfunctional anti-tumor immunity by proximal T cells. Viruses also have been observed to modulate the PD-1/PD-L1 pathway to inhibit immune host response. Hepatitis B virus has been shown to upregulate PD-L1 in infected hepatocytes, and PD-1 in associated T cells. PD- L1 protein (NCBI accession number NP 054862,1 ) is expressed from the cluster of differentiation 274 (CD274) transcript (NCBI accession number NM 014143.4). In some preferred embodiments, PD-L1 is the human PD-L1 protein, and CD274 is the human CD274 transcript or gene on chromosome 9. It should be understood that a person with ordinary skill in the art would view the terms PD-L1 and CD274 as often nominally interchangeable when considering the nucleic acid (DNA or RNA) or corresponding translated protein, or the sequences thereof.

Definitions

[0016] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art. All patents, applications, published applications and other publications referenced herein are incorporated by reference in their entirety unless stated otherwise. To the extent publications and patents or patent applications incorporated by reference contradict the disclosure contained in the specification, the specification is intended to supersede and/or take precedence over any such contradictory material. In the event that there are a plurality of definitions for a term herein, those in this section prevail unless stated otherwise.

[0017] Whenever a group is described as being “optionally substituted” that group may be unsubstituted or substituted with one or more of the indicated substituents. Likewise, when a group is described as being “unsubstituted or substituted” if substituted, the substituent(s) may be selected from one or more of the indicated substituents. If no substituents are indicated, it is meant that the indicated “optionally substituted” or “substituted” group maybe substituted with one or more group(s) (such as 1, 2 or 3 groups) individually and independently selected from deuterium, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl), (heterocyclyl)alkyl, hydroxy, alkoxy, acyl, cyano, halogen, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, O-carboxy, isocyanate, thiocyanate, isothiocyanate, nitro, azido, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, an amino, a mono-substituted amine and a di-substituted amine.

[0018] As used herein, “C a to GV’ or “C a -b” in which “a” and “b” are integers refer to the number of carbon atoms in an alkyl, alkenyl or alkynyl group, or the number of carbon atoms in the ring of a cycloalkyl, cycloalkenyl, aryl, heteroaryl or heterocyclyl group. That is, the alkyl, alkenyl, alkynyl, ring of the cycloalkyl, ring of the cycloalkenyl, ring of the aryl, ring of the heteroaryl or ring of the heterocyclyl can contain from “a” to “b”, inclusive, carbon atoms. Thus, for example, a “Ci to C4 alkyl” or a “C1-C4 alkyl” group refers to all alkyl groups having from I to 4 carbons, that is, CH 3 -, CH 3 CH 2 -, CH 3 CH 2 CH 2 -, (CH 3 ) 2 .CH-, CH 3 CH 2 CH 2 CH 2 -, CH 3 CH 2 CH (CH 3 )• and (CH 3 )3C-. If no “a” and “b” are designated with regard to an alkyl, alkenyl, alkynyl, cycloalkyl cycloalkenyl, aryl, heteroaryl or heterocyclyl group, the broadest range described in these definitions is to be assumed.

[0019] As used herein, “alkyl” refers to a straight or branched hydrocarbon chain that comprises a fully saturated (no double or triple bonds) hydrocarbon group. The alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as “1 to 20” refers to each integer in the given range; e.g., “1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated). The alkyl group may also be a medium size alkyl having 1 to 10 carbon atoms. The alkyl group could also be a lower alkyl having 1 to 6 carbon atoms. The alkyl group of the compounds may be designated as “C 1 -C 4 alkyl” or similar designations. By way of example only, “C 1 -C 4 alkyl” indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl and t-butyl . Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl and hexyl. The alkyl group may be substituted or unsubstituted.

[0020] As used herein, “alkenyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more double bonds. The length of an alkenyl can vary. For example, the alkenyl can be a C 2-4 alkenyl, C 2-6 alkenyl or C 2-8 alkenyl. Examples of alkenyl groups include allenyl, vinylmethyl and ethenyl. An alkenyl group may be unsubstituted or substituted.

[0021] As used herein, “alkynyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more triple bonds. The length of an alkynyl can vary. For example, the alkynyl can be a C 2-4 alkynyl, C 2-6 alkynyl or C 2-8 alkynyl. Examples of alkynyls include ethynyl and propynyl. An alkynyl group may be unsubstituted or substituted.

[0022] As used herein, “cycloalkyl” refers to a completely saturated (no double or triple bonds) mono- or multi- cyclic hydrocarbon ring system. When composed of two or more rings, the rings may be joined together in a fused fashion. Cycloalky I groups can contain 3 to 10 atoms in the rmg(s), 3 to 8 atoms in the ring(s) or 3 to 6 atoms in the ring(s). A cycloalkyl group may be unsubstituted or substituted. Typical cycloalkyl groups include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.

[0023] As used herein, “cycloalkenyl” refers to a mono- or multi- cyclic hydrocarbon ring system that contains one or more double bonds in at least one ring; although, if there is more than one, the double bonds cannot form a fully delocalized pi-electron system throughout all the rings (otherwise the group would be “aryl,” as defined herein). When composed of two or more rings, the rings may be connected together in a fused fashion. A cycloalkenyl can contain 3 to 10 atoms in the ring(s) or 3 to 8 atoms in the ring(s). A cycloalkenyl group may be unsubstituted or substituted.

[0024] As used herein, “aryl” refers to a carbocyclic (all carbon) monocyclic or multicyclic aromatic ring system (including fused ring systems where two carbocyclic rings share a chemical bond) that has a fully delocalized pi-electron system throughout all the rings. The number of carbon atoms in an aryl group can vary. For example, the aryl group can be a Ce-Cu aryl group, a Ce-Cio aryl group, or a Ce aryl group. Examples of aryl groups include, but are not limited to, benzene, naphthalene and azulene. An aryl group may be substituted or unsubstituted.

[0025] As used herein, “heteroaryl” refers to a monocyclic, bicyclic and tricyclic aromatic ring system (a ring system with fully delocalized pi-electron system) that contain(s) one or more heteroatoms (for example, 1 to 5 heteroatoms), that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur. The number of atoms in the rmg(s) of a heteroaryl group can vary'. For example, the heteroaryl group can contain 4 to 14 atoms in the ring(s), 5 to 10 atoms in the ring(s) or 5 to 6 atoms in the ring(s). Furthermore, the term “heteroaryl” includes fused ring systems where two rings, such as at least one aryl ring and at least one heteroaryl ring, or at least two heteroaryl rings, share at least one chemical bond. Examples of heteroaryl rings include, but are not limited to, furan, furazan, thiophene, benzothiophene, phthalazine, pyrrole, oxazole, benzoxazole, 1,2,3-oxadiazole, 1,2,4- oxadiazole, thiazole, 1,2,3-thiadiazole, 1,2,4-thiadiazole, benzothiazole, imidazole, benzimidazole, indole, indazole, pyrazole, benzopyrazole, isoxazole, benzoisoxazole, isothiazole, triazole, benzotriazole, thiadiazole, tetrazole, pyridine, pyridazine, pyrimidine, pyrazine, purine, pteridine, quinoline, isoquinoline, quinazoline, quinoxaline, cinnoline and triazine. A heteroaryl group may be substituted or unsubstituted.

[0026] As used herein, “heterocyclyl” refers to a monocyclic, bicyclic and tricyclic ring system wherein carbon atoms together with from I to 5 heteroatoms constitute said ring system. A heterocycle may optionally contain one or more unsaturated bonds situated in such a way, however, that a fully delocalized pi-electron system does not occur throughout all the rings. The number of atoms in the ring(s) of a heterocyclyl group can vary . For example, the heterocyclyl group can contain 4 to 14 atoms in the ring(s), 5 to 10 atoms in the ring(s) or 5 to 6 atoms in the ring(s). The heteroatom(s) is an element other than carbon including, but not limited to, oxygen, sulfur and nitrogen. A heterocycle may further contain one or more carbonyl or thiocarbonyl functionalities, so as to make the definition include oxo-systems and thio-systems such as lactams, lactones, cyclic imides, cyclic thioimides and cyclic carbamates. When composed of two or more rings, the rings may be joined together in a fused fashion. Additionally, any nitrogens in a heterocyclyl may be quaternized. Heterocyclyl groups may be unsubstituted or substituted. Examples of such “heterocyclyl groups include but are not limited to, 1,3-dioxin, 1,3-dioxane, 1,4-dioxane, 1 ,2-dioxolane, 1,3 -dioxolane, 1,4-dioxolane, 1,3-oxathiane, 1,4-oxathiin, 1,3 -oxathiolane, 1 ,3-dithiole, 1,3-dithiolane, 1,4-oxathiane, tetrahydro- 1,4-thiazme, 2H-1 ,2-oxazine, maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydro uracil, trioxane, hexahydro- 1 ,3,5- triazine, imidazoline, imidazolidine, isoxazoline, isoxazolidine, oxazoline, oxazolidine, oxazolidinone, thiazoline, thiazolidine, morpholine, oxirane, piperidine A'-Oxide, piperidine, piperazine, pyrrolidine, pyrrolidone, pyrrolidione, 4-piperidone, pyrazoline, pyrazolidine, 2- oxopyrrolidine, tetrahydropyran, 4H-pyran, tetrahydrothiopyran, thiamorpholine, thiamorpholine sulfoxide, thiamorpholine sulfone and their benzo-fused analogs (e.g., benzimidazolidinone, tetrahydroquinoline and 3,4-methylenedioxyphenyl).

[0027] As used herein, “aryl(alkyl)” refers to an aryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and aryl group of an aryl(alkyl) may be substituted or unsubstituted. Examples include but are not limited to benzyl, 2- phenyl(alkyl), 3-phenyl(alkyl), and naphthyl(alkyl).

[0028] As used herein, “heteroaryl(alkyl)” refer to a heteroaryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and heteroaryl group of heteroaryl(alkyl) may be substituted or unsubstituted. Examples include but are not limited to 2-thienyl(alkyl), 3-thienyl(alkyl), furyl(alkyl), thienyl(alkyl), pyrrolyl (alkyl), pyridyl(alkyl), isoxazolyl(alkyl), imidazolyl(alkyl) and their benzo-fused analogs.

[0029] A “(heterocyclyl)alkyl” refer to a heterocyclic group connected, as a substituent, via a lower alkylene group. The lower alkylene and heterocyclyl of a heterocyclyl(alkyl) may be substituted or unsubstituted. Examples include but are not limited tetrahydro- 2H-pyran-4-yl(methyl), piperidin-4-yl(ethyl), piperidin-4-yl(propyl), tetrahydro- 2H-thiopyran-4-yl(methyl) and 1 , 3 -thiazinan-4-yl(methyl).

[0030] “Lower alkylene groups” are straight-chained -CH2- tethering groups, forming bonds to connect molecular fragments via their terminal carbon atoms. In some embodiments, a lower alkylene can include 1, 2, 3, 4, 5 or 6 carbons. Examples include but are not limited to methylene (-CI I2-), ethylene (-CH2CH2-), propylene (-CH2CH2CH2-) and butylene (-CH2CH2CH2CH2-). A lower alkylene group can be substituted by replacing one or more hydrogen of the lower alkylene group with a substituent(s) listed under the definition of “substituted,”

[0031] As used herein, “alkoxy” refers to the formula -OR wherein R is an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl (alkyl) or heterocyclyl (alkyl) is defined herein. A non-limiting list of alkoxys are methoxy, ethoxy, n-propoxy, 1 -methylethoxy (isopropoxy), n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, phenoxy and benzoxy. In some instances, an alkoxy can be -OR wherein R is an unsubstituted C1-4 alkyl. An alkoxy may be substituted or unsubstituted.

[0032] As used herein, “acyl” refers to a hydrogen an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl (alkyl) or heterocyclyl(alkyl) connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl, benzoyl, and acryl. An acyl may be substituted or unsubstituted.

[0033] As used herein, “hydroxyalky!” refers to an alkyl group in which one or more of the hydrogen atoms are replaced by a hydroxy group. Exemplary hydroxyalkyl groups include but are not limited to, 2-hydroxy ethyl, 3 -hydroxypropyl, 2-hydroxypropyl and 2,2- dihydroxy ethyl. A hydroxyalkyl may be substituted or unsubstituted.

[0034] As used herein, “haloalkyl” refers to an alkyl group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkyl, di-haloalkyl and tri- haloalkyl). Such groups include but are not limited to, chloromethyl, fluoromethyl, difluoromethyl, trifluoromethyl, l-chloro-2-fluoromethyl and 2-fluoroisobutyl. A haloalkyl may be substituted or unsubstituted.

[0035] .As used herein, “haloalkoxy” refers to a O-alkyl group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkoxy, di- haloalkoxy and tn- haloalkoxy). Such groups include but are not limited to, chloromethoxy, fluoromethoxy, difluoromethoxy, trifluorom ethoxy, 1 -chloro-2-fluoromethoxy and 2-fluoroisobutoxy. In some instances, a haloalkoxy can be -OR, wherein R is a Ci-4 alkyl substituted by 1, 2 or 3 halogens. A haloalkoxy may be substituted or unsubstituted.

[0036] A “sulfenyl” group refers to an “-SR” group in which R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). A sulfenyl may be substituted or unsubstituted.

[0037] A “sulfinyl” group refers to an “-S(=O)-R” group in which R can be the same as defined with respect to sulfenyl. A sulfinyl may be substituted or unsubstituted.

[0038] A “sulfonyl” group refers to an “S(=O)2R” group in which R can be the same as defined with respect to sulfenyl. A sulfonyl may be substituted or unsubstituted.

[0039] An “O-carboxy” group refers to a “R.C(=O)O-” group in which R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl (alkyl) or heterocyclyl(alkyl), as defined herein. An O-carboxy may be substituted or unsubstituted.

[0040] The terms “ester” and “C-carboxy” refer to a “~C( :::: O)OR” group in which R can be the same as defined with respect to O-carboxy. An ester and C-carboxy may be substituted or unsubstituted.

[0041] A “thiocarbonyl” group refers to a “-C(= :: S)R” group in which R can be the same as defined with respect to O-carboxy. A thiocarbonyl may be substituted or unsubstituted.

[0042] A “tribal omethanesulfony I” group refers to an “XsCSfoO)?.---” group wherein each X is a halogen.

[0043] A “trihalomethanesulfonamido” group refers to an “X3CS(=O)2.N(RA)-” group wherein each X is a halogen, and RA is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl( alkyl) or heterocy cly I (alky I) .

[0044] The term “ammo” as used herein refers to a -NHz group.

[0045] .As used herein, the term “hydroxy” refers to a -OH group.

[0046] A “cyano” group refers to a “-CN” group.

[0047] The term “azido” as used herein refers to a -N? group.

[0048] An “isocyanate” group refers to a “-NCO” group.

[0049] A “thiocyanate” group refers to a “-SCN” group.

[0050] An “isothiocyanate” group refers to an “-NCS” group.

[0051] A “mercapto” group refers to an “-SH” group.

[0052] A “carbonyl” group refers to a -C(=O)- group.

[0053] An “S-suifonamido” group refers to a “-S(=O)2N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An S-sulfonamido may be substituted or unsubstituted.

[0054] An “N-sulfonamido” group refers to a “RS(=O)2N(RA)-” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl (alkyl) or heterocyclyl (alkyl). An N-sulfonamido may be substituted or unsubstituted,

[0055] An “O-carbamyl” group refers to a “~OC( :::: O)N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An O-carbamyl may be substituted or unsubstituted.

[0056] An “N-carbamyl” group refers to an “ROC( ::: O)N(RA)-” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-carbamyl may be substituted or unsubstituted.

[0057] An “O-thiocarbamyl” group refers to a “-OC(=S)-N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocy cly [(alky 1). An O-thiocarbamyl may be substituted or unsubstituted. [0058] An “N-thiocarbamyl” group refers to an “ROC( :=: S)N(RA)---” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-thiocarbamyl may be substituted or unsubstituted.

[0059] A “C-amido” group refers to a “---C( ; = : ())N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). A C-amido may be substituted or unsubstituted.

[0060] An “N-amido” group refers to a “RC(=O)N(RA)-” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-amido may be substituted or unsubstituted.

[0061] A “mono-substituted amine” refers to a “-NHRA” in which RA can be independently alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). A mono-substituted amine may be substituted or unsubstituted. In some instances, a mono-substituted amine can be -NHRA, wherein RA can be an unsubstituted Ci-6 alkyl or an unsubstituted or a substituted benzyl,

[0062] A “di-substituted amine” refers to a “-NRARB” in which RA and RB can be independently can be independently alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl (alkyl) or heterocyclyl(alkyl). A mono-substituted amine may be substituted or unsubstituted. In some instances, a mono-substituted amine can be -NRARB, wherein RA and RB can be independently an unsubstituted Ci-6 alkyl or an unsubstituted or a substituted benzyl.

[0063] The term “halogen atom” or “halogen” as used herein, means any one of the radio-stable atoms of column 7 of the Periodic Table of the Elements, such as, fluorine, chlorine, bromine and iodine.

[0064] Where the number of substituents is not specified (e.g., haloalkyl), there may be one or more substituents present. For example, “haloalkyl” may include one or more of the same or different halogens. As another example, “C1-C3 alkoxyphenyl” may include one or more of the same or different alkoxy groups containing one, two or three atoms. [0065] As used herein, the abbreviations for any protective groups, ammo acids and other compounds, are, unless indicated otherwise, in accord with their common usage, recognized abbreviations, or the IUPAC-IUB Commission on Biochemical Nomenclature (See, Biochem. 11 :942-944 (1972)).

[0066] The term “pharmaceutically acceptable salt” refers to a salt of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound. In some embodiments, the salt is an acid addition salt of the compound. Pharmaceutical salts can be obtained by reacting a compound with inorganic acids such as hydrohalic acid (e.g., hydrochloric acid or hydrobromic acid), sulfuric acid, nitric acid and phosphoric acid. Pharmaceutical salts can also be obtained by reacting a compound with an organic acid such as aliphatic or aromatic carboxylic or sulfonic acids, for example formic, acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic, methanesulfonic, ethanesulfonic, p-toluenesulfonic, salicylic or naphthalenesulfonic acid. Pharmaceutical salts can also be obtained by reacting a compound with a base to form a salt such as an ammonium salt (for example, ammonium or tri ethylammonium salt), an alkali metal salt, such as a lithium, a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, C1-C7 alkylamine, cyclohexylamine, triethanolamine, ethylenediamine, and salts with amino acids such as arginine and lysine.

[0067] Terms and phrases used in this application, and variations thereof, especially in the appended claims, unless otherwise expressly stated, should be construed as open ended as opposed to limiting. As examples of the foregoing, the term ‘including’ should be read to mean ‘including, without limitation,’ ‘including but not limited to,’ or the like; the term ‘comprising’ as used herein is synonymous with ‘including,’ ‘containing,’ or ‘characterized by,’ and is inclusive or open-ended and does not exclude additional, unrecited elements or method steps; the term ‘having’ should be interpreted as ‘having at least;’ the term ‘includes’ should be interpreted as ‘includes but is not limited to;’ the term ‘example’ is used to provide exemplary instances of the item in discussion, not an exhaustive or limiting list thereof. In addition, the term “comprising” is to be interpreted synonymously with the phrases "having at least" or "including at least". When used in the context of a compound or composition, the term "comprising" means that the compound or composition includes at least the recited features or components but may also include additional features or components.

[0068] With respect to the use of substantially any plural and/or singular terms herein, those having skill in the art can translate from the plural to the singular and/or from the singular to the plural as is appropriate to the context and/or application. The various singular/plural permutations may be expressly set forth herein for sake of clarity. The indefinite article “a” or “an” does not exclude a plurality.

[0069] It is understood that, in any compound described herein having one or more chiral centers, if an absolute stereochemistry’ is not expressly indicated, then each center may independently be of (Reconfiguration or (S)-configuration or a mixture thereof. Thus, the compounds provided herein may be enantiomerically pure, enantiomerically enriched, racemic mixture, diastereomerically pure, diastereomerically enriched, or a stereoisomeric mixture. In addition, it is understood that, in any’ compound described herein having one or more double bond(s) generating geometrical isomers that can be defined as E or Z, each double bond may’ independently be E or Z a mixture thereof. Likewise, it is understood that, in any compound described, all tautomeric forms are also intended to be included.

[0070] It is to be understood that where compounds disclosed herein have unfilled valencies, then the valencies are to be filled with hydrogens or isotopes thereof, e.g., hydrogen- 1 (protium) and hydrogen-2 (deuterium).

[0071 ] It is understood that the compounds described herein can be labeled isotopically. Substitution with isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements. Each chemical element as represented in a compound structure may include any isotope of said element. For example, in a compound structure a hydrogen atom may be explicitly disclosed or understood to be present in the compound. At any position of the compound that a hydrogen atom may be present, the hydrogen atom can be any isotope of hydrogen, including but not limited to hydrogen- 1 (protium) and hydrogen-2 (deuterium). Thus, reference herein to a compound encompasses all potential isotopic forms unless the context clearly dictates otherwise. [0072] Where a range of values is provided, it is understood that the upper and lower limit, and each intervening value between the upper and lower limit of the range is encompassed within the embodiments.

Compounds

[0073] Examples of embodiments of the present application include the following:

Embodiment 1

[0074] A compound of Formula (I), or a pharmaceutically acceptable salt thereof, having the structure: wherein: R la can be selected from -CM alkyl, -CM haloalkyl, -CH 2 (C3-6 monocyclic cycloalkyl), -CH 2 (4-6 membered monocyclic heterocyclyl) and -CH 2 (5-6 membered monocyclic heteroaryl); R lb can be selected from hydrogen, -CM alkyl and -CM haloalkyl; R !C can be selected from -N(R m1 )R nJ and -R xi ; R 2a , R 2b , R'\ R 2e , R 2g , R 2h can be independently selected from hydrogen and halogen; R 2d and R 2f can be independently selected from hydrogen, halogen, cyano, -CH 3 , -CH 2 CH 3 , -CH 2 OH, -OCH 3 and -SCH 3 ; R 3a can be selected from hydrogen, -CH 3 , -CF 3 and -CHF 2 ; R 3b can be selected from hydrogen, halogen, cyano, -CH 3 , -CF 3 , Cl IE'. -CH 2 CH 3 , -CH 2 OH, -OH, -OCH 3 and -SCHs; Y la can be selected from N (nitrogen) and -CH; Y ib can be selected from N (nitrogen) and C(R 4 ); Y 2a can be selected from N (nitrogen) and C(R 3 ); Y 2b can be selected from N (nitrogen) and C(R i d ), R f d can be selected from hydrogen and halogen; R 4 can be selected from hydrogen, halogen and -CH 3 ; R 5 can be selected from hydrogen, halogen and -CH 3 ; R ml can be selected from hydrogen, -CM alkyl, C 3-6 monocyclic cycloalkyl, C 5-12 bicyclic cycloalkyl, 5- or 6-membered monocyclic heteroaryl, 4-7 membered monocyclic heterocyclyl, 8-11 membered fused-bicyclic heteroaryl and 8-11 membered fused-bicyclic heterocyclyl; wherein the 5- or 6-membered monocyclic heteroaryl, the bicyclic heteroaryl, the 4-7 membered monocyclic heterocyclyl and the bicyclic heterocyclyl contain at least one atom or group of atoms independently selected from O (oxygen), S (sulfur), and N (nitrogen); wherein the can be optionally substituted with one or two or three substituents independently selected from monocyclic cycloalkyl, the C 5-12 bicyclic cycloalkyl, the 5- or 6-membered monocyclic heteroaryl, the 4-7 membered monocyclic heterocyclyl, the 8-1 1 membered fused-bicyclic heteroaryl and the 8-1 1 membered fused-bicyclic heterocyclyl can be optionally substituted with one or two or three substituents independently selected from halogen, cyano, -C 1-4 alkyl, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can have the structure:

(including pharmaceutically acceptable salts of any of the foregoing).

Embodiment 2

[0075] The compound of Embodiment 1, wherein Y 2a can be N (nitrogen).

Embodiment 3

[0076] The compound of Embodiment 1, wherein Y 2a can be C(R 5 ).

Embodiment 4

[0077] The compound of Embodiment 3, wherein R 5 can be hydrogen. Embodiment 5

[0078] The compound of Embodiment 3, wherein R 5 can be halogen, such as fluoro or chloro.

Embodiment 6

[0079] The compound of Embodiment 3, wherein R 5 can be ---CH 3 . Embodiment 7

[0080] The compound of any one of Embodiments 1-6, wherein R 3a can be hydrogen.

Embodiment 8

[0081] The compound of any one of Embodiments 1-6, wherein R 3a can be --CH 3 . Embodiment 9

[0082] The compound of any one of Embodiments 1-6, wherein R’ a can be -CF 3 . Embodiment 10

[0083] The compound of any one of Embodiments 1-6, wherein R' ;! can be -CHF 2 .

Embodiment 11

[0084] The compound of any one of Embodiments 1-10, wherein R 3b can be hydrogen.

Embodiment 12

[0085] The compound of any one of Embodiments 1-10, wherein R jb can be halogen, such as fluoro or chloro.

Embodiment 13

[0086] The compound of any one of Embodiments 1-10, wheremR 3b can be cyano.

Embodiment 14

[0087] The compound of any one of Embodiments 1-10, wherein R 3b can be -CH 3 or CH 2 CH 3 Embodiment 15

[0088] The compound of any one of Embodiments 1 -10, wherein R 5b can be --CF 3 or --CHF 2 .

Embodiment 16

[0089] The compound of any one of Embodiments 1-10, wherein R 3b can be -• CH 2 OH, OH, --OCH 3 or --SCH 3 .

Embodiment 17

[0090] The compound of any one of Embodiments 1-16, wherein Y la can be N (nitrogen).

Embodiment 18

[0091] The compound of any one of Embodiments 1 -16, wherein Y la can be ---CH. Embodiment 19

[0092] The compound of any one of Embodiments 1-18, wherein Y ib can be N (nitrogen).

Embodiment 20

[0093] The compound of any one of Embodiments 1-18, wherein Y ib can be C(R 4 ). Embodiment 21

[0094] The compound of Embodiment 20, wherein R 4 can be hydrogen.

Embodiment 22

[0095] The compound of Embodiment 20, wherein R 4 can be halogen. For example, R 4 can be fluoro or chloro.

Embodiment 23

[0096] The compound of Embodiment 20, wherein R 4 can be -CH 3 .

Embodiment 24

[0097] The compound of any one of Embodiments 1-23, wherein R La can be -C 1-4 alkyl. Examples of C 1-4 alkyls include methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl and tert-butyl.

Embodiment 25

[0098] The compound of any one of Embodiments 1 -23, wherein R la can be -Ci-4 haloalkyl, such as -CE 3 , -CHF 2 , -CH 2 F, -CCh, -CHCh, -CH 2 CI, -CH 2 CF 3 and -CH 2 CHF 2 . Embodiment 26

[0099] The compound of any one of Embodiments 1-23, wherein R La can be selected from -CH 2 (C 3-6 monocyclic cycloalkyl), -CH 2 ( 4-6 membered monocyclic heterocyclyl) and -CH 2 ( 5-6 membered monocyclic heteroaryl). Exemplary monocyclic cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. In some embodiments, the 4-6 membered monocyclic heterocyclyl of the -CH 2 ( 4-6 membered monocyclic heterocyclyl) and/or the 5-6 membered monocyclic heteroaryl of the -CH 2 ( 5-6 membered monocyclic heteroaryl) can include one or more ring atoms (such as 1, 2 or 3) selected from N (nitrogen), O (oxygen) and S (sulfur).

Embodiment 27

[0100] The compound of any one of Embodiments 1-26, wherein R ld can be hydrogen. Embodiment 28

[0101] The compound of any one of Embodiments 1-26, wherein R id can be halogen, such as fluoro or chloro.

Embodiment 29

[0102] The compound of any one of Embodiments 1-28, wherein R lc can be --- N(R ml )R n! .

Embodiment 30

[0103] The compound of Embodiment 29, wherein R nl can be hydrogen, such that R !C can be -NH(R ffi{ ).

Embodiment 31

[0104] The compound of Embodiment 2.9, wherein R r *’ ! can be -C 1-4 alkyl. For example, R nj can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl or tertbutyl.

Embodiment 32

[0105] The compound of Embodiment 29, wherein R n1 can be -C(=O)OR Z4 . In some embodiments, R Z4 can be a C 1-4 alkyl. In other embodiments, R 64 can be 5- to 6- membered monocyclic heterocyclyl(CH 2 )- optionally substituted with -CM alkyl. In other embodiments, the compound of Embodiment 29, wherein R nl can be C 1-4 haloalkyl In still other embodiments, the compound of Embodiment 29, wherein R nl can be C 3 -6 monocyclic cycloalkyl(CH 2 )-, such as cyclopropyl-(CH 2 )- cyclobutyl -(CTb)-, cyclopentyl-(CH 2 )- and cyclohexyl-(CIl2)-.

Embodiment 33

[0106] The compound of any one of Embodiments 29-32, wherein R ml : can be tetrahydrofuran or tetrahydro-2H-pyran, each optionally substituted with hydroxy. Embodiment 34

[0107] The compound of Embodiment 33, wherein R ml can be Embodiment 35

[0108] The compound of any one of Embodiments 29-32, wherein R ml can be The compound of Embodiment 37, wherein ~R xl can be selected from: Embodiment 41

[0114] The compound of any one of Embodiments 1-40, wherein R 2a , R 2b , R 2c , R 2 ®, R 2g and R 2h can be each hydrogen.

Embodiment 42

[0115] The compound of any one of Embodiments I -40, wherein R 2a , R 2c , R 2 ®, R 2g and R 2h can be each hydrogen: and R 2b can be halogen.

Embodiment 43

[0116] The compound of any one of Embodiments 1-40, wherein R 2b , R 2c , R 2 ®, R' e and R 2h can be each hydrogen; and R 2a can be halogen.

Embodiment 44

[0117] The compound of any one of Embodiments 1-43, wherein R 2d and R 2f can be each halogen. In some embodiments, R 2d and R 2f can be each fluoro. In other embodiments, R 2d and R 2f can be each chloro.

Embodiment 45

[0118] The compound of any one of Embodiments 1-43, wherein R 2d and R 2f can be each -CHs.

Embodiment 46

[0119] The compound of any one of Embodiments 1-43, wherein R 2d can be halogen; and R 21 can be -CH 3 .

Embodiment 47

[0120] The compound of any one of Embodiments 1 -43, wherein R 2d can be Ci b; and R 21 can be halogen.

Embodiment 48

[0121 ] The compound of any one of Embodiments 1 -43, wherein R 2Q can be cyano; and R 2f can be halogen.

Embodiment 49

[0122] The compound of any one of Embodiments 1-43, wherein R 2d can be - OCH?; and R 21 can be halogen.

Embodiment 50

[0123] The compound of any one of Embodiments 42-44 or 46-49, wherein the halogen can be chloro or fluoro.  

or a pharmaceutically acceptable salt of any of the foregoing.

Embodiment 54

[0127] In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, cannot be a compound or a pharmaceutically acceptable salt provided in WO 2021/076691,

Embodiment 55

[0128] A pharmaceutical composition that can include an effective amount of a compound of any one of Embodiments 1-54, or a pharmaceutically acceptable salt thereof, and excipient.

Embodiment 56

[0129] A method for treating hepatitis B in a subject that can include administering to the subject in need thereof an effective amount of a compound of any one of Embodiments 1-54, or a pharmaceutically acceptable salt thereof.

Embodiment 57

[0130] A method for treating hepatocellular carcinoma (HCC) in a subject that can include administering to the subject in need thereof an effective amount of a compound of any one of Embodiments 1-54, or a pharmaceutically acceptable salt thereof.

Embodiment 58

[0131] The method of any one of Embodiments 56-57, that can further include administering surgery, radiation therapy, chemotherapy, targeted therapy, immunotherapy, hormonal therapy, or antiviral therapy.

Embodiment 59

[0132] A compound of any one of Embodiments 1-54, or a pharmaceutically acceptable salt thereof, for use in treating hepatitis B. Embodiment 60

[0133] A compound of any one of Embodiments 1-54, or a pharmaceutically acceptable salt thereof, for use in treating hepatocellular carcinoma (HCC).

Embodiment 61

[0134] The compound of any one of Embodiments 59-60, that can further include administering surgery, radiation therapy, chemotherapy, targeted therapy, immunotherapy, hormonal therapy, or antiviral therapy.

Embodiment 62

[0135] Use of a compound of any one of Embodiments 1-54, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for use in treating hepatitis B. Embodiment 63

[0136] Use of a compound of any one of Embodiments 1-54, or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for use in treating hepatocellular carcinoma (HCC).

Embodiment 64

[0137] The use of any one of Embodiments 62-63, that can further include administering surgery, radiation therapy, chemotherapy, targeted therapy, immunotherapy, hormonal therapy, or antiviral therapy.

Methods for the Preparation

[0138] In this section, as in all other sections unless the context indicates otherwise, references to Formula (I), along with pharmaceutical acceptable salts thereof, include all other sub-groups and examples thereof as provided herein. The general preparations of some representative examples of compounds of Formula (I) are described herein, and are generally- prepared from starting materials which are either commercially available or prepared by standard synthetic processes used by those skilled in the art.

[0139] All variables shown in the schemes are defined as mentioned herein, unless otherwise is indicated or is clear from the context.

[0140] In general, compounds of Formula (I) (including pharmaceutically acceptable salts thereof) can be prepared according to Scheme 1. In Scheme 1 , LG can be Br or Cl. All other variables in Scheme 1 are according to Formula (I) provided herein. In Scheme 1, the following reaction conditions apply: (1) In the presence of suitable catalyst (for example, bis(triphenylphosphine)palladium(II) dichloride) in a suitable solvent, such as the mixture of 1,4-dioxane and water, with a suitable base (for example, K 2 CO 3 ) at a suitable temperature (for example, 100 °C); 2) In the presence of suitable base (for example, DIPEA), in a suitable solvent (for example, DCM), at a suitable temperature (for example, 100 °C); 3) In the presence of suitable catalyst (for example, bis(triphenylphosphine)palladium(II) dichloride) in a suitable solvent, such as the mixture of 1,4-dioxane and water, with a suitable base (for example, KO Ac) at a suitable temperature (for example, 90 °C); 4) In the presence of suitable catalyst (for example, bis(triphenylphosphine)palladium(Il) dichloride) in a suitable solvent, such as the mixture of 1,4-dioxane and water, with a suitable base (for example, K 2 CO 3 ) at a suitable temperature (for example, 100 °C) and 5) In the presence of suitable coupling reagents (for example, N,N,N',N'-tetramethylchloroformamidinium hexafluorophosphate and N- methylimidazole) in a suitable solvent (for example, acetonitrile) at a suitable temperature (for example, 50 °C).

[0141] In general, compounds of Formula (I) can be prepared according to Scheme 2. In Scheme 2, LG can be Br or Cl. All other variables in Scheme 2 are according to Formula (I) provided herein. In Scheme 2, the following reaction conditions apply: 1) In the presence of suitable catalyst (for example, bis(tnphenylphosphine)palladium(II) dichloride) in a suitable solvent, such as the mixture of 1,4-dioxane and water, with a suitable base (for example, K2CO3) at a suitable temperature (for example, 100 °C); 2 ) In the presence of suitable coupling reagents (for example, N,N,N',N' -tetramethylchloroformamidinium hexafluorophosphate and N-methylimidazole); 3) In the presence of suitable catalyst (for example, bis(triphenylphosphine)palladium(II) dichloride) in a suitable solvent, such as the mixture of 1,4-dioxane and water, with a suitable base (for example, KO Ac) at a suitable temperature (for example, 90 °C); 3) In a suitable solvent (for example, acetonitrile) at a suitable temperature (for example, 50 °C); (4) In the presence of suitable catalyst (for example, bis(triphenylphosphine)palladium(II) dichloride) in a suitable solvent, such as the mixture of 1,4-dioxane and water, with a suitable base (for example, K 2 CO 3 ) at a suitable temperature (for example, 100 °C); and 5) In the presence of an appropriate reductive reagent, such as sodium cyanoborohydride or sodium tnacetoxyborohydride, in a suitable solvent (for example, DCM or MeOH) at a suitable temperature, such as approximately 20 °C.

Pharmaceutical Compositions

[0142] Some embodiments described herein relate to pharmaceutical compositions that comprise, consist essentially of, or consist of an effective amount of a compound described herein (such as a compound of Formula (I), or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable carrier, excipient, or combination thereof. A pharmaceutical composition described herein is suitable for human and/or veterinary applications.

[0143] The terms “function’ 1 and “functional” as used herein refer to a biological, enzymatic, or therapeutic function.

[0144] The terms “effective amount” or “effective dose” is used to indicate an amount of an active compound, or pharmaceutical agent, which elicits the biological or medicinal response indicated. For example, an effective amount of compound can be the amount needed to alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated This response may occur in a tissue, system, animal or human and includes alleviation of the signs or symptoms of the disease being treated. Determination of an effective amount is well within the capability of those skilled in the art, in view' of the disclosure provided herein. The effective amount of the compounds disclosed herein required as a dose wall depend on the route of administration, the type of animal, including human, being treated, and the physical characteristics of the specific animal under consideration. The dose can be tailored to achieve a desired effect, but will depend on such factors as weight, diet, concurrent medication and other factors which those skilled in the medical arts wall recognize.

[0145] The term “pharmaceutically acceptable salts” includes relatively non-toxic, inorganic and organic acid, or base addition salts of compositions, including without limitation, analgesic agents, therapeutic agents, other materials, and the like. Examples of pharmaceutically acceptable salts include those derived from mineral acids, such as hydrochloric acid and sulfuric acid, and those derived from organic acids, such as ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, and the like. Examples of suitable inorganic bases for the formation of salts include the hydroxides, carbonates, and bicarbonates of ammonia, sodium, lithium, potassium, calcium, magnesium, aluminum, zinc, and the like. Salts may also be formed with suitable organic bases, including those that are non-toxic and strong enough to form such salts. For example, the class of such organic bases may include but are not limited to mono-, di-, and trialkylamines, including methylamine, dimethylamine, and triethylamine; mono-, di-, or trihydroxyalkylamines including mono-, di- , and triethanolamine; amino acids, including glycine, arginine and lysine; guanidine; N- methylglucosamine; N-methylglucamine; L-glutamine; N-methylpiperazme; morpholine; ethylenediamine; N-benzylphenethylamine; trihydroxymethyl aminoethane.

[0146] “Formulation”, “pharmaceutical composition”, and “composition” as used interchangeably herein are equivalent terms referring to a composition of matter for administration to a subject.

[0147] The term “pharmaceutically acceptable” means compatible with the treatment of a subject, and in particular, a human.

[0148] The terms “agent” refers to an active agent that has biological activity and may be used in a therapy. Also, an “agent” can be synonymous with “at least one agent,” “compound,” or “at least one compound,” and can refer to any form of the agent, such as a derivative, analog, salt or a prodrug thereof. The agent can be present in various forms, components of molecular complexes, and pharmaceutically acceptable salts (e.g., hydrochlorides, hydrobromides, sulfates, phosphates, nitrates, borates, acetates, maleates, tartrates, and salicy lates). The term “agent” can also refer to any pharmaceutical molecules or compounds, therapeutic molecules or compounds, matrix forming molecules or compounds, polymers, synthetic molecules and compounds, natural molecules and compounds, and any combination thereof.

[0149] The term “subject” as used herein has its ordinary meaning as understood m light of the specification and refers to an animal that is the object of treatment, inhibition, or amelioration, observation or experiment. “Animal” has its ordinary meaning as understood in light of the specification and includes cold- and warm-blooded vertebrates and/or invertebrates such as fish, shellfish, or reptiles and, in particular, mammals. “Mammal” has its ordinary meaning as understood in light of the specification, and includes but is not limited to mice, rats, rabbits, guinea pigs, dogs, cats, sheep, goats, cows, horses, primates, such as humans, monkeys, chimpanzees, or apes. In some embodiments, the subject is human.

[0150] Proper formulation is dependent upon the route of administration chosen. Techniques for formulation and administration of the compounds described herein are known to those skilled in the art. Multiple techniques of administering a compound exist in the art including, but not limited to, enteral, oral, rectal, topical, sublingual, buccal, intraaural, epidural, epi cutaneous, aerosol, parenteral delivery, including intramuscular, subcutaneous, intra-arterial, intravenous, intraportal, intra-articular, intradermal, peritoneal, intramedullary injections, intrathecal, direct intraventricular, intraperitoneal, intranasal or intraocular injections. Pharmaceutical compositions will generally be tailored to the specific intended route of administration. Pharmaceutical compositions can also be administered to isolated cells from a patient or individual, such as T cells, Natural Killer cells, B cells, macrophages, lymphocytes, stem cells, bone marrow cells, or hematopoietic stem cells.

[0151] The pharmaceutical compound can also be administered in a local rather than systemic manner, for example, via injection of the compound directly into an organ, tissue, cancer, tumor or infected area, often in a depot or sustained release formulation. Furthermore, one may administer the compound in a targeted drug delivery/ system, for example, in a liposome coated with a tissue specific antibody. The liposomes may be targeted to and taken up selectively by the organ, tissue, cancer, tumor, or infected area.

[0152] The pharmaceutical compositions disclosed herein may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes. As described herein, compounds used in a pharmaceutical composition may be provided as salts with pharmaceutically compatible counterions.

[0153] As used herein, a “carrier” refers to a compound, particle, solid, semi-solid, liquid, or diluent that facilitates the passage, delivery and/or incorporation of a compound to cells, tissues and/or bodily organs. For example, without limitation, a lipid nanoparticle (LNP) is a type of carrier that can encapsulate a compound, or a pharmaceutically acceptable salt thereof, as described herein to thereby protect the compound, or a pharmaceutically acceptable salt thereof, as described herein from degradation during passage through the bloodstream and/or to facilitate delivery’ to a desired organ, such as to the liver.

[0154] As used herein, a “diluent” refers to an ingredient in a pharmaceutical composition that lacks pharmacological activity but may be pharmaceutically necessary or desirable. For example, a diluent may be used to increase the bulk of a potent drug whose mass is too small for manufacture and/or administration. It may also be a liquid for the dissolution of a drug to be administered by injection, ingestion or inhalation. A common form of diluent in the art is a buffered aqueous solution such as, without limitation, phosphate buffered saline that mimics the composition of human blood,

[0155] The term “excipient” has its ordinary meaning as understood in light of the specification, and refers to inert substances, compounds, or materials added to a pharmaceutical composition to provide, without limitation, bulk, consistency, stability, binding ability, lubrication, disintegrating ability etc., to the composition. Excipients with desirable properties include but are not limited to preservatives, adjuvants, stabilizers, solvents, buffers, diluents, solubilizing agents, detergents, surfactants, chelating agents, antioxidants, alcohols, ketones, aldehydes, ethylenediaminetetraacetic acid (EDTA), citric acid, salts, sodium chloride, sodium bicarbonate, sodium phosphate, sodium borate, sodium citrate, potassium chloride, potassium phosphate, magnesium sulfate sugars, dextrose, fructose, mannose, lactose, galactose, sucrose, sorbitol, cellulose, serum, amino acids, polysorbate 20, polysorbate 80, sodium deoxycholate, sodium taurodeoxycholate, magnesium stearate, octylphenol ethoxylate, benzethonium chloride, thimerosal, gelatin, esters, ethers, 2-phenoxyethanol, urea, or vitamins, or any combination thereof. The amount of the excipient may be found in a pharmaceutical composition at a percentage of 0%, 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 100% w/w or any percentage by weight in a range defined by any two of the aforementioned numbers.

[0156] The term “adjuvant” as used herein refers to a substance, compound, or material that stimulates the immune response and increase the efficacy of protective immunity and is administered in conjunction with an immunogenic antigen, epitope, or composition. Adjuvants serve to improve immune responses by enabling a continual release of antigen, upregulation of cytokines and chemokines, cellular recruitment at the site of administration, increased antigen uptake and presentation in antigen presenting cells, or activation of antigen presenting cells and inflammasomes. Commonly used adjuvants include but are not limited to alum, aluminum salts, aluminum sulfate, aluminum hydroxide, aluminum phosphate, calcium phosphate hydroxide, potassium aluminum sulfate, oils, mineral oil, paraffin oil, oil-in-water emulsions, detergents, MF59®, squalene, AS03, a-tocopherol, polysorbate 80, AS04, monophosphoryl lipid A, virosomes, nucleic acids, polyinosinic: poly cytidylic acid, saponins, QS-21, proteins, flagellin, cytokines, chemokines, IL-1, IL-2, IL-12, IL-15, IL-21, imidazoquinolines, CpG oligonucleotides, lipids, phospholipids, dioleoyl phosphatidylcholine (DOPC), trehalose dimycolate, peptidoglycans, bacterial extracts, lipopolysaccharides, or Freund’s Adjuvant, or any combination thereof.

[0157] The term “purity” of any given substance, compound, or material as used herein refers to the actual abundance of the substance, compound, or material relative to the expected abundance. For example, the substance, compound, or material may be at least 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100% pure, including all decimals in between. Purity may be affected by unwanted impurities, including but not limited to side products, isomers, enantiomers, degradation products, solvent, carrier, vehicle, or contaminants, or any combination thereof. Purity can be measured technologies including but not limited to chromatography, liquid chromatography, gas chromatography, spectroscopy, UV-visible spectrometry, infrared spectrometry, mass spectrometry, nuclear magnetic resonance, gravimetry , or titration, or any combination thereof.

Methods of Use

[0158] Some embodiments disclosed herein related to selecting a subject or patient in need. In some embodiments, a patient is selected who is in need of treatment, inhibition, amelioration, prevention or slowing of diseases or conditions associated with PD-L1 dysregulation. In some embodiments, such diseases or conditions associated with PD-L1 dysregulation may include, for example, cancer, HCC, viral infections, or HBV. In some embodiments, a subject can be selected who has previously been treated for the disease or disorder described herein. In some embodiments, a subject can be selected who has previously been treated for being at risk for the disease or disorder described herein. In some embodiments, a subject can be selected who has developed a recurrence of the disease or disorder described herein. In some embodiments, a subject can be selected who has developed resistance to therapies for the disease or disorder described herein. In some embodiments, a subject can be selected who may have any combination of the aforementioned selection criteria.

[0159] Compounds, and pharmaceutically acceptable salts thereof, disclosed herein can be evaluated for efficacy and toxicity using known methods. A non-limiting list of potential advantages of a compound, or a pharmaceutically acceptable salt thereof, described herein include improved stability, increased safety profile, increased efficacy, increased binding to the target, increased specificity for the target (for example, a cancer cell or virally infected cell).

[0160] The terms “treating,” “treatment,” “therapeutic,” or “therapy” as used herein has its ordinary? meaning as understood in light of the specification, and do not necessarily mean total cure or abolition of the disease or condition. The term “treating” or “treatment” as used herein (and as well understood in the art) also means an approach for obtaining beneficial or desired results in a subject's condition, including clinical results. Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of the extent of a disease, stabilizing (i.e., not worsening) the state of disease, prevention of a disease's transmission or spread, delaying or slowing of disease progression, amelioration or palliation of the disease state, diminishment of the reoccurrence of disease, and remission, whether partial or total and whether detectable or undetectable. “Treating” and “treatment” as used herein also include prophylactic treatment. Treatment methods comprise administering to a subject a therapeutically effective amount of an active agent. The administering step may consist of a single administration or may comprise a series of administrations. The compositions are administered to the subject in an amount and for a duration sufficient to treat the subject. The length of the treatment period depends on a variety of factors, such as the severity of the condition, the age and genetic profile of the subject, the concentration of active agent, the activity of the compositions used in the treatment, or a combination thereof. It will also be appreciated that the effective dosage of an agent used for the treatment or prophylaxis may increase or decrease over the course of a particular treatment or prophylaxis regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art. In some instances, chronic administration may be required.

[0161] Some embodiments described herein relate to a method of treating, inhibiting, ameliorating, preventing, or slowing the disease or disorder described herein. In some embodiments, the methods include administering to a subject identified as suffering from the disease or disorder described herein an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein, or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein. Other embodiments described herein relate to using a compound, or a pharmaceutically acceptable salt thereof, as described herein in the manufacture of a medicament for treating, inhibiting ameliorating, preventing, or slowing the disease or disorder described herein. Still other embodiments described herein relate to the use of a compound, or a pharmaceutically acceptable salt thereof, as described herein or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein for treating, inhibiting ameliorating, preventing, or slowing the disease or disorder described herein.

[0162] Some embodiments described herein relate to a method for inhibiting replication of a cancer cell or a virus that can include contacting the cell or virus or administering to a subject identified as suffering from a cancer or a viral infection with an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein, or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein. Other embodiments described herein relate to the use of an effecti ve amount of a compound, or a pharmaceutically acceptable salt thereof, described herein, or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein in the manufacture of a medicament for inhibiting replication of a cancer cell or virus. Still other embodiments described herein relate to an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein, or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein for inhibiting replication of a cancer cell or virus. In some embodiments, the cancer cell is an HCC cell. In some embodiments, the virus is hepatitis B.

[0163] Some embodiments described herein relate to a method for inhibiting cell proliferation, such as inhibiting cell proliferation of a cancer cell or cell infected with a virus, that can include administering to a subject identified as suffering from a disease wherein inhibiting cell proliferation is desirable with an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein, or a pharmaceutical composition that includes effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein. Other embodiments described herein relate to the use of an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein, or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein in the manufacture of a medicament for inhibiting cell proliferation, such as inhibiting cell proliferation of a cancer cell or cell infected with a virus. Still other embodiments described herein relate to an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein, or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein for inhibiting cell proliferation, such as inhibiting cell proliferation of a cancer cell or cell infected with a virus. In some embodiments, the cancer cell is an HCC cell. In some embodiments, the cell infected with a virus is infected with hepatitis B virus.

[0164] Some embodiments described herein relate to a method of inducing apoptosis of a cell (for example, a cancer cell or cell infected with a virus) that can include contacting the cell with an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein, or a pharmaceutical composition that includes an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as described herein. Other embodiments described herein relate to using an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein in the manufacture of a medicament for inducing apoptosis of a cell, such as a cancer cell or cell infected with a virus. Still other embodiments described herein relate to the use of an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein for inducing apoptosis of a cell, such as a cancer cell or cell infected with a virus. In some embodiments, the cancer cell is an HCC cell. In some embodiments, the cell infected with a virus is infected with hepatitis B virus.

[0165] Some embodiments described herein relate to a method of decreasing the viability of a cell (for example, a cancer cell or cell infected with a virus) that can include contacting the cell with an effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein, or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein. Other embodiments described herein relate to using a compound, or a pharmaceutically acceptable salt thereof, as described herein in the manufacture of a medicament for decreasing the viability of a cell, such as a cancer cell or cell infected with a virus. Still other embodiments described herein relate to the use of an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein or a pharmaceutical composition that includes an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as described herein for decreasing the viability of a cell, such as a cancer cell or cell infected with a virus. In some embodiments, the cancer cell is an HCC cell In some embodiments, the cell infected with a virus is infected with hepatitis B virus.

[0166] Those of skill in the treatment of such diseases could determine the effective therapeutic daily amount from test results. An effective therapeutic daily amount would be from about 0.005 mg /kg to 50 mg/kg. in particular 0.01 mg/kg to 50 mg/kg body weight, more in particular from 0.01 mg/kg to 25 mg/kg body weight, preferably from about 0.01 mg/kg to about 15 mg/kg, more preferably from about 0.01 mg/kg to about 10 mg/kg, even more preferably from about 0.01 mg/kg to about 1 mg/kg, most preferably from about 0.05 mg/kg to about 1 mg/kg body weight. [0167] In some embodiments, the effective amount of a compound, or a pharmaceutically acceptable salt thereof, described herein is dosed more than one time. In some embodiments, the compound, or a pharmaceutically acceptable salt thereof, described herein can be administered every 1, 2, 3, 4, 5, 6, 7 days, or 1, 2, 3, 4 weeks, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 months, or 1, 2, 3, 4, 5 years, or any period or combination thereof within the range defined by any two aforementioned times. In some embodiments, at least one loading dose and at least one maintenance dose is administered to the subject, where the at least one loading dose is a higher dose of a compound, or a pharmaceutically acceptable salt thereof, described herein than the at least one maintenance dose.

[0168] As used herein, the term “combination therapy” is intended to define therapies which comprise the use of a combination of two or more pharmaceutical compounds/agents or therapies. Thus, references to “combination therapy”, “combinations” and the use of compounds/agents “in combination” in this application may refer to compounds/agents that are administered as part of the same overall treatment regimen. As such, the dosage or timing of each of the two or more compounds/agents may differ: each may be administered at the same time or at different times. Accordingly, the compounds/agents of the combination may be administered sequentially (e.g., before or after) or simultaneously, either in the same pharmaceutical formulation (i.e., together), or in different pharmaceutical formulations (i.e., separately). Each of the two or more compounds/agents in a combination therapy may also differ with respect to the route of administration.

[0169] The term “inhibitor”, as used herein, refers to an enzyme inhibitor or receptor inhibitor which is a molecule that binds to an enzyme or receptor, and decreases and/or blocks its activity. The term may relate to a reversible or an irreversible inhibitor.

[0170] Cancer may be treated with surgery, radiation therapy, chemotherapy, targeted therapies, immunotherapy or hormonal therapies. Any of these mentioned therapies may be used in conjunction with another therapy as a combination therapy. Chemotherapeutic compounds include but are not limited to alemtuzumab, altretamine, azacitidine, bendamustine, bleomycin, bortezomib, busulfan, cabazitaxel, capecitabine, carboplatin, carniofur, carmustine, chlorambucil, chlormethine, cisplatin, cladribine, clofarabme, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicm, decitabine, denosumab, docetaxel, doxorubicin, epirubicin, estraniustine, etoposide, everolimus, floxuridine, fludarabine, fluorouracil, fotemustine, gemcitabine, gemtuzumab, hydroxycarbamide, ibritumomab, idarubicin, ifosfamide, irinotecan, ixabepilone, lomustine, melphalan, mercaptopurine, methotrexate, mitomycin, mitoxantrone, nedaplatin, neiarabine, ofatumumab, oxaliplatin, paclitaxel, pemetrexed, pentostatin, pertuzumab, procarbazine, raltitrexed, streptozotocin, tegafur, temozolomide, temsirolimus, teniposide, tioguanine, topotecan, tositumomab, valrubicin, vinblastine, vincristine, vindesine, vmflunine, or vinorelbine, or any combination thereof.

[0171] As used herein, the term “protein kinase inhibitor” refers to inhibitors of protein kinases, serine/threonine kinases, tyrosine kinases, or dual-specificity’ kinases for the treatment of cancer or other illness. In some embodiments, the protein kinase inhibitor is a small molecule, compound, polysaccharide, lipid, peptide, polypeptide, protein, antibody, nucleoside, nucleoside analog, nucleotide, nucleotide analog, nucleic acid, or oligonucleotide. In some embodiments, the protein kinase inhibitor includes but is not limited to acalabrutinib, adavosertib, afatimb, alectinib, axitinib, binimetinib, bosutinib, brigatinib, cediranib, ceritinib, cetuximab, cobimetinib, crizotinib, cabozantinib, dacomitinib, dasatinib, entrectimb, erdafitinib, erlotinib, fostamatinib, gefitinib, ibrutinib, imatinib, lapatimb, lenvatinib, lestaurtinib, lortatinib, masitinib, momelotinib, mubritinib, neratinib, nilotinib, nintedamb, olmutinib, osimertinib, pacritinib, panitumumab, pazopanib, pegaptamb, ponatinib, radotinib, regorafenib, rociletmib, ruxolitinib, selumetinib, semaxanib, sorafenib, sunitmib, SU6656, tivozanib, toceranib, trametmib, trastuzumab, vandetanib, or vemurafenib, or any combination thereof.

[0172] As used herein, the term “checkpoint inhibitor” refers to an immunotherapy that targets immune checkpoints to stimulate immune function. In some embodiments, the checkpoint inhibitor is a small molecule, compound, polysaccharide, lipid, peptide, polypeptide, protein, antibody, nucleoside, nucleoside analog, nucleotide, nucleotide analog, nucleic acid, or oligonucleotide. In some embodiments, the immune checkpoint is the PD- 1/PD-L1 checkpoint. In some embodiments, the PD-1 checkpoint includes but is not limited to nivolumab, pembrolizumab, spartalizumab, cemiplimab, camrelizumab, sintilimab, tislelizumab, toripalimab, AMP-224 or AMP-514, or any combination thereof. In some embodiments, the PD-L1 checkpoint inhibitor includes but is not limited to atezolizumab, avelumab, durvalumab, KN035, AUNP12, CA-170, or BMS-986189, or any combination thereof. In some embodiments, the immune checkpoint is the CTLA-4 checkpoint. In some embodiments, the CTLA-4 checkpoint inhibitor includes but is not limited to ipilimumab or tremihmumab, or any combination thereof.

[0173] As used herein, the term “VEGF inhibitor” refers to inhibitors of vascular endothelial growth factor (VEGF) or a VEGF receptor (VEGFR). In some embodiments, the VEGF inhibitor is a small molecule, compound, polysaccharide, lipid, peptide, polypeptide, protein, antibody, nucleoside, nucleoside analog, nucleotide, nucleotide analog, nucleic acid, or oligonucleotide. In some embodiments, the VEGF inhibitor includes but is not limited to aflibercept, axitinib, bevacizumab, brivanib, cabozantinib, cediranib, lenvatinib, Imifinib, nintedanib, pazopanib, ponatinib, ramucirumab, regorafenib, semaxanib, sorafenib, sunitmib, tivozanib, toceranib, or vandetanib, or any combination thereof.

[0174] As used herein, the term “antiviral medication” refers to a pharmaceutical composition administered to treat a viral infection. In some embodiments, the viral infection is caused by adenovirus, Ebola virus, coronavirus, Epstein-Barr virus (EBV), Friend virus, hantavirus, hepatitis B virus (HBV), hepatitis C virus (HCV), herpes simplex virus, human immunodeficiency virus (HIV), human metapneumovirus, human papillomavirus (HPV), influenza virus, Japanese encephalitis virus, Kaposi’s sarcoma-associated herpesvirus, lymphocytic choriomeningitis virus, parainfluenza virus, rabies virus, respiratory' syncytial virus, rhinovirus, varicella zoster virus.

[0175] In some embodiments, the antiviral medication is a small molecule, compound, polysaccharide, lipid, peptide, polypeptide, protein, antibody, nucleoside, nucleoside analog, nucleotide, nucleotide analog, nucleic acid, or oligonucleotide. In some embodiments, the antiviral medication is an interferon, a capsid assembly modulator, a sequence specific oligonucleotide, an entry' inhibitor, or a small molecule immunomodulatory. In some embodiments, the antiviral medication includes but is not limited to AB-423, AB-506, ABI-H2158, vebicorvir (ABI-HO731), acyclovir, adapromme, adefovir, adefovir dipivoxil, alafenamide, amantadine, asunaprevir, baloxavir marboxil, beclabuvir, boceprevir, brivudine, cidofovir, ciluprevir, clevudine, cytarabine, daclatasvir, danoprevir, dasabuvir, deleobuvir, dipivoxil, edoxudine, elbasvir, entecavir, faldaprevir, famciclovir, favipiravir, filibuvir, fomivirsen, foscarnet, galidesivir, ganciclovir, glecaprevir, GLS4, grazoprevir, idoxuridine, imiquimod, IFN-a, interferon alfa 2b, JNJ-440, JNJ-6379 (JNJ-56136379), lamivudine, lanmamivir, ledipasvir, mericitabine, methisazone, MK-608, moroxydine, narlaprevir, NITD008, NZ-4, odalasvir, ombitasvir, oseltamivir, paritaprevir, peginterferon alfa-2a, penciclovir, peramivir, pibrentasvir, pimodivir, pleconaril, podophyllotoxin, presatovir, radalbuvir, ravidasvir, remdesivir, REP 2139, REP 2165, resiquimod, R07049389 (RG7907), ribavirin, rifampicin, rimantadine, ruzasvir, samatasvir, setrobuvir, simeprevir, sofosbuvir, sorivudine, sovaprevir, taribavirin, telaprevir, telbivudme, tenofovir, tenofovir disoproxil, tenofovir alfenamide, triazavirin, trifluridine, tromantadine, umifenovir, uprifosbuvir, valaciclovir, valgancicovir, vaniprevir, vedroprevir, velpatasvir, vidarabine, voxilaprevir, zanamivir, cledvudine, ANA-380/LB80380, thymalfasin (Zadaxin), AU-2173, VIR-2218, RG6346, JNJ-73763989 (JNJ-3989), .AB-729, BB-103, Hepcludex (Bulevirtide formerly Myrcludex B), hzVSF, morphothiadin, JNJ-56136379, EDP-514, QL- 007, ABI-H3733, ZM- H1505R, B-836, VNRX-9945, GLP-26, ABI-4334, IONIS-HBVRX (GSK 32.2.8836), EBT107, NASVAC, GS-4774, HepTcell, VBI-2601 (BRII-179), VVX001, VTP-300, CVLHBV-002, AIC-649, HB-110, JNJ-64300535, CARG-201, PRGN-2013, SA104, VRGN-0200, selgantolimod, RG7854, SBT-8230, YS-HBV-002, lenvervimab, Vir-3434, IMC-I109V, LTCR-H2-1, APG-1387, ASC42, EYP001, EDP-721, ENOB-HB-01, GV1001, CP101, DF- 006, ALG-000184, ALG-010133, ALG- 125097, ALG-020572, AEG- 125755, zanamivir or any combination thereof.

[0176] The term “% w/w” or “% wt/wf ’ as used herein has its ordinary meaning as understood in light of the specification and refers to a percentage expressed in terms of the weight of the ingredient or agent over the total weight of the composition multiplied by 100. The term “% v/v” or “% vol/vol” as used herein has its ordinary' meaning as understood in the light of the specification and refers to a percentage expressed in terms of the liquid volume of the compound, substance, ingredient, or agent over the total liquid volume of the composition multiplied by 100.

EXAMPLES

[0177] Some aspects of the embodiments discussed above are disciosed in further detail in the following examples, which are not in any way intended to limit the scope of the present disclosure. Those in the art will appreciate that many other embodiments also fall within the scope of the present disclosure, as it is described herein above and in the claims. [0178] Hereinafter, the term “rt”, “r.t.” or “RT” means room temperature: “h” means hours; “Me” means methyl; “MeOH” means methanol; “Et” means ethyl; “EtOH” means ethanol; “NaH” means sodium hydride; “NaBH(AcO)3” or “NaBH(OAc)3” means sodium triacetoxyborohydride; “EtOAc” means ethyl acetate; "TEA” or "EtsN” means triethylamine; “DCM” means dichloromethane; “MeCN” or “ACN” means acetonitrile; “DMF” means -dimethyl formamide; “GDI” means 1,1 ’ -carbonyldi imidazole; “DMA” means dimethyl acetamide; “PdfdppfsCh.” means [1.1 '-Bis(diphenylphosphino)ferrocene]- dichloropalladium(II); “PinjB?” means bis(pinacolato)diboron; “THF” means tetrahydrofuran; “i-PrOH” or “iPrOH” means 2-propanol; “LC” means liquid chromatography; “LCMS” means Liquid Chromatography/Mass spectrometry; “HPLC” means high-performance liquid chromatography; “prep-HPLC” means preparative high-performance liquid chromatography; “IF A” means trifluoroacetic acid; “RP” means reversed phase; "min" means minute(s); "h” means hour(s); “PE” means petroleum ether; “v/V” means volume per volume; “Celite ®” means diatomaceous earth; "DMSO” means dimethyl sulfoxide; "SFC” means Supercritical Fluid Chromatography; “DIPE” means diisopropyl ether; “DIPEA” or “DIEA” means N,N- diisopropylethylamine; “Pdsfdba)?” means Tns(dibenzylideneacetone)-dipalladium; “Pd(OAc)2” means palladium(II) acetate; “AcOH” means acetic acid; “DMAP” means 4- (dimethylamino)pyridine; “t-BuOK”, “BuO” or “KOtBu” means potassium tert-butoxide; “TLC” means thin layer chromatography; “prep-TLC” means preparative TLC; “KOAc” means potassium acetate.

[0179] For intermediates that were used in a next reaction step as a crude or as a partially purified intermediate, estimated mol amounts (in some cases indicated by ~) are indicated in the reaction protocols described below, or alternatively theoretical mol amounts are indicated.

[0180] The meanings of the abbreviations in the nuclear magnetic resonance spectra are provided as follows: s ::: singlet, d :::: doublet, dd :::: double doublet, dt :::: double triplet, ddd :=: doublet of doublets of doublets, Sept = septet, t = triplet, m = multiplet, br = broad, brs :=: broad singlet, q = quartet. Example Al

Intermediate 4

[0181] To a solution of 5-bromo-3-methoxypyrazine-2-carbaldehyde (55.0 g, 0.25 moi) and 2,5-diazaspiro[3.4] octan-6-one TFA salt (89.2 g, 0.28 mol) in DCM (800 mL) was added MgSCh (122 g, 1.01 mol) and EtiN (51.3 g, 0.51 moi, 70.5 mL). The mixture was stirred at 15 °C for 30 mms, and then NaBH(OAc)s (161 g, 0.76 moi) was added at 0 °C. The mixture was stirred at 15 °C for 1 h. The reaction was run in parallel 2 times. TLC (DCM:MeOH =10:1, Rj = 0.4) showed that the reaction was complete. The mixture was diluted with DCM (800 mL) and sat. NaHCCh solution (800 mL). The aqueous phase was separated and extracted with (CHCisti-PrOH = 3:1). The combined organic layers were washed with brine (1000 mL), dried over NajSCti, filtered and concentrated in vacuum to give a residue. The crude product was purified by re-crystallization from EtOAc (300 mL) at 25 °C to give Intermediate 1 (120 g, 69 % yield) as a white solid. ! H NMR: (400 MHz, CDCh) 8 8.24-8. 17 (m, 1H), 6.52 (br s, 1 H), 4.06-3.97 (m, 3FI), 3.81-3.71 (m, 2H), 3.58 (d. ./ 8.6 Hz, 2H), 3.39 (d, J ------ 8.6 Hz, 2H), 2.47-2.32 (m, 4H).

[0182] To a mixture of compound 2-chloro-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)phenol (94.1 g, 370 mmol), Intermediate 1 (110 g, 336 mmol) and K 2 CO 3 (139 g, 1.01 mol) in dioxane (1100 mL) and H 2 O (220 mL) was added Pd(dppf)Ch (24.6 g, 33.6 mmol). The brown mixture was purged with N? (3x) and stirred at 100°C for 1 h. The mixture was poured into H 2 .O (1000 mL) and extracted with ethyl acetate (3 x 1000 mL). The combined organic layers were washed with brine (1000 mL), dried over Na2SO4, filtered and concentrated in vacuum to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 330 g SepaFlash® Silica Flash Column, Eluent of 0 ~ 5% MeOH / DCM@ 100 mL/min) to give Intermediate 2 (123 g, 97 % yield) as a brown solid. i H NMR: (400 MHz, DMSO-rfe) 8 10.38 (br s, 1H), 8.33 (s, 1H), 8.14 (s, 1H), 7.39 - 7.17 (m, 1H), 7.06 (br dd, J = 7.8, 13.7 Hz, 2H), 3.94 (s, 3H), 3.72 (s, 2H), 3.44 (br d, J = 7.1 Hz, 2H), 3.25 (br d, J= 7.1 Hz, 2H), 2.28-2.07 (m, 4H).

[0183] To a mixture of Intermediate 2 (123 g, 328 mmol) and N-Phenyl- bis(trifluoromethane sulfonimide) (123 g, 344 mmol) in DCM (2260 ml.) was added DIEA (127 g, 984 mmol, 171 mL) dropwise at 15 °C. The brown mixture w'as stirred at 15 °C for 16 h. The residue was diluted with ice water (2000 mb) and extracted with DCM (2 x 1000 mL). The combined organic layers were washed with brine (2 x 1000 mL), dried over MgSCh, filtered and concentrated under reduced pressure to give a residue. The crude product was triturated with EtOAc (600 mL) for 2 h and filtered to give Intermediate 3 (136 g, 81 % yield) as a yellow' solid. i H NMR: (400 MHz, DMSOwL) 5 8.54-8.39 (m, 1H), 8.15 (s, 1H), 7.90- 7.60 (m, 3H), 3.96 (s, 3H), 3.75 (s, 2H), 3.45 (d, J ------ 8.1 Hz, 2H), 3.25 (d, ./ 8.0 Hz, 2H), 2.30-2.06 (m, 4H).

[0184] To the mixture of Intermediate 3 (50 g, 98.6 mmol), KOAc (29.0 g, 295 mmol) and dioxane (1000 mL) was added Pd(dppf)Ch (7.22 g, 9.86 mmol) and PinzBz (37.6 g, 148 mmol). The yellow/ mixture was stirred at 90 °C for 16 h. The mixture was filtered and concentrated. The residue was purified by flash silica gel chromatography to give Intermediate 4 (27 g, 70% purity) as a brown solid. MS: ES m/z calculated for C 24 H 31 BCIN 4 O 4 [M-i-H] + 485.2, found 485.0. Example A2 Intermediate 5

[0185] A mixture of Intermediate 4 (2.50 g, 5.6 mmol), 3-Bromo-2-chloroaniiine (1.38 g, 6.70 mmol), Pd(dppf)Ch (377 mg, 516 gmol) and K 2 CO 3 (2.14 g, 15.5 mmol) in dioxane (25 mL) and H 2 O (2.5 mL) was degassed and purged with N 2 . (3x), and then the mixture was stirred at 110 °C for 2 h under N 2 . atmosphere. The mixture was filtered and concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography and re-purified by prep-HPLC to give Intermediate 5 (687 mg, 98% purity) as a yellow oil, MS: ES m/z calculated for C 24 H 24 CI 2 N 5 O 2 [M+H] + 484.1, found 484. 1.

Example A3

Intermediate 6-1

[0186] A mixture of 5-bromo-3-methoxy-pyrazine-2-carbaldehyde (400 mg, 1,84 mmol), (5S)-5-(aminomethyl)pyrrolidin-2-one hydrochloride (416 mg, 2.76 mmol) and TEA (769.63 pL, 5.53 mmol) in DCM (10 ml.,) was stirred at 25 °C for 3 h. After adding NaBH(O Ac)3 (1 .17 g, 5.53 mmol) to the mixture, the mixture was stirred at 25 °C for 1 h. The reaction was quenched by addition water (50 mL), and then extracted with DCM (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over NasSOr, filtered and concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography to give Intermediate 6-1 (600 mg) as a yellow liquid. MS: ESm7Z calculated for C 11 H 16 BrN 4 O 2 , [M+Hf 315.0, found 315.0.

[0187] The intermediates shown in Table A-l were prepared by an analogous reaction protocol as was used for the preparation of Intermediate 6-1 using the appropriate starting materials.

Table A-l

Example A4

Intermediate 6A intermediate 6A

[0188] To a mixture of Methyl 6-chl oro-2, 3 -dihydro-3 -oxo-4- py ridazinecarboxy late (2,00 g, 10.6 mmol) in DMF (20 m.L) was added K 2 CO 3 (2.93 g, 21.2 mmol) and Mel (3.01 g, 2'1.2 mmol, 1.32 m.L) at rt. The mixture was stirred at 25 °C for 12 h to give a yellow suspension. The mixture was concentrated under reduced pressure to remove DMF. The residue was diluted with H 2 O (15 mL) and extracted with EtOAc (3 x 15 mL). The combined organic layers were washed with brine (2 x 15 mL), dried over NazSCL, filtered and concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography to give Intermediate 6A-2 (1.66 g, 95% purity) as a white solid. MS: ES m/z calculated for C 7 H 8 C 1 N 2 O 3 [ M H f 203.0, found 203.0.

[0189] A mixture of Intermediate 6A-2 (700.00 mg, 3.46 mmol) and L1iH’EhO (290 mg, 6.91 mmol) in THF (4.2 mL) and H2O (1.26 mL) was stirred at 40 °C for 2 h. The mixture was concentrated under reduced pressure to remove THF. The residue was purified by prep-HPLC to give Intermediate 6A-1 (451 .00 mg, 90% purity) as an off-white solid. MS: ES m/z calculated for C 6 H 6 CIN 2 O 3 [MvH] + 189.0, found 188.9.

[0190] To a solution of Intermediate 6A-1 (451 mg) in MeOH (10 mL) was added t-BuOK (671mg, 5.98 mmol) at rt. The mixture was stirred at 160 °C for 6 h. The reaction was quenched with IN aqueous HCI (10 mL) and the mixture extracted with DCM (20 mL). The organic phase was washed with 1 N aqueous HCI (2 x 10 mL). The organic phase was dried over anhydrous NazSCh, filtered and concentrated to give a residue, which was purified by flash silica gel chromatography to give Intermediate 6A (215 mg, 65% purity) as a yellow solid. MS: ES m/z calculated for C 7 H 9 N 2 O 4 [M+H] + 185.1, found 185.0.

[0191] A mixture of 2-Chloro-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)aniline (2.1 g, 8.28 mmol), l,3-Dibromo-2-chlorobenzene (4.48 g, 16.6 mmol), Pd(dppf)Ch (606 mg, 828 pmol) and K 2 CO 3 (3.43 g, 24.9 mmol) in dioxane (25 mL) and H 2 O (2.5 mL) was degassed and purged with Na (3x). After stirring the mixture at 100 °C for 2 h under Na atmosphere, the reaction was quenched with H 2 .O (30 mL). The mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were dried over NaaSCh, filtered and concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography to give Intermediate 7-1 (1.4 g) as a yellow' solid. MS: ES m/z calculated for C 12 H 9 B1-CI 2 N • H i ' 315.9, found 316.0.

[0192] The intermediates shown in Table A-2 were prepared by an analogous reaction protocol as was used for the preparation of Intermediate 7-1 using the appropriate starting materials.

Example A6

Intermediate 8-1

[0193] A mixture of Intermediate 7-1 (1.26 g, 3.98 mmol), 2-methyl-3-oxo- pyridazine-4-carboxylic acid (737 mg, 4.78 mmol), TCFH (2.24 g, 7,97 mmol) and NMI (818 mg, 9.96 mmol) in MeCN (12 mL) was stirred at 50 °C for 21 h. The mixture was filtered to give a crude product, which was triturated with EtOAc (2 x 15 mL) and dried to give Intermediate 8-1 (1.55 g, 96% purity) as a yellow solid. MS: ES rn/z calculated for C 18 H 13 BrCl 2 N 3 O 2 [M+ H i ' 552.0, found 551.8.

[0194] The intermediates shown in Table A-3 were prepared by an analogous reaction protocol as was used for the preparation of Intermediate 8-1 using the appropriate starting materials.

Example A7

Intermediate 9-1

[0195] A mixture of Intermediate 8-1 (1.55 g, 3.42 mmol), PinzBz (955.52 mg, 3.76 mmol), Pd(dppf)Cb. (250 mg, 342 jimol) and AcOK (1.01 g, 10.26 mmol) in dioxane (10 niL) was degassed and purged with N? (3x), and then the mixture was stirred at 110 °C for 2 h under N? atmosphere. The mixture was concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography to give Intermediate 9-1 (900 mg) as a yellow solid. MS: ES m/z calculated for C 24 H 25 BCI 2 N 3 O 4 [M+H] + 500.1, found 500.0. [0196] The intermediates shown in Table A-4 were prepared by an analogous reaction protocol as was used for the preparation of Intermediate 9-1 using the appropriate starting materials.

Example A8

Intermediate 10-1

[0197] To the solvent mixture of dioxane (8 mL) and H 2 O (0.8 m.L) were added Intermediate 9-1 (850 mg, 1.70 mmol), 6-chIoro-2-methoxy-pyridine-3-carbaldehyde (321 mg, 1.87 mmol), Pd(dppf)Ch (124mg, 170 nmol) and K 2 CO 3 (705 mg, 5.10 mmol). The mixture was degassed and purged with Nz (3x), and then the mixture was stirred at 110 °C for 2 li under N? atmosphere. The mixture was concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography to give Intermediate 10-1 (720 mg) as a yellow solid. MS: ES m/z calculated for C 25 H 19 CI 2 N 4 O 4 [M+H] + 509.1, found 509.1.

[0198] The compounds shown in Table A- 5 were prepared by an analogous reaction protocol as was used for the preparation of Intermediate 10-1 using the appropriate starting materials.

[0199] A mixture of Intermediate 5 (50 mg, 103 itniol), 2-methyl-3-oxo- pyridazine-4-carboxylic acid (19 mg, 124 timoi), chioro-N,N,N’,N'- tetramethylformamidinium hexafiuorophosphate (TCFH) (58 mg, 206 pmol) and N- methyl imidazole (NMI) (21 mg, 258 nmol) in MeCN (1.5 mL) was stirred at 50 °C for 16 h. The reaction was repeated using the same amount starting materials. The combined reaction mixture was diluted with H 2 O (10 mL) and extracted with DCM (3 x 5 mL). The combined organic layers were dried over anhydrous NajSCh, filtered and concentrated to give a residue, which was purified by prep-HPLC twice to give Compound A~1 (44 mg, 96% purity) as a yellow solid. NAIR (400 MHz, CDCh-d) 8 12.23 (br s, 1H), 8.66 (dd, J = 1.4, 8.3 Hz, 1H), 8.48 (s, 1H), 8.33 (d, J = 4.3 Hz, 1H), 8.06 (d, J = 4.3 Hz, 1H), 7.64 (dd, J = 1.7, 7.7 Hz, 1H), 7.52-7.33 (m, 3H), 7.13 (dd, J - 1.5, 7.6 Hz, 1H), 6.40 (br s, 1H), 4.05 (s, 3H), 3.97 (s, 3H), 3.92 (s, 2H), 3.69 (br d, J - 8.0 Hz, 2H), 3.61-3.53 (m, 2H), 2.45-2.33 (m, 4H).

[0200] The compounds shown in Table B-l were prepared by an analogous reaction protocol as was used for the preparation of Compound A-l using the appropriate starting materials.

Example 2 Compound B-l

[0201] To the solvent mixture of 1,4-dioxane (2 mL) and H 2 O (0.2 mL) were added

Intermediate 6-1 (50 mg, 159 gmol), Intermediate 9-1 (87 mg, 1751 gmol), Pd-118 (10 mg, 16 gmol) and K 2 CO 3 (66 mg, 476 gmol). The mixture was degassed and purged with N?. (3x). The mixture was stirred at 110 °C for 2 h under N?. atmosphere. The mixture was filtered and then concentrated to give a residue, which was purified by prep-HPLC to give Compound B- 1 (36 mg, 99 % purity) as a yellow solid.

[0202] The compounds shown in Table B-2 were prepared by an analogous reaction protocol as was used for the preparation of Compound JB-1 using the appropriate starting materials.

[0203] A mixture of (3R, 4R)-4-aminotetrahydropyran-3-ol HCI (74 mg, 485 pmol) and Intermediate 10-1 (170 mg, 334 gmol) in DCM (3 mL) was stirred at 25 °C for 2 li. To the mixture was added NaBH(OAc)? (202 mg, 954 pmol). The mixture was stirred at 25 °C for 12 h. The reaction was quenched by the addition water (0.5 mL). The mixture was concentrated under reduced pressure to give a residue, which was purified by prep-HPLC to give Compound C-l (125 mg, 99 % purity) as a white solid.

[0204] The compounds shown in Table B-3 were prepared by an analogous reaction protocol as was used for the preparation of Compound C-l using the appropriate starting materials. Example 3 Compound D-l

[0205] A mixture of Compound B-l (55 mg, 90 gmol), ethyl chloroformate (18 p1, 181 pmol) and K 2 CO 3 (37 mg, 271 gmol) in THF (2 mL) was stirred at 25°C for 1 h. The mixture concentrated to give a residue concentrated to give a residue, which was purified by prep-HPLC to give Compound D-l (20 mg, 99% purity) as a yellow solid.

[0206] The compound shown in Table B-4 was prepared by an analogous reaction protocol as was used for the preparation of Compound D-l using the appropriate starting materials. Example 4 Compound E-l

[0207] To a solution of Compound C-l (80 mg, 131 gmol) in THF (0.5 mL) was added NaH (15.72 mg, 393 umol, 60% purity) at 0 °C. .After the mixture was stirred at 0 °C for 0.5 h, DMAP (1.60 mg, 13.10 umol, 0.1 eq.) and (4-nitrophenyl)methyl carbonochloridate (28.25 mg, 131.04 umol, 1 eq.) were added into the mixture at 0 °C. The mixture was stirred at 25 °C for 4 h. The reaction was quenched by the addition water. The mixture was extracted with EtOAc (2 x 15 mL). The combined organic layers were dried with Na?S()4, filtrated and concentrated under reduced pressure to give a residue, which was purified by prep-HPLC to give Compound E-l (3.6 mg, 95% purity) as a white solid. [0208] To a solution of compound C-94 (448 mg, 734 pmol) and (5-Methyl-2-oxo- l,3-dioxol-4-yl)methyl 4-nitrophenyl carbonate (267.60 mg, 771 pmol) in DMF (5 mL) was added DIEA (383 gL, 2.20 mmol) at 0 °C. The mixture was stirred at 0 °C for 30 mm, then allowed to warm to 30 °C and stirred for 16 li. The mixture was filtered to give a filtrate, which was purified by prep-HPLC to give Compound F-l (172 mg, 99% purity) as a white solid. MS: ES m/z calculated for C 36 H 34 CI 2 N 5 O 10 [M+ Hf 766.2, found 766.1. NMR (400 MHz, CDCL3) 5 12.24 (s, IH), 8.65 (dd, J - 1.4, 8.3 Hz, H l). 8.33 (d, J == 4.3 Hz, 1 11). 8.06 (d, J == 4.1 Hz, IH), 7.69-7.64 (m, IH), 7.54-7.54 (m, IH), 7.54-7.37 (m, 3H), 7.31-7.28 (m, IH), 7.13 (br d, J = 7.6 Hz, IH), 4.89 (d, J = 17.0 Hz, 2H), 4.51 (d, J = 10.6 Hz, 2H), 4.13 (q, J = 7.2 Hz, 2H), 4.03 (s, 3H), 3.97 (s, 3H), 3.70-3.61 (m, 2H), 2.68-2.57 (m, 2H), 2.18 (d, J = 14.1 Hz, 3H), 1.26 (dt, J = 2.8, 7.1 Hz, 3H).

Compound G-1

[0209] A mixture of Compound C-38 (410 mg, 686 pmol) and 4- methyl benzenesulfonic acid;hydrate (300.24 mg, 1.58 mmol) in EtOH (8 mL) was stirred at 80 °C for 16 h. The mixture was concentrated under reduced pressure to give a residue, which was purified by prep-HPLC to give Compound G-l as an off-white solid. MS: ES ni/z calculated for CsoHioChNsOe [M+Hf 626.2 found 626.1. ! H NMR (400 MHz, CDCLi) 5 12.27-12.20 (m, 1H), 8.65 (dd, J = 1.5, 8.3 Hz, IH), 8.33 (d, J = 4.3 Hz, 1H), 8.06 (d, J = 4.3 Hz, 1H), 7.70-7.62 (m, 2H), 7.42 (td, J = 7.8, 14.4 Hz, 2H), 7.32-7.27 (m, 2H), 7.13 (dd, J = 1.5, 7.6 Hz, 1H), 4.42-4.35 (m, 1H), 4.27 (q, J == 7.1 Hz, 2H), 4.04 (s, 3H), 3.97 (s, 3H), 3.96- 3.95 (m, 1 H), 3.95-3.82 (m, 2H), 3.12 (dd, J == 3.8, 12.3 Hz, 1H), 2.96 (dd, J == 6.3, 12.4 Hz, 1H), 1.34-1.29 (m, 3H).

[0210] To a mixture of Compound G-l (70 mg, 89 pmol) in THF ( 1 mL) was added CDI (72 mg, 447 pmol) at 2.0 °C. The resulting mixture was stirred at 50 °C for 16 h to give a yellow solution. The yellow reaction solution was cooled to 20 °C. The resulting mixture was diluted with DCM (10 mL), and then was washed with H2O (5 mL). The aqueous phase was extracted with DCM (2 x 10 mL). The combined organic layers were filtered and concentrated under reduced pressure to give a residue, which was purified by prep-HPLC to give Compound H-l as a white solid. NMR (400 MHz, CDCLs) 8 = 12.24 (br s, 1H), 8.81-8.53 (m, 1H), 8.45-8.22 (m, 1H), 8.06 (br s, 1H), 7.65 (br s, 2H), 7.51-7.37 (m, 2H), 7.30 (br s, 2H), 7.13 (br s, 1H), 5.02-4.82 (m, 1H), 4.49 (br s, 2H), 4.36-4.22 (m, 2H), 4.03 (br s, 3H), 3.97 (br s, 3H), 3.85 (br s, 1H), 3.67 (br s, 1H), 1.36-1.24 (m, 3H). Example 5

Additional Compounds

[ 0211] Other compounds that can be prepared applying similar procedures as those described herein.

Example A

LCMS (Liquid chromatography/Mass spectrometry)

[0212] The High Performance Liquid Chromatography (HPLC) measurement was performed using a LC pump, a diode-array (DAD) or a LA’ detector and a column as specified in the respective methods. If necessary, additional detectors were included (see table of methods below). Flow from the column was brought to the Mass Spectrometer (MS) which was configured with an atmospheric pressure ion source. It is within the knowledge of the skilled person to set the tune parameters (e.g., scanning range, dwell time) in order to obtain ions allowing the identification of the compound's nominal monoisotopic molecular weight (MW). Data acquisition was performed with appropriate software. Compounds are described by their experimental retention times (Rt) and ions. If not specified differently in the table of data, the reported molecular ion corresponds to the [M+H] + (protonated molecule) and/or [M- H]’(deprotonated molecule). In case the compound was not directly ionizable the type of adduct is specified (i.e. [M+NI-Hp, [M-i-Nap, [M+HCOO]', etc.). For molecules with multiple isotopic patterns (Br, CI), the reported value is the one obtained for the lowest isotope mass. All results were obtained with experimental uncertainties that are commonly associated with the method used. Hereinafter, "SQD" means Single Quadrupole Detector, "MSD" Mass Selective Detector, "RT" room temperature, "BEH" bridged ethylsiloxane/ silica hybrid, "DAD" Diode Array Detector, "HSS" High Strength silica., "Q-Tof Quadrupole Time-off light mass spectrometers, "CLND", ChemiLuminescent Nitrogen Detector, "ELSD" Evaporative Light Scanning Detector.

Example A-l

NMR (Nuclear Magnetic Resonance)

[0213] Proton ( ! H) NMR spectra were recorded on a Bruker spectrometer operating at 400 MHz using CDCL? (deuterated chloroform) or DMSO-rfc (deuterated DMSO, dimethylde-sulfoxide) as solvents. Chemical shifts (d) are reported in parts per million (ppm) relative to tetramethylsilane (TMS), which was used as an internal standard.

[0214] Compound C-7: ] H NMR (400 MHz, CDCL?) 8 12.32-12.08 (m, IH), 8.65 (dd, J - 1 .4, 8.3 Hz, H I). 8.33 ( d. J - 4.3 Hz, 1 H ). 8.06 (d, J - 4. 1 Hz, IH), 7.74-7.61 (m, 2H), 7.41 (td, J = 7.8, 12.9 Hz, 2H), 7.31-7.28 (m, 211). 7.13 (dd, J 1.4, 7.6 Hz, 1H), 4.60-4.49 (m, 1H), 4.03 (s, 3H), 3.97 (s, 3H), 3.94-3.77 (m, 4H), 3.35 (br s, 2H).

[0215] Compound C-9: 1 H NMR (400 MHz, CDCL3) 5 12.23 (s, IH), 8.65 (dd, J = 1.3, 8.3 Hz, IH), 8.34-8.31 (m, IH), 8.06 (d, J = 4.3 Hz, IH), 7.93 (d, J = 7.6 Hz, HI), 7.65 (dd, J = 1.6, 7.8 Hz, IH), 7.54-7.28 (m, 4H), 7.11 (dd, J = 1.4, 7.5 Hz, IH), 4.70-4.33 (m, 2H), 4.27 (s, 2H), 4.23-4.09 (m, 2H), 4.08-4.02 (m, 3H), 3.99-3.94 (m, 3H),3.55 (br d, J - 10.3 Hz, IH).

[0216] Compound C-13: ! H NMR (400 MHz, CDCLs) 8 12.24 (s,lH), 8.65 (dd, J = 1.4, 8.3 Hz, IH), 8.33 (d, J = 4.1 Hz, IH), 8.06 (d, J = 4.3 Hz, IH), 7.72-7.57 (m, 2H), 7.41 (td, J = 7.8, 10.9 Hz, 2H), 7.29 (d, J = 1.6 Hz, 2H), 7.26 (s, IH), 7.12 (dd, J = 1.4, 7.6 Hz, IH), 4.23-4.14 (m, 2H), 4.02 (s, 3H), 3.97 (s, 3H), 3.69-3.61 (m, 4H), 3.46-3.32(m, 3H), 1.28 (t, J = 7.1 Hz, 3H). [0217] Compound C-81: 1 H-NMR(DMSO-d 6 , 400 MHz): 1.51-1.57 (m, 1H), 1.64- 1.72 (m, IH), 2.44-50 (m, IH), 2.70-2.74 (m, IH), 3.24-3.36 (m, 2H), 3.66-3.70 (m, 2H), 3.73- 3.78 (m, 3H), 3.86 (s, 3H), 3.99 (s, 3H), 4.65 (d, J - 3.6 Hz, IH), 7.22 (dd, J == 7.6 Hz, 1.5 Hz, 1H), 7.47-7.57 (m, 3H), 7.8 (dd, J - 7.6 Hz, 1.7 Hz, 1H), 8.26-8.29 (m, 2H), 8.62 (dd, J - 8.3 Hz, 1.5 Hz, IH), 8.67 (s, IH), 12.36 (s, IH).

Example B PDL1/PD1 Binding Assay

[0218] Compounds to be tested were serially diluted in DMSO, and further diluted m assay buffer (25 mM Hepes pH 7.4, 150 mM NaCl, 0.005% Tween 20, BSA 0.01%). Diluted compounds were added to the wells with final concentration of DMSO at 1%. PDLl-6xHis protein was added to the wells, mixed 'well with compound. The plates were incubated for 30 min at room temperature. PDl-Fc-Avi -Biotin protein was added to the wells. Final concentration of PDL1 and PD1 protein is 0.3 nM and 2.5 nM, respectively. After a binding time of 30 min at room temperature, Anti-6xHis Acceptor beads (final concentration 20 ug/ml) were added to the wells, and the incubation continued for 1 h. Streptavidin Donor beads (final concentration 20 ug/mL) were added at reduced light. The plates w'ere sealed with foil and incubated in the dark for additional 1 h or overnight before reading on an Envision reader. The ICso values were determined by fitting the curves using a four-parameter equation in Graphpad Prism 8,

Example C PD-1/PD-L1 NEAT Reporter Assay

[0219] Cellular activity of the compounds was assessed using a co-culture reporter assay in which TCR-mediated NEAT activity of Jurkat T cells is constitutively inhibited by the engagement of PD-1 by PD-L1 expressing CHO cells. Blocking the PD-1/ PD-L1 interaction will release the inhibitory signal and results in TCR signaling and NFAT-mediated luciferase activity.

[0220] CHO cells expressing surface-bound anti-CD3 antibodies and PD-L1 were first seeded overnight and treated with the compounds. Jurkat cells overexpressing PD-1 and a luciferase construct under NEAT promoter were then immediately seeded on the monolayer of CHO cells. The co-culture was then incubated for 6 hrs at 37 °C. Luciferase activity was assessed by adding the ONE-Glo reagent and measuring luminescence with a plate reader. ECso values were determined from the fit of the dose-response curves.

[0221] Compounds described herein, as exemplified in the Examples, showed ECso or IC50 values in the following ranges: A: ICso or ECso <10 nM; B: 10 nM < ICso or ECso <100 nM; C: 100 nM < ICso or ECso <1000 nM; D : 1000 nM < ICso or ECso <10000 nM; E : ICso or ECso > 10000 nM; n.d. - not determined; n.r. = ECso not reached in the range of tested concentrations starting from 1 nM to 5000 nM.

[0222] Although the foregoing has been described in some detail by way of illustrations and examples for purposes of clarity and understanding, it will be understood by those of skill in the art that numerous and various modifications can be made without departing from the spirit of the present disclosure. Therefore, it should be clearly understood that the forms disclosed herein are illustrative only and are not intended to limit the scope of the present disclosure, but rather to also cover all modification and alternatives coming with the true scope and spirit of the present disclosure.