Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INHIBITORS OF ANO6 AND THEIR USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2022/195522
Kind Code:
A1
Abstract:
The present invention relates to a composition comprising at least one active agent that can inhibit anoctamin 6 protein and a method of treating or preventing disease, disorder, or condition associated with virus infection by administering the composition to a subject. In addition, it relates to a method of disinfecting or sanitizing an object from virus contamination by contacting the composition with or applying the composition to an object.

Inventors:
OH CHANGMOK (KR)
KWON SUNGWOOK (KR)
LIM DAMI (KR)
LEE EUNJUNG (KR)
LEE SEOLHEE (KR)
YOON HONGCHUL (KR)
AN KYUNG MI (KR)
PARK JOONTAE (KR)
CHO SUNG MIN (KR)
CHOI WON GU (KR)
LEE MIN GOO (KR)
LEE JAE MYUN (KR)
NAMKUNG WAN (KR)
LEE JUNGWOO (KR)
Application Number:
PCT/IB2022/052412
Publication Date:
September 22, 2022
Filing Date:
March 17, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ILDONG PHARMACEUTICAL CO LTD (KR)
UIF UNIV INDUSTRY FOUNDATION YONSEI UNIV (KR)
International Classes:
A61K31/50; A61K31/44; A61K31/501; A61K31/505; A61K31/506; A61P31/14; C07D213/74; C07D237/20; C07D239/42; C07D405/04
Domestic Patent References:
WO2002046171A22002-06-13
WO2020011811A12020-01-16
WO2005026129A12005-03-24
Foreign References:
KR20150064730A2015-06-11
KR20150025531A2015-03-11
Attorney, Agent or Firm:
TAEWOOL PATENT LAW FIRM (KR)
Download PDF:
Claims:
CLAIMS

1. A composition for treating or preventing disease, disorder, or condition associated with virus infection, the composition comprising a therapeutically effective amount of a compound represented by Formula (I), a pharmaceutically acceptable salt of the compound, a solvate of the compound, or a hydrate of the compound: wherein ring A and ring B each are independently a monocyclic aliphatic ring, a polycyclic aliphatic ring, a monocyclic aromatic ring, or a polycyclic aromatic ring, which optionally contains at least one heteroatom selected from the group consisting of N, NO, NO2, S, SO, SO2, and O, wherein the ring A and ring B each are optionally and independently substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, ester, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic; wherein Ri and R3 each are independently hydrogen, halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, or aryl aliphatic, wherein Ri and R3 each are optionally and independently substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, ester, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic; wherein R2 is hydrogen, C1-5 alkyl or C3-6 cycloalkyl; wherein Li and L2 each are independently C1-C10 aliphatic, C3-C10 cycloaliphatic, or C3-C10 hetero cycloaliphatic, wherein Li and L2 each are optionally and independently substituted with at least one substituent selected from the group consisting of CN, C1-5 alkyl, and C3-6 cycloalkyl; and wherein m and n each are independently 0 or 1.

2. The composition of claim 1, wherein the ring A and ring B each are independently a 5- membered ring or a 6-membered ring.

3. The composition of claim 1 or 2, wherein the ring A is a monocyclic or polycyclic aliphatic ring which optionally contains at least one heteroatom selected from the group consisting of N, NO, NO2, S, SO, SO2, and O, or a monocyclic or polycyclic aromatic ring which optionally contains at least one heteroatom selected from the group consisting of N, NO, NO2, S, SO, SO2, and O; and/or wherein the ring B is a monocyclic or polycyclic aliphatic ring which optionally contains at least one heteroatom selected from the group consisting of N, O, and S, or a monocyclic or polycyclic aromatic ring which optionally contains at least one heteroatom selected from the group consisting of N, O, and S.

4. The composition of any one of claims 1-3, wherein the ring A is phenyl, pyridinyl, diazinyl, pyrimidinyl, triaziny, piperidinyl, oxadiazoline, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.

5. The composition of any one of claims 1-4, wherein the ring B is a phenyl, pyridinyl, diazinyl, cyclopentadienyl, cyclopentyl, cyclohexyl, adamantane, or bicyclo[2.2.1]heptane.

6. The composition of any one of claims

Xai, Xa2, Xa3, and Xa4 each are independently CH, N, or NH, or wherein the ring A is , in which Yai, Ya2, and Ya3 each are independently

CH, N, NH, S, SH or O.

7. The composition of any one of claims 1-6, wherein the ring B is , in which Xbi, Xb2, Xb3, and Xb4 each are independently CH, N, orNH.

8. The composition of any one of claims 1-7, wherein Ri is hydrogen; Ci-io alkyl; benzyl; alkoxy, CN, COOH, mono or bi aromatic ring which optionally contains at least one heteroatom selected from the group consisting of N, O, and S; mono or bi cycloaliphatic which optionally contains at least one heteroatom selected from the group consisting of N, O, and S; aryl which optionally contains at least one hetero atom selected from the group consisting of N, O, and S; an aromatic ring fused to a non-aromatic ring which optionally contains at least one heteroatom selected from the group consisting of N, O, and S; or an aromatic ring fused to an aromatic ring which optionally contains at least one heteroatom selected from the group consisting of N, O, and S.

9. The composition of any one of claims 1-8, wherein Ri is C1-4 alkyl, benzyl, phenyl, pyridinyl, diazinyl, triazinyl, piperidinyl, furanyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or thiophenyl, which is optionally substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, hetero cycloalkyl, hetero cycloalkenyl, hetero cycloalkynyl, alkoxy, aryl, aryloxy, diaryl, arylalkyl, arylalkyloxy, cycloalkylalkyl, cycloalkylalkyloxy, amino, hydroxy, hydroxyalkyl, acyl, heteroaryl, heteroaryloxy, heteroarylalkyl, heteroarylalkoxy, aryloxyalkyl, alkylthio, arylalkylthio, aryloxyaryl, alkylamido, alkanoylamino, arylcarbonylamino, nitro, cyano, thiol, haloalkyl, trihaloalkyl, alkyl ester, and alkylthio.

10. The composition of any one of claims 1-9, wherein the ring A, Ri, or both comprise a hetero aromatic ring which contains at least one N as the heteroatom.

11. The composition of any one of claims 1-10, wherein the R3 is hydrogen; halogen; halogen derivatives; CN; alkoxy, carboxyl, carbonyl, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, aryl aliphatic or fused ring.

12. The composition of any one of claims 1-11, wherein the group is one of the following groups:

13. The composition of any one of claims 1-12, wherein the group is one of the following groups:

14. The composition of any one of claims 1-13, wherein the group is one of the following groups:

15. The composition of any one of claims 1-14, wherein the group is one of the following groups:

16. The composition of any one of claims 1-15, wherein the disease, disorder, or condition associated with virus infection is at least one selected from the group consisting of cold, severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and COVID- 19.

17. The composition of any one of claims 1-16, wherein the virus is an RNA virus.

18. The composition of claim 17, wherein the RNA virus is selected from the group consisting of Coronaviridae, Amalgaviridae, Birnaviridae, Chrysoviridae, Cystoviridae, Endornaviridae, Hypoviridae, Megabirnaviridae, Partitiviridae, Picobirnaviridae, Reoviridae, Totiviridae, Quadriviridae, Arteriviridae, Mesoniviridae, Roniviridae, Dicistroviridae, Iflaviridae, Marnaviridae, Piconaviridae, Secoviridae, Alphaflexiviridae, Betaflexiviridae, Gammaflexiviridae, Tymoviridae, Bomaviridae, Filoviridae, Paramyxoviridae, Phabdoviridae, Nyamiviridae, Caliciviridae, Flaviviridae, Luteoviridae, Togaviridae, Pneumoviridae, Arenaviridae, Deltaviridae, and Orthomyxoviridae.

19. The composition of claim 1, wherein the compound represented by Formula (1) is selected from the group consisting of:

20. A composition for disinfecting or sanitizing an object from virus contamination by contacting the composition with or applying the composition to an object, the composition comprising a therapeutically effective amount of a compound represented by Formula (I), a pharmaceutically acceptable salt of the compound, a solvate of the compound, or a hydrate of the compound: wherein ring A and ring B each are independently a monocyclic aliphatic ring, a polycyclic aliphatic ring, a monocyclic aromatic ring, or a polycyclic aromatic ring, which optionally contains at least one heteroatom selected from the group consisting of N, NO, NO2, S, SO, SO2, and O, wherein the ring A and ring B each are optionally and independently substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, ester, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic; wherein Ri and R3 each are independently hydrogen, halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, or aryl aliphatic, wherein Ri and R3 each are optionally and independently substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, ester, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic; wherein R2 is hydrogen, C1-5 alkyl or C3-6 cycloalkyl; wherein Li and L2 each are independently C1-C10 aliphatic, C3-C10 cycloaliphatic, or C3-C10 hetero cycloaliphatic, wherein Li and L2 each are optionally and independently substituted with at least one substituent selected from the group consisting of CN, C1-5 alkyl, and C3-6 cycloalkyl; and wherein m and n each are independently 0 or 1.

Description:
INHIBITORS OF AN06 AND THEIR USES THEREOF

CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 63/162,511 filed March 17, 2021, which is incorporated herein in its entirety by reference.

TECHNICAL FIELD

The present invention relates to compounds capable of inhibiting anoctamin 6 (AN06) protein, compositions comprising the compounds, methods for preparing the compounds, and methods of using the compounds or compositions. More particularly, the present invention relates to using the compounds or compositions to treat virus infection.

BACKGROUND

Coronaviruses are enveloped RNA viruses that deliver their viral genome into the host cells by fusing the viral envelope with the host cell membrane. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), belongs to the b-coronavirus genera and has the spike (S) glycoprotein, a class I viral fusion protein, on the virion envelope. P. V’Kovski, A. Kratzel, S. Steiner, H. Stalder, V. Thiel, Nat Rev Microbiol 19, 155 (2021); Gordon, D.E., Jang, G.M., Bouhaddou, M. et al, Nature 583, 459-468 (2020). The SARS-CoV-2 S protein is the primary determinant of cell tropism and mediates binding to angiotensin-converting enzyme 2 (ACE2), the viral entry receptor on the host cells, which constitutes the initial step of the membrane fusion process. However, the viral and host cell membrane fusion process is not yet fully understood.

AN06, also known as TMEM16F, is a cell membrane protein functioning as a Ca 2+ - activated CT channel (CACC) and Ca 2+ -dependent phospholipid scramblase. H. Yang, A.

Kim, T. David, D. Palmer, T. Jin, J. Tien, F. Huang, T. Cheng, S. R. Coughlin, Y. N. Jan, L. Y. Jan, Cell 151, 111 (2012). Phosphatidylserine (PS), an anionic phospholipid, is mostly found in the inner leaflet of the cell membrane under normal conditions. The exposure of PS at the external surface of plasma membrane is associated with diverse physiological and pathological events, such as platelet aggregation, innate immunity and apoptotic cell death. A. Amara, J. Mercer, Nat Rev Microbiol 13, 461 (2015). This cell surface extemalization of PS is mediated by the inactivation of lipid flippases that build membrane PS asymmetry, or by the activation of phospholipid scramblases, such as AN06, that enhance the translocation of anionic phospholipids. A. Amara, J. Mercer, Nat Rev Microbiol 13, 461 (2015). Notably, a number of studies have shown that the cell surface exposure of PS is associated with membrane fusion events not only between mammalian cell membranes, such as myotube formation, but also between the viral envelope and host cell membrane, which enhances the virus entry into host cells. D. A. Coil, A. D. Miller, J Virol 79, 11496 (2005); P. Younan, M. Iampietro, R. I.

Santos, P. Ramanathan, V. L. Popov, A. Bukreyev, J Infect Dis 218, S335 (2018); and E. Zaitseva, E. Zaitsev, K. Melikov, A. Arakelyan, M. Marin, R. Villasmil, L. B. Margolis, G. B. Melikyan, L. V. Chernomordik, Cell Host Microbe 22, 99 (2017).

However, whether AN06-mediated PS externalization is involved in coronavirus entry into host cells has not been studied. We have discovered that AN06-mediated PS externalization in host cell membranes participates in the entry of enveloped viruses (e.g., SARS-CoV-2) and that AN06 inhibition is effective against the enveloped virus infection.

DEFINITIONS

As used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “an agent” includes a plurality of such agents, and reference to “a compound” or “the compound” includes reference to one or more compounds and equivalents thereof (e.g., plurality of compounds) known to those skilled in the art, and so forth. The term “about” when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary between 1% and 20% of the stated number or numerical range.

Aliphatic hydrocarbon compounds are saturated or unsaturated hydrocarbons based on chains of carbon atoms. They include alkyl, alkenyl, and alkynyl compounds, and their derivatives. The term “alkyl,” when used alone or as part of a larger moiety such as “arylalkyl,” or “cycloalkyl” refers to a straight- or branch-chained, saturated hydrocarbon containing a certain number of carbon atoms (e.g, 1-14 carbon atoms, 1-10 carbon atoms, 1-6 carbon atoms, or 1-4 carbon atoms). For example, “C 1 -C 6 alkyl” refers to alkyl having 1 to 6 carbon atoms and is intended to include Ci, C 2 , C 3 , C 4 , C 5 , Ce alkyl groups. Non-limiting examples of alkyl groups include methyl (Me), ethyl (Et), propyl (e.g, «-propyl and iso propyl), butyl (e.g, «-butyl, .vo-butyl, /-butyl), and pentyl (e.g, «-pentyl, /50-pentyl, neo pentyl), as well as chain isomers thereof. The term “alkenyl” when used alone or as part of a larger moiety such as “arylalkenyl,” or “cycloalkenyl” refers to a straight- or branch-chained hydrocarbon containing one or more double bonds and containing a certain number of carbon atoms (e.g., 2-14 carbon atoms, 2-10 carbon atoms, 2-6 carbon atoms, or 2-4 carbon atoms). For example, “C2-C6 alkenyl” refers to alkenyl having 2 to 6 carbon atoms and is intended to include C2, C3, C4, C5, Ce alkenyl groups. Non-limiting examples of alkenyl groups include ethenyl, propenyl, butenyl, l-methyl-2-buten-l-yl, heptenyl, octenyl, and the like, as well as chain isomers thereof.

The term “alkynyl” when used alone or as part of a larger moiety such as “arylalkynyl” or “cycloalkynyl” refers to a straight- or branch-chained hydrocarbon containing one or more triple bonds and containing a certain number of carbon atoms ( e.g ., 2-14 carbon atoms, 2-10 carbon atoms, 2-6 carbon atoms, or 2-4 carbon atoms). For example, “C2-C6 alkynyl” refers to alkynyl having 2 to 6 carbon atoms and is intended to include C2, C3, C4, C5, Ce alkynyl groups. Non-limiting examples of alkynyl groups include ethynyl, propynyl, butynyl, 1 -methyl -2-butyn-l-yl, heptynyl, octynyl, and the like, as well as chain isomers thereof.

Cycloaliphatic hydrocarbon compounds are saturated or unsaturated hydrocarbons containing one (i.e., monocyclic) or more (i.e., polycyclic) non-aromatic rings of carbons. They include cycloalkyl, cycloalkenyl, and cycloalkynyl compounds, and their derivatives Non-limiting examples of cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, cyclododecyl, cyclohexenyl, norbornyl,

The term “hetero” refers to the replacement of at least one carbon atom member in a ring system with at least one heteroatom such as nitrogen, sulfur, sulfoxide, sulfone, and oxygen. For example, the term “heterocyclo aliphatic” means an aliphatic compound having a non-aromatic monocyclic or polycyclic ring with a certain number of carbons (e.g, 2 to 20 carbon atoms, 2-15 carbon atoms, 2-10 carbon atoms, or 2-7 carbon atoms) in the ring and with one or more heteroatoms selected from nitrogen, oxidized nitrogen (e.g, NO and NO2), sulfur, oxidize sulfur (e.g, SO and SO2), and oxygen. The ring or ring system of a heterocyclo aliphatic group of a compound can be linked or fused to one or more different moieties (rings) of the compound via a carbon atom or a heteroatom of the ring. Non-limiting examples of the different ring include a substituted or unsubstituted cycloaliphatic, hetero cycloaliphatic, aromatic, and hetero aromatic ring. A bridged ring may occur when one or more atoms (i.e., C, O, N, or S) link two non-adjacent carbon or nitrogen atoms. Examples of bridged rings include, but are not limited to, one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms, and a carbon-nitrogen group. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.

As used herein, the term “aromatic,” or “aryl” refers to aromatic monocyclic or polycyclic groups. It includes carbocyclic aromatic groups ( e.g ., phenyl, naphthyl, and the like) and heteroaromatic groups (e.g., pyridyl, pyrimidinyl, and the like). The ring or ring system of an aromatic or heterocyclo aromatic group of a compound can be linked or fused to one or more different moieties (rings) of the compound via at least one carbon atom and/or at least one heteroatom of the ring, which results in fused rings (sharing two adjacent atoms), bridged rings (sharing two non-adjacent atoms), and spiro rings (sharing one atom). Non limiting examples of the different ring include a substituted or unsubstituted cycloaliphatic, hetero cycloaliphatic, aromatic, and hetero aromatic ring. For example, an aliphatic ring may be fused with an aromatic ring, as illustrated below. The arrowed lines drawn from the illustrated ring system indicate that the bond may be attached to any of the suitable ring atoms.

A bridged ring may occur when one or more atoms (e.g, C, O, N, or S) link two non- adjacent carbon, two non-adjacent heteroatoms, or one carbon and one heteroatom. Examples of bridged rings include, but are not limited to, one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms, and a carbon-nitrogen group. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.

Non-limiting examples of heterocyclic groups include azetidinyl, pyrrolidinyl, oxetanyl, imidazolinyl, oxazolidinyl, isoxazolinyl, thiazolidinyl, isothiazolidinyl, tetrahydrofuranyl, piperidyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidyl, 2-oxopyrrolodinyl, 2-oxoazepinyl, azepinyl, 4-piperidonyl, tetrahydropyranyl, morpholinyl, thiamorpholinyl, thiomorpholinyl sulfoxide, thiamorpholinyl sulfone, 1,3-dioxolane, tetrahydro-1,1- dioxothienyl, quinuclidinyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrroyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, thiophenyl, indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, purinyl, carbazolyl, benzimidazolyl, indolinyl, benzodioxolanyl, and benzodioxane.

As used herein, the term “alkoxy” refers to the alkyl groups above bound through oxygen, examples of which include methoxy, ethoxy, /.vo-propoxy, tert- butoxy, and the like. In addition, alkoxy also refers to polyethers such as -0-(0¾) 2 -0-(2H 3 , and the like.

As used herein, the term “hydroxyalkyl” refers to any hydroxyl derivative of alkyl radical. The term “hydroxyalkyl” includes any alkyl radical having one or more hydrogen atoms replaced by a hydroxy group.

As used herein, the term “aryl aliphatic” refers to aliphatic hydrocarbon compounds having one or more hydrogen atoms replaced by an aryl group. The term “arylalkyl,” or “alkylaryl” includes any alkyl radical having one or more hydrogen atoms replaced by an aryl group, e.g ., a benzyl group, a phenethyl group, and the like. The term “arylalkenyl” includes any alkenyl radical having one or more hydrogen atoms replaced by an aryl group. The term “arylalkynyl” includes any alkynyl radical having one or more hydrogen atoms replaced by an aryl group. The term “aryl aliphatic” is meant to include arylalkyl, arylalkenyl, and arylakynyl.

As used herein, the term “amine” refers to a derivative of ammonia in which one, two, or all three hydrogen atoms are replaced by hydrocarbon groups including aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, and hetero aromatic. The term “alkyl amine,” or “amine alkyl” refers to ammonia derivative having one, two, or all three hydrogen atoms replaced by an alkyl group. Unless otherwise specified, the term herein includes cyclic amines as well primary, secondary, tertiary amines. Non-limiting examples of amines include, but are not limited to, N(C2H5)2, N(CH3)2, N(C2H5)(benzyl), methyl piperazine, methyl piperidine, ethyl piperazine, and ethyl piperidine.

As used herein, the term “amide” refers to a carbonyl group bonded to a nitrogen. The simplest example is CONH2. Non-limiting examples of amines include the ones in which one or two of the hydrogen atoms are replaced by other groups including aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, and hetero aromatic.

As used herein, the term “sulfhydryl,” “sulfanyl,” or “thiol” refers to any organosulfur compound containing -SH group. The compounds are in the form R-SH, wherein R represents an aliphatic, aromatic ring or other organic substituent. Aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, heteroaromatic, alkoxy, aryl aliphatic (e.g., arylalkyl), carboxyl, carbonyl, hydroxyl, amine, amide, thioalkyl, and sulfhydryl each independently can be unsubstituted or substituted with one or more suitable substituents. Non-limiting examples of the substituents include halogen or halogen derivatives (e.g., F, Br, Cl, I, OCHF2, CF3, CHF2, or OCF3), alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, hetero cycloalkyl, hetero cycloalkenyl, hetero cycloalkynyl, alkoxy, aryl, aryloxy, diaryl, arylalkyl, arylalkyloxy, cycloalkylalkyl, cycloalkylalkyloxy, amino, hydroxy, hydroxyalkyl, acyl, heteroaryl, heteroaryloxy, heteroarylalkyl, heteroarylalkoxy, aryloxyalkyl, alkylthio, arylalkylthio, aryloxyaryl, alkylamido, alkanoylamino, arylcarbonylamino, nitro, cyano, thiol, haloalkyl, trihaloalkyl, and alkylthio. Also, non-limiting examples of the substituents include =0, -OR x , -SR X , =S, -NR x R y , -N(alkyl)3, -NR X S02, -NR x S02R y , -S02R X -, - S0 2 NR x R y , -S0 2 NR x COR y , -SO3H, -PO(OH) 2 , -COR x , -COOR x , COOC(alkyl) , -CONR x R y , - CO(C 1 -C 4 alkyl)NR x R y , -CONR x (S0 2 )R y , -C0 2 (Ci-C 4 alkyl)NR x R y , -NR x COR y , -NR x C0 2 R y , - NR X (C I -C 4 alkyl)C0 2 R y , =N-OH, and =N-0-alkyl. R x and R y each may be independently selected from hydrogen, alkyl, alkenyl, C3-C7 cycloalkyl, C 5 -C 11 aryl, benzyl, phenylethyl, naphthyl, a 3- to 7-membered heterocycloalkyl, and a 5- to 6-membered heteroaryl.

A “substituent” as used herein refers to a molecular moiety that is covalently bonded to an atom within a molecule of interest. For example, a ring substituent may be a moiety such as a halogen, alkyl group, haloalkyl group or other group that is covalently bonded to an atom (preferably a carbon or nitrogen atom) that is a ring member. Substituents of aromatic groups are generally covalently bonded to a ring carbon atom. The term “substitution” refers to replacing a hydrogen atom in a molecular structure with a substituent, such that the valence on the designated atom is not exceeded, and such that a chemically stable compound (i.e., a compound that can be isolated, characterized, and tested for biological activity) results from the substitution.

When the term “unsaturated” is used herein to refer to a ring or group, the ring or group may be fully unsaturated or partially unsaturated.

As described above, certain groups can be unsubstituted or substituted with one or more suitable substituents by other than hydrogen at one or more available positions, typically 1, 2,

3, 4, or 5 positions, by one or more suitable groups (which may be the same or different). Certain groups, when substituted, are substituted with 1, 2, 3 or 4 independently selected substituents. Suitable substituents include, but are not limited to, halo, alkyl, haloalkyl, aryl, hydroxy, alkoxy, hydroxyalkyl, amino, and the like. The term “compound” as used herein is meant to include all stereoisomers, geometric isomers, tautomers, isotopes, and prodrug of the chemical structures depicted.

The compounds herein described may have asymmetric centers, geometric centers (e.g., double bond), or both. All chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated. In some embodiments, the compounds described herein have one or more chiral centers. It is understood that if an absolute stereochemistry is not expressly indicated, then each chiral center may independently be of the R-configuration or the S- configuration or a mixture thereof. Thus, compounds described herein include enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions. Racemic mixtures of R-enantiomer and S-enantiomer, and enantio-enriched stereometric mixtures comprising of R- and S-enantiomers, as well as the individual optical isomers can be isolated or synthesized so as to be substantially free of their enantiomeric or diastereomeric partners, and these stereoisomers are all within the scope of the present technology.

Compounds of the present disclosure containing an asymmetrically substituted atom may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms, by synthesis from optically active starting materials, or through use of chiral auxiliaries.

Geometric isomers, resulting from the arrangement of substituents around a carbon- carbon double bond or arrangement of substituents around a cycloalkyl or heterocyclic ring, can also exist in the compounds of the present disclosure. Geometric isomers of olefins, C=N double bonds, or other types of double bonds may be present in the compounds described herein, and all such stable isomers are included in the present disclosure. Specifically, cis and trans geometric isomers of the compounds of the present disclosure may also exist and may be isolated as a mixture of isomers or as separated isomeric forms.

Compounds of the present disclosure also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Examples of prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, amide - imidic acid pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, such as, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.

The term “prodrug” refers to an agent which is converted into a biologically active drug in vivo by some physiological or chemical process. In some embodiments, a prodrug is converted to the desired drug form, when subjected to a biological system at physiological pH. In some embodiments, a prodrug is enzymatically converted to the desired drug form, when subjected to a biological system. Prodrug forms of any of the compounds described herein can be useful, for example, to provide particular therapeutic benefits as a consequence of an extension of the half-life of the resulting compound in the body, or a reduction in the active dose required. Pro-drugs can also be useful in some situations, as they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not. The prodrugs may also have improved solubility in pharmacological compositions over the parent drugs. Prodrug forms or derivatives of a compound of this disclosure generally include a promoiety substituent at a suitable labile site of the compound. The promoiety refers to the group that can be removed by enzymatic or chemical reactions, when a prodrug is converted to the drug in vivo. In some embodiments, the promoiety is a group (e.g., an optionally substituted Ci- 6 alkanoyl, or an optionally substituted Ci- 6 alkyl) attached via an ester linkage to a hydroxyl group or a carboxylic acid group of the compound or drug.

SUMMARY

An aspect of the present invention provides a method for treating or preventing diseases, disorders, or conditions associated with virus infection. The method comprises administering to a subject in need a composition containing a therapeutically effective amount of a compound that can inhibit AN06, a pharmaceutically acceptable salt of the compound, a solvate of the compound, or a hydrate of the compound.

Still another aspect of the present invention provides a method for disinfecting or sanitizing an object from virus contamination. The method comprises contacting with or applying to an object a composition containing a therapeutically effective amount of a compound that inhibits AN06, a pharmaceutically acceptable salt of the compound, a solvate of the compound, or a hydrate of the compound.

In some embodiments, the compound is represented by Formula (I).

Ring A and ring B each are independently a monocyclic aliphatic ring, a polycyclic aliphatic ring, a monocyclic aromatic ring, or a polycyclic aromatic ring, which optionally contains at least one heteroatom selected from the group consisting of N, NO, NO 2 , S, SO,

SO 2 , and O, wherein the ring A and ring B each are optionally and independently substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, ester, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic.

Ri and R 3 each are independently hydrogen, halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, or aryl aliphatic, wherein Ri and R 3 each are optionally and independently substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, ester, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic;

R 2 is hydrogen, C 1-5 alkyl or C 3-6 cycloalkyl.

Li and L2 each are independently C1-C1 0 aliphatic, C3-C10 cycloaliphatic, or C3-C10 hetero cycloaliphatic, wherein Li and L 2 each are optionally and independently substituted with at least one substituent selected from the group consisting of CN, C 1-5 alkyl, and C 3-6 cycloalkyl. m and n each are independently 0 or 1.

In some embodiments, the virus may be an RNA virus. Non-limiting examples of the RNA virus may include Coronaviridae, Amalgaviridae, Birnaviridae, Chrysoviridae, Cystoviridae, Endomaviridae, Hypoviridae, Megabimaviridae, Partitiviridae, Picobimaviridae, Reoviridae, Totiviridae, Quadriviridae, Arteriviridae, Mesoniviridae, Roniviridae, Dicistroviridae, Iflaviridae, Marnaviridae, Piconaviridae, Secoviridae, Alphaflexiviridae, Betaflexiviridae, Gammaflexiviridae, Tymoviridae, Bomaviridae, Filoviridae, Paramyxoviridae, Phabdoviridae, Nyamiviridae, Caliciviridae, Flaviviridae, Luteoviridae, Togaviridae, Pneumoviridae, Arenaviridae, Deltaviridae, and Orthomyxoviridae.

In some embodiments, the diseases, disorders, or conditions associated with virus infection comprises cold, severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and COVID-19.

A further aspect of the present invention provides a composition for treating or preventing diseases, disorders, or conditions associated with virus infection. The composition contains a therapeutically effective amount of a compound that can inhibit AN06, a pharmaceutically acceptable salt of the compound, a solvate of the compound, or a hydrate of the compound.

A still further aspect of the present invention provides a composition for disinfecting or sanitizing an object from virus contamination. The composition contains a therapeutically effective amount of a compound that inhibits AN06, a pharmaceutically acceptable salt of the compound, a solvate of the compound, or a hydrate of the compound.

Other aspects and embodiments will be described below.

BRIEF DESCRIPTION OF DRAWINGS

FIG. 1 represents YFP fluorescence traces showing inhibition of AN06 activity of a compound in accordance with an embodiment of the present invention.

FIGS. 2A-2E represent results of measurements of AN06 Ca 2+ -activated ion channel and phospholipid scramblase activities. FIGS 2A-2C show the inhibitory effect of a compound in accordance with an embodiment of the present invention on AN06 ion channel activity.

The pipette solution contained 10 mM free Ca 2+ Voltage ramps spanning a range of -100 to +100 mV were delivered from a holding potential of -60 mV every 20 s (FIG. 2A). The current-voltage (I-V) relationships at indicated times (1-5) with increasing concentrations of the compound are shown in FIG. 2B. The numbers in parentheses represent the time points to measure I-V relationships. The inward currents in (A) represent AN06 tail currents. The IC50 of the compound on AN06 ion channel activity is depicted in FIG. 2C (n = 4 at each concentration). FIGS. 2D and 2E show the dose responses of a compound in accordance with an embodiment of the present invention on AN06 phospholipid scramblase inhibition. PS externalization was assessed via the fluorescence intensity of Lact-C2-GFP. Representative images are shown in FIG. 2D, and the quantification of IC50 is presented in FIG. 2E (n = 6 at each concentration). ** P < 0.01 : difference from lane 2. Data were analyzed using one-way analysis of variance, followed by Tukey’s multiple comparison test. Scale bar: 10 pm.

FIGS. 3 A-3 J represent test results showing that AN06 is responsible for phosphatidylserine externalization evoked by pseudotyped SARS-CoV-2 S virus (SARS2- PsV). HeLa cells expressing ACE2 (HeLa-ACE2) were incubated with a lentivirus-based SARS2-PsV (100 ng p24/ml) for 15 min, and then with Lact-C2-mCherry for 45 min.

Chelation of cytosolic Ca 2+ (BAPTA-AM, 10 pM, 1 h) inhibits SARS2-PsVinduced PS externalization. Representative images are shown in FIG. 3 A, and the quantification results of multiple experiments are summarized in FIG. 3B (n = 3, each from 3 to 5 fields per experimental condition). FIGS. 3C and 3D are the results of control experiments in ACE2- negative HeLa cells. PS externalization was induced by ionomycin (10 pM, 10 min). Representative images are shown in FIG. 3C, and the quantification results of multiple experiments are summarized in FIG. 3D (n = 3). FIGS. 3E and 3F are test results showing that silencing of AN06 (siRNAs against AN06, 100 nM, 24 h) inhibits the phosphatidylserine (PS) externalization evoked by SARS2-PsV. Cell nuclei were stained with DAPI. Representative images are shown in FIG. 3E, and the quantification results of multiple experiments are summarized in FIG. 3F (n = 3, each from 3 to 5 fields per experimental condition). FIGS. 3G and 3H are test results showing that a compound in accordance with an embodiment of the present invention inhibits SARS2-PsVinduced PS externalization. Representative images are shown in FIG. 3G, and the quantification results of multiple experiments are summarized in FIG. 3H (n = 3-4). FIGS. 31 and 3J are test results showing that authentic SARS-CoV-2 virus evokes Ca 2+ and AN06-dependent PS externalization. The HeLa-ACE2 cells were pretreated with compounds (10 pM) for 1 h and infected with SARS-CoV-2 (10 MOI) for 15 min. Representative images are shown in FIG. 31, and the quantification results of multiple experiments are summarized in FIG. 3 J (n = 3). Bar graph data are shown as the mean ± SEM. *P < 0.05, **P < 0.01. Data were analyzed using one-way analysis of variance, followed by Tukey’s multiple comparison test. Scale bar: 50 pm.

FIGS. 4A-4H represent mechanisms involved in the SARS-CoV-2 Spike-mediated PS- scrambling and membrane fusion. FIGS. 4 A and 4B are test results showing that the trypsin inhibitor Camostat (100 pM, 1 h) inhibits the PS externalization evoked by SARS2-PsV, but not by trypsinized SARS2-PsV (trypsin 10 pg/ml, 10 min, 37°C). A compound in accordance with an embodiment of the present invention inhibits the PS externalization evoked by trypsinized SARS2-PsV. Representative images are shown in FIG. 4A, and the quantification results of multiple experiments are summarized in FIG. 4B (n = 3). FIGS 4C and 4D are test results showing the S1/S2 protease cleavage site defective mutation (R682S/R685G) reduces the PS scrambling effect of SARS2-PsV. Representative images are shown in FIG. 4C, and the quantification results of multiple experiments are summarized in FIG. 4D (n = 3). FIGS. 4E and 4F are test results showing that incubation with SARS2-PsV for 30 min evokes the appearance of multinucleated cells. A compound in accordance with an embodiment of the present invention inhibits the multinucleated cell formation. Representative images are shown in FIG. 4E, and the quantification results of multiple experiments are summarized in FIG. 4F (n = 3). FIGS. 4G and 4H are time-lapse imaging of membrane fusion events (8 h, 20 frame/min) between the SARS-CoV-2 Spike-expressing CHO cells labelled with the membrane lipid probe Vybrant-DiO (CHO-Spike, green) and the cytosolic mCherry-labelled ACE2-expressing HEK 293T cells (HEK 293T-ACE2-TMPRSS2, red). The expression of Spike induces the cell-cell adhesion and membrane fusion with ACE2-expressing cells. A compound in accordance with an embodiment of the present invention abolishes the Spike- ACE2 interaction-mediated membrane fusion events, while it does not affect the cell-cell adhesion. Representative images are shown in FIG. 4G, and the quantification results of multiple experiments are summarized in FIG. 4H (n = 3). Bar graph data are shown as the mean ± SEM. *P < 0.05, **P < 0.01. Data were analyzed using one-way analysis of variance, followed by Tukey’s multiple comparison test. Scale bar: 50 pm.

FIGS. 5A-5L represent test results showing that the pseudotyped SARS-CoV-2 S virus (SARS2-PsV) evokes a sustained intracellular Ca 2+ elevation. [Ca 2+ ]i was measured in HEK 293T cells expressing ACE2 and TMPRSS2 with a ratiometric Ca 2+ probe (Fura-2-AM, 5 pM, 30 min). Data are presented as fluorescence emission ratios from the 340/380 nm excitation. Representative [Ca 2+ ]i images with a rainbow scale and the [Ca 2+ ]i values of the indicated regions (arrows) are shown. The 0-second time represents the point of SARS2-PsV or vehicle application. FIGS. 5A and 5B show test results for mock vehicle. FIGS. 5C and 5D show test results for SARS2-PsV alone. FIGS. 5E and 5F show test results for BAPTA pretreatment (BAPTA-AM, 3 pM) plus SARS2-PsV. FIGS. 5G and 5H show test results for Camostat pretreatment (10 pM) plus SARS2-PsV. FIGS. 51 and 5J show test results for a compound in accordance with an embodiment of the present invention pretreatment (10 pM) plus SARS2- PsV. FIG. 5K represents a summary of maximum A[Ca 2+ ]i is presented (three cells from each experimental replicate). Data are shown as mean ± SEM (n = 12-27). FIG. 5L represents a summary of the time taken to reach the [Ca 2+ ]i peak after SARS2-PsV application. FIG. 5M represents a summary of the slope of the rising phase of [Ca 2+ ]i transients (D [Ca 2+ ]i/At, nM/s). **P < 0.01: difference from lane 2. n.s.: not significant. Data were analyzed using oneway analysis of variance, followed by Tukey’s multiple comparison test.

FIGS. 6A-6F represent test results showing that the single-round infection of pseudotyped SARS-CoV-2 S virus (SARS2PsV) is Ca 2+ - and AN06-dependent. FIGS. 6A and 6B are test results of the single-round infection of SARS2-PsV performed using a lentivirus- based SARS2-PsV encoding GFP. The cell viability was assessed using the mCherry fluorescence of HEK 293T-ACE2-TMPRSS2 cells. Silencing of AN06 (siRNAs against AN06, 100 nM, 24 h) diminished the single-round infection of SARS2-PsV (n = 3). **P <

0.01 : difference from lane 2. FIGS. 6C and 6D are test results showing that chelation of intracellular Ca 2+ (BAPTA-AM, 10 mM) reduced the single-round infection of SARS2-PsV (n = 3). **P < 0.01 : difference from lane 2. FIGS. 6E and 6F are test results showing that a compound in accordance with an embodiment of the present invention dose-dependently inhibited the single-round infection of SARS2-PsV (n = 4-5). The compound was administered during the transduction period (1-h pretreatment, 2-h incubation at 4°C, and 4-h incubation at 37°C). Then, cells were incubated without the compound throughout the 42-h post-transduction period. Data are shown as mean ± SEM. Data were analyzed using one-way analysis of variance followed by Tukey’s multiple comparison test. IC50: the half-maximal inhibitory concentration. Scale bar: 100 pm.

FIGS. 7A-7G represent test results showing that a compound in accordance with an embodiment of the present invention inhibits the viral replication of SARS-CoV-2 in Calu-3, Vero, and human nasal epithelial (HNE) cells. FIGS. 7A and 7B are results of assaying viral replication of authentic SARS-CoV-2 (0.001 MOI) in Calu-3 cells. The qPCR results of virion mRNAs with the indicated concentrations of the compound in accordance with an embodiment of the present invention are shown in FIG. 7A (*P < 0.05, **P < 0.01 : difference from 0 pM, one-way analysis of variance followed by Tukey’s multiple comparison test), and the IC50 values of the compound in accordance with an embodiment of the present invention at 48 h post infection are presented in FIG. 7B (n = 3 at each concentration). FIGS. 7C-7E are results of assaying viral replication of SARS-CoV-2 (0.001 MOI) in Vero cells. In the light microscopic analyses, the compound in accordance with an embodiment of the present invention dose-dependently reduced the virus-induced cytolysis (FIG. 7C, 48 h post infection, five independent experiments showed similar results, arrows indicate cytolytic cells). The qPCR results of virion mRNAs with the indicated concentrations of A6-001 are shown in FIG. 7D (**P < 0.01: difference from 0 mM, one-way analysis of variance followed by Tukey’s multiple comparison test), and the IC50 values of the compound in accordance with an embodiment of the present invention at 48-h post infection are presented in FIG. 7E (n = 5 at each concentration). Scale bar: 200 pm. FIGS. 7F and 7G are results of assaying viral replication of SARS-CoV-2 (1 MOI) in HNE cells. The passage #2 HNE cells were cultured under air-liquid interface conditions and apically infected with SARS-CoV-2. The compound in accordance with an embodiment of the present invention (10 mM) was administered to the basolateral compartments. Representative plaque assay results at 72-h post infection are shown in FIG. 7F, and the summary of multiple experiments are presented in FIG. 7G (n = 3, *P < 0.05, twotailed Student’s t-test). Bar graph data are shown as the mean ± SEM.

FIGS. 8A-8B represent expression of AN06 in FRT cells and effects of AN06 inhibitors on ANOl (FIG. 8 A) and AN02 (FIG. 8B).

FIGS. 9A-9C represent effects of AN06 inhibitors on cytosolic Ca 2+ levels, CFTR CT currents, and cell morphology. Intracellular calcium ([Ca 2+ ]i) was monitored using the Fluo-4 NW calcium indicator. The FRT cells were treated with the indicated concentrations of compounds for 10 min and then ionomycin (10 mM) was applied to evoke [Ca 2+ ]i elevation. Representative traces of intracellular calcium responses pretreated with the compound in accordance with an embodiment of the present invention are shown in FIG. 9 A. FIGS. 9B and 9C show effects of the compound in accordance with an embodiment of the present invention on CFTR CE channel activity. The apical membrane short-circuit currents (Isc) were measured in FRT-CFTR cells cultured on a permeable support. The basolateral membrane was permeabilized with amphotericin B (250 pg/ml). CFTR was stimulated using forskolin (20 pM) and blocked by CFTRinh-172 (20 mM). Representative Isc measurements are shown in FIG. 9B, and a summary of the current inhibition rates by the compound in accordance with an embodiment of the present invention (30 mM) and CFTRinh-172 (20 mM) is provided in FIG. 9C (mean ± SEM, n = 4, **P < 0.01 using a two-tailed Student’s t-test).

FIGS. 10A-10E represent additional test results supporting the test results represented by FIGS. 3 and 4. FIG. 10A represents mRNA quantification of AN06/TMEM16F using qPCR. The AN06 expression levels in HEK-293T, HeLa, and HNE cells are shown. FIG. 10B represents results of analyzing the silencing of AN06 mRNA via treatment with siRNAs against AN06 in HeLa- ACE2 cells and HEK 293 T - ACE2-TMPRS S2 cells using qPCR. Cells were treated with scrambled or AN06 siRNAs (100 nM in each case) for 24 h. FIGS. IOC and 10D represent effects of Camostat on ionomycin (10 pM, 10 min)-induced PS scrambling. The trypsin inhibitor Camostat (100 mM) was applied 1-h prior to ionomycin application. Representative images are shown in FIG. IOC, and the summarized results of multiple experiments are depicted in FIG. 10D (n = 3). Scale bar: 50 pm. FIG. 10E represents immunoblot analysis of the SARS-CoV-2 Spike glycoprotein. The cell lysates of wild-type and R682S/R685G SARS-PsV infected HEK 293T cells were blotted with primary antibodies against the S2 domain of Spike and aldolase A. The R682S/R685G mutant Spike proteins do not exhibit a protease cleaved form. Bar graph data are shown as the mean ± SEM. Data were analyzed using a two-tailed Student’s t-test (B) or one-way analysis of variance followed by Tukey’s multiple comparison test (D). **P < 0.01, ns: not significant.

FIGS. 11 A-l IF represent test results showing the compound in accordance with an embodiment of the present invention does not affect the ATP -induced Ca 2+ response. FIGS. Purinergic agonist (ATP, 100 pM)-induced Ca 2+ response in HEK-293T-ACE2-TMPRSS2 cells was monitored in the absence and presence of the compound in accordance with an embodiment of the present invention (10 pM). Representative traces (two each) are shown in FIGS. 11 A and 1 IB, respectively. A summary of A[Ca 2+ ]i increase is presented in FIG. 11C (n = 15). ATP-induced Ca 2+ response (100 pM) in Calu-3 cells was monitored in the absence and presence of the compound in accordance with an embodiment of the present invention (10 pM). Representative traces are shown in FIG. 10D and 10E, respectively. A summary of A[Ca 2+ ]i increase is presented in FIG. 1 IF (n = 12). Data were analyzed using a two-tailed Student’s t-test. ns: not significant.

FIGS. 12A-12C represent effects of the compound in accordance with an embodiment of the present invention on the viral replication of wild-type SARS-CoV-2 in Vero cells.

FIGS. 12A and 12B represent viral replication of SARS-CoV-2 in Vero cells with an infection dose of 0.01 MOI (ten times higher than that of FIGS. 7C-7E). In the light microscopic analysis, the compound in accordance with an embodiment of the present invention dose- dependently reduced the virus-induced cytolysis (FIG. 12A, 48 h post-infection, two independent experiments showed similar results. Arrows indicate cytolytic cells.). The IC 50 values of the compound in accordance with an embodiment of the present invention at 24 h and 48 h post-infection are presented in FIG. 12B (n = 2 at each concentration). Scale bar: 100 pm. Vero cells were infected with SARS-CoV-2 (0.01 MOI), and cellular viral RNAs were quantified using qPCR at 4 h post-infection (FIG. 12C). The compound in accordance with an embodiment of the present invention (10 pM) was treated only during the infection period alone (for 2-h pretreatment and 1-h post-infection, DDOΐ = 2.84 ± 0.16, n = 3) or during both the infection and the 4 h post-infection periods (AACt = 5.58 ± 0.15, n = 3). Ct: threshold cycle, GAPDH: glyceraldehyde 3-phosphate dehydrogenase. Bar graph data are shown as the mean ± SEM.

FIGS. 13A-13D represent analysis results of cell viability of the compound in accordance with an embodiment of the present invention. FRT cells (FIG. 13 A), Calu-3 cells (FIG. 13B), and Vero cells (FIG. 13C) were treated with the indicated concentrations of the compound in accordance with an embodiment of the present invention, abamectin, and ivermectin for 48 h. Cisplatin (50 mM) was used as a positive control. Cell viability was determined using the MTS assay (mean ± SEM, n = 6). The CC 50 (the half-maximal cytotoxic concentration) values are summarized in FIG. 13D.

DETAILED DESCRIPTION

1. Methods

An aspect of the present invention provides a method for treating or preventing diseases, disorders, or conditions associated with virus infection. The method comprises administering to a subject in need a composition comprising a therapeutically effective amount of a compound that can inhibit AN06 or, a pharmaceutically acceptable salt thereof, a diastereomer thereof, an enantiomer thereof, a racemate thereof, a solvate thereof, a hydrate thereof, a prodrug thereof, a crystalline thereof, or a combination thereof.

Still another aspect of the present invention provides a method for disinfecting or sanitizing an object from virus contamination. The method comprises contacting with or applying to an object a composition containing a therapeutically effective amount of a compound that inhibits AN06, a pharmaceutically acceptable salt thereof, a diastereomer thereof, an enantiomer thereof, a racemate thereof, a solvate thereof, a hydrate thereof, a prodrug thereof, a crystalline thereof, or a combination thereof.

The compound inhibits AN06 phospholipid scramblase activity and reduce phosphatidyl serine (PS) externalization. By inhibiting PS externalization, the compound or salt can inhibit the entry of viruses into host cells. In addition, the compound or salt inhibits viral replication. Thus, the compound or salt can treat or prevent diseases, disorders, or conditions associated with virus infection and can disinfect or sanitize an object from virus contamination. The compound or salt has AN06 inhibition activity, as illustrated in Examples 3 and 4. The compound or salt has anti-viral replication activity and anti-virus activity, as illustrated in Examples 6 and 7. In some embodiments, the virus of the present invention is an RNA virus. In some embodiments, the RNA virus may be an enveloped positive-strand RNA virus. In some embodiments, the enveloped positive-strand RNA virus may be a Coronaviridae. The Coronaviridae includes letovirinae and orthcoromavirinae (known as coronavirus) subfamily.

In some embodiments, the virus of the present invention may comprise all virus in the orthcoronavirinae (coronavirus). The orthcoronavirinae (coronavirus) may be selected from the group consisting of alphacoronavirus (Group 1 CoV), betacoronavirus (Group 2 CoV), gammacoronavirus (Group 3 CoV) and deltacoronavirus (Group 4 CoV) genus. In certain embodiments, the virus may be a Betacoronavirus. Specifically, the genus Betacoronavirus (Group 2 CoV) comprises five subgenera or lineages (A, B, C, and D): Embecovirus (lineage A), Sarbecovirus (lineage B), Merbecovirus (lineage C), Nobecovirus (lineage D) and Hibecovirus. The Embecovirus comprises Betacoronavirus 1 species (ex. Bovine coronavirus and Human coronavirus OC43), China Rattus coronavirus HKU24 species, Human coronavirus HKU1 species, Murine coronavirus species (ex. Mouse hepatitis virus) species, and My odes coronavirus 2JL14 species. The Sarbecovirus subgenera (lineage B) comprises severe acute respiratory syndrome-related coronavirus (SARSr-CoV). The severe acute respiratory syndrome-related coronavirus comprises severe acute respiratory syndrome coronavirus species (also known as SARS-CoV and SARS-CoV-1), severe acute respiratory syndrome coronavirus 2 species (SARS-CoV-2), Bat SARS-like coronavirus WIV1 species (Bat SL- CoV-WIVl), and Bat coronavirus RaTG13 species. The Merbecovirus subgenera (lineage C) comprises Hedgehog coronavirus 1 species, Middle East respiratory syndrome-related coronavirus species (MERS-CoV), Pipistrellus bat coronavirus HKU5 species, and Tylonycteris bat coronavirus HKU4 species. The Nobecovirus subgenera (lineage D) comprises Eidolon bat coronavirus C704 species, Rousettus bat coronavirus GCCDC1 species, and Rousettus bat coronavirus HKU9 species. The Hibecovirus subgenera comprises Bat Hp- betacoronavirus Zhejiang2013 species.

The diseases, disorders, or conditions associated with virus infection comprises cold, severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), COVID-19; and disorders or conditions thereof. The cold, also known as common cold, is a viral infectious disease of the upper respiratory tract that primarily affects the respiratory mucosa of the nose, throat, sinuses, and larynx. Signs and symptoms of the cold include coughing, sore throat, runny nose, sneezing, headache, and fever. The severe acute respiratory syndrome (SARS) is a viral respiratory disease of zoonotic origin caused by severe acute respiratory syndrome coronavirus (SARS-CoV or SARS-CoV-1). In December 2019, another strain of SARS-CoV was identified as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This new strain causes coronavirus disease 2019 (COVID-19), a disease which brought about the COVID-19 pandemic. The middle East respiratory syndrome (MERS) is a viral respiratory infection caused by Middle East respiratory syndrome-related coronavirus (MERS-CoV). Typical symptoms include fever, cough, diarrhea, and shortness of breath. The coronavirus disease 2019 (COVID-19) is a contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Symptoms of COVID-19 are variable, but often include fever, cough, headache, fatigue, breathing difficulties, loss of smell, and loss of taste.

As used herein, the term “treat,” “treating” or “treatment” refers to methods of alleviating, abating or ameliorating a disease or condition symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, inhibiting the disease or condition, arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactically and/or therapeutically.

As used herein, the term “disinfect,” “disinfecting” or “disinfection” refers to methods of inactivating or killing pathogenic microorganisms including virus.

As used herein, the term “sanitize,” “sanitizing” or “sanitization” refers to methods of removing or decreasing pathogenic microorganisms including virus from a surface of non living or living object.

As used herein, the term “subject” or “patient” encompasses mammals and non mammals. Examples of mammals include, but are not limited to, humans, chimpanzees, apes monkeys, cattle, horses, sheep, goats, swine; rabbits, dogs, cats, rats, mice, guinea pigs, and the like. Examples of non-mammals include, but are not limited to, birds, fishes and the like.

As used herein, the term “administration” or “administering” of the subject compound refers to providing a compound of the invention and/or a prodrug thereof to a subject in need of treatment.

As used herein, the term “contacting with” or “applying to” of an object refers to methods of allowing the compositions of the invention to be in contact with or be applied to an object by, for example, wiping, dipping, immersing, or spraying. As used herein, the term “effective amount” or “therapeutically effective amount” refer to a sufficient amount of an active ingredient(s) described herein being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result can be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms. An appropriate “effective” amount in any individual case may be determined using techniques, such as a dose escalation study. By way of example only, a therapeutically effective amount of a compound of the invention may be in the range of e.g ., about 0.01 mg/kg/day to about 1000 mg/kg/day, from about 0.1 mg/kg/day to about 500 mg/kg/day, from about 0.1 mg (x2)/kg/day to about 500 mg (x2)/kg/day.

In addition, such compounds and compositions may be administered singly or in combination with one or more additional therapeutic agents. The methods of administration of such compounds and compositions may include, but are not limited to, intravenous administration, inhalation, oral administration, rectal administration, parenteral, intravitreal administration, subcutaneous administration, intramuscular administration, intranasal administration, dermal administration, topical administration, ophthalmic administration, buccal administration, tracheal administration, bronchial administration, sublingual administration or optic administration. Compounds provided herein may be administered by way of known pharmaceutical formulations, including tablets, capsules or elixirs for oral administration, suppositories for rectal administration, sterile solutions or suspensions for parenteral or intramuscular administration, lotions, gels, ointments or creams for topical administration, and the like. In some embodiments, such pharmaceutical compositions are formulated as tablets, pills, capsules, a liquid, an inhalant, a nasal spray solution, a suppository, a solution, a gel, an emulsion, an ointment, eye drops, or ear drops.

The therapeutically effective amount may vary depending on, among others, the disease indicated, the severity of the disease, the age and relative health of the subject, the potency of the compound administered, the mode of administration and the treatment desired. The required dosage will also vary depending on the mode of administration, the particular condition to be treated and the effect desired.

The compound may be represented by Formula (I).

Ring A and ring B each may be independently a monocyclic or polycyclic aliphatic ring or a monocyclic or polycyclic aromatic ring, wherein the aliphatic ring and the aromatic ring each optionally and independently may contain at least one heteroatom selected from the group consisting of N, NO, NO2, S, SO, SO2, and O.

Ri R 2 , and R 3 each may be independently hydrogen, halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, or aryl aliphatic.

Li and L2 each may be independently aliphatic, cycloaliphatic, hetero cycloaliphatic, or alkoxy. m and n each are independently 0 or 1.

The ring A, the ring B, Ri , R 2 , R 3 , Li, and L 2 each may be optionally and independently substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, ester, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic.

In some embodiments, two or more of the polycyclic rings may be fused or linked with each other.

In some embodiments, the monocyclic or polycyclic aliphatic ring and the monocyclic or polycyclic aromatic ring of the ring A and the ring B each may be independently a 4- membered, 5-membered, 6-membered, 7-membered, 8-membered, 9-membered, 10- membered, 11-membered, or 12-membered ring.

In some embodiments, the monocyclic aliphatic ring and the monocyclic aromatic ring of the ring A may be a 5-membered ring or a 6-membered ring, and the monocyclic aliphatic ring and the monocyclic aromatic ring of the ring B may be a 5-membered ring or a 6- membered ring. In some embodiments, the monocyclic aliphatic ring and the monocyclic aromatic ring of the ring A may be 5-membered ring or a 6-membered ring, and the monocyclic aliphatic ring and the monocyclic aromatic ring of the ring B may be a 6-membered ring.

In some embodiments, -(Li) m -Ri may be connected to the ring A at the para, meta or ortho position. In some embodiments, -(Li) m -Ri may be connected to the ring A at the para position.

In some embodiments, the ring A may be a monocyclic or polycyclic aliphatic ring which optionally contains at least one heteroatom selected from the group consisting of N, NO, NO2, S, SO, S0 2 , and O.

In some embodiments, the ring A may be a monocyclic or polycyclic aromatic ring which optionally contains at least one heteroatom selected from the group consisting of N, NO, NO2, S, SO, S0 2 , and O.

In some embodiments, the ring A may be phenyl, pyridinyl, diazinyl, pyrimidinyl, triaziny, piperidinyl, oxadiazoline, cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.

In some embodiments, the ring A may be , in which X ai , X a2 , X a 3, and X a4 each are independently CH, N, NH, NO, or N0 2 . In certain embodiments, any one of X ai , X a2 , X a3 , and X a4 is N, NH, NO, or NO2, and the others are CH. In certain embodiments, two of Xai, X a 2, X a 3, and X a4 are N, NH, NO, or NO2, and the others are CH. In certain embodiments, three of X ai , X a2 , X a 3, and X a4 are N, NH, NO, or NO2, and the other one is CH. In certain embodiments, X ai and X a2 are N, and X a3 and X a4 are CH. In certain embodiments, X ai and X a3 are N, and X a2 and X a4 are CH. In certain embodiments, X ai and X a4 are N, and X a2 and X a3 are CH. In certain embodiments, X a2 and X a3 are N, and X ai and X a4 are CH. In certain embodiments, X a2 and X a4 are N, and X ai and X a3 are CH. In certain embodiments, X a3 and X a4 are N, and X ai and X a2 are CH. In certain embodiments, X ai , X a2 , and X a3 are N, and X a4 is

CH.

In some embodiments, the ring A may be , in which Y ai , Y a 2, and Y a3 each are independently CH, N, NH, NO, NO2, S, SH or O. In certain embodiments, any one of Y ai , Y a 2, and Y a3 is N, NH, NO, NO2, S, SH or O, and the others are CH. In certain embodiments, two of Y ai , Y a 2, and Y a3 are N, NH, NO, NO2, S, SH or O, and the other is CH. In certain embodiments, Y ai , and Y a2 are N, NO, NO 2 , or NH, and Y a3 is S, SH or O. In certain embodiments, Y a2 , and Y a3 are N, NO, NO 2 , or NH, and Y ai is S, SH or O.

In some embodiments, the ring B may be a monocyclic or polycyclic aromatic ring which optionally contains at least one heteroatom selected from the group consisting of N, O, and S.

In some embodiments, the ring B may be a monocyclic or polycyclic aliphatic ring which optionally contains at least one heteroatom selected from the group consisting of N, O, and S.

In certain embodiments, the ring B may be phenyl, pyridinyl, diazinyl, cyclopentadienyl, cyclopentyl, cyclohexyl, adamantane, or bicyclo[2.2.1]heptane.

In some embodiments, the ring B may be in which X bi , X b2 , X b3 , and X b4 each are independently CH, N, or NH. In certain embodiments, any one of X bi , X b2 , X b3 , and X b4 is N, NH, NO, or NO 2 , and the others are CH. In certain embodiments, two of X bi , X b2 , X b3 , and X b4 are N, NH, NO, or NO 2 , and the others are CH. In certain embodiments, three of X bi , X b2 , X b3 , and X b4 are N, NH, NO, or NO2, and the other one is CH. In certain embodiments, X bi and X b2 are N, and X b3 and X b4 are CH. In certain embodiments, X bi and X b3 are N, and X b2 and X b4 are CH. In certain embodiments, X bi and X b4 are N, and X b2 and X b3 are CH. In certain embodiments, X b2 and X b3 are N, and X bi and X b4 are CH. In certain embodiments, X b2 and X b4 are N, and X bi and X b3 are CH. In certain embodiments, X b3 and X b4 are N, and Xbi and Xb2 are CH. In certain embodiments, X bi , X b2 , and X b3 are N, and X b4 is CH.

In some embodiments, Li and L 2 each may be independently C 1 -C 10 aliphatic, C 3 -C 10 cycloaliphatic, or C 3 -C 10 hetero cycloaliphatic. In certain embodiments, Li and L 2 each may be independently C 1 -C 10 aliphatic. In certain embodiments, Li and L 2 each may be independently C 1 -C 10 alkyl or cyclopropyl. In certain embodiments, Li and L 2 each may be optionally and independently substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, hydroxyl, amine, amide, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic. In certain embodiments, Li and L 2 each may be optionally and independently substituted with at least one substituent selected from the group consisting of CN, C 1-5 alkyl, and C 3-6 cycloalkyl. In some embodiments, R2 may be hydrogen, C1-5 alkyl or C3-6 cycloalkyl. In certain embodiments, R2 may be hydrogen or C1-3 alkyl.

In some embodiments, Ri and R3 each may be optionally and independently hydrogen, benzyl, amide, amine, thioalkyl, alkoxy, CN, COOH, Ci-Cn aliphatic, C3-C11 cycloaliphatic, C3-C11 hetero cycloaliphatic, C3-C11 aromatic ring, or C3-C11 hetero aromatic ring.

In some other embodiments, Ri and R3 each may be optionally and independently 3- membered cycloaliphatic; 4-membered cycloaliphatic; 4-membered hetero cycloaliphatic; 5- membered cycloaliphatic; 5-membered hetero cycloaliphatic; 6-membered cycloaliphatic; 6- membered hetero cycloaliphatic; 5-membered aromatic ring; 5-membered hetero aromatic ring; 6-membered aromatic ring; 6-membered hetero aromatic ring; 7-membered cycloaliphatic; 7- membered hetero bicyclic aliphatic; 10-membered tricyclic aliphatic; 6-membered aromatic ring fused or linked with 5-membered cycloaliphatic, 5-membered hetero cycloaliphatic, 5- membered aromatic ring, or 5-membered aromatic ring; 6-membered aromatic ring fused or linked with 6-membered cycloaliphatic, 6-membered hetero cycloaliphatic, 6-membered aromatic ring, or 6-membered hetero aromatic ring; 6-membered cycloaliphatic fused or linked with 6-membered cycloaliphatic or 6-membered hetero cycloaliphatic; or 3-membered cycloaliphatic fused or linked with 5-membered aromatic ring, 5-membered hetero aromatic ring, 5-membered cycloaliphatic, 5-membered hetero cycloaliphatic, 6-membered aromatic ring, 6-membered hetero aromatic ring, 6-membered cycloaliphatic, or 6-membered hetero cycloaliphatic, wherein heteroatom is selected from the group consisting of N, O, and S.

In some other embodiments, Ri , and R3 each may be optionally and independently N(CH ) 2 , N(C 2 H 5 ) 2 , N(C 2 H 5 )(benzyl), or N(C 3 H 7 )(benzyl).

In some other embodiments, Ri , and R3 each may be independently hydrogen, Ci-10 alkyl, C2-5 alkenyl, C2-5 alkynyl, C3-11 cycloalkyl, C3-11 hetero-cycloalkyl, C3-11 cycloalkenyl, C3- 11 hetero-cycloalkenyl, C3-11 cycloalkynyl, C3-11 hetero-cycloalkynyl, C5-11 aryl, C5-11 hetero aryl, or CN.

In some other embodiments, Ri may be hydrogen; Ci-10 alkyl; benzyl; alkoxy; CN; COOH; mono or bi aromatic ring which optionally contains at least one heteroatom selected from the group consisting of N, O, and S; mono or bi cycloaliphatic which optionally contains at least one heteroatom selected from the group consisting of N, O, and S; aryl which optionally contains at least one hetero atom selected from the group consisting of N, O, and S; an aromatic ring fused to a non-aromatic ring which optionally contains at least one heteroatom selected from the group consisting of N, O, and S; or an aromatic ring fused to an aromatic ring which optionally contains at least one heteroatom selected from the group consisting of N, O, and S. Ri may be substituted or unsubstituted.

In certain embodiments, Ri may be Ci-4 alkyl, benzyl, phenyl, pyridinyl, diazinyl (such as pyrimidinyl, pyrazinyl, and pyridazinyl), triazinyl, piperidinyl, furanyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, thiophenyl or oxygen-containing fused heterocycle which is optionally substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, alkoxy, carboxyl, C 1-5 alkyl ester and C 1-5 alkyl. In certain embodiments, the substituent is selected from the group consisting of 0(CH 3 ), CH 3 , isopropyl, F, Cl, Br, CF3, NO2, NH 2 , OCHF2, CHF 2 , OCF3, SCH3, COOC(CH )3, COOCH 2 CH , OCH3, OCFFCFF, OCH 2 CH 2 CH 3 , N(C 2 H 5 ) 2 , 6-membered hetero cycloaliphatic, dimethyl amine, diethyl amine, and phenyl.

In some embodiments, one of the ring A and Ri may be or comprise a hetero aromatic ring which contains at least one N as the heteroatom.

In some other embodiments, both of the ring A and Ri may be or comprise a hetero aromatic ring which contains at least one N as the heteroatom.

In some embodiments, R 3 may be hydrogen, halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, aryl aliphatic or fused ring. In some other embodiments, R 3 may be hydrogen, Ci- 10 alkyl, alkyl amine, mono or bi aromatic ring, mono or bi hetero aromatic ring, mono or bi cycloaliphatic, mono or bi hetero cycloaliphatic, aryl, heteroaryl, aromatic ring fused to a non-aromatic ring which optionally contains at least one heteroatom, or aromatic ring fused to aromatic ring which optionally contains at least one heteroatom. Examples of the heteroatoms include N, O, and S. In some embodiments, R 3 may be bicycle, cycloaliphatic ring, aryl, or hetero aryl. In some embodiments, R 3 may be Ci- 10 alkyl, alkyl amine, benzyl, COOH, phenyl, pyridinyl, pyrimidinyl, piperidinyl, furanyl, thiophenyl, pyrrolyl, thiazolyl, C 3-7 cycloaliphatic, or oxygen-containing fused heterocycle. In some embodiments, R 3 may be optionally substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic. R 3 may be substituted or unsubstituted. In some embodiments, R I, R-2, and R3 each may be optionally and independently substituted with one or more groups selected from the group consisting of halogen, halogen derivatives ( e.g ., F, Br, Cl, I, OCHF 2 , CF 3 , CHF 2 , or OCF 3 ), alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, hetero cycloalkyl, hetero cycloalkenyl, hetero cycloalkynyl, alkoxy, aryl, aryloxy, diaryl, arylalkyl, arylalkyloxy, cycloalkylalkyl, cycloalkylalkyloxy, amino, hydroxy, hydroxyalkyl, acyl, heteroaryl, heteroaryloxy, heteroarylalkyl, heteroarylalkoxy, aryloxyalkyl, alkylthio, arylalkylthio, aryloxyaryl, alkylamido, alkanoylamino, arylcarbonylamino, nitro, cyano, thiol, haloalkyl, trihaloalkyl, alkyl ester, and alkylthio.

In some embodiments, R I, R2, and R3 each may be optionally and independently substituted with one or more groups selected from =0, -OR x , -SR X , =S, -NR x R y , -N(alkyl)3, - NR X S0 2 , -NR x S0 2 R y , -S0 2 R x -, -S0 2 NR x R y , -S0 2 NR x C0R y , -SO3H, -PO(OH) 2 , -COR x , - COORx, COOC(alkyl) , -CONR x R y , -CO(Ci-C 4 alkyl)NR x R y , -C0NR x (S0 2 )R y , -C0 2 (Ci-C 4 alkyl)NR x R y , -NR x COR y , -NR x C0 2 R y , -NR X (C I -C 4 alkyl)C0 2 R y , =N-OH, =N-0-alkyl. R x and R y each may be independently selected from hydrogen, alkyl, alkenyl, C3-C7 cycloalkyl, C 5 -C 11 aryl, benzyl, phenylethyl, naphthyl, a 3- to 7-membered heterocycloalkyl, and a 5- to 6- membered heteroaryl.

In some embodiments, Ri, and R 3 each may be optionally and independently substituted by at least one substituent selected from the group consisting of 0(CH3), CFF, CH2CH3 , isopropyl, F, Cl, Br, CF 3 , OCHF 2 , CHF 2 , OCF3, SCH3, COOH, COOC(CH )3, COOCH2CH3, COOCH3, OCH2CH3, OCH2CH2CH3, N(C 2 H 5 )2, NHCH3, NO2, NH 2 , CN, dimethyl amine, diethyl amine, phenyl, and 6-membered hetero cycloaliphatic.

In some embodiments, if Ri is a substituted cyclic compound, the substituent may be bound at the ortho, meta and/or para position of Ri. In some embodiments, the substituent may be bound at the meta, and/or para position of Ri.

In some embodiments, L2 may be aliphatic, cycloaliphatic, hetero cycloaliphatic, or alkoxy. In some embodiments, L 2 may be C 1-5 alkyl or C 1-5 cycloaliphatic. In still some other embodiments, L 2 may be C 1-3 alkyl or C 1-3 cycloaliphatic.

In some embodiments, the group may be one of the following groups:

In some embodiments, the group may be one of the following groups:

In some embodiments, the compound represented by Formula (I) may be selected from the following compounds.

In some other embodiments, compounds represented by Formula (II) are provided.

Ring A, Ring B, R 1 R 3 , Li, L2, m, and n are the same as defined with regard to Formula

(I).

In some other embodiments, compounds represented by Formula (III) are provided.

Ri and R 3 each may be independently hydrogen, halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, or aryl aliphatic. A’s and X’s each may be independently CH, N, NO, or NH. L 2 may be independently aliphatic, or cycloaliphatic. N may be 0 or 1. A’s, R I, R 3 , and L 2 each may be optionally and independently substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, ester, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic.

Some embodiments of Ri , R 3 , L 2 , and n are the same as defined with regard to Formula

(I).

In some other embodiments, compounds represented by Formula (IV) are provided.

Ri and R 3 each may be independently hydrogen, halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, or aryl aliphatic. A’s and X’s each may be independently CH, N, NO, or NH. L 2 may be independently aliphatic, or cycloaliphatic. N may be 0 or 1. A’s, R I, R 3 , and L 2 may be optionally and independently substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, ester, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic.

Some embodiments of Ri , R 3 , L 2 , and n are the same as defined with regard to Formula

(I)·

In some other embodiments, compounds represented by Formula (V) are provided.

Ri and R 3 each may be independently hydrogen, halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, or aryl aliphatic. A’s and X’s each may be independently CH, N, or NH. L2 may be independently aliphatic, or cycloaliphatic. N may be 0 or 1. A’s, R 1 ,R-3, and L2 may be optionally and independently substituted with at least one substituent selected from the group consisting of halogen, halogen derivatives, CN, alkoxy, carboxyl, carbonyl, ester, hydroxyl, amine, amide, nitro, phosphate, thioalkyl, sulfhydryl, oxo, aliphatic, cycloaliphatic, hetero cycloaliphatic, aromatic, hetero aromatic, and aryl aliphatic.

Some embodiments of Ri , R3, L2, and n are the same as defined with regard to Formula

(I)·

Non-limiting examples of the compounds of embodiments of the present invention are listed in Table 1 below.

The compounds described herein include all stereoisomers, geometric isomers, tautomers, isotopes, and prodrug of the structures depicted. The compounds described herein can be present in various forms including crystalline, powder and amorphous forms of those compounds, pharmaceutically acceptable salts, including, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, and amorphous forms of the compounds, as well as mixtures thereof.

As used herein, the term “pharmaceutically acceptable” refers a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compounds described herein. Such materials are administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.

As used herein, the term “pharmaceutically acceptable salt” refers to a formulation 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 compounds described herein.

Pharmaceutically acceptable salt forms may include pharmaceutically acceptable acidic/anionic or basic/cationic salts (UK Journal of Pharmaceutical and Biosciences Vol. 2(4), 01-04, 2014, which is incorporated herein by reference). Pharmaceutically acceptable acidic/anionic salts include acetate, benzenesulfonate, benzoate, bicarbonate, bitartrate, bromide, calcium edetate, camsylate, carbonate, chloride, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, glyceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, malate, maleate, malonate, mandelate, mesylate, methyl sulfate, mucate, napsylate, nitrate, pamoate, pantothenate, phosphate/diphosphate, polygalacturonate, salicylate, stearate, subacetate, succinate, sulfate, hydrogensulfate, tannate, tartrate, teoclate, tosylate, and triethiodide salts. Pharmaceutically acceptable basic/cationic salts include, the sodium, potassium, calcium, magnesium, diethanolamine, A -m ethyl -D-gl ucami ne, L.-lysine, L.-arginine, ammonium, ethanolamine, piperazine, and triethanolamine salts.

A pharmaceutically acceptable acid addition salt of a compound of the invention may be prepared by methods known in the art and may be formed by reaction of the free base form of the compound with a suitable inorganic or organic acid including, but not limited to, hydrobromic, hydrochloric, sulfuric, nitric, phosphoric, succinic, maleic, formic, acetic, propionic, fumaric, citric, tartaric, lactic, benzoic, salicylic, glutamic, aspartic, p- toluenesulfonic, benzenesulfonic, methanesulfonic, ethanesulfonic, naphthalenesulfonic such as 2-naphthalenesulfonic, and hexanoic acid. A pharmaceutically acceptable acid addition salt can comprise or be, for example, a hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, phosphate, succinate, maleate, formarate, acetate, propionate, fumarate, citrate, tartrate, lactate, benzoate, carbonate, benzathine, chloroprocaine, choline, histidine, meglumine, meglumine, procaine, triethylamine, besylate, decanoate, ethylenediamine, salicylate, glutamate, aspartate, /Mol uene sulfonate, benzenesulfonate, methanesulfonate, ethanesulfonate, naphthalenesulfonate e.g ., 2-naphthalenesulfonate), and hexanoate salt.

A pharmaceutically acceptable base addition salt of a compound of the invention may also be prepared by methods known in the art and may be formed by reaction of the free base form of the compound with a suitable inorganic or organic base including, but not limited to, hydroxide or other salt of sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, tromethamine, glycolate, hydrabamine, methylbromide, methylnitrate, octanoate, oleate, and the like.

A free acid or free base form of a compound of the invention may be prepared by methods known in the art (e.g., for further details see L.D. Bigley, S.M. Berg, D.C.

Monkhouse, in “ Encyclopedia of Pharmaceutical Technology" . Eds, J. Swarbrick and J.C. Boylam, Vol 13, Marcel Dekker, Inc., 1995, pp.453-499, which is incorporated herein by reference). For example, a compound of the invention in an acid addition salt form may be converted to the corresponding free base form by treating with a suitable base (e.g, ammonium hydroxide solution, sodium hydroxide, and the like). A compound of the invention in a base addition salt form may be converted to the corresponding free acid by treating with a suitable acid ( e.g ., hydrochloric acid, etc.).

Aspects of this disclosure include prodrug forms of any of the compounds described herein. Any convenient prodrug forms of the subject compounds can be prepared, for example, according to the strategies and methods described by Rautio et al. (“Prodrugs: design and clinical applications”, Nature Reviews Drug Discovery 7, 255-270 (February 2008)).

Prodrug derivatives of the compounds of the invention may be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et al, Bioorg. Med. Chem. Letters, 1994, 4, 1985, which is incorporated herein by reference). Protected derivatives of the compounds of the invention may be prepared by means known to those of ordinary skill in the art. A detailed description of techniques applicable to the creation of protecting groups and their removal can be found in T. W. Greene, “Protecting Groups in Organic Chemistry,” 3 rd edition, John Wiley and Sons, Inc., 1999 and “Design of Prodrugs”, ed. 11. Bundgaard, Elsevier, 1985, which are incorporated herein by reference.

The compounds of the present disclosure may be prepared as stereoisomers. Where the compounds have at least one chiral center, they may exist as enantiomers. Where the compounds possess two or more chiral centers, they may exist as diastereomers. The compounds of the invention may be prepared as racemic mixtures. Alternatively, the compounds of the invention may be prepared as their individual enantiomers or diastereomers by reaction of a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereo-isomeric compounds, separating the diastereomers, and recovering the optically pure enantiomers. Resolution of enantiomers may be carried out using covalent diastereomeric derivatives of the compounds of the invention, or by using dissociable complexes (e.g, crystalline diastereomeric salts). Diastereomers have distinct physical properties (e.g, melting points, boiling points, solubility, reactivity, etc.) and may be readily separated by taking advantage of these dissimilarities. The diastereomers may be separated by chromatography, or by separation/resolution techniques based upon differences in solubility. The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization. A more detailed description of the techniques applicable to the resolution of stereoisomers of compounds from their racemic mixture can be found in Jean Jacques, Andre Collet and Samuel H. Wilen, “Enantiomers, Racemates and Resolutions” John Wiley And Sons, Inc., 1981, which is incorporated herein by reference. The compounds of the invention may be prepared as solvates ( e.g ., hydrates). The term “solvate” refers to a complex of variable stoichiometry formed by a solute (for example, a compound of the invention or a pharmaceutically acceptable salt thereof) and a solvent. Such solvents for the purpose of the invention may not interfere with the biological activity of the solute. Non-limiting examples of suitable solvents include water, acetone, methanol, ethanol and acetic acid. Preferably the solvent used is a pharmaceutically acceptable solvent.

Furthermore, the compounds of the invention may be prepared as crystalline forms.

The crystalline forms may exist as polymorphs.

It should be noted that in view of the close relationship between compound of the invention and their other forms, whenever a compound is referred to in this context herein, a corresponding salt, diastereomer, enantiomer, racemate, crystalline, polymorph, prodrug, hydrate, or solvate is also intended, if it is possible or appropriate under certain circumstances.

2. Compositions

Another aspect of the present invention provides a composition for use in treating or preventing diseases, disorders, or conditions associated with virus infection. The composition contains a therapeutically effective amount of a compound that can inhibit AN06, a pharmaceutically acceptable salt thereof, a diastereomer thereof, an enantiomer thereof, a racemate thereof, a solvate thereof, a hydrate thereof, a prodrug thereof, a crystalline thereof, or a combination thereof.

Still further aspect of the present invention provides a composition for use in disinfecting or sanitizing an object from virus contamination. The composition contains a therapeutically effective amount of a compound that can inhibit AN06, a pharmaceutically acceptable salt thereof, a diastereomer thereof, an enantiomer thereof, a racemate thereof, a solvate thereof, a hydrate thereof, a prodrug thereof, a crystalline thereof, or a combination thereof.

The compound inhibits AN06 phospholipid scramblase activity and reduce phosphatidyl serine (PS) externalization. By inhibiting PS externalization, the compound or salt can inhibit the entry of virus in host cells. In addition, the compound or salt inhibits viral replication. Thus, the compound or salt can treat or prevent diseases, disorders, or conditions associated with virus infection and can disinfect or sanitize an object from virus contamination. The compound or salt has AN06 inhibition activity, as illustrated in Examples 3 and 4. The compound or salt has anti-virus activity and anti-viral replication activity, as illustrated in Examples 6 and 7. In some embodiments, the compound is represented by Formula (I) above.

In some embodiments, the virus of the present invention is an RNA virus. In some embodiments, the RNA virus may be an enveloped positive-strand RNA virus. In some embodiments, the enveloped positive-strand RNA virus may be a Coronaviridae. The Coronaviridae includes letovirinae and orthcoromavirinae (known as coronavirus) subfamily.

In some embodiments, the virus of the present invention may comprise all virus in the orthcoronavirinae (coronavirus). The orthcoronavirinae (coronavirus) may be selected from the group consisting of alphacoronavirus (Group 1 CoV), betacoronavirus (Group 2 CoV), gammacoronavirus (Group 3 CoV) and deltacoronavirus (Group 4 CoV) genus. In certain embodiments, the virus may be a Betacoronavirus. Specifically, the genus Betacoronavirus (Group 2 CoV) comprises five subgenera or lineages (A, B, C, and D): Embecovirus (lineage A), Sarbecovirus (lineage B), Merbecovirus (lineage C), Nobecovirus (lineage D) and Hibecovirus. The Embecovirus comprises Betacoronavirus 1 species (ex. Bovine coronavirus and Human coronavirus OC43), China Rattus coronavirus HKU24 species, Human coronavirus HKU1 species, Murine coronavirus species (ex. Mouse hepatitis virus) species, and My odes coronavirus 2JL14 species. The Sarbecovirus subgenera (lineage B) comprises severe acute respiratory syndrome-related coronavirus (SARSr-CoV). The severe acute respiratory syndrome-related coronavirus comprises severe acute respiratory syndrome coronavirus species (also known as SARS-CoV and SARS-CoV-1), severe acute respiratory syndrome coronavirus 2 species (SARS-CoV-2), Bat SARS-like coronavirus WIV1 species (Bat SL- CoV-WIVl), and Bat coronavirus RaTG13 species. The Merbecovirus subgenera (lineage C) comprises Hedgehog coronavirus 1 species, Middle East respiratory syndrome-related coronavirus species (MERS-CoV), Pipistrellus bat coronavirus HKU5 species, and Tylonycteris bat coronavirus HKU4 species. The Nobecovirus subgenera (lineage D) comprises Eidolon bat coronavirus C704 species, Rousettus bat coronavirus GCCDC1 species, and Rousettus bat coronavirus HKU9 species. The Hibecovirus subgenera comprises Bat Hp- betacoronavirus Zhejiang2013 species.

The diseases, disorders, or conditions associated with virus infection comprises cold, severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), COVID-19; and disorders or conditions thereof. The cold, also known as common cold, is a viral infectious disease of the upper respiratory tract that primarily affects the respiratory mucosa of the nose, throat, sinuses, and larynx. Signs and symptoms of the cold include coughing, sore throat, runny nose, sneezing, headache, and fever. The severe acute respiratory syndrome (SARS) is a viral respiratory disease of zoonotic origin caused by severe acute respiratory syndrome coronavirus (SARS-CoV or SARS-CoV-1). In December 2019, another strain of SARS-CoV was identified as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This new strain causes coronavirus disease 2019 (COVID-19), a disease which brought about the COVID-19 pandemic. The middle East respiratory syndrome (MERS) is a viral respiratory infection caused by Middle East respiratory syndrome-related coronavirus (MERS-CoV). Typical symptoms include fever, cough, diarrhea, and shortness of breath. The coronavirus disease 2019 (COVID-19) is a contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Symptoms of COVID-19 are variable, but often include fever, cough, headache, fatigue, breathing difficulties, loss of smell, and loss of taste.

As used herein, the term “composition” is intended to encompass a product comprising the claimed compound, salt, diastereomer, enantiomer, racemate, hydrate, solvate, or a pharmaceutical combination thereof in the therapeutically effective amount, as well as any other product which results, directly or indirectly, from claimed compound, salt, diastereomer, enantiomer, racemate, hydrate, solvate, or a pharmaceutical combination thereof.

As used herein, the term “pharmaceutical composition” refers to a mixture of a therapeutically active component (ingredient) with one or more other components, which may be chemically or biologically active or inactive. Such components may include, but not limited to, carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, excipients, and adjuvants.

As used herein, the term “pharmaceutical combination” means a product that results from the mixing or combining of more than one therapeutically active ingredient.

As used herein, the term “acceptable” with respect to a formulation, composition or ingredient, as used herein, means having no persistent detrimental effect on the general health of the subject being treated.

As used herein, the term “carrier” refers to chemical or biological material that can facilitate the incorporation of a therapeutically active ingredient(s) into cells or tissues.

Suitable excipients may include, for example, water, pharmaceutically acceptable organic solvents such as paraffins ( e.g ., petroleum fractions), vegetable oils (e.g. groundnut or sesame oil), mono- or polyfunctional alcohols (e.g, ethanol or glycerol), carriers such as natural mineral powders ( e.g ., kaoline, clays, talc, chalk), synthetic mineral powders (e.g., highly dispersed silicic acid and silicates), sugars (e.g, cane sugar, lactose and glucose), emulsifiers (e.g, lignin, spent sulphite liquors, methylcellulose, starch and polyvinylpyrrolidone), and lubricants (e.g, magnesium stearate, talc, stearic acid and sodium lauryl sulphate).

Any suitable pharmaceutically acceptable carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, excipients, and adjuvants known to those of ordinary skill in the art for use in pharmaceutical compositions may be selected and employed in the compositions described herein. The compositions described herein may be in the form of a solid, liquid, or gas (aerosol). For example, they may be in the form of tablets (coated tablets) made of, for example, collidone or shellac, gum Arabic, talc, titanium dioxide or sugar, capsules (gelatin), solutions (aqueous or aqueous-ethanolic solution), syrups containing the active substances, emulsions or inhalable powders (of various saccharides such as lactose or glucose, salts and mixture of these excipients with one another), and aerosols (propellant- containing or -free inhale solutions). Also, the compositions described herein may be formulated for sustained or slow release.

Other embodiments and uses will be apparent to one skilled in the art in light of the present disclosures. The following examples are provided merely as illustrative of various embodiments and shall not be construed to limit the invention in any way.

EXAMPLES

The present invention is further exemplified by the following examples. The examples are for illustrative purpose only and are not intended to limit the invention, nor should they be construed as limiting the invention in any manner. Those skilled in the art will appreciate that variations and modifications can be made without changing the scope of the invention.

'H and 13 C NMR spectra were recorded in CDCF (residual internal standard CHCF = d 7.26), DMSO-rA, (residual internal standard CD3SOCD2H = d 2.50), methanol-6/4 (residual internal standard CD2HOD = d 3.20), or acetone-rA, (residual internal standard CD3COCD2H = d 2.05). The chemical shifts (d) reported are given in parts per million (ppm) and the coupling constants (J) are in Hertz (Hz). The spin multiplicities are reported as s = singlet, bs = broad singlet, bm = broad multiplet, d = doublet, t = triplet, q = quartet, p = pentuplet, dd = doublet of doublet, ddd = doublet of doublet of doublet, dt = doublet of triplet, td = triplet of doublet, tt = triplet of triplet, and m = multiplet. Medium pressure liquid chromatography (MPLC) was performed with silica gel columns in both the normal phase and reverse phase.

EXAMPLE 1

SYNTHESIS OF COMMON INTERMEDIATES

In general, compounds used in the reactions described herein may be made according to organic synthesis techniques known to those skilled in this art, starting from commercially available chemicals and/or from compounds described in the chemical literature. “Commercially available chemicals” may be obtained from standard commercial sources including Aldrich Chemical (Milwaukee Wis., including Sigma Chemical and Fluka), Fisher Scientific Co. (Pittsburgh Pa.), and Wako Chemicals USA, Inc. (Richmond Va.), for example.

Methods known to one of ordinary skill in the art may be identified through various reference books and databases. Suitable reference books and treatises that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation, include for example, “Synthetic Organic Chemistry,”

John Wiley & Sons, Inc., New York; “Advanced Organic Chemistry: Reactions, Mechanisms and Structure”, 4th Ed., Wiley-Interscience, New York, 1992; “Organic Synthesis: Concepts, Methods, Starting Materials,” Second, Revised and Enlarged Edition (1994) John Wiley & Sons; and “Chemistry of Functional Groups” John Wiley & Sons, in 73 volumes.

Specific and analogous reactants may also be identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, Washington, D.C.. Chemicals that are known but not commercially available in catalogs may be prepared by custom chemical synthesis houses.

1. Synthesis of Formula Il-a

1) Suzuki coupling A

Suzuki coupling Formular ll-a

Synthesis of 6-(3-fluorophenyl)pyridazin-3-amine

(3-Fluorophenyl)boronic acid (300 mg, 2.15 mmol), 6-bromopyridazin-3-amine (310 mg, 1.79 mmol), Pd(PPh3)4 (103 mg, 0.089 mmol), and potassium carbonate (740 mg, 5.36 mmol) were mixed in H 2 0/Dimethylformamide (DMF) (4.3/1.3 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was concentrated. The residue was purified by MPLC to give 6-(3-fluorophenyl)pyridazin-3 -amine (255 mg, 75%) as a white solid.

¾ NMR (400 MHz, CDCh) d 7.77 - 7.71 (m, 2H), 7.64 (d, J= 9.3 Hz, 1H), 7.52 - 7.42 (m, 1H), 7.16 - 7.10 (m, 1H), 6.86 (d, J= 9.2 Hz, 1H), 4.90 (s, 2H).

Synthesis of [l,l’-biphenyl]-4-amine

Phenylboronic acid (255 mg, 2.09 mmol), 4-bromoaniline (300 mg, 1.74 mmol), Pd(PPh3)4 (100 mg, 0.087 mmol), and potassium carbonate (891 mg, 6.45 mmol) were mixed in H2O/DMF (3.5/3.5 mL) and heated in a microwave reactor for 30 minutes at 100°C. The reaction mixture was extracted by ethyl acetate (EA) and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC to give [I,G- biphenyl]-4-amine (257 mg, 87%) as a yellow solid.

¾ NMR (400 MHz, CDCh) d 7.58 - 7.55 (m, 2H), 7.49 - 7.38 (m, 4H), 7.32 - 7.27 (m, 1H), 6.81 - 6.76 (m, 2H), 3.75 (s, 2H).

Synthesis of 5-phenylpyrazin-2-amine

Phenylboronic acid (255 mg, 2.09 mmol), 5-bromopyrazin-2-amine (303 mg, 1.74 mmol), Pd(PPh3)4 (100 mg, 0.087 mmol), and potassium carbonate (891 mg, 6.45 mmol) were mixed in H2O/DMF (3.5/3.5 mL) and heated in a microwave reactor for 30 minutes at 100°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by MPLC to give 5- phenylpyrazin-2-amine (240 mg, 81%) as a yellow solid.

¾ NMR (400 MHz, CDCh) d 8.49 (s, 1H), 8.09 (d, 7=1.4 Hz, 1H), 7.90 (d, 7=7.4 Hz, 2H), 7.50 - 7.46 (m, 2H), 7.41 - 7.36 (m, 1H), 4.70 - 4.57 (m, 2H).

Synthesis of 5-phenylpyridin-2-amine

Phenylboronic acid (255 mg, 2.09 mmol), 6-bromopyridin-3 -amine (300 mg, 1.74 mmol), Pd(PPh3)4 (100 mg, 0.087 mmol), and potassium carbonate (891 mg, 6.45 mmol) were mixed in H2O/DMF (3.5/3.5 mL) and heated in a microwave reactor for 30 minutes at 100°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The residue was purified by MPLC to give 5- phenylpyridin-2-amine (251 mg, 84%) as an orange solid.

¾ NMR (400 MHz, CDCI3) d 8.35 (d, 7=1.9 Hz, 1H), 7.70 (dd, 7=2.4, 8.8 Hz, 1H),

7.56 - 7.51 (m, 2H), 7.45 (t, 7=7.6 Hz, 2H), 7.34 (t, 7=7.3 Hz, 1H), 6.61 (d, 7=8.5 Hz, 1H),

4.57 - 4.46 (m, 2H).

Synthesis of 5-(3-fluorophenyl)pyrimidin-2-amine

(3-Fluorophenyl)boronic acid (500 mg, 3.57 mmol), 5-bromopyrimidin-2-amine (518 mg, 2.98 mmol), Pd(PPh3)4 (172 mg, 0.15 mmol), and potassium carbonate (1.23 g, 8.93 mmol) were mixed in H2O/DMF (6/6 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by dichloromethane (DCM) and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The crude mixture was solidified by using EA and «-hexane (HEX) to give 5-(3-fluorophenyl)pyrimidin-2-amine (271 mg, 48%) as a grey solid. ¾ NMR (400 MHz, CDCI3) d 8.55 (s, 2H), 7.54 - 7.32 (m, 1H), 7.31 - 7.23 (m, 1H), 7.23 - 7.18 (m, 1H), 7.11 - 7.05 (m, 1H), 5.25 (s, 2H).

Synthesis of 6-phenylpyridazin-3-amine Phenylboronic acid (2.5 g, 20.7 mmol), 6-bromopyridazin-3 -amine (3 g, 17.2 mmol),

Pd(PPh 3 ) 4 (996 mg, 0.86 mmol), and potassium carbonate (8.3 g, 60.3 mmol) were mixed in H2O/DMF (34/39 mL) and stirred for 21 hours at 105°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was purified by MPLC to give 6-phenylpyridazin-3 -amine (1.9 g, 63%) as a white solid.

¾ NMR (400 MHz, CDCI3) d 8.00 - 7.98 (m, 2H), 7.66 (d, 7=9.1 Hz, 1H), 7.61 - 7.39 (m, 3H), 6.85 (d, 7=9.3 Hz, 1H), 4.76 (s, 2H).

Synthesis of 5-phenylpyrimidin-2-amine Phenylboronic acid (255 mg, 2.09 mmol), 5-bromopyrimidin-2-amine (303 mg, 1.74 mmol), Pd(PPh3)4 (100 mg, 0.087 mmol), and potassium carbonate (891 mg, 6.45 mmol) were mixed in H2O/DMF (3.5/3.5 mL) and heated in a microwave reactor for 30 minutes at 100°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by MPLC to give 5- phenylpyrimidin-2-amine (275 mg, 92%) as a yellow solid.

¾ NMR (400 MHz, CDCI3) d 8.56 (s, 2H), 7.53 - 7.46 (m, 4H), 7.42 - 7.36 (m, 1H), 5.13 (d, .7=1.8 Hz, 2H).

Synthesis of 5-(furan-3-yl)pyrimidin-2-amine Furan-3-ylboronic acid (617 mg, 5.52 mmol), 5-bromopyrimidin-2-amine (800 mg, 4.6 mmol), Pd(PPh3)4 (266 mg, 0.23 mmol), and potassium carbonate (1.9 g, 13.8 mmol) were mixed in H2O/DMF (9.2/9.2 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The crude mixture was solidified by using EA and HEX to give 5-(furan-3-yl)pyrimidin-2-amine (403 mg, 54%) as a grey solid.

'HNMR (400 MHz, DMSO-7/) d 8.52 (s, 2H), 8.11 (s, 1H), 7.73 (dd, 7=1.7, 1.7 Hz, 1H), 6.93 (d, 7=1.1 Hz, 1H), 6.71 (s, 2H).

Synthesis of 5-(3-fluorophenyl)pyridin-2-amine

(3-Fluorophenyl)boronic acid (873 mg, 6.24 mmol), 5-bromopyridin-2-amine (900 mg, 5.2 mmol), Pd(PPh3)4 (301 mg, 0.26 mmol), and potassium carbonate (2.2 g, 15.6 mmol) were mixed in H2O/DMF (10.4/10.4 mL) and heated in a microwave reactor for 60 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was purified by MPLC to give 5-(3- fluorophenyl)pyridin-2-amine (829 mg, 85%) as a white solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 8.30 (d, 7=2.0 Hz, 1H), 7.74 (dd, 7=2.6, 8.6 Hz, 1H), 7.66 - 7.53 (m, 1H), 7.45 - 7.41 (m, 2H), 7.11 - 7.05 (m, 1H), 6.53 - 6.50 (m, 1H), 6.17 (s, 2H).

Synthesis of 4-(pyridin-2-yl)aniline 2-Bromopyridine (1.16 mL, 12 mmol), 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)aniline (2.19 g, 10 mmol), Pd(PPh3)2Cl2 (0.70 g, 1 mmol), andNa 2 C0 3 (3.18 g, 30 mmol) were mixed in H 2 0/l,4-dioxane (12.5/37.5 mL) and stirred for 21 hours at 100°C. The reaction mixture was concentrated and purified by MPLC to give 4-(pyridin-2-yl)aniline (1.57 g, 92%) as an orange solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 8.54 - 8.51 (m, 1H), 7.81 - 7.72 (m, 4H), 7.17 - 7.13 (m, 1H), 6.66 - 6.60 (m, 2H), 5.44 (s, 2H).

Synthesis of 4-(pyridin-3-yl)aniline

3-Bromopyridine (1.17 mL, 12 mmol), 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)aniline (2.19 g, 10 mmol), Pd(PPh3)2Cl2 (0.70 g, 1 mmol), andNa 2 CC> 3 (3.18 g, 30 mmol) were mixed in H 2 0/l,4-dioxane (12.5/37.5 mL) and stirred for 16 hours at 100°C. The reaction mixture was concentrated and purified by MPLC to give 4-(pyri din-3 -yl)aniline (1.57 g, 92%) as a pale yellow solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 8.78 (d, 7=2.1 Hz, 1H), 8.41 (dd, 7=1.4, 4.7 Hz, 1H), 7.94 - 7.90 (m, 1H), 7.66 - 7.53 (m, 1H), 7.45 - 7.40 (m, 2H), 6.70 - 6.64 (m, 2H), 5.34 (s, 2H).

Synthesis of 4-(pyridin-4-yl)aniline

4-Bromopyridine hydrochloride (2.33 g, 12 mmol), 4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)aniline (2.19 g, 10 mmol), Pd(PPh3)2Cl2 (0.70 g, 1 mmol), andNa 2 C0 3 (3.18 g, 30 mmol) were mixed in H 2 0/l,4-dioxane (12.5/37.5 mL) and stirred for 16 hours at 100°C. The reaction mixture was concentrated and purified by MPLC to give crude 4-(pyridin- 4-yl)aniline (1.16 g, 67%) as a pale brown solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 8.49 - 8.47 (m, 2H), 7.58 - 7.52 (m, 4H), 6.69 - 6.64 (m, 2H), 5.53 (s, 2H). Synthesis of 4-(pyrimidin-2-yl)aniline

2-Chloropyrimidine (1.37 g, 12 mmol), 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)aniline (2.19 g, 10 mmol), Pd(PPh3)2Cl2 (0.70 g, 1 mmol), and Na 2 C0 3 (3.18 g, 30 mmol) were mixed in H 2 0/l,4-dioxane (12.5/37.5 mL) and stirred for 16 hours at 100°C. The reaction mixture was concentrated and purified by MPLC to give 4-(pyrimidin-2-yl)aniline (1.37 g, 80%) as a brown solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 8.73 (d, 7=4.9 Hz, 2H), 8.12 - 8.07 (m, 2H), 7.21 (t, 7=4.8 Hz, 1H), 6.65 - 6.60 (m, 2H), 5.68 (s, 2H).

Synthesis of 4-(pyrazin-2-yl)aniline

2-Chloropyrazine (1.07 mL, 12 mmol), 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)aniline (2.19 g, 10 mmol), Pd(PPh3)2Cl2 (0.70 g, 1 mmol), and Na 2 CC> 3 (3.18 g, 30 mmol) were mixed in H 2 0/l,4-dioxane (12.5/37.5 mL) and stirred for 16 hours at 100°C. The reaction mixture was concentrated and purified by MPLC to give 4-(pyrazin-2-yl)aniline (1.35 g, 78%) as a brown solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 9.06 (d, 7=1.5 Hz, 1H), 8.55 - 8.54 (m, 1H), 8.38 (d, 7=2.5 Hz, 1H), 7.89 - 7.83 (m, 2H), 6.69 - 6.64 (m, 2H), 5.63 (s, 2H).

Synthesis of 4-(pyrimidin-5-yl)aniline

5-Bromopyrimidine (1.91 g, 12 mmol), 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)aniline (2.19 g, 10 mmol), Pd(PPh3)2Cl2 (0.70 g, 1 mmol) and Na 2 C0 3 (3.18 g, 30 mmol) were combined in H 2 0/l,4-dioxane (12.5/37.5 mL) and stirred for 16 hours at 100°C. The reaction mixture was concentrated and purified by MPLC to give crude 4-(pyrimidin-5- yl)aniline (1.50 g, 87%) as a pale brown solid.

¾ NMR (400 MHz, DMSO-d 6 ) d 9.02 (s, 1H), 9.01 (s, 2H), 7.53 - 7.48 (m, 2H), 6.71 - 6.66 (m, 2H), 5.49 (s, 2H).

Synthesis of 4-(pyrimidin-4-yl)aniline

Step 1: 177-Pyrimidin-6-one (10 g, 104 mmol) and POCI3 (100 mL, 1.08 mol) were charged to a pressure flask. Flask was flushed with nitrogen and heated for 6 hours at 100°C. The reaction mixture was concentrated under reduced pressure to remove POCI3. The reaction mixture was poured into EA carefully and stirred for 30 minutes. The reaction mixture was filtered, and the filter cake was washed with ethyl acetate, dried to give 4-chloropyrimidine (3.50 g, crude) as a brown solid.

¾ NMR (400 MHz, DMSO-d 6 ) d 9.14 (s, 1H), 8.07 (d, 7=7.20 Hz, 1H), 6.62 (d, ,7=7.60 Hz, 1H).

Step 2: A mixture of 4-chloropyrimidine (1.80 g, 15.7 mmol), 4-(4, 4,5,5 -tetram ethyl- 1, 3, 2-dioxaborolan-2-yl)aniline (3.79 g, 17.3 mmol), CS2CO3 (20.5 g, 62.9 mmol), 1,1'- bis(diphenylphosphino)ferrocene]dichloropalladium(II) (575 mg, 0.79 mmol) in toluene (12 mL), ethanol (4 mL), and H2O (3.6 mL) and the mixture was degassed and purged with N2 for 3 times, and then the mixture was stirred for 12 hours at 100°C under N2 atmosphere. Thin- layer chromatography (TLC) indicated 4-chloropyrimidine was consumed completely, and one major new spot with larger polarity was detected. The reaction mixture was diluted with water and extracted with EA. The combined organic layers were washed with brine, dried over Na 2 SC> 4 , filtered, and concentrated under reduced pressure to give a residue. The reaction mixture was concentrated and purified by column chromatography to give 4-(pyrimidin-4- yl)aniline (1.70 g, crude) as a yellow solid.

¾ NMR (400 MHz, DMSO-d 6 ) d 9.02 (s, 1H), 8.62 (d, 7=5.20 Hz, 1H), 7.94 (d, ,7=8.80 Hz, 2H), 7.79 - 7.83 (m, 1H), 6.65 (d, 7=8.80 Hz, 2H), 5.80 (s, 2H)

2. Synthesis of Formula Il-b

Synthesis of 3-bromo-/V-((5-methylfuran-2-yl)methyl)benzamide

3-Bromobenzoyl chloride (2.17 mL, 16.5 mmol) and l-(5-methylfuran-2- yl)methanamine (1.5 mL, 13.7 mmol) were dissolved in DCM (137 mL), followed up by addition of A,/V-diisopropylethylamine (DIPEA) (5.14 mL, 29.6 mmol) and stirred for 18 hours at room temperature (r.t.). The reaction mixture was extracted by DCM and saturated aqueous (aq.) NaHCC . The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC to give 3 -bromo-A -((5-methylfuran-2-yl (methyl )benzamide (4 g, >99%) as an orange oil.

¾ NMR (400 MHz, CDCh) d 7.95 (dd, 7=1.8, 1.8 Hz, 1H), 7.74 - 7.71 (m, 1H), 7.65 (ddd, 7=1.0, 2.0, 8.0 Hz, 1H), 7.33 (dd, 7=7.9, 7.9 Hz, 1H), 6.31 (s, 1H), 6.20 (d, 7=3.0 Hz, 1H), 5.94 (dd, 7=1.0, 3.0 Hz, 1H), 4.59 (d, 7=5.3 Hz, 2H), 2.31 (s, 3H).

Synthesis of 3-bromo-/V-phenethylbenzamide

3-Bromobenzoyl chloride (1.3 mL, 9.9 mmol) and 2-phenylethan-l -amine (1 mL, 8.25 mmol) were dissolved in DCM (82 mL), followed up by addition of DIPEA (3 mL, 17.7 mmol) and stirred for 25 hours at room temperature. The reaction mixture was extracted by DCM and saturated aq. MLCl. The organic layer was dried over anhydrous NaiSCL and concentrated.

The crude mixture was solidified by using EA and HEX to give 3-bromo-A- phenethylbenzamide (1.8 g, 73%) as a yellowish white solid. ¾ NMR (400 MHz, DMSO-7 6 ) d 8.71 (t, 7=5.4 Hz, 1H), 7.99 (dd, 7=1.8, 1.8 Hz, 1H), 7.82 (dd, 7=1.3, 6.4 Hz, 1H), 7.75 - 7.72 (m, 1H), 7.44 (dd, 7=7.9, 7.9 Hz, 1H), 7.33 - 7.28 (m, 2H), 7.26 - 7.19 (m, 3H), 3.51 - 3.45 (m, 2H), 2.85 (t, 7=7.4 Hz, 2H).

Synthesis of 3-bromo-/V-(3-phenylpropyl)benzamide

3-Bromobenzoyl chloride (0.82 mL, 6.2 mmol) and 3-phenylpropan-l-amine (0.74 mL, 5.18 mmol) were dissolved in DCM (52 mL), followed up by addition of DIPEA (1.9 mL, 11 mmol) and stirred for 25 hours at room temperature. The reaction mixture was extracted by DCM and saturated aq. MLCl. The organic layer was dried over anhydrous NaiSCL and concentrated to give 3 -bromo-A -(3 -phenyl propyl )benzamide (2.17 g, >99%) as a brown oil.

¾ NMR (400 MHz, DMSO-7 6 ) d 8.66 - 8.60 (m, 1H), 8.03 - 8.01 (m, 1H), 7.86 - 7.84 (m, 1H), 7.75 - 7.72 (m, 1H), 7.46 - 7.42 (m, 1H), 7.31 - 7.26 (m, 2H), 7.24 - 7.18 (m, 3H), 3.28 (dd, 7=6.9, 12.8 Hz, 2H), 2.66 - 2.60 (m, 2H), 1.88 - 1.79 (m, 2H).

Synthesis of 3-bromo-/V-(2-cyclohexylethyl)benzamide

3-Bromobenzoyl chloride (0.12 mL, 0.9 mmol) and 2-cyclohexylethan-l -amine (0.13 mL, 0.9 mmol) were dissolved in DCM (9.1 mL), followed up by addition of DIPEA (0.34 mL, 1.9 mmol) and stirred for 23 hours at room temperature. The reaction mixture was extracted by DCM and saturated aq. MLCl. The organic layer was dried over anhydrous NaiSC o give crude 3-bromo-A-(2-cyclohexylethyl)benzamide (300 mg, >99%) as a yellow solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 8.54 (t, 7=5.3 Hz, 1H), 8.01 (t, 7=1.8 Hz, 1H), 7.85 - 7.83 (m, 1H), 7.74 - 7.71 (m, 1H), 7.43 (t, 7=7.9 Hz, 1H), 3.32 - 3.24 (m, 2H), 1.76 - 0.87 (m, 13H).

Synthesis of 3-bromo-A-((1R,2S)-2-phenylcyclopropyl)benzamide 3-Bromobenzoyl chloride (0.2 mL, 0.9 mmol) and (1R,2S)-2-phenylcyclopropan-l- amine hydrochloride (129 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.42 mL, 2.4 mmol) and stirred for 20 hours at room temperature. The reaction mixture was extracted by 10% methanol (MeOH) in DCM and saturated aq. MLCl. The organic layer was dried over anhydrous NaiSOHo give crude 3-bromo-/V-((1R,2S)-2- phenylcyclopropyl)benzamide (297 mg, >99%) as a beige solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 8.81 (d, 7=4.3 Hz, 1H), 8.03 (t, 7=1.8 Hz, 1H), 7.86 - 7.84 (m, 1H), 7.76 - 7.73 (m, 1H), 7.45 (t, 7=7.9 Hz, 1H), 7.29 (t, 7=7.4 Hz, 2H), 7.20 - 7.14 (m, 3H), 3.07 - 3.00 (m, 1H), 2.13 - 2.07 (m, 1H), 1.39 - 1.33 (m, 1H), 1.28 - 1.17 (m, 1H).

3. Synthesis of Formula II-c

1) Buchwald-Hartwig coupling

Synthesis of 3-((6-phenylpyridazin-3-yl)amino)benzoic acid

Step 1: To a solution of methyl 3-bromobenzoate (18 g, 83.7 mmol) in 1,4-dioxane (90 mL) was added 6-phenylpyridazin-3 -amine (15.1 g, 87.9 mmol), BrettPhos (8.99 g, 16.7 mmol), and cesium carbonate (68.2 g, 209 mmol). Pd 2 (dba) 3 (2.3 g, 2.51 mmol) was added into the solution. The solution was stirred for 6 hours at 100°C. The reaction was filtered, and the filter cake was triturated with tetrahydrofuran (THF) (180 mL) and MeOH (35 mL) for 2 hours at room temperature. Then the suspension was filtered, and filtrate was concentrated under reduced pressure to give a residue. The residue was dissolved in THF (200 mL). The solution was filtered through a pad of silica gel. The filtrate was concentrated under vacuum to give methyl 3-[(6-phenylpyridazin-3-yl)amino]benzoate (10.2 g, 40%) as a yellow solid. ¾ NMR (400 MHz, DMSO-7 6 ) d 9.93 (s, 1H), 8.48 (s, 1H), 8.15 (d, 7= 6.0 Hz, 1H), 8.05 (t, 7= 5.6 Hz, 3H), 7.61 (d, 7= 7.6 Hz, 1H), 7.53 (m, 7= 6.0 Hz, 4H), 7.38 (d, 7= 9.2 Hz, 1H), 3.89 (s, 3H).

Step 2: Methyl 3-[(6-phenylpyridazin-3-yl)amino]benzoate (9 g, 29.5 mmol) was dissolved in MeOH/THF (7/45 mL). aq. NaOH (2 M, 29.4 mL) was added into the solution. The solution was stirred for 12 hours at room temperature. The reaction mixture was concentrated under reduced pressure to remove MeOH and THF to give a residue. The H2O (80 mL) was added into the residue. The pH value of the suspension was adjusted to 2 by aq. HC1 (2 M). THF (30 mL) was added into the suspension. The suspension was filtered, and the filter cake was dried under vacuum to give 3-((6-phenylpyridazin-3-yl)amino)benzoic acid (5 g, 58%) as yellow solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 12.93 (s, 1H), 9.61 (s, 1H), 8.50 (s, 1H), 8.06 (t, 7=10.4 Hz, 4H), 7.57 - 7.46 (m, 5H), 7.24 (d, 7=9.2 Hz, 1H).

Synthesis of 3-((5-phenylpyrimidin-2-yl)amino)benzoic acid

Step 1: To a solution of 5-phenylpyrimidin-2-amine (22 g, 128 mmol) in 1,4-dioxane (130 mL) were added methyl 3-bromobenzoate (18.4 g, 85.7 mmol), cesium carbonate (83.7 g, 257 mmol), and XPhos (12.3 g, 25.7 mmol). Then Pd 2 (dba) 3 (2.35 g, 2.57 mmol) was added into the solution. Then solution was stirred for 12 hours at 100°C. The reaction solution was poured into H2O (500 mL). The suspension was filtered, and the filter cake was rinsed with H2O (100 mL). The filter cake was dried in vacuum to give the crude product. The crude product was diluted with THF (1 L). The resulting suspension was filtered, and the filter cake was washed with THF (200 mL). The filtrate was purified by column chromatography to give methyl 3-[(5-phenylpyrimidin-2-yl)amino]benzoate (9 g, 34%) as a white solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 10.03 (s, 1H), 8.88 (s, 2H), 8.05 (d, 7= 1.2 Hz, 1H), 8.05 (t, 7= 1.6 Hz, 1H), 7.75 (d, 7= 8.4 Hz, 2H), 7.57 - 7.38 (m, 5H), 3.86 (s, 3H).

Step 2: An aq. NaOH (2 M, 29.5 mL) was added into a solution of methyl 3-[(5- phenylpyrimidin-2-yl)amino]benzoate (9 g, 29.5 mmol) in THF (70 mL). Then MeOH (50 mL) was added into the reaction solution. The solution was stirred for 12 hours at 50°C. The reaction solution was concentrated to give a crude product. The crude product was added into H2O (500 mL). Then pH value of the solution was adjusted to 1-2 by aq. HC1 (1 M).

The suspension was filtered, and the filter cake was washed with H2O (200 mL). The filter cake was dried under vacuum to give 3-((5-phenylpyrimidin-2-yl)amino)benzoic acid (5 g, 58%) as white solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 12.89 (s, 1H), 9.99 (s, 1H), 8.88 (s, 2H), 8.46 (d, 7=1.6 Hz, 1H), 8.02 (d, 7=1.2 Hz, 1H), 7.74 (d, 7=8.4 Hz, 2H), 7.55 - 7.36 (m, 5H).

Synthesis of 3-((5-(3-fluorophenyl)pyridin-2-yl)amino)benzoic acid

Step 1: 5-(3-Fluorophenyl)pyridin-2-amine (700 mg, 3.72 mmol), methyl 3- bromobenzoate (1.2 g, 4.84 mmol), Pd 2 (dba) 3 (340 mg, 0.37 mmol), BrettPhos (339 mg, 0.74 mmol), and cesium carbonate (2.4 g, 7.44 mmol) were mixed in 1,4-dioxane (18.6 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The crude mixture was solidified by using DCM and HEX to give methyl 3-{[5-(3- fluorophenyl)pyridin-2-yl]amino}benzoate (539 mg, 45%) as a beige solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 9.52 (s, 1H), 8.60 (d, 7=2.3 Hz, 1H), 8.36 (dd, 7=1.9, 1.9 Hz, 1H), 8.07 (ddd, 7=1.1, 2.3, 8.1 Hz, 1H), 7.99 (dd, 7=2.6, 8.8 Hz, 1H), 7.61 - 7.40 (m, 5H), 7.19 - 7.13 (m, 1H), 6.94 (d, 7=8.8 Hz, 1H), 3.87 (s, 3H).

Step 2: Methyl 3-{[5-(3-fluorophenyl)pyridin-2-yl]amino}benzoate (400 mg, 1.24 mmol) and LiOH-H 2 0 (521 mg, 12.4 mmol) were mixed in H 2 0/l,4-dioxane (5.2/24.8 mL) and stirred for 18 hours at room temperature. The reaction mixture acidified by adding 1 N HC1 and extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The crude mixture was solidified by using EA to give 3-((5-(3- fluorophenyl)pyridin-2-yl)amino)benzoic acid (348 mg, 91%) as a beige solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 12.90 (s, 1H), 9.47 (s, 1H), 8.60 (d, 7=2.3 Hz, 1H), 8.35 (dd, 7=1.9, 1.9 Hz, 1H), 8.10 - 7.95 (m, 2H), 7.61 - 7.31 (m, 5H), 7.18 - 7.12 (m, 1H),

6.94 (d, 7=8.5 Hz, 1H).

Synthesis of 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)benzoic acid

Step 1: To a solution of 5-(3-fluorophenyl)pyrimidin-2-amine (30 g, 158 mmol) in 1,4- dioxane (210 mL) was methyl 3-bromobenzoate (31 g, 144 mmol), XPhos (20.6 g, 43.3 mmol), and cesium carbonate (141 g, 432 mmol). Then Pd 2 (dba) 3 (3.96 g, 4.32 mmol) was added into the solution. The solution was stirred for 12 hours at 100°C. The reaction solution was poured into H2O (500 mL), and the suspension was filtered. The filter cake was washed with H2O (100 mL) and dried under vacuum to give a crude product. The crude product was added into THF (1 L). The suspension was filtered, and the filter cake was washed with THF (200 mL). The filtrate was purified by column chromatography to give methyl 3-{[5-(3- fluorophenyl)pyrimidin-2-yl]amino}benzoate (10 g, 22%) as a white solid.

¾ NMR (400 MHz, DMSO-d 6 ) d 10.10 (s, 1H), 8.93 (s, 2H), 8.48 (d, J= 2 Hz, 1H), 8.05 (d, J= 8 Hz, 1H), 7.65-7.43 (m, 5H), 7.20 (m, 1H), 3.86 (s, 3H).

Step 2: An aq. NaOH (2 M, 30.9 mL) was added into a solution of methyl 3-{[5-(3- fluorophenyl)pyrimidin-2-yl]amino}benzoate (10 g, 30.9 mmol) in THF (70 mL). Then MeOH (50 mL) was added into the reaction solution. The solution was stirred for 12 hours at 50°C.

The reaction solution was concentrated to give a crude product. The crude product was added into H2O (500 mL). The pH value of the solution was adjusted to 1-2 by aq. HC1 (1 M).

The suspension was filtered. The filter cake was washed with H2O (200 mL) and dried under vacuum to give 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (5 g, 52%) as a white solid.

¾ NMR (400 MHz, DMSO-d 6 ) d 12.90 (s, 1H), 10.06 (s, 1H), 8.92 (s, 2H), 8.46 (s, 1H), 8.00 (d, ,7=8.0 Hz, 1H), 7.66 - 7.50 (m, 4H), 7.42 (t, ,7=9.2 Hz, 1H), 7.22 - 7.17 (m, 1H).

Synthesis of 3-((5-phenylpyridin-2-yl)amino)benzoic acid

Step 1: To a solution of methyl 3-bromobenzoate (20.8 g, 122 mmol) in 1,4-dioxane (125 mL) was added 5-phenylpyridin-2-amine (25.0 g, 116 mmol), XPhos (16.6 g, 34.8 mmol) and CS2CO3 (113 g, 348 mmol). The solution was degassed and purged with N2 for three times. Pd 2 (dba) 3 (3.19 g, 3.49 mmol) was added into the solution. The solution was degassed and purged with N2 for three times. The solution was stirred for 12 h at 100°C. The mixture suspension was filtered, and the filter cake was rinsed with EA. The filtrate was dried over sodium sulfate and filtered, concentrated under reduced pressure to give a residue. The residue was triturated with methyl tert-butyl ether (MTBE) for 1 hour at room temperature. The suspension was filtered, and the filter cake was rinsed with MTBE, and the filter cake was collected and dried under reduced pressure to give methyl 3-((5-phenylpyridin-2- yl)amino)benzoate (20.0 g, 56.5%) as a white solid.

¾ NMR (400 MHz, DMSO-Tf) d 9.44 (s, 1H), 8.53 (d, 7= 2.0 Hz, 1H), 8.34 (s, 1H), 8.07 (d, 7= 8.4 Hz, 1H), 7.94 (dd, 7= 8.8, 2.4 Hz, 1H), 7.66 (d, 7= 7.6 Hz, 2H), 7.49 - 7.40 (m, 4H), 7.33 (t, 7= 7.6 Hz, 1H), 6.94 (d, 7= 8.8 Hz, 1H), 3.86 (s, 3H).

Step 2: Methyl 3-((5-phenylpyridin-2-yl)amino)benzoate (20.0 g, 65.7 mmol) was dissolved in MeOH (100 mL) and THF (20 mL). aq. NaOH (2 M, 65.7 mL) was added into the solution. The solution was stirred for 12 hours at room temperature. The reaction mixture was concentrated under reduced pressure to remove MeOH and THF to give a residue. The ¾0 (80 mL) was added into the residue. The pH value of the suspension was adjusted to 5 by aq. HC1 (6 M). The suspension was filtered, and the filter cake was concentrated under reduced pressure to give 3-((5-phenylpyridin-2-yl)amino)benzoic acid (10 g, 52%) as white solid.

¾ NMR (400 MHz, DMSO-Tf) d 12.85 (s, 1H), 9.40 (s, 1H), 8.53 (d, 7= 2.4 Hz, 1H), 8.33 - 8.32 (m, 1H), 8.02 - 8.00 (m, 1H), 7.93 (dd, 7= 8.0, 2.4 Hz, 1H), 7.66 (d, 7= 7.2 Hz,

2H), 7.48 - 7.31 (m, 5H), 6.94 (d, 7= 8.8 Hz, 1H).

Synthesis of 3-((5-(furan-3-yl)pyrimidin-2-yl)amino)benzoic acid

Step 1: 5-(Furan-3-yl)pyrimidin-2-amine (400 mg, 2.48 mmol), methyl 3- bromobenzoate (807 mg, 3.23 mmol), Pd2(dba)3 (227 mg, 0.25 mmol), BrettPhos (267 mg, 0.5 mmol), and cesium carbonate (1.6 g, 4.96 mmol) were mixed in 1,4-dioxane (12 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by EA and brine. The crude mixture was solidified by using EA and HEX to give methyl 3-((5- (furan-3-yl)pyrimidin-2-yl)amino)benzoate (314 mg, 43%) as an orange solid.

Ή NMR (400 MHz, DMSO) d 10.01 (s, 1H), 8.84 (s, 2H), 8.51 (dd, 7=1.9, 1.9 Hz, 1H), 8.25 (s, 1H), 8.02 - 7.99 (m, 1H), 7.80 (dd, 7=1.7, 1.7 Hz, 1H), 7.55 (d, 7=7.8 Hz, 1H), 7.44 (dd, 7=7.9, 7.9 Hz, 1H), 7.06 (d, 7=1.0 Hz, 1H), 3.87 - 3.86 (m, 3H). Step 2: Methyl 3-((5-(furan-3-yl)pyrimidin-2-yl)amino)benzoate (300 mg, 1.02 mmol) and Li0H-H 2 0 (426 mg, 10.2 mmol) were mixed in H 2 0/l,4-dioxane (4.2/20.3 mL) and stirred for 18 hours at room temperature. Then pH value of the solution was adjusted to 1-2 by 1 N HC1. The reaction mixture was extracted by EA and brine. The crude mixture was solidified by using EA to give 3-((5-(furan-3-yl)pyrimidin-2-yl)amino)benzoic acid (199 mg, 70%) as a white solid.

'HNMR (400 MHz, DMSO) d 12.91 (s, 1H), 9.97 (s, 1H), 8.84 (s, 2H), 8.50 (dd,

7=1.8, 1.8 Hz, 1H), 8.25 (s, 1H), 7.96 (dd, 7=1.3, 8.1 Hz, 1H), 7.80 (dd, 7=1.7, 1.7 Hz, 1H), 7.54 (d, 7=7.8 Hz, 1H), 7.41 (dd, 7=7.9, 7.9 Hz, 1H), 7.06 (d, 7=1.0 Hz, 1H).

Synthesis of 3-((4-(pyridin-2-yl)phenyl)amino)benzoic acid

Step 1: To a solution of methyl 3-bromobenzoate (2.88 g, 13.4 mmol) in 1,4-dioxane (45 mL) was added 4-(pyridin-2-yl)aniline (1.52 g, 8.93 mmol), BrettPhos (0.96 g, 1.79 mmol), and cesium carbonate (11.64 g, 35.7 mmol). Pd 2 (dba) 3 (0.82 g, 0.89 mmol) was added into the solution. The solution was stirred for 15 hours at 100°C. The reaction mixture was concentrated and purified by MPLC to give methyl 3-((4-(pyridin-2-yl)phenyl)amino)benzoate (1.36 g, 49%) as a yellow solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 8.73 - 8.71 (m, 1H), 8.61 - 8.59 (m, 1H), 8.05 - 8.01 (m, 2H), 7.91 - 7.87 (m, 1H), 7.86 - 7.80 (m, 1H), 7.74 - 7.71 (m, 1H), 7.46 - 7.40 (m, 3H),

7.28 - 7.24 (m, 1H), 7.21 - 7.17 (m, 2H), 3.85 (s, 3H).

Step 2: Methyl 3-((4-(pyridin-2-yl)phenyl)amino)benzoate (1.35 g, 4.43 mmol) and LiOH-H 2 0 (0.75 g, 17.73 mmol) were mixed in THF/H2O (30/15 mL) and stirred for 117 hours at room temperature. The reaction mixture was extracted by EA and aq. HC1 (IN). The organic layer was dried over anhydrous MgSCE and concentrated. The residue was purified by MPLC to give 3-((4-(pyridin-2-yl)phenyl)amino)benzoic acid (321 mg, 25%) as a pale yellow solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 12.93 (s, 1H), 8.67 (s, 1H), 8.61 - 8.60 (m, 1H), 8.05 - 8.00 (m, 2H), 7.90 - 7.86 (m, 1H), 7.85 - 7.80 (m, 1H), 7.73 - 7.71 (m, 1H), 7.45 - 7.37 (m, 3H), 7.28 - 7.24 (m, 1H), 7.21 - 7.16 (m, 2H).

Synthesis of 3-((4-(pyridin-3-yl)phenyl)amino)benzoic acid

Step 1: To a solution of methyl 3-bromobenzoate (2.18 g, 10.14 mmol) in 1,4-dioxane (46 mL) was 4-(pyridin-3-yl)aniline (1.57 g, 9.22 mmol), XPhos (0.75 g, 1.56 mmol), and cesium carbonate (6.0 g, 18.44 mmol). Pd 2 (dba) 3 (0.68 g, 0.74 mmol) was added into the solution. The solution was stirred for 16 hours at 100°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using acetone to give methyl 3-((4-(pyridin-3-yl)phenyl)amino)benzoate (1.0 g, 36%) as a pale yellow solid.

¾ NMR (400 MHz, DMSO-d 6 ) S 8.89 - 8.85 (m, 1H), 8.67 (s, 1H), 8.53 - 8.48 (m,

1H), 8.05 - 8.00 (m, 1H), 7.73 - 7.65 (m, 3H), 7.48 - 7.37 (m, 4H), 7.24 - 7.19 (m, 2H), 3.85 (s, 3H).

Step 2: Methyl 3-((4-(pyridin-3-yl)phenyl)amino)benzoate (0.35 g, 1.15 mmol) and LiOH-HiO (0.19 g, 4.6 mmol) were mixed in THF/H2O (8/4 mL) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and aq. HC1 (IN). The organic layer was dried over anhydrous MgSCL and concentrated. The residue was purified by MPLC to give crude 3-((4-(pyridin-3-yl)phenyl)amino)benzoic acid (125 mg, 37%) as a yellow solid.

¾ NMR (400 MHz, DMSO-d 6 ) S 8.88 (s, 1H), 8.62 (s, 1H), 8.50 (d, J= 4.0 Hz, 1H), 8.06 - 8.02 (m, 1H), 7.70 - 7.64 (m, 2H), 7.47 - 7.30 (m, 5H), 7.24 - 7.18 (m, 2H).

Synthesis of 3-((4-(pyridin-4-yl)phenyl)amino)benzoic acid

Step 1: To a solution of methyl 3-bromobenzoate (2.18 g, 10.14 mmol) in 1,4-dioxane (46 mL) was 4-(pyridin-4-yl)aniline (1.57 g, 9.22 mmol), XPhos (0.75 g, 1.56 mmol), and cesium carbonate (6.0 g, 18.44 mmol). Pd 2 (dba) 3 (0.68 g, 0.74 mmol) was added into the solution. The solution was stirred for 16 hours at 100°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using acetone to give methyl 3-((4-(pyridin-4-yl)phenyl)amino)benzoate (1.0 g, 36%) as a pale yellow solid.

¾ NMR (400 MHz, DMSO-d 6 ) S 8.79 - 8.77 (m, 1H), 8.59 - 8.56 (m, 2H), 7.80 - 7.76 (m, 2H), 7.74 - 7.71 (m, 1H), 7.69 - 7.66 (m, 2H), 7.48 - 7.41 (m, 3H), 7.23 - 7.19 (m, 2H),

3.86 - 3.85 (m, 3H). Step 2: Methyl 3-((4-(pyridin-4-yl)phenyl)amino)benzoate (0.76 g, 2.5 mmol) and Li0H-H 2 0 (0.42 g, 10 mmol) were mixed in THF/H2O (17/8.5 mL) and stirred for 40 hours at room temperature. The reaction mixture was extracted by EA and aq. HC1 (IN). The organic layer was dried over anhydrous MgSCE and concentrated. The crude mixture was solidified by using EA and acetone to give 3-((4-(pyridin-4-yl)phenyl)amino)benzoic acid (244 mg, 34%) as a yellow solid.

¾ NMR (400 MHz, DMSO-d 6 ) S 12.99 (bs, 1H), 9.24 (s, 1H), 8.75 (s, 2H), 8.23 - 8.16 (m, 2H), 7.99 (d, J= 8.8 Hz, 2H), 7.79 (s, 1H), 7.61 - 7.40 (m, 3H), 7.26 (d, J=42A Hz, 2H).

Synthesis of 3-((4-(pyrimidin-2-yl)phenyl)amino)benzoic acid

Step 1: To a solution of methyl 3-bromobenzoate (1.89 g, 8.8 mmol) in 1,4-dioxane (40 mL) was 4-(pyrimidin-2-yl)aniline (1.37 g, 8.0 mmol), XPhos (0.65 g, 1.36 mmol), and cesium carbonate (5.21 g, 16 mmol). Pd 2 (dba) 3 (0.59 g, 0.64 mmol) was added into the solution. The solution was stirred for 16 hours at 100°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using acetone to give methyl 3-((4-(pyrimidin- 2-yl)phenyl)amino)benzoate (1.52 g, 62%) as a beige solid.

¾ NMR (400 MHz, DMSO-d 6 ) S 8.85 (s, 1H), 8.83 (d, J= 4.8 Hz, 2H), 8.33 - 8.29 (m, 2H), 7.76 - 7.74 (m, 1H), 7.52 - 7.42 (m, 3H), 7.33 (t, J= 4.8 Hz, 1H), 7.22 - 7.17 (m, 2H), 3.86 (s, 3H).

Step 2: Methyl 3-((4-(pyrimidin-2-yl)phenyl)amino)benzoate (1.50 g, 4.91 mmol) and LiOH-H 2 0 (0.83 g, 19.65 mmol) were mixed in THF/H2O (32/16 mL) and stirred for 40 hours at room temperature. The reaction mixture was extracted by EA and aq. HC1 (IN). The organic layer was dried over anhydrous MgSCE and concentrated. The crude mixture was solidified by using EA and acetone to give 3-((4-(pyrimidin-2-yl)phenyl)amino)benzoic acid (1.10 g, 77%) as a beige solid.

¾ NMR (400 MHz, DMSO-d 6 ) S 12.91 (bs, 1H), 8.88 - 8.79 (m, 3H), 8.32 - 8.29 (m, 2H), 7.74 (s, 1H), 7.51 - 7.46 (m, 1H), 7.46 - 7.39 (m, 2H), 7.32 (t, J= 4.8 Hz, 1H), 7.21 - 7.17 (m, 2H).

Synthesis of 3-((4-(pyrazin-2-yl)phenyl)amino)benzoic acid

Step 1: To a solution of methyl 3-bromobenzoate (1.80 g, 8.35 mmol) in 1,4-dioxane (38 mL) was 4-(pyrazin-2-yl)aniline (1.30 g, 7.59 mmol), XPhos (0.62 g, 1.29 mmol), and cesium carbonate (4.95 g, 15.2 mmol). Pd 2 (dba) 3 (0.56 g, 0.61 mmol) was added into the solution. The solution was stirred for 16 hours at 100°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using acetone to give methyl 3-((4-(pyrazin-2-yl)phenyl)amino)benzoate (1.60 g, 69%) as a brown solid.

¾ NMR (400 MHz, DMSO-d 6 ) S 9.19 (d, 7=1.4 Hz, 1H), 8.83 (s, 1H), 8.65 - 8.63 (m, 1H), 8.50 (d, J= 2.5 Hz, 1H), 8.11 - 8.07 (m, 2H), 7.76 - 7.74 (m, 1H), 7.53 - 7.40 (m, 3H), 7.22 (d, =8.8 Hz, 2H), 3.85 (s, 3H).

Step 2: Methyl 3-((4-(pyrazin-2-yl)phenyl)amino)benzoate (1.58 g, 5.74 mmol) and LiOH-HiO (0.87 g, 20.7 mmol) were mixed in THF/H2O (38/19 mL) and stirred for 64 hours at room temperature. The reaction mixture was extracted by EA and aq. HC1 (IN). The organic layer was dried over anhydrous MgSO 4 and concentrated. The residue was purified by MPLC to give 3-((4-(pyrazin-2-yl)phenyl)amino)benzoic acid (1.72 g, >99%) as a yellow solid.

¾ NMR (400 MHz, DMSO-d 6 ) S 12.83 (s, 1H), 9.18 (d, J= 1.5 Hz, 1H), 8.78 (s, 1H), 8.65 - 8.63 (m, 1H), 8.50 (d, J= 2.5 Hz, 1H), 8.08 (d, J= 8.8 Hz, 2H), 7.75 (s, 1H), 7.50 - 7.38 (m, 3H), 7.23 - 7.19 (m, 2H).

Synthesis of 3-((4-(pyrimidin-5-yl)phenyl)amino)benzoic acid

Step 1: To a solution of methyl 3-bromobenzoate (2.0 g, 9.32 mmol) in 1,4-dioxane (43 mL) was 4-(pyrimidin-5-yl)aniline (1.45 g, 8.47 mmol), XPhos (0.69 g, 1.44 mmol), and cesium carbonate (5.52 g, 16.94 mmol). Pd 2 (dba) 3 (0.62 g, 0.68 mmol) was added into the solution. The solution was stirred for 16 hours at 100°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using acetone to give methyl 3-((4-(pyrimidin-5-yl)phenyl)amino)benzoate (0.93 g, 36%) as a brown solid.

¾ NMR (400 MHz, DMSO-d 6 ) S 9.12 (s, 3H), 8.75 (s, 1H), 7.79 - 7.72 (m, 3H), 7.50 - 7.36 (m, 3H), 7.25 - 7.22 (m, 2H), 3.85 (s, 3H). Step 2: Methyl 3-((4-(pyrimidin-5-yl)phenyl)amino)benzoate (0.90 g, 2.95 mmol) and LiOH-H 2 0 (0.5 g, 20.7 mmol) were mixed in THF/H2O (20/10 mL) and stirred for 64 hours at room temperature. The reaction mixture was extracted by EA and aq. HC1 (IN). The organic layer was dried over anhydrous MgSCL and concentrated. The residue was purified by MPLC to give 3-((4-(pyrimidin-5-yl)phenyl)amino)benzoic acid (706 mg, 82%) as a yellow solid.

¾ NMR (400 MHz, DMSO-d 6 ) d 12.91 (bs, 1H), 9.12 - 9.11 (m, 3H), 8.70 (s, 1H), 7.79 - 7.70 (m, 3H), 7.49 - 7.33 (m, 3H), 7.26 - 7.20 (m, 2H).

Synthesis of 3-((4-(pyrimidin-4-yl)phenyl)amino)benzoic acid

Step 1: To a solution of methyl 3-bromobenzoate (2.14 g, 9.93 mmol) in 1,4-dioxane (15 mL) was 4-(pyrimidin-4-yl)aniline (1.70 g, 9.93 mmol), BrettPhos (1.07 g, 1.99 mmol), and cesium carbonate (8.09 g, 24.8 mmol). Pd 2 (dba) 3 (0.91 g, 0.99 mmol) was added into the solution. The solution was stirred for 12 hours at 100°C under N2 atmosphere. TLC indicated 4-(pyrimidin-4-yl)aniline was consumed completely and one new spot formed. The reaction was clean according to TLC. The reaction mixture was diluted with H2O and extracted with EA. The combined organic layers were washed with brine, dried over Na 2 SC> 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography to methyl 3-((4-(pyrimidin-4-yl)phenyl)amino)benzoate (1.30 g, 43%) as a yellow solid.

¾ NMR (400 MHz, DMSO-d 6 ) d 9.13 (s, 1H), 8.91 (s, 1H), 8.74 (d, J= 5.20 Hz, 1H), 8.15 (d, J= 8.80 Hz, 2H), 7.96 (d, J= 5.20 Hz, 1H), 7.76 (s, 1H), 7.52 - 7.49 (m, 1H), 7.46 - 7.43 (m, 2H), 7.20 (d, J= 8.80 Hz, 2H), 3.85 (s, 3H).

Step 2: Methyl 3-((4-(pyrimidin-4-yl)phenyl)amino)benzoate (1.30 g, 4.26 mmol) and KOH (478 mg, 8.52 mmol) were mixed in EtOH/H 2 0 (7/5 mL) and stirred for 4 hours at 100°C. TLC indicated methyl 3-((4-(pyrimidin-4-yl)phenyl)amino)benzoate was consumed completely and one new spot formed. The reaction was clean according to TLC. The reaction mixture was diluted with H2O and extracted with 2-methyltetrahydrofuran and the pH was adjusted to 5-6 with 0.5 M HC1 for aqueous phase. The resulting solution was extracted with 2- methyltetrahydrofuran. The combined organic layers were washed with brine, dried over Na 2 SC> 4 , filtered, and concentrated under reduced pressure to give a residue. The crude product was triturated with acetonitrile for 12 hours at room temperature. 3-((4-(Pyrimidin-4- yl)phenyl)amino)benzoic acid (1.01 g, 97%) as a yellow solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 12.96 (bs, 1H), 9.13 (s, 1H), 8.88 (s, 1H), 8.74 (d, 7= 5.60 Hz, 1H), 8.15 (d, 7= 8.80 Hz, 2H), 7.97 - 7.94 (m, 1H), 7.75 (s, 1H), 7.52 - 7.48 (m, 1H), 7.43 - 7.41 (m, 2H), 7.20 (d, 7= 8.80 Hz, 2H).

Synthesis of 2-((5-phenylpyridin-2-yl)amino)isonicotinic acid

Step 1: 5-Phenylpyridin-2-amine (350 mg, 2.1 mmol), methyl 2-bromoisonicotinate (620 mg, 2.47 mmol), Pd2(dba)3 (188 mg, 0.21 mmol), BrettPhos (221 mg, 0.41 mmol), and cesium carbonate (1.3 g, 4.1 mmol) were mixed in 1,4-dioxane (10 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by EA and brine. The crude mixture was solidified by using EA and HEX to give methyl 2-((5- phenylpyridin-2-yl)amino)isonicotinate(393 mg, 63%) as an orange solid.

Ή NMR (400 MHz, DMSO-7/) d 10.16 (s, 1H), 8.62 (d, 7= 2.5 Hz, 1H), 8.43 (d, 7=5.1 Hz, 1H), 8.33 (s, 1H), 8.04 (dd, 7=2.5, 8.8 Hz, 1H), 7.85 - 7.82 (m, 1H), 7.71 (d, 7=7.3 Hz, 2H), 7.48 (t, 7=7.7 Hz, 2H), 7.39 - 7.30 (m, 2H), 3.91 (s, 3H).

Step 2: Methyl 2-((5-phenylpyridin-2-yl)amino)isonicotinate (350 mg, 1.15 mmol) and LiOH-HiO (481 mg, 11.5 mmol) were mixed in H 2 0/l,4-dioxane (4.8/23 mL) and stirred for 18 hours at room temperature. Then pH value of the solution was adjusted to 1-2 by 1 N HC1. The yellow solid was precipitated out of the solution, and the solution was filtered to give 2- ((5-phenylpyridin-2-yl)amino)isonicotinic acid (190 mg, 57%) as a yellow solid.

Ή NMR (400 MHz, DMSO-7/) d 13.90 (s, 1H), 11.70 (s, 1H), 8.65 (d, 7=2.4 Hz, 1H), 8.51 (d, 7=5.4 Hz, 1H), 8.34 (d, 7=8.0 Hz, 1H), 8.06 (s, 1H), 7.75 - 7.71 (m, 3H), 7.56 - 7.49 (m, 3H), 7.43 (t, 7=7.4 Hz, 1H).

Synthesis of 2-((5-phenylpyrimidin-2-yl)amino)isonicotinic acid Step 1: 5-Phenylpyrimidin-2-amine (500 mg, 2.9 mmol), methyl 2-bromoisonicotinate (620 mg, 2.47 mmol), Pd2(dba)3 (267 mg, 0.29 mmol), BrettPhos (313 mg, 0.58 mmol), and cesium carbonate (1.9 g, 5.8 mmol) were mixed in 1,4-dioxane (15 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by DCM and brine. The crude mixture was solidified by using EA to give methyl 2-((5-phenylpyrimidin-2- yl)amino)isonicotinate (609 mg, 68%) as an yellow solid.

'H NMR (400 MHz, DMSO-7/) d 10.42 (s, 1H), 8.98 (s, 2H), 8.84 (s, 1H), 8.50 (d, ,7=5.0 Hz, 1H), 7.79 (d, ,7=7.4 Hz, 2H), 7.51 (dd, 7=7.7, 7.7 Hz, 2H), 7.46 - 7.39 (m, 2H), 3.93 (s, 3H).

Step 2: Methyl 2-((5-phenylpyrimidin-2-yl)amino)isonicotinate (550 mg, 1.8 mmol) and LiOH-HiO (753 mg, 18 mmol) were mixed in H 2 0/l,4-dioxane (7.5/36 mL) and stirred for 18 hours at room temperature. Then pH value of the solution was adjusted to 3 by 1 N HC1.

The reaction mixture was extracted by EA and brine. The crude mixture was solidified by using EA and HEX to give 2-((5-phenylpyrimidin-2-yl)amino)isonicotinic acid (153 mg, 29%) as a beige solid.

'H NMR (400 MHz, DMSO-Tf) d 13.60 (s, 1H), 10.34 (s, 1H), 8.97 (s, 2H), 8.84 (s,

1H), 8.47 (d, 7=5.0 Hz, 1H), 7.80 - 7.77 (m, 2H), 7.51 (dd, 7=7.6, 7.6 Hz, 2H), 7.44 - 7.38 (m, 2H).

Synthesis of 5-((5-phenylpyrimidin-2-yl)amino)nicotinic acid

Step 1: 5-Phenylpyrimidin-2-amine (500 mg, 2.9 mmol), methyl 5-bromonicotinate (757 mg, 3.5 mmol), Pd2(dba)3 (267 mg, 0.29 mmol), BrettPhos (313 mg, 0.58 mmol), and cesium carbonate (1.9 g, 5.8 mmol) were mixed in 1,4-dioxane (15 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by EA and brine. The grey solid was precipitated out of the solution, and the solution was filtered to give methyl 5-((5-phenylpyrimidin-2-yl)amino)nicotinate (630 mg, 70%) as a grey solid.

Ή NMR (400 MHz, DMSO-7/) d 10.28 (s, 1H), 9.16 (d, 7=2.6 Hz, 1H), 8.95 (s, 2H), 8.87 (dd, 7=2.3, 2.3 Hz, 1H), 8.70 (d, 7=1.9 Hz, 1H), 7.76 (d, 7=7.3 Hz, 2H), 7.50 (dd, 7=7.6, 7.6 Hz, 2H), 7.40 (t, 7=7.4 Hz, 1H), 3.91 (s, 3H). Step 2: Methyl 5-((5-phenylpyrimidin-2-yl)amino)nicotinate (620 mg, 2 mmol) and Li0H-H 2 0 (849 mg, 20 mmol) were mixed in H 2 0/l,4-dioxane (8.4/40 mL) and stirred for 18 hours at room temperature. Then pH value of the solution was adjusted to 2 by 1 N HC1. The grey solid was precipitated out of the solution, and the solution was filtered to give 5-((5- phenylpyrimidin-2-yl)amino)nicotinic acid (497 mg, 84%) as a grey solid.

Ή NMR (400 MHz, DMSO-d 6 ) d 10.22 (s, 1H), 9.11 (d, 7= 2.6 Hz, 1H), 8.94 (s, 2H), 8.83 (dd, 7=2.1, 2.1 Hz, 1H), 8.67 (d, 7=1.8 Hz, 1H), 7.76 (d, 7=7.4 Hz, 2H), 7.50 (dd, 7=7.6, 7.6 Hz, 2H), 7.40 (t, 7=7.4 Hz, 1H).

Synthesis of 4-((5-phenylpyrimidin-2-yl)amino)picolinic acid

Step 1: 5-Phenylpyrimidin-2-amine (500 mg, 2.9 mmol), methyl 4-bromopicolinate (757 mg, 3.5 mmol), Pd2(dba)3 (267 mg, 0.29 mmol), BrettPhos (313 mg, 0.58 mmol), and cesium carbonate (1.9 g, 5.8 mmol) were mixed in 1,4-dioxane (15 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by EA and brine. The beige solid was precipitated out of the solution, and the solution was filtered to give methyl 4-((5-phenylpyrimidin-2-yl)amino)picolinate (417 mg, 47%) as a beige solid.

Ή NMR (400 MHz, DMSO-7/) d 10.54 (s, 1H), 9.00 (s, 2H), 8.54 (d, 7=2.3 Hz, 1H), 8.51 (d, 7=5.6 Hz, 1H), 8.06 (dd, 7=2.3, 5.6 Hz, 1H), 7.78 (d, 7=7.4 Hz, 2H), 7.51 (dd, 7=7.6, 7.6 Hz, 2H), 7.42 (t, 7=7.3 Hz, 1H), 3.89 (s, 3H).

Step 2: Methyl 4-((5-phenylpyrimidin-2-yl)amino)picolinate (400 mg, 1.3 mmol) and LiOH-H 2 0 (548 mg, 13 mmol) were mixed in H 2 0/l,4-dioxane (5.4/26 mL) and stirred for 18 hours at room temperature. Then pH value of the solution was adjusted to 1 by 1 N HC1. The beige solid was precipitated out of the solution, and the solution was filtered to give 4-((5- phenylpyrimidin-2-yl)amino)picolinic acid (346 mg, 92%) as a beige solid.

Ή NMR (400 MHz, DMSO-7/) d 10.78 (s, 1H), 9.04 - 9.03 (m, 2H), 8.56 (d, 7=2.3 Hz, 1H), 8.47 (d, 7=5.8 Hz, 1H), 8.09 (dd, 7=2.3, 6.0 Hz, 1H), 7.80 (d, 7=7.3 Hz, 2H), 7.52 (t, 7=7.6 Hz, 2H), 7.43 (t, 7=7.3 Hz, 1H).

Synthesis of 3-((5-phenyl-l,3,4-oxadiazol-2-yl)amino)benzoic acid Step 1: To a solution of methyl 3-bromobenzoate (0.95 g, 4.4 mmol) in 1,4-dioxane (8 mL) was added 5-phenyl-l,3,4-oxadiazol-2-amine (0.65 g, 4.0 mmol), /-BuXPhos (0.29 g, 0.68 mmol), and /-BuONa (0.77 g, 8.0 mmol). Pd 2 (dba) 3 (0.29 g, 0.32 mmol) was added into the solution. The solution was stirred for 16 hours at 100°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using acetone to give methyl 3-((5-phenyl-l,3,4-oxadiazol-2-yl)amino)benzoate (0.33 g, 23%) as a beige solid.

¾ NMR (400 MHz, DMSO-7 6 ) d 10.99 (bs, 1H), 8.33 (dd, 7=1.8, 1.8 Hz, 1H), 7.94 - 7.90 (m, 2H), 7.89 - 7.85 (m, 1H), 7.68 - 7.58 (m, 4H), 7.54 (dd, 7=7.9, 7.9 Hz, 1H), 3.89 (s, 3H).

Step 2: Methyl 3-((5-phenyl-l,3,4-oxadiazol-2-yl)amino)benzoate (0.32 g, 1.08 mmol) and LiOH-HiO (0.18 g, 4.32 mmol) were mixed in THF/H2O (7.2/3.6 mL) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and aq. HC1 (IN). The organic layer was dried over anhydrous MgSCL and concentrated to give crude 3-((5- phenyl-l,3,4-oxadiazol-2-yl)amino)benzoic acid (125 mg, 41%) as a pale brown solid.

¾NMR (400 MHz, DMSO-7 6 ) d 10.95 (s, 1H), 8.29 (s, 1H), 7.97 - 7.88 (m, 2H), 7.85 (dd, 7=1.4, 8.0 Hz, 1H), 7.67 - 7.47 (m, 5H).

2) Substitution A

Synthesis of (1s,4s)-4-((6-phenylpyridazin-3-yl)amino)bicyclo[2.2.1]hepta ne-l- carboxylic acid

Step 1: In a sealed tube, 3-chloro-6-phenylpyridazine (500 mg, 2.6 mmol) and methyl (H,47)-4-aminobicyclo[2.2.1]heptane-l-carboxylate (578 mg, 3.4 mmol) were mixed in n- butanol (10 mL). To this reaction mixture, trifluoroacetic acid (75 mg, 0.65 mmol) was added at room temperature and allowed to stir for 72 hours at 150°C. Progress of the reaction was monitored by TLC. Reaction was cooled to r.t., water was added, and product was extracted with EA. The combined organic layer was washed with water and brine, dried over anhydrous Na 2 SC> 4, and concentrated under vacuum to provide crude product, which was purified by combi-flash column chromatography. Product was eluted out in 15% EA in HEX to provide methyl (n,4s)-4-((6-phenylpyridazin-3-yl)amino)bicyclo[2.2. l]heptane-l-carboxylate (170 mg, 20%) as a white solid m/z 324.

Ή NMR (400 MHz, methanol-7 4 ) d 7.88 (d, 7= 7.0 Hz, 1H), 7.73 (d, 7= 9.5 Hz, 1H),

7.55 - 7.38 (m, 3H), 6.96 (d, 7= 9.5 Hz, 1H), 3.71 (s, 3H), 2.34 - 2.08 (m, 6H), 2.03 - 1.95 (m, 2H), 1.87 - 1.74 (m, 2H).

Step 2: Methyl (ls,4s)-4-((6-phenylpyridazin-3-yl)amino)bicyclo[2.2.1]hepta ne-l- carboxylate (1.4 g, 4.3 mmol) was dissolved in tetrahydrofuran: H2O (2:1, 15mL) and lithium hydroxide (541 mg, 12.9 mmol) was added at 0°C and reaction was allowed to stir for 6 hours at room temperature. Progress of the reaction was monitored by TLC. After completion of reaction, 2N HC1 solution was added untill pH 4 adjusted and precipitates were filtered and dried to provide (ls,4s)-4-((6-phenylpyridazin-3-yl)amino)bicyclo[2.2.1]hepta ne-l-carboxylic acid (1.05 g, 78%) as an off white solid..

Ή NMR (400 MHz, DMSO-Tf) d 12.16 (s, 1H), 7.99 (d, 7=8.1 Hz, 2H), 7.87 (d, 7=9.6 Hz, 1H), 7.56 - 7.38 (m, 4H), 7.01 (d, 7=8.8 Hz, 1H), 2.21 - 1.97 (m, 6H), 1.90 - 1.79 (m, 2H), 1.76 - 1.63 (m, 2H).

3) Substitution B

Synthesis of 3-((6-phenylpyridazin-3-yl)amino)adamantane-l-carboxylic acid

Step 1: To a solution of 3 -aminoadamantane-1 -carboxylic acid hydrochloride (20 g, 86 mmol) in EtOH (140 mL) was added SOCE (10.3 g, 86.3 mmol) at room temperature. The reaction mixture was stirred for 4 hours at 80°C. Liquid chromatography-mass spectrometry (LCMS) showed the reaction was completed. The reaction mixture was concentrated under reduced pressure to give a residue. Petroleum ether was then added, and the mixture was once again concentrated under reduced pressure at which point a solid began to precipitate, the process was repeated three more times. The crude product was triturated with Petroleum ether for 30 minutes at room temperature and the suspension was filtered to give ethyl 3- aminoadamantane-l-carboxylate hydrochloride (21 g, 94%) as a white solid m z 224.

Ή NMR (400 MHz, DMSO-d 6 ) d 8.28 (s, 3H), 4.06 (q, J= 7.2 Hz, 2H), 2.18 (s, 2H), 1.90 (s, 2H), 1.77 (s, 6H), 1.67 - 1.55 (m, 4H), 1.17 (t, J= 7.2 Hz, 3H).

Step 2: To a solution of 3,6-dichloropyridazine (24 g, 162 mmol) in DMF (147 mL) was added ethyl 3-aminoadamantane-l-carboxylate hydrochloride (21.0 g, 80.8 mmol) and K 2 CO 3 (33.5 g, 243 mmol) at room temperature. The reaction mixture was stirred for 12 hours at 135°C. TLC showed the -50% of 3,6-dichloropyridazine remained and -20% of product was detected. The residue was diluted with water and extracted with EA. The combined organic layers were washed with brine, dried overNa 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography to give ethyl 3-((6-chloropyridazin-3-yl)amino)adamantane-l-carboxylate (1.80 g, 7%) as a white solid.

'HNMR (400 MHz, CDCh) d 7.11 (d, J= 9.2 Hz, 1H), 6.59 (d, J= 9.2 Hz, 1H), 4.44 (s, 1H), 4.14 - 3.93 (m, 2H), 2.25 (s, 4H), 2.18 - 2.08 (m, 4H), 1.92 - 1.88 (m, 4H), 1.70 - 1.68 (m, 2H), 1.26 - 1.22 (m, 3H).

Step 3: To a solution of ethyl 3-((6-chloropyridazin-3-yl)amino)adamantane-l- carboxylate (1.8 g, 5.4 mmol) in dimethyl ether (DME) (9 mL) and H2O (1.8 mL) was added phenylboronic acid (719 mg, 5.9 mmol) and Na 2 CC> 3 (2.84 g, 26.8 mmol) at room temperature. Pd(PPh3)2Cl2 (376 mg, 0.54 mmol) was added into above mixture at room temperature. The suspension was degassed under vacuum and purged with N2 three times, and the reaction mixture was stirred for 12 hours at 80°C. TLC showed the reaction was completed. The residue was diluted with H2O and extracted with EA. The combined organic layers were washed with brine, dried over Na 2 S0 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography to give product. The residue was purified by preparative HPLC(prep-HPLC) to give desired compound. Ethyl 3-((6-phenylpyridazin-3- yl)amino)adamantane-l-carboxylate (800 mg, 40%) was obtained as a white solid.

'HNMR (400 MHz, CDC1 3 ) d 7.97 (d, J= 7.2 Hz, 2H), 7.56 (d, J= 9.6 Hz, 1H), 7.48 - 7.44 (m 2H), 7.42 - 7.40 (m 1H), 6.70 (d, J= 9.2 Hz, 1H), 4.50 (bs, 1H), 4.15 - 4.09 (m, 2H), 2.33 (s, 2H), 2.28 (s, 2H), 2.19 (s, 3H), 1.90 (q, J= 12 Hz, 3H), 1.75 - 1.70 (m, 2H), 1.59 (s, 2H), 1.29 - 1.23 (m, 3H).

Step 4: To a solution of ethyl 3-((6-phenylpyridazin-3-yl)amino)adamantane-l- carboxylate (800 mg, 2.12 mmol) in EtOH (3.2 mL) was added H2O (1.6 mL) and LiOH-H 2 0 (445 mg, 10.6 mmol) at room temperature. The reaction mixture was stirred for 12 hours at 40 ~ 45°C. LCMS showed the reaction was completed. The reaction mixture was concentrated under reduced pressure to remove EtOH. The mixture was adjusted to pH 5 ~ 6 with citric acid solution and white solid was precipitated, the suspension was filtered and the filter cake was concentrated under reduced pressure to give 3-((6-phenylpyridazin-3-yl)amino)adamantane-l- carboxylic acid (450 mg, 60%) as a white solid m/z 350.

Ή NMR (400 MHz, DMSO-^) 5 12.11 (s, 1H), 7.96 (d, J= 7.2 Hz, 2H), 7.75 (d, J=

9.2 Hz, 1H), 7.48 - 7.44 (m, 2H), 7.40 - 7.38 (m, 1H), 6.90 (d, J= 9.6 Hz, 1H), 6.60 (s, 1H), 2.26 (s, 2H), 2.18 - 2.16 (m, 4H), 2.05 - 2.03 (m, 2H), 1.82 - 1.79 (m, 4H), 1.66 - 1.63 (m, 2H).

EXAMPLE 2

SYNTHESIS OF COMPOUNDS

1. Synthesis by Method A Synthesis of Compound 1

5-Phenylpyrazin-2-amine (10 mg, 0.058 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (14.5 mg, 0.049 mmol), Pd2(dba)3 (0.9 mg, 0.00098 mmol), BrettPhos (5.3 mg, 0.0098 mmol), and cesium carbonate (32 mg, 0.098 mmol) were mixed in 1,4-dioxane (0.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was purified by MPLC to give compound 1, A-[(5-methylfuran-2- yl)methyl]-3-[(5-phenylpyrazin-2-yl)amino]benzamide (13 mg, 69%) as a yellow solid.

Synthesis of Compound 2

5-Phenylpyrimidin-2-amine (10 mg, 0.058 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (14.5 mg, 0.049 mmol), Pd2(dba)3 (0.9 mg, 0.00098 mmol), BrettPhos (5.3 mg, 0.0098 mmol), and cesium carbonate (32 mg, 0.098 mmol) were mixed in 1,4-dioxane (0.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The residue was purified by MPLC to give compound 2, A-[(5-methylfuran-2- yl)methyl]-3-[(5-phenylpyrimidin-2-yl)amino]benzamide (14 mg, 74%) as a white solid.

Synthesis of Compound 3

[l,l’-Biphenyl]-4-amine (20 mg, 0.12 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (29 mg, 0.098 mmol), Pd2(dba)3 (1.8 mg, 0.002 mmol), BrettPhos (10.6 mg, 0.020 mmol), and cesium carbonate (64 mg, 0.2 mmol) were mixed in 1,4-dioxane (1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The residue was purified by MPLC to give compound 3, 3-({[l,l’-biphenyl]-4-yl}amino)-/V-[(5- methylfuran-2-yl)methyl]benzamide (36.5 mg, 97%) as a yellowish white solid.

Synthesis of Compound 4

5-Phenylpyridin-2-amine (20 mg, 0.12 mmol), 3-bromo-A-((5-methylfuran-2- yl)methyl)benzamide (29 mg, 0.098 mmol), Pd2(dba)3 (1.8 mg, 0.002 mmol), BrettPhos (10.6 mg, 0.020 mmol), and cesium carbonate (64 mg, 0.2 mmol) were mixed in 1,4-dioxane (1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by MPLC to give compound 4, /V-[(5-methylfuran-2-yl)methyl]-3-[(5- phenylpyridin-2-yl)amino]benzamide (24 mg, 64%) as a yellowish white solid.

Synthesis of Compound 5

Pyridazin-3 -amine (15 mg, 0.16 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (39 mg, 0.13 mmol), Pd2(dba)3 (2.4 mg, 0.0026 mmol), BrettPhos (14 mg, 0.026 mmol), and cesium carbonate (86 mg, 0.26 mmol) were mixed in 1,4-dioxane (0.7 mL) and heated in a microwave reactor for 60 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 5, A-[(5-methylfuran-2-yl)methyl]-3- [(pyridazin-3-yl)amino]benzamide (17 mg, 42%) as a beige solid.

Synthesis of Compound 6 Step 1: Phenylboronic acid (500 mg, 4.1 mmol), 6-bromopyri din-3 -amine (591 mg,

3.42 mmol), Pd(PPli3)4 (197 mg, 0.17 mmol), and potassium carbonate (1.7 g, 12.6 mmol) were mixed in H 2 O/DMF (7/7 mL) and heated in a microwave reactor for 60 minutes at 100°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The reaction mixture was concentrated and purified by MPLC to give 6-phenylpyri din-3 -amine (276 mg, 47%) as a yellow solid.

Step 2: 6-Phenylpyri din-3 -amine (20 mg, 0.12 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (29 mg, 0.098 mmol), Pd 2 (dba) 3 (1.8 mg, 0.002 mmol), BrettPhos (10.5 mg, 0.02 mmol), and cesium carbonate (64 mg, 0.2 mmol) were mixed in 1,4-dioxane (0.8 mL) and heated in a microwave reactor for 60 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 6, /V-[(5-methylfuran-2-yl)methyl]-3- [(6-phenylpyridin-3-yl)amino]benzamide (34 mg, 91%) as a yellowish white solid.

Synthesis of Compound 7

Step 1: Furan-3-ylboronic acid (250 mg, 2.23 mmol), 6-bromopyridazin-3 -amine (324 mg, 1.86 mmol), Pd(PPh3)4 (108 mg, 0.093 mmol), and potassium carbonate (982 mg, 6.9 mmol) were mixed in H 2 0/l,4-dioxane (1.6/6.2 mL) and heated in a microwave reactor for 60 minutes at 100°C. The reaction mixture was concentrated and purified by MPLC to give 6- (furan-3-yl)pyridazin-3 -amine (265 mg, 88%) as a yellow solid.

Step 2: 6-(Furan-3-yl)pyridazin-3 -amine (20 mg, 0.12 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (30 mg, 0.103 mmol), Pd2(dba)3 (1.9 mg, 0.002 mmol), BrettPhos (11 mg, 0.02 mmol), and cesium carbonate (67 mg, 0.21 mmol) were mixed in 1,4- dioxane (0.5 mL) and heated in a microwave reactor for 60 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 7, 3-{[6-(furan-3- yl)pyridazin-3-yl]amino}-/V-[(5-methylfuran-2-yl)methyl]benz amide (13 mg, 34%) as a yellowish white solid.

Synthesis of Compound 8

Step 1: Furan-2-ylboronic acid (250 mg, 2.23 mmol), 6-bromopyridazin-3 -amine (324 mg, 1.86 mmol), Pd(PPli3)4 (108 mg, 0.093 mmol), and potassium carbonate (952 mg, 6.9 mmol) were mixed in H 2 0/l,4-dioxane (1.6/6.2 mL) and heated in a microwave reactor for 60 minutes at 100°C. The reaction mixture was concentrated and purified by MPLC to give 6- (furan-2-yl)pyridazin-3 -amine (216 mg, 72%) as a yellow solid.

Step 2: 6-(Furan-2-yl)pyridazin-3 -amine (20 mg, 0.12 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (30 mg, 0.103 mmol), Pd2(dba)3 (1.9 mg, 0.002 mmol), BrettPhos (11 mg, 0.02 mmol), and cesium carbonate (67 mg, 0.21 mmol) were mixed in 1,4- dioxane (0.5 mL) and heated in a microwave reactor for 60 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 8, 3-{[6-(furan-2- yl)pyridazin-3-yl]amino}-/V-[(5-methylfuran-2-yl)methyl]benz amide (17 mg, 45%) as a yellowish white solid.

Synthesis of Compound 9

Step 1: Pyridin-4-ylboronic acid (600 mg, 4.9 mmol), 6-bromopyridazin-3 -amine (354 mg, 2.03 mmol), Pd(PPh3)4 (227 mg, 0.2 mmol), and potassium carbonate (1 g, 7.5 mmol) were mixed in H 2 0/l,4-dioxane (1.7/6.8 mL) and heated in a microwave reactor for 90 minutes at 150°C. The reaction mixture was concentrated and purified by MPLC to give 6-(pyridin-4- yl)pyridazin-3 -amine (63 mg, 18%) as a yellowish white solid. Step 2: 6-(Pyridin-4-yl)pyridazin-3-amine (20 mg, 0.12 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (29 mg, 0.097 mmol), Pd2(dba)3 (1.8 mg, 0.0019 mmol), BrettPhos (10.4 mg, 0.019 mmol), and cesium carbonate (63 mg, 0.19 mmol) were mixed in 1,4-dioxane (0.5 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 9, A-[(5-methylfuran-2- yl)methyl]-3-{[6-(pyridin-4-yl)pyridazin-3-yl]amino}benzamid e (18 mg, 48%) as an orange solid.

Synthesis of Compound 10

Step 1: Pyridin-3-ylboronic acid (250 mg, 2.03 mmol), 6-bromopyridazin-3 -amine (295 mg, 1.7 mmol), Pd(PPh3)4 (98 mg, 0.085 mmol), and potassium carbonate (867 mg, 6.3 mmol) were mixed in H 2 0/l,4-dioxane (1.4/5.6 mL) and heated in a microwave reactor for 60 minutes at 100°C. The reaction mixture was concentrated and purified by MPLC to give 6-(pyridin-3- yl)pyridazin-3 -amine (65 mg, 22%) as a yellowish white solid.

Step 2: 6-(Pyri din-3 -yl)pyridazin-3 -amine (20 mg, 0.12 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (29 mg, 0.097 mmol), Pd2(dba)3 (1.8 mg, 0.0019 mmol), BrettPhos (10.4 mg, 0.019 mmol), and cesium carbonate (63 mg, 0.19 mmol) were mixed in 1,4-dioxane (0.5 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 10, /V-[(5-methylfuran-2-yl)methyl]-3-{[6-(pyridin-3-yl)pyridazi n-3- yl]amino}benzamide (15 mg, 40%) as a pink solid.

Synthesis of Compound 11

Step 1: Phenylboronic acid (250 mg, 2.05 mmol), 2-bromopyrimidin-5-amine (297 mg, 1.71 mmol), Pd(PPh3)4 (99 mg, 0.085 mmol), and potassium carbonate (874 mg, 6.3 mmol) were mixed in H 2 0/l,4-dioxane (1.4/5.7 mL) and heated in a microwave reactor for 60 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give 2- phenylpyrimidin-5-amine (100 mg, 34%) as a beige solid.

Step 2: 2-Phenylpyrimidin-5-amine (20 mg, 0.12 mmol), 3-bromo-A-((5-methylfuran- 2-yl)methyl)benzamide (29 mg, 0.098 mmol), Pd2(dba)3 (1.8 mg, 0.002 mmol), BrettPhos (10.5 mg, 0.02 mmol), and cesium carbonate (64 mg, 0.2 mmol) were mixed in 1,4-dioxane (1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 11, /V-[(5-methylfuran-2-yl)methyl]-3-[(2-phenylpyrimidin-5- yl)amino]benzamide (12 mg, 32%) as a beige solid.

Synthesis of Compound 12

Step 1: Phenylboronic acid (300 mg, 2.5 mmol), 6-bromo-l,2,4-triazin-3-amine (359 mg, 2.05 mmol), Pd(PPh3)4 (119 mg, 0.103 mmol), and potassium carbonate (1 g, 7.59 mmol) were mixed in H 2 0/l,4-dioxane (1.7/6.8 mL) and heated in a microwave reactor for 60 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give 6-phenyl- l,2,4-triazin-3-amine (269 mg, 76%) as a yellowish white solid. Step 2: 6-Phenyl- 1, 2, 4-triazin-3 -amine (20 mg, 0.12 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (29 mg, 0.098 mmol), Pd2(dba)3 (1.8 mg, 0.002 mmol), BrettPhos (10.5 mg, 0.02 mmol), and cesium carbonate (64 mg, 0.2 mmol) were mixed in 1,4- dioxane (0.8 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 12, /V-[(5-methylfuran-2-yl)methyl]-3-[(6-phenyl-l,2,4-triazin-3 - yl)amino]benzamide (12 mg, 32%) as a yellow solid.

Synthesis of Compound 13

Step 1: (4-Methoxyphenyl)boronic acid (200 mg, 1.3 mmol), 6-bromopyridazin-3- amine (191 mg, 1.1 mmol), Pd(PPh3)4 (63 mg, 0.06 mmol), and potassium carbonate (561 mg, 4.06 mmol) were mixed in H 2 0/l,4-dioxane (0.9/3.7 mL) and heated in a microwave reactor for 60 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give 6-(4-methoxyphenyl)pyridazin-3-amine (189 mg, 86%) as a white solid.

Step 2: 6-(4-Methoxyphenyl)pyridazin-3-amine (20 mg, 0.1 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (24 mg, 0.08 mmol), Pd2(dba)3 (1.5 mg, 0.0017 mmol), BrettPhos (8.9 mg, 0.017 mmol), and cesium carbonate (54 mg, 0.17 mmol) were mixed in 1,4- dioxane (0.4 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 13, 3-{[6-(4-methoxyphenyl)pyridazin-3-yl]amino}-/V-[(5-methylfu ran- 2-yl)methyl]benzamide (16 mg, 47%) as a white solid.

Synthesis of Compound 15

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and 3-morpholinopropan-l- amine (0.11 mL, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 25 hours at room temperature. The reaction mixture was concentrated and purified by MPLC. And the mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated to give 3-bromo- A-(3-morpholi nopropyl )benzamide (338 mg, >99%) as a brownish oil.

Step 2: 6-Phenylpyridazin-3 -amine (20 mg, 0.12 mmol), 3-bromo-/V-(3- morpholinopropyl)benzamide (32 mg, 0.097 mmol), Pd2(dba)3 (8.9 mg, 0.0097 mmol), BrettPhos (10.5 mg, 0.019 mmol), and cesium carbonate (63 mg, 0.19 mmol) were mixed in 1,4-dioxane (0.5 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 15, /V-[3-(morpholin-4-yl)propyl]-3-[(6-phenylpyridazin-3- yl)amino]benzamide (9 mg, 23%) as a beige solid.

Synthesis of Compound 16

Step 1: (3,4-Dichlorophenyl)boronic acid (200 mg, 1.05 mmol), 6-bromopyridazin-3- amine (152 mg, 0.87 mmol), Pd(PPli3)4 (51 mg, 0.04 mmol), and potassium carbonate (447 mg, 3.23 mmol) were mixed in H 2 0/l,4-dioxane (0.7/2.9 mL) and heated in a microwave reactor for 60 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give 6-(3,4-di chi orophenyl)pyridazin-3 -amine (62 mg, 29%) as a yellowish white solid. Step 2: 6-(3,4-Dichlorophenyl)pyridazin-3 -amine (20 mg, 0.083 mmol), 3-bromo-A- ((5-methylfuran-2-yl)methyl)benzamide (20 mg, 0.07 mmol), Pd2(dba)3 (6.4 mg, 0.0069 mmol), BrettPhos (7.5 mg, 0.014 mmol), and cesium carbonate (45 mg, 0.14 mmol) were mixed in 1,4-dioxane (0.35 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 16, 3-{[6-(3,4-dichlorophenyl)pyridazin-3- yl]amino}-/V-[(5-methylfuran-2-yl)methyl]benzamide (3 mg, 10%) as a beige solid.

Synthesis of Compound 18

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and pyridin-4-ylmethanamine (0.08 mL, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.6 mmol) and stirred for 28 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NELCl. The organic layer was dried over anhydrous NaiSCL and concentrated to give 3 -bromo-A-(pyridin-4-yl methyl )benzamide (246 mg, >99%) as a brown oil.

Step 2: 6-Phenylpyridazin-3 -amine (30 mg, 0.18 mmol), 3-bromo-/V-(pyridin-4- ylmethyl)benzamide (46 mg, 0.16 mmol), Pd2(dba)3 (14.6 mg, 0.016 mmol), BrettPhos (17 mg, 0.032 mmol), and cesium carbonate (104 mg, 0.32 mmol) were mixed in 1,4-dioxane (1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by MeOH/DCM (10: 1) and ELO. The crude mixture was solidified by using DCM to give compound 18, 3-[(6-phenylpyridazin-3-yl)amino]-/V-[(pyridin-4-yl)methyl]b enzamide (17 mg, 28%) as a yellow solid.

Synthesis of Compound 19

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and pyridin-2-ylmethanamine (0.08 mL, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 28 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NELCl. The organic layer was dried over anhydrous Na2SCL and concentrated to give 3 -bromo-A-(pyri din-2-yl methyl )benzamide (268 mg, >99%) as a brown oil.

Step 2: 6-Phenylpyridazin-3 -amine (30 mg, 0.18 mmol), 3-bromo-/V-(pyridin-2- ylmethyl)benzamide (46 mg, 0.16 mmol), Pd2(dba)3 (14.6 mg, 0.016 mmol), BrettPhos (17 mg, 0.032 mmol), and cesium carbonate (104 mg, 0.32 mmol) were mixed in 1,4-dioxane (1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 19, 3-[(6-phenylpyridazin-3-yl)amino]- A-[(pyridin-2-yl)methyl]benzamide (39 mg, 65%) as a yellowish white solid.

Synthesis of Compound 20

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and pyridin-3-ylmethanamine (0.08 mL, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 26 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NELCl. The organic layer was dried over anhydrous Na2SCL and concentrated to give 3 -bromo-A-(pyridin-3-yl methyl )benzamide (268 mg, >99%) as a brown oil. Step 2: 6-Phenylpyridazin-3 -amine (30 mg, 0.18 mmol), 3-bromo-/V-(pyridin-3- ylmethyl)benzamide (46 mg, 0.16 mmol), Pd2(dba)3 (14.6 mg, 0.016 mmol), BrettPhos (17 mg, 0.032 mmol), and cesium carbonate (104 mg, 0.32 mmol) were mixed in 1,4-dioxane (1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 20, 3-[(6-phenylpyridazin-3-yl)amino]-/V-[(pyridin-3-yl)methyl]b enzamide (41 mg, 60%) as a beige solid.

Synthesis of Compound 21 Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and 3-(pyrrolidin-l-yl)propan-

1-amine (0.1 mL, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 26 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NELCl. The organic layer was dried over anhydrous Na2SCL. The mi xture(3-bromo-A-(3 -(pyrrol idin-1 -yl)propyl)benzamide) was concentrated and used in the next step without further purification.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-(3-(pyrrolidin-l- yl)propyl)benzamide (121 mg, 0.19 mmol), Pd2(dba)3 (18 mg, 0.019 mmol), BrettPhos (21 mg, 0.039 mmol), and cesium carbonate (127 mg, 0.39 mmol) were mixed in 1,4-dioxane (1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by EA and saturated aq. NELCl. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by preparative thin layer chromatography (PTLC). The crude mixture was solidified by using EA to give compound 21, 3-[(6-phenylpyridazin-3- yl)amino]-A-[3-(pyrrolidin-l-yl)propyl]benzamide (15 mg, 19%) as a beige solid.

Synthesis of Compound 22

Step 1: 4-Iodobenzoyl chloride (288 mg, 1.08 mmol) and (5-methylfuran-2- yl)methanamine (0.98 mL, 0.9 mmol) were dissolved in DCM (9 mL), followed up by addition of DIPEA (0.34 mL, 1.9 mmol) and stirred for 22 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NEECl. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC to give 4-iodo-A-((5- methylfuran-2-yl)methyl)benzamide (295 mg, 96%) as a beige solid.

Step 2: 6-Phenylpyridazin-3 -amine (100 mg, 0.58 mmol), 4-iodo-/V-((5-methylfuran-2- yl)methyl)benzamide (219 mg, 0.64 mmol), Pd2(dba)3 (53 mg, 0.058 mmol), BrettPhos (63 mg, 0.12 mmol), and cesium carbonate (381 mg, 1.17 mmol) were mixed in 1,4-dioxane (4 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using MeOH/DCM (1:10) to give compound 22, /V-[(5-methylfuran-2-yl)methyl]-4-[(6-phenylpyridazin-3- yl)amino]benzamide (21 mg, 9%) as a white solid.

Synthesis of Compound 23

6-(Pyridin-2-yl)pyridazin-3-amine (30 mg, 0.17 mmol), 3-bromo-A-((5-methylfuran-2- yl)methyl)benzamide (46 mg, 0.16 mmol), Pd2(dba)3 (14 mg, 0.016 mmol), BrettPhos (17 mg, 0.03 mmol), and cesium carbonate (103 mg, 0.32 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by MeOH/DCM (1:10) and brine. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using DCM and EA to give compound 23, /V-[(5-methylfuran-2-yl)methyl]-3-{[6-(pyridin-2- yl)pyridazin-3-yl]amino}benzamide (8 mg, 13%) as a beige solid.

Synthesis of Compound 24

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and 2,2-dimethylpropan-l- amine (0.08 mL, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 26 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3-bromo-A-neopentylbenzamide (166 mg, 66%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-A- neopentylbenzamide (63 mg, 0.23 mmol), Pd 2 (dba) 3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.046 mmol), and cesium carbonate (151 mg, 0.46 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 24, /V-(2,2-dimethylpropyl)-3-[(6-phenylpyridazin-3-yl)amino]ben zamide (20 mg, 24%) as a beige solid.

Synthesis of Compound 25

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and cyclobutanamine (54 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 25 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3-bromo-A-cyclobutylbenzamide (201 mg, >99%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V- cyclobutylbenzamide (54 mg, 0.21 mmol), Pd 2 (dba) 3 (20 mg, 0.021 mmol), BrettPhos (23 mg, 0.042 mmol), and cesium carbonate (138 mg, 0.42 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using DCM to give compound 25, /V-cyclobutyl-3-[(6-phenylpyridazin-3-yl)amino]benzamide (20 mg, 27%) as a white solid.

Synthesis of Compound 26

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and oxetan-3 -amine (56 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 23 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3-bromo-A-(oxetan-3-yl)benzamide (197 mg, >99%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-(oxetan-3- yl)benzamide (54 mg, 0.21 mmol), Pd2(dba)3 (20 mg, 0.021 mmol), BrettPhos (23 mg, 0.042 mmol), and cesium carbonate (138 mg, 0.42 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 26, /V-(oxetan-3-yl)-3-[(6-phenylpyridazin-3-yl)amino]benzamide (14 mg, 19%) as a beige solid.

Synthesis of Compound 27

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and 2-(pyridin-4-yl)ethan-l- amine (93 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 26 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NELCl. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by MPLC to give 3-bromo-A- (2-(pyridin-4-yl)ethyl)benzamide (170 mg, 73%) as a beige solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-(2-(pyridin-4- yl)ethyl)benzamide (65 mg, 0.21 mmol), Pd2(dba)3 (20 mg, 0.021 mmol), BrettPhos (23 mg, 0.042 mmol), and cesium carbonate (138 mg, 0.42 mmol) were mixed in 1,4-dioxane (1.4 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 27, 3-[(6-phenylpyridazin-3-yl)amino]- /V-[2-(pyridin-4-yl)ethyl]benzamide (49 mg, 83%) as a beige solid.

Synthesis of Compound 28

Step 1: 3-Bromobenzoyl chloride (200 mg, 0.91 mmol) and tetrahydro-2//-pyran-4- amine hydrochloride (104 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.42 mL, 2.4 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3-bromo-A-(tetrahydro-2//- pyran-4-yl)benzamide (206 mg, 96%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-A-(tetrahydro-2//- pyran-4-yl)benzamide (60 mg, 0.21 mmol), Pd2(dba)3 (20 mg, 0.021 mmol), BrettPhos (23 mg, 0.042 mmol), and cesium carbonate (138 mg, 0.42 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and DCM to give compound 28, /V-(oxan-4-yl)-3-[(6-phenylpyridazin-3-yl)amino]benzamide (31 mg, 39%) as a white solid.

Synthesis of Compound 29

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and 3-fluoroaniline (84 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 22 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3 -bromo-A-(3 -fluorophenyl )benzamide (223 mg, >99%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-(3- fluorophenyl)benzamide (62 mg, 0.21 mmol), Pd2(dba)3 (20 mg, 0.021 mmol), BrettPhos (23 mg, 0.042 mmol), and cesium carbonate (138 mg, 0.42 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 29, /V-(3-fluorophenyl)-3-[(6-phenylpyridazin-3-yl)amino]benzami de (25 mg, 30%) as a white solid.

Synthesis of Compound 30

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and cyclobutylmethanamine hydrochloride (92 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.42 mL, 2.4 mmol) and stirred for 19 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3-bromo-A- (cyclobutylmethyl)benzamide (210 mg, >99%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-A- (cyclobutylmethyl)benzamide (57 mg, 0.21 mmol), Pd 2 (dba) 3 (20 mg, 0.021 mmol), BrettPhos (23 mg, 0.042 mmol), and cesium carbonate (138 mg, 0.42 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 30, /V-(cyclobutylmethyl)-3-[(6-phenylpyridazin-3-yl)amino]benza mide (24 mg, 31%) as a white solid.

Synthesis of Compound 31

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and cyclohexylmethanamine (86 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 19 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3 -bromo-A-(cyclohexyl methyl )benzamide (192 mg, 86%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-A- (cyclohexylmethyl)benzamide (63 mg, 0.21 mmol), Pd 2 (dba) 3 (20 mg, 0.021 mmol), BrettPhos (23 mg, 0.042 mmol), and cesium carbonate (138 mg, 0.42 mmol) were mixed in 1,4-dioxane

(1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 31, A-(cyclohexylmethyl)-3-[(6- phenylpyridazin-3-yl)amino]benzamide (16 mg, 19%) as a yellowish white solid.

Synthesis of Compound 32 Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and cyclopropylmethanamine (54 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 19 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3 -bromo-A-(cyclopropyl methyl )benzamide (138 mg, 72%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-A- (cyclopropylmethyl)benzamide (59 mg, 0.23 mmol), Pd 2 (dba) 3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.046 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 32, A -(cycl opropyl methyl )-3 -[(6-phenyl pyridazin-3- yl)amino]benzamide (22 mg, 27%) as a white solid.

Synthesis of Compound 33

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and cyclopentylmethanamine hydrochloride (103 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.42 mL, 2.4 mmol) and stirred for 19 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3-bromo-A- (cyclopentylmethyl)benzamide (210 mg, 98%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-A- (cyclopentylmethyl)benzamide (66 mg, 0.23 mmol), Pd 2 (dba) 3 (21 mg, 0.023 mmol),

BrettPhos (25 mg, 0.046 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 33, N- (cyclopentylmethyl)-3-[(6-phenylpyridazin-3-yl)amino]benzami de (17 mg, 19%) as a yellowish white solid.

Synthesis of Compound 34

Step 1: 3-Bromobenzoyl chloride (200 mg, 0.91 mmol) and (tetrahydro-2//-pyran-4- yl)methanamine (88 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 21 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3-bromo-A-((tetrahydro-2//- pyran-4-yl)methyl)benzamide (163 mg, 72%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (49 mg, 0.29 mmol), 3-bromo-A-((tetrahydro-2//- pyran-4-yl)methyl)benzamide (85 mg, 0.29 mmol), Pd2(dba)3 (26 mg, 0.03 mmol), BrettPhos (31 mg, 0.06 mmol), and cesium carbonate (186 mg, 0.57 mmol) were mixed in 1,4-dioxane (1.4 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using HEX and EA to give compound 34, /V-[(oxan-4-yl)methyl]-3-[(6-phenylpyridazin-3-yl)amino]benz amide (28 mg, 25%) as a beige solid.

Synthesis of Compound 35

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and oxetan-3-ylmethanamine (66 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 23 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3 -bromo-A-(oxetan-3 -yl methyl )benzamide (195 mg, 95%) as a yellow oil.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-(oxetan-3- ylmethyl)benzamide (63 mg, 0.23 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.046 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 35, A-[(oxetan-3-yl)methyl]-3-[(6- phenylpyridazin-3-yl)amino]benzamide (27 mg, 28%) as a yellowish white solid. Synthesis of Compound 36

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and (3,4- dichlorophenyl)methanamine (134mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 22 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3-bromo-/V-(3,4- dichlorobenzyl)benzamide(268 mg, 98%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-(3,4- dichlorobenzyl)benzamide (94 mg, 0.23 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.046 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 36, /V-(3,4-dichlorobenzyl)-3-((6-phenylpyridazin-3-yl)amino)ben zamide (20 mg, 19%) as a beige solid.

Synthesis of Compound 37

6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-A-ethylbenzamide (64 mg, 0.28 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.046 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and DCM to give compound 37, /V-ethyl-3-[(6-phenylpyridazin-3-yl)amino]benzamide (14 mg, 19%) as a beige solid.

Synthesis of Compound 38

6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-cyclopropylbenzamide (67 mg, 0.28 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.046 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 38, N- cyclopropyl-3-[(6-phenylpyridazin-3-yl)amino]benzamide (25 mg, 33%) as a white solid.

Synthesis of Compound 39

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and thiophen-2- ylmethanamine (86 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 18 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3-bromo-A-(thiophen-2- ylmethyl)benzamide (206 mg, 92%) as a beige solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-(thiophen-2- ylmethyl)benzamide (76 mg, 0.26 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.046 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 39, 3-[(6-phenylpyridazin-3-yl)amino]-/V-[(thiophen-2-yl)methyl] benzamide (29 mg, 32%) as a white solid.

Synthesis of Compound 40

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and (5-methylthiophen-2- yl)methanamine hydrochloride (54 mg, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 2.4 mmol) and stirred for 25 hours at room temperature. The reaction mixture was concentrated and purified by MPLC to give 3-bromo-A- ((5-methylthiophen-2-yl)methyl)benzamide (236 mg, >99%) as a beige solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-((5- methylthiophen-2-yl)methyl)benzamide (80 mg, 0.26 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.046 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 40, /V-[(5-methylthiophen-2-yl)methyl]-3-[(6-phenylpyridazin-3- yl)amino]benzamide (20 mg, 21%) as a beige solid.

Synthesis of Compound 41

6-Phenylpyridazin-3 -amine (50 mg, 0.29 mmol), 3-bromo-A-methylbenzamide (188 mg, 0.88 mmol), Pd2(dba)3 (27 mg, 0.03 mmol), BrettPhos (31 mg, 0.06 mmol), and cesium carbonate (186 mg, 0.57 mmol) were mixed in 1,4-dioxane (1.5 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 41, A-methyl-3-[(6-phenylpyridazin-3- yl)amino]benzamide (10 mg, 11%) as a brown solid. Synthesis of Compound 42

5-Methylpyridazin-3-amine (35 mg, 0.32 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (123 mg, 0.42 mmol), Pd2(dba)3 (29 mg, 0.03 mmol), BrettPhos (34 mg, 0.06 mmol), and cesium carbonate (209 mg, 0.64 mmol) were mixed in 1,4-dioxane (1.6 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 42, /V-[(5-methylfuran-2-yl)methyl]-3-[(5-methylpyridazin-3-yl)a mino]benzamide (40 mg, 39%) as a beige solid.

Synthesis of Compound 43

6-Cy cl opropylpyridazin-3 -amine (40 mg, 0.3 mmol), 3-bromo-A-((5-methylfuran-2- yl)methyl)benzamide (111 mg, 0.38 mmol), Pd2(dba)3 (27 mg, 0.03 mmol), BrettPhos (32 mg, 0.06 mmol), and cesium carbonate (193 mg, 0.59 mmol) were mixed in 1,4-dioxane (1.5 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 43, 3-[(6-cyclopropylpyridazin-3-yl)amino]-A-[(5-methylfuran-2- yl)methyl]benzamide (47 mg, 45%) as a beige solid.

Synthesis of Compound 44

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and thiophen-3- ylmethanamine (0.075 mL, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 23 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NLLCl. The organic layer was dried over anhydrous NaiSCL and concentrated to give 3 -bromo-A-(thi ophen-3-yl methyl )benzamide (259 mg, >99%) as a brown solid.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-(thiophen-3- ylmethyl)benzamide (103 mg, 0.35 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 44, 3-[(6-phenylpyridazin-3-yl)amino]- /V-[(thiophen-3-yl)methyl]benzamide (10 mg, 11%) as a beige solid.

Synthesis of Compound 45

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and furan-3-ylmethanamine (0.082 mL, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 28 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NLLCl. The organic layer was dried over anhydrous NaiSCL and concentrated to give 3 -bromo-A-(furan-3 -yl methyl )benzamide (252 mg, >99%) as a brown oil.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-(furan-3- ylmethyl)benzamide (98 mg, 0.35 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 45, /V-[(furan-3-yl)methyl]-3-[(6-phenylpyridazin-3-yl)amino]ben zamide (33 mg, 38%) as a beige solid.

Synthesis of Compound 46

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and furan-2-ylmethanamine (0.08 mL, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 25 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NELCl. The organic layer was dried over anhydrous NaiSCL and concentrated to give 3 -bromo-A-(furan-2-yl methyl )benzamide (285 mg, >99%) as a brown oil.

Step 2: 6-Phenylpyridazin-3 -amine (40 mg, 0.23 mmol), 3-bromo-/V-(furan-2- ylmethyl)benzamide (98 mg, 0.35 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 46, /V-[(furan-2-yl)methyl]-3-[(6-phenylpyridazin-3-yl)amino]ben zamide (10 mg, 12%) as a beige solid.

Synthesis of Compound 47

6-Methylpyridazin-3 -amine (35 mg, 0.32 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (123 mg, 0.42 mmol), Pd2(dba)3 (29 mg, 0.03 mmol), BrettPhos (34 mg, 0.06 mmol), and cesium carbonate (209 mg, 0.64 mmol) were mixed in 1,4-dioxane (1.6 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 47, /V-[(5-methylfuran-2-yl)methyl]-3-[(6-methylpyridazin-3-yl)a mino]benzamide (45 mg, 44%) as a beige solid.

Synthesis of Compound 48

4-Methylpyridazin-3 -amine (35 mg, 0.32 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (123 mg, 0.42 mmol), Pd2(dba)3 (29 mg, 0.03 mmol), BrettPhos (34 mg, 0.06 mmol), and cesium carbonate (209 mg, 0.64 mmol) were mixed in 1,4-dioxane (1.6 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 48, /V-[(5-methylfuran-2-yl)methyl]-3-[(4-methylpyridazin-3-yl)a mino]benzamide (35 mg, 34%) as a beige solid.

Synthesis of Compound 49

6-(Tetrahydro-2iT-pyran-4-yl)pyridazin-3 -amine (40 mg, 0.22 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (85 mg, 0.29 mmol), Pd2(dba)3 (20 mg, 0.021 mmol), BrettPhos (23 mg, 0.042 mmol), and cesium carbonate (145 mg, 0.45 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 49, /V-[(5-methylfuran-2-yl)methyl]-3-{[6-(oxan-4-yl)pyridazin-3 - yl]amino}benzamide (42 mg, 48%) as a white solid.

Synthesis of Compound 50

[l,l’-Biphenyl]-4-amine (45 mg, 0.27 mmol), 3-bromo-A-phenethylbenzamide (97 mg, 0.32 mmol), Pd2(dba)3 (24 mg, 0.27 mmol), BrettPhos (29 mg, 0.053 mmol), and cesium carbonate (173 mg, 0.53 mmol) were mixed in 1,4-dioxane (1.3 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 50, 3- ({[l,l’-biphenyl]-4-yl}amino)-A-(2-phenylethyl)benzamide (29 mg, 28%) as a grey solid. Synthesis of Compound 51

5-Phenylpyrazin-2-amine (45 mg, 0.26 mmol), 3-bromo-A-phenethylbenzamide (96 mg, 0.32 mmol), Pd2(dba)3 (24 mg, 0.026 mmol), BrettPhos (28 mg, 0.053 mmol), and cesium carbonate (171 mg, 0.53 mmol) were mixed in 1,4-dioxane (1.3 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 51, N-( 2- phenylethyl)-3-[(5-phenylpyrazin-2-yl)amino]benzamide (20 mg, 19%) as a yellowish white solid.

Synthesis of Compound 52

5-Phenylpyrimidin-2-amine (45 mg, 0.26 mmol), 3-bromo-/V-phenethylbenzamide (96 mg, 0.32 mmol), Pd2(dba)3 (24 mg, 0.026 mmol), BrettPhos (28 mg, 0.053 mmol), and cesium carbonate (171 mg, 0.53 mmol) were mixed in 1,4-dioxane (1.3 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 52, N-( 2- phenylethyl)-3-[(5-phenylpyrimidin-2-yl)amino]benzamide (59 mg, 57%) as a beige solid.

Synthesis of Compound 53

[l,l’-Biphenyl]-4-amine (45 mg, 0.27 mmol), 3-bromo-/V-(3-phenylpropyl)benzamide (121 mg, 0.4 mmol), Pd2(dba)3 (24 mg, 0.027 mmol), BrettPhos (28 mg, 0.053 mmol), and cesium carbonate (173 mg, 0.53 mmol) were mixed in 1,4-dioxane (1.3 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 53, 3-({[l,l’-biphenyl]-4-yl}amino)-/V-(3-phenylpropyl)benzami de (25 mg, 23%) as a white solid. Synthesis of Compound 54

5-Phenylpyrazin-2-amine (45 mg, 0.27 mmol), 3 -bromo-A-(3 -phenyl propyl )benzamide (121 mg, 0.4 mmol), Pd2(dba)3 (24 mg, 0.027 mmol), BrettPhos (28 mg, 0.053 mmol), and cesium carbonate (173 mg, 0.53 mmol) were mixed in 1,4-dioxane (1.3 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 54, N-(3- phenylpropyl)-3-[(5-phenylpyrazin-2-yl)amino]benzamide (37 mg, 35%) as a yellowish white solid.

Synthesis of Compound 55

5-Phenylpyrimidin-2-amine (45 mg, 0.26 mmol), 3-bromo-/V-(3- phenylpropyl)benzamide (120 mg, 039 mmol), Pd2(dba)3 (24 mg, 0.026 mmol), BrettPhos (28 mg, 0.053 mmol), and cesium carbonate (171 mg, 0.53 mmol) were mixed in 1,4-dioxane (1.3 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 55, /V-(3-phenylpropyl)-3-[(5-phenylpyrimidin-2-yl)amino]benzami de (37 mg, 35%) as a beige solid.

Synthesis of Compound 57

Step 1: (3-Fluorophenyl)boronic acid (300 mg, 2.1 mmol), 4-bromoaniline (307 mg, 1.79 mmol), Pd(PPli3)4 (103 mg, 0.09 mmol) and potassium carbonate (740 mg, 5.36 mmol) were mixed in H2O/DMF (4.3/4.3 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was concentrated and purified by MPLC to give 3’-fluoro-[l,l’- biphenyl]-4-amine (276 mg, 82%) as a beige solid.

Step 2: 3’-Fluoro-[l,l’-biphenyl]-4-amine (40 mg, 0.21 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (82 mg, 0.28 mmol), Pd2(dba)3 (20 mg, 0.02 mmol), BrettPhos (23 mg, 0.042 mmol), and cesium carbonate (139 mg, 0.43 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 57, 3-({3’-fluoro-[l,l , -biphenyl]-4-yl}amino)-/V-[(5- methylfuran-2-yl)methyl]benzamide (30 mg, 35%) as a white solid.

Synthesis of Compound 58

Step 1: (3-Fluorophenyl)boronic acid (300 mg, 2.1 mmol), 5-bromopyrazin-2-amine (311 mg, 1.79 mmol), Pd(PPli3)4 (103 mg, 0.09 mmol) and potassium carbonate (740 mg, 5.36 mmol) were mixed in H2O/DMF (4.3/4.3 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was concentrated and purified by MPLC to give 5-(3- fluorophenyl)pyrazin-2-amine (277 mg, 82%) as a yellowish white solid.

Step 2: 5-(3-Fluorophenyl)pyrazin-2-amine (40 mg, 0.23 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (80 mg, 0.27 mmol), Pd2(dba)3 (20 mg, 0.021 mmol), BrettPhos (23 mg, 0.042 mmol), and cesium carbonate (138 mg, 0.42 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 58, 3-{[5-(3- fluorophenyl)pyrazin-2-yl]amino}-/V-[(5-methylfuran-2-yl)met hyl]benzamide (14 mg, 16%) as a brown solid.

Synthesis of Compound 60

5-(3-Fluorophenyl)pyrimidin-2-amine (45 mg, 0.24 mmol), 3-bromo-/V-(3- phenylpropyl)benzamide (114 mg, 0.36 mmol), Pd2(dba)3 (22 mg, 0.024 mmol), BrettPhos (26 mg, 0.048 mmol), and cesium carbonate (155 mg, 0.48 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 60, 3 - { [5-(3 -fluorophenyl)pyrimidin-2-yl] amino } -N-(3 -phenylpropyl)benzamide (30 mg, 30%) as a white solid.

Synthesis of Compound 61

6-Isobutylpyridazin-3 -amine (44 mg, 0.29 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (110 mg, 0.37 mmol), Pd2(dba)3 (26 mg, 0.03 mmol), BrettPhos (31 mg, 0.06 mmol), and cesium carbonate (188 mg, 0.58 mmol) were mixed in 1,4-dioxane (1.4 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 61, /V-[(5-methylfuran-2-yl)methyl]-3-{[6-(2-methylpropyl)pyrida zin-3- yl]amino}benzamide (49 mg, 47%) as a beige solid.

Synthesis of Compound 62

6-Cy cl opentylpyridazin-3 -amine (47 mg, 0.29 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (110 mg, 0.37 mmol), Pd2(dba)3 (26 mg, 0.03 mmol), BrettPhos (31 mg, 0.06 mmol), and cesium carbonate (188 mg, 0.58 mmol) were mixed in 1,4-dioxane (1.4 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 62, 3-[(6-cyclopentylpyridazin-3-yl)amino]-A-[(5-methylfuran-2- yl)methyl]benzamide (72 mg, 66%) as a beige solid. Synthesis of Compound 63

6-Cy cl ohexylpyridazin-3 -amine (51 mg, 0.29 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (110 mg, 0.37 mmol), Pd2(dba)3 (26 mg, 0.03 mmol), BrettPhos (31 mg, 0.06 mmol), and cesium carbonate (188 mg, 0.58 mmol) were mixed in 1,4-dioxane (1.4 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 63, 3-[(6-cyclohexylpyridazin-3-yl)amino]-/V-[(5-methylfuran-2- yl)methyl]benzamide (51 mg, 46%) as a beige solid. Synthesis of Compound 65

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and 2-methylpropan-2-amine (0.08 mL, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 31 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NH 4 CI. The organic layer was dried over anhydrous NaiSCL and concentrated to give 3-bromo-A-(/c/V-butyl)benzamide (209 mg, >99%) as a brown oil.

Step 2: 6-Phenylpyridazin-3 -amine (45 mg, 0.26 mmol), 3-bromo -N-(tert- butyl)benzamide (88 mg, 0.34 mmol), Pd 2 (dba) 3 (24 mg, 0.026 mmol), BrettPhos (28 mg, 0.053 mmol), and cesium carbonate (171 mg, 0.53 mmol) were mixed in 1,4-dioxane (1.3 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 65, A-/er/-butyl-3-[(6-phenylpyridazin-3-yl)amino]benzamide (14 mg, 15%) as a light orange solid.

Synthesis of Compound 66

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and pentan-3 -amine (0.09 mL, 0.76 mmol) were dissolved in DCM (7.6 mL), followed up by addition of DIPEA (0.28 mL, 1.63 mmol) and stirred for 31 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NELCl. The organic layer was dried over anhydrous NaiSCL and concentrated to give 3 -bromo-A -(pentan-3 -yl)benzamide (238 mg, >99%) as a brown oil.

Step 2: 6-Phenylpyridazin-3 -amine (45 mg, 0.26 mmol), 3-bromo-/V-(pentan-3- yl)benzamide (106 mg, 0.39 mmol), Pd2(dba)3 (24 mg, 0.026 mmol), BrettPhos (28 mg, 0.053 mmol), and cesium carbonate (171 mg, 0.53 mmol) were mixed in 1,4-dioxane (1.3 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 66, /V-(pentan-3-yl)-3-[(6-phenylpyridazin-3-yl)amino]benzamide (27 mg, 28%) as a beige solid.

Synthesis of Compound 69

/ert-Butyl 4-(6-aminopyridazin-3-yl)piperidine-l-carboxylate (146 mg, 0.52 mmol), 3- bromo-A-((5-methylfuran-2-yl)methyl)benzamide (200 mg, 0.68 mmol), Pd 2 (dba) 3 (48 mg,

0.05 mmol), BrettPhos (56 mg, 0.1 mmol), and cesium carbonate (341 mg, 1.05 mmol) were mixed in 1,4-dioxane (2.6 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 69, /ert-butyl 4-{6-[(3-{[(5-methylfuran-2- yl)methyl]carbamoyl}phenyl)amino]pyridazin-3-yl}piperidine-l -carboxylate (109 mg, 42%) as a beige solid.

Synthesis of Compound 70

6-(l-Methylpiperidin-4-yl)pyridazin-3 -amine (50 mg, 0.26 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (100 mg, 0.34 mmol), Pd2(dba)3 (24 mg, 0.03 mmol), BrettPhos (28 mg, 0.05 mmol), and cesium carbonate (170 mg, 0.52 mmol) were mixed in 1,4- dioxane (1.3 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 70, /V-[(5-methylfuran-2-yl)methyl]-3-{[6-(l-methylpiperidin-4- yl)pyridazin-3-yl]amino}benzamide (9 mg, 9%) as a beige solid.

Synthesis of Compound 71

/ <2/7 - Butyl 4-(6-((3-(((5-methylfuran-2-yl)methyl)carbamoyl)phenyl)amino )pyridazin-3- yl)piperidine-l-carboxylate (30 mg, 0.61 mmol) was dissolved in DCM (3 mL), followed up by addition of trifluoroacetic acid (TFA) (0.5 mL, 0.12 M) and stirred for 1 hour at room temperature. The reaction mixture was extracted by DCM and saturated aq. NaFlCCb. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated to give compound 71, A-[(5- methylfuran-2-yl)methyl]-3-{[6-(piperidin-4-yl)pyridazin-3-y l]amino}benzamide (18 mg,

75%) as a beige foam.

Synthesis of Compound 72

Step 1: (3,5-Dimethylisoxazol-4-yl)boronic acid (200 mg, 1.3 mmol), 6- bromopyridazin-3 -amine (150 mg, 0.86 mmol), Pd(PPli3)4 (50 mg, 0.04 mmol) and potassium carbonate (357 mg, 2.59 mmol) were mixed in H2O/DMF (1.7/1.7 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was concentrated and purified by MPLC to give 6-(3,5-dimethylisoxazol-4-yl)pyridazin-3-amine (51 mg, 31%) as a white solid.

Step 2: 6-(3,5-Dimethylisoxazol-4-yl)pyridazin-3-amine (45 mg, 0.24 mmol), 3-bromo- /V-((5-methylfuran-2-yl)methyl)benzamide (90 mg, 0.31 mmol), Pd2(dba)3 (22 mg, 0.024 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (153 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 72, 3-{[6-(3,5-dimethyl-l,2-oxazol-4-yl)pyridazin-3-yl]amino}- /V-[(5-methylfuran-2-yl)methyl]benzamide (28 mg, 29%) as a beige solid.

Synthesis of Compound 73

Step 1: Thiophen-3-ylboronic acid (132 mg, 1.03 mmol), 6-bromopyridazin-3 -amine (150 mg, 0.86 mmol), Pd(PPli3)4 (50 mg, 0.04 mmol) and potassium carbonate (357 mg, 2.59 mmol) were mixed in LLO/DMF (1.7/1.7 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was concentrated and purified by MPLC to give 6- (thiophen-3-yl)pyridazin-3 -amine (122 mg, 79%) as a yellowish white solid.

Step 2: 6-(Thiophen-3-yl)pyridazin-3 -amine (42 mg, 0.24 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (90 mg, 0.31 mmol), Pd2(dba)3 (22 mg, 0.024 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (153 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 73, /V-[(5-methylfuran-2-yl)methyl]-3-{[6-(thiophen-3-yl)pyridaz in-3- yl]amino}benzamide (30 mg, 33%) as a beige solid.

Synthesis of Compound 74

Step 1: (4-Methylthiophen-3-yl)boronic acid (147 mg, 1.03 mmol), 6-bromopyridazin- 3-amine (150 mg, 0.86 mmol), Pd(PPh3)4 (50 mg, 0.04 mmol), and potassium carbonate (357 mg, 2.59 mmol) were mixed in H2O/DMF (1.7/1.7 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was concentrated and purified by MPLC to give 6-

(4-methylthiophen-3-yl)pyridazin-3 -amine (70 mg, 42%) as a beige solid.

Step 2: 6-(4-Methylthiophen-3-yl)pyridazin-3 -amine (45 mg, 0.24 mmol), 3-bromo-/V- ((5-methylfuran-2-yl)methyl)benzamide (90 mg, 0.31 mmol), Pd2(dba)3 (22 mg, 0.024 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (153 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 74, /V-[(5-methylfuran-2-yl)methyl]-3-{[6-(4-methylthiophen-3- yl)pyridazin-3-yl]amino}benzamide (23 mg, 24%) as a beige solid.

Synthesis of Compound 75

Step 1: (4-Chlorophenyl)boronic acid (200 mg, 1.28 mmol), 6-bromopyridazin-3 -amine (290 mg, 1.66 mmol), Pd(PPli3)4 (74 mg, 0.064 mmol), and potassium carbonate (530 mg, 3.84 mmol) were mixed in H2O/DMF (2.6/2.6 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The crude mixture was solidified by using EA and HEX to give 6-(4-chlorophenyl)pyridazin-3 -amine (175 mg, 66%) as a yellow solid. Step 2: 6-(4-Chlorophenyl)pyridazin-3-amine (48 mg, 0.24 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (90 mg, 0.31 mmol), Pd2(dba)3 (22 mg, 0.024 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (153 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 75, 3-{[6-(4-chlorophenyl)pyridazin-3-yl]amino}-/V-[(5-methylfur an-2- yl)methyl]benzamide (29 mg, 29%) as a beige solid.

Synthesis of Compound 76

6-Phenethylpyridazin-3 -amine (47 mg, 0.24 mmol), 3-bromo-A-((5-methylfuran-2- yl)methyl)benzamide (90 mg, 0.31 mmol), Pd2(dba)3 (22 mg, 0.024 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (153 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 76, /V-[(5-methylfuran-2-yl)methyl]-3-{[6-(2-phenylethyl)pyridaz in-3- yl]amino}benzamide (37 mg, 38%) as a white solid.

Synthesis of Compound 77

6-(4-Fluorophenethyl)pyridazin-3-amine (51 mg, 0.24 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (90 mg, 0.31 mmol), Pd2(dba)3 (22 mg, 0.024 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (153 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 77, 3-({6-[2-(4-fluorophenyl)ethyl]pyridazin-3-yl}amino)-/V-[(5- methylfuran-2-yl)methyl]benzamide (33 mg, 33%) as a white solid.

Synthesis of Compound 78

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and 2-(3- methoxyphenyl)ethan-l -amine (0.13 mL, 0.91 mmol) were dissolved in DCM (9.1 mL), followed up by addition of DIPEA (0.34 mL, 1.96 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NH 4 C 1 . The organic layer was dried over anhydrous NaiSCL and concentrated to give 3-bromo-N-(3- methoxyphenethyl)benzamide (370 mg, >99%) as a yellow oil.

Step 2: 5-(3-Fluorophenyl)pyrimidin-2-amine (40 mg, 0.21 mmol), 3-bromo-/V-(3- methoxyphenethyl)benzamide (103 mg, 0.25 mmol), Pd2(dba)3 (20 mg, 0.021 mmol), BrettPhos (23 mg, 0.042 mmol), and cesium carbonate (138 mg, 0.42 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 78, 3-{[5-(3-fhuorophenyl)pyrimidin-2-yl]amino}-/V-[2-(3- methoxyphenyl)ethyl]benzamide (13 mg, 14%) as a white solid.

Synthesis of Compound 80

5-(3-Fluorophenyl)pyrimidin-2-amine (45 mg, 0.24 mmol), 3-bromo-/V-(2- cyclohexylethyl)benzamide (94 mg, 0.29 mmol), Pd 2 (dba) 3 (22 mg, 0.024 mmol), BrettPhos (26 mg, 0.048 mmol), and cesium carbonate (155 mg, 0.48 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 80, N -(2-cyclohexylethyl)-3-{[5-(3-fluorophenyl)pyrimidin-2-yl]am ino}benzamide (32 mg, 32%) as a white solid.

Synthesis of Compound 82

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and 2-(3,5- difluorophenyl)ethan-l -amine (0.12 mL, 0.91 mmol) were dissolved in DCM (9.1 mL), followed up by addition of DIPEA (0.34 mL, 1.96 mmol) and stirred for 22 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NELCl. The organic layer was dried over anhydrous Na2SCL and concentrated to give 3-bromo-A-(3,5- difluorophenethyl)benzamide (320 mg, >99%) as an orange solid.

Step 2: 5-Phenylpyrimidin-2-amine (40 mg, 0.23 mmol), 3-bromo-/V-(3,5- difluorophenethyl)benzamide (99 mg, 0.28 mmol), Pd 2 (dba) 3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane

(1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 82, /V-[2-(3,5-difluorophenyl)ethyl]-3-[(5-phenylpyrimidin-2-yl) amino]benzamide (26 mg, 26%) as a white solid. Synthesis of Compound 83

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and 2-(4- methoxyphenyl)ethan-l -amine (0.13 mL, 0.91 mmol) were dissolved in DCM (9.1 mL), followed up by addition of DIPEA (0.34 mL, 1.96 mmol) and stirred for 22 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NELCl. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated to give 3-bromo-A-(4- methoxyphenethyl)benzamide (325 mg, >99%) as a beige solid.

Step 2: 5-Phenylpyrimidin-2-amine (40 mg, 0.23 mmol), 3-bromo-/V-(4- methoxyphenethyl)benzamide (100 mg, 0.28 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 83, /V-[2-(4-methoxyphenyl)ethyl]-3-[(5-phenylpyrimidin-2- yl)amino]benzamide (28 mg, 28%) as a white solid.

Synthesis of Compound 84

Compound 84

6-Ethylpyridazin-3 -amine (28 mg, 0.23 mmol), 3-bromo-A-((5-methylfuran-2- yl)methyl)benzamide (80 mg, 0.27 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (24 mg, 0.045 mmol), and cesium carbonate (147 mg, 0.45 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 84, 3-[(6-ethylpyridazin-3-yl)amino]-/V-[(5-methylfuran-2- yl)methyl]benzamide (42 mg, 48%) as a beige solid.

Synthesis of Compound 85

6-Isopropylpyridazin-3 -amine (31 mg, 0.23 mmol), 3-bromo-A-((5-methylfuran-2- yl)methyl)benzamide (80 mg, 0.27 mmol), Pd2(dba)3 (21 mg, 0.023 mmol), BrettPhos (24 mg, 0.045 mmol), and cesium carbonate (147 mg, 0.45 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 85, /V-[(5-methylfuran-2-yl)methyl]-3-{[6-(propan-2-yl)pyridazin -3- yl]amino}benzamide (21 mg, 26%) as a beige solid. Synthesis of Compound 86

6-(Tetrahydrofuran-2-yl)pyridazin-3 -amine (40 mg, 0.24 mmol), 3-bromo-A-((5- methylfuran-2-yl)methyl)benzamide (98 mg, 0.29 mmol), Pd2(dba)3 (30 mg, 0.024 mmol), BrettPhos (26 mg, 0.048 mmol), and cesium carbonate (158 mg, 0.48 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 86, /V-[(5-methylfuran-2-yl)methyl]-3-{[6-(oxolan-2- yl)pyridazin-3-yl]amino}benzamide (20 mg, 22%) as a beige solid.

Synthesis of Compound 87

6-(Tetrahydrofuran-3-yl)pyridazin-3 -amine (40 mg, 0.24 mmol), 3-bromo-A-((5- methylfuran-2-yl)methyl)benzamide (98 mg, 0.29 mmol), Pd2(dba)3 (30 mg, 0.024 mmol), BrettPhos (26 mg, 0.048 mmol), and cesium carbonate (158 mg, 0.48 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 87, /V-[(5-methylfuran-2-yl)methyl]-3-{[6-(oxolan-3- yl)pyridazin-3-yl]amino}benzamide (44 mg, 48%) as a white solid.

Synthesis of Compound 88

[l,l’-Biphenyl]-3-amine (40 mg, 0.24 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (96 mg, 0.28 mmol), Pd2(dba)3 (30 mg, 0.024 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (154 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 88, 3-({[l,l’-biphenyl]-3-yl}amino)-A-[(5-methylfuran-2- yl)methyl]benzamide (38 mg, 42%) as a beige solid.

Synthesis of Compound 89

4-Phenylpyrimidin-2-amine (40 mg, 0.24 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (94 mg, 0.28 mmol), Pd2(dba)3 (30 mg, 0.024 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (154 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 89, /V-[(5-methylfuran-2-yl)methyl]-3-[(4-phenylpyrimidin-2- yl)amino]benzamide (27 mg, 30%) as a beige solid.

Synthesis of Compound 90

Step 1: 3-Bromobenzoyl chloride (0.12 mL, 0.91 mmol) and 2-(3-fluorophenyl)ethan- 1-amine (0.12 mL, 0.91 mmol) were dissolved in DCM (9.1 mL), followed up by addition of DIPEA (0.34 mL, 1.96 mmol) and stirred for 22 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NH 4 CI. The organic layer was dried over anhydrous NaiSCL and concentrated to give 3-bromo-A-(3-fluorophenethyl)benzamide (340 mg, >99%) as a yellow oil.

Step 2: 5-Phenylpyrimidin-2-amine (40 mg, 0.23 mmol), 3-bromo-/V-(3- fluorophenethyl)benzamide (105 mg, 0.28 mmol), Pd 2 (dba) 3 (29 mg, 0.023 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (152 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 90, /V-[2-(3-fluorophenyl)ethyl]-3-[(5-phenylpyrimidin-2-yl)amin o]benzamide (28 mg, 29%) as a white solid.

Synthesis of Compound 92

Step 1: (5-Methylfuran-2-yl)boronic acid (144 mg, 0.69 mmol), 5-bromopyrimidin-2- amine (100 mg, 0.57 mmol), Pd(PPli3)4 (33 mg, 0.03 mmol), and potassium carbonate (238 mg, 1.72 mmol) were mixed in H2O/DMF (1.1/1.1 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was purified by MPLC to give 5- (5-methylfuran-2-yl)pyrimidin-2-amine (66 mg, 66%) as a yellowish white solid.

Step 2: 5-(5-Methylfuran-2-yl)pyrimidin-2-amine (60 mg, 0.17 mmol), 3-bromo-A-((5- methylfuran-2-yl)methyl)benzamide (60 mg, 0.21 mmol), Pd2(dba)3 (21 mg, 0.017 mmol), BrettPhos (18 mg, 0.034 mmol), and cesium carbonate (112 mg, 0.34 mmol) were mixed in 1,4-dioxane (0.86 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 92, /V-[(5-methylfuran-2-yl)methyl]-3-{[5-(5- methylfuran-2-yl)pyrimidin-2-yl]amino}benzamide (15 mg, 22%) as a beige solid.

Synthesis of Compound 98

Step 1: (2-(Trifluoromethyl)phenyl)boronic acid (131 mg, 0.69 mmol), 5- bromopyrimidin-2-amine (100 mg, 0.57 mmol), Pd(PPh3)4 (33 mg, 0.03 mmol), and potassium carbonate (238 mg, 1.72 mmol) were mixed in H2O/DMF (1.1/1.1 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was purified by MPLC to give 5-(2-(trifluoromethyl)phenyl)pyrimidin-2-amine (31 mg, 23%) as a yellow solid.

Step 2: 5-(2-(Trifluoromethyl)phenyl)pyrimidin-2-amine (30 mg, 0.13 mmol), 3- bromo-/V-((5-methylfuran-2-yl)methyl)benzamide (44 mg, 0.15 mmol), Pd 2 (dba) 3 (12 mg, 0.013 mmol), BrettPhos (14 mg, 0.025 mmol), and cesium carbonate (82 mg, 0.25 mmol) were mixed in 1,4-dioxane (0.63 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 98, /V-[(5-methylfuran-2-yl)methyl]-3-({5-[2- (trifluoromethyl)phenyl]pyrimidin-2-yl}amino)benzamide (17 mg, 31%) as a white solid.

Synthesis of Compound 99

Step 1: (3-(Trifluoromethyl)phenyl)boronic acid (131 mg, 0.69 mmol), 5- bromopyrimidin-2-amine (100 mg, 0.57 mmol), Pd(PPh3)4 (33 mg, 0.03 mmol), and potassium carbonate (238 mg, 1.72 mmol) were mixed in H2O/DMF (1.1/1.1 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The crude mixture was solidified by using MeOH to give 5-(3-(trifluoromethyl)phenyl)pyrimidin-2- amine (54 mg, 40%) as a beige solid.

Step 2: 5-(3-(Trifluoromethyl)phenyl)pyrimidin-2-amine (40 mg, 0.17 mmol), 3- bromo-/V-((5-methylfuran-2-yl)methyl)benzamide (59 mg, 0.2 mmol), Pd 2 (dba) 3 (15 mg, 0.017 mmol), BrettPhos (18 mg, 0.034 mmol), and cesium carbonate (109 mg, 0.33 mmol) were mixed in 1,4-dioxane (0.84 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 99, /V-[(5-methylfuran-2-yl)methyl]-3-({5-[3- (trifluoromethyl)phenyl]pyrimidin-2-yl}amino)benzamide (21 mg, 28%) as a white solid.

Synthesis of Compound 100

Step 1: (4-(Trifluoromethyl)phenyl)boronic acid (131 mg, 0.69 mmol), 5- bromopyrimidin-2-amine (100 mg, 0.57 mmol), Pd(PPh3)4 (33 mg, 0.03 mmol), and potassium carbonate (238 mg, 1.72 mmol) were mixed in H2O/DMF (1.1/1.1 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The crude mixture was solidified by using MeOH to give 5-(4-(trifluoromethyl)phenyl)pyrimidin-2- amine (49 mg, 36%) as a beige solid.

Step 2: 5-(4-(Trifluoromethyl)phenyl)pyrimidin-2-amine (40 mg, 0.17 mmol), 3- bromo-/V-((5-methylfuran-2-yl)methyl)benzamide (59 mg, 0.2 mmol), Pd 2 (dba) 3 (15 mg, 0.017 mmol), BrettPhos (18 mg, 0.034 mmol), and cesium carbonate (109 mg, 0.33 mmol) were mixed in 1,4-dioxane (0.84 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 100, /V-[(5-methylfuran-2-yl)methyl]-3-({5-[4- (trifluoromethyl)phenyl]pyrimidin-2-yl}amino)benzamide (21 mg, 27%) as a white solid.

Synthesis of Compound 101

Step 1: (3-(Ethoxycarbonyl)phenyl)boronic acid (268 mg, 1.38 mmol), 5- bromopyrimidin-2-amine (200 mg, 1.15 mmol), Pd(PPli3)4 (66 mg, 0.06 mmol), and potassium carbonate (477 mg, 3.45 mmol) were mixed in H2O/DMF (2.3/2.3 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The crude mixture was solidified by using MeOH to give ethyl 3-(2-aminopyrimidin-5-yl)benzoate (130 mg, 47%) as a beige solid.

Step 2: Ethyl 3-(2-aminopyrimidin-5-yl)benzoate (100 mg, 0.41 mmol), 3-bromo-A- ((5-methylfuran-2-yl)methyl)benzamide (145 mg, 0.49 mmol), Pd2(dba)3 (38 mg, 0.041 mmol), BrettPhos (44 mg, 0.082 mmol), and cesium carbonate (268 mg, 0.82 mmol) were mixed in 1,4-dioxane (2.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 101, ethyl 3-{2-[(3-{[(5-methylfuran-2- yl)methyl]carbamoyl}phenyl)amino]pyrimidin-5-yl (benzoate (110 mg, 27%) as a white solid.

Synthesis of Compound 102

Step 1: (4-(Ethoxycarbonyl)phenyl)boronic acid (268 mg, 1.38 mmol), 5- bromopyrimidin-2-amine (200 mg, 1.15 mmol), Pd(PPli3)4 (66 mg, 0.06 mmol), and potassium carbonate (477 mg, 3.45 mmol) were mixed in H2O/DMF (2.3/2.3 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The crude mixture was solidified by using MeOH to give ethyl 4-(2-aminopyrimidin-5-yl)benzoate (117 mg, 42%) as a beige solid.

Step 2: Ethyl 4-(2-aminopyrimidin-5-yl)benzoate (100 mg, 0.41 mmol), 3-bromo-A- ((5-methylfuran-2-yl)methyl)benzamide (145 mg, 0.49 mmol), Pd2(dba)3 (38 mg, 0.041 mmol), BrettPhos (44 mg, 0.082 mmol), and cesium carbonate (268 mg, 0.82 mmol) were mixed in 1,4-dioxane (2.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 102, ethyl 4-{2-[(3-{[(5-methylfuran-2- yl)methyl]carbamoyl}phenyl)amino]pyrimidin-5-yl (benzoate (96 mg, 23%) as a white solid.

Synthesis of Compound 103

Step 1: Benzo[ ][l,3]dioxol-5-ylboronic acid (171 mg, 0.69 mmol), 5- bromopyrimidin-2-amine (100 mg, 0.57 mmol), Pd(PPh3)4 (33 mg, 0.03 mmol), and potassium carbonate (238 mg, 1.72 mmol) were mixed in H2O/DMF (1.1/1.1 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The crude mixture was solidified by using MeOH to give 5-(benzo[ ][l,3]dioxol-5-yl)pyrimidin-2-amine (72 mg, 58%) as a beige solid.

Step 2: 5-(Benzo[ ][l,3]dioxol-5-yl)pyrimidin-2-amine (40 mg, 0.19 mmol), 3-bromo- /V-((5-methylfuran-2-yl)methyl)benzamide (66 mg, 0.22 mmol), Pd2(dba)3 (17 mg, 0.019 mmol), BrettPhos (20 mg, 0.037 mmol), and cesium carbonate (121 mg, 0.37 mmol) were mixed in 1,4-dioxane (0.9 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 103, 3-{[5-(2iT-l,3-benzodioxol-5-yl)pyrimidin-2-yl]amino}-/V- [(5-methylfuran-2-yl)methyl]benzamide (28 mg, 15%) as an orange solid. Synthesis of Compound 104

Step 1: Quinolin-3-ylboronic acid (119 mg, 0.69 mmol), 5-bromopyrimidin-2-amine (100 mg, 0.57 mmol), Pd(PPh3)4 (33 mg, 0.03 mmol), and potassium carbonate (238 mg, 1.72 mmol) were mixed in H2O/DMF (1.1/1.1 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was purified by MPLC to give 5- (quinolin-3-yl)pyrimidin-2-amine (37 mg, 29%) as a white solid.

Step 2: 5-(Quinolin-3-yl)pyrimidin-2-amine (35 mg, 0.16 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (56 mg, 0.19 mmol), Pd2(dba)3 (14 mg, 0.016 mmol), BrettPhos (17 mg, 0.032 mmol), and cesium carbonate (103 mg, 0.31 mmol) were mixed in 1,4-dioxane (0.8 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 104, /V-[(5-methylfuran-2-yl)methyl]-3-{[5-(quinolin-3-yl)pyrimid in-2- yl]amino}benzamide (18 mg, 12%) as an orange solid.

Synthesis of Compound 107

6-Aminopyridazine-3-carbonitrile (40 mg, 0.33 mmol), 3-bromo-/V-((5-methylfuran-2- yl)methyl)benzamide (118 mg, 0.4 mmol), Pd2(dba)3 (41 mg, 0.03 mmol), BrettPhos (36 mg, 0.07 mmol), and cesium carbonate (217 mg, 0.67 mmol) were mixed in 1,4-dioxane (1.7 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 107, 3-[(6-cyanopyridazin-3-yl)amino]-/V-[(5-methylfuran-2-yl)met hyl]benzamide (27 mg, 24%) as a beige solid. Synthesis of Compound 108

Ethyl 3-(2-((3-(((5-methylfuran-2-yl)methyl)carbamoyl)phenyl)amino )pyrimidin-5- yl)benzoate (32 mg, 0.07 mmol) and LiOEbEEO (12 mg, 0.28 mmol) were mixed in THF/H2O (0.47/0.23 mL) and stirred for 5 hours at 40°C. The reaction mixture was extracted by EA and aq. HC1 (IN). The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 108 ,3-{2-[(3-{[(5-methylfuran- 2-yl)methyl]carbamoyl}phenyl)amino]pyrimidin-5-yl (benzoic acid (21 mg, 70%) as a yellow solid.

Synthesis of Compound 109

Ethyl 4-(2-((3-(((5-methylfuran-2-yl)methyl)carbamoyl)phenyl)amino )pyrimi din-5- yl)benzoate (32 mg, 0.07 mmol) and LiOH-tkO (12 mg, 0.28 mmol) were mixed in THF/H2O (0.47/0.23 mL) and stirred for 24 hours at 40°C. The reaction mixture was extracted by EA and aq. HC1 (IN). The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The crude mixture was solidified by using EA to give compound 109, 4-{2-[(3-{[(5-methylfuran- 2-yl)methyl]carbamoyl}phenyl)amino]pyrimidin-5-yl (benzoic acid (20 mg, 66%) as a yellow solid.

Synthesis of Compound 110 Step 1: Thiophen-2-ylboronic acid (132 mg, 1.03 mmol), 5-bromopyrimidin-2-amine (150 mg, 0.86 mmol), Pd(PPh3)4 (50 mg, 0.043 mmol) and potassium carbonate (357 mg, 2.59 mmol) were mixed in H2O/DMF (1.7/1.7 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was purified by MPLC to give 5- (thiophen-2-yl)pyrimidin-2-amine (87 mg, 57%) as a beige solid.

Step 2: 5-(Thiophen-2-yl)pyrimidin-2-amine (70 mg, 0.24 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (84 mg, 0.28 mmol), Pd2(dba)3 (29 mg, 0.024 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (154 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 110, /V-[(5-methylfuran-2-yl)methyl]-3-{[5-(thiophen-2-yl)pyrimid in-2- yl]amino}benzamide (34 mg, 36%) as a white solid.

Synthesis of Compound 111

Step 1: Benzofuran-2-ylboronic acid (167 mg, 1.03 mmol), 5-bromopyrimidin-2-amine (150 mg, 0.86 mmol), Pd(PPh3)4 (50 mg, 0.043 mmol), and potassium carbonate (357 mg, 2.59 mmol) were mixed in H2O/DMF (1.7/1.7 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The crude mixture was solidified by using EA to give 5-(benzofuran-2-yl)pyrimidin-2-amine (67 mg, 37%) as a yellowish white solid.

Step 2: 5-(Benzofuran-2-yl)pyrimidin-2-amine (50 mg, 0.21 mmol), 3-bromo-/V-((5- methylfuran-2-yl)methyl)benzamide (75 mg, 0.26 mmol), Pd2(dba)3 (26 mg, 0.021 mmol), BrettPhos (23 mg, 0.043 mmol), and cesium carbonate (154 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 111, 3-{[5-(l-benzofuran-2-yl)pyrimidin-2-yl]amino}-/V-[(5- methylfuran-2-yl)methyl]benzamide (33 mg, 37%) as a white solid.

Synthesis of Compound 112

Step 1: 4,4,5,5-Tetramethyl-2-(2-methylfuran-3-yl)-l,3,2-dioxaborola ne (167 mg, 1.03 mmol), 5-bromopyrimidin-2-amine (150 mg, 0.86 mmol), Pd(PPli3)4 (50 mg, 0.043 mmol), and potassium carbonate (357 mg, 2.59 mmol) were mixed in EEO/DMF (1.7/1.7 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC to give 5-(2-methylfuran-3-yl)pyrimidin-2-amine (154 mg, >99%) as a yellowish white solid.

Step 2: 5-(2-Methylfuran-3-yl)pyrimidin-2-amine (60 mg, 0.23 mmol), 3-bro o-A-((5- methylfuran-2-yl)methyl)benzamide (80 mg, 0.27 mmol), Pd2(dba)3 (28 mg, 0.023 mmol), BrettPhos (24 mg, 0.046 mmol), and cesium carbonate (148 mg, 0.45 mmol) were mixed in 1,4-dioxane (1.1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 112, A-[(5-methylfuran-2-yl)methyl]-3-{[5-(2-methylfuran-3- yl)pyrimidin-2-yl]amino}benzamide (6 mg, 6%) as a white solid.

Synthesis of Compound 113 Step 1: 2-Bromothiazole-5-carboxylic acid (416 mg, 2 mmol), 2-phenylethan-l -amine (0.28 mL, 2.2 mmol), and //-(benzotri azol - 1 -y 1 )-A f , N, N N -tetram ethyl uroni u tetrafluorob orate (1.2 g, 4 mmol) were dissolved in DMF (20 mL), followed up by addition of DIPEA (0.7 mL, 4 mmol) and stirred for 5 hours at room temperature. The reaction mixture was extracted by EA and 5% aq. LiCl. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by MPLC to give 2-bromo-A-phenethylthiazole-5- carboxamide (410 mg, 46%) as a white solid.

Step 2: 6-Phenylpyridazin-3 -amine (30 mg, 0.18 mmol), 2-bromo-A-phenethylthiazole- 5-carboxamide (65 mg, 0.21 mmol), Pd2(dba)3 (21 mg, 0.018 mmol), BrettPhos (19 mg, 0.035 mmol), and cesium carbonate (114 mg, 0.35 mmol) were mixed in 1,4-dioxane (1 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC to give compound 113, A-(2-phenylethyl)-2-[(6-phenylpyridazin-3- yl)amino]-l,3-thiazole-5-carboxamide (9 mg, 6%) as a brown foam.

Synthesis of Compound 114

5-Phenylpyridin-2-amine (40 mg, 0.24 mmol), 3-bromo-A-phenethylbenzamide (86 mg, 0.28 mmol), Pd2(dba)3 (22 mg, 0.024 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (153 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 114, N- (2-phenylethyl)-3-[(5-phenylpyridin-2-yl)amino]benzamide (18 mg, 20%) as a white solid.

Synthesis of Compound 115

5-Phenylpyridin-2-amine (40 mg, 0.24 mmol), 3-bromo-/V-((lf?,2A)-2- phenylcyclopropyl)benzamide (111 mg, 0.35 mmol), Pd2(dba)3 (22 mg, 0.024 mmol), BrettPhos (25 mg, 0.047 mmol), and cesium carbonate (153 mg, 0.47 mmol) were mixed in 1,4-dioxane (1.2 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using EA to give compound 115, A-[(li?,2,S)-2-phenylcyclopropyl]-3-[(5-phenylpyridin-2- yl)amino]benzamide (26 mg, 27%) as a white solid.

2. Synthesis by Method B

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (530 mg, 1.8 mmol), 2- phenylcyclopropan-1 -amine (267 mg, 2 mmol) and hexafluorophosphate benzotriazole tetramethyl uronium (HBTU) (1 g, 2.7 mmol) were dissolved in DMF (18 mL), followed up by addition of DIPEA (0.95 mL, 5.5 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 67, A-(2-phenylcyclopropyl)-3-[(6- phenylpyridazin-3-yl)amino]benzamide (167 mg, 23%) as a beige solid.

Synthesis of Compound 68

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (500 mg, 1.72 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (320 mg, 1.89 mmol), and HBTU (976 mg, 2.57 mmol) were dissolved in DMF (17 mL), followed up by addition of DIPEA (0.9 mL, 5.2 mmol) and stirred for 20 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The reaction mixture was solidified by using EA and DCM to give compound 68, N-[(lR,2S)-2- phenylcyclopropyl]-3-[(6-phenylpyridazin-3-yl)amino]benzamid e (376 mg, 54%) as a white solid.

Synthesis of Compound 79

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (550 mg, 1.78 mmol), 2-(3- (trifluoromethyl)phenyl)ethan-l -amine (308 mg, 1.96 mmol), and HBTU (1 g, 2.67 mmol) were dissolved in DMF (18 mL), followed up by addition of DIPEA (0.46 mL, 2.67 mmol) and stirred for 20 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA to give compound 79, 3-{[5-(3-fhiorophenyl)pyrimidin-2-yl]amino}-/V- {2-[3-(trifluoromethyl)phenyl]ethyl}benzamide (456 mg, 53%) as a white solid.

Synthesis of Compound 81

3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (600 mg, 2.06 mmol), 2- cyclohexylethan-1 -amine (288 mg, 2.27 mmol), and HBTU (1.2 g, 3.09 mmol) were dissolved in DMF (21 mL), followed up by addition of DIPEA (0.54 mL, 3.09 mmol) and stirred for 20 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA to give compound 81, /V-(2-cyclohexylethyl)-3-[(5-phenylpyrimidin-2- yl)amino]benzamide (395 mg, 48%) as a white solid.

Synthesis of Compound 93

3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (30 mg, 0.1 mmol), 2-(piperidin-l- yl)ethan-l -amine (14.5 mg, 0.11 mmol), and HBTU (59 mg, 0.15 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.027 mL, 0.15 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCC . The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 93, 3-[(5-phenylpyrimidin-2- yl)amino]-/V-[2-(piperidin-l-yl)ethyl]benzamide (24 mg, 59%) as a beige solid.

Synthesis of Compound 94

3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (30 mg, 0.1 mmol), 2-(pyrrolidin-l- yl)ethan-l -amine (13 mg, 0.11 mmol), and HBTU (59 mg, 0.15 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.027 mL, 0.15 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCCb. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 94, 3-[(5-phenylpyrimidin-2-yl)amino]-/V-[2- (pyrrolidin-l-yl)ethyl]benzamide (29 mg, 73%) as a beige solid.

Synthesis of Compound 95

3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (30 mg, 0.1 mmol), N^N 1 - dimethylethane- 1,2-diamine (12 mg, 0.11 mmol), and HBTU (59 mg, 0.15 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.027 mL, 0.15 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCCU The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 95, N-[2- (dimethylamino)ethyl]-3-[(5-phenylpyrimidin-2-yl)amino]benza mide (28 mg, 76%) as a white solid.

Synthesis of Compound 96 3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (30 mg, 0.1 mmol), N^N 1 - di ethyl ethane- 1,2-diamine (13 mg, 0.11 mmol), and HBTU (59 mg, 0.15 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.027 mL, 0.15 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCC . The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 96, A-[2-(diethylamino)ethyl]-3-[(5- phenylpyrimidin-2-yl)amino]benzamide (29 mg, 73%) as a white solid.

Synthesis of Compound 97

3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (30 mg, 0.1 mmol), 2-(4- methylpiperazin-l-yl)ethan-l -amine (16 mg, 0.11 mmol), and HBTU (59 mg, 0.15 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.027 mL, 0.15 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCC . The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 97, N-[2-(4- methylpiperazin-l-yl)ethyl]-3-[(5-phenylpyrimidin-2-yl)amino ]benzamide (36 mg, 83%) as a white solid.

Synthesis of Compound 105 3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (30 mg, 0.1 mmol), 2-(2- azabicyclo[2.2.1]heptan-2-yl)ethan-l -amine (16 mg, 0.11 mmol), and HBTU (59 mg, 0.15 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.027 mL, 0.15 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCCb. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA to give compound 105, N-( 2-{2- azabicyclo[2.2.1]heptan-2-yl}ethyl)-3-[(5-phenylpyrimidin-2- yl)amino]benzamide (13 mg,

31%) as a beige solid.

Synthesis of Compound 106

3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (30 mg, 0.1 mmol), 2- (benzo[ ][l,3]dioxol-5-yl)ethan-l-amine hydrochloride (23 mg, 0.11 mmol), and HBTU (59 mg, 0.15 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.027 mL, 0.15 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCCb. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA to give compound 106, N-[2-(2H-l,3- benzodioxol-5-yl)ethyl]-3-[(5-phenylpyrimidin-2-yl)amino]ben zamide (26 mg, 58%) as a beige solid.

Synthesis of Compound 116

3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (35 mg, 0.12 mmol), 3-fluoroaniline (15 mg, 0.13 mmol), and HBTU (68 mg, 0.18 mmol) were dissolved in DMF (1.2 mL), followed up by addition of DIPEA (0.03 mL, 0.18 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 116, /V-(3-fluorophenyl)-3-[(5-phenylpyrimidin-2-yl)amino]benzami de (32 mg, 69%) as a white solid. Synthesis of Compound 117

3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (35 mg, 0.12 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (45 mg, 0.26 mmol), and HBTU (68 mg, 0.18 mmol) were dissolved in DMF (1.2 mL), followed up by addition of DIPEA (0.03 mL, 0.18 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX. The crude mixture was purified by MPLC to give compound 117, /V-[(1R,2S)-2-phenylcyclopropyl]-3-[(5-phenylpyrimidin-2- yl)amino]benzamide (29 mg, 59%) as a white solid.

Synthesis of Compound 118

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (35 mg, 0.11 mmol), 3,4- dichloroaniline (20 mg, 0.12 mmol), and HBTU (64 mg, 0.17 mmol) were dissolved in DMF (1.1 mL), followed up by addition of DIPEA (0.03 mL, 0.17 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 118, /V-(3,4-dichlorophenyl)-3-{[5-(3-fluorophenyl)pyrimidin-2- yl]amino}benzamide (18 mg, 35%) as a beige solid. Synthesis of Compound 119

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (35 mg, 0.11 mmol), (1R,2S)-2-phenylcyclopropan-l -amine hydrochloride (21 mg, 0.12 mmol), and HBTU (64 mg, 0.17 mmol) were dissolved in DMF (1.1 mL), followed up by addition of DIPEA (0.05 mL, 0.28 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was solidified by using EA to give compound 119, 3-{[5-(3-fhiorophenyl)pyrimidin-2- yl]amino}-/V-[(1R,2S)-2-phenylcyclopropyl]benzamide (37 mg, 77%) as a white solid.

Synthesis of Compound 120

3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (35 mg, 0.12 mmol), 1- benzylpiperidin-4-amine (25 mg, 0.13 mmol), and HBTU (68 mg, 0.18 mmol) were dissolved in DMF (1.2 mL), followed up by addition of DIPEA (0.03 mL, 0.18 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 120, /V-(l-benzylpiperidin-4-yl)-3-[(5-phenylpyrimidin-2- yl)amino]benzamide (34 mg, 62%) as a beige solid.

Synthesis of Compound 121

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (35 mg, 0.11 mmol), 2- phenylethan-1 -amine (15 mg, 0.12 mmol), and HBTU (64 mg, 0.17 mmol) were dissolved in DMF (1.1 mL), followed up by addition of DIPEA (0.03 mL, 0.17 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 121, 3-{[5-(3-fluorophenyl)pyridin-2-yl]amino}-/V-(2- phenylethyl)benzamide (35 mg, 75%) as a white solid.

Synthesis of Compound 122 3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (35 mg, 0.11 mmol), 3- fluoroaniline (14 mg, 0.12 mmol), and HBTU (64 mg, 0.17 mmol) were dissolved in DMF (1.1 mL), followed up by addition of DIPEA (0.03 mL, 0.17 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 122, /V-(3-fluorophenyl)-3-{[5-(3-fluorophenyl)pyridin-2- yl]amino}benzamide (20 mg, 44%) as a beige solid.

Synthesis of Compound 123

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (35 mg, 0.11 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (19 mg, 0.12 mmol), and HBTU (64 mg, 0.17 mmol) were dissolved in DMF (1.1 mL), followed up by addition of DIPEA (0.06 mL, 0.34 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 123, 3-{[5-(3-fluorophenyl)pyridin-2- yl]amino}-/V-[(1R,2S)-2-phenylcyclopropyl]benzamide (31 mg, 64%) as a white solid.

Synthesis of Compound 124

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (35 mg, 0.12 mmol), A ; -benzyl -A ; - methylethane- 1,2-diamine (22 mg, 0.13 mmol), and HBTU (68 mg, 0.18 mmol) were dissolved in DMF (1.2 mL), followed up by addition of DIPEA (0.03 mL, 0.18 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using acetonitrile (ACN) to give compound 124, N-{ 2- [benzyl(methyl)amino]ethyl}-3-[(6-phenylpyridazin-3-yl)amino ]benzamide (8 mg, 15%) as a white solid.

Synthesis of Compound 125

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (60 mg, 0.19 mmol), (5- methylfuran-2-yl)methanamine (24 mg, 0.21 mmol), and HBTU (111 mg, 0.29 mmol) were dissolved in DMF (1.9 mL), followed up by addition of DIPEA (0.05 mL, 0.29 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 125, 3-{[5-(3-fluorophenyl)pyridin-2- yl]amino}-A-[(5-methylfuran-2-yl)methyl]benzamide (46 mg, 59%) as a yellowish white solid.

Synthesis of Compound 126

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (200 mg, 0.65 mmol), methyl 4-(2-aminoethyl)benzoate hydrochloride (153 mg, 0.71 mmol), and HBTU (368 mg, 0.97 mmol) were dissolved in DMF (6.5 mL), followed up by addition of DIPEA (0.34 mL, 1.94 mmol) and stirred for 18 h at room temperature. The residue was solidified by using EA to give compound 126, methyl 4-(2-(3-((5-(3-fluorophenyl)pyrimidin-2- yl)amino)benzamido)ethyl)benzoate (242 mg, 92%) as a white solid.

Synthesis of Compound 127

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (200 mg, 0.65 mmol), methyl 2-(2-aminoethyl)benzoate hydrochloride (153 mg, 0.71 mmol), and HBTU (368 mg, 0.97 mmol) were dissolved in DMF (6.5 mL), followed up by addition of DIPEA (0.34 mL, 1.94 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The white solid s precipitated out of the solution, and the solution was filtered to give compound 127, methyl 2-(2-(3-((5-(3-fluorophenyl)pyrimidin-2- yl)amino)benzamido)ethyl)benzoate (150 mg, 32%) as a white solid. Synthesis of Compound 128

3-((5-Phenylpyridin-2-yl)amino)benzoic acid (61 mg, 0.21 mmol), 3-fluoroaniline (26 mg, 0.23 mmol), and HBTU (119 mg, 0.32 mmol) were dissolved in DMF (2.1 mL), followed up by addition of DIPEA (0.055 mL, 0.32 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was solidified by using EA to give compound 128, N- (3-fluorophenyl)-3-((5-phenylpyridin-2-yl)amino)benzamide (38 mg, 47%) as a white solid.

Synthesis of Compound 129

Step 1: 3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (200 mg, 0.65 mmol), methyl 3-(2-aminoethyl)benzoate hydrochloride (153 mg, 0.71 mmol), and HBTU (368 g, 0.97 mmol) were dissolved in DMF (6.5 mL), followed up by addition of DIPEA (0.34 mL, 1.94 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA to give methyl 3-(2-(3-((5-(3-fluorophenyl)pyrimidin-2- yl)amino)benzamido)ethyl)benzoate (281mg, 107%) as a white solid.

Step 2: Methyl 3-(2-(3-((5-(3-fluorophenyl)pyrimidin-2- yl)amino)benzamido)ethyl)benzoate (100 mg, 0.21 mmol) and LiOH-HiO (89.2 mg, 2.13 mmol) were mixed in H 2 0/l,4-dioxane (0.89/4.25 mL) and stirred for 18 hours at 40°C. Then pH value of the solution was adjusted to 1-2 by 1 N HC1. The crude product was added into water. The suspension was filtered, and the filter cake was washed with water. The filter cake was dried under vacuum to give compound 129, 3-(2-(3-((5-(3-fluorophenyl)pyrimidin-2- yl)amino)benzamido)ethyl)benzoic acid (53 mg, 55%) as a yellowish white solid. Synthesis of Compound 130

Methyl 4-(2-(3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)benzamido)e thyl)benzoate (100 mg, 0.21 mmol) and Li0H-H 2 0 (89.2 mg, 2.13 mmol) were mixed in H 2 0/l,4-dioxane (0.89/4.25 mL) and stirred for 42 hours at 40°C. Then pH value of the solution was adjusted to 1-2 by 1 N HC1. The crude product was added into water. The suspension was filtered, and the filter cake was washed with water. The crude product was added into EA.

The suspension was filtered, and the filter cake was washed with EA. The filter cake was dried under vacuum to give compound 130, 4-(2-(3-((5-(3-fluorophenyl)pyrimidin-2- yl)amino)benzamido)ethyl)benzoic acid (84 mg, 87%) as a white solid.

Synthesis of Compound 131

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (150 mg, 0.48 mmol), (li?,2ri)-2-(4-chloro-3-fluorophenyl)cyclopropan-l-amine hydrochloride (118 mg, 0.53 mmol) and HBTU (276 mg, 0.73 mmol) were dissolved in DMF (4.8 mL), followed up by addition of DIPEA (0.25 mL, 1.45 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The white solid was precipitated out of the solution, and the solution was filtered to give compound 131, L -((///, 2k)-2-(4-chloro-3- fluorophenyl)cyclopropyl)-3-((5-(3-fluorophenyl)pyrimidin-2- yl)amino)benzamide (72 mg, 31%) as a white solid.

Synthesis of Compound 132

Methyl 2-(2-(3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)benzamido)e thyl)benzoate (100 mg, 0.21 mmol) and LiOH-HiO (89.2 mg, 2.13 mmol) were mixed in H 2 0/l,4-dioxane (0.89/4.25 mL) and stirred for 42 hours at 40°C. The reaction mixture acidified by adding 1 N HC1 and extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 132, 2-(2-(3- ((5-(3-fluorophenyl)pyrimidin-2-yl)amino)benzamido)ethyl)ben zoic acid (80 mg, 82%) as a white solid.

Synthesis of Compound 133

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 1- phenylcyclopropan-1 -amine (47 mg, 0.36 mmol), and HBTE1 (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.48 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 133, 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)- A-(l-phenylcyclopropyl)benzamide (83 mg, 60%) as a white solid.

Synthesis of Compound 134

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 1- phenylcyclopropan-1 -amine (47 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.48 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 134, 3-((5-(3-fluorophenyl)pyridin-2-yl)amino)-/V- (l-phenylcyclopropyl)benzamide (93 mg, 68%) as a white solid.

Synthesis of Compound 135 3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 4- ((4-methylpiperazin-l-yl)methyl)aniline (73 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.48 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCC . The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 135, 3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)-A-(4-((4-methylpiperazin- l -yl)methyl)phenyl)benzamide (116 mg, 73%) as a beige solid.

Synthesis of Compound 136

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 3- aminobenzonitrile (42 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.48 mmol) and stirred for 3 days at 45°C. The reaction mixture was extracted by EA and brine. The beige solid was precipitated out of the solution, and the solution was filtered to give compound 136, A-(3-cyanophenyl)-3- ((5-(3-fluorophenyl)pyrimidin-2-yl)amino)benzamide (56 mg, 42%) as a beige solid.

Synthesis of Compound 137

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 3- nitroaniline (49 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.48 mmol) and stirred for 3 days at 45°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 137, 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)-/V-(3-nitropheny l)benzamide (57 mg, 41%) as a yellow solid.

Synthesis of Compound 138

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), thiazol-2-amine (36 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.48 mmol) and stirred for 18 h at room temperature. The white solid was precipitated out of the solution. The crude product was added into EA, and the solution was filtered to give compound 138, 3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)-A-(thiazol-2-yl)benzamide (59 mg, 46%) as a white solid.

Synthesis of Compound 139

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 2-(l- methylpiperidin-4-yl)ethan-l -amine (51 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.49 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA and HEX to give compound 139, 3-((5-(3-fluorophenyl)pyridin-2- yl)amino)-A-(2-(l -methylpiperidin-4-yl)ethyl)benzamide (105 mg, 75%) as a yellowish white solid.

Synthesis of Compound 140

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), (1- methylpiperidin-4-yl)methanamine (46 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.49 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA and HEX to give compound 140, 3-((5-(3-fluorophenyl)pyridin-2- yl)amino)-A-(( l -methyl pi peri din-4-yl)methyl)benzamide (34 mg, 25%) as a white solid.

Synthesis of Compound 141

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (200 mg, 0.65 mmol), 3- nitroaniline (99 mg, 0.71 mmol), and HBTU (369 mg, 0.97 mmol) were dissolved in DMF (6.5 mL), followed up by addition of DIPEA (0.17 mL, 0.97 mmol) and stirred for 2 days at 50°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using ACN to give compound 141, 3-((5-(3-fluorophenyl)pyridin-2-yl)amino)-/V-(3-nitrophenyl) benzamide (49 mg, 16%) as a yellow solid.

Synthesis of Compound 142

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), l-(2- aminoethyl)adamantane (46 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.49 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using ACN to give compound 142, A-(2-(adamantan-l-yl)ethyl)-3-((5-(3-fluorophenyl)pyridin-2- yl)amino)benzamide (71 mg, 47%) as a beige solid.

Synthesis of Compound 143

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), benzene- 1,2-diamine (39 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.49 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 143, /V-(2-aminophenyl)-3-((5-(3-fluorophenyl)pyridin-2- yl)amino)benzamide (72 mg, 56%) as a white solid.

Synthesis of Compound 144

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 2-(l- methylpiperidin-4-yl)cyclopropan-l -amine (55 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.49 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA and HEX to give compound 144, 3-((5-(3- fluorophenyl)pyridin-2-yl)amino)-A-(2-(l -methyl pi peri din-4-yl)cy cl opropyl)benzamide (10 mg, 7%) as a white solid.

Synthesis of Compound 145

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), (1- methylpyrrolidin-3-yl)methanamine (41 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.49 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCCb. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using DCM and HEX to give compound 145, 3-((5-(3- fluorophenyl)pyridin-2-yl)amino)-A-((l -methylpyrrolidin-3-yl)methyl)benzamide (19 mg, 15%) as a beige solid.

Synthesis of Compound 146

3-((5-(3-Fluorophenyl)pyridin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), benzene- 1,4-diamine (39 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.49 mmol) and stirred for 2 days at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using DCM and MeOH to give compound 146, A-(4-aminophenyl)-3-((5-(3-fluorophenyl)pyridin-2- yl)amino)benzamide (15 mg, 12%) as a beige solid.

Synthesis of Compound 147

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 3- (2-aminoethyl)aniline (48 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.49 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCCU The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA and the solution was filtered. The filtrate was concentrated and solidified by using EA and HEX to give compound 147, A-(3-aminophenethyl)-3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)benzamide (36 mg, 26%) as a beige solid.

Synthesis of Compound 148

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 2- (2-aminoethyl)aniline (48 mg, 0.36 mmol) and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.49 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCCU The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA and the solution was filtered. The filtrate was concentrated and solidified by using EA and HEX to give compound 148, A-(2-aminophenethyl)-3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)benzamide (49 mg, 36%) as a beige solid.

Synthesis of Compound 149

2-((5-Phenylpyridin-2-yl)amino)isonicotinic acid (60 mg, 0.21 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (38 mg, 0.23 mmol), and HBTU (117 mg, 0.31 mmol) were dissolved in DMF (2.1 mL), followed up by addition of DIPEA (0.088 mL, 0.51 mmol) and stirred for 2 days at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA to give compound 149, /V-((1R,2S)-2-phenylcyclopropyl)-2-((5- phenylpyridin-2-yl)amino)isonicotinamide (51 mg, 60%) as a yellowish white solid.

Synthesis of Compound 150

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 2- (l,5-dimethyl-liT-pyrazol-4-yl)cyclopropan-l-amine dihydrochloride (87 mg, 0.39 mmol) and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.2 mL, 1.13 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCCb. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The crude mixture was solidified by using EA to give compound 150, A-(2-(l, 5-dimethyl- liT-pyrazol-4-yl)cyclopropyl)-3-((5-(3-fluorophenyl)pyrimidi n-2- yl)amino)benzamide (102 mg, 71%) as a white solid.

Synthesis of Compound 151 3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 2- (5-methylfuran-2-yl)ethan-l-amine (45 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.08 mL, 0.48 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 151, 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)- A-(2-(5-methylfuran-2-yl)ethyl)benzamide (65 mg, 49%) as a beige solid.

Synthesis of Compound 152

3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (100 mg, 0.34 mmol), 2-(l ,5- dimethyl- l.i -pyrazol-4-yl)cy clopropan- 1 -amine dihydrochloride (92 mg, 0.41 mmol), and HBTU (195 mg, 0.51 mmol) were dissolved in DMF (3.4 mL), followed up by addition of DIPEA (0.21 mL, 1.2 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and saturated aq. NaHCCb. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 152, N-( 2- (l,5-dimethyl-liT-pyrazol-4-yl)cyclopropyl)-3-((5-phenylpyri midin-2-yl)amino)benzamide (107 mg, 73%) as a beige solid.

Synthesis of Compound 153

3-((5-Phenylpyrimidin-2-yl)amino)benzoic acid (100 mg, 0.34 mmol), 2-(5- methylfuran-2-yl)ethan-l -amine (52 mg, 0.41 mmol), and HBTU (195 mg, 0.51 mmol) were dissolved in DMF (3.4 mL), followed up by addition of DIPEA (0.09 mL, 0.51 mmol) and stirred for 18 h at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 153, A-(2-(5-methylfuran-2-yl)ethyl)-3-((5- phenylpyrimidin-2-yl)amino)benzamide (87 mg, 64%) as a beige solid.

Synthesis of Compound 154

3-((4-(Pyridin-2-yl)phenyl)amino)benzoic acid (50 mg, 0.17 mmol), 3-fluoroaniline (0.018 mL, 0.19 mmol), and HBTU (98 mg, 0.26 mmol) were dissolved in DMF (1.7 mL), followed up by addition of DIPEA (0.045 mL, 0.26 mmol) and stirred for 15.5 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC to give compound 154, /V-(3-fluorophenyl)-3-((4-(pyridin-2-yl)phenyl)amino)benzami de (39 mg, 59%) as a white solid.

Synthesis of Compound 155

3-((4-(Pyridin-2-yl)phenyl)amino)benzoic acid (50 mg, 0.17 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (32 mg, 0.19 mmol), and HBTU (98 mg, 0.26 mmol) were dissolved in DMF (1.7 mL), followed up by addition of DIPEA (0.045 mL, 0.26 mmol) and stirred for 15.5 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC to give compound 155, A-((li?,2ri)-2-phenylcyclopropyl)-3- ((4-(pyridin-2-yl)phenyl)amino)benzamide (66 mg, 95%) as a yellow solid.

Synthesis of Compound 156

5-((5-Phenylpyrimidin-2-yl)amino)nicotinic acid (100 mg, 0.34 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (64 mg, 0.38 mmol), and HBTU (194 mg, 0.51 mmol) were dissolved in DMF (3.4 mL), followed up by addition of DIPEA (0.179 mL, 1 mmol) and stirred for 18 hours at room temperature. The white solid was precipitated out of the solution, and the solution was filtered to give compound 156, N-((lR,2S)-2- phenylcyclopropyl)-5-((5-phenylpyrimidin-2-yl)amino)nicotina mide (111 mg, 80%) as a white solid.

Synthesis of Compound 157

3-((5-(Furan-3-yl)pyrimidin-2-yl)amino)benzoic acid (60 mg, 0.21 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (40 mg, 0.23 mmol), and HBTU (121 mg, 0.32 mmol) were dissolved in DMF (2.1 mL), followed up by addition of DIPEA (0.11 mL, 0.64 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 157, 3-((5-(furan-3-yl)pyrimidin-2- yl)amino)-/V-((1R,2S)-2-phenylcyclopropyl)benzamide (60 mg, 71%) as a beige solid.

Synthesis of Compound 158

Compound 158

3-((5-Phenyl-l,3,4-oxadiazol-2-yl)amino)benzoic acid (50 mg, 0.18 mmol), 3- fluoroaniline (0.019 mL, 0.20 mmol), and HBTU (101 mg, 0.27 mmol) were dissolved in DMF (1.8 mL), followed up by addition of DIPEA (0.046 mL, 0.27 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCE and concentrated. The residue was purified by MPLC and solidified by using acetone to give compound 158, A-(3-fluorophenyl)-3-((5-phenyl-l,3,4- oxadiazol-2-yl)amino)benzamide (24 mg, 36%) as a white solid.

Synthesis of Compound 159

3-((5-Phenyl-l,3,4-oxadiazol-2-yl)amino)benzoic acid (50 mg, 0.18 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (34 mg, 0.20 mmol), and HBTU (101 mg, 0.27 mmol) were dissolved in DMF (1.8 mL), followed up by addition of DIPEA (0.046 mL, 0.27 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCE and concentrated. The crude mixture was solidified by using acetone to give compound 159, 3-((5-phenyl-l,3,4-oxadiazol- 2-yl)amino)-A-(( l A2A)-2-phenyl cyclopropyl )benzamide (33 mg, 47%) as a yellow solid.

Synthesis of Compound 160

3-((4-(Pyridin-3-yl)phenyl)amino)benzoic acid (50 mg, 0.17 mmol), 3-fluoroaniline (0.018 mL, 0.19 mmol), and HBTU (98 mg, 0.26 mmol) were dissolved in DMF (1.7 mL), followed up by addition of DIPEA (0.045 mL, 0.26 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCE and concentrated. The residue was purified by MPLC to give compound 160, A-(3-fluorophenyl)-3-((4-(pyri din-3 -yl)phenyl)amino)benzamide (21 mg, 30%) as a yellow solid.

Synthesis of Compound 161

3-((4-(Pyridin-3-yl)phenyl)amino)benzoic acid (50 mg, 0.17 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (32 mg, 0.19 mmol), and HBTU (98 mg, 0.26 mmol) were dissolved in DMF (1.7 mL), followed up by addition of DIPEA (0.045 mL, 0.26 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCE and concentrated. The residue was purified by MPLC to give compound 161, /V-((1R,2S)-2-phenylcyclopropyl)-3-((4- (pyridin-3-yl)phenyl)amino)benzamide (33 mg, 47%) as a yellow solid.

Synthesis of Compound 162

3-((4-(Pyridin-4-yl)phenyl)amino)benzoic acid (50 mg, 0.17 mmol), 3-fluoroaniline (0.018 mL, 0.19 mmol), and HBTU (98 mg, 0.26 mmol) were dissolved in DMF (1.7 mL), followed up by addition of DIPEA (0.045 mL, 0.26 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCE and concentrated. The residue was purified by MPLC to give compound 162, /V-(3-fluorophenyl)-3-((4-(pyridin-4-yl)phenyl)amino)benzami de (19 mg, 29%) as a brown solid.

Synthesis of Compound 163

3-((4-(Pyridin-4-yl)phenyl)amino)benzoic acid (50 mg, 0.17 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (32 mg, 0.19 mmol), and HBTU (98 mg, 0.26 mmol) were dissolved in DMF (1.7 mL), followed up by addition of DIPEA (0.045 mL, 0.26 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCE and concentrated. The residue was purified by MPLC to give compound 163, A-((li?,2ri)-2-phenylcyclopropyl)-3-((4- (pyridin-4-yl)phenyl)amino)benzamide (26 mg, 37%) as a yellow solid.

Synthesis of Compound 164

3-((4-(Pyrimidin-5-yl)phenyl)amino)benzoic acid (50 mg, 0.17 mmol), 3-fluoroaniline (0.018 mL, 0.19 mmol), and HBTU (98 mg, 0.26 mmol) were dissolved in DMF (1.7 mL), followed up by addition of DIPEA (0.045 mL, 0.26 mmol) and stirred for 18.5 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated. The crude mixture was solidified by using EA to give compound 164, /V-(3-fluorophenyl)-3-((4-(pyrimidin-5-yl)phenyl)amino)benza mide (50 mg, 75%) as an ivory solid.

Synthesis of Compound 165 3-((4-(Pyrimidin-5-yl)phenyl)amino)benzoic acid (50 mg, 0.17 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (32 mg, 0.19 mmol), and HBTU (98 mg, 0.26 mmol) were dissolved in DMF (1.7 mL), followed up by addition of DIPEA (0.045 mL, 0.26 mmol) and stirred for 18.5 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 165, N-((lR,2S)-2- phenylcyclopropyl)-3-((4-(pyrimidin-5-yl)phenyl)amino)benzam ide (66 mg, 94%) as a bright pink solid.

Synthesis of Compound 166 3 -((6-Phenylpyridazin-3-yl)amino)adamantane-l -carboxylic acid (100 mg, 0.26 mmol), (1R,2S)-2-phenylcyclopropan-l -amine hydrochloride (53 mg, 0.31 mmol), and HBTU (163 mg, 0.43 mmol) were dissolved in DMF (2.9 mL), followed up by addition of DIPEA (0.149 mL, 0.86 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was solidified by using EA and HEX to give compound 166, N-

((IR,2S)- 2-phenylcyclopropyl)-3-((6-phenylpyridazin-3-yl)amino)adaman tane-l-carboxamide (70 mg, 53%) as a beige solid.

Synthesis of Compound 167 4-((5-Phenylpyrimidin-2-yl)amino)picolinic acid (100 mg, 0.35 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (66 mg, 0.39 mmol), and HBTU (201 mg, 0.53 mmol) were dissolved in DMF (3.5 mL), followed up by addition of DIPEA (0.185 mL, 1.06 mmol) and stirred for 18 hours at room temperature. The white solid was precipitated out of the solution, and the solution was filtered to give compound 167, N-((lR,2S)-2- phenylcyclopropyl)-4-((5-phenylpyrimidin-2-yl)amino)picolina mide (97 mg, 67%) as a white solid.

Synthesis of Compound 168

(1.v,4.v)-4-((6-Phenylpyridazin-3-yl)amino)bicyclo[2.2. 1 Jheptane- 1 -carboxylic acid (100 mg, 0.32 mmol), (1R,2S)-2-phenylcyclopropan-l -amine hydrochloride (60 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.169 mL, 0.97 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was solidified by using EA and HEX to give compound 168, (1 s,4s)-iV-(( 1R,2S)-2-phenylcy clopropyl)-4-((6-phenylpyridazin-3 - yl)amino)bicyclo[2.2.1]heptane-l-carboxamide (91 mg, 66%) as a beige solid.

Synthesis of Compound 170

3-((4-(Pyrimidin-2-yl)phenyl)amino)benzoic acid (146 mg, 0.5 mmol), 3-fluoroaniline (0.053 mL, 0.55 mmol), and HBTU (284 mg, 0.75 mmol) were dissolved in DMF (5 mL), followed up by addition of DIPEA (0.13 mL, 0.75 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCE and concentrated. The residue was purified by MPLC to give compound 170, /V-(3-fluorophenyl)-3-((4-(pyrimidin-2-yl)phenyl)amino)benza mide (111 mg, 57%) as a pale yellow solid.

Synthesis of Compound 171

3-((4-(Pyrimidin-2-yl)phenyl)amino)benzoic acid (146 mg, 0.5 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (94 mg, 0.55 mmol), and HBTU (284 mg, 0.75 mmol) were dissolved in DMF (5 mL), followed up by addition of DIPEA (0.13 mL, 0.75 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCL and concentrated. The residue was purified by MPLC to give compound 171, /V-((1R,2S)-2-phenylcyclopropyl)-3-((4- (pyrimidin-2-yl)phenyl)amino)benzamide (98 mg, 48%) as a white solid.

Synthesis of Compound 172

3-((4-(Pyrazin-2-yl)phenyl)amino)benzoic acid (146 mg, 0.5 mmol), 3-fluoroaniline (0.053 mL, 0.55 mmol), and HBTU (284 mg, 0.75 mmol) were dissolved in DMF (5 mL), followed up by addition of DIPEA (0.13 mL, 0.75 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCE and concentrated. The residue was purified by MPLC to give compound 172, /V-(3-fluorophenyl)-3-((4-(pyrazin-2-yl)phenyl)amino)benzami de (29 mg, 15%) as a pale yellow solid.

Synthesis of Compound 173

3-((4-(Pyrazin-2-yl)phenyl)amino)benzoic acid (146 mg, 0.5 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (94 mg, 0.55 mmol), and HBTU (284 mg, 0.75 mmol) were dissolved in DMF (5 mL), followed up by addition of DIPEA (0.13 mL, 0.75 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCL and concentrated. The residue was purified by MPLC to give compound 173, /V-((li?,2ri)-2-phenylcyclopropyl)-3-((4- (pyrazin-2-yl)phenyl)amino)benzamide (66 mg, 32%) as a white solid.

Synthesis of Compound 174 3-((4-(Pyrimidin-4-yl)phenyl)amino)benzoic acid (64 mg, 0.22 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (41 mg, 0.24 mmol), and HBTU (125 mg, 0.33 mmol) were dissolved in DMF (2.2 mL), followed up by addition of DIPEA (0.057 mL, 0.33 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCL and concentrated. The residue was purified by MPLC to give compound 174, /V-((li?,2ri)-2-phenylcyclopropyl)-3-((4- (pyrimidin-4-yl)phenyl)amino)benzamide (44 mg, 50%) as a white solid.

Synthesis of Compound 175

3-((4-(Pyrimidin-4-yl)phenyl)amino)benzoic acid (64 mg, 0.22 mmol), 3-fluoroaniline (0.023 mL, 0.24 mmol), and HBTU (125 mg, 0.33 mmol) were dissolved in DMF (2.2 mL), followed up by addition of DIPEA (0.057 mL, 0.33 mmol) and stirred for 24 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous MgSCL and concentrated. The residue was purified by MPLC to give compound 175, /V-(3-fluorophenyl)-3-((4-(pyrimidin-4-yl)phenyl)amino)benza mide (23 mg, 27%) as an orange solid.

Synthesis of Compound 176

3-((5-(Furan-3-yl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.36 mmol), 3- fluoroaniline (44 mg, 0.39 mmol), and HBTU (202 mg, 0.53 mmol) were dissolved in DMF (3.6 mL), followed up by addition of DIPEA (0.093 mL, 0.53 mmol) and stirred for overnight at room temperature and stirred for overnight at 50°C and stirred for overnight at 70°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The crude mixture was solidified by using EA to give compound 176, /V-(3-fluorophenyl)-3-((5-(furan-3-yl)pyrimidin-2-yl)amino)b enzamide (34 mg, 26%) as an orange brown solid.

Synthesis of Compound 177

/tW- Butyl (3-(3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)benzamido)phe nyl)(methyl) carbamate (90 mg, 0.18 mmol) was dissolved in DCM (1.8 mL), followed up by addition of TFA (0.26 mL) and stirred for 1 hour at room temperature. The reaction mixture was extracted by DCM and aq. NaOH (1 M). The organic layer was dried over anhydrous NaiSCE and concentrated. The reaction mixture was solidified by using EA to give compound 177, 3-((5- (3-fluorophenyl)pyrimidin-2-yl)amino)-A-(3-(methylamino)phen yl)benzamide (52 mg, 72%) as a white solid.

Synthesis of Compound 178

2-((5-Phenylpyrimidin-2-yl)amino)isonicotinic acid (100 mg, 0.34 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (64 mg, 0.38 mmol), and HBTU (195 mg, 0.51 mmol) were dissolved in DMF (3.4 mL), followed up by addition of DIPEA (0.179 mL, 1.03 mmol) and stirred for overnight at room temperature. The white solid was precipitated out of the solution, and the solution was filtered and washed with EA to give compound 178, N- ((li?,2ri)-2-phenylcyclopropyl)-2-((5-phenylpyrimidin-2-yl)a mino)isonicotinamide (113 mg, 81%) as a white solid.

Synthesis of Compound 179

Step 1: (4-Methylthiophen-3-yl)boronic acid (902 mg, 6.35 mmol), 5-bromopyrimidin- 2-amine (850 mg, 4.88 mmol), Pd(PPh3)4 (282 mg, 0.244 mmol), and potassium carbonate (2.03 g, 14.65 mmol) were mixed in H2O/DMF (10/10 mL) and heated in a microwave reactor for 35 minutes at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was solidified by using EA and HEX to give 5-(4-methylthiophen-3-yl)pyrimidin-2-amine (496 mg, 53%) as a beige solid.

Step 2: 5-(4-Methylthiophen-3-yl)pyrimidin-2-amine (490 mg, 2.6 mmol), methyl 3- bromobenzoate (661 mg, 3.07 mmol), Pd 2 (dba) 3 (235 mg, 0.26 mmol), BrettPhos (275 mg,

0.51 mmol), and cesium carbonate (1.67 g, 5.12 mmol) were mixed in 1,4-dioxane (13 mL) and heated in a microwave reactor for 90 minutes at 120°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was solidified by using EA and HEX to give methyl 3-((5-(4-methylthiophen-3- yl)pyrimidin-2-yl)amino)benzoate (364 mg, 44%) as a white solid.

Step 3: Methyl 3-((5-(4-methylthiophen-3-yl)pyrimidin-2-yl)amino)benzoate (350 mg, 1.08 mmol) and LiOH-HiO (451 mg, 10.76 mmol) were mixed in H 2 0/l,4-dioxane (4.5/22 mL) and stirred for overnight at room temperature. Then pH value of the solution was adjusted to 3 by 1 N HC1. The reaction mixture was extracted by EA. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was solidified by using EA and HEX to give 3-((5-(4-methylthiophen-3-yl)pyrimidin-2-yl)amino)benzoic acid (311 mg, 93%) as a white solid.

Step 4: 3-((5-(4-Methylthiophen-3-yl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 3-fluoroaniline (0.039 mg, 0.35 mmol), and HBTE! (183 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.084 mL, 0.48 mmol) and stirred for overnight at 60°C, and stirred for overnight at 70°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The crude mixture was solidified by using EA and HEX to give compound 179, /V-(3-fluorophenyl)-3-((5-(4-methylthiophen-3-yl)pyrimidin-2 -yl)amino)benzamide (46 mg, 36%) as a beige solid.

Synthesis of Compound 181 tert- Butyl 5 -(3 -((5 -(3 -fluorophenyl)pyrimidin-2-yl)amino)benzamido)indoline- 1 - carboxylate (55 mg, 0.1 mmol) was dissolved in DCM (1 mL), followed up by addition of TFA (0.16 mL) and stirred for 1 hour at room temperature. The reaction mixture was extracted by DCM and saturated aq. NaOH (1 M). The organic layer was dried over anhydrous NaiSCL and concentrated. The reaction mixture was solidified by using EA and HEX to give compound 181, 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)-/V-(indolin-5-yl )benzamide (26 mg, 59%) as a grey solid.

Synthesis of Compound 182

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), tert- butyl (3-aminophenyl)(methyl)carbamate (79 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.85 mL, 0.48 mmol) and stirred for overnight at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated. The crude mixture was solidified by using EA and HEX to give compound 182, /ert-butyl (3-(3- ((5-(3-fluorophenyl)pyrimidin-2-yl)amino)benzamido)phenyl)(m ethyl)carbamate (109 mg, 66%) as a beige solid.

Synthesis of Compound 183 3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), tert- butyl 5-aminoisoindoline-2-carboxylate (83 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.085 mL, 0.48 mmol) and stirred for overnight at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA and HEX to give compound 183, /er/-butyl 5-(3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)benzamido)isoindoline-2-ca rboxylate (59 mg, 35%) as a white solid.

Synthesis of Compound 184

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), tert- butyl 5-aminoindoline-l-carboxylate (83 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.085 mL, 0.48 mmol) and stirred for overnight at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The crude mixture was solidified by using EA to give compound 184, /er/-butyl 5-(3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)benzamido)indoline-l-carbo xylate (117 mg, 69%) as a grey solid.

Synthesis of Compound 192

/c/7- Butyl 5-(3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)benzamido)isoi ndoline-2- carboxylate (55 mg, 0.1 mmol) was dissolved in DCM (1 mL), followed up by addition of TFA (0.16 mL) and stirred for 1 hour at room temperature. The reaction mixture was extracted by DCM and saturated aq. NaOH (1 M). The organic layer was dried over anhydrous NaiSCE and concentrated. The reaction mixture was solidified by using EA and HEX, and slurry with MeOH, and filtrate was concentrated to give compound 192, 3-((5-(3-fluorophenyl)pyrimidin- 2-yl)amino)-/V-(isoindolin-5-yl)benzamide (7 mg, 15%) as a white solid. Synthesis of Compound 193

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 5- phenyl-l,3,4-oxadiazol-2-amine (57 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.084 mL, 0.48 mmol) and stirred for overnight at 100°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC to give compound 193, 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)-/V-(5-phenyl- l,3,4-oxadiazol-2-yl)benzamide (15 mg, 10%) as a white solid.

Synthesis of Compound 194

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (100 mg, 0.32 mmol), 5- phenyl-4iT-l,2,4-triazol-3-amine (57 mg, 0.36 mmol), and HBTU (184 mg, 0.48 mmol) were dissolved in DMF (3.2 mL), followed up by addition of DIPEA (0.084 mL, 0.48 mmol) and stirred for overnight at 50°C. The white solid was precipitated out of the solution, and the solution was filtered, and washed with EA to give compound 194, 3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)-/V-(5-phenyl-4iT-l,2,4-tr iazol-3-yl)benzamide (45 mg,

31%) as a white solid.

Synthesis of Compound 195

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), (4-fluoro-3- (trifluoromethyl)phenyl)methanamine (0.02 mL, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 5 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 195, A-(4-fluoro-3-(trifluoromethyl (benzyl )-3 -((6-phenyl pyridazin-3-yl)amino)benzamide (23 mg, 49%) as a white solid.

Synthesis of Compound 196

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), l-(4- fluorophenyl)cyclopropan-l -amine (0.02 mL, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 5 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA, ether, and HEX to give compound 196, A-(l-(4-fluorophenyl)cyclopropyl)-3-((6-phenylpyridazin-3- yl)amino)benzamide (9 mg, 19%) as a white solid.

Synthesis of Compound 197

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (60 mg, 0.21 mmol), (4-(4- methylpiperazin-l-yl)phenyl)methanamine (51 mg, 0.25 mmol), and HBTU (117 mg, 0.31 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.11 mL, 0.62 mmol) and stirred for 18 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 197, /V-(4-(4-methylpiperazin-l-yl)benzyl)-3-((6-phenylpyridazin- 3- yl)amino)benzamide (80 mg, 81%) as a white solid.

Synthesis of Compound 198

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), 2-(4- fluorophenyl)ethan-l -amine (0.02 mL, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 5 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 198, /V-(4-fluorophenethyl)-3-((6-phenylpyridazin-3-yl)amino)benz amide (12 mg, 27%) as a white solid. Synthesis of Compound 199

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), ( \R,2S)-2 - phenylcyclopropan-1 -amine hydrochloride (23 mg, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 5 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 199, A-((li?,2ri)-2-phenylcyclopropyl)-3-((6-phenylpyridazin-3- yl)amino)benzamide (32 mg, 72%) as a white solid. Synthesis of Compound 200

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (100 mg, 0.34 mmol), (2-bromo-4- fluorophenyl)methanamine hydrochloride (99 mg, 0.41 mmol), and HBTU (195 mg, 0.52 mmol) were dissolved in DMF (4 mL), followed up by addition of DIPEA (0.30 mL, 1.72 mmol) and stirred for 20 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 200, /V-(2-bromo-4-fluorobenzyl)-3-((6-phenylpyridazin-3-yl)amino )benzamide (126 mg, 77%) as a white solid.

Synthesis of Compound 201

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (100 mg, 0.34 mmol), (3-bromo-4- fluorophenyl)methanamine (84 mg, 0.41 mmol), and HBTE! (195 mg, 0.52 mmol) were dissolved in DMF (4 mL), followed up by addition of DIPEA (0.18 mL, 1.03 mmol) and stirred for 20 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 201, A-(3-bromo-4-fluorobenzyl)-3 -((6-phenyl pyridazin-3-yl)amino)benzamide (145 mg,

88%) as a white solid.

Synthesis of Compound 202

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (4- (trifluoromethyl)phenyl)methanamine (34 mg, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.24 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 202, 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)-/V- (4-(trifluoromethyl)benzyl)benzamide (64 mg, 86%) as a white solid.

Synthesis of Compound 203

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (2- fluorophenyl)methanamine (0.02 mL, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.24 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 203, A-(2-fluorobenzyl)-3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)benzamide (57 mg, 86%) as a white solid.

Synthesis of Compound 204

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (2- (trifluoromethyl)phenyl)methanamine (0.03 mL, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.24 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 204, 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)-/V- (2-(trifluoromethyl)benzyl)benzamide (53 mg, 71%) as a white solid.

Synthesis of Compound 205

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (2,4,6- trifluorophenyl)methanamine (0.02 mL, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.24 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 205, 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)-/V- (2,4,6-trifluorobenzyl)benzamide (61 mg, 85%) as a white solid.

Synthesis of Compound 206

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (2,4- dimethylphenyl)methanamine (0.03 mL, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.24 mmol) and stirred for 20 hours at room temperature The reaction mixture was concentrated under reduced pressure and the residue was dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 206, /V-(2,4-dimethylbenzyl)-3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)benzamide (62 mg, 90%) as a white solid.

Synthesis of Compound 207

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (2,4- dichlorophenyl)methanamine (0.03 mL, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.24 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 207, A-(2,4-dichlorobenzyl)-3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)benzamide (57 mg, 76%) as a white solid.

Synthesis of Compound 208

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (2,4- bis(trifluoromethyl)phenyl)methanamine (47 mg, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.24 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 208, /V-(2,4-bis(trifluoromethyl)benzyl)-3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)benzamide (37 mg, 43%) as a white solid.

Synthesis of Compound 209

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (2,4,5- trifluorophenyl)methanamine (31 mg, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.24 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 209, 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)-/V- (2,4,5-trifluorobenzyl)benzamide (51 mg, 70%) as a white solid.

Synthesis of Compound 210

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), (4-fluoro-2- methylphenyl)methanamine (0.02 mL, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 5 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 210, /V-(4-fluoro-2-methylbenzyl)-3-((6-phenylpyridazin-3-yl)amin o)benzamide (31 mg, 72%) as a white solid.

Synthesis of Compound 211 3-((6-phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), (3-chloro-4- fluorophenyl)methanamine (0.02 mL, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 5 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 211, A-(3 -chi oro-4-fluorobenzyl)-3 -((6-phenyl pyridazin-3-yl)amino)benzamide (20 mg, 44%) as a white solid.

Synthesis of Compound 212

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), 2-amino-2-(4- fhiorophenyl)acetonitrile (19 mg, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 19 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 212, A-(cyano(4- fluorophenyl)methyl)-3-((6-phenylpyridazin-3-yl)amino)benzam ide (33 mg, 76%) as a white solid.

Synthesis of Compound 213

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), 2-(4- fluorophenyl)propan-2-amine (0.02 mL, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 19 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 213, /V-(2-(4-fluorophenyl)propan-2-yl)-3-((6-phenylpyridazin-3-y l)amino)benzamide (36 mg, 81%) as a white solid. Synthesis of Compound 214

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), (4-fluoro-2- methoxyphenyl)methanamine (0.02 mL, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 19 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 214, /V-(4-fluoro-2-methoxybenzyl)-3-((6-phenylpyridazin-3-yl)ami no)benzamide (18 mg, 41%) as a white solid.

Synthesis of Compound 215

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (2,4- difluorophenyl)methanamine (0.02 mL, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.24 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 215, A-(2,4-difluorobenzyl)-3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)benzamide (58 mg, 83%) as an off-white solid.

Synthesis of Compound 216

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (2- bromophenyl)methanamine (0.02 mL, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.24 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 216, /V-(2-bromobenzyl)-3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)benzamide (64 mg, 84%) as a white solid.

Synthesis of Compound 217

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (2,3,4- trifluorophenyl)methanamine (0.03 mL, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.24 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 217, 3-((5-(3-fluorophenyl)pyrimidin-2-yl)amino)-/V- (2,3,4-trifluorobenzyl)benzamide (58 mg, 81%) as a white solid.

Synthesis of Compound 218

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), 1- aminocyclopropane-l-carbonitrile hydrochloride (23 mg, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.08 mL, 0.48 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 218, A-(l-cyanocyclopropyl)-3-((5-(3- fluorophenyl)pyrimidin-2-yl)amino)benzamide (11 mg, 18%) as a white solid.

Synthesis of Compound 219

3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (50 mg, 0.16 mmol), (1- aminocyclopropyl)methanol hydrochloride (24 mg, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.08 mL, 0.48 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was dissolved in MeOH and sonicated. The slurry was filtered off and washed with MeOH to give compound 219, 3-((5-(3-fluorophenyl)pyrimidin- 2-yl)amino)-A-(l -(hydroxymethyl)cyclopropyl)benzamide (21 mg, 36%) as a white solid.

Synthesis of Compound 221

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), (lf?,2A)-2-(4- chloro-3-fluorophenyl)cyclopropan-l -amine hydrochloride (28 mg, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 22 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 221, A-((1R,2S)-2-(4-chloro-3-fluorophenyl)cyclopropyl)-3- ((6-phenylpyridazin-3-yl)amino)benzamide (47 mg, 99%) as a white solid.

Synthesis of Compound 223

3-((6-(3-Fluorophenyl)pyridazin-3-yl)amino)benzoic acid (50 mg, 0.16 mmol), 1- aminocyclopropane-l-carbonitrile hydrochloride (23 mg, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.08 mL, 0.48 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was purified by MPLC to give compound 223, A-(l- cyanocyclopropyl)-3-((6-(3-fluorophenyl)pyridazin-3-yl)amino )benzamide (19 mg, 31%) as a white solid.

Synthesis of Compound 224

3-((6-(3-Fluorophenyl)pyridazin-3-yl)amino)benzoic acid (50 mg, 0.16 mmol), (1- aminocyclopropyl)methanol hydrochloride (24 mg, 0.19 mmol), and HBTU (92 mg, 0.24 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.08 mL, 0.48 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 224, 3-((6-(3-fluorophenyl)pyridazin-3- yl )ami ho)-l-( 1 -(hydroxy ethyl )cycl opropyl )benzami de (35 mg, 58%) as a white solid.

Synthesis of Compound 225

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (50 mg, 0.17 mmol), 1- aminocyclopropane-l-carbonitrile hydrochloride (24 mg, 0.21 mmol), and HBTU (98 mg, 0.26 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.09 mL, 0.51 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was purified by MPLC to give compound 225, A-(l- cyanocyclopropyl)-3-((6-phenylpyridazin-3-yl)amino)benzamide (21 mg, 35%) as a white solid.

Synthesis of Compound 228

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (50 mg, 0.17 mmol), (1- aminocyclopropyl)methanol hydrochloride (25 mg, 0.21 mmol), and HBTU (98 mg, 0.26 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.09 mL, 0.51 mmol) and stirred for 20 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was purified by MPLC to give compound 228, A-(l- (hydroxymethyl)cyclopropyl)-3-((6-phenylpyridazin-3-yl)amino )benzamide (29 mg, 47%) as a white solid.

Synthesis of Compound 229 3-((6-(3-Fluorophenyl)pyridazin-3-yl)amino)benzoic acid (300 mg, 0.97 mmol), (4- fluoro-2-methoxyphenyl)methanamine (166 mg, 1.07 mmol), and HBTU (405 mg, 1.07 mmol) were dissolved in DCM (10 mL), followed up by addition of DIPEA (0.33 mL, 1.94 mmol) and stirred for 16 hours at room temperature. The reaction mixture was extracted by DCM and H2O. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 229, A-(4-fluoro-2-methoxybenzyl)-3-((6-(3-fluorophenyl)pyridazin -3-yl)amino)benzamide (435 mg, >100%) as a yellow solid.

Synthesis of Compound 230 3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), (3,4- difluorophenyl)methanamine (0.02 mL, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 4 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 230, /V-(3,4-difluorobenzyl)-3-((6-phenylpyridazin-3-yl)amino)ben zamide (14 mg, 31%) as a white solid.

Synthesis of Compound 231 3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), (4-fluoro-3- methylphenyl)methanamine (17 mg, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 4 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 231, /V-(4-fluoro-3-methylbenzyl)-3-((6-phenylpyridazin-3-yl)amin o)benzamide (11 mg, 25%) as a white solid.

Synthesis of Compound 232

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), (4-fluoro-3- methoxyphenyl)methanamine (0.02 mL, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 4 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 232, /V-(4-fluoro-3-methoxybenzyl)-3 -((6-phenyl pyridazin-3-yl)amino)benzamide (39 mg, 89%) as a white solid.

Synthesis of Compound 233

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), l-(4- fluorophenyl)ethan-l -amine (0.02 mL, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 4 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 233, A-(l -(4-fluorophenyl)ethyl)-3 -((6-phenyl pyridazin-3-yl)amino)benzamide (20 mg, 47%) as a white solid. Synthesis of Compound 234

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), (1R,2S)-2-(3,4- difluorophenyl)cyclopropan-l -amine hydrochloride (26 mg, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 25 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 234, /V-((1R,2S)-2-(3,4-difluorophenyl)cyclopropyl)-3-((6-phenylp yridazin-3- yl)amino)benzamide (40 mg, 87%) as a white solid.

Synthesis of Compound 235

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), (1R,2S)-2-(p- tolyl)cyclopropan-l -amine (23 mg, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 25 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 235, 3-((6-phenylpyridazin-3-yl)amino)-/V-((1R,2S)-2-(p-tolyl)cyc lopropyl)benzamide (36 mg, 84%) as a white solid.

Synthesis of Compound 236 3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (30 mg, 0.10 mmol), (1R,2S)-2-(4- methoxyphenyl)cyclopropan-l -amine (25 mg, 0.12 mmol), and HBTU (59 mg, 0.16 mmol) were dissolved in DMF (1 mL), followed up by addition of DIPEA (0.05 mL, 0.31 mmol) and stirred for 25 hours at room temperature. The reaction mixture was extracted by EA and aq. NH 4 CI. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 236, /V-((1R,2S)-2-(4-methoxyphenyl)cyclopropyl)-3-((6-phenylpyri dazin-3- yl)amino)benzamide (25 mg, 56%) as a white solid.

Synthesis of Compound 237

3-((6-Phenylpyridazin-3-yl)amino)benzoic acid (50 mg, 0.17 mmol), 1- (aminomethyl)cyclopropan-l-ol (18 mg, 0.21 mmol), and HBTU (98 mg, 0.26 mmol) were dissolved in DMF (2 mL), followed up by addition of DIPEA (0.04 mL, 0.26 mmol) and stirred for 23 hours at room temperature. The reaction mixture was concentrated under reduced pressure and the residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 237, /V-((l-hydroxycyclopropyl)methyl)-3-((6-phenylpyridazin-3- yl)amino)benzamide (15 mg, 24%) as a white solid.

3. Synthesis by Method C 3-Chloro-6-phenylpyridazine (75 mg, 0.39 mmol), 3-aminobenzamide (54 mg, 0.39 mmol), Pd 2 (dba) 3 (36 mg, 0.039 mmol), XantPhos (46 mg, 0.079 mmol), and cesium carbonate (256 mg, 0.79 mmol) were mixed in 1,4-dioxane (2 mL) and heated in a microwave reactor for 60 minutes at 110°C. The reaction mixture was concentrated and purified by MPLC to give compound 17, 3-[(6-phenylpyridazin-3-yl)amino]benzamide (10 mg, 9%) as a white solid.

4. Synthesis by Method D

Step 1: 2-Bromo-4-fluorobenzonitrile (300 mg, 1.50 mmol), 1-ethylpiperazine (0.23 mL, 1.80 mmol), cesium carbonate (977 mg, 3.00 mmol), Pd 2 (dba) 3 (137 mg, 0.15 mmol), and 2,2'-bis(diphenylphosphino)-l,l '-binaphthyl (BINAP) (93 mg, 0.15 mmol) were mixed in

Toluene (15 mL) and stirred for 21 hours at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by MPLC to give 2-(4-ethylpiperazin-l-yl)-4-fluorobenzonitrile (294 mg, 84%) as a pale-yellow solid. Step 2: 2-(4-Ethylpiperazin-l-yl)-4-fluorobenzonitrile (290 mg, 1.24 mmol) was dissolved in THF (12 mL) followed up by dropwise addition of L1AIH4 (2.0 M in THF) (1.87 mL, 3.73 mmol) at 0°C. Then the reaction mixture was stirred for 5 hours at 66°C. The reaction mixture was extracted by EA and aq. NaHCCb and concentrated to give (2-(4- ethylpiperazin-l-yl)-4-fluorophenyl)methanamine (212 mg, 72%) as a yellow liquid.

Step 3: To a solution of 3-((6-phenylpyridazin-3-yl)amino)benzoic acid (60 mg, 0.21 mmol) in chloroform (2 mL), DMF (catalytic amount), and SOCE (1.0 M in DCM) (1.03 mL, 1.03 mmol) were added and stirred for 5 hours at 60°C. The mixture was concentrated to give 3 -((6-phenyl pyridazin-3-yl)amino)benzoyl chloride (64 mg, >100%) as a yellow solid.

Step 4: To a solution of (2-(4-ethylpiperazin-l-yl)-4-fluorophenyl)methanamine (49 mg, 0.21 mmol) and pyridine (0.05 mL, 0.62 mmol) in chloroform (2 mL), 3-((6- phenylpyridazin-3-yl)amino)benzoyl chloride (64 mg, 0.21 mmol) dissolved in chloroform (2 mL) was added dropwise and stirred for 19 hours at room temperature. The reaction mixture was extracted by DCM and aq. NH 4 CI. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 220, /V-(2-(4-ethylpiperazin-l-yl)-4-fluorobenzyl)-3- ((6-phenylpyridazin-3-yl)amino)benzamide (12 mg, 11%) as a white solid.

Synthesis of Compound 222

Step 1: 2-Bromo-4-fluorobenzonitrile (300 mg, 1.50 mmol), 1-methylpiperazine (0.20 mL, 1.80 mmol), cesium carbonate (977 mg, 3.00 mmol), Pd 2 (dba) 3 (137 mg, 0.15 mmol), and BINAP (93 mg, 0.15 mmol) were mixed in Toluene (15 mL) and stirred for 17 hours at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by MPLC to give 4-fluoro-2-(4- methylpiperazin-l-yl)benzonitrile (266 mg, 81%) as a pale yellow solid.

Step 2: 4-Fluoro-2-(4-methylpiperazin-l-yl)benzonitrile (266 mg, 1.22 mmol) was dissolved in THF (12 mL) followed up by dropwise addition of L1AIH4 (2.0 M in THF) (1.82 mL, 3.64 mmol) at 0°C. Then the reaction mixture was stirred for 3 hours at 66°C. The reaction mixture was extracted by DCM and aq. NaHCCb and concentrated to give (4-fluoro-2- (4-methylpiperazin-l-yl)phenyl)methanamine (249 mg, 92%) as a brown liquid.

Step 3: To a solution of 3-((6-phenylpyridazin-3-yl)amino)benzoic acid (100 mg, 0.34 mmol) in chloroform (3 mL), DMF (catalytic amount), and SOCL (1.0 M in DCM) (1.72 mL, 1.72 mmol) were added and stirred for 4 hours at 60°C. The mixture was concentrated to give 3 -((6-phenyl pyridazin-3-yl)amino)benzoyl chloride (106 mg, >100%) as a yellow solid.

Step 4: To a solution of (4-fluoro-2-(4-methylpiperazin-l-yl)phenyl)methanamine (77 mg, 0.34 mmol) and pyridine (0.08 mL, 1.03 mmol) in chloroform (3 mL), 3-((6- phenylpyridazin-3-yl)amino)benzoyl chloride (106 mg, 0.34 mmol) dissolved in chloroform (3 mL) was added dropwise and stirred for 4 hours at room temperature. The reaction mixture was extracted by DCM and aq. NaHCCb. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 222, /V-(4-fluoro-2-(4-methylpiperazin-l-yl)benzyl)-3- ((6-phenylpyridazin-3-yl)amino)benzamide (25 mg, 15%) as a pale yellow solid.

Synthesis of Compound 226 Step 1: 3-Bromo-4-fluorobenzonitrile (300 mg, 1.50 mmol), 1-ethylpiperazine (0.23 mL, 1.80 mmol), cesium carbonate (977 mg, 3.00 mmol), Pd 2 (dba) 3 (137 mg, 0.15 mmol), and BINAP (93 mg, 0.15 mmol) were mixed in Toluene (15 mL) and stirred for 18 hours at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC to give 3-(4- ethylpiperazin-l-yl)-4-fluorobenzonitrile (278 mg, 79%) as a pale yellow solid.

Step 2: 3-(4-Ethylpiperazin-l-yl)-4-fluorobenzonitrile (277 mg, 1.19 mmol) was dissolved in THF (12 mL) followed up by dropwise addition of LiAlEL (2.0 M in THF) (1.78 mL, 3.56 mmol) at 0°C. Then the reaction mixture was stirred for 4 hours at 66°C. The reaction mixture was extracted by EA and aq. NaHCCb and concentrated to give (3-(4- ethylpiperazin-l-yl)-4-fluorophenyl)methanamine (186 mg, 66%) as a brown liquid.

Step 3: To a solution of 3-((6-phenylpyridazin-3-yl)amino)benzoic acid (100 mg, 0.34 mmol) in chloroform (3 mL), DMF (catalytic amount), and SOCL (1.0 M in DCM) (1.72 mL, 1.72 mmol) were added and stirred for 8 hours at 60°C. The mixture was concentrated to give 3 -((6-phenyl pyridazin-3-yl)amino)benzoyl chloride (106 mg, >100%) as a yellow solid.

Step 4: To a solution of (3-(4-ethylpiperazin-l-yl)-4-fluorophenyl)methanamine (81 mg, 0.34 mmol) and pyridine (0.08 mL, 1.03 mmol) in chloroform (3 mL), 3-((6- phenylpyridazin-3-yl)amino)benzoyl chloride (106 mg, 0.34 mmol) dissolved in chloroform (3 mL) was added dropwise and stirred for 17 hours at room temperature. The reaction mixture was extracted by DCM and aq. NaHCCb. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 226, /V-(3-(4-ethylpiperazin-l-yl)-4-fluorobenzyl)-3- ((6-phenylpyridazin-3-yl)amino)benzamide (4 mg, 2%) as a pale yellow solid.

Synthesis of Compound 227

Step 1: 3-Bromo-4-fluorobenzonitrile (300 mg, 1.50 mmol), 1-methylpiperazine (0.20 mL, 1.80 mmol), cesium carbonate (977 mg, 3.00 mmol), Pd 2 (dba) 3 (137 mg, 0.15 mmol), and BINAP (93 mg, 0.15 mmol) were mixed in Toluene (15 mL) and stirred for 18 hours at 110°C. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC to give 4-fluoro-3-(4- methylpiperazin-l-yl)benzonitrile (208 mg, 63%) as a pale yellow solid.

Step 2: 4-Fluoro-3-(4-methylpiperazin-l-yl)benzonitrile (207 mg, 0.94 mmol) was dissolved in THF (9 mL) followed up by dropwise addition of LiAlLL (2.0 M in THF) (1.42 mL, 2.83 mmol) at 0°C. Then the reaction mixture was stirred for 4 hours at 66°C. The reaction mixture was extracted by EA and aq. NaHCCb and concentrated to (4-fluoro-3-(4- methylpiperazin-l-yl)phenyl)methanamine (143 mg, 68%) as a brown liquid.

Step 3: To a solution of 3-((6-phenylpyridazin-3-yl)amino)benzoic acid (100 mg, 0.34 mmol) in chloroform (3 mL), DMF (catalytic amount), and SOCL (1.0 M in DCM) (1.72 mL, 1.72 mmol) were added and stirred for 8 hours at 60°C. The mixture was concentrated to give

3 -((6-phenyl pyridazin-3-yl)amino)benzoyl chloride (106 mg, >100%) as a yellow solid.

Step 4: To a solution of (4-fluoro-3-(4-methylpiperazin-l-yl)phenyl)methanamine (77 mg, 0.34 mmol) and pyridine (0.08 mL, 1.03 mmol) in chloroform (3 mL), 3-((6- phenylpyridazin-3-yl)amino)benzoyl chloride (106 mg, 0.34 mmol) dissolved in chloroform (3 mL) was added dropwise and stirred for 17 hours at room temperature. The reaction mixture was extracted by DCM and aq. NaHCCb. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by MPLC. The crude mixture was solidified by using EA and HEX to give compound 227, /V-(4-fluoro-3-(4-methylpiperazin-l-yl)benzyl)-3- ((6-phenylpyridazin-3-yl)amino)benzamide (6 mg, 3%) as a white solid.

5. Synthesis by Method E

Synthesis of Compound 180

Step 1: 3-((5-(3-Fluorophenyl)pyrimidin-2-yl)amino)benzoic acid (618 mg, 2 mmol) and cesium carbonate (1,955 mg, 6 mmol) were mixed in DMF (10 mL) followed up by addition of methyl iodide (0.274 mL, 2.2 mmol) and stirred for 5 days at room temperature.

The reaction mixture was concentrated and purified by MPLC. The crude mixture was solidified by using DCM to give methyl 3-((5-(3-fluorophenyl)pyrimidin-2- yl)(methyl)amino)benzoate (512 mg, 76%) as a beige solid.

Step 2: Methyl 3-((5-(3-fluorophenyl)pyrimidin-2-yl)(methyl)amino)benzoate (400 mg, 1.18 mmol) and LiOFFFLO (496 mg, 11.8 mmol) were mixed in H2O/THF (5/10 mL) and stirred for 8 hours at room temperature. The reaction mixture acidified by adding 1 N HC1 and the suspension was filtered. The filter cake was washed with FLO (100 mL) and dried under vacuum to give 3-((5-(3-fluorophenyl)pyrimidin-2-yl)(methyl)amino)benzoic acid (369 mg, 97%) as a white solid.

Step 3: 3-((5-(3-Fluorophenyl)pyrimidin-2-yl)(methyl)amino)benzoic acid (100 mg, 0.35 mmol), (li?,2ri)-2-phenylcyclopropan-l -amine hydrochloride (58 mg, 0.34 mmol) and HBTU (176 mg, 0.46 mmol) were dissolved in DMF (3.1 mL), followed up by addition of DIPEA (0.16 mL, 0.93 mmol) and stirred for 16 hours at room temperature. The reaction mixture was extracted by EA and brine. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was solidified by using EA and HEX to give compound 180, 3- ((5-(3-fluorophenyl)pyrimidin-2-yl)(methyl)amino)-/V-((1R,2S )-2- phenylcyclopropyl)benzamide (90 mg, 67%) as a beige solid.

Synthesis of Compound 185

Step 1: 3-Bromobenzoic acid (0.2 g, 0.995 mmol) and hexafluorophosphate azabenzotriazole tetramethyl uronium (0.57 g, 1.492 mmol) in DMF (2 mL) was added DIPEA (0.52 mL, 2.984 mmol) at room temperature. After 15 minutes of stirring, 3-flouroaniline (0.133 g, 1.193 mmol) was added and the reaction mixture was stirred for 2 hours at room temperature. The reaction mixture was extracted by EA and water. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC to give 3- bromo-A-(3-fluorophenyl)benzamide (160 mg, 55%).

Step 2: 3-Bromo-/V-(3-fluorophenyl)benzamide (0.205 g, 0.697 mmol) and ( \R,2S)-2 - phenylcyclopropan-1 -amine (0.102 g, 0.767 mmol), /-Butyl BrettPhos Pd G3 (0.032 g, 0.035 mmol), and cesium carbonate (0.68 g, 2.091 mmol) were mixed in 1,4-dioxane (2 mL) and heated in a microwave reactor for 2 hours at 130°C. The reaction mixture was poured into water, and extracted by EA. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by prep. HPLC to give compound 185, N -(3- fluorophenyl)-3-(((l A, 2,V)-2-phenyl cyclopropyl )amino)benzamide (24 mg, 10%) as a white solid. Synthesis of Compound 186 Step 1: 3-Phenylcyclobutan-l-amine HC1 salt (0.475 g, 2.586 mmol), methyl 3- bromobenzoate (0.612 g, 2.845 mmol), /-Butyl BrettPhos Pd G3 (117 mg, 0.129 mmol), and cesium carbonate (2.528 g, 7.758 mmol) were mixed in 1,4-dioxane (9.5 mL) and heated in a microwave reactor for 1 hour at 130°C. The reaction mixture was poured into water, and extracted by EA. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was purified by MPLC to give methyl 3-((3-phenylcyclobutyl)amino)benzoate (0.41 g, 56%).

Step 2: Methyl 3-((3-phenylcyclobutyl)amino)benzoate (0.40 g, 1.421 mmol) and LiOH-TBO (0.24 g, 5.7 mmol) were mixed in MeOH/ THF/H2O (1/2/1 mL) and stirred for 4 hours at room temperature. The reaction mixture acidified by adding 1 N HC1, precipitates were filtered and washed with water to give 3-((3 phenylcyclobutyl)amino)benzoic acid (0.23 g, 61%).

Step 3: 3-((3-Phenylcyclobutyl)amino)benzoic acid (0.23 g, 0.860 mmol), 3- flouroaniline (0.105 g, 0.946 mmol), and EDC-HC1 (0.660 g, 3.44 mmol) were mixed in pyridine (2.3 mL) and stirred for 1 hour at room temperature. The reaction mixture was poured into water, and extracted by EA. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by prep. HPLC to give compound 186, N-(3- fluorophenyl)-3-((3-phenylcyclobutyl)amino)benzamide (17 mg, 5%) as a white solid.

Synthesis of Compound 187

Compound 187

Step 1: 3-Phenylcyclopentan-l-one (0.41 g, 2.58 mmol), methyl 3-aminobenzoate (0.39 g, 2.58 mmol), and acetic acid (1.24 g, 20.64 mmol) were mixed in THF/MeOH (18/9 mL) and heated for 30 minutes at 70°C. The reaction mixture was cooled to room temperature and

NaBEECN (0.32 g, 5.16 mmol) was added and stirred for 2 hours at room temperature. The reaction mixture was poured into water and extracted by EA. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC to give methyl 3-((3- phenylcyclopentyl)amino)benzoate (0.2 g, 26%). Step 2: Methyl 3-((3-phenylcyclopentyl)amino)benzoate (0.1 g, 0.338 mmol) and LiOHTEO (0.056 g, 1.35 mmol) were mixed in THF/Me0H/H 2 0 (0.33/0.33/0.33 mL) and stirred for 3 hours at room temperature. The reaction mixture acidified by adding 1 N HC1, precipitates were filtered and washed with water to give 3-((3- phenylcyclopentyl)amino)benzoic acid (0.089 g, 93%).

Step 3: 3-((3-Phenylcyclopentyl)amino)benzoic acid (0.075 g, 0.266 mmol), 3- flouroaniline (0.033 g, 0.293 mmol), and 1 -ethyl-3 -(3 -dimethylaminopropyl)carbodiimide hydrochloride (EDC-HC1) (0.2 g, 1.06 mmol) were mixed in pyridine (0.5 mL) and stirred for 1 hour at room temperature. The reaction mixture was poured into water and extracted by EA. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by prep. HPLC to give compound 187, /V-(3-fluorophenyl)-3-((3- phenylcyclopentyl)amino)benzamide (17 mg, 17%) as a white solid.

Synthesis of Compound 188

Step 1: 4-Phenylcyclohexan-l-one (0.60 g, 3.44 mmol), methyl 3-aminobenzoate (0.52 g, 3.44 mmol), and acetic acid (1.65 g, 27.55 mmol) were mixed in THF/MeOH (12/6 mL) and heated for 30 minutes at 70°C. The reaction mixture was cooled to room temperature and NaBEECN (0.43 g, 6.89 mmol) was added and stirred for 1 hour at room temperature. The reaction mixture was poured into water, and extracted by EA. The organic layer was dried over anhydrous Na 2 SC> 4 and concentrated. The residue was purified by MPLC to give methyl 3-((4- phenylcyclohexyl)amino)benzoate (0.44 g, 41%).

Step 2: Methyl 3-((4-phenylcyclohexyl)amino)benzoate (0.44 g, 1.42 mmol) and LiOH-H 2 0 (0.26 g, 5.68 mmol) were mixed in MeOH/H 2 0 (2.5/2.5 mL) and stirred for 4 hours at room temperature. The reaction mixture acidified by adding 1 N HC1, precipitates were filtered and washed with water to give 3-((4-phenylcyclohexyl)amino)benzoic acid (0.16 g, 38%). Step 3: 3-((4-Phenylcyclohexyl)amino)benzoic acid (0.26 g, 0.880 mmol), 3- flouroaniline (0.108 g, 0.968 mmol), and EDC-HC1 (0.675 g, 3.521 mmol) were mixed in pyridine (5.2 mL) and stirred for 1.5 hours at room temperature. The reaction mixture was poured into water and extracted by EA. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by prep. HPLC to give compound 188, N-(3- fluorophenyl)-3-((4-phenylcyclohexyl)amino)benzamide (Diastereomer A: 18 mg, 5% as a yellow solid / Diastereomer B: 12 mg, 3.5% as a brown solid)

Synthesis of Compound 189

Step 1: l-Phenylpiperidin-4-one (0.60 g, 3.42 mmol), methyl 3-aminobenzoate (0.41 g, 2.74 mmol), and acetic acid (1.64 g, 27.39 mmol) were mixed in THF/MeOH (20/10 mL) and heated for 30 minutes at 50°C. The reaction mixture was cooled to room temperature and

NaBEECN (0.43 g, 6.85 mmol) was added and stirred for 2 hours at room temperature. The reaction mixture was poured into water, and extracted by EA. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC to give methyl methyl 3-((l-phenylpiperidin-4-yl)amino)benzoate (0.54 g, 50%).

Step 2: Methyl 3-((l-phenylpiperidin-4-yl)amino)benzoate (0.53 g, 1.72 mmol) and LiOEhELO (0.29 g, 6.88 mmol) were mixed in TElF/MeOEl/EEO (2.5/1.25/1.25 mL) and stirred for 4 hours at room temperature. The reaction mixture acidified by adding 1 N HC1, precipitates were filtered and washed with water to give 3-((l-phenylpiperidin-4- yl)amino)benzoic acid (0.27 g, 53%).

Step 3: 3-((l-Phenylpiperidin-4-yl)amino)benzoic acid (0.36 g, 1.215 mmol), 3- flouroaniline (0.13 g, 1.215 mmol), and EDC-HC1 (0.93 g, 4.859 mmol) were mixed in pyridine (3.6 mL) and stirred for 1 hour at room temperature. The reaction mixture was poured into water and extracted by EA. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by prep. HPLC to give compound 189, N-( 3- fluorophenyl)-3-((l-phenylpiperidin-4-yl)amino)benzamide (0.016 g, 3%) as a white solid.

Synthesis of Compound 190

Step 1: 3,6-Dichloropyridazine (0.550 g, 3.692 mmol) and methyl 3- aminocyclopentane-l-carboxylate-HCl (0.730 g, 4.061 mmol) was dissolved in /V-Methyl-2- Pyrrolidone (NMP) (5.5 mL), followed up by addition of DIPEA (3.2 mL, 18.460 mmol) and heated in a microwave reactor for 2 hours at 130°C. The reaction mixture was poured into water, and extracted by EA. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC to give 3-((6-chloropyridazin-3- yl)amino)cyclopentane-l-carboxylate (0.37 g, 39%).

Step 2: Methyl 3-((6-chloropyridazin-3-yl)amino)cyclopentane-l-carboxylate (0.590 g, 2.31 mmol), phenyl boronic acid (0.338 g, 2.77 mmol), cesium carbonate (1.88 g, 5.77 mmol) and l,l'-bis(diphenylphosphino)fenOcene]dichloropalladium(II)-DC M complex (0.188 g,

0.231 mmol) were mixed in l,4-dioxane/H 2 0 (5.9 mL/1.2 mL) and heated in a microwave reactor for 2 hours at 100°C. The reaction mixture was poured into water, and extracted by EA. The organic layer was dried over anhydrous NaiSCL and concentrated. The residue was purified by MPLC to give methyl 3-((6-phenylpyridazin-3-yl)amino)cyclopentane-l- carboxylate (0.15 g, 22%).

Step 3: Methyl 3-((6-phenylpyridazin-3-yl)amino)cyclopentane-l-carboxylate (0.15 g , 0.504 mmol) and LiOH-EEO (0.086 g, 2.01 mmol) were mixed in THF/MeOH/ELO (0.75/0.375/0.375 mL) and stirred for 4 hours at room temperature. The reaction mixture acidified by adding 1 N HC1, precipitates were filtered and washed with water to give 3-((6- phenylpyridazin-3-yl)amino)cyclopentane-l -carboxylic acid (0.08 g, 56 %).

Step 4: 3 -((6-Phenylpyridazin-3-yl)amino)cyclopentane-l -carboxylic acid (0.08 g, 0.282 mmol), 3-flouroaniline (0.034 g, 0.311 mmol), and EDC-HC1 (0.217 g, 1.129 mmol) were mixed in pyridine (0.8 mL) and stirred for 1 hour at room temperature. The reaction mixture was poured into water, and extracted by EA. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The residue was purified by prep. HPLC to give compound 190, /V-(3-fluorophenyl)-3-((6-phenylpyridazin-3-yl)amino)cyclope ntane-l- carboxamide (13 mg, 12%) as a white solid.

Synthesis of Compound 191

Step 1: 3,6-Dichloropyridazine (0.550 g, 3.692 mmol) and methyl 3- aminocyclohexane-l-carboxylate-HCl (0.786 g, 4.061 mmol) was dissolved inNMP (5.5 mL), followed up by addition of DIPEA (3.2 mL, 18.460 mmol) and heated in a microwave reactor for 3 hours at 160°C. The reaction mixture was poured into water, and extracted by EA. The organic layer was dried over anhydrous NaiSCE and concentrated. The residue was purified by MPLC to give methyl 3-((6-chloropyridazin-3-yl)amino)cyclohexane-l-carboxylate. (0.32 g, 32%).

Step 2: Methyl 3-((6-chloropyridazin-3-yl)amino)cyclohexane-l-carboxylate (0.50 g , 1.854 mmol), phenyl boronic acid (0.271 g, 2.224 mmol), cesium carbonate (1.51 g, 4.634 mmol) and l,l'-bis(diphenylphosphino)ferrocene]dichloropalladium(II)-D CM complex (0.151 mg, 0.185 mmol) were mixed in l,4-dioxane/H 2 0 (5.4 mL/0.3 mL) and heated in a microwave reactor for 2 hours at 100°C. The reaction mixture was poured into water, and extracted by EA. The organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The residue was purified by MPLC to give methyl 3-((6-phenylpyridazin-3-yl)amino)cyclohexane-l- carboxylate (0.12 g, 21%).

Step 3: Methyl 3-((6-phenylpyridazin-3-yl)amino)cyclohexane-l-carboxylate (0.12 g , 0.385 mmol) and LiOH-EEO (0.065 g, 1.54 mmol) were mixed in THF/MeOH/LLO (0.5/0.25/0.25 mL) and stirred for 3 hours at room temperature. The reaction mixture acidified by adding 1 N HC1, precipitates were filtered and washed with water to give 3-((6- phenylpyridazin-3-yl)amino)cyclohexane-l -carboxylic acid (0.06 g, 52%).

Step 4: 3 -((6-Phenylpyridazin-3-yl)amino)cyclohexane-l -carboxylic acid (0.225 g, 0.757 mmol), 3-flouroaniline (0.084 g, 0.757 mmol), and EDC-HC1 (0.580 g, 3.027 mmol) were mixed in pyridine (4.5 mL) and stirred for 1 hour at room temperature. The reaction mixture was poured into water, and extracted by EA. The organic layer was dried over anhydrous NaiSCri and concentrated. The residue was purified by prep. HPLC to give compound 191, /V-(3-fluorophenyl)-3-((6-phenylpyridazin-3-yl)amino)cyclohe xane-l- carboxamide (11 mg, 4%) as a white solid. Table 1 lists the chemical structures, characterization data, and preparation methods of the above-described compounds.

Table 1: Compound Structure, Characterization Data, and Preparation Method

Table 2 lists the chemical structures, catalog numbers, and purchase source of the compounds that were used for the assay described in the following examples. Table 2: Compound Structure, Catalog Number, and Purchase Source

EXAMPLE 3

YFP QUENCHING ASSAY

1. Materials and Instruments Ionomycin (Alomonelab cat. #1-700), FLUOstar Omega microplate reader (BMG Labtech, Ortenberg, Germany), and MARS Data Analysis Software (BMG Labtech)

2. Cell Culture

Fisher rat thyroid (FRT) cells stably expressing human AN06 (GenBank accession no. NP_001191732.1, provided by J.H. Nam, Dongguk University College of Medicine, Korea) and halide sensor mutant YFP-H148Q/I152L/F46L) were constructed and grown in Dulbecco's modified Eagle's medium Nutrient Mixture F-12 (DMEM/F-12) supplemented 10% FBS, lOOunits/mL penicillin, 500 pg/mL hygromycin B and 100 pg/mL neomycin.

3. Assay Procedure

Fisher rat thyroid (FRT) cells stably expressing human AN06 and halide sensor mutant YFP (H148Q/I152L/F46L) were seeded in black walled 96 well plates and incubated in a 37°C, 5% C02 incubator to reach about 100% cell confluency. Then, each well of the 96 well plates were washed for several times with phosphate buffered saline (PBS), and 50 pL of PBS was added to each well. Test compounds (100X in DMSO) were added to each well to be 1% v/v DMSO. After incubation for 10 minutes in 40°C, the 96 well plates were transferred to a plate reader, and YFP fluorescence changed by SCN- introduced into cells through activated AN06 were measured by the following steps.

(1) YFP fluorescence signals were recorded in every 0.4 seconds.

(2) Basal YFP fluorescence signals were recorded for 1 second.

(3) 140 mM SCN (50 pL) containing 10 pM ionomycin was injected to each well, and YFP fluorescence signals were recorded.

The inhibitory activity (%) of each of the test compounds was obtained by the following steps.

(1) Background signals were subtracted from recording values, and the resulting values were converted to relative percentages. The values at 0 second were set to be 100%.

(2) Differences between the values at 3.6 seconds and those of at 7.6 seconds were calculated.

(3) In each row of 96 well plate, the inhibitory activity of each of the test compounds was calculated as percentages. The inhibitory activity of a negative control group to which neither compounds nor ionomycin were treated was set to be 100%, and the inhibitory activity of a positive control group to which ionomycin was treated and compounds were not treated was set to be 0%.

(4) The assay was performed in duplicate or triplicate, and the results were averaged.

The assay result is shown in Table 3, where ‘A’ means that the compound showed 60% or more inhibitory activities at each concentration (A > 60%), ‘B’ means that the compound showed inhibitory activities of 30% or more to less than 60% (60% > B > 30%) at each concentration, and ‘C’ means that the compound shows less than 30% inhibitory activities (30% > C) at each concentration.

Table 3: YFP Quenching Assay Result

EXAMPLE 4

LACT C2 (PHOSPHATIDYLSERINE SCRAMBLASE FUNCTION) ASSAY

1. Materials and Instruments

Ionomycin (Alomonelab, cat. # 1-700), DAPI (Sigma- Aldrich, cat. # D8417), paraformaldehyde (Biosesang, cat. # P2031), Lionheart FX Automated Microscope (BioTek, Winooski), Python3 (Python Software Foundation), and OpenCV (Open Source Computer Vision Library).

2. Cell Culture

Fisher rat thyroid (FRT) cells stably expressing human AN06 (GenBank accession no. NP_001191732.1, provided by J.H. Nam, Dongguk University College of Medicine, Korea) were grown in DMEM/Ham's F-12 (1 : 1) medium with 10% FBS, 2 mM L-549 glutamine, 100 units/mL penicillin, and 100 pg/mL streptomycin at 37°C and 5% CO2.

3. Assay Procedure

FRT cells stably expressing human AN06 were plated in 96 well black-walled microplates at a density of 2 c 10 4 cells/well. After 24 hours incubation, cells were treated with the test compound (dissolved in DMSO) were treated to each well to become 1% v/v DMSO for 10 minutes, then 10 pM of ionomycin was applied. Finally each well was washed with 200 pL PBS after 10 minutes. After washout, the phosphatidylserine and nuclei were stained with PBS containing 500 nM Lactadherin-C2 (Lact-C2)-GFP, then cells were washed with 200 pL PBS. Cells were fixed with 4% paraformaldehyde for 5 minutes at room temperature. For morphological analysis, some cells were stained with fluorescently labelled DAPI for 15 minutes at room temperature. Quantitative analysis of the fluorescence intensity of Lact-C2- GFP was performed with Python3 and OpenCV. OpenCV was used to remove background and noise pixels, and the sum of all remaining pixel values was used to evaluate the fluorescence intensity of Lact-C2-GFP.

The inhibitory activity (%) of each of the test compounds was obtained by the following steps. In each row of 96 well plate, the inhibitory activity of each of the test compounds was calculated as percentages. The inhibitory activity of a negative control group to which neither compounds nor ionomycin were treated was set to be 100%, and the inhibitory activity of a positive control group to which ionomycin was treated and compounds were not treated was set to be 0%. The assay was performed in triplicate, and the results were averaged.

The assay result is shown in Table 4, where ‘A’ means that the compound showed 60% or more inhibitory activities at each concentration (A > 60%), ‘B’ means that the compound showed inhibitory activities of 30% or more to less than 60% (60% > B > 30%) at each concentration, and ‘C’ means that the compound showed less than 30% inhibitory activities (30% > C) at each concentration.

Table 4: LACT C2 Assay Result

EXAMPLE 5

CYTOTOXICITY ASSAY

1. Materials and Instruments Vero cell lines (Ministry of Food and Drug Safety, Republic of Korea, batch #

VEROOlWCB-1201), CellTiter 96® AQueous One Solution Cell Proliferation Assay kit (Promega, cat. # G3582), Infinite M200 microplate reader (Tecan).

2. Cell Culture

Vero cells were grown in DMEM supplemented with 10% FBS, 100 units/mL penicillin and 100 pg/mL streptomycin in an 5% CO2 incubator at 37°C.

3. Assay Procedure

Vero cells were seeded at -20% confluency in 96-well plates. After a 24 h incubation, cells were treated with different concentrations (1, 3, and 10 mM) of compounds. After incubation for 48 h, 100 pL of culture medium was added to each well before treatment with 20 pL of 3- (4,5-dimethythizol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sul fophenyl)-2H-tetrazolium salt (MTS) assay solution per well (CellTiter 96® AQueous One Solution Cell Proliferation Assay kit). Cells were incubated for 1 h at 37 °C. The soluble formazan produced by the cellular reduction of MTS was quantified by measuring the absorbance at 490 nm or 690 nm with a microplate reader (Infinite M200).

The viability (%) of each of the test compounds was obtained by the following steps. Background signals (at 690nm) were subtracted from recording values at 490 nm, and the resulting values were converted to relative percentages. The values treated with DMSO were set to be 100%. The viability of each test compound was calculated as percentages. The assay was performed in replicated (n=2~6) and the results were averaged.

The assay result is shown in Table 5, where ‘A’ is marked with compounds having viabilities that are equal to or higher than 80% (A > 80%) at each concentration, ‘B’ is marked with compounds having viabilities that are equal to or higher than 50% and are lower than 80% (50% < B < 80%) at each concentration, and ‘C’ is marked with compounds having viabilities that are lower than 50% at each concentration (C < 50%).

Table 5: Cytotoxicity Assay Result

EXAMPLE 6

VIRAL REPLICATION

1. Materials and Instruments SARS-CoV-2 KUMC-2 strain (Korea University, Republic of Korea), NCCP-43344 strain (National Culture Collection for Pathogens, Republic of Korea), Vero cell lines (Ministry of Food and Drug Safety, Republic of Korea, batch # VEROOlWCB-1201), Calu-3 (Korean Cell Line Bank, KCLB #30055), Minimum Essential Medium (MEM, HyClone #SH30024.01, Logan, UT), Fetal Bovine Serum (FBS, HyClone #SH30084.03), penicillin- streptomycin (HyClone #SV300010), Phosphate Buffered Saline (PBS, HyClone, Logan, UT), QIAamp Viral RNA Mini kit (QIAGEN #52906, Valencia, CA), Luna® Universal One-Step RT-qPCR Kit (NEW ENGLAND BioLabs® Inc, Ipswich, MA, USA), SARS-CoV-2 nsp targeting forward primer, 5’- TGG GGY TTT ACR GGT AAC CT-3’; SARS-CoV-2 nsp targeting reverse primer, 5’- AAC RCG CTT AAC AAA GCA CTC -3’; probe, 5’-56-FAM- TAG TTG TGA /ZEN/ TGC WAT CAT GAC TAG-3 IABkFQ-3 ’ (Chu, D.K. W. et al, 2020),

Applied Biosystem Quantstudio 3 detection system (Applied Biosystems, Waltham, MA,

USA)

2. Cell Culture

Calu-3 and Vero cells were maintained in MEM with Earle’s Balanced Salts containing 2.2 g/L sodium bicarbonate supplemented with 10% FBS and 1% penicillin-streptomycin.

Cells were grown at 37°C in a 5% C02 incubator. 3. Assay Procedure

A clinically isolated SARS-CoV-2 KUMC-2 and NCCP-43344 strains were used. To evaluate compound efficacy against virus infection, Vero cell was pretreated with the indicated concentrations of compounds for 2 h, prior to infection with KUMC-2 (0.001 MOI) for 1 h. After washing with PBS, cells were replenished with culture media supplemented with 2%

FBS together with 5 test compounds (1, 3 mM) and then incubated for 48 h. Calu-3 cell was pretreated with the indicated concentrations of compounds for 2 h, prior to infection with NCCP-43344 (0.01 or 0.05 MOI) for 1 h. After washing with PBS, cells were replenished with culture media supplemented with 2% FBS together with 6 test compounds (1, 10 mM) and then incubated for 48 h. All experiments with infectious virus were approved by the Institutional Biosafety Committee of the Yonsei University Health System (IBC 2020-003) and conducted in the Biosafety Level 3 facility at Yonsei University College of Medicine. For the quantification of the SARS-CoV-2 virus secretion, viral RNAs in cell culture supernatant were extracted using a QIAamp Viral RNA Mini kit, as per the manufacturer’s instructions. Quantitative PCR was performed with a Luna® Universal One- Step RT-qPCR Kit using the Applied Biosystem Quantstudio 3 detection system. For amplification, 2 pL of extracted RNA was added to the mixture of 10 pL of 2X Luna Universal One-Step Reaction Mix, 0.4 pL of 10 pM primers, 0.2 pL of 10 pM probe and 1 pL of 20X Luna WarmStart ® RT Enzyme Mix adjusted to a total reaction volume of 20 pL with RNase-free water. Amplification was carried out using the optimal thermocycling conditions described in the manufacturer’s protocol for 40 cycles. The standard curve was generated by conducting qRT-PCR using serially diluted virus stock.

The inhibitory activity (%) of each of the test compounds was obtained by the following steps.

(1) Test compound signals were subtracted from negative control (DMSO) values, and the resulting values were converted to relative percentages. The value of negative control was set to be 0 %. Inhibitory activity was then calculated by the following formula:

Inhibitory activity (%) = [negative control - test compounds] ÷ [negative control] x 100

(2) The assay was performed in duplicate and the results were averaged.

The assay result is shown in Table 6, where ‘A’ is marked with compounds having inhibitory activities that are equal to or higher than 60% (A > 60%) at each concentration, ‘B’ is marked with compounds having inhibitory activities that are equal to or higher than 40% and are lower than 60% (40% < B < 60%) at each concentration, ‘C’ is marked with compounds having inhibitory activities that are equal to or higher than 20% and are lower than 40% (20%

< C < 40%) at each concentration and ‘D’ is marked with compounds having inhibitory activities that are lower than 20% (D < 20%) at each concentration.

Table 6: Viral replication results

* Drug treatment with a concentration of 5 mM

EXAMPLE 7

MECHANISM OF ANTI-VIRUS ACTIVITY 1. Materials and Methods

Cell culture, plasmids, cloning, siRNA, and transfection FRT cells were stably transfected with both a yellow fluorescent protein (YFP) variant (H148Q/I152L/F46L) and human ANOl (abc isoform), mouse AN02 or human AN06 (variant 5), and the cells were cultured in DMEM/Ham's F-12 (1 : 1) medium with 10% FBS, 2 mM L-glutamine, 100 units/ml penicillin, and 100 pg/ml streptomycin at 37°C and 5% CO2. HEK293T cells and HeLa cells were obtained from the American Type Culture Collection, Manassas, VA, USA. For the generation of stable ACE2 overexpressing HeLa and HEK293T cells, hACE2 and P2A-BSD encoding sequences were amplified from pcDNA3.1-hACE2 (Addgene plasmid #145033) and lentiCas9-Blast (Addgene plasmid #52962), respectively. The amplicons were cloned into the lentiviral pLV-mCherry vector using a NEBuilder HiFi DNA Assembly Kit (NEB Biolabs). HeLa cells were transduced with lentiviral particles encoding hACE- P2A-BSD in the presence of 8 pg /ml of polybrene (Merck-Millipore, Burlington, MA, USA) for one day. Cells were maintained at a low density in the presence of 10 pg/ml of blasticidin for the selection. CHO cells, HEK293T cells, HeLa cells, and their derivatives, were cultured in Dulbecco’s modified Eagle’s medium-high glucose (Gibco #11995-065, Carlsbad, CA) and supplemented with 10% heat-inactivated fetal bovine serum (Gibco #26140-079), and 100 U/ml penicillin and 100 pg/ml streptomycin at 37°C in a 5 % CO2- humidified incubator. The HEK293T-ACE2-TMPRSS2 cells bearing mCherry fluorescence were commercially purchased from Genecopoeia, Rockville, MD, USA, and maintained in culture media in the presence of 1 pg/mL of puromycin and 100 pg/mL of Hygromycin B (USA #10687010; Invitrogen, Carlsbad, CA).

Calu-3 and Vero cells were maintained in Minimum Essential Medium (MEM) with Earle's Balanced Salts containing 2.2 g/L sodium bicarbonate (HyClone #SH30024.01, Logan, UT) supplemented with 10% FBS (HyClone #SH30084.03) and 1% penicillin-streptomycin (HyClone #SV300010). Cells were grown at 37°C in a 5% CO2 incubator. BHK-21/WI-2 (Kerafast, Boston, MA) cells were maintained in Dulbecco’s Modified Eagle Medium (DMEM) (Gibco) supplemented with 10% FBS (Gibco), Penicillin (100 units/ml)(Gibco), and Streptomycin (100 ug/ml) (Gibco) in an 8% C02 incubator at 37°C and passaged every 2-3 days.

The mammalian expressible pcDNA3.1 -Spike plasmid was generated by inserting a stop codon before the C9 tag of a pcDNA3.1-SARS2-Spike plasmid (Addgene #145032). To generate the plasmids encoding the S1/S2 cleavage mutants of the Spike protein, the cleavage site was abolished by amino acid substitution at R682S and R685G using an NEBuilder HiFi DNA Assembly Kit. The lactadherin-C2 (Lact-C2-eGFP) encoding plasmid (Addgene plasmid # 22852) was subcloned into a pET28c bacterial expression vector for His 6 -fusion protein purification. For the generation of mcherry-LactC2, fragments of mCherry encoding sequences (Addgene# 36084) and Lact-C2 encoding sequences were PCR amplified and cloned into the pET28c plasmid using an NEBuilder HiFi DNA Assembly Kit. AccuTarget™ pre-designed human 7MEM76F-specific (gene ID: 196527) or control scrambled siRNAs were commercially purchased from Bioneer (Daejeon, Korea). Transfection of siRNA into HEK293T or HeLa cells was performed using the TransIT-X2 Dynamic Delivery System (#MIR6006, Mirus Bio LLC, Madison, WI, US) according to the manufacturer’s protocol.

Chemical reagents, lentiviral particles and antibodies

Compound A24 (ChemDiv; Cat# F575-0482, 3-{[l-benzyl-2-(thiophen-2-yl)-lH-indol- 3-yl]methyl}-l-[(oxolan-2-yl)methyl]urea), Compound A24 (ChemDiv; Cat# F474-2423, ethyl 8-methyl-4-((4-(methylthio)benzyl)amino)-2-oxo-l,2-dihydroqu inoline-3-carboxylate), abamectin (Cayman Chemical, Ann Arbor, MI, USA), ivermectin (Sigma-Aldrich, St. Louis, MO, USA), camostat mesylate (Sigma-Aldrich, #SML0057), BAPTA-AM (Sigma-Aldrich, #A1076), amphotericin B solution (Sigma-Aldrich, #A2942), and trypsin (Sigma-Aldrich, #T6567) were purchased commercially. LentifectTM SARS-CoV-2 Spike-pseudotyped Lentivirus used in the single-round infection (GFP reporter) was commercially acquired from Genecopoeia (cat# SP001). 7-nitro-2-l,3-benzoxadiazol-4-yl (NBD)-PS (16:0-06:0) was purchased from Avanti Polar Lipids (# 810192). The following antibodies were acquired commercially: anti-SARS-CoV-2 S2 (GeneTex #GTX632604), anti-aldolase A (Santa Cruz Biotechnology #SC-390733), and anti-HIV-p24 (GeneTex #GTX64128).

Generation of pseudotyped SARS-CoV-2 S virus particles

For generation of lentivirus-based SARS2-PsV used in the PS extemalization and [Ca 2+ ]i measurements, HEK 293T cells were plated in a 6-well plate and transfected the next day when they were approximately 80% confluent with a combination of the following plasmids: 1 pg of pEGIP (Addgene #26777), 0.75 pg of psPAX2, and 2.25 pg of pcDNA3.1- SARS2-Spike (Addgene #145032). The TransIT-X2 Dynamic Delivery System (#MIR6006; Mirus Bio LLC, Madison, WI, US) was used for transfection, following the manufacturer’s protocol. The next day, the transfection medium was replaced with fresh culture medium, and cells were cultured for 48 h. The pseudotyped virus-containing culture medium was collected 48 h post transfection, centrifuged at 500 xg for 5 min, and stored at -80°C.

Western blot analysis For immunoblotting of AN06, FRT, FRT-AN06, and Calu-3, cells were lysed in lysis buffer (50 mM Tris-HCl (pH 7.4), 1 mM EDTA, 1 mM NasVCH, 150 mM NaCl, 1% Nonidet P-40, 0.25% sodium deoxycholate, protease inhibitor mixture). The cell lysates were centrifuged at 13,000 rpm for 20 min at 4°C and protein concentrations were measured with a Bradford assay kit (Sigma) in accordance with the manufacturer’s instructions. The supernatant proteins of FRT (40 pg), FRT-AN06 (40 pg), Calu-3 (120 pg) were separated using 4-12% Tris-glycine precast gel (KOMA BIOTECH) and transferred to a PVDF membrane and then blocked with 5% non-fat skim milk in Tris-buffered saline containing 0.1% Tween 20 (TBST) for 1 h. Membranes were incubated in a solution containing primary antibodies against AN06 or b-actin. Antibodies against mAN06 were generated using the same epitope (67-81 a.a, DFRTPEFEEFNGKPD-C) appearing in a previous study (Yang et ak, 2012), which retains a high homology among human, rat, and mouse AN06s. Membranes were then incubated with HRP-conjugated anti-secondary IgG antibody (Enzo Life Sciences) and visualized using the ECL Plus Western Blotting detection system (Pierce).

For immunoblotting of the SARS-CoV-2 Spike glycoprotein, the cell lysates of wild- type and R682S/R685G SARS-PsV infected HEK 293T cells were homogenized with sonication for 20 s (1-second pulses) followed by centrifugation at 16,000 x g for 20 min at 4°C. The samples were separated by a 4-12% SDS-PAGE gel electrophoresis and transferred to nitrocellulose filter membranes. After being blocked by 5% skim milk, the membranes were blotted with primary antibodies against the S2 domain of the Spike protein (GeneTex #GTX632604) and aldolase A (Santa Cruz #SC-390733), then incubated with horseradish peroxidase (HRP) conjugated secondary antibodies (1:2000) and detected with Chemiluminescent Reagent (Amersham #RPN2105).

YFP quenching assay

FRT cells expressing a YFP variant (H148Q/I152L/F46L) with ANOl, AN02 or AN06 were plated in 96-well black- walled microplates (Corning Inc., Corning, NY, USA) at a density of 2 x 10 4 cells/well. After a 48-h incubation, each well of 96-well plates was washed twice in phosphate-buffered solution (PBS, 200 mΐ/wash) and test compounds containing 50 pi PBS were added to each well. For the high-throughput screening of a library of 54,000 drug like small-molecule compounds (Chemdiv, San Diego, CA, USA) and a library of 1,700 approved drugs (TargetMol, Boston, MA, USA), FRT-AN06 cells were treated with 25 mM compounds. After a 10-min incubation at 37°C, the 96-well plates were transferred to a FLUOstar Omega microplate reader (BMG Labtech, Ortenberg, Germany) for fluorescence assay. Each well was assayed individually for AN06-mediated thiocyanate (SCN) influx by monitoring YFP fluorescence continuously (0.4 s/point) for 1 s (baseline), then 140 mM SCN solution containing 10 mM ionomycin was added at 1 s before YFP fluorescence was recorded for 7 s. The initial rate of SCN influx was computed from fluorescence data by nonlinear regression.

Cytoplasmic calcium measurements

FRT cells were plated in 96-well black- walled microplates at a density of 2 x 10 4 cells/well. After 48 h incubation, the cells were loaded with Fluo4 NW (Invitrogen) as per the manufacturer’s protocol. Cells were incubated with 100 pi assay buffer containing Fluo-4 NW. After a 1-h incubation, cells were treated with test compounds for 10 min and the 96-well plates were transferred to a FLUOstar Omega microplate reader (BMG Labtech) equipped with syringe pumps and custom Fluo-4 excitation/emission filters (485/538 nm). Intracellular calcium signaling was induced via application of 10 mM ionomycin.

The [Ca 2+ ]i imaging in HEK 293T-ACE2-TMPRSS2 cells was performed with a ratiometric Ca 2+ probe, Fura-2 (Fura-2 AM; Molecular Probes, Eugene, Oregon, USA). The ACE2 and TMRPSS2 stable cells grown on glass cover slips were loaded with cell permeable Fura-2 AM (5 mM, 30 min, 37°C) in HEPES-buffered physiological salt solution (PSS) containing (mM) 145 NaCl, 5 KC1, 10 HEPES, 5 D-Glucose, 1 MgC12 and 1 CaC12 (adjusted to pH 7.4 NaOH). Some cells were additionally treated with BAPTA-AM (3 mM, 20 min) for the chelation of intracellular Ca 2+ . Glass coverslips were placed into an experimental chamber (Live Cell Instrument, Seoul, Korea) with a perfusion system at 37°C and then the cells were transferred to the perfusion chamber. Ca 2+ measurements were performed using a Zeiss Axio microscope attached to a charge coupled device camera in a temperature-controlled chamber set to 37°C. For the illumination of Fura-2, an ultra-fast switching monochromator (Polychrome V, Till Photonics) was used. Fura-2 was excited alternatively at 340 nm and 380 nm and the emission light was simultaneously collected at 510 nm. Images were acquired at 2 frames/s using MetaFlour (Molecular devices). In each recording, three cells showing the maximum cytosolic Ca 2+ response were chosen for the analysis of [Ca 2+ ]i dynamics. A region of interest (ROI) of [Ca 2+ ]i increasing microdomain in each cell was monitored to determine the cytoplasmic Fura-2 fluorescence changes during SARS2-PsV or ATP application. The [Ca 2+ ]i values were estimated from the Fura-2 fluorescence ratiometric values using the following equation (Grynkiewicz et ah, 1985). where Kdis the indicator’s dissociation constant for Ca 2+ and R is the intensity ratio for fluorescence at the two chosen wavelengths (340, 380 nm). The Rmax and Rminare ratios at zero and saturating [Ca 2+ ]i, respectively, and Sf2/Sb2is the ratio of excitation efficiencies for free and bound Fura-2. For calibration, 20 mM EGTA was added to obtain the minimum fluorescence ratio and 5 mM ionomycin with 10 mM CaCk was added to obtain the maximum fluorescence ratio. In each recording, three cells showing the maximum Ca 2+ response were chosen for statistical analyses.

Electrophysiology

The Ca 2+ -activated CT channel activities were measured in HEK 293T cells using the whole-cell clamp techniques. Cells were transferred into a bath mounted on a stage with an inverted microscope (Ti-2, Nikon). The whole-cell clamp was achieved by rupturing the patch membrane after forming a giga-seal. The bath solution was perfused at 5 mL/min. The voltage and current recordings were performed at room temperature (22-25°C). Patch pipettes with a free-tip resistance of approximately 2-5 MW were connected to the head stage of a patch- clamp amplifier (Axopatch-200B, Axon Instruments). pCLAMP software v.10.7 and Digidata- 1440A (Axon Instruments) were used to acquire data and apply command pulses. Voltage and current traces were stored and analyzed using Clampfit v. 10.7 and Origin v. 8.0 (Origin Lab Inc.). To record AN06 (TMEM16F) currents, voltage ramps spanning a range of -100 to +100 mV were delivered from a holding potential of -60 mV every 20 s. Currents were sampled at 5 kHz. All data were low-pass filtered at 1 kHz. The standard pipette solution for whole-cell clamp contained (in mM) 141.8 NMDG-C1, 5 HEPES, 0.5 MgCk, 10 HEDTA, and 1 Mg-ATP. An appropriate amount of CaCk was added to the pipette solution to obtain a pipette Ca 2+ concentration of 10 mM (adjusted to pH 7.2 with NMDG-OH). The bath solution contained (in mM) 146 NMDG-Cl, 1 CaCk, 1 MgCk, 10 HEPES, and 5 Glucose (adjusted to pH 7.2 with NMDG-OH).

For the CFTR short-circuit current measurements, Snapwell cell culture inserts containing FRT cells expressing human WT-CFTR were mounted in Ussing chambers (Physiological Instruments, San Diego, CA). The apical hemichamber was filled with buffer solution containing (in mM): 60 NaCl, 60 Na-gluconate, 5 KC1, 1 MgCk, 1 CaCk, 10 D- glucose, 2.5 HEPES, and 25 NaHC03 (pH 7.4). The basolateral hemichamber was filled with buffer solution containing (in mM): 120 NaCl, 5 KC1, 1 MgCk, 1 CaCk, 10 D-glucose, 2.5 HEPES, and 25 NaHC03 (pH 7.4). The basolateral membrane was permeabilized using 250 pg/mL amphotericin B, and then the cells were incubated for 20 min and aerated with 95% 02/5% CO2 at 37°C. The apical membrane current was recorded using an EVC4000 Multi- Channel V/I Clamp (World Precision Instruments, Sarasota, FL) and PowerLab 4/35 (AD Instruments, Castle Hill, Australia).

PS externalization assay and confocal microscopy

The LactC2-GFP and LactC2-mCherry fusion proteins were produced in the BL-21 (DE3) Escherichia coli strain. Cells were grown in an LB medium at 30°C in 120 pg/ml kanamycin until the culture reached A600 = 0.6. After the addition of 0.5 mM IPTG, the culture was incubated for 6 h at 30°C. Cells were lysed in lysis buffer containing 50 mM sodium phosphate (pH 7.4), 200 mM NaCl, 0.5% NP-40, 10 mM imidazole and protease inhibitor cocktail (Roche Applied Science, Mannheim, Germany). The cell lysates were homogenized with sonication for 3 min (1-s pulse) followed by centrifugation at 12,000 x g for 20 min at 4°C. After centrifugation, His6-mcherry-LactC2 was purified with a nickel- nitrilotriacetic acid (Ni-NTA) protein purification system (QIAGEN) by following the manufacturer’s instructions. Stock solutions of 1-3 mg/ml were stored in the dialysis buffer (100 mM KC1, 1 M HEPES-KOH pH 8.0, 0.2 M EDTA, 10% glycerol, 1 M DTT, 200 mM PMSF) and were used at 1-3 pg/ml.

FRT cells stably expressing AN06 (variant 5) were plated in 96-well black-walled microplates at a density of 2 x 10^ cells/well. After a 48-h incubation, cells were treated with the test compound for 10 min, then 10 pM of ionomycin was applied and finally each well was washed with 200 pi PBS after 10 min. Cells were fixed with 4% paraformaldehyde (100 pl/well) for 10 min at room temperature. After washout, the phosphatidyl serine and nuclei were stained with PBS containing 500 nM Lact-C2-GFP, then cells were washed with 200 pi PBS and microscopic images were acquired with a Lionheart FX Automated Microscope (BioTek, Winooski, VT, USA). For morphological analysis, some cells were stained with fluorescently labelled phalloidin and DAPI (Sigma-Aldrich) for 15 min at room temperature. Quantitative analysis of the fluorescence intensity of Lact-C2-GFP was performed with Python3 (Python Software Foundation, Wilmington, DE, USA) and OpenCV (Open Source Computer Vision Library). OpenCV was used to remove background and noise pixels, and the sum of all remaining pixel values was used to evaluate the fluorescence intensity of Lact-C2- GFP. The original microscopic images contained approximately 130 to 160 cells per image, and these images were used for the calculation of fluorescence intensity. FIG. 2D shows magnified images cropped to show the cells more clearly, and these images contain approximately 50 cells per image.

For SARS2-PsV-induced PS externalization assay, HeLa-ACE2 cells were plated on 12-mm round coverslips and incubated with the lentivirus-based SARS2-PsV (p24 100 ng/mL) for 15 min. For authentic SARS-CoV-2 virus-induced PS externalization, the cells were infected with SARS-CoV-2 with an MOI of 10 for 15 min. After infection, the cells were fixed with 2% formaldehyde for 5 min at room temperature, and then incubated with solutions containing LactC2-mCherry for 45 min at 37°C. For nucleus staining, cells were also stained with DAPI. Fluorescent images were obtained with a Zeiss LSM confocal microscope (LSM 780; Carl Zeiss, Berlin, Germany) with a 20x or 63 x (1.4 numerical aperture) objective lens. For image quantification using the open-source software ImageJ Fiji, 24-bit confocal images including red, green, and blue components were converted into three 8-bit mono-channel images. To quantify PS externalization, pixels above a threshold level of 50 were defined as externalized PS. The percentage of Lact-C2 labeled cells in DAPI-stained cells (100 to 200 cells) was calculated. To minimize variations between experimental replicates, the recorded fluorescence values were normalized to a maximum value in each set of experiments. For example, in Fig 3B, the maximum Lact-C2-mCherry fluorescence value was observed in cells treated with SARS2-PsV alone (lane 2). Then, the fluorescence values of Control (lane 1) and SARS2-PsV + BAPTA-AM (lane 3) were normalized to the value of SARS2-PsV alone (lane 2) in each experimental replication set.

Membrane fusion assay

For analysis of SARS2-PsV-induced cell fusion, HeLa-ACE2 cells were plated on 12- mm round coverslips and incubated with SARS2-PsV (p24 100 ng/mL) for 30 min. To quantify the number of fusion events, LactC2-positive multinucleated cells containing more than two nuclei were considered to be membrane-fused multinucleated cells. We analyzed micrograph images of 3 to 5 fields (-150 cells per field) in each experimental replication set and counted the number of LactC2-positive multinucleated cells per field. The percentage of multinucleated cells/total cells (D API-positive) was calculated.

For analysis of Spike-induced membrane fusion mechanisms, time-lapse imaging of membrane fusion events occurring between the Spike-expressing CHO cells and ACE2- expressing HEK 293T-ACE2-TMPRSS2 cells was performed. CHO cells were transfected with plasmids expressing Spike envelope proteins (2 pg/2 ml, pcDNA3.1-SARS2-Spike). Two days after transfection, Spike-expressing CHO cells at a density of 2 x 10 5 cells/well were labeled with the fluorescent lipophilic tracer Vybrant DiO (2 mM) in serum-free medium for 20 min at 37°C, washed, and added to the cytosolic mCherry-expressing HEK 293T-ACE2-

TMPRSS2 cells (2 x 10^ cells/well) labeled with Hoechst 33342 (1 pg/mL). For the examination of the multistep processes of Spike-mediated cell-cell fusion, live imaging was performed using Thunder Imager Live cell fluorescence microscopy (Leica microsystems, Wetzlar, Germany). An 8-h time-lapse image acquired at 20 frame/min was analyzed using the Leica Application Suite X software. The cell-cell adhesion was quantified by counting the number of GFP-positive cells in contact with mCherry-positive cells during the initial 1 h co incubation period, and the cell-cell fusion was quantified by counting the membrane fusion events between the GFP-positive and mCherry-positive cells during the overall 8-h coincubation period.

Single-round infection

The cytosolic mCherry-expressing HEK293T-ACE2-TMPRSS2 cells were seeded to

96-well plates at a density of 5 x 10^ cells/well. After a 24-h incubation, the cells were pretreated with different concentrations of inhibitors for 1 h. Subsequently, 5 pi of Lentifect

SARS-CoV-2 Spike-pseudotyped lentivirus (1.88 x 10^ IFU/mL) was added to the cells and incubated first at 4°C for 2 h and subsequently at 37° C for 4 h. After the 6-h incubation with pseudotyped virus, cells were fed with fresh culture medium and further incubated for 42 h.

We quantified the amount of infection by counting the number of GFP-positive cells (threshold level of 32 in 8-bit mono channel images) using fluorescence microcopy with a software package (NIS-elements AR; Nikon, Tokyo, Japan); and the percentage of GFP-positive cells/total cells (mCherry-positive) was calculated.

Reverse-transcription PCR (RT-PCR) and quantitative PCR (qPCR) analysis

Twenty-four hours after transfection with siRNAs, the total cellular RNA was extracted using Tri-RNA reagent (#FATRR 001; Favorgen Biotech Corp., Taiwan) according to the manufacturer’s protocol. Complementary DNA (cDNA) was synthesized using an RNA to cDNA EcoDry premix (#639549; Takara Bio Inc., Shiga, Japan). Mixtures were incubated at 42°C for 1 h, followed by 70°C for 10 min. qPCR was performed with an Applied Biosystem StepOne System (Applied Biosystems, Forster City, CA, USA). Cell viability assay

FRT, HEK 293 T, Calu-3 and Vero cells were seeded at -20% confluency in 96-well plates. After a 24-h incubation, cells were treated with different concentrations (0.3, 1, 3, 10,

30 and 100 mM) of compounds. After incubation for 48 h, the effects of the compounds on cell viability were evaluated via 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)2-(4- sulfophenyl)-2H-tetrazolium (MTS) assay using a CellTiter 96® AQueous One Solution Cell Proliferation Assay kit (Promega) following the manufacturer’s instructions. The soluble formazan produced by the cellular reduction of MTS was quantified by measuring the absorbance at 490 nm with a microplate reader (Infinite M200; Tecan, Grodig, Austria).

Virus propagation, quantification, and infection of SARS-CoV-2

A clinically isolated SARS-CoV-2 KUMC-2 strain (GISAID accession#:

EPI ISL 413018) was used. For viral propagation, Vero cells were infected with SARS-CoV- 2 (0.01 MOI), then culture supernatant was harvested at 48 h post-infection. The harvested supernatant was filtered and stocked at -80°C. For plaque formation assay, one of the stock vials was serially diluted and used to infect a Vero cell for 1 h, which was then incubated with culture media containing 1 % low melting temperature agarose (#50101; Lonza, Rockland,

ME) for 48 h. Viral plaques were visualized with Neutral Red Solution (Sigma #2889) for an additional 24 h and were counted to determine the plaque-forming unit of the viral stock. To evaluate drug efficacy against virus infection, Calu-3 and Vero cells were pretreated with the indicated concentrations of drugs for 2 h, prior to infection with the virus (0.01 or 0.001 MOI) for 1 h. After washing with PBS, cells were replenished with culture media supplemented with 2% FBS together with the indicated concentrations of drugs and then incubated for 48 h. All experiments with infectious virus were approved by the Institutional Biosafety Committee of the Yonsei University Health System (IBC 2020-003) and conducted in the Biosafety Level 3 facility at Yonsei University College of Medicine.

For the quantification of the SARS-CoV-2 virus secretion, viral RNAs in cell culture supernatant were extracted using a QIAamp Viral RNA Mini kit (QIAGEN #52906, Valencia, CA), as per the manufacturer's instructions. Quantitative PCR was performed with a Luna ® Universal One-Step RT-qPCR Kit (NEW ENGLAND BioLabs" Inc, Ipswich, MA, USA) using the Applied Biosystem Quantstudio 3 detection system (Applied Biosystems, Waltham, MA, USA). For amplification, 2 pL of extracted RNA was added to the mixture of 10 pL of 2X Luna Universal One-Step Reaction Mix, 0.4 pi of 10 pM primers, 0.2 pi of 10 pM probe and 1 μL of 20X Luna WarmStart ® RT Enzyme Mix adjusted to a total reaction volume of 20 pL with RNase-free water. Amplification was carried out using the optimal thermocycling conditions described in the manufacturer’s protocol for 40 cycles. The standard curve was generated by conducting qRT-PCR using serially diluted virus stock.

Authentic virus infection of primary human nasal epithelial (HNE) cells

All experiments using HNE cells were approved by the Institutional Review Board of Yonsei University College of Medicine (4-2016-0902). The cells were isolated from nasal polyps obtained from patients with chronic rhinosinusitis and who had no clinical history of asthma, aspirin sensitivity, or cystic fibrosis; and cultured under air-liquid interface (ALI) culture conditions as previously described (Yoon et al., 1999). The passage #2 HNE cells were seeded at a density of 1 c 10 5 cells/well on a 12-mm, 0.45-pm pore transwell-clear culture insert (3460; Coming, Kennebunk, ME) and cultured in a 1:1 mixture of bronchial epithelial cell growth media (BEGM) and DMEM, supplemented with growth factors according to the manufacturer’s instructions (Lonza, Walkersville, MD).

The cells were cultured using the media in both the apical and basolateral compartments until the cells became confluent (approximately 3 days). Once confluent, the cells were placed under ALI culture conditions; the apical compartments were exposed to air and only the basolateral compartments were supported by the media. The media were changed on alternate days for 21 days. AN06 inhibiting drugs were administered to the basolateral compartments of the fully differentiated HNE cells 2 h prior to infection with the vims. SARS- CoV-2 vims (1.0 MOI) was added to the apical compartments and removed after incubation for 3 h. At 3 days after infection, 150 pL of lx PBS was added to the apical compartments, incubated for 10 min at 37°C, and collected to recover progeny viruses. The plaque assay for viral quantification was performed using Vero cell culture at 85-95% confluence in 12-well plates.

Statistical analysis

The results of multiple experiments are presented as the mean ± standard error of the mean (SEM). For biochemical and in vitro assays, statistical analysis was performed using a two-tailed Student’s t-test or one-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison test, as appropriate, using GraphPad Prism8 (GraphPad Software, Inc., La Jolla, CA, USA). A P value < 0.05 was considered statistically significant.

2. Inhibitory Effect on AN06 Ion Channel Activity As shown in FIG. 1, the YFP fluorescence quenching -based anion channel assay with ionomycin-induced Ca 2+ activation in FRT cells expressing AN06 showed that Compound A24 has an inhibitory effect on AN06. The IC 50 value of Compound A24 was identified as 91nM.

The effect (FIGS. 8A and 8B) of Compound A24 on ANOl and AN02 CACCs and the effect (FIGS. 9A-9C) of Compound A24 on cytosolic Ca 2+ signals and CFTR, and a major Cl channel in the respiratory epithelial cells were investigated. Compound A24 was identified to have a significantly high AN06 selectivity. The IC 50 values of Compound A24 on ANOl, AN02, and ANO 6 were identified as about 41.9mM, 8.20mM, and 0.091mM, respectively, which means that the selectivity of Compound A24 against AN06 was identified to be about 460-fold and about 90-fold selectivity against ANOl and AN02, respectively. Compound A24 did not affect ionomycin-induced cytosolic Ca 2+ signals up to 30mM. Compound A24 up to a 30 mM concentration did not affect CFTR currents.

The inhibitory effect of Compound A24 was further verified by whole-cell current measurements, showing that it potently inhibits AN06 CACC with an IC 50 of 73 nM in HEK 293T-AN06 cells (FIGS. 2A-2C), which is comparable to that of YFP-based assay in FRT- AN06 cells.

3. Inhibitory Effect on ANO6 Phospholipid Scramblase Activity

Using the fluorescent protein-labelled C2 domain of lactadherin (Lact-C2), a sensitive probe for phosphatidylserine (PS)(Yeung et al., 2008), whether Compound A24 also inhibits AN06 phospholipid scramblase activity was investigated. At 10 mM, Compound A24 significantly reduced the ionomycin-induced PS extemalization in FRT-AN06 cells. The IC 50 of Compound A24 for AN06 scramblase was 0.91 mM (FIGS. 2D and 2E).

4. Inhibitory Effect on PS Extemalization

Using the SARS-CoV-2 S pseudotyped lentivirus (PsV), whether SARS-CoV-2 induces PS extemalization was investigated. In the ACE2 expressing HeLa (HeLa-ACE2) cells, application of a lentivirus-based SARS2-PsV evoked PS scrambling with the PS extemalization being nullified by the intracellular Ca 2+ chelator BAPTA-AM (FIGS. 3 A and B). The same SARS2-PsV did not evoke PS scrambling in ACE2 -negative cells (FIGS. 3C and 3D), indicating that Spike-ACE2 interaction is required for this to occur. Notably, the SARS2-PsV-induced PS scrambling was significantly reduced by AN06 silencing (FIGS. 3E and 3F). siRNA-mediated AN06 knockdown was verified by qPCR analysis (FIGS. 10A and 10B). This SARS2-PsV-induced, Ca 2+ - and AN06- dependent PS externalization was strongly inhibited by Compound A24 (10 M, FIGS. 3G and 3H). Furthermore, treatment with the authentic SARS-CoV-2 virus again evoked PS scrambling, which was in turn suppressed by B APT A- AM and Compound A24 (FIGS. 31 and 3J).

A serine protease inhibitor, camostat, prevents activation of the SARS-CoV-2 S fusion peptide (FP) by inhibiting the proteolytic cleavage of the S protein, which is mediated by TMPRSS2 and other trypsin-like enzymes (Hoffmann et ah, 2021; Hoffmann et al., 2020; Lee et al., 1996). Treatment with camostat (100 mM) reduced the SARS2-PsV-induced PS externalization; yet this was reversed by the application of trypsin-pretreated SARS2-PsV (FIGS. 4 A and 4B), suggesting that interaction between FP with the host cell membrane contributes to the AN06-mediated PS externalization. Camostat did not affect the PS scrambling induced by ionomycin (FIGS. IOC and 10D), implying that camostat reduces SARS2-PsV-induced PS scrambling by inhibiting FP-mediated Ca 2+ signaling. Also of note, Compound A24 inhibited the PS externalization evoked by the trypsin-pretreated SARS2-PsV (FIGS. 4A and 4B). In addition, the R682S/R685G SARS2-PsV mutant bearing a defective protease cleavage site (S1/S2 site, FIG. 10E) significantly lost its PS scrambling activity (FIGS. 4C and 4D), further indicating that AN06-mediated PS scrambling is located downstream of the FP activation.

5. Inhibitory Effect on Membrane Fusion

Incubation with SARS2-PsV for 30 min resulted in the appearance of multinucleated cells, a sign of membrane fusion. Treatment with Compound A24 significantly reduced the multinucleated cell formation (FIGS. 4E and 4F), implying that AN06 inhibition downregulates the SARS2-PsV-induced membrane fusion events. The membrane fusion process comprises a series of events involving virus-host adhesion, hemifusion, and fusion pore formation. In order to more precisely analyze the Spike-induced membrane fusion mechanisms, an 8-h time-lapse imaging of the membrane fusion events occurring between the Spike-expressing cells labelled with the membrane lipid probe Vybrant-DiO (CHO-Spike, green) and the cytosolic mCherry-labelled ACE2-expressing cells (HEK 293T-ACE2- TMPRSS2, red) was performed. As shown in FIGS. 4G and 4H, expression of Spike increased cell adhesion and membrane fusion between these two types of cells. Notably, Compound A24 significantly reduced the occurrence of membrane fusion events, despite not affecting cell-cell adhesion. In time-lapse imaging of HEK 293 T-ACE2-TMPRSS2/CHO- Spike co-cultures, all cells showing initial lipid-mixing, a sign of membrane hemifusion, proceeded to develop into fully fused cells. Treatment with Compound A24 eliminated the initial lipid-mixing event as well as the cell-cell fusion. Collectively, these results imply that the PS-dependent stage takes place after Spike-ACE2 binding (virus-host adhesion) but before the membrane hemifusion process.

6. Ca 2+ - and ANOό-Dependency of Viral Entry

Using the ratiometric Ca 2+ probe Fura-2, whether SARS-CoV-2 induces cytosolic Ca 2+ signals was investigated. In the ACE2- and TMPRSS2 expressing HEK 293T (HEK 293T- ACE2-TMPRSS2) cells, application of SARS2-PsV evoked a sustained elevation of cytosolic Ca 2+ levels (FIGS. 5C, 5D, and 5K), which was nullified by pretreatment with the intracellular Ca 2+ chelator BAPTA-AM (FIGS. 5E, 5F, and 5K). Camostat significantly reduced the intensity SARS2-PsV-induced Ca 2+ signals (FIGS. 5G, 5H, and 5K), implying that viral FP activation is responsible for cytosolic Ca 2+ signaling in the host cells. Notably, Compound A24 did not affect SARS2-PsV-induced cytosolic Ca 2+ signaling in regards to A[Ca 2+ ]i change, lag time (time to peak), or [Ca 2+ ]i increase slope (A[Ca 2+ ]i/At) (FIGS. 5I-5M). Furthermore, Compound A24 did not alter the purinergic agonist (ATP)-induced Ca 2+ response in HEK 293T-ACE2-TMPRSS2 and Calu-3 cells (FIGS. 11 A-l IF). These results indicate that alterations in cytosolic Ca 2+ signals are not responsible for the inhibitory effects of Compound A24 on SARS2-PsV-induced PS scrambling.

7. Anti-Viral Activity

Next, the importance of PS externalization for viral infectivity was investigated via analysis of single-round SARS2-PsV infections. Transduction with a commercially available SARS2-PsV for 48 h in HEK 293 T - ACE2-TMPRS S2 cells produced a stable level of expression of the viral reporter protein GFP, which was significantly reduced by AN06 silencing (FIGS. 6A, 6B, and 10B) and pretreatment with BAPTA-AM (FIGS. 6C and 6D), indicating that AN06 and cytosolic Ca 2+ signals are involved in the viral infection. Furthermore, treatment with Compound A24 dose dependently inhibited the single-round infection with an IC50 of 0.42 mM (FIGS. 6E and 6F).

The anti-viral effects of Compound A24 was further investigated via viral replication assays using authentic SARS-CoV-2 virus. The anti-viral effects in Calu-3 human airway epithelial cells that are TMPRSS2-positive were analyzed. The qPCR quantification of virion mRNAs revealed that treatment with Compound A24 (10 mM, 48 h) induced a 1/10,000 reduction in viral titers (AC t = -13.5, FIG. 7A). The IC50 value of Compound A24 in Calu-3 cells was estimated as 0.97 mM (FIG. 7B).

Compound A24 exhibited anti-viral activity against SARS-CoV-2 in the TMPRSS2- negative Vero cells, suggesting that AN06 inhibition is also effective in FP activation by other enzymes. In the light microscopic examinations, treatment with Compound A24 for 48 h dose- dependently reduced the virus-induced cytolysis in Vero cells (FIGS. 7C and 12A).

Compound A24 (10 mM) induced 1/100 - 1/3,500 reductions in viral titers (AC t = -6.4 ~ -11.8), with the magnitude of the reduction depending on the drug treatment time (24-48 h) and viral infection dose (0.001 and 0.01 multiplicity of infection, MOI) (FIGS. 7D, 7E, andl2A-12C). The IC50 values of Compound A24 under these conditions were 0.62-0.93 mM. The half- maximal cytotoxic concentration (CC50) of Compound A24 was calculated as >100 mM in Calu-3 and Vero cells as well as in FRT cells, indicating that the safety margin of Compound A24 is >100, as shown in FIGS.13A-13D.

Lastly, the antiviral effects of Compound A24 on SARS-CoV-2 were investigated in primary cultures of human nasal epithelial (HNE) cells (Yoon et al., 1999). The passage #2 HNE cells were cultured under air-liquid interface conditions to retain airway epithelial properties including endogenous expressions of ANOl and AN06 (FIG. 10A). Notably, treatments with Compound A24 (10 mM) to the basolateral compartment of HNE cells for 72 h, in which the apical side was infected with authentic SARS-CoV-2 virus, induced an average reduction of 94% in viral replications when determined by the plaque assay (FIGS. 7F and 7G).