CN117980290A - Cyanopyridine and cyanopyrimidine BCL6 degrading agents - Google Patents

Cyanopyridine and cyanopyrimidine BCL6 degrading agents Download PDF

Info

Publication number
CN117980290A
CN117980290A CN202280061397.8A CN202280061397A CN117980290A CN 117980290 A CN117980290 A CN 117980290A CN 202280061397 A CN202280061397 A CN 202280061397A CN 117980290 A CN117980290 A CN 117980290A
Authority
CN
China
Prior art keywords
alkyl
compound
amino
methyl
hydroxy
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280061397.8A
Other languages
Chinese (zh)
Inventor
L·H·琼斯
车健为
黄皇
尼基·孔若曦
S·费劳
刘颖鹏
J·克鲁特
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Dana Farber Cancer Institute Inc
Original Assignee
Dana Farber Cancer Institute Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Dana Farber Cancer Institute Inc filed Critical Dana Farber Cancer Institute Inc
Priority claimed from PCT/US2022/074387 external-priority patent/WO2023015164A1/en
Publication of CN117980290A publication Critical patent/CN117980290A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Compounds, compositions, and methods for treating diseases or disorders characterized by aberrant B-cell lymphoma 6 (BCL 6) activity are described.

Description

Cyanopyridine and cyanopyrimidine BCL6 degrading agents
RELATED APPLICATIONS
The present application claims priority to U.S. patent application Ser. No. 63/228,303, filed on 8/2/2021, and U.S. patent application Ser. No. 63/352,063, filed on 14/6/2022, each of which are hereby incorporated by reference in their entireties, in accordance with 35 U.S. C. ≡119 (e).
Background
As demonstrated by the clinical efficacy of thalidomide analogs in the treatment of hematological malignancies, small molecule-induced protein degradation has become a powerful therapeutic strategy. Thalidomide analogs (including lenalidomide and pomalidomide) regulate the activity of the novel gene (RING) ligase 4-Cereblon (CRBN) (CRL 4 CRBN) E3 ubiquitin ligase of real interest to Cullin to recruit and ubiquitinate novel substrates including zinc finger 1 (IKZF 1), IKZF3 and casein kinase 1 a (CK 1 a) of the Ikaros family, which results in proteasome degradation (Kronke et al.,Science 343:301-305(2014);Lu et al.,Science 343:305-309(2014);Kronke et al.,Nature 523:183-188(2015)). other small molecules that induce protein degradation including aryl sulfonamides that promote disruption of RNA binding motif protein 39 (RBM 39) in CRL4-DNA damage binding protein 1 (DDB 1) and cull 4-related factor 15 (DCAF 15) (CRL 4 DCAF15) dependent manner (Han et al, science 356: eaal3755 (2017)).
Other types of small molecules, including heterobifunctional degradants (also known as PROTAC) (Toure et al, angew.chem.int.ed.Engl.55:1966-1973 (2016)), have been developed for a variety of targets, including kinases (Huang et al, cell chem.biol.25:88-99 (2018)), nuclear receptors (Bondeson et al, nat.chem.biol.11:611-617 (2015)), and epigenetic enzymes (Winter et al, science 348:1376-1381 (2015)). These small molecule degradants bind to the E3 ligase and target protein substrate, promoting the formation of substrate-drug-ligase ternary complex (Nowak et al.,Nat.Chem.Biol.14:706-714(2018);Petzold et al.,Nature 532:127-130(2016);Sievers et al.,Science 362:aat0572(2018)).
While degradants exhibit significant efficacy and sustained consumption of certain target proteins, other proteins have been demonstrated to be resistant to this approach. One such example is the B cell lymphoma 6 (BCL 6) protein for which the heterobifunctional degradants show insufficient target regulation to induce growth inhibition (McCoull et al., ACS chem. Biol.13:3131-3141 (2018)).
BCL6 was originally identified as a locus affected by chromosomal translocation in diffuse large B-cell lymphoma (DLBCL). It is now known to be widely expressed in many lymphomas. Its role in lymphogenesis stems from its function in the humoral immune system, where BCL6 upregulation is necessary for Germinal Center (GC) formation during the humoral immune response (Ye et al, nat. Genet.16:161-170 (1997); dent et al, science 276:89-92 (1997)). GC is a transient structure formed in response to antigenic stimulation. In GC, B cells are resistant to massive proliferation and the mutagenic effects of DNA editing enzymes AICDA for immunoglobulin affinity maturation (Klein et al, nat. Rev. Immunol.8:22-33 (2008)). These activities are coordinated by BCL 6and depend on BCL6, and BCL6 is a powerful transcriptional repressor that silences hundreds of genes. Some of the target genes control DNA damage induction (i.e., ATR, CHEK1, TP53, ARF) and proliferation checkpoints (i.e., CDKN1A, CDKN1B, CDKN2A, CDKN2B, PTEN) (Hatzi et al., trends mol. Med.20:343-352 (2014)). BCL6 also inhibits genes (e.g., IRF4, PRDM 1) required for withdrawal from GC reaction and plasma cell differentiation. This ensures that GC B cells have enough time to acquire somatic hypermutations of their immunoglobulin genes. Thus, deregulation of the inhibition of these target genes may lead to malignant transformation of B cells.
BCL6 also inhibits many oncogenes in GC B cells, including MYC, BCL2, BMI1, and CCND1 (Ci et al, blood 113:5536-5548 (2009)). By this function, BCL6 can alleviate its own pro-oncogene checkpoint inhibition and thereby reduce the likelihood of malignant transformation of GC B cells. This effect is eliminated in the presence of BCL2 or MYC translocations that drive the expression of these protooncogenes through aberrant regulatory elements. The simultaneous presence of MYC and/or BCL2 with BCL6 (regardless of translocation) is clearly detrimental in that it provides both an inhibitory checkpoint effect and a growth and survival promoting effect of MYC and BCL6 for B cells through BCL6 (CARDENAS ET al., clin. Cancer res.23:885-893 (2017)). In a normal immune response, the BCL6 transcriptional complex is disrupted by CD 40-induced ERK signaling and down-regulated BCL6 mRNA by IRF4 and PRDM1, thereby terminating BCL6 function (Polo et al, blood 112:644-651 (2008)). B cells exit the GC reaction requiring termination of BCL6 function.
BCL6 is a promising non-hodgkin lymphoma drug target, such as diffuse large B-cell lymphoma (DLBCL)(Cerchietti et al.,Cancer Cell 17:400-411(2010);Cardenas et al.,J.Clin.Invest.126:3351-3362(2016)) and follicular lymphoma (Bosga-Bouwer et al Genes Chromosomes Cancer 44:44:301-304 (2005)). The increase in pathological BCL6 expression by somatic BCL6 translocation, exon mutations, promoter mutations, or regulatory pathway mutations is a common driver of B-cell malignancies (Hatzi et al., trends mol. Med.20:343-352 (2014)). In genetically engineered mice, the overexpression of BCL6 is sufficient to drive the progression of lymphomas (Cattoretti et al, CANCER CELL 7:445-455 (2005)). BCL6, a major transcription repressor, can rapidly express Germinal Center (GC) B cells and tolerate genomic instability caused by immunoglobulin gene hypermutation and class switching recombination (Hatzi et al., trends mol. Med.20:343-352 (2014)). BCL6 inhibits a range of genes involved in DNA damage reactions (Ranuncolo et al., blood Cells mol. Dis.41:95-99 (2008)), cell cycle checkpoints (Tunyaplin et al., j. Immunol.173:1158-1165 (2004)) and differentiation (Phan et al., nat. Immunol.6:1054-1060 (2005)). As expected, the knockout of BCL6 in lymphoma cells results in tumor arrest (SCHLAGER ET al, oncotarget 11:875-890 (2020)). Several BCL 6-targeting peptides and small molecule inhibitors show efficacy in vivo, but only at high concentrations, this limits their conversion to clinical therapeutic agents (CERCHIETTI ET al, CANCER CELL 17:400-411 (2010); CARDENAS ET al, j.clin.invest.126:3351-3362 (2016)).
The Broad complex/TRAMTRACK/Bric-a-brac (BTB) proteins are distinct protein families characterized by the presence of a common protein-protein interaction domain called the BTB domain. BTB proteins have a variety of functions, ranging from transcriptional regulation and chromatin remodeling to protein degradation and cytoskeletal regulation. The specificity of function is determined in part by the other domains present in a given BTB protein and the interaction partners. Studies of BTB proteins in Drosophila and mammalian systems have revealed the importance of these proteins in a variety of developmental environments, as well as in cancer, neurological and musculoskeletal diseases. BTB proteins play a key role in transcriptional regulation and chromatin remodeling (Chaharbakhshi et al., genesis54:505-518 (2016)).
The BTB domain mediates multiple functions of BCL6, like type dimerization and interaction with co-repressor proteins (Ghetu et al., mol. Cell 29:384-391 (2008); ahmad et al., mol. Cell12:1551-1564 (2003)). Techniques that disrupt the protein-protein interactions between the BTB domain of BCL6 and its co-repressor may be helpful against BCL 6-related diseases.
Summary of The Invention
A first aspect of the present invention relates to a compound having a structure represented by formula (I):
Wherein A, X 1、X2 and R 1 are as described herein.
Another aspect of the invention relates to pharmaceutical compositions comprising a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt or stereoisomer thereof, and a pharmaceutically acceptable carrier.
Another aspect of the invention relates to a method of treating a disease or condition characterized or mediated by aberrant BCL6 activity, which involves administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments, the disease or disorder is malignant lymphoma. In some embodiments, the malignant lymphoma is peripheral T-cell lymphoma (PTCL), diffuse large B-cell lymphoma (DLBCL), mantle Cell Lymphoma (MCL), follicular Lymphoma (FL), chronic Lymphocytic Leukemia (CLL), acute lymphoblastic leukemia/lymphoma (ALL), cutaneous T-cell lymphoma, chronic myeloid leukemia, or B-cell non-hodgkin's lymphoma. In some embodiments, the disease or disorder is cancer.
Brief description of the drawings
Fig. 1A is a graph showing the antiproliferative effect of a treatment with tazistat (Taz). Fig. 1B is a graph showing the antiproliferative effect of the rimet tolstat (Lirametostat) (Lira) treatment. Fig. 1C is a heat map showing calculated excess over Bliss (eob) scores for tazistat treatments. Fig. 1D is a heat map showing calculated excess over Bliss (eob) scores for the rimet tostat treatment. Fig. 1E is a plan view showing that both the tazistat and the rimet tostat treatment reduced the dose requirements of BCL6 degradants.
Detailed Description
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this subject matter belongs. Unless indicated to the contrary, the following terms, as used in the specification and the appended claims, have the indicated meanings to facilitate an understanding of the invention.
As used in this description and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a composition" includes a mixture of two or more such compositions, reference to "an inhibitor" includes a mixture of two or more such inhibitors, and the like.
Unless otherwise indicated, the term "about" refers to within 10% (e.g., within 5%, 2%, or 1%) of a particular value modified by the term "about".
The transitional term "comprising" is synonymous with "including", "containing" or "characterized by … …" and is inclusive or open-ended and does not exclude other unrecited elements or method steps. In contrast, the transitional phrase "consisting of … …" excludes any element, step, or component not specified in the claims. The transitional phrase "consisting essentially of … …" limits the scope of the claims to a specified material or step "as well as a material or step that does not substantially affect the essential and novel features of the claimed invention.
Regarding the compounds of the present invention, and to the extent that the following terms are used herein to further describe these compounds, the following definitions apply.
As used herein, the term "alkyl" refers to a saturated straight or branched chain monovalent hydrocarbon group. In one embodiment, the alkyl group is a C 1-C18 group. In other embodiments, the alkyl group is C0-C6、C0-C5、C0-C3、C1-C12、C1-C8、C1-C6、C1-C5、C1-C4 or a C 1-C3 group (where C 0 alkyl refers to a bond). Examples of alkyl groups include methyl, ethyl, 1-propyl, 2-propyl, isopropyl, 1-butyl, 2-methyl-1-propyl, 2-butyl, 2-methyl-2-propyl, 1-pentyl, n-pentyl, 2-pentyl, 3-pentyl, 2-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, 3-dimethyl-2-butyl, 3-dimethyl-2-butyl, heptyl, octyl, nonyl, decyl, undecyl and dodecyl. In some embodiments, the alkyl is a C 1-C3 alkyl. In some embodiments, the alkyl is a C 3-C5 branched alkyl.
As used herein, the term "alkylene" refers to a straight or branched divalent hydrocarbon chain connecting the rest of the molecule to one group, consisting of carbon and hydrogen only, free of unsaturation and having 1 to 12 carbon atoms, such as methylene, ethylene, propylene, butylene, and the like. The alkylene chain may be linked to the remainder of the molecule by a single bond and to the radical by a single bond. In some embodiments, the alkylene contains 1 to 8 carbon atoms (C 1-C8 alkylene). In other embodiments, the alkylene group contains 1 to 5 carbon atoms (C 1-C5 alkylene). In other embodiments, the alkylene group contains 1 to 4 carbon atoms (C 1-C4 alkylene). In other embodiments, the alkylene group contains 1 to 3 carbon atoms (C 1-C3 alkylene). In other embodiments, the alkylene group contains 1 to 2 carbon atoms (C 1-C2 alkylene). In other embodiments, the alkylene group contains 1 carbon atom (C 1 alkylene).
As used herein, the term "alkenyl" refers to a straight or branched chain monovalent hydrocarbon radical having at least one carbon-carbon double bond. Alkenyl includes groups having "cis" and "trans" directions, or alternatively "E" and "Z" directions. In one example, the alkenyl group is a C 2-C18 group. In other embodiments, the alkenyl group is a C 2-C12、C2-C10、C2-C8、C2-C6 or C 2-C3 group. Examples include vinyl (ethyl) or vinyl (vinyl), prop-1-enyl, prop-2-enyl, 2-methylprop-1-enyl, but-2-enyl, but-3-enyl, but-1, 3-dienyl, 2-methylbut-1, 3-dienyl, hex-1-enyl, hex-2-enyl, hex-3-enyl, hex-4-enyl and hex-1, 3-dienyl.
The term "alkoxy" or "alkoxy" as used herein refers to an alkyl group as described above having an oxygen group attached thereto. Representative alkoxy groups include methoxy, ethoxy, propoxy, t-butoxy, and the like. An "ether" is two hydrocarbon groups covalently linked by oxygen. Thus, the substituent of the alkyl group which makes the alkyl group an ether is or is similar to an alkoxy group, and may be represented by, for example, one of-O-alkyl, -O-alkenyl and-O-alkynyl groups.
As used herein, the term "alkyleneoxy" refers to a saturated monovalent aliphatic radical of the general formula (-O-C nH2n -) wherein n represents an integer (e.g., 1,2,3,4, 5, 6, or 7) and includes straight and branched chain groups. The alkyleneoxy chain may be linked to the remainder of the molecule by a single bond and to the group by a single bond. In some embodiments, the alkyleneoxy group contains from 1 to 3 carbon atoms (-O-C 1-C3 alkyleneoxy). In other embodiments, the alkyleneoxy group contains from 1 to 5 carbon atoms (-O-C 1-C5 alkyleneoxy).
As used herein, the term "cyclic group" refers broadly to any group containing a saturated, partially saturated, or aromatic ring system (e.g., carbocyclyl (cycloalkyl, cycloalkenyl), heterocyclyl (heterocycloalkyl, heterocycloalkenyl), aryl, and heteroaryl), either alone or as part of a larger moiety. The ring groups may have one or more (e.g., fused) ring systems. Thus, for example, a ring group may contain one or more carbocyclyl, heterocyclyl, aryl, or heteroaryl groups.
As used herein, the term "carbocycle" (also referred to as "carbocyclyl") refers to a group having 3 to 20 carbon atoms (e.g., an alkanecarbocycle (alkcarbocyclic)) that contains a saturated, partially unsaturated, or aromatic system, alone or as part of a larger moiety. The term carbocyclyl includes monocyclic, bicyclic, tricyclic, fused, bridged, and spiro ring systems, and combinations thereof. In one embodiment, the carbocyclyl group includes 3 to 15 carbon atoms (C 3-C15). In one embodiment, the carbocyclyl group includes 3 to 12 carbon atoms (C 3-C12). In another embodiment, carbocyclyl includes C 3-C8、C3-C10 or C 5-C10. In another embodiment, the carbocyclyl group as a single ring includes C 3-C8、C3-C6 or C 5-C6. In some embodiments, the carbocyclyl group as a bicyclic ring includes C 7-C12. In another embodiment, the carbocyclyl group as the spiro ring system includes C 5-C12. Representative examples of monocyclic carbocyclyl include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, perdeuterated cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, phenyl and cyclododecyl; bicyclic carbocyclyl groups having 7 to 12 ring atoms include [4,3], [4,4], [4,5], [5,6] or [6,6] ring systems, such as bicyclo [2.2.1] heptane, bicyclo [2.2.2] octane, naphthalene and bicyclo [3.2.2] nonane. Representative examples of spirocarbocyclyl groups include spiro [2.2] pentane, spiro [2.3] hexane, spiro [2.4] heptane, spiro [2.5] octane, and spiro [4.5] decane. The term carbocyclyl includes aryl ring systems as defined herein. The term carbocyclyl also includes cycloalkyl rings (e.g., saturated or partially unsaturated mono-, bi-or spiro carbocycles). The term carbocyclic group also includes carbocycles fused to one or more (e.g., 1, 2, or 3) different ring groups (e.g., aromatic or heterocyclic), wherein the radical or point of attachment is on the carbocycle.
As used herein, the term "heterocyclyl" refers to a "carbocyclyl", alone or as part of a larger moiety, containing a saturated, partially unsaturated, or aromatic ring system in which one or more (e.g., 1, 2, 3, or 4) carbon atoms are replaced with heteroatoms (e.g., O, N, N (O), S, S (O), or S (O) 2). The term heterocyclyl includes monocyclic, bicyclic, tricyclic, fused, bridged, and spiro ring systems, and combinations thereof. In some embodiments, heterocyclyl refers to a 3-to 15-membered heterocyclic ring system. In some embodiments, heterocyclyl refers to a 3-to 12-membered heterocyclic ring system. In some embodiments, heterocyclyl refers to a saturated ring system, e.g., a 3-to 12-membered saturated heterocyclic system. In some embodiments, heterocyclyl refers to a heteroaromatic ring system, e.g., a 5-to 14-membered heteroaromatic ring system. The term heterocyclyl also includes C 3-C8 heterocycloalkyl, which is a saturated or partially unsaturated monocyclic, bicyclic or spiro ring system containing 3 to 8 carbons and one or more (1, 2, 3 or 4) heteroatoms.
In some embodiments, heterocyclyl includes 3 to 12 ring atoms and includes monocyclic, bicyclic, tricyclic, and spiro ring systems, wherein a ring atom is carbon, and 1 to 5 ring atoms are heteroatoms, such as nitrogen, sulfur, or oxygen. In some embodiments, heterocyclyl includes 3-to 7-membered monocyclic rings having one or more heteroatoms selected from nitrogen, sulfur, or oxygen. In some embodiments, heterocyclyl includes 4-to 6-membered monocyclic rings having one or more heteroatoms selected from nitrogen, sulfur, or oxygen. In some embodiments, the heterocyclyl includes a 3-membered monocyclic ring. In some embodiments, the heterocyclyl includes a 4-membered monocyclic ring. In some embodiments, the heterocyclyl includes a 5-to 6-membered monocyclic ring. In some embodiments, the heterocyclyl includes 0 to 3 double bonds. In any of the preceding embodiments, the heterocyclyl includes 1,2,3, or 4 heteroatoms. Any nitrogen or sulfur heteroatoms may optionally be oxidized (e.g., NO, SO 2), and any nitrogen heteroatoms may optionally be quaternized (e.g., [ NR 4]+Cl-、[NR4]+OH-). Representative examples of heterocyclyl groups include oxiranyl, aziridinyl, thiiranyl, azetidinyl, oxetanyl, thietanyl, 1, 2-dithiatidinyl, 1, 3-dithiatidinyl, pyrrolidinyl, dihydro-1H-pyrrolyl, dihydrofuranyl, tetrahydropyranyl, dihydrothienyl, tetrahydrothienyl, imidazolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, 1-dioxothiomorpholinyl, dihydropyranyl, tetrahydropyranyl, hexahydrothiopyranyl, hexahydropyrimidinyl, oxazinyl, thiazinyl, thiazanyl, homopiperazinyl, homopiperidinyl, azepanyl, oxacycloheptyl, thietanyl, oxazepinyl (oxazepanyl), diazepinyl, 1, 4-diazacycloheptyl, oxazepinyl diazacyclyl, thiazepinyl, tetrahydrothiopyranyl, oxazolidinyl, thiazolidinyl, isothiazolidinyl, 1-dioxoisothiazolidinyldinonyl, oxazolidindinonyl, imidazolidinyldinonyl, 4,5,6, 7-tetrahydro [2H ] indazolyl, tetrahydrobenzimidazolyl, 4,5,6, 7-tetrahydrobenzo [ D ] imidazolyl, 1, 6-dihydroimidazo [4,5-D ] pyrrolo [2,3-b ] pyridinyl, thiazinyl, thiophenyl, oxazinyl, thiadiazinyl, oxadiazinyl, dithiazinyl, oxathiazinyl, thiotriazinyl (thiatriazinyl), oxatriazinyl, dithiadiazinyl, imidazolinyl, dihydropyrimidinyl, tetrahydropyrimidinyl, 1-pyrrolinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, thiopyranyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1, 3-dioxolanyl, pyrazolinyl, pyrazolidinyl, dithianyl, dithiopentyl, pyrimidinonyl, pyrimidindionyl, pyrimidinyl-2, 4-dione, piperazinonyl, piperazindinonyl, pyrazolidinyl imidazolinyl, 3-azabicyclo [3.1.0] hexy, 3, 6-diazabicyclo [3.1.1] heptyl, 6-azabicyclo [3.1.1] heptyl, 3-azabicyclo [4.1.0] heptyl, azabicyclo [2.2.2] hexyl 2-azabicyclo [3.2.1] octanyl, 8-azabicyclo [3.2.1] octanyl, 2-azabicyclo [2.2.2] octanyl, 8-azabicyclo [2.2.2] octanyl, 7-oxabicyclo [2.2.1] heptane, azaspiro [3.5] nonyl, azaspiro [2.5] octanyl, azaspiro [4.5] decanyl, 1-azaspiro [4.5] decan-2-onyl, azaspiro [5.5] undecyl, tetrahydroindolyl, octahydroindolyl, tetrahydroisoindolyl, tetrahydroindazolyl, 1-dioxohexahydrothiopyranyl. Examples of 5-membered heterocyclic groups containing sulfur or oxygen atoms and 1 to 3 nitrogen atoms are thiazolyl (including thiazol-2-yl and thiazol-2-yl N-oxide), thiadiazolyl (including 1,3, 4-thiadiazol-5-yl and 1,2, 4-thiadiazol-5-yl), oxazolyl (e.g., oxazol-2-yl) and oxadiazolyl (e.g., 1,3, 4-oxadiazol-5-yl and 1,2, 4-oxadiazol-5-yl). Example 5-membered ring heterocyclyl containing 2 to 4 nitrogen atoms include imidazolyl, such as imidazol-2-yl; triazolyl, such as 1,3, 4-triazol-5-yl; 1,2, 3-triazol-5-yl, 1,2, 4-triazol-5-yl and tetrazolyl, for example 1H-tetrazol-5-yl. Representative examples of benzo-fused 5-membered heterocyclyl groups are benzooxazol-2-yl, benzothiazol-2-yl and benzimidazol-2-yl. Example 6 membered heterocyclyl contains 1 to 3 nitrogen atoms and optionally sulphur or oxygen atoms, for example pyridinyl, such as pyridin-2-yl, pyridin-3-yl and pyridin-4-yl; pyrimidinyl, such as pyrimidin-2-yl and pyrimidin-4-yl; triazinyl groups such as 1,3, 4-triazin-2-yl and 1,3, 5-triazin-4-yl; pyridazinyl, in particular pyridazin-3-yl and pyrazinyl.
Thus, the term heterocycle includes N-heterocyclyl, as used herein, N-heterocyclyl refers to a heterocyclyl containing at least one nitrogen, and wherein the point of attachment of the heterocyclyl to the remainder of the molecule is through a nitrogen atom in the heterocyclyl. Representative examples of N-heterocyclyl include 1-morpholinyl, 1-piperidinyl, 1-piperazinyl, 1-pyrrolidinyl, pyrazolidinyl, imidazolinyl, and imidazolidinyl. The term heterocycle also includes C-heterocyclyl, as used herein, C-heterocyclyl refers to a heterocyclyl containing at least one heteroatom, and wherein the point of attachment of the heterocyclyl to the remainder of the molecule is through a carbon atom in the heterocyclyl. Representative examples of C-heterocyclyl include 2-morpholinyl, 2-or 3-or 4-piperidinyl, 2-piperazinyl, and 2-or 3-pyrrolidinyl. The term heterocycle also includes heterocycloalkyl, as disclosed above, which refers to a group of the formula-R c -heterocyclyl, wherein R c is an alkylene chain. The term heterocycle also includes heterocycloalkoxy, as used herein, which refers to a radical bonded through an oxygen atom of a heterocyclic group of the formula-O-R c -wherein R c is an alkylene chain.
As used herein, the term "aryl" (e.g., "aralkyl" wherein the terminal carbon atom on the alkyl group is the point of attachment, such as benzyl, "aralkoxy" wherein the oxygen atom is the point of attachment, or "aryloxyalkyl" wherein the point of attachment is on the aryl group) used alone or as part of a larger moiety refers to a group comprising a monocyclic, bicyclic, or tricyclic carbocyclic ring system, including fused rings, wherein at least one ring in the system is aromatic. In some embodiments, the aralkoxy is benzoyloxy. The term "aryl" may be used interchangeably with the term "aromatic ring". In one embodiment, aryl groups include groups having 6 to 18 carbon atoms. In another embodiment, aryl groups include groups having 6 to 10 carbon atoms. Examples of aryl groups include phenyl, naphthyl, anthracenyl, biphenyl, phenanthryl, NAPHTHACENYL, 1,2,3, 4-tetrahydronaphthyl, 1H-indenyl, 2, 3-dihydro-1H-indenyl, naphthyridinyl (NAPHTHYRIDINYL), and the like, which may be substituted with one or more substituents described herein or independently. One particular aryl group is phenyl. In some embodiments, an aryl group includes an aromatic ring fused to one or more (e.g., 1,2, or 3) different cyclic groups (e.g., carbocyclic or heterocyclic), wherein the radical or point of attachment is on the aromatic ring.
Thus, the term aryl includes aralkyl (e.g., benzyl), as described above, aralkyl refers to a group of the formula-R c -aryl, wherein R c is an alkylene chain, such as methylene or ethylene. In some embodiments, the aralkyl is an optionally substituted benzyl. The term aryl also includes aralkoxy, as used herein, which refers to a group bonded through an oxygen atom of the formula-O-R c -aryl, wherein R c is an alkylene chain, such as methylene or ethylene.
As used herein, the term "heteroaryl", used alone or as part of a larger moiety, for example, "heteroarylalkyl (heteroarylalkyl)" (also referred to as "heteroarylalkyl (heteroaralkyl)") or "heteroarylalkoxy (heteroarylalkoxy)" (also referred to as "heteroarylalkoxy (heteroaralkoxy)") refers to a monocyclic, bicyclic or tricyclic ring system having 5 to 14 ring atoms, wherein at least one ring is aromatic and contains at least one heteroatom. In one embodiment, heteroaryl groups include 5-to 6-membered monocyclic aromatic groups in which one or more ring atoms are independently optionally substituted nitrogen, sulfur or oxygen. Representative examples of heteroaryl groups include thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, tetrazolyl, thiatriazolyl, oxazolyl, pyridyl, pyrimidinyl, imidazopyridyl, pyrazinyl, pyridazinyl, triazinyl, tetrazinyl, tetrazolo [1,5-b ] pyridazinyl, purinyl, deazapurine, benzoxazolyl, benzofuranyl, benzothiazolyl, benzothiadiazolyl, benzotriazolyl, benzimidazolyl, indolyl, 1, 3-thiazol-2-yl, 1,3, 4-triazol-5-yl, 1,3, 4-oxazol-5-yl, 1,2, 4-oxadiazol-5-yl, 1,3, 4-thiadiazol-5-yl, 1H-tetrazol-5-yl, 1,2, 3-5-yl and pyridin-2-yl N-oxide. The term "heteroaryl" also includes groups in which the heteroaryl group is fused to one or more cyclic (e.g., carbocyclyl or heterocyclyl) rings, wherein the radical or point of attachment is on the heteroaryl ring. Non-limiting examples include indolyl, indolazinyl, isoindolyl, benzothienyl, methylenedioxyphenyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzodioxazolyl, benzothiazolyl, quinolinyl, isoquinolinyl, cinnolazinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroisoquinolinyl, and pyrido [2,3-b ] -1, 4-oxazin-3 (4H) -one. Heteroaryl groups may be monocyclic, bicyclic or tricyclic. In some embodiments, heteroaryl groups include heteroaryl rings fused to one or more (e.g., 1,2, or 3) different cyclic groups (e.g., carbocyclic or heterocyclic), wherein the radical or point of attachment is on the heteroaryl ring, and in some embodiments, wherein the point of attachment is a heteroatom contained in the heterocyclic ring.
The term heteroaryl also includes N-heteroaryl, which as used herein refers to heteroaryl as described above, and which comprises at least one nitrogen atom and wherein the point of attachment of the N-heteroaryl to the remainder of the molecule is through a nitrogen atom in the heteroaryl. The term heteroaryl also includes C-heteroaryl, which as used herein refers to heteroaryl as described above, and wherein the point of attachment of the heteroaryl to the remainder of the molecule is through a carbon atom in the heteroaryl. The term heteroaryl also includes heteroarylalkyl, which as described above refers to a group of the formula- -R c - -heteroaryl, wherein R c is an alkylene chain as described above. The term heteroaryl also includes heteroarylalkoxy (heteroaralkoxy) (or heteroarylalkoxy (heteroarylalkoxy)) groups, which as used herein refers to groups bonded through an oxygen atom of a formula- -O- -R c - -heteroaryl, wherein R c is alkylene as described above.
Unless otherwise indicated, and to the extent that no particular group is further defined, any group described herein may be substituted or unsubstituted. As used herein, the term "substituted" broadly refers to all permissible substituents, provided that such substituents conform to the valences permitted by the substituents and substituents, and the substitution results in stable compounds, i.e., compounds that do not spontaneously undergo transformations of rearrangement, cyclization, elimination, and the like. Representative substituents include halogen, hydroxy, and any other organic group containing any number of carbon atoms (e.g., 1 to 14 carbon atoms), and which may include one or more (e.g., 1,2,3, or 4) heteroatoms, such as oxygen, sulfur, and nitrogen, which are aggregated in a linear, branched, or cyclic structure.
Representative examples of substituents within the scope of any particular group not otherwise disclosed may thus include alkyl (e.g., substituted C1-C6、C1-C5、C1-C4、C1-C3、C1-C2、C1)、 alkoxy (e.g., substituted C1-C6、C1-C5、C1-C4、C1-C3、C1-C2、C1)、 haloalkyl (e.g., CF 3), alkenyl (e.g., C2-C6、C2-C5、C2-C4、C2-C3、C2)、 substituted alkenyl (e.g., substituted C2-C6、C2-C5、C2-C4、C2-C3、C2)、 alkynyl (e.g., ,C2-C6、C2-C5、C2-C4、C2-C3、C2)、 substituted alkynyl (e.g., substituted C2-C6、C2-C5、C2-C4、C2-C3、C2)、 cyclic group (e.g., C 3-C12、C5-C6)), substituted cyclic group (e.g., C 3-C12、C5-C6), carbocycle (e.g., C 3-C12、C5-C6), substituted carbocycle (e.g., substituted C 3-C12、C5-C6), heterocycle (e.g., 3-to 12-membered, 5-to 6-membered), substituted heterocycle (e.g., substituted 3-to 12-membered, 5-to 6-membered), aryl (e.g., benzyl and phenyl), substituted aryl (e.g., substituted benzyl or substituted phenyl), heteroaryl (e.g., substituted pyridyl or pyrimidinyl), aralkyl (e.g., benzyl), substituted aralkyl (e.g., substituted benzyl), substituted hydroxy (e.g., substituted benzyl), aryloxy (e.g., substituted aryloxy (e.g., C3783), aryloxy (e.g., substituted thio) (e.g., C5428), aryloxy (e.g., C5454), thio (e.g., substituted alkylthio) (e.g., C5426), thio (e.g., C5428), cyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted amino, amido, substituted amido, thio, substituted thio, sulfinyl, substituted sulfinyl, sulfonyl, substituted sulfonyl, sulfenamide, substituted sulfenamide, sulfonamide, substituted sulfonamide, urea, substituted urea, carbamate, substituted carbamate, amino acid, and peptide group.
In one aspect, the compounds of the present invention are represented by formula (I):
Or a pharmaceutically acceptable salt or stereoisomer thereof,
Wherein:
X 1 is N, CH, CCl, CF or CCN;
Each X 2 is independently CH 2, S, CHF, CHCl, CHOH or CF 2;
R 1 is hydrogen, =o, -CN, -c≡ch, -OH, -SH, -NH 2, -COOH, halo, (C 1-C6) alkyl, -O- (C 1-C6) alkyl, (C 1-C6) haloalkyl, amido, carboxyl, carbamoyl, sulfamoyl, phenyl, 5-to 8-membered heterocyclyl, -NR 7R8、–C(O)R9、–C(O)NR10R11 or L 1Y1, wherein the alkyl, phenyl or heterocyclyl is optionally substituted with one or more groups selected from: halo, -COOH, -OH, -NH 2、(C1-C6) alkyl, -C (O) O- (C 1-C6) alkyl, -C (O) N (C 1-C6 alkyl) 2、–O–(C1-C6) alkyl, -N (C 1-C3 alkyl) 2, phenyl and 4-to 6-membered heterocyclyl optionally substituted with one or more groups selected from halo and (C 1-C6) alkyl;
r 7 is hydrogen, (C 1-C4) alkyl or (C 3-C6) cycloalkyl;
R 8 is hydrogen, (C 1-C4) alkyl, (C 1-C4) haloalkyl, (C 3-C6) cycloalkyl or six membered heterocyclyl;
R 9 is- (C 1-C3) alkyl-N (C 1-C3 alkyl) 2、(C3-C6) cycloalkyl or a 5-to 6-membered heterocyclyl, wherein the heterocyclyl is optionally substituted with (C 1-C3) alkyl;
R 10 is hydrogen, (C 1-C3) alkyl or (C 3-C6) cycloalkyl;
R 11 is optionally substituted by-NH 2、–O-(C1-C6) alkyl-O- (C 1-C6) alkyl-NH 2 or-O- (C 1-C6) alkyl-O- (C 1-C6) alkyl-NH 2 substituted (C 3-C6) cycloalkyl or (C 1-C6) alkyl;
L 1 is absent, (C 1-C6) alkylene or (C 3-C7) carbocyclyl; wherein the alkylene or carbocyclyl is further optionally substituted with one or more R A groups, which are the same or different;
each R A is independently oxo, alkyl, alkenyl, alkynyl, halo, haloalkyl, carbocyclyl, heterocyclyl, hydroxy, alkoxy, cycloalkoxy, heterocycloalkoxy, haloalkoxy, aryloxy, heteroaryloxy, aralkyloxy, alkenyloxy, alkynyloxy, amino, alkylamino, cycloalkylamino, heterocycloalkylamino, arylamino, heteroarylamino, aralkylamino, N-alkyl-N-arylamino, N-alkyl-N-heteroarylamino, N-alkyl-N-aralkylamino, hydroxyalkyl, aminoalkyl, alkylthio, haloalkylthio, alkylsulfonyl, haloalkylsulfonyl, cycloalkylsulfonyl, heterocycloalkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, alkylaminosulfonyl, cycloalkylaminosulfonyl, heteroarylsulfonyl, heterocyclylalkylaminosulfonyl, arylaminoculfonyl, heteroarylaminosulfonyl, N-alkyl-N-arylaminoculfonyl, N-alkyl-N-heteroarylaminosulfonyl, formyl, alkylcarbonyl, haloalkylcarbonyl, alkenylcarbonyl, alkynylcarbonyl, carboxy, alkoxycarbonyl, alkylcarbonyloxy, amino, alkylsulfonylamino, haloalkylsulfonylamino, cycloalkylsulfonylamino, heterocyclylalkylsulfonylamino, arylsulfonylamino, heteroarylsulfonylamino, aralkylsulfonylamino, alkylcarbonylamino, haloalkylcarbonylamino, cycloalkylcarbonylamino, heterocyclylcarbonylamino, arylcarbonylamino, heteroarylcarbonylamino, aralkylsulfonylamino, aminocarbonyl, alkylaminocarbonyl, cycloalkylaminocarbonyl, heterocyclylalkylaminocarbonyl, arylaminocarbonyl, heteroarylaminocarbonyl, N-alkyl-N-arylaminocarbonyl, N-alkyl-N-heteroarylaminocarbonyl, cyano, nitro, azido or phosphino;
Y 1 is-CN, -OH, halo, (C 1-C4) alkoxy, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, 4-to 7-membered heterocyclyl, (C 3-C6) carbocyclyl, -NR 7'R8'、–C(O)R9、–C(O)NR10R11; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more of the same or different groups selected from (C 1-C4) alkyl, halo, (C 1-C4) haloalkyl, -CN, -OH and-NH 2;
R 7' and R 8' are each independently hydrogen, (C 1-C6) alkyl, (C 3-C7) carbocyclyl, 4-to 7-membered heterocyclyl, (C 6-C10) aryl, or mono-or bi-cyclic 5-to 10-membered heteroaryl; wherein the alkyl, carboxyl, heterocyclyl, aryl or heteroaryl groups are further optionally substituted with one or more identical or different R A groups, or
R 7' and R 8' form, with the nitrogen atom to which they are attached, a 3-to 7-membered heterocyclyl, wherein the heterocyclyl is further optionally substituted with one or more identical or different R A groups, or L 1 is (C 2-C4) alkylene which combines with R 7' to form a 4-to 6-membered heterocyclyl;
R 1' is absent, hydrogen, -CN, -C≡CH, -OH, -SH, -NH 2, -COOH, halo, (C 1-C6) alkyl, -O- (C 1-C6) alkyl, (C 1-C6) haloalkyl, amido, carboxyl, carbamoyl, sulfamoyl, phenyl, 5-to 8-membered heterocyclyl, -NR 7R8、-C(O)R9 or-C (O) NR 10R11; wherein the alkyl, phenyl or heterocyclyl is further optionally substituted with one or more identical or different R A groups, or
R 1' and L 1 together with the same carbon atom to which they are attached form a spiro (C 3-C7) carbocyclyl or a 4-to 7-membered heterocyclyl; wherein the carbocyclyl or heterocyclyl is further optionally substituted with one or more R A groups, which may be the same or different;
is/>
X 3 and X 4 are independently CR 12 or N;
x 5 is CH or N;
R 12 is hydrogen, (C 1-C4) alkyl, halo, hydroxy, amino, (C 1-C4) alkoxy, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, (C 2-C4) alkenyl, (C 2-C4) alkynyl, nitro, cyano, NH (C 1-C4) alkyl or N (C 1-C4) alkyl) 2;
R 2 is hydrogen, (C 1-C6) alkyl, (C 3-C6) carbocyclyl, 4-to 7-membered heterocyclyl, (C 3-C7) carbocyclyl (C 1-C6) alkyl, or 4-to 7-membered heterocyclyl (C 1-C6) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups, wherein R 13 is (C 1-C6) alkyl, (C 1-C6) alkoxy, halo, amino, hydroxy, haloalkyl, NH (C 1-C6) alkyl or N ((C 1-C6) alkyl) 2、(C3-C6) carbocyclyl or 4-to 7-membered heterocyclyl, or R 2 is-L 2-Y2 -Z;
L 2 is (C 1-C5) alkylene which is absent or optionally substituted by one or more substituents selected from (C 1-C2) alkyl and oxo;
Y 2 is absent 、O、S、S(O)、S(O)2、NR'、C(O)、C(O)O、OC(O)、C(O)N(R')、N(R')C(O)、N(R')C(O)N(R')、N(R')C(O)O、OC(O)N(R')、S(O)2N(R') or N (R') S (O) 2;
Each R' is independently hydrogen or (C 1-C4) alkyl;
Z is hydrogen, (C 1-C6) alkyl, (C 2-C6) alkenyl, (C 2-C6) alkynyl, (C 3-C10) carbocyclyl or 3-to 10-membered heterocyclyl; wherein Z is optionally substituted with one or more substituents independently selected from (C 1-C4) alkyl, halo, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, amino, (C 1-C4) aminoalkyl, cyano, hydroxy, carboxy, carbamoyl, sulfamoyl, mercapto, ureido 、NRrRs、ORr、C(O)Rr、C(O)ORr、OC(O)Rr、C(O)NRrRs、N(Rr)C(O)Rr、S(O)0-2Rr、S(O)2NRrRs、N(Rr)SO2Rr、Si(Rr)(Rs)Rt, and (CH 2)1-3NRrRs), wherein R r、Rs and R t are each independently hydrogen, (C 1-C6) alkyl, or (C 3-C6) cycloalkyl, or R r and R s together with the nitrogen atom to which they are attached form a 4-to 9-membered heterocyclyl optionally substituted with one or more substituents selected from (C 1-C4) alkyl, halo, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, (C 1-C4) alkoxy, (C 1-C4) alkylamino, amino, cyano, and hydroxy;
R 3 is-L 3CR14R15R16 or-ch=ch-R 16;
L 3 is absent, O, S, (C 1-C4) alkylene, -O- (C 1-C4) alkylene or-S- (C 1-C4) alkylene;
R 14 is hydrogen or (C 1-C4) alkyl;
R 15 is hydrogen or (C 1-C4) alkyl, or
R 14 and R 15 together with the carbon atom to which they are attached form (C 3-C5) carbocyclyl, 4-to 7-membered heterocyclyl or c=o;
R 16 is (C 1-C6) alkyl 、–NR17R18、–OR17、–C(O)R17、–C(O)OR17、–N(R18)C(O)R17、–C(O)NR17R18、–S(O)–(C1-C6) alkyl, -S (O) 2–(C1-C6 alkyl, -P (O) - (C 1-C6 alkyl) 2、–C(NH)NH2、–(C1-C4) alkyl-NR 18C(O)R17 or 4-to 7-membered heterocyclyl;
R 17 is hydrogen, 3-to 6-membered heterocyclyl or (C 1-C4) alkyl optionally substituted with one or more identical or different groups selected from OH, cl, F, CF 3、N(C1-C4 alkyl) 2、(C3-C6) carbocyclyl, 3-to 6-membered heterocyclyl, (C 2-C4) alkenyl and (C 2-C4) alkynyl;
R 18 is hydrogen or (C 1-C4) alkyl;
R 4 is hydrogen, methyl, - (CH 2)1-3W1W2) or
W 1 is CR 19R19' or C (O);
R 19 and R 19' are independently hydrogen, (C 1-C2) alkyl, fluoro, hydroxy, cyano, nitro, (C 1-C2) alkoxy, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, amino, NH (C 1-C2) alkyl or N (C 1-C2) alkyl) 2, or
R 19 and R 19' together with the carbon atom to which they are attached form a C (O), (C 3-C6) carbocyclyl or a 3-to 6-membered heterocyclyl optionally substituted with one or more substituents independently selected from (C 1-C2) alkyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, (C 1-C2) alkylamino, amino, cyano and hydroxy;
W 2 is cyano, hydroxy, 5-or 6-membered heteroaryl, phenyl, C (O) - (C 1-C2) alkyl, S (O) 2-(C1-C2) alkyl, C (O) OCH 3、C(O)NHCH3、CR20R21R22, amino, NH (C 1-C2) alkyl or N (C 1-C2 alkyl) 2:
R 20 is hydrogen, (C 1-C2) alkyl, fluoro, chloro, bromo, hydroxy, amino, cyano, nitro, (C 1-C2) alkoxy, (C 1-C2) haloalkyl or (C 1-C2) haloalkoxy;
R 21 is hydrogen, (C 1-C2) alkyl, fluoro, chloro, bromo, hydroxy, cyano, nitro, (C 1-C2) alkoxy, (C 1-C2) haloalkyl or (C 1-C2) haloalkoxy or-Y 3-L4-Z2;
Y 3 is absent, O, S, S (O), S (O) 2、NR'、C(O)、C(O)O、OC(O)、C(O)N(R')、N(R')C(O)、S(O)2 N (R ') or N (R') S (O) 2;
L 4 is absent or (C 1-C2) alkylene;
Z 2 is hydrogen, (C 1-C6) alkyl, (C 2-C4) alkenyl, (C 2-C4) alkynyl, phenyl, (C 3-C6) carbocyclyl, OR 4-to 6-membered heterocyclyl, wherein Z 2 is optionally substituted with one OR more substituents independently selected from (C 1-C4) alkyl, halo, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, (C 1-C4) alkoxy, (C 1-C4) alkylamino, amino, cyano, hydroxy, C (O) R ', C (O) OR', OC (O) R ', C (O) NR' R ', and N (R') C (O) R ', wherein each R' is independently hydrogen OR (C 1-C4) alkyl;
Or R 20 and R 21 together with the carbon atom to which they are attached form a (C 3-C6) carbocyclyl or 3-to 6-membered heterocyclyl, optionally substituted with one or more substituents selected from (C 1-C2) alkyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, (C 1-C2) alkylamino, amino, cyano and hydroxy;
R 22 is (C 1-C2) alkyl, -C (O) OR ', -C (O) NR ', -NR ' R ', phenyl, OR a 5 membered heteroaryl, wherein each R ' is independently hydrogen OR (C 1-C2) alkyl;
A "is (C 4-C6) carbocyclyl or 4-to 6-membered heterocyclyl optionally substituted with one or more substituents independently selected from (C 1-C2) alkyl, halo, hydroxy, oxo, cyano and (C 1-C2) alkoxy;
W 3 is NR 23 or CR 24R24';
R 23 is hydrogen, (C 1-C2) alkyl, (C 1-C4) haloalkyl, (C 1-C4) hydroxyalkyl, -C (O) CH 3 or-C (O) O- (C 1-C4) alkyl;
r 24 and R 24' are independently hydrogen, (C 1-C2) alkyl, cyclopropyl, fluoro, chloro, bromo, hydroxy, amino, cyano, nitro, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, -C (O) OR ', NR ' R ', phenyl OR 5 membered heteroaryl;
R 5 is hydrogen, (C 1-C4) alkyl, (C 3-C6) cycloalkyl, (C 1-C4) haloalkyl or cyano, wherein the alkyl or cycloalkyl is optionally substituted with one or more substituents selected from (C 1-C4) alkyl, (C 3-C6) cycloalkyl, hydroxy, (C 1-C2) alkoxy, amino, NH (C 1-C2) alkyl, N ((C 1-C2) alkyl) 2、(C1-C2) aminoalkyl and halo;
R 5' is hydrogen, (C 1-C4) alkyl, cyano, (C 1-C4) haloalkyl or-Y 4-L5-Z3;
Y 4 is absent, C (O) O or C (O) N (R ");
L 5 is absent or (C 1-C2) alkylene;
Z 3 is hydrogen, (C 1-C6) alkyl, phenyl, (C 3-C6) cycloalkyl or 4-to 6-membered heterocyclyl, wherein Z 3 is optionally substituted with one or more substituents independently selected from (C 1-C2) alkyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, amino, nitro, cyano and hydroxy, or
R 5 and R 5' together with the carbon atom to which they are attached form a (C 4-C6) carbocyclyl or a 4-to 6-membered heterocyclyl;
A 'is a 6-or 7-membered heterocyclyl, wherein in addition to R 5 and R 5', the heterocyclyl is optionally substituted with one or more substituents independently selected from oxo, (C 1-C2) alkyl, cyclopropyl, spiro-cyclopropyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, amino, cyano and hydroxy;
X 6 is CR 25 or N;
R 25 is hydrogen, fluoro, chloro or methyl;
R 6 is hydrogen, (C 1-C2) alkyl, (C 3-C4) cycloalkyl, (C 1-C2) haloalkyl, cyano, (C 2-C4) alkenyl, or (C 2-C4) alkynyl;
R 6' is (C 1-C4) alkyl, cyano, (C 1-C4) haloalkyl or-Y 5-L6-Z4;
Y 5 is absent, C (O) O, OC (O), C (O) N (R ') or S (O) 2 N (R');
l 6 is (C 1-C2) alkylene which is absent or optionally substituted by one or more substituents selected from (C 1-C2) alkyl and oxo;
Z 4 is hydrogen, (C 1-C6) alkyl, (C 2-C4) alkenyl, (C 2-C4) alkynyl, phenyl, (C 3-C6) carbocyclyl, (C 3-C6) cycloalkenyl, or 4-to 6-membered heterocyclyl, wherein Z 4 is optionally substituted with one or more substituents independently selected from oxo, (C 1-C4) alkyl, (C 3-C6) cycloalkyl, halo, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, (C 1-C4) alkoxy, (C 1-C4) alkylamino, amino, nitro, cyano, hydroxy, C (O) R u、C(O)ORu、OC(O)Ru、C(O)NRuRu, and N (R u)C(O)Ru), wherein each R u is independently hydrogen, (C 1-C4) alkyl, or (C 3-C6) cycloalkyl, or;
Z 4 is-Q-L 7-W4, where
Q is absent, O, NH or N (C 1-C2) alkyl;
L 7 is (C 1-C2) alkylene which is absent or optionally substituted by one or more substituents selected from oxo and (C 1-C2) alkyl;
W 4 is (C 1-C4) alkyl, phenyl, (C 3-C6) cycloalkyl, (C 3-C6) cycloalkenyl, or a 5-or 6-membered heterocyclyl, wherein W 4 is optionally substituted with one or more substituents independently selected from (C 1-C4) alkyl, halo, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, (C 1-C4) alkoxy, (C 1-C4) alkylamino, amino, nitro, cyano, and hydroxy, or
R 6 and R 6' together with the carbon atom to which they are attached form a (C 3-C10) carbocyclyl or 4-to 10-membered heterocyclyl, optionally substituted with one or more substituents independently selected from oxo, (C 1-C2) alkyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, (C 1-C2) alkylamino, amino, nitro, cyano or hydroxy; or (C 3-C10) carbocyclyl or 4-to 10-membered heterocyclyl optionally fused to a 5-or 6-membered heteroaryl or benzene ring, and the 5-or 6-membered heteroaryl or benzene ring is optionally substituted with (C 1-C2) alkyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, (C 1-C2) alkylamino, amino, nitro, cyano or hydroxy; and is also provided with
R 6 "is hydrogen, (C 1-C4) alkyl, (C 1-C2) haloalkyl, (C 1-C2) alkoxy, (C 1-C2) haloalkoxy, cyano, nitro, ethynyl, phenyl, or 5-or 6-membered heteroaryl, wherein the alkyl, phenyl, or heteroaryl is optionally substituted with one or more substituents independently selected from halo, hydroxy, and amino.
In some embodiments, R 1 is methyl, -OH, -NH 2、-COOH、-CH2CH2OH、-CH2CH2NH2,In some embodiments, R 1 is methyl. In some embodiments, R 1 is —oh. In some embodiments, R 1 is-NH 2. In some embodiments, R 1 is-COOH. In some embodiments, R 1 is-CH 2CH2 OH. In some embodiments, R 1 is-CH 2CH2NH2.
In some embodiments, X 1 is N, CH, CCl, or CF. In some embodiments, X 1 is N. In some embodiments, X 1 is CH. In some embodiments, X 1 is CCl. In some embodiments, X 1 is CF.
In some embodiments, X 2 is CH 2, CHF, CHCl, or CF 2. In some embodiments, X 2 is CH 2. In some embodiments, X 2 is CHF. In some embodiments, X 2 is CHCl. In some embodiments, X 2 is CF 2.
In some embodiments of the present invention, in some embodiments,Is/>And the compound of formula (I) has the structure of formula I-1,Or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments of formula I-1, X 3 is N. In some embodiments of formula I-1, X 3 is CR 12. In some embodiments of formula I-1, X 3 is CH. In some embodiments of formula I-1, X 3 is CF. In some embodiments of formula I-1, X 3 is COMe.
In some embodiments of formula I-1, X 4 is N. In some embodiments of formula I-1, X 4 is CR 12. In some embodiments of formula I-1, X 4 is CH. In some embodiments of formula I-1, X 4 is CF. In some embodiments of formula I-1, X 4 is COMe.
In some embodiments of formula I-1, X 5 is N. In some embodiments of formula I-1, X 5 is CH.
In some embodiments of formula I-1, R 2 is (C 1-C2) alkyl, 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl, or 4 membered heterocyclyl (C 1) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups. In some embodiments, R 2 is methyl. In some embodiments, R 2 is 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl or 4 membered heterocyclyl (C 1) alkyl, wherein the heterocyclyl contains 1 heteroatom selected from N and O.
In some embodiments of formula I-1, R 3 is-L 3CR14R15R16. In some embodiments of formula I-1, L 3 is (C 1-C4) alkylene, -O- (C 1-C4) alkylene, or-S- (C 1-C4) alkylene. In some embodiments of formula I-1, L 3 is-O- (C 1-C4) alkylene. In some embodiments of formula I-1, L 3 is-O- (C 1) alkylene.
In some embodiments of formula I-1, R 14 and R 15 together with the carbon atom to which they are attached form (C 3-C5) carbocyclyl, 4 to 7 membered heterocyclyl, or c=o. In some embodiments of formula I-1, R 14 and R 15 together with the same carbon atom to which they are attached form c=o. In some embodiments of formula I-1, R 14 and R 15 together with the same carbon atom to which they are attached form a 4 to 7 membered heterocyclyl. In some embodiments of formula I-1, R 14 and R 15 together with the same carbon atom to which they are attached form an oxetane ring.
In some embodiments of formula I-1, R 16 is (C 1-C6) alkyl, -NR 17R18, OR-OR 17. In some embodiments of formula I-1, R 16 is methyl, hydroxy, amino, or NHMe. In some embodiments, R 16 is methyl. In some embodiments, R 16 is hydroxy. In some embodiments, R 16 is amino. In some embodiments, R 16 is NHMe.
In some embodiments, the compound of formula I-1 has the formula I-1a, I-1b, I-1c, I-1d, I-1e, I-1f, I-1g, I-1h, I-1I, or I-1j:
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments, the compound of formula I-1 has the formula I-1a ', I-1b ', I-1c ', I-1d ' I-1e ':
Or a pharmaceutically acceptable salt or stereoisomer thereof. In some embodiments of formulas I-1a ', I-1b ', I-1c ', I-1d ' and I-1e ', R 2 is methyl. In some embodiments of formulas I-1a ', I-1b ', I-1C ', I-1d ', and I-1e ', R 14 and R 15 form c=o, and R 16 is-NR 17R18. In some embodiments of formulas I-1a ', I-1b ', I-1c ', I-1d ' and I-1e ', R 16 is NHMe.
In some embodiments, the compound of formula I-1 has the formula I-1k, I-1l, I-1m, I-1n, I-1o, I-1p, I-1q, I-1r, I-1s, I-1t, I-1u, I-1v, I-1w, I-1x, I-1y, I-1z, I-1aa, I-1bb, I-1cc, or I-1dd:
/>
/>
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments of the present invention, in some embodiments,Is/>And the compound of formula (I) has the structure of formula I-2,Or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments of formula I-2, X 3 is CR 12. In some embodiments of formula I-2, X 3 is CH. In some embodiments of formula I-2, X 3 is N.
In some embodiments of formula I-2, X 4 is CR 12. In some embodiments of formula I-2, X 4 is CH. In some embodiments of formula I-2, X 4 is N.
In some embodiments of formula I-2, R 2 is (C 1-C2) alkyl, 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl, or 4 membered heterocyclyl (C 1) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups. In some embodiments, R 2 is methyl. In some embodiments, R 2 is 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl or 4 membered heterocyclyl (C 1) alkyl, wherein the heterocyclyl contains 1 heteroatom selected from N and O.
In some embodiments of formula I-2, R 4 is- (CH 2)1-3W1W2. In some embodiments of formula I-2, R 4 is- (CH 2)2W1W2. In some embodiments of formula I-2, R4 isWherein W 3 is NR 23 and A' is an optionally substituted 4-to 6-membered heterocyclyl. In some embodiments of formula I-2, R 4 is/>
In some embodiments of formula I-2, W 1 is CR 19R19'. In some embodiments of formula I-2, R 19 and R 19' are independently hydrogen or (C 1-C2) alkyl. In some embodiments of formula I-2, R 19 and R 19' are both (C 1-C2) alkyl. In some embodiments of formula I-2, R 19 and R 19' are both methyl. In some embodiments of formula I-2, R 19 and R 19' together with the carbon atom to which they are attached form a (C 3-C6) carbocyclyl group. In some embodiments of formula I-2, R 19 and R 19' together with the carbon atom to which they are attached form cyclopropyl.
In some embodiments of formula I-2, W 2 is cyano, hydroxy, or amino. In some embodiments of formula I-2, W 2 is hydroxy.
In some embodiments, the compound of formula I-2 has the formula I-2a, I-2b, I-2c, I-2d, I-2e, I-2f, I-2g, I-2h, I-2I, or I-2j:
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments, the compound of formula I-2 has the formula I-2k, I-2l, I-2m, I-2n, I-2o, I-2p, I-2q, I-2r, I-2s, I-2t, I-2u, I-2v, I-2w, I-2x, I-2y, I-2z, I-2aa, I-2bb, I-2cc, or I-2dd:
/>
/>
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments of the present invention, in some embodiments,Is/>And the compound of formula (I) has the structure of formula I-3,/>Or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments of formula I-3, X 2 is CH 2. In some embodiments of formula I-3, X 2 is CF 2.
In some embodiments of formula I-3, X 3 is CR 12. In some embodiments of formula I-3, X 3 is CH. In some embodiments of formula I-3, X 3 is N.
In some embodiments of formula I-3, X 4 is CR 12. In some embodiments of formula I-3, X 4 is CH. In some embodiments of formula I-3, X 4 is N.
In some embodiments of formula I-3, R 2 is (C 1-C2) alkyl, 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl, or 4 membered heterocyclyl (C 1) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups. In some embodiments, R 2 is methyl. In some embodiments, R 2 is 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl or 4 membered heterocyclyl (C 1) alkyl, wherein the heterocyclyl contains 1 heteroatom selected from N and O.
In some embodiments of formula I-3, R 5 is (C 3-C6) cycloalkyl and R 5' is H. In some embodiments of formula I-3, R 5 is cyclopropyl and R 5' is H.
In some embodiments of formula I-3, a 'is a 7 membered heterocyclyl, wherein the heterocyclyl comprises 2 heteroatoms selected from N and O, and in addition to R 5 and R 5', the heterocyclyl is optionally substituted with one or more substituents independently selected from oxo, (C 1-C2) alkyl, cyclopropyl, spiro-cyclopropyl, halogen, (C 1-C2) haloalkyl, (C 1-C2) alkoxy, amino, cyano, and hydroxy.
In some embodiments, the compound of formula I-3 has the formula I-3a, I-3b, I-3c, I-3d, I-3e, I-3f, I-3g, I-3h, I-3I, I-3j, I-3k, I-3l, I-3m, I-3n, I-3o, I-3p, I-3q, I-3r, I-3s, or I-3t:
/>
/>
/>
Or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each R 25 is independently oxo, (C 1-C2) alkyl, cyclopropyl, spirocyclopropyl, halo, (C 1-C2) haloalkyl, (C 1-C2) alkoxy, amino, cyano, and hydroxy; and n is 0 to 3.
In some embodiments, the compound of formula I-3 has formula-3 u or I-3v:
Or a pharmaceutically acceptable salt or stereoisomer thereof. /(I)
In some embodiments of the present invention, in some embodiments,Is/>And a compound of formula (I)
Has the structure of the formula I-4,Or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments of formula I-4, X 3 is CR 12. In some embodiments of formula I-4, X 3 is CH. In some embodiments of formula I-4, X 3 is N.
In some embodiments of formula I-4, X 4 is CR 12. In some embodiments of formula I-4, X 4 is CH. In some embodiments of formula I-4, X 4 is N.
In some embodiments of formula I-4, X 6 is N. In some embodiments of formula I-4, X 6 is CH.
In some embodiments of formula I-4, R 2 is (C 1-C2) alkyl, 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl, or 4 membered heterocyclyl (C 1) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups. In some embodiments, R 2 is methyl. In some embodiments, R 2 is 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl or 4 membered heterocyclyl (C 1) alkyl, wherein the heterocyclyl contains 1 heteroatom selected from N and O.
In some embodiments, the compound of formula I-4 has the formula I-4a, I-4b, I-4c, I-4d, I-4e, I-4f, I-4g, I-4h, I-4I, or I-4j:
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments of the present invention, in some embodiments,Is/>And the compound of formula (I) has the structure of formula I-5,Or a pharmaceutically acceptable salt or stereoisomer thereof.
In some embodiments of formula I-5, X 3 is N. In some embodiments of formula I-5, X 3 is CR 12. In some embodiments of formula I-5, X 3 is CH. In some embodiments of formula I-5, X 3 is CF. In some embodiments of formula I-5, X 3 is COMe.
In some embodiments of formula I-5, X 4 is N. In some embodiments of formula I-5, X 4 is CR 12. In some embodiments of formula I-5, X 4 is CH. In some embodiments of formula I-5, X 4 is CF. In some embodiments of formula I-5, X 4 is COMe.
In some embodiments of formula I-5, R 2 is 4-membered heterocyclyl or 4-membered heterocyclyl (C 2) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups. In some embodiments, R 2 is 4 membered heterocyclyl or 4 membered heterocyclyl (C 2) alkyl, wherein the heterocyclyl contains 1 heteroatom selected from N and O.
In some embodiments of formula I-5, R 2 is (C 1-C6) alkyl, (C 1-C6) alkyl-OH, (C 1-C6) alkylnh (C 1-C6) alkyl, or (C 1-C6) alkyl-N ((C 1-C6) alkyl) 2.
In some embodiments of formula I-5, R 3 is-L 3CR14R15R16. In some embodiments of formula I-5, L 3 is (C 1-C4) alkylene, -O- (C 1-C4) alkylene, or-S- (C 1-C4) alkylene. In some embodiments of formula I-5, L 3 is-O- (C 1-C4) alkylene. In some embodiments of formula I-5, L 3 is-O- (C 1) alkylene.
In some embodiments of formula I-5, R 14 and R 15 together with the carbon atom to which they are attached form (C 3-C5) carbocyclyl, 4-to 7-membered heterocyclyl, or c=o. In some embodiments of formula I-5, R 14 and R 15 together with the same carbon atom to which they are attached form c=o. In some embodiments of formula I-5, R 14 and R 15 together with the same carbon atom to which they are attached form a 4-to 7-membered heterocyclyl. In some embodiments of formula I-5, R 14 and R 15 together with the same carbon atom to which they are attached form an oxetane ring.
In some embodiments of formula I-5, R 16 is (C 1-C6) alkyl, -NR 17R18, OR-OR 17. In some embodiments of formula I-5, R 16 is methyl, hydroxy, amino, or NHMe. In some embodiments, R 16 is methyl. In some embodiments, R 16 is hydroxy. In some embodiments, R 16 is amino. In some embodiments, R 16 is NHMe.
In some embodiments, the compound of formula I-5 has the formula I-5a, I-5b, I-5c, I-5d, I-5e, I-5f, I-5g, I-5h, I-5I, or I-5j:
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
Representative examples of compounds of the present invention include:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
The compounds of the invention may be in the form of the free acid or free base or a pharmaceutically acceptable salt. As used herein, the term "pharmaceutically acceptable" in the context of salts refers to salts of compounds that do not abrogate the biological activity or properties of the compound and are relatively non-toxic, i.e., the compound in salt form may be administered to a subject without causing adverse biological effects (such as dizziness or gastric discomfort) or interacting in a deleterious manner with any of the other components of the composition in which it is contained. The term "pharmaceutically acceptable salt" refers to the product obtained by reacting a compound of the invention with a suitable acid or base. Examples of pharmaceutically acceptable salts of the compounds of the present invention include those derived from suitable inorganic bases such as Li, na, K, ca, mg, fe, cu, al, zn and Mn salts. Examples of pharmaceutically acceptable non-toxic acid addition salts are salts of amino groups with inorganic acids, such as hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisate, fumarate, gluconate, glucuronate, gluconate, formate, benzoate, glutamate, mesylate, ethanesulfonate, benzenesulfonate, 4-methylbenzenesulfonate, p-toluenesulfonate and the like. Certain compounds of the invention may form pharmaceutically acceptable salts with various organic bases such as lysine, arginine, guanidine, diethanolamine, or metformin. Suitable base salts include aluminum, calcium, lithium, magnesium, potassium, sodium or zinc salts.
The compounds of the invention may have at least one chiral center and thus may be in the form of stereoisomers, which as used herein, include all isomers of the individual compounds, differing only in the orientation of their atoms in space. The term stereoisomers includes mirror image isomers (enantiomers, including the (R-) or (S-) configuration of the compounds), mirror image isomer mixtures of the compounds (physical mixtures of enantiomers and racemates or racemic mixtures), geometric (cis/trans or E/Z, R/S) isomers of the compounds, and isomers of the compounds having more than one chiral center and which are not mirror images of each other (diastereomers). Chiral centers of compounds may undergo epimerization in vivo; thus, for these compounds, administration of the (R-) form of the compound is considered equivalent to administration of the (S-) form of the compound. Thus, the compounds of the present invention may be manufactured and used in the form of individual isomers and substantially free of other isomers, or in the form of mixtures of various isomers, e.g., racemic mixtures of stereoisomers.
In some embodiments, the compound is an isotopic derivative in that it has at least one desired isotopic substitution of an atom, the amount of substitution being higher than the natural abundance of the isotope, i.e., enriched. In one embodiment, the compound includes deuterium or multiple deuterium atoms. Substitution with heavier isotopes such as deuterium (i.e., 2 H), may be advantageous for certain therapeutic advantages due to higher metabolic stability, for example increased in vivo half-life or reduced dosage requirements and therefore may be advantageous in certain circumstances.
The compounds of formula (I) may also be in N-oxide form, crystalline form (also known as polymorphs), active metabolites, prodrugs, tautomers and unsolvated forms of compounds having the same type of activity, as well as solvated (e.g., hydrated) forms using pharmaceutically acceptable solvents (e.g., water, ethanol, etc.).
The compounds of the present invention may be prepared by crystallization under different conditions and may exist as one or a combination of polymorphs of the compound. For example, different polymorphs can be identified and/or prepared by performing crystallization at different temperatures, or by performing recrystallization using different solvents or different solvent mixtures during crystallization using various cooling modes (from very fast to very slow cooling). Polymorphs can also be obtained by heating or melting the compound and then gradually or rapidly cooling. The presence of polymorphs may be determined by solid probe NMR spectroscopy, IR spectroscopy, differential scanning calorimetry, powder X-ray diffraction patterns, and/or other known techniques.
In some embodiments, the pharmaceutical composition comprises a co-crystal of a compound of the invention. As used herein, the term "co-crystal" refers to a stoichiometric multicomponent system comprising a compound of the invention and a co-crystal former, wherein the compound of the invention and the co-crystal former are linked by a non-covalent interaction. As used herein, the term "co-crystal former" refers to a compound capable of forming intermolecular interactions with and co-crystallizing with a compound of the present invention. Representative examples of co-crystal formers include benzoic acid, succinic acid, fumaric acid, glutaric acid, trans-cinnamic acid, 2, 5-dihydroxybenzoic acid, glycolic acid, trans-2-hexanoic acid, 2-hydroxycaproic acid, lactic acid, sorbic acid, tartaric acid, ferulic acid, suberic acid, picolinic acid, salicylic acid, maleic acid, saccharin, 4' -bipyridine para-aminosalicylic acid, nicotinamide, urea, isonicotinamide, methyl 4-hydroxybenzoate, adipic acid, terephthalic acid, resorcinol, pyrogallol, phloroglucine, pyrogallol, isoniazid, theophylline, adenine, theobromine, phenacetin, antipyrine, ethoxytheophylline, and phenobarbital.
Synthesis method
In another aspect, the present invention relates to a process for the manufacture of a compound of formula (I), or a pharmaceutically acceptable salt or stereoisomer thereof. In a broad sense, the compounds of the present invention, or pharmaceutically acceptable salts or stereoisomers thereof, may be prepared by any process known to be suitable for preparing chemically related compounds. The compounds of the present invention can be better understood in conjunction with the synthetic schemes described in the various working examples, and these schemes illustrate non-limiting methods by which the compounds of the present invention can be prepared.
Pharmaceutical composition
Another aspect of the invention relates to pharmaceutical compositions comprising a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt or stereoisomer thereof, and a pharmaceutically acceptable carrier. The term "pharmaceutically acceptable carrier" as known in the art refers to a pharmaceutically acceptable material, composition or carrier suitable for administration of a compound of formula (I) to a mammal. Suitable carriers can include, for example, liquids (both aqueous and non-aqueous, and combinations thereof), solids, encapsulating materials, gases, and combinations thereof (e.g., semi-solids) and gases that function to carry or transport the compounds from one organ or body part to another organ or body part. The carrier is "acceptable", i.e., physiologically inert and compatible with the other ingredients of the formulation, and not deleterious to the subject or patient. Depending on the type of formulation, the composition may also include one or more pharmaceutically acceptable excipients.
In a broad sense, the compounds of formula (I) and pharmaceutically acceptable salts and stereoisomers thereof may be formulated in accordance with conventional pharmaceutical practice, e.g. conventional mixing, dissolving, granulating, dragee-making, pulverizing, emulsifying, encapsulating and tabletting processes, into compositions of a given type (see, e.g. ,Remington:The Science and Practice of Pharmacy(20th ed.),ed.A.R.Gennaro,Lippincott Williams&Wilkins,2000 and Encyclopedia of Pharmaceutical Technology,eds.J.Swarbrick and J.C.Boylan,1988-1999,Marcel Dekker,New York). formulations, depending on the mode of administration, which may include enteral (e.g. oral, buccal, sublingual and rectal), parenteral (e.g. subcutaneous (s.c.), intravenous (i.v.), intramuscular (i.m.) and intrasternal injection, or infusion techniques, intraocular, intraarterial, intramedullary, intrathecal, intraventricular, transdermal, intradermal, intravaginal, intraperitoneal, mucosal, nasal, intratracheal instillation, bronchial instillation and inhalation), generally the most suitable route of administration will depend on a variety of factors including, for example, the nature of the agent (e.g. its stability in the gastrointestinal environment) and/or the condition of the subject (e.g. whether the subject is capable of being administered, e.g. orally, alone or as well as an acute dose, or as a relatively rapid, e.g. oral dose.
In some embodiments, the compounds are formulated for oral or intravenous administration (e.g., systemic intravenous injection).
Thus, the compounds of formula (I) may be formulated as solid compositions (e.g., powders, tablets, dispersible granules, capsules, cachets, and suppositories), liquid compositions (e.g., solutions that dissolve the compound, suspensions that disperse the compound solid particles, emulsions, and solutions containing liposomes, micelles or nanoparticles, syrups, and elixirs); semisolid compositions (e.g., gels, suspensions, and creams); and a gas (e.g., a propellant for an aerosol composition). The compounds may also be formulated for immediate, medium or slow release.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In such solid dosage forms, the active compound is admixed with carriers such as sodium citrate or dicalcium phosphate and other carriers or excipients, for example a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as methyl cellulose, microcrystalline cellulose, hydroxypropyl methylcellulose, carboxymethylcellulose, sodium carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrants such as crosslinked polymers (e.g., crosslinked polyvinylpyrrolidone (crospovidone), crosslinked sodium carboxymethylcellulose (croscarmellose sodium), sodium starch glycolate, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarders such as paraffin, f) absorption promoters such as quaternary ammonium compounds, g) wetting agents such as cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. Solid compositions of a similar type may also be used as fill for soft and hard filled gelatin capsules using excipients such as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings. They may also contain opacifying agents.
In some embodiments, the compounds of formula (I) may be formulated as hard gelatin or soft gelatin capsules. Representative excipients that may be used include pregelatinized starch, magnesium stearate, mannitol, sodium stearyl fumarate, lactose anhydrous, microcrystalline cellulose, and croscarmellose sodium. The gelatin shell may comprise gelatin, titanium dioxide, iron oxide, and a colorant.
Liquid dosage forms for oral administration include solutions, suspensions, emulsions, microemulsions, syrups and elixirs. In addition to the compounds, the liquid dosage forms may contain aqueous or non-aqueous carriers commonly used in the art (depending on the solubility of the compound), for example, water or other solvents, solubilizing agents and emulsifiers such as ethanol, isopropanol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. The oral compositions may also include excipients such as wetting agents, suspending agents, colorants, sweeteners, flavoring agents, and flavoring agents.
Injectable formulations for parenteral administration may include sterile aqueous solutions or oily suspensions. They may be formulated according to standard techniques using suitable dispersing or wetting agents and suspending agents. Sterile injectable preparations are also sterile injectable solutions, suspensions or emulsions in non-toxic parenterally acceptable diluents or solvents, such as solutions in 1, 3-butanediol. Acceptable carriers and solvents that can be used are water, ringer's solution, u.s.p. And isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. In addition, fatty acids (such as oleic acid) are useful in the preparation of injectables. The injectable formulation may be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which may be dissolved or dispersed in sterile water or other sterile injectable medium prior to use. The effect of the compound can be prolonged by slowing its absorption, which can be achieved by using liquid suspensions or crystalline or amorphous materials with poor water solubility. Prolonged absorption of the compound from the parenterally administered formulation may also be accomplished by suspending the compound in an oily vehicle.
In certain embodiments, the compounds of formula (I) may be administered in a local rather than systemic manner, for example, by direct injection of the conjugate into the organ, typically in the form of a depot or slow release formulation. In particular embodiments, the depot is administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Injectable depot forms are made by forming the compound into a microcapsule matrix in a biodegradable polymer (e.g., polylactic acid-polyglycolic acid, polyorthoesters, and polyanhydrides). The release rate of the compound can be controlled by varying the ratio of compound to polymer and the nature of the particular polymer employed. Injectable depot formulations are also prepared by encapsulating the compound in liposomes or microemulsions which are compatible with body tissues. Furthermore, in other embodiments, the compounds are delivered in targeted drug delivery systems, e.g., in liposomes coated with organ-specific antibodies. In such embodiments, the liposome targets the organ and is selectively taken up by the organ.
The compositions may be formulated for buccal or sublingual administration, examples of which include tablets, troches and gels.
The compounds of formula (I) may be formulated for administration by inhalation. Various forms suitable for inhaled administration include aerosols, sprays or powders. The pharmaceutical composition may be delivered from a pressurized package or nebulizer in the form of an aerosol spray using a suitable propellant (e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas). In some embodiments, the dosage unit of the pressurized aerosol may be determined by providing a valve to deliver a metered amount. In some embodiments, capsules and cartridges (e.g., for inhalers or insufflators) containing gelatin may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
The compounds of formula (I) may be formulated for topical administration, as used herein, refers to intradermal administration of the formula (I) formulation to the epidermis. These types of compositions are typically in the form of ointments, pastes, creams, lotions, gels, solutions and sprays.
Representative examples of carriers for formulating the topical compounds include solvents (e.g., alcohols, polyols, water), creams, lotions, ointments, oils, plasters, liposomes, powders, emulsions, microemulsions, and buffers (e.g., hypotonic or buffered saline). For example, a cream may be formulated using saturated or unsaturated fatty acids such as stearic acid, palmitic acid, oleic acid, palmitoleic acid, cetyl alcohol or oleyl alcohol. The cream may also contain nonionic surfactants such as polyoxyl (40) stearate.
In some embodiments, the surface preparation may further comprise an excipient, an example of which is a penetration enhancer. These formulations are capable of transporting and transporting the pharmacologically active compound through the stratum corneum into the epidermis or dermis, preferably with little or no systemic absorption. Various compounds have been evaluated for their effectiveness in increasing the skin permeation rate of drugs. See, e.g., percutaneous Penetration Enhancers, maibach h.i. and Smith h.e. (eds.), CRC Press, inc., boca Raton, fla (1995) (which investigated the use and testing of various skin penetration enhancers) and Buyuktimkin et al, CHEMICAL MEANS of Transdermal Drug Permeation ENHANCEMENT IN TRANSDERMAL and Buyuktimkin et al Topical Drug Delivery Systems,Gosh T.K.,Pfister W.R.,Yum S.I.(Eds.),Interpharm Press Inc.,Buffalo Grove,Ill.
(1997). Representative examples of penetration enhancers include triglycerides (e.g., soybean oil), aloe compositions (e.g., aloe vera gel), ethanol, isopropanol, octylphenyl polyethylene glycol (octylphenylpolyethylene glycol), oleic acid, polyethylene glycol 400, propylene glycol, N-decyl methyl sulfoxide, fatty acid esters (e.g., isopropyl myristate, methyl laurate, glycerol monooleate, and propylene glycol monooleate), and N-methyl pyrrolidone.
Representative examples of other excipients that may be included in the surface formulations, as well as in other types of formulations, to the extent that they are compatible include preservatives, antioxidants, moisturizers, emollients, buffers, solubilizers, skin protectants, and surfactants. Suitable preservatives include alcohols, quaternary amines, organic acids, benzoates and phenols. Suitable antioxidants include ascorbic acid and its esters, sodium bisulphite, butyl hydroxy toluene, butyl hydroxy anisole, tocopherols and chelating agents such as EDTA and citric acid. Suitable humectants include glycerin, sorbitol, polyethylene glycol, urea and propylene glycol. Suitable buffers include citric acid, hydrochloric acid and lactic acid buffers. Suitable solubilizing agents include quaternary ammonium salts, cyclodextrins, benzyl benzoate, lecithin and polysorbates. Suitable skin protectants include vitamin E oil, allantoin (allantoin), dimethicone, glycerin, petrolatum, and zinc oxide.
Transdermal formulations typically employ transdermal delivery devices and transdermal delivery patches, wherein the compound is formulated as a lipophilic emulsion or buffered aqueous solution dissolved and/or dispersed in a polymer or adhesive. Patches may be constructed for continuous, pulsed (pulsatile) or on-demand delivery of agents. Transdermal delivery of the compound may be achieved by iontophoretic patches. Transdermal patches can provide controlled delivery of compounds, where absorption is slowed by the use of rate controlling membranes or by entrapment of the compound in a polymer matrix or gel. Absorption enhancers may be used to increase absorption, examples of which include absorbable pharmaceutically acceptable solvents that aid in passage through the skin.
Ophthalmic formulations include eye drops.
Formulations for rectal administration include enemas, rectal gels, rectal foams, rectal aerosols and retention enemas, which may contain conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG and the like. Compositions for rectal or vaginal administration may also be formulated as suppositories, which may be prepared by mixing the compounds with suitable non-irritating carriers and excipients such as cocoa butter, mixtures of fatty acid glycerides, polyethylene glycols, suppository waxes and combinations thereof, all of which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the compound.
Dosage of
As used herein, the term "therapeutically effective amount" refers to an amount of a compound of formula (I), or a pharmaceutically acceptable salt or stereoisomer thereof, effective to produce a desired therapeutic response in a particular patient suffering from a disease or disorder mediated by aberrant BCL6 activity. Thus, the term "therapeutically effective amount" includes a compound, or a pharmaceutically acceptable salt or stereoisomer thereof, that, when administered, induces a positive change in the disease or disorder to be treated, or is sufficient to prevent the development or progression of the disease or disorder, or to alleviate to some extent one or more symptoms of the disease or disorder being treated in a subject, or simply kills or inhibits the growth of diseased (e.g., cancer) cells, or reduces the amount of BCL6 in diseased cells.
The total daily dose of the compound and its use may be determined according to standard medical practice, for example, by the attending physician using sound medical judgment. The specific therapeutically effective dose of any particular subject may depend on a variety of factors, including the disease or disorder being treated and its severity (e.g., its status); age, body weight, general health, sex, and diet of the subject; the time of administration, route of administration and rate of excretion of the particular compound being used; duration of treatment; a medicament for use in combination or simultaneously with a compound; and similar factors known in the medical arts (see, e.g. ,Goodman and Gilman's,The Pharmacological Basis of Therapeutics,10th Edition,A.Gilman,J.Hardman and L.Limbird,eds.,McGraw-Hill Press,155-173,2001).
The compounds of formula (I) and pharmaceutically acceptable salts and stereoisomers thereof may be effective over a wide dosage range. In some embodiments, the total daily dose (e.g., for an adult) may range from about 0.001mg to about 1600mg, 0.01mg to about 500mg, about 0.01mg to about 100mg, about 0.5mg to about 100mg, about 1mg to about 100-400mg per day, about 1mg to about 50mg per day, about 5mg to about 40mg per day, or in other embodiments about 10mg to about 30mg per day. In some embodiments, the total daily dose may range from 400mg to 600mg. Depending on the number of times the compound is administered per day, a single dose containing the desired dose may be formulated. As an example, capsules may be formulated with about 1mg to about 200mg of a compound (e.g., 1mg, 2mg, 2.5mg, 3mg, 4mg, 5mg, 10mg, 15mg, 20mg, 25mg, 50mg, 100mg, 150mg, and 200 mg). In some embodiments, the compound may be administered at a dose in the range of about 0.001mg/kg to about 200mg/kg body weight per day. In some embodiments, a dose of 0.1 to 100, for example, one or more doses of 1mg/kg to 30mg/kg per day may be effective. As an example, a suitable dose for oral administration may be in the range of 1mg/kg-30mg/kg body weight per day, while a suitable dose for intravenous administration may be in the range of 1mg/kg-10mg/kg body weight per day.
In some embodiments, the compounds of formula (I) and pharmaceutically acceptable salts and stereoisomers thereof may be administered at dosage levels of from about 0.001mg/kg to about 50mg/kg, from about 0.01mg/kg to about 25mg/kg, or from about 0.1mg/kg to about 10mg/kg of subject body weight, one or more times per day, to achieve the desired therapeutic effect.
Application method
In certain aspects, the invention is directed to treating diseases or conditions characterized or mediated by aberrant (e.g., elevated BCL6 levels or other dysfunctions, e.g., dysfunctional BCL6 levels) BCL6 activity relative to non-pathological conditions. The method entails administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt or stereoisomer thereof. A "disease" is generally considered to be a state of health of a subject, wherein the subject is unable to maintain homeostasis, and wherein the subject's health continues to deteriorate if the disease is not improved. In contrast, a subject's "discomfort" is a health condition in which the subject is able to maintain homeostasis, but the subject's health condition is not as good as in the absence of the disorder. If not treated, the discomfort does not necessarily lead to a further decline in the health condition of the subject.
As used herein, the term "subject" (or "patient") includes all members of the kingdom animalia that are susceptible to or suffering from the disease or disorder. In some embodiments, the subject is a mammal, e.g., a human or non-human mammal. The method is also applicable to companion animals such as dogs and cats, as well as livestock such as cows, horses, sheep, goats, pigs and other domestic and wild animals. According to the present invention, a subject in need of "treatment may" have or be suspected of having "a particular disease or condition, may have been positively diagnosed or otherwise exhibited a sufficient number of risk factors or a sufficient number of signs or symptoms, or a combination thereof, such that a medical professional may diagnose or suspected the subject as having the disease or condition. Thus, a subject having a particular disease or disorder and a subject suspected of having a particular disease or disorder are not necessarily two distinct populations.
In some embodiments, the compounds of the invention are useful for treating cell proliferative diseases and disorders (e.g., cancer or benign tumors). As used herein, the term "cell proliferative disease or disorder" refers to a disease characterized by abnormal cell growth or both, which includes non-cancerous diseases such as tumors, pre-cancerous diseases, benign tumors, and cancers.
In some embodiments, the methods relate to treating a subject having cancer. Including adult tumors/cancers and pediatric tumors/cancers. The cancer may be vascularized, or a tumor that has not been substantially vascularized or non-vascularized.
In some embodiments, the methods of the invention relate to treating a subject suffering from a cell proliferative disease or disorder of the blood system.
As used herein, "cell proliferative disease or disorder of the blood system" includes lymphomas, leukemias, bone marrow tumors, mast cell tumors, myelodysplasias, benign monoclonal gammaglobuloses, lymphomatoid papuloses, polycythemia vera, chronic granulocytic leukemia, idiopathic myelometaplasia, and primary thrombocythemia. Representative examples of hematological cancers may thus include multiple myeloma, lymphoma (including T-cell lymphoma, hodgkin's lymphoma, non-Hodgkin's lymphoma (diffuse large B-cell lymphoma (DLBCL)), follicular Lymphoma (FL), mantle Cell Lymphoma (MCL), and ALK+ anaplastic large cell lymphoma (e.g., B-cell non-Hodgkin's lymphoma selected from diffuse large B-cell lymphoma (e.g., germinal center B-cell like diffuse large B-cell lymphoma or activated B-cell like diffuse large B-cell lymphoma), burkitt's lymphoma/leukemia, mantle cell lymphoma, mediastinal (thymus) large B-cell lymphoma, follicular lymphoma, marginal zone lymphoma, lymphoplasmacytic lymphoma/macroglobulinemia, metastatic pancreatic cancer, refractory B-cell non-Hodgkin's lymphoma, and recurrent B-cell non-Hodgkin's lymphoma, childhood lymphoma, and lymphomas of lymphocytic and cutaneous origin, for example, small lymphocytic lymphomas, leukemias including childhood leukemia, hairy cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia (e.g., acute monocytic leukemia), chronic lymphocytic leukemia, small lymphocytic leukemia, chronic myelogenous leukemia and mast cell leukemia, bone marrow tumors and mast cell tumors.
In some embodiments, the methods relate to treating a subject with malignant lymphoma.
In some embodiments, the malignant lymphoma is peripheral T-cell lymphoma (PTCL), diffuse large B-cell lymphoma (DLBCL), mantle Cell Lymphoma (MCL), follicular Lymphoma (FL), chronic Lymphocytic Leukemia (CLL), acute lymphoblastic leukemia/lymphoma (ALL), cutaneous T-cell lymphoma, chronic myeloid leukemia, or B-cell non-hodgkin's lymphoma.
In some embodiments, the cancer is melanoma, breast cancer, or non-small cell lung cancer.
The compounds of formula (I) may be administered to a patient, such as a cancer patient, as monotherapy or by combination therapy. The treatment may be "front/first-line", i.e., as an initial treatment of a patient who has not previously received an anti-cancer treatment regimen, whether alone or in combination with other treatments; or "two-wire", as a treatment of a patient who has previously received an anti-cancer treatment regimen, whether alone or in combination with other treatments; or as "three-wire", "four-wire" or the like, whether alone or in combination with other therapies. Treatment may also be performed on patients who have failed or partially failed past treatment but who have failed or are intolerant to the particular treatment. Treatment may also be administered as an adjunct therapy, i.e., to prevent cancer recurrence in patients who are not currently detected with disease or after surgical removal of the tumor. Thus, in some embodiments, the compound may be administered to a patient who has received another treatment, such as chemotherapy, radioimmunotherapy, surgical treatment, immunotherapy, radiation therapy, targeted therapy, or any combination thereof.
The methods of the invention may entail administering a compound of formula (I) or a pharmaceutical composition thereof to a patient in a single dose or in multiple doses (e.g., 1,2, 3, 4, 5, 6, 7, 8, 10, 15, 20, or more doses). For example, the frequency of administration may range from once per day to about once every eight weeks. In some embodiments, the frequency of administration ranges from about once a day for 1,2, 3, 4, 5, or 6 weeks, while in other embodiments, at least one 28 day period is required, including daily administration for 3 weeks (21 days), followed by a 7 day "off" period. In other embodiments, the compound may be administered twice daily (BID) for a two-and-a-half day course (total 5 doses) or once daily (QD) for a two-day course (total 2 doses). In other embodiments, the compound may be administered once daily (QD) over a five day course of treatment.
Combination therapy
The compounds of formula (I) and pharmaceutically acceptable salts or stereoisomers thereof may be used in combination or simultaneously with at least one other active agent, such as an anti-cancer agent or regimen to treat diseases and conditions. In this context, the terms "combination" and "simultaneously" refer to the administration of agents by the same or separate dosage forms, as well as by the same or different modes of administration, or sequentially, e.g., as part of the same therapeutic regimen, or by successive therapeutic regimens, which include administration substantially simultaneously. Thus, if administered sequentially, in some cases, at the beginning of administration of the second compound, the first of the two compounds will remain detectable at an effective concentration at the treatment site. The order and time intervals may be determined so that they may act together (e.g., synergistically) to provide a greater benefit than would otherwise be administered. For example, the therapeutic agents may be administered simultaneously or sequentially in any order at different time points; however, if not administered simultaneously, they may be administered in close enough time to provide the desired therapeutic effect, which may be in a synergistic manner. Thus, these terms are not limited to the administration of active agents at the same time.
In some embodiments, a treatment regimen may include administering a compound of formula (I) in combination with one or more other therapeutic agents known to be useful in treating a disease or disorder (e.g., cancer). The dosage of the other therapeutic agent may be the same as or even lower than the known or recommended dosage. See Hardman et al.,eds.,Goodman&Gilman's the Pharmacological Basis of Basis of Therapeutics,10th ed.,McGraw-Hill,New York,2001;Physician's Desk Reference 60th ed.,2006. for example, anticancer agents that may be suitable for use in combination with the compounds of the present invention are known in the art. See, for example, U.S. patent 9,101,622 (section 5.2 therein) and U.S. patent 9,345,705B2 (columns 12-18 therein). Representative examples of other anti-cancer agents and treatment regimens include radiation therapy, chemotherapy (e.g., mitotic inhibitors, angiogenesis inhibitors, anti-hormones, autophagy inhibitors, alkylating agents, intercalating antibiotics, growth factor inhibitors, anti-androgens, signal transduction pathway inhibitors, anti-microtubule agents, platinum coordination complexes, HDAC inhibitors, proteasome inhibitors, and topoisomerase inhibitors), immunomodulators, therapeutic antibodies (e.g., monospecific and bispecific antibodies), and CAR-T therapies.
In some embodiments, the compounds of formula (I) and other (e.g., anti-cancer) therapeutic agents can be administered less than 5 minutes apart, less than 30 minutes apart, less than 1 hour apart, about 1 hour to about 2 hours apart, about 2 hours to about 3 hours apart, about 3 hours to about 4 hours apart, about 4 hours to about 5 hours apart, about 5 hours to about 6 hours apart, about 6 hours to about 7 hours apart, about 7 hours to about 8 hours apart, about 8 hours to about 9 hours apart, about 9 hours to about 10 hours apart, about 10 hours to about 11 hours apart, about 11 hours to about 12 hours apart, about 12 hours to 18 hours apart, 18 hours to 24 hours apart, 24 hours to 36 hours apart, 36 hours to 48 hours, 48 hours to 52 hours apart, 52 hours to 60 hours apart, 60 hours to about 9 hours apart, about 9 hours to about 10 hours apart, about 10 hours to about 11 hours apart, about 12 hours to about 12 hours apart, about 12 hours to 18 hours apart, 18 hours to 24 hours apart, 24 hours to 36 hours apart, 36 hours to 48 hours to 52 hours apart, about 52 hours to 60 hours apart, about 60 hours to about 84 hours to 96 hours apart, or 96 hours to about 120 hours apart. Two or more (e.g., anti-cancer) therapeutic agents may be administered in the same visit of the patient.
When the active ingredients of the composition are not administered in the same pharmaceutical composition, it is understood that they may be administered to a subject in need thereof in any order. For example, a compound of the invention can be administered to a subject in need thereof prior to, simultaneously with, or after (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before) administration of the other therapeutic agent (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after). In various aspects, these therapeutic agents are administered 1 minute apart, 10 minutes apart, 30 minutes apart, less than 1 hour apart, 1 hour to 2 hours apart, 2 hours to 3 hours apart, 3 hours to 4 hours apart, 4 hours to 5 hours apart, 5 hours to 6 hours apart, 6 hours to 7 hours apart, 7 hours to 8 hours apart, 8 hours to 9 hours apart, 9 hours to 10 hours apart, 10 hours to 11 hours apart, 11 hours to 12 hours apart, no more than 24 hours apart, or no more than 48 hours apart. In another example, the (e.g., anti-cancer) therapeutic agent is administered in the same visit. In another example, the combination anti-cancer therapeutic may be administered between 1 minute and 24 hours apart.
In some embodiments involving cancer treatment, the compound of formula (I) and the other anti-cancer agent or therapeutic agent are administered cyclically. Cycling therapy involves the administration of one anti-cancer therapeutic agent over a period of time followed by the administration of a second anti-cancer therapeutic agent over a period of time and repeating such sequential administration, i.e., cycling, to reduce the occurrence of resistance to one or both anti-cancer therapeutic agents, to avoid or reduce side effects of one or both anti-cancer therapeutic agents, and/or to increase the efficacy of the treatment. In one example, the circulatory therapy comprises administering a first anti-cancer therapeutic agent over a period of time, then administering a second anti-cancer therapeutic agent over a period of time, optionally, then administering a third anti-cancer therapeutic agent over a period of time, and so forth, and repeating such sequential administration, i.e., circulatory, to reduce the occurrence of resistance to one anti-cancer therapeutic agent, to avoid or reduce side effects of one anti-cancer therapeutic agent, and/or to increase the efficacy of multiple anti-cancer therapeutic agents.
In some embodiments, the compounds of the invention may be used in combination with other anticancer agents, examples of which include etoposide (e.g., lymphomas and non-lymphoblastic leukemias), vincristine (e.g., leukemia), daunorubicin (e.g., acute Myelogenous Leukemia (AML), acute Lymphoblastic Leukemia (ALL), chronic Myelogenous Leukemia (CML) and kaposi's sarcoma), rituximab (e.g., non-hodgkin's lymphoma), alemtuzumab (e.g., chronic Lymphoblastic Leukemia (CLL), cutaneous T-cell lymphoma (CTCL) and T-cell lymphoma), bortezomib (e.g., multiple myeloma and mantle cell lymphoma), pekine (e.g., acute lymphoblastic leukemia), curida (e.g., hodgkin's lymphoma), and dexamethasone (e.g., acute multiple myeloma).
In some embodiments, the additional anticancer agent is an enhancer of zeste homolog 2 (EZH 2) inhibitors, examples of which include tazistat, GSK126, li Lamei tostat (CPI-1205), CPI-0209, PF-06821497, SHR2554, HH2853, valmotostat (DS 3201), MAK-683, and FTX-6058.
Pharmaceutical kit
The composition can be assembled into a kit or pharmaceutical system. According to this aspect of the invention, a kit or pharmaceutical system comprises a carrier or package, such as a box, carton, tube or the like, in which one or more containers, such as vials, tubes, ampoules or bottles, containing a compound of the invention or a pharmaceutical composition containing the compound and a pharmaceutically acceptable carrier are tightly enclosed, wherein the compound and carrier may be placed in the same or separate containers. Kits or pharmaceutical systems of the invention may also include printed instructions for using the compounds and compositions.
These and other aspects of the invention will be further appreciated upon consideration of the following examples, which are intended to illustrate certain specific embodiments of the invention, but are not intended to limit the scope thereof as defined by the claims.
Examples
Example 1: synthesis of 2- ((6- (5-cyano-4- ((3 s,5 r) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyrimidin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (1)
2, 4-Dimethyl-3-oxoglutarate dimethyl ester
K 2CO3 (39.68 g,287.11 mmol) was added to a solution of dimethyl 3-oxoglutarate (20 g,114.84 mmol) in THF (240 mL) at 20deg.C and the resulting mixture was stirred at 45deg.C for 20 min. CH 3 I (32.60 g,229.69 mmol) was then added and the reaction mixture was heated to 60℃and stirred for 100 minutes. The reaction mixture was cooled to 20 ℃, filtered and the filter cake was washed with THF (400 mL). The filtrate was dried in vacuo to give the title compound (40 g) as a crude yellow oil. 1H NMR(400MHz,CDCl3 ) Delta 3.80-3.71 (m, 6H), 1.44-1.34 (m, 6H).
1-Benzyl-3, 5-dimethyl-4-oxopiperidine-3, 5-dicarboxylic acid dimethyl ester
Aqueous HCl (1M, 21.76 mL), benzylamine (11.66 g,108.80 mmol) and formaldehyde (17.66 g,217.60mmol, 37% purity) were added to a solution of dimethyl 2, 4-dimethyl-3-oxoglutarate (22 g,108.80 mmol) in MeOH (400 mL) at 0deg.C. The reaction mixture was stirred at 15 ℃ for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=100/1 to 20/1) to give the title compound as a yellow oil (15g,41%).1H NMR(400MHz,CDCl3)δ7.37-7.27(m,5H),3.67(s,6H),3.65(s,2H),3.53(d,J=11.6Hz,2H),1.30(s,6H).
(3S, 5R) -1-benzyl-3, 5-dimethylpiperidin-4-one
A solution of 1-benzyl-3, 5-dimethyl-4-oxopiperidine-3, 5-dicarboxylic acid dimethyl ester (15 g,44.99 mmol) in aqueous hydrochloric acid (150 mL, 4M) was stirred at 100deg.C for 24 hours. The reaction mixture was concentrated under reduced pressure and pH was adjusted to 8 with 2M NaOH, then the mixture was extracted with ethyl acetate (200 ml x 3). The organic phases were combined, dried over anhydrous Na 2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=20/1 to 5/1) to give the title compound as a yellow oil (7.2g,74%).1H NMR(400MHz,CDCl3)δ7.33-7.27(m,5H),3.57(s,2H),3.12(dd,J=8.8,5.2Hz 2H),2.69(dd,J=10.8,5.6Hz 2H),2.05-1.99(m,2H),0.93(d,J=6.8Hz,6H).
(3S, 5R) -1-benzyl-4, 4-difluoro-3, 5-dimethylpiperidine
DAST (55.63 g,345.14 mmol) was added to a solution of (3S, 5R) -1-benzyl-3, 5-dimethylpiperidin-4-one (5 g,23.01 mmol) in DCM (50 mL) and the reaction mixture stirred at 50℃for 12 h. The pH was adjusted to 7-8 with saturated NaHCO 3 and extracted with ethyl acetate (500 mL x 3). The organic phases were combined and dried over anhydrous Na 2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=1/0 to 20/1) to give the title compound as a yellow oil (2.5g,45%).1H NMR(400MHz,CDCl3)δ7.28-7.21(m,5H),3.46(s,2H),2.71(d,J=10.8Hz,2H),2.09-2.03(m,2H),1.96-1.90(m,2H),0.93(d,J=6.8Hz,6H).
(3S, 5R) -4, 4-difluoro-3, 5-dimethylpiperidine
TFA (4.76 g,41.79 mmol) and Pd/C (0.8 g,10% purity) were added to a solution of (3S, 5R) -1-benzyl-4, 4-difluoro-3, 5-dimethylpiperidine (2.5 g,10.45 mmol) in MeOH (25 mL) under nitrogen and the reaction mixture was stirred at 60℃under H 2 (15 Psi) for 12H. The reaction mixture was filtered and concentrated under reduced pressure to give the title compound (2.3 g,84%, TFA salt) as a white solid ).1H NMR(400MHz,CDCl3)δ9.64-9.51(m,2H),3.34(d,J=12.4Hz,2H),2.88-2.80(m,2H),2.46-2.37(m,2H),1.12(d,J=6.8Hz,6H).
2-Chloro-4- ((3 s,5 r) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyrimidine-5-carbonitrile
A mixture of 2, 4-dichloropyrimidine-5-carbonitrile (300 mg,1.72 mmol), (3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidine (453.83 mg,1.72mmol, TFA salt) and DIEA (557.11 mg,4.31 mmol) in DMF (5 mL) was stirred at 100deg.C under N 2 for 1 hr. The reaction mixture was concentrated under reduced pressure to give a residue which was purified by reverse phase HPLC (neutral conditions: column: 80g Agela C18; mobile phase: [ water-ACN ]; B%:30-60%15min;60%5 min) to give 4-chloro-2- [ (3R, 5S) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] pyrimidine-5-carbonitrile (150 mg,30%, LCMS: [ M+H + ] = 287, retention time = 0.928 min) as a white solid and the title compound (60 mg,12%, LCMS: [ M+H+ ] = 287, retention time = 0.885 min) as a white solid.
2- ((6- (5-Cyano-4- ((3 s,5 r) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyrimidin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (1)
A mixture of 2- [ (6-amino-1-methyl-2-oxo-3-quinolinyl) oxy ] -N-methyl-acetamide (30 mg, 114.82. Mu. Mol), 2-chloro-4- [ (3R, 5S) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] pyrimidine-5-carbonitrile (36.21 mg, 126.30. Mu. Mol) and 4-methylbenzenesulfonic acid hydrate (26.21 mg, 137.79. Mu. Mol) in DMF (1 mL) was stirred at 130℃under N 2 for 12 hours. H 2 O (15 mL) was added to the reaction mixture, and the precipitated solid was filtered and washed with H 2 O (30 mL) and ethyl acetate (30 mL). The solid was dried under reduced pressure. Trituration of the solid with ethanol (5 mL) at 15℃for 30 min gave the title compound as a yellow solid (21.2mg,35%).1H NMR(400MHz,DMSO-d6)δ10.05(s,1H),8.49(s,1H),7.90-7.89(m,2H),7.68(dd,J=8.8,2.0Hz,1H),7.46(d,J=9.2Hz,1H),7.18(s,1H),4.62(d,J=13.6Hz,2H),4.56(s,2H),3.66(s,3H),2.93(t,J=12.8Hz,2H),2.66(d,J=4.8Hz,3H),2.33-2.27(m,2H),1.00(d,J=6.8Hz,6H).LCMSL:[M+H+]=512.2.
Example 2: synthesis of 2- ((6- (3-chloro-5-cyano-6- ((3 s,5 r) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (2)
2- ((6- ((3, 6-Dichloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
DIEA (197.86 mg,1.53 mmol) was added to a mixture of 2,5, 6-trichloropyridine-3-carbonitrile (158.79 mg, 765.47. Mu. Mol) and 2- [ (6-amino-1-methyl-2-oxo-3-quinolinyl) oxy ] -N-methyl-acetamide (200 mg, 765.47. Mu. Mol) in DMF (3 mL) and the reaction mixture was stirred at 100℃for 4h under N 2. The reaction mixture was cooled to 15 ℃, water (about 5 mL) was added and a precipitate formed. The reaction mixture was filtered and the filter cake was washed with EtOAc (about 20 mL) and concentrated in vacuo to give the title compound as a yellow solid (250mg,76%).1H NMR(400MHz,DMSO-d6)δ9.64(s,1H),8.39(s,1H),7.93(s,1H),7.73(s,1H),7.66(d,J=8.4Hz,1H),7.53(d,J=8.8Hz,1H),7.22(s,1H),4.58(s,2H),3.69(s,3H),2.67(s,3H).
2- ((6- (3-Chloro-5-cyano-6- ((3 s,5 r) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (2)
DIPEA (298.99 mg,2.31 mmol) was added to a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methyl-acetamide (100 mg, 231.34. Mu. Mol) and (3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidine (304.45 mg,1.16mmol, TFA salt) in DMSO (3 mL) and the reaction mixture was stirred at 130℃for 3 hours. The reaction mixture was then cooled to 15 ℃ and treated with water (8 mL) to form a precipitate. The mixture was filtered and washed with water (10 mL), etOAc (10 mL) and EtOH (20 mL), respectively. The filter cake was concentrated under reduced pressure to give the title compound as a pale brown solid (80.6mg,62%).1H NMR(400MHz,DMSO-d6)δ9.14(s,1H),7.99(s,1H),7.92(br s,1H),7.78(s,1H),7.63(br d,J=8.8Hz,1H),7.47(br d,J=9.2Hz,1H),7.23(s,1H),4.54(s,2H),4.13(br d,J=13.2Hz,2H),3.68(s,3H),2.78(br t,J=12.8Hz,2H),2.66(br d,J=4.4Hz,3H),2.15-1.98(m,2H),0.84(br d,J=6.8Hz,6H).
Example 3: synthesis of 2- ((6- ((5-cyano-4-4- ((3 s,5 r) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methylpiperidin-1-yl) pyrimidin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (3) and 2- ((6- ((5-cyano-4- ((3 r,5 s) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methylpiperidin-1-yl) pyrimidin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (4)
2- (1-Benzyl-5-methyl-4-oxo-3-piperidinyl) acetic acid methyl ester
LDA (2M, 73.80 mL) was added dropwise to a solution of 1-benzyl-3-methyl-4-piperidone (30 g,147.58 mmol) in THF (300 mL) at-78deg.C, and the reaction was stirred at-78deg.C for 30 min. Methyl 2-bromoacetate (33.86 g,221.38 mmol) and HMPA (31.74 g,177.10 mmol) were then added dropwise at-78℃and the reaction mixture stirred at-78℃for 2.5 hours. The reaction mixture was allowed to warm to 20 ℃ and stirred for an additional 9 hours. The reaction mixture was quenched with saturated NH 4 Cl (100 mL), diluted with H 2 O (500 mL), and then extracted with EtOAc (300 mL x 3). The combined organic layers were washed with brine (200 mL), dried over Na 2SO4, filtered and concentrated under reduced pressure to give a residue which was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=1/0 to 0/1) to give the title compound (8.3 g, 17%) as a yellow oil. LCMS: m+h + ] = 276.2.
2- (1-Benzyl-4, 4-difluoro-5-methyl-3-piperidinyl) acetic acid methyl ester
DAST (77.74 g,482.31 mmol) was added to a solution of methyl 2- (1-benzyl-5-methyl-4-oxo-3-piperidinyl) acetate (8.3 g,30.14 mmol) in DCM (85 mL) and the reaction mixture stirred at 50℃for 12 h. The reaction mixture was diluted with DCM (60 mL) and then saturated aqueous NaHCO 3 (200 mL) was added dropwise. The resulting mixture was stirred at 20 ℃ for 20min and then extracted with EtOAc (100 ml x 3). The combined organic layers were washed with brine (50 mL), dried over Na 2SO4, filtered and concentrated under reduced pressure to give a residue which was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=1/0 to 0/1) to give the title compound as a yellow oil (3.7g,41%).1H NMR(400MHz,CDCl3)δ7.31-7.21(m,5H),3.64-3.60(m,3H),3.57-3.54(m,1H),3.42(d,J=13.2Hz,1H),2.97-2.87(m,1H),2.78-2.66(m,2H),2.65-2.48(m,1H),2.25-2.05(m,2H),2.04-1.87(m,2H),0.92(d,J=6.8Hz,3H).
2- (1-Benzyl-4, 4-difluoro-5-methyl-3-piperidinyl) ethanol
LiAlH 4 (255.29 mg,6.73 mmol) was slowly added to a solution of methyl 2- (1-benzyl-4.4-difluoro-5-methyl-3-piperidinyl) acetate (1 g,3.36 mmol) in THF (10 mL) at 0deg.C and the reaction mixture was stirred at 20deg.C for 1 hr. The reaction mixture was quenched with saturated aqueous NH 4 Cl (100 mL), diluted with H 2 O (10 mL), and then extracted with EtOAc (10 mL x 3). The combined organic layers were washed with brine (10 mL), dried over Na 2SO4, filtered and concentrated under reduced pressure to give the title compound (700 mg, 77%) as a yellow oil. LCMS: [ m+h + ] =270.1.
2- (4, 4-Difluoro-5-methylpiperidin-3-yl) ethanol
TFA (3.39 g,29.70 mmol) and Pd/C (0.5 g,10% purity) were added to a solution of 2- (1-benzyl-4, 4-difluoro-5-methyl-3-piperidinyl) ethanol (2 g,7.43 mmol) in MeOH (20 mL) and the reaction mixture stirred at 60℃under H 2 (15 Psi) for 6H. The reaction mixture was filtered and concentrated under reduced pressure to give the title compound (2 g,92%, TFA salt) as a white solid ).1H NMR(400MHz,methanol-d4)δ3.66(d,J=6.4Hz,2H),3.62-3.60(m,1H),3.45-3.39(m,1H),2.97-2.88(m,2H),2.51-2.26(m,2H),2.11-2.03(m,1H),1.53-1.44(m,1H),1.11(d,J=6.8Hz,3H).
2-Chloro-4- (4, 4-difluoro-3- (2-hydroxyethyl) -5-methylpiperidin-1-yl) pyrimidine-5-carbonitrile
A mixture of 2- (4, 4-difluoro-5-methyl-3-piperidinyl) ethanol (1 g,3.41mmol, TFA salt), 2, 4-dichloropyrimidine-5-carbonitrile (593.35 mg,3.41 mmol) and DIEA (1.10 g,8.53 mmol) in DMF (10 mL) was stirred at 50℃under N 2 atmosphere for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by reverse phase HPLC (neutral condition: column: 80g Agela C18; mobile phase: [ water-ACN ];B%:35-65%25min;65%5 min) to give the title compound (120 mg, 11%) as a yellow solid. LCMS: m+h + ] = 317.0.
2-Chloro-4- ((3 s,5 r) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methylpiperidin-1-yl) pyrimidine-5-carbonitrile and 2-chloro-4- ((3 r,5 s) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methylpiperidin-1-yl) pyrimidine-5-carbonitrile
2-Chloro-4- [4, 4-difluoro-3- (2-hydroxyethyl) -5-methyl-1-piperidinyl ] pyrimidine-5-carbonitrile (170 mg, 536.73. Mu. Mol) was purified by SFC (column: DAICEL)OJ (250 mm. Times.30 mm,10 μm); mobile phase: [ Neu-MeOH ]; b%:10% -40%,15 min) to give 2-chloro-4- [ (3 s,5 r) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methyl-1-piperidinyl ] pyrimidine-5-carbonitrile (70 mg,41%, retention time = 1.894 min) as a yellow solid and 2-chloro-4- [ (3 r,5 s) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methyl-1-piperidinyl) pyrimidine-5-carbonitrile (70 mg,40%, retention time = 2.013 min) as a yellow solid.
2- ((6- (5-Cyano-4- ((3 s,5 r) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methylpiperidin-1-yl) pyrimidin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (3)
A mixture of 2-chloro-4- [ (3S, 5R) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methyl-1-piperidinyl ] pyrimidine-5-carbonitrile (70 mg, 221.01. Mu. Mol), 2- [ (6-amino-1-methyl-2-oxo-3-quinolinyl) oxy ] -N-methylacetamide (52.49 mg, 200.91. Mu. Mol) and 4-toluenesulfonic acid monohydrate (42.04 mg, 221.01. Mu. Mol) in DMF (1 mL) was stirred at 130℃under N 2 for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue which was purified by preparative HPLC (column: waters TM Xbridge BEH C18.times.30 mm.times.10 μm; mobile phase: [ water (10 mM NH 4HCO3) -ACN ];B%:25% -55%,8 min) to give the title compound as a white solid (38mg,34%).1H NMR(400MHz,DMSO-d6)δ10.06(s,1H),8.49(s,1H),7.93(d,J=4.4Hz,1H),7.85(s,1H),7.71(d,J=9.2Hz,1H),7.46(d,J=9.2Hz,1H),7.20(s,1H),4.83(d,J=13.2Hz,1H),4.62(t,J=5.2Hz,2H),4.56(s,2H),3.66(s,3H),3.52-3.47(m,2H),2.96-2.87(m,2H),2.66(d,J=4.8Hz,3H),2.21-2.14(m,2H),1.91-1.83(m,1H),1.42-1.33(m,1H),1.00(d,J=6.8Hz,3H).LCMS:[M+H+]=542.2.
2- ((6- (5-Cyano-4- ((3 r,5 s) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methylpiperidin-1-yl) pyrimidin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (4)
A mixture of 2-chloro-4- [ (3R, 5S) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methyl-1-piperidinyl ] pyrimidine-5-carbonitrile (70 mg, 221.01. Mu. Mol), 2- [ (6-amino-1-methyl-2-oxo-3-quinolinyl) oxy ] -N-methyl-acetamide (52.49 mg, 200.91. Mu. Mol) and 4-toluenesulfonic acid monohydrate (45.86 mg, 241.10. Mu. Mol) in DMF (2 mL) was stirred at 130℃under N 2 atmosphere for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue which was purified by preparative HPLC (column: waters TM Xbridge BEH C18.times.30 mm.times.10 μm; mobile phase: [ water (10 mM NH 4HCO3) -ACN ];B%:25% -55%,8 min) to give the title compound as a white solid (16mg,14%).1H NMR(400MHz,DMSO-d6)δ10.07(s,1H),8.49(s,1H),7.94-7.86(m,2H),7.71(d,J=9.2Hz,1H),7.46(d,J=8.8Hz,1H),7.20(s,1H),4.83(d,J=11.6Hz,1H),4.62(t,J=5.2Hz,2H),4.56(s,2H),3.67(s,3H),3.50-3.49(m,2H),2.96-2.87(m,2H),2.66(d,J=4.4Hz,3H),2.20(s,2H),1.86(s,1H),1.37(s,1H),1.00(d,J=6.8Hz,3H).LCMS:[M+H+]=542.2.
Example 4: synthesis of 2- ((6- ((4- (3- (2-aminoethyl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-cyanopyrimidin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (mixture of 14 and 15)
2- [2- (1-Benzyl-4, 4-difluoro-5-methyl-3-piperidinyl) ethyl ] isoindoline-1, 3-dione
PPh 3 (886.20 mg,3.38 mmol) was added to a solution of 2- (1-benzyl-4, 4-difluoro-5-methyl-3-piperidinyl) ethanol (700 mg,2.60 mmol) and isoindole-1, 3-dione (458.88 mg,3.12 mmol) in DCM (7 mL) at 0deg.C. A solution of DEAD (588.43 mg,3.38 mmol) in DCM (1 mL) was then added dropwise at 0deg.C. The reaction mixture was then stirred at 20 ℃ for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=1/0 to 0/1) to give the title compound (600 mg, 58%) as a colorless oil. LCMS: m+h + ] = 399.1.
2-Chloro-4- (3- (2- (1, 3-dioxoisoindolin-2-yl) ethyl) -4, 4-difluoro-5-methylpiperidin-1-yl) pyrimidine-5-carbonitrile
A mixture of 2- [2- (4, 4-difluoro-5-methyl-3-piperidinyl) isoindoline-1, 3-dione (1 g,2.37mmol, TFA salt), 2, 4-dichloropyrimidine-5-carbonitrile (494.35 mg,3.41 mmol) and DIEA (612.03 mg,4.74 mmol) in DMF (10 mL) was stirred at 100deg.C under N 2 for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue which was purified by reverse phase HPLC (neutral conditions: column: 120g Agela C18; mobile phase: [ water-ACN ]; gradient B%:30-60%20min;60%10 min) to give the title compound (200 mg, 19%) as a yellow solid. LCMS: [ m+h + ] =446.1.
2- ((6- ((5-Cyano-4- (3- (2- (1, 3-dioxoisoindolin-2-yl) ethyl) -4, 4-difluoro-5-methylpiperidin-1-yl) pyrimidin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
A mixture of 2-chloro-4- [3- [2- (1, 3-dioxoisoindolin-2-yl) ethyl ] -4, 4-difluoro-5-methyl-1-piperidinyl ] pyrimidine-5-carbonitrile (200 mg, 448.58. Mu. Mol), 2- [ (6-amino-1-methyl-2-oxo-3-quinolinyl) oxy ] -N-methylacetamide (106.55 mg, 407.80. Mu. Mol) and DIEA (105.41 mg, 815.60. Mu. Mol) in NMP (3 mL) was stirred at 130℃for 2 hours under an atmosphere of N 2. H 2 O (5 mL) was added to the reaction mixture, and the precipitated solid was filtered and washed with H 2 O (5 mL) and EtOAc (5 mL). The solid was dried under reduced pressure to give the title compound as a brown solid (200mg,64%).1H NMR(400MHz,DMSO-d6)δ10.06(s,1H),8.50(s,1H),7.87-7.82(m,5H),7.75-7.66(m,2H),7.46(d,J=9.2Hz,1H),7.17(s,1H),4.83(d,J=12.8Hz,1H),4.6(d,J=12.4Hz,1H),4.52(s,2H),3.60(s,3H),3.30-3.28(m,2H),2.69(s,3H),2.65(d,J=4.4Hz,2H),2.00-1.86(m,3H),1.57-1.48(m,1H),0.98(d,J=6.4Hz,3H).
2- ((6- ((4- (3- (2-Aminoethyl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-cyanopyrimidin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (mixture of 14 and 15)
NH 2NH2.H2 O (279.91 mg,5.59 mmol) was added to a mixture of 2- [ [6- [ [ 5-cyano-4- [3- [2- (1, 3-dioxoisoindolin-2-yl) ethyl ] -4, 4-difluoro-5-methyl-1-piperidinyl ] pyrimidin-2-yl ] amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methyl-acetamide (250 mg, 372.76. Mu. Mol) in NMP (0.5 mL) and the reaction was heated to 100deg.C and stirred for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by preparative HPLC (column: phenomnex)80 X 30mm x 3 μm; mobile phase: [ water (TFA) -ACN ]; b%:10% -40%,8 min) to give the title compound as a yellow solid (25mg,12%).1H NMR(400MHz,DMSO-d6)δ10.12(s,1H),8.52(s,1H),7.95(d,J=4.8Hz,1H),7.88(s,1H),7.74-7.69(m,3H),7.47(d,J=9.2Hz,1H),7.18(s,1H),4.72-4.64(m,2H),4.57(s,2H),3.67(s,3H),3.02(d,J=12.8Hz,1H),2.91-2.84(m,3H),2.66(d,J=4.4Hz,3H),2.25-2.15(m,2H),2.03-1.94(m,1H),1.59-1.49(m,1H),1.01(d,J=6.8Hz,3H).
Example 5: synthesis of 5-chloro-2- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-3- ((3-methyloxetan-3-yl) methoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) nicotinonitrile (9)
3-Hydroxy-1-methyl-6-nitroquinolin-2 (1H) -one
Trimethylsilanized diazomethane (2M, 29.10 mL) is added dropwise to a mixture of 1-methyl-5-nitroindoline-2, 3-dione (10 g,48.51 mmol) and TEA (9.82 g,97.01 mmol) in EtOH (300 mL) and the reaction mixture is stirred at 20deg.C for 12 h. The reaction mixture was concentrated under reduced pressure, 1N HCl (100 mL) was added and the mixture was stirred at 20℃for 2 hours. The mixture was filtered and washed with DMF/ethyl acetate (1/10, 50 mL) and the filter cake was dried under reduced pressure to give the title compound as a yellow solid (2g,19%).1H NMR(400MHz,DMSO-d6)δ10.09(s,1H),8.66(d,J=2.4Hz,1H),8.19(dd,J=9.2,2.4Hz,1H),7.65(d,J=9.2Hz,1H),7.34(s,1H),3.74(s,3H).
1-Methyl-3- ((3-methyloxetan-3-yl) methoxy) -6-nitroquinolin-2 (1H) -one
A mixture of 3-hydroxy-1-methyl-6-nitro-quinolin-2-one (500 mg,2.27 mmol), 3-iodomethyl-3-methyloxetane (577.78 mg,2.73 mmol) and Cs 2CO3 (1.48 g,4.54 mmol) in DMF (5 mL) was stirred at 60℃under N 2 for 12 hours. H 2 O (5 mL) was added and the precipitated solid was filtered and washed with H 2 O (10 mL) and EtOAc (5 mL). The solid was dried under reduced pressure to give the title compound as a brown solid (350mg,51%).1H NMR(400MHz,DMSO-d6)δ8.57(d,J=2.4Hz,1H),8.24(dd,J=9.2,2.4Hz,1H),7.66(d,J=9.2Hz,1H),7.65(s,1H),4.50(d,J=6.0Hz,2H),4.34(d,J=6.0Hz,2H),4.13(s,2H),3.70(s,3H),1.40(s,3H).
6-Amino-1-methyl-3- ((3-methyloxetan-3-yl) methoxy) quinolin-2 (1H) -one
Pd/C (10 mg, purity 10%) was added to a solution of 1-methyl-3- [ (3-methyloxetan-3-yl) methoxy ] -6-nitro-quinolin-2-one (350 mg,1.15 mmol) in DMF (0.5 mL) under an atmosphere of N 2. The reaction mixture was stirred at 20℃under H 2 (15 Psi) for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give the title compound as a brown solid (250mg,79%).1H NMR(400MHz,DMSO-d6)δ7.18(d,J=9.2Hz,1H),7.10(s,1H),6.78(dd,J=8.8,2.4Hz,1H),6.72(d,J=2.4Hz,1H),5.03(s,2H),4.49(d,J=5.6Hz,2H),4.31(d,J=6.0Hz,2H),4.06(s,2H),3.56(s,3H),1.38(s,3H).
2, 5-Dichloro-6- ((1-methyl-3- ((3-methyloxetan-3-yl) methoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) nicotinonitrile
A mixture of 6-amino-1-methyl-3- [ (3-methyloxetan-3-yl) methoxy ] quinolin-2-one (250 mg, 911.36. Mu. Mol), 2,5, 6-trichloropyridine-3-carbonitrile (207.96 mg,1.00 mmol) and DIEA (235.57 mg,1.82 mmol) in DMF (3 mL) was stirred at 100deg.C under an atmosphere of N 2 for 4 hours. H 2 O (3 mL) was added and the precipitated solid was filtered and washed with H 2 O (3 mL) and EtOAc (5 mL). The solid was dried under reduced pressure to give the title compound as a brown solid (300mg,74%).1H NMR(400MHz,DMSO-d6)δ9.42(s,1H),8.29(s,1H),7.78(s,1H),7.65(d,J=8.8Hz,1H),7.49(d,J=9.2Hz,1H),7.27(s,1H),4.55(d,J=5.6Hz,2H),4.33(d,J=6.0Hz,2H),4.16(s,2H),3.68(s,3H),1.42(s,3H).
5-Chloro-2- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-3- ((3-methyloxetan-3-yl) methoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) nicotinonitrile (9)
A mixture of 2, 5-dichloro-6- [ [ 1-methyl-3- [ (3-methyloxetan-3-yl) methoxy ] -2-oxo-6-quinolinyl ] amino ] pyridine-3-carbonitrile (100 mg,224.57 mmol), (3S, 5R) -4, 4-difluoro-3, 5-dimethylpiperidine (76.84 mg,291.94mmol, TFA salt) and DIEA (72.56 mg, 561.42. Mu. Mol) in DMSO (1 mL) was stirred at 130℃under an atmosphere of N 2 for 3 hours. The reaction mixture was concentrated under reduced pressure to give a residue which was purified by preparative HPLC (column: phenomnex C1880. Times.40 mm. Times.3 μm; mobile phase: [ water (NH 4HCO3) -ACN ];: B%:35% -65%,8 min) to give the title compound as a yellow solid (40mg).1H NMR(400MHz,DMSO-d6)δ9.16(s,1H),8.0(s,1H),7.84(d,J=2.4Hz,1H),7.59(dd,J=9.2,2.4Hz,1H),7.46(d,J=9.2Hz,1H),7.29(s,1H),4.48(d,J=6.0Hz,2H),4.33(d,J=6.0Hz,2H),4.13(d,J=12.8Hz,2H),4.09(s,2H),3.66(s,3H),2.78(t,J=12.8Hz,2H),2.11-2.04(m,2H),1.39(s,3H),0.85(t,J=6.8Hz,6H).
Example 6: synthesis of 2- ((3S, 5R) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (48) and 2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (49)
3- (1, 3-Dioxoisoindolin-2-yl) -4-oxopiperidine-1-carboxylic acid tert-butyl ester
A mixture of tert-butyl 3-bromo-4-oxopiperidine-1-carboxylate (100 g,359.53 mmol) and potassium (1, 3-dioxoisoindolin-2-yl) in DMF (800 mL) was stirred at 20℃for 2hr. The mixture was slowly added to water (2.5L) to form a precipitate, which was filtered and the filter cake was washed with water (500 mL) and dried in vacuo to give the title compound as a white solid (80 g, 59% yield, purity 91%).1H NMR(400MHz,DMSO-d6)δ=7.97-7.82(m,4H),4.88(dd,J=7.2,11.6Hz,1H),4.41-4.11(m,2H),3.76-3.51(m,1H),3.29-3.16(m,1H),2.85-2.71(m,1H),2.47(t,J=3.0Hz,1H),1.47-1.43(s,9H).
3- (1, 3-Dioxoisoindolin-2-yl) -5-methyl-4-oxopiperidine-1-carboxylic acid tert-butyl ester
To a solution of 3- (1, 3-dioxoisoindolin-2-yl) -4-oxo-piperidine-1-carboxylic acid tert-butyl ester (25 g,72.60 mmol) in THF (250 mL) was added LDA (2M, 43.56 mL) dropwise at-70℃and the mixture was stirred at-70℃for 45min, then HMPA (15.61 g,87.12mmol,15.31 mL) was added dropwise and stirred for a further 30min. Finally, methyl iodide (15.46 g,108.90mmol,6.78 mL) was added dropwise at-70℃and the resulting mixture was stirred at 20℃for 11hr. The reaction mixture was quenched by addition of saturated aqueous NH 4 Cl (400 mL) and then extracted with DCM (500 mL x 3). The combined organic layers were dried over Na 2SO4, filtered and concentrated under reduced pressure to give a residue which was purified by flash chromatography (SILICA FLASH column, 25% -50% ethyl acetate/petroleum ether gradient) to give the title compound as a pink solid (4.3 g, yield 7%, purity 87%).1HNMR(400MHz,CDCl3)δ=7.90-7.83(m,2H),7.77–7.74(m,2H),4.86(dd,J=7.2,11.2Hz,1H),4.65-4.29(m,2H),3.97-3.81(m,1H),2.90(s,1H),2.67(s,1H),1.52(s,9H),1.13(d,J=6.8Hz,3H).
2- (5-Methyl-4-oxopiperidin-3-yl) isoindoline-1, 3-dione
A mixture of 3- (1, 3-dioxoisoindolin-2-yl) -5-methyl-4-oxopiperidine-1-carboxylic acid tert-butyl ester (4.3 g,12.00 mmol) in HCl/EtOAc (4M, 21.50 mL) was stirred at 20deg.C for 1hr. The reaction mixture was concentrated under reduced pressure to give the title compound (3.5 g, crude, HCl) as a white solid.
2- (1-Benzyl-5-methyl-4-oxopiperidin-3-yl) isoindoline-1, 3-dione
A mixture of 2- (5-methyl-4-oxo-3-piperidinyl) isoindoline-1, 3-dione (3.5 g,9.50mmol,80% purity, HCl), benzaldehyde (1.21 g,11.40mmol,1.15 mL) and AcOH (285.25 mg,4.75mmol, 271.67. Mu.L) in DMF (35 mL) was stirred at 20℃for 2hr, then sodium triacetoxyborohydride (4.03 g,19.00mmol,2 eq.) was added to the mixture, and the reaction mixture was stirred at 20℃for 10hr. The reaction mixture was treated with water (50 mL), a precipitate formed, which was filtered, and the solid was collected and dried in vacuo. Trituration of the crude product at 20deg.C with (PE: etOAc=15:1, 60 mL) for 30min afforded the title compound as a white solid (3 g, 75% yield, purity 83%).1H NMR(400MHz,CDCl3)δ=7.76-7.70(m,2H),7.65-7.59(m,2H),7.29-7.16(m,5H),5.01-4.92(m,1H),3.69-3.54(m,2H),3.21-3.04(m,3H),2.76-2.64(m,1H),2.34-2.26(m,1H),0.98(d,J=6.8Hz,3H).
2- (1-Benzyl-4, 4-difluoro-5-methylpiperidin-3-yl) isoindoline-1, 3-dione
To a solution of 2- (1-benzyl-5-methyl-4-oxo-3-piperidinyl) isoindoline-1, 3-dione (3 g,8.61 mmol) in DCM (30 mL) was added DAST (20.82 g,129.16mmol,17.07 mL) in one portion, and the reaction mixture was then stirred at 50℃for 12h. The reaction mixture was diluted with DCM (50 mL) and then saturated aqueous NaHCO 3 (200 mL) was added to the stirred mixture. The organic phase was separated and the aqueous phase was extracted with EtOAc (100 ml x 3). The combined organic phases were washed with brine (150 mL), dried over anhydrous Na 2SO4, filtered and the filtrate concentrated in vacuo and purified by flash chromatography on silica gel (SILICA FLASH column, 5% -30% ethyl acetate/petroleum ether gradient) to give the title compound (2 g,3.78mmol, 44% purity) as a white solid 70%).1H NMR(400MHz,CDCl3)δ=7.81-7.74(m,2H),7.69-7.62(m,2H),7.27-7.15(m,5H),4.71-4.55(m,1H),3.67-3.59(m,1H),3.55-3.46(m,2H),2.85-2.76(m,2H),2.35-2.15(m,2H),0.97(d,J=6.4Hz,3H).
2- (4, 4-Difluoro-5-methylpiperidin-3-yl) isoindoline-1, 3-dione
To a mixture of 2- (1-benzyl-4, 4-difluoro-5-methyl-3-piperidinyl) isoindoline-1, 3-dione (2 g,5.40 mmol) in H 2 O (20 mL) and ACN (80 mL) was added CAN (29.60 g,54.00mmol,26.91 mL) and the reaction mixture was stirred at 40℃for 12hr. The reaction mixture was treated with saturated aqueous K 2CO3 (50 mL) to form a precipitate, which was filtered and the filter cake was washed with EtOAc (80 mL). The filtrate was extracted with EtOAc (100 mL x 3), the combined organic phases were washed with brine (150 mL), dried over anhydrous Na 2SO4, filtered, and the filtrate concentrated in vacuo. The residue was purified by flash chromatography on silica gel (SILICA FLASH column, 80% -100% ethyl acetate/petroleum ether gradient). Further purification was then performed by preparative HPLC (column: waters Xbridge Prep OBD C18:150:40 mm:10 μm; mobile phase: [ water (NH 3H2O+NH4HCO3) -ACN ]; B%:20% -50%,8 min) to give the title compound (550 mg,1.95mmol, 36% yield, 99% purity) as a yellow solid ).1H NMR(400MHz,DMSO-d6)δ=7.94-7.83(m,4H),4.50-4.34(m,1H),3.72(t,J=12.4Hz,1H),3.15-3.03(m,1H),2.98-2.88(m,1H),2.54(s,1H),2.41-2.31(m,1H),2.22-2.01(m,1H),0.90(d,J=6.4Hz,3H).
2- ((3 S,5 r) -4, 4-difluoro-5-methylpiperidin-3-yl) isoindoline-1, 3-dione and 2- ((3 r,5 s) -4, 4-difluoro-5-methylpiperidin-3-yl) isoindoline-1, 3-dione
2- (4, 4-Difluoro-5-methyl-3-piperidinyl) isoindoline-1, 3-dione (550 mg) was isolated by SFC (column: DAICEL CHIRALPAK IG (250 mm. Times.50 mm,10 μm); mobile phase: [0.1% NH 3H2 O ETOH ]; B%:40% -40%,3.8 min) to give 2- [ (3S, 5R) -4, 4-difluoro-5-methyl-3-piperidinyl ] isoindoline-1, 3-dione (200 mg, 520.93. Mu. Mol, yield 27%) and 2- [ (3R, 5S) -4, 4-difluoro-5-methyl-3-piperidinyl ] isoindoline-1, 3-dione (190 mg, 460.99. Mu. Mol, yield 23%) as white solids.
Intermediate products 1:1H NMR(400MHz,DMSO-d6)δ=7.95-7.81(m,4H),4.52-4.32(m,1H),3.73(t,J=12.4Hz,1H),3.14-3.03(m,1H),2.96–2.90(m,1H),2.58(s,1H),2.36(t,J=12.6Hz,1H),2.20-2.03(m,1H),0.90(d,J=6.8Hz,3H).
Intermediate products 2:1H NMR(400MHz,DMSO-d6)δ=7.93-7.82(m,4H),4.55-4.30(m,1H),3.73(t,J=12.4Hz,1H),3.14-3.04(m,1H),2.96–2.91(m,1H),2.70(s,1H),2.36(t,J=12.0Hz,1H),2.21-2.02(m,1H),0.91(d,J=6.8Hz,3H).
3-Hydroxy-3-methylbutyl 4-methylbenzenesulfonate
DMAP (5.87 g,48.01 mmol) and TEA (97.16 g,960.18mmol, 133.64 mL) were added to a mixture of 3-methylbutane-1, 3-diol (50 g,480.09mmol,51.23 mL) in DCM (1L) at 15℃under nitrogen. 4-Methylbenzenesulfonyl chloride (91.53 g,480.09 mmol) was slowly added at 0deg.C. The reaction was stirred at 15℃for 12 hours and then poured into saturated aqueous NaHCO 3 (1000 mL). The biphasic layer was separated and the aqueous layer was extracted with DCM (2000 mL). The combined organic layers were washed with brine (1500 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica gel, eluting with petroleum ether/ethyl acetate=10/1 to 2/1) to give the title compound (250 g,953.23mmol, yield 99%, purity 98%) as a colorless oil, which was used without further purification .1H NMR(400MHz,CDCl3)δppm 7.75-7.80(m,2H),7.34(d,J=7.99Hz,2H),4.19(t,J=6.91Hz,2H),2.44(s,3H),1.84(t,J=6.85Hz,2H),1.68(s,1H),1.20(s,6H).
1-Methyl-5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one
A mixture of N-1-methyl-4-nitrobenzene-1, 2-diamine (50 g,299.11 mmol) and bis (imidazol-1-yl) methanone (48.50 g,299.11 mmol) in DMF (500 mL) was stirred at 15℃for 12 hours. The resulting suspension was filtered and the filter cake was washed with EtOAc (100 mL) and concentrated under reduced pressure to give the crude title compound (92 g,468.15mmol, yield 78%, purity 98%) as a red solid, which was used without further purification .1H NMR(400MHz,DMSO-d6)δ11.38(br s,1H),7.99(dd,J=8.70,2.15Hz,1H),7.71(d,J=2.03Hz,1H),7.26(d,J=8.70Hz,1H),3.34(s,3H).
3- (3-Hydroxy-3-methylbutyl) -1-methyl-5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one
Cs 2CO3 (50.60 g,155.31 mmol) was added in one portion to a mixture of 3-hydroxy-3-methylbutyl 4-methylbenzenesulfonate (24.07 g,93.19 mmol) and 1-methyl-5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one (15 g,77.66 mmol) in DMSO (400 mL) under nitrogen. The mixture was stirred at 100℃for 12 hours. (3-hydroxy-3-methyl-butyl) 4-methylbenzenesulfonate (6.02 g,23.30 mmol) was added. The mixture was stirred at 100℃for 12 hours. Water (600 mL) was then added and the biphasic mixture extracted with ethyl acetate (500 mL x 2). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated to give the crude title compound (46 g,154.92mmol, 99% yield, 94% purity) as a black oil, which was used without further purification .1H NMR(400MHz,DMSO-d6)δ8.02(dd,J=8.70,2.15Hz,1H),7.96(d,J=2.15Hz,1H),7.32(d,J=8.70Hz,1H),4.52(s,1H),3.91-4.00(m,2H),3.38(s,3H),1.67-1.75(m,2H),1.16(s,6H).M+H+=280.0.
5-Amino-3- (3-hydroxy-3-methylbutyl) -1-methyl-1H-benzo [ d ] imidazol-2 (3H) -one
Pd/C (10 g, 10%) was added to a solution of 3- (3-hydroxy-3-methylbutyl) -1-methyl-5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one (15 g,53.71 mmol) in DMF (200 mL) under argon. The suspension was degassed under vacuum and purged several times with H 2. The mixture was stirred at H 2 (50 psi) and 50℃for 96 hours. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (silica gel, eluting with DCM: meoh=20:1 to 5:1) to give the title compound as a pink solid (13 g,51.44mmol, 96% yield, purity 99%).1H NMR(400MHz,DMSO-d6)δ6.78(d,J=8.31Hz,1H),6.37(d,J=1.83Hz,1H),6.30(dd,J=8.25,1.90Hz,1H),4.79(s,2H),4.45(s,1H),3.73-3.83(m,2H),3.21(s,3H),1.59-1.72(m,2H),1.16(s,6H).M+H+=250.1.
2, 5-Dichloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
DIEA (3.11 g,24.07mmol,4.19 mL) was added in one portion to a mixture of 5-amino-3- (3-hydroxy-3-methyl-butyl) -1-methylbenzimidazol-2-one (3 g,12.03 mmol) and 2,5, 6-trichloropyridine-3-carbonitrile (2.50 g,12.03 mmol) in DMSO (30 mL) under nitrogen. The mixture was stirred at 100℃for 12 hours. To the mixture was added water (20 mL), the precipitated solid was filtered, and washed with H 2 O (30 mL) and ethyl acetate (20 mL), and then the solid was dried under reduced pressure to give the title compound (4 g,6.53mmol, yield 54% purity as a black solid) 69%).1H NMR(400MHz,DMSO-d6)δppm 9.50(s,1H),8.28(s,1H),7.35(m,1H),7.20(dd,J=8.44,1.59Hz,1H),7.11(m,1H),4.44(s,1H),3.87(m,2H),2.54(s,3H),1.72(m,2H),1.17(s,6H).
5-Chloro-2- (3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
DIEA (922.52 mg,7.14mmol,1.24 mL) was added in one portion to a mixture of 2, 5-dichloro-6- [ [3- (3-hydroxy-3-methyl-butyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (1.5 g,3.57 mmol) and 2- (4, 4-difluoro-5-methyl-3-piperidinyl) isoindoline-1, 3-dione (1.10 g,3.93 mmol) in DMSO (15 mL) under N 2. The mixture was stirred at 100℃for 12 hours. The mixture was cooled to 15 ℃ and poured into water (30 mL). The aqueous phase was extracted with ethyl acetate (20 mL x 3). The combined organic phases were washed with brine (50 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo to give the title compound as a yellow solid (2.25 g,2.35mmol, 66% yield, purity) 69%).1H NMR(400MHz,DMSO-d6)δ9.13(s,1H),8.01(s,1H),7.45-7.95(m,4H),7.11-7.20(m,1H),7.00-7.08(m,1H),6.81(d,J=8.34Hz,1H),4.12-4.54(m,4H),3.83-3.93(m,1H),3.58-3.77(m,1H),3.07-3.25(m,1H),2.82(s,2H),2.54(s,3H),2.08-2.28(m,1H),1.67-1.76(m,1H),1.05-1.21(m,6H),0.80-1.02(m,3H).
2- (3-Amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a mixture of 5-chloro-2- [3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methyl-1-piperidinyl ] -6- [ [3- (3-hydroxy-3-methyl-butyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (2.25 g,3.39 mmol) in EtOH (25 mL) was added N 2H4.H2 O (2.99 g,50.82mmol,2.91mL, 85% purity) in one portion under N 2. The mixture was stirred at 60℃for 1 hour. The reaction mixture was concentrated under reduced pressure. The residue was purified by preparative HPLC (neutral condition: column: welch Xtimate C18:250:70 mm#10μm; mobile phase: [ water (NH 4HCO 3) -ACN ];% B: 25-60%,20 min) to give the title compound (1.5 g, yield 83%, purity) as a yellow solid 88%).1H NMR(400MHz,DMSO-d6)δ9.13(s,1H),8.01(s,1H),7.45-7.95(m,4H),7.11-7.20(m,1H),7.00-7.08(m,1H),6.81(d,J=8.34Hz,1H),4.12-4.54(m,4H),3.83-3.93(m,1H),3.58-3.77(m,1H),3.07-3.25(m,1H),2.82(s,2H),2.54(s,3H),2.08-2.28(m,1H),1.67-1.76(m,1H),1.05-1.21(m,6H),0.80-1.02(m,3H).
2- ((3 R,5 s) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile and 2- ((3 s,5 r) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
2- (3-Amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile mixture was separated by SFC (conditions: column: DAICEL CHIRALCEL OJ (250 mm. Mu.m; 50 mm; 10 μm); mobile phase: [0.1% NH 3H2 O MEOH ];: 20% -20%,5.3 min) to give 2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile as a white solid and 2- ((3R, 5R) -3-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-hydroxy-3-imidazol-5-yl) amino) nicotinonitrile as a white solid Nicotine.
2- ((3 S,5 r) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (48):1HNMR(400MHz,DMSO-d6)δppm 9.00(s,1H),7.95(s,1H),7.29(d,J=1.63Hz,1H),7.22(dd,J=8.32,1.81Hz,1H),7.11(d,J=8.38Hz,1H),4.47(s,1H),4.10-4.25(m,1H),3.93-4.02(m,1H),3.84-3.93(m,2H),2.58-2.94(m,3H),1.92-2.12(m,1H),1.61-1.76(m,4H),1.15(s,6H),0.78(d,J=6.75Hz,3H).
2- ((3 R,5 s) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (49):1HNMR(400MHz,DMSO-d6)δ9.00(s,1H),7.95(s,1H),7.29(d,J=1.75Hz,1H),7.22(dd,J=8.38,1.75Hz,1H),7.11(d,J=8.38Hz,1H),4.47(s,1H),4.11-4.25(m,1H),3.78-4.04(m,3H),2.75-2.95(m,2H),2.68(br t,J=12.69Hz,1H),1.88-2.13(m,1H),1.50-1.80(m,4H),1.15(s,6H),0.78(d,J=6.75Hz,3H).
The absolute configuration of compounds 48 and 49 is randomly assigned to the cis conformation based on amino and methyl.
Example 7: synthesis of 5-chloro-2- [ (3S, 4R, 5R) -4-fluoro-3-hydroxy-5-methyl-1-piperidinyl ] -6- [ [3- (3-hydroxy-3-methyl-butyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (50) and 5-chloro-2- [ (3R, 4S, 5S) -4-fluoro-3-hydroxy-5-methyl-1-piperidinyl ] -6- [ [3- (3-hydroxy-3-methyl-butyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (51)
1-Benzyl-3-methyl-1, 2,3, 6-tetrahydropyridin-4-yl triflate
LiHMDS (1M, 295.16 mL) (1M in THF) was added dropwise to a solution of 1-benzyl-3-methyl-piperidin-4-one (50 g,245.97 mmol) in THF (500 mL) at-65 ℃. After the addition, the mixture was stirred at-65℃for 1hr, and then 1, 1-trifluoro-N-phenyl-N- (trifluoromethylsulfonyl) methanesulfonamide (96.66 g,270.56 mmol) in THF (400 mL) was added dropwise at-65 ℃. The resulting mixture was stirred at 15℃for 3hr. The mixture (mixed with another batch of the same scale) was slowly quenched with saturated NH 4 Cl (800 mL) and water (800 mL) was added. The mixture was extracted with ethyl acetate (800 ml x 3). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated to give a residue. The residue was purified by column chromatography on silica gel (silica gel, petroleum ether/ethyl acetate=1/0, 20/1) to give 180g of crude product. The crude product was dissolved in ethyl acetate (1L), then water (1L) was added to the mixture, followed by 1N HCl until ph=3. A white precipitate formed and the mixture was filtered to give a filter cake. To the filter cake was added water (1L), and then saturated Na 2CO3 was added until ph=8. The mixture was extracted with ethyl acetate (1 l×3). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated to give the title compound as a yellow oil (140 g, purity 93%).1H NMR(400MHz,CDCl3)δppm 7.33-7.36(m,4H),7.27-7.32(m,1H),5.69-5.74(m,1H),3.57-3.67(m,2H),3.06-3.18(m,2H),2.79(dd,J=11.31,4.95Hz,1H),2.62-2.72(m,1H),2.37(dd,J=11.37,5.87Hz,1H),1.16(d,J=6.85Hz,3H).
1-Benzyl-3-methyl-1, 2,3, 6-tetrahydropyridine
A mixture of 1-benzyl-3-methyl-1, 2,3, 6-tetrahydropyridin-4-yl trifluoromethanesulfonate (30 g,89.46 mmol), pd (OAc) 2 (401.69 mg,1.79 mmol), PPh3 (938.58 mg,3.58 mmol) and TEA (27.16 g,268.38mmol,37.36 mL) in DMF (300 mL) was stirred at 60℃under N 2 for 10min, and then formic acid (4.12 g,89.46mmol,3.37 mL) was added in one portion to the mixture. The mixture was stirred at 60℃under an atmosphere of N 2 for 50min. To the mixture (combined with the other 3 batches of the same scale) was added water (2L) and then extracted with ethyl acetate (3×800 mL). The combined organic phases were washed with brine (1L), dried over anhydrous Na 2SO4, filtered, and concentrated in vacuo. The residue was purified by column chromatography (silica gel, petroleum ether/ethyl acetate=1/0 to 20/1) to give the title compound (32 g,148.65mmol, yield 42%, purity) as a yellow oil 87%).1H NMR(400MHz,CDCl3)δppm 7.30-7.39(m,4H),7.27(s,2H),7.23-7.26(m,1H),5.59-5.68(m,2H),3.52-3.66(m,2H),3.02-3.14(m,1H),2.74-2.88(m,2H),2.37-2.48(m,1H),1.98(dd,J=10.94,8.13Hz,1H),0.96(d,J=7.09Hz,3H).
3-Benzyl-5-methyl-7-oxa-3-azabicyclo [4.1.0] heptane
To a solution of H 2O2 (15.14 g,133.49mmol,12.83mL, purity 30%) in DCM (50 mL) was added dropwise a solution of trifluoroacetic anhydride (TFAA) (84.11 g,400.47mmol,55.70 mL) at 0deg.C, and the resulting suspension was stirred at 0deg.C for 2hr. Simultaneously, TFA (3.04 g,26.70mmol,1.98 mL) was added dropwise to a solution of 1-benzyl-3-methyl-1, 2,3, 6-tetrahydropyridine (5 g,26.70 mmol) in DCM (100 mL) at 0deg.C and stirred for 2hr. The cold TFA/piperidine solution was added dropwise to the H 2O2/TFAA solution and stirred at 0deg.C for 1hr. The mixture was slowly washed with saturated sodium sulfite solution (300 mL) at 0 ℃ and N 2, then the system ph=8 was adjusted with saturated NaHCO 3. The mixture was extracted with DCM (3X 200 mL). The peroxide was detected with wet starch potassium iodide paper and the combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography (silica gel, petroleum ether/ethyl acetate=1/0, 10/1) to give the title compound (2.9 g,13.12mmol, yield 49%, purity) as a yellow oil 92%).1H NMR(400MHz,CDCl3)δppm 7.30-7.33(m,4H),7.22-7.27(m,1H),3.47(s,2H),3.22(t,J=3.70Hz,1H),2.97(d,J=3.93Hz,1H),2.84-2.92(m,1H),2.76-2.83(m,1H),2.46(dd,J=11.50,4.71Hz,1H),2.20(t,J=6.44Hz,1H),1.88(dd,J=11.44,6.08Hz,1H),1.10(d,J=7.15Hz,3H).
(3R, 4R, 5R) -1-benzyl-4-fluoro-5-methylpiperidin-3-ol
To a solution of 3-benzyl-5-methyl-7-oxa-3-azabicyclo [4.1.0] heptane (2.9 g,14.27 mmol) in DCM (45 mL) at 15deg.C and N 2 was added ethoxyethane in one portion; boron trifluoride; hydrofluoride (9.24 g,28.53mmol,7.83mL, purity 50%). The mixture was stirred at 15℃for 10min. Saturated NaHCO 3 (20 mL) was added to the mixture. The mixture was extracted with DCM (3X 15 mL). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated to give a residue. The residue was purified by column chromatography (silica gel, petroleum ether/ethyl acetate=1/0 to 5/1) to give the title compound (2 g,8.51mmol, yield 60%, purity) as a yellow solid 95%).1H NMR(400MHz,CDCl3)δppm 7.27-7.38(m,5H),4.35-4.52(m,1H),3.87(br s,1H),3.54(s,2H),2.81-3.01(m,1H),2.74(br d,J=10.37Hz,1H),2.48-2.59(m,2H),1.99-2.25(m,2H),0.99(d,J=6.79Hz,3H).
(4R, 5R) -1-benzyl-4-fluoro-5-methylpiperidin-3-one
To a solution of (COCl) 2 in DCM (50 mL) (1.71 g,13.44mmol,1.18 mL) was added dropwise DMSO (2.10 g,26.87mmol,2.10 mL) in DCM (5 mL) at-65 ℃. After stirring for 15min at-65 ℃, a solution of (3 r,4r,5 r) -1-benzyl-4-fluoro-5-methyl-piperidin-3-ol (2 g,8.96mmol,1 eq.) in DCM (20 mL) was slowly added dropwise at-65 ℃. The mixture was stirred at-65℃for another 15min, and TEA (4.53 g,44.79mmol,6.23 mL) was added to the mixture. After the addition, the mixture was stirred at 15℃for 0.5h. Water (150 mL) was slowly added at 0deg.C and the mixture was extracted with DCM (3X 100 mL). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated to give the title compound (2 g,5.69mmol, 64% yield, 64% purity) as a yellow oil (in 20mL THF).
(3S, 4R, 5R) -1-benzyl-4-fluoro-5-methylpiperidin-3-ol
To a solution of (4R, 5R) -1-benzyl-4-fluoro-5-methylpiperidin-3-one (1 g,4.52 mmol) in EtOH (10 mL) and THF (10 mL) at 0deg.C, N 2 was added NaBH 4 (205.17 mg,5.42 mmol). The mixture was stirred at 0deg.C for 1hr. The reaction was quenched with cold water (50 mL) and extracted with ethyl acetate (50 mL. Times.3). The organic layer was collected, washed with water, brine, dried over Na 2SO4, and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel20g/>SILICA FLASH chromatographic column, 0% -25% ethyl acetate/petroleum ether gradient eluent @120 mL/min) to give 500mg of crude product. The crude product was purified by preparative HPLC (column: phenomenex C18:30 mm 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:25% -45%,8 min) to give the title compound (400 mg,1.68mmol, 37% yield, purity) as a white solid 94%).1H NMR(400MHz,CDCl3)δppm 7.24-7.36(m,5H),4.50-4.71(m,1H),3.70-3.84(m,1H),3.55(d,J=1.71Hz,2H),2.90(br dd,J=10.39,5.01Hz,1H),2.49-2.59(m,1H),2.07-2.18(m,1H),1.81-2.03(m,3H),1.02(d,J=6.72Hz,3H).
(3S, 4R, 5R) -4-fluoro-5-methylpiperidin-3-ol
To a mixture of Pd/C (0.2 g, purity 10%) in THF (20 mL) under Ar was added (3S, 4R, 5R) -1-benzyl-4-fluoro-5-methylpiperidin-3-ol (400 mg,1.79 mmol) and TFA (612.79 mg,5.37mmol, 397.92. Mu.L). The mixture was stirred at 15℃for 10hr under H 2 (15 psi). The mixture was filtered and the filtrate concentrated in vacuo without further purification to give the title compound as a white solid (450 mg, crude ,TFA).1H NMR(400MHz,DMSO-d6)δppm 4.52-4.76(m,1H),3.70-3.91(m,1H),3.02-3.21(m,2H),2.70-2.86(m,1H),2.59(q,J=12.15Hz,1H),1.94-2.11(m,1H),0.95-1.01(m,3H).
5-Chloro-2- ((3 s,4r,5 r) -4-fluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a solution of 2, 5-dichloro-6- [ [3- (3-hydroxy-3-methyl-butyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (100 mg, 237.93. Mu. Mol) in DMSO (1 mL) was added DIEA (153.75 mg,1.19mmol, 207.22. Mu.L) and 4-fluoro-5-methyl-piperidin-3-ol (60.54 mg, 356.89. Mu. Mol, HCl). The mixture was stirred at 100deg.C for 12hr. The solution was filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: phenomenex C18 x 30mm x 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:25% -55%,8 min) to give the title compound (80 mg,154.59 μmol, 65% yield, purity) as a white solid 99%).1H NMR(400MHz,DMSO-d6)δ=8.94(s,1H),7.92(s,1H),7.32(d,J=1.7Hz,1H),7.27-7.22(m,1H),7.10(d,J=8.4Hz,1H),5.26(d,J=6.0Hz,1H),4.65-4.58(m,1H),4.51-4.40(m,2H),4.10(br dd,J=4.3,12.9Hz,1H),3.96-3.75(m,4H),3.59-3.43(m,1H),2.94(t,J=11.9Hz,1H),2.74-2.60(m,2H),1.94-1.76(m,1H),1.74-1.66(m,2H),1.18-1.14(m,6H),0.84-0.78(m,3H).
5-Chloro-2- ((3S, 4R, 5R) -4-fluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (50) and 5-chloro-2- ((3R, 4S, 5S) -4-fluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (51)
Racemic 5-chloro-2- ((3 s,4r,5 r) -4-fluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (80 mg, 154.59. Mu. Mol) was purified by SFC (column: CHIRALPAK IH,250 x 30mm,10 μm; mobile phase: [ Neu-IPA ]; B%:45% -45%,10 min) to give 5-chloro-2- ((3 s,4r,5 r) -4-fluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (30 mg, 57.45. Mu. Mol, 30%, purity 99%) and 5-chloro-2- ((3 r,4s,5 s) -4-fluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (30 mg,56.00 μmol, yield 29%, purity 96%) as a white solid.
50:1H NMR(400MHz,DMSO-d6)δ=8.94(s,1H),7.92(s,1H),7.32(d,J=1.5Hz,1H),7.28-7.21(m,1H),7.10(d,J=8.3Hz,1H),5.76(s,1H),5.27(d,J=5.9Hz,1H),4.64-4.48(m,1H),4.47(s,1H),4.17-4.05(m,1H),3.95-3.80(m,3H),3.59-3.42(m,1H),3.30(br s,1H),2.95(t,J=12.0Hz,1H),2.74-2.65(m,1H),2.34(s,1H),1.96-1.76(m,1H),1.74-1.61(m,2H),1.17(s,6H),0.82(d,J=6.9Hz,3H).
51:1H NMR(400MHz,DMSO-d6)δ=8.93(s,1H),7.92(s,1H),7.32(d,J=1.6Hz,1H),7.24(dd,J=1.7,8.3Hz,1H),7.09(d,J=8.4Hz,1H),5.26(d,J=6.0Hz,1H),4.65-4.47(m,1H),4.46(s,1H),4.15-4.06(m,1H),3.94-3.79(m,3H),3.60-3.42(m,1H),3.29(s,1H),2.94(t,J=11.9Hz,1H),2.76-2.62(m,2H),2.36-2.29(m,1H),1.98-1.76(m,1H),1.75-1.64(m,2H),1.16(s,6H),0.81(d,J=6.9Hz,3H).
The absolute configuration of compounds 50 and 51 is randomly assigned to the cis-conformation based on all substituents of the piperidine ring.
Example 8: synthesis of 5-fluoro-2- [ (3S, 4R, 5R) -4-fluoro-3-hydroxy-5-methyl-1-piperidinyl ] -6- [ [3- (3-hydroxy-3-methyl-butyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (52) and synthesis of 5-fluoro-2- [ (3R, 4S, 5S) -4-fluoro-3-hydroxy-5-methyl-1-piperidinyl ] -6- [ [3- (3-hydroxy-3-methyl-butyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (53)
2-Chloro-5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
DIEA (10.37 g,80.22mmol,13.97 mL) was added in one portion to a mixture of 5-amino-3- (3-hydroxy-3-methylbutyl) -1-methyl-1H-benzo [ d ] imidazol-2 (3H) -one (10 g,40.11 mmol) and 2, 6-dichloro-5-fluoro-pyridine-3-carbonitrile (7.66 g,40.11 mmol) in DMSO (100 mL) under N 2. The mixture was stirred at 100℃for 1.3 hours. To the mixture was added water (150 mL), and the resulting suspension was filtered, and the filter cake was dried under reduced pressure to give the crude title compound (16 g, crude) as a white solid, which was used without further purification .1H NMR(400MHz,DMSO-d6)δppm 9.95(s,1H),8.02-8.24(m,1H),7.51(d,J=3.00Hz,1H),7.25-7.35(m,1H),7.13(dd,J=8.38,5.13Hz,1H),4.44(d,J=5.25Hz,1H),3.79-3.96(m,2H),3.24-3.40(m,3H),1.64-1.80(m,2H),1.18(d,J=5.00Hz,5H).M+H+=404.1.
5-Fluoro-2- ((3 s,4r,5 r) -4-fluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a suspension of 2-chloro-5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (300 mg, 742.87. Mu. Mol), 4-fluoro-5-methyl-piperidin-3-ol (367.26 mg,1.49mmol, TFA) and Cs 2CO3 (2.42G, 7.43 mmol) in dioxane (6 mL) was added rac-BINAP-Pd-G3 (73.72 mg, 74.29. Mu. Mol) under a nitrogen atmosphere. The mixture was stirred at 100deg.C for 12hr. The mixture was filtered, and the filtrate was concentrated, and then purified by preparative HPLC to give the title compound as a white solid (60 mg,114.01 μmol, 15% yield, purity 95%).1H NMR(400MHz,DMSO-d6)δppm 9.41(s,1H),7.77(d,J=10.88Hz,1H),7.44(s,1H),7.38(d,J=8.31Hz,1H),7.08(d,J=8.44Hz,1H),5.27(d,J=5.87Hz,1H),4.50-4.66(m,1H),4.47(s,1H),4.02(dd,J=12.41,4.71Hz,1H),3.85-3.91(m,2H),3.80(br dd,J=12.72,3.30Hz,1H),3.50-3.66(m,1H),2.96(t,J=11.80Hz,1H),2.74(t,J=12.47Hz,1H),1.85-2.02(m,1H),1.66-1.73(m,2H),1.16(s,6H),0.91(d,J=6.85Hz,3H).
5-Fluoro-2- ((3S, 4R, 5R) -4-fluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (52)
5-Fluoro-2- ((3S, 4R, 5R) -4-fluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (70 mg, 139.85. Mu. Mol) was isolated by SFC to give the title compound (25.7 mg, 50.83. Mu. Mol, 36% yield, purity as a yellow solid 99%).1H NMR(400MHz,MeCN-d3)δppm 7.76(s,1H),7.55(d,J=1.83Hz,1H),7.44(d,J=10.64Hz,1H),7.20(dd,J=8.44,1.96Hz,1H),7.00(d,J=8.44Hz,1H),4.55-4.72(m,1H),4.10(dd,J=12.10,3.18Hz,1H),3.90-4.02(m,2H),3.82-3.90(m,1H),3.61-3.77(m,2H),3.33(s,3H),2.97-3.06(m,2H),2.84(t,J=12.53Hz,1H),1.98-2.11(m,1H),1.80(t,J=7.95Hz,2H),1.22(d,J=2.81Hz,6H),0.99(d,J=6.97Hz,3H).M+H+=501.3.
5-Fluoro-2- ((3R, 4S, 5S) -4-fluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (53)
5-Fluoro-2- ((3S, 4R, 5R) -4-fluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (70 mg, 139.85. Mu. Mol) was isolated by SFC to give the title compound (24.8 mg, 49.05. Mu. Mol, 35% yield, purity as a yellow solid 99%).1H NMR(400MHz,MeCN-d3)δppm 7.76(s,1H),7.55(d,J=1.83Hz,1H),7.45(d,J=10.76Hz,1H),7.20(dd,J=8.38,1.90Hz,1H),7.00(d,J=8.31Hz,1H),4.54-4.72(m,1H),4.06-4.15(m,1H),3.90-4.02(m,2H),3.86(dd,J=13.20,3.18Hz,1H),3.59-3.78(m,2H),3.33(s,3H),2.99-3.06(m,1H),2.98(s,1H),2.84(t,J=12.53Hz,1H),1.98-2.12(m,1H),1.77-1.84(m,2H),1.22(d,J=2.69Hz,6H),0.99(d,J=6.97Hz,3H).M+H+=501.3.
The absolute configuration of compounds 52 and 53 is randomly assigned to the cis-conformation based on all substituents of the piperidine ring.
Example 9: synthesis of 5-fluoro-2- ((3S, 4R, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((3- ((S) -3-hydroxybutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (54) and 5-fluoro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((3- ((R) -3-hydroxybutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (55)
/>
3-Hydroxybutyl 4-methylbenzenesulfonate
To a mixture of butane-1, 3-diol (25 g,277.41mmol,51.23 mL) in DCM (500 mL) was added DMAP (3.39 g,27.74 mmol) and TEA (56.14 g,554.81mmol, 77.22 mL) in one portion under N 2. 4-Methylbenzenesulfonyl chloride (52.89 g,277.41 mmol) was then added slowly at 0deg.C. The mixture was stirred at 20℃for 12 hours. The mixture was poured into saturated NaHCO 3 (500 mL) and extracted with DCM (500 mL x 3). The combined organic layers were washed with brine (1.2L), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=10/1 to 2/1) to give the title compound (58 g,227.71mmol, yield 41%, purity) as a violet oil 96%).1H NMR(400MHz,CDCl3)δ7.73(br d,J=7.83Hz,2H),7.30(br d,J=7.82Hz,2H),4.11-4.25(m,1H),4.00-4.11(m,1H),3.78-3.93(m,1H),2.39(s,3H),1.58-2.06(m,3H),1.11(br d,J=6.11Hz,3H).
3-Hydroxybutyl 4-methylbenzenesulfonate
DMAP (3.39 g,27.74 mmol) and TEA (56.14 g,554.81mmol, 77.22 mL) were added in one portion to a mixture of butane-1, 3-diol (25 g,277.41mmol,51.23 mL) in DCM (500 mL) under N 2. 4-Methylbenzenesulfonyl chloride (52.89 g,277.41 mmol) was then added slowly at 0deg.C. The mixture was stirred at 20℃for 12 hours. The mixture was poured into saturated NaHCO 3 (500 mL) and extracted with DCM (500 mL x 3). The combined organic layers were washed with brine (1.2L), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=10/1 to 2/1) to give the title compound (58 g,227.71mmol, yield 41%, purity) as a violet oil 96%).1H NMR(400MHz,CDCl3)δ7.73(br d,J=7.83Hz,2H),7.30(br d,J=7.82Hz,2H),4.11-4.25(m,1H),4.00-4.11(m,1H),3.78-3.93(m,1H),2.39(s,3H),1.58-2.06(m,3H),1.11(br d,J=6.11Hz,3H).
3- (3-Hydroxybutyl) -1-methyl-5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one
To a solution of 3-methyl-6-nitro-1H-benzimidazol-2-one (3 g,15.53 mmol) and 3-hydroxybutyl 4-methylbenzenesulfonate (7.59 g,31.06 mmol) in DMSO (30 mL) was added Cs 2CO3 (10.12 g,31.06 mmol) and KI (1.29 g,7.77 mmol). The flask was degassed and purged 3 times with N 2 and then the mixture was stirred at 100 ℃ for 12hr under an atmosphere of N 2. Water (50 mL) was added to the mixture. The mixture was extracted with ethyl acetate (50 ml x 3). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated to give a residue which was purified by column chromatography (silica gel, petroleum ether/ethyl acetate=10:1 to 1:1) and then triturated with petroleum ether/ethyl acetate=3:1 (15 mL) at 15 ℃ for 15min to give the title compound as a yellow solid (2 g,6.26mmol, 40% yield, 83% purity ).1H NMR(400MHz,DMSO-d6)δppm 1.09(d,J=6.25Hz,3H),1.61-1.77(m,2H),3.40(s,3H),3.57-3.67(m,1H),3.93-4.03(m,2H),4.63(d,J=4.88Hz,1H),7.35(d,J=8.63Hz,1H),8.04-8.07(m,1H),8.08(d,J=2.13Hz,1H).
5-Amino-3- (3-hydroxybutyl) -1-methyl-1H-benzo [ d ] imidazol-2 (3H) -one
To a solution of Pd/C (0.5 g, purity 10%) in DMF (20 mL) was added 3- (3-hydroxybutyl) -1-methyl-5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one (2 g,7.54 mmol) under Ar. The mixture was stirred at 50deg.C under H 2 (15 psi) for 12hr. The mixture was filtered and the filtrate concentrated in vacuo to give the title compound (1.7 g,6.86mmol, 91% yield, 95% purity) as a grey solid, which was used without further purification .1H NMR(400MHz,DMSO-d6)δppm 1.09(d,J=6.11Hz,3H),1.56-1.68(m,2H),3.22(s,3H),3.61(br s,1H),3.68-3.85(m,2H),4.64(br d,J=3.55Hz,1H),4.78(br s,2H),6.31(dd,J=8.31,1.71Hz,1H),6.41(d,J=1.59Hz,1H),6.79(d,J=8.19Hz,1H).
2-Chloro-5-fluoro-6- ((3- (-3-hydroxybutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a mixture of 5-amino-3- (3-hydroxybutyl) -1-methylbenzimidazol-2-one (200 mg, 850.05. Mu. Mol) and 2, 6-dichloro-5-fluoropyridine-3-carbonitrile (194.82 mg,1.02 mmol) in DMSO (2 mL) was added DIPEA (219.72 mg,1.70mmol, 296.13. Mu.L). The mixture was stirred at 100deg.C for 12hr. The mixture was poured into water (5 mL) and stirred for 10min. The mixture was filtered and the filter cake was washed with ethyl acetate (3 mL). The filter cake was dried under vacuum to give the title compound as a yellow solid (300 mg, 644.26. Mu. Mol, yield 84%, purity) 84%).1H NMR(400MHz,DMSO-d6)δ9.94(s,1H),8.11(d,J=10.63Hz,1H),7.50(d,J=1.38Hz,1H),7.29(dd,J=8.44,1.56Hz,1H),7.12(d,J=8.38Hz,1H),4.63(d,J=4.63Hz,1H),3.74-3.96(m,2H),3.57-3.71(m,1H),3.32(s,3H),1.59-1.80(m,2H),1.10(d,J=6.13Hz,3H).
(S) -2-chloro-5-fluoro-6- ((3- (3-hydroxybutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile and (R) -2-chloro-5-fluoro-6- ((3- (3-hydroxybutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
The residue was purified by preparative HPLC (neutral conditions: column: phenomenex C18 x 40mm x 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ];: B%:15% -45%,8 min). The residue was further separated by SFC (conditions: column: REGIS (S, S) WHELK-O1 (250 mm. 25mm,10 μm); mobile phase: [ 0.1% NH 3H2 O in IPA;:. B%:45% -45%,7 min) to give (S) -2-chloro-5-fluoro-6- ((3- (3-hydroxybutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (40 mg, 97.47. Mu. Mol, yield 15%, purity 95%) as a white solid and (R) -2-chloro-5-fluoro-6- ((3- (3-hydroxybutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (40 mg, 92.14. Mu. Mol, yield 14%, purity as a white solid 90%).1H NMR(400MHz,MeOD-d4)δppm 7.72-7.80(m,1H),7.68(s,1H),7.35(br d,J=8.25Hz,1H),7.13(br d,J=8.25Hz,1H),4.02(br t,J=7.00Hz,2H),3.78(br d,J=4.38Hz,1H),3.43(s,3H),1.97-2.10(m,1H),1.74-1.97(m,2H),1.27-1.39(m,1H),1.22(br d,J=6.00Hz,3H).
5-Fluoro-2- ((3S, 4R, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((3- ((S) -3-hydroxybutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (54)
To a solution of (S) -2-chloro-5-fluoro-6- ((3- (3-hydroxybutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (40 mg, 102.61. Mu. Mol) and (3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidine (37.74 mg, 153.92. Mu. Mol, TFA) in DMSO (0.5 mL) was added DIEA (26.52 mg, 205.23. Mu. Mol, 35.75. Mu.L) under N 2. The mixture was stirred at 100deg.C for 12hr. The residue was purified by preparative HPLC (neutral conditions: chromatography column: phenomenex C18 x 75 x 30mm x 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:25% -55%,8 min) to give the title compound (16 mg,33.02 μmol, 32% yield, purity) as a white solid 100%).1H NMR(400MHz,DMSO-d6)δ9.37(s,1H),7.76(d,J=11.00Hz,1H),7.50(d,J=1.71Hz,1H),7.37(dd,J=8.44,1.83Hz,1H),7.09(d,J=8.44Hz,1H),4.54-4.64(m,2H),4.45(s,1H),3.76-3.98(m,4H),3.55-3.68(m,1H),2.80(t,J=12.59Hz,2H),1.77-1.98(m,2H),1.54-1.77(m,2H),1.08(d,J=6.11Hz,3H),0.90(dd,J=6.85,2.57Hz,6H).
5-Fluoro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((3- ((R) -3-hydroxybutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (55)
To a solution of (R) -2-chloro-5-fluoro-6- ((3- (3-hydroxybutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (40 mg, 102.61. Mu. Mol) and (3S, 4S, 5R) -4-fluoro-3, 5-dimethyl-piperidine (37.74 mg, 153.92. Mu. Mol, TFA) in DMSO (0.5 mL) was added DIEA (26.52 mg, 205.22. Mu. Mol, 35.75. Mu. L) under N 2. The mixture was stirred at 100deg.C for 12hr. The residue was purified by preparative HPLC (neutral conditions: chromatography column: phenomenex C18 x 75 x 30mm x3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:25% -55%,8 min) to give the title compound (9.5 mg,19.61 μmol, 19% yield, purity) as a white solid 100%).1H NMR(400MHz,DMSO-d6)δ9.38(s,1H),7.76(d,J=11.00Hz,1H),7.50(d,J=1.83Hz,1H),7.37(dd,J=8.44,1.83Hz,1H),7.09(d,J=8.44Hz,1H),4.55-4.68(m,2H),4.45(s,1H),3.76-3.99(m,4H),3.55-3.70(m,1H),2.80(br t,J=12.59Hz,2H),1.55-2.03(m,4H),1.05-1.11(m,3H),0.90(dd,J=6.91,2.51Hz,6H).
The absolute configuration of compounds 54 and 55 is randomly assigned to the cis-conformation based on all of the piperidine ring substituents.
Example 10: synthesis of 5-fluoro-2- [ (3R, 4R, 5S) -4-fluoro-3- (2-hydroxyethyl) -5-methyl-1-piperidinyl ] -6- [ [3- (3-hydroxybutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (56) and 5-fluoro-2- [ (3S, 4S, 5R) -4-fluoro-3- (2-hydroxyethyl) -5-methyl-1-piperidinyl ] -6- [ [3- (3-hydroxybutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (57)
2- (1-Benzyl-5-methyl-4-oxopiperidin-3-yl) acetic acid methyl ester
LDA (2M, 79.94 mL) was added dropwise to a mixture of 1-benzyl-3-methylpiperidin-4-one (25 g,122.98 mmol) in THF (250 mL) at-65℃and the mixture was stirred at-65℃for 30min, then HMPA (28.65 g,159.88mmol,28.09 mL) was added dropwise and the mixture was stirred at-65℃for 30min. Methyl 2-bromoacetate (24.46 g,159.88mmol,15.10 mL) was then added dropwise. The mixture was heated to 15 ℃ and stirred for 12hr. To the mixture (combined with another batch of the same scale) was then added water (600 mL) and extracted with EtOAc (800 mL x 3). The combined organic layers were dried over Na 2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=1/0 to 3/1) to give the title compound (40 g,116.22mmol, yield 47% purity) as a yellow solid 80%).(1H NMR(400MHz,DMSO-d6)δ7.37-7.27(m,5H),3.66-3.62(m,2H),3.59-3.54(m,3H),3.20-3.01(m,3H),2.78(td,J=6.1,11.8Hz,1H),2.56(d,J=7.6Hz,1H),2.24-1.99(m,3H),0.83(d,J=6.6Hz,3H).
1-Benzyl-3- (2-hydroxyethyl) -5-methylpiperidin-4-ol
To a solution of methyl 2- (1-benzyl-5-methyl-4-oxo-3-piperidinyl) acetate (10 g,36.32 mmol) in THF (100 mL) was added LAH (2.76 g,72.64 mmol) at 0 ℃. The mixture was stirred at 20deg.C for 1hr. To the mixture was added water (3 mL) and 2M NaOH (3 mL) at 0deg.C. The mixture was dried over anhydrous Na 2SO4 and stirred at 15 ℃ for 20min. The mixture was then filtered and the filtrate concentrated under reduced pressure to give the title compound (9 g, crude) as a yellow oil ).1H NMR(400MHz,DMSO-d6)δ7.35-7.20(m,7H),4.62(d,J=6.5Hz,1H),4.44(t,J=5.0Hz,1H),3.52-3.42(m,2H),2.85(br d,J=10.3Hz,1H),2.70-2.64(m,1H),1.87-1.77(m,1H),1.69-1.41(m,5H),1.14-1.07(m,1H),0.83(d,J=6.4Hz,3H).
1-Benzyl-3- (2- ((tert-butyldiphenylsilyl) oxy) ethyl) -5-methylpiperidin-4-ol
To a solution of 1-benzyl-3- (2-hydroxyethyl) -5-methylpiperidin-4-ol (5.00 g,20.05 mmol) in DCM (60 mL) was added TBDPSCl (6.06 g,22.06mmol,5.66 mL), DMAP (734.93 mg,6.02 mmol) and TEA (10.15 g,100.26mmol,13.96 mL). The mixture was stirred at 15℃for 4hr. After addition of water (100 mL), the solution was extracted with EtOAc (100 mL. Times.3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo to give a residue which was purified by flash column chromatography (SILICA FLASH column, gradient of 0% -30% ethyl acetate/petroleum ether) to give the title compound (7.5 g,14.11mmol, 70% purity) as a white oil 92%).1H NMR(400MHz,DMSO-d6)δ7.71-7.54(m,4H),7.46-7.36(m,6H),7.33-7.18(m,4H),4.50(d,J=6.7Hz,1H),4.03(q,J=7.1Hz,1H),3.70-3.55(m,2H),3.41-3.29(m,3H),2.90-2.63(m,2H),2.06-1.95(m,1H),1.68-1.48(m,3H),1.29-1.12(m,1H),1.01-0.91(m,9H),0.83(d,J=6.4Hz,3H).
1-Benzyl-3- (2- ((tert-butyldiphenylsilyl) oxy) ethyl) -4-fluoro-5-methylpiperidine
DAST (3.30 g,20.50mmol,2.71 mL) was added to a solution of 1-benzyl-3- (2- ((tert-butyldiphenylsilyl) oxy) ethyl) -5-methylpiperidin-4-ol (5.00 g,10.25 mmol) in DCM (50 mL) at-65 ℃. The mixture was stirred at-70deg.C for 0.5hr and at 15deg.C for 11.5hr. Saturated NaHCO 3 (50 mL) was added to the mixture and extracted with DCM (50 mL x 3). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel (SILICA FLASH column, gradient of 0% -30% ethyl acetate/petroleum ether) to give the title compound (3.3 g,6.71mmol, 65% yield, purity) as a yellow oil 99%).1H NMR(400MHz,CDCl3)δ7.69-7.62(m,4H),7.44-7.36(m,5H),7.33-7.27(m,5H),3.81-3.59(m,3H),3.56-3.39(m,2H),3.06-2.76(m,2H),2.08-1.88(m,3H),1.80-1.62(m,2H),1.44-1.31(m,1H),1.04(s,9H),0.96(d,J=6.5Hz,3H).
2- [ (3R, 4R, 5S) -1-benzyl-4-fluoro-5-methyl-3-piperidinyl ] ethoxy-tert-butyl-diphenylsilane (intermediate 3) and 2- [ (3S, 4S, 5R) -1-benzyl-4-fluoro-5-methyl-3-piperidinyl ] ethoxy-tert-butyl-diphenylsilane (intermediate 4)
The racemic 1-benzyl-3- (2- ((tert-butyldiphenylsilyl) oxy) ethyl) -4-fluoro-5-methylpiperidine was purified by SFC (column: DAICEL CHIRALCEL OD (250 mm. Times.50 mm,10 μm); mobile phase: [0.1% NH 3H2 O IPA ]; B%:25% -25%,5 min) to give 2- [ (3R, 4R, 5S) -1-benzyl-4-fluoro-5-methyl-3-piperidinyl ] ethoxy-tert-butyl-diphenylsilane (1.5 g,3.03mmol, 45% yield), purity 99%).1H NMR(400MHz,DMSO-d6)δ7.60-7.54(m,4H),7.48-7.37(m,6H),7.33-7.21(m,5H),3.89-3.68(m,1H),3.64(t,J=6.3Hz,2H),3.48-3.38(m,2H),2.96-2.85(m,1H),2.80-2.70(m,1H),1.98-1.75(m,3H),1.74-1.64(m,2H),1.40-1.29(m,1H),0.95(s,9H),0.88(d,J=6.2Hz,3H) and 2- [ (3S, 4S, 5R) -1-benzyl-4-fluoro-5-methyl-3-piperidinyl ] ethoxy-tert-butyl-diphenylsilane (1.5 g,3.03mmol, 45% purity) as a white solid 99%).1H NMR(400MHz,DMSO-d6)δ7.60-7.53(m,4H),7.48-7.37(m,6H),7.33-7.20(m,5H),3.88-3.69(m,1H),3.64(t,J=6.3Hz,2H),3.43(d,J=0.7Hz,2H),2.96-2.70(m,2H),2.00-1.75(m,3H),1.74-1.66(m,2H),1.41-1.28(m,1H),0.95(s,9H),0.88(d,J=6.3Hz,3H).
2- [ (3R, 4R, 5S) -1-benzyl-4-fluoro-5-methyl-3-piperidinyl ] ethanol
A solution of 2- [ (3R, 4R, 5S) -1-benzyl-4-fluoro-5-methyl-3-piperidinyl ] ethoxy-tert-butyldiphenylsilane (1.5 g,3.06 mmol) in HCl/MeOH (50 mL) was stirred at 15℃for 8hr. After addition of saturated aqueous NaHCO 3 (30 mL), the solution was extracted with EtOAc (30 mL. Times.3). The combined organic phases were washed with brine (30 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo to give the title compound as a yellow oil (600 mg,2.39mmol, 78% yield, purity 100%).1H NMR(400MHz,DMSO-d6)δ7.35-7.21(m,5H),4.40(t,J=5.1Hz,1H),3.88-3.63(m,1H),3.55-3.46(m,1H),3.44-3.31(m,4H),2.99-2.85(m,1H),2.78-2.65(m,1H),1.81-1.61(m,4H),1.30-1.13(m,1H),0.88(d,J=6.4Hz,3H).
2- [ (3R, 4R, 5S) -4-fluoro-5-methyl-3-piperidinyl ] ethanol
Pd/C (300 mg, purity 10%) was added to a solution of 2- [ (3R, 4R, 5S) -1-benzyl-4-fluoro-5-methyl-3-piperidinyl ] ethanol (600 mg,2.39 mmol) in EtOH (40 mL) under Ar. The flask was degassed and purged 3 times with H 2. The mixture was stirred at H 2 (15 psi) and 60℃for 12hr. Passing the reaction mixture throughPad filtration and concentration of the filtrate in vacuo gave the title compound (200 mg, 372.17. Mu. Mol, 16% yield, purity) as a grey oil 30%).1H NMR(400MHz,DMSO-d6)δ4.43(br dd,J=6.5,11.8Hz,1H),3.54-3.35(m,3H),3.34-3.21(m,1H),2.82-2.61(m,2H),2.13-1.88(m,2H),1.81(dtd,J=4.2,6.7,13.8Hz,1H),1.39-1.29(m,1H),1.22(t,J=7.3Hz,1H),1.05(t,J=7.0Hz,1H),1.01-0.95(m,3H).
5-Fluoro-2- [ (3R, 4R, 5S) -4-fluoro-3- (2-hydroxyethyl) -5-methyl-1-piperidinyl ] -6- [ [3- (3-hydroxybutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (50)
To a solution of 2-chloro-5-fluoro-6- [ [3- (3-hydroxybutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (20 mg, 51.31. Mu. Mol) in DMSO (1 mL) was added 2- [ (3R, 4R, 5S) -4-fluoro-5-methyl-3-piperidinyl ] ethanol (8.27 mg, 51.31. Mu. Mol) and DIEA (26.52 mg, 205.24. Mu. Mol, 35.75. Mu.L). The mixture was stirred at 100deg.C for 2hr. The solution was filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: phenomenex C18 x 30mm x 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:10% -40%,8 min) to give the title compound (3 mg,5.36 μmol, yield 10%, purity) as a yellow solid 92%).1H NMR(400MHz,DMSO-d6)δ9.39(br s,1H),7.78(br d,J=10.9Hz,1H),7.52-7.31(m,2H),7.14-7.00(m,1H),4.62(br d,J=4.0Hz,1H),4.49-4.37(m,1H),4.29-4.14(m,1H),4.08-4.00(m,1H),3.98-3.90(m,1H),3.89-3.78(m,2H),3.62(br d,J=1.5Hz,1H),3.47-3.36(m,3H),2.78-2.69(m,1H),2.67-2.60(m,1H),1.82-1.62(m,6H),1.37-1.25(m,2H),1.09(br d,J=5.5Hz,3H),0.92(br s,3H).
2- [ (3S, 4S, 5R) -1-benzyl-4-fluoro-5-methyl-3-piperidinyl ] ethanol
A solution of 2- [ (3S, 4S, 5R) -1-benzyl-4-fluoro-5-methyl-3-piperidinyl ] ethoxy-tert-butyl-diphenyl-silane (1.5 g,3.06 mmol) in HCl/MeOH (1.25M, 50 mL) was stirred at 15℃for 8hr. After addition of saturated aqueous NaHCO 3 (30 mL), the solution was extracted with EtOAc (30 mL. Times.3). The combined organic phases were washed with brine (30 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo to give the crude title compound (400 mg,1.59mmol, 52% yield, 100% purity) as a yellow oil, which was used without further purification .1H NMR(400MHz,DMSO-d6)δ7.39-7.20(m,5H),4.40(t,J=5.1Hz,1H),3.90-3.65(m,1H),3.56-3.47(m,1H),3.42-3.31(m,4H),2.98-2.67(m,2H),1.81-1.61(m,4H),1.30-1.14(m,1H),0.89(d,J=6.4Hz,3H).M+H+=252.1.
2- [ (3S, 4S, 5R) -4-fluoro-5-methyl-3-piperidinyl ] ethanol
Pd/C (50 mg, purity 10%) was added to a solution of 2- [ (3S, 4S, 5R) -1-benzyl-4-fluoro-5-methyl-3-piperidinyl ] ethanol (400 mg,1.59 mmol) in EtOH (5 mL) under an argon atmosphere. The suspension was degassed and purged 3 times with H 2. The mixture was stirred at H 2 (15 psi) and 60℃for 12hr. Passing the reaction mixture throughPad filtration and concentration of the filtrate under reduced pressure gave the crude title compound (200 mg, crude) as a pale oil, which was used without further purification .1H NMR(400MHz,DMSO-d6)δ4.48-4.38(m,1H),4.23-4.02(m,1H),3.53-3.38(m,3H),3.33-3.22(m,1H),2.85-2.60(m,2H),2.12-1.90(m,2H),1.81(dtd,J=4.2,6.8,13.8Hz,1H),1.41-1.29(m,1H),1.26-1.19(m,1H),1.06(t,J=7.0Hz,1H),1.02-0.96(m,3H).
5-Fluoro-2- [ (3S, 4S, 5R) -4-fluoro-3- (2-hydroxyethyl) -5-methyl-1-piperidinyl ] -6- [ [3- (3-hydroxybutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (56)
To a solution of 2-chloro-5-fluoro-6- [ [3- (3-hydroxybutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (20 mg, 51.31. Mu. Mol) in DMSO (1 mL) was added 2- [ (3S, 4S, 5R) -4-fluoro-5-methyl-3-piperidinyl ] ethanol (14.12 mg, 87.60. Mu. Mol) and DIEA (26.52 mg, 205.24. Mu. Mol, 35.75. Mu.L). The mixture was stirred at 100deg.C for 2hr. The solution was filtered and the filtrate concentrated in vacuo to give a residue which was purified by prep HPLC to give the title compound as a yellow solid (5 mg,8.94 μmol, 17% purity 92%).1H NMR(400MHz,DMSO-d6)δ9.39(s,1H),7.77(d,J=10.9Hz,1H),7.49(dd,J=1.9,2.9Hz,1H),7.39(br d,J=8.4Hz,1H),7.09(d,J=8.5Hz,1H),4.62(d,J=4.6Hz,1H),4.48-4.40(m,1H),4.28-4.18(m,1H),4.08-4.01(m,1H),3.98-3.91(m,1H),3.90-3.78(m,2H),3.62(br s,1H),3.46-3.37(m,3H),2.73(br t,J=12.3Hz,1H),2.68-2.60(m,1H),1.84-1.62(m,6H),1.37-1.22(m,2H),1.09(d,J=6.1Hz,3H),0.93(dd,J=2.4,6.4Hz,3H).M+H+=515.3.
The absolute configuration of compounds 56 and 57 is randomly assigned to the cis conformation based on amino and methyl.
Example 11: synthesis of 2- ((6- ((3-chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2- (dimethylamino) ethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (11)
3-Hydroxy-6-nitroquinolin-2 (1H) -one
TMSCHN 2 (2M, 62.46 mL) was added dropwise to a mixture of 5-nitroindoline-2, 3-dione (20 g,104.10 mmol) and TEA (21.07 g,208.19mmol,28.98 mL) in EtOH (600 mL) at 20 ℃. The reaction mixture was stirred at 20 ℃ for 12hr and concentrated in vacuo. 1N HCl (600 mL) was then added to the concentrated reaction mixture and stirred for 3hr. The resulting suspension was filtered and the filter cake was collected and dried in vacuo to give the title compound as an orange-red solid (40 g, crude product) ).1HNMR(400MHz,DMSO-d6)δ12.54(s,1H),10.38-9.71(m,1H),8.51(d,J=2.4Hz,1H),8.13(dd,J=2.4,9.2Hz,1H),7.39(d,J=9.2Hz,1H),7.31(s,1H).
2- ((6-Nitro-2-oxo-1, 2-dihydro-quinolin-3-yl) oxy) acetic acid methyl ester
To a solution of 3-hydroxy-6-nitro-1H-quinolin-2-one (20 g,97.01 mmol) in DMSO (200 mL) was added DBU (17.72 g,116.42mmol,17.55 mL) and then methyl 2-bromoacetate (17.81 g,1161.42mmol, 10.99 mL) and the resulting mixture was stirred at 100deg.C for 3hr. The reaction mixture was treated with water (600 mL) to yield a precipitate. The suspension was filtered and the filter cake washed with EtOAc (800 mL) and the solid was dried in vacuo to give the title compound (26 g, crude) as an orange-red solid ).1H NMR(400MHz,DMSO-d6)δ12.51(s,1H),8.54(d,J=2.4Hz,1H),8.20-8.17(m,1H),7.49(s,1H),7.39(d,J=9.2Hz,1H),4.89(s,2H),3.77-3.71(s,3H).
2- ((6-Amino-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid methyl ester
Pd/C (5 g, purity 10%) was added to a solution of methyl 2- [ (6-nitro-2-oxo-1H-quinolin-3-yl) oxy ] acetate (16 g,57.51 mmol) in DMF (200 mL) under Ar. The flask was degassed and purged 3 times with H 2. The mixture was stirred at H 2 (50 psi) and 50deg.C for 12hr. The reaction mixture was filtered and the filtrate concentrated in vacuo to give the title compound as an off-black solid (12 g, crude product) ).1H NMR(400MHz,DMSO-d6)δ11.60(s,1H),7.95(s,1H),7.03-6.98(m,2H),6.72(dd,J=2.4,8.8Hz,1H),6.68(d,J=2.0Hz,1H),6.05-5.39(m,2H),4.83(s,2H),3.71(s,3H).
2- ((6- ((3, 6-Dichloro-5-cyanopyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid methyl ester
To a mixture of methyl 2- [ (6-amino-2-oxo-1H-quinolin-3-yl) oxy ] acetate (3 g,12.09 mmol) and 2,5, 6-trichloropyridine-3-carbonitrile (2.51 g,12.09 mmol) in DMF (30 mL) was added DIPEA (3.12 g,24.17mmol,4.21 mL) and the reaction mixture was stirred at 100deg.C for 4hr. The mixture was cooled to 15 ℃ and water (about 50 mL) was added to form a precipitate. The mixture was filtered and the filter cake was washed with EtOAc (about 80 mL) and dried in vacuo to give the title compound (15 g, crude) as a brown solid ).1H NMR(400MHz,DMSO-d6)δ12.02(s,1H),9.58(s,1H),8.35(s,1H),7.65(d,J=2.0Hz,1H),7.48(dd,J=2.0,8.8Hz,1H),7.27(d,J=8.8Hz,1H),7.21(s,1H),4.88(s,2H),3.72(s,3H).
2- ((6- ((3-Chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid methyl ester
To a solution of methyl 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] acetate (5 g,11.93 mmol) and (3 s,5 r) -4, 4-difluoro-3, 5-dimethyl-piperidine (4.08 g,15.51mmol, tfa) in DMSO (50 mL) was added DIPEA (7.71 g,59.63mmol,10.39 mL) and the reaction mixture was stirred at 100 ℃ for 12H. The mixture was cooled to 15 ℃ and water (about 50 mL) was added to form a precipitate. The mixture was filtered, and the filter cake was washed with EtOAc (about 80 mL) and dried in vacuo. The residue was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=1/1 to 0/1) to give a residue, which was then triturated with a solvent mixture (PE: etoac=1:1, 40 mL) at 20 ℃ for 30min to give the title compound (2.3 g,3.98mmol, 33% yield, 92% purity as a pale brown solid ).1H NMR(400MHz,DMSO-d6)δ11.96(s,1H),9.08(s,1H),7.97(s,1H),7.66(s,1H),7.49(dd,J=2.0,8.8Hz,1H),7.25(d,J=8.4Hz,1H),7.20(s,1H),4.85(s,2H),4.10(d,J=13.2Hz,2H),3.72(s,3H),2.77(t,J=12.4Hz,2H),2.14-1.93(m,2H),0.82(br d,J=6.4Hz,6H).
2- ((6- ((3-Chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
To a mixture of EtOH (90 mL) and methyl 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3S, 5R) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] acetate (10 g,23.85 mmol) in NMP (10 mL) was added MeNH 2 (14.82 g,190.83mmol, purity 40%) and the reaction mixture was stirred at 70℃for 12hr. The mixture was cooled to 20 ℃, then the reaction mixture was filtered, and the filter cake was collected and dried under reduced pressure to give the title compound (9.2 g,21.12mmol, 89% yield, 96% purity) as a pale brown solid.
2- ((6- ((3-Chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2- (dimethylamino) ethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid methyl ester
A mixture of methyl 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3S, 5R) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] acetate (350 mg, 657.97. Mu. Mol), 2-chloro-N, N-dimethylethylamine (123.21 mg, 855.36. Mu. Mol, HCl), cs 2CO3 (643.14 mg,1.97 mmol), KI (54.61 mg, 328.99. Mu. Mol) in DMSO (5 mL) was stirred at 60℃for 2hr under an atmosphere of N 2. To the mixture was added water (5 mL), the precipitated solid was filtered, and washed with H 2 O (20 mL) and petroleum ether/ethyl acetate (1/1.5 mL), then purified by preparative HPLC (column: phenomnex C18 80 x 40mm x3 μm; mobile phase: [ water (NH 4HCO3) -ACN ];: B%:50% -70%,8 min) to give the title compound (50 mg, 78.36. Mu. Mol, 12% yield, purity) as a yellow solid 95%).1H NMR(400MHz,DMSO-d6)δ9.14(s,1H),7.99(s,1H),7.75(d,J=2.0Hz,1H),7.63(dd,J=2.4,9.2Hz,1H),7.46(d,J=9.2Hz,1H),7.22(s,1H),4.86(s,2H),4.38(t,J=6.8Hz,2H),4.12(br d,J=12.4Hz,2H),3.72(s,3H),2.78(br t,J=12.4Hz,2H),2.52(d,J=1.6Hz,2H),2.23(s,6H),2.10-1.99(m,2H),0.84(d,J=6.8Hz,6H).
2- ((6- ((3-Chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2- (dimethylamino) ethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (11)
To a mixture of methyl 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3 s,5 r) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -1- [2- (dimethylamino) ethyl ] -2-oxo-3-quinolinyl ] oxy ] acetate (50 mg,82.91 μmol) in EtOH (5 mL) was added hexamethylenetetramine (purity 40% in 5mL, h 2 O). The mixture was stirred at 70℃for 12hr. The reaction mixture was concentrated under reduced pressure to give a residue which was purified by preparative HPLC (column: waters Xbridge BEH C18.100.30 mm.10 μm; mobile phase: [ water (NH 4HCO3) -ACN ];B%:35% -65%,10 min) to give the title compound (6 mg, 9.25. Mu. Mol, 11% yield, purity) as a white solid 93%).1HNMR(400MHz,DMSO-d6)δ9.14(s,1H),7.99(s,1H),7.74(s,1H),7.66(d,J=8.8Hz,1H),7.47(d,J=9.2Hz,1H),7.22(s,2H),4.53(s,2H),4.39(t,J=6.8Hz,2H),4.13(d,J=12.4Hz,2H),2.78(t,J=12.8Hz,2H),2.67(d,J=4.4Hz,3H),2.63-2.58(m,1H),2.23(s,6H),2.09–2.03(m,3H),0.84(d,J=6.8Hz,6H).
Example 12: synthesis of 2- ((6- ((3-chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (7) and 2- ((6- ((3-chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2- (oxetan-3-ylmethoxy) quinolin-3-yl) oxy) -N-methylacetamide (58)
Methyl 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3S, 5R) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -1- (oxetan-3-ylmethyl) -2-oxo-3-quinolinyl ] oxy ] acetate and methyl 2- ((6- ((3-chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2- (oxetan-3-ylmethoxy) quinolin-3-yl) oxy) acetate
To a solution of methyl 2- ((6- ((3-chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetate (500 mg, 939.96. Mu. Mol) and K 2CO3 (259.82 mg,1.88 mmol) in DMSO (6 mL) was added 3- (iodomethyl) oxetan (223.34 mg,1.13 mmol) and the reaction mixture was stirred at 80℃for 12hr. The reaction mixture was treated with water (9 mL), then a precipitate formed, the suspension was filtered, and the filter cake was collected and dried in vacuo to give a mixture of methyl 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3 s,5 r) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -1- (oxetan-3-ylmethyl) -2-oxo-3-quinolinyl ] oxy ] acetate and methyl 2- ((6- ((3-chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2- (oxetan-3-ylmethoxy) quinolin-3-yl) oxy) acetate as a brown solid (500 mg, crude product).
2- ((6- ((3-Chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide and 2- ((6- ((3-chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2- (oxetan-3-ylmethoxy) quinolin-3-yl) oxy) -N-methylacetamide
A mixture of methyl 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3S, 5R) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -1- (oxetan-3-ylmethyl) -2-oxo-3-quinolinyl ] oxy ] acetate and methyl 2- ((6- ((3-chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2- (oxetan-3-ylmethoxy) quinolin-3-yl) oxy) acetate (200 mg, 332.21. Mu. Mol) and 40% purity in MeNH 2(25.79mg,332.21μmol,10mL,H2 O in EtOH (10 mL) was stirred at 70℃for 12hr. The reaction mixture was concentrated in vacuo and then dissolved in DMSO (1.5 mL) and purified by preparative HPLC (column: waters Xbridge Prep OBD C, 150 x 40mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ];: B%:35% -65%,8 min) to give the two title compounds.
2- ((6- ((3-Chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (7)
The title compound (45 mg, 74.79. Mu. Mol, yield 23%, purity) was isolated as a white solid 99%).1H NMR(400MHz,DMSO-d6)δ9.13(s,1H),7.99(s,1H),7.98-7.92(d,J=4.4Hz,1H),7.80-7.74(d,J=2.4Hz,1H),7.64-7.58(m,1H),7.57-7.51(m,1H),7.22(m,1H),4.66(d,J=7.0Hz,2H),4.62-4.56(m,2H),4.55-4.49(m,4H),4.17-4.08(m,2H),3.43(m,1H),2.86-2.72(m,2H),2.67(d,J=4.8Hz,3H),2.15-1.97(m,2H),0.84(d,J=6.4Hz,6H).
2- ((6- ((3-Chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2- (oxetan-3-ylmethoxy) quinolin-3-yl) oxy) -N-methylacetamide (58)
The title compound (45 mg, 74.79. Mu. Mol, yield 23%, purity) was isolated as a white solid 99%).1H NMR(400MHz,DMSO-d6)δ9.16(m,1H),8.01(m,1H),7.91-7.85(m,2H),7.74-7.64(m,2H),7.49(s,1H),4.78-4.72(m,2H),4.67(d,J=6.8Hz,2H),4.63(s,2H),4.51(t,J=6.0Hz,2H),4.20-4.12(d,J=12.4Hz,2H),3.51-3.42(m,1H),2.80(t,J=12.4Hz,2H),2.66(d,J=4.8Hz,3H),2.15-1.99(m,2H),0.84(d,J=6.8Hz,6H).
Example 13: synthesis of 2- ((3S, 5R) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (2- (1-hydroxycyclopropyl) ethyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (59)
2, 5-Dichloro-6- ((1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A flask containing a mixture of 5-amino-1-methyl-1H-benzo [ d ] imidazol-2 (3H) -one (2 g,12.26 mmol), 2,5, 6-trichloropyridine-3-carbonitrile (2.54 g,12.26 mmol) and DIEA (3.17 g,24.51mmol,4.27 mL) in DMSO (20 mL) was degassed and purged 3 times with N 2, and then the mixture was stirred at 80℃under an atmosphere of N 2 for 2hr. To the mixture was added water (50 mL) to form a precipitate, which was filtered, and the filter cake was dried under reduced pressure to give the title compound (4 g, purity) as a brown solid 88%).1H NMR(400MHz,DMSO-d6)δ3.28(s,3H),7.06-7.09(m,1H),7.13-7.17(m,1H),7.20(d,J=1.71Hz,1H),8.32(s,1H),9.43(s,1H),10.90(s,1H).
1- (2-Bromoethyl) cyclopropan-1-ol
EtMgBr (3M, 43.91 mL) (3M solution in diethyl ether) was added dropwise to a solution of methyl 3-bromopropionate (10 g,59.88mmol,6.54 mL) and titanium tetraisopropoxide (17.02 g,59.88mmol,17.67 mL) in THF (400 mL) at 0℃and N 2. The mixture was stirred at 20deg.C for 3hr. The reaction mixture was quenched with saturated ammonium chloride solution (600 mL). The mixture is then addedAnd filtering to obtain filtrate. The filtrate was extracted with EtOAc (3X 500 mL). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=0/1 to 10/1) to give the title compound (7 g,42.42mmol, yield) as a yellow oil 35%).1H NMR(400MHz,DMSO-d6)δ5.15(s,1H),3.61(t,J=7.6Hz,2H),1.99(t,J=7.6Hz,2H),0.56(dd,J=6.8,4.8Hz,1H),0.43(dd,J=6.4,4.4Hz,1H).
2, 5-Dichloro-6- ((3- (2- (1-hydroxycyclopropyl) ethyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A flask containing a solution of 2, 5-dichloro-6- ((1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (1.5 g,4.49 mmol), 1- (2-bromoethyl) cyclopropan-1-ol (1.48 g,8.98 mmol), K 2CO3 (1.24 g,8.98 mmol) and KI (372.58 mg,2.24 mmol) in DMSO (15 mL) was degassed and purged 3 times with N 2 and the mixture was stirred at 100℃under an atmosphere of N 2 for 12hr. The mixture was concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: xtimate C, 10 μm 250mm 80mm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:35% -65%,20 min) to give the title compound (600 mg,1.42mmol, 32% yield, purity) as a yellow solid 99%).1H NMR(400MHz,DMSO-d6)δ0.27-0.33(m,2H),0.51-0.57(m,2H),1.76-1.82(m,2H),3.34(s,3H),3.95-4.03(m,2H),5.22(s,1H),7.14-7.17(m,1H),7.18-7.21(m,1H),7.36(d,J=1.63Hz,1H),8.35(s,1H),9.53(s,1H).
5-Chloro-2- ((3 s,5 r) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -6- ((3- (2- (1-hydroxycyclopropyl) ethyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A flask charged with a solution of 2, 5-dichloro-6- ((3- (2- (1-hydroxycyclopropyl) ethyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (150 mg, 358.61. Mu. Mol), 2- [ (3R, 5S) -4, 4-difluoro-5-methyl-3-piperidinyl ] isoindoline-1, 3-dione (150.76 mg, 537.92. Mu. Mol) and DIEA (231.74 mg,1.79mmol, 312.31. Mu.L) in DMSO (1.5 mL) was degassed and purged 3 times with N 2, and the mixture was then stirred at 80℃under an atmosphere of N 2 for 6hr. The mixture was concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: waters Xbridge BEH C100 x 30mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:40-70%,8 min) to give the title compound (90 mg, yield 28%, purity) as a yellow solid 99%).1H NMR(400MHz,DMSO-d6)δ0.21-0.29(m,2H),0.49(br d,J=5.13Hz,2H),0.84-0.89(m,3H),1.17(t,J=7.09Hz,2H),1.62-1.76(m,2H),2.13-2.28(m,1H),2.80-2.86(m,3H),3.11-3.20(m,1H),3.77-3.84(m,2H),4.14-4.38(m,4H),5.19(s,1H),6.82(d,J=8.34Hz,1H),7.05(dd,J=8.34,1.55Hz,1H),7.19(d,J=1.79Hz,1H),7.70-7.83(m,2H),7.85-7.91(m,2H),8.02(s,1H),9.11(s,1H).
2- ((3 S,5 r) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (2- (1-hydroxycyclopropyl) ethyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (59)
To a solution of 5-chloro-2- ((3 r,5 s) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -6- ((3- (2- (1-hydroxycyclopropyl) ethyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (90 mg, 135.93. Mu. Mol) in EtOH (4 mL), was added MeNH 2/H2 O (4 mL, 40% in water). The mixture was stirred at 70℃for 1hr. The mixture was concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: waters Xbridge BEH C, 100 x 30mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:30% -60%,8 min) to give the title compound (25 mg, 46.52. Mu. Mol, 34% yield, purity) as a white solid 99%).1H NMR(400MHz,DMSO-d6)δ0.22-0.31(m,2H),0.46-0.56(m,2H),0.79(d,J=6.72Hz,3H),1.60-1.71(m,2H),1.73-1.80(m,2H),1.94-2.13(m,1H),2.76-2.92(m,2H),3.33(s,3H),4.00(br t,J=7.64Hz,2H),4.14-4.24(m,1H),5.27(s,1H),7.13(d,J=8.44Hz,1H),7.23(dd,J=8.44,1.83Hz,1H),7.35(d,J=1.83Hz,1H),7.96(s,1H),8.97(s,1H).
The absolute configuration of compound 59 is randomly assigned to the cis-conformation based on amino and methyl groups.
Example 14: synthesis of 2- [ (3R, 5S) -3-amino-4, 4-difluoro-5-methyl-1-piperidinyl ] -5-fluoro-6- [ [ 1-methyl-2-oxo-3- [ [ (R) -2-oxooxazolidin-4-yl ] methyl ] benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (60) and 2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((1-methyl-2-oxo-3- (((S) -2-oxooxazolidin-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (61)
(2-Oxazolidin-4-yl) methyl 4-methylbenzenesulfonate
To a solution of 4- (hydroxymethyl) oxazolidin-2-one (12.5 g,106.74 mmol) and DMAP (26.08 g,213.49 mmol) in DCM (500 mL) was added 4-methylbenzenesulfonyl chloride (24.42 g,128.09 mmol) at 0deg.C. The mixture was stirred at 0deg.C for 1hr, and then warmed to 15deg.C and stirred for 1hr. The reaction mixture was washed with 1N HCl (1L), H 2 O (1L), saturated NaHCO 3 (1L), saturated NaCl (1L), dried over anhydrous Na 2SO4 and concentrated in vacuo to give a residue which was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=5/1 to 0/1) to give the title compound as a white solid (45 g,159.24mmol, yield 75%, purity 96%).1H NMR(400MHz,MeCN-d3)δ7.79(d,J=8Hz,2H),7.45(d,J=8Hz,2H),5.99(br s,1H),4.32-4.40(m,1H),3.93-4.07(m,4H),2.45(s,3H).
(S) - (2-Oxazolidin-4-yl) methyl 4-methylbenzenesulfonate and (R) - (2-Oxazolidin-4-yl) methyl 4-methylbenzenesulfonate
Racemic (2-oxooxazolidin-4-yl) methyl 4-methylbenzenesulfonate (10 g,36.86 mmol) is separated by SFC (column: phenomenex-Cellulose-2 (250 mm. Times.50 mm,10 μm); mobile phase: [0.1% NH 3H2 O EtOH ];: 60% -60%,7.7 min) to afford two enantiomerically pure compounds. (S) - (2-Oxazolidin-4-yl) methyl 4-methylbenzenesulfonate (4.9 g,17.70mmol, yield 48%, purity 98%) as a yellow solid .1H NMR(400MHz,MeCN-d3)δ7.80(d,J=8Hz,2H),7.46(d,J=8Hz,2H),5.95(br s,1H),4.32-4.40(m,1H),3.93-4.07(m,4H),2.45(s,3H).(R)-(2- Oxazolidin-4-yl) methyl 4-methylbenzenesulfonate (4.9 g,17.70mmol, yield 48%, purity 98%) as a yellow solid .1H NMR(400MHz,MeCN-d3)δ7.79(d,J=8.34Hz,2H),7.45(d,J=8.11Hz,2H),5.98(br s,1H),4.31-4.41(m,1H),3.92-4.08(m,4H),2.45(s,3H).
(R) -4- [ (3-methyl-6-nitro-2-oxo-benzimidazol-1-yl) methyl ] oxazolidin-2-one
A flask containing a mixture of 3-methyl-6-nitro-1H-benzimidazol-2-one (500 mg,2.59 mmol), (S) - (2-oxooxazolid-4-yl) methyl 4-methylbenzenesulfonate (842.69 mg,3.11 mmol), K 2CO3 (715.52 mg,5.18 mmol) and KI (214.85 mg,1.29 mmol) in DMSO (10 mL) was degassed and purged 3 times with N 2 and then the mixture stirred at 80℃under an atmosphere of N 2 for 3hr. The reaction mixture was treated with water (20 mL) to form a precipitate, the precipitate was filtered and the filter cake was washed with EtOAc (10 mL) and dried under reduced pressure to give the title compound as a black solid (600 mg,2.03mmol, 79% yield, 99% purity).
(R) -4- [ (6-amino-3-methyl-2-oxo-benzimidazol-1-yl) methyl ] oxazolidin-2-one
To a solution of (R) -4- [ (3-methyl-6-nitro-2-oxo-benzimidazol-1-yl) methyl ] oxazolidin-2-one (600 mg,2.05 mmol) in DMF (2 mL) was added Pd/C (200 mg, purity 10%) under Ar. The suspension was degassed under vacuum and purged 3 times with H 2. The mixture was stirred at H 2 (50 psi) and 60℃for 12 hours. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue, which was purified by column chromatography (SiO 2, ethyl acetate/ethanol=1/0 to 0/1) to give the title compound (500 mg,1.91mmol, yield) as a yellow solid 93%).1H NMR(400MHz,DMSO-d6)δ7.84(s,1H),6.89(d,J=8Hz,1H),6.61(d,J=1.8Hz,1H),6.48(dd,J=8,2Hz,1H),6.15(br s,2H),4.31-4.40(m,1H),4.09-4.19(m,2H),3.77-3.85(m,2H),3.25(s,3H).
2-Chloro-5-fluoro-6- [ [ 1-methyl-2-oxo-3- [ [ (R) -2-oxooxazolidin-4-yl ] methyl ] benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile
To a solution of (R) -4- [ (6-amino-3-methyl-2-oxo-benzimidazol-1-yl) methyl ] oxazolidin-2-one (500 mg,1.91 mmol) and 2, 6-dichloro-5-fluoro-pyridine-3-carbonitrile (364.12 mg,1.91 mmol) in DMSO (10 mL) was added DIPEA (492.79 mg,3.81mmol, 664.13. Mu.L). The mixture was stirred at 100deg.C for 2hr. The mixture was treated with water (20 mL) to form a precipitate, which was filtered and dried under reduced pressure to give the title compound (550 mg,1.17mmol, 62% yield, purity) as a brown solid 89%).1H NMR(400MHz,DMSO-d6)δ9.99(s,1H),8.14(d,J=10.4Hz,1H),7.83(s,1H),7.49(d,J=1.2Hz,1H),7.25(br dd,J=8,1.6Hz,1H),7.15(d,J=8Hz,1H),4.32-4.44(m,1H),4.14-4.24(m,2H),3.80-3.97(m,2H),3.33(br s,3H).
2- ((3R, 5 s) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((1-methyl-2-oxo-3- (((R) -2-oxooxazolidin-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a solution of 2-chloro-5-fluoro-6- [ [ 1-methyl-2-oxo-3- [ [ (R) -2-oxooxazolidin-4-yl ] methyl ] benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (500 mg,1.20 mmol), 2- [ (3R, 5S) -4, 4-difluoro-5-methyl-3-piperidinyl ] isoindoline-1, 3-dione (336.22 mg,1.20 mmol) and Cs 2CO3 (781.73 mg,2.40 mmol) in dioxane (5 mL) was added rac-BINAP-Pd-G3 (119.05 mg, 119.96. Mu. Mol). The mixture was stirred at 90℃for 4hr. Passing the reaction mixture throughPad filtration and concentration of the filtrate under reduced pressure gave a residue which was purified by preparative HPLC (column: waters Xbridge BEH C18:100:30 mm:10 μm; mobile phase: [ water (NH 4HCO3) -ACN ];B%:40-60%,8 min) to give the title compound (150 mg, yield 19%, purity) 98%).1H NMR(400MHz,MeCN-d3)δ7.87(br s,1H),7.83(dd,J=5.46,3.07Hz,2H),7.67-7.76(m,2H),7.51(d,J=10.54Hz,1H),7.09-7.22(m,2H),6.75(d,J=8.28Hz,1H),6.08(s,1H),4.48-4.57(m,1H),4.33-4.47(m,2H),4.24-4.32(m,3H),4.17-4.23(m,2H),3.69-3.87(m,2H),3.21(dd,J=14.12,12.11Hz,1H),2.84(s,3H),0.97(d,J=6.78Hz,3H).
2- [ (3R, 5S) -3-amino-4, 4-difluoro-5-methyl-1-piperidinyl ] -5-fluoro-6- [ [ 1-methyl-2-oxo-3- [ [ (R) -2-oxooxazolid-4-yl ] methyl ] benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (60)
To a solution of 2- [ (3R, 5 s) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methyl-1-piperidinyl ] -5-fluoro-6- [ [ 1-methyl-2-oxo-3- [ [ (R) -2-oxooxazolid in-4-yl ] methyl ] benzoimidazol-5-yl ] amino ] pyridine-3-carbonitrile (100 mg, 151.38. Mu. Mol) in EtOH (3.5 mL) was added MeNH 2 (40% purity). The mixture was stirred at 70℃for 1hr. The reaction mixture was concentrated under reduced pressure to give a residue which was purified by preparative HPLC (column: waters Xbridge BEH C18.100.30 mm. 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ];B%:25% -45%,8 min) to give the title compound (10 mg, 18.66. Mu. Mol, yield 12%, purity) as a white solid 99%).1H NMR(400MHz,MeCN-d3)δ7.81(br d,J=1.13Hz,1H),7.55(d,J=1.88Hz,1H),7.46-7.50(m,1H),7.22(dd,J=8.44,1.94Hz,1H),7.04(d,J=8.50Hz,1H),6.36(br s,1H),4.36-4.46(m,1H),4.22-4.32(m,2H),4.14-4.21(m,1H),4.01-4.08(m,1H),3.94-4.00(m,2H),3.36(s,3H),3.00-3.16(m,1H),2.78-2.90(m,2H),2.18-2.25(m,1H),0.98(d,J=6.88Hz,3H).
(S) -4- ((3-methyl-6-nitro-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-1-yl) methyl) oxazolidin-2-one
A flask charged with a mixture of 3-methyl-6-nitro-1H-benzimidazol-2-one (500 mg,2.59 mmol), [ (4R) -2-oxooxazolid-4-yl) methyl 4-methylbenzenesulfonate (842.69 mg,3.11 mmol), K 2CO3 (715.52 mg,5.18 mmol), and KI (214.85 mg,1.29 mmol) in DMSO (10 mL) was degassed and purged 3 times with N 2 and then the mixture stirred at 80℃under an atmosphere of N 2 for 3hr. The reaction mixture was treated with water (20 mL) to form a precipitate, the precipitate was filtered and the filter cake was washed with EtOAc (10 mL) and then dried under reduced pressure to give the title compound as a black solid (600 mg,2.03mmol, 79% yield, 99% purity).
(S) -4- ((6-amino-3-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-1-yl) methyl) oxazolidin-2-one
To a solution of (4R) -4- [ (3-methyl-6-nitro-2-oxo-benzimidazol-1-yl) methyl ] oxazolidin-2-one (600 mg,2.05 mmol) in DMF (2 mL) was added Pd/C (200 mg, purity 10%) under Ar. The flask was degassed and purged 3 times with H 2. The mixture was stirred at H 2 (50 psi) and 60℃for 10 hours. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give a residue, which was purified by column chromatography (SiO 2, ethyl acetate/ethanol=1/0 to 0/1) to give the title compound (500 mg,1.70mmol, yield 83%, purity) as a yellow solid 89%).1H NMR(400MHz,DMSO-d6)δ7.85(s,1H),6.80(d,J=8.23Hz,1H),6.47(d,J=1.67Hz,1H),6.34(dd,J=8.23,1.79Hz,1H),4.76(br s,2H),4.30-4.39(m,1H),4.07-4.20(m,2H),3.78(br d,J=5.72Hz,2H),3.23(s,3H).
(S) -2-chloro-5-fluoro-6- ((1-methyl-2-oxo-3- ((2-oxooxazolidin-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a solution of (4S) -4- [ (6-amino-3-methyl-2-oxo-benzimidazol-1-yl) methyl ] oxazolidin-2-one (500 mg,1.91 mmol) and 2, 6-dichloro-5-fluoro-pyridine-3-carbonitrile (364.12 mg,1.91 mmol) in DMSO (10 mL) was added DIEA (492.79 mg,3.81mmol, 664.13. Mu.L). The mixture was stirred at 100deg.C for 2hr. The mixture was treated with water (20 mL) to form a precipitate, which was filtered and dried under reduced pressure to give the title compound (560 mg,1.25mmol, 66% purity) as a brown solid 93%).1H NMR(400MHz,DMSO-d6)δ9.98(br s,1H),8.13(d,J=10.61Hz,1H),7.83(s,1H),7.49(s,1H),7.10-7.33(m,2H),4.31-4.46(m,1H),4.19(br d,J=5.25Hz,2H),3.88(br dd,J=12.22,5.30Hz,2H),3.33(s,3H).
2- ((3 R, 5S) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((1-methyl-2-oxo-3- (((S) -2-oxooxazolidin-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a solution of 2-chloro-5-fluoro-6- [ [ 1-methyl-2-oxo-3- [ [ (4S) -2-oxooxazolidin-4-yl ] methyl ] benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (500 mg,1.20 mmol), 2- [ (3R, 5S) -4, 4-difluoro-5-methyl-3-piperidinyl ] isoindoline-1, 3-dione (336.22 mg,1.20 mmol) and Cs 2CO3 (781.73 mg,2.40 mmol) in dioxane (5 mL) was added rac-BINAP-Pd-G3 (119.05 mg, 119.96. Mu. Mol). The mixture was stirred at 90℃for 4hr. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue which was purified by preparative HPLC (column: waters Xbridge BEH C18.100.30 mm.10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:30% -60%,8 min) to give the title compound (120 mg, 174.39. Mu. Mol, yield 15%, purity) as a yellow solid 96%).1H NMR(400MHz,MeCN-d3)δ7.90(br d,J=1.00Hz,1H),7.83(dd,J=5.46,3.07Hz,2H),7.74(br s,2H),7.51(d,J=10.54Hz,1H),7.21(d,J=1.88Hz,1H),7.10(dd,J=8.28,1.76Hz,1H),6.81(d,J=8.41Hz,1H),5.94(s,1H),4.45-4.57(m,1H),4.32-4.44(m,2H),4.22-4.32(m,3H),4.11-4.22(m,2H),3.75(dd,J=14.56,6.15Hz,1H),3.60-3.68(m,1H),3.15-3.25(m,1H),2.98(s,3H),0.97(d,J=6.78Hz,3H).
2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((1-methyl-2-oxo-3- (((S) -2-oxooxazolidin-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (61)
To a solution of 2- [ (3R, 5S) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methyl-1-piperidinyl ] -5-fluoro-6- [ [ 1-methyl-2-oxo-3- [ [ (4S) -2-oxooxazolid in-4-yl ] methyl ] benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (100.00 mg, 151.38. Mu. Mol) in EtOH (5 mL) was added MeNH 2 (5 mL, purity 40%). The mixture was stirred at 70℃for 1hr. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by preparative HPLC (column: phenomnex C1875X 30mm X3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:25% -45%,8 min) to give the title compound (10 mg, 18.66. Mu. Mol, yield 12%, purity) as a white solid 99%).1H NMR(400MHz,DMSO-d6)δ9.45(br s,1H),7.78-7.90(m,2H),7.55(s,1H),7.26-7.33(m,1H),7.12(d,J=8.34Hz,1H),4.33-4.41(m,1H),4.16-4.25(m,2H),4.06-4.15(m,1H),3.94-3.99(m,1H),3.91(br s,2H),3.33(br s,3H),2.89-3.04(m,1H),2.70-2.84(m,2H),2.08-2.20(m,1H),1.65-1.79(m,2H),0.88(br d,J=6.68Hz,3H).
The absolute configuration of compounds 60 and 61 is randomly assigned to the cis conformation based on amino and methyl.
Example 15: synthesis of 5-chloro-2- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-3- ((3-methyloxetan-3-yl) methoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) nicotinonitrile (6)
4-Methylbenzenesulfonic acid oxetane-2-methyl ester
To a solution of oxetan-2-ylmethanol (200 mg,2.27 mmol) and 4-methylbenzenesulfonyl chloride (519.33 mg,2.72 mmol) in DCM (3 mL) was added DMAP (27.73 mg, 227.00. Mu. Mol) and TEA (459.40 mg,4.54mmol, 631.92. Mu.L). The mixture was stirred at 20deg.C for 12hr. The mixture was concentrated under reduced pressure to give a residue, which was purified by preparative TLC (SiO 2, petroleum ether: ethyl acetate=2:1) to give the title compound as a white solid (440 mg,1.82mmol, yield 80%).1H NMR(400MHz,CDCl3)δ7.83(d,J=8.4Hz,2H),7.36(d,J=8.0Hz,2H),4.96-4.90(m,1H),4.64-4.58(m,1H),4.54-4.89(m,1H),4.16(d,J=4.0Hz,2H),2.76-2.67(m,1H),2.62-2.53(m,1H),2.46(s,3H).
2- ((6- ((3-Chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (oxetan-2-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid methyl ester
A mixture of methyl 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3S, 5R) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] acetate (100 mg, 187.99. Mu. Mol), oxetan-2-methyl 4-methylbenzenesulfonate (136.65 mg, 563.98. Mu. Mol), K 2CO3 (77.94 mg, 563.98. Mu. Mol), KI (94.00. Mu. Mol) in DMSO (1 mL) was stirred at 80℃under an atmosphere of N 2 for 12hr. Water (3 mL) was added to form a precipitate, the precipitate was filtered and washed with H 2 O (3 mL) and ethyl acetate (5 mL), and the solid was then dried under reduced pressure to give the title compound (100 mg, crude) as a brown solid.
2- ((6- ((3-Chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (oxetan-2-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (6)
To a mixture of methyl 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3 s,5 r) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -1- (oxetan-2-ylmethyl) -2-oxo-3-quinolinyl ] oxy ] acetate (100 mg,166.11 μmol) in EtOH (5 mL), hexamethylenetetramine (purity 40% in h 2 O) was added. The mixture was stirred at 70℃for 12hr. The reaction mixture was concentrated under reduced pressure to give a residue which was purified by preparative HPLC (column: waters Xbridge BEH C18:100 x 25mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ];% B: 45% -75%,10 min) to give the title compound (5 mg, 7.51. Mu. Mol, yield 5%, purity) as a white solid 90%).1HNMR(400MHz,DMSO-d6)δ9.12(S,1H),7.99(S,1H),7.94(d,J=4.4Hz,1H),7.74(d,J=2.4Hz,1H),7.69(d,J=9.2Hz,1H),7.61(dd,J=2.4,9.2Hz,1H),7.24(s,1H),5.05-5.01(m,1H),4.68-4.50(m,2H),4.54(s,2H),4.50-4.40(m,2H),4.14(d,J=12.8Hz,2H),2.82-2.74(m,2H),2.67-2.66(m,4H),2.12–2.00(m,2H),1.23(s,1H),0.84(d,J=6.8Hz,6H).
Example 16: synthesis of 2- ((6- (3-chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (3-hydroxypropyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (18)
2- ((1- (3- ((Tert-Butyldimethylsilanyloxy) propyl) -6- ((3-chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid methyl ester
A mixture of methyl 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3S, 5R) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] acetate (100 mg, 187.99. Mu. Mol), tert-butyl- (3-iodopropoxy) -dimethyl-silane (56.44 mg, 187.99. Mu. Mol, 5.41. Mu. L), K 2CO3 (51.96 mg, 375.98. Mu. Mol) in DMSO (2 mL) was stirred at 80℃for 12hr. The mixture was cooled to 20 ℃ and water (1 mL) was added to form a precipitate, which was filtered and the filter cake was washed with H 2 O (10 mL x 2), etOAc (10 mL x 2) and then concentrated to give the title compound (150 mg, crude) as a yellow solid, which was used without purification.
2- ((1- (3- ((Tert-butyldimethylsilyloxy) propyl) -6- ((3-chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
A mixture of methyl 2- [ [1- [3- [ tert-butyl (dimethyl) silyl ] oxypropyl ] -6- [ [ 3-chloro-5-cyano-6- [ (3S, 5R) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -2-oxo-3-quinolinyl ] oxy ] acetate (150 mg, 85.19. Mu. Mol, purity 40%), purity 40% in MeNH 2(370.59μmol,9mL,H2 O) in EtOH (10 mL) was stirred at 70℃for 12hr. The mixture was concentrated to give the title compound (100 mg, 56.88. Mu. Mol, 67% yield, 40% purity) as a yellow solid, which was used without further purification.
2- ((6- ((3-Chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (3-hydroxypropyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (18)
To a solution of 2- [ [1- [3- [ tert-butyl (dimethyl) silyl ] oxypropyl ] -6- [ [ 3-chloro-5-cyano-6- [ (3 s,5 r) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -2-oxo-3-quinolinyl ] oxy ] -N-methylacetamide (100 mg,142.19 μmol) in THF (2 mL), and then the mixture was stirred at 20 ℃ for 12hr. The mixture was concentrated and the residue was purified directly by preparative HPLC (column: phenomenex Luna 80 x 30mm x 3 μm; mobile phase: [ water (HCl) -ACN ]; B%:30% -60%,8 min) to give the title compound (6 mg, 9.50. Mu. Mol, yield 7%, purity) as a white solid 99%,HCl).1H NMR(400MHz,DMSO-d6)δppm 9.08-9.18(m,1H),7.95-8.04(m,2H),7.78(d,J=2.38Hz,1H),7.61-7.68(m,1H),7.50-7.57(m,1H),7.22-7.28(m,1H),4.63-4.78(m,1H),4.50-4.60(m,2H),4.27-4.40(m,2H),4.13(br d,J=12.51Hz,2H),3.54(br t,J=5.69Hz,2H),2.79(br t,J=12.76Hz,2H),2.67(d,J=4.50Hz,3H),1.96-2.15(m,2H),1.75-1.86(m,2H),0.82-0.86(m,6H).
Example 17: synthesis of 2- ((6- ((3-chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (62) and 2- ((6- ((3-chloro-5-cyano-6- ((3S, 5R) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (63)
2- ((3 R,5 s) -1-benzyl-4, 4-difluoro-5-methylpiperidin-3-yl) ethanol and 2- ((3 s,5 r) -1-benzyl-4, 4-difluoro-5-methylpiperidin-3-yl) ethanol
2- (1-Benzyl-4, 4-difluoro-5-methyl-3-piperidinyl) ethanol (1.2 g,4.46 mmol) was isolated by SFC (column: phenomenex-Cellulose-2 (250 mm. Times.30 mm,5 μm); mobile phase; [ n-heptane-IPA (0.1% NH3.H 2 O) ];B%:10% -10%,12 min) to give 2- [ (3R, 5S) -1-benzyl-4, 4-difluoro-5-methyl-3-piperidinyl ] ethanol (540 mg,1.98mmol, 44% yield), 99%);1H NMR(400MHz,DMSO-d6)δ=7.33–7.25(m,5H),4.50(t,J=5.2Hz,1H),3.58–3.50(m,1H),3.47–3.33(m,3H),2.95(d,J=10Hz,2H),2.72(d,J=10Hz,2H),2.20–2.04(m,2H),1.91-1.81(m,3H),1.25-1.19(m,1H),0.87(d,J=6.8Hz,3H); purity and colorless oily 2- [ (3S, 5R) -1-benzyl-4, 4-difluoro-5-methyl-3-piperidinyl ] ethanol (510 mg,1.79mmol, 40% purity) 94%);1HNMR(400MHz,DMSO-d6)δ7.35–7.25(m,5H),4.50(t,J=5.2Hz,1H),3.58–3.50(m,1H),3.47–3.33(m,3H),2.95(d,J=10Hz,2H),2.72(d,J=10Hz,2H),2.20–2.04(m,2H),1.91-1.81(m,3H),1.25-1.19(m,1H),0.87(d,J=6.8Hz,3H).
2- ((3R, 5S) -4, 4-difluoro-5-methylpiperidin-3-yl) ethanol
To a solution of 2- [ (3R, 5S) -1-benzyl-4, 4-difluoro-5-methyl-3-piperidinyl ] ethanol (200 mg, 742.59. Mu. Mol) in MeOH (2 mL) was added TFA (338.69 mg,2.97mmol, 219.93. Mu.L) and Pd/C (10%, 50 mg) under Ar. The mixture was stirred at H 2 (15 psi) and 60℃for 12 hours. The reaction mixture was filtered and the filtrate was concentrated to give the title compound (140 mg, crude) as a yellow oil ,TFA).1H NMR(400MHz,MeOD-d4)δ3.66-3.63(m,2H),3.62-3.60(m,1H),3.41(d,J=12Hz,1H),2.99-2.86(m,2H),2.50-2.33(m,2H),2.09–2.00(m,1H),1.52-1.43(m,1H),1.09(d,J=6.8Hz,3H).
2- ((6- ((3-Chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (62)
To a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methylacetamide (40 mg,92.54 μmol) and 2- [ (3 r,5 s) -4, 4-difluoro-5-methyl-3-piperidinyl ] ethanol (54.27 mg,185.07 μmol, TFA) in DMSO (0.5 mL) was added DIPEA (47.84 mg,370.15 μmol,64.47 μL). The mixture was stirred at 100deg.C for 3hr. The mixture was concentrated under reduced pressure. The residue was purified by preparative HPLC (column: phenomenex C1880 x 40mm x 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:30% -60%,8 min) to give the title compound (8 mg,13.63 μmol, 15% yield, purity) as a white solid 98%).1H NMR(400MHz,DMSO-d6)δ9.19(s,1H),8.06(s,1H),8.00(d,J=4.4Hz,1H),7.83(d,J=2.4Hz,1H),7.71(dd,J=2.4,8.8Hz,1H),7.39(d,J=9.2Hz,1H),7.30(s,1H),4.61(s,2H),4.60-4.57(m,1H),4.42(d,J=13.2Hz,1H),4.17(d,J=12.4Hz,1H),3.74(s,3H),2.90-2.76(m,2H),2.73(d,J=4.8Hz,3H),2.23–2.01(m,3H),1.89-1.80(m,1H),1.36-1.27(m,2H),0.89(d,J=6.4Hz,3H).
2- ((3 S,5 r) -4, 4-difluoro-5-methylpiperidin-3-yl) ethanol
To a solution of 2- [ (3S, 5R) -1-benzyl-4, 4-difluoro-5-methyl-3-piperidinyl ] ethanol (200 mg, 742.59. Mu. Mol) in MeOH (2 mL) was added TFA (338.69 mg,2.97mmol, 219.93. Mu.L) and Pd/C (10%, 50 mg) under Ar. The mixture was stirred at H 2 (15 psi) and 60℃for 12 hours. The reaction mixture was filtered and the filtrate was concentrated to give the title compound (120 mg, crude) as a yellow oil ,TFA).1H NMR(400MHz,MeOD-d4)δ4.43-4.34(m,2H),3.71-3.68(m,1H),3.45–3.42(m,1H),3.08-2.93(m,2H),2.57-2.43(m,2H),2.08–2.00(m,1H),1.51–1.42(m,1H),1.07(d,J=6.8Hz,3H).
2- ((6- ((3-Chloro-5-cyano-6- ((3 s,5 r) -4, 4-difluoro-3- (2-hydroxyethyl) -5-methylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (63)
To a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methyl-acetamide (40 mg, 92.54. Mu. Mol) and 2- [ (3 s,5 r) -4, 4-difluoro-5-methyl-3-piperidinyl ] ethanol (54.27 mg, 185.07. Mu. Mol, TFA) in DMSO (0.5 mL) was added DIPEA (35.88 mg, 277.61. Mu. Mol, 48.35. Mu.l). The mixture was stirred at 100deg.C for 3hr. The mixture was purified by preparative HPLC (column: waters Xbridge BEH C, 100 x 30mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:25% -55%,8 min) to give the title compound (11 mg, 18.78. Mu. Mol, 20% yield, purity) as a white solid 98%).1H NMR(400MHz,DMSO-d6)δ9.13(s,1H),8.00(s,1H),7.92(d,J=4.4Hz,1H),7.77(d,J=2.4Hz,1H),7.64(dd,J=2.4,9.2Hz,1H),7.48(d,J=9.2Hz,1H),7.24(s,1H),4.55(s,2H),4.52(t,J=5.2Hz,1H),4.36(d,J=14Hz,1H),4.11(d,J=11.2Hz,1H),3.68(s,3H),2.83-2.72(m,2H),2.66(d,J=4.8Hz,3H),2.16-1.94(m,3H),1.82-1.74(m,1H),1.28-1.23(m,2H),0.82(d,J=6.8Hz,3H).
The absolute configuration of compounds 62 and 63 is randomly assigned to the cis conformation based on the aliphatic alcohol group and methyl group.
Example 18: synthesis of 2- ((6- ((3-chloro-5-cyano-6- ((3R, 4r, 5S) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (64)
N-methyl-2- ((1-methyl-6-nitro-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetamide
A mixture of 3-hydroxy-1-methyl-6-nitroquinolin-2 (1H) -one (5 g,22.71 mmol), 2-bromo-N-methylacetamide (4.14 g,27.25 mmol) and Cs 2CO3 (14.80 g,45.42 mmol) in DMF (100 mL) was stirred at 20℃for 12hr. The mixture was then poured into water (150 mL), the precipitated solid was filtered and washed with H 2 O (200 mL) and MTBE (200 mL), and the solid was then dried under reduced pressure to give the title compound (3.3 g,10.51mmol, yield 46%, purity 93%) as a yellow solid.
2- ((6-Amino-1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
Pd/C (1 g,11.33mmol, purity 10%) was added to a solution of N-methyl-2- ((1-methyl-6-nitro-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetamide (3.3 g,11.33 mmol) in DMF (60 mL) under an argon atmosphere. The mixture was stirred at H 2 (50 psi) and 50deg.C for 12hr. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to give the title compound (1.8 g,6.13mmol, yield 54%, purity 89%) as a yellow oil.
2- ((6- ((3, 6-Dichloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
A mixture of 2- ((6-amino-1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (1 g,3.83 mmol), 2,5, 6-trichloropyridine-3-carbonitrile (793.97 mg,3.83 mmol), DIEA (989.32 mg,7.65mmol,1.33 mL) in DMF (20 mL) was degassed and purged 3 times with N 2, and then the mixture was stirred at 100℃under an atmosphere of N 2 for 12hr. The mixture was cooled to 20 ℃. Water (10 mL) was added to form a precipitate, which was filtered and the filter cake was washed with EtOAc (20 mL) and dried in vacuo to give the title compound as a white solid (1.1 g,1.35mmol, yield 35%, purity 53%).1H NMR(400MHz,DMSO-d6)δ9.64(s,1H),8.38(s,1H),7.94(dd,J=6.60,1.59Hz,1H),7.74(s,1H),7.66(d,J=8.93Hz,1H),7.53(d,J=9.29Hz,1H),7.22(s,1H),4.58(s,2H),3.69(s,3H),2.66(s,3H).
2- ((6- ((3-Chloro-5-cyano-6- ((3 r,4r,5 s) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (64)
To a solution of 2- ((6- ((3, 6-dichloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (50 mg, 115.67. Mu. Mol) and (3 s,4R, 5R) -4-fluoro-3, 5-dimethylpiperidine (38.78 mg, 231.34. Mu. Mol, HCl) in DMSO (1 mL) was added DIEA (74.75 mg, 578.36. Mu. Mol, 100.74. Mu.L) under N 2. The mixture was stirred at 100deg.C for 12hr. The mixture was purified by preparative HPLC (column: phenomenex C18.80.40 mm.3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:35% -65%,8 min) to give the title compound (12 mg,22.07 μmol, 19% yield, purity) as a white solid 97%).1H NMR(400MHz,DMSO-d6)δ9.09(s,1H),7.92-7.97(m,2H),7.81(d,J=2.20Hz,1H),7.64(dd,J=8.99,2.38Hz,1H),7.46(d,J=9.05Hz,1H),7.23(s,1H),4.53(s,2H),4.09-4.18(m,2H),3.80-3.99(m,1H),3.67(s,3H),2.64-2.71(m,5H),1.73(dd,J=9.29,4.65Hz,2H),0.87(d,J=6.48Hz,6H).
Example 19: synthesis of 2- ((6- ((3-chloro-5-cyano-6- ((3R, 4r, 5S) -4-hydroxy-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (47) and 2- ((6- ((3-chloro-5-cyano-6- ((3R, 4s, 5S) -4-hydroxy-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (46)
(3R, 5S) -benzyl 3, 5-dimethyl-4-oxopiperidine-1-carboxylic acid
To a solution of (3S, 5R) -1-benzyl-3, 5-dimethyl-piperidin-4-one (400 mg,1.84 mmol) in toluene (4 mL) was added benzyl chloroformate (480.44 mg,2.82mmol, 400.37. Mu.L). The mixture was stirred at 110deg.C for 12hr. The residue was purified by flash column chromatography on silica gel (SILICA FLASH column, 0% -30% ethyl acetate/petroleum eluent) to give the title compound as a white solid (300 mg,1.04mmol, 57% yield, purity) 91%).1H NMR(400MHz,CDCl3)δ7.41-7.32(m,5H),5.20(d,J=4.4Hz,2H),4.50-4.41(m,2H),2.73-2.58(m,4H),1.03(d,J=6.4Hz,6H).
(3S, 4R, 5R) -4-hydroxy-3, 5-dimethylpiperidine-1-carboxylic acid benzyl ester
To a solution of (3 s,5 r) -3, 5-dimethyl-4-oxo-piperidine-1-carboxylic acid benzyl ester (4.1 g,15.69 mmol) in MeOH (45 mL) was added NaBH 4 (712.30 mg,18.83 mmol) at 0 ℃. The mixture was stirred at 20deg.C for 12hr. The reaction mixture was quenched by addition of 1N HCl (10 mL) at 0deg.C, and the mixture was concentrated under reduced pressure. The residue was dissolved in ethyl acetate (20 mL) and washed with water (10 mL x 3), the combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: agela DuraShell C18:250:80 mm 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:25% -50%,20 min) to give benzyl (3S, 4S, 5R) -4-hydroxy-3, 5-dimethyl-piperidine-1-carboxylate (1.4 g,5.26mmol, yield 34%, purity 99%);1H NMR(400MHz,MeOD-d4)δ7.36-7.31(m,5H),5.11(s,2H),4.09-4.04(m,2H),2.74(t,J=9.6Hz,1H),2.52-2.46(m,2H),1.50-1.39(m,2H),0.99(d,J=6.0Hz,6H); and benzyl (3S, 4R, 5R) -4-hydroxy-3, 5-dimethyl-piperidine-1-carboxylate (0.9 g,3.35mmol, yield 21%, purity) as a colorless oil 98%);1H NMR(400MHz,MeOD-d4)δ=7.36-7.29(m,5H),5.10(s,2H),3.77(dd,J=4.4,13.2Hz,2H),3.54(s,1H),2.77-2.72(m,2H),1.70-1.60(m,2H),0.94(d,J=6.0Hz,6H).
(3R, 4R, 5S) -3, 5-dimethylpiperidin-4-ol
To a solution of benzyl (3S, 5R) -4-hydroxy-3, 5-dimethylpiperidine-1-carboxylate (200 mg, 759.50. Mu. Mol) in MeOH (2 mL) under Ar was added TFA (346.39 mg,3.04mmol, 224.93. Mu.L) and Pd/C (50 mg, purity 10%). The mixture was stirred at H 2 (15 psi) and 20deg.C for 12hr. The mixture was filtered, and the filtrate was concentrated under reduced pressure to give the title compound (150 mg, crude product) as a yellow oil ,TFA).1H NMR(400MHz,DMSO-d6)δ4.33(s,2H),3.18-3.16(m,2H),2.76-2.71(m,1H),2.62-2.53(m,2H),1.68-1.57(m,2H),0.92(d,J=6.8Hz,6H).
2- ((6- ((3-Chloro-5-cyano-6- ((3 r,4r,5 s) -4-hydroxy-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (47)
To a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methyl-acetamide (50 mg,115.67 μmol) and (3 s,4r,5 r) -3, 5-dimethylpiperidin-4-ol (45.00 mg,185.02 μmol, TFA) in DMSO (1 mL) was added DIPEA (74.75 mg,578.36 μmol,100.74 μl). The mixture was stirred at 100deg.C for 3hr. To the reaction mixture was added water (2 mL) to form a precipitate, the precipitate was filtered, and the filter cake was washed with water (5 mL) and EtOAc (5 mL x 2), and the filter cake was dried under reduced pressure to give the title compound (30 mg, 56.21. Mu. Mol, yield 49% purity) as a white solid 98%).1H NMR(400MHz,DMSO-d6)δ9.04(s,1H),7.95-7.92(m,2H),7.83(s,1H),7.66-7.64(m,1H),7.46-7.44(m,1H),7.21(s,1H),4.61(d,J=4.8Hz,1H),4.53(s,2H),4.14(d,J=11.2Hz,2H),3.67(s,3H),2.66-2.61(m,6H),1.42(s,2H),0.83(s,6H).
(3R, 4S, 5S) -3, 5-dimethylpiperidin-4-ol
To a solution of benzyl (3 s,4s,5 r) -4-hydroxy-3, 5-dimethyl-piperidine-1-carboxylate (200 mg, 759.50. Mu. Mol) in MeOH (2 mL) was added TFA (346.39 mg,3.04mmol, 224.93. Mu.l) and Pd/C (50 mg, purity 10%) under Ar atmosphere. The mixture was stirred at H 2 (15 psi) and 20deg.C for 12hr. The mixture was filtered, and the filtrate was concentrated under reduced pressure to give the title compound (150 mg, crude product) as a yellow oil ,TFA).1H NMR(400MHz,MeOD-d4)δ3.05(s,1H),2.74-2.70(m,2H),2.61–2.55(m,2H),1.69–1.65(m,2H),0.74(d,J=6.8Hz,6H).
2- ((6- ((3-Chloro-5-cyano-6- ((3 r,4s,5 s) -4-hydroxy-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (46)
To a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methyl-acetamide (25 mg, 57.84. Mu. Mol) and (3 s,4s,5 r) -3, 5-dimethylpiperidin-4-ol (21.10 mg, 86.75. Mu. Mol, TFA) in DMSO (0.5 mL) was added DIPEA (37.37 mg, 289.18. Mu. Mol, 50.37. Mu.l). The mixture was stirred at 100deg.C for 3hr. To the reaction mixture was added water (2 mL) to form a precipitate, the precipitate was filtered, and the filter cake was washed with water (5 mL), etOAc (5 mL x 2), and dried under reduced pressure to give the title compound (9.4 mg, 17.52. Mu. Mol, yield 30% purity as a white solid 98%).1H NMR(400MHz,DMSO-d6)δ9.00(s,1H),7.95(s,1H),7.89(s,1H),7.83(s,1H),7.67-7.64(m,1H),7.46-7.44(m,1H),7.20(s,1H),4.57-4.53(m,3H),3.87(d,J=11.2Hz,2H),3.67(s,3H),3.43(s,1H),2.83(t,J=12.3Hz,2H),2.67(m,3H),1.66(s,2H),0.78(d,J=6.4Hz,6H).
Example 20: synthesis of 2- ((6- ((3-chloro-5-cyano-6- ((3R, 4S, 5S) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (45)
(3R, 4S, 5S) -benzyl 4-fluoro-3, 5-dimethylpiperidine-1-carboxylate
DAST (367.27 mg,2.28mmol, 301.04. Mu.L) was added to a solution of (3S, 4S, 5R) -4-hydroxy-3, 5-dimethyl-piperidine-1-carboxylic acid benzyl ester (300.00 mg,1.14 mmol) in DCM (3 mL) at-65 ℃. The mixture was then stirred at 20℃for 12hr. Saturated NaHCO 3 was added to the mixture until pH 8-9 was reached, and then extracted with ethyl acetate (10 ml x 2). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel (SILICA FLASH column, 0% -10% ethyl acetate/petroleum eluent). The product was further purified by p-TLC (25% ethyl acetate/petroleum eluent) to give the title compound (50 mg, 150.76. Mu. Mol, 13% yield, purity) as a yellow oil 80%).1H NMR(400MHz,MeOD-d4)δ7.40-7.28(m,5H),5.11(s,2H),4.44(d,J=10Hz,1H),4.10-4.06(m,1H),3.90(dd,J=4.4,13.2Hz,1H),2.71-2.53(m,2H),1.82-1.68(m,2H),1.00-0.98(m,6H).
(3R, 4S, 5S) -4-fluoro-3, 5-dimethylpiperidine
Pd/C (10%, 8 mg) was added to a solution of benzyl (3S, 4S, 5R) -4-fluoro-3, 5-dimethyl-piperidine-1-carboxylate (40 mg, 150.76. Mu. Mol) in EtOH (1 mL) under Ar. The mixture was stirred at H 2 (15 psi) and 20℃for 12 hours. The reaction mixture was filtered, and HCl/dioxane (2 mL) was added to the filtrate and stirred at 20deg.C for 0.5hr. The solution was concentrated under reduced pressure to give the title compound (20 mg, 57.26. Mu. Mol, yield 38%, purity) as a white oil 48%,HCl).1H NMR(400MHz,MeOD-d4)δ4.61-4.49(m,1H),3.37-3.33(m,2H),3.24-3.16(m,2H),2.32-2.13(m,2H),1.11-1.10(m,6H).
2- ((6- ((3-Chloro-5-cyano-6- ((3R, 4S, 5S) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (45)
To a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methyl-acetamide (30 mg,69.40 μmol) and (3 s,4s,5 r) -4-fluoro-3, 5-dimethyl-piperidine (18.62 mg,111.04 μmol, HCl) in DMF (0.5 mL) was added DIPEA (26.91 mg,208.20 μmol,36.27 μl). The mixture was stirred at 100deg.C for 3hr. The mixture was purified by preparative HPLC (column: phenomenex C18 x 30mm x 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:35% -65%,8 min) to give the title compound (4.5 mg,8.38 μmol, 12% yield, purity) as a white solid 98%).1H NMR(400MHz,DMSO-d6)δ9.07(s,1H),7.95(s,2H),7.82(d,J=2.0Hz,1H),7.64(dd,J=2.0,9.2Hz,1H),7.46(d,J=9.2Hz,1H),7.22(s,1H),4.56-4.43(m,3H),3.98(br dd,J=3.6,13.2Hz,2H),3.67(s,3H),2.78(t,J=12.4Hz,2H),2.66(d,J=4.4Hz,3H),1.89-1.76(m,2H),0.84(d,J=6.8Hz,6H).
Example 21: synthesis of 2- ((6- ((6- ((3S, 5R) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (43) and 2- ((6- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (44)
2- ((6- ((3-Chloro-5-cyano-6- (3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
To a solution of 2- (4, 4-difluoro-5-methyl-3-piperidinyl) isoindoline-1, 3-dione (250.00 mg,892.00 μmol) and 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methylacetamide (296.60 mg,686.15 μmol) in DMSO (3 mL) was added DIPEA (177.36 mg,1.37mmol,239.03 μmol), and the mixture was then stirred at 100 ℃ for 3hr. The reaction mixture was treated with water (10 mL) to form a precipitate, which was filtered, and the filter cake was washed, collected and dried in vacuo to give the title compound (450 mg, crude) as a yellow solid.
2- ((6- ((6- (3-Amino-4, 4-difluoro-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
To a solution of 2- [ [6- [ [ 3-chloro-5-cyano-6- [3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methyl-1-piperidinyl ] -2-pyridinyl ] amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methyl-acetamide (400 mg,591.66 μmol) in EtOH (20 mL) was added 40% pure in MeNH 2(86.13mg,1.11mmol,20mL,H2 O and the reaction mixture was stirred at 70 ℃ for 12hr. The reaction mixture (combined with another batch at 50mg scale) was concentrated in vacuo and purified by preparative HPLC (column: phenomenex C18 80 x 40mm x 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ];: 25% -45%,8 min) to give the title compound as a white solid (68 mg, 98.90% purity).
2- ((6- ((6- ((3 S,5 r) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (43) and 2- ((6- ((3 r,5 s) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (44)
The racemate was separated by SFC (column: DAICEL CHIRALPAK AD (250 mm. Times.30 mm,10 μm); mobile phase: [0.1% NH3H2O ETOH ]; B%:50% -50%,15 min) and by preparative HPLC (column: C18-1.150. Times.30 mm. Times.10 μm); mobile phase: [ water (NH 4HCO3) -ACN ]; b%:20% -60%,20 min) to give 2- [ [6- [ [6- [ (3 r,5 s) -3-amino-4, 4-difluoro-5-methyl-1-piperidinyl ] -3-chloro-5-cyano-2-pyridinyl ] amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methyl-acetamide (8.5 mg,15.38 μmol, yield 12%, purity 99%);1H NMR(400MHz,DMSO-d6)δ9.08(s,1H),8.00(s,1H),7.94(m,2H),7.65(dd,J=2.4,9.2Hz,1H),7.48(d,J=9.2Hz,1H),7.36(s,1H),4.55(s,2H),4.31-4.22(d,J=9.2Hz,1H),4.16-4.06(d,J=11.6Hz,1H),3.67(s,3H),3.05-2.91(m,1H),2.87-2.74(m,2H),2.66(d,J=4.4Hz,3H),2.16-2.01(m,1H),1.94-1.61(m,2H),0.85(d,J=6.4Hz,3H); and 2- [ [6- [ (3 s,5 r) -3-amino-4, 4-difluoro-5-methyl-1-piperidinyl ] -3-chloro-5-cyano-2-pyridinyl ] amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methyl-acetamide (13 mg,23.25 μmol, yield 19%, purity) as a white solid 98%);1H NMR(400MHz,DMSO-d6)δ9.08(s,1H),8.00(s,1H),7.94(m,2H),7.65(dd,J=2.4,9.2Hz,1H),7.48(d,J=9.2Hz,1H),7.36(s,1H),4.55(s,2H),4.26(d,J=12.8Hz,1H),4.11(dd,J=2.4,14.0Hz,1H),3.67(s,3H),3.05-2.91(m,1H),2.86-2.74(m,2H),2.66(d,J=4.4Hz,3H),2.16-2.00(m,1H),1.76(s,2H),0.85(d,J=6.8Hz,3H).
The absolute configuration of compounds 43 and 44 is randomly assigned to the cis conformation based on amino and methyl.
Example 22: synthesis of 2- ((1- (azetidin-3-ylmethyl) -6- ((3-chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (65)
3- ((6- ((3-Chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -3- (2-methoxy-2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) methyl) azetidine-1-carboxylic acid tert-butyl ester
A mixture of methyl 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3S, 5R) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] acetate (100 mg, 187.99. Mu. Mol), tert-butyl 3- (iodomethyl) azetidine-1-carboxylate (55.86 mg, 187.99. Mu. Mol, 5.41. Mu. L), K 2CO3 (51.96 mg, 375.98. Mu. Mol) in DMSO (1.5 mL) was stirred at 80℃for 12hr. The mixture (combined with another batch of the same scale) was cooled to 20 ℃ and water (2 mL) was added to form a precipitate, which was filtered and washed with H 2 O (50 mL) and ethyl acetate (20 mL), then the solid was dried under reduced pressure to give the title compound (200 mg, crude) as a brown solid. LCMS: m+h + =701.3.
3- ((6- ((3-Chloro-5-cyano-6- ((3R, 5S) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -3- (2- (methylamino) -2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) methyl) azetidine-1-carboxylic acid tert-butyl ester
A mixture of 3- [ [6- [ [ 3-chloro-5-cyano-6- [ (3S, 5R) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -3- (2-methoxy-2-oxo-ethoxy) -2-oxo-1-quinolinyl ] methyl ] azetidine-1-carboxylic acid tert-butyl ester (200 mg, 285.24. Mu. Mol), meNH 2/H2 O (10 mL, purity 40%) in EtOH (10 mL) was stirred at 70℃for 12hr. The mixture was concentrated to give the title compound (250 mg, crude) as a yellow solid, which was used without further purification. LCMS: m+h + =700.2.
2- ((1- (Azetidin-3-ylmethyl) -6- ((3-chloro-5-cyano-6- ((3 r,5 s) -4, 4-difluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (65)
A mixture of 3- [ [6- [ [ 3-chloro-5-cyano-6- [ (3S, 5R) -4, 4-difluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -3- [2- (methylamino) -2-oxoethoxy ] -2-oxo-1-quinolinyl ] methyl ] azetidine-1-carboxylic acid tert-butyl ester (250 mg, 357.05. Mu. Mol) in DCM (2 mL) and TFA (1 mL) was stirred at 20℃for 1hr. The mixture was concentrated in vacuo and purified by p-HPLC (column: waters Xbridge BEH C, 100 x 25mm x 5 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:30% -60%,10 min) to give the title compound (25 mg, 37.34. Mu. Mol, yield 10%, purity) as a white solid 90%).1H NMR(400MHz,DMSO-d6)δ9.01-9.21(m,1H),8.00(s,1H),7.95(br d,J=4.63Hz,1H),7.75(d,J=2.13Hz,1H),7.61(dd,J=8.94,1.81Hz,1H),7.51(br d,J=9.01Hz,1H),7.22(s,1H),4.49-4.61(m,4H),4.12(br d,J=12.51Hz,2H),3.41(br d,J=5.00Hz,4H),2.99-3.10(m,1H),2.78(br t,J=13.01Hz,2H),2.67(d,J=4.50Hz,4H),1.96-2.17(m,2H),0.83(d,J=6.63Hz,6H).
Example 23: synthesis of 2- ((3R, 5S) -3-amino-5-methylpiperidin-1-yl) -5-chloro-6- (((S) -2-cyclopropyl-3, 3-difluoro-7-methyl-6-oxo-1, 2,3,4,6, 7-hexahydro- [1,4] oxazepan [2,3-c ] quinolin-10-yl) amino) nicotinonitrile (24) and 2- ((3S, 5R) -3-amino-5-methylpiperidin-1-yl) -5-chloro-6- (((S) -2-cyclopropyl-3, 3-difluoro-7-methyl-6-oxo-1, 2,3,4,6, 7-hexahydro- [1,4] oxazepan [2,3-c ] quinolin-10-yl) amino) nicotinonitrile (25)
4-Hydroxy-6-nitro-2-oxo-1, 2-dihydroquinoline-3-carboxylic acid ethyl ester
To a solution of 6-nitro-1H-benzo [ d ] [1,3] oxazine-2, 4-dione (20 g,96.09 mmol) and diethyl malonate (23.09 g,144.14mmol,21.78 mL) in DMF (500 mL) was added NaH (7.69 g,192.19mmol, 60% purity) at 0deg.C. The reaction mixture was warmed to 20 ℃ and stirred for 12hr. The reaction mixture was cooled to 0deg.C and water (1000 mL) was added. The aqueous mixture was neutralized to ph=7 with 1N aqueous HCI solution and the resulting mixture was stirred at 20 ℃ for 30min. The resulting yellow precipitate was filtered and washed with water (1L). The yellow precipitate was dried under reduced pressure to give the title compound (46 g,160.49mmol, yield 84%, purity of yellow solid) 97%).1H NMR(400MHz,DMSO-d6)δ12.08(s,1H),8.73(d,J=1.9Hz,1H),8.47-8.36(m,1H),7.95(s,1H),7.41(d,J=9.0Hz,1H),4.32(q,J=7.1Hz,2H),2.89(s,2H),2.73(s,3H),1.30(t,J=7.0Hz,3H).
2, 4-Dichloro-6-nitroquinoline-3-carboxylic acid ethyl ester
A mixture of 4-hydroxy-6-nitro-2-oxo-1, 2-dihydroquinoline-3-carboxylic acid ethyl ester (46 g,165.34 mmol) in POCl 3 (450 mL) was prepared and the mixture was stirred at 80℃under N 2 for 4hr. The reaction mixture was concentrated under reduced pressure to give a residue which was triturated with MTBE (100 mL) at 25℃for 30min to give the title compound as a yellow solid (37 g,114.07mmol, 69% yield, purity) 97%).1H NMR(400MHz,DMSO-d6)δ8.99(d,J=2.5Hz,1H),8.67(dd,J=2.5,9.2Hz,1H),8.31(d,J=9.2Hz,1H),4.53(q,J=7.0Hz,2H),1.39(t,J=7.1Hz,3H).
4-Chloro-6-nitro-2-oxo-1, 2-dihydroquinoline-3-carboxylic acid ethyl ester
A mixture of ethyl 2, 4-dichloro-6-nitroquinoline-3-carboxylate (35 g,111.07 mmol), naOAc (10.02 g,122.18 mmol) in AcOH (350 mL) was prepared and then the mixture was stirred at 120deg.C under N 2 atmosphere for 12hr. To the reaction mixture (combined with another batch of 2g size) was added water (500 mL) to form a precipitate, which was filtered and washed with H 2 O (1L), and the solid was dried under reduced pressure to give the title compound (25 g, purity) as a yellow solid 92%).1H NMR(400MHz,DMSO-d6)δ12.99(s,1H),8.66(d,J=2.5Hz,1H),8.49(dd,J=2.5,9.0Hz,1H),7.55(d,J=9.0Hz,1H),4.38(q,J=7.0Hz,2H),1.32(t,J=7.1Hz,3H).
4-Chloro-1-methyl-6-nitro-2-oxo-1, 2-dihydroquinoline-3-carboxylic acid ethyl ester
To a mixture of ethyl 4-chloro-6-nitro-2-oxo-1, 2-dihydroquinoline-3-carboxylate (15 g,50.56 mmol) in DMF (150 mL) was added NaH (2.83 g,70.79mmol, 60% purity) and the mixture was stirred at 0deg.C for 0.5hr, then methyl iodide (35.88 g,252.81mmol,15.74 mL) was added to the mixture and the mixture was stirred at 20deg.C for 6hr. To the mixture was added water (50 mL), a precipitate was formed, and the precipitate was filtered and washed with H 2 O (100 mL) and ethyl acetate (30 mL), and then the solid was dried under reduced pressure. The residue was purified by column chromatography (SiO 2, petroleum ether/ethyl acetate=30/1 to 2/1) to give a semi-purified product, which was further purified by reverse phase MPLC (neutral condition: chromatography column: 330g of agola c18; mobile phase: [ water-ACN ]; gradient B%:25% -50%,20min;50% -50%,20 min) to give the title compound (9.3 g,29.68mmol, yield 59%, purity) as a yellow solid 99%).1H NMR(400MHz,DMSO-d6)δ8.71(d,J=2.6Hz,1H),8.54(dd,J=2.6,9.3Hz,1H),7.87(d,J=9.4Hz,1H),4.39(q,J=7.1Hz,2H),3.69(s,3H),1.32(t,J=7.1Hz,3H).
(S) -ethyl 4- ((1-cyclopropyl-2, 2-difluoro-3-hydroxypropyl) amino) -1-methyl-6-nitro-2-oxo-1, 2-dihydroquinoline-3-carboxylic acid
To a solution of 4-chloro-1-methyl-6-nitro-2-oxo-1, 2-dihydroquinoline-3-carboxylic acid ethyl ester (500 mg,1.61 mmol) and (S) -3-amino-3-cyclopropyl-2, 2-difluoropropane-1-ol hydrochloride (413.65 mg,2.20 mmol) in MeCN (5 mL) was added DIEA (519.40 mg,4.02mmol, 700.00. Mu.L) under N 2. The mixture was stirred at 160℃for 32hr under microwaves. The mixture was concentrated in vacuo to give the title compound as a brown solid (680 mg, 799.28. Mu. Mol, yield 50%, purity 50%). LCMS: [ m+h ] + =426.1.
(S) -4- ((1-cyclopropyl-2, 2-difluoro-3-hydroxypropyl) amino) -1-methyl-6-nitroquinolin-2 (1H) -one
To a solution of (S) -ethyl 4- ((1-cyclopropyl-2, 2-difluoro-3-hydroxypropyl) amino) -1-methyl-6-nitro-2-oxo-1, 2-dihydroquinoline-3-carboxylic acid (680 mg,1.60 mmol) in MeCN (5 mL) was added NaOH (2 m,25.00 mL). The mixture was stirred at 85deg.C for 2hr. Water (20 mL) was then added followed by 3N HCl until ph=6. The precipitate formed was filtered off and the filter cake was dried under reduced pressure to give the title compound (400 mg, 520.77. Mu. Mol, 33% yield, purity) as a yellow solid 46%).1H NMR(400MHz,DMSO-d6)δ9.30(d,J=2.50Hz,1H),8.39(dd,J=9.36,2.44Hz,1H),7.62(d,J=9.42Hz,1H),7.47(d,J=8.70Hz,1H),5.72(s,1H),5.59(t,J=6.14Hz,1H),3.72-3.89(m,2H),3.55(s,3H),3.46-3.53(m,1H),1.29-1.38(m,1H),0.64-0.72(m,1H),0.60(dq,J=9.40,4.77Hz,1H),0.46-0.54(m,1H),0.24(dq,J=9.55,4.84Hz,1H).
(S) -3-bromo-4- ((1-cyclopropyl-2, 2-difluoro-3-hydroxypropyl) amino) -1-methyl-6-nitroquinolin-2 (1H) -one
To a solution of (S) -4- ((1-cyclopropyl-2, 2-difluoro-3-hydroxypropyl) amino) -1-methyl-6-nitroquinolin-2 (1H) -one (400 mg,1.13 mmol) and NBS (201.49 mg,1.13 mmol) in DCM (8 mL) was added TFA (645.42 mg,5.66mmol, 419.10. Mu.L) dropwise at 0deg.C and N 2. The mixture was stirred at 0deg.C for 0.5hr. Saturated NaHCO 3 (50 mL) was added to the mixture and extracted with DCM (20 mL x 3). The combined organic phases were washed with brine (20 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. Passing the residue through flash silica gel column chromatography20g/>SILICA FLASH column, 0% -30% ethyl acetate/petroleum ether gradient eluent @100 mL/min) to give the title compound (400 mg, 832.92. Mu. Mol, 74% yield, purity) as a white solid 90%).1H NMR(400MHz,DMSO-d6)δ8.95(d,J=2.45Hz,1H),8.43(dd,J=9.35,2.51Hz,1H),7.75(d,J=9.41Hz,1H),5.85(d,J=11.13Hz,1H),5.62(t,J=5.14Hz,1H),3.96-4.06(m,1H),3.77-3.90(m,2H),3.71(s,3H),1.20-1.33(m,1H),0.52-0.70(m,3H),0.41-0.51(m,1H).
(S) -2-cyclopropyl-3, 3-difluoro-7-methyl-10-nitro-1, 2,3, 4-tetrahydro- [1,4] oxazepino [2,3-c ] quinolin-6 (7H) -one
To a solution of ((S) -3-bromo-4- ((1-cyclopropyl-2, 2-difluoro-3-hydroxypropyl) amino) -1-methyl-6-nitroquinolin-2 (1H) -one (400 mg, 925.46. Mu. Mol) in THF (10 mL) at 20 ℃ C., N 2) was added t-BuLi (1.3M, 1.14 mL) (1.3M in pentane.) the mixture was stirred at 60 ℃ C. For 0.25hr, water (40 mL) was added and the aqueous mixture was extracted with CH 2Cl2 (20 mL x 2.) the organic extracts were combined, washed with brine (50 mL), dried over anhydrous Na 2SO4, filtered, and concentrated in vacuo the residue was purified by flash column chromatography20g/>SILICA FLASH column, 0% -30% ethyl acetate/Petroleum ether gradient eluent @100 mL/min) to afford the title compound as a yellow solid (250 mg, 711.63. Mu. Mol, yield 77%).1H NMR(400MHz,DMSO-d6)δ9.11(d,J=2.41Hz,1H),8.34(dd,J=9.32,2.52Hz,1H),7.65(d,J=9.43Hz,1H),7.01(d,J=3.73Hz,1H),4.34-4.56(m,2H),3.61(s,3H),3.22-3.30(m,1H),1.29-1.40(m,1H),0.68-0.77(m,1H),0.49-0.59(m,2H),0.30-0.38(m,1H).
(S) -10-amino-2-cyclopropyl-3, 3-difluoro-7-methyl-1, 2,3, 4-tetrahydro- [1,4] oxazepino [2,3-c ] quinolin-6 (7H) -one
To a Pd/C (0.05 g, 768.56. Mu. Mol, purity 10%) mixture in THF (10 mL) under Ar was added (S) -2-cyclopropyl-3, 3-difluoro-7-methyl-10-nitro-1, 2,3, 4-tetrahydro- [1,4] oxaazepino [2,3-C ] quinolin-6 (7H) -one (250 mg, 711.63. Mu. Mol). The suspension was degassed under vacuum and purged several times with H 2. The mixture was stirred at H 2 (15 psi) and 60℃for 2 hours. The mixture was filtered, and the filtrate was concentrated in vacuo to give the title compound as a yellow solid (150 mg, 261.42. Mu. Mol, 37% yield, 56% purity). LCMS: [ m+h ] + =322.1.
(S) -2, 5-dichloro-6- ((2-cyclopropyl-3, 3-difluoro-7-methyl-6-oxo-1, 2,3,4,6, 7-hexahydro- [1,4] oxazepino [2,3-c ] quinolin-10-yl) amino) nicotinonitrile
To a solution of (S) -10-amino-2-cyclopropyl-3, 3-difluoro-7-methyl-1, 2,3, 4-tetrahydro- [1,4] oxazepino [2,3-c ] quinolin-6 (7H) -one (150 mg, 466.82. Mu. Mol) and 2,5, 6-trichloropyridine-3-carbonitrile (96.84 mg, 466.82. Mu. Mol) in DMF (5 mL) was added DIEA (120.67 mg, 933.65. Mu. Mol, 162.62. Mu.L). The mixture was stirred at 100deg.C for 12hr. The mixture was cooled to 20 ℃ and water (10 mL) was added to form a precipitate, which was filtered and the filter cake concentrated in vacuo to give the title compound as a yellow solid (150 mg,152.34 μmol, 33% yield, 50% purity). LCMS: [ m+h ] + =492.0.
(1- (5-Chloro-3-cyano-6- (((S) -2-cyclopropyl-3, 3-difluoro-7-methyl-6-oxo-1, 2,3,4,6, 7-hexahydro- [1,4] oxazepino [2,3-c ] quinolin-10-yl) amino) pyridin-2-yl) -5-methylpiperidin-3-yl) carbamic acid tert-butyl ester
To a solution of (S) -2, 5-dichloro-6- ((2-cyclopropyl-3, 3-difluoro-7-methyl-6-oxo-1, 2,3,4,6, 7-hexahydro- [1,4] oxazepino [2,3-c ] quinolin-10-yl) amino) nicotinonitrile (150 mg, 304.69. Mu. Mol) and tert-butyl N- (5-methyl-3-piperidinyl) carbamate (78.36 mg, 365.63. Mu. Mol) in DMSO (2 mL) was added DIEA (78.76 mg, 609.38. Mu. Mol, 106.14. Mu. L). The mixture was stirred at 100deg.C for 1hr. The mixture was cooled to 20 ℃ and water (5 mL) was added to form a precipitate, which was filtered and the filter cake concentrated in vacuo to give the title compound as a yellow solid (150 mg,111.92 μmol, 37% yield, 50% purity). LCMS: [ m+h ] + =670.2.
2- (3-Amino-5-methylpiperidin-1-yl) -5-chloro-6- (((S) -2-cyclopropyl-3, 3-difluoro-7-methyl-6-oxo-1, 2,3,4,6, 7-hexahydro- [1,4] oxazepino [2,3-c ] quinolin-10-yl) amino) nicotinonitrile
To a solution of tert-butyl (1- (5-chloro-3-cyano-6- (((S) -2-cyclopropyl-3, 3-difluoro-7-methyl-6-oxo-1, 2,3,4,6, 7-hexahydro- [1,4] oxazepino [2,3-c ] quinolin-10-yl) amino) pyridin-2-yl) -5-methylpiperidin-3-yl) carbamate (150 mg, 223.83. Mu. Mol) in HCl/EtOAc (4M, 7.50 mL). The mixture was stirred at 20deg.C for 1hr. The mixture was concentrated in vacuo and used without further manipulation. The residue was separated by preparative HPLC (column: waters Xbridge BEH C100X 30mm X10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:30% -60%,10 min) to give the title compound (40 mg, 70.07. Mu. Mol, 31% yield, purity) as a yellow solid 99%).1H NMR(400MHz,DMSO-d6)δ9.02(s,1H),8.09-8.13(m,1H),7.90(s,1H),7.64(dd,J=8.99,1.65Hz,1H),7.44(dd,J=9.05,1.10Hz,1H),6.26-6.33(m,1H),4.26-4.51(m,3H),4.14(d,J=11.74Hz,1H),3.98-4.06(m,1H),3.56(s,3H),2.37-2.45(m,2H),2.23-2.35(m,2H),1.76-1.85(m,2H),1.52(s,1H),1.28-1.37(m,1H),0.72-0.82(m,2H),0.68(d,J=7.09Hz,3H),0.52(d,J=5.14Hz,2H),0.28-0.36(m,1H).
2- ((3R, 5S) -3-amino-5-methylpiperidin-1-yl) -5-chloro-6- (((S) -2-cyclopropyl-3, 3-difluoro-7-methyl-6-oxo-1, 2,3,4,6, 7-hexahydro- [1,4] oxazepino [2,3-c ] quinolin-10-yl) amino) nicotinonitrile (24)
2- (3-Amino-5-methylpiperidin-1-yl) -5-chloro-6- (((S) -2-cyclopropyl-3, 3-difluoro-7-methyl-6-oxo-1, 2,3,4,6, 7-hexahydro- [1,4] oxazepino [2,3-c ] quinolin-10-yl) amino) nicotinonitrile (70.00 mg, 122.80. Mu. Mol) was separated by SFC (column: DAICEL CHIRALPAK IC (250 mm. Times.30 mm, 10. Mu.m); mobile phase: [ ACN/IPA (0.1% NH 3H2 O) ]; B%:65% -65%,30 min) to give the title compound (8 mg, 13.31. Mu. Mol, 11% yield, 95% purity) as a yellow solid ).1H NMR(400MHz,DMSO-d6)δ9.16(s,1H),8.09(d,J=1.67Hz,1H),8.02(s,1H),7.92(s,2H),7.68(dd,J=8.94,1.79Hz,1H),7.45(d,J=9.06Hz,1H),6.29(s,1H),4.24-4.48(m,3H),3.94(d,J=12.64Hz,1H),3.57(s,3H),3.18-3.26(m,1H),3.06(t,J=11.32Hz,1H),2.82(t,J=11.98Hz,1H),2.28-2.36(m,1H),1.95(d,J=12.16Hz,1H),1.59-1.71(m,1H),1.30-1.37(m,1H),1.23(s,1H),1.05(q,J=11.96Hz,1H),0.68(d,J=6.44Hz,3H),0.53(t,J=5.66Hz,2H),0.28-0.35(m,1H).
2- ((3S, 5 r) -3-amino-5-methylpiperidin-1-yl) -5-chloro-6- (((S) -2-cyclopropyl-3, 3-difluoro-7-methyl-6-oxo-1, 2,3,4,6, 7-hexahydro- [1,4] oxazino [2,3-c ] quinolin-10-yl) amino) nicotinonitrile (25)
2- (3-Amino-5-methylpiperidin-1-yl) -5-chloro-6- (((S) -2-cyclopropyl-3, 3-difluoro-7-methyl-6-oxo-1, 2,3,4,6, 7-hexahydro- [1,4] oxazepino [2,3-c ] quinolin-10-yl) amino) nicotinonitrile (70.00 mg, 122.80. Mu. Mol) was separated by SFC (column: DAICEL CHIRALPAK IC (250 mm. Times.30 mm, 10. Mu.m); mobile phase: [ ACN/IPA (0.1% NH 3H2 O) ]; B%:65% -65%,30 min) to give the title compound (8.2 mg, 14.18. Mu. Mol, 12% yield, purity) as a yellow solid 99%).1H NMR(400MHz,DMSO-d6)δ9.22(s,1H),8.14(s,1H),8.09(s,1H),7.94(d,J=2.38Hz,2H),7.73-7.78(m,1H),7.51(d,J=9.06Hz,1H),6.33(s,1H),4.29-4.53(m,3H),3.99(d,J=12.52Hz,1H),3.63(s,3H),3.24-3.32(m,1H),3.07-3.16(m,1H),2.86(t,J=11.86Hz,1H),2.39(t,J=12.16Hz,1H),2.02(d,J=11.21Hz,1H),1.70(dt,J=5.27,2.67Hz,1H),1.35-1.42(m,1H),1.29(s,1H),1.12(q,J=12.20Hz,1H),0.76(d,J=6.44Hz,3H),0.59(d,J=5.01Hz,2H),0.38(d,J=4.41Hz,1H).
The absolute configuration of compounds 24 and 25 is randomly assigned to the cis conformation based on amino and methyl.
Example 24: synthesis of 2- ((6- ((6- ((3R, 5S) -3-amino-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (39) and 2- ((6- ((6- ((3S, 5R) -3-amino-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (40)
(1- (5-Chloro-3-cyano-6- ((1-methyl-3- (2- (methylamino) -2-oxoethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) pyridin-2-yl) -5-methylpiperidin-3-yl) carbamic acid tert-butyl ester
A flask containing a mixture of 2- ((6- ((3, 6-dichloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (300 mg, 694.03. Mu. Mol), tert-butyl N- (5-methyl-3-piperidinyl) carbamate (178.48 mg, 832.83. Mu. Mol) and DIEA (179.40 mg,1.39mmol, 241.77. Mu. L) in DMSO (6 mL) was degassed and purged 3 times with N 2, and then the mixture was stirred at 100℃under an atmosphere of N 2 for 12hr. The mixture was cooled to 20 ℃ and water (10 mL) was added to form a precipitate, which was filtered and the filter cake washed with EtOAc (20 mL) and concentrated in vacuo to give the title compound as a grey solid (300 mg,418.95 μmol, 60% yield, 85% purity). LCMS: m+h + =610.2.
2- ((6- ((6- (3-Amino-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
A mixture of tert-butyl 1- (5-chloro-3-cyano-6- ((1-methyl-3- (2- (methylamino) -2-oxoethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) pyridin-2-yl) -5-methylpiperidin-3-yl) carbamate (300 mg, 491.72. Mu. Mol) and HCl/EtOAc (4N HCl in EtOAc, 20 mL) was stirred at 20deg.C for 1hr. The mixture was concentrated in vacuo without further manipulation and the residue was purified by preparative HPLC (column: phenomenex Luna C, 80 x 40mm x 3 μm; mobile phase: [ water (HCl) -ACN ];: 20% -50%,7 min) to give the title compound as a yellow solid (150 mg, HCl, purity 99%).1H NMR(400MHz,DMSO-d6,26℃)δ9.12(s,1H),8.26(s,2H),8.00-8.08(m,2H),7.86(d,J=2.20Hz,1H),7.71(dd,J=9.05,2.20Hz,1H),7.54(d,J=9.17Hz,1H),7.31(s,1H),4.56-4.62(m,2H),4.32(d,J=9.17Hz,1H),4.01(d,J=11.49Hz,1H),3.69(s,3H),3.10-3.21(m,1H),2.84(t,J=11.74Hz,1H),2.68(d,J=4.52Hz,3H),2.39-2.46(m,1H),2.03-2.11(m,1H),1.72-1.86(m,1H),1.16(q,J=11.86Hz,1H),0.84(d,J=6.48Hz,3H).
2- ((6- ((6- ((3R, 5S) -3-amino-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (39)
2- ((6- ((6- (3-Amino-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (150 mg, 274.50. Mu. Mol, HCl) was separated by SFC (column: DAICEL CHIRALPAK AD (250 mm. Times.30 mm, 10. Mu.); mobile phase: 0.1% NH 3H2 O EtOH; B%:47% -47%,7 min) to give the title compound (10 mg, 18.50. Mu. Mol, 7% yield, purity) as a white solid 94%).1H NMR(400MHz,DMSO-d6,24℃)δ8.98(s,1H),7.98(s,2H),7.92(s,1H),7.67(dd,J=8.80,1.83Hz,1H),7.47(d,J=8.93Hz,1H),7.29(s,1H),4.54(s,2H),4.24(d,J=11.62Hz,1H),4.13(d,J=12.72Hz,1H),3.67(s,3H),2.66(d,J=4.52Hz,4H),2.32-2.45(m,1H),1.88(d,J=12.23Hz,1H),1.68-1.75(m,1H),1.64(d,J=9.29Hz,1H),1.18-1.30(m,1H),0.79(d,J=6.48Hz,3H).
2- ((6- ((6- ((3 S,5 r) -3-amino-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (40)
2- ((6- ((6- (3-Amino-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (150 mg, 274.50. Mu. Mol, HCl) was separated by SFC (column: DAICEL CHIRALPAK AD (250 mm. Times.30 mm, 10. Mu.); mobile phase: 0.1% NH 3H2 O EtOH; B%:47% -47%,7 min) to give the title compound (10 mg, 18.50. Mu. Mol, 7% yield, purity) as a white solid 94%).1H NMR(400MHz,DMSO-d6,24℃)δ9.13(s,1H),8.25(s,2H),8.01-8.11(m,2H),7.86(s,1H),7.70(d,J=8.93Hz,1H),7.54(d,J=9.17Hz,1H),7.31(s,1H),4.59(s,2H),4.31(d,J=10.88Hz,1H),4.00(d,J=11.25Hz,1H),3.68(s,3H),3.15(t,J=10.58Hz,1H),2.83(t,J=11.80Hz,1H),2.67(d,J=4.40Hz,3H),2.37-2.45(m,1H),2.02-2.10(m,1H),1.78(d,J=3.55Hz,1H),1.09-1.29(m,2H),0.83(d,J=6.48Hz,3H).
The absolute configuration of compounds 39 and 40 is randomly assigned to the cis conformation based on amino and methyl.
Example 25: synthesis of 2- ((3R, 5S) -3-amino-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (35) and 2- ((3S, 5R) -3-amino-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (36)
(1- (5-Chloro-3-cyano-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) pyridin-2-yl) -5-methylpiperidin-3-yl) carbamic acid tert-butyl ester
A flask containing a mixture of 2, 5-dichloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (100 mg, 237.93. Mu. Mol), (5-methylpiperidin-3-yl) carbamic acid tert-butyl ester (61.19 mg, 285.52. Mu. Mol) and DIEA (61.50 mg, 475.86. Mu. Mol, 82.89. Mu.L) in DMSO (1 mL) was degassed and purged 3 times with N 2, and then the mixture was stirred at 100℃under an atmosphere of N 2 for 1hr. Water (5 mL) was added to form a precipitate, which was filtered, and the filter cake was dried under reduced pressure to give the title compound as a yellow solid (150 mg, 213.51. Mu. Mol, yield 90%, purity 85%). LCMS: [ M-boc+h ] + =498.2.
2- (3-Amino-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A mixture of tert-butyl (1- (5-chloro-3-cyano-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) pyridin-2-yl) -5-methylpiperidin-3-yl) carbamate (130 mg, 217.34. Mu. Mol) and HCl/EtOAc (4M, 13 mL) was stirred at 20℃under N 2 for 1hr. The mixture was concentrated in vacuo to give the title compound as a yellow solid (25 mg, 46.43. Mu. Mol, 21% yield, purity 99%,HCl).1H NMR(400MHz,DMSO-d6)δ0.78(d,J=6.60Hz,3H),1.17(s,7H),1.67-1.75(m,3H),2.02(br d,J=11.98Hz,1H),2.34-2.42(m,1H),2.83(t,J=11.92Hz,1H),3.07-3.17(m,1H),3.32(s,2H),3.85-3.90(m,2H),3.93-3.99(m,1H),4.26-4.32(m,1H),7.12(d,J=8.31Hz,1H),7.25-7.33(m,2H),7.98(s,1H),8.09(br s,3H),8.99(s,1H).
2- ((3R, 5S) -3-amino-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (35) and 2- ((3S, 5R) -3-amino-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (36)
Racemic 2- (3-amino-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (50 mg, HCl) was separated by SFC (column: DAICEL CHIRALPAK AD (250 mm. Times.30 mm,10 μm); mobile phase: [0.1% NH 3H2 O IPA ]; B%:30% -30%,10 min) to give 2- ((3S, 5R) -3-amino-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (11.6 mg, 22.90. Mu. With a yield of 24%, 98%);1H NMR(400MHz,DMSO-d6)δ0.78(d,J=6.60Hz,3H),1.08-1.16(m,2H),1.17(s,6H),1.67-1.74(m,3H),2.04(br d,J=11.49Hz,1H),2.40(br d,J=11.86Hz,1H),2.85(br t,J=11.86Hz,1H),3.10(br t,J=10.82Hz,1H),3.32(br s,2H),3.84-3.91(m,2H),3.96(br d,J=11.49Hz,1H),4.28(br d,J=9.66Hz,1H),7.14(d,J=8.44Hz,1H),7.28-7.31(m,1H),7.32(s,1H),7.98(s,1H),8.22(br s,3H),8.99(s,1H); purity and 2- ((3R, 5S) -3-amino-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (12.2 mg, 24.27. Mu. Mol, 26% purity) as a white solid 99%).1H NMR(400MHz,DMSO-d6)δ0.76(d,J=6.60Hz,3H),0.79-0.87(m,1H),1.16(s,6H),1.57-1.65(m,1H),1.67-1.73(m,1H),1.86(br d,J=12.23Hz,1H),2.33(br t,J=12.10Hz,2H),2.60-2.69(m,2H),3.32(br s,2H),3.86-3.92(m,2H),4.04(br d,J=11.74Hz,1H),4.16-4.22(m,1H),7.10(d,J=8.44Hz,1H),7.25(dd,J=8.44,1.59Hz,1H),7.38(d,J=1.47Hz,1H),7.89(s,1H),8.87(s,1H).
The absolute configuration of compounds 35 and 36 is randomly assigned to the cis conformation based on amino and methyl.
Example 26: synthesis of chloro-5-cyanopyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (41) and 2- ((6- ((6- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (42)
2- ((6- ((3, 6-Dichloro-5-cyanopyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid methyl ester
To a solution of methyl 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] acetate (3.5 g,8.35 mmol) in DMSO (35 mL) was added K 2CO3 (2.31 g,16.70 mmol) and 3- (iodomethyl) oxetane (1.65 g,8.35 mmol). The mixture was stirred at 80℃for 12hr. The reaction mixture was cooled to 15 ℃ and water (20 mL) was added to form a precipitate, which was filtered and the filter cake dried in vacuo to give the title compound (4 g, crude) as a yellow solid. LCMS: m+h + = 489.0.
2- ((6- ((3, 6-Dichloro-5-cyanopyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid
To a solution of methyl 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -1- (oxetan-3-ylmethyl) -2-oxo-3-quinolinyl ] oxy ] acetate (4 g,8.17 mmol) in EtOH (40 mL) and H 2 O (40 mL) was added LiOH.H 2 O (1.72 g,40.87 mmol). The mixture was stirred at 25℃for 1hr. The mixture was concentrated in vacuo to give the title compound as a yellow solid (4 g, crude). LCMS: m+h + = 474.9.
2- ((6- ((3, 6-Dichloro-5-cyanopyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
To a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -1- (oxetan-3-ylmethyl) -2-oxo-3-quinolinyl ] oxy ] acetic acid (4 g,8.42 mmol) and MeNH 2 (1.14 g,16.83mmol, HCl) in DMF (30 mL) was added HATU (6.40 g,16.83 mmol) and DIPEA (4.35 g,33.66mmol,5.86 mL). The mixture was stirred at 25℃for 12hr. Water (60 mL) was added, and the mixture was then extracted with ethyl acetate (50 mL x 2). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. The crude product was triturated with EtOAc (20 mL) at 25℃for 10min to give the title compound as a yellow solid (1.2 g,1.72mmol, 20% yield, 70% purity). LCMS: m+h + = 488.2.
2- ((6- ((3-Chloro-5-cyano-6- (3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) pyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
To a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -1- (oxetan-3-ylmethyl) -2-oxo-3-quinolinyl ] oxy ] -N-methylacetamide (200 mg, 409.57. Mu. Mol) and 2- (4, 4-difluoro-5-methyl-3-piperidinyl) isoindoline-1, 3-dione (114.79 mg, 409.57. Mu. Mol) in DMSO (2 mL) was added DIPEA (105.87 mg, 819.13. Mu. Mol, 142.68. Mu. L). The mixture was stirred at 100deg.C for 12hr. The mixture was cooled to 20 ℃ and water (3 mL) was added. The mixture was filtered and the filter cake was concentrated in vacuo to give a residue which was purified by preparative HPLC (column: waters Xbridge BEH C, 100, 30mm, 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:40% -65%,8 min) to give the title compound as a yellow solid (100 mg, 136.59. Mu. Mol, 25% yield).
2- ((6- ((6- (3-Amino-4, 4-difluoro-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (41)
To a solution of 2- [ [6- [ [ 3-chloro-5-cyano-6- [3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methyl-1-piperidinyl ] -2-pyridinyl ] amino ] -1- (oxetan-3-ylmethyl) -2-oxo-3-quinolinyl ] oxy ] -N-methylacetamide (100 mg, 136.59. Mu. Mol) in EtOH (4 mL) was added NH 2NH2.H2 O (102.56 mg,2.05mmol, 99.58. Mu.L). The mixture was stirred at 60℃for 1hr. The mixture was cooled to 20 ℃ and concentrated in vacuo. The residue was purified by preparative HPLC (column: phenomenex C18.80 x 40mm x 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:15% -45%,8 min) to give the title compound (40 mg,61.58 μmol, yield 45%, purity) as a white solid 93%).1H NMR(400MHz,DMSO-d6)δ9.08(s,1H),8.00(s,1H),7.91-7.97(m,2H),7.60-7.66(m,1H),7.52-7.57(m,1H),7.36(s,1H),4.66(br d,J=7.00Hz,2H),4.57-4.63(m,2H),4.48-4.56(m,4H),4.21-4.31(m,1H),4.11(br d,J=12.01Hz,1H),3.43(dt,J=14.07,7.10Hz,1H),2.89-3.06(m,1H),2.74-2.87(m,2H),2.67(d,J=4.63Hz,3H),1.98-2.18(m,1H),1.77(br d,J=1.63Hz,2H),0.86(d,J=6.63Hz,3H).
2- ((6- ((6- ((3S, 5R) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (41) and 2- ((6- ((6- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -3-chloro-5-cyanopyridin-2-yl) amino) -1- (oxetan-3-ylmethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (42)
2- [ [6- [ [6- (3-Amino-4, 4-difluoro-5-methyl-1-piperidinyl) -3-chloro-5-cyano-2-pyridinyl ] amino ] -1- (oxetan-3-ylmethyl) -2-oxo-3-quinolinyl ] oxy ] -N-methyl-acetamide (40 mg, 66.44. Mu. Mol) was separated by SFC (column: DAICEL CHIRALCEL OD (250 mm. Times.30 mm,10 μm); mobile phase: [0.1% nh 3H2 O EtOH ]; B%:55% -55%,10 min) to give 2- [ [6- [ [6- [ (3 s,5 r) -3-amino-4, 4-difluoro-5-methyl-1-piperidinyl ] -3-chloro-5-cyano-2-pyridinyl ] amino ] -1- (oxetan-3-ylmethyl) -2-oxo-3-quinolinyl ] oxy ] -N-methylacetamide (12 mg, 19.73. Mu. Mol, 30%, 99%);1H NMR(400MHz,DMSO-d6)δ9.07(s,1H),8.00(s,1H),7.93(br d,J=1.79Hz,2H),7.61-7.65(m,1H),7.52-7.56(m,1H),7.36(s,1H),4.66(br d,J=6.91Hz,2H),4.57-4.62(m,2H),4.47-4.56(m,4H),4.08-4.30(m,2H),3.43(dt,J=14.04,6.99Hz,1H),3.30(br s,1H),2.92-3.05(m,1H),2.80(td,J=12.58,7.27Hz,2H),2.67(d,J=4.53Hz,3H),1.99-2.17(m,1H),1.77(br s,1H),0.86(d,J=6.68Hz,3H); purity and white solid of 2- [ [6- [ [6- [ (3R, 5S) -3-amino-4, 4-difluoro-5-methyl-1-piperidinyl ] -3-chloro-5-cyano-2-pyridinyl ] amino ] -1- (oxetan-3-ylmethyl) -2-oxo-3-quinolinyl ] oxy ] -N-methyl-acetamide ;1H NMR(400MHz,DMSO-d6)δ9.07(s,1H),8.00(s,1H),7.91-7.97(m,2H),7.60-7.66(m,1H),7.52-7.57(m,1H),7.36(s,1H),4.66(br d,J=7.03Hz,2H),4.57-4.63(m,2H),4.49-4.56(m,4H),4.07-4.30(m,2H),3.39-3.48(m,1H),3.30(br s,1H),2.92-3.05(m,1H),2.80(td,J=12.55,7.21Hz,2H),2.67(d,J=4.53Hz,3H),1.99-2.15(m,1H),1.72-1.90(m,1H),0.86(d,J=6.68Hz,3H).
The absolute configuration of compounds 41 and 42 is randomly assigned to the cis-conformation based on amino and methyl.
Example 27: synthesis of 2- ((3S, 5R) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1- (oxetan-3-ylmethyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (37) and 2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1- (oxetan-3-ylmethyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (38)
4-Nitro-N1- (oxetan-3-ylmethyl) benzene-1, 2-diamine
To a solution of 2-fluoro-5-nitroaniline (2 g,12.81 mmol) and oxetan-3-ylmethylamine (1.34 g,15.37 mmol) in DMSO (20 mL) was added K 2CO3 (2.66 g,19.22 mmol). The mixture was stirred at 100deg.C for 12hr. Water (200 mL) was added and the mixture was then extracted with DCM (300 mL x 4). The combined organic phases were dried over anhydrous Na2SO4, filtered and concentrated to give a residue which was purified by column chromatography (silica gel, petroleum ether/ethyl acetate=5/1 to 0/1) to give the title compound (2.5 g,10.53mmol, yield 82% purity) as a white solid 94%).1H NMR(400MHz,DMSO-d6,25℃)δ7.51(dd,J=8.82,2.62Hz,1H),7.40(d,J=2.62Hz,1H),6.52(d,J=8.82Hz,1H),5.93(t,J=5.01Hz,1H),5.15(s,2H),4.69(dd,J=7.51,6.08Hz,2H),4.31(t,J=5.90Hz,2H),3.50(dd,J=7.27,5.36Hz,2H),3.20-3.28(m,1H).
6-Nitro-3- (oxetan-3-ylmethyl) -1H-benzimidazol-2-one
A flask containing a mixture of 4-nitro-N1- (oxetan-3-ylmethyl) benzene-1, 2-diamine (1 g,4.48 mmol) and CDI (1.09 g,6.72 mmol) in DMF (10 mL) was degassed and purged 3 times with N 2 and the mixture stirred at 15℃under an atmosphere of N 2 for 12hr. Water (80 mL) was then added and the mixture extracted with ethyl acetate (45 mL x 3). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated to give a residue which was purified by column chromatography (silica gel, petroleum ether/ethyl acetate=4/1 to 0/1) to give the title compound (1.1 g,4.10mmol, 92% purity) as a yellow solid 93%).1H NMR(400MHz,DMSO-d6,25℃)δ8.00(dd,J=8.70,2.26Hz,1H),7.74(d,J=2.15Hz,1H),7.41(d,J=8.70Hz,1H),4.61(dd,J=7.63,6.20Hz,2H),4.41(t,J=6.08Hz,2H),4.18(d,J=7.15Hz,2H),3.33-3.44(m,1H).
3- (3-Hydroxy-3-methyl-butyl) -5-nitro-1- (oxetan-3-ylmethyl) benzimidazol-2-one
A flask containing a mixture of 6-nitro-3- (oxetan-3-ylmethyl) -1H-benzimidazol-2-one (1.3 g,5.22 mmol), butyl 3-hydroxy-3-methyl 4-methylbenzenesulfonate (2.02 g,7.83 mmol) and Cs 2CO3 (5.10 g,15.66 mmol) in DMF (10 mL) was degassed and purged 3 times with N 2 and the mixture stirred at 100deg.C under an atmosphere of N 2 for 2hr. Water (60 mL) was then added and the mixture extracted with ethyl acetate (50 mL x 3). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated to give a residue which was purified by column chromatography (silica gel, petroleum ether/ethyl acetate=3/1 to 0/1) to give the title compound as a yellow solid (1 g, purity 92%).1H NMR(400MHz,DMSO-d6,25℃)δ8.05(dd,J=8.69,2.06Hz,1H),8.00(d,J=2.00Hz,1H),7.48(d,J=8.63Hz,1H),4.61(dd,J=7.63,6.25Hz,2H),4.51(s,1H),4.40(t,J=6.07Hz,2H),4.23(d,J=7.13Hz,2H),3.96-4.02(m,2H),3.39(dt,J=13.91,6.86Hz,1H),1.70-1.76(m,2H),1.16(s,6H).
5-Amino-3- (3-hydroxy-3-methyl-butyl) -1- (oxetan-3-ylmethyl) benzimidazol-2-one
To a mixture of Pd/C (200 mg, purity 10%) in DMF (10 mL) was added 3- (3-hydroxy-3-methyl-butyl) -5-nitro-1- (oxetan-3-ylmethyl) benzimidazol-2-one (1 g,2.98 mmol). The mixture was stirred at 15℃for 12hr under H 2 (15 psi). Adding the mixture to And filtered to give a filtrate, which was concentrated in vacuo to give the title compound as a red solid (900 mg,2.71mmol, 91% yield, purity) 92%).1H NMR(400MHz,DMSO-d6,25℃)δ6.85(d,J=8.25Hz,1H),6.38(d,J=1.88Hz,1H),6.30(dd,J=8.25,2.00Hz,1H),4.81(br s,2H),4.58(dd,J=7.75,6.13Hz,2H),4.47(s,1H),4.38(t,J=6.07Hz,2H),4.01(d,J=7.00Hz,2H),3.74-3.86(m,2H),3.27-3.35(m,1H),1.58-1.73(m,2H),1.16(s,6H).
2, 5-Dichloro-6- [ [3- (3-hydroxy-3-methyl-butyl) -1- (oxetan-3-ylmethyl) -2-oxo-benzoimidazol-5-yl ] amino ] pyridine-3-carbonitrile
A flask containing a mixture of 5-amino-3- (3-hydroxy-3-methyl-butyl) -1- (oxetan-3-ylmethyl) benzimidazol-2-one (800 mg,2.62 mmol), 2,5, 6-trichloropyridine-3-carbonitrile (543.46 mg,2.62 mmol) and DIEA (677.17 mg,5.24mmol, 912.63. Mu.L) in DMF (9 mL) was degassed and purged 3 times with N 2, then the mixture was stirred at 100℃under an atmosphere of N 2 for 1hr. The mixture was concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: welch Xtimate C18:250:70 mm#10μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:30% -60%,20 min) to give the title compound (700 mg,1.40mmol, 53% purity) as a white solid 95%).1H NMR(400MHz,DMSO-d6,25℃)δ9.54(s,1H),8.35(s,1H),7.35(d,J=1.67Hz,1H),7.22-7.26(m,1H),7.16-7.19(m,1H),4.62(dd,J=7.69,6.14Hz,2H),4.42-4.44(m,2H),4.40(s,1H),4.14(d,J=7.03Hz,2H),3.85-3.92(m,2H),3.35-3.45(m,1H),1.65-1.77(m,2H),1.17(s,6H).
5-Chloro-2- ((3 s,5 r) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1- (oxetan-3-ylmethyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A flask containing a mixture of 2, 5-dichloro-6- [ [3- (3-hydroxy-3-methyl-butyl) -1- (oxetan-3-ylmethyl) -2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (150 mg, 314.89. Mu. Mol), 2- ((3S, 5R) -4, 4-difluoro-5-methylpiperidin-3-yl) isoindoline-1, 3-dione (88.25 mg, 314.89. Mu. Mol) and DIEA (203.49 mg,1.57mmol, 274.24. Mu.L) in DMSO (1.5 mL) was degassed and purged 3 times with N 2, and the mixture was then stirred at 100℃under an atmosphere of N 2 for 12hr. The mixture was concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: waters Xbridge BEH C, 100 x 30mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:40% -60%,8 min) to give the title compound as a yellow solid (170 mg,228.98 μmol, 73% yield, 97% purity). LCMS: m+h + =720.0.
2- ((3S, 5R) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1- (oxetan-3-ylmethyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (37)
To a solution of 5-chloro-2- ((3 s,5 r) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1- (oxetan-3-ylmethyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (170 mg, 236.06. Mu. Mol) in EtOH (10 mL) was added MeNH 2/H2 O (purity 40%). The mixture was stirred at 70℃for 1hr. The mixture was concentrated in vacuo and purified by preparative HPLC (column: phenomnex C18.80X 40mm X3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:20% -50%,8 min) to give the title compound as a white solid (35 mg, 55.76. Mu. Mol, 24% yield, purity) 94%).1H NMR(400MHz,DMSO-d6,22℃)δ7.97(s,1H),7.29(s,1H),7.23(s,2H),4.61(s,2H),4.50(s,1H),4.42(s,2H),4.15(s,3H),3.97(br s,1H),3.89(br s,2H),3.36-3.46(m,2H),2.77-2.91(m,2H),2.64-2.71(m,1H),1.92-2.09(m,1H),1.68-1.73(m,2H),1.66(br s,1H),1.16(d,J=0.73Hz,6H),0.76(d,J=6.72Hz,3H).
5-Chloro-2- ((3R, 5S) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1- (oxetan-3-ylmethyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A flask containing a mixture of 2, 5-dichloro-6- [ [3- (3-hydroxy-3-methyl-butyl) -1- (oxetan-3-ylmethyl) -2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (150 mg, 314.89. Mu. Mol), 2- ((3R, 5S) -4, 4-difluoro-5-methylpiperidin-3-yl) isoindoline-1, 3-dione (88.25 mg, 314.89. Mu. Mol) and DIEA (203.49 mg,1.57mmol, 274.24. Mu.L) in DMSO (1.5 mL) was degassed and purged 3 times with N 2, and the mixture was then stirred at 100℃under an atmosphere of N 2 for 12hr. The mixture was concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: waters Xbridge BEH C, 100 x 30mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:40% -60%,8 min) to give the title compound as a white solid (150 mg,202.04 μmol, yield 64%, purity 97%). LCMS: m+h + =720.0.
2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-chloro-6- ((3- (3-hydroxy-3-methylbutyl) -1- (oxetan-3-ylmethyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (38)
To a solution of 5-chloro-2- ((3 r,5 s) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1- (oxetan-3-ylmethyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (150 mg, 208.29. Mu. Mol) in EtOH (10 mL) was added MeNH 2/H2 O (10 mL, purity 40%). The mixture was stirred at 70℃for 1hr. The mixture was concentrated in vacuo and purified by preparative HPLC (column: phenomnex C18.cndot.80X 40mm X3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:20% -50%,8 min) to give the title compound as a white solid (30 mg, 50.33. Mu. Mol, 24% yield, purity) 99%).1H NMR(400MHz,DMSO-d6,22℃)δ9.02(s,1H),7.96(s,1H),7.28(s,1H),7.22(s,2H),4.60(dd,J=7.76,6.05Hz,2H),4.49(s,1H),4.41(t,J=6.05Hz,2H),4.11-4.19(m,3H),3.94(br dd,J=13.51,2.14Hz,1H),3.85-3.91(m,2H),3.35-3.43(m,2H),2.78-2.91(m,2H),2.63-2.70(m,1H),1.94-2.09(m,1H),1.67-1.72(m,2H),1.65(br s,1H),1.15(s,6H),0.75(d,J=6.72Hz,3H).
The absolute configuration of compounds 37 and 38 is randomly assigned to the cis conformation based on amino and methyl.
Example 28: synthesis of 2- ((1- (2-aminoethyl) -6- ((3-chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (66)
2- ((1- (2- ((Tert-Butoxycarbonyl) amino) ethyl) -6-nitro-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid methyl ester
A flask containing a mixture of methyl 2- [ (6-nitro-2-oxo-1H-quinolin-3-yl) oxy ] acetate (500 mg,1.80 mmol), tert-butyl N- (2-bromoethyl) carbamate (2.01 g,8.99 mmol), K 2CO3 (496.77 mg,3.59 mmol) and KI (149.16 mg, 898.58. Mu. Mol,0.5 eq.) in DMSO (10 mL) was degassed and purged 3 times with N 2 and then the mixture stirred at 80℃under an atmosphere of N 2 for 2hr. Water (60 mL) was added and the mixture extracted with ethyl acetate (50 mL x 3). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated to give the title compound (1 g, crude) as a brown oil. LCMS: m+h + = 422.2.
2- ((1- (2- ((Tert-butoxycarbonyl) amino) ethyl) -6-nitro-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid
To a solution of methyl 2- ((1- (2- ((tert-butoxycarbonyl) amino) ethyl) -6-nitro-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetate (800 mg,1.90 mmol) in EtOH (4 mL) and H 2 O (4 mL) was added LiOH.H 2 O (159.33 mg,3.80 mmol), and the mixture was then stirred at 20℃for 1hr. The mixture was concentrated in vacuo to give the title compound as a yellow solid (800 mg, crude). LCMS: m+h + = 408.0.
(2- (3- (2- (Methylamino) -2-oxoethoxy) -6-nitro-2-oxoquinolin-1 (2H) -yl) ethyl) carbamic acid tert-butyl ester
To a solution of 2- ((1- (2- ((tert-butoxycarbonyl) amino) ethyl) -6-nitro-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid (800 mg,1.96 mmol), hexamethylenetetramine (530.37 mg,7.86mmol, hcl), HATU (1.49 g,3.93 mmol) and DIEA (1.02 g,7.86mmol,1.37 mL) in DMF (6 mL) and the mixture was stirred at 20 ℃ for 1hr. Water (40 mL) was then added and the mixture extracted with ethyl acetate (30 mL x 2). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated to give the title compound as a yellow oil (800 mg,1.09mmol, 56% yield, 57% purity). LCMS: m+h + = 421.2.
(2- (6-Amino-3- (2- (methylamino) -2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) ethyl) carbamic acid tert-butyl ester
To a solution of Pd/C (0.1 g, purity 10%) in DMF (15 mL) was added tert-butyl (2- (3- (2- (methylamino) -2-oxoethoxy) -6-nitro-2-oxoquinolin-1 (2H) -yl) ethyl) carbamate (600 mg,1.43 mmol) under Ar. The mixture was stirred at 50deg.C under H 2 (50 psi) for 12hr. The mixture was filtered and the filtrate concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: waters Xbridge BEH C18:250:50 mm:10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:15% -45%,10 min) to give the title compound (90 mg, 187.91. Mu. Mol, yield 13%, purity) as a white solid 82%).1H NMR(400MHz,DMSO-d6)δ1.37(s,9H),2.67-2.72(m,3H),3.37(br d,J=5.87Hz,2H),4.31(br t,J=5.44Hz,2H),4.56(s,2H),5.22(br s,2H),6.70(d,J=2.45Hz,1H),6.88(dd,J=8.80,2.45Hz,1H),7.01(br t,J=5.44Hz,1H),7.25(s,1H),7.38(d,J=8.80Hz,1H),7.84(br d,J=4.03Hz,1H).
(2- (6-Amino-3- (2- (methylamino) -2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) ethyl) carbamic acid tert-butyl ester
A flask containing a mixture of tert-butyl (2- (6-amino-3- (2- (methylamino) -2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) ethyl) carbamate (60 mg, 153.68. Mu. Mol), 2,5, 6-trichloropyridine-3-carbonitrile (31.88 mg, 153.68. Mu. Mol) and DIEA (39.72 mg, 307.35. Mu. Mol, 53.54. Mu. L) in DMF (1 mL) was degassed and purged 3 times with N 2 and the mixture stirred at 100℃under an N 2 atmosphere for 12hr. Water (15 mL) was then added and the mixture was filtered to give a filter cake, which was dried in vacuo to give the title compound as a grey solid (60 mg, 56.98. Mu. Mol, 37% yield, 53% purity). LCMS: m+h + = 561.2.
(2- (6- ((3-Chloro-5-cyano-6- ((3S, 4S, 5R) -4-4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -3- (2- (methylamino) -2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) ethyl) carbamic acid tert-butyl ester
A flask containing a mixture of tert-butyl (2- (6-amino-3- (2- (methylamino) -2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) ethyl) carbamate (60.00 mg, 106.87. Mu. Mol), (3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidine (53.75 mg, 320.62. Mu. Mol, HCl) and DIEA (138.13 mg,1.07mmol, 186.15. Mu.L) in DMSO (1 mL) was degassed and purged 3 times with N 2 and the mixture was stirred at 100℃under an atmosphere of N 2 for 2hr. Water (20 mL) was then added and the mixture was filtered to give a filter cake, which was dried in vacuo to give the title compound as a grey solid (60 mg, 62.78. Mu. Mol, 59% yield, 69% purity). LCMS: [ M-56] +=600.2,[M-100]+ = 556.3.
2- ((1- (2-Aminoethyl) -6- ((3-chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (66)
A solution of tert-butyl (2- (6- ((3-chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -3- (2- (methylamino) -2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) ethyl) carbamate (60 mg, 91.44. Mu. Mol) in HCl/EtOAc (4M, 6 mL) was stirred at 20℃for 1hr. The mixture was concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: phenomnex Luna 80X 30mm X3 μm; mobile phase: [ water (HCl) -ACN ]; B%:20% -50%,8 min) to give the title compound (45 mg, 74.12. Mu. Mol, 11% yield, purity) as a yellow solid 98%,HCl).1H NMR(400MHz,DMSO-d6)δ0.86(d,J=6.85Hz,6H),1.74-1.95(m,2H),2.67(d,J=4.65Hz,3H),2.79(t,J=12.65Hz,2H),3.08(d,J=5.87Hz,2H),3.99(dd,J=12.59,3.79Hz,2H),4.53-4.58(m,4H),7.25-7.27(m,1H),7.63-7.66(m,2H),7.85-7.88(m,1H),7.95-8.01(m,2H),8.12(s,3H),9.07-9.12(m,1H).
Example 29: synthesis of 2- ((6- ((3-chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2-hydroxy-3- (methylamino) propyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (67)
2- ((6- ((3, 6-Dichloro-5-cyanopyridin-2-yl) amino) -3- (2-methoxy-2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) methyl) morpholine-4-carboxylic acid tert-butyl ester
To a solution of methyl 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] acetate (2 g,4.77 mmol) and tert-butyl 2- (bromomethyl) morpholine-4-carboxylate (1.47 g,5.25 mmol) in DMSO (25 mL) was added K 2CO3 (1.32 g,9.54 mmol) and KI (395.98 mg,2.39 mmol). The mixture was stirred at 80℃for 4hr. To the mixture was added water (50 mL) and extracted with ethyl acetate (50 mL x 3). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated under reduced pressure to give the title compound (2.9 g, crude) as a white solid. LCMS: m+h + =618.1.
2- ((1- ((4- (Tert-butoxycarbonyl) morpholin-2-yl) methyl) -6- ((3, 6-dichloro-5-cyanopyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid
To a solution of tert-butyl 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -3- (2-methoxy-2-oxoethoxy) -2-oxo-1-quinolinyl ] methyl ] morpholine-4-carboxylate (2.9 g,4.69 mmol) in EtOH (20 mL) and H 2 O (20 mL) was added LiOH.H 2 O (255.80 mg,6.10 mmol). The mixture was stirred at 20deg.C for 12hr. The mixture was purified by preparative HPLC (column: agela DuraShell C 18 x 80mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ];% B30% -60%,20 min) to give the title compound as a white solid .1H NMR(400MHz,DMSO-d6)=9.67(s,1H),8.38(s,1H),7.73(s,1H),7.63(s,2H),7.16(s,1H),4.52-4.14(m,4H),3.84-3.56(m,4H),3.17-2.67(m,4H),1.38(s,9H).
2- ((6- ((3, 6-Dichloro-5-cyanopyridin-2-yl) amino) -3- (2- (methylamino) -2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) methyl) morpholine-4-carboxylic acid tert-butyl ester
To a solution of 2- [ [1- [ (4-tert-butoxycarbonylmorpholin-2-yl) methyl ] -6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-3-quinolinyl ] oxy ] acetic acid (1 g,1.65 mmol) and hexamethylenetetramine (223.41 mg,3.31mmol, HCl) in DMF (10 mL) was added HATU (1.26 g,3.31 mmol) and DIPEA (855.29 mg,6.62mmol,1.15 mL). The mixture was stirred at 25℃for 12hr. To the mixture was added water (10 mL), a precipitate was formed, the precipitate was filtered, and the filter cake was washed with water (20 mL x 2), PE (20 mL x 2) and dried under reduced pressure to give the title compound as a white solid. LCMS: m+h + = 617.2.
(3- (6- ((3-Chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -3- (2- (methylamino) -2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) -2-methylpropyl) carbamic acid tert-butyl ester
To a solution of tert-butyl 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -3- [2- (methylamino) -2-oxoethoxy ] -2-oxo-1-quinolinyl ] methyl ] morpholine-4-carboxylate (100 mg, 161.95. Mu. Mol) and (3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidine (32.58 mg, 194.34. Mu. Mol, HCl) in DMSO (1 mL) was added DIPEA (62.79 mg, 485.85. Mu. Mol, 84.63. Mu.L). The mixture was stirred at 100deg.C for 2hr. To the mixture was added water (5 mL), a precipitate formed, the precipitate was filtered and the filter cake was washed with water (10 mL x 2) and EA (10 mL x 2). The filter cake was dried under reduced pressure to give the title compound (90 mg, 126.37. Mu. Mol, yield 78%) as a white solid. LCMS: m+h + = 712.3.
2- ((6- ((3-Chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2-hydroxy-3- (methylamino) propyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (67)
A solution of tert-butyl 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3S, 4S, 5R) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -3- [2- (methylamino) -2-oxoethoxy ] -2-oxo-1-quinolinyl ] methyl ] morpholine-4-carboxylate (330 mg, 463.35. Mu. Mol) in HCl/EtOAc (4 mL, 4M) was stirred at 25℃for 1hr. The reaction mixture was concentrated under reduced pressure and the residue was purified by preparative HPLC (column: phenomnex Luna 80 x 30mm x3 μm; mobile phase: [ water (HCl) -ACN ];% B: 20% -50%,8 min) to give the title compound (30 mg, 45.74. Mu. Mol, yield 10%, purity) as a white solid 99%,HCl).1H NMR(400MHz,DMSO-d6)δ9.61-9.59(m,1H),9.52-9.50(m,1H),9.08-9.05(m,1H),8.03(d,J=4.4Hz,1H),7.95-7.94(m,1H),7.82-7.81(m,1H),7.65-7.62(m,1H),7.58-7.56(m,1H),7.25-7.24(m,1H),4.55(s,2H),4.43-4.41(m,1H),4.18-4.15(m,2H),3.99(d,J=13.2Hz,2H),3.88(d,J=12.8Hz,1H),3.68-3.65(m,1H),3.34(d,J=12.4Hz,1H),3.13(br d,J=12.0Hz,1H),3.03-2.93(m,2H),2.83-2.72(m,2H),2.67(d,J=4.8Hz,3H),1.90-1.73(m,2H),0.89-0.85(m,6H).
Example 30: synthesis of 2- ((6- ((3-chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2-morpholinoethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (68)
2- (4, 4-Difluoro-3-hydroxy-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a solution of methyl 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] acetate (3 g,7.16 mmol) in NMP (3 mL) and EtOH (27 mL) was added MeNH 2 (4.44 g,57.25mmol, 40% purity). The mixture was stirred at 70℃for 12hr. The reaction mixture was cooled to 20℃and EtOH (90 mL) was added. The suspension was filtered and the filter cake was dried under reduced pressure to give the title compound (2.6 g,5.41mmol, yield 76%, purity 87%) as a yellow solid. LCMS: m+h + = 418.0.
2- ((6- ((3-Chloro-5-cyano-6- (4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
To a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] -N-methylacetamide (500 mg,1.20 mmol) and (3R, 5S) -4-fluoro-3, 5-dimethylpiperidine (300.64 mg,1.79mmol, HCl) in DMSO (1 mL) was added DIEA (618.04 mg,4.78mmol,832.94 μl). The mixture was stirred at 100deg.C for 2hr. The reaction mixture was treated with water (20 mL) to form a precipitate, which was filtered. The filter cake was washed with EtOAc (30 mL) and then dried under reduced pressure to give the title compound as a brown solid (600 mg,818.77 μmol, 68% yield, 70% purity). LCMS: m+h + =513.3.
2- [ [6- [ [ 3-Chloro-5-cyano-6- [ (3R, 5S) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -1- (2-morpholinoethyl) -2-oxo-3-quinolinyl ] oxy ] -N-methylacetamide (68)
To a solution of 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3R, 5S) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] -N-methylacetamide (200 mg, 389.89. Mu. Mol) and 4- (2-bromoethyl) morpholine (189.17 mg, 974.73. Mu. Mol) in DMSO (2 mL) was added Cs 2CO3 (254.07 mg, 779.78. Mu. Mol) and KI (32.36 mg, 194.95. Mu. Mol). The mixture was stirred at 80℃for 5hr. The reaction mixture was treated with water (30 mL) to form a precipitate, which was filtered. The filter cake was washed with EtOAc (30 mL), collected and dried under reduced pressure to give a residue which was further separated by preparative HPLC (column: phenomnex Luna 80X 30mm X3 μm; mobile phase: [ water (HCl) -ACN ]; B%:25% -50%,8 min) and SFC (column: DAICEL CHIRALPAK IG (250 mm X30 mm,10 μm); mobile phase: [0.1% NH 3H2 O EtOH ];B%:50% -50%,10 min) to give the title compound (18 mg, 23.29. Mu. Mol, 29% yield, purity) as a white solid 81%).1H NMR(400MHz,DMSO-d6)δ9.07(s,1H),7.88-8.01(m,2H),7.78(br s,1H),7.66(dd,J=9.13,2.38Hz,1H),7.49(br d,J=9.13Hz,1H),7.22(s,1H),4.52-4.58(m,2H),4.43(br s,2H),3.98(br dd,J=12.94,3.69Hz,2H),3.58(br s,4H),3.30(br s,1H),2.79(br t,J=12.69Hz,2H),2.67(d,J=4.63Hz,3H),2.52-2.53(m,4H),1.75-1.93(m,2H),1.23(s,2H),0.85(d,J=6.88Hz,6H).
Example 31: synthesis of 2- ((1- (3-aminopropyl) -6- ((3-chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (69)
2- ((1- (3- ((Tert-Butoxycarbonyl) amino) propyl) -6- ((3, 6-dichloro-5-cyanopyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid methyl ester
To a solution of methyl 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] acetate (1 g,2.39 mmol) and tert-butyl N- (3-bromopropyl) carbamate (852.02 mg,3.58 mmol) in DMSO (10 mL) was added K 2CO3 (659.35 mg,4.77mmol, 359.55. Mu.L). The mixture was stirred at 80℃for 12hr. Another batch of tert-butyl N- (3-bromopropyl) carbamate (568.01 mg,2.39 mmol) was added and stirred at 80℃for 12hr. The mixture was treated with water (10 mL) to form a precipitate, the precipitate was filtered, and the filter cake was collected and dried in vacuo to give the title compound (1.4 g, crude) as a yellow solid. LCMS: m+h + = 576.0.
2- ((1- (3- ((Tert-butoxycarbonyl) amino) propyl) -6- ((3, 6-dichloro-5-cyanopyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) acetic acid
To a solution of methyl 2- [ [1- [3- (tert-butoxycarbonylamino) propyl ] -6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-3-quinolinyl ] oxy ] acetate (1.4 g,2.43 mmol) in EtOH (16 mL) and H 2 O (16 mL) was added LiOH.H 2 O (203.84 mg,4.86 mmol). The mixture was stirred at 25℃for 12hr. The reaction mixture was concentrated under reduced pressure to give the title compound (1.5 g, crude) as a yellow solid. LCMS: m+h + =562.1.
(3- (6- ((3, 6-Dichloro-5-cyanopyridin-2-yl) amino) -3- (2- (methylamino) -2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) propyl) carbamic acid tert-butyl ester
To a solution of 2- [ [1- [3- (tert-butoxycarbonylamino) propyl ] -6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-3-quinolinyl ] oxy ] acetic acid (2.2 g,3.91 mmol) and hexamethylenetetramine (396.17 mg,5.87mmol, HCl) in DMF (22 mL) was added HATU (2.97 g,7.82 mmol) and DIPEA (2.02 g,15.65mmol,2.73 mL). The mixture was stirred at 25℃for 12hr. The reaction mixture was treated with water (30 mL) and extracted with EtOAc (30 mL. Times.3). The combined organic layers were washed with brine, dried over anhydrous Na 2SO4, filtered and concentrated under reduced pressure to give a residue which was purified by flash column chromatography (SILICA FLASH column, gradient elution of 0% -100% DMF/ethyl acetate ether) and further purified by preparative HPLC (column: welch Xtimate C, 250 x 70mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ];: B%:30% -60%,20 min) to give the title compound as a yellow solid (180 mg,303.42 μmol, yield 8%, purity 97%).
(3- (6- ((3-Chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -3- (2- (methylamino) -2-oxoethoxy) -2-oxoquinolin-1 (2H) -yl) propyl) carbamic acid tert-butyl ester
To a solution of tert-butyl N- [3- [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -3- [2- (methylamino) -2-oxoethoxy ] -2-oxo-1-quinolinyl ] propyl ] carbamate (130 mg, 225.91. Mu. Mol) and (3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidine (75.75 mg, 451.83. Mu. Mol, HCl) in DMSO (2 mL) was added DIPEA (116.79 mg, 903.65. Mu. Mol, 157.40. Mu.L). The mixture was stirred at 100deg.C for 12hr. The reaction mixture was treated with water (2 mL) to form a precipitate, which was filtered and dried under reduced pressure to give the title compound as an orange-red oil (130mg).1H NMR(400MHz,DMSO-d6)=9.07(s,1H),7.94(s,1H),7.80(br s,1H),7.63(br dd,J=9.00,2.09Hz,1H),7.48(br d,J=9.18Hz,1H),7.21(s,1H),6.93(br t,J=5.19Hz,1H),4.53(s,2H),4.28(br t,J=7.27Hz,2H),3.97(br dd,J=13.17,3.76Hz,2H),3.04(q,J=6.20Hz,2H),2.78(br t,J=12.58Hz,2H),2.66(d,J=4.53Hz,3H),1.87(br s,1H),1.72-1.82(m,3H),1.39(s,9H),0.92-1.03(m,1H),0.80-0.89(m,6H).
2- ((1- (3-Aminopropyl) -6- ((3-chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (69)
To a solution of tert-butyl N- [3- [6- [ [ 3-chloro-5-cyano-6- [ (3S, 4S, 5R) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -3- [2- (methylamino) -2-oxoethoxy ] -2-oxo-1-quinolinyl ] propyl ] carbamate (100 mg, 149.22. Mu. Mol) in HCl/EtOAc (10 mL). The mixture was stirred at 20deg.C for 0.5hr. The reaction mixture was concentrated under reduced pressure to give a residue which was purified by preparative HPLC (column: phenomnex C18.40 mm. Times.3 μm; mobile phase: [ water (NH 4HCO3) -ACN ];B%:28% -58%,8 min) to give the title compound (26 mg, 43.33. Mu. Mol, 29% yield, purity) as a yellow solid 95%).1H NMR(400MHz,DMSO-d6)δ8.94-9.19(m,1H),7.92-8.00(m,2H),7.80(br s,1H),7.64(br d,J=8.23Hz,1H),7.55(br d,J=8.34Hz,1H),7.22(br s,1H),4.53(br s,2H),4.34(br s,2H),3.97(br d,J=11.68Hz,2H),2.78(br t,J=12.52Hz,2H),2.60-2.68(m,5H),1.66-1.93(m,5H),0.84(br d,J=6.44Hz,6H).
Example 32: synthesis of 5-chloro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-3- ((S) -morpholin-2-ylmethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) nicotinonitrile (70) and 5-chloro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-3- ((R) -morpholin-2-ylmethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) nicotinonitrile (71)
3-Hydroxy-1-methyl-6-nitroquinolin-2 (1H) -one
Azimethyl (trimethyl) silane (2M, 72.76 mL) was added dropwise to a solution of 1-methyl-5-nitroindoline-2, 3-dione (25 g,121.27 mmol) and TEA (24.54 g,242.54mmol,33.76 mL) in EtOH (750 mL) under N 2. The mixture was stirred at 20deg.C for 12hr. The mixture was concentrated under reduced pressure, and 1N aqueous HCl (300 mL) was added. The mixture was then stirred at 20deg.C for 2hr, filtered and washed with DMF/ethyl acetate (10/1, 150 mL) and the filter cake dried under reduced pressure to give the title compound as a brown solid (9.2 g,35.18mmol, 15% yield, purity) 84%).1H NMR(400MHz,DMSO-d6)δ10.08(s,1H),8.55(d,J=2.69Hz,1H),8.19(dd,J=9.29,2.69Hz,1H),7.65(d,J=9.41Hz,1H),7.34(s,1H),3.74(s,3H).
2- (((1-Methyl-6-nitro-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) methyl) morpholine-4-carboxylic acid tert-butyl ester
To a solution of 3-hydroxy-1-methyl-6-nitroquinolin-2 (1H) -one (1 g,4.54 mmol) and tert-butyl 2- (bromomethyl) morpholine-4-carboxylate (1.53 g,5.45 mmol) in DMSO (20 mL) was added DBU (829.70 mg,5.45mmol, 821.49. Mu.L) under N 2. The mixture was stirred at 100deg.C for 3hr. The mixture (combined with another batch at 200mg scale) was cooled to 20 ℃ and water (ca. 30 mL) was added to form a precipitate, which was filtered and the filter cake washed with EtOAc (20 mL) and concentrated in vacuo to give the title compound as a yellow solid (1.2 g,2.00mmol, 44% yield, 70% purity). LCMS: [ M-100] + =320.1.
2- (((6-Amino-1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) methyl) morpholine-4-carboxylic acid tert-butyl ester
To a mixture of Pd/C (0.1 g, purity 10%) in THF (20 mL) under Ar was added tert-butyl 2- (((1-methyl-6-nitro-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) methyl) morpholine-4-carboxylate (0.6 g,1.43 mmol). The flask was degassed under vacuum and purged several times with H 2. The mixture was stirred at H 2 (15 psi) and 20℃for 12 hours. The mixture was filtered and the filtrate concentrated in vacuo to give the title compound as a yellow solid (600 mg,1.39mmol, 97% yield, purity 90%).1H NMR(400MHz,DMSO-d6)δ7.17(d,J=8.82Hz,1H),7.05(s,1H),6.77(dd,J=8.88,2.56Hz,1H),6.71(d,J=2.50Hz,1H),5.03(s,2H),4.00(d,J=5.01Hz,2H),3.94(d,J=13.35Hz,1H),3.86(d,J=10.25Hz,1H),3.69-3.77(m,2H),3.56(s,3H),3.46(td,J=11.56,2.62Hz,1H),2.89(d,J=3.22Hz,2H),1.41(s,9H).
2- (((6- ((3, 6-Dichloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) methyl) morpholine-4-carboxylic acid tert-butyl ester
A flask charged with a mixture of tert-butyl 2- (((6-amino-1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) methyl) morpholine-4-carboxylate (1 g,2.57 mmol), 2,5, 6-trichloronicotinonitrile (532.67 mg,2.57 mmol) and DIEA (663.73 mg,5.14mmol, 894.52. Mu.L) in DMF (20 mL) was degassed and purged 3 times with N 2 and then the mixture stirred at 100deg.C under an atmosphere of N 2 for 12hr. The mixture was cooled to 20 ℃ and water (20 mL) was added to form a precipitate, the precipitate was filtered and the filter cake was dried in vacuo to give the title compound as a yellow solid (1 g,1.48mmol, 58% yield, 83% purity). LCMS: [ m+h ] + =560.2.
2- (((6- ((3-Chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) methyl) morpholine-4-carboxylic acid tert-butyl ester
To a solution of tert-butyl 2- (((6- ((3, 6-dichloro-5-cyanopyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) methyl) morpholine-4-carboxylate (200 mg, 356.87. Mu. Mol) and (3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidine hydrochloride (119.66 mg, 713.74. Mu. Mol, HCl) in DMSO (4 mL) was added DIEA (230.61 mg,1.78mmol, 310.80. Mu.L). The mixture was stirred at 100deg.C for 12hr. The mixture (combined with another batch at 100mg scale) was cooled to 20 ℃ and water (10 mL) was added to form a precipitate, which was filtered and the filter cake was concentrated in vacuo to give the title compound as a white solid (300 mg, purity 70%). LCMS: m+h + = 655.3.
5-Chloro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-3- (morpholin-2-ylmethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) nicotinonitrile
A mixture of tert-butyl 2- (((6- ((3-chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1-methyl-2-oxo-1, 2-dihydroquinolin-3-yl) oxy) methyl) morpholine-4-carboxylate (250 mg, 381.59. Mu. Mol) and HCl/EtOAc (4M, 10.00 mL) (4M HCl in EtOAc) was stirred at 20℃for 1hr under an atmosphere of N 2. The mixture was concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: phenomenex Luna C, 80 x 40mm x 3 μm; mobile phase: [ water (HCl) -ACN ];% B: 30% -60%,7 min) to give the title compound (110 mg, 184.11. Mu. Mol, yield 48%, purity) as a yellow solid 99%,HCI).1H NMR(400MHz,DMSO-d6)δ9.27-9.41(m,2H),9.09(s,1H),7.96(s,1H),7.86(d,J=2.32Hz,1H),7.63(dd,J=9.05,2.45Hz,1H),7.47(d,J=9.05Hz,1H),7.25(s,1H),4.11-4.20(m,2H),4.08(d,J=4.65Hz,2H),3.96-4.05(m,3H),3.78-3.88(m,2H),3.67(s,3H),2.94-3.04(m,2H),2.79(t,J=12.59Hz,2H),1.77
-1.94(m,2H),0.86(d,J=6.85Hz,6H)。
5-Chloro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-3- ((S) -morpholin-2-ylmethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) nicotinonitrile (70)
Racemic 5-chloro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-3- (morpholin-2-ylmethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) nicotinonitrile (110 mg, 185.97. Mu. Mol, HCl) was separated by SFC (column: REGIS (S, S) WHELK-O1 (250 mm. Times.25 mm, 10. Mu.m); mobile: [0.1% NH 3H2 O MeOH ];: 60% -60%,40 min) to give the title compound (40 mg, 71.92. Mu. Mol, 39% purity) as a yellow solid 99%).1H NMR(400MHz,DMSO-d6)δ9.04(s,1H),7.93-7.95(m,1H),7.88(d,J=2.32Hz,1H),7.59(dd,J=9.05,2.32Hz,1H),7.43(d,J=9.05Hz,1H),7.20(s,1H),4.41-4.59(m,1H),3.85-4.04(m,5H),3.74(s,2H),3.65(s,3H),3.48(td,J=10.64,3.42Hz,1H),2.89(dd,J=12.17,1.41Hz,1H),2.78(t,J=12.65Hz,2H),2.64-2.72(m,3H),1.76-1.96(m,2H),0.86(d,J=6.97Hz,6H).
5-Chloro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-3- ((R) -morpholin-2-ylmethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) nicotinonitrile (71)
Racemic 5-chloro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-3- (morpholin-2-ylmethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) nicotinonitrile (110 mg, 185.97. Mu. Mol, HCl) was separated by SFC (column: REGIS (S, S) WHELK-O1 (250 mm. Times.25 mm, 10. Mu.m); mobile: [0.1% NH 3H2 O MEOH ]; B%:60% -60%,40 min) to give the title compound (25 mg, 44.56. Mu. Mol, 24% purity) as a yellow solid 99%).1H NMR(400MHz,DMSO-d6)δ9.05(s,1H),7.94(s,1H),7.87(d,J=2.20Hz,1H),7.58(dd,J=8.99,2.26Hz,1H),7.43(d,J=9.05Hz,1H),7.20(s,1H),4.42-4.59(m,1H),3.87-4.03(m,5H),3.75(d,J=10.51Hz,2H),3.65(s,3H),3.48(td,J=10.64,3.30Hz,1H),2.89(d,J=10.76Hz,1H),2.78(t,J=12.65Hz,2H),2.61-2.72(m,2H),1.76-1.93(m,2H),0.86(d,J=6.85Hz,6H).
Example 33: synthesis of 2- ((6- ((3-chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2- (dimethylamino) ethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (72)
2- ((6- ((3, 6-Dichloro-5-cyanopyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
To a solution of methyl 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] acetate (3 g,7.16 mmol) in NMP (3 mL) and EtOH (27 mL) was added MeNH 2 (4.44 g,57.25mmol, 40% purity). The mixture was stirred at 70℃for 12hr. The reaction mixture was cooled to 20 ℃ and EtOH (90 mL) was added and the suspension filtered. The filter cake was dried under reduced pressure to give the title compound (2.6 g,5.41mmol, yield 76%, purity 87%) as a yellow solid.
2- ((6- ((3-Chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
To a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] -N-methylacetamide (500 mg,1.20 mmol) and (3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidine (300.64 mg,1.79mmol, HCl) in DMSO (1 mL) was added DIEA (618.04 mg,4.78mmol, 832.94. Mu.L). The mixture was stirred at 100deg.C for 2hr, then cooled to room temperature and treated with water (1 mL) to form a precipitate and filtered. The filter cake was washed with EtOAc (2 mL) and then dried under reduced pressure to give the title compound as a brown solid (600 mg,818.77 μmol, yield 68%, purity 70%).
2- ((6- ((3-Chloro-5-cyano-6- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2- (dimethylamino) ethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (72)
To a solution of 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3 s,4s,5 r) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] -N-methylacetamide (100 mg,194.95 μmol) and 2-chloro-N, N-dimethylethylamine (42.12 mg,292.42 μmol, HCl) in DMSO (1 mL) was added KI (16.18 mg,97.47 μmol) and Cs 2CO3 (190.55 mg,584.84 μmol). The mixture was stirred at 60 ℃ and then heated to 80 ℃ for 2hr. The reaction mixture was treated with water (3 mL) to form a precipitate, which was filtered and the filter cake was washed with EtOAc (6 mL). The filter cake was collected and dried in vacuo to give the title compound as an orange solid (2.6 mg, 4.23. Mu. Mol, yield 2%, purity) 95%).1H NMR(400MHz,MeOD-d4)δ8.02(d,J=2.38Hz,1H),7.76-7.80(m,1H),7.75(s,1H),7.58(d,J=9.18Hz,1H),7.39-7.43(m,1H),4.83-4.84(m,2H),4.61(s,2H),4.41-4.58(m,1H),4.11(dd,J=12.87,3.93Hz,2H),3.61(t,J=6.02Hz,2H),3.09(s,6H),2.85-2.93(m,5H),1.80-2.00(m,2H),0.98(d,J=6.91Hz,6H).
Example 34: synthesis of 2- ((6- ((3-chloro-5-cyano-6- ((3S, 4S, 5R) -4-hydroxy-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2- (dimethylamino) ethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (73)
2- ((6- ((3-Chloro-5-cyano-6- ((3S, 4S, 5R) -4-hydroxy-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
To a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] -N-methylacetamide (300 mg, 717.30. Mu. Mol) and (3R, 5S) -3, 5-dimethylpiperidin-4-ol (178.24 mg,1.08mmol, HCl) in DMSO (1 mL) was added DIPEA (370.82 mg,2.87mmol, 499.75. Mu.L). The mixture was stirred at 100deg.C for 12hr. Water (5 mL) was then added to form a precipitate, the precipitate was filtered, and the filter cake was washed with water (10 mL) and EtOAc (20 mL) and then dried in vacuo to give the title compound as a brown solid (250 mg, 457.95. Mu. Mol, 64% yield, purity) 94%).1H NMR(400MHz,DMSO-d6)δ12.04(s,1H),8.99(s,1H),7.99(m,1H),7.92(s,1H),7.76(d,J=2Hz,1H),7.61(dd,J=8.8,2.4Hz,1H),7.30(d,J=8.8Hz,1H),7.25(s,1H),4.61-4.58(m,3H),3.92(dd,J=13.2,3.2Hz,2H),3.49(d,J=4.4Hz,1H),2.91–2.83(m,2H),2.74(d,J=4.8Hz,3H),1.72-1.69(m,2H),0.84(d,J=6.8Hz,6H).
2- ((6- ((3-Chloro-5-cyano-6- ((3S, 4S, 5R) -4-hydroxy-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2- (dimethylamino) ethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (73)
To a solution of 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3R, 5S) -4-hydroxy-3, 5-dimethyl-1-piperidinyl) -2-pyridinyl ] amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] -N-methylacetamide (150 mg, 293.56. Mu. Mol) and 2-chloro-N, N-dimethylethylamine (63.43 mg, 440.34. Mu. Mol, HCl) in DMSO (3 mL) was added Cs 2CO3 (286.94 mg, 880.68. Mu. Mol) and KI (24.37 mg, 146.78. Mu. Mol). The mixture was stirred at 60℃for 2hr. Water (2 mL) was added to the mixture (combined with another batch at 70mg scale) and filtered. The filter cake was dried in vacuo to give a residue which was purified by preparative HPLC (column: waters Xbridge BEH C.sup.18X105 mm. Sup.10μm; mobile phase: [ water (NH 4HCO3) -ACN ];B%:35% -65%,8 min) to give the title compound as a white solid (purity) 96%).1H NMR(400MHz,DMSO-d6)δ9.00(br s,1H),7.96(br d,J=4.41Hz,1H),7.88(s,1H),7.77(d,J=2.15Hz,1H),7.69(dd,J=9.06,2.15Hz,1H),7.45(d,J=9.18Hz,1H),7.19(s,1H),4.52(s,3H),4.39(br t,J=7.09Hz,2H),3.87(br dd,J=12.64,3.10Hz,2H),3.43(br s,1H),3.21-3.29(m,2H),2.83(br t,J=12.46Hz,2H),2.67(d,J=4.53Hz,3H),2.23(s,6H),1.59-1.73(m,2H),0.79(d,J=6.79Hz,6H).
Example 35: synthesis of 5-chloro-2- ((3S, 5R) -4, 4-difluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (74) and 5-chloro-2- ((3R, 5S) -4, 4-difluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (75)
3-Hydroxy-4, 4-dimethoxy-5-methylpiperidine-1-carboxylic acid tert-butyl ester
To a solution of KOH (133.48 g,2.38 mol) in MeOH (590 mL) at 0deg.C was added tert-butyl 3-methyl-4-oxopiperidine-1-carboxylate (118 g,553.28 mmol) in portions, and the mixture was stirred at 0deg.C for 20min. Then [ phenyl- (2, 2-trifluoroacetyl) oxy- λ3-indanyl ]2, 2-trifluoroacetate (356.90 g,829.92mmol,1.5 eq.) is added in portions at 0deg.C. The resulting mixture was stirred at 20deg.C for 12hr. The mixture (combined with the other 4 batches of the same scale) was concentrated in vacuo. Water (2L) was then added and extracted with ethyl acetate (1L x 3). The combined organic phases were washed with brine (2L), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (SILICA FLASH column, gradient of 0% -50% ethyl acetate/petroleum ether) to give the title compound as a yellow oil (750g).1H NMR(400MHz,CDCl3)δ4.04-4.29(m,1H),3.72-3.93(m,2H),3.26(s,3H),3.21(s,3H),2.93-3.13(m,2H),1.95-2.10(m,2H),1.44-1.47(m,9H),1.13(d,J=7.34Hz,3H).
3-Benzyloxy-4, 4-dimethoxy-5-methylpiperidine-1-carboxylic acid tert-butyl ester
To a solution of NaH (29.85 g,746.23mmol, 60% purity) in THF (1L) was added dropwise tert-butyl 3-hydroxy-4, 4-dimethoxy-piperidine-1-carboxylate (150 g,574.02 mmol) in THF (500 mL) at 0deg.C. After the addition, the mixture was stirred at 0deg.C for 30min, and bromomethylbenzene (108.00 g,631.42mmol,75.00 mL) was then added dropwise at 0deg.C. The resulting mixture was stirred at 20deg.C for 11.5hr (4 batches). The reaction mixture (combined with the other 3 batches of the same scale) was quenched with saturated NH 4 Cl (1L). The mixture was concentrated and extracted with ethyl acetate (500 ml x 3). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel (SILICA FLASH column, gradient of 0% -50% ethyl acetate/petroleum ether) to give the title compound as a yellow oil (80g).1H NMR(400MHz,CDCl3)δ7.20-7.44(m,5H),4.74-4.89(m,1H),4.26-4.60(m,2H),3.71-4.01(m,1H),3.43-3.57(m,1H),3.14-3.25(m,6H),2.86-3.12(m,2H),1.98-2.16(m,1H),1.36-1.51(m,9H),1.15(br dd,J=12.04,7.27Hz,3H).
3-Benzyloxy-5-methylpiperidin-4-one
A mixture of 3-benzyloxy-4, 4-dimethoxy-5-methyl-piperidine-1-carboxylic acid tert-butyl ester (84.5 g,231.21 mmol) in H 2 O (300 mL) and TFA (600 mL) was stirred at 20℃for 2H. The reaction mixture was concentrated under reduced pressure to give the title compound (75 g,225.02mmol, 97% yield, TFA) as a yellow oil. LCMS: m+h + =220.2.
(3R, 5S) -3-benzyloxy-5-methylpiperidin-4-one
3-Benzyloxy-5-methyl-piperidin-4-one (60.00 g,180.02mmol, TFA salt) was mixed with saturated aqueous NaHCO 3 (500 mL) and extracted with EtOAc (300 mL x 2). The combined organic phases were washed with brine (500 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (SILICA FLASH column, 30% -100% ethyl acetate/petroleum ether gradient eluent) to give 40g of product mixture as two close spots on TLC. 40g of the mixture was purified by preparative HPLC (column: waters Xbridge BEH C18.100.30 mm.10 μm; mobile phase: [ water (NH 3H2O+NH4HCO3) -ACN ]; B%:15% -50%,8 min) to give the title compound (16 g,66.40mmol, 37% yield, purity) as a colourless oil 91%).1H NMR(400MHz,CDCl3)δ7.28-7.43(m,5H),4.88(d,J=11.92Hz,1H),4.51(d,J=11.80Hz,1H),3.95(dd,J=10.67,6.74Hz,1H),3.56(ddd,J=12.28,6.79,2.03Hz,1H),3.25-3.38(m,1H),2.79(dd,J=12.10,10.91Hz,1H),2.43-2.55(m,2H),0.99-1.11(m,3H).
(3R, 5S) -benzyl 3- (benzyloxy) -5-methyl-4-oxopiperidine-1-carboxylic acid ester
To a mixture of (3R, 5S) -3-benzyloxy-5-methylpiperidin-4-one (10.00 g,45.60 mmol) in DCM (100 mL) under N 2 was added DIEA (17.68 g,136.81mmol,23.83 mL) in one portion. CbzCl (8.56 g,50.16mmol,7.13 mL) was then added dropwise at 0deg.C under N 2. The mixture was stirred at 15℃for 3 hours. The mixture was washed with 10% citric acid (100 mL) and extracted with ethyl acetate (120 mL x 2). The combined organic phases were washed with brine (200 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. Through flash silica gel column chromatography40g/>SILICA FLASH chromatography, 0% -10% ethyl acetate/petroleum ether gradient eluent @100 mL/min) to give the title compound (10.5 g,27.80mmol, 61% purity) as a yellow oil 94%).1H NMR(400MHz,CDCl3)δ7.28-7.47(m,10H),5.18(br s,2H),4.87(d,J=11.63Hz,1H),4.28-4.81(m,3H),3.97(br s,1H),3.00(br s,1H),2.47-2.82(m,2H),1.08(d,J=6.38Hz,3H).
(3R, 5S) -benzyl 3- (benzyloxy) -4, 4-difluoro-5-methylpiperidine-1-carboxylic acid ester
To a solution of (3R, 5S) -3-benzyloxy-5-methyl-4-oxopiperidine-1-carboxylic acid benzyl ester (3 g,8.49 mmol) in DCM (40 mL) was added DAST (2.74 g,16.98mmol,2.24 mL) at-70 ℃. The mixture was stirred at 15℃for 12hr. The mixture was then quenched with saturated NaHCO 3 (about 60 mL) to ph=8 and the mixture was extracted with DCM (40 mL x 2). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. The residue was purified by flash column chromatography on silica gel (SILICA FLASH column, gradient of 0% -30% ethyl acetate/petroleum ether @100 mL/min) to give the title compound (2 g,5.33mmol, yield) as a colourless oil 63%).1HNMR(400MHz,CDCl3)δ7.27-7.46(m,10H),5.11(br s,2H),4.59-4.89(m,2H),3.84-4.39(m,2H),3.54(br s,1H),3.07(br t,J=11.44Hz,1H),2.83(br d,J=7.51Hz,1H),1.90-2.07(m,1H),1.09(br d,J=6.68Hz,3H).
(3R, 5S) -tert-butyl 4, 4-difluoro-3-hydroxy-5-methylpiperidine-1-carboxylate
To a solution of benzyl (3R, 5S) -3-benzyloxy-4, 4-difluoro-5-methylpiperidine-1-carboxylate (1 g,2.66 mmol) in t-BuOH (5 mL) under Ar was added Pd/C (0.3 g, purity 10%) and Boc 2 O (1.16 g,5.33mmol,1.22 mL). The suspension was degassed under vacuum and purged several times with H 2. The mixture was stirred at H 2 (15 psi) and 60℃for 12 hours. By passing throughThe mixture was filtered and the filtrate concentrated in vacuo to give the title compound (1.5 g, crude) as a colourless oil ).1H NMR(400MHz,MeOD-d4)δ4.04-4.17(m,1H),3.94(br d,J=10.01Hz,1H),3.61-3.75(m,1H),2.57-2.95(m,2H),1.89-2.10(m,1H),1.47(s,9H),1.04(d,J=6.88Hz,3H).
(3R, 5S) -4, 4-difluoro-5-methylpiperidine-3-ol hydrochloride
A solution of (3R, 5S) -4, 4-difluoro-3-hydroxy-5-methylpiperidine-1-carboxylic acid tert-butyl ester (1.5 g,5.97 mmol) in 4N HCl/EtOAc (20 mL) was stirred at 15℃for 1 hour. The mixture was concentrated in vacuo to give the title compound as a white solid (1.1 g,5.86mmol, yield 98%,HCl).1HNMR(400MHz,MeOD-d4)δ4.03-4.18(m,1H),3.47(br d,J=12.38Hz,1H),3.39(br d,J=12.63Hz,1H),3.04(br t,J=11.63Hz,1H),2.94(br t,J=12.26Hz,1H),2.30-2.52(m,1H),1.15(d,J=6.75Hz,3H).
5-Chloro-2- (4, 4-difluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a solution of 2, 5-dichloro-6- [ [3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (100 mg, 237.93. Mu. Mol), 4-difluoro-5-methylpiperidin-3-ol (66.96 mg, 356.89. Mu. Mol, HCl) in DMSO (1 mL) was added DIPEA (123.00 mg, 951.72. Mu. Mol, 165.77. Mu.L). The mixture was stirred at 100deg.C for 12hr. The mixture was cooled to 15 ℃ and purified by preparative HPLC (column: waters Xbridge Prep OBD C, 150 x 40mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:25% -55%,8 min) to give the title compound (50 mg,91.22 μmol, 38% yield, purity) as a white solid 98%).1H NMR(400MHz,DMSO-d6)δ9.01(s,1H),7.95(s,1H),7.29(s,1H),7.22(br d,J=8.13Hz,1H),7.11(br d,J=8.25Hz,1H),5.72(br d,J=5.50Hz,1H),4.46(s,1H),4.23(br d,J=12.26Hz,1H),3.96(br d,J=12.38Hz,1H),3.83-3.91(m,2H),3.57-3.73(m,1H),3.32(br s,3H),2.93(br t,J=11.94Hz,1H),2.71(br t,J=12.63Hz,1H),1.93-2.14(m,1H),1.63-1.75(m,2H),1.16(s,6H),0.79(br d,J=6.50Hz,3H).
5-Chloro-2- ((3S, 5R) -4, 4-difluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (74) and 5-chloro-2- ((3R, 5S) -4, 4-difluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (75)
Racemic 5-chloro-2- (4, 4-difluoro-3-hydroxy-5-methyl-1-piperidinyl) -6- [ [3- (3-hydroxy-3-methyl-butyl) -1-methyl-2-oxo-benzoimidazol-5-yl ] amino ] pyridine-3-carbonitrile (60 mg, 112.15. Mu. Mol) was separated by SFC (column: DAICEL CHIRALPAK IG (250 mm. Times.30 mm, 10. Mu.); mobile phase: [0.1% NH 3H2 O IPA ]; B%:40% -40%,20 min) to give 5-chloro-2- ((3S, 5R) -4, 4-difluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methyl-butyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile as a white solid (18 mg, purity 99%,ee100%);1H NMR(400MHz,MeCN-d3)δ7.72(s,2H),7.34(d,J=1.88Hz,1H),7.16(dd,J=8.38,1.88Hz,1H),7.02(d,J=8.38Hz,1H),4.27-4.36(m,1H),4.04-4.12(m,1H),3.91-3.98(m,2H),3.70-3.88(m,2H),3.34(s,3H),2.94-3.03(m,1H),2.80-2.92(m,2H),2.02-2.12(m,1H),1.77-1.83(m,2H),1.22(s,6H),0.93(d,J=6.75Hz,3H); and white solid 5-chloro-2- ((3R, 5S) -4, 4-difluoro-3-hydroxy-5-methylpiperidin-1-yl) -6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (17 mg, purity 99%,ee99%);1H NMR(400MHz,MeCN-d3)δ7.72(s,2H),7.34(d,J=1.75Hz,1H),7.16(dd,J=8.32,1.94Hz,1H),7.02(d,J=8.38Hz,1H),4.24-4.37(m,1H),4.03-4.12(m,1H),3.95(dd,J=8.82,7.19Hz,2H),3.70-3.88(m,2H),3.34(s,3H),2.94-3.03(m,1H),2.90(s,1H),2.80-2.89(m,1H),2.01-2.12(m,1H),1.77-1.84(m,2H),1.22(s,6H),0.93(d,J=6.88Hz,3H).
The absolute configuration of compounds 74 and 75 is randomly assigned to the cis conformation based on the hydroxyl and methyl groups.
Example 36: synthesis of 2- ((6- ((3-chloro-5-cyano-6- (3-hydroxy-5-methylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2- (dimethylamino) ethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (76)
2- ((6- ((3-Chloro-5-cyano-6- (3-hydroxy-5-methylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
To a solution of 5-methylpiperidin-3-ol (315.40 mg,1.80mmol, HOAc) and 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] -N-methylacetamide (500 mg,1.20 mmol) in DMSO (5 mL) was added DIEA (620.35 mg,4.80mmol, 836.05. Mu.L). The mixture was stirred at 100deg.C for 1hr. The reaction mixture was treated with water (20 mL) to form a precipitate, which was filtered and the filter cake was washed with EtOAc (30 mL). The cake was collected and dried under reduced pressure to give the title compound (600 mg, 567.47. Mu. Mol, yield 47%, purity 47%) as a yellow solid. LCMS: m+h + = 497.1.
2- ((6- ((3-Chloro-5-cyano-6- (3-hydroxy-5-methylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2- (dimethylamino) ethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (76)
To a solution of 2- [ [6- [ [ 3-chloro-5-cyano-6- (3-hydroxy-5-methyl-1-piperidinyl) -2-pyridinyl ] amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] -N-methylacetamide (500 mg,1.01 mmol) and 2-chloro-N, N-dimethylethylamine (217.39 mg,1.51mmol, HCl) in DMSO (5 mL) was added Cs 2CO3 (983.47 mg,3.02 mmol) and KI (83.51 mg, 503.07. Mu. Mol). The mixture was stirred at 60℃for 2hr. The reaction mixture was treated with water (30 mL) to form a precipitate, which was filtered and the filter cake was washed with EtOAc (30 mL). The filter cake was collected and dried under reduced pressure to give a residue which was purified by preparative HPLC (column: phenomnex Luna 80 x 30mm x 3 μm; mobile phase: [ water (HCl) -ACN ];% B: 15% -35%,8 min) to give the title compound (8 mg, 12.53. Mu. Mol, yield 1%, purity) as a yellow solid 95%,HCI).1H NMR(400MHz,DMSO-d6)δ10.07(br s,1H),9.03(s,1H),8.04(d,J=2.03Hz,1H),7.92-7.99(m,2H),7.67-7.71(m,1H),7.61-7.65(m,1H),7.32(s,1H),4.66(br t,J=6.44Hz,2H),4.54(s,2H),4.33(br dd,J=12.10,3.99Hz,1H),4.12(br d,J=12.76Hz,1H),3.51(br dd,J=10.55,4.47Hz,2H),2.91(d,J=4.65Hz,6H),2.68(d,J=4.65Hz,3H),2.57(br d,J=12.28Hz,1H),2.54(s,1H),2.41-2.45(m,1H),1.98(br d,J=12.04Hz,1H),1.59-1.72(m,1H),1.00(q,J=11.88Hz,1H),0.82(d,J=6.56Hz,3H).
Example 37: synthesis of 2- ((6- ((3-chloro-5-cyano-6- ((3R, 4R, 5S) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2- (dimethylamino) ethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (77)
(3R, 4S, 5S) -1-benzyl-3, 5-dimethylpiperidin-4-ol
To a solution of (3R, 5S) -1-benzyl-3, 5-dimethylpiperidin-4-one (4 g,18.41 mmol) in MeOH (45 mL) at 0deg.C was added NaBH 4 (835.67 mg,22.09 mmol) for 10 min. The mixture was stirred at 20deg.C for 12hr. The reaction mixture was quenched by addition of 1N HCl (40 mL) at 20deg.C, and the mixture was concentrated under reduced pressure. The residue was dissolved in ethyl acetate (40 mL) and washed with water (40 mL x 3), the combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel (SILICA FLASH column, 0% -20% ethyl acetate/petroleum eluent) to give the title compound as a white solid (1 g,3.65mmol, 20% yield, 80% purity). LCMS: m+h + =220.2.
(3R, 4R, 5S) -1-benzyl-4-fluoro-3, 5-dimethylpiperidine
DAST (955.43 mg,5.93mmol, 783.14. Mu.L) was added to a solution of (3R, 4S, 5S) -1-benzyl-3, 5-dimethylpiperidin-4-ol (650 mg,2.96 mmol) in DCM (7 mL) at-55℃and N 2. The mixture was stirred at 20deg.C for 12hr. Saturated NaHCO 3 (30 mL) was added to the mixture, and the mixture was extracted with DCM (3X 20 mL). The combined organic phases were dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography (silica gel, petroleum ether/ethyl acetate=1/0 to 10 to 1) to give the title compound (500 mg,2.09mmol, yield 70%, purity) as a yellow oil 92%).1H NMR(400MHz,DMSO-d6)δ7.23-7.35(m,5H),3.59-3.78(m,1H),3.45(s,2H),2.72-2.80(m,2H),1.75-1.84(m,2H),1.67-1.74(m,2H),0.90(d,J=6.36Hz,6H).
(3R, 4R, 5S) -4-fluoro-3, 5-dimethylpiperidine
To a Pd/C (0.1 g, purity 10%) mixture in EtOH (5 mL) under Ar was added (3R, 4R, 5S) -1-benzyl-4-fluoro-3, 5-dimethylpiperidine (500 mg,2.26 mmol). The flask was degassed under vacuum and purged several times with H 2. The mixture was stirred at H 2 (15 psi) and 50℃for 3 hours. By usingThe mixture was pad filtered and washed with EtOH (500 mL). To the filtrate was added HCl/EtOAc (4M in EtOAc, 3 mL). After stirring at 20℃for 12h, the mixture was concentrated in vacuo to give the title compound as a white solid (360 mg,2.15mmol, yield 95%,HCl).1H NMR(400MHz,DMSO-d6)δ3.91-4.13(m,1H),3.17-3.29(m,2H),2.60-2.69(m,2H),1.98-2.14(m,2H),0.97-1.00(m,6H).
2- ((6- ((3-Chloro-5-cyano-6- ((3 r,4r,5 s) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide
A flask containing a mixture of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] -N-methylacetamide (200 mg, 478.20. Mu. Mol), (3R, 4R, 5S) -4-fluoro-3, 5-dimethylpiperidine (120.26 mg, 717.30. Mu. Mol, HCl) and DIEA (432.63 mg,3.35mmol, 583.06. Mu. L) in DMSO (2 mL) was degassed and purged 3 times with N 2, and then the mixture was stirred at 100℃under an atmosphere of N 2 for 12hr. Water (10 mL) was added to the mixture to form a precipitate, and the precipitate was filtered. The filter cake was dried under reduced pressure and the residue triturated with petroleum ether/ethyl acetate=3:1 (5 mL) at 15 ℃ for 10min to give the title compound as a grey solid (280 mg,436.68 μmol, 91% yield, 80% purity). LCMS: m+h + =513.2.
2- ((6- ((3-Chloro-5-cyano-6- ((3R, 4R, 5S) -4-fluoro-3, 5-dimethylpiperidin-1-yl) pyridin-2-yl) amino) -1- (2- (dimethylamino) ethyl) -2-oxo-1, 2-dihydroquinolin-3-yl) oxy) -N-methylacetamide (77)
A flask containing a mixture of 2- [ [6- [ [ 3-chloro-5-cyano-6- [ (3S, 5R) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -2-pyridinyl ] amino ] -2-oxo-1H-quinolin-3-yl ] oxy ] -N-methylacetamide (100 mg, 194.95. Mu. Mol), 2-chloro-N, N-dimethylethylamine (30.89 mg, 214.44. Mu. Mol, HCl), cs 2CO3 (190.55 mg, 584.84. Mu. Mol) and KI (16.18 mg, 97.47. Mu. Mol) in DMSO (1 mL) was degassed and purged 3 times with N 2, and then the mixture was stirred at 60℃under an atmosphere of N 2 for 2hr. The mixture was concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: phenomenex C18 x 40mm x 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:25% -60%,8 min) to give the title compound (25 mg, purity) as a white solid 92.27%).1H NMR(400MHz,DMSO-d6)δ0.87(d,J=6.48Hz,6H),1.66-1.78(m,2H),2.24(s,6H),2.66-2.68(m,5H),3.30(br s,2H),3.79-3.99(m,1H),4.09-4.19(m,2H),4.39(br t,J=7.15Hz,2H),4.53(s,2H),7.22(s,1H),7.46(d,J=9.17Hz,1H),7.66(dd,J=9.05,2.32Hz,1H),7.75(d,J=2.32Hz,1H),7.92-7.97(m,2H),9.09(s,1H).
Example 38: synthesis of (3R, 5S) -1- (5-chloro-3-cyano-6- ((1-methyl-3- (2- (methylamino) -2-oxoethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) pyridin-2-yl) -5-methylpiperidine-3-carboxylic acid (78) and (3S, 5R) -1- (5-chloro-3-cyano-6- ((1-methyl-3- (2- (methylamino) -2-oxoethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) pyridin-2-yl) -5-methylpiperidine-3-carboxylic acid (79)
5-Methylpiperidine-3-carboxylic acid methyl ester hydrochloride
To a solution of methyl 5-methylpyridine-3-carboxylate (20 g,132.31 mmol) in 1.25N HCl/MeOH (150 mL) was added PtO 2 (3.00 g,13.23 mmol) under Ar. The flask was degassed and purged several times with H 2. The mixture was stirred at H 2 (50 psi) and 25℃for 12 hours. By usingThe reaction mixture was pad filtered and washed with MeOH (500 mL). The filtrate was concentrated under reduced pressure to give the title compound (21 g, crude, HCl) as a colourless oil. LCMS: m+h + =158.3.
1- (5-Chloro-3-cyano-6- ((1-methyl-3- (2- (methylamino) -2-oxoethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) pyridin-2-yl) -5-methylpiperidine-3-carboxylic acid methyl ester
To a solution of 2- [ [6- [ (3, 6-dichloro-5-cyano-2-pyridinyl) amino ] -1-methyl-2-oxo-3-quinolinyl ] oxy ] -N-methylacetamide (300 mg,694.03 μmol) and methyl 5-methylpiperidine-3-carboxylate (201.62 mg,1.04mmol, hcl) in DMSO (5 mL) was added DIPEA (358.78 mg,2.78mmol,483.54 μl). The mixture was stirred at 100deg.C for 12hr. The mixture was cooled to 20 ℃ and water (3 mL) was added to form a precipitate, the precipitate was filtered and the filter cake was concentrated to give the title compound as a yellow solid (300 mg,542.49 μmol, 78% yield). LCMS: m+h + = 553.2.
1- (5-Chloro-3-cyano-6- ((1-methyl-3- (2- (methylamino) -2-oxoethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) pyridin-2-yl) -5-methylpiperidine-3-carboxylic acid (cis)
To a solution of methyl 1- [ 5-chloro-3-cyano-6- [ [ 1-methyl-3- [2- (methylamino) -2-oxoethoxy ] -2-oxo-6-quinolinyl ] amino ] -2-pyridinyl ] -5-methylpiperidine-3-carboxylate (250 mg, 452.07. Mu. Mol) in H 2 O (2 mL), meOH (2 mL), and DMSO (2 mL) was added LiOH.H 2 O (189.71 mg,4.52 mmol). The mixture was stirred at 25℃for 12hr. To the mixture at ph=4 (combined with another batch at 50mg scale) was then added 1N HCl (10 mL) and the mixture was extracted with ethyl acetate (10 mL x 2). The combined organic phases were washed with brine (20 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo. The residue was purified by preparative HPLC (column: phenomenex Luna80 x 30mm x 3 μm; mobile phase: [ water (TFA) -ACN ]; B%:30% -60%,8 min) to give the title compound (60 mg, purity 95%, cis) as a white solid ).1H NMR(400MHz,DMSO-d6)δ9.08-9.18(m,1H),7.95-8.04(m,2H),7.78(d,J=2.38Hz,1H),7.61-7.68(m,1H),7.50-7.57(m,1H),7.22-7.28(m,1H),4.63-4.78(m,1H),4.50-4.60(m,2H),4.27-4.40(m,2H),4.13(br d,J=12.51Hz,2H),3.54(br t,J=5.69Hz,2H),2.79(br t,J=12.76Hz,2H),2.67(d,J=4.50Hz,3H),1.96-2.15(m,2H),1.75-1.86(m,2H),0.82-0.86(m,6H).
(3R, 5S) -1- (5-chloro-3-cyano-6- ((1-methyl-3- (2- (methylamino) -2-oxoethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) pyridin-2-yl) -5-methylpiperidine-3-carboxylic acid (78) and (3S, 5R) -1- (5-chloro-3-cyano-6- ((1-methyl-3- (2- (methylamino) -2-oxoethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) pyridin-2-yl) -5-methylpiperidine-3-carboxylic acid (79)
Racemic 1- [ 5-chloro-3-cyano-6- [ [ 1-methyl-3- [2- (methylamino) -2-oxoethoxy ] -2-oxo-6-quinolinyl ] amino ] -2-pyridinyl ] -5-methylpiperidine-3-carboxylic acid (60 mg, 111.32. Mu. Mol) was separated by SFC (column: DAICEL CHIRALCEL OJ (250 mm. Mu. 30mm, 10. Mu.); mobile phase: [0.1% NH 3H2 O MeOH ]; B%:36% -36%,10 min) to give (3R, 5S) -1- (5-chloro-3-cyano-6- ((1-methyl-3- (2- (methylamino) -2-oxoethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) pyridin-2-yl) -5-methylpiperidine-3-carboxylic acid (10.7 mg, 19.14. Mu. Mol, 17%, purity 96%);1H NMR(400MHz,DMSO-d6)δ9.00(s,1H),8.15(br s,1H),7.89-7.96(m,2H),7.68(br d,J=9.01Hz,1H),7.41(br d,J=9.13Hz,1H),7.21(s,1H),4.59(s,2H),4.46(br d,J=12.13Hz,1H),4.16(br d,J=12.38Hz,1H),3.66(s,3H),2.81(br t,J=12.38Hz,1H),2.66(br d,J=4.38Hz,3H),2.44(br s,1H),2.02(br d,J=12.38Hz,1H),1.54-1.73(m,1H),0.82(br d,J=6.38Hz,3H); and (3 s,5 r) -1- (5-chloro-3-cyano-6- ((1-methyl-3- (2- (methylamino) -2-oxoethoxy) -2-oxo-1, 2-dihydroquinolin-6-yl) amino) pyridin-2-yl) -5-methylpiperidine-3-carboxylic acid (10.5 mg, 18.84. Mu. Mol, 17% yield, purity as a white solid 97%);1HNMR(400MHz,DMSO-d6)δ9.02(s,1H),8.01(br s,1H),7.93(s,1H),7.87(d,J=1.88Hz,1H),7.69(dd,J=9.07,2.19Hz,1H),7.41(d,J=9.13Hz,1H),7.20(s,1H),4.56(s,2H),4.44(br d,J=12.76Hz,1H),4.16(br d,J=12.51Hz,1H),3.66(s,3H),2.82(br t,J=12.38Hz,1H),2.67(d,J=4.50Hz,3H),2.43(br s,1H),2.02(br d,J=12.38Hz,1H),1.56-1.71(m,1H),0.82(d,J=6.50Hz,3H).
The absolute configuration of compounds 78 and 79 is randomly assigned to the cis-conformation based on the carboxy and methyl groups.
Example 39: synthesis of 2- ((3S, 5R) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile hydrochloride (80) and 2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile hydrochloride (81)
2- ((3 S,5 r) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A flask containing a mixture of 2-chloro-5-fluoro-6- [ [3- (3-hydroxy-3-methyl-butyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (200 mg, 495.25. Mu. Mol), 2- [ (3S, 5R) -4, 4-difluoro-5-methyl-3-piperidinyl ] isoindoline-1, 3-dione (138.80 mg, 495.25. Mu. Mol) and DIEA (320.04 mg,2.48mmol, 431.32. Mu. L) in DMSO (3 mL) was degassed and purged 3 times with N 2, and then the mixture was stirred at 130℃under an atmosphere of N 2 for 16hr. To the mixture was added water (15 mL), a precipitate was formed, the precipitate was filtered and washed with H 2 O (3 mL), and then the solid was dried under reduced pressure to give the title compound as a red solid (260 mg,148.54 μmol, yield 30%, purity 37%). LCMS: m+h + = 648.1.
2- ((3 S,5 r) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile hydrochloride (80)
To a solution of 2- ((3 s,5 r) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (260 mg, 401.45. Mu. Mol) in EtOH (16 mL) was added MeNH 2/H2 O (16 mL, purity 40%). The mixture was stirred at 70℃for 1hr. The mixture was concentrated in vacuo and purified by preparative HPLC (column: phenomnex Luna 80 x 30mm x 3 μm; mobile phase: [ water (HCl) -ACN ]; B%:15% -45%,8 min) to give the title compound (50 mg, 84.84. Mu. Mol, 21% yield, purity) as a yellow solid 94%,HCl).1H NMR(400MHz,MeOD-d4,25℃)δ7.60(d,J=10.49Hz,1H),7.44(d,J=1.67Hz,1H),7.37(dd,J=8.40,1.85Hz,1H),7.15(d,J=8.46Hz,1H),4.28-4.45(m,1H),3.97-4.08(m,3H),3.74-3.89(m,1H),3.43(s,3H),3.23(t,J=12.28Hz,1H),2.89(t,J=12.70Hz,1H),2.33-2.56(m,1H),1.86(dd,J=8.88,7.57Hz,2H),1.29(d,J=2.50Hz,6H),1.06(d,J=6.68Hz,3H).
2- ((3 R,5 s) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino)
A flask containing a mixture of 2-chloro-5-fluoro-6- [ [3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (200 mg, 495.25. Mu. Mol), 2- [ (3R, 5S) -4, 4-difluoro-5-methyl-3-piperidinyl ] isoindoline-1, 3-dione (138.80 mg, 495.25. Mu. Mol) and DIEA (320.04 mg,2.48mmol, 431.32. Mu.L) in DMSO (3 mL) was degassed and purged 3 times with N 2, and then the mixture was stirred at 130℃under an atmosphere of N 2 for 16hr. To the mixture was added water (15 mL), a precipitate was formed, the precipitate was filtered and washed with H 2 O (3 mL), and then the solid was dried under reduced pressure to give the title compound as a red solid (270 mg,154.25 μmol, yield 31%, purity 37%). LCMS: m+h + = 648.1.
2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile hydrochloride (81)
To a solution of 2- ((3 r,5 s) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (270 mg, 416.89. Mu. Mol) in EtOH (16 mL) was added MeNH 2/H2 O (16 mL, purity 40%). The mixture was stirred at 70℃for 1hr. The mixture was concentrated in vacuo and purified directly by preparative HPLC (column: phenomnex Luna 80 x 30mm x3 μm; mobile phase: [ water (HCl) -ACN ]; B%:15% -45%,8 min) to give the title compound (40 mg, 67.87. Mu. Mol, 16% yield, purity) as a yellow solid 94%,HCl).1H NMR(400MHz,MeOD-d4,25℃)δ7.60(d,J=10.49Hz,1H),7.44(s,1H),7.37(d,J=8.34Hz,1H),7.15(d,J=8.46Hz,1H),4.32-4.44(m,1H),3.94-4.12(m,3H),3.73-3.89(m,1H),3.43(s,3H),3.23(t,J=12.22Hz,1H),2.89(t,J=12.64Hz,1H),2.35-2.54(m,1H),1.86(t,J=8.11Hz,2H),1.29(d,J=2.03Hz,6H),1.05(d,J=6.68Hz,3H).
The absolute configuration of compounds 80 and 81 is randomly assigned to the cis conformation based on amino and methyl.
Example 40: synthesis of 2- ((3S, 5R) -4, 4-difluoro-3-hydroxy-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (82) and 2- ((3R, 5S) -4, 4-difluoro-3-hydroxy-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (83)
2- (4, 4-Difluoro-3-hydroxy-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A flask charged with a mixture of 2-chloro-5-fluoro-6- [ [3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (300 mg, 742.87. Mu. Mol), (3R, 5S) -4, 4-difluoro-5-methylpiperidin-3-ol (278.75 mg,1.49mmol, HCl), DIEA (480.06 mg,3.71mmol, 646.98. Mu.L) in DMSO (3 mL) was degassed and purged 3 times with N 2, and then the mixture was stirred at 130℃under an atmosphere of N 2 for 12hr. The mixture was cooled to 15 ℃ and purified directly by preparative HPLC (column: waters Xbridge Prep OBD C, 150 x 40mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:35% -55%,8 min) to give the title compound (150 mg,286.39 μmol, 39% yield, purity) as a yellow solid 99%).1H NMR(400MHz,MeCN-d3)δ7.80(br s,1H),7.45-7.50(m,2H),7.20(dd,J=8.40,1.97Hz,1H),7.01(d,J=8.34Hz,1H),4.19-4.28(m,1H),3.99-4.06(m,1H),3.90-3.98(m,3H),3.76-3.90(m,1H),3.34(s,3H),2.96-3.05(m,1H),2.94(s,1H),2.88(t,J=12.64Hz,1H),2.14-2.26(m,1H),1.77-1.85(m,2H),1.22(d,J=2.03Hz,6H),0.99(d,J=6.79Hz,3H).
2- ((3 S,5 r) -4, 4-difluoro-3-hydroxy-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (82) and 2- ((3 r,5 s) -4, 4-difluoro-3-hydroxy-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (83)
2- [ (3R, 5S) -4, 4-difluoro-3-hydroxy-5-methyl-1-piperidinyl ] -5-fluoro-6- [ [3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (150 mg, 289.28. Mu. Mol) was separated with SFC (column: DAICEL CHIRALPAK IG (250 mm. Mu.m), mobile phase: [ heptane-EtOH ]; B%:20% -20%,10 min) to give 2- [ (3S, 5R) -4-difluoro-3-hydroxy-5-methyl-1-piperidinyl ] -5-fluoro-6- [ [3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile as a white solid (45 mg, 85.92. Mu. Mol, yield 30%, purity 99%);1H NMR(400MHz,MeOD-d4)δ7.34-7.43(m,2H),7.26(br d,J=8.44Hz,1H),7.01(br d,J=8.44Hz,1H),4.16(br d,J=12.47Hz,1H),3.88-3.98(m,3H),3.71(ddd,J=15.28,10.45,5.20Hz,1H),3.20(br s,3H),2.91(br t,J=11.98Hz,1H),2.75(br t,J=12.53Hz,1H),1.96-2.12(m,1H),1.71-1.79(m,2H),1.17(s,6H),0.89(br d,J=6.72Hz,3H); and white solid 2- [ (3R, 5S) -4, 4-difluoro-3-hydroxy-5-methyl-1-piperidinyl ] -5-fluoro-6- [ [3- (3-hydroxy-3-methylbutyl) -1-methyl-2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile, 85.92. Mu. Mol, yield 30%, purity 99%).1H NMR(400MHz,MeOD-d4)δ7.35-7.43(m,2H),7.25(br d,J=8.31Hz,1H),7.01(br d,J=8.19Hz,1H),4.16(br d,J=12.10Hz,1H),3.93(br d,J=8.19Hz,3H),3.64-3.78(m,1H),3.20(br s,3H),2.70-2.96(m,2H),1.95-2.12(m,1H),1.69-1.80(m,2H),1.17(s,6H),0.88(br d,J=6.60Hz,3H).
The absolute configuration of compounds 82 and 83 is randomly assigned to the cis-conformation based on the hydroxyl and methyl groups.
Example 41: synthesis of 2- ((3S, 5R) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((1-methyl-2-oxo-3- (((R) -2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (84) and 2- [ (3R, 5S) -3-amino-4, 4-difluoro-5-methyl-1-piperidinyl ] -5-fluoro-6- [ [ 1-methyl-2-oxo-3- [ [ (5S) -2-oxooxazolidin-5-yl ] methyl ] benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (85)
5- ((3-Methyl-6-nitro-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-1-yl) methyl) oxazolidin-2-one
A flask containing a mixture of 1-methyl-5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one (5 g,25.89 mmol), 5- (chloromethyl) oxazolidin-2-one (5.26 g,38.84 mmol), K 2CO3 (7.16 g,51.78 mmol) and KI (2.15 g,12.95 mmol) in DMSO (50 mL) was degassed and purged 3 times with N 2 and then the mixture stirred at 80℃under an atmosphere of N 2 for 6hr. To the mixture was added water (80 mL), a precipitate formed, and the precipitate was filtered and washed with H 2 O (20 mL) and petroleum ether/ethyl acetate= (1:1, 30 mL). The solid was dried under reduced pressure to give the title compound (5.5 g,18.07mmol, yield 70%, purity 96%) as a yellow solid. LCMS: m+h + = 293.1.
5- ((6-Amino-3-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-1-yl) methyl) oxazolidin-2-one
To a mixture of Pd/C (1 g, purity 10%) in DMF (300 mL) was added 5- ((3-methyl-6-nitro-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-1-yl) methyl) oxazolidin-2-one (5.5 g,18.82 mmol). The mixture was stirred at 50℃under H 2 (50 psi) for 20 hours. Will beAdded to the mixture and filtered to give a filtrate, which was concentrated and purified by column chromatography (SiO 2, ethyl acetate/etoh=1/0 to 1/1) to give the title compound (3.3 g,11.83mmol, 63% yield, purity) as a yellow solid 94%).1H NMR(400MHz,DMSO-d6)δ7.53(s,1H),6.80(d,J=8.23Hz,1H),6.46(d,J=1.19Hz,1H),6.33(dd,J=8.23,1.55Hz,1H),4.75-4.89(m,3H),3.89-4.00(m,2H),3.57(t,J=8.82Hz,1H),3.31(dd,J=8.58,6.56Hz,1H),3.23(s,3H).
(R) -5- ((6-amino-3-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-1-yl) methyl) oxazolidin-2-one
Isolation of racemic 5- ((6-amino-3-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-1-yl) methyl) oxazolidin-2-one (2.7 g,10.29 mmol) by SFC (column: DAICEL CHIRALPAK AD (250 mm. Times.50 mm,10 μm); mobile phase; [0.1% NH 3H2 O MeOH ];% B: 50% -50%,3.9 min) afforded the title compound (1.2 g,4.26 mmol) as a yellow solid in 41% yield and purity 93%).1H NMR(400MHz,DMSO-d6)δ7.55(s,1H),6.80(d,J=8.4Hz,1H),6.47(s,1H),6.35(dd,J=8.0,2.4Hz,1H),4.83-4.88(m,2H),3.93-4.00(m,2H),3.58(t,J=8.8Hz,1H),3.32(dd,J=8.8Hz,6.4Hz,1H),3.23(s,3H).
(R) -2-chloro-5-fluoro-6- ((1-methyl-2-oxo-3- ((2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A flask containing a mixture of 5- [ (6-amino-3-methyl-2-oxo-benzimidazol-1-yl) methyl ] oxazolidin-2-one (600 mg,2.29 mmol), 2, 6-dichloro-5-fluoropyridine-3-carbonitrile (436.94 mg,2.29 mmol) and DIEA (591.34 mg,4.58mmol, 796.96. Mu.L) in DMSO (6 mL) was degassed and purged 3 times with N 2 and then the mixture stirred at 100deg.C under an atmosphere of N 2 for 2hr. To the mixture was added water (30 mL), a precipitate formed, and the precipitate was filtered and washed with H 2 O (20 mL) and ethyl acetate (10 mL). The solid was dried under reduced pressure to give the title compound (700 mg,1.55mmol, yield 68%, purity) as a yellow solid 92%).1H NMR(400MHz,DMSO-d6)δ9.96(s,1H),8.09(d,J=10.27Hz,1H),7.45-7.60(m,2H),7.27(d,J=7.70Hz,1H),7.11(d,J=7.95Hz,1H),4.86(s,1H),3.92-4.12(m,2H),3.56(t,J=7.83Hz,1H),3.30(s,4H).
2- ((3 S, 5R) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((1-methyl-2-oxo-3- (((R) -2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A flask containing a mixture of (R) -2-chloro-5-fluoro-6- ((1-methyl-2-oxo-3- ((2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (500 mg,1.20 mmol), 2- [ (3R, 5S) -4, 4-difluoro-5-methyl-3-piperidinyl ] isoindoline-1, 3-dione (336.22 mg,1.20 mmol) and DIEA (310.08 mg,2.40mmol, 417.90. Mu.L) in DMSO (5 mL) was degassed and purged 3 times with N 2 and then the mixture was stirred under an atmosphere of N 2 at 130℃for 10hr. The mixture was concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: phenomenex C18 x 40mm x 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:25% -45%,8 min) to give the title compound as a white solid (230 mg,344.69 μmol, 29% yield, 99% purity). LCMS: m+h + = 661.3.
2- ((3S, 5R) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((1-methyl-2-oxo-3- (((R) -2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (84)
A flask containing a mixture of 2- ((3S, 5R) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((1-methyl-2-oxo-3- (((R) -2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (230 mg, 348.17. Mu. Mol) in EtOH (15 mL) and MeNH 2 (15 mL, purity 40%) was degassed and purged 3 times with N 2 and then the mixture was stirred under an atmosphere of N 2 at 70℃for 1hr. The mixture was concentrated in vacuo and purified directly by preparative HPLC (column: phenomnex C18.80X 40mm X3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:20% -40%,8 min) to give the title compound as a white solid (40 mg, 74.65. Mu. Mol, 21% yield, purity) 99%).1H NMR(400MHz,DMSO-d6,25℃)δ9.46(s,1H),7.82(d,J=10.85Hz,1H),7.63(d,J=1.31Hz,1H),7.53(s,1H),7.34(d,J=7.03Hz,1H),7.13(d,J=8.34Hz,1H),4.90(d,J=2.38Hz,1H),4.03-4.22(m,3H),3.95-4.02(m,1H),3.59(t,J=8.94Hz,1H),3.35(s,3H),2.90-3.07(m,1H),2.78(q,J=12.12Hz,2H),2.08-2.22(m,1H),1.69(s,2H),0.90(d,J=6.68Hz,3H).
(S) -5- ((3-methyl-6-nitro-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-1-yl) methyl) oxazolidin-2-one
A flask containing a mixture of 3-methyl-6-nitro-1H-benzimidazol-2-one (5 g,25.89 mmol), (5S) -5- (chloromethyl) oxazolidin-2-one (4.21 g,31.06 mmol), K 2CO3 (7.16 g,51.77 mmol) and KI (2.15 g,12.94 mmol) in DMSO (50 mL) was degassed and purged 3 times with N 2 and then the mixture stirred at 80℃under an atmosphere of N 2 for 6hr. To the mixture was added water (40 mL), a precipitate formed, and the precipitate was filtered and washed with H 2 O (15 mL) and ethyl acetate (30 mL). The solid was dried under reduced pressure to give the title compound (4 g,12.87mmol, yield 50%, purity of yellow solid) 94%).1H NMR(400MHz,DMSO-d6)δ8.22(d,J=2.0Hz,1H),8.08(dd,J=8.4,2.0Hz,1H),7.55(s,1H),7.38(d,J=8.8Hz,1H),4.88-4.98(m,1H),4.15-4.34(m,2H),3.59-3.68(m,1H),3.43(s,3H),3.33(dd,J=9.05,6.24Hz,1H).
(S) -5- ((6-amino-3-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-1-yl) methyl) oxazolidin-2-one
To a suspension of Pd/C (300 mg, purity 10%) in DMF (100 mL) was added (S) -5- ((3-methyl-6-nitro-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-1-yl) methyl) oxazolidin-2-one (4 g,13.69 mmol). The mixture was stirred at 50deg.C under H 2 (50 psi) for 20hr. By usingThe mixture was filtered, and the filtrate was concentrated to give a residue, which was purified by silica gel column chromatography (silica gel, ethyl acetate/etoh=1/0 to 1/1) to give the title compound (2 g,6.71mmol, yield 49% purity) as a yellow solid 88%).1H NMR(400MHz,DMSO-d6)δ7.53(s,1H),6.80(br d,J=8.0Hz,1H),6.47(s,1H),6.34(br d,J=8.0Hz,1H),4.65-5.07(m,3H),3.87-4.03(m,2H),3.57(br t,J=8.8Hz,1H),3.28-3.35(m,1H),3.24(s,3H).
(S) -2-chloro-5-fluoro-6- ((1-methyl-2-oxo-3- ((2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A flask charged with a mixture of (S) -5- ((6-amino-3-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-1-yl) methyl) oxazolidin-2-one (1 g,3.81 mmol), 2, 6-dichloro-5-fluoropyridine-3-carbonitrile (728.24 mg,3.81 mmol) and DIEA (985.59 mg,7.63mmol,1.33 mL) in DMSO (6 mL) was degassed and purged 3 times with N 2, and the mixture was stirred at 100deg.C under an atmosphere of N 2 for 2hr. To the mixture was added water (25 mL), a precipitate formed, and the precipitate was filtered and washed with H 2 O (10 mL) and ethyl acetate (15 mL). The solid was dried under reduced pressure to give the title compound (1.1 g,2.51mmol, yield 66%, purity 95%) as a yellow solid. LCMS: m+h + = 417.1.
2- ((3 R, 5S) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((1-methyl-2-oxo-3- (((S) -2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
A flask containing a mixture of (S) -2-chloro-5-fluoro-6- ((1-methyl-2-oxo-3- ((2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (600 mg,1.44 mmol), 2- ((3R, 5S) -4, 4-difluoro-5-methylpiperidin-3-yl) isoindoline-1, 3-dione (403.47 mg,1.44 mmol) and DIEA (372.11 mg,2.88mmol, 501.49. Mu.L) in DMSO (6 mL) was degassed and purged 3 times with N 2, then the mixture was stirred under an atmosphere of N 2 at 130℃for 10hr. The mixture was concentrated in vacuo without further manipulation. The residue was purified by preparative HPLC (column: waters Xbridge BEH C, 100 x 30mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:40% -60%,8 min) to give the title compound (270 mg, 396.46. Mu. Mol, yield 28%, purity) as a white solid 97%).1H NMR(400MHz,DMSO-d6)δ9.62(s,1H),7.87-7.95(m,4H),7.52(s,1H),7.46(d,J=2.0Hz,1H),7.18(dd,J=8.0,2.0Hz,1H),6.94(d,J=8.4Hz,1H),4.81-4.91(m,1H),4.44-4.57(m,1H),4.20-4.39(m,3H),3.87-3.94(m,1H),3.76-3.84(m,1H),3.57(t,J=8.0Hz,1H),3.15-3.30(m,2H),3.05(s,3H),2.18-2.40(m,1H),0.96(d,J=6.8Hz,3H).
2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((1-methyl-2-oxo-3- (((S) -2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (85)
To a solution of 2- ((3 r, 5S) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((1-methyl-2-oxo-3- (((S) -2-oxooxazolid in-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (270 mg, 408.72. Mu. Mol) in EtOH (20 mL) was added MeNH 2/H2 O (31.73 mg, 408.72. Mu. Mol,20mL, purity 40%). The mixture was stirred at 70℃for 1hr. The mixture was concentrated in vacuo and purified directly by preparative HPLC (column: phenomnex C18 75X 30mm X3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:25% -45%,8 min) to give the title compound as a white solid (40 mg, 75.40. Mu. Mol, yield 18%, purity) 100%).1H NMR(400MHz,DMSO-d6)δ9.48(s,1H),7.83(d,J=10.8Hz,1H),7.65(d,J=2.0Hz,1H),7.54(s,1H),7.35(dd,J=8.0,4.0Hz,1H),7.14(d,J=8.0Hz,1H),4.86-4.99(m,1H),4.06-4.19(m,3H),3.99(br d,J=16.0Hz,1H),3.61(t,J=8.0Hz,1H),3.36(s,3H),2.93-3.11(m,1H),2.70-2.88(m,2H),2.07-2.23(m,1H),1.70(br s,2H),0.91(d,J=8.0Hz,3H).
The absolute configuration of compounds 84 and 85 is randomly assigned to the cis conformation based on amino and methyl.
Example 42: synthesis of 5-fluoro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-2-oxo-3- (((R) -2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (86) and 5-fluoro-2- [ (3S, 4S, 5R) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -6- [ [ 1-methyl-2-oxo-3- [ [ (5S) -2-oxooxazolidin-5-yl ] methyl ] benzimidazol-5-yl ] amino ] pyridin-3-carbonitrile (87)
2-Chloro-5-fluoro-6- ((1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a solution of 6-amino-3-methyl-1H-benzimidazol-2-one (1 g,6.13 mmol) and 2, 6-dichloro-5-fluoropyridine-3-carbonitrile (1.17 g,6.13 mol) in DMSO (10 mL) was added DIPEA (1.58 g,12.26mmol,2.13 mL). The mixture was stirred at 100deg.C for 2hr. The mixture was cooled to 15 ℃ and water (10 mL) was added to form a precipitate, which was filtered and the filter cake was washed with EtOAc (50 mL). The filter cake was dried in vacuo to give the title compound as a yellow solid (1.8 g,5.10mmol, yield 83%, purity 90%).1H NMR(400MHz,DMSO-d6)δ10.89(s,1H),9.86(s,1H),8.12(d,J=,10.8Hz,1H),7.38(d,J=1.6Hz,1H),7.26(dd,J=8.4,1.6Hz,1H),7.06(d,J=8.4Hz,1H),2.54(s,3H).
5-Fluoro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a solution of 2-chloro-5-fluoro-6- [ (1-methyl-2-oxo-3H-benzimidazol-5-yl) amino ] pyridine-3-carbonitrile (1 g,3.15 mmol) and (3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidine (1.54 g,6.30mmol, TFA) in DMSO (10 mL) was added DIPEA (2.03 g,15.74mmol,2.74 mL). The mixture was stirred at 100deg.C for 12hr. To the mixture was added water (5 mL) to form a precipitate, the precipitate was filtered, and the filter cake was washed with water (10 mL) and EtOAc (20 mL), and then the filter cake was dried in vacuo to give the title compound (1 g,2.34mmol, yield 74%, purity as a yellow solid 96%).1H NMR(400MHz,DMSO-d6)δ10.85(s,1H),9.31(s,1H),7.73(d,J=10.4Hz,1H),7.32(s,2H),7.01(d,J=7.6Hz,1H),4.42-4.65(m,1H),3.90(d,J=11.6Hz,2H),3.26(s,3H),2.82(t,J=12Hz,2H),1.73-1.97(m,2H),0.91(d,J=5.2Hz,6H).
5-Fluoro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-2-oxo-3- (((R) -2-oxooxazolidin-5-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (86)
To a mixture of 5-fluoro-2- [ (3S, 4S, 5R) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -6- [ (1-methyl-2-oxo-3H-benzimidazol-5-yl) amino ] pyridine-3-carbonitrile (100 mg, 242.46. Mu. Mol) and (R) -5- (chloromethyl) oxazolidin-2-one (49.30 mg, 363.69. Mu. Mol) in DMSO (1 mL) was added K 2CO3 (67.02 mg, 484.92. Mu. Mol) and KI (20.12 mg, 121.23. Mu. Mol), and the reaction mixture was stirred at 100℃for 12hr. The reaction mixture was filtered and the filtrate was then purified by preparative HPLC (column: waters Xbridge Prep OBD C, 150 x 40mm x10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:35% -55%,8 min) to give the title compound (40 mg,77.96 μmol, 32% yield, purity) as a white solid 99%).1H NMR(400MHz,DMSO-d6)δ9.40(s,1H),7.77(d,J=10.8Hz,1H),7.59-7.51(m,2H),7.38(dd,J=1.6,8.4Hz,1H),7.11(d,J=8.4Hz,1H),4.96-4.83(m,1H),4.61-4.40(m,1H),4.12-4.04(m,1H),4.02-3.95(m,1H),3.87(dd,J=4.0,12.8Hz,2H),3.59(t,J=8.8Hz,1H),3.34(s,3H),3.31-3.29(m,1H),2.85-2.74(m,2H),2.00-1.75(m,2H),0.89(d,J=6.8Hz,6H).
5-Fluoro-2- [ (3S, 4S, 5R) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -6- [ [ 1-methyl-2-oxo-3- [ [ (5S) -2-oxooxazolid in-5-yl ] methyl ] benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (87)
To a solution of 5-fluoro-2- [ (3S, 4S, 5R) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -6- [ (1-methyl-2-oxo-3H-benzimidazol-5-yl) amino ] pyridine-3-carbonitrile (150 mg, 363.69. Mu. Mol) in DMSO (1.5 mL) was added K 2CO3 (100.53 mg, 727.39. Mu. Mol,2 eq), KI (30.19 mg, 181.85. Mu. Mol) and (S) -5- (chloromethyl) oxazolidin-2-one (49.30 mg, 363.69. Mu. Mol). The mixture was stirred at 60℃for 2hr. The reaction mixture was filtered and the filtrate was purified by preparative HPLC (column: waters Xbridge Prep OBD C, 150 x 40mm x 10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:35% -50%,8 min) to give the title compound (30 mg,58.65 μmol, yield) as a white solid 48%).1H NMR(400MHz,DMSO-d6)δ9.40(s,1H),7.77(d,J=10.97Hz,1H),7.50-7.61(m,2H),7.38(dd,J=8.46,1.79Hz,1H),7.11(d,J=8.46Hz,1H).4.82-4.97(m,1H),4.42-4.62(m,1H),4.03-4.15(m,1H),3.96-4.02(m,1H),3.87(br dd,J=12.99,4.05Hz,2H),3.59(t,J=8.82Hz,1H),3.34(s,3H),3.31-3.29(m,1H),2.74-2.85(m,2H),2.00-1.75(m,2H),0.89(d,J=6.91Hz,6H).
The absolute configuration of compounds 86 and 87 is randomly assigned to the cis-conformation based on all substituents of the piperidine ring.
Example 43: synthesis of 5-fluoro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-2-oxo-3- (((R) -2-oxooxazolidin-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (88) and 5-fluoro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-2-oxo-3- (((S) -2-oxooxazolidin-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (89)
5-Fluoro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-2-oxo-3- (((R) -2-oxooxazolidin-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (88)
A flask containing a mixture of 5-fluoro-2- [ (3S, 5R) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -6- [ (1-methyl-2-oxo-3H-benzimidazol-5-yl) amino ] pyridine-3-carbonitrile (100 mg, 242.46. Mu. Mol), [ (4S) -2-oxooxazolid-4-yl ] methyl-4-methylbenzenesulfonate (98.67 mg, 363.69. Mu. Mol), K 2CO3 (67.02 mg, 484.92. Mu. Mol) and KI (20.12 mg, 121.23. Mu. Mol) in DMSO (2 mL) was degassed and purged 3 times with N 2, and then the mixture was stirred at 80℃under an atmosphere of N 2 for 6hr. The reaction mixture was filtered and the filtrate was purified by preparative HPLC (column: phenomnex C18:30 mm 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:35% -55%,8 min) to give the title compound (37 mg, 72.33. Mu. Mol, yield 30%, purity) as a white solid 100%).1H NMR(400MHz,DMSO-d6)δ9.38(s,1H),7.83(s,1H),7.77(d,J=10.88Hz,1H),7.49(d,J=1.34Hz,1H),7.34(dd,J=8.38,1.65Hz,1H),7.10(d,J=8.44Hz,1H),4.42-4.59(m,1H),4.33-4.40(m,1H),4.18(q,J=4.69Hz,2H),3.80-3.95(m,4H),3.33(s,3H),2.78(br t,J=12.41Hz,2H),1.77-1.98(m,2H),0.88(dd,J=6.85,3.42Hz,6H).
5-Fluoro-2- ((3S, 4S, 5R) -4-fluoro-3, 5-dimethylpiperidin-1-yl) -6- ((1-methyl-2-oxo-3- (((S) -2-oxooxazolidin-4-yl) methyl) -2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (89)
A flask containing a mixture of 5-fluoro-2- [ (3S, 5R) -4-fluoro-3, 5-dimethyl-1-piperidinyl ] -6- [ (1-methyl-2-oxo-3H-benzimidazol-5-yl) amino ] pyridine-3-carbonitrile (100 mg, 242.46. Mu. Mol), [ (4R) -2-oxooxazolid-4-yl ] methyl-4-methylbenzenesulfonate (98.67 mg, 363.69. Mu. Mol), K 2CO3 (67.02 mg, 484.93. Mu. Mol) and KI (20.12 mg, 121.23. Mu. Mol) in DMSO (2 mL) was degassed and purged 3 times with N 2, and then the mixture was stirred at 80℃under an atmosphere of N 2 for 6hr. The reaction mixture was filtered and the filtrate was purified by preparative HPLC (column: phenomnex C18:30 mm 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:35% -60%,8 min) to give the title compound (25 mg, 48.87. Mu. Mol, 20% yield, purity) as a white solid 100%).1H NMR(400MHz,DMSO-d6)δ9.39(s,1H),7.84(s,1H),7.78(d,J=10.88Hz,1H),7.49(d,J=1.71Hz,1H),7.35(dd,J=8.44,1.71Hz,1H),7.11(d,J=8.44Hz,1H),4.44-4.59(m,1H),4.34-4.41(m,1H),4.16-4.21(m,2H),3.80-3.95(m,4H),3.34(s,3H),2.74-2.83(m,2H),1.79-1.97(m,2H),0.89(dd,J=6.85,3.42Hz,6H).
The absolute configuration of compounds 88 and 89 is randomly assigned to the cis-conformation based on all substituents of the piperidine ring.
Example 44: synthesis of 2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1- ((S) -2-hydroxypropyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (90) and 2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1- ((S) -2-hydroxypropyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (91)
(S) -1- ((2-amino-4-nitrophenyl) amino) propan-2-ol
A mixture of 2-fluoro-5-nitroaniline (6 g,38.43 mmol) and (S) -1-aminopropan-2-ol (11.55 g,153.73mmol,12.10 mL) in NMP (120 mL) was stirred at 100deg.C for 12hr. The reaction mixture (mixed with another batch of the same scale) was treated with H 2 O (300 mL) to form a precipitate, which was filtered and the solid was washed with solvent (petroleum ether/ethyl acetate=2:1, 500 mL). The filter cake was collected and dried in vacuo to give the title compound as a yellow solid (15 g,65.34mmol, 85% yield, purity 92%).1H NMR(400MHz,DMSO-d6)δ7.51(d,J=8.8Hz,1H),7.42(d,J=2.0Hz,1H),6.50(d,J=8.8Hz,1H),5.90(s,1H),5.17(s,2H),4.86(s,1H),3.81-3.93(m,1H),3.05-3.21(m,2H),1.13(d,J=6.0Hz,3H).LCMSL:[M+H]+=212.3.
(S) -1- (2-hydroxypropyl) -5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one
To a solution of (S) -1- ((2-amino-4-nitrophenyl) amino) propan-2-ol (15 g,71.02 mmol) in MeCN (150 mL) and NMP (15 mL) was added DSC (19.10 g,74.57 mmol). The mixture was stirred at 15℃for 12hr. The reaction mixture was filtered to give a filter cake which was washed with solvent (petroleum ether/ethyl acetate=1:1, 80 mL), collected and dried in vacuo to give the title compound (9 g,37.18mmol, 52% yield, purity) as a yellow solid 98%).1H NMR(400MHz,DMSO-d6)δ11.40(s,1H),7.99(dd,J=8.8,2.0Hz,1H),7.74(d,J=2.0Hz,1H),7.35(d,J=8.8Hz,1H),4.91(d,J=4.8Hz,1H),3.96(m,1H),3.71-3.79(m,2H),1.10(d,J=6.0Hz,3H).
(S) -3- (3-hydroxy-3-methylbutyl) -1- (2-hydroxypropyl) -5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one
To a solution of (S) -1- (2-hydroxypropyl) -5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one (4 g,16.86 mmol) and 3-hydroxy-3-methylbutyl 4-methylbenzenesulfonate (6.97 g,26.98 mmol) in DMF (50 mL) was added Cs 2CO3 (10.99 g,33.73 mmol). The mixture was stirred at 120deg.C for 2hr. Water (80 mL) was then added to the mixture (combined with another batch at 1g scale) and extracted with EtOAc (100 mL. Times.3). The combined organic phases were washed with brine (150 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo to give the title compound as a yellow oil (6.5 g, 91% purity). LCMSL: [ m+h ] + =324.3.
(S) -5-amino-3- (3-hydroxy-3-methylbutyl) -1- (2-hydroxypropyl) -1H-benzo [ d ] imidazol-2 (3H) -one
Pd/C (2 g, purity 10%) was added to a solution of (S) -3- (3-hydroxy-3-methylbutyl) -1- (2-hydroxypropyl) -5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one (6.5 g,20.10 mmol) in DMF (100 mL) under Ar. The flask was degassed under vacuum and purged three times with H 2. The mixture was stirred at H 2 (50 psi) and 50deg.C for 12hr. The reaction mixture was filtered, and the filtrate was concentrated in vacuo to give the title compound as a yellow oil (5 g,15.00mmol, 75% yield, 88% purity). LCMSL: [ m+h ] + =294.1.
(S) -2-chloro-5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1- (2-hydroxypropyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a solution of (S) -5-amino-3- (3-hydroxy-3-methylbutyl) -1- (2-hydroxypropyl) -1H-benzo [ d ] imidazol-2 (3H) -one (1 g,3.41 mmol) and 2, 6-dichloro-5-fluoronicotinonitrile (651.04 mg,3.41 mmol) in DMF (10 mL) was added DIEA (881.12 mg,6.82mmol,1.19 mL). The mixture was stirred at 100deg.C for 1hr. The reaction mixture was treated with H 2 O (50 mL) to form a precipitate, which was filtered and the solid was washed with solvent (petroleum ether/ethyl acetate=2:1, 50 mL). The filter cake was collected and dried in vacuo to give the title compound as a yellow solid (1.3 g,2.67mmol, 78% yield, 92% purity). LCMSL: m+h + = 448.1.
2- ((3 R, 5S) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1- ((S) -2-hydroxypropyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a solution of (S) -2-chloro-5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1- (2-hydroxypropyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (300 mg, 669.81. Mu. Mol) and 2- ((3R, 5S) -4, 4-difluoro-5-methylpiperidin-3-yl) isoindoline-1, 3-dione (187.73 mg, 669.81. Mu. Mol) in DMSO (3 mL) was added DIEA (432.84 mg,3.35mmol, 583.34. Mu. L). The mixture was stirred at 130℃for 10hr. The mixture (combined with another batch at 50mg scale) was directly purified without further manipulation. The residue was purified by preparative HPLC (column: waters Xbridge Prep OBD C18:150:40 mm:10 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:35% -65%,8 min) to give the title compound (90 mg,123.61 μmol, 18% yield, 95% purity) as a yellow solid. LCMS: m+h + = 692.3.
2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1- ((S) -2-hydroxypropyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (90)
To a solution of 2- ((3 r, 5S) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1- ((S) -2-hydroxypropyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (70 mg, 101.20. Mu. Mol) in EtOH (3.5 mL) was added MeNH 2 (7.86 mg, 101.20. Mu. Mol,3.5mL, purity 40%) under N 2 (40% aqueous solution). The mixture was stirred at 70℃for 1hr. The mixture (combined with another batch on a 20mg scale) was concentrated in vacuo. The residue was purified by preparative HPLC (column: phenomenex C18 x 30mm x 3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:35% -65%,8 min) to give the title compound (20 mg,35.35 μmol, yield 35%, purity) as a white solid 99%).1H NMR(400MHz,DMSO-d6)δ9.44(s,1H),7.81(d,J=10.85Hz,1H),7.40(d,J=1.91Hz,1H),7.34(dd,J=8.46,1.91Hz,1H),7.15(d,J=8.46Hz,1H),4.86(d,J=5.01Hz,1H),4.51(s,1H),4.11(d,J=12.16Hz,1H),3.93-4.01(m,2H),3.89(dd,J=10.55,6.62Hz,2H),3.71(dd,J=5.84,2.03Hz,2H),2.88-3.02(m,1H),2.69-2.87(m,2H),2.03-2.19(m,1H),1.70(t,J=8.05Hz,4H),1.16(d,J=2.62Hz,6H),1.06(d,J=6.20Hz,3H),0.90(d,J=6.68Hz,3H).
(R) -1- ((2-amino-4-nitrophenyl) amino) propan-2-ol
A mixture of 2-fluoro-5-nitroaniline (10 g,64.06 mmol) and (R) -1-aminopropan-2-ol (19.25 g,256.24mmol,20.17 mL) in NMP (100 mL) was stirred at 100deg.C for 12hr. The reaction mixture was treated with H 2 O (300 mL) to form a precipitate, the precipitate was filtered and the solid was washed with solvent (EtOAc: pe=1:2,500 mL), the filter cake was collected and dried in vacuo to give the title compound (12 g,55.68mmol, 87% yield, 98% purity) as a yellow solid. LCMS: m+h + =212.3.
(R) -1- (2-hydroxypropyl) -5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one
To a solution of (R) -1- (2-amino-4-nitro-anilino) propan-2-ol (10 g,47.34 mmol) in ACN (100 mL) was added DSC (12.13 g,47.34 mmol) followed by NMP (10 mL). The reaction mixture was stirred at 20℃for 12hr. The reaction mixture was filtered and the solid was then washed with PE: etoac=1:1, 80mL, collected and dried in vacuo to give the title compound as a yellow solid (9.3 g,38.81mmol, 82% yield, purity 99%).1H NMR(400MHz,DMSO-d6)δ11.40(s,1H),7.99(dd,J=2.4,8Hz,1H),7.74(d,J=2.4Hz,1H),7.35(d,J=8.8Hz,1H),4.91(d,J=4.8Hz,1H),4.02-3.91(m,1H),3.81-3.69(m,2H),1.10(d,J=6.4Hz,3H).
(R) -3- (3-hydroxy-3-methylbutyl) -1- (2-hydroxypropyl) -5-nitro-1H-benzo [ d ] imidazol-2 (3H) -one
To a mixture of 3- [ (R) -2-hydroxypropyl ] -6-nitro-1H-benzimidazol-2-one (4.3 g,18.13 mmol) and (3-hydroxy-3-methyl-butyl) 4-methylbenzenesulfonate (7.49 g,29.00 mmol) in DMF (50 mL) was added Cs 2CO3 (11.81 g,36.25 mmol) and stirred at 120℃for 2hr. The reaction mixture was treated with H 2 O (80 mL) and extracted with EtOAc (100 mL x 3), then the combined organic phases were washed with brine (150 mL), dried over anhydrous Na 2SO4, filtered and concentrated in vacuo to give the title compound (6.5 g, crude) as a yellow oil ).1H NMR(400MHz,CDCl3)δ8.06(dd,J=2.0,8.8Hz,1H),7.92(d,J=2.0Hz,1H),7.19(d,J=8.8Hz,1H),4.26-4.17(m,1H),4.14-4.06(m,2H),4.02-3.93(m,1H),3.91-3.82(m,1H),2.51(s,1H),1.91(t,J=7.6Hz,2H),1.48(s,1H),1.30(s,6H),1.28(d,J=6.0Hz,3H).
(R) -5-amino-3- (3-hydroxy-3-methylbutyl) -1- (2-hydroxypropyl) -1H-benzo [ d ] imidazol-2 (3H) -one
To a solution of 3- (3-hydroxy-3-methyl-butyl) -1- [ (R) -2-hydroxypropyl ] -5-nitrobenzimidazol-2-one (6.5 g,20.10 mmol) in DMF (100 mL) was added Pd/C (3 g,21.65mmol, purity 10%) under Ar. The flask was degassed under vacuum and purged three times with H 2. The mixture was stirred at H 2 (50 psi) and 50℃for 12 hours. The reaction mixture was filtered and the filtrate concentrated in vacuo and purified by column chromatography (SiO 2, etOAc/etoh=1/0 to 3/1) to give the title compound as a yellow solid (3.6 g,11.54mmol, 57% yield, 94% purity). LCMS: [ m+h ] + =294.1.
(R) -2-chloro-5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1- (2-hydroxypropyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a mixture of 5-amino-3- (3-hydroxy-3-methyl-butyl) -1- [ (R) -2-hydroxypropyl ] benzimidazol-2-one (1 g,3.41 mmol) and 2, 6-dichloro-5-fluoropyridine-3-carbonitrile (651.04 mg,3.41 mmol) in DMF (10 mL) was added DIPEA (881.12 mg,6.82mmol,1.19 mL) and the reaction mixture was stirred at 100deg.C for 1hr. The reaction mixture was treated with H 2 O (20 mL) to form a precipitate, which was filtered and the solid was washed with PE: etoac=2:1, 40 mL. The solid was dried in vacuo to give the title compound as a reddish brown solid (1.3 g,2.61mmol, 77% yield, 90% purity). LCMS: m+h + = 448.2.
2- ((3R, 5 s) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1- ((R) -2-hydroxypropyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile
To a mixture of 2-chloro-5-fluoro-6- [ [3- (3-hydroxy-3-methyl-butyl) -1- [ (R) -2-hydroxypropyl ] -2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (319.61 mg, 713.60. Mu. Mol) and 2- [ (3R, 5 s) -4, 4-difluoro-5-methyl-3-piperidinyl ] isoindoline-1, 3-dione (200 mg, 713.60. Mu. Mol) in DMSO (3 mL) was added DIPEA (184.46 mg,1.43mmol, 248.59. Mu.l), and the reaction mixture was stirred at 130 ℃ for 10hr. The reaction mixture was filtered (combined with one batch at 100mg scale and another at 200mg scale) and purified by preparative HPLC (column: C18 (250 x 50mm x 10 μm), mobile phase: [ water (NH 4HCO3) -ACN ];: B%:40% -65%,10 min) to give the title compound as a yellow solid (250 mg, 90% purity). LCMS: m+h + = 692.3.
2- ((3R, 5S) -3-amino-4, 4-difluoro-5-methylpiperidin-1-yl) -5-fluoro-6- ((3- (3-hydroxy-3-methylbutyl) -1- ((R) -2-hydroxypropyl) -2-oxo-2, 3-dihydro-1H-benzo [ d ] imidazol-5-yl) amino) nicotinonitrile (91)
To a solution of 2- [ (3R, 5 s) -3- (1, 3-dioxoisoindolin-2-yl) -4, 4-difluoro-5-methyl-1-piperidinyl ] -5-fluoro-6- [ [3- (3-hydroxy-3-methyl-butyl) -1- [ (R) -2-hydroxypropyl ] -2-oxo-benzimidazol-5-yl ] amino ] pyridine-3-carbonitrile (100 mg,144.57 μmol) in EtOH (7.5 mL) was added MeNH 2 (11.22 mg,144.57 μmol,7.5mL, purity 40%) and stirred at 70 ℃ for 1hr. The reaction mixture was concentrated in vacuo and then purified by preparative HPLC (column: phenomnex C18X 30mm X3 μm; mobile phase: [ water (NH 4HCO3) -ACN ]; B%:15% -60%,8 min) to give the title compound (30 mg, 53.42. Mu. Mol, 37% yield, purity) as a white solid 100%).1H NMR(400MHz,DMSO-d6)δ9.44(s,1H),7.81(d,J=10.8Hz,1H),7.40(d,J=2.0Hz,1H),7.33(dd,J=1.6,8.8Hz,1H),7.15(d,J=8.4Hz,1H),4.86(d,J=5.2Hz,1H),4.52(s,1H),4.12(d,J=12.0Hz,1H),4.03-3.85(m,4H),3.75-3.66(m,2H),3.00-2.87(m,1H),2.87-2.79(m,1H),2.74(t,J=12.4Hz,1H),2.22-2.02(m,1H),1.80-1.62(m,4H),1.16(d,J=2.0Hz,6H),1.06(d,J=6.4Hz,3H),0.89(d,J=6.4Hz,3H).
The absolute configuration of compounds 90 and 91 is randomly assigned to the cis conformation based on amino and methyl.
Example 45: degradation Activity
TABLE 1 degradation Activity of Compounds
/>
HiBiT scheme
The DC 50 (concentration to 50% degradation) values were determined according to the cell degradation assay (HiBiT, promega TM) in Su-DHL-4 cells (Table 1). Endogenous BCL6 was labeled with 11-amino acid SmBiT by CRISPR/Cas9 gene editing and single cell clonal selection. After 24 hours of treatment with the compound, the cells were lysed and incubated with LgBiT protein to reconstitute the complete nanoluciferase. The substrate is then added and the relative luciferase units measured. The degradation level of each treatment is expressed as a percentage of 100% DMSO (Prism) relative to the control.
Example 46: synergistic effects of BCL6 degradation and EZH2 inhibition
Zeste homolog enhancer 2 (EZH 2), a member of the multicomb repression complex 2 (PRC 2), is a marker associated with gene repression, PRC2 methylates histone 3 lysine 27. EZH2 expression is upregulated in both the normal germinal center B cell development and Germinal Center (GC) subtype of B cell lymphomas, and (Caganova et al.,J.Clin.Invest.123(12):5009-5022(2013);Beguelin et al.,Cancer Cell 23(5):677-692(2013))., very similar to BCL6, describes the cooperation of BCL6 and PRC2 in negative regulation of transcription in both normal B cell development and lymphogenesis (Beguelin et al., CANCER CELL (2): 197-213 (2016)). Su-DHL-4 cells were treated (ranging from 10. Mu.M to 5 nM) with different concentrations of either tazistat (Taz) or rimet tostat (Lira). After 48 hours, cells were treated with different concentrations of compound 1 (ranging from 50nM to 2 nM). Five days after treatment with Compound 1, through CellTiter-The assay (Promega TM) monitors cell proliferation. The antiproliferative effects of Taz (fig. 1A) and Lira treatments (fig. 1B) are plotted. The excess over Bliss (eob) scores were calculated over at least three concentrations (fig. 1C-1D) and significant synergy was observed over most concentration ranges (eob >1 with an average synergy score exceeding 10, synergy finder (IANEVSKI ET al., bioenformatics 33 (15): 2413-2415 (2017)). Since anti-proliferative effects were observed in single compound experiments only at BCL6 degradation agent treated DC 99, dose reduction at 60% growth inhibition (GI 60) was calculated by addition of two EZH2 inhibitors BCL6 degradation agents for Taz and Lira treatments reduced BCL6 degradation agent dose requirements to GI 60 by about 4.3 fold at 480nM (E). These results indicate that EZH2 inhibition exerts synergistic anti-proliferative effects with BCL6 degradation agents.
Example 47: proliferation protocol
To ensure that the decrease in HiBiT signal was due to BCL6 degradation rather than dying cells, cellTiter-(CTG, promega TM). To investigate the antiproliferative effect of BCL6 degrading agents and their synergistic effect with EZH2 inhibitors, su-DHL-4 and other DLBCL cells were treated with DC 99、DC75、DC50 or 1 μm compounds, followed by 5 days of culture, at which point CTG values and/or Caspase3/7 activity were measured as percent DMSO treatment.
All patent publications and non-patent publications are indicative of the level of skill of those skilled in the art to which this invention pertains. All publications cited herein are incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference.
Although the invention herein has been described with reference to particular embodiments, it is to be understood that these embodiments are merely illustrative of the principles and applications of the present invention. It is therefore to be understood that numerous modifications may be made to the illustrative embodiments and that other arrangements may be devised without departing from the spirit and scope of the present invention as defined by the appended claims.

Claims (72)

1. A compound having a structure represented by formula I:
Or a pharmaceutically acceptable salt or stereoisomer thereof,
Wherein:
X 1 is N, CH, CCl, CF or CCN;
Each X 2 is independently CH 2, S, CHF, CHCl, CHOH or CF 2;
R 1 is hydrogen, =o, -CN, -c≡ch, -OH, -SH, -NH 2, -COOH, halo, (C 1-C6) alkyl, -O- (C 1-C6) alkyl, (C 1-C6) haloalkyl, amido, carboxyl, carbamoyl, sulfamoyl, phenyl, 5-to 8-membered heterocyclyl, -NR 7R8、–C(O)R9、–C(O)NR10R11 or L 1Y1, wherein the alkyl, phenyl or heterocyclyl is optionally substituted with one or more groups selected from: halo, -COOH, -OH, -NH 2、(C1-C6) alkyl, -C (O) O- (C 1-C6) alkyl, -C (O) N (C 1-C6 alkyl) 2、–O–(C1-C6) alkyl, -N (C 1-C3 alkyl) 2, phenyl and 4-to 6-membered heterocyclyl optionally substituted with one or more groups selected from halo and (C 1-C6) alkyl;
r 7 is hydrogen, (C 1-C4) alkyl or (C 3-C6) cycloalkyl;
R 8 is hydrogen, (C 1-C4) alkyl, (C 1-C4) haloalkyl, (C 3-C6) cycloalkyl or six membered heterocyclyl;
R 9 is- (C 1-C3) alkyl-N (C 1-C3 alkyl) 2、(C3-C6) cycloalkyl or a 5-to 6-membered heterocyclyl, wherein the heterocyclyl is optionally substituted with (C 1-C3) alkyl;
R 10 is hydrogen, (C 1-C3) alkyl or (C 3-C6) cycloalkyl;
R 11 is optionally substituted by-NH 2、–O-(C1-C6) alkyl-O- (C 1-C6) alkyl-NH 2 or-O- (C 1-C6) alkyl-O- (C 1-C6) alkyl-NH 2 substituted (C 3-C6) cycloalkyl or (C 1-C6) alkyl;
L 1 is absent, (C 1-C6) alkylene or (C 3-C7) carbocyclyl; wherein the alkylene or carbocyclyl is further optionally substituted with one or more R A groups, which are the same or different;
each R A is independently oxo, alkyl, alkenyl, alkynyl, halo, haloalkyl, carbocyclyl, heterocyclyl, hydroxy, alkoxy, cycloalkoxy, heterocycloalkoxy, haloalkoxy, aryloxy, heteroaryloxy, aralkyloxy, alkenyloxy, alkynyloxy, amino, alkylamino, cycloalkylamino, heterocycloalkylamino, arylamino, heteroarylamino, aralkylamino, N-alkyl-N-arylamino, N-alkyl-N-heteroarylamino, N-alkyl-N-aralkylamino, hydroxyalkyl, aminoalkyl, alkylthio, haloalkylthio, alkylsulfonyl, haloalkylsulfonyl, cycloalkylsulfonyl, heterocycloalkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, alkylaminosulfonyl, cycloalkylaminosulfonyl, heteroarylsulfonyl, heterocyclylalkylaminosulfonyl, arylaminoculfonyl, heteroarylaminosulfonyl, N-alkyl-N-arylaminoculfonyl, N-alkyl-N-heteroarylaminosulfonyl, formyl, alkylcarbonyl, haloalkylcarbonyl, alkenylcarbonyl, alkynylcarbonyl, carboxy, alkoxycarbonyl, alkylcarbonyloxy, amino, alkylsulfonylamino, haloalkylsulfonylamino, cycloalkylsulfonylamino, heterocyclylalkylsulfonylamino, arylsulfonylamino, heteroarylsulfonylamino, aralkylsulfonylamino, alkylcarbonylamino, haloalkylcarbonylamino, cycloalkylcarbonylamino, heterocyclylcarbonylamino, arylcarbonylamino, heteroarylcarbonylamino, aralkylsulfonylamino, aminocarbonyl, alkylaminocarbonyl, cycloalkylaminocarbonyl, heterocyclylalkylaminocarbonyl, arylaminocarbonyl, heteroarylaminocarbonyl, N-alkyl-N-arylaminocarbonyl, N-alkyl-N-heteroarylaminocarbonyl, cyano, nitro, azido or phosphino;
Y 1 is-CN, -OH, halo, (C 1-C4) alkoxy, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, 4-to 7-membered heterocyclyl, (C 3-C6) carbocyclyl, -NR 7'R8'、–C(O)R9、–C(O)NR10R11; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more of the same or different groups selected from (C 1-C4) alkyl, halo, (C 1-C4) haloalkyl, -CN, -OH and-NH 2;
R 7' and R 8' are each independently hydrogen, (C 1-C6) alkyl, (C 3-C7) carbocyclyl, 4-to 7-membered heterocyclyl, (C 6-C10) aryl, or mono-or bi-cyclic 5-to 10-membered heteroaryl; wherein the alkyl, carboxyl, heterocyclyl, aryl or heteroaryl groups are further optionally substituted with one or more identical or different R A groups, or
R 7' and R 8' form, with the nitrogen atom to which they are attached, a 3-to 7-membered heterocyclyl, wherein the heterocyclyl is further optionally substituted with one or more identical or different R A groups, or L 1 is (C 2-C4) alkylene which combines with R 7' to form a 4-to 6-membered heterocyclyl;
R 1' is absent, hydrogen, -CN, -C≡CH, -OH, -SH, -NH 2, -COOH, halo, (C 1-C6) alkyl, -O- (C 1-C6) alkyl, (C 1-C6) haloalkyl, amido, carboxyl, carbamoyl, sulfamoyl, phenyl, 5-to 8-membered heterocyclyl, -NR 7R8、-C(O)R9 or-C (O) NR 10R11; wherein the alkyl, phenyl or heterocyclyl is further optionally substituted with one or more identical or different R A groups, or
R 1' and L 1 together with the same carbon atom to which they are attached form a spiro (C 3-C7) carbocyclyl or a 4-to 7-membered heterocyclyl; wherein the carbocyclyl or heterocyclyl is further optionally substituted with one or more R A groups, which may be the same or different;
is/>
X 3 and X 4 are independently CR 12 or N;
x 5 is CH or N;
R 12 is hydrogen, (C 1-C4) alkyl, halo, hydroxy, amino, (C 1-C4) alkoxy, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, (C 2-C4) alkenyl, (C 2-C4) alkynyl, nitro, cyano, NH (C 1-C4) alkyl or N (C 1-C4) alkyl) 2;
R 2 is hydrogen, (C 1-C6) alkyl, (C 3-C6) carbocyclyl, 4-to 7-membered heterocyclyl, (C 3-C7) carbocyclyl (C 1-C6) alkyl, or 4-to 7-membered heterocyclyl (C 1-C6) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups, wherein R 13 is (C 1-C6) alkyl, (C 1-C6) alkoxy, halo, amino, hydroxy, haloalkyl, NH (C 1-C6) alkyl or N ((C 1-C6) alkyl) 2、(C3-C6) carbocyclyl or 4-to 7-membered heterocyclyl, or R 2 is-L 2-Y2 -Z;
L 2 is (C 1-C5) alkylene which is absent or optionally substituted by one or more substituents selected from (C 1-C2) alkyl and oxo;
Y 2 is absent 、O、S、S(O)、S(O)2、NR'、C(O)、C(O)O、OC(O)、C(O)N(R')、N(R')C(O)、N(R')C(O)N(R')、N(R')C(O)O、OC(O)N(R')、S(O)2N(R') or N (R') S (O) 2;
Each R' is independently hydrogen or (C 1-C4) alkyl;
Z is hydrogen, (C 1-C6) alkyl, (C 2-C6) alkenyl, (C 2-C6) alkynyl, (C 3-C10) carbocyclyl or 3-to 10-membered heterocyclyl; wherein Z is optionally substituted with one or more substituents independently selected from (C 1-C4) alkyl, halo, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, amino, (C 1-C4) aminoalkyl, cyano, hydroxy, carboxy, carbamoyl, sulfamoyl, mercapto, ureido 、NRrRs、ORr、C(O)Rr、C(O)ORr、OC(O)Rr、C(O)NRrRs、N(Rr)C(O)Rr、S(O)0-2Rr、S(O)2NRrRs、N(Rr)SO2Rr、Si(Rr)(Rs)Rt, and (CH 2)1-3NRrRs), wherein R r、Rs and R t are each independently hydrogen, (C 1-C6) alkyl, or (C 3-C6) cycloalkyl, or R r and R s together with the nitrogen atom to which they are attached form a 4-to 9-membered heterocyclyl optionally substituted with one or more substituents selected from (C 1-C4) alkyl, halo, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, (C 1-C4) alkoxy, (C 1-C4) alkylamino, amino, cyano, and hydroxy;
R 3 is-L 3CR14R15R16 or-ch=ch-R 16;
L 3 is absent, O, S, (C 1-C4) alkylene, -O- (C 1-C4) alkylene or-S- (C 1-C4) alkylene;
R 14 is hydrogen or (C 1-C4) alkyl;
R 15 is hydrogen or (C 1-C4) alkyl, or
R 14 and R 15 together with the carbon atom to which they are attached form (C 3-C5) carbocyclyl, 4-to 7-membered heterocyclyl or c=o;
R 16 is (C 1-C6) alkyl 、–NR17R18、–OR17、–C(O)R17、–C(O)OR17、–N(R18)C(O)R17、–C(O)NR17R18、–S(O)–(C1-C6) alkyl, -S (O) 2–(C1-C6 alkyl, -P (O) - (C 1-C6 alkyl) 2、–C(NH)NH2、–(C1-C4) alkyl-NR 18C(O)R17 or 4-to 7-membered heterocyclyl;
R 17 is hydrogen, 3-to 6-membered heterocyclyl or (C 1-C4) alkyl optionally substituted with one or more identical or different groups selected from OH, cl, F, CF 3、N(C1-C4 alkyl) 2、(C3-C6) carbocyclyl, 3-to 6-membered heterocyclyl, (C 2-C4) alkenyl and (C 2-C4) alkynyl;
R 18 is hydrogen or (C 1-C4) alkyl;
R 4 is hydrogen, methyl, - (CH 2)1-3W1W2) or
W 1 is CR 19R19' or C (O);
R 19 and R 19' are independently hydrogen, (C 1-C2) alkyl, fluoro, hydroxy, cyano, nitro, (C 1-C2) alkoxy, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, amino, NH (C 1-C2) alkyl or N (C 1-C2) alkyl) 2, or
R 19 and R 19' together with the carbon atom to which they are attached form a C (O), (C 3-C6) carbocyclyl or a 3-to 6-membered heterocyclyl optionally substituted with one or more substituents independently selected from (C 1-C2) alkyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, (C 1-C2) alkylamino, amino, cyano and hydroxy;
W 2 is cyano, hydroxy, 5-or 6-membered heteroaryl, phenyl, C (O) - (C 1-C2) alkyl, S (O) 2-(C1-C2) alkyl, C (O) OCH 3、C(O)NHCH3、CR20R21R22, amino, NH (C 1-C2) alkyl or N (C 1-C2 alkyl) 2:
R 20 is hydrogen, (C 1-C2) alkyl, fluoro, chloro, bromo, hydroxy, amino, cyano, nitro, (C 1-C2) alkoxy, (C 1-C2) haloalkyl or (C 1-C2) haloalkoxy;
R 21 is hydrogen, (C 1-C2) alkyl, fluoro, chloro, bromo, hydroxy, cyano, nitro, (C 1-C2) alkoxy, (C 1-C2) haloalkyl or (C 1-C2) haloalkoxy or-Y 3-L4-Z2;
Y 3 is absent, O, S, S (O), S (O) 2、NR'、C(O)、C(O)O、OC(O)、C(O)N(R')、N(R')C(O)、S(O)2 N (R ') or N (R') S (O) 2;
L 4 is absent or (C 1-C2) alkylene;
Z 2 is hydrogen, (C 1-C6) alkyl, (C 2-C4) alkenyl, (C 2-C4) alkynyl, phenyl, (C 3-C6) carbocyclyl, OR 4-to 6-membered heterocyclyl, wherein Z 2 is optionally substituted with one OR more substituents independently selected from (C 1-C4) alkyl, halo, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, (C 1-C4) alkoxy, (C 1-C4) alkylamino, amino, cyano, hydroxy, C (O) R ', C (O) OR', OC (O) R ', C (O) NR' R ', and N (R') C (O) R ', wherein each R' is independently hydrogen OR (C 1-C4) alkyl;
Or R 20 and R 21 together with the carbon atom to which they are attached form a (C 3-C6) carbocyclyl or 3-to 6-membered heterocyclyl, optionally substituted with one or more substituents selected from (C 1-C2) alkyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, (C 1-C2) alkylamino, amino, cyano and hydroxy;
R 22 is (C 1-C2) alkyl, -C (O) OR ', -C (O) NR ', -NR ' R ', phenyl, OR a 5 membered heteroaryl, wherein each R ' is independently hydrogen OR (C 1-C2) alkyl;
A "is (C 4-C6) carbocyclyl or 4-to 6-membered heterocyclyl optionally substituted with one or more substituents independently selected from (C 1-C2) alkyl, halo, hydroxy, oxo, cyano and (C 1-C2) alkoxy;
W 3 is NR 23 or CR 24R24';
R 23 is hydrogen, (C 1-C2) alkyl, (C 1-C4) haloalkyl, (C 1-C4) hydroxyalkyl, -C (O) CH 3 or-C (O) O- (C 1-C4) alkyl;
r 24 and R 24' are independently hydrogen, (C 1-C2) alkyl, cyclopropyl, fluoro, chloro, bromo, hydroxy, amino, cyano, nitro, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, -C (O) OR ', NR ' R ', phenyl OR 5 membered heteroaryl;
R 5 is hydrogen, (C 1-C4) alkyl, (C 3-C6) cycloalkyl, (C 1-C4) haloalkyl or cyano, wherein the alkyl or cycloalkyl is optionally substituted with one or more substituents selected from (C 1-C4) alkyl, (C 3-C6) cycloalkyl, hydroxy, (C 1-C2) alkoxy, amino, NH (C 1-C2) alkyl, N ((C 1-C2) alkyl) 2、(C1-C2) aminoalkyl and halo;
R 5' is hydrogen, (C 1-C4) alkyl, cyano, (C 1-C4) haloalkyl or-Y 4-L5-Z3;
Y 4 is absent, C (O) O or C (O) N (R ");
L 5 is absent or (C 1-C2) alkylene;
Z 3 is hydrogen, (C 1-C6) alkyl, phenyl, (C 3-C6) cycloalkyl or 4-to 6-membered heterocyclyl, wherein Z 3 is optionally substituted with one or more substituents independently selected from (C 1-C2) alkyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, amino, nitro, cyano and hydroxy, or
R 5 and R 5' together with the carbon atom to which they are attached form a (C 4-C6) carbocyclyl or a 4-to 6-membered heterocyclyl;
A 'is a 6-or 7-membered heterocyclyl, wherein in addition to R 5 and R 5', the heterocyclyl is optionally substituted with one or more substituents independently selected from oxo, (C 1-C2) alkyl, cyclopropyl, spiro-cyclopropyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, amino, cyano and hydroxy;
X 6 is CR 25 or N;
R 25 is hydrogen, fluoro, chloro or methyl;
R 6 is hydrogen, (C 1-C2) alkyl, (C 3-C4) cycloalkyl, (C 1-C2) haloalkyl, cyano, (C 2-C4) alkenyl, or (C 2-C4) alkynyl;
R 6' is (C 1-C4) alkyl, cyano, (C 1-C4) haloalkyl or-Y 5-L6-Z4;
Y 5 is absent, C (O) O, OC (O), C (O) N (R ') or S (O) 2 N (R');
l 6 is (C 1-C2) alkylene which is absent or optionally substituted by one or more substituents selected from (C 1-C2) alkyl and oxo;
Z 4 is hydrogen, (C 1-C6) alkyl, (C 2-C4) alkenyl, (C 2-C4) alkynyl, phenyl, (C 3-C6) carbocyclyl, (C 3-C6) cycloalkenyl, or 4-to 6-membered heterocyclyl, wherein Z 4 is optionally substituted with one or more substituents independently selected from oxo, (C 1-C4) alkyl, (C 3-C6) cycloalkyl, halo, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, (C 1-C4) alkoxy, (C 1-C4) alkylamino, amino, nitro, cyano, hydroxy, C (O) R u、C(O)ORu、OC(O)Ru、C(O)NRuRu, and N (R u)C(O)Ru), wherein each R u is independently hydrogen, (C 1-C4) alkyl, or (C 3-C6) cycloalkyl, or;
Z 4 is-Q-L 7-W4, where
Q is absent, O, NH or N (C 1-C2) alkyl;
L 7 is (C 1-C2) alkylene which is absent or optionally substituted by one or more substituents selected from oxo and (C 1-C2) alkyl;
W 4 is (C 1-C4) alkyl, phenyl, (C 3-C6) cycloalkyl, (C 3-C6) cycloalkenyl, or a 5-or 6-membered heterocyclyl, wherein W 4 is optionally substituted with one or more substituents independently selected from (C 1-C4) alkyl, halo, (C 1-C4) haloalkyl, (C 1-C4) haloalkoxy, (C 1-C4) alkoxy, (C 1-C4) alkylamino, amino, nitro, cyano, and hydroxy, or
R 6 and R 6' together with the carbon atom to which they are attached form a (C 3-C10) carbocyclyl or 4-to 10-membered heterocyclyl, optionally substituted with one or more substituents independently selected from oxo, (C 1-C2) alkyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, (C 1-C2) alkylamino, amino, nitro, cyano or hydroxy; or (C 3-C10) carbocyclyl or 4-to 10-membered heterocyclyl optionally fused to a 5-or 6-membered heteroaryl or benzene ring, and the 5-or 6-membered heteroaryl or benzene ring is optionally substituted with (C 1-C2) alkyl, halo, (C 1-C2) haloalkyl, (C 1-C2) haloalkoxy, (C 1-C2) alkoxy, (C 1-C2) alkylamino, amino, nitro, cyano or hydroxy; and is also provided with
R 6 "is hydrogen, (C 1-C4) alkyl, (C 1-C2) haloalkyl, (C 1-C2) alkoxy, (C 1-C2) haloalkoxy, cyano, nitro, ethynyl, phenyl, or 5-or 6-membered heteroaryl, wherein the alkyl, phenyl, or heteroaryl is optionally substituted with one or more substituents independently selected from halo, hydroxy, and amino.
2. The compound of claim 1, wherein R 1 is methyl, -OH, -NH 2、-CH2CH2OH、-CH2CH2NH2,
3. The compound of claim 2, wherein R 1 is methyl.
4. The compound of claim 2, wherein R 1 is-OH or-CH 2CH2 OH.
5. The compound of claim 2, wherein R 1 is-NH 2 or-CH 2CH2NH2.
6. The compound of any one of claims 1-5, wherein X 1 is N, CH, CCl, or CF.
7. The compound of claim 6, wherein X 1 is N.
8. The compound of claim 6, wherein X 1 is CH.
9. The compound of claim 6, wherein X 1 is CCl.
10. The compound of claim 6, wherein X 1 is CF.
11. The compound of any one of claims 1-10, wherein X 2 is CH 2, CHF, CHCl, or CF 2.
12. The compound of claim 11, wherein X 2 is CH 2.
13. The compound of claim 11, wherein X 2 is CHF.
14. The compound of claim 11, wherein X 2 is CHCl.
15. The compound of claim 11, wherein X 2 is CF 2.
16. The compound of any one of claims 1-15, whereinIs/>And the compound of formula (I) has the structure of formula I-1,/>Or a pharmaceutically acceptable salt or stereoisomer thereof.
17. The compound of claim 16, wherein X 3 is CH.
18. The compound of claim 16, wherein X 3 is N.
19. The compound of claim 16, wherein X 3 is CF.
20. The compound of claim 16, wherein X 3 is COMe.
21. The compound of any one of claims 16-20, wherein X 4 is CH.
22. The compound of any one of claims 16-21, wherein R 2 is (C 1-C2) alkyl, 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl, or 4 membered heterocyclyl (C 1) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups.
23. The compound of claim 22, wherein R 2 is methyl.
24. The compound of claim 22, wherein the heterocyclyl contains 1 heteroatom selected from N and O.
25. The compound of any one of claims 16-24, wherein R 3 is-L 3CR14R15R16.
26. The compound of claim 25, wherein L 3 is-O- (C 1) alkylene.
27. The compound of claim 25, wherein R 14 and R 15 together with the carbon atom to which they are attached form (C 3-C5) carbocyclyl, 4-to 7-membered heterocyclyl, or c=o.
28. The compound of claim 27, wherein R 14 and R 15 together with the same carbon atom to which they are attached form c=o.
29. The compound of claim 27, wherein R 14 and R 15 together with the same carbon atom to which they are attached form an oxetane ring.
30. The compound of claim 25, wherein R 16 is C 1-C6 alkyl, -NR 17R18, OR-OR 17.
31. The compound of claim 30, wherein R 16 is methyl, hydroxy, amino, or NHMe.
32. The compound of claim 16, wherein the compound has the formula I-1a, I-1b, I-1c, I-1d, I-1e, I-1f, I-1g, I-1h, I-1I, or I-1j:
or a pharmaceutically acceptable salt or stereoisomer thereof.
33. The compound of claim 32, wherein the compound has the formula I-1a ', I-1b ', I-1c ', I-1d ', or I-1e ':
or a pharmaceutically acceptable salt or stereoisomer thereof.
34. The compound of claim 32, wherein the compound has the formula I-1k, I-1l, I-1m, I-1n, I-1o, I-1p, I-1q, I-1r, I-1s, I-1t, I-1u, I-1v, I-1w, I-1x, I-1y, I-1z, I-1aa, I-1bb, I-1cc, or I-1dd:
/>
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
35. The compound of any one of claims 1-15, whereinIs/>And the compound of formula (I) has the structure of formula I-2,/>Or a pharmaceutically acceptable salt or stereoisomer thereof.
36. The compound of claim 35, wherein X 3 is CH.
37. The compound of claim 35 or 36, wherein X 4 is CH.
38. The compound of any one of claims 35-37, wherein R 2 is (C 1-C2) alkyl, 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl, or 4 membered heterocyclyl (C 1) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups.
39. The compound of claim 38, wherein R 2 is methyl.
40. The compound of claim 38, wherein the heterocyclyl contains 1 heteroatom selected from N and O.
41. The compound of any one of claims 35-40, wherein R 4 is- (CH 2)2W1W2.
42. A compound according to claim 41, wherein W 1 is CR 21R21'.
43. A compound according to claim 42, wherein R 21 and R 21' are both methyl.
44. The compound of any one of claims 41-43, wherein W 2 is cyano, hydroxy, or amino.
45. The compound of claim 44, wherein W 2 is hydroxy.
46. The compound of claim 35, wherein the compound has the formula I-2a, I-2b, I-2c, I-2d, I-2e, I-2f, I-2g, I-2h, I-2I, or I-2j:
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
47. The compound of claim 46, wherein the compound has the formula I-2k, I-2l, I-2m, I-2n, I-2o, I-2p, I-2q, I-2r, I-2s, I-2t, I-2u, I-2v, I-2w, I-2x, I-2y, I-2z, I-2aa, I-2bb, I-2cc, or I-2dd:
/>
/>
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
48. The compound of any one of claims 1-15, whereinIs/>And the compound of formula (I) has the structure of formula I-3,/>
Or a pharmaceutically acceptable salt or stereoisomer thereof.
49. The compound of claim 48, wherein X 3 is CH.
50. The compound of claim 48 or 49, wherein X 4 is CH.
51. The compound of any one of claims 48-50, wherein R 2 is (C 1-C2) alkyl, 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl, or 4 membered heterocyclyl (C 1) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups.
52. The compound of claims 48-51 wherein a 'is a 7 membered heterocyclyl wherein the heterocyclyl contains 2 heteroatoms selected from N and O and the heterocyclyl is further optionally substituted with one or more substituents independently selected from oxo, (C 1-C2) alkyl, cyclopropyl, spiro-cyclopropyl, halo, (C 1-C2) haloalkyl, (C 1-C2) alkoxy, amino, cyano and hydroxy, in addition to R 5 and R 5'.
53. The compound of claim 48, wherein the compound has the formula I-3a, I-3b, I-3c, I-3d, I-3e, I-3f, I-3g, I-3h, I-3I, I-3j, I-3k, I-3l, I-3m, I-3n, I-3o, I-3p, I-3q, I-3r, I-3s, or I-3t:
/>
/>
/>
Or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each R 25 is independently oxo, (C 1-C2) alkyl, cyclopropyl, spirocyclopropyl, halo, (C 1-C2) haloalkyl, (C 1-C2) alkoxy, amino, cyano, and hydroxy; and n is 0 to 3.
54. The compound of claim 48, wherein the compound has formula I-3u or I-3v:
or a pharmaceutically acceptable salt or stereoisomer thereof.
55. The compound of any one of claims 1-15, whereinIs/>And the compound of formula (I) has the structure of formula I-4,/>Or a pharmaceutically acceptable salt or stereoisomer thereof. /(I)
56. The compound of claim 55, wherein X 3 is CH.
57. The compound of claim 55 or 56, wherein X 4 is CH.
58. The compound of any one of claims 55-57, wherein R 2 is (C 1-C2) alkyl, 4 membered heterocyclyl, (C 3) carbocyclyl (C 1) alkyl, or 4 membered heterocyclyl (C 1) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups.
59. The compound of claim 55, wherein the compound has the formula I-4a, I-4b, I-4c, I-4d, I-4e, I-4f, I-4g, I-4h, I-4I, or I-4j:
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
60. The compound of any one of claims 1-15, whereinIs/>And the compound of formula (I) has the structure of formula I-4,/>Or a pharmaceutically acceptable salt or stereoisomer thereof.
61. The compound of claim 60, wherein X 3 is CH.
62. The compound of claim 60 or 61, wherein X 4 is CH.
63. The compound of any one of claims 60-62, wherein R 2 is 4-membered heterocyclyl or 4-membered heterocyclyl (C 2) alkyl; wherein the alkyl, carbocyclyl or heterocyclyl is further optionally substituted with one or more identical or different R 13 groups.
64. The compound of claim 60, wherein the compound has the formula I-5a, I-5b, I-5c, I-5d, I-5e, I-5f, I-5g, I-5h, I-5I, or I-5j:
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
65. The compound according to claim 1, which is:
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
or a pharmaceutically acceptable salt or stereoisomer thereof.
66. A pharmaceutical composition comprising a therapeutically effective amount of a compound or pharmaceutically acceptable salt or stereoisomer of any one of claims 1-65, and a pharmaceutically acceptable carrier.
67. The pharmaceutical composition of claim 66, which is in liquid or solid form.
68. A method of treating a disease or disorder characterized by aberrant B-cell lymphoma 6 (BCL 6) activity comprising administering to a subject in need thereof a therapeutically effective amount of a compound according to any one of claims 1-65 or a pharmaceutically acceptable salt or stereoisomer thereof.
69. The method of claim 68, wherein the disease or disorder is malignant lymphoma.
70. The method of claim 69, wherein the malignant lymphoma is peripheral T-cell lymphoma (PTCL), diffuse large B-cell lymphoma (DLBCL), mantle Cell Lymphoma (MCL), follicular Lymphoma (FL), chronic Lymphocytic Leukemia (CLL), acute lymphoblastic leukemia/lymphoma (ALL), or cutaneous T-cell lymphoma.
71. The method of claim 67 or 70, further comprising administering an additional anticancer agent.
72. The method of claim 71, wherein the other anti-cancer agent is an enhancer of a zeste homolog 2 (EZH 2) inhibitor.
CN202280061397.8A 2021-08-02 2022-08-01 Cyanopyridine and cyanopyrimidine BCL6 degrading agents Pending CN117980290A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/228,303 2021-08-02
US202263352063P 2022-06-14 2022-06-14
US63/352,063 2022-06-14
PCT/US2022/074387 WO2023015164A1 (en) 2021-08-02 2022-08-01 Cyanopyridine and cyanopyrimidine bcl6 degraders

Publications (1)

Publication Number Publication Date
CN117980290A true CN117980290A (en) 2024-05-03

Family

ID=90848311

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280061397.8A Pending CN117980290A (en) 2021-08-02 2022-08-01 Cyanopyridine and cyanopyrimidine BCL6 degrading agents

Country Status (1)

Country Link
CN (1) CN117980290A (en)

Similar Documents

Publication Publication Date Title
JP7010935B2 (en) Cyano-substituted heterocycle with activity as an inhibitor of USP30
JP7389856B2 (en) 1-Cyano-pyrrolidine compounds as USP30 inhibitors
EP3429994B1 (en) 2-cyanoisoindoline derivatives for treating cancer
TWI736566B (en) Tlr7/8 antagonists and uses thereof
US20210188857A1 (en) Sos1 inhibitors
ES2742843T3 (en) ROR quinolinyl modulators (gamma) t
WO2017009798A1 (en) Indazole and azaindazole compounds as irak-4 inhibitors
KR20240042620A (en) Cyanopyridine and cyanopyrimidine BCL6 degrader
CN110099901A (en) Pyrazolo [3,4-b] pyridine and imidazo [1,5-b] pyridazine as PDE1 inhibitor
KR20210049895A (en) Highly active STING protein agonist compounds
AU2022214618A1 (en) Cdk2 inhibitors and methods of using the same
JP2023540548A (en) Compounds with antitumor activity and their uses
CN111527090A (en) Novel pyrazolo-pyrrolo-pyrimidine-dione derivatives as P2X3 inhibitors
TWI730027B (en) Fused quadracyclic compounds, compositions and uses thereof
CN115209952A (en) Substituted pyrazolopyrimidines and use thereof
CN117980290A (en) Cyanopyridine and cyanopyrimidine BCL6 degrading agents
TW202146398A (en) Compounds targeting rna-binding proteins or rna-modifying proteins
WO2024035688A1 (en) Macrocyclic bcl6 degraders
WO2024086094A1 (en) Alkylamine-containing small molecule degraders of bcl6
TW202400578A (en) Novel compound and its use for inhibiting checkpoint kinase 2
WO2021218997A1 (en) Substituted five-membered aza-ring compound and use thereof in drug
CN117412959A (en) Substituted 1-aryl-1 '-heteroaryl compounds, substituted 1,1' -biaryl compounds, and methods of use thereof
WO2023183437A1 (en) Tead inhibitors and methods of use
CA3213823A1 (en) Selective modulators of ataxia telangiectasia mutated (atm) kinase and uses thereof
WO2023187677A1 (en) N-(pyrrolidin-3-yl or piperidin-4-yl)acetamide derivatives

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication