COMPOUNDS AND THEIR USES AS GPR183 INHIBITORS

Abstract
The disclosure relates to the compounds as GPR183 inhibitors, the methods for preparing these compounds, and the compositions and their uses as treatment or prevention of cancers, autoimmune diseases, pain, and osteoporosis using GPR 183 inhibitors targeted immune cells.
Description
FILED OF THE INVENTION

The present invention relates to the compounds as GPR183 inhibitors, the methods for preparing these compounds, and the compositions and their uses as treatment or prevention of cancers. autoimmune diseases, pain, and osteoporosis using GPR183 inhibitors targeted immune cells.


BACKGROUND OF THE INVENTION

The seven-transmembrane G protein-coupled receptor EBV-induced gene 2 (EBI2), also known as GPR183, was identified, when its expression was found to be highly up-regulated in B cells upon infection with EBV. After the initial identification of EBI2 expression in B cells, it became clear that EBI2 is expressed in several cells of the hematopoietic lineage, including (in addition to B cells) T cells, natural killer cells, monocytes, macrophages, dendritic cells (DCs), neutrophils, cosinophils. platelets, and osteoclasts. Moreover, EBI2 expression has been characterized in astrocytes and in carly developmental stages of immune cells, including hematopoictic stem and progenitor cells and thymocytes.


GPR183 ligands were identified as oxysterols which arise from oxidation of cholesterol. 7α,25-dihydroxycholesterol (7α,25-diHC) has the strongest affinity to GPR183, 7α,27-diHC shows second highest affinity whereas other oxysterols (monohydroxylated oxysterols 25-HC and 7α-HC) show substantially lower activity. Synthesis of the GPR183 ligand 7α,25-diHC requires two hydroxylation steps at position 25 by the enzyme cholesterol 25-hydroxylase (CH25H) and at position 7α, by cytochrome P450 family 7 subfamily member B1 (CYP7B1). Degradation of 7α,25-diHC is catalyzed by the enzyme hydroxy-δ-5-steroid dehydrogenase. 3 β- and steroid δ isomerase.


Binding of oxysterols to GPR183 leads to the release of intracellular calcium, suppression of CAMP and internalization of GPR183. The most important consequence of GPR183 activation is migration of GPR 183- expressing cells towards higher 7α,25-diHC concentrations. GPR183 contributes at various levels to the coordination of cell-cell encounters by modulating the migration and positioning of DCs, T cells and B cells. Besides supporting the migration of lymphocytes and DCs, GPR183 coordinates the migration of innate lymphoid cells (ILCs). GPR183 also contributes to the organization of intestinal lymphoid tissue.


Genome wide association studies (GWAS) have linked the GPR183-oxysterol system to inflammatory bowel diseases (IBD). Experimental observations indicate an important role of GPR183 in inflammation and pathogenesis of colitis such as anti-CD40 murine colitis model. Similarly, GPR183 confers pro-inflammatory effects in the IL-10 murine model of chronic colitis. There is strong and consistent evidence for increased expression of the enzymes synthesizing the GPR183 ligand 7α,25-diHC in human samples and different mouse models of colitis and enzyme levels correlate with severity of inflammation. A few studies so far have tested directly that EBI2 plays a role in the pathogenesis of autoimmune disease. One of those studies indicates a role for EBI2 in the recruitment of pathogenic T cells into the CNS in the EAE model, suggesting EBI2 as an important regulator of autoimmune disease


GPR183-oxysterol also promotes osteoclast precursor migration to bone surfaces and regulates bone mass homeostasis. The study shows that GPR183 enhances the development of large osteoclasts by promoting osteoclast precursor motility, facilitating cell-cell interactions and fusion in vitro and in vivo. GPR183 is also necessary and sufficient for guiding osteoclast precursors (OCPs) toward bone surfaces. Defective GPR183 signaling led to increased bone mass in male mice and protected female mice from age-and estrogen deficiency-induced osteoporosis.


GPR183-oxysterol axis in the spinal cord also contributes to neuropathic pain. In an in-silicon modeling, a library of 5 million compounds was screened to identify several novel small-molecule antagonists of GPR183 with nanomolar potency. These compounds were able to antagonize 7α,25-diHC-induced calcium mobilization in vitro with IC50 values below 50 nM. In vivo intrathecal injections of these antagonists during peak pain after chronic constriction injury (CCI) surgery reversed allodynia in male and female mice. Acute intrathecal injection of the GPR183 ligand, 7α,25-diHC in naïve mice induced dose dependent allodynia. Importantly, this effect was blocked using GPR183 antagonists, suggesting spinal GPR183 activation as pro-nociceptive. The study reveals a role for GPR183 in neuropathic pain and identifies GPR183 as a potential target for therapeutic intervention.


GPR183-oxysterol axis is also implicated in playing a role in Non-alcoholic fatty liver disease. GPR183 is expressed in human hepatoma cell lines and its expression is induced in vivo in mouse livers after high-fat diet feeding. Activation of GPR183 inhibits fat accumulation in primary mouse hepatocytes and HepG2 cells through Gi/o proteins, p38 MAPKs, PI3K, and AMPK.


Francois Gessier et. al. isolated a compound 4m, i.e., (E)-3-(4-Bromophenyl)-1-(4-(4-methoxybenzoyl)piperazin-1-yl)-prop-2-en-1-one, which was suggested to play a functional role of the oxysterol/EBI2 pathway in these immune cells (J. Med. Chem. 2014, 57, 3358-3368). However, Compound 4m in the Francois Gessier et. al. reference was found to have a very poor solubility and poor pharmacokinetics (e.g., poor liver microsome stability, poor hepatocyte stability, poor human plasma stability, poor clearance, shorter half-life, lower Vss and lower AUC exposure), higher CYP inhibition and higher hERG channel inhibition.


In light of the role that GPR183 plays in the pathogenesis of various diseases, it is desirable to prepare compounds that inhibit GPR183 activity so as to be used in the treatment of diseases mediated by GPR183, such as cancer, autoimmune diseases, liver diseases, osteoporosis and neuropathic pain; and have better solubility, good permeability better pharmacokinetics (e.g., liver microsome stability, hepatocyte stability, human plasma stability, better clearance, longer half-life, higher Vss and higher AUC exposure), lower or no CYP inhibition and lower or no hERG channel inhibition so as to have good druggability such as low hepatotoxicity, more tolerance, more safety, and more efficacy.


SUMMARY OF THE INVENTION

Provided are a series of novel compounds as GPR183 inhibitors. The inventors of the instant invention have found that structural modifications to the prior art compounds, especially the change of ring B in formula (I) of the instant invention resulted in a series of new compounds which show better solubility, better pharmacokinetics (e.g., liver microsome stability, hepatocyte stability, human plasma stability, better clearance, longer half-life, higher Vss and higher AUC exposure), lower or no CYP inhibition and lower or no hERG channel inhibition, low hepatotoxicity, more tolerance, more safety, and more efficacy and comparable chemotaxis and Ca+ mobilization potency.


Provided is a compound of formula (I) or (II)




embedded image


or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof,


wherein

    • ring A is phenyl, monocyclic 5- to 9-membered heteroaryl, bicyclic 7- to 12-membered heteroaryl, monocyclic 5- to 9-membered heterocyclyl, bicyclic 7- to 12-membered heterocyclyl, monocyclic 3- to 8-membered cycloalkyl, or bicyclic 7- to 12-membered cycloalkyl;
    • p is 0, 1, or 2;
    • R1 is halogen, cyano, C1-6alkyl, or haloC1-6alkyl;
    • L1 is a direct bond, C1-6alkylene, C2-6alkenylene, C2-6alkynylene or C3-6cycloalkylene;
    • X is CRa or N, and Y is CRb or N, wherein Ra and Rb are each independently hydrogen, halogen, hydroxy, or C1-6alkyl;
    • m and n are each independently 0, 1, or 2;
    • t is 0, 1, or 2;
    • R3 is halogen, C1-6alkyl, or haloC1-6alkyl;
    • L2 is a direct bond, —C(O)—, *1—NRc—C(O)—*2, *1—C(O)—NRc—*2, *1—CRdRe—NRc—C(O)—*2, *1—NRc—C(O)—CRdRe—*2, or —NRc—, wherein Rc, Rd and Re are each independently hydrogen or C1-6alkyl, and wherein the symbol *2 refers to the position attached to ring B and the symbol *1 refers to the position opposite to ring B in formula (I);
    • ring B is phenyl, monocyclic 5- to 9-membered heteroaryl, bicyclic 7- to 12-membered heteroaryl, monocyclic 5- to 9-membered heterocyclyl, bicyclic 7- to 12-membered heterocyclyl, monocyclic 3- to 8-membered cycloalkyl, or bicyclic 7- to 12-membered cycloalkyl;
    • q is 0, 1 or 2;
    • R2 is halogen, oxo, C1-6alkyl, haloC1-6alkyl, or —ORf, wherein Rf is hydrogen, C1-6alkyl, haloC1-6alkyl, C1-6alkoxy-C1-6alkyl-, or monocyclic 5- to 9-membered heterocyclyl,


      Provided that said compound is not any of the following compounds:


      (2E)-3-(4-bromophenyl)-1-{4-[(4-methoxyphenyl)carbonyl]piperazinyl}prop-2-en-1-one;


      3-(4-bromophenyl)-1-{4-[(4-methoxyphenyl)carbonyl]piperazinyl}prop-2-en-1-one;


      3-(3,4-dichlorophenyl)-1-{4-[(4-methoxyphenyl)carbonyl]piperazinyl}prop-2-en-1-one;


      6-chloroindol-2-yl 4-[(4-methoxyphenyl)carbonyl]piperazinyl ketone;


      3-(2H-benzo[d][1,3]dioxolen-5-yl)-1-{4-[(4-methoxyphenyl)carbonyl]piperazinyl}prop-2-en-1-one;


      (E)-1-(4-(1,4-oxazepane-4-carbonyl)piperidin-1-yl)-3-(4-chlorophenyl)prop-2-en-1-one;


      (E)-3-(4-chlorophenyl)-1-(4-morpholinoazepan-1-yl)prop-2-en-1-one;


      (E)-3-(4-bromophenyl)-1-(4-cyclobutylpiperazin-1-yl) prop-2-en-1-one;


      (E)-3-(4-chlorophenyl)-1-(4-cyclobutylpiperazin-1-yl)prop-2-en-1-one;


      (E)-3-(4-fluorophenyl)-2-methyl-1-(4-(tetrahydro-2H-pyran-4-yl)-1,4-diazepan-1-yl) prop-2-en-1-one;


      (E)-1-(4-(azetidin-3-yl)piperazin-1-yl)-3-(4-chlorophenyl)prop-2-en-1-one;


      (E)-1-(4-(azetidin-3-yl)piperazin-1-yl)-3-(4-fluorophenyl)prop-2-en-1-one;


      (E)-3-(2-bromophenyl)-1-(4-cyclopentylpiperazin-1-yl)prop-2-en-1-one;


      (E)-3-(4-bromophenyl)-1-(4-(2,3-dihydro benzofuran-5-carbonyl)piperazin-1-yl)prop-2-en-1-one;


      (4-(1H-indole-2-carbonyl)piperazin-1-yl)(4-methoxyphenyl)methanone; or


      (5-chloro-1H-indol-2-yl)(4-(4-methoxy benzoyl)piperazin-1-yl)methanone.


The Definition of Ring A

In some embodiments, ring A is phenyl, monocyclic 5- to 9-membered heteroaryl, bicyclic 7- to 12-membered heteroaryl, monocyclic 5- to 9-membered heterocyclyl, bicyclic 7- to 12-membered heterocyclyl, monocyclic 3- to 8-membered cycloalkyl, or bicyclic 7- to 12-membered cycloalkyl, each of which is unsubstituted or substituted with one or two substituents selected from halogen, cyano, C1-6alkyl, or haloC1-6alkyl.


In some further embodiments, ring A is phenyl which is unsubstituted or substituted with one or two halogens. In some even further embodiments, ring A is phenyl which is substituted with one halogen at position 4, preferably ring A is 4-bromophenyl.


In some further embodiments, ring A is monocyclic 5- to 9-membered heteroaryl selected from pyridinyl, pyridinyl, or pyrimidinyl, preferably pyridin-3-yl, pyridin-4-yl, or pyrimidin-5-yl, each of which is unsubstituted or substituted with one or two substituents selected from halogen, cyano or C1-6alkyl.


In some further embodiments, ring A is bicyclic 7- to 12-membered heteroaryl selected from indolyl, pyrrolopyridinyl, or benzoimidazolyl, preferably 1H-indol-2-yl, 1H-indol-3-yl, 1H-pyrrolo[2,3-b]pyridine-2-yl, 1H-pyrrolo[3,2-b]pyridine-2-yl, 1H-pyrrolo[3,2-c]pyridine-2-yl, pyrazolo[1,5-a]pyridine-2-yl, or 1H-benzo[d]imidazol-2-yl, each of which is unsubstituted or substituted with one or two halogens.


In some embodiments, ring A is bicyclic 7- to 12-membered heterocyclyl which is benzo fused heterocyclyl. In some further embodiments, the benzo fused heterocyclyl is indolinyl, isoindolinyl, benzopyranyl, dihydrothiazolopyrimidinyl, tetrahydroquinolyl, tetrahydroisoquinolyl, dihydrobenzofuranyl, dihydrobenzoxazinyl, dihydrobenzoimidazolyl, tetrahydrobenzothienyl, tetrahydrobenzofuranyl, benzodioxolyl, benzodioxonyl, chromanyl, chromenyl, octahydrochromenyl, dihydrobenzodioxynyl, dihydrobenzoxezinyl, dihydrobenzodioxepinyl, dihydrothienodioxynyl, dihydrobenzooxazepinyl, tetrahydrobenzooxazepinyl, dihydrobenzoazepinyl, tetrahydrobenzoazepinyl, isochromanyl, or chromanyl.


In some further embodiments, ring A is bicyclic 7- to 12-membered heterocyclyl selected from benzodioxolyl or dihydrobenzofuranyl, preferably benzo[d][1,3]dioxol-5-yl or 2,3-dihydrobenzofuran-6-yl, each of which is unsubstituted or substituted with one or two halogens.


In some further embodiments, ring A is bicyclic 7- to 12-membered cycloalkyl selected from dihydro-1H-inden-2-yl, which is unsubstituted or substituted with one or two halogens.


In some further embodiments, the moiety




embedded image


is 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, 4-cyanophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, pyridin-3-yl, pyridin-4-yl, pyrimidin-5-yl, 5-chloro-1H-indol-2-yl, 1H-indol-2-yl, 5-fluoro-1H-indol-2-yl, 5-fluoro-1H-indol-3-yl, 1H-pyrrolo[2,3-b]pyridine-2-yl, 1H-pyrrolo[3,2-b]pyridine-2-yl, 1H-pyrrolo[3,2-c]pyridine-2-yl, pyrazolo[1,5-a]pyridine-2-yl, 2,3-dihydrobenzofuran-6-yl, 5-chloro-1H-benzo[d]imidazol-2-yl, 2,2-difluorobenzo[d][1,3]dioxol-5-yl or 5-bromo-2,3-dihydro-1H-inden-2-yl. In some preferred embodiments, the moiety




embedded image


is 4-bromophenyl, 4-fluorophenyl, 4-chlorophenyl, 5-chloro-1H-indol-2-yl, 5-fluoro-1H-indol-2-yl or 2,2-difluorobenzo[d][1,3]dioxol-4-yl, 2,2-difluorobenzo[d][1,3]dioxol-5-yl.


The Definition of L1

In some embodiments, L1 is a direct bond, C1-6alkylene, C2-6alkenylene, or C3-6cycloalkylene. In some further embodiments, L1 is a direct bond, —CH2—CH2—, —CH═CH—, or




embedded image


In some preferred embodiments, L1 is a direct bond or —CH═CH—. In some preferred embodiments, L1 is —CH═CH—.


The Definition of



embedded image


In some embodiments, X is CRa or N, and Y is CRb or N, wherein Ra and Rb are each independently hydrogen, halogen, hydroxy, or C1-6alkyl. In some embodiments, m is 1, and n is 1; or m is 2, and n is 1; or m is 1, and n is 2; or m is 0, and n is 0; or m is 0, and n is 1; or m is 1, and n is 0. In some further embodiments, X is N and Y is N; and m is 1 and n is 1. In some embodiments, t is 0, 1, or 2, preferably 0.


In some further embodiments, the moiety




embedded image


wherein the symbol ** refers to the position attached to L2 and the symbol




embedded image


refers to the position opposite to L2 in formula (I). In some preferred embodiments, the moiety is




embedded image


In some embodiments, two R3 attached to on the same carbon atom in the ring form a spiro C3-C6 carbon ring. In some embodiments, the moiety




embedded image


The Definition of



embedded image


In some embodiments, X is N, and Y is N. In some embodiments, m is 1, and n is 1; or m is 2, and n is 1; or m is 1, and n is 2; or m is 0, and n is 0; or m is 0, and n is 1; or m is 1, and n is 0. In some further embodiments, X is N and Y is N; and m is 0 and n is 0.


In some further embodiments, the moiety




embedded image


The Definition of L2

In some embodiments, L2 is a direct bond, —C(O)—, *1—NRc—C(O)—*2, *1—C(O)—NRc—*2, *1—CRdRe—NRc—C(O)—*2, *1—NRc—C(O)—CRdRe—*2, or —NRc—, wherein Rc, Rd and Re are each independently hydrogen or C1-6alkyl, and wherein the symbol *2 refers to the position attached to ring B and the symbol *1 refers to the position opposite to ring B in formula (I).


In some further embodiments. L2 is a direct bond, —C(O)—, *1—NH—C(O)—*2, *1—C(O)—NH—*2, *1—CH2—NH—C(O)—*2, or —NH—, wherein the symbol *2 refers to the position attached to ring B and the symbol *1 refers to the position opposite to ring B in formula (I). In some preferred embodiments, L2 is a direct bond or —C(O)—, more preferably —C(O)—.


The Definition of Ring B

In some embodiments, ring B is phenyl, monocyclic 5- to 9-membered heteroaryl, bicyclic 7- to 12-membered heteroaryl, monocyclic 5- to 9-membered heterocyclyl, bicyclic 7- to 12-membered heterocyclyl, monocyclic 3- to 8-membered cycloalkyl, or bicyclic 7- to 12-membered cycloalkyl, each of which is unsubstituted or substituted with one or two substituents selected from halogen, oxo, C1-6alkyl, haloC1-6alkyl, or —ORf, wherein Rf is hydrogen, C1-6alkyl, haloC1-6alkyl, C1-6alkoxy-C1-6alkyl-, or monocyclic 5- to 9-membered heterocyclyl.


In some further embodiments, ring B is phenyl which is substituted with —ORf, wherein Rf is hydrogen, C1-6alkyl, haloC1-6alkyl, or monocyclic 5- to 9-membered heterocyclyl, preferably monocyclic 5- to 9-membered heterocyclyl comprising one nitrogen or oxygen heteroatom, more preferably azetidinyl or tetrahydrofuranyl, e.g., azetidin-3-yl or tetrahydrofuran-3-yl. Preferably, ring B is phenyl which is methoxy, difluoromethoxy, azetidin-3-yloxy, or (tetrahydrofuran-3-yl)oxy.


In some further embodiments, ring B is monocyclic 3- to 8-membered cycloalkyl, which is unsubstituted or substituted with one or two substituents selected from halogen, C1-6alkyl, haloC1-6alkyl, or —ORf, wherein Rf is hydrogen or C1-6alkyl. Preferably, ring B is cyclopropyl or cyclohexyl, which is unsubstituted or substituted with hydroxy or methoxy.


In some further embodiments, ring B is monocyclic 5- to 9-membered heteroaryl, which is unsubstituted or substituted with one or two substituents selected from oxo, halogen, or —ORf, wherein Rf is hydrogen, C1-6alkyl or C1-6alkoxy-C1-6alkyl-. Preferably, ring B is pyridinyl, pyrimidinyl, or pyrazinyl, which is unsubstituted or substituted as defined above. More preferably, ring B is pyridin-3-yl, pyridin-2-yl, pyridin-4-yl, pyridin-5-yl, pyrimidin-5-yl, pyrimidin-4-yl, or pyrazine-2-yl, which is unsubstituted or substituted with oxo, methoxy, or 2-methoxyethoxy.


In some further embodiments, ring B is monocyclic 5- to 9-membered heterocyclyl or bicyclic 7- to 12-membered heterocyclyl, each of which is unsubstituted or substituted with oxo, halogen, or C1-6alkyl. Preferably, ring B is piperidinyl, tetrahydropyranyl, oxetanyl, morpholino, benzodioxolyl, or dihydrobenzofuranyl; more preferably, ring B is piperidine-4-yl, tetrahydro-2H-pyran-4-yl, oxetan-3-yl, morpholino, 2,2-difluorobenzo[d][1,3]dioxol-4-yl, 2,2-difluorobenzo[d][1,3]dioxol-5-yl or 2,3-dihydrobenzofuran-5-yl.


In some embodiments, ring B is monocyclic 5- to 9-membered heteroaryl or bicyclic 7- to 12-membered heteroaryl, which is unsubstituted or substituted with one or two substituents selected from oxo, halogen, or C1-6alkyl. Preferably, ring B is imidazolyl, indolinyl, benzofuranyl, or benzoimidazolyl, which is unsubstituted or substituted with oxo; more preferably, ring B is 1H-imidazol-4-yl, 1H-indol-5-yl, benzofuran-5-yl, or 1H-benzo[d]imidazol-5-yl, each of which is unsubstituted or substituted with oxo.


In some further embodiments, the moiety




embedded image


is 4-methoxyphenyl, 4-(difluoro methoxy)phenyl, 4-(azetidin-3-yloxy)phenyl, 4-((tetrahydrofuran-3-yl)oxy)phenyl, 4-((tetrahydrofuran-3-yl)oxy)phenyl, 4-methoxycyclohexyl, 4-hydroxycyclohexyl, cyclopropyl, 6-methoxypyridin-3-yl, 5-methoxypyridin-2-yl, 6-oxo-1,6-dihydropyridin-3-yl, 6-(2-methoxyethoxy)pyridin-3-yl, 2-methoxypyridin-4-yl, 2-oxo-pyridin-5-yl, 2-methoxypyrimidin-5-yl, 2-methoxy pyrimidin-4-yl, 5-methoxypyrazine-2-yl, piperidine-4-yl, tetrahydro-2H-pyran-4-yl, oxetan-3-yl, morpholino, 1H-imidazol-4-yl, 2,2-difluorobenzo[d][1,3]dioxol-5-yl, 2,3-dihydrobenzo[b][1,4]dioxine-6-yl, 2,2-difluoro benzo[d][1,3]dioxol-4-yl, 2,3-dihydrobenzofuran-5-yl, 1,3-dihydro-2H-2-oxo-benzo[d]imidazol-5-yl, 1H-benzo[d]imidazol-5-yl, 2-oxo-indolin-5-yl, 1H-indol-5-yl, or benzofuran-5-yl. In some preferred embodiments, the moiety




embedded image


is 4-methoxycyclohexyl, oxetan-3-yl, tetrahydro-2H-pyran-4-yl, 2,2-difluorobenzo[d][1,3]dioxol-5-yl, 2,3-dihydrobenzo[b][1,4]dioxine-6-yl, 2-oxo-pyridin-5-yl, 2-oxo-indolin-5-yl, 6-methoxypyridin-3-yl, 1H-indol-5-yl, benzofuran-5-yl, 1H-benzo[d]imidazol-5-yl, 2-methoxypyrimidin-5-yl, 6-(2-methoxyethoxy)pyridin-3-yl or 2,2-difluoro benzo[d][1,3]dioxol-4-yl, more preferably oxetan-3-yl, tetrahydro-2H-pyran-4-yl, 6-methoxypyridin-3-yl, 2-methoxypyrimidin-5-yl, or 6-methoxypyridin-3-yl.


Provided is a compound of formula (IA)




embedded image


or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof,


wherein

    • ring A is bicyclic 7- to 12-membered heterocyclyl;
    • p is 0, 1, or 2;
    • R1 is halogen, cyano, C1-6alkyl, or haloC1-6alkyl;
    • L1 is C1-6alkylene, C2-6alkenylene, C2-6alkynylene or C3-6cycloalkylene;
    • X is CRa or N, and Y is CRb or N, wherein Ra and Rb are each independently hydrogen, halogen, hydroxy, or C1-6alkyl;
    • m and n are each independently 0, 1, or 2;
    • t is 0, 1, or 2;
    • R3 is halogen, C1-6alkyl, or haloC1-6alkyl;
    • L2 is a direct bond, —C(O)—, *1—NRc—C(O)—*2, *1—C(O)—NRc—*2, *1—CRdRe—NRc—C(O)—*2, *1—NRc—C(O)—CRdRe—*2, or —NRc—, wherein Rc, Rd and Re are each independently hydrogen or C1-6alkyl, and wherein the symbol *2 refers to the position attached to ring B and the symbol *1 refers to the position opposite to ring B in formula (I);
    • ring B is phenyl, monocyclic 5- to 9-membered heteroaryl, bicyclic 7- to 12-membered heteroaryl, monocyclic 5- to 9-membered heterocyclyl, bicyclic 7- to 12-membered heterocyclyl, monocyclic 3- to 8-membered cycloalkyl, or bicyclic 7- to 12-membered cycloalkyl;
    • q is 0, 1 or 2;
    • R2 is halogen, oxo, C1-6alkyl, haloC1-6alkyl, or —ORf, wherein Rf is hydrogen, C1-6alkyl, haloC1-6alkyl, C1-6alkoxy-C1-6alkyl-, or monocyclic 5- to 9-membered heterocyclyl.


In some embodiments, ring A is bicyclic 7- to 12-membered heterocyclyl which is benzo fused heterocyclyl. In some further embodiments, the benzo fused heterocyclyl is indolinyl, isoindolinyl, benzopyranyl, dihydrothiazolopyrimidinyl, tetrahydroquinolyl, tetrahydroisoquinolyl, dihydrobenzofuranyl, dihydrobenzoxazinyl, dihydrobenzoimidazolyl, tetrahydrobenzothienyl, tetrahydrobenzofuranyl, benzodioxolyl, benzodioxonyl, chromanyl, chromenyl, octahydrochromenyl, dihydrobenzodioxynyl, dihydrobenzoxezinyl, dihydrobenzodioxepinyl, dihydrothienodioxynyl, dihydrobenzooxazepinyl, tetrahydrobenzooxazepinyl, dihydrobenzoazepinyl, tetrahydrobenzoazepinyl, isochromanyl, or chromanyl.


In some further embodiments, ring A is bicyclic 7- to 12-membered heterocyclyl selected from benzodioxolyl or dihydrobenzofuranyl, preferably benzo[d][1,3]dioxol-5-yl or 2,3-dihydrobenzofuran-6-yl, each of which is unsubstituted or substituted with one or two halogens.


In some embodiments, the variables L1, R3, t, X, Y, n, m, X, Y, L2, ring B, R2, and q are defined as for formula (I).


Provided is a compound of formula (III)




embedded image


or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein the variables L1, R3, t, X, Y, n, m, X, Y, L2, ring B, R2, and q are defined as for formula (I).


In some embodiments for formula (IA) or (III), L1 is C2-6alkenylene. In some further embodiments, L1 is —CH═CH—.


In some embodiments for formula (IA) or (III), X is N and Y is N; and m is 1 and n is 1. In some embodiments, t is 0. In some embodiments, L2 is —C(O)—.


In some embodiments for formula (IA) or (III), ring B is phenyl, monocyclic 5- to 9-membered heteroaryl, bicyclic 7- to 12-membered heteroaryl, monocyclic 5- to 9-membered heterocyclyl, bicyclic 7- to 12-membered heterocyclyl, monocyclic 3- to 8-membered cycloalkyl, or bicyclic 7- to 12-membered cycloalkyl, each of which is unsubstituted or substituted with one or two substituents selected from halogen, oxo, C1-6alkyl, haloC1-6alkyl, or —ORf, wherein Rf is hydrogen, C1-6alkyl, haloC1-6alkyl, C1-6alkoxy-C1-6alkyl-, or monocyclic 5- to 9-membered heterocyclyl.


In some further embodiments for formula (IA) or (III), ring B is phenyl which is substituted with —ORf, wherein Rf is hydrogen, C1-6alkyl, haloC1-6alkyl, or monocyclic 5- to 9-membered heterocyclyl, preferably monocyclic 5- to 9-membered heterocyclyl comprising one nitrogen or oxygen heteroatom, more preferably azctidinyl or tetrahydrofuranyl, e.g., azctidin-3-yl or tetrahydrofuran-3-yl. Preferably, ring B is phenyl which is methoxy, difluoromethoxy, azetidin-3-yloxy, or (tetrahydrofuran-3-yl)oxy.


In some further embodiments for formula (IA) or (III), ring B is monocyclic 3- to 8-membered cycloalkyl, which is unsubstituted or substituted with one or two substituents selected from halogen, C1-6alkyl, haloC1-6alkyl, or —ORf, wherein Rf is hydrogen or C1-6alkyl. Preferably, ring B is cyclopropyl or cyclohexyl, which is unsubstituted or substituted with hydroxy or methoxy.


In some further embodiments for formula (IA) or (III), ring B is monocyclic 5- to 9-membered heteroaryl, which is unsubstituted or substituted with one or two substituents selected from oxo, halogen, or —ORf, wherein Rf is hydrogen, C1-6alkyl or C1-6alkoxy-C1-6alkyl-. Preferably, ring B is pyridinyl, pyrimidinyl, or pyrazinyl, which is unsubstituted or substituted as defined above. More preferably, ring B is pyridin-3-yl, pyridin-2-yl, pyridin-4-yl, pyridin-5-yl, pyrimidin-5-yl, pyrimidin-4-yl, or pyrazine-2-yl, which is unsubstituted or substituted with oxo, methoxy, or 2-methoxyethoxy.


In some further embodiments for formula (IA) or (III), ring B is monocyclic 5- to 9-membered heterocyclyl or bicyclic 7- to 12-membered heterocyclyl, each of which is unsubstituted or substituted with oxo, halogen, or C1-6alkyl. Preferably, ring B is piperidinyl, tetrahydropyranyl, oxctanyl, morpholino, benzodioxolyl, or dihydrobenzofuranyl; more preferably, ring B is piperidine-4-yl, tetrahydro-2H-pyran-4-yl, oxctan-3-yl, morpholino, 2,2-difluorobenzo[d][1,3]dioxol-4-yl, 2,2-difluorobenzo[d][1,3]dioxol-5-yl or 2,3-dihydrobenzofuran-5-yl.


In some embodiments for formula (IA) or (III), ring B is monocyclic 5- to 9-membered heteroaryl or bicyclic 7- to 12-membered heteroaryl, which is unsubstituted or substituted with one or two substituents selected from oxo, halogen, or C1-6alkyl. Preferably, ring B is imidazolyl, indolinyl, benzofuranyl, or benzoimidazolyl, which is unsubstituted or substituted with oxo; more preferably, ring B is 1H-imidazol-4-yl, 1H-indol-5-yl, benzofuran-5-yl, or 1H-benzo[d]imidazol-5-yl, each of which is unsubstituted or substituted with oxo.


In some further embodiments for formula (IA) or (III), the moiety




embedded image


is 4-methoxyphenyl, 4-(difluoro methoxy)phenyl, 4-(azctidin-3-yloxy)phenyl, 4-((tetrahydrofuran-3-yl)oxy)phenyl, 4-((tetrahydrofuran-3-yl)oxy)phenyl, 4-methoxycyclohexyl, 4-hydroxycyclohexyl, cyclopropyl, 6-methoxypyridin-3-yl, 5-methoxypyridin-2-yl, 6-oxo-1,6-dihydropyridin-3-yl, 6-(2-methoxyethoxy)pyridin-3-yl, 2-methoxypyridin-4-yl, 2-oxo-pyridin-5-yl, 2-methoxypyrimidin-5-yl, 2-methoxy pyrimidin-4-yl, 5-methoxypyrazine-2-yl, piperidine-4-yl, tetrahydro-2H-pyran-4-yl, oxetan-3-yl, morpholino, 1H-imidazol-4-yl, 2,2-difluorobenzo[d][1,3]dioxol-5-yl, 2,3-dihydrobenzo[b][1,4]dioxine-6-yl, 2,2-difluoro benzo[d][1,3]dioxol-4-yl, 2,3-dihydrobenzofuran-5-yl, 1,3-dihydro-2H-2-oxo-benzo[d]imidazol-5-yl, 1H-benzo[d]imidazol-5-yl, 2-oxo-indolin-5-yl, 1H-indol-5-yl, or benzofuran-5-yl. In some preferred embodiments, the moiety




embedded image


methoxycyclohexyl, oxetan-3-yl, tetrahydro-2H-pyran-4-yl, 2,2-difluorobenzo[d][1,3]dioxol-5-yl, 2,3-dihydrobenzo[b][1,4]dioxine-6-yl, 2-oxo-pyridin-5-yl, 2-oxo-indolin-5-yl, 6-methoxypyridin-3-yl, 1H-indol-5-yl, benzofuran-5-yl, 1H-benzo[d]imidazol-5-yl, 2-methoxypyrimidin-5-yl, 6-(2-methoxyethoxy)pyridin-3-yl or 2,2-difluoro benzo[d][1,3]dioxol-4-yl, more preferably oxetan-3-yl, tetrahydro-2H-pyran-4-yl, 6-methoxypyridin-3-yl, 2-methoxypyrimidin-5-yl, or 6-methoxypyridin-3-yl.


In alternative embodiments, the compound is selected from




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


Provided is a pharmaceutical composition comprising a compound disclosed herein or a stereoisomer thereof or a pharmaceutically acceptable salt thereof, optionally together with a pharmaceutically acceptable excipient.


Provided is a method of treating a disease mediated by GPR183, comprising administering a subject in need thereof a compound disclosed herein or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof. In some embodiments, the disease mediated by GPR183 is cancer, autoimmune diseases, liver diseases, osteoporosis, and neuropathic pain. In one embodiment, the cancer is blood, brain, breast, colorectal, gastrointestinal, liver, lung, ovarian, pancreatic, prostate, skin, or uterine cancer. In some embodiments, the cancer produces molecules involved in Epstein-Barr virus (EBV)-induced G-protein coupled receptor 2 (EBI2) mediated signaling.







DETAILED DESCRIPTION OF THE INVENTION

The following terms have the indicated meanings throughout the specification:


Unless specifically defined elsewhere in this document, all other technical and scientific terms used herein have the meaning commonly understood by one of ordinary skill in the art to which this invention belongs.


The following terms have the indicated meanings throughout the specification:


As used herein, including the appended claims, the singular forms of words such as “a”, “an”, and “the”, include their corresponding plural references unless the context clearly indicates otherwise.


The term “or” is used to mean, and is used interchangeably with, the term “and/or” unless the context clearly dictates otherwise.


The term “alkyl” includes a hydrocarbon group selected from linear and branched, saturated hydrocarbon groups comprising from 1 to 18, such as from 1 to 12, further such as from 1 to 10, more further such as from 1 to 8, or from 1 to 6, or from 1 to 4, carbon atoms. Examples of alkyl groups comprising from 1 to 6 carbon atoms (i.e., C1-6 alkyl) include, but not limited to, methyl, ethyl, 1-propyl or n-propyl (“n-Pr”), 2-propyl or isopropyl (“i-Pr”), 1-butyl or n-butyl (“n-Bu”), 2-methyl-1-propyl or isobutyl (“i-Bu”), 1-methylpropyl or s-butyl (“s-Bu”), 1,1-dimethylethyl or t-butyl (“t-Bu”), 1-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl-3-pentyl, 2,3-dimethyl-2-butyl and 3,3-dimethyl-2-butyl groups.


The term “alkylene” refers to a bivalent alkyl as defined above.


The term “halogen” includes fluoro (F), chloro (Cl), bromo (Br) and iodo (I).


The term “haloalkyl” includes an alkyl group in which one or more hydrogen is/are replaced by one or more halogen atoms such as fluoro, chloro, bromo, and iodo. Examples of the haloalkyl include haloC1-8alkyl, haloC1-6alkyl or halo C1-4alkyl, but not limited to —CF3, —CH2Cl, —CH2CF3, —CHCl2, —CF3, and the like.


The term “alkenyl” includes a hydrocarbon group selected from linear and branched hydrocarbon groups comprising at least one C═C double bond and from 2 to 18, such as from 2 to 8, further such as from 2 to 6, carbon atoms. Examples of the alkenyl group, e.g., C2-6 alkenyl, include, but not limited to ethenyl or vinyl, prop-1-enyl, prop-2-enyl, 2-methylprop-1-enyl, but-1-enyl, but-2-enyl, but-3-enyl, buta-1,3-dienyl, 2-methylbuta-1,3-dienyl, hex-1-enyl, hex-2-enyl, hex-3-enyl, hex-4-enyl, and hexa-1,3-dienyl groups.


The term “alkenylene” refers to a bivalent alkenyl as defined above.


The term “alkynyl” includes a hydrocarbon group selected from linear and branched hydrocarbon group, comprising at least one C═C triple bond and from 2 to 18, such as 2 to 8, further such as from 2 to 6, carbon atoms. Examples of the alkynyl group, e.g., C2-6 alkynyl, include, but not limited to ethynyl, 1-propynyl, 2-propynyl (propargyl), 1-butynyl, 2-butynyl, and 3-butynyl groups.


The term “alkynylene” refers to a bivalent alkynyl as defined above.


The term “cycloalkyl” includes a hydrocarbon group selected from saturated cyclic hydrocarbon groups, comprising monocyclic and polycyclic (e.g., bicyclic and tricyclic) groups including fused, bridged or spiro cycloalkyl.


For example, the cycloalkyl group may comprise from 3 to 12, such as from 3 to 10, further such as 3 to 8, further such as 3 to 6, 3 to 5, or 3 to 4 carbon atoms. Even further for example, the cycloalkyl group may be selected from monocyclic group comprising from 3 to 12, such as from 3 to 10, further such as 3 to 8, 3 to 6 carbon atoms. Examples of the monocyclic cycloalkyl group include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, and cyclododecyl groups. In particular, Examples of the saturated monocyclic cycloalkyl group, e.g., C3-8cycloalkyl, include, but not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl groups. In a preferred embedment, the cycloalkyl is a monocyclic ring comprising 3 to 6 carbon atoms (abbreviated as C3-6 cycloalkyl), including but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Examples of the bicyclic cycloalkyl groups include those having from 7 to 12 ring atoms arranged as a fused bicyclic ring selected from [4,4], [4,5], [5,5], [5,6] and [6,6] ring systems, or as a bridged bicyclic ring selected from bicyclo[2.2.1]heptane. bicyclo[2.2.2]octane, and bicyclo[3.2.2]nonane. Further Examples of the bicyclic cycloalkyl groups include those arranged as a bicyclic ring selected from [5,6] and [6,6] ring systems.


The term “heteroaryl” includes a group selected from:

    • 5- to 9-membered (e.g., 5-, 6-, 7-, 8- or 9-membered) aromatic, monocyclic rings comprising at least one heteroatom, for example, from 1 to 4, or, in some embodiments, from 1 to 3, in some embodiments, from 1 to 2, heteroatoms, selected from nitrogen (N), sulfur (S) and oxygen (O), with the remaining ring atoms being carbon;
    • 7- to 12-membered bicyclic rings comprising at least one heteroatom, for example, from 1 to 4, or, in some embodiments, from 1 to 3, or, in other embodiments, 1 or 2, heteroatoms, selected from N, O, and S, with the remaining ring atoms being carbon and wherein at least one ring is aromatic and at least one heteroatom is present in the aromatic ring; and
    • 11- to 14-membered tricyclic rings comprising at least one heteroatom, for example, from 1 to 4, or in some embodiments, from 1 to 3, or, in other embodiments, 1 or 2, heteroatoms, selected from N, O, and S, with the remaining ring atoms being carbon and wherein at least one ring is aromatic and at least one heteroatom is present in an aromatic ring.


When the total number of S and O atoms in the heteroaryl group exceeds 1, those heteroatoms are not adjacent to one another. In some embodiments, the total number of S and O atoms in the heteroaryl group is not more than 2. In some embodiments, the total number of S and O atoms in the aromatic heterocycle is not more than 1. When the heteroaryl group contains more than one heteroatom ring member, the heteroatoms may be the same or different. The nitrogen atoms in the ring(s) of the heteroaryl group can be oxidized to form N-oxides.


“Heterocyclyl”, “heterocycle” or “heterocyclic” are interchangeable and include a non-aromatic heterocyclyl group comprising one or more, e.g., 1 to 3, heteroatoms selected from nitrogen, oxygen or optionally oxidized sulfur as ring members, with the remaining ring members being carbon, including monocyclic, fused, bridged, and spiro ring, i.e., containing monocyclic heterocyclyl, bridged heterocyclyl, spiro heterocyclyl, and fused heterocyclic groups.


The term “fused heterocyclyl” refers to a 5 to 20-membered polycyclic heterocyclyl group, wherein each ring in the system shares an adjacent pair of atoms (carbon and carbon atoms or carbon and nitrogen atoms) with another ring, comprising one or more heteroatoms selected from nitrogen, oxygen or optionally oxidized sulfur as ring members, with the remaining ring members being carbon. One or more rings of a fused heterocyclic group may contain one or more double bonds, but the fused heterocyclic group does not have a completely conjugated pi-electron system. Preferably, a fused heterocyclyl is 6 to 14-membered, and more preferably 7 to 12-membered, or 7- to 10-membered. According to the number of membered rings, a fused heterocyclyl is divided into bicyclic, tricyclic, tetracyclic, or polycyclic fused heterocyclyl. The group can be attached to the remainder of the molecule through cither ring.


Specifically, the term “bicyclic fused heterocyclyl” refers to a 7 to 12-membered (also referred to as bicyclic 7- to 12-membered heterocyclyl), preferably 7- to 10-membered, more preferably 9- or 10-membered fused heterocyclyl as defined herein comprising two fused rings and comprising 1 to 4 heteroatoms selected from nitrogen, oxygen or optionally oxidized sulfur as ring members. Typically, a bicyclic fused heterocyclyl is 5-membered/5-membered, 5-membered/6-membered, 6-membered/6-membered, or 6-membered/7-membered bicyclic fused heterocyclyl. Representative examples of (bicyclic) fused heterocycles include, but not limited to, the following groups octahydrocyclopenta[c]pyrrolc, octahydropyrrolo[3, 4-c]pyrrolyl, octahydroisoindolyl, isoindolinyl, octahydro-benzo[b][1, 4]dioxin, indolinyl, isoindolinyl, benzopyranyl, dihydrothiazolopyrimidinyl, tetrahydroquinolyl, tetrahydroisoquinolyl (or tetrahydroisoquinolinyl), dihydrobenzofuranyl, dihydrobenzoxazinyl, dihydrobenzoimidazolyl, tetrahydrobenzothienyl, tetrahydrobenzofuranyl, benzodioxolyl, benzodioxonyl, chromanyl, chromenyl, octahydrochromenyl, dihydrobenzodioxynyl, dihydrobenzoxczinyl, dihydrobenzodioxepinyl, dihydrothienodioxynyl, dihydrobenzooxazepinyl, tetrahydrobenzooxazepinyl, dihydrobenzoazepinyl, tetrahydrobenzoazepinyl, isochromanyl, chromanyl, or tetrahydropyrazolopyrimidinyl (e.g., 4, 5, 6, 7-tetrahydropyrazolo[1, 5-a]pyrimidin-3-yl).


The term “benzo fused heterocyclyl” is a bicyclic fused heterocyclyl in which a monocyclic 4 to 9-membered heterocyclyl as defined herein (preferably 5- or 6-membered) fused to a benzene ring. Representative examples of benzo fused heterocyclyl includes indolinyl, isoindolinyl, benzopyranyl, dihydrothiazolopyrimidinyl, tetrahydroquinolyl, tetrahydroisoquinolyl, dihydrobenzofuranyl, dihydrobenzoxazinyl, dihydrobenzoimidazolyl, tetrahydrobenzothienyl, tetrahydrobenzofuranyl, benzodioxolyl, benzodioxonyl, chromanyl, chromenyl, octahydrochromenyl, dihydrobenzodioxynyl, dihydrobenzoxczinyl, dihydrobenzodioxepinyl, dihydrothienodioxynyl, dihydrobenzooxazepinyl, tetrahydrobenzooxazepinyl, dihydrobenzoazepinyl, tetrahydrobenzoazepinyl, isochromanyl, or chromanyl.


The term “stercoisomer” refers to all isomers of individual compounds that differ only in the orientation of their atoms in space. The term stercoisomer includes mirror image isomers (enantiomers), mixtures of mirror image isomers (racemates, racemic mixtures), geometric (cis/trans or syn/anti or E/Z) isomers, and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereoisomers).


Compounds disclosed herein may contain an asymmetric center and may thus exist as enantiomers. “Enantiomers” refer to two stereoisomers of a compound which are non-superimposable mirror images of one another. Where the compounds disclosed herein possess two or more asymmetric centers, they may additionally exist as diastereomers. Enantiomers and diastercomers fall within the broader class of stereoisomers. All such possible stereoisomers as substantially pure resolved enantiomers, racemic mixtures thereof, as well as mixtures of diastercomers are intended to be included. All stercoisomers of the compounds disclosed herein and/or pharmaceutically acceptable salts thereof are intended to be included. Unless specifically mentioned otherwise, reference to one isomer applies to any of the possible isomers. Whenever the isomeric composition is unspecified, all possible isomers are included.


When compounds disclosed herein contain olefinic double bonds, unless specified otherwise, such double bonds are meant to include both E and Z geometric isomers.


When compounds disclosed herein contain a di-substituted cyclic ring system, substituents found on such ring system may adopt cis and trans formations. Cis formation means that both substituents are found on the upper side of the 2 substituent placements on the carbon, while trans would mean that they were on opposing sides. For example, the di-substituted cyclic ring system may be cyclohexyl or cyclobutyl ring.


It may be advantageous to separate reaction products from one another and/or from starting materials. The desired products of each step or series of steps is separated and/or purified (hereinafter separated) to the desired degree of homogeneity by the techniques common in the art. Typically such separations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (“SMB”) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography. One skilled in the art could select and apply the techniques most likely to achieve the desired separation.


“Diastercomers” refer to stercoisomers of a compound with two or more chiral centers but which are not mirror images of one another. Diastercomeric mixtures can be separated into their individual diastercomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastercomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Enantiomers can also be separated by use of a chiral HPLC column.


A single stereoisomer, e.g., a substantially pure enantiomer, may be obtained by resolution of the racemic mixture using a method such as formation of diastercomers using optically active resolving agents (Eliel, E. and Wilen, S. Stereochemistry of Organic Compounds. New York: John Wiley & Sons, Inc., 1994; Lochmuller, C. H., et al. “Chromatographic resolution of enantiomers: Selective review.” J. Chromatogr., 113(3) (1975): pp. 283-302). Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastercomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastercomeric compounds with chiral derivatizing reagents, separation of the diastercomers, and conversion to the pure stercoisomers, and (3) separation of the substantially pure or enriched stercoisomers directly under chiral conditions. See: Wainer, Irving W., Ed. Drug Stereochemistry: Analytical Methods and Pharmacology. New York: Marcel Dekker. Inc., 1993.


“Pharmaceutically acceptable salts” refer to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without unduc toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio. A pharmaceutically acceptable salt may be prepared in situ during the final isolation and purification of the compounds disclosed herein, or separately by reacting the free base function with a suitable organic acid or by reacting the acidic group with a suitable base.


In addition, if a compound disclosed herein is obtained as an acid addition salt, the free base can be obtained by basifying a solution of the acid salt. Conversely, if the product is a free base, an addition salt, such as a pharmaceutically acceptable addition salt, may be produced by dissolving the frec base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds. Those skilled in the art will recognize various synthetic methodologies that may be used without undue experimentation to prepare non-toxic pharmaceutically acceptable addition salts.


As defined herein, “a pharmaceutically acceptable salt thereof” includes salts of at least one compound of Formula (I), and salts of the stereoisomers of the compound of Formula (I), such as salts of enantiomers, and/or salts of diastercomers.


The terms “administration”, “administering”, “treating” and “treatment” herein, when applied to an animal, human, experimental subject, cell, tissuc, organ, or biological fluid, mean contact of an exogenous pharmaceutical, therapeutic, diagnostic agent, or composition to the animal, human, subject, cell, tissue, organ, or biological fluid. Treatment of a cell encompasses contact of a reagent to the cell, as well as the contact of a reagent to a fluid, where the fluid is in contact with the cell. The term “administration” and “treatment” also means in vitro and ex vivo treatments, e.g., of a cell, by a reagent, diagnostic, binding compound, or by another cell. The term “subject” hercin includes any organism, preferably an animal, more preferably a mammal (e.g., rat, mouse, dog, cat, and rabbit) and most preferably a human.


The term “effective amount” or “therapeutically effective amount” refers to an amount of the active ingredient, such as a compound that, when administered to a subject for treating a disease, or at least one of the clinical symptoms of a disease or disorder, is sufficient to affect such treatment for the disease, disorder, or symptom. The term “therapeutically effective amount” can vary with the compound, the disease, disorder, and/or symptoms of the disease or disorder, severity of the disease, disorder, and/or symptoms of the disease or disorder, the age of the subject to be treated, and/or the weight of the subject to be treated. An appropriate amount in any given instance can be apparent to those skilled in the art or can be determined by routine experiments. In some embodiments, “therapeutically effective amount” is an amount of at least one compound and/or at least one stereoisomer thereof, and/or at least one pharmaceutically acceptable salt thereof disclosed herein effective to “treat” as defined herein, a disease or disorder in a subject. In the case of combination therapy, the term “therapeutically effective amount” refers to the total amount of the combination objects for the effective treatment of a disease, a disorder or a condition.


The term “disease” refers to any disease, discomfort, illness, symptoms or indications, and can be interchangeable with the term “disorder” or “condition”.


Throughout this specification and the claims which follow, unless the context requires otherwise, the term “comprise”, and variations such as “comprises” and “comprising” are intended to specify the presence of the features thereafter, but do not exclude the presence or addition of one or more other features. When used herein the term “comprising” can be substituted with the term “containing”, “including” or sometimes “having”.


Throughout this specification and the claims which follow, the term “Cn-m” indicates a range which includes the endpoints, wherein n and m are integers and indicate the number of carbons. Examples include C1-8, C1-6, and the like.


Unless specifically defined elsewhere in this document, all other technical and scientific terms used herein have the meaning commonly understood by one of ordinary skill in the art to which this invention belongs.












ABBREVIATIONS


















ACN
Acetonitrile



DCM
Dichloromethane



DIEA
N,N-Diisopropylethylamine



EA
Ethyl Acrylate



HATU
2-(1H-7-Azabenzotriazol-1-yl)-1,1,3,3-tetramethyl




uroniumhexafluorophosphate Methanaminium



HPLC
high performance liquid chromatography



LCMS
liquid chromatograph mass spectrometer



PE
Petroleum Ether



RT
Room temperature



TCFH
N,N,N′,N′-tetramethylchloroformamidinium




Hexafluorophosphate



TFA
Trifluoroacetic Acid



T3P
Propylphosphonic anhydride










EXAMPLES

Example 1: (E)-1-(4-(1H-benzo[d]imidazole-5-carbonyl)piperazin-1-yl)-3-(4-bromophenyl)prop-2-en-1-one (33)




embedded image


Step 1: tert-butyl (E)-4-(3-(4-bromophenyl)acryloyl)piperazine-1-carboxylate (33-3)

To a solution of (E)-3-(4-bromophenyl)acrylic acid (33-2) (1.00 g, 4.40 mmol) in DCM (15 mL) were added DIEA (2.92 mL, 17.6 mmol) and T3P (8.40 g, 13.2 mmol, 50% in EA). The reaction was stirred at room temperature for 30 min before tert-butyl piperazine-1-carboxylate (33-1) (0.98 g, 5.28 mmol) was added. The reaction mixture was stirred at room temperature for 1.5 hr. The mixture was concentrated in vacuum. The residue was mixed with water (30 mL) and the resulting mixture was extracted with EA (30 mL*2). The organic layers were combined, washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was slurried with EA (2 mL) and PE (15 mL) to afford the subtitled compound (33-3) (1.33 g, 3.36 mmol, 76.4% yield) as a white solid. LC-MS (ESI): m/z 341.0 [M−55]+


Step 2: (E)-3-(4-bromophenyl)-1-(piperazin-1-yl)prop-2-en-1-one hydrochloride (33-4)

A mixture of Compound 33-3 (800 mg, 2.02 mmol) and HCl/dioxane (6 mL, 4.0 M) was stirred at room temperature for 2 hr. LCMS showed that the reaction was completed. The mixture was concentrated in vacuum to afford the subtitled compound (33-4) as a white solid. LC-MS (ESI): m/z 297.0 [M+H]+


Step 3: (E)-1-(4-(1H-benzo[d]imidazole-5-carbonyl)piperazin-1-yl)-3-(4-bromophenyl)prop-2-en-1-one (33)

To a solution of 1H-benzo[d]imidazole-5-carboxylic acid (33-5) (104 mg, 0.64 mmol) in DCM (10 mL) were added DIEA (0.27 mL, 1.61 mmol), HATU (244 mg, 0.64 mmol) and Compound 33-4 (150 mg, 0.54 mmol). Then the reaction mixture was stirred at room temperature for 2 h. The mixture was diluted with H2O (25 mL) and the resulting mixture was extracted with DCM (25 mL*2). The organic layers were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by prep-HPLC (Waters 2767/2545/2489/Qda, Waters sunfire C18 10 um OBD 19*250 mm, Mobile Phase A: 0.1% TFA in water, Mobile Phase B: CH3CN, Flow: 20 mL/min, Column temp: RT) to afford the titled compound (33) (1.5 mg, 0.00 mmol, 0.6% yield). LC-MS (ESI): m/z 439.1/441.1 [M−H]. 1H NMR (400 MHz, DMSO-d6) δ 12.64 (s, 1H), 8.49-8.24 (m, 2H), 7.72-7.58 (m, 5H), 7.52-7.46 (m, 1H), 7.36-7.23 (m, 2H), 3.85-3.73 (m, 2H), 3.67-3.48 (m, 6H).


The compounds below were synthesized following the procedures similar to those for Compound 33:

















MS ESI



No.
Structure/Name
[M + H]+

1H NMR (400 MHz, DMSO-d6) δ



















1


embedded image


389.2
7.41-7.39 (m, 1H), 7.39-7.37 (m, 1H), 7.36-7.27 (m, 2H), 7.12-7.07 (m, 1H), 7.02-6.99 (m, 1H), 6.99-6.96 (m, 1H), 3.80 (s, 3H), 3.49 (s, 8H), 2.81 (t, J = 7.6 Hz, 2H), 2.66 (t, J = 7.5 Hz, 2H).






3-(3,4-difluoropheny1)-1-(4-(4-





methoxybenzoyl)piperazin-1-y1)propan-1-one







2


embedded image


387.2
8.00-7.91 (m, 1H), 7.57 (s, 1H), 7.53- 7.45 (m, 2H), 7.43 (t, J = 7.3 Hz, 2H), 7.31 (d, J = 15.3 Hz, 1H), 7.00 (d, J = 8.7 Hz, 2H), 3.82-3.75 (m, 5H), 3.70-3.45 (m, 6H)






(E)-3-(3,4-difluorophenyl)-1-(4-(4-





methoxybenzoyl)piperazin-1-y1)prop-2-en-1-





one







3


embedded image


441.1
7.67-7.64 (m, 2H), 7.62-7.61 (m, 2H), 7.61-7.58 (m, 2H), 7.43 (s, 0.5H), 7.39 (s, 0.5H), 7.01-6.99 (m, 1H), 6.98-6.91 (m, 1H), 6.76-6.72 (m,1H), 4.51-4.43 (m, 4H), 4.20 (s, 2H), 4.12 (s, 2H), 3.81 (s, 3H).






(E)-3-(4-bromophenyl)-1-(6-(4-





methoxybenzoyl)-2,6-diazaspiro[3.3]heptan-2-





y1)prop-2-en-1-one







4


embedded image


445.1
7.65 (t, J = 14.6 Hz, 1H), 7.51 (t, J = 6.9 Hz, 2H), 7.42-7.28 (m, 4H), 6.99- 6.74 (m, 3H), 3.89 (s, 1H), 3.86-3.59 (m, 9H), 3.57-3.42 (m, 1H), 2.09 (s, 1H), 1.79 (s, 1H).






(E)-3-(4-bromophenyl)-1-(4-(4-





methoxybenzoyl)-1,4-diazepan-1-y1)prop-2-en-





1-one







5


embedded image


443.1
7.70 (d, J = 8.5 Hz, 2H), 7.61 (d, J = 8.5 Hz, 2H), 7.54-7.46 (m, 1H), 7.40- 7.36 (m, 2H), 7.36-7.21 (m, 1H), 7.02-7.00 (m, 1H), 7.00-6.97 (m, 1H), 4.49-4.10 (m, 3H), 4.06-3.75 (m, 4H), 3.42 (d, J = 12.3 Hz, 1H), 3.26- 3.07 (m, 1H), 3.00-2.76 (m, 1H), 1.12 (dd, J = 14.5, 6.1 Hz, 3H).






(E)-3-(4-bromophenyl)-1-(4-(4-





methoxybenzoyl)-3-methylpiperazin-1-y1)prop-





2-en-1-one







6


embedded image


443.1/ 445.1
8.15 (d, J = 7.7 Hz, 1H), 7.61 (d, J = 8.5 Hz, 2H), 7.51 (d, J = 8.5 Hz, 2H), 7.40 (d, J = 15.8 Hz, 1H), 7.37-7.33 (m, 2H), 7.01-6.97 (m, 2H), 6.63 (d, J = 15.8 Hz, 1H), 4.37-4.03 (m, 1H), 4.00-3.90 (m, 1H), 3.88-3.64 (m, 4H), 3.21-2.97 (m, 2H), 1.88- 1.78 (m, 2H), 1.43-1.31(m, 2H).






(E)-3-(4-bromophenyl)-N-(1-(4-





methoxybenzoyl)piperidin-4-y1)acrylamide







7


embedded image


443.1
7.70 (d, J = 8.5 Hz, 2H), 7.61 (d, J = 8.5 Hz, 2H), 7.54-7.46 (m, 1H), 7.40-7.36 (m, 2H), 7.36-7.21 (m, 1H), 7.02-7.00 (m, 1H), 7.00-6.97 (m, 1H), 4.49-4.10 (m, 3H), 4.06- 3.75 (m, 4H), 3.42 (d, J = 12.3 Hz, 1H), 3.26-3.07 (m, 1H), 3.00-2.76 (m, 1H), 1.12 (dd, J = 14.5, 6.1 Hz, 3H).






(E)-N-(1-(3-(4-





bromophenyl)acryloy1)piperidin-3-y1)-4-





methoxybenzamide







8


embedded image


445.1
8.15 (d, J = 7.6 Hz, 1H), 7.62 (d, J = 8.5 Hz, 2H), 7.51 (d, J = 8.4 Hz, 2H), 7.36 (d, J = 8.5 Hz, 3H), 6.94 (d, J = 7.1 Hz, 2H), 6.67 (d, J = 15.8 Hz, 1H), 3.78 (s, 5H), 3.31-2.89 (m, 2H), 1.95-1.69 (m, 2H), 1.52 (t, J = 8.9 Hz, 2H).






(E)-3-(4-bromophenyl)-N-(1-(4-





methoxybenzoyl)piperidin-3-y1)acrylamide







9


embedded image


415.1
8.89 (d, J = 6.7 Hz, 1H), 7.87 (d, J = 8.8 Hz, 2H), 7.67 (d, J = 8.6 Hz, 2H), 7.60 (d, J = 8.4 Hz, 2H), 7.43 (d, J = 15.7 Hz, 1H), 7.01 (d, J = 8.9 Hz, 2H), 6.81 (d, J = 15.7 Hz, 1H), 4.79- 4.68 (m, 1H), 4.62 (t, J = 8.4 Hz, 1H), 4.29-4.18 (m, 2H), 3.98 (dd, J = 10.5, 5.2 Hz, 1H), 3.81 (s, 3H).






(E)-N-(1-(3-(4-bromophenyl)acryloyl)azetidin-





3-y1)-4-methoxybenzamide







11


embedded image


437.1
7.70 (d, J = 8.5 Hz, 2H), 7.61 (d, J = 8.4 Hz, 2H), 7.48 (d, J = 15.4 Hz, 1H), 7.31 (d, J = 15.4 Hz, 1H), 3.80-3.63 (m, 2H), 3.61-3.43 (m, 6H), 3.23 (s,3H), 3.13-3.02 (m, 1H), 2.59 (td, J = 11.6, 6.2 Hz, 1H), 2.01 (dd, J = 11.9, 2.3 Hz,2H), 1.70 (d, J = 12.3 Hz, 2H), 1.45-1.32 (m, 2H), 1.24-1.10 (m,2H).






(E)-3-(4-bromophenyl)-1-(4-(4-





methoxycyclohexane-1-carbonyl)piperazin-1-





y1)prop-2-en-1-one







12


embedded image


437.1
7.70 (d, J = 8.5 Hz, 2H), 7.61 (d, J = 8.5 Hz, 2H), 7.48 (d, J = 15.4 Hz, 1H), 7.31 (d, J = 15.4 Hz, 1H), 3.77-3.63 (m, 2H), 3.62-3.44 (m, 6H), 3.41-3.37 (m, 1H), 3.25- 3.17 (m, 3H), 2.71-2.61 (m, 1H), 1.93-1.78 (m, 2H), 1.71-1.56 (m, 2H), 1.50-1.34 (m, 4H).






(E)-3-(4-bromophenyl)-1-(4-(4-





methoxycyclohexane-1-carbonyl)piperazin-1-





y1)prop-2-en-1-one







13


embedded image


431.1
8.43 (dd, J = 18.6, 6.5 Hz, 1H), 7.86 (d, J = 8.1 Hz, 2H), 7.73-7.66 (m, 2H), 7.60 (t, J = 8.7 Hz, 2H), 7.46 (dd, J = 15.6, 6.5 Hz, 1H), 7.09-6.96 (m, 3H), 4.62-4.41 (m, 1H), 4.03-3.96 (m, 0.5H), 3.89- 3.83 (m, 0.5H), 3.81 (s, 3H), 3.77-3.67 (m, 1H), 3.64-3.57 (m, 1H), 3.51-3.42 (m, 1H), 2.24-1.95 (m, 2H).






(E)-N-(1-(3-(4-





bromophenyl)acryloyl)pyrrolidin-3-y1)-4-





methoxybenzamide







14


embedded image


403.1/ 405.1
8.41 (s, 1H), 8.27 (s, 1H), 7.85- 7.80 (m, 2H), 7.63-7.58 (m, 2H), 7.54-7.49 (m, 2H), 7.41 (d, J = 15.9 Hz, 1H), 7.01-6.96 (m, 2H), 6.64 (d, J = 15.8 Hz, 1H), 3.80 (s, 3H), 3.38-3.33 (m, 4H).






(E)-N-(2-(3-(4-





bromophenyl)acrylamido )ethy1)-4-





methoxybenzamide







16


embedded image


412.1
11.85 (s, 1H), 7.66 (d, J = 1.7 Hz, 1H), 7.42 (dd, J = 14.7, 8.7 Hz, 3H), 7.19 (dd, J = 8.7, 2.0 Hz, 1H), 7.01 (s, 1H), 6.99 (s, 1H), 6.82 (s, 1H), 4.64-4.30 (m, 2H), 4.24 (d, J = 13.4 Hz, 1H), 4.10- 3.84 (m, 1H), 3.80 (s, 3H), 3.33 (s, 3H), 1.18 (d, J = 6.7 Hz, 3H).






(5-chloro-1H-indol-2-y1)(4-(4-





methoxybenzoyl)-3-methylpiperazin-1-





y1)methanone







17


embedded image


445.1
9.76 (s, 1H), 7.70 (d, J = 8.5 Hz, 2H), 7.60 (d, J = 8.5 Hz, 2H), 7.49 (t, J = 8.0 Hz, 2H), 7.44 (s, 1H), 7.33 (d, J = 15.4 Hz, 1H), 6.86 (d, J = 9.0 Hz, 2H), 4.42 (dd, J = 65.4, 11.9 Hz, 2H), 3.71 (s, 3H), 3.14 (s, 1H), 2.74 (s, 1H), 2.60 (t, J = 11.4 Hz, 1H), 1.85 (d, J = 12.3 Hz, 2H), 1.65-1.40 (m, 2H).






(E)-1-(3-(4-bromophenyl)acryloyl)-N-(4-





methoxyphenyl)piperidine-4-carboxamide







18


embedded image


457.1
8.42-8.30 (m, 1H), 7.82 (t, J = 6.3 Hz, 2H), 7.68 (d, J = 7.9 Hz, 1H), 7.58 (d, J = 8.0 Hz, 1H), 7.51 (s, 2H), 7.44-7.26 (m, 1.5H), 7.16 (d, J = 15.6 Hz, 0.5H), 7.03-6.91 (m, 2H), 4.37 (d, J = 12.0 Hz, 0.5H), 4.25-





3.93 (m, 1.5H), 3.82 (s, 3H),



(E)-N-((1-(3-(4-

3.25-3.11 (m,2H), 3.00-2.85



bromophenyl)acryloyl)piperidin-3-yl)methy1)-

(m, 1H), 2.59-2.51 (m, 1H),



4-methoxybenzamide

1.88-1.58 (m, 3H), 1.41-1.22





(m, 2H).





19


embedded image


431.0
8.47-8.39 (m, 1H), 7.65-7.34 (m, 7H), 6.97 (d, J = 7.2 Hz, 1H), 6.69-6.57 (m, 1H), 4.44- 4.24 (m, 1H), 3.82-3.70 (m, 4H), 3.65-3.48(m, 2H), 3.43- 3.36 (m, 1H), 2.21-2.05 (m, 1H), 1.95-1.77 (m, 1H).






(E)-3-(4-bromophenyl)-N-(1-(4-





methoxybenzoyl)pyrrolidin-3-y1)acrylamide







20


embedded image


403.1
7.66 (d, J = 8.3 Hz, 2H), 7.39 (dd, J = 8.5, 1.9 Hz, 4H), 6.99 (d, J = 8.7 Hz, 2H), 3.79 (s, 3H), 3.74-3.34 (m, 8H).






(4-(4-bromobenzoyl)piperazin-1-y1)(4-





methoxyphenyl)methanone







21


embedded image


465.0/ 467.0
7.69 (d, J = 7.9 Hz, 2H), 7.61 (d, J = 8.5 Hz, 2H), 7.55-7.49 (m, 3H), 7.47 (s, 0.75H), 7.33 (s, 1H), 7.2-7.24(m, 2H), 7.15 (s, 0.26H), 3.86-3.44 (m, 8H).






(E)-3-(4-bromophenyl)-1-(4-(4-





(difluoromethoxy)benzoyl)piperazin-1-y1)prop-





2-en-1-one







22


embedded image


421.1/ 423.1
7.70 (d, J = 8.5 Hz, 2H), 7.61 (d, J = 8.5 Hz, 2H), 7.48 (d, J = 15.4 Hz, 1H), 7.32 (d, J = 15.4 Hz, 1H), 4.28 (d, J = 2.9 Hz, 1H), 3.80 (s, 1H), 3.73- 3.44 (m, 8H), 2.65-2.56 (m, 1H), 1.82-1.71 (m, 2H), 1.70- 1.61 (m, 2H), 1.53-1.43 (m, 2H), 1.39-1.32 (m, 2H).






(E)-3-(4-bromophenyl)-1-(4-(4-





hydroxycyclohexane-1-carbonyl)piperazin-1-





y1)prop-2-en-1-one







27


embedded image


457.1/ 459.1
7.69 (d, J = 8.4 Hz, 2H), 7.61 (d, J = 8.5 Hz, 2H), 7.48 (d, J = 15.2 Hz, 1H), 7.31 (d, J = 15.2 Hz, 1H), 6.93 (dd, J = 10.7, 1.1 Hz, 3H), 4.28 (s, 4H), 3.75 (s, 2H), 3.68- 3.41 (m, 6H).






(E)-3-(4-bromophenyl)-1-(4-(2,3-





dihydrobenzo[b][1,4]dioxine-6-





carbonyl)piperazin-1-y1)prop-2-en-1-one







28


embedded image


454.0/ 456.0
10.57 (s, 1H), 7.73-7.66 (m, 2H), 7.64-7.59 (m, 2H), 7.49 (d, J = 15.4 Hz, 1H), 7.36-7.26 (m, 3H), 6.86 (d, J = 7.8 Hz, 1H), 3.76 (s, 2H), 3.66-3.42 (m, 8H).






(E)-5-(4-(3-(4-





bromophenyl)acryloyl)piperazine-1-





carbonyl)indolin-2-one







29


embedded image


430.0/ 432.0
8.30 (d, J = 2.3 Hz, 1H), 7.75- 7.57 (m, 5H), 7.55-7.44 (m, 2H), 7.39-7.25(m, 1H), 3.89 (s, 3H), 3.82-3.72 (m, 2H), 3.71-3.53 (m, 6H).






(E)-3-(4-bromophenyl)-1-(4-(5-





methoxypicolinoyl)piperazin-1-y1)prop-2-en-1-







30


embedded image


430.0/ 432.0
8.34-8.28 (m, 1H), 7.81 (dd, J = 8.5, 2.4 Hz, 1H), 7.70 (d, J = 8.4 Hz, 2H), 7.65-7.58 (m, 2H), 7.53-7.47 (m, 1H), 7.38- 7.27 (m, 1H), 6.90 (dd, J = 8.5, 0.6 Hz, 1H), 3.90 (s, 3H), 3.85-3.73 (m, 2H), 3.69-3.45 (m, 6H).






(E)-3-(4-bromophenyl)-1-(4-(6-





methoxynicotinoyl)piperazin-1-y1)prop-2-en-1-







31


embedded image


438.0/ 440.0
11.31 (s, 1H), 7.71-7.66 (m, 3H), 7.63-7.59 (m, 2H), 7.49 (d, J = 15.4 Hz, 1H), 7.46-7.42 (m, 2H), 7.32 (d, J = 15.4 Hz, 1H), 7.18 (dd, J = 8.4, 1.5 Hz, 1H), 6.53-6.49 (m, 1H), 3.78 (s, 2H), 3.68-3.50 (m, 6H).






(E)-1-(4-(1H-indole-5-carbonyl)piperazin-1-





y1)-3-(4-bromophenyl)prop-2-en-1-one







32


embedded image


439.1/ 441.1
8.09 (d, J = 2.2 Hz, 1H), 7.77 (d, J = 1.2 Hz, 1H), 7.73-7.65 (m, 3H), 7.63-7.56 (m, 2H), 7.49 (d, J = 15.4 Hz, 1H), 7.43- 7.25 (m, 2H), 7.06-7.00 (m, 1H), 3.91-3.46 (m, 8H).






(E)-1-(4-(benzofuran-5-carbonyl)piperazin-1-





y1)-3-(4-bromophenyl)prop-2-en-1-one







33


embedded image


439.1/ 441.1
12.64 (s, 1H), 8.51-8.24 (m, 2H), 7.72-7.59 (m, 5H), 7.49 (d, J = 15.4 Hz, 1H), 7.36-7.23 (m, 2H), 3.85-3.73 (m, 2H), 3.67-3.48 (m, 6H).






(E)-1-(4-(1H-benzo[d]imidazole-5-





carbonyl)piperazin-1-y1)-3-(4-





bromophenyl)prop-2-en-1-one







34


embedded image


484.0/ 486.0
7.64 (d, J = 15.4 Hz, 1H), 7.55- 7.48 (m, 2H), 7.43-7.36 (m, 4H), 7.00-6.91 (m, 2H), 6.86 (d, J = 15.4 Hz, 1H), 3.93-3.86 (m, 4H), 3.72 (s, 8H), 3.28-3.20 (m, 4H).






(E)-3-(4-bromophenyl)-1-(4-(4-





morpholinobenzoyl)piperazin-1-y1)prop-2-en-1-





one







37


embedded image


416.0/ 418
11.90 (s, 1H), 7.70 (d, J = 8.5 Hz, 2H), 7.65-7.58 (m, 3H), 7.56-7.44 (m, 2H), 7.32 (d, J = 15.4 Hz, 1H), 6.36 (d, J = 9.5 Hz, 1H), 3.77 (s, 2H), 3.62 (s, 2H), 3.55 (s, 4H).






(E)-5-(4-(3-(4-





bromophenyl)acryloyl)piperazine-1-





carbonyl)pyridin-2(1H)-one







38


embedded image


365.1
11.85 (s, 1H), 8.43 - 8.37 (m, 1H), 7.79 (d, J = 8.2 Hz, 1H), 7.44 (d, J = 8.7 Hz, 2H), 7.20 (dd, J = 8.3, 4.5 Hz, 1H), 7.01 (d, J = 8.7 Hz, 2H), 6.96 (s, 1H), 3.93-3.74 (m, 7H), 3.69-3.54 (m, 4H).



(E)-3-(4-bromophenyl)-1-(4-(5-





methoxypyrazine-2-carbonyl)piperazin-1-





y1)prop-2-en-1-one







39


embedded image


365.1
12.14 (s, 1H), 8.32 (dd, J = 4.6, 1.6 Hz, 1H), 8.03 (dd, J = 7.9, 1.1 Hz, 1H), 7.45-7.40 (m, 2H), 7.12 (dd, J = 7.9, 4.6 Hz, 1H), 7.02-6.98 (m, 2H), 6.79 (d, J = 1.7 Hz, 1H), 3.80 (s, 3H), 3.79- 3.72 (m, 4H), 3.61-3.60 (m, 4H)






(4-(1H-pyrrolo[2,3-b]pyridine-2-





carbonyl)piperazin-1-y1)(4-





methoxyphenyl)methanone







40


embedded image


365.1
11.85 (s, 1H), 8.43-8.37 (m, 1H), 7.79 (d, J = 8.2 Hz, 1H), 7.44 (d, J = 8.7 Hz, 2H), 7.20 (dd, J = 8.3, 4.5 Hz, 1H), 7.01 (d, J = 8.7 Hz, 2H), 6.96 (s, 1H), 3.93-3.74 (m, 7H), 3.69-3.54 (m, 4H).






(4-(1H-pyrrolo[3,2-b]pyridine-2-





carbonyl)piperazin-1-y1)(4-





methoxyphenyl)methanone







41


embedded image


388.9/ 390.9
12.53 (s, 1H), 7.75-7.68 (m, 3H), 7.67-7.56 (m, 3H), 7.49 (d, J = 15.4 Hz, 1H), 7.33 (d, J = 15.4 Hz, 1H), 4.52-3.96 (m, 2H), 3.83 - 3.57(m, 6H).






(E)-1-(4-(1H-imidazole-4-carbony1)piperazin-





1-y1)-3-(4-bromophenyl)prop-2-en-1-one







42


embedded image


448.1
11.82 (s, 1H), 7.66 (d, J = 1.4 Hz, 1H), 7.57 (d, J = 1.4 Hz, 1H), 7.51 (d, J = 8.2 Hz, 1H), 7.44 (d, J = 8.7 Hz, 1H), 7.33 (dd, J = 8.2, 1.5 Hz, 1H), 7.20 (dd, J = 8.7, 2.1 Hz, 1H), 6.83 (s, 1H), 3.91-3.46 (m, 8H).






(5-chloro-1H-indol-2-y1)(4-(2,2-





difluorobenzo[d][1,3]dioxole-5-





carbonyl)piperazin-1-y1)methanone







45


embedded image


415.1
11.85 (s, 1H), 8.40 (dd, J = 4.5, 1.2 Hz, 1H), 7.79 (d, J = 8.1 Hz, 1H), 7.57 (d, J = 1.3 Hz, 1H), 7.51 (d, J = 8.2 Hz, 1H), 7.34 (dd, J = 8.3, 1.5 Hz, 1H), 7.21 (dd, J = 8.3, 4.5 Hz, 1H), 6.96 (s, 1H), 3.89-3.47 (m, 8H). 19F NMR (400 MHz, DMSO-d6) δ −48.87/48.89.






(4-(1H-pyrrolo[3,2-b]pyridine-2-





carbonyl)piperazin-1-y1)(2,2-





difluorobenzo[d][1,3]dioxol-5-y1)methanone







46


embedded image


449.1/ 451.1
7.71 (d, J = 8.3 Hz, 2H), 7.61 (d, J = 8.2 Hz, 2H), 7.54-7.44 (m, 1H), 7.38-7.22 (m, 1H), 4.66-3.74 (m, 4H), 3.30-3.13 (m, 4H), 3.13-2.51 (m, 4H), 2.09-1.93 (m, 2H), 1.80-1.59 (m, 2H), 1.50-0.91 (m, 7H).






(E)-3-(4-bromophenyl)-1-(4-(4-





methoxycyclohexane-1-carbonyl)-3-





methylpiperazin-1-y1)prop-2-en-1-one







47


embedded image


449.1/ 451.1
7.71 (d, J = 8.4 Hz, 2H), 7.61 (d, J = 8.4 Hz, 2H), 7.54-7.44 (m, 1H), 7.39-7.21 (m, 1H), 4.64-3.77 (m, 4H), 3.39 (s, 1H), 3.20 (s, 3H), 2.96-2.55 (m, 3H), 1.94-1.78(m, 2H), 1.74-1.20 (m, 7H), 1.19- 0.91 (m, 3H).






(E)-3-(4-bromophenyl)-1-(4-(4-





methoxycyclohexane-1-carbonyl)-3-





methylpiperazin-1-y1)prop-2-en-1-one







48


embedded image


393.1
8.02-7.91 (m, 1H), 7.63-7.54 (m, 1H), 7.53-7.43 (m, 2H), 7.31 (d, J = 15.4 Hz, 1H), 3.78-3.65 (m, 2H), 3.63-3.43 (m, 6H), 3.21 (d, J = 12.4 Hz, 3H), 3.13-3.02 (m, 1H), 2.66- 2.55 (m, 1H), 2.06-1.82 (m, 2H), 1.75-1.60 (m, 2H), 1.44- 1.32 (m, 2H), 1.28-1.11 (m, 2H).






19F NMR (400 MHz, DMSO-d6)




(E)-3-(3,4-difluorophenyl)-1-(4-(4-

δ −136.86/136.92,



methoxycyclohexane-1-carbonyl)piperazin-1-

−138.37/ 138.43.



yl)prop-2-en-1-one







49


embedded image


447.1/ 449.1
7.78-7.53 (m, 4H), 7.52-7.37 (m, 1H), 7.30-7.11 (m, 1H), 3.90-3.48 (m, 7H), 3.44-3.39 (m, 1H), 3.23-3.13(m, 3H), 3.09-2.93((m, 1H), 2.47- 0.88 (m, 11H).






(E)-3-(4-bromophenyl)-1-(4-(4-





methoxycyclohexane-1-carbonyl)-1,4-





diazepan-1-y1)prop-2-en-1-one







50


embedded image


447.1/ 449.1
7.80-7.54 (m, 4H), 7.53-7.35 (m, 1H), 7.32-7.08 (m, 1H), 3.90-3.48 (m, 7H), 3.45-3.35 (m,2H), 3.23-3.11 (m, 3H), 1.89-1.20 (m, 11H).






(E)-3-(4-bromophenyl)-1-(4-(4-





methoxycyclohexane-1-carbonyl)-1,4-





diazepan-1-y1)prop-2-en-1-one







51


embedded image


444.1/ 446.1
8.23-8.07 (m, 1H), 7.81-7.15 (m, 7H), 6.96-6.57 (m, 1H), 3.91-3.37 (m, 11H), 1.87-1.58 (m, 2H)






(E)-3-(4-bromophenyl)-1-(4-(6-





methoxynicotinoy1)-1,4-diazepan-1-y1)prop-2-





en-1-one







52


embedded image


435.1
7.76 (d, J = 7.6 Hz, 2H), 7.60- 7.42 (m, 5H), 7.32 (dd, J = 8.2, 1.3 Hz, 2H), 3.94-3.36 (m, 8H). 19F NMR (400 MHz, DMSO-d6) δ −48.88






(E)-3-(4-chloropheny1)-1-(4-(2,2-





difluorobenzo[d][1,3]dioxole-5-





carbonyl)piperazin-1-y1)prop-2-en-1-one







55


embedded image


437.1
7.82-7.36 (m, 6H), 7.32 (dd, J = 8.2, 1.4 Hz, 1H), 7.29-7.20 (m, 1H), 3.91-3.37 (m, 8H).






(E)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-





carbonyl)piperazin-1-y1)-3-(3,5-





difluorophenyl)prop-2-en-1-one







57


embedded image


402.1
8.65-8.58 (m, 2H), 7.68 (d, J = 4.5 Hz, 2H), 7.57-7.43 (m, 4H), 7.32 (dd, J = 8.2, 1.5 Hz, 1H), 3.84- 3.39 (m, 8H).






(E)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-





carbonyl)piperazin-1-y1)-3-(pyridin-4-y1)prop-





2-en-1-one







58


embedded image


430.0
11.71 (s, 1H), 7.57 (s, 1H), 7.50 (dd, J = 8.1, 2.6 Hz, 1H), 7.42 (dd, J = 8.5, 4.3 Hz, 1H), 7.39-7.29 (m, 2H), 7.09-7.00 (m, 1H), 6.82 (s, 1H), 3.90- 3.48 (m, 8H).






(2,2-difluorobenzo[d][1,3]dioxol-5-y1)(4-(5-





fluoro-1H-indole-2-carbonyl)piperazin-1-





yl)methanone







59


embedded image


414.1
11.61 (s, 1H), 7.60 (d, J = 7.9 Hz, 1H), 7.57 (d, J = 1.0 Hz, 1H), 7.51 (d, J = 8.2 Hz, 1H), 7.43 (d, J = 8.2 Hz, 1H), 7.34 (dd, J = 8.2, 1.1 Hz, 1H), 7.19 (t, J = 7.4 Hz, 1H), 7.05 (t, J = 7.4 Hz, 1H), 6.84 (s, 1H), 3.92-3.43(m, 8H).






19F NMR (400 MHz,




(4-(1H-indole-2-carbonyl)piperazin-1-y1)(2,2-

DMSO-d6) δ −48.88.



difluorobenzo[d][1,3]dioxo1-5-y1)methanone







60


embedded image


432.1
11.75 (s, 1H), 7.82 (d, J = 2.8 Hz, 1H), 7.55 (d, J = 1.4 Hz, 1H), 7.50 (d, J = 8.2 Hz, 1H), 7.47-7.42 (m, 2H), 7.31 (dd, J = 8.2, 1.5 Hz, 1H), 7.01 (td, J = 9.2, 2.5 Hz, 1H), 3.74-3.44 (m, 8H). 19F NMR (400 MHz, DMSO-d6) δ −48.88, −123.15.






(2,2-difluorobenzo[d][1,3]dioxol-5-y1)(4-(5-





fluoro-1H-indole-3-carbonyl)piperazin-1-





y1)methanone







61


embedded image


415.1
12.15 (s, 1H), 8.32 (dd, J = 4.6, 1.5 Hz, 1H), 8.04 (d, J = 7.1 Hz, 1H), 7.57 (d, J = 1.4 Hz, 1H), 7.51 (d, J = 8.2 Hz, 1H), 7.33 (dd, J = 8.3, 1.5 Hz, 1H), 7.12 (dd, J = 7.9, 4.6 Hz, 1H), 6.80 (s, 1H), 3.82- 3.46 (m, 8H).






19F NMR (400 MHz,




(4-(1H-pyrrolo[2,3-b]pyridine-2-

DMSO-d6) δ −48.88 .



carbonyl)piperazin-1-y1)(2,2-





difluorobenzo[d][1,3]dioxo1-5-y1)methanone







63


embedded image


415.1
12.03 (s, 1H), 8.91 (s, 1H), 8.23 (d, J = 5.8 Hz, 1H), 7.57 (d, J = 1.3 Hz, 1H), 7.51 (d, J = 8.2 Hz, 1H), 7.38 (d, J = 5.8 Hz, 1H), 7.34 (dd, J = 8.3, 1.5 Hz, 1H), 7.00 (s, 1H)., 3.95-3.44 (8, 1H).






(4-(1H-pyrrolo[3,2-c]pyridine-2-





carbonyl)piperazin-1-y1)(2,2-





difluorobenzo[d][1,3]dioxo1-5-y1)methanone







64


embedded image


415.1
8.69 (s, 1H), 7.75 (d, J = 8.9 Hz, 1H), 7.55 (d, J = 1.3 Hz, 1H), 7.49 (d, J = 8.2 Hz, 1H), 7.34- 7.26 (m, 2H), 7.00 (t, J = 6.5 Hz, 1H), 6.89 (s, 1H), 3.96-3.55(m, 8H). 19F NMR (400 MHz, DMSO-d6) δ −48.88.






(2,2-difluorobenzo[d][1,3]dioxo1-5-y1)(4-





(pyrazolo[1,5-a]pyridine-2-carbonyl)piperazin-





1-yl)methanone







65


embedded image


393.3
7.54-7.47 (m, 4H), 7.32-7.26 (m, 3H), 3.71-3.36 (m, 8H). 19F NMR (400 MHz, DMSO-d6) δ −48.94, −110.92.






(2,2-difluorobenzo[d][1,3]dioxo1-5-y1)(4-(4-





fluorobenzoyl)piperazin-1-y1)methanone







72


embedded image


407.0/ 409.0
δ 8.05 (dd, J = 17.5, 6.7 Hz, 1H), 7.71-7.65 (m, 2H), 7.63- 7.56 (m, 2H), 7.45 (dd, J = 15.5, 6.9 Hz, 1H), 7.02 (dd, J = 15.5, 11.5 Hz, 1H), 4.36- 4.18 (m, 1H), 3.93-3.67 (m, 4H), 3.60-3.42 (m, 2H), 3.29- 3.24 (m, 2H), 2.40-2.30 (m,





1H), 2.17-1.96 (m, 1H), 1.89-



(E)-N-(1-(3-(4-

1.72 (m, 1H), 1.61-1.52



bromophenyl)acryloyl)pyrrolidin-3-

(m, 4H).



yl)tetrahydro-2H-pyran-4-carboxamide







77


embedded image


399.1
δ 11.82 (s, 1H), 8.32 (d, J = 1.9 Hz, 1H), 7.82 (dd, J = 8.5, 2.4 Hz, 1H), 7.66 (d, J = 1.8 Hz, 1H), 7.44 (d, J = 8.7 Hz, 1H), 7.20 (dd, J = 8.7, 2.1 Hz, 1H), 6.90 (d, J = 8.5 Hz, 1H), 6.83 (s, 1H), 3.90 (s, 3H), 3.86-3.79 (m, 3H), 3.69-3.54 (m, 4H).






N-(1-(5-chloro-1H-indole-2-





carbonyl)pyrrolidin-3-y1)-6-





methoxynicotinamide







80


embedded image


353.1
δ 8.76 (d, J = 5.0 Hz, 2H), 8.31 (d, J = 2.1 Hz, 1H), 7.99 (d, J = 5.0 Hz, 2H), 7.81 (dd, J = 8.5, 2.4 Hz, 1H), 7.67 (d, J = 15.3 Hz, 1H), 7.55 (d, J = 15.4 Hz, 1H), 6.91 (d, J = 8.5 Hz, 1H), 3.91 (s, 3H), 3.71-3.50 (s, 8H).






(E)-1-(4-(6-methoxynicotinoyl)piperazin-1-y1)-





3-(pyridin-4-y1)prop-2-en-1-one







83


embedded image


402.2
δ 8.89 (s, 1H), 8.55 (s, 1H), 8.18 (s, 1H), 7.64-7.25 (m, 6H), 3.96-3.38 (m, 8H). 19F NMR (400 MHz, DMSO-d6) δ −46.88.






(E)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-





carbonyl)piperazin-1-y1)-3-(pyridin-3-y1)prop-





2-en-1-one







90


embedded image


442.1/ 444.1
8.47 (d, J = 2.1 Hz, 1H), 7.94 (dd, J = 8.7, 2.4 Hz, 1H), 7.68-7.64 (m, 2H), 7.63-7.58 (m, 2H), 7.45- 7.38 (m, 1H), 6.89 (d, J = 8.6 Hz, 1H), 6.75 (d, J = 15.7 Hz, 1H), 4.59-4.42 (m, 4H), 4.27-4.09 (m, 4H), 3.91 (s, 3H).






(E)-3-(4-bromophenyl)-1-(6-(6-





methoxynicotinoy1)-2,6-diazaspiro[3.3]heptan-





2-y1)prop-2-en-1-one







91


embedded image


443.1/ 445.1
8.85 (s, 2H), 7.66 (d, J = 8.6 Hz, 2H), 7.60 (d, J = 8.5 Hz, 2H), 7.41 (d, J = 15.7 Hz, 1H), 6.75 (d, J = 15.7 Hz, 1H), 4.57 (s, 2H), 4.51-4.40 (m, 2H), 4.24 (s, 2H), 4.18-4.07 (m, 2H), 3.98 (s, 3H).






(E)-3-(4-bromophenyl)-1-(6-(2-





methoxypyrimidine-5-carbony1)-2,6-





diazaspiro[3.3]heptan-2-y1)prop-2-en-1-one







96


embedded image


444.1/ 446.1
8.28 (d, J = 1.9 Hz, 1H), 7.78 (dd, J = 8.5, 2.4 Hz, 1H), 7.45 (d, J = 8.4 Hz, 2H), 7.16 (d, J = 8.4 Hz, 2H), 6.88 (d, J = 8.5 Hz, 1H), 3.89 (s, 3H), 3.83- 3.44 (m, 8H), 2.38-2.28 (m, 2H), 1.46-1.39 (m, 1H), 1.26-1.19 (m, 1H).






(4-(2-(4-bromophenyl)cyclopropane-1-





carbonyl)piperazin-1-y1)(6-methoxypyridin-3-





y1)methanone







97


embedded image


445.1/ 447.1
8.71 (s, 2H), 7.45 (d, J = 8.3 Hz, 2H), 7.16 (d, J = 8.3 Hz, 2H), 3.97 (s, 3H), 3.79-3.44 (m, 8H), 2.40-2.29 (m, 2H), 1.47-1.40 (m, 1H), 1.27-1.20 (m, 1H).






(4-(2-(4-bromophenyl)cyclopropane-1-





carbonyl)piperazin-1-y1)(2-methoxypyrimidin-





5-y1)methanone







103


embedded image


493.1/ 495.1
7.52 (d, J = 1.5 Hz, 1H), 7.48 (d, J = 8.2 Hz, 1H), 7.45 (d, J = 8.3 Hz, 2H), 7.29 (dd, J = 8.2, 1.6 Hz, 1H), 7.15 (d, J = 8.2 Hz, 2H), 3.88 - 3.36 (m, 8H), 2.40-2.24 (m, 2H), 1.46- 1.39 (m, 1H), 1.25-1.20 (m, 1H). 19H NMR (400 MHz, DMSO-d6) δ −48.88.






(4-(2-(4-bromophenyl)cyclopropane-1-





carbonyl)piperazin-1-y1)(2,2-





difluorobenzo[d][1,3]dioxo1-5-y1)methanone







104


embedded image


432.0/ 434.0
8.28 (d, J = 2.2 Hz, 1H), 7.78 (dd, J = 8.5, 2.3 Hz, 1H), 7.46 (d, J = 8.3 Hz, 2H), 7.22 (d, J = 8.3 Hz, 2H), 6.89 (d, J = 8.5 Hz, 1H), 3.89 (s, 3H), 3.59- 3.35 (m, 8H), 2.80 (t, J = 7.5 Hz, 2H), 2.65 (t, J = 6.8 Hz, 2H).






3-(4-bromopheny1)-1-(4-(6-





methoxynicotinoyl)piperazin-1-y1)propan-1-one







105


embedded image


481.0/ 483.0
7.52 (d, J = 1.2 Hz, 1H), 7.49 (d, J = 8.3 Hz, 1H), 7.45 (d, J = 8.3 Hz, 2H), 7.29 (dd, J = 8.2, 1.3 Hz, 1H), 7.22 (d, J = 8.2 Hz, 2H), 3.66-3.34 (m, 8H), 2.79 (t, J = 7.5 Hz, 2H), 2.69- 2.59 (m, 2H). 19F NMR (400 MHz, DMSO-d6) δ −48.89 .






3-(4-bromopheny1)-1-(4-(2,2-





difluorobenzo[d][1,3]dioxole-5-





carbonyl)piperazin-1-y1)propan-1-one







110


embedded image


449.1
13.35 (d, J = 22.0 Hz, 1H), 7.78 (s, 1H), 7.62-7.45 (m, 3H), 7.39-7.25 (m, 2H), 4.50 (s, 2H), 3.88-3.64 (m, 4H), 3.62-3.45 (m, 2H). 19F NMR (400 MHz, DMSO-d6) δ −48.85.






(5-chloro-1H-benzo[d]imidazol-2-y1)(4-(2,2-





difluorobenzo[d][1,3]dioxole-5-





carbonyl)piperazin-1-y1)methanone







112


embedded image


400.1
13.36 (s, 1H), 8.33 (d, J = 1.9 Hz, 1H), 7.82 (dd, J = 8.5, 2.2 Hz, 1H), 7.76- 7.55 (m, 2H), 7.32 (dd, J = 8.6, 1.0 Hz, 1H), 6.90 (d, J = 8.5 Hz, 1H), 4.51 (s, 2H), 3.91 (s, 3H), 3.79 (s, 2H), 3.74-3.53 (s, 4H).






(5-chloro-1H-benzo[d]imidazol-2-y1)(4-(6-





methoxynicotinoyl)piperazin-1-y1)methanone







114


embedded image


481.0/ 479.0
7.74-7.66 (m, 2H), 7.61 (d, J = 8.0 Hz, 2H), 7.54 (dd, J = 7.3, 1.8 Hz, 1H), 7.49 (d, J = 15.4 Hz, 1H), 7.39-7.26 (m, 3H), 3.84- 3.58 (m, 6H), 3.39 (s, 2H). 19F NMR (400 MHz, DMSO-d6) δ −48.78.






(E)-3-(4-bromophenyl)-1-(4-(2,2-





difluorobenzo[d][1,3]dioxole-4-





carbonyl)piperazin-1-y1)prop-2-en-1-one









Example 2: (E)-3-(4-bromophenyl)-1-(4-(cyclopropanecarbonyl)piperazin-1-yl)prop-2-en-1-one (26)



embedded image


To a mixture of (E)-3-(4-bromophenyl)-1-(piperazin-1-yl)prop-2-en-1-one hydrochloride (Compound 33-4 prepared in Example 1, Step 2) (80 mg, 0.24 mmol), TEA (0.10 mL, 0.72 mmol) in DCM (10 mL) was added cyclopropanecarbonyl chloride (26-2) (0.03 mL, 0.29 mmol), then the reaction was stirred at room temperature for 2 hr. The mixture was diluted with H2O (50 mL) and the resulting mixture was extracted with DCM (25 mL*2). The organic layers were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by silica gel column chromatography (EA/PE=1/10-1/1) to afford the titled compound (55 mg, 0.15 mmol, 62.8% yield). LC-MS (ESI): m/z 363.0/365.0 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 7.74-7.67 (m, 2H), 7.64-7.57 (m, 2H), δ 7.49 (d, J=15.4 Hz, 1H), 7.33 (d, J=15.3 Hz, 1H), 3.83-3.44 (m, 8H), 2.07-1.95 (m, 1H), 0.80-0.66 (m, 4H).


The compounds below were synthesized following the procedures similar to those for Compound 26:

















MS ESI



No.
Structure/Name
[M + H]+

1H NMR (400 MHz, DMSO-d6) δ








68


embedded image


303.1
7.81 (dd, J = 8.7, 5.7 Hz, 2H), 7.52 (d, J = 15.4 Hz, 1H), 7.30-7.20 (m, 3H), 3.78-3.48 (m, 8H), 2.06-1.95 (m, 1H), 0.81-0.69 (m, 4H). 19F NMR (400 MHz, DMSO-d6) δ −111.38.






(E)-1-(4-





(cyclopropanecarbonyl)piperazin-1-y1)-3-





(4-fluorophenyl)prop-2-en-1-one







71


embedded image


332.1
11.76 (s, 1H), 8.40 (t, J = 6.8 Hz, 1H), 7.69 (s, 1H), 7.46 (d, J = 8.7 Hz, 1H), 7.20 (dd, J = 8.7, 1.7 Hz, 1H), 6.95 (d, J = 29.5 Hz, 1H), 4.39-4.27 (m, 1H), 4.12-3.84 (m, 2H), 3.76-3.68 (m, 1H), 3.68-3.60 (m, 1H), 2.23- 2.05 (m, 1H), 1.99-1.80 (m, 1H), 1.59-1.49 (m, 1H), 0.71-0.62 (m, 4H).






N-(1-(5-chloro-1H-indole-2-





carbonyl)pyrrolidin-3-





y1)cyclopropanecarboxamide







74


embedded image


363.0/ 365.0
8.35 (dd, J = 19.4, 6.7 Hz, 1H), 7.69 (dd, J = 8.4, 4.8 Hz, 2H), 7.60 (t, J = 8.0 Hz, 2H), 7.45 (dd, J = 15.5, 7.0 Hz, 1H), 7.03 (dd, J = 15.5, 11.7 Hz, 1H), 4.38-4.21 (m, 1H), 3.87 (dd, J = 10.7, 6.2 Hz, 0.5H), 3.81-3.69 (m, 1H), 3.60-3.52(m, 1H), 3.50-3.44 (m, 1H), 3.29 (d, J = 4.1 Hz, 0.5H), 2.18 - 1.99 (m, 1H), 1.91-1.76 (m, 1H), 1.59-1.50 (m, 1H), 0.73-0.58 (m, 4H).






(E)-N-(1-(3-(4-





bromophenyl)acryloyl)pyrrolidin-3-





y1)cyclopropanecarboxamide









Example 3: (E)-N-(3-(3-(4-bromophenyl)acrylamido)cyclobutyl)-4-methoxybenzamide (10)



embedded image


Step 1: tert-butyl (3-((2,4-dimethoxybenzyl)amino)cyclobutyl)carbamate (10-3)

A mixture of tert-butyl (3-oxocyclobutyl)carbamate (10-1) (1.0 g, 5.39 mmol), (2,4-dimethoxyphenyl) methanamine (10-2) (0.89 mL, 5.93 mmol), HOAc (15 mL), NaBH3CN (0.68 g, 10.7 mmol) and MeOH (20 mL) was degassed with argon for 3 times, then the reaction mixture was stirred at room temperature overnight. The mixture was diluted with DCM (50 mL), washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by silica gel chromatography (PE/EA=100/1 to 2/1) to afford the subtitled compound (980 mg, 2.91 mmol, 54.0% yield) as a white solid. LC-MS (ESI): m/z 337.2 [M+H]+


Step 2: tert-butyl (3-(N-(2,4-dimethoxybenzyl)-4-methoxybenzamido) cyclobutyl)carbamate (10-5)

To a solution of Compound 10-3 (980 mg, 2.91 mmol) in DMF (20 mL) were added DIEA (0.48 mL, 2.91 mmol), HATU (1107 mg, 2.91 mmol) and 4-methoxybenzoic acid (10-4) (0.64 mL, 5.82 mmol). Then the reaction mixture was stirred at room temperature for 3 h. The mixture reaction was diluted with DCM (50 mL), washed with brine (50 mL). The organic layer was separated, filtered and concentrated. The residue was purified by silica gel chromatography (PE/EA=50/1 to 5/1) to afford the subtitled compound (330 mg, 0.70 mmol, 24.1% yield) as a white solid. LC-MS (ESI): m/z 471.3 [M+H]+


Step 3: N-(3-aminocyclobutyl)-4-methoxybenzamide trifluoroacetic salt (10-6)

A solution of Compound 10-5 (270 mg, 0.574 mmol) in DCM (5 mL) and TFA (2 mL, 0.106 mmol) was stirred at room temperature for 2 h. The mixture was concentrated in vacuum to afford the subtitled compound (300 mg, 0.78 mmol, 136.1% yield) as a colorless oil. LC-MS (ESI): m/z 221.2 [M+H]+


Step 4: (E)-N-(3-(3-(4-bromophenyl)acrylamido)cyclobutyl)-4-methoxybenzamide (10)

To a solution of Compound 10-6 (163 mg, 0.718 mmol) in DCM (10 mL) were added DIEA (0.59 mL, 3.59 mmol), HATU (272 mg, 0.718 mmol) and (E)-3-(4-bromophenyl)acrylic acid (10-7) (200 mg, 0.908 mmol). The resulting reaction mixture was stirred at room temperature for 3 h. The solution was mixed with water (20 mL) and the resulting mixture was extracted with DCM (20 mL*2). The organic layers were combined, washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by Prep-HPLC (Waters 2767/2545/2489, Waters Xbridge C18 10 um OBD 19*250 mm, Mobile Phase A: 0.1% FA in water, Mobile Phase B: CH3CN, Flow: 20 mL/min, Column temp: RT) to afford the titled compound (2.2 mg, 0.01 mmol, 0.9% yield). LC-MS (ESI): m/z 429.1/431.1 [M+H]+



1H NMR (400 MHz, DMSO-d6) δ 8.64-8.32 (m,2H), 7.89-7.81 (m, 2H), 7.65-7.59 (m, 2H), 7.55-7.48 (m, 2H), 7.43-7.35 (m, 1H), 7.03-6.95 (m, 2H), 6.69-6.60 (m, 1H), 4.58-3.98 (m, 2H), 3.85-3.79 (m, 3H), 2.64-2.36 (m, 2H), 2.30-1.97 (m, 2H).


Example 4: (E)-3-(4-bromophenyl)-1-(4-(oxetan-3-yl)piperazin-1-yl)prop-2-en-1-one (15)



embedded image


To a solution of 1-(oxetan-3-yl) piperazine (15-1) (383 mg, 1.69 mmol), DIEA (0.70 mL, 4.22 mmol) and HATU (642 mg, 1.69 mmol) in DCM (10 mL) weas added (E)-3-(4-bromophenyl)acrylic acid (15-2) (200 mg, 1.41 mmol). Then the reaction mixture was stirred at room temperature for 2 hr. The mixture was diluted with water (30 mL) and the resulting mixture was extracted with DCM (30 mL*2). The organic layers were combined, washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by Prep-HPLC (Method: Waters 2767/2545/2489/Qame: Inertsil ODS-3 10 um 20*250 nm, Mobile Phase A: 0.1% FA in water, Mobile Phase B: CH3CN, Flow: 20 mL/min: Column temp: RT) to afford the titled compound (200 mg, 0.57 mmol, 40.5% yield). LC-MS (ESI): m/z 351.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 7.69 (d, J=8.5 Hz, 2H), 7.60 (d, J=8.5 Hz, 2H), 7.45 (d, J=15.4 Hz, 1H), 7.30 (d, J=15.4 Hz, 1H), 4.54 (t, J=6.5 Hz, 2H), 4.46 (t, J=6.1 Hz, 2H), 3.72 (s, 2H), 3.59 (s, 2H), 3.42 (dt, J=12.6, 6.3 Hz, 1H), 2.27 (s, 4H)


The compounds below were synthesized following the procedures similar to those for Comnonnd 15:

















MS ESI



NO.
Structure/Name
[M + H]+

1H NMR (400 MHz, DMSO-d6) δ



















81


embedded image


291.2
7.79 (dd, J = 8.4, 5.5 Hz, 2H), 7.49 (d, J = 15.4 Hz, 1H), 7.28-7.19 (m, 3H), 4.55 (t, J = 6.5 Hz, 2H), 4.47 (t, J = 6.1 Hz, 2H), 3.72 (s, 2H), 3.60 (s, 2H), 3.46-3.39 (m, 1H), 2.27 (s, 4H). 19F NMR (400 MHz, DMSO-d6) δ −111.51 .






(E)-3-(4-fluorophenyl)-1-(4-(oxetan-3-y1)piperazin-1-





y1)prop-2-en-1-one







82


embedded image


274.1
8.63-8.57 (dd, J = 4.6, 1.4 Hz, 2H), 7.68 (dd, J = 4.6, 1.4 Hz, 2H), 7.48 (q, J = 15.5 Hz, 2H), 4.55 (t, J = 6.5 Hz, 2H), 4.46 (t, J = 6.1 Hz, 2H), 3.77- 3.69 (m, 2H), 3.64-3.56 (m, 2H), 3.47-3.39 (m, 1H), 2.32-2.24 (m, 4H).






(E)-1-(4-(oxetan-3-y1)piperazin-1-y1)-3-(pyridin-4-





y1)prop-2-en-1-one







84


embedded image


304.1
11.68 (s, 1H), 7.40 (dd, J = 8.9, 4.7 Hz, 1H), 7.35 (dd, J = 9.9, 2.5 Hz, 1H), 7.04 (td, J = 9.3, 2.6 Hz, 1H), 6.76 (d, J = 1.5 Hz, 1H), 4.55 (t, J = 6.5 Hz, 2H), 4.46 (t, J = 6.1 Hz, 2H), 3.77 (s, 4H), 3.51-3.40 (m, 1H), 2.39-2.26 (m, 4H). 19F NMR (400 MHz, DMSO-d6) δ −123.89 .






(5-fluoro-1H-indol-2-y1)(4-(oxetan-3-y1)piperazin-1-





y1)methanone







194


embedded image


379.1/ 381.1
7.71-7.67 (m, 2H), 7.61-7.57 (m, 2H), 7.45 (d, J = 15.4 Hz, 1H), 7.30 (d, J = 15.4 Hz, 1H), 3.92- 3.85 (m, 2H), 3.68 (s, 2H), 3.55 (s, 2H), 3.32- 3.23 (m, 3H), 2.49-2.38(m, 4H), 1.72-1.66 (m, 2H), 1.46-1.36 (m, 2H).






(E)-3-(4-bromophenyl)-1-(4-(tetrahydro-2H-pyran-4-





y1)piperazin-1-y1)prop-2-en-1-one







95


embedded image


379.1/ 381.1
7.69 (d, J = 8.6 Hz, 2H), 7.59 (d, J = 8.5 Hz, 2H), 7.43 (d, J = 15.4 Hz, 1H), 7.31 (d, J = 15.5 Hz, 1H), 4.46 (d, J = 13.1 Hz, 1H), 4.29 (d, J = 13.6 Hz, 1H), 3.56 (t, J = 4.6 Hz, 4H), 3.07 (t, J = 12.3 Hz, 1H), 2.67 (t, J = 12.4 Hz, 1H), 2.48-2.35 (m, 5H), 1.88-1.77 (m, 2H), 1.35-1.21 (m, 2H).






(E)-3-(4-bromophenyl)-1-(4-morpholinopiperidin-1-





y1)prop-2-en-1-one









Example 5: (E)-3-(4-bromophenyl)-1-(4-(piperidine-4-carbonyl)piperazin-1-yl)prop-2-en-1-one (24)



embedded image


A solution of tert-butyl (E)-4-(4-(3-(4-bromophenyl)acryloyl)piperazine-1-carbonyl)piperidine-1-carboxylate (24-1, prepared synthsized following the procedures similar to those for compound 33) (150 mg, 0.30 mmol) in 4M HCl/1,4-dioxane (10 mL) was stirred at room temperature for 2 h. LCMS showed that the reaciton was completed. The mixtured was mixed with water (20 mL) and the resulting mixture was extracted with DCM (20 mL×2). The water was combined, 1M NaOH (5 mL) was used to adjust the water pH to 7˜8, and the water was extracted with DCM (20 mL×2). The organic layers were combined, washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum to afford the titled compound (95 mg, 0.23 mmol, 78.9% yield). LC-MS (ESI): m/z 406.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 7.70 (d, J=8.5 Hz, 2H), 7.61 (d, J=8.5 Hz, 2H), 7.48 (d, J=15.4 Hz, 1H), 7.32 (d, J=15.5 Hz, 1H), 3.77-3.45 (m, 8H), 3.00-2.90 (m, 2H), 2.78-2.64 (m, 1H), 2.58-2.51 (m, 2H), 1.63-1.37 (m, 4H).


Example 6: (E)-1-(4-(4-(azetidin-3-yloxy)benzoyl)piperazin-1-yl)-3-(4-bromophenyl)prop-2-en-1-one (35)



embedded image


Step 1: (E)-3-(4-bromophenyl)-1-(4-(4-hydroxybenzoyl)piperazin-1-yl)prop-2-en-1-one (35-3)

To a solution of 4-hydroxybenzoic acid (35-2) (296 mg, 2.14 mmol), HATU (815 mg, 2.14 mmol) and DIEA (0.89 mL, 5.36 mmol) in DCM (10 mL) was added (E)-3-(4-bromophenyl)-1-(piperazin-1-yl)prop-2-en-1-one hydrochloride salt (35-1, i.e., Compound 33-4 prepared in Example 1, Step 2) (500 mg, 1.79 mmol). Then the reaction mixture was stirred at room temperature for 3 hr. The mixture was mixed with water (60 mL) and the resulting mixture was extracted with EA (50 mL*2). The organic layers were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was slurried with EA (4 mL) and PE (20 mL) to afford the subtitled compound (307 mg, 0.74 mmol, 41.4% yield) as a white solid. LC-MS (ESI): m/z 413.1/415.1 [M+H]+


Step 2: tert-butyl (E)-3-(4-(4-(3-(4-bromophenyl)acryloyl)piperazine-1-carbonyl)phenoxy)azetidine-1-carboxylate (35-5)

To a solution of Compound 35-3 (207 mg, 0.50 mmol) in DMF (8 mL) were added tert-butyl 3-iodoazetidine-1-carboxylate (35-4) (169 mg, 0.60 mmol) and Cs2CO3 (325 mg, 1.00 mmol), and the mixture was heated at 80° C. overnight under N2 atmosphere. The solution was mixed with water (80 mL) and the resulting mixture was extracted with EA (40 mL*2). The organic layers were combined, washed with brine (40 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel colum chromatography (PE/EA=50/1 to 1/1) to afford the subtitled compound (183 mg, 0.32 mmol, 64.5% yield) as a white solid. LC-MS (ESI): m/z 570.1/572.1 [M+H]+


Step 3: (E)-1-(4-(4-(azetidin-3-yloxy)benzoyl)piperazin-1-yl)-3-(4-bromophenyl) prop-2-en-1-one (35)

A solution of tert-butyl (E)-3-(4-(4-(3-(4-bromophenyl)acryloyl)piperazine-1-carbonyl)phenoxy)azetidine-1-carboxylate (35-5) (272 mg, 0.48 mmol) in TFA (2 mL) and DCM (8 mL) was stirred at room temperature for 3 hr. The mixture was mixed with water (20 mL) and the resulting mixture was extracted with DCM (20 mL*2). The water was combined, then the water was adjusted to pH=7˜8, and the water was extracted with DCM (20 mL×2). The organic layers were combined, washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum to afford the titled compound (170 mg, 0.36 mmol, 75.8% yield). LC-MS (ESI): m/z 470.2/472.2 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 7.73-7.66 (m, 2H), 7.63-7.58 (m, 2H), 7.51-7.46 (m, 1H), 7.42-7.37 (m, 2H), 7.31 (d, J=15.8 Hz, 1H), 6.93-6.83 (m, 2H), 5.07-4.97 (m, 1H), 3.85-3.69 (m, 4H), 3.66-3.38 (m. 8H).


Example 7: (E)-3-(4-bromophenyl)-1-(4-(4-((tetrahydrofuran-3-yl)oxy)benzoyl)piperazin-1-yl)prop-2-en-1-one (36)



embedded image


To a solution of Compound 35-3 (50.0 mg, 0.12 mmol) in DMSO (5 mL) were added Cs2CO3 (78.2 mg, 0.24 mmol) and tetrahydrofuran-3-yl methanesulfonate (19.94 mg, 0.12 mmol), and the reaction was stirred at 100° C. for 2 hr. The mixture was purified by Prep-HPLC (Waters 2767/2545/2489, Waters Xbridge C18 10 um OBD 19*250 mm, Mobile Phase A: 0.1% NH4HCO3 in water, Mobile Phase B: CH3CN, Flow: 20 mL/min, Column temp: RT) to afford the titled compound (13.08 mg, 0.03 mmol, 22.4% yield). LC-MS (ESI): m/z 485.1/487.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 7.70 (d, J=8.3 Hz,2H), 7.61 (d, J=8.5 Hz, 2H), 7.49 (d, J=15.4 Hz, 1H), 7.41 (d, J=8.7 Hz, 2H), 7.32 (d, J=15.6 Hz, 1H), 6.99 (d, J=8.7 Hz, 2H), 5.15-4.99 (m, 1H), 3.93-3.80 (m, 3H), 3.80-3.72 (m, 3H), 3.67-3.43 (m, 6H), 2.31-2.20 (m, 1H), 2.02-1.93 (m, 1H).


Example 8: (E)-4-(3-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)-3-oxoprop-1-en-1-yl)benzonitrile (56)



embedded image


Step 1: (E)-3-(4-cyanophenyl)acrylic acid (56-3)

A mixture of 4-formylbenzonitrile (56-1) (500 mg, 3.81 mmol), malonic acid (1190.35 mg, 11.44 mmol) (56-2) and Pyridine (2 mL) was stirred at 100° C. for 3 h. After cooling the mixture was poured into aqueous sulfuric acid (6 mL, 1 M) and the white precipitate filtered and dried to afford the subtitled compound (500 mg, 2.89 mmol, 75.7% yield) as a white solid. LC-MS (ESI): m/z 172.2 [M+H]+


Step 2: (E)-4-(3-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)-3-oxoprop-1-en-1-yl) benzonitrile (56)

To a solution of Compound 56-3 (100 mg, 0.58 mmol) in DMF (5 mL) were added DIEA (0.29 mL, 1.73 mmol), HATU (439 mg, 1.16 mmol) and the reaction was stirred at room temperature for 0.5 h before (2,2-difluorobenzo[d][1,3]dioxol-5-yl)(piperazin-1-yl) methanone hydrochloride (56-4) (176 mg, 0.69 mmol) was added. Then the reaction mixture was stirred at room temperature overnight. The mixture was diluted with H2O (20 mL) and the resulting mixture was extracted with EA (50 mL*2). The organic layers were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel column chromatography (EA/PE=1/40 to 1/1) to afford the titled compound (47.44 mg, 0.11 mmol, 19.3% yield). LC-MS (ESI): m/z 426.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 7.99-7.85 (m, 4H), 7.60-7.41 (m, 4H), 7.32 (dd, J=8.3, 1.5 Hz, 1H), 3.95-3.40 (m, 8H). 19F NMR (400 MHz, DMSO-d6) δ −48.87.


Example 9: (E)-3-(4-bromophenyl)-1-(4-(2-methoxypyrimidine-5-carbonyl)piperazin-1-yl)prop-2-en-1-one (62)



embedded image


Step 1: methyl 2-methoxypyrimidine-5-carboxylate (62-2)

To a solution of methyl 2-chloropyrimidine-5-carboxylate (62-1) (3.00 g, 17.4 mmol) in MeOH (15 mL) were added Sodium methoxide (4.70 g, 86.9 mmol) at rt, and the reaction was stirred at 80° C. for 2 h. The reaction was poured into water (200 mL), then EA (300 mL) was added. The organic layer was separated, dried and concentrated in vacuod to afford the subtitled compound (1.14 g, 6.76 mmol, 38.9% yield) as a yellow solid. LC-MS (ESI): m/z 169.2 [M+H]+


Step 2: 2-methoxypyrimidine-5-carboxylic acid (62-3)

To a solution of Compound 62-2 (100 mg, 0.60 mmol) in MeOH (5 mL) and H2O (5 mL) was added NaOH (35.7 mg, 0.89 mmol), and the reaction mixture was stirred at rt for 2 h, then cooled to rt, adjusted to pH=2˜3 with con. HCl. The mixture was extracted with EA (30 mL×2). The organic layers were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum to afford the subtitled compound (31.0 mg, 0.20 mmol, 33.8% yield) as a white solid. LC-MS (ESI): m/z 155.2 [M+H]+


Step 3: (E)-3-(4-bromophenyl)-1-(4-(2-methoxypyrimidine-5-carbonyl)piperazin-1-yl)prop-2-en-1-one (62)

To a solution of Compound 62-3 (31.0 mg, 0.20 mmol) in DCM (4 mL) were added DIEA (0.03 mL, 0.17 mmol), T3P (79.94 mg, 0.25 mmol, 50% in EA) and the reaction was stirred at room temperature for 0.5 hr before (2E)-3-(4-bromophenyl)-1-(piperazin-1-yl)prop-2-en-1-one hydrochloride (62-4) (55.6 mg, 0.17 mmol) was added. Then the reaction mixture was stirred at room temperature for 2.5 hr. The mixture was diluted with H2O (20 mL) and the resulting mixture was extracted with EA (50 mL*2). The organic layers were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel column chromatography (EA/PE=1/40-1/2) to afford the titled compound (28.0 mg, 0.06 mmol, 38.7% yield). LC-MS (ESI): m/z 431.0/433.0 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 8.74 (s, 2H), 7.70 (d, J=8.2 Hz, 2H), 7.61 (d, J=8.4 Hz, 2H), 7.49 (d, J=15.4 Hz, 1H), 7.33 (d, J=13.6 Hz, 1H), 3.98 (s, 3H), 3.82-3.47 (m, 8H).


The compounds below were synthesized following the procedures similar to those for Compound 62:

















MS ESI



No.
Structure/Name
[M + H]+

1H NMR (400 MHz, DMSO-d6) δ



















52


embedded image


443.8/ 445.8
8.29 (d, J = 1.8 Hz, 1H), 7.79 (dd, J = 8.6, 2.4 Hz, 1H), 7.71 (d, J = 8.4 Hz, 2H), 7.62 (d, J = 8.5 Hz, 2H), 7.55-7.48 (m, 1H), 7.39-7.23 (m, 1H), 6.91 (d, J = 8.4 Hz, 1H), 4.44-4.15 (m, 3H), 3.90 (s, 3H), 3.04-2.83 (m, 2H), 1.19-1.11 (m, 3H).






(E)-3-(4-bromophenyl)-1-(4-(6-





methoxynicotiney1)-3-methylpiperazin-1-





yl)prop-2-en-1-one







54


embedded image


456.0/ 458.0
8.33 (d, J = 2.0 Hz, 1H), 7.82 (dd, J = 8.4, 1.8 Hz, 1H), 7.72-7.67 (m, 2H), 7.64-7.59 (m, 2H), 7.50 (d, J = 15.4 Hz, 1H), 7.35 (d, J = 15.4 Hz, 0.5H), 7.21 (d, J = 15.4 Hz, 0.5H), 6.91-6.85 (m, 1H), 3.91 (s, 3H), 3.87-3.57 (m, 6H), 0.88-0.65 (m, 4H).






(E)-3-(4-bromophenyl)-1-(4-(6-





methoxynicotinoy1)-4,7-diazaspiro[2.5]octan-7-





y1)prop-2-en-1-one







66


embedded image


444.1/ 446.1
δ 8.29 (d, J = 1.8 Hz, 1H), 7.79 (dd, J = 8.5, 2.4 Hz, 1H), 7.58 (d, J = 8.6 Hz, 2H), 7.53 (d, J = 8.5 Hz, 2H), 6.89 (d, J = 8.6 Hz, 1H), 6.52 (s, 1H), 3.89 (d, J = 3.6 Hz, 3H), 3.85-3.48 (m, 8H), 2.17 (d, J = 1.0 Hz, 3H).






(E)-3-(4-bromophenyl)-1-(4-(6-





methoxynicotinoyl)piperazin-1-y1)but-2-en-1-





one







73


embedded image


430.0/ 432.0
δ 8.67 (t, J = 1.8 Hz, 1H), 8.59 (dd, J = 20.4, 6.6 Hz, 1H), 8.14 (ddd, J = 8.7, 2.4, 0.8 Hz, 1H), 7.69 (dd, J = 8.6, 2.8 Hz, 2H), 7.60 (t, J = 8.4 Hz, 2H), 7.46 (dd, J = 15.5, 6.8 Hz, 1H), 7.05 (dd, J = 15.5, 10.8 Hz, 1H), 6.89 (dd, J = 8.7, 3.1 Hz, 1H), 4.61-4.43 (m, 1H), 4.04-3.97





(m, 0.5H), 3.90 (s, 3H), 3.88-3.82



(E)-N-(1-(3-(4-

(m, 0.5H), 3.81-3.66 (m, 1H), 3.66-



bromophenyl)acryloy1)pyrrolidin-3-y1)-6-

3.57 (m, 1H), 3.54-3.43(m, 1H),



methoxynicotinamide

2.29-2.08 (m, 1H), 2.07-1.91 (m, 1H).





76


embedded image


444.0/ 446.0
δ 9.94 (s, 1H), 8.36 (d, J = 1.9 Hz, 1H), 7.91 (dd, J = 8.8, 2.3 Hz, 1H), 7.70 (d, J = 8.3 Hz, 2H), 7.60 (d, J = 8.3 Hz, 2H), 7.46 (d, J = 15.4 Hz, 1H), 7.33 (d, J = 15.4 Hz, 1H), 6.78 (d, J = 8.8 Hz, 1H), 4.60-4.27 (m, 2H), 3.81 (s, 3H), 3.16 (t, J = 12.9 Hz, 1H), 2.82-2.57 (m, 2H), 1.88 (d, J = 11.6 Hz, 2H), 1.65- 1.45 (m, 2H).






(E)-1-(3-(4-bromophenyl)acryloyl)-N-(6-





methoxypyridin-3-y1)piperidine-4-carboxamide







87


embedded image


475.2
8.73 (s, 2H), 7.70 (d, J = 8.3 Hz, 2H), 7.61 (d, J = 8.4 Hz, 2H), 7.49 (d, J = 15.4 Hz, 1H), 7.33 (d, J = 14.4 Hz, 1H), 4.51-4.46 (m, 2H), 3.79 (s, 2H), 3.72- 3.46 (m, 8H), 3.31 (s, 3H).






(E)-3-(4-bromophenyl)-1-(4-(2-(2-





methoxyethoxy )pyrimidine-5-





carbonyl)piperazin-1-yl)prop-2-en-1-one







109


embedded image


446.1
8.66 (d, J = 2.2 Hz, 1H), 8.30 (d, J = 7.7 Hz, 1H), 8.12 (dd, J = 8.7, 2.5 Hz, 1H), 7.71 (d, J = 8.5 Hz, 2H), 7.60 (d, J = 8.5 Hz, 2H), 7.47 (d, J = 15.4 Hz, 1H), 7.36 (d, J = 15.4 Hz, 1H), 6.88 (d, J = 8.7 Hz, 1H), 4.44 (d, J = 12.5 Hz, 1H), 4.30 (d, J = 13.6 Hz, 1H), 4.12-4.03 (m, 1H), 3.90 (s, 3H), 3.24 (t, J = 12.6 Hz, 1H), 2.85 (t, J = 11.5 Hz, 1H),





1.94-1.84 (m, 2H), 1.52-1.40 (m, 2H)



(E)-N-(1-(3-(4-





bromophenyl)acryloyl)piperidin-4-y1)-6-





methoxynicotinamide







116


embedded image


445.1
8.72 (s, 2H), 7.70 (d, J = 8.3 Hz, 2H), 7.61 (d, J = 8.4 Hz, 2H), 7.49 (d, J = 15.4 Hz, 1H), 7.33 (d, J = 15.7 Hz, 1H), 4.42 (q, J = 7.0 Hz, 2H), 3.83-3.50 (m, 8H), 1.36 (t, J = 7.0 Hz, 3H).






(E)-3-(4-bromophenyl)-1-(4-(2-





ethoxypyrimidine-5-carbonyl)piperazin-1-





y1)prop-2-en-1-one









Example 10: (E)-3-(4-bromophenyl)-1-(4-(6-methoxynicotinoyl)piperazin-1-yl)-2-methylprop-2-en-1-one (67)



embedded image


Step 1: ethyl (E)-3-(4-bromophenyl)-2-methylacrylate (67-2)

To a solution of ethyl 2-(diethoxyphosphoryl) propanoate (0.70 mL, 3.24 mmol) in THF (10 mL) was added NaH (130 mg, 3.24 mmol) at 0° C. under N2 atmosphere. The resulting mixture was stirred at this temperature for 0.5 h before 4-bromobenzaldehyde (67-1) (500 mg, 2.70 mmol) was added. The mixture was stirred at RT overnight under N2 atmosphere. The mixture was diluted with H2O (20 mL) and the resulting mixture was extracted with EA (50 mL*2). The organic layers were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum to afford the subtitled compound (700 mg, 2.03 mmol, 75.1% yield) as a yellow oil. LC-MS (ESI): m/z 269.1/271.1 [M+H]+


Step 2: (E)-3-(4-bromophenyl)-2-methylacrylic acid (67-3)

To a solution of Compound 67-2 (700 mg, 2.60 mmol) in MeOH/H2O (1/1, 12 mL) was added NaOH (208 mg, 5.20 mmol) and the reaction mixture was stirred at 40° C. overnight. Then cooled to rt, the mixture was adjusted to pH=2˜3 with con HCl. The mixture was extracted with EA (50 mL*2). The organic layers were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum to afford the subtitled compound (600 mg, 2.49 mmol, 95.7% yield) as a white solid. LC-MS (ESI): m/z 239.0/241.0 [M−H]


Step 3, Step 4 were Synthesized Following the Procedures Similar to Those for Compound 33
Step 5: (E)-3-(4-bromophenyl)-1-(4-(6-methoxynicotinoyl)piperazin-1-yl)-2-methylprop-2-en-1-one (67)

To a solution of 6-methoxypyridine-3-carboxylic acid (67-7) (79.7 mg, 0.52 mmol) in DCM (8 mL) were added DIEA (0.22 mL, 1.30 mmol), TCFH (183 mg, 0.65 mmol) and (2E)-3-(4-bromophenyl)-2-methyl-1-(piperazin-1-yl)prop-2-en-1-one hydro chloride (150 mg, 0.43 mmol). Then the reaction mixture was stirred at room temperature for 2 hr. The mixture was mixed with H2O (50 mL) and the resulting mixture was extracted with DCM (50 mL*2). The organic layers were combined, washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by silica gel column chromatography (EA/PE=1/20-100/1) to afford the titled compound (67) (115 mg, 0.26 mmol, 59.6% yield). LC-MS (ESI): m/z 444.0/446.0 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 8.29 (d, J=2.2 Hz, 1H), 7.79 (dd, J=8.5, 2.4 Hz, 1H), 7.59 (d, J=8.4 Hz, 2H), 7.35 (d, J=8.4 Hz, 2H), 6.89 (d, J=8.5 Hz, 1H), 6.50 (s, 1H), 3.90 (s, 3H), 3.70-3.46 (m, 8H), 2.00 (d, J=1.2 Hz, 3H).


Example 11: (E)-3-(4-bromophenyl)-1-(4-(6-(2-methoxyethoxy)nicotinoyl)piperazin-1-yl) prop-2-en-1-one (69) and (E)-3-(4-bromophenyl)-1-(4-(6-chloronicotinoyl)piperazin-1-yl)prop-2-en-1-one (70)



embedded image


Step 1: 6-(2-methoxyethoxy)pyridine-3-carboxylic acid (69-3) and 6-chloropyridine-3-carboxylic acid (69-4)

To a solution of 2-methoxyethan-1-ol (69-2) (0.19 mL, 2.46 mmol) in THF (8 mL) was added NaH (103 mg, 60% in oil) at 0° C. under N2 atmosphere. The resulting mixture was stirred at this temperature for 0.5 h before 6-chloronicotinic acid (69-1) (400 mg, 1.23 mmol) was added. The mixture was stirred at rt for overnight under N2 atmosphere. The mixture was diluted with H2O (20 mL) and the resulting mixture was extracted with EA (50 mL*2). The organic layers were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum to afford 6-(2-methoxyethoxy)pyridine-3-carboxylic acid (69-3) and 6-chloropyridine-3-carboxylic acid (69-4) (154.18 mg, 0.43 mmol, 35.4% yield) as a white solid. LC-MS (ESI): m/z 198.2/158.2 [M+H]+


Step 2: (E)-3-(4-bromophenyl)-1-(4-(6-(2-methoxyethoxy)nicotinoyl)piperazin-1-yl)prop-2-en-1-one (69) and (E)-3-(4-bromophenyl)-1-(4-(6-chloronicotinoyl)piperazin-1-yl)prop-2-en-1-one (70)

To a solution of 6-(2-methoxyethoxy)pyridine-3-carboxylic acid (69-3) and 6-chloropyridine-3-carboxylic acid (69-4) (257 mg, 0.72 mmol) in DCM (10 mL) were added DIEA (0.30 mL, 1.81 mmol), T3P (576 mg, 0.91 mmol, 50% in EA) and the reaction was stirred at room temperature for 0.5 h before (2E)-3-(4-bromophenyl)-1-(piperazin-1-yl)prop-2-en-1-one hydrochloride (200 mg, 0.603mmol) was added. Then the reaction mixture was stirred at room temperature for 2.5 h. The mixture was diluted with water (30 mL) and the resulting mixture was extracted with DCM (30 mL*2). The organic layers were separated, washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by Prep-HPLC (Method: Waters 2767/2545/2489, Waters Xbridge C18 10 um OBD 19*250 mm, Mobile Phase A: 0.1% NH3H2O in water, Mobile Phase B: CH3CN, Flow: 20 mL/min: Column temp: RT) to afford (E)-3-(4-bromophenyl)-1-(4-(6-(2-methoxyethoxy)nicotinoyl)piperazin-1-yl)prop-2-en-1-one (69) (4.00 mg, 0.01 mmol, 1.4% yield) and (E)-3-(4-bromophenyl)-1-(4-(6-chloronicotinoyl)piperazin-1-yl)prop-2-en-1-one (72) (65 mg, 0.15 mmol, 24.8% yield).


(E)-3-(4-bromophenyl)-1-(4-(6-(2-methoxyethoxy)nicotinoyl)piperazin-1-yl)prop-2-en-1-one (69): LC-MS (ESI): m/z 474.1/476.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 8.28 (d, J=2.2 Hz, 1H), 7.80 (dd, J=8.5, 2.3 Hz, 1H), 7.70 (d, J=8.6 Hz, 2H), 7.61 (d, J=8.5 Hz, 2H), 7.49 (d, J=15.5 Hz, 1H), 7.32 (d, J=14.2 Hz, 1H), 6.91 (d, J=8.5 Hz, 1H), 4.44-4.41 (m, 2H), 3.82-3.74 (m, 2H), 3.69-3.50 (m, 8H), 3.30 (s, 3H).


(E)-3-(4-bromophenyl)-1-(4-(6-chloronicotinoyl)piperazin-1-yl)prop-2-en-1-one (70): LC-MS (ESI): m/z 434.0/436.0 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 8.52 (d, J=2.0 Hz, 1H), 7.96 (dd, J=8.2, 2.3 Hz, 1H), 7.74-7.67 (m, 2H), 7.66-7.59 (m, 3H), 7.49 (d, J=15.4 Hz, 1H), 7.41-7.26 (m, 1H), 3.85-3.59 (m, 6H), 3.49-3.35 (m, 2H).


Example 12: (E)-3-(4-bromophenyl)-1-(3-(oxetan-3-ylamino)pyrrolidin-1-yl)prop-2-en-1-one (75)



embedded image


To a solution of (2E)-1-(3-aminopyrrolidin-1-yl)-3-(4-bromophenyl)prop-2-en-1-one hydrochloride (75-1, prepared following the procedures similar to those of Compound 1) (100 mg, 0.30 mmol) in DCM (8 mL) were added oxetan-3-one (88-2) (26.1 mg, 0.36 mmol), NaBH(CN)3 (32.6 mg, 0.42 mmol) and AcOH (0.13 mL, 0.76 mmol). The mixture was stirred at rt for 18 h. The mixture was diluted with H2O (20 mL) and the resulting mixture was extracted with EA (50 mL*2). The organic layers were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel column chromatography (MeOH/DCM=1/60-1/10) to afford the titled compound (28.0 mg, 0.08 mmol, 26.4% yield). LC-MS (ESI): m/z 351.1/353.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 7.69-7.64 (m, 2H), 7.62-7.58 (m, 2H), 7.43 (d, J=15.5 Hz, 1H), 7.00 (dd, J=15.5, 4.3 Hz, 1H), 4.64 (dd, J=11.8, 5.5 Hz, 2H), 4.36-4.27 (m, 2H), 3.99-3.90 (m, 1H), 3.78-3.70 (m, 1H), 3.65-3.41 (m, 2H), 3.38-3.24 (m, 1H), 3.20-3.11 (m, 1H), 2.88-2.62 (m, 1H), 2.01-1.88 (m, 1H), 1.76-1.57 (m, 1H).


The compound below was synthesized following the procedures similar to those for compound 88

















MS ESI



NO.
Structure/Name
[M + H]+

1H NMR (400 MHz, DMSO-d6) δ








78


embedded image


304.2
11.63 (d, J = 2.4 Hz, 1H), 7.43 (dd, J = 8.9, 4.7 Hz, 1H), 7.41-7.35 (m, 1H), 7.09-7.02 (m, 1H), 6.92 (d, J = 13.1 Hz, 1H), 4.69-4.59 (m, 2H), 4.38-4.27 (m, 2H), 4.01-3.88 (m, 2H), 3.87-3.43 (m, 3H), 3.31- 3.23 (m, 2H), 2.06-1.90 (m, 1H), 1.84-1.64 (m, 1H). 19F NMR (400 MHz, DMSO-d6) δ −123.97/124.01.






(5-fluoro-1H-indol-2-y1)(3-(oxetan-3-





ylamino)pyrrolidin-1-y1)methanone









Example 13: (E)-3-(4-bromophenyl)-1-(4-(6-methoxypyridin-3-yl)piperazin-1-yl)prop-2-en-1-one (98)



embedded image


Step 1: tert-butyl 4-(6-methoxypyridin-3-yl)piperazine-1-carboxylate (98-3)

To a solution of 5-iodo-2-methoxypyridine (550 mg, 2.34 mmol) in toluene (10 mL) were added Xphos (223.13 mg, 0.47 mmol), Sodium tert-butoxide (674.69 mg, 7.02 mmol), Pd2(dba)3 (134.56 mg, 0.23 mmol) and tert-butyl piperazine-1-carboxylate (871.73 mg, 4.68 mmol). Then the reaction mixture was stirred at 110° C. for 2 hr. The mixture was diluted with H2O (100 mL) and the resulting mixture was extracted with EA (100 mL*2). The organic layers were combined, washed with brine (100 mL), dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by silica gel column chromatography (EA/PE=1/100 to 3/1) to afford the subtitled compound (859 mg, 2.93 mmol, 125.1% yield) as a yellow solid. LC-MS (ESI): m/z 294.2 [M+H]+.


Step 2:1-(6-methoxypyridin-3-yl)piperazine trifluoroacetic acid salt

A solution of tert-butyl 4-(6-methoxypyridin-3-yl)piperazine-1-carboxylate (550 mg, 1.88 mmol) in DCM (10 mL) and TFA (10 mL) was stirred at RT for 2 h. The mixture was concentrated under vacuum to afford the subtitled compound (652 mg, 1.55 mmol, 82.5% yield) as a yellow oil. LC-MS (ESI): m/z 194.2 [M+H]+.


Step 3: (E)-3-(4-bromophenyl)-1-(4-(6-methoxypyridin-3-yl)piperazin-1-yl)prop-2-en-1-one (98)

To a solution of 1-(6-methoxypyridin-3-yl) piperazine trifluoroacetic acid salt (50 mg, 0.16 mmol) in DMF (10 mL) were added DIEA (0.13 mL, 0.81 mmol), HATU (74.25 mg, 0.20 mmol), and (2E)-3-(4-bromophenyl)prop-2-enoic acid (44.34 mg, 0.20 mmol). Then the reaction mixture was stirred at room temperature for 2 hr. The mixture was diluted with H2O (100 mL) and the resulting mixture was extracted with EA (100 mL*2). The organic layers were combined, washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by silica gel column chromatography (EA/PE=1/100 to 1/0) to afford the titled compound (27.90 mg, 0.07 mmol, 42.6% yield). LC-MS (ESI): m/z 402.0 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 7.82 (d, J=2.8 Hz, 1H), 7.72 (d, J=8.4 Hz, 2H), 7.61 (d, J=8.4 Hz, 2H), 7.52-7.46 (m, 2H), 7.37 (d, J=15.4 Hz, 1H), 6.74 (d, J=9.0 Hz, 1H), 3.86 (s, 2H), 3.78 (s, 3H), 3.72 (s, 2H), 3.07 (s, 4H).


The compound below was synthesized following the procedures similar to those for Compound 98:

















MS ESI



No.
Structure/Name
[M + H]+

1H NMR (400 MHz, DMSO-d6) δ



















99


embedded image


403.1/ 405.1
8.39 (s, 2H), 7.72 (d, J = 8.5 Hz, 2H), 7.61 (d, J = 8.5 Hz, 2H), 7.49 (d, J = 15.4 Hz, 1H), 7.38 (d, J = 15.4 Hz, 1H), 3.91- 3.81(m, 5H), 3.73 (s, 2H), 3.14 (s, 4H).






(E)-3-(4-bromophenyl)-1-(4-(2-methoxypyrimidin-5-





y1)piperazin-1-y1)prop-2-en-1-one







102


embedded image


402.1/ 404.1
7.83 (d, J = 6.1 Hz, 1H), 7.71 (d, J = 8.5 Hz, 2H), 7.61 (d, J = 8.5 Hz, 2H), 7.49 (d, J = 15.4 Hz, 1H), 7.35 (d, J = 15.4 Hz, 1H), 6.60 (dd, J = 6.1, 2.3 Hz, 1H), 6.16 (d, J = 2.0 Hz, 1H), 3.83 (s, 2H), 3.78 (s, 3H), 3.68 (s, 2H), 3.36 (s, 4H).






(E)-3-(4-bromophenyl)-1-(4-(2-methoxypyridin-4-





y1)piperazin-1-y1)prop-2-en-1-one









Example 14: (E)-3-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-1-(4-(2-methoxypyrimidine-5-carbonyl)piperazin-1-yl)prop-2-en-1-one (92)



embedded image


Step 1: methyl 2-methoxypyrimidine-5-carboxylate (92-7-b)

To a solution of methyl 2-chloropyrimidine-5-carboxylate (92-7-a) (15 g, 86.92 mmol) in MeOH (15 mL) were added sodium methoxide (5.79 mL, 28.97 mmol, 5 mol/L in MeOH) at RT, and the reaction was stirred at 80° C. for 2 h. The reaction was poured into DCM 200 mL, then 600 mL H2O was added. The organic layer was separated, dried and concentrated in vacuum to afford the subtitled compound (11.84 g, 70.40 mmol, 81%) as a white solid. LC-MS (ESI): m/z 169.1 [M+H]+


Step 2: 2-methoxypyrimidine-5-carboxylic acid (92-7)

To a solution of methyl 2-methoxypyrimidine-5-carboxylate (92-7-b) (12.77 g, 75.94 mmol) in MeOH (20 mL) and H2O (20 mL) was added NaOH (4.56 g, 113.92 mmol) and the reaction mixture was stirred at RT for 3 hr, then the mixture was adjusted to pH=2˜3 with con HCl solution. The mixture was extracted with DCM (60 mL×2). The organic layers were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum to afford the subtitled compound (11.00 g, 71.37 mmol, 94%) as a yellow solid. LC-MS (ESI): m/z 153.00 [M−H]−


Step 3: (2E)-3-(2,2-difluoro-2H-1,3-benzodioxol-5-yl)prop-2-enoic acid (92-3)

To a solution of propanedioic acid (307.52 mg, 2.96 mmol) in pyridine (15 mL) were added piperidine (0.3 mL), 2,2-difluoro-2H-1,3-benzodioxole-5-carbaldehyde (500 mg, 2.69 mmol), and the reaction was stirred at room temperature for 3 hr. The reaction was diluted with Water and con HCl (10 mL) was added. The mixture was filtered. The cake was collected, dried under vacuum to afford the subtitled compound (609 mg, 2.67 mmol, 99.4%) as white solid. LC-MS (ESI): m/z 227.0 [M−H].


Step 4: tert-butyl 4-[(2E)-3-(2,2-difluoro-2H-1,3-benzodioxol-5-yl)prop-2-enoyl]piperazine-1-carboxylate (92-5)

To a solution of (2E)-3-(2,2-difluoro-2H-1,3-benzodioxol-5-yl)prop-2-enoic acid (92-3) (300 mg, 1.32 mmol) in DCM (10 mL) were added DIEA (0.74 mL, 4.47 mmol), HATU (749.97 mg, 1.97 mmol) and tert-butyl piperazine-1-carboxylate (304.09 mg, 1.63 mmol). Then the reaction mixture was stirred at RT for 3 hr. The mixture was diluted with H2O (50 mL) and the resulting mixture was extracted with DCM (50 mL*2). The organic layers were combined, washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by silica gel column chromatography (EA/PE=1/20-1/1) to afford the subtitled compound (174 mg, 0.44 mmol, Y=40.1%) as a yellow solid. LC-MS (ESI): m/z 341.1 [M−56+H]+.


Step 5: (2E)-3-(2,2-difluoro-2H-1,3-benzodioxol-5-yl)-1-(piperazin-1-yl)prop-2-en-1-one hydrochloride (92-6)

A solution of tert-butyl 4-[(2E)-3-(2,2-difluoro-2H-1,3-benzodioxol-5-yl)prop-2-enoyl]piperazine-1-carboxylate (92-5) (174 mg, 0.44 mmol) in 4M HCl/dioxane (8 mL) was stirred at room temperature for 2 hr. The mixture was concentrated under vacuum to afford the crude subtitled compound (158 mg, 0.47 mmol, 108.2%) as a white solid. The product was used for next step without further purification. LC-MS (ESI): m/z 297.1 [M+H]+.


Step 6: (2E)-3-(2,2-difluoro-2H-1,3-benzodioxol-5-yl)-1-[4-(2-methoxypyrimidine-5-carbonyl)piperazin-1-yl]prop-2-en-1-one (92)

To a solution of 2-methoxypyrimidine-5-carboxylic acid (92-7) (33.42 mg, 0.22 mmol) in DCM (6 mL) were added DIEA (0.09 mL, 0.54 mmol), TCFH (75.89 mg, 0.27 mmol) and (2E)-3-(2,2-difluoro-2H-1,3-benzodioxol-5-yl)-1-(morpholin-4-yl)prop-2-en-1-one (92-6) (60 mg, 0.18 mmol), and the reaction was stirred at RT for 3 hr. The reaction was diluted with DCM (50 mL) and H2O (50 mL) and the resulting mixture was extracted with DCM (30 mL*2). The organic layers were combined, washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified using silica gel column chromatography eluting with (EA/PE=1/50-3/1) to afford the titled compound (20 mg, 0.05 mmol, 25.6%). LC-MS (ESI): m/z 433.2 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 8.74 (s, 2H), 7.95 (s, 1H), 7.58-7.51 (m, 2H), 7.45 (d, J=8.3 Hz, 1H), 7.31 (d, J=14.5 Hz, 1H), 3.98 (s, 3H), 3.80 (s, 2H), 3.70-3.44 (m, 6H). 19F NMR (400 MHz, DMSO-d6) δ −49.20.


The compound below was synthesized following the procedures similar to those for compound 92:

















MS ESI



No.
Structure/Name
[M + H]+

1H NMR (400 MHz, DMSO-d6) δ



















93


embedded image


432.2
8.31 (s, 1H), 7.94 (s, 1H), 7.83- 7.77 (m, 1H), 7.58-7.49 (m, 2H), 7.45 (dd, J = 8.3, 2.1 Hz, 1H), 7.29 (d, J = 17.9 Hz, 1H), 6.90 (dd, J = 8.5, 1.9 Hz, 1H), 3.91 (d, J = 2.0 Hz, 3H), 3.79 (s, 2H), 3.66-3.51 (m, 6H). 19F NMR (400 MHz, DMSO-d6) δ −49.19.






(E)-3-(2,2-difluorobenzo[d][1,3]dioxo1-5-y1)-1-





(4-(6-methoxynicotinoy1)piperazin-1-y1)prop -2-





en-1-one







117


embedded image


409.2
7.95 (s, 1H), 7.56 (dd, J = 8.4, 1.7 Hz, 1H), 7.52 (d, J = 15.2 Hz, 1H), 7.45 (d, J = 8.3 Hz, 1H), 7.30 (d, J = 15.3 Hz, 1H), 3.89-3.81 (m, 2H), 3.76-3.66 (m, 2H), 3.62-3.46 (m, 6H), 3.43-3.36 (m, 2H), 2.99-2.85 (m, 1H), 1.64-1.50 (m, 4H).






(E)-3-(2,2-difluorobenzo[d][1,3]dioxol-5-y1)-1-





(4-(tetrahydro-2H-pyran-4-carbonyl)piperazin-1-





y1)prop-2-en-1-one







118


embedded image


353.2
7.95 (d, J = 1.4 Hz, 1H), 7.54 (dd, J = 8.4, 1.4 Hz, 1H), 7.49 (d, J = 15.4 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.28 (d, J = 15.3 Hz, 1H), 4.55 (t, J = 6.5 Hz, 2H), 4.46 (t, J = 6.1 Hz, 2H), 3.76-3.68 (m, 2H), 3.63-3.55 (m, 2H), 3.47-3.40 (m, 1H), 2.32- 2.22 (m, 4H)






(E)-3-(2,2-difluorobenzo[d][1,3]dioxol-5-y1)-1-





(4-(oxetan-3-y1)piperazin-1-y1)prop-2-en-1-one







119


embedded image


365.1
7.96 (d, J = 1.0 Hz, 1H), 7.60-7.49 (m, 2H), 7.45 (d, J = 8.3 Hz, 1H), 7.31 (d, J = 15.3 Hz, 1H), 3.83- 3.46 (m, 8H), 2.02 (s, 1H), 0.81- 0.69 (m, 4H).






(E)-1-(4-(cyclopropanecarbonyl)piperazin-1-y1)-





3-(2,2-difluorobenzo[d][1,3]dioxol-5-y1)prop-2-





en-1-one







120


embedded image


381.2
7.62 (d, J = 15.4 Hz, 1H), 7.28- 7.21 (m, 2H), 7.06 (d, J = 8.2 Hz, 1H), 6.79 (d, J = 15.3 Hz, 1H), 4.04 (dd, J = 11.3, 4.4 Hz, 2H), 3.73 (d, J = 34.0 Hz, 4H), 3.38 (td, J = 11.8, 2.0 Hz, 2H), 2.62 (t, J = 5.0 Hz, 4H), 2.58-2.46 (m, 1H), 1.81-1.72 (m, 2H), 1.66- 1.54 (m, 2H).






(E)-3-(2,2-difluorobenzo[d][1,3]dioxo1-5-y1)-1-





(4-(tetrahydro-2H-pyran-4-y1)piperazin-1-





y1)prop-2-en-1-one







121


embedded image


381.1
7.60 (d, J = 15.3 Hz, 1H), 7.29- 7.20 (m, 2H), 7.06 (d, J = 8.2 Hz, 1H), 6.81 (d, J = 15.4 Hz, 1H), 4.71 (d, J = 12.7 Hz, 1H), 4.13 (d, J = 13.1 Hz, 1H), 3.73 (t, J = 4.6 Hz, 4H), 3.24-3.07 (m, 1H), 2.83-2.67 (m, 1H), 2.58 (t, J = 4.7 Hz, 4H), 2.55-2.43 (m, 1H), 1.99-1.89 (m, 2H), 1.57-1.44 (m, 2H).






(E)-3-(2,2-difluorobenzo[d][1,3]dioxo1-5-y1)-1-





(4-morpholinopiperidin-1-y1)prop-2-en-1-one







122


embedded image










(E)-3-(2,2-difluorobenzo[d][1,3]dioxo1-5-y1)-1-





(4-(2-(2-methoxyethoxy)pyrimidine-5-





carbonyl)piperazin-1-y1)prop-2-en-1-one









Example 15: (E)-3-(4-bromophenyl)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl) prop-2-en-1-one (25)



embedded image


To a solution of (E)-3-(4-bromophenyl)-1-(piperazin-1-yl)prop-2-en-1-one hydrochloride (Compound 33-4 prepared in Example 1, Step 2) (200 mg, 0.714 mmol) in ACN (10 mL) were added DIEA (0.24 mL, 1.428 mmol), TCFH (239 mg, 0.857 mmol) and 2,2-difluorobenzo[d][1,3]dioxole-5-carboxylic acid (25-1) (173 mg, 0.857 mmol). The resulting reaction mixture was stirred at room temperature for 2 h. The solution was mixed with water (20 mL) and the resulting mixture was extracted with DCM (20 mL*2). The organic layers were combined, washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by Prep-HPLC (Waters 2767/2545/2489, Waters Xbridge C18 10 um OBD 19*250 mm, Mobile Phase A: 0.1% FA in water, Mobile Phase B: CH3CN, Flow: 20 mL/min, Column temp: RT) to afford the titled compound (10.67 mg, 0.02 mmol, 3.1% yield). LC-MS (ESI): m/z 479.0/481.0 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 7.74-7.53 (m, 5H), 7.52-7.46 (m, 2H), 7.35-7.28 (m, 2H), 3.80-3.35 (m, 8H).


The compound below was synthesized following the procedures similar to those for compound 25:

















MS ESI



No.
Structure/Name
[M + H]+

1H NMR (400 MHz, DMSO-d6) δ








43


embedded image


437.0
8.04-.787 (m, 1H), 7.63-7.43 (m, 5H), 7.32 (dd, J = 8.2, 1.4 Hz, 2H), 3.88-3.36 (m, 8H). 19F NMR (400 MHz, DMSO-d6) δ −48.86, −136.82/136.88, 138.34/138.43.






(E)-1-(4-(2,2-difluorobenzo[d] [1,3]dioxole-5-





carbonyl)piperazin-1-y1)-3-(3,4-





difluorophenyl)prop-2-en-1-one







44


embedded image


418.9
7.86-7.74 (m, 2H), 7.59-7.46 (m, 3H), 7.32 (dd, J = 8.2, 1.5 Hz, 1H), 7.30-7.18 (m, 3H), 3.94-3.38 (m, 8H). 19F NMR (400 MHz, DMSO-d6) δ −48.89, 111.33/111.34






(E)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-





carbonyl)piperazin-1-y1)-3-(4-





fluorophenyl)prop-2-en-1-one







79


embedded image


403.2
9.17 (s, 2H), 9.15 (s, 1H), 7.57-7.47 (m, 4H), 7.32 (dd, J = 8.2, 1.2 Hz, 1H), 3.90-3.46 (m, 8H). 19F NMR (400 MHz, DMSO-d6) δ −48.87.






(E)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-





carbonyl)piperazin-1-y1)-3-(pyrimidin-5-





yl)prop-2-en-1-one









Example 16: (E)-3-(4-bromophenyl)-1-(4-(tetrahydro-2H-pyran-4-carbonyl)piperazin-1-yl)prop-2-en-1-one (23)



embedded image


To a solution of tetrahydro-2H-pyran-4-carboxylic acid (23-2) (83.6 mg, 0.642 mmol) in DCM (10 mL) were added DIEA (0.27 mL, 1.60 mmol), I-3-(4-bromophenyl)-1-(piperazin-1-yl)prop-2-en-1-one hydrochloride (23-1, i.e., Compound 33-4 prepared in Example 1, Step 2) (150 mg, 0.535 mmol) and HATU (244 mg, 0.642 mmol), and the reaction was stirred at room temperature for 2 h. The reaction was diluted with DCM (20 mL) and saturated NaCl solution (40 mL). The organic layer was separated and concentrated in vacuum. The residue was purified by silica gel column (EA/PE=1/50 to 1/1) to afford the titled compound (10 mg, 0.02 mmol, 4.6% yield). LC-MS (ESI): m/z 407.1/409.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ 7.70 (d, J=8.5 Hz, 2H), 7.61 (d, J=8.4 Hz, 2H), 7.48 (d, J=15.4 Hz, 1H), 7.32 (d, J=15.4 Hz, 1H), 3.91-3.80 (m, 2H), 3.79-3.65 (m, 2H), 3.64-3.45 (m, 6H), 3.44-3.35 (m, 2H), 2.98-2.84 (m, 1H), 1.68-1.49 (m, 4H).


The Chemotaxis Biological Function Assay of GPR183 Antagonists

A chemotaxis assay is conducted to analyze whether or not a cell type directly orients and migrates towards a defined chemoattractant. Since 7α,25-OHC-GPR183 axis play an important role in mediating immune cells migration in autoimmune diseases, the function of GPR183 antagonists were evaluated using cell-based chemotaxis assay in vitro.


The U937 cell line was used to monitor the inhibitory effect of antagonists on 7α,25-OHC-mediated chemotaxis. All the compounds are investigated based on the following assay methods.


U937 cells were passaged in complete medium (RPMI1640, 10% FBS, 1% penicillin-streptomycin) in incubator (37° C., 5% CO2).


Prior to the migration assay, U937 cells were kept for 2 h in lipid-depleted media (RPMI 1640, 1% lipid-free FBS, 1% penicillin-streptomycin).


Chemotaxis was performed using HTS Transwell-96 plates with 5.0 μm pore polycarbonate membranes according to the manufacturer's protocol. In brief, lower chambers were filled with 100 μL of RPMI1640 media with 0.5% BSA containing 20 nM 7α,25-OHC. After insertion of the filters, 1*105 cells which were pre-treated with compounds at different concentrations in 75 μL RPMI1640 media with 0.5% BSA were added to the upper chamber. After 3 h at 37° C., cell numbers in the lower chamber were analyzed by flow cytometry. Chemotaxis is expressed as the total number of cells in the lower chambers. The cell numbers of each well were plotted against various antagonists concentrations and analyzed in GraphPad Prism for concentration curve generation.


The Ca2+ Mobilization Assay of GPR183 Antagonists

Calcium mobilization assay is a cell-based second messenger assay to measure the calcium flux associated with G-protein coupled receptor activation or inhibition. The change in the fluorescence intensity is directly correlated to the amount of intracellular calcium that is released into cytoplasm in response to ligand activation of the receptor of interest.


The GPR183-Gqi5-CHO K1 cells (constructed by Genomeditech) passaged in complete medium (F12K medium, 10% FBS, 1% penicillin-streptomycin, 4 μg/ml puromycin) in the incubator (37° C., 5% CO2) were used in the Ca2+ mobilization assay.


The fluorescent membrane permeable calcium binding dye (the FLIPR Calcium 6 Assay Kit) was dissolved in assay buffer (20 mM HEPES buffer+1*Hank's Balanced Salt Solution (HBSS), pH 7.4). The loading buffer was prepared with the dye solution containing 5 mM probenecid (The probenecid stock solution was prepared with 500 mM solution in 1 N NaOH, which then diluted to 250 mM in HBSS buffer).


Approximately 1.5*104 GPR183-Gqi5-CHO K1 cells were seeded into a 384-well plate with a 25 μL starving medium (1% lipid-free FBS, 1% penicillin-streptomycin) the day before the assay. On the assay day, the starving medium was completely changed with 25 μL assay buffer, then 25 μL loading buffer was added into the desired wells. After adding dye, the cell plate was incubated for 2 hours at 37° C. with 5% CO2 and then kept at room temperature until used. The compounds in 12.5 μL assay buffer at desired concentration (5*) were added to each well and incubated with cells for 30 min at room temperature. After incubation, transfer the microplates to the FLIPR instrument and start the calcium assay as described in the user guide for the instrument. 12.5 μL assay buffer with or without 7α,25-OHC was added during the assay. The MAX ratio values of each well were plotted against various antagonists' concentrations and analyzed in GraphPad Prism for concentration curve generation.


The results of Ca2+ mobilization IC50 and Chemtaxis IC50 of the compounds disclosed hercin are listed in the following table














Compound No.
Ca2+ mobilization IC50 (nM)
Chemtaxis IC50 (nM)


Reference*
38.52
1.236

















1
Not available
Not available


2
Not available
Not available


3
1800
Not available


4
24.92
9.507


5
26.22
6.764


6
Not available
Not available


7
Not available
Not available


8
Not available
Not available


9
Not available
Not available


10
Not available
Not available


11
Not available
9.33


12
Not available
Not available


13
3341
Not available


14
Not available
Not available


15
39.97
6.74


16
Not available
Not available


17
92.64
Not active


18
378.7
Not available


19
Not available
Not available


20
Not available
Not available


21
406.8
Not available


22
548.4
Not available


23
39.45
10.46


24
1020
Not available


25
3.97
0.98


26
24.89
2.226


27
17.49
Not available


28
295.2
Not available


29
106.7
51.2


30
30.11
1.62


31
66.03
3.99


32
16.10
0.83


33
213.5
Not available


34
Not available
Not available


35
Not available
Not available


36
Not available
Not available


37
45.79
68.66


38
51.38
Not available


39
Not active
Not available


40
Not active
Not available


41
Not available
Not available


42
158.1
343.5


43
901.4
Not available


44
21.84
Not available


45
Not available
Not available


46
169.9
Not available


47
700.4
Not available


48
239.8
Not available


49
Not available
Not available


50
Not available
Not available


51
65.17
277.2


52
Not available
Not available


53
91.54
7.308


54
497.7
Not active


55
123.1
Not active


56
179.7
Not active


57
2590
Not active


58
435.6
Not active


59
232.3
Not available


60
Not active
Not available


61
Not active
Not available


62
38.57
Not available


63
Not active
Not active


64
Not active
Not available


65
Not active
Not available


66
564.5
Not available


67
Not active
Not active


68
69.55
Not active


69
64.85
1.239


70
34.67
1.26


71
Not active
Not available


72
824.3
71.01


73
Not active
Not available


74
420.5
52.85


75
70.4
Not active


76
297.4
Not active


77
240.2
687.4


78
Not active
Not active


79
Not active
Not active


80
2040
Not active


81
4800
Not active


82
Not active
Not active


83
Not active
Not active


84
Not active
Not active


85
Not available
Not available


86
Not available
Not available


87
15.91
Not available


88
Not available
Not available


89
Not available
Not available


90
105.5
Not available


91
35.04
Not available


92
31.64
Not available


93
18.7
Not available


94
21.5
Not available


95
28.8
Not available


96
Not active
Not available


97
Not active
Not available


98
106.6
Not available


99
152
Not available


100
Not available
Not available


101
Not available
Not available


102
1940
Not available


103
Not active
Not available


104
179.9
Not available


105
128.8
Not available


106
Not available
Not available


107
Not available
Not available


108
Not available
Not available


109
865
Not available


110
Not active
Not available


111
Not available
Not available


112
Not active
Not available


113
Not available
Not available


114
163.6
Not available


115
23.71
4.642


116
32.5
Not available


117
562
Not available


118
645.6
Not available


119
36.4
Not available


120
57.6
Not available


121
260.9
Not available





*Reference indicates a compound having a structure of




embedded image








Pharmacokinetics Study

Compounds disclosed herein were further studied with respect to CYP inhibition, hERG inhibition, kinetic solubility, permeability, PPB, LMS, and other pharmacokinetics.


Results of the representative compounds and the reference compound




















hERG
Kinetic





Compound

IC50
Solubility
mPPB
hPPB



No.
CYP IC50 (μM)
(μM)
(μM)
(% fu)
(% fu)
hLMS





















Reference
1A2 > 10, 2B6 > 10
1.648
7
6.4

> 186.4



2C8 > 10, 2C9 > 10








2C19:0.99, 2D6 > 10








3A4-M > 10, 3A4-T > 10







15
1A2 > 10, 2B6 > 10
> 30
225.5
17.8
15.2
> 186.4



2C8 > 10, 2C9 > 10








2C19 > 10, 2D6 > 10








3A4-M > 10, 3A4-T > 10







62
1A2 > 10, 2B6 > 10
4.98
108
20.0
11.1
> 186.4



2C8 > 10, 2C9 > 10








2C19: 6.60, 2D6 > 10








3A4-M > 10, 3A4-T > 10







23
1A2 > 10, 2B6 > 10
18.32
68
23.7
18.4
> 186.4



2C8 > 10, 2C9 > 10








2C19 > 10 2D6 > 10








3A4-M > 10 3A4-T > 10







44
1A2 > 10, 2B6 > 10
5.51
45.8
3.6
2.2
> 186.4



2C8 > 10, 2C9 > 10








2C19 > 10, 2D6 > 10








3A4-M > 10, 3A4-T > 10







25
1A2 > 10, 2B6: 19.04
2.587
34.9
1.2
0.8
> 186.4



2C8 > 10, 2C9 > 18.54








2C19: 6.62, 2D6:3.45








3A4-M:14.62, 3A4-








T:7.64







53
1A2 > 10, 2B6 > 10
2.746
22.1
1.3
0.9
> 186.4



2C8 > 10, 2C9 > 10








2C19 > 5.07, 2D6 > 4.45








3A4-M > 10, 3A4-T > 10







92
A2 > 10, 2B6 > 10
7.9
114
22.0
21.6
> 186.4



2C8 > 10, 2C9 > 10








2C19 > 10, 2D6 > 10








3A4-M > 10, 3A4-T > 10









CYP inhibition: compared with the reference compound Reference (2C19: 0.99 μM), representative compounds disclosed herein, e.g., Compound 15, 23, 44, 92 showed no or very weak CYP inhibition. In conclusion, the compounds disclosed herein show little or no CYP inhibition compared with the reference compound, suggesting little or no drug-drug interaction.


hERG inhibition: compared with the reference compound (1.648 μM, representative compounds disclosed herein, e.g., Compound 15 and 23 showed no or weak inhibition, and representative compounds disclosed herein, e.g., Compound 62, 23, 44, 25, 53, 92 showed poor or weak inhibition compared with the strong inhibtion of the reference compound.


Solubility and permeability: compared with the reference compound (7 μM), representative compounds disclosed herein were unexpectedly found to have improved solubility. These representative compounds showed high permeability.


PPB on mouse and human: representative compounds disclosed herein, e.g., Compound 15, Compound 62, Compound 23 and Compound 92 showed improved plasma protein binding, indicating the improved concentration of the free active compound that actually works in the body.


Stability of human liver microsomes: Compounds disclosed herein were also tested with respect to the stability of human liver microsomes. The results showed that all the tested compounds are greater than 186.4 min, showing good stability.


Compounds disclosed herein were tested with respect to pharmacokinetics in mice as shown in the table below:


















Parameters
CL
T1/2
Cmax
AUC(0-t)
Vss
F (%)







Unit
mL/min/kg
h
ng/mL
h*ng/ml
L/kg



Reference IV 1 mg/kg
16.86
0.87
866
972
1.32



Reference PO 50 mg/kg

3.72
8,931
45,663

119


Compound 15 IV 1 mg/kg
32.56
0.69
934.25
513.00
1.53



Compound 15 PO 10 mg/kg

1.25
3,026.21
6,819.57

133.21


Compound 62 IV 1 mg/kg

2.18
920.44
1,877
1.43



Compound 62 PO 10 mg/kg

3.74
4900
27,242




Compound 23 IV 1 mg/kg
10.17
1.91
965
1,575
1.50



Compound 23 PO 10 mg/kg

4.58
5,108
28,042

224.50


Compound 44 IV 1 mg/kg
13.41
2.18
517.84
1,156
2.43



Compound 44 PO 10 mg/kg

3.30
3205
13948

119


Compound 25 IV 1 mg/kg
3.86
3.57
474.31
1,341
3.43



Compound 25 PO 10 mpk

2.80
1,197
13,184




Compound 53 IV 1 mg/kg
17.38
2.31
437.00
875.34
2.31



Compound 53 PO 10 mg/kg

3.39
2,348
16,384

186.87


Compound 92 IV 1 mg/kg
3.98
2.97
832
4,186
1.13



Compound 92 PO 10 mg/kg

2.46
5,895
4,9654

118.94









Half-life: compared with the reference compound (3.72 h), compounds disclosed herein showed better or comparable half-life.


Cmax and AUC: compounds disclosed herein, e.g., Compound 15, Compound 62, Compound 23, Compound 44, Compound 25, and Compound 53 showed better pharmacokinetics such as Cmax (ng/ml) and AUClast (h*ng/mL) if converted into the same dose (PO 50 mpk), indicating that the compounds disclosed herein show much better in vivo exposure.


Vss: compared with the reference compound, compounds disclosed herein, e.g., Compound 15, Compound 62, Compound 23, Compound 44, Compound 25, Compound 53 showed improved Vss, indicating better tissue distribution.


Compounds disclosed herein were tested with respect to pharmacokinetics in rats as shown in the table below:


















Parameters
CL
T1/2
Cmax
AUC(0-t)
Vss
F (%)







Unit
mL/min/kg
h
ng/ml
h*ng/ml
L/kg



Reference IV 1 mg/kg
14.14
0.78
1470
1156
0.82



Reference PO10 mg/kg

0.65
4,273
12,756

110.3


Compound 25 IV 1 mg/kg
3.86
5.87
873
4,188
1.72



Compound 25 PO 10 mg/kg

1.92
5,208
45,906

109.56


Compound 92 IV 1 mg/kg
3.40
1.56
2,111
4,840
0.46



Compound 92 PO 10 mg/kg

2.60
17,394
87,585

201.11









Clearance and half-life: Compared with the reference compound, Compounds 25 and 92 showed a much-improved clearance rate and half-life.


Cmax and AUC: Compounds 25 and 92 showed much better pharmacokinetics such as Cmax (ng/ml) and AUClast (h*ng/ml) in the same dose.


Vss: compared with the reference compound, Compound 25 showed an improved Vss.


It is to be understood that, if any prior art publication is referred to herein; such reference does not constitute an admission that the publication forms a part of the common general knowledge in the art in any country.


The disclosures of all publications, patents, patent applications, and published patent applications referred to herein by an identifying citation are hereby incorporated herein by reference in their entirety.


Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it is apparent to those skilled in the art that certain minor changes and modifications will be practiced. Therefore, the description and examples should not be construed as limiting the scope of the invention.

Claims
  • 1-30. (canceled)
  • 31. A compound of formula (III)
  • 32. The compound of claim 31, wherein L1 is C2-6alkenylene.
  • 33. The compound of claim 31, wherein L1 is —CH═CH—.
  • 34. The compound of claim 31, wherein X is N and Y is N; and m is 1 and n is 1.
  • 35. The compound of claim 31, wherein t is 0.
  • 36. The compound of claim 31, wherein L2 is —C(O)—.
  • 37. The compound of claim 31, wherein ring B is phenyl, monocyclic 5- to 9-membered heteroaryl, bicyclic 7- to 12-membered heteroaryl, monocyclic 5- to 9-membered heterocyclyl, bicyclic 7- to 12-membered heterocyclyl, monocyclic 3- to 8-membered cycloalkyl, or bicyclic 7- to 12-membered cycloalkyl, each of which is unsubstituted or substituted with one or two substituents selected from halogen, oxo, C1-6alkyl, haloC1-6alkyl, or —ORf, wherein Rf is hydrogen, C1-6alkyl, haloC1-6alkyl, C1-6alkoxy-C1-6alkyl-, or monocyclic 5- to 9-membered heterocyclyl.
  • 38. The compound of claim 37, wherein (a) ring B is phenyl which is substituted with —ORf, wherein Rf is hydrogen, C1-6alkyl, haloC1-6alkyl, or monocyclic 5- to 9-membered heterocyclyl;(b) ring B is monocyclic 3- to 8-membered cycloalkyl, which is unsubstituted or substituted with one or two substituents selected from halogen, C1-6alkyl, haloC1-6alkyl, or —ORf, wherein Rf is hydrogen or C1-6alkyl;(c) ring B is monocyclic 5- to 9-membered heteroaryl, which is unsubstituted or substituted with one or two substituents selected from oxo, halogen, or —ORf, wherein Rf is hydrogen, C1-6alkyl or C1-6alkoxy-C1-6alkyl-;(d) ring B is monocyclic 5- to 9-membered heterocyclyl or bicyclic 7- to 12-membered heterocyclyl, each of which is unsubstituted or substituted with oxo, halogen, or C1-6alkyl; or(e) ring B is bicyclic 7- to 12-membered heteroaryl, which is unsubstituted or substituted with one or two substituents selected from oxo, halogen, or C1-6alkyl.
  • 39. The compound of claim 7, wherein (a) ring B is phenyl which is substituted with methoxy, difluoromethoxy, azetidin-3-yloxy, or (tetrahydrofuran-3-yl)oxy;(b) ring B is cyclopropyl or cyclohexyl, which is unsubstituted or substituted with hydroxy or methoxy;(c) ring B is pyridin-3-yl, pyridin-2-yl, pyridin-4-yl, pyridin-5-yl, pyrimidin-5-yl, pyrimidin-4-yl, or pyrazine-2-yl, which is unsubstituted or substituted with oxo, methoxy, or 2-methoxyethoxy;(d) ring B is piperidinyl, tetrahydropyranyl, oxetanyl, morpholino, benzodioxolyl, or dihydrobenzofuranyl;(e) ring B is piperidine-4-yl, tetrahydro-2H-pyran-4-yl, oxetan-3-yl, morpholino, 2,2-difluorobenzo[d][1,3]dioxol-4-yl, 2,2-difluorobenzo[d][1,3]dioxol-5-yl or 2,3-dihydrobenzofuran-5-yl; or(f) ring B is 1H-imidazol-4-yl, 1H-indol-5-yl, benzofuran-5-yl, or 1H-benzo[d]imidazol-5-yl, each of which is unsubstituted or substituted with oxo.
  • 40. The compound of claim 37, wherein the moiety
  • 41. The compound of claim 37, wherein the moiety
  • 42. The compound of claim 37, wherein the moiety
  • 43. A compound, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, which is selected from 3-(3,4-difluorophenyl)-1-(4-(4-methoxybenzoyl)piperazin-1-yl)propan-1-one (1);(E)-3-(3,4-difluorophenyl)-1-(4-(4-methoxybenzoyl) piperazin-1-yl)prop-2-en-1-one (2);(E)-3-(4-bromophenyl)-1-(6-(4-methoxybenzoyl)-2,6-diazaspiro[3.3]heptan-2-yl)prop-2-en-1-one (3);(E)-3-(4-bromophenyl)-1-(4-(4-methoxybenzoyl)-1,4-diazepan-1-yl)prop-2-en-1-one (4);(E)-3-(4-bromophenyl)-1-(4-(4-methoxybenzoyl)-3-methylpiperazin-1-yl)prop-2-en-1-one (5);(E)-3-(4-bromophenyl)-N-(1-(4-methoxybenzoyl) piperidin-4-yl)acrylamide (6);(E)-N-(1-(3-(4-bromophenyl)acryloyl)piperidin-3-yl)-4-methoxybenzamide (7);(E)-3-(4-bromophenyl)-N-(1-(4-methoxybenzoyl)piperidin-3-yl)acrylamide (8);(E)-N-(1-(3-(4-bromophenyl)acryloyl)azetidin-3-yl)-4-methoxybenzamide (9);(E)-N-(3-(3-(4-bromophenyl)acrylamido)cyclobutyl)-4-methoxybenzamide (10);(E)-3-(4-bromophenyl)-1-(4-(4-methoxy-(cisortrans)-cyclohexane-1-carbonyl) piperazin-1-yl)prop-2-en-1-one (11);(E)-3-(4-bromophenyl)-1-(4-(4-methoxy-(transorcis)-cyclohexane-1-carbonyl) piperazin-1-yl)prop-2-en-1-one (12);(E)-N-(1-(3-(4-bromophenyl)acryloyl)pyrrolidin-3-yl)-4-methoxybenzamide (13);(E)-N-(2-(3-(4-bromophenyl)acrylamido)ethyl)-4-methoxybenzamide (14);(E)-3-(4-bromophenyl)-1-(4-(oxctan-3-yl)piperazin-1-yl)prop-2-en-1-one (15);(5-chloro-1H-indol-2-yl)(4-(4-methoxybenzoyl)-3-methylpiperazin-1-yl) methanone (16);(E)-1-(3-(4-bromophenyl)acryloyl)-N-(4-methoxyphenyl)piperidine-4-carboxamide (17);(E)-N-((1-(3-(4-bromophenyl)acryloyl)piperidin-3-yl)methyl)-4-methoxybenzamide (18);(E)-3-(4-bromophenyl)-N-(1-(4-methoxybenzoyl)pyrrolidin-3-yl)acrylamide (19);(4-(4-bromobenzoyl)piperazin-1-yl)(4-methoxyphenyl)methanone (20);(E)-3-(4-bromophenyl)-1-(4-(4-(difluoromethoxy)benzoyl)piperazin-1-yl)prop-2-en-1-one (21);(E)-3-(4-bromophenyl)-1-(4-(4-hydroxycyclohexane-1-carbonyl)piperazin-1-yl)prop-2-en-1-one (22);(E)-3-(4-bromophenyl)-1-(4-(tetrahydro-2H-pyran-4-carbonyl)piperazin-1-yl)prop-2-en-1-one (23);(E)-3-(4-bromophenyl)-1-(4-(piperidine-4-carbonyl)piperazin-1-yl)prop-2-en-1-one (24);(E)-3-(4-bromophenyl)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)prop-2-en-1-one (25);(E)-3-(4-bromophenyl)-1-(4-(cyclopropanecarbonyl)piperazin-1-yl)prop-2-en-1-one (26);(E)-3-(4-bromophenyl)-1-(4-(2,3-dihydrobenzo[b][1,4]dioxine-6-carbonyl)piperazin-1-yl)prop-2-en-1-one (27);(E)-5-(4-(3-(4-bromophenyl)acryloyl)piperazine-1-carbonyl)indolin-2-one (28);(E)-3-(4-bromophenyl)-1-(4-(5-methoxypicolinoyl)piperazin-1-yl)prop-2-en-1-one (29);(E)-3-(4-bromophenyl)-1-(4-(6-methoxynicotinoyl)piperazin-1-yl)prop-2-en-1-one (30);(E)-1-(4-(1H-indole-5-carbonyl)piperazin-1-yl)-3-(4-bromophenyl)prop-2-en-1-one (31);(E)-1-(4-(benzofuran-5-carbonyl)piperazin-1-yl)-3-(4-bromophenyl)prop-2-en-1-one (32);(E)-1-(4-(1H-benzo[d]imidazole-5-carbonyl)piperazin-1-yl)-3-(4-bromophenyl)prop-2-en-1-one (33);(E)-3-(4-bromophenyl)-1-(4-(4-morpholinobenzoyl)piperazin-1-yl)prop-2-en-1-one (34);(E)-1-(4-(4-(azetidin-3-yloxy)benzoyl)piperazin-1-yl)-3-(4-bromophenyl)prop-2-en-1-one (35);(E)-3-(4-bromophenyl)-1-(4-(4-((tetrahydrofuran-3-yl)oxy)benzoyl)piperazin-1-yl)prop-2-en-1-one (36);(E)-5-(4-(3-(4-bromophenyl)acryloyl)piperazine-1-carbonyl)pyridin-2 (1H)-one (37);(E)-3-(4-bromophenyl)-1-(4-(5-methoxypyrazine-2-carbonyl)piperazin-1-yl)prop-2-en-1-one (38);(4-(1H-pyrrolo[2,3-b]pyridine-2-carbonyl)piperazin-1-yl)(4-methoxyphenyl)methanone (39);(4-(1H-pyrrolo[3,2-b]pyridinc-2-carbonyl)piperazin-1-yl)(4-methoxyphenyl)methanone (40);(E)-1-(4-(1H-imidazole-4-carbonyl)piperazin-1-yl)-3-(4-bromophenyl)prop-2-en-1-one (41);(5-chloro-1H-indol-2-yl)(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)methanone (42);(E)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)-3-(3,4-difluorophenyl) prop-2-en-1-one (43);(E)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)-3-(4-fluorophenyl)prop-2-en-1-one (44);(4-(1H-pyrrolo[3,2-b]pyridine-2-carbonyl)piperazin-1-yl)(2,2-difluorobenzo[d][1,3]dioxol-5-yl)methanone (45);(E)-3-(4-bromophenyl)-1-(4-(4-methoxy-(cisortrans)-cyclohexane-1-carbonyl)-3-methylpiperazin1-yl)prop-2-en-1-one (46);(E)-3-(4-bromophenyl)-1-(4-(4-methoxy-(transorcis)-cyclohexane-1-carbonyl)-3-methylpiperazin-1-yl)prop-2-en-1-one (47);(E)-3-(3,4-difluorophenyl)-1-(4-(4-methoxycyclohexane-1-carbonyl)piperazin-1-yl)prop-2-en-1-one (48);(E)-3-(4-bromophenyl)-1-(4-(4-methoxycyclohexane-1-carbonyl)-1,4-diazepan-1-yl)prop-2-en-1-one (49);(E)-3-(4-bromophenyl)-1-(4-(4-methoxycyclohexane-1-carbonyl)-1,4-diazepan-1-yl)prop-2-en-1-one (50);(E)-3-(4-bromophenyl)-1-(4-(6-methoxynicotinoyl)-1,4-diazepan-1-yl) prop-2-en-1-one (51);(E)-3-(4-bromophenyl)-1-(4-(6-methoxynicotinoyl)-3-methylpiperazin-1-yl)prop-2-en-1-one (52);(E)-3-(4-bromophenyl)-1-(4-(6-methoxynicotinoyl)-3-methylpiperazin-1-yl)prop-2-en-1-one (53);(E)-3-(4-bromophenyl)-1-(4-(6-methoxynicotinoyl)-4,7-diazaspiro[2.5]octan-7-yl)prop-2-en-1-one (54);(E)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)-3-(3,5-difluorophenyl)prop-2-en-1-one (55);(E)-4-(3-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)-3-oxoprop-1-en-1-yl)benzonitrile (56);(E)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)-3-(pyridin-4-yl)prop-2-en-1-one (57);(2,2-difluorobenzo[d][1,3]dioxol-5-yl)(4-(5-fluoro-1H-indole-2-carbonyl)piperazin-1-yl)methanone (58);(4-(1H-indole-2-carbonyl)piperazin-1-yl)(2,2-difluorobenzo[d][1,3]dioxol-5-yl)methanone (59);(2,2-difluorobenzo[d][1,3]dioxol-5-yl)(4-(5-fluoro-1H-indole-3-carbonyl)piperazin-1-yl)methanone (60);(4-(1H-pyrrolo[2,3-b]pyridine-2-carbonyl)piperazin-1-yl)(2,2-difluorobenzo[d][1,3]dioxol-5-yl)methanone (61);(E)-3-(4-bromophenyl)-1-(4-(2-methoxypyrimidine-5-carbonyl)piperazin-1-yl)prop-2-en-1-one (62);(4-(1H-pyrrolo[3,2-c]pyridine-2-carbonyl)piperazin-1-yl)(2,2-difluorobenzo[d][1,3]dioxol-5-yl)methanone (63);(2,2-difluorobenzo[d][1,3]dioxol-5-yl)(4-(pyrazolo[1,5-a]pyridine-2-carbonyl)piperazin-1-yl)methanone (64);(2,2-difluorobenzo[d][1,3]dioxol-5-yl)(4-(4-fluorobenzoyl)piperazin-1-yl)methanone (65);(E)-3-(4-bromophenyl)-1-(4-(6-methoxynicotinoyl)piperazin-1-yl)but-2-en-1-one (66);(E)-3-(4-bromophenyl)-1-(4-(6-methoxynicotinoyl)piperazin-1-yl)-2-methylprop-2-en-1-one (67);(E)-1-(4-(cyclopropanecarbonyl)piperazin-1-yl)-3-(4-fluorophenyl)prop-2-en-1-one (68);(E)-3-(4-bromophenyl)-1-(4-(6-(2-methoxyethoxy)nicotinoyl)piperazin-1-yl)prop-2-en-1-one (69);(E)-3-(4-bromophenyl)-1-(4-(6-chloronicotinoyl) piperazin-1-yl)prop-2-en-1-one (70);N-(1-(5-chloro-1H-indole-2-carbonyl)pyrrolidin-3-yl)cyclopropanecarboxamide (71);(E)-N-(1-(3-(4-bromophenyl)acryloyl)pyrrolidin-3-yl)tetrahydro-2H-pyran-4-carboxamide (72);(E)-N-(1-(3-(4-bromophenyl)acryloyl)pyrrolidin-3-yl)-6-methoxynicotinamide (73);(E)-N-(1-(3-(4-bromophenyl)acryloyl)pyrrolidin-3-yl)cyclopropanecarboxamide (74);(E)-3-(4-bromophenyl)-1-(3-(oxctan-3-ylamino)pyrrolidin-1-yl)prop-2-en-1-one (75);(E)-1-(3-(4-bromophenyl) acryloyl)-N-(6-methoxypyridin-3-yl) piperidine-4-carboxamide (76);N-(1-(5-chloro-1H-indole-2-carbonyl)pyrrolidin-3-yl)-6-methoxynicotinamide (77);(5-fluoro-1H-indol-2-yl)(3-(oxetan-3-ylamino)pyrrolidin-1-yl)methanone (78);(E)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)-3-(pyrimidin-5-yl)prop-2-en-1-one (79);(E)-1-(4-(6-methoxynicotinoyl)piperazin-1-yl)-3-(pyridin-4-yl)prop-2-en-1-one (80);(E)-3-(4-fluorophenyl)-1-(4-(oxctan-3-yl)piperazin-1-yl)prop-2-en-1-one (81);(E)-1-(4-(oxctan-3-yl)piperazin-1-yl)-3-(pyridin-4-yl)prop-2-en-1-one (82);(E)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)-3-(pyridin-3-yl)prop-2-en-1-one (83);(5-fluoro-1H-indol-2-yl)(4-(oxctan-3-yl)piperazin-1-yl)methanone (84);(E)-N-(1-(3-(4-fluorophenyl)acryloyl)pyrrolidin-3-yl)-6-oxo-1,6-dihydropyridine-3-carboxamide (85);(E)-3-(4-chlorophenyl)-1-(4-(6-(2-methoxyethoxy)nicotinoyl)piperazin-1-yl)prop-2-en-1-one (86);(E)-3-(4-bromophenyl)-1-(4-(2-(2-methoxyethoxy)pyrimidine-5-carbonyl)piperazin-1-yl)prop-2-en-1-one (87);(E)-3-(2,3-dihydrobenzofuran-6-yl)-1-(4-(6-methoxynicotinoyl)piperazin-1-yl)prop-2-en-1-one (88);(E)-3-(1H-indol-6-yl)-1-(4-(6-methoxynicotinoyl)piperazin-1-yl)prop-2-en-1-one (89);(E)-3-(4-bromophenyl)-1-(6-(6-methoxynicotinoyl)-2,6-diazaspiro[3.3]heptan-2-yl)prop-2-en-1-one (90);(E)-3-(4-bromophenyl)-1-(6-(2-methoxypyrimidine-5-carbonyl)-2,6-diazaspiro[3.3]heptan-2-yl)prop-2-en-1-one (91);(E)-3-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-1-(4-(2-methoxypyrimidine-5-carbonyl)piperazin-1-yl)prop-2-en-1-one (92);(E)-3-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-1-(4-(6-methoxynicotinoyl)piperazin-1-yl)prop-2-en-1-one (93);(E)-3-(4-bromophenyl)-1-(4-(tetrahydro-2H-pyran-4-yl)piperazin-1-yl)prop-2-en-1-one (94);(E)-3-(4-bromophenyl)-1-(4-morpholinopiperidin-1-yl)prop-2-en-1-one (95);(4-(2-(4-bromophenyl)cyclopropane-1-carbonyl)piperazin-1-yl)(6-methoxypyridin-3-yl)methanone (96);(4-(2-(4-bromophenyl)cyclopropane-1-carbonyl)piperazin-1-yl)(2-methoxypyrimidin-5-yl)methanone (97);(E)-3-(4-bromophenyl)-1-(4-(6-methoxypyridin-3-yl)piperazin-1-yl)prop-2-en-1-one (98);(E)-3-(4-bromophenyl)-1-(4-(2-methoxypyrimidin-5-yl)piperazin-1-yl)prop-2-en-1-one (99);(E)-3-(4-bromophenyl)-1-(4-(5-methoxypyridin-2-yl)piperazin-1-yl)prop-2-en-1-one (100);(E)-3-(4-bromophenyl)-1-(4-(2-methoxypyrimidin-4-yl)piperazin-1-yl)prop-2-en-1-one (101);(E)-3-(4-bromophenyl)-1-(4-(2-methoxypyridin-4-yl)piperazin-1-yl)prop-2-en-1-onc (102);(4-(2-(4-bromophenyl)cyclopropane-1-carbonyl)piperazin-1-yl)(2,2-difluorobenzo[d][1,3]dioxol-5-yl) methanone (103);3-(4-bromophenyl)-1-(4-(6-methoxynicotinoyl)piperazin-1-yl)propan-1-one (104);3-(4-bromophenyl)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl)propan-1-one (105);(5-bromo-2,3-dihydro-1H-inden-2-yl)(4-(6-methoxynicotinoyl)piperazin-1-yl)methanone (106);(6-bromo-1H-inden-2-yl)(4-(6-methoxynicotinoyl)piperazin-1-yl)methanone (107);(E)-3-(4-bromophenyl)-1-(1-(6-methoxynicotinoyl)-1,6-diazaspiro[2.5]octan-6-yl)prop-2-en-1-one (108);(E)-N-(1-(3-(4-bromophenyl)acryloyl)piperidin-4-yl)-6-methoxynicotinamide (109);(5-chloro-1H-benzo[d]imidazol-2-yl)(4-(2,2-difluorobenzo[d][1,3]dioxole-5-carbonyl)piperazin-1-yl) methanone (110);(E)-5-(4-(3-(4-bromophenyl)acryloyl)piperazine-1-carbonyl)-1,3-dihydro-2H-benzo[d]imidazol-2-one (111);(5-chloro-1H-benzo[d]imidazol-2-yl)(4-(6-methoxynicotinoyl)piperazin-1-yl)methanone (112);(E)-3-(4-bromophenyl)-1-(4-(2,2-difluoro-[1,3]dioxolo[4,5-c]pyridine-6-carbonyl)piperazin-1-yl)prop-2-en-1-one (113);(E)-3-(4-bromophenyl)-1-(4-(2,2-difluorobenzo[d][1,3]dioxole-4-carbonyl)piperazin-1-yl)prop-2-en-1-one (114);(E)-3-(4-bromophenyl)-1-(4-(6-(tetrahydrofuran-3-yloxy)nicotinoyl)piperazin-1-yl)prop-2-en-1-one (115);(E)-3-(4-bromophenyl)-1-(4-(2-ethoxypyrimidine-5-carbonyl)piperazin-1-yl)prop-2-en-1-one (116);(E)-3-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-1-(4-(tetrahydro-2H-pyran-4-carbonyl)piperazin-1-yl)prop-2-en-1-one (117);(E)-3-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-1-(4-(oxetan-3-yl)piperazin-1-yl)prop-2-en-1-one (118);(E)-1-(4-(cyclopropanecarbonyl)piperazin-1-yl)-3-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)prop-2-en-1-one (119);(E)-3-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-1-(4-(tetrahydro-2H-pyran-4-yl)piperazin-1-yl)prop-2-en-1-one (120);(E)-3-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-1-(4-morpholinopiperidin-1-yl)prop-2-en-1-one (121); or(E)-3-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)-1-(4-(2-(2-methoxyethoxy)pyrimidine-5-carbonyl)piperazin-1-yl)prop-2-en-1-one (122).
  • 44. A pharmaceutical composition comprising a compound of claim 31 or a stereoisomer hereof or a pharmaceutically acceptable salt thereof, optionally together with a pharmaceutically acceptable excipient.
  • 45. A method of treating a disease mediated by GPR183, comprising administering a subject in need thereof a compound of claim 31 or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof.
  • 46. The method of claim 45, wherein the disease mediated by GPR183 is cancer, autoimmune diseases, liver diseases, osteoporosis, and neuropathic pain.
  • 47. The method of claim 46, wherein the cancer is blood, brain, breast, colorectal, gastrointestinal, liver, lung, ovarian, pancreatic, prostate, skin, or uterine cancer.
  • 48. The method of claim 47, wherein the cancer produces molecules involved in Epstein-Barr virus (EBV)-induced G-protein coupled receptor 2 (EBI2) mediated signaling.
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2022/125632 10/17/2022 WO
Continuations (2)
Number Date Country
Parent PCT/CN2022/114934 Aug 2022 WO
Child 18702236 US
Parent PCT/CN2021/124341 Oct 2021 WO
Child PCT/CN2022/114934 US