Sulfonylureas and related compounds and uses thereof

文档序号:163053 发布日期:2021-10-29 浏览:118次 中文

阅读说明:本技术 磺酰脲和相关化合物及其用途 (Sulfonylureas and related compounds and uses thereof ) 是由 卢克·奥尼尔 丽贝卡·科尔 马修·库珀 埃夫里尔·罗伯逊 凯特·施罗德 于 2016-02-16 设计创作,主要内容包括:本申请涉及磺酰脲和相关化合物及其用途。本发明提供了某些磺酰脲和相关化合物,它们具有有利的特性并且在抑制NLRP3炎性体的活化中显示出有用的活性。这样的化合物可用于治疗其中炎症过程,或更具体地,NLRP3炎性体已被认为是关键因素的各种病症。(The present application relates to sulfonylureas and related compounds and uses thereof. The present invention provides certain sulfonylureas and related compounds that have advantageous properties and exhibit useful activity in inhibiting the activation of the NLRP3 inflammasome. Such compounds are useful in the treatment of various disorders in which the inflammatory process, or more specifically, the NLRP3 inflammasome, has been identified as a key factor.)

1. A compound of formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein W is selected from O, S and Se;

j is selected from S and Se;

R1selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted;

R2selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted; and

R1is directly bonded to J via a carbon atom, and R2Directly bonded to the adjacent nitrogen via a carbon atom.

2. The compound of claim 1, wherein R1Is selected from C5Or C6Cycloalkyl, 5-or 6-membered heteroaryl, bicyclic heteroaryl, wherein at least one ring is heteroaryl, phenyl, biphenyl, phenylheterocyclyl, 5-or 6-membered heterocyclyl and heterocyclylcycloalkyl, all of which may be optionally substituted.

3. A compound as claimed in claim 1 or claim 2 wherein R is 1Selected from the group consisting of pyrazole, furan, tetrahydrofuran, tetrahydropyran, pyran, pyrrolidine, pyrrole, triazole, tetrazole, imidazole, pyridine, morpholine, piperazine, piperidine, substituted phenyl, phenyl heteroaryl, phenyl heterocyclyl, biphenyl, quinoline, isoquinoline, naphthyl, pyrazine and pyrimidine, all of which may be optionally substituted as appropriate.

4. The compound of any one of the preceding claims, wherein R1Is a 5-membered heterocyclyl or heteroaryl, each of which may be optionally substituted, containing at least one ring heteroatom selected from N, O and S.

5. The compound of claim 4, wherein R1Is a 5-membered azaheterocyclyl or 5-membered azaaryl, each of which may be optionally substituted.

6. A compound as claimed in claim 4 or claim 5 wherein R is1Is a 5-membered heterocyclyl or 5-membered heteroaryl, each of which may be optionally substituted, comprising at least two ring nitrogen atoms.

7. The compound of any one of the preceding claims, wherein R2Selected from bicyclic and tricyclic hydrocarbons, 5-, 6-and 7-membered heterocycles or heteroaryls, each of which may be optionally substituted, and substituted phenyl.

8. The compound of any one of the preceding claims, wherein R 2Selected from:

wherein each occurrence of Y is independently selected from C, N, S and O, which may be optionally substituted as appropriate;

R5、R11、R12、R13、R14and R15Independently selected from: hydrogen, halo, cyano, amide, sulfonamide, acyl, hydroxy, C1-C6Alkyl radical, C1-C6Haloalkyl, C3-C5Cycloalkyl radical, C1-C6Alkoxy, all of which may be substituted, where appropriate, by halogen, cyano or C1-C6Alkoxy is optionally substituted; and is

Wherein R is11And R12Can be combined to form a phenyl, a 5-or 6-membered oxa-ring or a 5-or 6-membered aza-ring, each of which can be optionally substituted;

R12and R13Can be combined to form a 5-or 6-membered azaaryl group, which can be optionally substituted; and

R14and R15Can be combined to form a 5-or 6-membered cycloalkyl ring, phenyl, a 5-or 6-membered oxa ring, or a 5-or 6-membered azaaryl, each of which can be optionally substituted.

9. The compound of any one of the preceding claims, wherein R1Selected from:

and

and with each such R1Group combination of R2Can be independently selected from:

and

10. the compound of any one of the preceding claims, wherein R2Selected from: substituted or hydrogenated indacenes,2, 6-dialkylphenyl, 2, 6-dialkyl-4-halophenyl, 2, 6-dicycloalkylphenyl, and 2, 6-dicycloalkyl-4-halophenyl.

Technical Field

The present invention relates to the field of medical treatment and diagnosis of diseases. More particularly, the invention relates to novel sulfonylureas and related compounds and their use in treating or identifying diseases or conditions that respond to modulation of NLRP3 or inhibition of activation of NLRP3 or a component associated with inflammatory processes.

Background

Any reference herein to background art is not to be construed as an admission that such prior art forms part of the common general knowledge in australia or elsewhere.

The NOD-like receptor (NLR) family, the protein 3 containing the pyrogenic protein domain (NLRP3) inflammasome, is a component of inflammatory processes, and its aberrant activation is pathogenic in genetic disorders such as cryptotherm-related periodic syndrome (CAPS) and complex diseases such as multiple sclerosis, type 2 diabetes, Alzheimer's disease and atherosclerosis.

NLRP3 is an intracellular signaling molecule that senses many pathogen-, environmental-, and host-derived factors. NLRP3 binds after activation to apoptosis-associated spot-like proteins containing the caspase activation and recruitment domain (ASC). The ASCs then polymerize to form large aggregates called ASC blobs. The polymerized ASC in turn interacts with the cysteine protease caspase-1 to form a complex called the inflammasome. This results in the activation of caspase-1, which cleaves the pro-inflammatory cytokines IL-1 β and IL-18 into their active forms and mediates a type of inflammatory cell death known as cell apoptosis. ASC spots can also recruit and activate caspase-8, which can process pre-IL-1 β and pre-IL-18 and trigger apoptotic cell death.

Caspase-1 cleaves pre-IL-1 β and pre-IL-18 into their active forms, which are secreted from the cell. Active caspase-1 also cleaves gasdermin-D, triggering cell apoptosis. Caspase-1 also mediates the release of alarm molecules such as IL-33 and the high mobility histobox 1 protein (HMGB1) by controlling the cell pyrophoric cell death pathway. Caspase-1 also cleaves intracellular IL-1R2, causing it to degrade and allowing IL-1 α to be released. In human cells, caspase-1 may also control the processing and secretion of IL-37. Many other caspase-1 substrates, such as the cytoskeleton and components of the glycolytic pathway, may contribute to caspase-1 dependent inflammation.

NLRP 3-dependent ASC spots are released into the extracellular environment where they can activate caspase-1, induce processing of caspase-1 substrates and spread inflammation.

Active cytokines derived from the inflammatory activation of NLRP3 are important drivers of inflammation and interact with other cytokine pathways to develop an immune response to infection and injury. For example, IL-1 β signaling induces the secretion of the proinflammatory cytokines IL-6 and TNF. IL-1 β and IL-18 act synergistically with IL-23 to induce IL-17 production by memory CD4 Th17 cells and γ δ T cells in the absence of T cell receptor involvement. IL-18 and IL-12 can also act synergistically to induce IFN- γ production by memory T cells and NK cells that drive a Th1 response.

Other intracellular Pattern Recognition Receptors (PRRs) are also capable of forming inflammasomes. These include other NLR family members such as NLRP1 and NLRC4, as well as non-NLR PRRs such as melanoma 2(AIM2) and double-stranded dna (dsdna) receptors not present in interferon, gamma inducible protein 16(IFI 16). NLRP 3-dependent IL-1. beta. processing can also be activated by an indirect non-canonical pathway downstream of caspase-11.

Hereditary CAPS disease Muckle-Wells syndrome (MWS), familial cold autoinflammatory syndrome and neonatal onset multisystem inflammatory disease are caused by gain of function mutations in NLRP3, defining NLRP3 as a key component of the inflammatory process. NLRP3 has also been shown to be involved in the pathogenesis of many complex diseases, including in particular metabolic disorders such as type 2 diabetes, atherosclerosis, obesity and gout.

A role for NLRP3 in central nervous system disease is emerging, and pulmonary disease is also shown to be affected by NLRP 3. In addition, NLRP3 plays a role in the development of liver disease, kidney disease, and aging. Many of these associations are using Nlrp3-/-Mice are defined, but there is also insight into the specific activation of NLRP3 in these diseases. In type 2 diabetes, deposition of islet amyloid polypeptide in the pancreas activates NLRP3 and IL-1 β signaling, leading to cell death and inflammation.

Several small molecules have been shown to inhibit the NLRP3 inflammasome. Glibenclamide inhibits IL-1 β production at micromolar concentrations in response to activation of NLRP3 but not NLRC4 or NLRP 1. Other previously characterized NLRP3 inhibitors include parthenolide, 3, 4-methylenedioxy-beta-nitrostyrene, and dimethyl sulfoxide (DMSO), although these agents have limited potency and are non-specific

Current treatments for NLRP 3-related diseases include biologies targeting IL-1. These are recombinant IL-1 receptor antagonists, anakinra, the neutralizing IL-1. beta. antibody kanamycin, and the soluble decoy IL-1 receptor riloncept (rilonacept). These approaches have proven successful in the treatment of CAPS, and these biologies have been used in clinical trials for other IL-1 β related diseases.

Several small molecules have been shown to inhibit the NLRP3 inflammasome. Glibenclamide inhibits IL-1 β production at micromolar concentrations in response to activation of NLRP3 but not NLRC4 or NLRP 1. Other previously characterized NLRP3 inhibitors include parthenolide, 3, 4-methylenedioxy- β -nitrostyrene, and Dimethylsulfoxide (DMSO), although these agents are limited in potency and are non-specific.

Certain diarylsulfonylurea-containing compounds have been identified as Cytokine Release Inhibitory Drugs (CRIDs) (Perregaux et al; J.Pharmacol. Exp.Ther.299,187-197,2001). CRIDs are a class of diarylsulfonylurea-containing compounds that inhibit IL-1 β post-translational processing. The post-translational processing of IL-1 β is accompanied by caspase-1 activation and cell death. CRIDs arrest activated monocytes, leaving caspase-1 inactive and retaining plasma membrane latency.

There is a need to provide compounds having improved pharmacological and/or physiological and/or physicochemical properties and/or to provide useful alternatives to known compounds.

Summary of The Invention

According to a first aspect of the present invention there is provided a compound of formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein W is selected from O, S and Se;

J is selected from S and Se;

R1selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted;

R2selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted; and

R1is directly bonded to J via a carbon atom, and R2Directly bonded to the adjacent nitrogen via a carbon atom.

According to a second aspect of the present invention there is provided a pharmaceutical composition comprising a compound of the first aspect or a pharmaceutically acceptable salt, solvate or prodrug thereof together with a pharmaceutically acceptable carrier, diluent and/or excipient.

A third aspect of the invention is a method of treating or preventing a disease, disorder or condition comprising the steps of: administering an effective amount of a compound of the first aspect or a pharmaceutically effective salt, solvate or prodrug thereof, or a pharmaceutical composition of the second aspect, thereby treating or preventing a disease, disorder or condition.

A fourth aspect of the invention provides a compound of the first aspect or a pharmaceutically effective salt, solvate or prodrug thereof or a pharmaceutical composition of the second aspect for use in the treatment or prevention of a disease, disorder or condition.

A fifth aspect of the invention provides the use of a compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof, in the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition.

In one embodiment, the disease, disorder or condition is responsive to inhibition of activation of the NLRP3 inflammasome.

In some particular non-limiting embodiments of the above aspects, the disease, disorder or condition is a disease, disorder or condition of the immune system, cardiovascular system, endocrine system, gastrointestinal tract, renal system, respiratory system, central nervous system, is a cancer or other malignancy and/or is caused by or associated with a pathogen.

In a sixth aspect of the invention, there is provided a method of diagnosing a disease, disorder or condition in a mammal, comprising the step of administering to said mammal or a biological sample obtained from said mammal a labeled compound of formula (I), (Ia), (Ib), (Ic) or (II), or a pharmaceutically effective salt, solvate or prodrug thereof, to facilitate diagnosis of said disease, disorder or condition in a mammal.

A seventh aspect of the present invention is directed to a method of modulating the activity of a biological target comprising the step of exposing the biological target to a compound of the first aspect or a pharmaceutically acceptable salt thereof.

The biological target can be selected from NLRP3 inflammasome, IL-1 beta, IL-17, IL-18, IL-1 alpha, IL-37, IL-33 and Th17 cells.

The invention may include the following features:

1. a compound of formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein W is selected from O, S and Se;

j is selected from S and Se;

R1selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted;

R2selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted; and

R1is directly bonded to J via a carbon atom, and R2Directly bonded to the adjacent nitrogen via a carbon atom.

2. A compound according to feature 1, wherein R1Is selected from C5Or C6Cycloalkyl, 5-or 6-membered heteroaryl, bicyclic heteroaryl, wherein at least one ring is heteroaryl, phenyl, biphenyl, phenylheterocyclyl, 5-or 6-membered heterocyclyl and heterocyclylcycloalkyl, all of which may be optionally substituted.

3. The compound according to feature 1 or feature 2, wherein R1Selected from the group consisting of pyrazole, furan, tetrahydrofuran, tetrahydropyran, pyran, pyrrolidine, pyrrole, triazole, tetrazole, imidazole, pyridine, morpholine, piperazine, piperidine, substituted phenyl, phenyl heteroaryl, phenyl heterocyclyl, biphenyl, quinoline, isoquinoline, naphthyl, pyrazine and pyrimidine, all of which may be optionally substituted as appropriate.

4. The compound of any one of the preceding features, wherein R1Is a 5-membered heterocyclyl or heteroaryl, each of which may be optionally substituted, containing at least one ring heteroatom selected from N, O and S.

5. The compound of feature 4 wherein R1Is a 5-membered azaheterocyclyl or 5-membered azaaryl, each of which may be optionally substituted.

6. The compound of feature 4 or feature 5, wherein R1Is a 5-membered heterocyclyl or 5-membered heteroaryl, each of which may be optionally substituted, comprising at least two ring nitrogen atoms.

7. The compound of any one of the preceding features, wherein R2Selected from bicyclic and tricyclic hydrocarbons, 5-, 6-and 7-membered heterocycles or heteroaryls, each of which may be optionally substituted, and substituted phenyl.

8. The compound of any one of the preceding features, wherein R2Selected from:

wherein each occurrence of Y is independently selected from C, N, S and O, which may be optionally substituted as appropriate;

R5、R11、R12、R13、R14and R15Independently selected from: hydrogen, halo, cyano, amide, sulfonamide, acyl, hydroxy, C1-C6Alkyl radical, C1-C6Haloalkyl, C3-C5Cycloalkyl radical, C1-C6Alkoxy, all of which may be substituted, where appropriate, by halogen, cyano or C1-C6Alkoxy is optionally substituted; and is

Wherein R is11And R12Can be combined to form a phenyl, a 5-or 6-membered oxa-ring or a 5-or 6-membered aza-ring, each of which can be optionally substituted;

R12and R13Can be combined to form a 5-or 6-membered azaaryl group, which can be optionally substituted; and

R14and R15Can be combined to form a 5-or 6-membered cycloalkyl ring, phenyl, a 5-or 6-membered oxa ring, or a 5-or 6-membered azaaryl, each of which can be optionally substituted.

9. The compound of any one of the preceding features, wherein R1Selected from:

and

and with each such R1Group combination of R2Can be independently selected from:

and

10. the compound of any one of the preceding features, wherein R2Selected from: substituted or hydrogenated indacenes, 2, 6-dialkylphenyl, 2, 6-dialkyl-4-halophenyl, 2, 6-dicycloalkylphenyl, and 2, 6-dicycloalkyl-4-halophenyl.

11. The compound of any one of the preceding features, wherein R2Selected from: hexahydroindacene, 2, 6-diisopropylphenyl, 2, 6-diisopropyl-4-chlorophenyl, 2, 6-dicyclopropylphenyl and 2, 6-dicyclopropyl-4-chlorophenyl.

12. The compound of any one of the preceding features, wherein J is a sulfur atom.

13. The compound of any one of the preceding features, wherein W is an oxygen atom.

14. The compound of any one of the preceding features, wherein the compound is a compound of formula (Ia), (Ib), or (Ic), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein R is1As defined in any of the preceding features.

15. As described in feature 14A compound of formula (I) wherein R1Selected from the group consisting of pyrazole, furan, tetrahydrofuran, tetrahydropyran, pyran, pyrrolidine, pyrrole, triazole, tetrazole, imidazole, pyridine, morpholine, piperazine, piperidine, substituted phenyl, phenyl heteroaryl, phenyl heterocyclyl, biphenyl, quinoline, isoquinoline, naphthyl, pyrazine and pyrimidine, all of which may be optionally substituted as appropriate.

16. The compound of feature 14 or feature 15, wherein R1Selected from:

17. the compound of any one of the preceding features, wherein the compound is a compound of formula (II), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein A, B, D and E are independently selected from C, N, O, S and Se, but at least one of which is C;

each dotted line may represent a bond

R2As defined in any of the preceding features, or R2May be a fluorophore;

R6independently at each occurrence, is selected from: hydrogen, halide, cyano, C 1-C6Alkyl radical, C1-C6Alkylamino radical, C1-C6Alkyl hydroxy, C3-C6Cycloalkyl, alkylphenyl, phenyl, benzyl, C1-C6Esters, C2-C6Alkenyl radical, C1-C6Trifluoroalkyl and C1-C6Alkoxy, each of which may be optionally substituted as appropriate, or R6Can be used forIs a fluorophore.

18. The compound of feature 17, wherein the compound of formula (II) is selected from:

wherein R is40Selected from H, alkyl and halo;

R41selected from H, alkyl and cycloalkyl;

p is independently at each occurrence selected from C, O and S; and

wherein R is6At each occurrence, when present, is independently selected from those groups as defined in feature 17.

19. The compound of any one of the preceding features, wherein the compound is selected from a compound of formula (IIa), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein R is11、R12、R13、R14And R15As defined in any one of features 8 to 11;

A. b, D and E are selected from N and C, and at least two of A, B, D and E are N;

R6independently at each occurrence, is selected from: hydrogen, halide, cyano, C1-C6Alkyl radical, C1-C6Alkylamino radical, C1-C6Alkyl hydroxy, C3-C6Cycloalkyl, alkylphenyl, phenyl, benzyl, C1-C6Esters, C2-C6Alkenyl radical, C1-C6Trifluoroalkyl and C1-C6Alkoxy groups, each of which may be optionally substituted.

20. The compound of any one of the preceding features, wherein the compound is a compound of formula (IIb), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein Y and R5As defined in any one of features 8 to 11;

A. b, D and E are selected from N and C, and at least two of A, B, D and E are N;

R6independently at each occurrence, is selected from: hydrogen, halide, cyano, C1-C6Alkyl radical, C1-C6Alkylamino radical, C1-C6Alkyl hydroxy, C3-C6Cycloalkyl, alkylphenyl, phenyl, benzyl, C1-C6Esters, C2-C6Alkenyl radical, C1-C6Trifluoroalkyl and C1-C6Alkoxy groups, each of which may be optionally substituted.

21. The compound of any one of the preceding features, wherein the compound is a compound of formula (IIIa), (IIIb), or (IIIc), or a pharmaceutically acceptable salt, solvate, or prodrug thereof:

wherein R is21Selected from H, alkyl, perhaloalkyl or hydroxyalkyl;

R22selected from H, alkyl, perhaloalkyl, C3-C6Cycloalkyl, phenyl or benzyl;

R18is H or halogen;

R16and R17Is H, alkyl or cycloalkyl; or R16And R17Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturatedOptionally, the ring contains one or two heteroatoms selected from N, O and S;

R19And R20Is H or alkyl; or R19And R20Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

provided that R is21And R22Are not all H; and

provided that R is16、R17、R18、R19And R20Not all are H.

22. The compound of feature 21, wherein:

R21selected from H, alkyl, perhaloalkyl or hydroxyalkyl;

R22selected from H, alkyl, perhaloalkyl, C3-C6Cycloalkyl, phenyl or benzyl;

R16and R17Together with the atoms to which they are attached form a cyclopentyl ring;

R19and R20Together with the atoms to which they are attached form a cyclopentyl ring;

R18is H or halogen; and is

Provided that R is21And R22Not all are H.

23. The compound of feature 21, wherein:

R21selected from H, alkyl, perhaloalkyl or hydroxyalkyl;

R22selected from H, alkyl, perhaloalkyl, C3-C6Cycloalkyl, phenyl and benzyl;

R16and R20Is C1-6Alkyl or C3-5A cycloalkyl group;

R17and R19Is a compound of formula (I) wherein the compound is H,

R18is H or halogen; and is

Provided that R is21And R22Not all are H.

24. The compound of any one of the preceding features, wherein the compound is a compound of formula (IVa), (IVb) or (IVc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

Wherein R is21And R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl, or R21And R22Together with the carbon atom to which they are attached may form a cyclopentyl or cyclohexyl ring;

R18is H or halogen;

R16and R17Is H, alkyl or cycloalkyl; or R16And R17Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

R19and R20Is H or alkyl; or R19And R20Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

provided that R is21And R22Are not all H; and

provided that R is16、R17、R18、R19And R20Not all are H.

25. The compound of feature 24 wherein:

R21and R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl;

R16and R17Together with the atoms to which they are attached form a cyclopentyl ring;

R19and R20Together with the atoms to which they are attached form a cyclopentyl ring;

R18is H or halogen; and is

Provided that R is21And R22Not all are H.

26. The compound of feature 24 wherein:

R21and R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl; preferably, the perhaloalkyl and hydroxyalkyl groups are C1-6Perhaloalkyl and hydroxyalkyl;

R16and R20Is C1-6Alkyl or C3-5A cycloalkyl group;

R17and R19Is a compound of formula (I) wherein the compound is H,

R18is H or halogen;

provided that R is21And R22Not all are H.

27. A compound according to any one of the preceding features, wherein the compound is a compound of formula (Va), (Vb) or (Vc):

wherein R is21And R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl;

R18is H or halogen;

R16and R17Is H, alkyl or cycloalkyl; or R16And R17Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

R19and R20Is H or alkyl; or R19And R20Together with the carbon atom to which they are attachedForming a 5-or 6-membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, O and S;

Provided that R is21And R22Are not all H; and is

Provided that R is16、R17、R18、R19And R20Not all are H.

28. The compound of feature 27, wherein:

R21and R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl;

R16and R17Together with the atoms to which they are attached form a cyclopentyl ring;

R19and R20Together with the atoms to which they are attached form a cyclopentyl ring;

R18is H or halogen; and is

Provided that R is21And R22Not all are H.

29. The compound of feature 27, wherein:

R21and R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl;

R16and R20Is C1-6Alkyl or C3-5A cycloalkyl group;

R17and R19Is H;

R18is H or halogen; and is

Provided that R is21And R22Not all are H.

30. The compound of any one of the preceding features, wherein the compound is a compound of formula (VIa) or (VIb), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein R is22Selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl;

R18is H or halogen;

R16and R17Is H, alkyl or cycloalkyl; or R16And R17Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

R19And R20Is H or alkyl; or R19And R20Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S; and is

Provided that R is16、R17、R18、R19And R20Not all are H.

31. The compound of feature 30, wherein:

R22selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl;

R16and R17Together with the atoms to which they are attached form a cyclopentyl ring;

R19and R20Together with the atoms to which they are attached form a cyclopentyl ring; and is

R18Is H or halogen.

32. The compound of feature 30, wherein:

R22selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl;

R16and R20Is C1-6Alkyl or C3-5Cycloalkyl, preferably isopropyl or cyclopentyl;

R17and R19Is H; and

R18is H or halogen.

33. The compound of any one of the preceding features, wherein the compound is a compound of formula (VII):

wherein Q is O or S;

R30independently at each occurrence, selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl and alkylamino;

R18Is H or halogen;

R16and R17Is H or alkyl; or R16And R17Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

R19and R20Is H or alkyl; or R19And R20Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

provided that R is16、R17、R18、R19And R20Are not all H; and is

Provided that when Q is O, and R16And R17And R19And R20When taken alone with each carbon atom to which they are attached to form a cyclopentyl ring, then R30Is not C-3A hydroxyalkyl group.

34. The compound of feature 33, wherein:

q is O or S;

R30independently at each occurrence, selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl and alkylamino.

R16And R17Together with the atoms to which they are attached form a cyclopentyl ring;

R19and R20Together with the atoms to which they are attached form a cyclopentyl ring; and

R18is H or halogen; and is

Provided that when Q is O, then R30Is not C-3A hydroxyalkyl group.

35. The compound of feature 33 or feature 34, wherein:

Q is O or S;

R30independently at each occurrence, selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl and alkylamino;

R16and R20Is C1-6Alkyl, preferably isopropyl;

R17and R19Is H; and

R18is H or halogen.

36. The compound of any one of the preceding features, wherein the compound is selected from:

and

37. the compound of any one of the preceding features, wherein the compound, or a pharmaceutically effective salt, solvate or prodrug thereof, is an inhibitor of the NLRP3 inflammasome.

38. A pharmaceutical composition comprising a compound of any one of features 1 to 37, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier, diluent and/or excipient.

39. A method of treating or preventing a disease, disorder or condition comprising the steps of: administering an effective amount of a compound according to any one of features 1 to 37 or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition according to feature 38, thereby treating or preventing the disease, disorder or condition.

40. The method of feature 39, wherein the disease, disorder or condition is one that responds to inhibition of activation of the NLRP3 inflammasome.

41. The method of feature 39 or feature 40, wherein the disease, disorder or condition is responsive to modulation of one or more of IL-1 β, IL-17, IL-18, IL-1 α, IL-37, IL-33 and Th17 cells.

42. The method of any one of features 39 to 41, wherein the disease, disorder or condition is a disease, disorder or condition of the immune system.

43. The method of any one of features 39 to 41, wherein the disease, disorder or condition is an inflammatory disease, disorder or condition or an autoimmune disease, disorder or condition.

44. The method of any one of features 39-41, wherein the disease, disorder or condition is a disease, disorder or condition of the skin.

45. The method of any one of features 39 to 41, wherein the disease, disorder or condition is a disease, disorder or condition of the cardiovascular system.

46. The method of any one of features 39-41, wherein the disease, disorder or condition is a cancer, tumor or other malignancy.

47. The method of any one of features 39 to 41, wherein the disease, disorder or condition is a disease, disorder or condition of the renal system.

48. The method of any one of features 39 to 41, wherein the disease, disorder or condition is a disease, disorder or condition of the gastrointestinal tract.

49. The method of any one of features 39 to 41, wherein the disease, disorder or condition is a disease, disorder or condition of the respiratory system.

50. The method of any one of features 39 to 41, wherein the disease, disorder or condition is a disease, disorder or condition of the endocrine system.

51. The method of any one of features 39 to 41, wherein the disease, disorder or condition is a disease, disorder or condition of the Central Nervous System (CNS).

52. The method of any one of features 39 to 41, wherein the disease, disorder or condition is selected from constitutive inflammation, including cryptotropin-associated periodic syndrome (CAPS): moore-weirs syndrome (MWS), Familial Cold Autoinflammatory Syndrome (FCAS), and neonatal onset multiple system inflammatory disease (NOMID); including auto-inflammatory diseases: familial Mediterranean Fever (FMF), TNF receptor-related cycle syndrome (TRAPS), Mevalonate Kinase Deficiency (MKD), hyperimmunoglobulinemia D and periodic fever syndrome (HIDS), interleukin 1 receptor (DIRA) antagonist deficiency, magenis syndrome, septic arthritis, pyoderma gangrenosum and acne (PAPA), a20 deficiency (HA20), granulomatous arthritis in children (PGA), PLCG 2-associated antibody deficiency and immune dysregulation (PLAID), PLCG 2-associated autoinflammation, antibody deficiency and immune dysregulation (aploid), sideroblastic anemia (SIFD) with B-cell immune deficiency, periodic fever and developmental delay; swiner's syndrome, Chronic Nonbacterial Osteomyelitis (CNO), Chronic Relapsing Multifocal Osteomyelitis (CRMO) and synovitis, acne, impetigo, hyperostosis, osteomyelitis Syndrome (SAPHO); autoimmune diseases, including Multiple Sclerosis (MS), type 1 diabetes, psoriasis, rheumatoid arthritis, behcet's disease, sjogren's syndrome, and schnitz's syndrome; respiratory diseases including Chronic Obstructive Pulmonary Disorder (COPD), steroid-resistant asthma, asbestosis, silicosis and cystic fibrosis; central nervous system diseases including parkinson's disease, alzheimer's disease, motor neuron disease, huntington's disease, cerebral malaria and brain damage caused by pneumococcal meningitis; metabolic diseases including type 2 diabetes, atherosclerosis, obesity, gout, pseudogout; ocular diseases including those of the ocular epithelium, age-related macular degeneration (AMD), corneal infections, uveitis, and dry eye; renal diseases, including chronic kidney disease, oxalate kidney disease, and diabetic nephropathy; liver diseases including non-alcoholic steatohepatitis and alcoholic liver disease; inflammatory reactions of the skin, including contact hypersensitivity and sunburn; inflammatory response of joints including osteoarthritis, systemic juvenile idiopathic arthritis, adult onset steve's disease, recurrent polychondritis; viral infections including alphaviruses including chikungunya and ross river, and flaviviruses including dengue and zaka viruses, influenza, HIV; hidradenitis Suppurativa (HS) and other skin diseases that cause cysts; cancers including lung cancer metastasis, pancreatic cancer, gastric cancer, myelodysplastic syndrome, leukemia; polymyositis; stroke; myocardial infarction; graft versus host disease; hypertension; colitis; infection by worms; bacterial infection; abdominal aortic aneurysm; healing of the wound; depression, psychological stress; including pericarditis of delayer's syndrome, ischemia reperfusion injury, and any disease in which an individual has been determined to carry germline or somatic non-silent mutations in NLRP 3.

53. The method of any one of features 39 to 52, wherein the treatment or prevention of the disease, disorder or condition is performed on a mammal.

54. The method of feature 53, wherein the mammal is a human subject.

55. A method of diagnosing a disease, disorder or condition in a mammal comprising the steps of: administering to the mammal or a biological sample obtained from the mammal a labeled compound as described in any one of features 1 to 37, or a pharmaceutically effective salt, solvate or prodrug thereof, or a metal ion chelating complex thereof, to facilitate diagnosis of the disease, disorder or condition in the mammal.

56. A method of modulating an activity of a biological target comprising the step of exposing the biological target to a compound of any one of features 1 to 37, or a pharmaceutically effective salt, solvate or prodrug thereof.

57. The method of feature 56, wherein the biological target can be selected from the group consisting of NLRP3 inflammasome, IL-1 β, IL-17, IL-18, IL-1 α, IL-37, IL-33, and Th17 cells.

The various features and embodiments of the invention mentioned in the various sections above apply appropriately to the other sections, as appropriate. Thus, features specified in one section may be combined with features specified in other sections as appropriate.

Other features and advantages of the present invention will become apparent from the detailed description that follows.

Drawings

In order that the invention may be readily understood and put into practical effect, preferred embodiments will now be described, by way of example, with reference to the accompanying drawings, in which:

figures 1A to 1C are a series of graphical representations of plasma concentrations of known sulfonylureas (MCC950) following different levels of administration to mice; and

figures 2A to 2C are a series of graphical representations of plasma concentrations of sulfonylureas of the invention (MCC7840) following different levels of administration to mice.

Detailed Description

The present invention is predicated, at least in part, on the discovery that certain sulfonylureas and related compounds have advantageous properties and are in inhibiting the activation of the NLRP3 inflammasome and/or inhibiting IL-1 β and/or IL-17 and/or IL-18, and/or IL-1 α, and/or IL-37 and/or IL-33, and interfering with or modulating the activity of T helper cells such as Th 17. In particular, the compounds of the invention are useful for the treatment of a number of disorders in which inflammatory processes or NLRP3 inflammasome and/or IL-1 β and/or IL-17 and/or IL-18 and/or IL-1 α, and/or IL-37, and/or IL-33 and/or Th17 cells play a role.

Evidence from human CAPS patients and CAPS mouse models led the present inventors to believe that NLRP3 inhibition would be an excellent treatment over IL-1 biologies, as inhibition of all NLRP 3-dependent processes would be more effective than inhibition of a single NLRP 3-dependent process (e.g., IL-1 signaling).

Individuals with CAPS show dysregulated secretion of both IL-1 β and IL-18, and CAPS patients treated with anti-IL-1 biologies present with residual disease. IL-1 biologics do not prevent symptoms such as bony overgrowth and joint deformities. In addition, symptoms related to the central nervous system (e.g., hearing loss) are difficult to control with IL-1 biologics, which seem to be poor at penetrating the central nervous system. Studies in the CAPS mouse model have shown that IL-1 signaling or a deficiency in IL-18 alone is not sufficient to block systemic inflammation, particularly in older animals. In a severe model of CAPS, only complete loss of caspase-1 signaling could completely rescue the disease.

Specific inhibition of NLRP3 by sulfourea-containing compounds (e.g., those of the first aspect) can block all processes downstream of NLRP3, including ASC spot formation and caspase-8 and caspase-1 activation. Thus, NLRP3 inhibition will block all caspase-1 dependent processes, such as IL-1 β, IL-18 and IL-37 processing and secretion; gasdermin D cleavage; apoptosis of cells and release of IL-1 alpha, IL-33 and HMGB. Furthermore, NLRP 3-dependent extracellular release of ASC spots will be blocked and caspase-8-dependent pre-IL-1 β and pre-IL-18 cleavage and apoptotic cell death will be prevented. Thus, specific inhibition of NLRP3 by the compounds of the first aspect will prevent multiple downstream inflammatory signaling and should therefore prove to be a more effective anti-inflammatory therapy than IL-1 blockade alone.

anti-IL-1 biologics block IL-1 derived from NLRP 3-independent sources, and such IL-1 produced by other inflammatories (e.g., NLRC4, NLRP1, NLRP6, AIM2) and IL-1 produced by the latter pathway may be important for the host to defend against pathogens. For example, patients receiving IL-1/IL-1R antagonists exhibit an increased incidence of upper respiratory tract infections. The specific inhibition of NLRP3 by the compounds of the invention may therefore exert a less general immunosuppressive effect (less produced immunosuppression) compared to anti-IL-1 biologicals.

IL-1. beta. and IL-18 produced by Nlrp 3/caspase-1 axis play a key role in driving IL-17 production by CD4 Th17 cells and γ δ T cells. IL-1 β and IL-18 act synergistically with IL-23 to induce IL-17 production by memory CD4 Th17 cells and γ δ T cells in the absence of TCR involvement. IL-1 driven IL-17 has also been shown to be involved in psoriasis, type I diabetes, rheumatoid arthritis, type 2 diabetes, atherosclerosis, obesity, gout and, more recently, asthma.

Essentially, each of these diseases has been shown to involve activation of tissue macrophages, dendritic cells or brain microglia, driven by soluble mutant proteases or by unfortunate phagocytosis of extracellularly accumulated metabolites. NLRP3 senses these events, resulting in IL-1 release, triggering inflammation to clear the aggressive substances. If this process becomes chronic or overactive, disease results, explaining why so many diseases have been shown to involve NLRP 3. Inhibitors that act to prevent activation of NLRP3 may therefore be useful in IL-17 driven as well as IL-1 driven diseases.

In this patent specification, the terms "comprises," "comprising," "includes," "including," or similar terms, are intended to cover a non-exclusive inclusion, such that a method or composition that comprises a list of elements does not include only those elements but may include other elements not expressly listed.

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 invention belongs.

The term "pharmaceutically acceptable salt" as used herein refers to salts that are toxicologically safe for systemic or topical administration, e.g., salts prepared from pharmaceutically acceptable non-toxic bases or acids, including inorganic or organic bases and inorganic or organic acids. The pharmaceutically acceptable salt may be selected from the group comprising: alkali and alkaline earth metals, ammonium, aluminum, iron, amines, glucosamine, chloride, sulfate, sulfonate, bisulfate, nitrate, citrate, tartrate, piclorate, phosphate, carbonate, bicarbonate, malate, maleate, naphthalenesulfonate, fumarate, succinate, acetate, benzoate, terephthalate, palmitate, piperazine, pectate, and S-methylmethionate, and the like.

The term "alkyl" refers to a straight or branched alkyl substituent containing, for example, from 1 to about 12 carbon atoms, preferably from 1 to about 9 carbon atoms, more preferably from 1 to about 6 carbon atoms, even more preferably from 1 to about 4 carbon atoms, and still more preferably from 1 to 2 carbon atoms. Examples of such substituents may be selected from methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, pentyl, isopentyl, 2-methylbutyl, 3-methylbutyl, hexyl, heptyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2-ethylbutyl, 3-ethylbutyl, octyl, nonyl, decyl, undecyl, dodecyl and the like. The carbon numbers mentioned relate to the carbon skeleton and the carbon branches, but do not include carbon atoms belonging to any substituents, for example carbon atoms of alkoxy substituents branching from the main carbon chain. Substituted alkyl groups include alkyl groups substituted with one or more moieties selected from: halo (e.g., Cl, F, Br, and I); halogenated alkyls (e.g., CF)32-Br-ethyl, CH2F、CH2Cl、CH2CF3Or CF2CF3) (ii) a A hydroxyl group; an amino group; a carboxylate; a carboxamide group; an alkylamino group; an arylamino group; an alkoxy group; an aryloxy group; a nitro group; an azide group; a cyano group; thio; a sulfonic acid; a sulfate ester; a phosphonic acid; a phosphate ester; and phosphonates and those described under the definition of "optionally substituted".

The term "alkenyl" refers to an optionally substituted unsaturated straight or branched chain hydrocarbon group having 2 to 12 carbon atoms, preferably 2 to 9 carbon atoms, more preferably 2 to 6 carbon atoms, and having at least one carbon-carbon double bond. Where appropriate, the alkenyl radicals may have the indicated number of carbon atoms, for example C2-C6Alkenyl groups, which include alkenyl groups having 2, 3, 4, 5, or 6 carbon atoms in a straight or branched chain arrangement. The carbon numbers mentioned relate to the carbon skeleton and the carbon branches, but do not include carbon atoms belonging to any substituents. Examples of such substituents may be selected from the group consisting of ethenyl, propenyl, isopropenyl, butenyl, secondary and tertiary butenyl, pentenyl, hexenyl, hepta-1, 3-diene, hexa-1, 3-diene, non-1, 3, 5-triene and the like. Substituted alkenyl includes one or more moieties selected from the group consisting ofSubstituted alkenyl groups: halo (e.g., Cl, F, Br, and I); halogenated alkyls (e.g., CF)32-Br-ethyl, CH2F、CH2Cl、CH2CF3Or CF2CF3) (ii) a A hydroxyl group; an amino group; a carboxylic acid ester; a carboxamide group; an alkylamino group; an arylamino group; an alkoxy group; an aryloxy group; a nitro group; an azide group; a cyano group; thio; a sulfonic acid; a sulfate ester; a phosphonic acid; a phosphate ester; and phosphonates and those described under the definition of "optionally substituted".

The term "alkoxy" as used herein means a straight or branched chain alkyl group (i.e., -O-alkyl) attached through an oxygen atom, wherein the alkyl group is as described above. In particular embodiments, alkoxy refers to an oxygen linking group containing 1 to 10 carbon atoms ("C1-10 alkoxy"). In other embodiments, alkoxy refers to an oxygen linking group containing 1 to 8 carbon atoms ("C1-8 alkoxy"), 1 to 6 carbon atoms ("C1-6 alkoxy"), 1 to 4 carbon atoms ("C1-4 alkoxy"), or 1 to 3 carbon atoms ("C1-3 alkoxy").

The terms "cycloalkyl" and "cycloalkenyl" refer to optionally substituted saturated and unsaturated monocyclic, bicyclic or tricyclic carbon groups. Cycloalkyl or cycloalkenyl radicals may have the indicated number of carbon atoms, as appropriate, for example C3-C6Cycloalkyl or cycloalkenyl includes within its scope carbocyclic groups having 3, 4, 5 or 6 carbon atoms. Examples of such substituents may be selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl and the like. Substituted cycloalkyl or cycloalkenyl includes substitution with one or more moieties selected from: halo (e.g., Cl, F, Br, and I); halogenated alkyls (e.g., CF) 32-Br-ethyl, CH2F、CH2Cl、CH2CF3Or CF2CF3) (ii) a A hydroxyl group; an amino group; a carboxylic acid ester; a carboxamide group; an alkylamino group; an arylamino group; an alkoxy group; an aryloxy group; a nitro group; an azide group; a cyano group; thio; a sulfonic acid; a sulfate ester; a phosphonic acid; a phosphate ester; and phosphonates and those described under the definition of "optionally substituted".

The term "alkylthio" as used herein means a thio group having one or more alkyl substituents, wherein alkyl is as defined above.

The term "amino" as used herein means a compound represented by the structure NR23And include primary and secondary and tertiary amines substituted with alkyl (i.e., alkylamino). Thus, R23May represent, for example, two hydrogen atoms, two alkyl moieties or one hydrogen atom and one alkyl moiety.

The term "aryl" refers to a stable monocyclic, bicyclic, or tricyclic carbocyclic ring of up to 8 members in each ring, wherein at least one ring is aromatic as defined by the Huckel 4n +2 rule. The term includes polycyclic ring systems containing saturated carbocyclic or heteroaryl groups or heterocyclic groups, so long as at least one ring is aryl as described.

The terms "aralkyl" and "arylalkyl" as used herein mean an aryl group, as defined above, attached to the molecule through an alkyl group, as defined above.

The term "heteroaryl" refers to an aryl group containing one or more (particularly one to four) non-carbon atoms (particularly N, O or S), or combinations thereof, said heteroaryl group being optionally substituted on one or more carbon or nitrogen atoms. The heteroaryl ring may also be fused to one or more cyclic hydrocarbon, heterocyclic, aryl, or heteroaryl rings. Heteroaryl groups include, but are not limited to, 5-membered heteroaryl groups having one heteroatom (e.g., thiophene, pyrrole, furan); 5-membered heteroaryl having two heteroatoms in the 1,2 or 1,3 positions (e.g., oxazole, pyrazole, imidazole, thiazole, purine); 5-membered heteroaryl having three heteroatoms (e.g., triazole, thiadiazole); five-membered heteroaryl having four heteroatoms (e.g., tetrazole); 6-membered heteroaryl having one heteroatom (e.g., pyridine, quinoline, isoquinoline, phenanthridine, 5, 6-cycloheptene pyridine); 6-membered heteroaryl having two heteroatoms (e.g., pyridazine, cinnoline, phthalazine, pyrazine, pyrimidine, quinazoline); 6-membered heteroaryl groups having three heteroatoms (e.g., 1,3, 5-triazine) and 6-membered heteroaryl groups having four heteroatoms. "substituted heteroaryl" means a heteroaryl having one or more non-interfering groups as substituents and including those defined under "optionally substituted".

As used herein, "heterocyclyl" refers to a non-aromatic ring having from 5 to 8 atoms in the ring, and from 1 to 4 of those atoms are heteroatoms. The heterocyclic ring may also be fused to one or more cyclic hydrocarbon, heterocyclic, aryl or heteroaryl rings. Heterocycles include partially and fully saturated heterocyclic groups. The heterocyclic ring system may be attached to another moiety through any number of the carbon or heteroatoms of the group, and may be both saturated and unsaturated. Non-limiting examples of heterocycles include C4-C6Selenocycle, pyrrolidinyl, pyrrolinyl, pyranyl, piperidinyl, piperazinyl, morpholinyl, tetrahydrofuranyl, tetrahydrothienyl, pyrazolinyl, dithiacyclopentadienyl, oxathiolanyl, dioxanyl, oxazinyl, aza-cyclopentadienylRadical diazaRadical, sulfur nitrogen heteroOxygen radical and oxygen radicalRadicals and sulfur heteroAnd a group such as an imidazolinyl group and a thiomorpholinyl group.

Reference to a substituent "optionally substituted" refers to a substituent optionally substituted with one or more moieties, such as those selected from: optionally substituted C1-10 alkyl (e.g., optionally substituted C1-6 alkyl); optionally substituted C3-6 cycloalkyl (e.g., optionally substituted cyclopropyl); optionally substituted hydroxyalkyl; optionally substituted C1-10 alkoxy (e.g., optionally substituted C1-6 alkoxy); optionally substituted C2-10 alkenyl; optionally substituted C2-10 alkynyl (ii) a An optionally substituted C6-C12 aryl group; an aryloxy group; optionally substituted heteroaryl; an optionally substituted heterocyclic group; halo (e.g., Cl, F, Br, and I); a hydroxyl group; halogenated alkyls (e.g., CF)32 Br-ethyl, CH2F、CH2CF3And CF2CF3) (ii) a Amino (e.g. NH)2、NR12H and NR12R13) (ii) a An alkylamino group; an arylamino group; an acyl group; an amide group; CN; NO2;N3;CH2OH;CONH2;CONR24R25;CO2R24;CH2OR24;NHCOR24;NHCO2R24(ii) a C1-3 alkylthio; a sulfate ester; a sulfonic acid; sulfonates such as alkyl or aralkylsulfonyl including methanesulfonyl; a phosphonic acid; a phosphate ester; a phosphonate ester; mono-, di-, or tri-phosphate; trityl or monomethoxytrityl; r24SO;R24SO2;CF3S; and CF3SO2(ii) a Trialkylsilyl groups such as dimethyl-tert-butylsilyl or diphenylmethylsilyl; and R is24And R25Each independently selected from H or optionally substituted C1-10 alkyl, C1-6 alkyl or C1-4 alkyl.

Whenever a range of atomic numbers is indicated in a structure (e.g., C)1-C12、C1-C10、C1-C9、C1-C6、C1-C4Or C2-C20、C2-C12、C2-C10、C2-C9、C2-C8、C2-C6、C2-C4Alkyl, alkenyl, etc.), it is specifically contemplated that any subrange or single number of carbon atoms falling within the stated ranges can also be used. Thus, for example, 1 to 12 carbon atoms (e.g., C) are recited for use in view of any chemical group (e.g., alkyl, etc.) mentioned herein1-C12) 1 to 9 carbon atoms (e.g. C) 1-C9) 1 to 6 carbon atoms (e.g. C)1-C6) 1 to 4 carbon atoms (e.g. C)1-C4) 1 to 3 carbon atoms (e.g. C)1-C3) Or 2 to 8 carbon atoms (e.g. C)2-C8) Suitable 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and/or 12 carbon atoms and any subrange thereof are contemplated and specifically described (e.g., 1 to 2 carbon atoms, 1 to 3 carbon atoms, 1 to 4 carbon atoms, 1 to 5 carbon atoms, 1 to 6 carbon atoms, 1 to 7 carbon atoms, 1 to 8 carbon atoms, 1 to 9 carbon atoms, 1 to 10 carbon atoms, 1 to 11 carbon atoms, 1 to 12 carbon atoms, 2 to 3 carbon atoms, 2 to 4 carbon atoms, 2 to 5 carbon atoms, 2 to 6 carbon atoms, 2 to 7 carbon atoms, 2 to 8 carbon atoms, 2 to 9 carbon atoms, 2 to 10 carbon atoms, 2 to 11 carbon atoms, 2 to 12 carbon atoms, 3 to 4 carbon atoms, 3 to 5 carbon atoms, 3 to 6 carbon atoms, 3 to 7 carbon atoms, 2 to 9 carbon atoms, 2 to 10 carbon atoms, 2 to 11 carbon atoms, 2 to 12 carbon atoms, 3 to 4 carbon atoms, 3 to 5 carbon atoms, 3 to 6 carbon atoms, 3 to 7 carbon atoms, 2 to 5 carbon atoms, and any subrange thereof are suitable for each occurrence, 3 to 8 carbon atoms, 3 to 9 carbon atoms, 3 to 10 carbon atoms, 3 to 11 carbon atoms, 3 to 12 carbon atoms, 4 to 5 carbon atoms, 4 to 6 carbon atoms, 4 to 7 carbon atoms, 4 to 8 carbon atoms, 4 to 9 carbon atoms, 4 to 10 carbon atoms, 4 to 11 carbon atoms, and/or 4 to 12 carbon atoms, etc., as appropriate).

According to a first aspect of the present invention there is provided a compound of formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein W is selected from O, S and Se;

j is selected from S and Se;

R1selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted;

R2selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted; and

R1is directly bonded to J via a carbon atom, and R2Directly bonded to the adjacent nitrogen via a carbon atom.

In a preferred embodiment, W is O.

In a preferred embodiment, J is S.

In a particularly preferred embodiment, W is O and J is S.

In one embodiment, R1Is selected from C5Or C6Cycloalkyl, 5-or 6-membered heteroaryl, bicyclic heteroaryl, wherein at least one ring is heteroaryl, phenyl, biphenyl, phenylheterocyclyl, 5-or 6-membered heterocyclyl and heterocyclylcycloalkyl, all of which may be optionally substituted.

In certain embodiments, W is O, J is S, and R1Selected from the group consisting of pyrazole, furan, tetrahydrofuran, tetrahydropyran, pyran, pyrrolidine, pyrrole, triazole, tetrazole, imidazole, pyridine, morpholine, piperazine, piperidine, substituted phenyl, phenyl heteroaryl, phenyl heterocyclyl, biphenyl, quinoline, isoquinoline, naphthyl, pyrazine and pyrimidine, all of which may be optionally substituted as appropriate.

In one embodiment, when W is O, J is S, and R1When it is 2-furan or 2-thiophene, it is selected from unsubstituted 2-furan or 2, 5-substituted furan and unsubstituted 2-thiophene or 2, 5-substituted thiophene.

In one embodiment, when W is O, J is S, and R1In the case of a 2, 5-substituted furan or a 2, 5-substituted thiophene, the 2, 5-substituted furan or 2, 5-substituted thiophene is not substituted with a tertiary alcohol group.

In certain embodiments, it is found that when R1Is an unsubstituted furan, it has a level of crossing the blood-brain barrier about 10 times higher than the sulfonylurea CRID3 of the prior art.

In the above embodiments, reference to 2, 5-substitution does not exclude the presence of other substitutions on the ring, but merely indicates that the numbered substitution must be present. For example, 2,4, 5-substitution is considered within the scope of such terms.

Reference to 2-furan and 2-thiophene means that the ring is attached to the sulfonyl sulfide at the 2-ring position as shown below:

in one embodiment, R1Is a 5-membered heterocyclyl or heteroaryl, each of which may be optionally substituted, containing at least one, preferably at least two ring heteroatoms selected from N, O and S.

In certain embodiments, R1Is azaheterocyclyl or azaaryl, each of which may be optionally substituted.

In one embodiment, R1Is a 5-membered azaheterocyclyl or 5-membered azaaryl, each of which may be optionally substituted.

In one embodiment, R1Is a 5-membered heterocyclyl or 5-membered heteroaryl, each comprising at least two ring nitrogen atoms and each of said rings may be optionally substituted.

In one embodiment, W is O, J is S, and R1Selected from the group consisting of quinoline, isoquinoline, naphthyl, pyrazine, tetrazole, imidazole, pyrrolidine, pyrrole, tetrahydropyran, pyran, piperidine, piperazine, pyrazole, pyridine, pyrimidine, and triazole, each of which may be optionally substituted.

In one embodiment, R1And/or R2May comprise a selenium ring.

In one embodiment, R2Can be selected from bicyclic and tricyclic hydrocarbons, 5-, 6-and 7-membered heterocycles or heteroaryls, each of which rings can be optionally substituted, and substituted phenyl.

Suitably, the tricyclic hydrocarbon may be indacene.

In one embodiment, R2May be selected from 5-, 6-or 7-membered nitrogen heterocycles, 6-membered nitrogen heteroaryl and aryl fused to a cycloalkyl ring.

In one embodiment of the compounds of formula (I), W is O, J is S, and R is1Can be selected from:

and

and for these R1Each of the radicals R 2Can be independently selected from:

and

in any embodiment of the first aspect, when J is S, W is O, and is identical to any R described above1When the radicals are combined, R2Can be selected from:

wherein Y is independently selected for each occurrence from C, N, S and O, and may be optionally substituted where appropriate;

R5、R11、R12、R13、R14and R15Independently selected from: hydrogen, halo, cyano, amide, sulfonamide, acyl, hydroxy, C1-C6Alkyl radical, C1-C6Haloalkyl, C3-C5Cycloalkyl and C1-C6Alkoxy, all of which may be substituted, where appropriate, by halo, cyano orC1-C6Alkoxy is optionally substituted; and

wherein R is11And R12Can be combined to form a phenyl, 5-or 6-membered oxa-ring or 5-or 6-membered aza-ring, each of which can be optionally substituted;

R12and R13Can be combined to form a 5-or 6-membered azaaryl group, which can be optionally substituted; and

R14and R15Can be combined to form a 5-or 6-membered cycloalkyl ring, phenyl, a 5-or 6-membered oxa ring, or a 5-or 6-membered azaaryl, all of which can be optionally substituted.

Suitably, each occurrence of Y is carbon, R5Is H or halo.

In one embodiment, R12And R14Is hydrogen, R11And R15Is C1-C6Alkyl, and R13Is H or halo.

Preferably, R2Selected from: substituted or hydrogenated indacenes, 2, 6-dialkylphenyl, 2, 6-dialkyl-4-halophenyl, 2, 6-dicycloalkylphenyl, and 2, 6-dicycloalkyl-4-halophenyl.

In certain preferred embodiments, and with any of the formulae for the first aspect1Combination of radicals R2Selected from hexahydroindacene, 2, 6-diisopropylphenyl, 2, 6-diisopropyl-4-chlorophenyl, 2, 6-dicyclopropylphenyl and 2, 6-dicyclopropyl-4-chlorophenyl.

In one embodiment, W is O and J is S, R1Is heteroaryl, and R2Is that

Wherein each Y is CH, and R5Is H or halo, preferably R5Is H.

In one embodiment, W is O and J is S, R1Is heteroaryl, and R2Is that

Wherein

R11And R15Is C1-6Alkyl, preferably isopropyl;

R12and R14Is a compound of formula (I) wherein the compound is H,

R13is H or halo, preferably H or Cl.

In one embodiment, W is O and J is S, R1Is heteroaryl, and R2Is that

Wherein R is11And R15Is isopropyl, R12And R14Is H, and R13Is H or Cl.

In certain embodiments, the compound of formula (I) may be selected from compounds of formulae (Ia), (Ib), and (Ic), or pharmaceutically acceptable salts, solvates, or prodrugs thereof:

wherein R is1As previously described for any of the embodiments of formula (I).

In one embodiment of the compounds of formulae (Ia), (Ib) and (Ic), R1Selected from the group consisting of pyrazole, furan, tetrahydrofuran, tetrahydropyran, pyran, pyrrolidine, pyrrole, triazole, tetrazole, imidazole, pyridine, morpholine, piperazine, piperidine, substituted phenyl, phenyl heteroaryl, phenyl heterocyclyl, biphenyl, quinoline, isoquinoline, naphthyl, pyrazine and pyrimidine, all of which may be optionally substituted as appropriate.

In one embodiment of the compounds of formulae (Ia), (Ib) and (Ic), R1Selected from:

and

in one embodiment, the compound of formula (I) may be selected from compounds of formula (II), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein A, B, D and E are independently selected from C, N, O, S and Se, but at least one of which is C;

each dashed line may represent a bond;

R2as previously defined for any embodiment of formula (I), (Ia), (Ib) or (Ic), or may be a fluorophore;

R6independently at each occurrence, selected from hydrogen, halo, cyano, C1-C6Alkyl radical, C1-C6Alkylamino radical, C1-C6Alkyl hydroxy, C3-C6Cycloalkyl, alkylphenyl, phenyl, benzyl, C1-C6Esters, C2-C6Alkenyl radical, C1-C6Trifluoroalkyl and C1-C6Alkoxy, each of which may be optionally substituted, or R6May be a fluorophore.

In a preferred embodiment of the compound of formula (II), at least one of A, B, D and E is N (i.e., nitrogen).

In another preferred embodiment of the compound of formula (II), at least two of A, B, D and E are N.

In one embodiment of the compound of formula (II), A, B, D and E are selected from N and C.

In another embodiment of the compounds of formula (II), at least two of a is C, B, D and E are N.

In one embodiment, A, B, D and E form a ring selected from pyrazole, imidazole, triazole and tetrazole.

Preferably A, B, D and E form a ring selected from pyrazole or imidazole rings, most preferably a pyrazole ring.

In one embodiment, A, B, D and E and/or R2May contain a selenium ring.

In one embodiment, the compound of formula (I) may be selected from compounds of formula (IIa), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein R is11、R12、R13、R14And R15As defined hereinbefore;

A. b, D and E are selected from N and C, and at least two of A, B, D and E are N;

R6independently at each occurrence, selected from hydrogen, halide, cyano, C1-C6Alkyl radical, C1-C6Alkylamino radical, C1-C6Alkyl hydroxy, C3-C6Cycloalkyl, alkylphenyl, phenyl, benzyl, C1-C6Esters, C2-C6Alkenyl radical, C1-C6Trifluoroalkyl and C1-C6Alkoxy groups, each of which may be optionally substituted.

In one embodiment, the compound of formula (I) may be selected from compounds of formula (IIb), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein Y and R5As defined hereinbefore;

A. b, D and E are selected from N and C, and at least two of A, B, D and E are N;

R6independently at each occurrence, selected from hydrogen, halide, cyano, C1-C6Alkyl radical, C 1-C6Alkylamino radical, C1-C6Alkyl hydroxy, C3-C6Cycloalkyl, alkylphenyl, phenyl, benzyl, C1-C6Esters, C2-C6Alkenyl radical, C1-C6Trifluoroalkyl and C1-C6Alkoxy groups, each of which may be optionally substituted.

In one embodiment, the compound of formula (II) is selected from:

wherein R is40Selected from H, alkyl and halo;

R41selected from H and alkyl;

p is independently at each occurrence selected from C, O or S; and

wherein R is6Each occurrence, when present, is independently selected from those defined for formula (II).

It will be understood that R extends from the center of each ring6The moiety may represent a group bonded to a ring carbon atom or a ring heteroatom as appropriate in view of the valence state, or may be absent.

In one embodiment of formula (II), R6Is C1-C6Alkyl or C1-C6An alkyl hydroxy group.

In certain embodiments of the compounds of formula (II), for example, when R2Is hexahydro indacene and R1When it is furan, R6May not be a tertiary alcohol substituent.

In one embodiment, the compound of the first aspect may be selected from compounds of formula (IIIa), (IIIb) or (IIIc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein R is21Selected from H, alkyl, perhaloalkyl or hydroxyalkyl;

R22selected from H, alkyl, perhaloalkyl, C 3-C6Cycloalkyl, phenyl or benzyl;

R18is H or halogen;

R16and R17Is hydrogen or alkyl; or R16And R17Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

R19and R20Is H or alkyl; or R19And R20Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

provided that R is21And R22Are not all H; and

provided that R is16、R17、R18、R19And R20Not all are H.

In a preferred embodiment of the compounds of formulae (IIIa), (IIIb) and (IIIc):

R21selected from H, alkyl, perhaloalkyl or hydroxyalkyl; preferably C1-6Perhaloalkyl or hydroxyalkyl;

R22selected from H, alkyl, perhaloalkyl, C3-C6Cycloalkyl, phenyl or benzyl;

R16and R17Together with the atoms to which they are attached form a cyclopentyl ring;

R19and R20Together with the atoms to which they are attached form a cyclopentyl ring;

R18is H or halogen, preferably R18Is H; and is

Provided that R is21And R22Not all are H.

In another preferred embodiment of the compounds of formulae (IIIa), (IIIb) and (IIIc):

R21Selected from H, alkyl, perhaloalkyl or hydroxyalkyl; preferably C1-6Perhaloalkyl or hydroxyalkyl;

R22selected from H, alkyl, perhaloalkyl, C3-C6Cycloalkyl, phenyl and benzyl;

R16and R20Is C1-6Alkyl, preferably isopropyl;

R17and R19Is a compound of formula (I) wherein the compound is H,

R18is H or halogen; preferably R18Is H or Cl; and is

Provided that R is21And R22Not all are H.

In one embodiment, the compound of the first aspect may be selected from compounds of formula (IVa), (IVb) or (IVc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein R is21And R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl, or R21And R22Together with the carbon atom to which they are attached may form a cyclopentyl or cyclohexyl ring;

R18is H or halogen;

R16and R17Is H or alkyl; or R16And R17Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

R19and R20Is H or alkyl; or R19And R20Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

Provided that R is21And R22Are not all H; and

provided that R is16、R17、R18、R19And R20Not all are H.

In a preferred embodiment of the compounds of formulae (IVa), (IVb) and (IVc):

R21and R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl; preferably, the perhaloalkyl and hydroxyalkyl groups are C1-6Perhaloalkyl and hydroxyalkyl;

R16and R17Together with the atoms to which they are attached form a cyclopentyl ring;

R19and R20Together with the atoms to which they are attached form a cyclopentyl ring;

R18is H or halogen; preferably R18Is H; and is

Provided that R is21And R22Not all are H.

In another preferred embodiment of the compounds of formulae (IVa), (IVb) and (IVc):

R21and R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl; preferably, the perhaloalkyl and hydroxyalkyl groups are C1-6Perhaloalkyl and hydroxyalkyl;

R16and R20Is C1-6Alkyl, preferably isopropyl;

R17and R19Is a compound of formula (I) wherein the compound is H,

R18is H or halogen; preferably R18Is H or Cl;

provided that R is21And R22Not all are H.

In one embodiment, the compound of the first aspect may be selected from compounds of formula (Va), (Vb) or (Vc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

Wherein R is21And R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl;

R18is H or halogen;

R16and R17Is H or alkyl; or R16And R17Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

R19and R20Is H or alkyl; or R19And R20Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

provided that R is21And R22Are not all H; and is

Provided that R is16、R17、R18、R19And R20Not all are H.

In a preferred embodiment of the compounds of formulae (Va), (Vb) and (Vc):

R21and R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl; preferably, the perhaloalkyl and hydroxyalkyl groups are C1-6Perhaloalkyl and hydroxyalkyl;

R16and R17Together with the atoms to which they are attached form a cyclopentyl ring;

R19and R20Together with the atoms to which they are attached form a cyclopentyl ring;

R18is H or halogen; preferably R18Is H; and is

Provided that R is21And R22Not all are H.

In another preferred embodiment of the compounds of formulae (Va), (Vb) and (Vc):

R21and R22Selected from H, alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl; preferably, the perhaloalkyl and hydroxyalkyl groups are C1-6Perhaloalkyl and hydroxyalkyl;

R16and R20Is C1-6Alkyl, preferably isopropyl;

R17and R19Is H;

R18is H or halogen; preferably R18Is H or Cl; and is

Provided that R is21And R22Not all are H.

In one embodiment, the compound of the first aspect may be selected from compounds of formula (VIa) or (VIb), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein R is22Selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl;

R18is H or halogen;

R16and R17Is H or alkyl; or R16And R17Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

R19and R20Is H or alkyl; or R19And R20Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S; and is

Provided that R is16、R17、R18、R19And R20Not all are H.

In a preferred embodiment of the compounds of formulae (VIa) and (VIb):

R22selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl; preferably, the perhaloalkyl and hydroxyalkyl groups are C1-6Perhaloalkyl and hydroxyalkyl;

R16and R17Together with the atoms to which they are attached form a cyclopentyl ring;

R19and R20Together with the atoms to which they are attached form a cyclopentyl ring; and

R18is H or halogen; preferably R18Is H.

In another preferred embodiment of the compounds of formulae (VIa) and (VIb):

R22selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl, phenyl and benzyl; preferably, the perhaloalkyl and hydroxyalkyl groups are C1-6Perhaloalkyl and hydroxyalkyl;

R16and R20Is C1-6Alkyl, preferably isopropyl;

R17and R19Is H; and

R18is H or halogenA peptide; preferably R18Is H or Cl.

In one embodiment, the compound of the first aspect may be selected from compounds of formula (VII), or a pharmaceutically acceptable salt, solvate or prodrug thereof:

wherein Q is O or S;

R30independently at each occurrence, selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl and alkylamino.

R18Is H or halogen;

R16And R17Is H or alkyl; or R16And R17Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

R19and R20Is H or alkyl; or R19And R20Together with the carbon atom to which they are attached form a 5-or 6-membered ring, which ring is saturated, partially unsaturated or unsaturated, which optionally contains one or two heteroatoms selected from N, O and S;

provided that R is16、R17、R18、R19And R20Are not all H; and is

Provided that when Q is O, and R16And R17And R19And R20When taken alone together with each carbon atom to which they are attached form a cyclopentyl ring, then R30Is not C-3A hydroxyalkyl group.

In a preferred embodiment of the compound of formula (VII):

q is O or S;

R30independently at each occurrence, selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl and alkylamino; preferably C1-6Alkyl, perhaloalkyl, hydroxyalkyl and alkylamino;

R16and R17Together with the atoms to which they are attached form a cyclopentyl ring;

R19and R20Together with the atoms to which they are attached form a cyclopentyl ring; and

R18is H or halogen halo; preferably R 18Is H and

provided that when Q is O, then R30Is not C-3A hydroxyalkyl group.

In another preferred embodiment of the compound of formula (VII):

q is O or S;

R30independently at each occurrence, selected from alkyl, perhaloalkyl, hydroxyalkyl, C3-C6Cycloalkyl and alkylamino; preferably C1-6Alkyl, perhaloalkyl, hydroxyalkyl and alkylamino;

R16and R20Is C1-6Alkyl, preferably isopropyl;

R17and R19Is H; and

R18is H or halogen; preferably R18Is H or Cl.

The compounds of the first aspect, in particular the compounds of formulae (II) to (VI), provide a series of unexpected benefits over those of the prior art, which benefits may be selected from: improved microsomal stability; improved permeability; reduced Pgp adverse factor (liability); reduced plasma protein binding; an extended half-life; improved oral bioavailability; improved AUC; an improved Cmax; reduced Cyp inhibition; improved inhibition of NLRP3 inflammasome activation; and improved solubility. In particular, solubility and some other improvements can be seen in aqueous environments.

In one embodiment, the compound of the first aspect provides improved pharmacokinetic profiles. The known half-life of the sulfonylurea CRID3 is 3.2 hours (mouse), which may result in significant trough levels relative to QD or BD administration when t1/2 is extrapolated to humans. The compounds of the first aspect may differ in, for example, their protein binding, metabolism and oral availability.

In particular, it has been found that the compounds of the first aspect, especially those in which A, B, D and E form a 5-membered azaaryl group (e.g. a pyrazole ring), are less metabolically stable and/or have improved pharmacokinetic properties relative to other structurally similar furans and thiophenes found in the prior art.

In one embodiment, the compound of the first aspect has a structure of less thanThe tPSA of (1).

One advantage of the compounds of the present invention of the first aspect is that they can show a significantly reduced polar surface area compared to prior art sulfonylureas (e.g. CRID 3).

In another embodiment, the compound of the first aspect has a chemical structure of less thanAnd a molecular weight of less than 405.

In some embodiments, the absence of tertiary alcohol groups increases plasma concentration and helps to reduce both MW and polar surface area, resulting in overall improvement in blood brain barrier penetration.

In any of the embodiments described for the compounds of the first aspect, including the compounds of formulas (I) through (VII), one or more hydrogens of the substituents, or optional substitutions thereon, may be deuterated.

Deuterated analogs of the compounds of the invention may exhibit increased metabolic stability due to kinetic isotope effects.

In one embodiment, the compound of the first aspect is selected from:

in certain embodiments, the compounds of the first aspect may exhibit improved properties compared to known antidiabetic drugs. Examples of such compounds may include those of the following:

and

these four compounds of formula (I) can be considered as very effective forms of the current sulfonylurea antidiabetic drugs. The IC50 data shown in the experimental section reflect this view. Known drugs are not believed to target NLRP3 to any therapeutically significant extent, so it is necessary to use very high doses to have any significant effect on the inflammasome of NLRP 3. The four compounds shown above and the other compounds of the first aspect show advantageously improved properties in significantly reducing the IC50 vs NLRP3 inflammasome and additionally have benefits not achieved by existing diabetes and other drugs associated with NLRP3 inhibition, e.g. improved wound healing and other advantages described herein.

In any one or more embodiments of the first aspect, and in relation to any one or more compounds of formulae (I) to (VII), the compounds are inhibitors of activation of the NLRP3 inflammasome.

It will therefore be appreciated that the present invention provides sulphonylureas and related drugs which show significantly lower NLRP3 IC than the above mentioned comparative compounds in a cell-based assay using HMDM (see protocol in experimental section)50The value is obtained. The currently known diabetes drugs are not effective in inhibiting NLRP3 inflammasome at therapeutic dosesAgents, and achieving any such inhibition would require administration outside of the recommended levels. The compounds of the invention allow the use of lower doses, thus limiting the risk of toxic effects.

In another embodiment, one or more compounds of the first aspect may be used as a photoconvertible (photoswitchable) compound, which may be applied in a range of uses including, but not limited to, insulin release. In one embodiment, such compounds may be selected from:

wherein R is2As defined in any one or more embodiments of the compounds of formulae (I) to (VII) above.

In certain embodiments of the invention, one or more compounds of the first aspect may be suitable for use as probes, for example photoaffinity probes, or as reactive intermediates, fluorescent or photoaffinity probes that may be modified directly or via a linking moiety to generate biotinylation, including but not limited to those shown below:

Wherein R is2As defined in any one or more embodiments described for formulas (I) through (VII).

In particular, compounds such as probes or reactive intermediates may be selected from those such as:

it will be appreciated that the compounds of the first aspect may be modified or derivatised by means well known in the art to allow attachment to molecules such as biotin or fluorophores or photoaffinity labels, as shown for certain of the compounds above.

In one embodimentWherein the compound of formula (I) or (II) does not comprise a moiety selected from the group consisting of a sulfonyl moiety (i.e., as R)1Group) the structure of the following groups shown below:

in one embodiment, wherein the compounds of the first aspect, including any of the compounds of formulae (I) to (VII), have J as S, W as O, and R2Selected from hexahydroindacene, 2, 6-diisopropylphenyl and 2, 6-diisopropyl-4-chlorophenyl, then R is1Is not one of 2, 4-disubstituted furan, 2, 4-disubstituted thiophene, 2, 5-disubstituted furan, and 2, 5-disubstituted thiophene.

In one embodiment, wherein the compounds of the first aspect, including any of the compounds of formulas (I) to (VII), have J as S, have W as O, and R1Selected from the group consisting of substituted triazoles, thiadiazoles, 4-substituted pyridines and 1, 2-disubstituted imidazoles, then R 2Is not unsubstituted phenyl, 2-or 4-chlorophenyl or 3, 4-substituted phenyl, substituted with one or more of halo, trifluoromethyl, nitro or thiomethyl.

In one embodiment, wherein the compounds of the first aspect, including any of the compounds of formulas (I) to (VII), have J as S, have W as O, and R1Selected from the group consisting of substituted triazoles, thiadiazoles, benzothiazoles, and substituted pyrimidines, then R2Is not thiophene, 3-chlorophenyl, 4-ethoxyphenyl, substituted benzimidazole or substituted benzothiazole.

In one embodiment, wherein the compounds of the first aspect, including any of the compounds of formulas (I) to (VII), have J as S, have W as O, and R1Is benzothiazole substituted by ethoxy, then R2Is not 2, 6-diisopropylphenyl.

In one embodiment, wherein the compounds of the first aspect, including any of the compounds of formulas (I) to (VII), have J as S, have W as O, and R1Selected from benzofuran, benzothiophene and indole, then R2Phenyl which is not 3-or 3, 4-halo, methyl, ethyl or trifluoromethyl substituted.

In one embodiment, wherein the compounds of the first aspect, including any of the compounds of formulas (I) to (VII), have J as S, have W as O, and R 2Is a substituted pyrimidine, then R1Pyrazoles not substituted by ester or carboxyl groups.

In one embodiment, wherein the compound of the first aspect, including any compound of formulas (I) through (VII), has J as S, has W as O, then R directly bonded to the urea nitrogen2Is not a carbonyl carbon.

In one embodiment, wherein the compound of the first aspect, including any compound of formulas (I) through (VII), has J as S, has W as O, then R directly bonded to the urea nitrogen2Is an aryl, heteroaryl or heterocyclic carbon.

In one embodiment, wherein the compounds of the first aspect, including any of the compounds of formulas (I) to (VII), have J as S, have W as O, and R2Is substituted phenyl, R1Is pyrazole, then R1The pyrazoles are not substituted by aryl or heteroaryl groups.

In one embodiment, wherein the compounds of the first aspect, including any of the compounds of formulas (I) to (VII), have J as S, have W as O, and R1Is pyrazole, and the sulfonylurea linker is branched at its 4-position, pyrazole is not fused with a 6-membered heterocyclic ring at the 1-and 5-positions to form pyrazolopyrimidine derivatives.

In one embodiment, the compounds of the first aspect, including any compounds of formulae (I) to (VII), are not compounds selected from the group consisting of:

1- (4-chloro-2, 6-diisopropyl-phenyl) -3- [3- (1-hydroxy-1-methyl-ethyl) -benzenesulfonyl ] -urea;

1- (1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) -3- [4- (1-hydroxy-1-methyl-ethyl) -furan-2-sulfonyl ] -urea;

1- (1,2,3,5,6, 7-hexahydro-4-aza-s-indacen-8-yl) -3- [4- (1-hydroxy-1-methyl-ethyl) -furan-2-sulfonyl ] -urea;

1- (1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) -3- [4- (1-hydroxy-1-methyl-ethyl) -thiophene-2-sulfonyl ] -urea;

1- (4- [1,3] dioxolan-2-yl-furan-2-sulfonyl) -3- (1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) -urea;

1- (2, 6-diisopropyl-phenyl) -3- [4- (1-hydroxy-1-methyl-ethyl) -furan-2-sulfonyl ] -urea;

1- (2, 6-diisopropyl-phenyl) -3- [4- (1-hydroxy-1-methyl-ethyl) -thiophene-2-sulfonyl ] -urea;

1- (4-acetyl-thiophene-2-sulfonyl) -3- (1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) -urea;

1- (1H-benzoimidazole-5-sulfonyl) -3- (1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) -urea;

1- (1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) -3- [4- (1-hydroxy-1-methyl-ethyl) -thiophene-2-sulfonyl ] -urea;

1- (8-chloro-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) -3- [4- (1-hydroxy-1-methyl-ethyl) -furan-2-sulfonyl ] -urea;

1- (4-acetyl-furan-2-sulfonyl) -3- (1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) -urea;

1- (8-fluoro-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) -3- [4- (1-hydroxy-1-methyl-ethyl) -furan-2-sulfonyl-urea;

1- (4-fluoro-2, 6-diisopropyl-phenyl) -3- [3- (1-hydroxy-1-methyl-ethyl) -benzenesulfonyl ] -urea; and

1- (6-fluoro-1H-benzoimidazole-5-sulfonyl) -3- (1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) -urea;

1- (4-chloro-2, 6-diisopropyl-phenyl) -3- (1H-indole-6-sulfonyl) -urea;

1- (4-chloro-2, 6-diisopropyl-phenyl) -3- (5-fluoro-1H-indole-6-sulfonyl) -urea;

1- [1,2,3,5,6, 7-hexahydro-s-indacen-u-yl) -3- (1H-indole-6-sulfonyl) -urea;

1- (5-fluoro-1H-indole-6-sulfonyl) -3- (1,2,3,5,6, 7-hexahydro-5-indacen-4-yl) -urea;

1- [ 4-chloro-2, 6-diisopropyl-phenyl ] -3- [ 2-fluoro-5- (2-methyl- (1,3) dioxolan-2-yl) -benzenesulfonyl ] -urea;

3- [3- [ 4-chloro-2, 6-diisopropyl-phenyl ] -ureidosulfonyl ] -N-methyl-benzenesulfonamide;

1- [ 2-fluoro-5- (2-methyl- (1,3) dioxolan-2-yl) benzenesulfonyl ] -3-1,2,3,5,6, 7-hexahydro-indacen-4-yl) -urea;

3- [3- (1,2,3,5,6, 7-hexahydro-S-indacen-4-yl) -ureidosulfonyl ] -N-methyl-benzenesulfonamide;

4- (1-hydroxy-1-methyl-ethyl) -furan-2-sulfonamide.

In some embodiments of the invention, there are provided therapeutically inactive prodrugs of the compounds of the first aspect. Prodrugs are compounds that are converted, in whole or in part, to the compounds of the present invention when administered to a mammal. In most embodiments, prodrugs are pharmacologically inert chemical derivatives that can be converted in vivo to active drug molecules to exert a therapeutic effect. Any of the compounds described herein can be administered as prodrugs to improve the activity, bioavailability, or stability of the compounds, or to otherwise alter the properties of the compounds. Typical examples of prodrugs include compounds having a biologically labile protecting group on a functional moiety of the active compound. Prodrugs include, but are not limited to, compounds that can be oxidized, reduced, aminated, desamino, hydroxylated, dehydroxylated, hydrolyzed, dehydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated, and/or dephosphorylated to yield the active compound.

Many prodrug ligands are known. Typically, alkylation, acylation, or other lipophilic modification of one or more heteroatoms (e.g., free amine or carboxylic acid residues) of a compound can reduce polarity and allow the compound to enter a cell. Examples of substituents that may replace one or more hydrogen atoms on the free amine and/or carboxylic acid moiety include, but are not limited to, the following: an aryl group; a steroid; carbohydrates (including sugars), 1, 2-diacylglycerols; an alcohol; acyl (including lower acyl); alkyl (including lower alkyl); sulfonates (including alkyl or arylalkylsulfonyl groups, such as methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents, as provided in the aryl definition given herein); optionally substituted arylsulfonyl; lipids (including phospholipids); phosphatidylcholine; phosphorylcholine; an amino acid residue or derivative; an amino acid acyl residue or derivative; a peptide; cholesterol; or other pharmaceutically acceptable leaving group which provides the free amine upon in vivo administration. Any of these moieties can be used in combination with the disclosed active agents to achieve the desired effect.

In some embodiments, compounds having one or more chiral centers are provided. While racemic mixtures of the compounds of the present invention can be active, selective, and bioavailable, isolated isomers are also of interest.

The compound of the first aspect may contain a chiral centre, which may be in the (R) or (S) configuration, or it may comprise a mixture thereof. Thus, the present invention also includes stereoisomers of the compounds described herein, which may be mixed individually or in any proportion, as applicable. Stereoisomers may include, but are not limited to, enantiomers, diastereomers, racemic mixtures, and combinations thereof. Such stereoisomers may be prepared and isolated by reacting the enantiomeric starting materials, or by isolating isomers of the compounds and prodrugs of the invention, using conventional techniques. Isomers may include geometric isomers. Examples of geometric isomers include, but are not limited to, cis-isomers or trans-isomers at the double bond. The compounds of the invention encompass other isomers. Isomers may be used in pure form or in admixture with other isomers of the compounds described herein.

Various methods are known in the art for preparing optically active forms and measuring activity. Such methods include standard assays as described herein and other similar assays known in the art. Examples of methods that can be used to obtain optical isomers of the compounds of the present invention include the following:

i) Physical separation of crystals, wherein macroscopic crystals of the individual enantiomers are separated manually. This technique can be used in particular when there are crystals of the separated enantiomers (i.e. the material is conglomerate) and the crystals are visually distinct;

ii) simultaneous crystallization, wherein the individual enantiomers are crystallized separately from the solution of the racemate, only the latter being possible as solid aggregates;

iii) enzymatic resolution, in which the racemates are partially or completely separated due to the different reaction rates of the enantiomers with the enzyme;

iv) enzymatic asymmetric synthesis, a synthetic technique in which at least one step of the synthesis utilizes an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer;

v) chemically asymmetric synthesis, where the desired enantiomer is synthesized from an achiral precursor under conditions that produce asymmetry (i.e., chirality) in the product, which can be achieved using a chiral catalyst or chiral auxiliary;

vi) diastereoisomeric separation, in which the racemic compound is reacted with an enantiomerically pure reagent (chiral auxiliary) which converts the individual enantiomers into diastereomers. The resulting diastereomers are then separated by chromatography or crystallization, whereby they now show a more pronounced structural difference, followed by removal of the chiral auxiliary to obtain the desired enantiomer;

vii) first and second order asymmetric transformations, in which the diastereomer from the racemate is balanced to give an advantage in the diastereomer solution from the desired enantiomer, or preferential crystallization of the diastereomer from the desired enantiomer disturbs the balance so that ultimately in principle all the material is converted from the desired enantiomer to the crystalline diastereomer. The desired enantiomer is then released from the diastereomer;

viii) kinetic resolution, including partial or complete resolution of the racemate (or further resolution of partially resolved compounds) by virtue of unequal reaction rates of the enantiomers with chiral non-racemic reagents or catalysts under kinetic conditions;

ix) enantiomer-specific synthesis from non-racemic precursors, wherein the desired enantiomer is obtained from achiral starting materials, wherein the stereochemical integrity is not impaired or is only impaired to a minimal extent during the synthesis;

x) chiral liquid chromatography, in which the enantiomers of the racemate are separated in a liquid mobile phase by virtue of their different interactions with a stationary phase. The stationary phase may be made of chiral material, or the mobile phase may contain additional chiral material to initiate different interactions;

xi) chiral gas chromatography, in which the racemate is volatilized and the enantiomers are separated by means of different interactions in a gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase;

xii) extraction with a chiral solvent, wherein the enantiomers are separated by preferential dissolution of one enantiomer into a particular chiral solvent; and

xiii) transport through a chiral membrane, wherein the racemate is brought into contact with a thin membrane barrier. Barriers typically separate two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential results in preferential transport across the membrane barrier. The separation occurs as a result of the non-racemic chiral nature of the membrane, which allows only one enantiomer of the racemate to pass through.

The compounds optionally may be provided in enantiomerically enriched compositions, for example mixtures of enantiomers in which one enantiomer is present in an excess to an extent of, in particular, 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more, including 100%.

The terms (R), (S), (R, R), (S, S), (R, S) and (S, R) as used herein indicate that the composition contains a greater proportion of the named compound isomer relative to the other isomers. In a preferred embodiment, these terms mean that the composition contains at least 90% by weight of the named isomer and 10% or less by weight of one or more other isomers; or more preferably about 95% by weight of the named isomer and 5% or less by weight of one or more other isomers. In some embodiments, the composition may contain at least 99% by weight of the named isomer and 1% or less by weight of the one or more other isomers, or may contain 100% by weight of the named isomer and 0% by weight of the one or more other isomers. These percentages are based on the total amount of the compounds of the present invention present in the composition.

The compounds of the first aspect may be used as such or in the form of pharmaceutically acceptable esters, amides, salts, solvates, prodrugs or isomers where appropriate. For example, the compounds may be provided as pharmaceutically acceptable salts. If used, salts of the pharmaceutical compounds should be pharmacologically and pharmaceutically acceptable, but non-pharmaceutically acceptable salts may be conveniently used to prepare the free active compounds or pharmaceutically acceptable salts thereof and are not excluded from the scope of the present invention. Such pharmacologically and pharmaceutically acceptable salts can be prepared by reaction of the drug with an organic or inorganic acid using standard procedures as detailed in the literature.

Examples of pharmaceutically acceptable salts of compounds useful according to the invention include acid addition salts. However, salts of non-pharmaceutically acceptable acids are useful, for example, in the preparation and purification of compounds. Suitable acid addition salts according to the present invention include organic and inorganic acids. Preferred salts include those formed from: hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, citric acid, tartaric acid, lactic acid, pyruvic acid, acetic acid, succinic acid, fumaric acid, maleic acid, oxaloacetic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, and isethionic acid. Other useful acid addition salts include propionic acid, glycolic acid, oxalic acid, malic acid, malonic acid, benzoic acid, cinnamic acid, mandelic acid, salicylic acid, and the like. Specific examples of pharmaceutically acceptable salts include, but are not limited to, sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, caprate, caprylate, acrylate, formate, isobutyrate, hexanoate, heptanoate, propionate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-1, 4-dioate, hexyne-1, 6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, sulfonate, xylenesulfonate, phenylacetate, phenylpropionate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, and the like, Phenylbutyrate, citrate, lactate, gamma-hydroxybutyrate, glycolate, tartrate, mesylate, propanesulfonate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, and mandelate.

Acid addition salts can be reconverted to the free base by treatment with a suitable base. The preparation of base salts of acid moieties that may be present on a compound or prodrug useful according to the present invention may be prepared in an analogous manner using pharmaceutically acceptable bases such as sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, triethylamine and the like.

Esters of the activator compounds of the present invention may be prepared by functionalizing hydroxyl and/or carboxyl groups that may be present within the molecular structure of the compound. Amides and prodrugs can also be prepared using techniques known to those skilled in the art. For example, amides may be prepared from esters using suitable amine reactants, or they may be prepared from acid anhydrides or acid chlorides by reaction with ammonia or lower alkylamines. In addition, esters and amides of the compounds of the present invention can be prepared by reaction with a carbonylating agent (e.g., ethyl formate, acetic anhydride, methoxyacetyl chloride, benzoyl chloride, methyl isocyanate, ethyl chloroformate, methanesulfonyl chloride) and a suitable base (e.g., 4-dimethylaminopyridine, pyridine, triethylamine, potassium carbonate) in a suitable organic solvent (e.g., tetrahydrofuran, acetone, methanol, pyridine, N-dimethylformamide) at a temperature of 0 ℃ to 60 ℃. Prodrugs are generally prepared by covalently linking moieties that result in a therapeutically inactive compound until modified by the individual's metabolic system. Examples of pharmaceutically acceptable solvates include, but are not limited to, combinations of the compounds of the invention with water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, or ethanolamine.

In the case of solid compositions, it will be appreciated that the compounds used in the process of the invention may exist in different forms. For example, these compounds may exist in stable and metastable crystalline forms and isotropic and amorphous forms, all of which are intended to be within the scope of the present invention.

If the compound useful as an active agent of the invention is a base, the desired salt can be prepared by any suitable method known in the art, including treating the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, pyranosyl acids, such as glucuronic and galacturonic acids, alpha-hydroxy acids, such as citric and tartaric acids, amino acids, such as aspartic and glutamic acids, aromatic acids, such as benzoic and cinnamic acids, sulfonic acids, such as p-toluenesulfonic or ethanesulfonic acid, and the like.

If the compound described herein as an active agent is an acid, the desired salt can be prepared by any suitable method known in the art, including treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali or alkaline earth metal hydroxide, and the like. Illustrative examples of suitable salts include organic salts derived from amino acids (e.g., glycine and arginine), ammonia, primary, secondary and tertiary amines, and cyclic amines such as piperidine, morpholine and piperazine, as well as inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.

According to a second aspect of the present invention there is provided a pharmaceutical composition comprising a compound of formulae (I) to (VII), or a pharmaceutically acceptable salt, solvate or prodrug thereof, together with a pharmaceutically acceptable carrier, diluent and/or excipient.

Suitably, the pharmaceutically acceptable carrier, diluent and/or excipient may be or include one or more of: diluents, solvents, pH buffers, binders, fillers, emulsifiers, disintegrants, polymers, lubricants, oils, fats, waxes, coatings, viscosity modifiers, glidants, and the like.

Salt forms of the compounds of the invention may be particularly useful due to their improved solubility.

In one embodiment, the pharmaceutical composition comprises a cyclodextrin.

The cyclodextrin may be selected from alpha, beta or gamma cyclodextrin.

In one embodiment, the cyclodextrin is selected from the group consisting of methyl cyclodextrin, hydroxypropyl cyclodextrin, and sulfobutyl ether cyclodextrin.

It has been found that cyclodextrins provide significant advantages in the formulation and delivery of the compounds of the present invention.

Cyclodextrin formulations such as one or more compounds of the invention having hydroxypropyl beta cyclodextrin or methyl beta-cyclodextrin can be useful in cholesterol sequestration/lowering or inhibition of nonalcoholic steatohepatitis (NASH), alcoholic liver disease, atherosclerosis, and Alzheimer's Disease (AD) by NLRP 3.

The diluent may include one or more of microcrystalline cellulose, lactose, mannitol, calcium phosphate, calcium sulfate, kaolin, dry starch, powdered sugar, and the like. The binder may include one or more of povidone, starch, stearic acid, gums, hydroxypropyl methylcellulose, and the like. Disintegrants may include one or more of starch, croscarmellose sodium, crospovidone, sodium starch glycolate, and the like. The solvent may include one or more of ethanol, methanol, isopropanol, chloroform, acetone, methyl ethyl ketone, dichloromethane, water, and the like. Lubricants may include one or more of magnesium stearate, zinc stearate, calcium stearate, stearic acid, sodium stearyl fumarate, hydrogenated vegetable oils, glyceryl behenate, and the like. The glidant may be one or more of colloidal silicon dioxide, talc, corn starch, or the like. The buffer may include, but is not limited to, a phosphate buffer, a borate buffer, and a carbonate buffer. The filler may include one or more gels including, but not limited to, gelatin, starch, and synthetic polymer gels. The coating may include one or more of a film former, solvent, plasticizer, and the like. Suitable film forming agents may be one or more of hydroxypropyl methylcellulose, methyl hydroxyethyl cellulose, ethyl cellulose, hydroxypropyl cellulose, povidone, sodium carboxymethyl cellulose, polyethylene glycol, acrylates, and the like. Suitable solvents may be one or more of water, ethanol, methanol, isopropanol, chloroform, acetone, methyl ethyl ketone, methylene chloride, and the like. The plasticizer may be one or more of propylene glycol, castor oil, glycerin, polyethylene glycol, polysorbate, and the like.

Reference is made to the Handbook of Excipients 6 th edition, Rowe, Sheskey & Quinn edition (Pharmaceutical Press) which provides non-limiting examples of Excipients which may be useful according to the present invention.

It will be appreciated that the selection of a pharmaceutically acceptable carrier, diluent and/or excipient will depend, at least in part, on the mode of administration of the formulation. By way of example only, the composition may be in the form of a tablet, capsule, caplet, powder, injection, suppository, sustained release formulation, osmotic pump formulation, or any other form that is effective and safe for administration.

Suitably, the pharmaceutical composition is for treating or preventing a disease, disorder or condition in a mammal.

A third aspect of the invention is a method of treating or preventing a disease, disorder or condition comprising the steps of: administering an effective amount of a compound of formulae (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof, or a pharmaceutical composition of the second aspect, thereby treating or preventing a disease, disorder or condition.

A fourth aspect of the present invention provides a compound of formulae (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof, or a pharmaceutical composition of the second aspect, for use in the treatment or prevention of a disease, disorder or condition.

A fifth aspect of the invention provides the use of a compound of formulae (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof, in the manufacture of a medicament for the treatment or prophylaxis of a disease, disorder or condition.

As generally used herein, the terms "administering," and the like, describe the introduction of a compound or composition into a mammal, for example, by a particular route or carrier. Routes of administration may include topical, parenteral, and enteral, including oral, buccal, sublingual, nasal, anal, gastrointestinal, subcutaneous, intramuscular, and intradermal routes of administration, but are not limited thereto.

By "treating" is meant administering a compound or composition to a subject to at least ameliorate, reduce, or inhibit an existing sign or symptom of a disease, disorder, or condition experienced by the subject.

By "preventing" or "prophylactic" is meant prophylactic administration of a formulation to a non-prophylactic subject who does not exhibit signs or symptoms of a disease, disorder or condition, but who is expected or predicted to develop such signs or symptoms. Prophylactic treatment can at least reduce or partially ameliorate the desired symptoms or signs.

As used herein, "effective amount" refers to an amount of a relevant compound or composition administered sufficient to prevent the onset of symptoms of the condition being treated, or to stop worsening of symptoms, or to treat and alleviate, or at least reduce the severity of, symptoms. The effective amount will vary in the manner of the patient's age, sex, weight, etc., as will be appreciated by those skilled in the art. Appropriate dosages or dosage regimens may be determined by routine experimentation.

The term "subject" or "individual" or "patient" as used herein may refer to any subject, in particular a vertebrate subject, even more particularly a mammalian subject in need of treatment. Suitable vertebrates include, but are not limited to, primates, birds, livestock (e.g., sheep, cattle, horses, donkeys, pigs), laboratory test animals (e.g., rabbits, mice, rats, guinea pigs, hamsters), companion animals (e.g., cats, dogs), and wild animals (e.g., foxes, deer, wild dogs). Preferred subjects are humans in need of treatment for the diseases, disorders, or conditions described herein. It should be understood, however, that the foregoing terms do not imply that symptoms are necessarily present.

In a particular embodiment, the disease, disorder or condition is one that responds to inhibition of NLRP3 inflammatory body activation.

According to this embodiment, the compound of the first aspect or a pharmaceutically effective salt, solvate or prodrug thereof is a specific inhibitor of NLRP 3.

In another embodiment, the disease, disorder or condition is responsive to modulation of one or more of IL-1 β, IL-17, IL-18, IL-1 α, IL-37, IL-33 and Th17 cells.

In one embodiment, the modulation is inhibition of one or more of IL-1 β, IL-17, IL-18, IL-1 α, IL-37, and IL-33.

In one embodiment, the modulation of Th17 cells is by inhibiting the production and/or secretion of IL-17.

In general embodiments, the disease, disorder or condition is a disease, disorder or condition of the immune system, cardiovascular system, endocrine system, gastrointestinal tract, renal system, respiratory system, central nervous system, is a cancer or other malignancy and/or is caused by or associated with a pathogen.

It is to be understood that these general embodiments, defined in terms of a broad class of diseases, conditions and disorders, are not mutually exclusive. In this regard, any particular disease, disorder or condition may be classified according to more than one of the above general embodiments. A non-limiting example is type I diabetes, which is an autoimmune disease and a disease of the endocrine system.

In one embodiment, the disease, disorder or condition is a disease, disorder or condition of the immune system. In particular embodiments, the disease condition or disorder is an inflammatory disease condition or disorder or an autoimmune disease condition or disorder.

In one embodiment, the disease, disorder or condition is a disease, disorder or condition of the skin.

In one embodiment, the disease, disorder or condition is a disease, disorder or condition of the cardiovascular system.

In one embodiment, the disease, disorder or condition is a cancer, tumor or other malignancy. Cancer tumors and malignancies as used herein refer to diseases, disorders or conditions, or cells or tissues associated with diseases, disorders or conditions, characterized by abnormal or abnormal cell proliferation, differentiation and/or migration, typically accompanied by an abnormal or abnormal molecular phenotype including one or more genetic mutations or other genetic changes associated with neoplasia, expression of tumor markers, loss of tumor suppressor gene expression or activity and/or abnormal cell surface marker expression. In general embodiments, cancers, tumors, and malignancies can include, but are not limited to, sarcomas, lymphomas, leukemias, solid tumors, blastomas, gliomas, carcinomas, melanomas, and metastatic cancers. A more comprehensive list of Cancer tumors and malignancies can be found in the National Cancer institute website http:// www.cancer.gov/Cancer/types/alphalist.

In one embodiment, the disease, disorder or condition is a disease, disorder or condition of the renal system.

In one embodiment, the disease, disorder or condition is a disease, disorder or condition of the gastrointestinal tract.

In one embodiment, the disease, disorder or condition is a disease, disorder or condition of the respiratory system.

In another embodiment, the disease, disorder or condition is a disease, disorder or condition of the endocrine system.

In one embodiment, the disease, disorder or condition is a disease, disorder or condition of the Central Nervous System (CNS).

In one embodiment, the disease, disorder or condition is caused by or associated with a pathogen. The pathogen may be a virus, bacteria, protist, worm or fungus or any other organism capable of infecting a mammal, but is not limited thereto.

Non-limiting examples of viruses include influenza Virus, cytomegalovirus, Epstein-Barr Virus (Epstein Barr Virus), Human Immunodeficiency Virus (HIV), alphaviruses such as Chikungunya Virus (Chikungunya Virus) and Ross River Virus (Ross River Virus), flaviviruses such as dengue Virus, Zika Virus (Zika Virus), and papilloma Virus, but are not limited thereto.

Non-limiting examples of pathogenic bacteria include Staphylococcus aureus (Staphylococcus aureus), Helicobacter pylori (Helicobacter pylori), Bacillus anthracis (Bacillus antrhraxis), Bordetella pertussis (Bordetella pertussis), Aspergillus niger (Corynebacterium dipentaire), Clostridium tetani (Clostridium tetani), Clostridium botulinum (Clostridium botuli), Streptococcus pneumoniae (Streptococcus pneumae), Streptococcus pyogenes (Streptococcus pyelones), Listeria monocytogenes (Listeria monocytogenes), Haemophilus influenzae (Hemophilus influenzae), Pasteurella multocida (Pasteurella multocida), Shigella dysenteriae (Shigella senteriae), Mycobacterium tuberculosis (Mycobacterium tuberculosis), Neisseria meningitidis (Mycobacterium meningitidis), Mycobacterium meningitidis (Mycobacterium tuberculosis), Mycobacterium meningitidis (Mycobacterium tuberculosis (Streptococcus pneumoniae), Mycobacterium meningitidis (Streptococcus pneumoniae (Mycobacterium meningitidis), Mycobacterium meningitidis (Mycobacterium meningitidis), Mycobacterium tuberculosis (Streptococcus pneumoniae (Mycobacterium meningitidis), Mycobacterium tuberculosis (Mycobacterium), Mycobacterium tuberculosis (Mycobacterium), and Mycobacterium tuberculosis (Mycobacterium), and Mycobacterium tuberculosis (Mycobacterium tuberculosis) in-infection), and Mycobacterium tuberculosis (Mycobacterium), and other, Klebsiella pneumoniae (Klebsiella pneumoniae), Pseudomonas aeruginosa (Pseudomonas aeruginosa), Propionibacterium acnes (Propionibacterium acnes), Treponema pallidum (Treponema pallidum), Chlamydia trachomatis (Chlamydia trachoromatis), Vibrio cholerae (Vibrio cholerae), Salmonella typhimurium (Salmonella typhimurium), Salmonella typhi (Salmonella typhi), Borrelia burgdorferi (Borrelia burgdorferi) and Yersinia pestis (Yersinia pestis), but are not limited thereto.

Non-limiting examples of protists include, but are not limited to, plasmodium, babesia, giardia, amoeba, leishmania, and trypanosoma.

Non-limiting examples of worms include intestinal worms, including schistosomes (schistosismies), roundworms, tapeworms, and trematodes, but are not limited thereto.

Non-limiting examples of fungi include, but are not limited to, the genera Candida and Aspergillus.

Other related diseases, disorders or conditions may be selected from those listed in the following journal: http:// onlineibrary. wire.com/store/10.1111/j.1365-2249.2011.04440. x/asset/j.1365-2249.2011.04440.x.pdfv ═ 1& t ═ i60c1phf & s ═ d26f50a2622926cc6b4bc855bd911ae9dc9750 cf.

In particular embodiments, the disease, disorder or condition is selected from constitutive inflammation, including cryptotropin-associated periodic syndrome (CAPS): moore-weirs syndrome (MWS), Familial Cold Autoinflammatory Syndrome (FCAS), and neonatal onset multiple system inflammatory disease (NOMID); including auto-inflammatory diseases: familial Mediterranean Fever (FMF), TNF receptor-related periodic syndrome (TRAPS), Mevalonate Kinase Deficiency (MKD), hyperimmunoglobulinemia D and periodic fever syndrome (HIDS), interleukin 1 receptor (DIRA) antagonist deficiency, magedrid syndrome (Majeed syndrome), septic arthritis, pyoderma gangrenosum and acne (PAPA), a20 haploinsufficiency (HA20), granulomatous arthritis in children (PGA), PLCG 2-associated antibody deficiency and immune disorders (plai), PLCG 2-associated autoinflammation, antibody deficiency and immune disorders (aploid), sideroblastic anemia (SIFD) with B-cell immunodeficiency, periodic fever and developmental delay; swart's syndrome, Chronic Nonbacterial Osteomyelitis (CNO), Chronic Relapsing Multifocal Osteomyelitis (CRMO) and synovitis, acne, impetigo, hypertrophic bone, osteitis Syndrome (SAPHO); autoimmune diseases, including Multiple Sclerosis (MS), type 1 diabetes, psoriasis, rheumatoid arthritis, Behcet's disease, Sjogren's syndrome, and Schnitzler syndrome; respiratory diseases including Chronic Obstructive Pulmonary Disorder (COPD), steroid-resistant asthma, asbestosis, silicosis and cystic fibrosis; central nervous system diseases including Parkinson's disease, alzheimer's disease, motor neuron disease, Huntington's disease, cerebral malaria and brain injury caused by pneumococcal meningitis; metabolic diseases including type 2 diabetes, atherosclerosis, obesity, gout, pseudogout; ocular diseases including those of the ocular epithelium, age-related macular degeneration (AMD), corneal infections, uveitis, and dry eye; renal diseases, including chronic kidney disease, oxalate kidney disease, and diabetic nephropathy; liver diseases including non-alcoholic steatohepatitis and alcoholic liver disease; inflammatory reactions of the skin, including contact hypersensitivity and sunburn; arthritic reactions including osteoarthritis, systemic juvenile idiopathic arthritis, adult onset steve's disease, relapsing polychondritis; viral infections including alphaviruses (chikungunya, ross river) and flaviviruses (dengue and zaka viruses), influenza, HIV; hidradenitis Suppurativa (HS) and other skin diseases that cause cysts; cancers including lung cancer metastasis, pancreatic cancer, gastric cancer, myelodysplastic syndrome, leukemia; polymyositis; stroke; myocardial infarction; graft versus host disease; hypertension; colitis; infection by worms; bacterial infection; abdominal aortic aneurysm; healing of the wound; depression, psychological stress; pericarditis including Dressler's syndrome, ischemia reperfusion injury, and any disease in which an individual has been determined to carry germline or somatic non-silent mutations in NLRP 3.

In one non-limiting example of those described, the disease, disorder or condition being treated is NASH. NLRP3 inflammatory body activation is central to the inflammatory recruitment of NASH, and inhibition of NLRP3 may both prevent and reverse liver fibrosis. The compounds of the invention can lead to a histological reduction of liver inflammation, a reduction in macrophage and neutrophil recruitment, and inhibition of NF- κ B activation by interfering with the function of the NLRP3 inflammasome in liver tissue. Inhibition of NLRP3 can reduce hepatic expression of pre-IL-1 β and normal hepatic and circulating IL-1 β, IL-6 and MCP-1 levels, thereby contributing to the treatment of disease.

In another non-limiting example of those described, the disease, disorder or condition being treated is severe steroid-resistant (SSR) asthma. Respiratory infection induces the NLRP3 inflammasome/caspase-1/IL-1 β signaling axis in the lungs that contribute to SSR asthma. The NLRP3 inflammasome recruits and activates pre-caspase-1 to induce an IL-1 β response. Therefore, NLRP3 inflammasome-induced IL-1 β responses are important in controlling infection, however, over-activation leads to abnormal inflammation and is associated with the pathogenesis of SSR asthma and COPD. Administration of a compound of the first aspect which targets a particular disease process is more therapeutically attractive than non-specific inhibition of the inflammatory response of steroids or IL-1 β. Thus, targeting the NLRP3 inflammasome/caspase-1/IL-1 β signaling axis with the compounds of the first aspect may be useful in the treatment of SSR asthma and other steroid-resistant inflammatory disorders.

In another non-limiting example of those described, the disease, disorder or condition being treated is Parkinson's disease. Parkinson's disease is the most common neurodegenerative dyskinesia, characterized by selective loss of dopaminergic neurons, with accumulation of misfolded α -synuclein (Syn) into the lewy body that is the pathological feature of the disease. Chronic microglial neuroinflammation is evident in early stages of the disease and has been proposed to drive pathology.

Microglia NLRP3 is postulated to play a key role in the progression of parkinson's disease. NLRP3 inflammasome is activated by the fibrillar Syn through a Syk kinase dependent mechanism and also occurs in the absence of Syn pathology at the early stages of dopaminergic degeneration and drives neuronal loss. The compounds of the first aspect may block NLRP3 inflammasome activation by fibrous Syn or mitochondrial dysfunction, thereby conferring potent neuroprotective effects on the nigrostriatal dopaminergic system and contributing to the treatment of parkinson's disease.

In a sixth aspect of the invention, there is provided a method of diagnosing a disease, disorder or condition in a mammal, comprising the steps of: administering a labeled compound of formulae (I) to (VII), or a pharmaceutically effective salt, solvate, or prodrug thereof, to a mammal or a biological sample obtained from the mammal to facilitate diagnosis of a disease, disorder, or condition in the mammal.

It is known that inflammatory body activation, particularly NLRP3 inflammatory body activation, can drive the initiation, progression, and chronic development of a number of inflammatory diseases. The sulfonylureas and related compounds of the first aspect are potent and specific direct inhibitors of NLRP 3. Therefore, chemical probes specific for NLRP3, which are present in immune cells during inflammation, have potential uses in the diagnosis of inflammatory and other related diseases. NRLP3 activation probes comprising the compounds of the first aspect may be useful as surrogate biomarkers for ex vivo (blood) or in vivo (MRI, PET, etc.) diagnosis of inflammatory diseases.

The use of compounds of the first aspect in the diagnosis of inflammatory and other related diseases (e.g., those listed above) can be characterized by near infrared fluorescence imaging and ex vivo characterization of immune cells by inhibition of IL-1 βDegree of cleavage by pro-caspase 1 and IL-18 levels. In particular, peripheral blood mononuclear cells (PMBC), macrophages, dendritic cells, CD4+T cells, Th17 cells, Th1 cells and Th2 cells are related. Magnetic Resonance Imaging (MRI) is used in vivo diagnostics. By IV, IM, SC, PO, topical, IT, etc. [ Compound classes]H2 (deuterium), 13C、19F、15The N-labeled variant is administered to the patient.

In vivo diagnostics using Positron Emission Tomography (PET) are also suitable. PET is a molecular imaging technique that requires specific probes radiolabeled with short-lived positron emitting radionuclides. Typical isotopes include11C、13N、15O、18F、64Cu、62Cu、124I、76Br、82Rb and68ga, wherein18F is the most widely used clinically. In particular, stable ion exchange with one or more compounds of formula (I) can be simply generated by simple ion exchange with a sodium (or other monovalent cation) salt of the compound64Cu or62A Cu salt. This enables rapid preparation of diagnostic probes for radioimaging, PET, etc., whereby the intensity, location and time accretion of the diagnostic probes enables identification of the extent and/or location of immune cells with activated NLRP3 as a surrogate biomarker for the inflammatory state of a patient and the site of inflammation in vivo. They can also be used for biological samples taken from the body, i.e. in vitro diagnostics.

A seventh aspect of the present invention resides in a method of modulating the activity of a biological target comprising the step of exposing the biological target to a compound of formulae (I) to (VII) or a pharmaceutically effective salt, solvate or prodrug thereof.

The biological target may be selected from the group consisting of NLRP3 inflammasome, IL-1 β, IL-17, IL-18, IL-1 α, IL-37, IL-33, and Th17 cells.

The adjustment may be as described previously for the third to fifth aspects.

As generally used herein, a biological sample may include cells, tissues, fluids, molecules, or other biological materials obtained or obtainable from a mammal. Non-limiting examples include urine, blood and fractions thereof such as serum, plasma, lymphocytes and erythrocytes, cerebrospinal fluid, PAP smears, nasal and ocular secretions, amniotic fluid, feces, semen, tissue and/or organ biopsies and nucleic acid (e.g., DNA, RNA) or protein samples, but are not limited thereto.

The following experimental section describes in more detail the characteristics of certain compounds of the invention and their efficacy. It is intended to illustrate certain embodiments of the compounds of the invention and their effectiveness, without limiting the invention in any way.

Experiment of

General synthetic method

The method A comprises the following steps:

a1: to a solution of the R2 amine intermediate (1 equivalent) in an anhydrous aprotic solvent such as but not limited to tetrahydrofuran or dichloromethane with or without a base such as but not limited to triethylamine (1.2 equivalents) is added triphosgene (0.4 to 1.1 equivalents). the reaction is stirred at ambient temperature or heated at reflux if necessary until complete, typically 2 to 18 hours.

A2: DMAP (15-100 mol%) was added to anhydrous acetonitrile or di-tert-butyl dicarbonate (1.2 to 1.4 equivalents) in THF, and after 5 minutes, a solution of the R2 amine intermediate (1.0 equivalent) in acetonitrile was added. The reaction mixture was stirred at room temperature for 30-60 minutes.

The method B comprises the following steps:

b1: the R2 carboxylic acid intermediate (1 eq) was dissolved in an aprotic solvent (e.g., toluene with or without 2 drops of DMF) and a chlorinating agent such as thionyl chloride (2 eq) was added. The reaction mixture was heated at reflux until completion, then concentrated in vacuo. The corresponding R2 acid chloride intermediate was obtained.

Alternative methods or formation of acid chlorides are also useful herein, for example the above procedure can be carried out in the absence of toluene and DMF, thereby using thionyl chloride as both the solvent and the chlorinating agent.

The R2 acid chloride intermediate was dissolved in acetone and sodium azide (1.5 equivalents) at 0 ℃ in water: acetone (50:50) solution. Ice water was added to precipitate the resulting R2 acyl azide intermediate, which was dissolved in toluene and dried (MgSO4), and then the solution was added to anhydrous toluene in a dropwise fashion while maintaining a constant inert gas flow. The reaction was heated to completion, typically for 2 hours, to give R2 isocyanate.

B2: at 0 ℃ in anhydrous CH2Cl2To R2 acid chloride (formed as shown in method B1) was added NaN3(2.0 equiv.). The reaction mixture was stirred at room temperature for 1 hour and extracted into EtOAc. Subjecting the organic layer to H2O (15mL) was washed, dried (MgSO4) and carefully evaporated to give the acyl azide. The acyl azide was dissolved in anhydrous toluene and heated to 100 ℃ for 2 hours. The solvent was removed to give crude R2 isocyanate.

The method C comprises the following steps:

c1: the R1 sulfonamide intermediate (1 eq) was dissolved in anhydrous THF and treated with NaH (1 eq) under reduced pressure. The mixture was heated at reflux for 2 hours, then cooled to room temperature and the R2 isocyanate intermediate in THF was added under a nitrogen atmosphere. The reaction mixture was stirred at reflux until completion.

C2: the R1 sulfonamide intermediate (1 eq) was dissolved in anhydrous THF or anhydrous methanol and treated with NaH (1 eq) under reduced pressure. Once effervescence ceased, R2 isocyanate intermediate was added and the reaction mixture was stirred at ambient temperature overnight.

C3: to the R1 sulfonamide intermediate (1 eq) in anhydrous THF (5mL/mmol) at 0 ℃, NaH (1 eq) was added and stirred at ambient temperature under nitrogen for 30 minutes to 2 hours, or until completion. Cooled again to 0 ℃, R2 isocyanate (1.0 eq) in THF was added and stirred at ambient temperature until completion, typically 2 to 16 hours.

C4: to crude R2 isocyanate (1.0 equiv.) in dry THF or DCM (5-11mL/mmol) was added R1 sulfonamide (1.0 equiv.) followed by a base such as triethylamine, DIPEA or DBU (1-2 equiv.) and the reaction mixture was stirred at ambient temperature overnight.

C5: to the R1 sulfonamide intermediate (1 eq) in dry MeOH (5mL/mmol) was added NaOMe (1 eq) [ alternatively, 1.0mM freshly prepared sodium methoxide solution (1 eq) was added to a solution of 1.0mM R1 sulfonamide (1 eq) in dry methanol ]. The solvent was then removed in vacuo. The salt was suspended in an anhydrous aprotic solvent such as acetonitrile or THF, R2 isocyanate (1.0 eq) in an anhydrous aprotic solvent such as acetonitrile or THF was added, and the mixture was stirred at ambient temperature overnight. The solution is then heated at reflux until completion, typically for 90 minutes.

C6: the R1 sulfonamide (1.0 equivalent) was dissolved in anhydrous THF under a nitrogen atmosphere. Solid sodium methoxide (1.0 equivalent mmol) was added in one portion. The mixture was stirred at ambient temperature for 3 hours. A solution of R2 isocyanate (1.17 equivalents) in THF was added dropwise. The reaction mixture was stirred at room temperature overnight.

The method D comprises the following steps:

using concentrated HCl (1.25-2.25mL/mmol) in H at 0 deg.C2A solution of amine (1.0 equiv.) in acetonitrile (7-12mL/mmol) was treated with O (0.5-1.2mL/mmol) and then NaNO 2(1.2 equiv.) in H2O(0.3-0.5mL/mmol NaNO2) The solution treatment of (1). The resulting solution was stirred at 0 ℃ for 45 minutes. AcOH (0.5-1.2mL/mmol), CuCl2·2H2O (0.5 equiv.) and CuCl (0.05 equiv.) are added to the mixture in succession and with SO at 0 deg.C2Gas purge for 20 minutes. The resulting reaction mixture was stirred at 0 ℃ to 10 ℃ until completion.

Method E

E1: a solution of sulfonyl chloride (1 eq) in THF (10-20mL/mmol) was cooled to-78 deg.C and ammonia gas bubbled through the solution for 15 minutes, stirring was continued for an additional 30 minutes, then allowed to warm to ambient temperature and stirred for 2 hours or until completion.

E2: at ambient temperature with NH dissolved in water4HCO3(4 equiv) (1.5mL/mmol NH)4HCO3) The solution treated a solution of sulfonyl chloride (1 eq) in acetone (20mL/mmol) and stirred for 4 hours or until completion.

E3: at 0 ℃ with NH3(3.5mL/mmol,NH4OH in H2In O, 28% NH3Base) a solution of sulfonyl chloride (1 eq) in acetone (2.5mL/mmol) was treated and stirred for 2 hours or until completion.

Method F

General procedure for the Synthesis of triazoles

Alkyne (1 equivalent) and azide (1.2 equivalents), 5 mol% CuSO4A solution of 10 mol% NaAsc in DMSO (500 μ L) was stirred at room temperature until completion, typically 12 hours.

Synthesis of R1 sulfonamide intermediate:

Cyclohexanesulfonamides

To a solution of cyclohexanesulfonyl chloride (0.1g, 0.54mmol) in acetone (1mL) at 0 deg.C was added NH3Aqueous solution (2mL, 28% NH)4OH in H2O) and the reaction mixture was stirred at room temperature for about 2 hours. The solvent was removed in vacuo and MeOH/dichloromethane (1:9) (5mL) added and the NH removed by filtration4Cl by-product, and the remaining solution was concentrated in vacuo. Chromatography on silica gel using 0.2% MeOH-CH2Cl2The crude product was purified with eluent to give the cyclohexanesulfonamide as an off-white solid (30mg, 34%).1H NMR(400MHz,DMSO-d6):δ=6.61(br s,2H),2.76-2.70(m,1H),2.09-2.04(m,2H),1.80-1.76(m,2H),1.65-1.60(m,1H),1.31-1.19(m,4H),1.16-1.06(m,1H)。

Cyclopentane sulfonamides

To a solution of cyclopentanesulfonyl chloride (0.1g, 0.59mmol) in acetone (1mL) at 0 deg.C was added NH3Aqueous solution (1mL, 28% NH)4OH in H2O) and the reaction mixture was stirred at room temperature for about 2 hours. The solvent was removed in vacuo and MeOH/dichloromethane (1:9) (5mL) added and the NH removed by filtration4Cl by-product, and the remaining solution was concentrated in vacuo. The crude product was purified by silica gel column chromatography using 35% EtOAc-hexanes eluent to give the cyclopentane sulfonamide as an off-white solid (72mg, 81%).1H NMR(400MHz,DMSO-d6):δ=6.69(br s,2H),3.42-3.32(m,1H),1.89-1.84(m,4H),1.68-1.64(m,2H),1.61-1.52(m,2H)。

5- ((dimethylamino) methyl) furan-2-sulfonamide

Furan-2-carboxylic acid (5g, 44.6mmol) was dissolved in ethanol (100mL), concentrated sulfuric acid (1.0mL) was added, and the solution was heated to reflux overnight. The reaction mixture was concentrated in vacuo, then taken up in ethyl acetate (100mL) and saturated NaHCO 3(100 mL). The organic phase was washed with water and then brine, dried (MgSO)4) And concentrated in vacuo to give ethyl furan-2-carboxylate (4.5g, 80%).1H NMR(400MHz,CDCl3):δ=7.57(d,J=1.2Hz,1H),7.18(d,J=3.5Hz,1H),6.51(dd,J=3.5,1.2Hz,1H),4.37(q,J=7.1Hz,2H),1.38(t,J=7.1Hz,3H)。

Ethyl furan-2-carboxylate (9.0g, 64.3mmol) was dissolved in dichloromethane (200mL) and chlorosulfonic acid (7.5g, 64.3mmol) was added. The reaction is stirred at ambient temperature for 6 hours, orTo completion, pyridine (5.6g, 70.7mmol) and PCl5(14.7g, 70.7mmol) were then added portionwise. The reaction mixture was stirred at ambient temperature for 16 hours, then quenched with ice water and stirred for 30 minutes. The mixture was extracted with DCM and the combined organics were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The crude product ethyl 5- (chlorosulfonyl) furan-2-carboxylate (7g, 46%) was used without further purification.1H NMR(300MHz,CDCl3)δ=7.33(d,J=3.9Hz,1H),7.27(d,J=3.9Hz,1H),4.44(q,J=7.1Hz,1H),1.42(t,J=7.1Hz,2H)。

Crude ethyl 5- (chlorosulfonyl) furan-2-carboxylate (7g) was converted by general method E1 to give ethyl 5-sulfamoylfuran-2-carboxylate (5g, 78%).1H NMR(300MHz,DMSO-d6)δ=8.05(s,2H),7.38(d,J=3.7Hz,1H),7.09(d,J=3.7Hz,1H),4.32(q,J=7.1Hz,2H),1.29(t,J=7.1Hz,3H)。

Ethyl 5-sulfamoylfuran-2-carboxylate (2g, 9.13mmol) was cooled to 0 ℃ in dry THF (40mL) and lithium aluminum hydride (1.05g, 27.3mmol) was added portionwise over a period of 30 minutes. The reaction was heated to 70 ℃ for 4 hours. The reaction was cooled to 0 ℃ and saturated NH was very carefully added dropwise over a period of 30 minutes 4Cl (20 mL). The reaction mixture was diluted with ethyl acetate (100mL) and filtered through a pad of celite. The organic phase was washed with water (100mL), brine (100mL) and dried (MgSO)4) And concentrated in vacuo to give 5- (hydroxymethyl) furan-2-sulfonamide (1.25g, 78%) as a pale brown liquid. 1H NMR (400MHz, DMSO-d6) δ 7.71(s,2H),6.88(d, J ═ 3.5Hz,1H),6.44(d, J ═ 3.4Hz,1H),5.47(s,1H),4.44(d, J ═ 5.7Hz,2H),3.36(s,7H),2.51(q, J ═ 1.8Hz,5H),1.36(s, 1H).

5- (hydroxymethyl) furan-2-sulfonamide (0.3g, 1.7mmol) in THF (5mL) was cooled to 0 deg.C and POCl was slowly added3(0.4g, 2.54 mmol). The reaction mixture was stirred at 75 ℃ for 2 hours and then cooled to ambient temperature. The crude mixture was dissolved in ethyl acetate (50mL) and saturated NaHCO3The aqueous solution (50mL) was partitioned, and the organic phase was washed with water (50mL), brine (50mL), dried (Na)2SO4) And concentrated in vacuo. Passing through a silica gel columnChromatography the crude product was purified using 30% EtOAc-hexanes eluent to give 5- (chloromethyl) furan-2-sulfonamide (0.25g, 76%) as a light brown semisolid.1H NMR(300MHz,DMSO-d6)δ=7.85(s,2H),6.93(dd,J=3.5,1.3Hz,1H),6.69(dd,J=3.5,1.3Hz,1H),4.89(d,J=1.3Hz,2H)。

5- (chloromethyl) furan-2-sulfonamide (0.4g, 2.05mmol) in THF (20mL) was cooled to 0 deg.C, concentrated HCl (7.5mg, 2.05mmol) was added, and the solution was stirred at the same temperature for 20 minutes. 5.6M N, N-dimethylamine in ethanol (0.28g, 6.15mmol, 3 equivalents) was added at 0 deg.C and the reaction tube was sealed and then stirred at room temperature overnight. The solvent was removed in vacuo and azeotroped with toluene (× 2) to give 5- ((dimethylamino) methyl) furan-2-sulfonamide (0.25g, 60%) as a gum. The crude product was used directly without further purification.

Furan-2-sulfonamides

Furan-2-sulfonyl chloride (0.30g, 1.8mmol) was added to aqueous ammonia (1.0mL) at 0 ℃ and the mixture was stirred at ambient temperature for 1 hour. After completion of the reaction, excess ammonia was removed in vacuo. The residue was azeotroped with isopropanol and triturated with pentane to give the title compound as a light brown solid (0.21g, 79%).1H NMR(400MHz,DMSO-d6):δ=7.91(s,1H),7.45(br.s.,2H),6.95(d,J=3.6Hz,1H),6.63(dd,J=2.8,1.6Hz,H)。LC-MS 97.4%(ELSD);m/z 146.11[M-H]+

5-methylfuran-2-sulfonamide

To a solution of 2-methylfuran (2.0g, 24.3mmol) in anhydrous acetonitrile (4mL) was added SO3Py complex (5.0g, 31.6mmol) and the reaction mixture was heated at 40 ℃ under nitrogen overnight. The reaction mixture was diluted with EtOAc (5mL) and stirred at 0 deg.CThe resulting precipitate was removed by filtration and dried for 2 hours to give 5-methylfuran-2-pyridinesulfonate (2.93g, 50%) as an off-white solid.1H NMR(400MHz,DMSO):δ=8.90(dd,J1=4Hz,J2=8Hz,2H),8.57(tt,J1=1.5Hz,J2=8.1Hz,1H),8.04(dd,J1=4Hz,J2=8Hz,2H),6.27(d,J=3.0Hz,1H),5.98-5.94 9m,1H),2.19(s,3H)。

A slurry of 5-methylfuran-2-sulfonic acid pyridinium salt (1.0g, 4.41mmol) in anhydrous DME was treated with oxalyl chloride (0.53mL, 6.21mmol) followed by DMF (0.32mL, 4.41mmol) at 0 deg.C under argon and the reaction stirred at room temperature until complete. The reaction was quenched with ice water and extracted with toluene (2X 50mL), and the combined organics were extracted with saturated NaHCO3Washed with aqueous solution (20mL), brine (20mL), and dried (MgSO) 4) And concentrated in vacuo to give 5-methylfuran-2-sulfonyl chloride as a pale yellow oil (350mg, 47%).1H NMR(400MHz,CDCl3):δ=7.23-7.21(m,1H),6.27-6.25(m,1H),2.47(s,3H)。

To a solution of 5-methylfuran-2-sulfonyl chloride (0.2g, 1.10mmol) in acetone (1mL) at 0 deg.C was added NH3Aqueous solution (1mL, 28% NH)4OH in H2O) in (A). The reaction mixture was stirred at ambient temperature for about 2 hours, then concentrated in vacuo. The residue was suspended in dichloromethane (5mL) and NH was removed by filtration4Cl by-product, and the remaining solution was concentrated in vacuo. The crude product was purified by silica gel column chromatography using 40% EtOAc-hexanes eluent to give 5-methylfuran-2-sulfonamide as an off-white solid (130mg, 73%).1H NMR(400MHz,DMSO-d6):δ=7.60(s,2H),6.83-6.82(d,J=4.0Hz,1H),6.26-6.25(d,J=4.0Hz,1H),2.34(s,3H)。

5-ethyl-N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) furan-2-sulfonamide

To 2-ethylfuran (2.0g, 20.8mmol) in the absence ofTo a solution in acetonitrile water (3mL) was added SO3Py complex (4.30g, 27.0 mmol). The resulting reaction mixture was heated at 40 ℃ under nitrogen atmosphere for 23 hours or until completion. EtOAc (5mL) was added and the solution was stirred at 0 ℃ for 2 h. The resulting precipitate was removed by filtration and dried to give pyridin-1-ium 5-ethylfuran-2-sulfonate (3.2g, 60%) as a brown hygroscopic solid, which was used directly in the next step without purification.

To a slurry of 5-ethylfuran-2-sulfonic acid pyridinium (3.2g, 12.5mmol) in DME (15mL) was added oxalyl chloride (1.62mL, 27.0mmol) followed by DMF (0.97mL, 12.5mmol) at 0 deg.C under argon and the resulting reaction mixture was stirred at room temperature until complete. The reaction mixture was quenched with ice water, then extracted with toluene (2X 50mL), and the organic layer was washed with saturated NaHCO3The aqueous solution (20mL) and brine (20mL) were washed and dried (MgSO4) And concentrated in vacuo to give 5-ethylfuran-2-sulfonyl chloride as a light brown oil (510mg, 21%).1H NMR(400MHz,CDCl3):δ7.23(d,J=4Hz,1H),6.26(d,J=8Hz,1H),2.80(q,J=8Hz,2H),1.33(t,J=8Hz,3H)。

To a solution of 5-ethylfuran-2-sulfonyl chloride in acetone (1mL) at 0 deg.C was added NH3Aqueous solution (1.5mL, NH)4OH in H2In O, 28% NH3Base), the resulting reaction mixture was stirred at room temperature for 2 hours or until completion. The solvent was removed in vacuo and azeotroped with toluene (. times.2). The residue was purified by silica gel column chromatography using 1% MeOH in DCM eluent to give 5-ethylfuran-2-sulfonamide (0.36g, 78%) as a brown gum.1H NMR(400MHz,DMSO-d6):δ7.63(bs,2H),6.85(d,J=4Hz,1H),6.28(d,J=4Hz,1H),2.70(q,J=8Hz,2H),1.21(t,J=6Hz,3H)。

4- (prop-1-en-2-yl) furan-2-sulfonamide

Synthesis of 4- (prop-1-en-2-yl) furan-2-sulfonamide from ethyl furan-3-carboxylate was carried out using the procedure described in detail in Urban et al Synth. Commun.2003,33(12), 2029-.

d6-4- (prop-1-en-2-yl) furan-2-sulfonamide

The procedure contained in Urban et al, Synth. Commun.2003,33(12),2029-3Magnesium iodide instead of methyl magnesium chloride to give the corresponding d6-4- (prop-1-en-2-yl) furan-2-sulfonamide.

4- (prop-1-en-2-yl) furan-2-sulfonamide

To a solution of triphenylphosphine (0.3g, 1.16mmol) in anhydrous THF (5.0mL) was added iodine (1.0 equiv.), and the mixture was stirred at room temperature for 10 min. A solution of 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide in THF (3.0ml) was added slowly and stirring was continued for 2 hours or until completion. The solution was diluted with EtOAc (20mL) and washed with 10% aqueous sodium bisulfite (20mL) and water (20 mL). The organic layer was separated and dried (MgSO)4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 20% EtOAc in hexanes eluent to give the title compound as a white solid (0.1g, 58%).1H NMR(400MHz,CDCl3)δ=7.51(s,1H),7.16(s,1H),5.27(s,2H),2.02(s,3H)。13C NMR(101MHz,CDCl3)δ149.3,140.7,140.7,132.3,127.8,112.1,111.9,76.0,7.7,28.7,19.8。

4- (2-hydroxy-2-yl) -5-methylfuran-2-sulphonamide

At-10 deg.C, byChlorosulfonic acid (23.8g, 0.204M) was added dropwise to a solution of ethyl 2-methyl-3-furoate (30g, 0.195M) in DCM (300mL) for 15 minutes. The reaction was allowed to warm to ambient temperature and stirred for 72 hours. The solution was cooled to-10 ℃ and anhydrous pyridine (16.9g, 0.214M) was added dropwise, followed by phosphorus pentachloride (44.6g, 0.214M) in about 10g portions over 10 minutes. In that <Stir at 0 ℃ for 30 minutes and then at ambient temperature overnight. The reaction mixture was added dropwise to water (550mL) with stirring and stirring was continued for 2 hours. The organic phase was separated and the aqueous phase was extracted with DCM (150 mL). The combined organics were washed with water (300mL) and dried (Na)2SO4) And concentrated in vacuo to give 44g of a dark red oil. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes eluent to give ethyl 5- (chlorosulfonyl) -2-methylfuran-3-carboxylate (36g, 73%) as an orange oil.1H NMR(400MHz,CDCl3)δ=7.55(s,1H),4.63(q,J=7.2Hz,2H),2.75(s,3H),1.38(t,J=7.2Hz,3H)。

To a solution of ammonium bicarbonate (37.6g, 0.475M) in water (630mL) was added 5- (chlorosulfonyl) -2-methylfuran-3-carboxylic acid ethyl ester (30g, 0.12M) in acetone (200mL) dropwise over 15 minutes. The reaction mixture was stirred at ambient temperature until completion (about 3 hours). EtOAc (250mL) was added and the pH was adjusted to about pH 2 with dropwise addition of concentrated HCl. The organics were separated and the remaining aqueous phase was saturated with sodium chloride and re-extracted with EtOAc (250 mL). The combined organics were washed with brine (300mL) and dried (Na)2SO4) And concentrated in vacuo to give a brown oily solid which was recrystallized from EtOAc-hexanes to give ethyl 2-methyl-5-sulfamoylfuran-3-carboxylate (11.4g, 41%) as a beige solid. 1H NMR(400MHz,DMSO-d6)δ=7.8(s,2H),7.02(s,1H),4.26(q,J=7.2Hz,2H),2.62(s,3H),1.3(t,J=7.2Hz,3H)。

Ethyl 2-methyl-5-sulfamoylfuran-3-carboxylate (10g, 0.043M) in anhydrous THF (400mL) was treated dropwise with a solution of methyl magnesium chloride (3.0M in THF, 64.3mL) under vigorous stirring at-10 ℃ over 5 minutes. The solution was then stirred at ambient temperature for 6 hours, then cooled to-5 ℃ and treated with ammonium chloride solution (51.8g in 265mL of water)Middle) was treated dropwise. The aqueous solution was extracted with EtOAc (2X 250mL), and the combined organics were washed with brine (250mL) and dried (Na)2SO4) And concentrated in vacuo to an orange oil (10 g). The crude product was purified by silica gel column chromatography using 40% EtOAc-hexanes eluent to give the title compound as a white solid (6.1g, 42%).1H NMR(400MHz,DMSO-d6)δ=7.54(br.s.,2H),6.78(s,1H),4.95(s,1H),2.42(s,3H),1.4(s,6H)。

d6-4- (2-hydroxypropan-2-yl) -5-methylfuran-2-sulphonamide

Methyl 2-methyl-5-sulfamoylfuran-3-carboxylate can be prepared by modifying the procedure used for the synthesis of ethyl 2-methyl-5-sulfamoylfuran-3-carboxylate, but using methyl 2-methylfuran-3-carboxylate as starting material instead of ethyl 2-methylfuran-3-carboxylate. Methyl 2-methyl-5-sulfamoylfuran-3-carboxylate (3g, 29%) was obtained as a white solid. 1H NMR (400MHz, DMSO-d)6)δ=7.89(s,2H),7.03(s,1H),3.79(s,3H),2.61(s,3H)。

At-10 ℃ for 10 minutes with vigorous stirring3Solution of-methyl magnesium iodide (1.0M in Et) 2O, 26mL) methyl 2-methyl-5-sulfamoylfuran-3-carboxylate (0.7g, 3.2mmol) in anhydrous THF (20mL) was treated dropwise. The solution was then stirred at ambient temperature for 12 hours, then cooled to 0 ℃ and treated dropwise with a saturated ammonium chloride solution. The aqueous solution was extracted with EtOAc (2X 25mL), and the combined organics were washed with brine (25mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using a gradient of 40-70% EtOAc-hexanes eluent to give the title compound as a white solid (0.37g, 51%).1H NMR(400MHz,DMSO-d6)δ=7.57(s,2H),6.79(s,1H),4.99(s,1H),2.4(s,3H)。13C NMR(100MHz,CD3OD) δ 150.3,147.3,128.4,113.4,67.3,28.5 (multiplet), 12.2.

1-benzyl-1H-1, 2, 4-triazole-3-sulfonamides

A solution of 1H-1,2, 4-triazole-3-thiol (1g, 9.90mmol) in DMF (20mL) was taken with K2CO3(4.8g, 34.7mmol), cooled to 0 deg.C, and benzyl bromide (4.2g, 24.8mmol) added dropwise over 5 minutes. The resulting reaction mixture was warmed to ambient temperature and stirred for 12 hours. The reaction mixture was diluted with water (25mL) and extracted with ethyl acetate (2X 25 mL). The combined organics were washed with water (20mL), brine (20mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 20% EtOAc-hexanes eluent to give 1-benzyl-3- (benzylsulfanyl) -1H-1,2, 4-triazole (1.5g, 54%) as a white solid. 1H NMR(400MHz,DMSO-d6):δ=8.67(s,1H),7.39-7.32(m,5H),7.27-7.21(m,5H),5.36(s,2H),4.29(s,2H)。

To a solution of 1-benzyl-3- (benzylsulfanyl) -1H-1,2, 4-triazole, 2(0.5g, 1.77mmol) in acetonitrile (5mL) at 0 deg.C was added AcOH (3mL) and H2O (2mL), then Cl2Gas was bubbled through the solution for 45 minutes. Stirring was continued at 0 ℃ for 30 minutes and then at 20 ℃ for 1.5 hours. The reaction mixture was diluted with water (20mL) and extracted with EtOAc (2X 20 mL). The combined organics were washed with water (20mL), brine (20mL) and dried (Na)2SO4) And concentrated in vacuo to give a colorless liquid. The residue was diluted with THF and cooled to-78 ℃. Ammonia gas was bubbled through the solution for 20 minutes and stirring was continued for another 30 minutes, then warmed to ambient temperature and stirred for 1 hour. The reaction mixture was diluted with water (25mL) and extracted with ethyl acetate (2X 25 mL). The combined organics were washed with water (20mL), brine (20mL) and dried (Na)2SO4) And concentrated in vacuo. The residue was triturated with diethyl ether to give 1-benzyl-1H-1, 2, 4-triazole-3-sulfonamide as an off-white solid (0.25g, 60%).1H NMR(400MHz,DMSO-d6):δ=8.88(s,1H),7.77(s,2H),7.39-7.33(m,5H),5.45(s,2H)。

1-isopropyl-1H-1, 2, 3-triazole-4-sulfonamide

Sodium 1H-1,2, 3-triazole-5-thiolate (500mg, 4.06mmol) is dissolved in EtOH (5mL) and cooled to 0 ℃. Benzyl bromide (0.69g, 4.06mmol) was added dropwise over a period of 5 minutes. The resulting reaction mixture was warmed to room temperature and stirred for 1 hour. After completion, the reaction mixture was concentrated in vacuo and the resulting residue was taken up with saturated NaHCO 3The solution was diluted and extracted with EtOAc (2X 20 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The resulting residue was stirred with n-pentane (30mL), filtered and dried in vacuo to give 4- (benzylsulfanyl) -1H-1,2, 3-triazole (0.7g, 90%) as a white solid, which was used without further purification.1H NMR(400MHz,CDCl3):δ=7.40-7.38(m,1H),7.35-7.21(m,5H),4.12(s,2H)。LCMS(m/z):192.0[M+H]+

A solution of 4- (benzylsulfanyl) -1H-1,2, 3-triazole (5g, 26.1mmol) in DMF (50mL) was cooled to 0 deg.C and treated with K2CO3(9.03g, 65.4 mmol). The reaction mixture was stirred at the same temperature for 5 minutes. Isopropyl iodide (8.89g, 52.3mmol) was added dropwise to the above mixture over 5 minutes. The resulting reaction mixture was warmed to room temperature and stirred for 2 hours. Upon completion, the reaction mixture was diluted with water (30mL) and extracted with ethyl acetate (50 mL). The organic extracts were washed with water, brine and over anhydrous Na2SO4And (5) drying. The crude product was purified by silica gel column chromatography with 8% EtOAc-hexanes eluent to give 5- (benzylthio) -1-isopropyl-1H-1, 2, 3-triazole a (0.9g), 4- (benzylthio) -2-isopropyl-2H-1, 2, 3-triazole B (1g), and the desired product 4- (benzylthio) -1-isopropyl-1H-1, 2, 3-triazole C (1.4g, 23%).1H NMR(400MHz,DMSO-d6):δ=7.29-7.18(m,5H),4.78-4.71(m,1H),4.09(s,2H),1.4(d,J=6.8Hz,6H)。LCMS(m/z):234.30[M+H]+

4- (benzylsulfanyl) -1-isopropyl-1H-1, 2, 3-triazole (75mg, 0.32mmol) in acetic acid (2.25 mL) ) And H2The solution in O (1.12mL) was cooled to 0 ℃. N-chlorosuccinamide (170mg, 1.28mmol) was added at 0 ℃. The resulting reaction mixture was warmed to room temperature and stirred for 1 hour. After completion, the reaction mixture was diluted with water and extracted with ethyl acetate (2 × 10 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 8% EtOAc-hexanes eluent to give 1-isopropyl-1H-1, 2, 3-triazole-4-sulfonyl chloride (0.1g, 100%) as a light brown liquid, which was used without further purification. LCMS (M/z) 210.10[ M + H]+。

A solution of 1-isopropyl-1H-1, 2, 3-triazole-4-sulfonyl chloride (100mg) in THF (5mL) was cooled to-40 ℃. Ammonia gas was purged through the solution for 15 minutes. The reaction mixture was warmed to room temperature and stirred for 2 hours. Upon completion, the reaction mixture was concentrated in vacuo and the resulting residue was diluted with ethyl acetate (25mL) and water (10 mL). The organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo to give 1-isopropyl-1H-1, 2, 3-triazole-4-sulfonamide (0.07g, 78%) as a brown solid.1H NMR(400MHz,DMSO-d6):δ=8.71(s,1H),7.66(s,2H),4.91-4.87(m,1H),1.5(d,J=6.8Hz,6H)。LCMS(m/z):191.30[M+H]+

1-methyl-1H-pyrazole-3-sulfonamides

Using general procedure D, 1-methyl-1H-pyrazol-3-amine hydrochloride (0.7g, 38%) was reacted with 1-methyl-1H-pyrazole-3-sulfonyl chloride (light yellow liquid). 1H NMR(300MHz,CDCl3):δ=7.51-7.50(d,J=2.1Hz,1H),6.89-6.88(d,J=2.4Hz,1H),4.06(s,3H)。LCMS(m/z):160.9(M-1)-. Sulfonyl chloride was converted using general method E1 to give the title compound as an off-white solid (0.4g, 69%).1H NMR(400MHz,DMSO-d6):δ=7.80(d,J=2.1Hz,1H),7.36(s,2H),6.53(d,J=2.1Hz,1H),3.88(s,3H)。LCMS(m/z):162.05(M+1)+

1- (trifluoromethyl) -1H-pyrazole-3-sulfonamides

1- (trifluoromethyl) -1H-pyrazol-3-amine was reacted with 1- (trifluoromethyl) -1H-pyrazole-3-sulfonyl chloride (brown liquid) using general procedure D (0.4g, 43%).1H NMR(300MHz,CDCl3)δ=8.02(d,J=2.8Hz,1H),7.06(d,J=2.8Hz,1H)。19F NMR(282MHz,CDCl3)δ=-60.46。

Sulfonyl chloride was converted using general procedure E1 to give the title compound (0.22g, 46%).1H NMR(300MHz,CDCl3)δ=7.92(dd,J=2.8,0.3Hz,1H),6.91(dd,J=2.8,0.7Hz,1H),5.28(s,2H)。19F NMR(282MHz,CDCl3)δ=-60.41。

1-isopropyl-1H-pyrazole-3-sulfonamides

1-isopropyl-1H-pyrazol-3-amine was reacted with 1-isopropyl-1H-pyrazole-3-sulfonyl chloride (brown liquid) using general procedure D (0.5g, 43%).1H NMR(400MHz,CDCl3):δ=7.55(s,1H),6.88(s,1H),4.66-4.63(m,1H),3.6(br.s.,2H),1.59(d,J=6.8Hz,6H)。LCMS(m/z):209.0(M+1)+. Sulfonyl chloride was converted using general procedure E1 to give the title compound as a yellow solid (0.45g, 82%).1H NMR(300MHz,DMSO-d6):δ=7.9(d,J=2.4Hz,1H),7.36(s,2H),6.55(d,J=2.1Hz,1H),4.57-4.53(m,1H),1.42(d,J=6.9Hz,6H)。LCMS(m/z):190.0(M+1)+

1-isopropyl-1H-pyrazole-4-sulfonamides

At ambient temperature under a nitrogen atmosphere,by K2CO3A solution of 4-iodo-1H-pyrazole (1g, 5.15mmol) in DMF (20mL) was treated (1.42g, 10.30mmol) and isopropyl iodide (1.05g, 6.19 mmol). The resulting reaction mixture was heated to 90 ℃ and stirred for 12 hours. The mixture was cooled, diluted with water (50mL), and extracted with diethyl ether (2X 50 mL). The combined organics were washed with water (2X 50mL), brine (50mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes eluent to give 4-iodo-1-isopropyl-1H-pyrazole as a colorless liquid (1.1g, 92%). 1H NMR(400MHz,CDCl3):δ=7.50-7.46(m,2H),4.53-4.47(m,1H),1.50(d,J=6.8Hz,6H)。LCMS(m/z):237.2(M+1)+

A solution of 4-iodo-1-isopropyl-1H-pyrazole (1g, 4.24mmol) in dioxane (20mL) was treated with benzylthiol (0.8g, 6.35mmol) and DIPEA (1.1g, 8.47mmol) in succession under nitrogen atmosphere. The solution was degassed by purging with argon and held for 15 minutes. Adding Pd under argon atmosphere2(dba)3(40mg, 0.0423mmol) and Xantphos (50mg, 0.0847mmol), and the resulting mixture was sealed in a reaction vessel and heated at 75 ℃ for 6 hours. The reaction mixture was cooled, concentrated in vacuo, diluted with water (20mL), and extracted with EtOAc (2X 20 mL). The combined organics were washed with water (2X 50mL), brine (50mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes eluent to give 4- (benzylsulfanyl) -1-isopropyl-1H-pyrazole as a yellow liquid (650mg, 66%).1H NMR(400MHz,CDCl3):δ=7.36(s,1H),7.26-7.22(m,4H),7.11-7.09(m,2H),4.41-4.36(m,1H),3.76(s,2H),1.42(d,J=6.8Hz,6H)。LCMS(m/z):233.3(M+1)+

To a solution of 4- (benzylsulfanyl) -1-isopropyl-1H-pyrazole, 3(0.35g, 1.508mmol) in acetonitrile (10mL) at 0 deg.C was added AcOH (0.7mL) and H2O (0.35mL), then DCDMH (0.6g, 3.017mmol) was added portionwise over 5 minutes. The solution was stirred for 30 minutes, then warmed to ambient temperature and stirred for an additional 2 hours. The reaction mixture was diluted with water (20mL) and extracted with EtOAc (2X 20 mL). The combined organics were treated with water (20mL), Washed with brine (20mL) and dried (Na)2SO4) And concentrated in vacuo to give 1-isopropyl-1H-pyrazole-4-sulfonyl chloride as a colorless liquid. The sulfonyl chloride was diluted with THF and cooled to-78 deg.C, then the NH was added3Gas was bubbled through the solution for 15 minutes. The reaction mixture was stirred at-78 ℃ for 1 hour and at ambient temperature for 2 hours. The reaction mixture was diluted with water and the compound was extracted with ethyl acetate (2X 25 mL). The combined organic extracts were washed with water (20mL), brine (20mL) and dried (Na)2SO4) And concentrated in vacuo. The resulting residue was triturated with diethyl ether and dried under reduced pressure to give 1-isopropyl-1H-pyrazole-4-sulfonamide (0.2g, 71%) as a light brown solid.1H NMR(400MHz,DMSO-d6):δ=8.21(s,1H),7.71(s,1H),7.22(s,2H),4.59-4.53(m,1H),1.4(d,J=6.8Hz,6H)。LCMS(m/z):190.2(M+1)+

1-cyclopropyl-1H-pyrazole-3-sulfonamides

1-cyclopropyl-1H-pyrazol-3-amine was reacted with 1-cyclopropyl-1H-pyrazole-3-sulfonyl chloride using general procedure D, then transformed using general procedure E1 to give the title compound as a light brown solid (0.2g, 33%).1H NMR(400MHz,CDCl3)δ=7.51(d,J=2.4Hz,1H),6.69(d,J=2.4Hz,1H),5.04(s,2H),3.67(m,1H),1.28–1.05(m,4H)。

1- (tert-butyl) -1H-pyrazole-3-sulfonamides

1- (tert-butyl) -1H-pyrazol-3-amine was reacted with 1- (tert-butyl) -1H-pyrazole-3-sulfonyl chloride using general procedure D and then transformed using general procedure E1 to give the title compound as a light brown solid (150mg, 26%). 1H NMR(400MHz,DMSO-d6):δ=7.56(d,J=3.6Hz,1H),6.7(d,J=3.2Hz,1H),4.75(br.s.,1H),1.60(s,9H)。LCMS(m/z):204.15(M+1)+

1-cyclohexyl-1H-pyrazole-3-sulfonamides

1-cyclohexyl-1H-pyrazol-3-amine was reacted with 1-cyclohexyl-1H-pyrazole-3-sulfonyl chloride using general procedure D, then converted using general procedure E1 to give the title compound as a white solid (0.35mg, 50%).1H NMR(300MHz,DMSO-d6)δ=7.89(d,J=2.3Hz,1H),7.36(s,2H),6.55(d,J=2.3Hz,1H),4.28–4.08(m,1H),2.0 1.1(m,6H)。

1-phenyl-1H-pyrazole-3-sulfonamides

Using general procedure D (0.5g, 47%), 1-phenyl-1H-pyrazol-3-amine was reacted with 1-phenyl-1H-pyrazole-3-sulfonyl chloride (yellow liquid).1H NMR(400MHz,CDCl3) δ is 8.04(d, J is 2.4Hz,1H),7.73(d, J is 9.2Hz,2H),7.58(t, J is 7.6Hz,2H),7.47(t, J is 7.2Hz,1H),7.08(d, J is 2.8Hz, 1H). Sulfonyl chloride was converted using general procedure E1 to give the title compound as a yellow solid (0.4g, 87%).1H NMR(400MHz,DMSO-d6):δ=8.62(d,J=2.7Hz,1H),7.86(d,J=8.7Hz,2H),7.61(br.s.,2H),7.57(t,J=7.8Hz,2H),7.41(t,J=7.2Hz,1H),6.85(d,J=2.4Hz,1H)。LCMS(m/z):224.1(M+1)+

1-benzyl-1H-pyrazole-3-sulfonyl chloride

Using general procedure D (0.2g, 45%), 1-benzyl-1H-pyrazol-3-amine was reacted with 1-benzyl-1H-pyrazole-3-sulfonyl chloride (light brown liquid).1H NMR(300MHz,CDCl3):δ7.42-7.38(m,3H),7.33-7.28(m,3H),6.8(d,J=2.4Hz,1H),5.42(s,2H). Sulfonyl chloride was converted using general procedure E1 to give the title compound as a light brown liquid (0.15g, 81%).1H NMR(400MHz,CDCl3):δ=7.42-7.36(m,4H),7.24(d,J=1.6Hz,2H),6.7(d,J=2.4Hz,1H),5.35(s,2H),5.10(s,2H)。LCMS(m/z):238.10(M+1)+

1- (1-phenylethyl) -1H-pyrazole-3-sulfonamide

1- (1-phenylethyl) -1H-pyrazol-3-amine was reacted with 1- (1-phenylethyl) -1H-pyrazole-3-sulfonyl chloride using general method D and then converted using general method E1 to give the title compound as a white solid (0.25mg, 68%). 1H NMR(300MHz,CDCl3)δ=7.43–7.18(m,6H),6.72(d,J=2.4Hz,1H),5.57(q,J=7.1Hz,1H),5.02(s,2H),1.92(d,J=7.1Hz,3H)。

1- (2- (piperidin-1-yl) ethyl) -1H-pyrazole-3-sulfonamide

1- (2- (piperidin-1-yl) ethyl) -1H-pyrazol-3-amine was reacted with 1- (2- (piperidin-1-yl) ethyl) -1H-pyrazole-3-sulfonyl chloride (light brown liquid) using general procedure D and then converted using general procedure E1 to give the title compound as an off-white solid (0.3g, 46%).1H NMR(300MHz,DMSO-d6):δ=7.84(d,J=2.1Hz,1H),7.36(s,2H),6.54(s,J=2.4Hz,1H),4.26(t,J=6.9Hz,2H),2.66(t,J=6.6Hz,2H),2.36(s,4H),1.46-1.34(m,6H)。LCMS(m/z):259.10(M+1)+

1, 5-dimethyl-1H-pyrazole-3-sulfonamides

1, 5-dimethyl-1H-pyrazol-3-amine and 1, 5-dimethyl-1H-pyri-dine were reacted using general procedure D (0.45g, 26%)Oxazole-3-sulfonyl chloride (yellow liquid) reaction.1H NMR(300MHz,CDCl3):δ=5.92(s,1H),3.71(s,3H),2.23(s,3H)。LCMS(m/z):217(M+Na)+. Sulfonyl chloride was converted using general method E1 to give the title compound as an off-white solid (0.25g, 55%).1H NMR(400MHz,DMSO-d6):δ=7.30(s,2H),6.36(s,1H),3.76(s,3H),2.27(s,3H)。LCMS(m/z):175.9(M+1)+

1-methyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonamide

Using general procedure D (1.1g, 37%), 1-methyl-5- (trifluoromethyl) -1H-pyrazol-3-amine was reacted with 1-methyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonyl chloride (light brown liquid).1H NMR(300MHz,CDCl3) δ is 7.21(s,1H),4.16(s, 3H). Sulfonyl chloride was converted using general method E1 to give the title compound as a yellow solid (0.45mg, 82%).1H NMR(300MHz,CDCl3):δ=7.06(s,1H),5.02(br.s.,2H),4.03(s,3H)。

1-isopropyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonamide

A mixture of NaH (2.14g, 89.3mmol) in DMF (20mL) was cooled to-10 ℃. A solution of 1,1, 1-trifluoropropan-2-one (5g, 44.6mmol) in DMF (80mL) was added very carefully to the above mixture and stirred at-10 ℃ for 5 min. (iii) adding CS over 30 minutes 2(10.2g, 133.9mmol) was added dropwise to the above mixture, and the reaction mixture was then warmed to ambient temperature and stirred for 1 hour. The reaction mixture was cooled to 0 ℃ and over 10 minutes with CH3I (7.5 mL). The resulting reaction mixture was warmed to ambient temperature and stirred for 12 hours. The reaction mixture was diluted with cold water (50mL) and extracted with diethyl ether (2X 100 mL). The combined organic extracts were washed with water (50mL), brine (50 m)L) washing and drying (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 5% EtOAc-hexanes eluent to give 1,1, 1-trifluoro-4, 4-bis (methylthio) but-3-en-2-one (3.5g, 36%) as a light brown solid.1H NMR(300MHz,CDCl3):δ=6.24(s,1H),2.57(m,6H)。LCMS(m/z):217.20(M+1)+

A solution of 1,1, 1-trifluoro-4, 4-bis (methylthio) but-3-en-2-one (2.5g, 11.6mmol) in EtOH (25mL) was treated with isopropylhydrazine hydrochloride (2g, 13.9mmol) at 0 deg.C and Et was added3N (2.4g, 40.98mmol) and the mixture was heated at 80 ℃ for 12 h. The reaction mixture was concentrated in vacuo and saturated NaHCO3The aqueous solution was diluted and extracted with EtOAc (2X 250 mL). The combined organics were washed with water (200mL), brine (200mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 100% EtOAc eluent to give 1-isopropyl-3- (methylthio) -5- (trifluoromethyl) -1H-pyrazole as a light brown liquid (1.5g, 58%). 1H NMR(400MHz,CDCl3):δ=6.47(s,1H),4.58-4.53(m,1H),2.49(s,3H),1.50(d,J=6.8Hz,6H)。LCMS(m/z):225.20(M+1)+

A solution of 1-isopropyl-3- (methylthio) -5- (trifluoromethyl) -1H-pyrazole (0.5g, 2.23mmol) in chloroform (10mL) was treated with mCPBA (0.38g, 2.23mmol) at 0 deg.C and stirred at 10 deg.C for 1H. The reaction mixture was washed with saturated NaHCO3The solution (10mL) was diluted and diluted with CHCl3(2X 30 mL). The combined organics were washed with water (30mL), brine (30mL) and dried (Na)2SO4) And concentrated in vacuo. The resulting residue was dissolved in CHCl3(10mL) and treated with trifluoroacetic anhydride (1.4g, 6.7 mmol). The reaction mixture was heated at 50 ℃ for 3 hours, cooled to ambient temperature and concentrated in vacuo. The resulting residue was taken up in MeOH (5mL) -THF (5mL) -H2Diluted with O (5mL), cooled to 0 deg.C, and Na2CO3(0.7g, 6.7mmol) and stirred for 3 hours. The solution was diluted with water (30mL) and CHCl3(2X 50 mL). The combined organics were washed with water (50mL), brine (50mL) and dried (Na)2SO4) And concentrated in vacuo. The crude residue containing 1-isopropyl-3- ((1-isopropyl-5- (trifluoromethyl) -1H-imidazol-4-yl) disulfanyl) -5- (trifluoromethyl) -1H-pyrazole (0.2g) was used in the next step without further purification.1H NMR(400MHz,CDCl3):δ=6.85(s,2H),6.70(s,1H),6.60(s,1H),4.6(m,2H),1.53(m,6H)。LCMS(m/z):416.75(M-1)-

A solution of crude 1-isopropyl-3- ((1-isopropyl-5- (trifluoromethyl) -1H-imidazol-4-yl) disulfanyl) -5- (trifluoromethyl) -1H-pyrazole (0.2g crude, 0.478mmol) in acetonitrile (10mL) was cooled to 0 deg.C and treated with AcOH (1mL) and H 2O (1.5 mL). DCDMH (0.19g, 0.956mmol) was added portionwise over 5 minutes and stirred for 2 hours. The mixture was diluted with water (20mL) and extracted with DCM (2X 20 mL). The combined organics were washed with water (50mL), brine (50mL) and dried (Na)2SO4) And concentrated in vacuo to give 1-isopropyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonyl chloride as a colorless liquid. (1-isopropyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonyl chloride) was diluted with THF, cooled to-78 deg.C, and the NH was added3The gas was bubbled through the solution for 10 minutes, followed by stirring for 1 hour, then warmed to ambient temperature and stirred for an additional 1 hour. The reaction mixture was diluted with water and extracted with ethyl acetate (2 × 25 mL). The combined organics were washed with water (50mL), brine (50mL) and dried (Na)2SO4) And concentrated in vacuo. The resulting residue was triturated with diethyl ether and n-pentane to give 1-isopropyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonamide (75mg, 61%) as a white solid.1H NMR(400MHz,CDCl3):δ=7.01(s,1H),5.06(s,2H),4.73-4.70(m,1H),1.5(d,J=6.8Hz,6H)。LCMS(m/z):256.0(M-1)-

5-isopropyl-1-methyl-1H-pyrazole-3-sulfonamide

A solution of 1-methyl-5- (prop-1-en-2-yl) -1H-pyrazol-3-amine (0.25g, 1.824mmol) in acetonitrile (10mL) was used for H at 0 deg.C2O(0.5mL) Concentrated HCl (1.2mL) in (g) was treated and then dissolved in H2NaNO in O (2mL) 2Aqueous solution (0.15g, 2.19mmol) was treated. The resulting solution was stirred at 0 ℃ for 45 minutes. AcOH (0.25mL), CuCl2·2H2O (0.15g, 0.91mmol) and CuCl (10mg, 0.091mmol) are added to the mixture in succession and treated with SO at 0 deg.C2Gas purge for 20 minutes. The resulting reaction mixture was stirred at 0 ℃ to 10 ℃ for 60 minutes. Upon completion, the reaction mixture was diluted with water (20mL) and extracted with EtOAc (2X 20 mL). The combined organics were washed with water (20mL), brine (20mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 20% EtOAc-hexanes eluent to give 1-methyl-5- (prop-1-en-2-yl) -1H-pyrazole-3-sulfonyl chloride as a colorless liquid (0.15g, 38%).1H NMR(300MHz,CDCl3):δ=6.77(s,1H),5.51(s,1H),5.28(s,1H),4.02(s,3H),2.11(s,3H)。

A solution of 1-methyl-5- (prop-1-en-2-yl) -1H-pyrazole-3-sulfonyl chloride (0.075g, 0.34mmol) in THF (7mL) was cooled to-78 ℃ and ammonia gas was bubbled through the solution for 15 minutes, stirring was continued for an additional 30 minutes, then warmed to ambient temperature and stirred for 2 hours or until completion. The reaction mixture was diluted with ethyl acetate (25mL) and filtered through a pad of celite. The filtrate was dried (Na)2SO4) And concentrated in vacuo to give 0.04g (crude) of 1-methyl-5- (prop-1-en-2-yl) -1H-pyrazole-3-sulfonamide as an off-white solid, which was used without purification.

A solution of crude 1-methyl-5- (prop-1-en-2-yl) -1H-pyrazole-3-sulfonamide (0.12g, 0.6mmol) in MeOH (10mL) -EtOAc (4mL) was treated with 10% palladium on carbon (30mg) under nitrogen. The reaction flask was evacuated, filled with hydrogen (balloon) and stirred for 4 hours. The reaction mixture was diluted with ethyl acetate (25mL), filtered through a pad of celite, and dried (Na)2SO4) And concentrated in vacuo. The resulting solid was further washed with diethyl ether to give 5-isopropyl-1-methyl-1H-pyrazole-3-sulfonamide as an off-white solid (0.11g, 91%).1H NMR(400MHz,CDCl3):δ=6.50(s,1H),5.00(br.s.,2H),3.87(s,3H),2.97-2.93(m,1H),1.28(d,J=7.2Hz,6H)。

5- (2-hydroxypropan-2-yl) -1-methyl-1H-pyrazole-3-sulfonamide

Ethyl 3-amino-1-methyl-1H-pyrazole-5-carboxylate was reacted with ethyl 3- (chlorosulfonyl) -1-methyl-1H-pyrazole-5-carboxylate (pale yellow liquid) using general procedure D (0.35g, 47%).1H NMR (300MHz, chloroform-d) δ 7.39(s,1H),4.40(q, J ═ 7.1Hz,2H),4.32(s,3H),1.40(t, J ═ 7.1Hz, 3H). Sulfonyl chloride was converted using general procedure E2 to give ethyl 1-methyl-3-sulfamoyl-1H-pyrazole-5-carboxylate (0.3g, 94%) as an off-white solid.1H NMR(300MHz,DMSO-d6)δ=7.59(s,2H),7.09(s,1H),4.33(q,J=7.1Hz,2H),4.14(s,3H),1.31(t,J=7.1Hz,3H)。

To a solution of ethyl 1-methyl-3-sulfamoyl-1H-pyrazole-5-carboxylate (0.25g, 1.07mmol) in anhydrous THF (10mL) at 0 ℃ was added dropwise magnesium methyl chloride (3M in THF, 5 equivalents). The resulting reaction mixture was gradually warmed to ambient temperature and stirred for 6 hours or until completion. The solution was cooled to 0 ℃ with saturated NH 4Aqueous Cl (2.0mL) was quenched, then diluted with cold water (20mL) and extracted with EtOAc (2X 25 mL). The combined organics were washed with brine (50mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using a gradient of 50% EtOAc in hexanes to give the title compound as a white solid (0.2g, 87%).1H NMR(400MHz,DMSO-d6)δ=7.34(s,2H),6.40(s,1H),5.48(s,1H),4.0(s,3H),1.50(s,6H)。

1-benzyl-5- (2-hydroxypropan-2-yl) -1H-pyrazole-3-sulfonamide

Using method D (0.35g, 47%) ethyl 3-amino-1-benzyl-1H-pyrazole-5-carboxylate with 1-benzyl-3- (chlorosulfonyl) -1H-pyrazole-5-carboxylic acidEthyl ester (light brown liquid) reaction.1H NMR(400MHz,CDCl3) δ 7.41(s,1H),7.34-7.26(m,5H),5.87(s,2H),4.37(q, J7.2 Hz,2H),1.38(t, J7.2 Hz, 3H). Sulfonyl chloride was converted using general procedure E2 to give ethyl 1-benzyl-3-sulfamoyl-1H-pyrazole-5-carboxylate (0.7g, 88%) as a white solid.1H NMR(400MHz,DMSO-d6):δ=7.66(s,2H),7.39-7.27(m,3H),7.2-7.18(m,3H),5.77(s,2H),4.33(q,J=7.2Hz,2H),1.29(t,J=7.2Hz,3H)。LCMS(m/z):310.05(M+1)+

To a solution of ethyl 1-benzyl-3-sulfamoyl-1H-pyrazole-5-carboxylate (0.5g, 1.62mmol) in anhydrous THF (10mL) at 0 deg.C was added dropwise methylmagnesium chloride (3M in THF, 2.77mL, 8.1 mmol). The resulting reaction mixture was gradually warmed to ambient temperature and stirred for 4 hours or until completion. The solution was cooled to 0 ℃ with saturated aqueous NH 4Cl (2.0mL) was quenched, then diluted with cold water (20mL) and extracted with EtOAc (2X 25 mL). The combined organics were washed with brine (50mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using a gradient of 70-100% EtOAc in hexanes to give the title compound as a white solid (0.27g, 57%).1H NMR(300MHz,DMSO-d6):δ=7.37(s,2H),7.39-7.27(m,3H),7.2-7.18(m,2H),6.45(s,1H),5.66(s,2H),5.60(s,1H),1.44(s,6H)。LCMS(m/z):296.1(M+1)+

5- (2-hydroxypropan-2-yl) -1-phenyl-1H-pyrazole-3-sulfonamide

2- (3-amino-1-phenyl-1H-pyrazol-5-yl) propan-2-ol was reacted with 5- (2-hydroxypropan-2-yl) -1-phenyl-1H-pyrazole-3-sulfonyl chloride (yellow liquid) using method D (0.4g, 36%).1H NMR(300MHz,CDCl3) δ is 7.55-7.45(m,5H),6.91(s,1H),1.51(s, 6H). Sulfonyl chloride was converted using general procedure E2 to give the title compound as a yellow solid (0.32g, 87%).1H NMR(300MHz,DMSO-d6):δ=7.5(s,5H),7.47(s,2H),6.65(s,1H),5.41(s,1H),1.30(s,6H)。

5- (dimethylamino) naphthalene-1-sulfonamide

5- (dimethylamino) naphthalene-1-sulfonamide 3-azidobenzenesulfonamide was synthesized according to the procedures contained in Satish K.Nair, Daniel Elbaum and David W.Christianson.J.biol.chem.1996,271: 1003-.

A solution of 5- (dimethylamino) naphthalene-1-sulfonyl chloride (0.12g, 0.44mmol) in acetone (5mL) was added dropwise to a solution of ammonium bicarbonate (0.17g, 1.76mmol) in water (1.0mL) and the reaction was stirred for 2 hours at ambient temperature or until completion. The pH was adjusted to pH 2.0 using concentrated HCl. The organic phase was separated and the aqueous phase was saturated with NaCl and extracted with ethyl acetate. The combined organic phases were dried with brinem (MgSO) 4) Washed and concentrated in vacuo to give the title compound as a white solid (0.075g, 67% yield).1H NMR(600MHz,CD3OD)δ=8.54(d,J=8.5Hz,0H),8.36(d,J=8.7Hz,1H),8.23(d,J=7.3Hz,1H),7.58(ddd,J=17.0,8.6,7.4Hz,2H),7.28(d,J=7.6Hz,1H),2.89(s,6H)。13C NMR(151MHz,CD3OD)δ=151.6,138.9,129.7,129.4,129.2,127.4,126.6,122.9,119.5,114.8,44.4。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -2, 3-dihydrobenzo [ b ] thiophene-6-sulfonamide 1, 1-dioxide

At room temperature under vigorous stirring m-chloroperbenzoic acid (77%, 6.35g, 27.9mmol) was added portionwise to benzo [ b ]]Thiophene (1.50g, 11.1mmol) in anhydrous dichloromethane (100mL) and the resulting reaction mixture was stirred at the same temperature for 16 hours. Adding saturated NaHCO3Aqueous solution(250mL), the aqueous layer was extracted with dichloromethane (2X 100mL), the organic layer was separated and dried (MgSO4) The combined organic layers were concentrated in vacuo. Crystallization from ethanol gave benzo [ b ] as an off-white solid]Thiophene 1, 1-dioxide (1.56g, 84%).1H NMR(600MHz,CDCl3):δ=7.73(d,J=6Hz,1H),7.58-7.53(m,2H),7.38(d,J=12Hz,1H),7.23(d,J=6Hz,1H),6.73(d,J=6Hz,1H)。LCMS(m/z):167[M+H]+

A benzene ring [ b ]]A solution of thiophene 1, 1-dioxide (0.75g, 4.51mmol) in ethanol (55mL) was degassed with nitrogen for 10 minutes, then 10% Pd/C (10mg) was added, and the mixture was stirred under a hydrogen atmosphere (1atm) for 24 hours. The reaction mixture was filtered through a pad of celite, and the filtrate was concentrated to give 2, 3-dihydrobenzo [ b ] as an off-white solid]Thiophene 1, 1-dioxide (0.74g, 97%).1H NMR(600MHz,CDCl3):δ=7.75(d,J=6Hz,1H),7.59(t,J=9Hz,1H),7.49(t,J=6Hz,1H),7.40(d,J=6Hz,1H),3.51(t,J=6Hz,2H),3.41(t,J=6Hz,2H)。LCMS(m/z):169[M+H]+

At 80 ℃,2, 3-dihydrobenzo [ b ] ]Thiophene 1, 1-dioxide (0.75g, 4.45mmol) was heated in chlorosulfonic acid (1.5mL, 22.2mmol) for 4 hours. The reaction mixture was poured onto crushed ice and stirred for 5 minutes. The aqueous solution was extracted with dichloromethane (2X 50mL) and the combined organics were dried (MgSO)4) And concentrated in vacuo to give 2, 3-dihydrobenzo [ b ] as a light brown oil]Thiophene-6-sulfonyl chloride 1, 1-dioxide (0.45g, 38%). The crude product was used directly in the next step without purification.1H NMR(600MHz,CDCl3):δ=8.42(s,1H),8.25(d,J=12Hz,1H),7.69(d,J=6Hz,1H),3.64(t,J=9Hz,2H),3.55(t,J=6Hz,2H)。

At 0 deg.C, to 2, 3-dihydrobenzo [ b ]]To a solution of thiophene-6-sulfonyl chloride 1, 1-dioxide (0.45g, 1.68mmol) in acetone (1mL) was added aqueous NH3(2mL,28%NH4OH in H2O), the resulting reaction mixture was stirred at room temperature for 2 hours or until completion. The solvent was removed in vacuo and azeotroped with toluene (. times.2). Chromatography on silica gel using 4% MeOH/CH2Cl2Eluent purification of the crude residue to give an off-white solid2, 3-dihydrobenzo [ b ]]Thiophene-6-sulfonamide 1, 1-dioxide (0.16mg, 39%).1H-NMR(DMSO-d6):δ=8.09(s,1H),8.06(d,J=12Hz,1H),7.75(d,J=6Hz,1H),7.60(bs,2h)3.70(t,J=6Hz,2H),3.44(t,J=9Hz,2H)。

3-azidobenzenesulfonamides

Synthesized according to the procedure contained in Pawan Kumar, Navneet Chandak, Poul Nielsen, Pawan k.sharma.bioorg.med.chem.2012,20, 3843-. 3-Aminobenzenesulfonamide (0.3g, 1.7mmol) in CH 3The solution in CN (8mL) was cooled to 0 ℃. To the stirred mixture was added t-BuONO (250. mu.L, 2.1mmol) followed by TMSN3(276. mu.L, 2.1 mmol). The resulting solution was stirred at room temperature for 1 hour. The reaction mixture was concentrated in vacuo and the crude product was purified by silica gel column chromatography using 100% hexane eluent to give the title compound (0.31g, 91%) as a light yellow solid.1H NMR(600MHz,CD3OD)δ7.68–7.62(m,1H),7.56(d,J=1.8Hz,1H),7.47(d,J=7.8Hz,1H),7.19–7.15(m,1H)。13C NMR(151MHz,DMSO-d6)δ146.2,140.8,131.2,122.9,122.4,116.5。

N- (3-sulfamoylphenyl) pent-4-ynylamides

To a solution of pent-4-ynoic acid (0.1g, 1.02mmol) and 3-aminobenzenesulfonamide (0.21g, 1.22mmol) in anhydrous DMF (5.0mL) was added HBTU (0.46g, 1.22mmol) followed by DIPEA (212uL, 1.22 mmol). The reaction mixture was stirred at ambient temperature for 2 hours, or until completion. The mixture was diluted with EtOAc (30mL) and washed with H2O (20mL), brine (20mL) and then dried (MgSO)4) The organics were concentrated in vacuo. The crude product was purified by silica gel column chromatography using 100% hexane eluent to give the title compound as a pale yellow solid (0.2g,79%)。1H NMR(400MHz,CD3OD)δ=8.22(dd,J=2.2,1.7Hz,1H),7.75–7.68(m,1H),7.65–7.58(m,1H),7.51–7.42(m,2H),2.64–2.59(m,2H),2.58–2.54(m,2H),2.32–2.25(m,1H)。13C NMR(101MHz,CD3OD)δ=171.3,143.8,138.9,129.2,122.9,121.0,117.1,82.1,69.1,35.4,14.0。

benzene-1, 3-disulfonamides

Benzene-1, 3-disulfonyl chloride (0.50g, 0.726mmol) was dissolved in tetrahydrofuran (4mL) and the solution was cooled to 0 ℃. Aqueous ammonia (0.4mL) was added at 0 ℃ and the mixture was stirred at ambient temperature for 1 hour. After completion of the reaction, the mixture was poured into cooling water and extracted with ethyl acetate. The combined organic extracts were washed with brine and dried (Na) 2SO4) And concentrated in vacuo. The resulting solid was triturated with pentane to give the title compound as a light brown solid (0.16g, 87%).1H NMR(400MHz,DMSO-d6):δ=8.27(t,J=2.0Hz,1H),8.06(dd,J=2.0,8.0Hz,2H),7.81(t,J=8.0Hz,1H),7.64(s,4H)。

N1,N1-dimethylbenzene-1, 3-disulfonamides

The 3-amino-N, N-dimethylbenzenesulfonamide was converted to 3- (N, N-dimethylsulfamoyl) benzenesulfonyl chloride (0.45g, 80%) using method D.1H NMR(300MHz,CDCl3) δ is 8.42(t, J ═ 2.0Hz,1H),8.27(d, J ═ 7.9Hz,1H),8.14(d, J ═ 7.9Hz,1H),7.85(t, J ═ 7.9Hz,1H),2.79(s, 6H). Sulfonyl chloride was converted using general procedure E1 to give the title compound as a yellow solid (0.45g, 93%).1H NMR(400MHz,DMSO-d6)δ=8.13(m,2H),7.98(d,J=7.9Hz,1H),7.87(t,J=7.9Hz,1H),7.65(s,2H),2.65(s,6H)。

3-sulfamoylbenzoic acid methyl ester

Methyl 3- (chlorosulfonyl) benzoate (1.00g, 4.26mmol) was dissolved in anhydrous tetrahydrofuran (15mL) and the solution was cooled to 0 ℃. Aqueous ammonia (5.0mL) was added dropwise and the mixture was stirred at ambient temperature for 2 hours. After completion, the reaction mixture was poured into cooling water and extracted with ethyl acetate. The combined organic extracts were washed with brine and dried (Na)2SO4) And concentrated in vacuo. The resulting solid was triturated with pentane to give the title compound as a light brown solid (0.75g, 82%).1H NMR(400MHz,DMSO-d6):δ=8.40(s,1H),8.19(d,J=8Hz,1H),8.1(d,J=8Hz,1H),7.77(t,J=8Hz,1H),7.6(s,2H),3.92(s,3H);m/z 214.0[M–H+]-

3- (4-phenyl-1H-1, 2, 3-triazol-1-yl) benzenesulfonamide

Ethynylbenzene (1 equivalent) and 3-azidobenzenesulfonamide (1.2 equivalents), 5 mol% CuSO, were reacted at room temperature 410 mol% NaAsc solution in DMSO (500. mu.L) was stirred for 12 hours. The crude product was directly purified from the reaction mixture by reverse phase column chromatography (revieris flash column chromatography, 4g, 18mL/min) and lyophilized to give the product as a white solid (32mg, 70%).1H NMR(400MHz,DMSO-d6)δ=9.57–9.36(m,1H),8.46(d,J=5.7Hz,1H),8.20(s,1H),7.98(d,J=8.1Hz,3H),7.88(d,J=7.6Hz,1H),7.62(s,2H),7.53(d,J=7.4Hz,2H),7.42(d,J=7.5Hz,1H)。

N- (prop-2-yn-1-yl) -3- (4-sulfamoylphenyl) propanamide

To 3- (4-sulfamoylphenyl) propionic acid (0.3g, 1.5)mmol) and propargylamine (0.11g, 1.5mmol) in anhydrous DMF (5.0ml) HBTU (0.74g, 1.5mmol) was added followed by DIPEA (342uL, 1.22 mmol). The reaction mixture was stirred at room temperature for 2 hours. The reaction was monitored by LCMS and, upon completion, it was diluted with EtOAc (30mL) and washed with H2O (20mL), brine (20 mL). Separating the organic layer; drying (MgSO)4) And evaporated to give the crude product. The crude product was purified by silica gel column chromatography (1:1, EtOAc: hexanes) to isolate the title compound (0.22g, 63%) as a white solid.1H NMR(400MHz,CD3OD)δ=7.85(d,J=7.9Hz,2H),7.38(d,J=7.9Hz,2H),3.97(t,J=2.4Hz,2H),3.04(t,J=7.6Hz,2H),2.55(t,J=7.6Hz,2H),2.33(d,J=2.8Hz,1H)。

Benzo [ d ] [1,3] dioxole-5-sulfonamides

Sulfuryl chloride (2.18ml, 26.7mmol) was added to anhydrous DMF (2.10ml, 26.7mmol) under nitrogen at 0 deg.C, then the ice bath was removed and the solution was stirred for 15 minutes. The solution was cooled again to 0 ℃ and benzo [ d ] was added ][1,3]Dioxoles. The reaction mixture was allowed to reach room temperature and then heated at 100 ℃ for 2 hours. The reaction mixture was poured onto crushed ice, stirred for 5 minutes and then extracted with dichloromethane (100mL then 2 x 50 mL). The combined organics were dried (MgSO)4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 15% DCM-hexane eluent to give benzo [ d ] as an off-white solid][1,3]Dioxol-5-sulfonyl chloride (1.78g, 33%).1H NMR(400MHz,CDCl3):δ=7.64(d,J=8.0Hz,1H),7.43(s,1H),6.95(d,J=8.0Hz,1H),6.16(s,2H)。

At 0 ℃ to benzo [ d][1,3]To a solution of dioxole-5-sulfonyl chloride (0.30g, 1.35mmol) in acetone (1mL) was added aqueous ammonia (1.5mL, 28% NH)4OH in H2O), the reaction mixture is stirred at room temperature until completion, typically for 2 hours, and then concentrated in vacuo. Washing by silica gel column chromatography with 2% MeOH-DCMThe crude product was purified by elimination of the solvent to give the title compound as an off-white solid (210mg, 77%).1H NMR(400MHz,DMSO-d6):δ=7.32(d,J=8.0Hz,1H),7.25(s,1H),7.21(bs,2H),7.02(d,J=8.0Hz,1H),6.11(s,2H)。

Pyridine-4-sulfonamides

Pyridine-4-sulfonyl chloride was converted using general method E3 to give the title compound as a pale yellow solid (50mg, 56%).1H NMR(300MHz,DMSO-d6):δ=8.56(d,J=4.5Hz,1H),7.49(d,J=4.5Hz,1H),7.24(br.s.,2H)。

Pyridine-3-sulfonamides

Pyridine-3-sulfonyl chloride was converted using general method E3 to give the title compound as a light yellow solid (0.7g, 79%). 1H NMR(300MHz,DMSO-d6)δ=8.96(dd,J=2.5,0.9Hz,1H),8.77(dd,J=4.8,1.6Hz,1H),8.17(ddd,J=8.0,2.4,1.6Hz,1H),7.67–7.56(m,3H)。

Pyridine-2-sulfonamides

A solution of 1.0M HCl (45mL) and DCM (45mL) was cooled to-10 ℃ and pyridine-2-thiol (1.0g, 9.0mmol) was added. After 10 minutes, NaOCl (6% solution, 47mL, 3.3 equiv.) was added dropwise over 5 minutes and stirring continued at-10 ℃ for 10 minutes. Separating the organic phase with Na2SO4Dried and filtered. The resulting solution was added dropwise to a pre-cooled solution of saturated methanolic ammonia and DCM (1:1,40mL) at 0 ℃, then allowed to warm to ambient temperature and stirred to completion, typically for 2 hours. The solvent was removed in vacuo to give a white solid, which was dissolved in heatEtOAc and filtered to remove solid impurities. The solvent was removed in vacuo and recrystallized from EtOAc-hexanes to give the title compound as a yellow solid (0.5g, 35%).1H NMR(300MHz,DMSO-d6)δ=8.70(ddd,J=4.7,1.7,0.9Hz,1H),8.05(td,J=7.7,1.7Hz,1H),7.91(dt,J=7.9,1.1Hz,1H),7.62(ddd,J=7.6,4.7,1.2Hz,1H),7.45(s,2H)。

4- (trifluoromethyl) pyridine-2-sulfonamides

The 4- (trifluoromethyl) pyridine-2-sulfonamide was synthesized according to the procedure used to synthesize pyridine-2-sulfonamide but using 4- (trifluoromethyl) pyridine-2-thiol instead of pyridine-2-thiol. The product, 4- (trifluoromethyl) pyridine-2-sulfonamide (0.7g, 56%) was obtained as a solid.1H NMR(300MHz,DMSO-d6)δ=9.02(d,J=5.0Hz,1H),8.16(s,1H),8.07(d,J=5.0Hz,1H),7.68(s,2H)。

3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide

3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) aniline was converted to 3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonyl chloride as a yellow liquid using general procedure D (1.1g, 52%). 1H NMR(300MHz,CDCl3)δ=8.15–8.08(m,1H),7.82–7.77(m,1H),7.76–7.68(m,1H),7.68–7.61(m,1H)。19F NMR(282MHz,CDCl3)δ-65.06。

3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonyl chloride was converted using general method E2 to the title compound as a white solid (0.6g, 60%).1H NMR(300MHz,CDCl3)δ=7.99(dt,J=7.9,1.5Hz,1H),7.71(t,J=2.0Hz,1H),7.60(t,J=7.9Hz,1H),7.49(d,J=7.9Hz,1H),4.87(s,2H)。19F NMR(282MHz,CDCl3)δ-65.13。

2- (methyl (7-nitrobenzo [ c ] [1,2,5] oxadiazol-4-yl) amino) -N- (4-sulfamoylphenethyl) acetamide

2- (methylamino) acetic acid (0.24g, 2.75mmol) and sodium bicarbonate (0.694g, 8.26mmol) were dissolved in a mixture of water (10mL) and MeOH (20 mL). 4-chloro-7-nitrobenzo [ c ] [1,2,5] oxadiazole (0.50g, 2.50mmol) was then added and the mixture was stirred at 60 ℃ for 2 h. After completion of the reaction, the volatiles were removed under reduced pressure and the resulting crude residue was purified by silica gel column chromatography eluting with a gradient of 0-5% methanol in dichloromethane to give 2- (methyl (7-nitrobenzo [ c ] [1,2,5] oxadiazol-4-yl) amino) acetic acid as a brick-red solid (1.10g, 87%).

Under nitrogen atmosphere, 2- (methyl (7-nitrobenzo [ c ]) is reacted][1,2,5]Oxadiazol-4-yl) amino) acetic acid (1.00g, 3.96mmol) was dissolved in anhydrous tetrahydrofuran (25mL) and the solution was cooled to 0 ℃. Diisopropylethylamine (0.76g, 5.55mmol) and 1,1' -carbonyldiimidazole (0.90g, 4.75mmol) were added and the mixture was stirred at 50 ℃ until all of the 2- (methyl (7-nitrobenzo [ c ] s) ][1,2,5]Oxadiazol-4-yl) amino) acetic acid. The reaction mixture is then cooled to 0 ℃, 4- (2-aminoethyl) benzenesulfonamide (0.95g, 4.75mmol) is added, and stirring is continued at ambient temperature until completion, typically for 6 hours. The solvent was removed in vacuo and the residue was purified by reverse phase preparative HPLC to give the title compound as a brick red solid (1.20g, 70%). LCMS (M/z):435.4(M +1)+

4- (2- (7-nitrobenzo [ c ] [1,2,5] oxadiazol-4-ylamino) ethyl) benzenesulfonamide

4- (2-aminoethyl) benzenesulfonamide (0.55g, 2.75mmol) and diisopropylethylamine (0.64g, 2.75mmol) were dissolved in ethanol (20mL) and the solution was cooled to 0 ℃. Adding 4-chloro-7-nitrobenzo [ c ] at 0 deg.C][1,2,5]Oxadiazole (0.50g, 2.50mmol) and the mixture was stirred at ambient temperature for 16 h. After completion of the reaction, the reaction was poured into brine and extracted with ethyl acetate. The solvent was evaporated from the combined organic extracts under reduced pressure and the resulting crude product was purified by reverse phase preparative HPLC to give the title product as a dark yellow solid (0.250g, 7%).1H NMR(400MHz,CD3OD)δ=8.5(d,J=8.8Hz,1H),7.82(d,J=8.4Hz,2H),7.47(d,J=8.4Hz,2H),6.35(d,J=8.8Hz,1H),3.83(m,2H),3.15(t,J=7.6Hz,2H)。

2- (7- (dimethylamino) -2-oxo-2H-chroman-4-yl) -N- (4-sulfamoylphenethyl) acetamide (17 l):

2- (7- (dimethylamino) -2-oxo-2H-chroman-4-yl) acetic acid (0.50g, 2.02mmol), EDC. HCl (0.47g, 3.03mmol), HOBt (0.464g, 3.03mmol) and N-methylmorpholine (0.409g, 4.04mmol) were mixed in dry tetrahydrofuran (5mL) at 0 deg.C and stirred for 30 min. 4- (2-aminoethyl) benzenesulfonamide (0.445g, 2.224mmol) was added and stirring continued at ambient temperature for 18 h. After completion, the reaction was poured into cooling water and extracted with ethyl acetate. The solvent was removed in vacuo and the residue was purified by silica gel column chromatography using a 0-5% gradient of methanol in dichloromethane to give 2- (7- (dimethylamino) -2-oxo-2H-chroman-4-yl) -N- (4-sulfamoylphenethyl) acetamide as a green-yellow solid (0.25g, 29%). LCMS (M/z):430.2(M +1)+

6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazole-2-sulfonamides

A solution of 3-chlorobutyronitrile (20g, 193.1mmol) in diethyl ether (100mL) was treated with MeOH (7.41g, 231.7mmol) and cooled to 0 deg.C. HCl gas was bubbled into the reaction mixture for 4 hours at 0 ℃. The reaction mixture was stirred at-20 ℃ for 24 hours and concentrated in vacuo. The resulting solid residue was washed with diethyl ether (3X 100mL), n-pentane (2X 100mL) and dried under vacuum at 45 ℃ to give 4-chlorobutyrimido hydrochloride as a white solid.

Methyl 4-chlorobutyrimidoate hydrochloride (25g, 146.1mmol) was dissolved in Et3N (44.3g, 4.38mmol) in DCM (250mL) and the resulting solution cooled to 0 ℃. To the above mixture was added dropwise 2, 2-dimethoxyethan-1-amine (12.2g, 116.9mmol) over a period of 5 minutes. The resulting reaction mixture was warmed to 60 ℃ and stirred for 3 hours. The reaction mixture was concentrated in vacuo, and the resulting residue was treated with formic acid (150mL) and heated at 80 ℃ for 24 hours. After completion, the reaction mixture was concentrated in vacuo and the residue obtained was azeotroped with toluene (2 × 100 mL). The crude mixture was taken up with saturated NaHCO3The solution was basified and extracted with DCM (3X 200 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And vacuum concentrating to obtain 6, 7-dihydro-5H-pyrrolo [1,2-a ] as a low melting black solid]Imidazole (8g, over 3 steps 39%).1H NMR(300MHz,CDCl3):δ=7.0(s,1H),6.83(s,1H),3.95(t,J=7.2Hz,2H),2.84(t,J=7.2Hz,2H),2.61-2.51(m,2H)。

Reacting 6, 7-dihydro-5H-pyrrolo [1,2-a ]]A solution of imidazole (4g, 37.0mmol) in acetonitrile (120mL) was cooled to 0 ℃. N-iodosuccinimide (9.16g, 40.7mmol) was added portionwise at 0 ℃. The resulting reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was taken up with saturated Na2S2O3The solution was diluted and extracted with ethyl acetate (2X 50 mL). The combined organic extracts were washed with water, brine and dried (Na) 2SO4) And concentrated in vacuo. Purifying the crude product by silica gel column chromatography using 4-40% EtOAc-hexane eluent to obtain 2-iodo-6, 7-dihydro-5H-pyrrolo [1,2-a ] as a white solid]Imidazole (1.0g, 19%).1H NMR(300MHz,CDCl3):δ=7.03(s,1H),3.89(t,J=7.2Hz,2H),3.02(t,J=7.2Hz,2H),2.65-2.55(m,2H)。LCMS(m/z):235[M+H]+。

2-iodo-6, 7-dihydro-5H-pyrrolo [1,2-a ] was treated with DIPEA (0.41g, 3.20mmol) in a 50mL resealable reaction tube under nitrogen at room temperature]A solution of imidazole (0.3g, 1.28mmol) and phenylmethanethiol (0.24g, 1.92mmol) in 1, 4-dioxane (10 mL). Nitrogen was purged through the solution for 5 minutes. Xantphos (74mg, 0.128mmol) and Pd2(dba)3(60mg, 0.064mmol) was added to the above solution in order, and the tube was purged with nitrogen for 5 minutes. The resulting mixture was stirred at 110 ℃ for 12 hours. After completion, the mixture was cooled to room temperature, diluted with EtOAc (25mL) and filtered through celite. The filtrate was dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 50-70% EtOAc-hexane eluent to give 2- (benzylsulfanyl) -6, 7-dihydro-5H-pyrrolo [1,2-a ] as a brown liquid]Imidazole (0.15g, 51%).1H NMR(300MHz,CDCl3):δ=7.26-7.22(m,3H),7.12(s,1H),7.05-7.02(m,2H),3.73(s,2H),3.14(t,J=6.9Hz,2H),2.81(t,J=7.2Hz,2H),2.32-2.27(m,2H)。LCMS(m/z):231.3[M+H]+

Reacting 2- (benzylthio) -6, 7-dihydro-5H-pyrrolo [1,2-a ]]Imidazole (250mg, 1.08mmol) in acetonitrile (2.5mL), acetic acid (0.5mL) and H 2The solution in O (1.2mL) was cooled to 0 ℃. DCDMH (170mg, 0.869mmol) was added at 0 deg.C and the resulting reaction mixture was stirred at 0 deg.C for 2 hours. The reaction mixture was diluted with water and extracted with ethyl acetate (2X 20 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo to give 6, 7-dihydro-5H-pyrrolo [1,2-a ] as a pale brown liquid]Imidazole-2-sulfonyl chloride, which was used directly in the next step.

Reacting 6, 7-dihydro-5H-pyrrolo [1,2-a ]]A solution of imidazole-2-sulfonyl chloride (300mg) in THF (15mL) was cooled to-40 ℃. Ammonia gas was purged through the above solution for 15 minutes and the solution was stirred at-40 ℃ for 1 hour. The reaction mixture was warmed to room temperature, stirred for 1 hour, and then concentrated in vacuo after completion. Column chromatography on silica gel (60-120 mesh) using 10% MeOH-CHCl3Purifying the crude product with an eluent to obtain 6, 7-dihydro-5H-pyrrolo [1,2-a ] as a white solid]Imidazole-2-sulfonamide (117mg,88%)。1H NMR(300MHz,DMSO-d6):δ=7.54(bs,2H),7.26(s,1H),4.06(s,2H),2.80(s,2H),2.32(s,2H)。LCMS(m/z):187.95[M+H]+

4-Nitrobenzenesulfonamides

4-Nitrobenzenesulfonyl chloride (1.0 equiv.) dissolved in acetone (0.8mL/mmol) was added dropwise to ammonium bicarbonate (4.0 equiv.) dissolved in water (0.8 mL/mmol). The reaction mixture was stirred at room temperature for 2 hours and then acidified with 1M HCl (pH 2). The mixture was extracted with ethyl acetate (3X 10mL) and dried (MgSO) 4) And concentrated in vacuo to give the title compound as a pale orange solid (157mg, 57%).1H NMR(600MHz,DMSO-d6)δ=8.42(d,J=8.8Hz,2H),8.06(d,J=8.8Hz,2H),7.74(s,2H)。C6H5N2O4S1[M-H]-HRMS calculated of (a): 200.9976, found: 200.9984.

5-methyl-N- (4-sulfamoylphenethyl) isoxazole-3-carboxamide

5-Methylisoxazole-3-carbonyl chloride (1.0 eq.) (prepared using general method B1) was dissolved in anhydrous THF (4mL/mmol) and treated with triethylamine (1.0 eq.). After stirring for 5 minutes, 4- (2-aminoethyl) benzenesulfonamide (1.0 eq) was added to the acid chloride solution. The reaction was stirred at room temperature under argon overnight. The solvent was removed in vacuo and the residue was purified by reverse phase column chromatography with acetonitrile/10 mM ammonium bicarbonate (aq) as the mobile phase to give the title compound as a white solid (205mg, 48%).1H NMR(600MHz,DMSO-d6)δ=8.79(t,J=5.8Hz,1H),7.74(d,J=8.3Hz,2H),7.41(d,J=8.3Hz,2H),7.30(s,2H),6.50(q,J=0.6Hz,1H),3.52–3.46(m,2H),2.91(t,J=7.2Hz,2H),2.45(d,J=0.6Hz,3H)。C13H14N3O4S1[M-H]-HRMS calculated of (a): 308.0711, found: 308.0708.

4- (2- (7-methoxy-4, 4-dimethyl-1, 3-dioxo-3, 4-dihydroisoquinolin-2 (1H) -yl) ethyl) benzenesulfonamide

N- (cyclohexylcarbamoyl) -4- (2- (7-methoxy-4, 4-dimethyl-1, 3-dioxo-3, 4-dihydroisoquinolin-2 (1H) -yl) ethyl) benzenesulfonamide (1.0 eq) dissolved in anhydrous pyridine (8mL/mmol) was treated with phthalic anhydride (1 eq) and DMAP (0.1 eq) and heated to reflux under an inert atmosphere for 4 hours. The solvent was removed in vacuo and the residue was purified by reverse phase column chromatography with acetonitrile/10 mM ammonium bicarbonate (aq) as the mobile phase to give the title compound as a white solid (291mg, 75%). 1H NMR(600MHz,DMSO-d6)δ=7.72(d,J=8.4Hz,2H),7.61(d,J f=8.7Hz,1H),7.53(d,J=2.8Hz,1H),7.40(d,J=8.2Hz,2H),7.33–7.25(m,3H),4.13(d,J=7.5Hz,2H),3.83(s,3H),2.93(t,J=7.3Hz,2H),1.45(s,6H)。C20H21N2O5S1[M-H]-HRMS calculated of (a): 401.1177, found: 401.1174.

synthesis of R1 and R2 amine intermediates:

1-methyl-1H-pyrazol-3-amine HCl

In a 20mL microwave vial, a solution of 2-chloroacrylonitrile (2g, 22.85mmol) in EtOH (10mL) was treated with methylhydrazine (1.93g, 41.13 mmol). The resulting reaction mixture was heated in a Biotage microwave synthesizer at 100 ℃ for 10 minutes. The reaction mixture is added in<The mixture was left at 5 ℃ for 12 hours, during which time the solid precipitated. The precipitate was removed by filtration and dried in vacuo to give the title compound as a white solid (0.12g, 55%).1H NMR(400MHz,DMSO-d6):δ7.76(s,1H),6.13(s,1H),3.80(s,3H),2.58(s,2H)。LCMS(m/z):98.3(M+1)+

1- (trifluoromethyl) -1H-pyrazol-3-amine

3-Nitro-1H-pyrazole (5g, 44mmol) was dissolved in N, N-dimethylformamide (100mL), cooled to-5 ℃ and NaH (3.8g, 93.6mmol) was added portionwise. The reaction mixture was stirred for 15 min, then dibromodifluoromethane (8.6g, 44mmol) was added and allowed to warm to ambient temperature overnight. The reaction mixture was quenched with ice water and extracted with ethyl acetate. The organic phase was washed with water, brine and dried (Na)2SO4) And concentrated in vacuo to give 3-nitro-1- (trifluoromethyl) -1H-pyrazole (2.1g, 22%), which was used without further purification.19F NMR(282MHz,CDCl3)δ=-34.20。

1- (bromodifluoromethyl) -3-nitro-1H-pyrazole (2.1g, 9.76mmol) was dissolved in DCM (50mL) and cooled to-78 deg.C, then AgBF was added 4(5.7g, 28.3, 3 equivalents). The reaction mixture was allowed to warm to ambient temperature overnight, then cooled to 0 ℃ and quenched by the addition of saturated aqueous NaHCO3 (50 mL). The aqueous phase was extracted with DCM and the combined organics were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo to give 3-nitro-1- (trifluoromethyl) -1H-pyrazole (0.9g, 51%).19F NMR(282MHz,CDCl3)δ=-60.96。

3-Nitro-1- (trifluoromethyl) -1H-pyrazole (1.0g) was dissolved in THF EtOAc (1:1,50mL), Pd/C (200mg) was added, and the mixture was stirred under a hydrogen atmosphere (balloon) overnight. The mixture was filtered through celite and washed by using ethyl acetate. The solvent was removed in vacuo and the residue was purified by silica gel column chromatography using 40% EtOAc in hexanes to give the title product (0.75g, 87%).1H NMR(300MHz,CDCl3)δ=7.54(d,J=2.7Hz,1H),5.84(d,J=2.7Hz,1H)。19F NMR(282MHz,CDCl3)δ=-61.13。

1-isopropyl-1H-pyrazol-3-amine

In turn with K2CO3(12.5g, 91mmol) and 2-chloroacrylonitrile (4g, 45.45mmol) Isopropylhydrazine hydrochloride (5g, 45.45mmol) in water (40mL) was treated. The resulting reaction mixture was stirred at 50 ℃ for 1 hour, cooled to room temperature, and extracted with ethyl acetate (50 mL). The organic extracts were washed with water (40mL), brine (40mL) and dried (Na)2SO4) And concentrated in vacuo to give the title compound as a yellow solid (3.5g, 62%). 1H NMR(400MHz,CDCl3):δ=7.15(s,1H),5.56(s,1H),4.27-4.23(m,1H),3.6(br.s.,2H),1.43(d,J=6.4Hz,6H)。LCMS(m/z):126.0(M+1)+

1-cyclopropyl-1H-pyrazol-3-amines

A solution of 1H-pyrazol-3-amine (2g, 24.1mmol) in AcOH (20mL) was treated with 2, 5-hexanedione (5.7g, 50.6mmol) at ambient temperature under a nitrogen atmosphere. The resulting reaction mixture was heated to 100 ℃ for 6 hours. The reaction mixture was concentrated under reduced pressure and azeotroped with toluene. The crude product was purified by silica gel column chromatography using a gradient of 50-100% EtOAc-hexanes eluent to give 3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1H-pyrazole as a red solid (2.25g, 59%).1H NMR(400MHz,DMSO-d6)δ12.92(s,1H),7.85(t,J=1.8Hz,1H),6.28(t,J=2.1Hz,1H),5.75(s,2H),2.00(s,6H)。

Copper (II) acetate (0.56g, 3.1mmol), 2; bipyridine (0.48g, 3.1mmol) and dichloroethane (10mL) were heated to 75 ℃ for 20 minutes. 5mL of this pre-solution was added to a mixture of 3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1H-pyrazole (0.5g, 3.1mmol), potassium cyclopropyltrifluoroborate (2 equiv.) and sodium carbonate (2 equiv.) in dichloroethane (5mL)The reaction was then stirred at 75 ℃ for 6 hours. The reaction mixture was diluted with DCM, washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes eluent to give 1-cyclopropyl-3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1H-pyrazole as a yellow liquid (0.2g, 32%). 1H NMR (300MHz, chloroform-d) δ 7.48(dd, J ═ 2.3,0.5Hz,1H),6.12(d, J ═ 2.3Hz,1H),5.84(s,2H),3.61(tt, J ═ 7.3,3.6Hz,1H),2.09(s,6H), 1.22-0.95 (m, 4H).

To a solution of ammonium hydroxide hydrochloride (1.64g, 11.8mmol) in ethanol ((10mL) at 0 deg.C was added a solution of potassium hydroxide (0.66g) in water (10mL) after stirring for 10 minutes, a solution of 1-cyclopropyl-3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1H-pyrazole (0.95g) in ethanol (10mL) was added and the reaction was heated at 100 deg.C for 20 hours, the solvent was removed in vacuo and the residue was partitioned between ethyl acetate and water, the organic phase was washed with water, brine, dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using a gradient of 70-100% EtOAc-hexanes eluent to give the title product as a brown solid (0.4g, 69%).1H NMR(400MHz,CDCl3)δ=7.51(d,J=2.4Hz,1H),6.69(d,J=2.4Hz,1H),5.04(s,2H),3.67(m,1H),1.19(m,2H),1.12(m,2H)。

1- (tert-butyl) -1H-pyrazol-3-amine

Isopropylhydrazine hydrochloride (1.42g, 11.4mmol) in water (25mL) was sequentially reacted with K at 0 deg.C2CO3(1.57g,11.4mmol)、NaHCO3(1.91g, 22.9mmol) and 2-chloroacrylonitrile (1g, 11.4mmol) and then warmed to ambient temperature and stirred for 12 hours. The reaction mixture was diluted with water (20mL) and extracted with ethyl acetate (2X 25 mL). The combined organics were washed with water (30mL), brine (30mL) and dried (Na) 2SO4) And concentrated in vacuo to give the title compound (0.9g, 60%) as a brown liquid.1H NMR(400MHz,DMSO-d6):δ=7.34(s,1H),5.35(s,1H),4.51(br.s.,2H),1.40(s,9H)。LCMS(m/z):140.10(M+1)+

1-cyclohexyl-1H-pyrazol-3-amines

3-Nitro-1H-pyrazole (1g, 8.85mmol) was dissolved in N, N-dimethylformamide (20mL) and treated with potassium carbonate (1.47g, 10.62mmol) and bromocyclohexane (1.8g, 10.62 mmol). The mixture was heated to 100 ℃ for 16 hours (or until completion), then cooled to ambient temperature, diluted with water (100mL), and extracted with ethyl acetate (2 × 75 mL). The combined organics were washed with water (100mL), brine (100mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using a 10% gradient of EtOAc-hexanes eluent to give 1-cyclohexyl-3-nitro-1H-pyrazole as a colorless liquid (1.3g, 76%).1H NMR(300MHz,CDCl3)δ=7.47(d,J=2.5Hz,1H),6.88(d,J=2.5Hz,1H),4.26–4.11(m,1H),2.18(m,2H),1.93(m,2H),1.82-1.20(m,6H)。

A solution of 1, 5-dimethyl-3-nitro-1H-pyrazole (0.65g, 3.3mmol) in MeOH (4mL) and EtOAc (20mL) was treated with 10% palladium on carbon (200mg) under a nitrogen atmosphere in a 100mL Parr's shaker reaction vessel. The flask was evacuated and then filled with hydrogen (60psi) and stirred at ambient temperature for 12 hours. The reaction mixture was diluted with ethyl acetate (50mL) and filtered through a celite bed. The filtrate was dried (Na)2SO4) And concentrated in vacuo to give 1-cyclohexyl-1H-pyrazol-3-amine (0.3g, 55%) as a light brown solid. 1H NMR(300MHz,CDCl3)δ=7.15(d,J=2.3Hz,1H),5.56(d,J=2.3Hz,1H),3.85(m,1H),3.62(s,1H),2.1(m,2H),1.8(m,2H),1.77–1.10(m,6H)。

1-phenyl-1H-pyrazol-3-amines

Potassium tert-butoxide (11.9g, 106.3mmol) was dissolved in tBuOH (100mL) and the solution was heated to 100 ℃. Phenylhydrazine (5g, 46.2mmol) and 3-ethoxyacrylonitrile (4.5g, 46.2mmol) were added successively and heating was continued for 16 h. The mixture was concentrated in vacuo. The resulting residue was partitioned between water (500mL) and ethyl acetate (500 mL). The organic extracts were washed with water (250mL), brine (250mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 25% EtOAc-hexanes eluent to give 1-phenyl-1H-pyrazol-3-amine (3.5g, 48%) as a light brown solid.1H NMR(400MHz,CDCl3):δ=7.69(s,1H),7.57(d,J=8.0Hz,2H),7.41(d,J=8.0Hz,2H),7.2(t,J=7.6Hz,1H),5.85(s,1H),3.83(br.s.,2H)。LCMS(m/z):160.3(M+1)+

1-benzyl-1H-pyrazol-3-amines

A solution of 3-nitro-1H-pyrazole (1g, 8.85mmol) in THF (20mL) was cooled to 0 deg.C and NaH (0.53g, 13.27mmol) was added. The suspension was stirred for 20 min, then benzyl bromide (1.5g, 8.85mmol) was added dropwise. The reaction was stirred until completion, about 6 hours, with saturated NaHCO3The solution (20mL) was diluted and extracted with EtOAc (2X 50 mL). The organics were washed with water (30mL), brine (30mL) and dried (Na)2SO4) And concentrated in vacuo to give 1-benzyl-3-nitro-1H-pyrazole (1.5g, 84%) as a white solid.1H NMR(300MHz,CDCl3):δ=7.40-7.36(m,4H),7.31-7.27(m,2H),6.90(d,J=2.7Hz,1H),5.37(s,2H)。LCMS(m/z):204.20(M+1)+

A solution of 1-benzyl-3-nitro-1H-pyrazole (1.5g, 7.39mmol) in THF (20mL) and MeOH (5mL) was cooled to 0 ℃. Zinc powder (2.4g, 36.9mmol) and NH were added 4Cl solution (1.97g, 36.94 mmol; in 5mL water). The resulting reaction mixture was heated at 70 ℃ for 12 hours. The reaction mixture was cooled to ambient temperature, diluted with EtOAc (50mL) and filtered through a celite bed. Drying the filtrate (a)Na2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 50% EtOAc-hexanes eluent to give 1-benzyl-1H-pyrazol-3-amine (0.85g, 67%) as a light brown liquid.1H NMR(300MHz,CDCl3):δ=7.34-7.26(m,3H),7.14-7.11(m,2H),7.05(d,J=2.4Hz,1H),5.59(d,J=2.4Hz,1H),5.14(s,2H)。LCMS(m/z):174.10(M+1)+

1- (1-phenylethyl) -1H-pyrazol-3-amine

A solution of 3-nitro-1H-pyrazole (1g, 8.85mmol) in THF (20mL) was cooled to 0 deg.C and NaH (0.7g, 17.7mmol) was added. The suspension was stirred for 30 min, then (1-bromoethyl) benzene (1.96g, 10.6mmol) was added dropwise. The reaction was heated to 80 ℃ overnight or until completion, cooled to ambient temperature, diluted with water (40mL), and extracted with EtOAc (2X 50 mL). The organics were washed with water (30mL), brine (30mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 20% EtOAc-hexanes eluent to give 3-nitro-1- (1-phenylethyl) -1H-pyrazole as a yellow liquid (1.2g, 63%).1H NMR(300MHz,CDCl3)δ7.46–7.18(m,6H),6.88(d,J=2.5Hz,1H),5.59(q,J=7.1Hz,1H),1.96(d,J=7.1Hz,3H)。

A solution of 3-nitro-1- (1-phenylethyl) -1H-pyrazole (1g, 4.6mmol) in THF (20mL) and MeOH (5mL) was cooled to 0 ℃. Zinc powder (1.49g, 23.04mmol) and NH were added 4Cl solution (1.23g, 23.04 mmol; in 5mL water). The resulting reaction mixture was stirred for 30 minutes and then heated at 80 ℃ for 6 hours. The reaction mixture was cooled to ambient temperature, diluted with EtOAc (50mL) and filtered through a celite bed. The organic phase was washed with water (20mL), brine (20mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 50% EtOAc-hexanes eluent to give 1- (1-phenylethyl) -1H-pyrazol-3-amine (0.85g, 67%) as a yellow liquid.1H NMR(300MHz,CDCl3)δ=7.39–7.04(m,6H),5.59(d,J=2.4Hz,1H),5.35(q,J=7.1Hz,1H),3.83(s,2H),1.78(d,J=7.1Hz,3H)。

1- (2- (piperidin-1-yl) ethyl) -1H-pyrazol-3-amine

A solution of 3-nitro-1H-pyrazole (2g, 17.7mmol) in DMF (20mL) was treated with 1- (2-chloroethyl) piperidine hydrochloride (4.8g, 26.5mmol) at ambient temperature. The solution was cooled to 0 ℃ and over a period of 5 minutes with K2CO3(6.1g, 44.27mmol) was processed batchwise. The resulting reaction mixture was stirred at ambient temperature for 4 hours. The reaction mixture was diluted with water and extracted with ethyl acetate (2X 40 mL). The combined organics were washed with water (40mL), brine (40mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 25% EtOAc-hexanes eluent to give 1- (2- (3-nitro-1H-pyrazol-1-yl) ethyl) piperidine (2.5g, 64%) as a pale yellow solid. 1H NMR(400MHz,CDCl3):δ=7.60(d,J=2.4Hz,1H),6.81(d,J=2.4Hz,1H),4.29(t,J=6.4Hz,2H),2.78(t,J=6.4Hz,2H),2.41(s,4H),1.57-1.53(m,4H),1.45(t,J=6Hz,2H)。LCMS(m/z):225.10(M+1)+

A solution of 1- (2- (3-nitro-1H-pyrazol-1-yl) ethyl) piperidine 3(2.5g, 11.16mmol) in THF (20mL) and MeOH (5mL) was cooled to 0 ℃. The solution was washed successively with zinc dust (3.6g, 55.8mmol) and NH4Aqueous Cl (3g, 55.8mmol) was treated and then warmed to ambient temperature and stirred for 5 hours. The reaction mixture was diluted with ethyl acetate (50mL), filtered through a celite bed and concentrated in vacuo. The residue was diluted with ethyl acetate (60mL) and washed with water (40mL), brine (40mL), dried (Na)2SO4) And concentrated in vacuo to give 1- (2- (piperidin-1-yl) ethyl) -1H-pyrazol-3-amine (1.75g, 81%) as a pale yellow liquid.1H NMR(400MHz,DMSO-d6):δ=7.28(d,J=2Hz,1H),5.30(s,J=2Hz,1H),4.50(s,2H),3.90(t,J=6.8Hz,2H),2.5-2.53(m,4H),2.39-2.33(m,6H),1.2(s,2H)。LCMS(m/z):195.10(M+1)+

1, 5-dimethyl-1H-pyrazol-3-amine

A solution of 5-methyl-3-nitro-1H-pyrazole (2g, 15.7mmol) in THF (20mL) was cooled to 0 deg.C. NaH (0.7g, 17.32mmol) was added portionwise over 10 minutes under nitrogen. The resulting suspension was stirred for 10 min, then treated with MeI (2.2g, 15.7mmol), warmed to ambient temperature and stirred for 4 h. The reaction mixture was washed with saturated NH4The Cl solution (20mL) was diluted and extracted with EtOAc (2X 30 mL). The organics were washed with water (30mL), brine (30mL) and dried (Na)2SO4) And concentrated in vacuo to give 1, 5-dimethyl-3-nitro-1H-pyrazole (2g, 91%) as a white solid. 1H NMR(300MHz,CDCl3):δ=6.71(s,1H),3.87(s,3H),2.34(s,3H)。

A solution of 1, 5-dimethyl-3-nitro-1H-pyrazole (2g, 14.18mmol) in MeOH (4mL) and EtOAc (20mL) was treated with 10% palladium on carbon (400mg) under a nitrogen atmosphere in a 100mL Parr's shaker reaction vessel. The flask was evacuated and then filled with hydrogen (60psi) and stirred at ambient temperature for 12 hours. The reaction mixture was diluted with ethyl acetate (50mL) and filtered through a celite bed. The filtrate was dried (Na)2SO4) And concentrated in vacuo to give 1, 5-dimethyl-1H-pyrazol-3-amine (1.36g, 87%) as a light brown solid.1H NMR(400MHz,DMSO-d6):δ=5.19(s,1H),4.33(br.s.,2H),3.43(s,3H),2.07(s,3H)。LCMS(m/z):112.3(M+1)+

1-methyl-5- (trifluoromethyl) -1H-pyrazol-3-amine

A solution of 1,1, 1-triethoxyethane (20g, 123mmol) in DCM (250mL) and pyridine (20.5g, 259mmol) was cooled to 0 ℃. Trifluoroacetic anhydride (52g, 246mmol) in DCM (50) was added dropwise over 30 minmL). The reaction mixture was warmed to ambient temperature, stirred for 12 hours, and then saturated NaHCO3The aqueous solution was diluted and extracted with DCM (2X 250 mL). The combined organics were washed with water (20mL), brine (20mL) and dried (Na)2SO4) And concentrated in vacuo to give 4, 4-diethoxy-1, 1, 1-trifluorobut-3-en-2-one (20g, 76%) as a light brown liquid.1H NMR(400MHz,CDCl3):δ=4.93(s,1H),4.39(q,J=7.2Hz,2H),4.18(q,J=7.2Hz,4H),1.46-1.40(m,6H)。

By NH at 0 deg.C3A solution of 4, 4-diethoxy-1, 1, 1-trifluorobut-3-en-2-one (10g, 47.16mmol) in acetonitrile (100mL) was treated with an aqueous solution (15mL) and then stirred at room temperature for 12 hours. The reaction mixture was concentrated in vacuo, then the residue was treated with water (250mL) and extracted with DCM (2X 250 mL). The combined organics were washed with water (250mL), brine (250mL) and dried (Na) 2SO4) And concentrated in vacuo to give (E) -4-amino-4-ethoxy-1, 1, 1-trifluorobut-3-en-2-one (7.5g, 87%) as an off-white solid.1H NMR(300MHz,CDCl3):δ=5.6(br.s.,1H),4.17(q,J=7.2Hz,2H),1.42(t,J=7.2Hz,3H)。

At ambient temperature, with hydrazine methylsulfate (4.72g, 32.8mmol) and Et3A solution of (E) -4-amino-4-ethoxy-1, 1, 1-trifluorobut-3-en-2-one (5g, 27.3mmol) in EtOH (30mL) was treated with N (4.1g, 41.0 mmol). The resulting reaction mixture was heated at 85 ℃ for 12 hours, then cooled to ambient temperature and concentrated in vacuo. The residue obtained is taken up in saturated NaHCO3Aqueous (250mL) was diluted and extracted with EtOAc (2X 250 mL). The combined organics were washed with water (250mL), brine (250mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 20% EtOAc-hexanes eluent to give 1-methyl-5- (trifluoromethyl) -1H-pyrazol-3-amine (0.17g, 38%) as a light brown liquid.1H NMR(400MHz,CDCl3):δ=5.93(s,1H),3.79(s,3H),3.68(br.s.,2H)。

1-methyl-5- (prop-1-en-2-yl) -1H-pyrazol-3-amine

A solution of 1-methyl-1H-pyrazol-3-amine (2g, 20.6mmol) in AcOH (50mL) was treated with 2, 5-hexanedione (4.9g, 43.29mmol) at ambient temperature under a nitrogen atmosphere. The resulting reaction mixture was heated to 100 ℃ for 1 hour and then stirred at ambient temperature for 5 hours. The reaction mixture was concentrated under reduced pressure and azeotroped with toluene. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes eluent to give 3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1-methyl-1H-pyrazole as a liquid (2.5g, 69%). 1H NMR(300MHz,CDCl3):δ=7.39(d,J=2.1Hz,1H),6.15(d,J=2.4Hz,1H),5.84(s,2H),3.92(s,3H),2.10(s,6H)。

A solution of 3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1-methyl-1H-pyrazole (1g, 5.71mmol) in anhydrous THF (10mL) was cooled to-78 deg.C under a nitrogen atmosphere, n-BuLi (1.6M in hexane, 4.4mL, 6.86mmol) was added dropwise over a period of 10 minutes to the solution, which was then stirred at-78 deg.C for 1 hour, then treated with I at-78 deg.C2(1.54g, 5.71mmol) in THF (5mL) and stirring continued at this temperature until completion (2 h). The reaction mixture was washed with saturated NH4Aqueous Cl was quenched and extracted with ethyl acetate (2X 25 mL). The combined organics were washed with water (20mL), brine (20mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 50% EtOAc-hexanes eluent to give 3- (2, 5-dimethyl-1H-pyrrol-1-yl) -5-iodo-1-methyl-1H-pyrazole as an off-white solid (0.75g, 43.6%).1H NMR(400MHz,CDCl3):δ=6.33(s,1H),5.84(s,2H),3.95(s,3H),2.09(s,6H)。

Dioxolanyl borane (0.67g, 3.98mmol) and Na were used at ambient temperature under nitrogen with 4,4,5, 5-tetramethyl-2- (prop-1-en-2-yl) -1,3, 2-cyclopentylborane2CO3(0.52g, 4.98mmol) A solution of 3- (2, 5-dimethyl-1H-pyrrol-1-yl) -5-iodo-1-methyl-1H-pyrazole (1g, 3.32mmol) in DME: water (8:2,10mL) was treated. The resulting solution was degassed by purging with argon for 15 minutes, then with Pd (P) under argon atmosphere Ph3)4(190mg, 0.166 mmol). The resulting mixture was heated at 90 ℃ for 24 hours, then cooled to ambient temperature and concentrated in vacuo. The resulting residue was diluted with cold water (20mL) and extracted with EtOAc (2X 20 mL). The combined organics were washed with water (20mL), brine (20mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 5% EtOAc-hexanes eluent to give 3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1-methyl-5- (prop-1-en-2-yl) -1H-pyrazole as a light yellow liquid (0.765g, 92%).1H NMR(300MHz,CDCl3):δ=6.08(s,1H),5.84(s,2H),5.39(s,1H),5.23(s,1H),3.92(s,3H),2.12(s,9H)。

By NH2Treatment of 3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1-methyl-5- (prop-1-en-2-yl) -1H-pyrazole (0.7g, 3.25mmol) in EtOH-H with OH & HCl (2.26g, 32.55mmol) and KOH (1.8g, 32.55mmol)2Solution in O (8:2,12 mL). The resulting reaction mixture was heated at 100 ℃ for 48 hours. The reaction mixture was cooled and concentrated in vacuo. With saturated NaHCO3The residue was worked up to give a solution of pH 8, then extracted with EtOAc (2X 50 mL). The combined organics were washed with water (20mL), brine (20mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 100% EtOAc eluent to give 1-methyl-5- (prop-1-en-2-yl) -1H-pyrazol-3-amine (0.4g, 91%) as a light brown liquid. 1H NMR(400MHz,CDCl3):δ=5.54(s,1H),5.27(s,1H),5.08(s,1H),3.71(s,3H),2.41(s,2H),1.92(s,3H)。

1-benzyl-3-nitro-1H-pyrazole-5-carboxylic acid ethyl ester

A solution of 3-nitro-1H-pyrazole-5-carboxylic acid (5g, 31.8mmol) in ethanol (50mL) was treated dropwise with thionyl chloride (4.5g, 38.2mmol) at 0 ℃ over a period of 10 minutes under a nitrogen atmosphere. The resulting mixture was stirred at 80 ℃ for 6 hours, then cooled to ambient temperature and concentrated in vacuo. The residue obtained is taken up in saturated NaHCO3The solution was basified to pH 8 and then extracted with ethyl acetate (2X 100 mL). The combined organics were washed with water (100mL), brine (100mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was triturated with diethyl ether, filtered and dried under reduced pressure to give ethyl 3-nitro-1H-pyrazole-5-carboxylate (5g, 85%) as a white solid.1H NMR(400MHz,DMSO-d6):δ=7.44(s,1H),4.36(q,J=6.8Hz,2H),1.33(t,J=7.2Hz,3H)。LCMS(m/z):184(M-1)-

3-Nitro-1H-pyrazole-5-carboxylic acid ethyl ester (1g, 5.4mmol) was dissolved in DMF (10mL) at ambient temperature and washed with K2CO3(1.34g, 9.7 mmol). The resulting mixture was cooled to 0 ℃ and methyl iodide (1.15g, 8.1mmol) was added dropwise, the reaction mixture was sealed, allowed to warm to ambient temperature and stirred for 12 hours. The reaction mixture was diluted with water (20mL) and extracted with EtOAc (2X 30 mL). The combined organics were washed with water (30mL), brine (30mL) and dried (Na) 2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 5% EtOAc-hexanes eluent to give 1-methyl-3-nitro-1H-pyrazole-5-carboxylic acid ethyl ester (0.65g, 61%) as a white solid.1H NMR(400MHz,DMSO-d6)δ=7.54(s,J=1.1Hz,1H),4.35(q,J=7.1Hz,2H),4.19(d,J=1.2Hz,3H),1.33(t,J=7.1Hz,3H)。

Ethyl 1-methyl-3-nitro-1H-pyrazole-5-carboxylate (0.65g, 3.3mmol) was dissolved in THF (20mL) and MeOH (5mL) at 0 ℃. Zinc powder (1.0g, 16.3mmol) and NH were added in this order4Aqueous Cl (0.87g, 16.3 mmol). The resulting reaction mixture was stirred at ambient temperature for 4 hours, then heated to 70C for 1 hour. The solvent was removed in vacuo. The resulting residue was dissolved in EtOAc (30mL) and filtered through a bed of celite. The filtrate was washed with water (30mL), brine (30mL) and dried (Na)2SO4) And concentrated in vacuo to give ethyl 3-amino-1-methyl-1H-pyrazole-5-carboxylate (0.5g, 91%) as a white solid.1H NMR(400MHz,DMSO-d6)δ5.95(s,1H),4.24(q,J=7.1Hz,2H),3.83(s,3H),3.41(s,2H),1.27(t,J=7.1Hz,4H)。

1-benzyl-3-nitro-1H-pyrazole-5-carboxylic acid ethyl ester

A solution of 3-nitro-1H-pyrazole-5-carboxylic acid (5g, 31.8mmol) in ethanol (50mL) was treated dropwise with thionyl chloride (4.5g, 38.2mmol) at 0 ℃ over a period of 10 minutes under a nitrogen atmosphere. The resulting mixture was stirred at 80 ℃ for 6 hours, then cooled to ambient temperature and concentrated in vacuo. The residue obtained is taken up in saturated NaHCO 3The solution was basified to pH 8 and then extracted with ethyl acetate (2X 100 mL). The combined organics were washed with water (100mL), brine (100mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was triturated with diethyl ether, filtered and dried under reduced pressure to give ethyl 3-nitro-1H-pyrazole-5-carboxylate (5g, 85%) as a white solid.1H NMR(400MHz,DMSO-d6):δ=7.44(s,1H),4.36(q,J=6.8Hz,2H),1.33(t,J=7.2Hz,3H)。LCMS(m/z):184(M-1)-

Ethyl 3-nitro-1H-pyrazole-5-carboxylate (2g, 10.8mmol) was dissolved in DMF (10mL) at ambient temperature and washed with K2CO3(3g, 21.6 mmol). The resulting mixture was cooled to 0 ℃ and benzyl bromide (2.7g, 16.2mmol) was added dropwise and the reaction mixture was allowed to warm to ambient temperature and stirred for 2 hours. The reaction mixture was diluted with water (20mL) and extracted with EtOAc (2X 30 mL). The combined organics were washed with water (30mL), brine (30mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 5% EtOAc-hexanes eluent to give ethyl 1-benzyl-3-nitro-1H-pyrazole-5-carboxylate (1.2g, 40%) as a white solid.1H NMR(400MHz,CDCl3):δ=7.44(s,1H),7.34-7.31(m,5H),5.83(s,2H),4.39(q,J=7.2Hz,2H),1.38(t,J=7.2Hz,3H)。LCMS(m/z):276.15(M+1)+

Ethyl 1-benzyl-3-nitro-1H-pyrazole-5-carboxylate (1.2g, 4.36mmol) was dissolved in THF (20mL) and MeOH (5mL) at 0 ℃. Zinc powder (1.4g, 21.8mmol) and NH were added successively4Aqueous Cl (1.16g, 21.8 mmol). Subjecting the obtained product to The reaction mixture was stirred at ambient temperature for 4 hours and then concentrated in vacuo. The resulting residue was dissolved in EtOAc (30mL) and filtered through a bed of celite. The filtrate was washed with water (30mL), brine (30mL) and dried (Na)2SO4) And concentrated in vacuo to give ethyl 3-amino-1-benzyl-1H-pyrazole-5-carboxylate (1g, 94%) as a white solid.1H NMR(300MHz,DMSO-d6):δ=7.30-7.19(m,3H),7.11-7.08(m,2H),6.00(s,1H),5.43(s,2H),4.91(s,2H),4.23(q,J=7.2Hz,2H),1.24(t,J=7.2Hz,3H)。LCMS(m/z):245.9(M+1)+

3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1-phenyl-1H-pyrazole

A solution of 1-phenyl-1H-pyrazol-3-amine (3.5g, 21.9mmol) in acetic acid (20mL) was treated with 2, 5-hexanedione (5.2g, 45.9mmol) and heated to 100 ℃ for 4 hours. The mixture was allowed to cool and concentrated in vacuo. The crude product was purified by silica gel column chromatography using 5% EtOAc-hexanes eluent to give 3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1-phenyl-1H-pyrazole as a colorless liquid (2.8g, 54%).1H NMR(400MHz,CDCl3):δ=7.98(s,1H),7.74(d,J=8.4Hz,2H),7.49(d,J=8.4Hz,2H),7.32(t,J=7.6Hz,1H),6.39(s,1H),5.9(s,2H),2.19(s,6H)。

A solution of 3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1-phenyl-1H-pyrazole (2.7g, 11.4mmol) in THF (70mL) was treated dropwise with n-BuLi (1.6M in THF, 10mL, 23.91mmol) at-78 deg.C over 10 min. The reaction mixture was stirred at-78 ℃ for 1.5 h, then treated with fresh dry acetone (1g, 17.0mmol) and stirred at-78 ℃ for a further 1.5 h. The reaction mixture was quenched with saturated ammonium chloride (2mL), concentrated in vacuo, and partitioned between water (100mL) and ethyl acetate (100 mL). The organic extracts were washed with water (100mL), brine (100mL) and dried (Na) 2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 20% EtOAc-hexanes eluent to give 2- (3- (2, 5-bis) as an off-white solidmethyl-1H-pyrrol-1-yl) -1-phenyl-1H-pyrazol-5-yl) propan-2-ol (1.4g, 42%).1H NMR(400MHz,CDCl3):δ=7.57-7.56(m,2H),7.47-7.46(m,3H),6.23(s,1H),5.85(s,2H),2.19(s,6H),1.52(s,6H)。LCMS(m/z):296.1(M+1)+

2- (3- (2, 5-dimethyl-1H-pyrrol-1-yl) -1-phenyl-1H-pyrazol-5-yl) propan-2-ol (1.4g, 4.74mmol) was dissolved in EtOH-H in a 100mL resealable reaction tube at ambient temperature2O (1:1,50 mL). Hydroxylamine hydrochloride (3.3g, 47.45mmol) and KOH (2.6g, 47.45mmol) were added successively and the resulting reaction mixture was heated at 120 ℃ for 16 hours. The reaction mixture was concentrated in vacuo, diluted with water (50mL), and extracted with ethyl acetate (2X 50 mL). The organic extracts were washed with water (50mL), brine (50mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 50% EtOAc-hexanes eluent to give 2- (3-amino-1-phenyl-1H-pyrazol-5-yl) propan-2-ol (0.8g, 78%) as a colorless liquid. LCMS (M/z):218.1(M +1)+

8-bromo-1, 2,3,5,6, 7-hexahydro-s-indacen-4-amine

N-bromosuccinimide (1.02g, 5.78mmol) was added portionwise to a solution of 1,2,3,5,6, 7-hexahydro-s-indacen-4-amine (1g, 5.78mmol) in DCM (20mL) at 0 ℃. The solution was gradually warmed to ambient temperature and stirred for 12 hours. The reaction mixture was saturated with Na 2S2O3The solution (50mL) was diluted and extracted with DCM (2X 25 mL). The combined organic extracts were washed with water (25mL), brine (25mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 5% EtOAc-hexanes eluent to give 8-bromo-1, 2,3,5,6, 7-hexahydro-s-indacen-4-amine as a brown solid (1.2g, 83%).1H NMR(300MHz,CDCl3):δ=3.45(br.s.,2H),2.92-2.88(m,4H),2.81-2.77(m,4H),2.16-2.09(m,4H);LC-MS 94%(210nM);m/z 252.15[M+H]+

8-chloro-1, 2,3,5,6, 7-hexahydro-s-indacen-4-amine

N-chlorosuccinimide (0.46g, 3.46mmol) was added portionwise to 1,2,3,5,6, 7-hexahydro-s-indacen-4-amine, 1(0.6g, 3.46mmol) in CHCl at 0 deg.C3(10 mL). The solution was gradually warmed to ambient temperature and stirred for 10 hours. The reaction mixture was saturated with Na2S2O3Aqueous solution (50mL) was diluted and extracted with DCM (2X 25 mL). The combined organic extracts were washed with water (25mL), brine (25mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes eluent to give 8-chloro-1, 2,3,5,6, 7-hexahydro-s-indacen-4-amine as a brown solid (0.45g, 63%).1H NMR(300MHz,CDCl3):δ=2.94(t,J=7.2Hz,4H),2.77(t,J=8.1Hz,4H),2.18(m,4H);m/z 207.8[M+H]+

8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacen-4-amine

8-bromo-1, 2,3,5,6, 7-hexahydro-s-4-amine (400mg, 1.59mmol) was dissolved in 1, 4-dioxane-water (8:2,10mL) and the reaction flask was purged with argon for 15 minutes. Adding K in turn under argon atmosphere 2CO3(650mg, 4.78mmol), methylboronic acid (100mg, 1.75mmol) and Pd (PPh)3)4(100mg, 0.079 mmol). The resulting mixture was sealed and heated at 100 ℃ for 2 hours. The reaction mixture was cooled, diluted with water and extracted with EtOAc (2X 20 mL). The combined organic extracts were washed with water (25mL), brine (25mL) and dried (Na)2SO4) And concentrated in vacuo. Purify the crude product by silica gel column chromatography using 5% EtOAc-hexanes eluent to give 8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacen-4-amine (b) as a colorless liquid0.220g,76%)。1H NMR(400MHz,CDCl3):δ=3.41(br.s.,2H),2.88-2.8(m,J=7.5Hz,4H),2.75-2.67(m,4H),2.18-2.09(m,7H);m/z 188.2[M+H]+

3,5,6, 7-tetrahydro-2H-indeno [5,6-b ] furan-8-amine

A solution of 2, 3-dihydrobenzofuran-5-carbaldehyde (10g, 67.6mmol), malonic acid (10.5g, 101.35mmol), and piperidine (0.47mL, 4.73mmol, 0.07 equiv.) was heated in pyridine (60mL) at 100 ℃ for 5 hours. The reaction mixture was acidified with 1N HCl topH 3, and the product extracted with 10% IPA/chloroform (2X 250 mL). The combined organic extracts were washed with water (250mL), brine (250mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was triturated with diethyl ether to give (E) -3- (2, 3-dihydrobenzofuran-5-yl) acrylic acid (10g, 78%) as a yellow solid.1H NMR (300MHz, chloroform-d) δ 7.73(d, J15.9 Hz,1H),7.43(s,1H),7.33(dd, J8.1, 1.8Hz,1H),6.80(d, J8.1 Hz,1H),6.29(d, J15.9 Hz,1H),4.64(t, J8.7 Hz,2H),3.24(t, J8.7 Hz, 2H).

A solution of (E) -3- (2, 3-dihydrobenzofuran-5-yl) acrylic acid (8.0g, 42.1mmol) in acetic acid (80mL) and water (1.0mL) was treated with 10% palladium on carbon (1.0g) in two portions. The reaction mixture is stirred under atmosphere or hydrogen (balloon) until completion, typically for 4 hours. The mixture was diluted with ethyl acetate (100mL) and filtered through a celite bed, followed by washing with ethyl acetate. The solvent was removed in vacuo and the crude residue azeotroped with toluene (2X 50mL) to give an off-white solid, which was triturated with diethyl ether (50mL) to give 3- (2, 3-dihydrobenzofuran-5-yl) propionic acid as a white solid (6.5g, 80%).1H NMR(400MHz,CDCl3)δ=7.04(s,1H),6.93(d,J=8.4,1H),6.7(d,J=8.4Hz,1H),4.55(t,J=8.4Hz,2H),3.18(t,J=8.4Hz,2H),2.89(t,J=7.6Hz,2H),2.64(t,J=7.6Hz,2H)。

A solution of 3- (2, 3-dihydrobenzofuran-5-yl) propionic acid (6.0g, 31mmol) in thionyl chloride (8mL) was heated at 80 ℃ for 1 hour. After completion of the reaction, thionyl chloride was removed in vacuo and the crude 3- (2, 3-dihydrobenzofuran-5-yl) propionyl chloride was dissolved in dry 1, 2-dichloroethane (30 mL). In a separate flask, aluminum trichloride (2g, 15mmol) was added to anhydrous 1, 2-dichloroethane (40mL) at 0 deg.C, then the acid chloride solution (10mL) was added dropwise over 5 minutes, and the resulting solution was stirred at 0 deg.C for 30 minutes. Another portion of aluminum trichloride (3g, 22.5mmol) was added, followed by dropwise addition of the remaining acid chloride solution (20mL) at 0 ℃. The reaction mixture was stirred at room temperature for 1 hour or until completion, diluted with water and extracted with EtOAc (2 × 50 mL). The combined organic extracts were washed with 1N HCl (50mL), 1N NaOH (50mL), water (25mL) and brine (25mL) and dried (Na) 2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes eluent to give 2,3,5, 6-tetrahydro-7H-indeno [5,6-b ] as a white solid]Furan-7-one (3.8g, 70%).1H NMR(300MHz,CD3OD)δ=7.36(s,1H),6.91(s,1H),4.61(t,J=8.6Hz,3H),3.26(t,J=8.6Hz,2H),3.05(t,J=5.5Hz,3H),2.68(t,J=5.5Hz,2H)。

At 0 ℃, reacting 2,3,5, 6-tetrahydro-7H-indeno [5,6-b ]]Furan-7-one (1.5g, 8.61mmol) was dissolved in concentrated H2SO4(6.0mL), then f.HNO3: concentrated H was added dropwise at 0 deg.C2SO41:1(1.2mL), stirring was continued for 1 hour. The reaction mixture was added to ice-cold water (60mL) and stirred for 10 min, and the resulting light brown ppt was removed by filtration, washed with ice-cold water (20mL) and dried in vacuo to give 8-nitro-2, 3,5, 6-tetrahydro-7H-indeno [5,6-b]Furan-7-one (1.2g, 64%).1H NMR(300MHz,CD3OD)δ=7.54(s,1H),4.80(t,J=8.6Hz,2H),3.42(t,J=8.6Hz,2H),3.09(t,J=5.6Hz,2H),2.74(t,J=5.6Hz,2H)。

8-Nitro-2, 3,5, 6-tetrahydro-7H-indeno [5,6-b ] is treated with methanesulfonic acid (0.2mL) followed by 20% palladium hydroxide (0.5g) at 0 deg.C]Solution of furan-7-one (1.0g, 4.57mmol) in methanol (20 mL). The reaction mixture was stirred under atmosphere or under hydrogen (60psi)Stirring until the stirring is finished. The reaction mixture was filtered through a celite bed, washed with methanol (50mL) and concentrated in vacuo. The residue was diluted with ethyl acetate (50mL) and washed with saturated aqueous NaHCO3 (50mL), water (20mL), brine (20mL), dried (Na) 2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes eluent to give 3,5,6, 7-tetrahydro-2H-indeno [5,6-b ] as a white solid]Furan-8-amine (0.5g, 63%).1H NMR(300MHz,CDCl3)δ=6.54(s,1H),5.30(s,2H),4.61(t,J=8.7Hz,2H),3.21(t,J=8.7Hz,2H),2.95(t,J=5.5Hz,2H),2.66(t,J=5.5Hz,2H)。

4-bromo-3, 5,6, 7-tetrahydro-2H-indeno [5,6-b ] furan-8-amine

3,5,6, 7-tetrahydro-2H-indeno [5,6-b ] in dichloromethane (6.0mL) was treated dropwise with a solution of pivaloyl chloride (0.41g, 3.43mmol) in DCM (4.0mL) at 0 deg.C]Furan-8-amine (0.5g, 2.86mmol) and triethylamine (0.51mL, 3.71 mmol). The reaction was stirred at ambient temperature for 6 hours. The reaction mixture was added to saturated NaHCO3Aqueous (30mL) and extracted with DCM (2X 25 mL). The combined organics were washed with water (25mL), brine (25mL) and dried (Na)2SO4) And concentrated in vacuo to give N- (3,5,6, 7-tetrahydro-2H-indeno [5,6-b ] as a white solid]Furan-8-yl) pivaloamide (0.55g, 74%).1H NMR (300MHz, chloroform-d) δ 6.91(s,1H),4.56(t, J8.6 Hz,2H),3.17(t, J8.6 Hz,2H),2.83(t, J7.4 Hz,2H),2.75(t, J7.4 Hz,2H),2.04(p, J7.4 Hz,2H),1.32(s, 9H).

N- (3,5,6, 7-tetrahydro-2H-indeno [5,6-b ] in acetic acid (10mL) was treated dropwise with a solution of bromine (0.4g, 2.55mmol) in acetic acid (2.0mL) ]Furan-8-yl) pivalamide (0.55g, 2.12mmol) and the reaction was stirred at ambient temperature for 3 hours. Ice-cold water was added to the reaction mixture and stirred for 10 minutes. The resulting precipitate was removed by filtration, washed with water (20mL), and dried in vacuo to afford N-(4-bromo-3, 5,6, 7-tetrahydro-2H-indeno [5, 6-b)]Furan-8-yl) pivaloamide (0.65g, 91%).1H NMR (300MHz, chloroform-d) δ 6.94(s,1H),4.61(t, J8.7 Hz,2H),3.18(t, J8.7 Hz,2H), 2.92-2.80 (m,4H),2.06(p, J7.4 Hz,2H),1.31(s, 9H).

N- (4-bromo-3, 5,6, 7-tetrahydro-2H-indeno [5,6-b ] in EtOH (10mL) and concentrated HCl (15mL) was heated at 90 deg.C]Furan-8-yl) pivaloamide (0.6g, 1.78mmol) for 36 hours. The solution was concentrated in vacuo and then treated with NH4Basifying the OH aqueous solution. The aqueous phase was extracted with ethyl acetate (2 × 20mL) and the combined organics were dried (Na)2SO4) And concentrated in vacuo to give 4-bromo-3, 5,6, 7-tetrahydro-2H-indeno [5,6-b ] as a brown solid]Furan-8-amine (0.3g, 67%).1H NMR (400MHz, chloroform-d) δ 4.61(t, J8.6 Hz,2H),3.49(s,2H),3.17(t, J8.6 Hz,2H),2.84(t, J7.4 Hz,2H),2.78(t, J7.4 Hz,2H),2.12(p, J7.4 Hz, 2H).

3,5,6, 7-tetrahydro-2H-indeno [5,6-b ] furan-4-amine

3,5,6, 7-tetrahydro-2H-indeno [5,6-b ] in ethanol (10mL) and acetic acid (1.5mL) was treated with a solution of sodium nitrate (1.3g, 19.8mmol) in water (3.0mL)]Furan-8-amine (0.5g, 1.98mmol) and the reaction stirred at ambient temperature for 4 hours. Ethanol was removed in vacuo, then the residue was diluted with water (30mL), extracted with 10% IPA/chloroform (2X 25mL), and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 5% EtOAc-hexanes eluent to give 4-bromo-3, 5,6, 7-tetrahydro-2H-indeno [5,6-b ] as a yellow solid]Furan (0.28g, 60%).

4-bromo-3, 5,6, 7-tetrahydro-2H-indeno [5,6-b ] in DMSO (10mL) was treated with copper iodide (0.22g, 1.18mmol), L-proline (0.21g, 1.88mmol), and sodium azide (0.19g, 2.94mmol)]Furan (0.28g, 1.18 mmol). The reaction mixture was heated in a sealed tube at 135 ℃ for 36 hours. The reaction mixture was cooled, diluted with water and extracted with EtOAc (2X 25 mL). The combined organic extracts are usedWashed with water (25mL), brine (25mL), and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes eluent to give 3,5,6, 7-tetrahydro-2H-indeno [5,6-b ] as a gray solid ]Furan-4-amine (0.17g, 85%).1H NMR (300MHz, chloroform-d) δ 6.21(s,1H),4.59(t, J ═ 8.5Hz,2H),3.51(s,1H),2.98(t, J ═ 8.5Hz,2H),2.83(t, J ═ 7.5Hz,2H),2.64(t, J ═ 7.5Hz,2H),2.10(p, J ═ 7.5Hz, 2H).

Benzo [1,2-b:4,5-b' ] difuran-4-amines

2,3,6, 7-tetrahydrobenzo [1,2-b:4,5-b 'in anhydrous dioxane (20 mL)']Furan-4-carboxylic acid (0.8g, 3.88mmol), 2, 3-dichloro-5, 6-dicyanobenzoquinone (2.64g, 11.65mmol) were heated in a sealed tube at 120 ℃ for 18 hours. The reaction mixture was cooled to room temperature and saturated Na was added2S2O3Aqueous solution (30mL) and extracted with ethyl acetate (2X 25 mL). The combined organics were dried (Na)2SO4) And concentrated in vacuo to give crude benzo [1,2-b:4,5-b']Difuran-4-carboxylic acid (1.5 g). The crude acid (1.5g), triethylamine (2.05mL) and diphenylphosphorylazide (4.08g, 14.85mmol) in t-butanol (20mL) were heated in a sealed tube at 90 ℃ for 12 hours. The solution was cooled to room temperature, diluted with water (50mL), and extracted with EtOAc (2X 50 mL). The combined organic extracts were washed with water (25mL), brine (25mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes eluent to afford benzo [1,2-b:4,5-b' ]Tert-butyl difuran-4-ylcarbamate (0.75g), containing a small amount of impurities from the phosphine reagent, was dissolved in DCM (10mL) and TFA (3.0mL) was added dropwise over 5 min at 0 ℃. The reaction was stirred at ambient temperature for 2 hours, then saturated NaHCO was added carefully3Aqueous solution (50 mL). The aqueous phase was extracted with DCM (2X 30mL) and the combined organic extracts were washed with water (25mL), brine (25mL) and dried (Na)2SO4) Is true ofAnd (4) concentrating in air. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexane eluent to afford benzo [1,2-b:4,5-b 'as an off-white solid']Difuran-4-amine (0.2g, 30% in three steps).1H NMR(400MHz,CDCl3):δ=7.6(d,J=2.2Hz,1H),7.53(d,J=2.2Hz,1H),7.12(s,1H),6.78(m,2H),4.17(br.s.,1H)。

3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) aniline

To 2,2, 2-trifluoro-1-phenyleth-1-one (5g, 28.7mmol) in concentrated H at-5 deg.C2SO4(10mL) to the solution was added concentrated H2SO4HNO3(1:1,16mL) and the reaction mixture was stirred for 3 hours. The resulting solution was poured onto ice/water (100mL) and extracted with ethyl acetate (2X 100 mL). The combined organics were washed with water (100mL), brine (100mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 20% EtOAc-hexanes eluent to give 2,2, 2-trifluoro-1- (3-nitrophenyl) ethan-1-one (4.2g, 67%) as a yellow liquid. 1H NMR(400MHz,CDCl3)δ=8.92(s,1H),8.59(dd,J=8.1,1.4Hz,1H),8.41(d,J=7.8Hz,1H),7.82(t,J=8.1Hz,1H)。19F NMR(233.33MHz,CDCl3):-71.82(s,3F)。

A solution of 2,2, 2-trifluoro-1- (3-nitrophenyl) ethan-1-one (4.2g, 19.2mmol), hydroxylamine hydrochloride (4.0g, 57.5mmol), and pyridine (25mL) in ethanol (25mL) was heated at reflux for 3 hours, or until completion. The solvent was removed in vacuo and the crude product was purified by silica gel column chromatography using 40% EtOAc-hexanes eluent to give 2,2, 2-trifluoro-1- (3-nitrophenyl) ethan-1-one oxime (4.0g, 89%) as a colorless liquid.19F NMR(233.33MHz,CDCl3) -66.42 and 62.28(E oxime and Z oxime).

A solution of 2,2, 2-trifluoro-1- (3-nitrophenyl) ethan-1-one oxime (4.0g, 17.1mmol) in dichloromethane (20mL) was cooled to 0 deg.C with triethylamine (1.5 equiv.), N-dimethylaminePyridine (0.5 equiv), tosyl chloride (1.1 equiv) and stirred at ambient temperature until complete, typically 16 hours. The reaction mixture was diluted with dichloromethane (50mL) and saturated NH4Washed with aqueous Cl (100mL), water (100mL), brine (100mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 5% EtOAc-hexanes eluent to give 2,2, 2-trifluoro-1- (3-nitrophenyl) ethan-1-one o-toluenesulfonyl oxime (4.0g, 60%) as a white solid.1H NMR(300MHz,CDCl3)δ=8.41(ddd,J=5.6,3.5,2.2Hz,1H),8.21(t,J=1.5Hz,1H),7.89(d,J=8.3Hz,2H),7.81–7.65(m,2H),7.42(d,J=7.8Hz,2H),2.50(s,3H)。19F NMR(282MHz,cdcl3)δ-61.55,-66.90。

A solution of 2,2, 2-trifluoro-1- (3-nitrophenyl) ethan-1-one o-toluenesulfonyl oxime (4.0g, 10.3mmol) in diethyl ether was cooled to-78 deg.C and an ammonia solution was bubbled through the solution for 30 minutes. The reaction mixture was sealed, allowed to warm to ambient temperature, and then stirred for 16 hours. The mixture was filtered through a pad of celite and concentrated in vacuo. The crude product was purified by silica gel column chromatography using 7% EtOAc-hexanes eluent to give 3- (3-nitrophenyl) -3- (trifluoromethyl) bisaziridine (2.4g, 100%) as a colorless liquid. 1H NMR(300MHz,CDCl3)δ=8.52(t,J=2.0Hz,1H),8.33(ddd,J=8.3,2.3,1.1Hz,1H),7.99(d,J=8.0Hz,1H),7.65(tt,J=7.8,0.4Hz,1H),2.95(d,J=8.8Hz,1H),2.31(d,J=8.9Hz,1H)。19F NMR(282MHz,CDCl3)δ=-75.10。

A solution of 3- (3-nitrophenyl) -3- (trifluoromethyl) bisaziridine (2.4g, 10.3mmol) in methanol (30mL) was treated with triethylamine (2 equivalents) and iodine (1 equivalent) and the reaction mixture was stirred to completion, typically for 2 hours. The solution was diluted with diethyl ether, washed with 10% aqueous citric acid, water, aqueous sodium thiosulfate, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes eluent to give 3- (3-nitrophenyl) -3- (trifluoromethyl) -3H-bisaziridine (2.1g, 88%) as a colorless liquid.1H NMR(300MHz,CDCl3)δ8.30(ddd,J=7.9,2.2,1.4Hz,1H),8.09–8.01(m,1H),7.70–7.54(m,2H)。19F NMR(282MHz,CDCl3)δ=-65.14。

3- (3-Nitrophenyl) -3- (trifluoromethyl) -3H-bisaziridine (3.0g, 13mmol) in THF (70mL) was treated with a solution of sodium dithionate (10 equivalents) in water (30mL) and the mixture was stirred at ambient temperature until completion, typically overnight. The solution was diluted with water, extracted with ethyl acetate (× 2), washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 40% EtOAc-hexanes eluent to give the title compound 3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) aniline as a yellow solid (1.5g, 58%).1H NMR(300MHz,CDCl3)δ=7.16(t,J=7.9Hz,1H),6.70(ddd,J=8.1,2.3,0.9Hz,1H),6.52(ddt,J=7.9,1.9,0.9Hz,1H),6.45br.(.s,1H),3.77(s,2H)。19F NMR(282MHz,CDCl3)δ-65.07。

4, 6-di-tert-butylpyrimidin-2-amine

2,4, 6-trichloropyrimidine (2.7g, 14.7mmol) was dissolved in anhydrous THF (30mL) at 0 ℃ under a nitrogen atmosphere. CuI (280mg, 1.47mmol) was added to the above solution, which was then treated with 2M tert-butylmagnesium chloride in THF (3.78g, 16.15mL, 32.3mmol) at 0 deg.C under a nitrogen atmosphere. The resulting mixture was stirred at room temperature for 3 hours. After completion, the reaction mixture was taken up with saturated NH4The Cl solution was diluted and extracted with EtOAc (2X 50 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 100% hexane eluent to give 4, 6-di-tert-butyl-2-chloropyrimidine (1.3g, 39%) as a pale brown liquid.1H NMR(300MHz,CDCl3):δ=7.20(s,1H),1.33(s,18H)。LCMS(m/z):227.3[M+H]+

In a 100mL resealable reaction tube, 4, 6-di-tert-butylA solution of the group-2-chloropyrimidine (1.3g) in EtOH (15mL) was cooled to-50 ℃. Ammonia gas was purged through the solution for 15 minutes. The reaction mixture was warmed to 70 ℃ and stirred for 12 hours. Upon completion, the reaction mixture was concentrated in vacuo, and the resulting residue was diluted with water and extracted with ethyl acetate (50 mL). The organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo to give 4, 6-di-tert-butylpyrimidin-2-amine (0.7g, 59%) as a white solid. 1H NMR(300MHz,CDCl3):δ=6.64(s,1H),4.83(s,2H),1.26(s,18H)。LCMS(m/z):208.4[M+H]+

4-chloro-2, 6-diisopropylaniline

2, 6-diisopropylaniline (5.0g, 28.2mmol) in DMF (100mL) was treated with N-chlorosuccinimide (3.97g, 29.7mmol) and the reaction mixture was stirred at room temperature overnight. The solution was poured into water (500mL) and extracted with diethyl ether (2X 150 mL). The combined organics were washed with water (2X 200mL), brine (200mL), dried (MgSO)4) And concentrated in vacuo. The product was purified by short path distillation to give the title compound as a red oil (3.0g, 50%).1H NMR(600MHz,DMSO-d6):δ=6.84(s,2H),4.75(s,2H),3.01(hept,J=6.8Hz,2H),1.13(d,J=6.8Hz,12H)。13C NMR(151MHz,DMSO-d6):δ=141.1,133.8,122.5,120.5,27.2,22.8。

4-chloro-2, 6-dicyclopropylaniline

In a 50mL resealable reaction tube, 2, 6-dibromo-4-chloroaniline (0.25g, 0.88mmol) and cyclopropylboronic acid (0.22g, 2.62mmol) and K3PO4(0.74g, 3.50mmol) of the solution was dissolved in toluene: water (10 mL: 1 mL). The resulting solution was degassed by purging with nitrogen for 5 minutes. Adding intoPd(OAc)2(20mg, 0.087mmol) and tricyclohexylphosphine (25mg, 0.087mmol), and the solution was purged with nitrogen for an additional 5 minutes. The resulting mixture was stirred at 100 ℃ for 12 hours. After completion of the reaction, the mixture was diluted with water (25mL), extracted with EtOAc (2 × 25mL), and the combined organic extracts were washed with water, brine, dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 5% EtOAc-hexanes eluent to give 4-chloro-2, 6-dicyclopropylaniline (150mg, 83%) as a brown liquid. 1H NMR(300MHz,DMSO-d6):δ=6.69(s,2H),4.98(s,2H),1.74-1.64(m,2H),0.90-0.84(m,4H),0.52-0.47(m,4H)。LCMS(m/z):208.30[M+H]+

Synthesis of 4-chloro-2-methyl-6- (trifluoromethyl) aniline

A solution of 2-methyl-6- (trifluoromethyl) aniline (0.4g, 2.20mmol) in acetonitrile (4mL) and AcOH (0.3mL) was cooled to 0 ℃. N-chlorosuccinimide (0.36g, 2.70mmol) was added at 0 deg.C, then the solution was warmed to room temperature and stirred for 12 hours. After completion of the reaction, the reaction mixture was diluted with ice-cold water, and the resulting precipitate was removed by filtration and successively saturated NaHCO3、Na2S2O3The solution, washed with n-pentane and dried in vacuo to give 4-chloro-2-methyl-6- (trifluoromethyl) aniline as a white solid (0.25g, 52%).1H NMR(400MHz,CDCl3):δ=7.30(d,J=2.4Hz,1H),7.18(d,J=2.0Hz,1H),2.17(s,3H)。19F NMR(400MHz,CDCl3):δ=-63.03

4-chloro-2, 6-diethylaniline

Using K at room temperature under argon3PO4(1.5g, 7.0mmol) 2, 6-dibromo-4-chloroaniline (0.5g, 1.75 mm)ol) and ethyl boronic acid (0.4g, 5.25mmol) in toluene (15mL) and water (4 mL). The solution was purged with argon for 5 minutes, then Pd (OAc)2(40mg, 0.175mmol) and tricyclohexylphosphine (50mg, 0.175 mmol). The reaction mixture was again purged with argon for 5 minutes. The resulting mixture was stirred at 100 ℃ for 12 hours. After completion, the reaction mixture was diluted with water, extracted with EtOAc (2 × 25mL), and the combined organic extracts were washed with water, brine, and dried (Na) 2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 8% EtOAc-hexanes eluent to give 4-chloro-2, 6-dicyclopropylaniline (100mg, 31%) as a yellow liquid.1H NMR(300MHz,CDCl3):δ=6.94(s,2H),3.61(s,2H),2.53(q,J=7.5Hz,4H),1.27(t,J=7.5Hz,6H)。LCMS(m/z):184.00[M+H]+

4-chloro-2, 6-dimethoxyaniline

2, 6-dibromo-4-chloroaniline (4g, 14.0mmol) was dissolved in 25% NaOMe solution in MeOH (48mL) and treated with CuI (2.9g, 15.4mmol) at room temperature under nitrogen. The resulting mixture was stirred at 70 ℃ for 12 hours under nitrogen atmosphere. After completion, the reaction mixture was cooled to room temperature and concentrated in vacuo. The residue obtained is taken up in saturated NH4The Cl solution was diluted, extracted with EtOAc (2 × 50mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 1% EtOAc-hexanes to give 4-chloro-2, 6-dimethoxyaniline (1.0g, 38%) as a light yellow liquid.1H NMR(300MHz,CDCl3):δ=6.52(s,2H),3.83(s,6H)。LCMS(m/z):187.9[M+H]+

2-amino-5-chloro-3-cyclopropyl-N, N-dimethylbenzamide

In a 50mL resealable reaction tube, 2-amino-3-bromobenzoic acid (2.0g, 9.25mmol) was dissolved in DMF (20mL) and cooled to 0 ℃. EDC-HCl (2.1g, 11.0mmol), HOBt (1.49g, 11.0mmol), DIPEA (2.8mL, 27.7mmol) and dimethylamine hydrochloride (1.13g, 13.8mmol) were added sequentially at 0 ℃. The reaction mixture was warmed to 70 ℃ and stirred for 12 hours. Upon completion, the reaction mixture was diluted with water, extracted with EtOAc (2 × 50mL), and the combined organic extracts were washed with water, brine, and dried (Na) 2SO4) And concentrated in vacuo to give 2-amino-3-bromo-N, N-dimethylbenzamide (2.0g, 89%) as a white solid.1H NMR(300MHz,CDCl3):δ=7.44(dd,J=7.8,1.5Hz,1H),7.06(dd,J=7.8,1.5Hz,1H),6.61(t,J=7.8Hz,1H),4.82(bs,2H),3.05(s,6H)。LCMS(m/z):243.10,245.10[M+H]+

In a 50mL resealable reaction tube, 2-amino-3-bromo-N, N-dimethylbenzamide (2g, 8.23mmol), cyclopropylboronic acid (850mg, 9.87mmol) and K were added3PO4A solution of (5.23g, 24.06mmol) was dissolved in toluene (30mL) and water (3 mL). The solution was degassed by purging with nitrogen for 5 minutes, then Pd (OAc) was added2(184mg, 0.823mmol) and tricyclohexylphosphine (230mg, 0.823mmol), and the solution was again purged with nitrogen for 5 minutes. The resulting mixture was stirred at 100 ℃ for 12 hours. After completion, the reaction mixture was taken up with saturated NH4The Cl solution was diluted, extracted with EtOAc (2X 50mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 20% EtOAc-hexanes to give 2-amino-3-cyclopropyl-N, N-dimethylbenzamide (1.0g, 60%) as a light brown solid.1H NMR(300MHz,CDCl3):δ=7.06(d,J=7.5Hz,1H),6.99(dd,J=7.8,1.5Hz,1H),6.66(t,J=7.8Hz,1H),4.70(bs,2H),3.05(s,6H),1.68-1.61(m,1H),0.94-0.88(m,2H),0.62-0.57(m,2H)。LCMS(m/z):205.3[M+H]+

A solution of 2-amino-3-cyclopropyl-N, N-dimethylbenzamide (0.5g, 2.44mmol) in acetonitrile (10mL) and AcOH (0.3mL) was cooledTo 0 ℃. N-chlorosuccinimide (0.5g, 3.67mmol) was added at 0 deg.C and the resulting reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was taken up with saturated Na 2S2O3The solution was diluted and extracted with ethyl acetate (2X 50 mL). The combined organic extracts were extracted with NaHCO3The solution was washed with brine and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 25% EtOAc-hexanes eluent to give 2-amino-5-chloro-3-cyclopropyl-N, N-dimethylbenzamide (0.2g, 34%) as a brown solid.1H NMR(300MHz,CDCl3):δ=7.02(d,J=1.5Hz,1H),6.96(d,J=2.7Hz,1H),4.66(bs,2H),3.05(s,6H),1.68-1.61(m,1H),0.94-0.88(m,2H),0.62-0.57(m,2H)。LCMS(m/z):239.0[M+H]+

4-chloro-2-methoxy-6- (trifluoromethyl) aniline

A solution of 2-bromo-4-chloro-6- (trifluoromethyl) aniline (0.5g, 1.82mmol), bis (pinacoldiborane) (0.92g, 3.64mmol) and KOAc (0.44g, 4.55mmol) in 1, 4-dioxane (10mL) was degassed by purging with nitrogen in a 50mL resealable reaction tube for 5 minutes. Adding Pd (dppf) Cl2(015g, 0.182mmol), and the solution was again purged with nitrogen for 5 minutes. The resulting mixture was stirred at 110 ℃ for 12 hours. After completion, the reaction mixture was diluted with water, extracted with EtOAc (2 × 50mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 2% EtOAc-hexanes eluent to give 4-chloro-2- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -6- (trifluoromethyl) aniline (0.5g, 85%). LCMS (M/z) 324.10[ M + H ]+。

4-chloro-2- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -6- (trifluoromethyl) aniline (500mg, 1.55mmol) was dissolved in THF (5mL)) and H at room temperature2O (2 mL). Adding NaBO in batches3·H2O (0.62g, 6.23mmol), and the reaction was stirred at room temperature for 4 hours. After completion, the reaction mixture was diluted with water, extracted with EtOAc (2 × 50mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 15% EtOAc-hexanes eluent to give 2-amino-5-chloro-3- (trifluoromethyl) phenol (0.5g, 100%) as a yellow liquid.1H NMR(300MHz,DMSO-d6):δ=10.50(s,1H),6.85(s,2H),5.12(bs,2H)。19F NMR(300MHz,DMSO-d6):δ=-61.46.LCMS(m/z):211.6[M+H]+

2-amino-5-chloro-3- (trifluoromethyl) phenol (250mg, 1.18mmol) was dissolved in anhydrous DMF (5mL) and washed with K2CO3(240mg, 1.77 mmol). The resulting mixture was stirred at room temperature for 30 minutes. Methyl iodide (185mg, 1.303mmol) was added dropwise and the reaction was stirred at room temperature for 2 hours. After completion, the reaction mixture was diluted with water, extracted with EtOAc (2 × 50mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 10% EtOAc-hexanes eluent to give 4-chloro-2-methoxy-6- (trifluoromethyl) aniline as a pale brown solid (0.2g, 75%). 1H NMR(400MHz,DMSO-d6):δ=7.10(d,J=2.0Hz,1H),6.98(d,J=2.0Hz,1H),5.34(bs,2H)3.85(s,3H)。19F NMR(400MHz,DMSO-d6):δ=-61.45。

7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-amine

A solution of 2, 3-dihydro-1H-inden-4-amine, 1(500mg, 3.75mmol) in EtOH (5mL) was cooled to 0 deg.C and treated dropwise with acetic anhydride (0.95g, 9.37mmol) under nitrogen. The resulting reaction mixture was warmed to room temperature and stirred for 3 hours. After completion of the reaction, (TLC, 30% EtOAc-hexane, Rf0.2), concentration in vacuoThe reaction mixture. The resulting residue was diluted with diethyl ether, filtered and dried in vacuo to give N- (2, 3-dihydro-1H-inden-4-yl) acetamide as a white solid (0.3g, 45%).1H NMR(400MHz,DMSO-d6):δ=9.29(s,1H),7.41(d,J=8.0Hz,1H),7.08(t,J=7.6Hz,1H),6.99(d,J=6.8Hz,1H),2.87(t,J=7.2Hz,2H),2.80(t,J=7.2Hz,2H),2.04(s,3H),1.99-1.95(m,2H)。LCMS(m/z):176.40[M+H]+

N- (2, 3-dihydro-1H-inden-4-yl) acetamide (200mg, 1.11mmol) was dissolved in AcOH (5mL) and cooled to 0 ℃. N-chlorosuccinimide (220mg, 1.69mmol) was added and the reaction mixture was warmed to room temperature and stirred overnight. After completion, the reaction mixture was diluted with ice-cold water and the solid formed was removed by filtration and saturated NaHCO3、Na2S2O3The solution was washed and dried in vacuo to give N- (7-chloro-2, 3-dihydro-1H-inden-4-yl) acetamide as a white solid (0.12g, 50%).1H NMR(300MHz,CDCl3):δ=7.73(d,J=8.4Hz,1H),7.14(d,J=8.4Hz,1H),6.86(s,1H),3.02-2.85(m,4H),2.19(s,3H),1.99(m,2H)。LCMS(m/z):209.80[M+H]+

N- (7-chloro-2, 3-dihydro-1H-inden-4-yl) acetamide (120mg, 0.57mmol) was dissolved in 3M HCl (5mL), warmed to 90 ℃ and held for 4 hours. After completion, the reaction mixture was cooled to room temperature and saturated NaHCO 3Basification of the solutionThen extracted with EtOAc (2X 20 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo to give 7-chloro-2, 3-dihydro-1H-inden-4-amine (70mg, 74%) as a white solid.1H NMR(300MHz,DMSO-d6):δ=6.85(d,J=8.4Hz,1H),6.40(d,J=8.4Hz,1H),4.97(s,2H),2.82(t,J=8.1Hz,2H),2.71(t,J=7.5Hz,2H),2.01-1.96(m,2H)。LCMS(m/z):168.20[M+H]+

A solution of 7-chloro-2, 3-dihydro-1H-inden-4-amine (0.8g, 4.79mmol) in acetonitrile (10mL) was cooled to 0 deg.C and treated with N-iodosuccinimide (1.61g, 7.18 mmol) at 0 deg.C) And (6) processing. The resulting reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was taken up with saturated Na2S2O3The solution was diluted and extracted with ethyl acetate (2X 50 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 4-5% EtOAc-hexanes eluent to give 7-chloro-5-iodo-2, 3-dihydro-1H-inden-4-amine (0.45g, 32%) as a light brown solid.1H NMR(300MHz,DMSO-d6):δ=7.36(s,1H),5.04(s,2H),2.82-2.72(m,4H),2.03-1.98(m,2H)。LCMS(m/z):293.7[M+H]+

In a 50mL resealable reaction tube, under nitrogen atmosphere at room temperature, with Cs2CO3(1.16g, 3.57mmol) A solution of 7-chloro-5-iodo-2, 3-dihydro-1H-inden-4-amine (0.35g, 1.19mmol) and cyclopropylboronic acid (0.41g, 4.77mmol) in 1, 4-dioxane (14mL) and water (4mL) was treated. Nitrogen was purged through the solution for 5 minutes under a nitrogen atmosphere with Pd (OAc) 2(26mg, 0.119mmol) and Catacxium-A (42mg, 0.119 mmol). The resulting mixture was degassed with nitrogen for 5 minutes. The resulting mixture was stirred at 100 ℃ for 24 hours. Upon completion, the reaction mixture was diluted with water and extracted with EtOAc (2 × 25 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 5% EtOAc-hexanes eluent to give 7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-amine (70mg, 28%) as a light brown solid.1H NMR(300MHz,CDCl3):δ=6.86(s,1H),2.94(t,J=7.5Hz,2H),2.80(t,J=7.5Hz,2H),2.15-2.10(m,2H),1.44-1.43(m,1H),0.91-0.88(m,2H),0.58-0.55(m,2H)。LCMS(m/z):208.3[M+H]+

Synthesis of R2 acid intermediate:

2,3,6, 7-tetrahydrobenzo [1,2-b:4,5-b' ] difuran-4-carboxylic acid

2,3,6, 7-tetrahydrobenzo [1,2-b:4,5-b 'was performed with hydroquinone using the procedure described in detail in Monte et al, J.Med.chem.1996,39, 2953-2961']Synthesis of Difuran-4-carboxylic acid to give 2,3,6, 7-tetrahydrobenzo [1,2-b:4,5-b 'as a bright yellow solid']Difuran-4-carbaldehyde;1H NMR(400MHz,CDCl3):δ=10.27(s,1H),6.87(s,1H),4.67(t,J=8.8Hz,2H),4.59(t,J=8.8Hz,2H),4.59(t,J=8.8Hz,2H),3.46(t,J=8.8Hz,2H),3.18(t,J=8.8Hz,2H)。

aldehyde (0.68g, 3.58mmol) was oxidized using silver (I) oxide (1.5 equiv.) in 5% aqueous sodium hydroxide at room temperature for 20 days. The crude reaction mixture was filtered through celite, extracted with diethyl ether (2 × 50mL) to remove unreacted aldehyde, and the aqueous phase was then acidified dropwise to pH 1 at 0 ℃ using 3.0M aqueous HCl. The product was extracted with dichloromethane (2X 50mL) and the combined organics were washed with brine (50mL) and dried (MgSO) 4) And concentrated in vacuo to give 2,3,6, 7-tetrahydrobenzo [1,2-b:4,5-b 'as a white solid']Difuran-4-carboxylic acid (0.44 g; 60%).

Alternatively, aldehyde (0.5g, 2.77mmol) in acetone (5.0mL) was treated with sulfamic acid (0.4g, 4.17mmol) in two portions at 0 deg.C. After 2 minutes, a solution of sodium chlorite (0.32g, 3.6mmol) in water (1.0mL) was added dropwise and stirring continued at 0 ℃ for 4 hours. The reaction mixture was diluted with water (20mL) and extracted with 10% IPA/chloroform (2X 20 mL). The combined organics were washed with water (25mL), brine (25mL) and dried (Na)2SO4) And concentrated in vacuo. Triturating the crude solid with diethyl ether to give 2,3,6, 7-tetrahydrobenzo [1,2-b:4,5-b']Difuran-4-carboxylic acid (0.4 g; 70%).1H NMR(400MHz,DMSO-d6):δ=6.86(s,1H),4.52(t,J=8.8Hz,2H),4.47(t,J=8.8Hz,2H),3.30(t,J=8.8Hz,2H),3.10(t,J=8.8Hz,2H)。13C(100MHz,DMSO-d6):δ=166.4,154.2,153.9,128.9,127.2,111.4,110.43,71.9,71.6,31.5,29.5。

Benzo [ d ] [1,3] dioxole-4-carboxylic acid

Synthesized using a procedure modified from Plee et al J.Med.chem.2004,47,871- & 887 as follows:

2, 3-Dihydroxybenzoic acid (5.0g, 32.4mmol) in dry methanol (50mL) was treated with concentrated sulfuric acid (10 drops) and heated at reflux overnight. The reaction mixture was concentrated in vacuo, diluted with EtOAc (100mL), and saturated NaHCO3Aqueous solution (2X 50mL), brine (50mL) and then dried (MgSO)4) And concentrated in vacuo to give methyl 2, 3-dihydroxybenzoate (2.92 g; 54%). 1H NMR(400MHz,CDCl3):δ=10.9(s,1H),7.32(dd,J=8.0,1.2Hz,1H),7.09(m,1H),6.78(t,J=8.0Hz,1H),5.65(s,3H)。13C NMR(100Hz,CDCl3)170.7,148.8,145.0,120.5,119.8,119.2,112.4,52.4。

Methyl 2, 3-dihydroxybenzoate (1.0g, 5.95mmol) in DMF (16mL) was treated with KF (1.79g, 30.9mmol) and stirred at ambient temperature for 30 min. Diiodomethane (1.79g, 6.7mmol) was added and the reaction was heated at 100 ℃ for 5 hours. The reaction mixture was cooled to room temperature, poured onto water (100mL), and extracted with diethyl ether (2X 50 mL). The combined organics were washed with water (50mL), brine (50mL), and dried (MgSO)4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 10% EtOAc-petroleum ether eluent to afford benzo [ d ] as a white crystalline solid][1,3]Dioxole-4-carboxylic acid methyl ester (0.56 g; 52%);1H NMR(400MHz,CDCl3):δ=7.41(dd,J=8.0,1.2Hz,1H),6.97(dd,J=8.0,1.2Hz,1H),6.86(t,J=8.0Hz,1H),6.1(s,2H),3.93(s,3H)。

treatment of benzo [ d ] with 2.0M aqueous KOH (2.2mL)][1,3]A solution of methyl dioxole-4-carboxylate (0.4g, 2.22mmol) in methanol (8.0mL) was stirred at room temperature for 3 hours. The mixture was concentrated to a volume of-3 mL, diluted with water (5mL) and acidified to pH-3 with 2.0M HCl. The resulting precipitate was removed by filtration, washed with water then diethyl ether and dried in vacuo to give benzo [ d ] a beige solid][1,3]Dioxole-4-carboxylic acid (0.38g, 97%).1H NMR(400MHz,DMSO-d6):δ=7.28(dd,J=8.0,1.2Hz,1H),6.97(dd,J=8.0,1.2Hz,1H),6.89(t,J=8.0Hz,1H),6.12(s,2H);13C NMR(100Hz,DMSO-d6)165.5,148.9,148.5,122.9,121.6,113.8,112.5,102.1。

Compounds synthesized by substituents

Aliphatic compounds

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) cyclohexanesulfonamides

The title compound was obtained as a white solid (12mg, 41%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and cyclohexane sulfonamide in general method C3.1H NMR(400MHz,CD3OD):δ=6.97(s,1H),3.50-3.43(m,1H),2.87(t,4H,J=8.0Hz),2.78(t,4H,J=8.0Hz),2.22-2.18(m,2H),2.10-2.02(m,4H),1.94-1.71(m,2H),1.63-1.59(m,1H),1.64-1.53(m,2H);1.41-1.21(m,3H);13C NMR(100MHz,CD3OD): δ 143.7,137.8,126.4,118.4,110.2,59.9,35.5,30.0,28.5,25.8,25.1, 24.8; LCMS purity:>95%;LCMS(m/z):363[M+H]+;C19H26N2O3S[M+H]+HRMS calculated of (a): 363.1737, found: 363.1729.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) cyclopentane sulphonamide

Use of 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method A2) and cyclopentanesulfonamide in general procedure C3 to give the title compound as a white solid (26mg, 42%)1H NMR(400MHz,CD3OD):δ=6.97(s,1H),4.08-4.02(m,1H),2.83(t,J=8.0Hz,4H),2.80(t,J=8.0Hz,4H),2.13-2.01(m,8H),1.84-1.77(m,2H),1.71-1.65(m,2H);13C NMR(100MHz,CD3OD): δ 145.1,139.2,127.8,119.8,111.7,62.2,33.9,31.4,29.9,28.6, 26.9; LCMS purity:>95%;LCMS(m/z):349[M+H]+;C18H24N2O3HRMS calculated value of S: 349.1580, found: 349.1588.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) tetrahydro-2H-pyran-4-sulfonamide.

Using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and tetrahydro-2H-pyran-4-sulfonamide in general method C2, the title compound is obtained as a white solid (12mg, 57%). 1H NMR(400MHz,CD3OD):δ=7.00(s,1H),4.09(dd,J1=4Hz,J2=12Hz,2H),3.82-3.76(m,1H),3.49-3.43,(m,2H),2.89(t,J=8Hz,4H),2.81(t,J=8Hz,4H),2.12-2.05(m,6H),1.98-1.87(m,2H);13C NMR(100MHz,CD3OD): δ 154.0,143.1,137.7,126.5,110.4,66.0,57.0,32.5,28.5,25.9, 25.1; LCMS purity:>95%;LCMS(m/z):365[M+H]+;C18H24N2O4HRMS calculated value of S: 365.1530, found: 365.1541.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) tetrahydrofuran-3-sulfonamide

The title compound was obtained as a white solid (12mg, 60%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and tetrahydrofuran-3-sulfonamide in general method C2.1H NMR(400MHz,DMSO-d6):δ=8.04(s,1H),6.93(s,1H),4.33-4.27(m,1H),4.04-4.00(m,1H),3.91-3.89(m,1H),3.85-3.79(m,1H),3.72-3.66(m,1H),2.80(t,J=16.0Hz,4H),2.70(t,J=16.0Hz,4H),2.24-2.17(m,2H),1.99-1.95(m,4H)。13C NMR(100MHz,DMSO-d6) δ 142.4,139.6,136.6,124.7,108.2,68.7,61.7,32.5,30.3,28.8,28.1, 24.9; LCMS purity:>95%;LCMS(m/z):351[M+H]+;C17H22N2O4HRMS calculated value of S: 351.1373, found: 351.1389.

furan compounds

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) furan-2-sulfonamide

The title compound was obtained as a white solid (75mg, 16%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and furan-2-sulfonamide in general method C4.1H NMR(400MHz,DMSO-d6):δ=11.08(br.s,1H),8.08(s,1H),8.02(s,1H),7.22(q,J=2.0Hz,1H),6.94(s,1H),6.71(q,J=2.0Hz,1H),2.78(t,J=7.2Hz,4H),2.59(t,J=7.2Hz,4H),1.94(quin,J=7.2Hz,4H)。13C NMR(100MHz,DMSO-d6) Delta-148.9,147.9,147.3,143.1,137.3,128.7,118.0,117.5,111.7, 54.9,32.5,30.1,25.1.LCMS, purity 96.26%; m/z 345.1 (M-H)+)。C17H18N2O4S[M-H]-HRMS (FAB)-) Calculated values: 345.0987, found: 345.0866.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -5-methylfuran-2-sulfonamide

The title compound was obtained as a white solid (28mg, 53%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 5-methylfuran-2-sulfonamide in general method C2.1H NMR(400MHz,DMSO-d6):δ=7.96(s,1H),7.00-6.99(d,J=4.0Hz,1H),6.91(s,1H),6.29-6.28(d,J=4.0Hz,1H),1H),2.78(t,J=8.0Hz,4H),2.61(t,J=8.0Hz,4H),2.34(s,3H),2(t,J=8.0Hz,4H),1.98-1.90(m,4H);13C NMR(100MHz,DMSO-d6) δ 143.3,137.6,129.9,125.2,118.0,114.6,108.7,108.2,107.8,32.9,30.6,25.4, 13.8; LCMS purity:>95%;LCMS(m/z):361[M+H]+;C18H20N2O4S[M+H]+HRMS calculated of (a): 361.1216, found: 361.1217.

5-ethyl-N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) furan-2-sulfonamide

The title compound was obtained as a white solid (51mg, 47%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 5-ethylfuran-2-sulfonamide in general method C2.1H NMR(400MHz,DMSO-d6):δ=7.97(bs,1H),7.02(s,1H),6.91(d,J=4.0Hz,1H),6.31(d,J=4.0Hz,1H),2.78(t,J=8.0Hz,4H),2.68(q,J=8.0Hz,2H),2.59(t,J=6.0Hz,4H),1.97-1.90(m,4H),1.19(t,J=8.0Hz,3H)。13C NMR(150MHz,DMSO-d6) δ 143.5,143.3,142.9,137.6,129.8,118.0,108.7,106.8,106.3,32.9,30.5,25.4,21.3, 12.1; LCMS purity:>95%;LCMS(m/z):375[M+H]+;C19H22N2O4S[M+H]+HRMS calculated of (a): 375.13730, found: 375.13910.

5- ((dimethylamino) methyl) -N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) furan-2-sulfonamide

Use of 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure A1) and 5- ((dimethylamino) methyl) furan-2-sulfonamide in general procedure C2 to afford a compound as Title compound as white solid (25mg, 6%).1H NMR(400MHz,CD3OD)δ=7.17(d,J=3.5Hz,1H),6.96(s,1H),6.86(d,J=3.5Hz,1H),4.43(s,2H),2.86(s,3H),2.86(t,J=7.4Hz,4H),2.73(t,J=7.4Hz,4H),2.04(p,J=7.4Hz,4H)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

The title compound was obtained as a white solid (2.5g, 63%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide in general method C5.1H NMR(600MHz,DMSO-d6):δ=7.61(br.s.,1H),7.37(d,J=0.9Hz,1H),6.77(s,1H),6.58(d,J=0.9Hz,1H),2.74(t,J=7.3Hz,4H),2.65(t,J=7.3Hz,4H),1.89(tt,J=7.3,7.3Hz,4H),1.34(s,6H)。13C NMR(101Hz,DMSO-d6):δ=157.4,155.7,142.2,137.3,136.7,135.7,132.4,115.7,109.3,66.6,32.6,31.1,30.6,25.1。

N- ((8-bromo-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

The title compound was obtained as a white solid (40mg, 7%) using 4-bromo-8-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide in general method C1.1H NMR(400MHz,CD3OD):δ=7.68(s,1H),7.23(s,1H),2.91(t,J=7.6Hz,4H),2.85(t,J=7.6Hz,4H),2.11(m,4H),1.51(s,6H)。LCMS(m/z):482.9[M-H]-;97.64%(210nm),99%(254nm)。HPLC:96.70%(210nm),97.22%(254nm)。

N- ((8-chloro-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

The title compound was obtained as a white solid (50mg, 16%) using 4-chloro-8-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide in general method C1.1H NMR(400MHz,CD3OD)δ=7.55(s,1H),7.02(s,1H),2.91(t,J=7.2Hz,4H),2.85(t,J=7.2Hz,4H),2.09(m,4H),1.5(s,6H)。LCMS(m/z):460.9(M+Na)-;95.16%(210nm),95.07%(254nm)。HPLC:97.91%(210nm),98.04%(254nm)。

4- (2-hydroxypropan-2-yl) -N- ((8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) furan-2-sulfonamide

Using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide in general method C1 to give the title compound as a white solid (15mg, 3%).1H NMR(400MHz,CD3OD):δ=7.58(s,1H),7.07(s,1H),6.46(s,1H),2.82-2.73(m,J=7.5Hz,8H),2.12(s,3H),2.05-2.02(m,4H),1.508(s,6H)。LCMS(m/z):417.10(M-1)-;99.59%(210nm),99.33%(254nm)。HPLC:97.92%(210nm),97.53%(254nm)。

5- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) furan-3-carboxylic acid

Treatment of 5- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) in THF (8mL) with a solution of LiOH (0.1g, 2.4mmol) in water (2mL) at 0 deg.CYl) sulfonamide) furan-3-carboxylic acid ethyl ester (0.1g, 0.24 mmol). The cooling bath was removed and the reaction mixture was stirred for 3 hours. The solution was acidified with 10% citric acid and immediately extracted with ethyl acetate (2 × 25 mL). The organics were washed with water (20mL), brine (20mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by reverse phase HPLC to give the title compound as a white solid (5.0mg, 5%).1H NMR(400MHz,CD3OD):δ=8.14(s,1H),7.28(s,1H),6.93(s,1H),2.85(t,J=7.6Hz,4H),2.74(t,J=7.6Hz,4H),2.04(quin,J=7.6Hz,4H)。

5- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) furan-3-carboxylic acid ethyl ester

In general procedure C3, 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general procedure A2) and ethyl 5-sulfamoylfuran-3-carboxylate were used. The reaction mixture was quenched with water (50mL), extracted with ethyl acetate (2 × 25mL), and the organics were washed with brine (25mL), dried (Na) 2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography with 50% EtOAc-hexanes eluent to give the title compound as a white solid (0.45g, 63%).1H NMR(300MHz,DMSO-d6)δ=8.31(s,1H),7.59(s,1H),6.77(s,1H),4.22(q,J=7.2Hz,2H),2.75(t,J=7.3Hz,4H),2.65(t,J=7.3Hz,4H)1.90(pent,J=7.6Hz,4H),1.26(t,J=7.2Hz,3H)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (prop-1-en-2-yl) furan-2-sulfonamide

The titled compound is obtained as a white solid by using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general method A2) and 4- (prop-1-en-2-yl) furan-2-sulfonamide in general method C6Compound (85mg, 51%).1H NMR(400MHz,CDCl3)δ7.54(s,1H),7.28(s,1H),7.00(s,1H),5.26(s,1H),5.05(s,1H),2.86(t,J=7.4Hz,4H),2.69(t,J=7.5Hz,4H),2.09–1.98(m,7H)。13C NMR(101MHz,CDCl3)δ144.4,142.8,137.8,132.8,129.2,127.2,119.4,115.4,113.6,32.9,30.5,25.5,20.9.C20H23N2O4S[M+H]Hrms (esi) calculated value of (a): 387.1373, found: 387.1379.

4- (2-hydroxypropan-2-yl) -N- ((3,5,6, 7-tetrahydro-2H-indeno [5,6-b ] furan-8-yl) carbamoyl) furan-2-sulfonamide

Use of 8-Isocyanato-3, 5,6, 7-tetrahydro-2H-indeno [5,6-b ] in general procedure C3]Furan (prepared by general procedure A1) and ethyl 5-sulfamoylfuran-3-carboxylate to give 5- (N- ((3,5,6, 7-tetrahydro-2H-indeno [5, 6-b) as a light brown solid]Furan-8-yl) carbamoyl) sulfamoyl) furan-3-carboxylic acid ethyl ester (0.25g, 50%).1H NMR(300MHz,DMSO-d6)δ8.32(s,1H),7.17(s,1H),6.77(d,J=5.2Hz,1H),4.43(t,J=8.6Hz,2H),4.23(q,J=7.1Hz,2H),3.07(t,J=8.6Hz,2H),2.71(t,J=7.3Hz,2H),2.63(t,J=7.3Hz,2H),1.89(p,J=7.4Hz,2H),1.26(t,J=7.1Hz,3H)。

At 0 ℃ with vigorous stirring over 5 minutes with a solution of methyl magnesium chloride (3.0M in Et)2O, 6 equiv.) 5- (N- ((3,5,6, 7-tetrahydro-2H-indeno [5, 6-b) in anhydrous THF (10mL) was treated dropwise ]Furan-8-yl) carbamoyl) sulfamoyl) furan-3-carboxylic acid ethyl ester (0.25g, 0.6 mmol). The solution was then stirred at 0 ℃ for 30 minutes and then at ambient temperature for 4 hours, followed by dropwise quenching with saturated ammonium chloride solution. The aqueous solution was extracted with EtOAc (2X 25mL) and the combined organics were washed with brine (20mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was triturated with diethyl ether then purified by reverse phase preparative HPLC to give the title compound as a white solid (32mg, 13%).1H NMR(400MHz,DMSO-d6)δ7.58(s,1H),7.08(s,1H),6.84(s,1H),5.02(s,1H),4.47(t,J=8.6Hz,2H),3.10(t,J=8.6Hz,2H),2.73(t,J=7.3Hz,2H),2.59(t,J=7.5Hz,2H),1.90(d,J=7.4Hz,2H),1.36(s,6H)。

N- ((4-bromo-3, 5,6, 7-tetrahydro-2H-indeno [5,6-b ] furan-8-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

Use of 4-bromo-8-isocyanato-3, 5,6, 7-tetrahydro-2H-indeno [5,6-b ] in general procedure C1]Furan (prepared using general method a 1) and 4- (prop-1-en-2-yl) furan-2-sulfonamide to give the title compound as a white solid (20mg, 9%).1H NMR(400MHz,CD3OD)δ=7.48(d,J=1.2Hz,1H),6.93(d,J=1.2Hz,1H),4.60(t,J=8.7Hz,2H),3.16(t,J=8.6Hz,2H),2.85(m,4H),2.03(p,J=7.5Hz,2H),1.50(s,6H)。

4- (2-hydroxypropan-2-yl) -N- ((3,5,6, 7-tetrahydro-2H-indeno [5,6-b ] furan-4-yl) carbamoyl) furan-2-sulfonamide

Use of 4-Isocyanato-3, 5,6, 7-tetrahydro-2H-indeno [5,6-b ] in general procedure C1]Furan (prepared using general method a 1) and 4- (prop-1-en-2-yl) furan-2-sulfonamide to give the title compound as a white solid (20mg, 9%). 1H NMR(400MHz,CD3OD)δ=7.58(s,1H),7.07(s,1H),6.46(s,1H),4.49(d,J=8.9Hz,2H),3.05(t,J=8.7Hz,2H),2.82(t,J=7.4Hz,2H),2.70(t,J=7.4Hz,2H),2.04(p,J=7.4Hz,2H),1.51(d,J=1.9Hz,6H)。

4- (2-hydroxypropan-2-yl) -N- ((2,3,6, 7-tetrahydrobenzo [1,2-b:4,5-b' ] difuran-4-yl) carbamoyl) furan-2-sulfonamide

Use of 4-isocyanato-2, 3,6, 7-tetrahydrobenzo [1,2-b:4,5-b 'in general method C6']Difuran (prepared by general method a 1) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide to give the title compound as a white solid (285mg, 96%).1H NMR(600MHz,DMSO-d6)δ7.76(s,1H),7.66(s,1H),7.01(s,1H),6.45(s,1H),5.04(s,1H),4.46(t,J=8.6Hz,2H),4.39(t,J=8.6Hz,2H),3.08(t,J=8.6Hz,2H),2.94(t,J=8.6Hz,2H),1.37(s,6H)。

N- (benzo [1,2-b:4,5-b' ] difuran-4-ylcarbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

Use of 4-Isocyanatobenzo [1,2-b:4,5-b 'in general procedure C3']Difuran (prepared by general method A1) and ethyl 5-sulfamoylfuran-3-carboxylate to give 5- (N- (benzo [1,2-b:4,5-b ') as a white solid']Difuran-4-ylcarbamoyl) sulfamoyl) furan-3-carboxylic acid ethyl ester (0.05g, 53%).1H NMR(300MHz,CD3OD)δ=8.25(s,1H),7.72(d,J 2.1Hz,1H),7.63(d,J 2.1Hz,1H),7.46(s,1H),7.27(s,1H),6.93(s,1H),6.89(d,J 2.1Hz,1H),6.86(d,J 2.1Hz,1H),4.30(q,J 6.9Hz,2H),1.4(t,J 6.9Hz,3H)。

With vigorous stirring over 10 min with a solution of methyl magnesium chloride (3.0M in Et2O, 10 equivalents) 5- (N- (benzo [1,2-b:4,5-b ') in dry THF (10mL) was treated dropwise']Difuran-4-ylcarbamoyl) sulfamoyl) furan-3-carboxylic acid ethyl ester (0.25g, 0.6 mmol). The solution was then stirred at 0-10 ℃ for 3 hours and then quenched dropwise with saturated ammonium chloride solution. The aqueous solution was extracted with EtOAc (2X 20mL), and the combined organics were washed with brine (20mL) and dried (Na) 2SO4) And concentrated in vacuo. The crude product was triturated with diethyl ether then purified by reverse phase preparative HPLC to give the title compound as a white solid (15mg, 6%).1H NMR(400MHz,CD3OD)δ=7.76(d,J 2.0Hz,1H),7.65(d,J 2.4Hz,1H),7.55(s,1H),7.47(s,1H),7.05(s,1H),6.93(d,J 2.0Hz,1H),6.89(d,J 2.4Hz,1H),1.5(s,6H)。

N- (Anthracene-9-ylcarbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

9-Isocyanatoanthracene (prepared using general procedure B2) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide were used in general procedure C6 to give the title compound as a white solid (24mg, 23%).1H NMR(400MHz,CD3OD)δ=8.49(s,1H),8.07–7.98(m,4H),7.75(s,1H),7.55–7.44(m,4H),7.27–7.22(m,1H),1.49(s,6H)。13C NMR(101MHz,CD3OD)δ=153.8,149.4,141.4,136.6,131.7,128.8,128.2,127.4,126.4,125.9,124.9,122.8,115.2,111.1,67.2,29.6.8,115.2,111.1,67.2,29.6。

4- (2-hydroxypropan-2-yl) -N- (quinolin-8-ylcarbamoyl) furan-2-sulfonamide

Phenyl chloroformate (1.5 equiv.) was slowly added to a solution of quinolin-8-amine (1g, 6.9mmol) in THF (10mL) and triethylamine (2 equiv.) to 0 deg.C. The solution was stirred at room temperature for 2 hours or until completion. The solution was diluted with saturated aqueous NaHCO3, extracted with ethyl acetate (2 × 50mL), washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using 10% EtOAc-hexanes to give quinolin-8-yl phenyl carbamate as a white solid (1.5g, 83%) which was used directly in the next reaction step.

4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (0.2g, 0.98mmol) in THF (5mL) was treated portionwise with sodium hydride (3 equivalents) at 0 deg.C, and the suspension was stirred at ambient temperature for 45 minutes (until effervescence ceased). Crude quinolin-8-yl phenyl carbamate is dissolved in THF (5mL) and then slowly added to the reaction and the solution is stirred at ambient temperature until completion, usually It was 4 hours. The reaction mixture is saturated with NH4Aqueous Cl solution quenching, extraction with ethyl acetate (× 2), washing with water, brine and drying (Na)2SO4) And concentrated in vacuo. The crude product was purified by reverse phase HPLC to give the title compound 4- (2-hydroxypropan-2-yl) -N- (quinolin-8-ylcarbamoyl) furan-2-sulfonamide (40mg, 11%) as a white solid.1H NMR(400MHz,DMSO-d6)δ9.63(s,1H),8.89(d,J=4.3Hz,1H),8.37(m,2H),7.80–6.76(m,5H),5.09(s,1H),1.38(s,6H)。

4- (2-hydroxypropan-2-yl) -N- ((6-methoxy-8-yl) carbamoyl) furan-2-sulfonamide

4- (2-hydroxypropan-2-yl) -N- ((6-methoxyquinolin-8-yl) carbamoyl) furan-2-sulfonamide was synthesized using a modification of the procedure used to prepare 4- (2-hydroxypropan-2-yl) -N- ((quinolin-8-ylcarbamoyl) furan-2-sulfonamide but using 6-methoxyquinolin-8-amine instead of quinolin-8-amine the title compound was obtained as an off-white solid (75mg, 38%).1H NMR(400MHz,DMSO-d6)δ8.79(s,1H),8.63(m,1H),8.17(m,1H),8.09(d,J=2.7Hz,1H),7.49(dd,J=8.3,4.2Hz,1H),7.40(s,1H),6.79(d,J=2.8Hz,1H),6.69(s,1H),4.96(s,1H),3.84(s,3H),1.36(s,6H)。

N- ((2, 3-dihydrobenzo [ b ] [1,4] dioxin-5-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

In general procedure C6 use was made of 5-isocyanato-2, 3-dihydrobenzo [ b ]][1,4]Dioxine (prepared using general method A1) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide to give the title compound as a white solid (49mg, 39%). 1H NMR (600MHz, acetonitrile-d)3)δ=7.56(dd,J=8.4,1.5Hz,1H),7.45(d,J=1.0Hz,1H),6.98(d,J=1.0Hz,1H),6.7(t,J=8.4Hz,1H),6.48(dd,J=8.4,1.5Hz,1H),4.22(m,4H),1.43(s,6H)。

N- ((2, 3-dihydrobenzofuran-7-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

7-Isocyanato-2, 3-dihydrobenzofuran (prepared using general procedure A1) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide were used in general procedure C6 to give the title compound as a white solid (32mg, 39%).1H NMR (600MHz, acetonitrile-d)3)δ7.64(d,J=7.7Hz,1H),7.48(d,J=1.1Hz,1H),7.00(d,J=1.1Hz,1H),6.89(m,1H),6.74(t,J=7.7Hz,1H),4.56(t,J=8.7Hz,1H),3.2(t,J=8.7Hz,1H),1.43(s,6H)。

N- ((2, 4-bis (trifluoromethyl) phenyl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

1-Isocyanato-2, 4-bis (trifluoromethyl) benzene (prepared using general procedure A1) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide were used in general procedure C4 to give the title compound as an off-white solid (0.12g, 33%).1H NMR(400MHz,DMSO-d6):δ=8.59(d,J=8.8Hz,1H),7.87(d,J=9.2Hz,1H),7.81(s,1H),7.67(s,1H),7.43(s,1H),6.68(s,1H),4.94(s,1H),1.36(s,6H)。13C NMR(100MHz,DMSO-d6) δ 156.0,154.4,142.5,138.1,135.8,129.9,125.2,124.9,123.0,122.5,121.3,120.7,120.4,115.5,115.2,110.2,66.5,31.0, LCMS, purity 90.47%, tr 3.84 min, M/z 459.25 (M-H-459.25)+)。C16H14F6N2O5S[M-H]-HRMS (FAB)-) Calculated values: 459.0528, found: 459.0512.

n- ((2, 5-bis (trifluoromethyl) phenyl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

2-Isocyanato-1, 4-bis (trifluoromethyl) benzene (prepared using general procedure A1) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide were used in general procedure C4 to give the title compound as an off-white solid (55mg, 12%). 1H NMR(400MHz,CD3OD):δ=8.61(s,1H),7.75(d,J=7.6Hz,1H),7.48(s,1H),7.37(d,J=8.4Hz,1H),6.95(s,1H),1.41(s,6H)。13C NMR(100MHz,DMSO-d6) Delta 156.4,154.5,139.7,138.1,132.9,127.2,124.9,122.3,118.9,117.6,117.0,110.0,66.5,31.0.LCMS, purity 95.02%, tr 2.09min, M/z 558.94 (M-H)+)。C16H14F6N2O5S[M-H]-HRMS (FAB)-) Calculated values: 459.0528, found: 459.0224.

4- (2-hydroxypropan-2-yl) -N- ((2-methoxyphenyl) carbamoyl) furan-2-sulfonamide

1-Isocyanato-2-methoxybenzene (prepared using general procedure A2) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide were used in general procedure C2 to give the title compound as an off-white solid (30mg, 38%).

N- ((2, 5-dimethoxyphenyl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

2-Isocyanato-1, 4-dimethoxybenzene and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide were used in general procedure C2 to give the title compound as an off-white solid (52mg, 55%).

N- ((4-chloro-2, 6-dimethylphenyl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

4-chloro-2, 6-dimethylaniline, 1(300mg, 1.92mmol) was dissolved in THF (50mL) and cooled to 0 ℃. NaH (100mg, 2.49mmol) was added portionwise to the above solution under nitrogen and the mixture was stirred for 15 min. Phenyl chloroformate (0.33mL, 0.72mmol) was added dropwise to the above solution at 0 ℃. The reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 30% EtOAc-hexanes eluent to give (4-chloro-2, 6-dimethylphenyl) aminomethylbenzoate (0.2g, 85%) as a white solid. 1H NMR(300MHz,CDCl3):δ=7.41-7.36(m,2H),7.21-7.19(m,2H),7.11-7.10(m,3H),6.30(bs,1H),2.33(s,6H)。

4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (133mg, 0.64mmol) was dissolved in anhydrous THF (5mL) and treated cautiously with NaH (65mg, 1.63mmol) at 0 deg.C under a nitrogen atmosphere. The resulting mixture was stirred at room temperature for 45 minutes, then treated with a solution of phenyl (4-chloro-2, 6-dimethylphenyl) carbamate (200mg, 0.73mmol) in THF (3mL) at 0 ℃ under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred for 2 hours. After completion, the reaction mixture was washed with saturated NH4The Cl solution dilution was extracted with EtOAc (2X 50mL) and the combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluent to give N- ((4-chloro-2, 6-dimethylphenyl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (20mg, 31%) as a white solid.1H NMR(400MHz,CDCl3):δ=7.83(s,1H),7.56(s,1H),7.19(s,1H),7.10-7.05(m,2H),2.16(s,6H),1.55(s,6H)。LCMS(m/z):385.05[M-H]-,94.12%(210nm)。HPLC:92.60%(210nm)。C16H18Cl1N2O5S1[M-H]-HRMS calculated of (a): 385.0630, found: 365.0621.

n- ((2, 4-dimethyl-6- (trifluoromethyl) phenyl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

4-chloro-2-methyl-6- (trifluoromethyl) aniline (230mg, 1.1mmol) was dissolved in anhydrous THF (20mL) and Et at room temperature 3N (0.17mL, 1.32 mmol). The solution was treated with triphosgene (130mg, 0.44mmol) and the resulting mixture was stirred at 60 ℃ for 4 h, then concentrated in vacuo. The resulting residue was stirred with n-pentane (20mL) for 10 minutes, filtered through a pad of celite and concentrated in vacuo to give 5-chloro-2-isocyanato-1-methyl-3- (trifluoromethyl) benzene (0.2g) as a white solid. The product was used in the next step without further purification.

4- (2-hydroxypropan-2-yl) furan-2-sulfonamide, 3(150mg, 0.731mmol) was dissolved in anhydrous THF (50mL) and carefully treated with NaH (44mg, 1.096mmol) at 0 ℃ under a nitrogen atmosphere. The resulting reaction mixture was stirred at room temperature for 30 minutes and treated with a solution of 5-chloro-2-isocyanato-1-methyl-3- (trifluoromethyl) benzene (0.2g) in THF (30mL) under a nitrogen atmosphere. The resulting reaction mixture was stirred at room temperature for 2 hours. After completion, the reaction mixture was taken up with saturated NH4The Cl solution was diluted, extracted with EtOAc (2 × 50mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluent. The product was then triturated with diethyl ether and N-pentane to give N- ((2, 4-dimethyl-6- (trifluoromethyl) phenyl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (15mg, 5%) as a white solid. 1H NMR(400MHz,CD3OD):δ=7.52-7.48(m,3H),6.99(s,1H),2.19(s,3H),1.47(s,6H)。19F NMR(400MHz,CD3OD):δ=-63.09.LCMS(m/z):439.05[M-H]-;94.86%(210nm),96.92%(254nm)。HPLC:98.90%(210nm)。C16H15Cl1F3N2O5S1[M-H]-HRMS calculated of (a): 439.0348, found: 439.0339.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

The title compound was obtained as a white solid in general method C2 using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general method a 2) and 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (161mg, 34%).1H NMR(600MHz,DMSO-d6)δ=7.82(s,1H),7.61(s,1H),7.09(s,2H),6.93(s,1H),5.04(s,1H),3.05–2.99(m,2H),1.35(s,6H),1.05(d,J=6.9Hz,12H)。C20H26Cl1N2O5S1[M-H]-HRMS calculated of (a): 441.1256, found: 441.1264.

n- ((4-chloro-2, 6-dicyclopropylphenyl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

4-chloro-2, 6-dicyclopropylaniline, 1(250mg, 1.20mmol) was dissolved in THF (50mL) and cooled to 0 ℃. NaH (72mg, 1.80mmol) was added portionwise and the resulting mixture was stirred under nitrogen for 20 min. Phenyl chloroformate (370mg, 2.40mmol) was added dropwise at 0 deg.C, and the reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 8% EtOAc-hexanes eluent to give phenyl (4-chloro-2, 6-dicyclopropylphenyl) carbamate as a white solid (0.2g, 51%). 1H NMR(300MHz,CDCl3):δ=7.38-7.35(m,2H),7.21-7.19(m,3H),6.84-6.83(m,2H),2.06-2.04(m,2H),1.04-1.02(m,4H),0.69-0.68(m,4H)。

4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (75mg, 0.365mmol) was dissolved in anhydrous THF (50mL) and treated carefully with NaH (36mg, 0.914mmol) at 0 deg.C under a nitrogen atmosphere. The resulting mixture was stirred at room temperature for 30 minutes, then treated with a solution of phenyl (4-chloro-2, 6-dicyclopropylphenyl) carbamate (135mg, 0.402mmol) in THF (3mL) at 0 ℃ under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred for 4 hours. After completion, the reaction mixture was taken up with saturated NH4The Cl solution was diluted, extracted with EtOAc (2 × 50mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 50-100% EtOAc-hexanes to give N- ((4-chloro-2, 6-dicyclopropylphenyl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (10mg, 6%) as a white solid.1H NMR(400MHz,CD3OD):δ=7.59(s,1H),7.13(s,1H),6.76(s,2H),1.88-1.86(m,2H),1.47(s,6H),0.89-0.84(m,4H),0.55-0.54(m,4H)。LCMS(m/z):436.95[M-H]-;96.29%(210nm)。HPLC:98.29%(210nm)。C20H22Cl1N2O5S1[M-H]-HRMS calculated of (a): 437.0943, found: 437.0945.C20H22Cl1N2O5S1[M-H]-HRMS calculated of (a): 437.0943, found: 437.0945.

4- (2-hydroxypropan-2-yl) -N- ((5-methoxy-2, 3-dihydro-1H-inden-4-yl) carbamoyl) furan-2-sulfonamide

5-methoxy-2, 3-dihydro-1H-inden-4-amine (150mg, 0.59mmol) was dissolved in THF (15mL) and cooled to 0 ℃. NaH (35mg, 0.89mmol) was added to the above solution and stirred for 20 min. Phenyl chloroformate (150mg, 0.932mmol) was added dropwise at 0 ℃ and the solution was warmed To room temperature overnight. After completion, the reaction mixture was taken up with saturated NaHCO3Diluted and extracted with EtOAc (30 mL). The organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo to give phenyl (5-methoxy-2, 3-dihydro-1H-inden-4-yl) carbamate as a white solid (100mg, 59%).1H NMR(300MHz,CDCl3):δ=7.40-7.35(m,2H),7.22-7.18(m,3H),7.06(d,J=8.1Hz,1H),6.73(d,J=8.4Hz,1H),3.86(s,3H),2.98-2.84(m,4H),2.08(t,J=7.5Hz,2H)。LCMS(m/z):284.3[M+H]+

4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (87mg, 0.424mmol) was dissolved in anhydrous THF (5mL) and treated carefully with NaH (44mg, 1.097mmol) at 0 ℃ under a nitrogen atmosphere. The resulting mixture was stirred at room temperature for 1H, then treated with a solution of phenyl (5-methoxy-2, 3-dihydro-1H-inden-4-yl) carbamate (120mg, 0.424mmol) in THF (5mL) at 0 deg.C under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred for 6 hours. After completion, the reaction mixture was taken up with saturated NH4The Cl solution was diluted and extracted with EtOAc (2X 30 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. Preparative HPLC on reverse phase [ column: x bridge (150 mm. times.19 mm particle size 5 μm)); flow rate: 15 mL/min; eluent: 10mM ammonium bicarbonate in water (A)&In MeCN (B); gradient: t/% B0/10, 2/10,9/70]The crude product was purified. The fractions were lyophilized to give 4- (2-hydroxypropan-2-yl) -N- ((5-methoxy-2, 3-dihydro-1H-inden-4-yl) carbamoyl) furan-2-sulfonamide as a white solid (45mg, 17%). 1H NMR(400MHz,CD3OD):δ=7.67(s,1H),7.21(s,1H),7.04(d,J=8Hz,1H),6.77(d,J=8Hz,1H),3.79(s,3H),2.84(t,J=7.2Hz,2H),2.69(t,J=7.2Hz,2H),2.01-1.97(m,2H),1.49(s,6H)。LCMS(m/z):393.10[M-H]-;98.97%(210nm),99.47%(254nm)。HPLC:92.07%(210nm),93.87%(254nm)。C18H21N2O6S1[M-H]+HRMS calculated of (a): 393.1126, found: 392.1113.

n- ((7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-amine, 6(70mg, 0.33mmol) was dissolved in THF (5mL) and cooled to 0 ℃. NaH (20mg, 0.505mmol) was added to the above solution under nitrogen and stirred for 15 minutes, then phenyl chloroformate (100mg, 0.674mmol) was added dropwise at 0 ℃. The reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 10% EtOAc-hexanes eluent to give phenyl (7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-yl) carbamate (80mg, 73%) as a brown solid.1H NMR(300MHz,CDCl3):δ=7.39-7.37(m,3H),7.25-7.24(m,2H),6.85(s,1H),3.0-2.94(m,4H),2.12-2.10(m,2H),1.34(m,1H),0.96-0.95(m,2H),0.59-0.57(m,2H)。LCMS(m/z):328.30[M+H]+

4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (56mg, 0.274mmol) was dissolved in anhydrous THF (5mL) and treated carefully with NaH (27mg, 0.685mmol) at 0 deg.C under a nitrogen atmosphere. The resulting mixture was stirred at room temperature for 15 minutes and treated with a solution of phenyl (7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-yl) carbamate (100mg, 0.244mmol) in THF (2mL) at 0 deg.C under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred for 3 hours. After completion, the reaction mixture was taken up with saturated NH 4The Cl solution was diluted, extracted with EtOAc (2 × 30mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. Preparative HPLC on reverse phase [ column: gemini NX C18(21.2mm X150 mm particle size 5 μm)); flow rate: 18 mL/min; eluent: 10mM ammonium bicarbonate in water (A)&In MeCN (B); gradient: t/% B0/20, 2/30,10/50]The crude product was purified. The fractions were lyophilized to give N- ((7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (45mg, 38%) as a white solid.1H NMR(400MHz,DMSO-d6):δ=7.95(d,J=2.4Hz,1H),6.74(s,1H),6.69(d,J=2.4Hz,1H),4.49(m,1H),2.84(t,J=7.6Hz,2H),2.66(t,J=7.6Hz,2H),1.95(m,2H),1.78(m,1H),1.41(d,J=6.4Hz,6H),0.82(m,2H),0.54(m,2H)。LCMS(m/z):437.0[M-H]-,97.99%(210nm)。HPLC:98.26%(210nm)。C20H22Cl1N2O5S1[M-H]+HRMS calculated of (a): 437.0943, found: 437.0927.

4- (2-hydroxypropan-2-yl) -N- ((3-oxo-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) furan-2-sulfonamide

A solution of 8-nitro-3, 5,6, 7-tetrahydro-s-indacen-1 (2H) -one (200mg, 0.92mmol) in MeOH (5mL) was degassed with nitrogen for 5 min, 10% Pd/C (20mg, 10% wt/wt) was added, and the mixture was stirred at room temperature under a hydrogen atmosphere for 2H. The reaction mixture was filtered through celite, and the filtrate was concentrated in vacuo to give 8-amino-3, 5,6, 7-tetrahydro-s-indacen-1 (2H) -one as an off-white solid (160mg, 93%).1H NMR(600MHz,DMSO-d6)δ6.49(s,1H),6.34(s,2H),2.90-2.84(m,2H),2.80(t,J=7.5Hz,2H),2.62(t,J=7.4Hz,2H),2.56-2.51(m,2H),2.04-1.99(m,2H)。13C NMR(150MHz,DMSO-d6):206.6,155.4,153.7,144.1,125.3,118.6,109.4,36.8,33.7,28.6,25.0,24.9。LCMS(m/z):188[M+H]+。C2H14N1O1[M+H]+HRMS calculated of (a): 188.1070, found: 188.1077.

DMAP (26.1mg, 0.21mmol) was added to di-tert-butyl dicarbonate (163mg, 0.74mmol) in dry acetonitrile (1mL) at room temperature, stirred for 5 min and a solution of 8-amino-3, 5-6, 7-tetrahydro-s-indacen-1 (2H) -one (100mg, 0.53mmol) in acetonitrile was added. The reaction mixture was stirred at room temperature for 30 minutes. The reaction mixture was used directly in the next step without work-up.

4- (2) in anhydrous THF (1mL) at 0 deg.C-hydroxypropan-2-yl) furan-2-sulfonamide intermediate (100mg, 0.48mmol) was added NaH (18.3mg, 0.48mmol) and stirred at ambient temperature under nitrogen for 30 minutes. Cooled again to 0 ℃, 8-isocyanato-3, 5,6, 7-tetrahydro-s-indacen-1 (2H) -one (previous reaction mixture) was added and stirred at ambient temperature for 16 hours. To the reaction mixture was added 0.5mL of H2O, directly loaded on a C18 column to be purified with 10mM (NH)4)HCO3The aqueous solution and acetonitrile were purified as mobile phases to give 4- (2-hydroxypropan-2-yl) -N- ((3-oxo-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) furan-2-sulfonamide as a white solid (150mg, 67%).1H NMR(600MHz,DMSO-d6):δ8.79(s,1H),7.37(s,1H),6.94(s,1H),6.61(s,1H),4.92(s,1H),2.92(t,J=5.6Hz,2H),2.82(t,J=7.5Hz,2H),2.75(t,J=7.5Hz,2H),2.63-2.57(m,2H),1.97-1.80(m,2H),1.34(s,6H);LCMS(m/z):417[M-H]-。C20H23N2O6S1[M+H]+HRMS calculated of (a): 419.1271, found: 419.1291

To a solution of 4- (2-hydroxypropan-2-yl) -N- ((3-oxo-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) furan-2-sulfonamide (70mg, 0.16mmol) in MeOH (2mL) at 0 deg.C under a nitrogen atmosphere was added NaBH 4(63mg, 1.67mmol) and the resulting reaction mixture was stirred at room temperature for 3 hours. Subjecting the reaction mixture to hydrogenation with H2O (2mL) quench, distill off MeOH, load the aqueous layer directly onto a C18 column using 10mM (NH)4)HCO3The aqueous solution and acetonitrile were purified as mobile phases to give N- ((3-hydroxy-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (60mg, 86%) as an off-white solid.1H NMR(600MHz,DMSO-d6)δ7.73(bs,1H),7.38(s,1H),6.81(s,1H),6.60(s,1H),5.63(bs,1H),4.92(bs,1H),4.87(d,J=6.0Hz,1H),3.00-2.84(m,2H),2.77(t,J=7.4Hz,2H),2.64-2.53(m,2H),2.07-2.00(m,1H),1.97-1.92(m,1H),1.91-1.81(m,2H),1.35(s,6H);13C NMR(150MHz,DMSO-d6):159.5,156.0,144.9,143.3,138.3,137.7,136.8,136.0,133.0,72.6,67.0,35.4,33.1,31.5,31.4,31.0,30.4,25.5;LCMS(m/z):419[M-H]-;C20H23N2O6S1[M-H]-HRMS calculated of (a): 419.1282, found: 419.1263.

n- ((1-hydroxy-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

A solution of 4-nitro-3, 5,6, 7-tetrahydro-s-indacen-1 (2H) -one (110mg, 0.50mmol) in MeOH (5mL) was degassed with nitrogen for 5 minutes, 10% Pd/C (11mg, 10% wt/wt) was added, and the mixture was stirred at room temperature under a hydrogen atmosphere for about 2 hours. The reaction mixture was filtered through a pad of celite and the filtrate was concentrated to give 4-amino-3, 5,6, 7-tetrahydro-s-indacen-1 (2H) -one as an off-white solid (75mg, 80%).1H NMR(600MHz,DMSO-d6)δ6.72(s,1H),5.11(s,2H),2.87-2.74(m,4H),2.70(t,J=7.4Hz,2H),2.62-2.54(m,2H),2.06-1.99(m,2H);13C NMR(150MHz,DMSO-d6)δ206.7,144.6,141.7,139.9,136.9,136.8,104.2,39.9,36.7,30.6,30.5,24.5,23.7.LCMS(m/z):188[M+H]+;C12H14N1O1[M+H]+HRMS calculated of (a): 188.1070, found: 188.1074.

to di-tert-butyl dicarbonate (81.6mg, 0.37mmol) in anhydrous acetonitrile (1mL) was added DMAP (13.0mg, 0.04mmol) at room temperature, stirred for 5 minutes, and a solution of 4-amino-3, 5,6, 7-tetrahydro-s-indacen-1 (2H) -one (50mg, 0.26mmol) in acetonitrile (1mL) was added. The reaction mixture was stirred at room temperature for 30 minutes. The reaction mixture was used directly in the next step without work-up.

To 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide intermediate (50mg, 0.24mmol) in anhydrous THF (1mL) was added NaH (9.3mg, 0.24mmol) at 0 deg.C and stirred at ambient temperature under nitrogen for 30 minutes. Cooled again to 0 ℃, 4-isocyanato-3, 5,6, 7-tetrahydro-s-indeno-1 (2H) -one (previous reaction mixture) was added and stirred at ambient temperature for 16 hours. To the reaction mixture was added 0.5mLH2O, directly loaded on a C18 column to be purified with 10mM (NH)4)HCO3The aqueous solution and acetonitrile were purified as mobile phases to give 4- (2-hydroxypropan-2-yl) -N- ((1-oxo-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) furan-2-sulfonamide (70mg, 63%).1H NMR(600MHz,DMSO-d6)δ7.94(s,1H),7.38(s,1H),7.17(s,1H),6.60(s,1H),4.92(s,1H),2.94-2.89(m,2H),2.85(t,J=7.4Hz,2H),2.80(t,J=7.4Hz,2H),2.56-2.52(m,2H),2.00-1.94(m,2H),1.35(s,6H);LCMS(m/z):417[M-H]-;C20H21N2O6S1[M-H]-HRMS calculated of (a): 417.1126, found: 417.1113.

to a solution of 4- (2-hydroxypropan-2-yl) -N- ((1-oxo-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) furan-2-sulfonamide (50mg, 0.11mmol) in MeOH (2mL) at 0 deg.C under a nitrogen atmosphere was added NaBH4(45mg, 1.19mmol) and the resulting reaction mixture was stirred at room temperature for 3 hours. By H2O (1mL) quench the reaction mixture, evaporate the MeOH, load the aqueous layer directly onto a C18 column using 10mM (NH)4)HCO3The aqueous solution and acetonitrile were purified as mobile phases to give N- ((1-hydroxy-1, 2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (20mg, 40%). 1H NMR(600MHz,DMSO-d6)δ7.77(s,1H),7.51(s,1H),6.93(s,1H),6.79(s,1H),6.55(s,1H),5.05(d,J=5.8Hz,1H),4.99(s,1H),4.94(q,J=6.4Hz,1H),2.79(t,J=7.5Hz,2H),2.70-2.61(m,3H),2.52-2.49(m,1H),2.2-2.21(m,1H),1.95-1.90(m,2H),1.74-1.59(m,1H),1.36(s,6H);13C NMR(150MHz,DMSO-d6):163.5,146.0,143.1,140.6,138.5,136.5,136.2,122.0,116.5,112.5,108.5,74.9,67.0,36.0,33.0,31.5,30.9,27.8,25.6;LCMS(m/z):419[M-H]-.C20H23N2O6S1[M-H]-HRMS calculated of (a): 419.1282, found: 419.1265.

n- ((4, 6-dimethylpyrimidin-2-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

A solution of 4, 6-dimethylpyrimidin-2-amine (200mg, 1.62mmol) in THF (5mL) was cooled to 0 deg.C and treated with NaH (130mg, 3.24mmol) under a nitrogen atmosphere. The reaction mixture was stirred for 15 minutes and treated with phenyl chloroformate (380mg, 2.43mmol) at 0 ℃ under a nitrogen atmosphere. The reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluent to give phenyl (4, 6-dimethylpyrimidin-2-yl) carbamate as a white solid (200mg, 51%).1H NMR(300MHz,CDCl3):δ=7.91(s,1H),7.40-7.35(m,2H),7.24-7.19(m,3H),6.78(s,1H),2.41(s,6H)。LCMS(m/z):244.20[M+H]+

4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (150mg, 0.731mmol) was dissolved in anhydrous THF (5mL) and treated carefully with NaH (75mg, 1.83mmol) at 0 deg.C under nitrogen. The resulting mixture was warmed to 60 ℃ and stirred for 2 hours. The solution was cooled to 0 ℃ and treated with a solution of phenyl (4, 6-dimethylpyrimidin-2-yl) carbamate (195mg, 0.804mmol) in THF (5mL) at 0 ℃ under a nitrogen atmosphere. The reaction mixture was warmed to 50 ℃ for 4 hours and then stirred at room temperature for 4 hours. After completion, the reaction mixture was taken up with saturated NH 4The Cl solution was diluted, extracted with EtOAc (2 × 30mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. Preparative HPLC on reverse phase [ column: x-bridge (150 mm. times.19 mm particle size 5 μm)); flow rate: 15 mL/min; eluent: 10mM ammonium acetate in 0.1% AcOH in water (A)&In MeCN (B); gradient: t/% B0/15, 2/25,8/40]The crude product was purified. The fractions were lyophilized to give N- ((4, 6-dimethylpyrimidin-2-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (25mg, 7%) as a white solid.1H NMR(400MHz,CDCl3):δ=8.18(s,1H),7.51(d,J=0.8Hz,1H),7.40(d,J=1.2Hz,1H),6.78(s,1H),2.48(s,6H),1.57(s,6H)。LCMS(m/z):355.0[M+H]+,100%(210nm),100%(254nm)。HPLC:96.49%(210nm),98.76%(254nm)。C14H17N4O5S1[M-H]-HRMS calculated of (a): 353.0925, found: 353.0921.

n- ((4-cyclopropyl-6-methylpyrimidin-2-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

4-cyclopropyl-6-methylpyrimidin-2-amine (50mg, 0.33mmol) was dissolved in THF (2mL) and cooled to 0 ℃. NaH (16mg, 0.40mmol) was carefully added to the above solution and stirred for 20 min. Phenyl chloroformate (80mg, 0.503mmol) was added dropwise at 0 ℃. The reaction mixture was warmed to room temperature and stirred at room temperature for 12 hours. Reaction completion (TLC, 50% EtOAc-hexane, R)f0.4), the reaction mixture was diluted with EtOAc and filtered through a pad of Celite. The filtrate was concentrated in vacuo and the crude product was purified by column chromatography on silica gel (60-120 mesh) using 30% EtOAc-hexanes eluent to give phenyl (4-cyclopropyl-6-methylpyrimidin-2-yl) carbamate as an off-white solid (40mg, 44%). 1H NMR(300MHz,CDCl3):δ=8.06(s,1H),7.40-7.37(m,2H),7.24-7.15(m,3H),6.73(s,1H),2.4(s,3H),1.94-1.86(m,1H),1.15-1.1(m,2H),1.0-0.99(m,2H)。LCMS(m/z):270.3[M+H]+4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (150mg, 0.731mmol) was dissolved in anhydrous THF (5mL) and treated carefully with NaH (73mg, 1.829mmol) at 0 deg.C under nitrogen. The resulting mixture was stirred at room temperature for 1 hour and treated with a solution of phenyl (4-cyclopropyl-6-methylpyrimidin-2-yl) carbamate (190mg, 0.731mmol) in THF (5mL) at 0 deg.C under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred at room temperature for 6 hours. After completion, the reaction mixture was taken up with saturated NH4The Cl solution was diluted and extracted with EtOAc (2X 30 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. Preparative HPLC on reverse phase [ column: x bridge (150mm is multiplied by 19 mm)Particle size 5 μm)); flow rate: 15 mL/min; eluent: 10mM ammonium bicarbonate in water (A)&In MeCN (B); gradient: t/% B0/15, 2/25,8/40]The crude product was purified. The fractions were lyophilized to give N- ((4-cyclopropyl-6-methylpyrimidin-2-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (30mg, 11%) as a white solid.1H NMR(400MHz,CD3OD):δ=7.53(s,1H),7.07(s,1H),6.80(s,1H),2.35(s,3H),1.97-1.93(m,1H),1.42(s,6H),1.08-1.02(m,4H)。LCMS(m/z):381.00[M+H]+;98.60%(210nm),99.49%(254nm)。HPLC:98.05%(210nm),99.01%(254nm)。C16H19N4O5S1[M-H]+HRMS calculated of (a): 379.1082, found: 379.1082.

n- ((4, 6-di-tert-butylpyrimidin-2-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

4, 6-di-tert-butylpyrimidin-2-amine (0.15g, 0.72mmol) was dissolved in THF (5mL) and cooled to 0 ℃. To the above solution was added NaH (37mg, 0.93mmol), and the resulting mixture was stirred under nitrogen for 15 minutes. Phenyl chloroformate (0.16g, 1.08mmol) was added dropwise to the above solution at 0 ℃. The reaction mixture was warmed to room temperature and stirred for 12 hours. Reaction completion (TLC, 10% EtOAc-hexane, R)f0.5), the reaction mixture is concentrated in vacuo. The resulting residue was taken up in 10% IPA/CHCl3Dilute, filter through a pad of celite and concentrate in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 4% EtOAc-hexanes eluent to give phenyl (4, 6-di-tert-butylpyrimidin-2-yl) carbamate as a white solid (0.1g, 43%).1H NMR(300MHz,CDCl3):δ=7.65(s,1H),7.38(m,2H),7.22(m,3H),7.01(s,1H),1.32(s,18H)。LCMS(m/z):327.80[M+H]+

4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (65mg, 0.305mmol) was dissolved in anhydrous THF (8mL) and treated carefully with NaH (30mg, 0.764mmol) at 0 deg.C under nitrogen at 0 deg.C. The resulting mixture was stirred at room temperature for 45 minutes and treated dropwise with a solution of phenyl (4, 6-di-tert-butylpyrimidin-2-yl) carbamate (100mg, 0.305mmol) in THF (5mL) at 0 ℃ under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred for 3 hours. After completion of the reaction, (TLC, 50% EtOAc-hexane, R f0.5), the reaction mixture was washed with saturated NH4The Cl solution was diluted and extracted with EtOAc (2X 20 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluent to give N- ((4, 6-di-tert-butylpyrimidin-2-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (0.07g, 52%) as a white solid.1H NMR(400MHz,DMSO-d6):δ=13.75(s,1H),10.71(s,1H),7.83(s,1H),7.37(s,1H),7.20(s,1H),5.17(s,1H),1.39(s,6H),1.31(s,18H)。LCMS(m/z):439.55[M+H]+;94.58%(210nm),97.94%(254nm)。HPLC:98.51%(210nm),99.27%(254nm)。C20H29N4O5S1[M-H]+HRMS calculated of (a): 437.1864, found: 437.1846.

methyl furan

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) -5-methylfuran-2-sulfonamide

Using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 4- (2-hydroxypropan-2-yl) -5-methylfuran-2-sulfonamide in general method C2 to give the title compound as a white solid (52mg, 51%).

N- ((2, 6-diisopropylphenyl) carbamoyl) -4- (2-hydroxypropan-2-yl) -5-methylfuran-2-sulfonamide

2-Isocyanato-1, 3-diisopropylbenzene (prepared using general procedure A1) and 4- (2-hydroxypropan-2-yl) -5-methylfuran-2-sulfonamide were used in general procedure C1 to give the title compound as an off-white solid (14mg, 4%). 1H NMR(400MHz,CD3OD):δ=7.24(t,J=7.6Hz,1H),7.15(d,J=7.6Hz,2H),7.04(s,1H),3.10(sept.,J=6.8Hz,2H),2.50(s,3H),1.50(s,6H),1.17(d,J=6.8Hz,12H)。

Deuterated furans

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl-1, 1,1,3,3,3-d6) Furan-2-sulfonamides

4-Isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general procedure A1) and d can be used in general procedure C16-4- (2-hydroxypropan-2-yl) -5-methylfuran-2-sulfonamide synthesis of N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide.

Alternatively, d is used at-10 ℃ under vigorous stirring for 10 minutes3Solution of methyl magnesium iodide (1.0M in Et)2O, 10 equivalents) ethyl 2-methyl-5-sulfamoylfuran-3-carboxylate (0.4g, 0.96mmol) in anhydrous THF (30mL) was treated dropwise. The solution was then stirred at ambient temperature for 12 hours, then cooled to 0 ℃ and quenched dropwise with saturated ammonium chloride solution. The aqueous solution was extracted with EtOAc (2X 20mL), and the combined organics were washed with brine (20mL) and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC to give the title compound as a white solid (5mg, 1%).1H NMR(300MHz,CD3OD):δ=7.50(d,J=1.2Hz,1H),6.95(d,J=1.2Hz,1H),6.89(s,1H),2.83(t,J=7.2Hz,4H),2.75(t,J=7.2Hz,4H),2.02(quin,J=7.2Hz,4H)。

N- ((1,2,3,5,6, 7-hexahydro-s-Indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl-1, 1,1,3,3,3-d6) -5-methylfuran-2-sulfonamide

In general procedure C1 using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general procedure A1) and d6-4- (2-hydroxypropan-2-yl) -5-methylfuran-2-sulfonamide to give the title compound as a white solid (10mg, 3%).1H NMR(400MHz,CD3OD)δ=7.03(s,1H),6.95(s,1H),2.86(t,J=7.4Hz,4H),2.73(t,J=7.4Hz,4H),2.48(s,3H),2.04(p,J=7.4Hz,4H)。

4- (2-hydroxypropan-2-yl-1, 1,1,3,3,3-d6) -5-methyl-N- ((3,5,6, 7-tetrahydro-2H-indeno [5, 6-b)]Furan-4-yl) carbamoyl) furan-2-sulphonamides

Use of 4-Isocyanato-3, 5,6, 7-tetrahydro-2H-indeno [5,6-b ] in general procedure C1]Furan (prepared by general procedure A1) and d6-4- (2-hydroxypropan-2-yl) -5-methylfuran-2-sulfonamide to give the title compound as a white solid (20mg, 5%).1H NMR(400MHz,CD3OD)δ=7.13(s,1H),6.52(s,1H),4.51(t,J=8.6Hz,2H),3.03(t,J=8.6Hz,2H),2.84(t,J=7.4Hz,2H),2.68(t,J=7.4Hz,2H),2.50(s,3H),2.05(p,J=7.4Hz,2H)。

N- ((4-bromo-3, 5,6, 7-tetrahydro-2H-indeno [5, 6-b)]Furan-8-yl) carbamoyl) -4- (2-hydroxypropan-2-yl-1, 1,1,3,3,3-d6) -5-methylfuran-2-sulfonamide

Use of 4-bromo-8-isocyanato-3, 5,6, 7-tetrahydro-2H-indeno [5,6-b ] in general procedure C1]Furan (using general procedure)Preparation of A1) and d6-4- (2-hydroxypropan-2-yl) -5-methylfuran-2-sulfonamide to give the title compound as a white solid (32mg, 39%).1H NMR(400MHz,DMSO-d6)δ=8.01(s,1H),7.07(s,1H),5.04(s,1H),4.59(t,J=8.7Hz,2H),3.14(t,J=8.7Hz,2H),2.77(t,J=7.4Hz,2H),2.69(t,J=7.4Hz,2H),2.43(s,3H),1.98(q,J=7.4Hz,2H)。

Thiophene(s)

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) thiophene-2-sulfonamide

The title compound was obtained as a white solid (11mg, 11%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and thiophene-2-sulfonamide in general method C2.1H NMR(400MHz,CD3OD):δ=7.79(d,J=4.0Hz,1H),7.76(d,J=4.0Hz,1H),7.73(t,J=4.0Hz,1H),6.93(s,1H),2.83(t,J=12Hz,4H),2.66(t,J=12Hz,4H),2.04-1.96(m,4H)。13C NMR(100MHz,CD3OD) delta 143.5,143.2,137.8,132.7,132.2,126.6,126.4,118.2,110.3,32.5,29.9, 25.1; LCMS purity:>95%;LCMS(m/z):363[M+H]+;C17H18N2O3S2(M+H)+HRMS calculated of (a): 363.0832, found: 363.0819.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -5-methylthiophene-2-sulphonamide

4-Isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure A2) 5-methylthiophene-2-sulfonamide was used in general method C2 to give the title compound as a white solid (12mg, 18%).1H NMR(400MHz,DMSO-d6):δ=7.88(s,1H),7.43(d,J=4.0Hz,1H),6.89(s,1H),6.82(d,J=4.0Hz,1H),2.78(t,J=12Hz,4H),2.61(t,J=12Hz,4H),2.47(s,3H),1.97-1.89(m,4H)。13C NMR(100MHz,DMSO-d6) δ 143.2,142.9,137.3,130.6,126.0,125.6,125.2,117.5,108.7,32.69,30.7,25.5, 15.4; LCMS purity:>95%;LCMS(m/z):377[M+H]+;C18H20N2O3S2,(M+H)+HRMS calculated of (a): 377.0988, found: 377.0994.

thiazoles

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) thiazole-2-sulfonamide

The title compound was obtained as a white solid (8mg, 20%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and thiazole-2-sulfonamide in general method C2.1H NMR(400MHz,CD3OD):δ=7.02(d,1H,J=4.0Hz),6.99(s,1H),6.60(d,1H,J=4.0Hz),2.88(t,4H,J=8.0Hz),2.76(t,4H,J=8.0Hz),2.08-2.02(m,4H);13C NMR(100MHz,CD3OD):δ=169.6,144.2,144.1,137.7,137.5,132.4,118.1,106.9,32.4,29.9,25.2。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -2-methylthiazole-5-sulfonamide

Using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 2-methylthiazole-5-sulfonamide in general method C3 to give the title compound as a white solid (35mg, 65%);1H NMR(600MHz,DMSO-d6)δ=7.73(s,1H),7.53(s,1H),6.78(s,1H),2.75(t,J=7.4Hz,4H),2.66(t,J=7.4Hz,4H),2.59(s,3H),1.93-1.88(m,4H);13C NMR(150MHz,DMSO-d6):166.8,158.3,143.9,142.0,141.4,136.6,132.3,115.5,32.5,30.4,25.0, 18.6; LCMS purity:>95%;LCMS(m/z):378[M+H]+;C17H18N3O3S2[M-H]+HRMS calculated of (a): 376.0795, found: 376.0791.

triazole compounds

1-benzyl-N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1H-1,2, 4-triazole-3-sulfonamide

Use of 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method A1) and 1-benzyl-1H-1, 2, 4-triazole-3-sulfonamide in general procedure C3 to give the title compound as a white solid (40mg, 15%)1H NMR(400MHz,DMSO-d6):δ=8.9(s,1H),8.0(s,1H),7.35-7.28(m,5H),6.90(s,1H),5.48(s,2H),2.77(t,J=7.2Hz,4H),2.59(t,J=7.2Hz,4H),1.95-1.90(m,4H)。LCMS(m/z):438.10(M+1)+95.84%(210nm),97.84%(254nm)。HPLC:95.99%(210nm),95.31%(254nm)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4H-1,2, 4-triazole-3-sulfonamide

Using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 4H-1,2, 4-triazole-3-sulfonamide in general method C3 to give the title compound as a white solid (31mg, 62%);1H NMR(600MHz,DMSO-d6)δ=9.83(bs,1H),7.99(s,1H),7.62(s,1H),6.77(s,1H),2.73(t,J=7.4Hz,4H),2.64(t,J=7.4Hz,4H),1.91-1.86(m,4H);13C NMR(150MHz,DMSO-d6) 159.2,149.8,148.7,142.5,137.2,132.8,116.1,33.0,30.9, 25.6; LCMS purity:>95%;LCMS(m/z):348[M+H]+;C15H16N5O3S1[M-H]-HRMS calculated of (a): 346.0979, found: 346.0983.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1-isopropyl-1H-1, 2, 3-triazole-4-sulfonamide

1,2,3,5,6, 7-hexahydro-s-indacen-4-amine, 7(100mg, 0.578mmol) was dissolved in anhydrous THF (5mL) and treated with Et at room temperature3N (70mg, 0.693 mmol). The solution was treated with triphosgene (70mg, 0.231mmol) and the resulting mixture was stirred at 70 ℃ for 3 hours. The reaction mixture was concentrated in vacuo. The resulting residue was stirred with n-pentane (20mL) for 10 minutes and filtered through celite. The filtrate was concentrated in vacuo to give the isocyanate as a white solid. In another 50mL round bottom flask, 1-isopropyl-1H-1, 2, 3-triazole-4-sulfonamide (95mg, 0.50mmol) was dissolved in anhydrous THF (5mL) and carefully treated with NaH (42mg, 1.05mmol) under nitrogen at 0 ℃. It was stirred at room temperature for 45 minutes and treated with a solution of the above isocyanate in THF under nitrogen. The resulting reaction mixture was stirred at room temperature for 5 hours. After completion (TLC, 70% EtOAc-hexane, R)f0.3), the reaction mixture is saturated with NH4The Cl solution was diluted and extracted with EtOAc (2X 25 mL). The combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. Preparative HPLC on reverse phase [ column: gemini NX C18(21.5mm X150 mm particle size 5 μm)); flow rate: 15 mL/min; eluent: 10mM ammonium bicarbonate in water (A) &In MeCN (B); gradient: t/% B0/10, 2/20,8/65]The crude product was purified. The fractions were lyophilized to give N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1-isopropyl-1H-1, 2, 3-triazole-4-sulfonamide (25mg, 12%) as a white solid.1H NMR(400MHz,DMSO-d6):δ=8.75(s,1H),7.91(s,1H),6.89(s,1H),4.9(m,1H),2.79(t,J=7.2Hz,4H),2.60(t,J=7.2Hz,4H),1.96-1.89(m,4H),1.5(d,J=6.8Hz,6H)。LCMS(m/z):390.10[M+H]+;100%(210nm),100%(254nm)。HPLC:96.05%(210nm),96.13%(254nm)。C18H22N5O3S1[M-H]-HRMS calculated of (a): 388.1449, found: 388.1457.

pyrazoles

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1-methyl-1H-pyrazole-5-sulfonamide

4-Isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure A2) and 1-methyl-1H-pyrazole-5-sulfonamide were used in general procedure C2 to give the title compound (8mg) as a white solid, 20%.1H NMR(400MHz,DMSO-d6):δ=7.95(bs,1H),7.45(s,1H),6.88(s,1H),6.66(s,1H),4.02(s,3H),2.77(t,J=16Hz,4H),2.60(t,J=16Hz,4H),1.96-1.88(m,4H)。13C NMR (150MHz, DMSO-d6) δ 143.1,142.9,137.2,125.2,117.4,110.0,109.0,108.7,38.6,33.0,30.7, 25.5; LCMS purity:>95%;LCMS(m/z):361[M+H]+;C17H20N4O3S(M+H)+HRMS calculated of (a): 361.13289, found: 361.13213.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1-methyl-1H-pyrazole-3-sulfonamide

The title compound was obtained as a white solid (40mg, 8%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1-methyl-1H-pyrazole-3-sulfonamide in general method C1. 1H NMR(400MHz,DMSO-d6):δ=10.8(brs,1H),8.02(s,1H),7.86(s,1H),6.92(s,1H),6.69(s,1H),3.91(s,3H),2.80(t,J=7.2Hz,4H),2.62(t,J=7.2Hz,4H),1.96(t,J=7.2Hz,4H)。LCMS(m/z):383.10(M+Na)+;96.00%(210nm),93.44%(254nm)。HPLC:97.86%(210nm),97.44%(254nm)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1- (trifluoromethyl) -1H-pyrazole-3-sulfonamide

The title compound was obtained as a white solid (5mg, 1%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1- (trifluoromethyl) -1H-pyrazole-3-sulfonamide in general method C2.1H NMR(400MHz,CD3OD)δ=8.28(d,J=2.8Hz,1H),6.96(d,J=2.8,1H),6.91(s,1H),2.84(t,J=7.4Hz,4H),2.75(t,J=7.4Hz,4H),2.03(m,J=7.4Hz,4H)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide

The title compound was obtained as an off-white solid (40mg, 9%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1-isopropyl-1H-pyrazole-3-sulfonamide in general method C1.1H NMR(400MHz,DMSO-d6):δ=10.92(s,1H),8.02(s,1H),8.0(s,1H),6.94(s,1H),6.74(s,1H),4.67-4.59(m,1H),2.78(t,J=7.2Hz,4H),2.58(t,J=7.2Hz,4H),1.95-1.91(m,4H),1.44(d,J=6.8Hz,6H)。LCMS(m/z):387.1(M-1)-;97.14%(210nm),95.11%(254nm)。HPLC:95.57%(210nm),93.53%(254nm)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1-isopropyl-1H-pyrazole-4-sulfonamide

In general procedure C3, 4-isocyanato-8-methyl-1, 2,3,5,6,7-hexahydro-s-indacene (prepared by general method A1) and 1-isopropyl-1H-pyrazole-4-sulfonamide to give the title compound as a white solid (40mg, 10%)1H NMR(400MHz,DMSO-d6):δ=10.6(s,1H),8.44(s,1H),8.05(s,1H),7.86(s,1H),6.94(s,1H),4.63-4.57(m,1H),2.80(t,J=7.2Hz,4H),2.57(t,J=7.6Hz,4H),1.94-1.89(m,4H),1.42(d,J=6.8Hz 6H)。LCMS(m/z):389.20(M+1)+;97.25%(210nm),94.22%(254nm)。HPLC:97.13%(210nm),95.06%(254nm)。

1-cyclopropyl-N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1H-pyrazole-3-sulfonamide

Using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1-cyclopropyl-1H-pyrazole-3-sulfonamide in general method C3 to give the title compound as a white solid (20mg, 6%). 1H NMR(400MHz,DMSO-d6)δ=7.83(s,1H),7.8(s,1H),6.84(s,1H),6.48(s,1H),3.81–3.71(m,1H),2.77(t,J=7.4Hz,4H),2.64(t,J=7.4Hz,4H),2.02–1.86(m,4H),1.09–0.93(m,4H)。

1- (tert-butyl) -N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1H-pyrazole-3-sulfonamide

Using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1- (tert-butyl) -1H-pyrazole-3-sulfonamide in general method C3 to give the title compound as a light yellow solid (120mg, 51%).1H NMR(400MHz,DMSO-d6):δ=10.85(br.s.,1H),7.95(s,1H),7.88(br.s.,1H),6.88(s,1H),6.63(s,1H),2.79(t,J=7.2Hz,4H),2.61(t,J=7.2Hz,4H),1.96(m,4H),1.55(s,9H)。LCMS(m/z):403.15(M+1)+;97.86%(210nm),96.50%(254nm)。HPLC:96.45%(210nm),95.89%(254nm)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1- (1-methylpiperidin-4-yl) -1H-pyrazole-3-sulfonamide

The title compound was obtained as a white solid (20mg, 6%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1-cyclohexyl-1H-pyrazole-3-sulfonamide in general method C3.1H NMR(400MHz,DMSO-d6)δ=10.8(s,1H),8.03(s,1H),7.99(d,J=2.4,1H),6.95(s,1H),6.75(d,J=2.4Hz,1H),4.33–4.20(m,1H),2.79(t,J=7.4Hz,4H),2.58(t,J=7.4Hz,4H),2.05–1.88(m,6H),1.86–1.63(m,6H),1.48–1.33(m,2H)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1-phenyl-1H-pyrazole-3-sulfonamide

The title compound was obtained as an off-white solid (110mg, 27%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1-phenyl-1H-pyrazole-3-sulfonamide in general method C1.1H NMR(400MHz,DMSO-d6):δ=10.92(s,1H),8.61(d,J=2.4Hz,1H),7.95(br.s.,1H),7.86(d,J=8.4Hz,2H),7.56(t,J=7.6Hz,2H),7.41(t,J=7.2Hz,1H),6.9(d,J=2.0Hz,1H),6.86(s,1H),2.77(t,J=7.2Hz,4H),2.62(t,J=7.2Hz,4H),1.91-1.83(m,4H)。LCMS(m/z):421.05(M-1)-;96.62%(210nm),95.12%(254nm)。HPLC:95.2%(210nm),95.77%(254nm)。

1-benzyl-N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1H-pyrazole-3-sulfonamide

The title compound was obtained as a white solid (85mg, 34%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1-benzyl-1H-pyrazole-3-sulfonamide in general method C3. 1H NMR (400MHz, DMSO-d) 6):δ=10.85(s,1H),8.05(d,J=2.4Hz,1H),7.99(s,1H),7.32-7.31(m,3H),7.24-7.22(m,2H),6.93(s,1H),6.78(d,J=2.4Hz,1H),5.44(s,2H),2.80(t,J=7.6Hz,4H),2.57(t,J=7.2Hz,4H),1.96(m,4H)。LCMS(m/z):437.15(M+1)+;97.70%(210nm),96.86%(254nm)。HPLC:98.05%(210nm),97.56%(254nm)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1- (1-phenylethyl) -1H-pyrazole-3-sulfonamide

The title compound was obtained as a white solid (0.13g, 38%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1- (1-phenylethyl) -1H-pyrazole-3-sulfonamide in general method C3.1H NMR(400MHz,DMSO-d6)δ=7.94(d,J=2.4Hz,1H),7.80(s,1H),7.34–7.18(m,5H),6.85(s,1H),6.62(d,J=2.3Hz,1H),5.68(q,J=7.0Hz,1H),2.76(t,J=7.4Hz,4H),2.58(t,J=7.4Hz,4H),1.90(p,J=7.4Hz,4H),1.8(d,J7.1Hz,3H)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1- (2- (piperidin-1-yl) ethyl) -1H-pyrazole-3-sulfonamide

The title compound was obtained as a white solid (110mg, 25%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1- (2- (piperidin-1-yl) ethyl) -1H-pyrazole-3-sulfonamide in general method C3.1H NMR(400MHz,CD3OD):δ=7.76(d,J=2.4Hz,1H),6.91(s,1H),6.73(d,J=2.4Hz 1H),4.55(t,J=6.4Hz,2H),3.41(t,J=6.0Hz,2H),3.02(s,4H),2.86(t,J=7.2Hz,4H),2.78(t,J=7.2Hz,4H),2.06-1.99(m,4H),1.74-1.70(m,4H),1.51(d,J=5.2Hz,2H)。LCMS(m/z):458.20(M+1)+;100%(210nm),100%(254nm)。HPLC:98.70%(210nm),98.31%(254nm)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1, 5-dimethyl-1H-pyrazole-3-sulfonamide

Using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1, 5-dimethyl-1H-pyrazole-3-sulfonamide in general method C1 to give the title compound as a white solid (15mg, 4%).1H NMR(400MHz,DMSO-d6):δ=10.7(br.s.,1H),7.98(s,1H),6.93(s,1H),6.52(s,1H),3.79(s,3H),2.80(t,J=7.2Hz,4H),2.62(t,J=7.6Hz,4H),2.28(s,3H),1.98-1.93(m,4H)。LCMS(m/z):397.10(M+Na)+;97.75%(210nm),88.23%(254nm)。HPLC:94.42%(210nm),95.19%(254nm)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1-methyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonamide

The title compound was obtained as a white solid (200mg, 48%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1-methyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonamide in general method C3.1H NMR(400MHz,CD3OD):δ=7.10(s,1H),6.87(s,1H),4.03(s,3H),2.83(t,J=7.2Hz,4H),2.74-(t,J=7.2Hz,4H),2.03-1.99(m,4H)。LCMS(m/z):429.10(M+1)+;97.73%(210nm),95.71%(254nm)。HPLC:94.95%(210nm),93.52%(254nm)。

N- ((2, 6-diisopropylphenyl) carbamoyl) -1-methyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonamide

2-Isocyanato-1, 3-diisopropylbenzene (prepared using general procedure A1) and 1-methyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonamide were used in general procedure C3 to give the title compound as a white solid (70mg, 39%).1H NMR(400MHz,CD3OD):δ=7.18-7.16(m,1H),7.10-7.08(m,3H),4.03(s,3H),3.17-3.13(m,2H),1.03(d,J=6.0Hz,12H)。LCMS(m/z):433.15(M+1)+;99.73%(210nm),98.16%(254nm)。HPLC:97.51%(210nm),95.47%(254nm)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1-isopropyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonamide

The title compound was obtained as a white solid (15mg, 12%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1-isopropyl-5- (trifluoromethyl) -1H-pyrazole-3-sulfonamide in general method C3.1H NMR(400MHz,DMSO-d6):δ=8.54(s,1H),6.90(s,1H),6.77(s,1H),4.62-4.56(m,1H),2.76(t,J=7.2Hz,4H),2.67(t,J=7.6Hz,4H),1.92-1.84(m,4H),1.44(d,J=6.4Hz,6H)。LCMS(m/z):455.05(M-1)-;96.13%(210nm),95.41%(254nm)。HPLC:95.71%(210nm),95.12%(254nm)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -5-isopropyl-1-methyl-1H-pyrazole-3-sulfonamide

In general procedure C3 use was made of 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general procedure A1) and 5 -isopropyl-1-methyl-1H-pyrazole-3-sulfonamide to give the title compound as a white solid (10mg, 17%).1H NMR(400MHz,CD3OD):δ=6.88(s,1H),6.50(s,1H),3.82(s,3H),3.08–3.03(m,1H),2.83(t,J=7.2Hz,4H),2.71(t,J=7.2Hz,4H),2.04-1.96(m,4H),1.21(d,J=6.8Hz,6H)。LCMS(m/z):403.20(M+1)+;98.39%(210nm),94.19%(254nm)。HPLC:95.62%(210nm),93.00%(254nm)。

N- ((2, 6-diisopropyl) carbamoyl) -5- (2-hydroxypropan-2-yl) -1-methyl-1H-pyrazole-3-sulfonamide

2-Isocyanato-1, 3-diisopropylbenzene (prepared using general procedure A1) and 5- (2-hydroxypropan-2-yl) -1-methyl-1H-pyrazole-3-sulfonamide were used in general procedure C1 to give the title compound as a white solid (90mg, 26%).1H NMR(400MHz,CD3OD):δ=7.25-7.24(m,1H),7.16-7.14(m,2H),6.67(s,1H),4.13(s,3H),3.11-3.08(m,2H),1.61(s,6H),1.16(d,J=6.8Hz,12H)。LCMS(m/z):423.20(M+1)+;99.16%(210nm),97.19%(254nm)。HPLC:98.16%(210nm),97.09%(254nm)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -5- (2-hydroxypropan-2-yl) -1-methyl-1H-pyrazole-3-sulfonamide.

The title compound was obtained as a white solid (70mg, 15%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 5- (2-hydroxypropan-2-yl) -1-methyl-1H-pyrazole-3-sulfonamide in general method C1.1H NMR(400MHz,DMSO-d6)δ8.01(s,1H),6.92(s,1H),6.53(s,1H),5.51(s,1H),4.02(s,3H),2.79(t,J=7.4Hz,4H),2.62(t,J=7.4Hz,4H),1.95(p,J=7.4Hz,4H),1.50(s,6H)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -5- (2-hydroxypropan-2-yl) -1-phenyl-1H-pyrazole-3-sulfonamide

The title compound was obtained as a white solid (10mg, 2%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 5- (2-hydroxypropan-2-yl) -1-phenyl-1H-pyrazole-3-sulfonamide in general method C1. 1H NMR(400MHz,CD3OD):δ=7.54(s,5H),6.59(s,1H),6.91(s,1H),2.86(t,J=7.2Hz,4H),2.69(t,J=7.2Hz,4H),2.05-1.96(m,4H),1.44(s,6H)。LCMS(m/z):481.20(M-1)-;93.76%(210nm),93.24%(254nm)。HPLC:95.86%(210nm),93.93%(254nm)。

1-benzyl-N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -5- (2-hydroxypropan-2-yl) -1H-pyrazole-3-sulfonamide

The title compound was obtained as a white solid (40mg, 7%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 1-benzyl-5- (2-hydroxypropan-2-yl) -1H-pyrazole-3-sulfonamide in general method C1.1H NMR(400MHz,CD3OD):δ=7.20-7.14(m,5H),6.95(s 2H),6.73(s,1H),5.77(s,2H),2.86(t,J=7.2Hz,4H),2.68(t,J=7.6Hz,4H),2.01-1.94(m,4H),1.51(s,6H)。LCMS(m/z):494.7(M+1)+;98.74%(210nm),96.05%(254nm)。HPLC:95.11%(210nm),95.08%(254nm)。

N- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) -5- (2-hydroxypropan-2-yl) -1-methyl-1H-pyrazole-3-sulfonamide

Using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and 5- (2-hydroxypropan-2-yl) -1-methyl-1H-pyrazole-3-sulfonamide in general procedure C2 to give the title compound as a white solid (83mg, 17%).1H NMR(600MHz,DMSO-d6)δ=7.81(s,1H),7.10(s,2H),6.42(s,1H),5.45(s,1H),3.99(s,3H),3.03(hept,J=7.0Hz,2H),1.47(s,6H),1.05(d,J=1.8Hz,12H)。C20H28Cl1N4O4S1[M-H]-HRMS calculated of (a): 455.1525, found: 455.1515.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) -5- (2-hydroxypropan-2-yl) -1-phenyl-1H-pyrazole-3-sulfonamide

Using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and 5- (2-hydroxypropan-2-yl) -1-phenyl-1H-pyrazole-3-sulfonamide in general procedure C2 to give the title compound as a white solid (168mg, 31%). 1H NMR(600MHz,DMSO-d6)δ=7.87(s,1H),7.52(s,5H),7.10(s,2H),6.71(s,1H),5.42(s,1H),3.10–2.92(m,2H),1.31(s,6H),1.02(d,J=7.0Hz,12H)。C25H30Cl1N4O4S1[M-H]-HRMS calculated of (a): 517.1682, found: 517.1671.

n- ((4-chloro-2, 6-dimethylphenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide

A solution of 4-chloro-2, 6-dimethylaniline (50mg, 0.321mmol) in DCM (5mL) was treated with Et3N (50mg, 0.48mmol), cooled to 0 ℃ and phenyl chloroformate (60mg, 0.39mmol) was added dropwise at 0 ℃. The reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was washed with saturated NaHCO3The solution was diluted, extracted with DCM (2 × 20mL) and the combined organic extracts were washed with water, brine and dried (Na)2SO4) And concentrated in vacuo. The resulting residue was washed with n-pentane and dried in vacuo to give phenyl (4-chloro-2, 6-dimethylphenyl) carbamate (75mg, 85%) as a brown solid.1H NMR(300MHz,CDCl3):δ=7.46-7.37(m,4H),7.26-7.20(m,2H),7.12-7.11(m,2H),2.33(s,6H)。LCMS(m/z):275.9[M+H]+

1-isopropyl-1H-pyrazole-3-sulfonamide (150mg, 0.79mmol) was dissolved in anhydrous THF (5mL) and treated carefully with NaH (80mg, 1.98mmol) at 0 deg.C under nitrogen. The resulting mixture was stirred at room temperature for 30 minutes, then treated with a solution of phenyl (4-chloro-2, 6-dimethylphenyl) carbamate (240mg, 0.87mmol) in THF (3mL) at 0 ℃ under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred for 3 hours. After completion, the reaction mixture was taken up with saturated NH 4The Cl solution was diluted, extracted with EtOAc (2 × 30mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluent to give N- ((4-chloro-2, 6-dimethylphenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide (90mg, 31%) as a white solid.1H NMR(400MHz,DMSO-d6):δ=11.05(s,1H),7.99(d,J=2.8Hz,1H),7.93(s,1H),7.13(s,2H),6.73(d,J=2.4Hz,1H),4.64-4.57(m,1H),2.03(s,6H),1.43(d,J=6.8Hz,6H)。LCMS(m/z):370.95[M+H]+.;97.62%(210nm),97.48%(254nm)。HPLC:97.20%(210nm)。C15H18Cl1N4O3S1[M-H]-HRMS calculated of (a): 369.0794, found: 369.0785.

n- ((4-chloro-2, 6-dimethoxyphenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide

4-chloro-2, 6-A solution of dimethoxyaniline (200mg, 1.06mmol) in THF (8mL) was cooled to 0 deg.C and treated with NaH (62mg, 1.59mmol) under a nitrogen atmosphere. The resulting mixture was stirred for 15 minutes, after which phenyl chloroformate (330mg, 2.13mmol) was added dropwise at 0 ℃. The reaction mixture was then warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was diluted with EtOAc and filtered through a pad of celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 10% EtOAc-hexanes eluent to give phenyl (4-chloro-2, 6-dimethoxyphenyl) carbamate as a white solid (0.2g, 61%). 1H NMR(300MHz,CDCl3):δ=7.35-7.33(m,2H),7.20-7.19(m,3H),6.61(s,2H),3.83(s,6H)。

1-isopropyl-1H-pyrazole-3-sulfonamide (100mg, 0.53mmol) was dissolved in anhydrous THF (5mL) and treated carefully with NaH (52mg, 1.32mmol) at 0 deg.C under nitrogen. The resulting mixture was stirred at room temperature for 40 minutes, then treated with a solution of phenyl (4-chloro-2, 6-dimethoxyphenyl) carbamate (180mg, 0.58mmol) in THF (3mL) at 0 ℃ under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was taken up with saturated NH4The Cl solution was diluted, extracted with EtOAc (2 × 50mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The combined organic extracts were washed with water, brine and anhydrous Na2SO4Dried and concentrated in vacuo. Preparative HPLC on reverse phase [ column: gemini NX C18(21.2mm X150 mm particle size 5 μm)); flow rate: 20 mL/min; eluent: 10mM ammonium bicarbonate in water (A)&In MeCN (B); gradient: t/% B0/20, 2/20,8/70]The crude product was purified. The fractions were lyophilized to give N- ((4-chloro-2, 6-dimethoxyphenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide (20mg, 9%) as a white solid.1H NMR(400MHz,DMSO-d6):δ=7.90(d,J=2.0Hz,1H),7.28(s,1H),6.72(s,2H),6.63(d,J=2.0Hz,1H),4.60-4.54(m,1H),3.70(s,6H),1.43(d,J=6.8Hz,6H)。LCMS(m/z):403.0[M+H]+.;90.61%(210nm)。HPLC:91.63%(210nm)。C15H18Cl1N4O5S1[M-H]-HRMS calculated of (a): 401.0692, found: 401.0684.

N- ((4-chloro-2-methyl-6- (trifluoromethyl) phenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide

4-chloro-2-methyl-6- (trifluoromethyl) aniline (50mg, 0.24mmol) was dissolved in anhydrous THF (5mL) and Et at room temperature3N (30mg, 0.29 mmol). The solution was treated with triphosgene (30mg, 0.095mmol) and the resulting mixture was stirred at 60 ℃ for 4 hours. The reaction mixture was concentrated in vacuo. The resulting residue was stirred with n-pentane (20mL) for 10 minutes, filtered through a pad of celite and concentrated in vacuo to give the corresponding isocyanate as a white solid. In another 50mL round bottom flask, 1-isopropyl-1H-pyrazole-3-sulfonamide (40mg, 0.212mmol) was dissolved in anhydrous THF (5mL) and carefully treated with NaH (22mg, 0.529mmol) at 0 deg.C under nitrogen. It was stirred at room temperature for 30 minutes. The isocyanate solution was added to THF under nitrogen atmosphere. The resulting reaction mixture was stirred at room temperature for 2 hours. After completion of the reaction, the reaction mixture was washed with saturated NH4The Cl solution was diluted, extracted with EtOAc (2 × 25mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluent to give N- ((4-chloro-2-methyl-6- (trifluoromethyl) phenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide. This was triturated with diethyl ether and N-pentane to give N- ((4-chloro-2-methyl-6- (trifluoromethyl) phenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide (35mg, 35%) as a white solid. 1H NMR(400MHz,DMSO-d6):δ=11.05(s,1H),8.09(s,1H),7.96(s,1H),7.70(s,1H),7.61(s,1H),6.67(d,J=0.4Hz,1H),4.62(m,1H),2.05(s,3H),1.43(d,J=6.8Hz,6H)。19F NMR(400MHz,DMSO-d6):δ=-60.82.LCMS(m/z):425.00[M+H]+;94.05%(210nm)。HPLC:98.03%(210nm)。C15H15Cl1F3N4O3S1[M-H]-HRMS calculated of (a): 423.0511, found: 423.0513.

n- ((4-chloro-2-methoxy-6- (trifluoromethyl) phenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide

4-chloro-2-methoxy-6- (trifluoromethyl) aniline (50mg, 0.22mmol) was dissolved in anhydrous THF (2mL) and Et at room temperature3N (27mg, 0.27 mmol). The solution was treated with triphosgene (32mg, 0.11mmol) and the resulting mixture was stirred at 70 ℃ for 3 hours. The reaction mixture was concentrated in vacuo, and the resulting residue was stirred with 5% EtOAc-hexanes (20mL) for 10 minutes, filtered through celite and concentrated in vacuo to afford the desired isocyanate as a white solid. In another 50mL round-bottom flask, 1-isopropyl-1H-pyrazole-3-sulfonamide (42mg, 0.22mmol) was dissolved in anhydrous THF (5mL) and carefully treated with NaH (18mg, 0.44mmol) at 0 deg.C under nitrogen. The mixture was stirred at 0 ℃ for 20 minutes and treated with a solution of the above isocyanate in THF under a nitrogen atmosphere. The resulting reaction mixture was stirred at 0-10 ℃ for 2 hours. After completion, the reaction mixture was taken up with saturated NH4The Cl solution was diluted, extracted with EtOAc (2 × 50mL), and the combined organic extracts were washed with water, brine, and dried (Na) 2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 50% EtOAc-hexanes eluent to give N- ((4-chloro-2-methoxy-6- (trifluoromethyl) phenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide (10mg, 10%) as a white solid.1H NMR(400MHz,CDCl3):δ=7.81(s,1H),7.53(d,J=2.4Hz,1H),7.22(d,J=1.6Hz,1H),7.10(s,1H),6.82(d,J=2.0Hz,1H),4.64-4.57(m,1H),3.84(s,3H),1.54(d,J=6.8Hz,6H)。19F NMR(400MHz,CDCl3):δ=-61.55.LCMS(m/z):441.05[M+H]+.;94.58%(210nm)。HPLC:92.16%(210nm)。C15H15Cl1F3N4O4S1[M-H]-HRMS calculated of (a): 439.0460, found: 439.0478.

n- ((4-chloro-2, 6-diethylphenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide

A solution of 4-chloro-2, 6-dicyclopropylaniline (100mg, 0.546mmol) in THF (5mL) was cooled to 0 deg.C and treated with NaH (30mg, 0.66mmol) under a nitrogen atmosphere and stirred for 15 min. Phenyl chloroformate (130mg, 0.819mmol) was added dropwise to the above solution at 0 ℃. The reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the mixture was diluted with EtOAc, filtered through a pad of celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 10% EtOAc-hexanes to give phenyl (4-chloro-2, 6-diethylphenyl) carbamate as a white solid (0.15g, 91%).1H NMR(300MHz,CDCl3):δ=7.38-7.33(m,2H),7.23-7.18(m,3H),7.13(m,2H),6.27(s,1H),2.75-2.64(m,4H),1.28-1.22(m,6H)。

1-isopropyl-1H-pyrazole-3-sulfonamide (75mg, 0.40mmol) was dissolved in anhydrous THF (5mL) and treated carefully with NaH (40mg, 0.99mmol) at 0 deg.C under nitrogen. The resulting mixture was stirred at room temperature for 30 minutes, then treated with the resulting solution of phenyl (4-chloro-2, 6-diethylphenyl) carbamate (130mg, 0.44mmol) in THF (3mL) at 0 ℃ under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred for 4 hours. After completion, the reaction mixture was taken up with saturated NH 4The Cl solution was diluted, extracted with EtOAc (2 × 30mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 30-40% EtOAc-hexanes eluent, followed by trituration with diethyl ether and N-pentane to give N- ((4-chloro-2, 6-diethylphenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide (40mg, 24%) as a white solid.1H NMR(400MHz,DMSO-d6):δ=11.05(s,1H),7.99(d,J=2.4Hz,1H),7.89(s,1H),7.12(s,2H),6.72(d,J=2.4Hz,1H),4.62-4.59(m,1H),2.42(q,J=7.6Hz,4H),1.43(d,J=6.8Hz,6H),1.02(t,J=7.6Hz,6H)。LCMS(m/z):399.0[M+H]+,96.72%(210nm)。HPLC:97.13%(210nm)。C17H22Cl1N4O3S1[M-H]-HRMS calculated of (a): 397.1107, found: 397.1090.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide

The title compound was obtained as a white solid in general procedure C2 using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and 1-isopropyl-1H-pyrazole-3-sulfonamide (221mg, 49%).1H NMR(600MHz,DMSO-d6)δ=7.70(d,J=2.3Hz,1H),7.49(s,1H),7.00(s,2H),6.36(s,1H),4.62–4.29(m,1H),3.11(d,J=6.4Hz,2H),1.38(d,J=6.8Hz,6H),1.01(d,J=6.8Hz,12H)。13C NMR(151MHz,DMSO)δ=160.96,156.43,150.19,135.25,131.49,128.53,123.27,105.21,54.23,28.80,24.15,23.55.C19H26Cl1N4O3S1[M-H]-HRMS calculated of (a): 425.1420, found: 425.1409.

n- ((4-chloro-2, 6-dicyclopropylphenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide

A solution of 4-chloro-2, 6-dicyclopropylaniline (150mg, 0.724mmol) in THF (5mL) was cooled to 0 deg.C. NaH (35mg, 0.87mmol) was added portionwise to the above solution and stirred for 20 min. Phenyl chloroformate (170mg, 1.08mmol) was added dropwise to the above solution at 0 ℃. The reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the mixture was taken up in Et The OAc was diluted, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 15% EtOAc-hexanes eluent to give phenyl (4-chloro-2, 6-dicyclopropylphenyl) carbamate as a white solid (195mg, 83%).1H NMR(300MHz,CDCl3):δ=7.37-7.35(m,2H),7.21-7.19(m,3H),6.84-6.83(m,2H),2.08-2.04(m,2H),1.04-1.02(m,4H),0.69-0.68(m,4H)。LCMS(m/z):328.2[M+H]+

1-isopropyl-1H-pyrazole-3-sulfonamide (100mg, 0.53mmol) was dissolved in anhydrous THF (5mL) and treated carefully with NaH (53mg, 1.32mmol) at 0 deg.C under nitrogen. The resulting mixture was stirred at room temperature for 30 minutes, then treated with a solution of phenyl (4-chloro-2, 6-dicyclopropylphenyl) carbamate (190mg, 0.582mmol) in THF (3mL) at 0 ℃ under a nitrogen atmosphere. The reaction mixture was warmed to room temperature and stirred for 4 hours. After completion of the reaction, the reaction mixture was washed with saturated NH4The Cl solution was diluted, extracted with EtOAc (2 × 30mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluent to give N- ((4-chloro-2, 6-dicyclopropylphenyl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide. This material was triturated with diethyl ether and n-pentane to give a white solid (25mg, 11%). 1H NMR(400MHz,DMSO-d6):δ=11.05(s,1H),8.01-7.98(m,2H),6.74(s,3H),4.59-4.56(m,1H),1.77-1.76(m,2H),1.41(d,J=6.8Hz,6H),0.77-0.75(m,4H),0.56-0.55(m,4H)。LCMS(m/z):423.00[M+H]+.;93.58%(210nm)。HPLC:92.87%(210nm)。C19H22Cl1N4O3S1[M-H]-HRMS calculated of (a): 421.1107, found: 421.1107.

n- ((7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-yl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide

Will 7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-amine, 6(70mg, 0.33mmol) was dissolved in THF (5mL) and cooled to 0 ℃. NaH (20mg, 0.505mmol) was added to the above solution under nitrogen and stirred for 15 minutes, then phenyl chloroformate (100mg, 0.674mmol) was added dropwise at 0 ℃. The reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 10% EtOAc-hexanes eluent to give phenyl (7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-yl) carbamate (80mg, 73%) as a brown solid.1H NMR(300MHz,CDCl3):δ=7.39-7.37(m,3H),7.25-7.24(m,2H),6.85(s,1H),3.0-2.94(m,4H),2.12-2.10(m,2H),1.34(m,1H),0.96-0.95(m,2H),0.59-0.57(m,2H)。LCMS(m/z):328.30[M+H]+

1-isopropyl-1H-pyrazole-3-sulfonamide (41mg, 0.219mmol) was dissolved in anhydrous THF (3mL) and treated carefully with NaH (21mg, 0.549mmol) at 0 deg.C under nitrogen. The resulting mixture was stirred at room temperature for 30 minutes and treated with a solution of phenyl (7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-yl) carbamate (80mg, 0.244mmol) in THF (2mL) at 0 deg.C under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred for 4 hours. After completion, the reaction mixture was taken up with saturated NH 4The Cl solution was diluted, extracted with EtOAc (2 × 30mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. Preparative HPLC on reverse phase [ column: gemini NX C18(21.2mm X150 mm particle size 5 μm); flow rate: 20 mL/min; eluent: 10mM ammonium bicarbonate in water (A)&In MeCN (B); gradient: t/% B0/20, 2/30,8/70]The crude product was purified. The fractions were lyophilized to give N- ((7-chloro-5-cyclopropyl-2, 3-dihydro-1H-inden-4-yl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide (20mg, 20%) as a white solid.1H NMR(400MHz,DMSO-d6):δ=7.95(d,J=2.4Hz,1H),6.74(s,1H),6.69(d,J=2.4Hz,1H),4.60-4.55(m,1H),2.84(t,J=7.6Hz,2H),2.66(t,J=7.6Hz,2H),1.95-1.92(m,2H),1.79-1.76(m,1H),1.41(d,J=6.4Hz,6H),0.82-0.78(m,2H),0.54-0.50(m,2H)。LCMS(m/z):421.15[M-H]-;94.19%(210nm)。HPLC:95.46%(210nm)。C19H22Cl1N4O3S1[M-H]-HRMS calculated of (a): 421.1107, found: 421.1110.

5-chloro-3-cyclopropyl-2- (3- ((1-isopropyl-1H-pyrazol-3-yl) sulfonyl) ureido) -N, N-dimethylbenzamide

2-amino-5-chloro-3-cyclopropyl-N, N-dimethylbenzamide (200mg, 0.84mmol) was dissolved in anhydrous THF (5mL) and carefully treated with NaH (50mg, 1.26mmol) at 0 deg.C under nitrogen. The resulting reaction mixture was stirred for 15 minutes. Phenyl chloroformate (262mg, 1.68mmol) was then added dropwise at 0 ℃. The reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 15% EtOAc-hexanes eluent to give phenyl (4-chloro-2-cyclopropyl-6- (dimethylcarbamoyl) phenyl) carbamate (0.14, 47%) as a dark brown liquid. 1H NMR(300MHz,CDCl3):δ=7.39-7.34(m,2H),7.23-7.15(m,3H),7.08-7.02(m,2H),3.09(s,3H),2.96(s,3H),2.05-2.0(m,1H),1.05-1.02(m,2H),0.71-0.69(m,2H)。LCMS(m/z):358.60[M+H]+

1-isopropyl-1H-pyrazole-3-sulfonamide (57mg, 0.30mmol) was dissolved in anhydrous THF (5mL) and treated carefully with NaH (18mg, 0.451mmol) at 0 deg.C under nitrogen. The resulting mixture was stirred at room temperature for 45 minutes and then treated with a solution of phenyl (4-chloro-2-cyclopropyl-6- (dimethylcarbamoyl) phenyl) carbamate (120mg, 0.335mmol) in THF (3mL) at 0 deg.C under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred for 2 hours. After completion, the reaction mixture was taken up with saturated NH4The Cl solution was diluted, extracted with EtOAc (2 × 30mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. Preparative HPLC on reverse phase [ column: gemini NX C18110A AXIA (21.2mm × 150mm particle size 5 μm)); flow rate: 18 mL/min; eluent: 10mM ammonium bicarbonate in water (A)&In MeCN (B); gradient: t/% B0/20, 2/20,10/60]The crude product was purified. The fractions were lyophilized to give 5-chloro-3-cyclopropyl-2- (3- ((1-isopropyl-1H-pyrazol-3-yl) sulfonyl) ureido) -N, N-dimethylbenzamide as a white solid (10mg, 7%).1H NMR(400MHz,CD3OD):δ=7.77(s,1H),7.11(d,J=2.4Hz,1H),7.05(d,J=2.0Hz,1H),6.76(s,1H),4.62-4.53(m,1H),2.96(s,3H),2.84(s,3H),1.88-1.87(m,1H),1.50(d,J=6.8Hz,6H),0.90-0.88(m,2H),0.63-0.61(m,2H)。LCMS(m/z):454.0[M+H]+.;97.52%(210nm)。HPLC:92.05%(210nm)。C19H23Cl1N5O4S1[M-H]-HRMS calculated of (a): 452.1165, found: 452.1180.

n- ((4, 6-dimethylpyrimidin-2-yl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide

A solution of 4, 6-dimethylpyrimidin-2-amine (300mg, 2.43mmol) in THF (10mL) was cooled to 0 deg.C and treated with NaH (140mg, 3.64mmol) under a nitrogen atmosphere. The resulting mixture was stirred for 15 minutes, then treated with phenyl chloroformate (0.6mL, 4.87mmol) at 0 ℃. The reaction mixture was warmed to room temperature and stirred for 12 hours. Upon completion, the reaction mixture was diluted with EtOAc (30mL), filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 30% EtOAc-hexanes eluent to give phenyl (4, 6-dimethylpyrimidin-2-yl) carbamate as a white solid (250mg, 42%).1H NMR(300MHz,CDCl3):δ=8.14(s,1H),7.45-7.34(m,2H),7.25-7.18(m,3H),6.78(s,1H),2.46(s,6H)。LCMS(m/z):244.30[M+H]+

1-isopropyl-1H-pyrazole-3-sulfonamide (75mg, 0.396mmol) was dissolved in anhydrous THF (50mL) and treated carefully with NaH (40mg, 0.99mmol) at 0 deg.C under nitrogen. The resulting mixture was warmed to room temperature and stirred for 30 minutes. Will be reversedThe mixture was cooled to 0 ℃ and then treated with a solution of phenyl (4, 6-dimethylpyrimidin-2-yl) carbamate (100mg, 0.436mmol) in THF (5mL) at 0 ℃ under a nitrogen atmosphere. The reaction mixture was warmed to room temperature and stirred for 3 hours. After completion, the reaction mixture was taken up with saturated NH4The Cl solution was diluted, extracted with EtOAc (2 × 50mL), and the combined organic extracts were washed with water, brine, and dried (Na) 2SO4) And concentrated in vacuo. Preparative HPLC on reverse phase [ column: gemini NX-bridge (150mm X21.2 mm particle size 5 μm)); flow rate: 15 mL/min; eluent: 10mM ammonium bicarbonate in water (A)&In MeCN (B); gradient: t/% B0/10, 2/20,10/60]The crude product was purified. The fractions were lyophilized to give N- ((4, 6-dimethylpyrimidin-2-yl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide (10mg, 13%) as a white solid.1H NMR(400MHz,CDCl3):δ=13.0(s,1H),7.49-7.46(m,2H),7.02(d,J=2.4Hz,1H),6.74(s,1H),4.62-4.57(m,1H),2.45(s,6H),1.53(d,J=6.8Hz,6H)。LCMS(m/z):339.10[M+H]+.99.70%(210nm),100%(254nm)。HPLC:97.22%(210nm)。C13H17N6O3S1[M-H]-HRMS calculated of (a): 337.1088, found: 337.1099.

n- ((4, 6-di-tert-butylpyrimidin-2-yl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide

A solution of 4, 6-di-tert-butylpyrimidin-2-amine (0.15g, 0.72mmol) in THF (5mL) was cooled to 0 deg.C and treated with NaH (35mg, 0.86mmol) under a nitrogen atmosphere. The resulting mixture was stirred for 15 minutes, and phenyl chloroformate (0.17g, 1.08mmol) was added dropwise to the above solution at 0 ℃. The reaction mixture was warmed to room temperature and stirred for 12 hours. After completion, the reaction mixture was concentrated in vacuo and the resulting residue was diluted with ethyl acetate, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 15% EtOAc-hexanes eluent to give (4, 6-di-tert-butylpyrimidine as a white solid Pyridin-2-yl) carbamic acid phenyl ester (140mg, 59%).1H NMR(400MHz,CDCl3):δ=7.95(s,1H),7.65-7.61(m,2H),7.50-7.45(m,4H),1.32(s,18H)。LCMS(m/z):328.40[M+H]+

1-isopropyl-1H-pyrazole-3-sulfonamide (50mg, 0.264mmol) was dissolved in anhydrous THF (40mL) and treated carefully with NaH (27mg, 0.661mmol) at 0 deg.C under nitrogen. The resulting mixture was stirred for 30 minutes and then treated with a solution of phenyl (4, 6-di-tert-butylpyrimidin-2-yl) carbamate (95mg, 0.29mmol) in THF (5mL) at 0 deg.C under a nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred for 4 hours. After completion, the reaction mixture was taken up with saturated NH4The Cl solution was diluted, extracted with EtOAc (2 × 50mL), and the combined organic extracts were washed with water, brine, and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluent to give N- ((4, 6-di-tert-butylpyrimidin-2-yl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide (38mg, 25%) as a white solid.1H NMR(400MHz,DMSO-d6):δ=13.75(s,1H),10.71(s,1H),8.01(d,J=2.4Hz,1H),7.18(s,1H),6.81(d,J=2.4Hz,1H),4.61-4.54(m,1H),1.38(d,J=6.8Hz,6H),1.31(s,18H)。LCMS(m/z):423.50[M+H]+;99.88%(210nm)。HPLC:98.49%(210nm)。C19H29N6O3S1[M-H]-HRMS calculated of (a): 421.2027, found: 421.2008.

phenyl/bicyclic ring

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) benzenesulfonamide

The title compound was obtained as a white solid (50mg, 13%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and phenylsulfonamide in general method C1. 1H NMR(400MHz,CD3OD):δ=8.05(d,J=7.6Hz,2H),7.72(t,J=7.6Hz,1H),7.63(t,J=7.6Hz,2H),6.96(s,1H),2.84(t,J=7.2Hz,4H),2.59(t,J=7.2Hz,4H),2.00(quin,J=7.2Hz,4H)。

5- (dimethylamino) -N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) naphthalene-1-sulfonamide

A solution of 5- (dimethylamino) naphthalene-1-sulfonamide (20mg, 0.08mmol) in THF (5.0mL) was treated with DIPEA (17 μ L, 0.09mmol), stirred at ambient temperature for 15 minutes, and then a solution of 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method A2) (19mg, 0.09mmol) in THF (1.0mL) was added dropwise. The reaction mixture was stirred at ambient temperature overnight and then concentrated in vacuo. The crude product was purified by HPLC to give the title compound as a pale yellow solid (24mg, 66%).1H NMR(400MHz,CD3OD)δ=8.62(d,J=8.4Hz,1H),8.36(dd,J=9.5,8.1Hz,2H),7.67–7.56(m,2H),7.26(d,J=7.8Hz,1H),6.91(s,1H),2.91(s,6H),2.79(t,J=7.4Hz,4H),2.40(t,J=7.4Hz,4H),1.92(quin,J=7.4Hz,4H)。13C NMR(101MHz,CD3OD)δ=152.5,150.3,144.4,138.3,131.9,131.7,131.1,130.3,130.1,129.9,128.9,127.2,123.6,119.4,119.1,118.9,115.7,78.4,78.0,77.7,49.6,48.3,45.6,33.4,33.2,30.5,29.3,25.9,25.8;C25H27N3O3S[M+H]Hrms (esi) calculated value of (a): 450.1846, found: 450.1859.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -2, 3-dihydrobenzo [ b ] thiophene-6-sulfonamide 1, 1-dioxide

In general procedure C2, 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general procedure A2) and 2, 3-dihydrobenzo [ b ] are used]Thiophene-6-sulfonamide 1, 1-dioxide to give a target as a white solidTitle compound (33mg, 28%).1H NMR(600MHz,DMSO-d6):δ=8.17(bs,1H),8.15(s,1H),8.13(d,J=9Hz,1H),7.73(d,J=12Hz,1H),6.89(s,1H),3.68(t,J=9Hz,2H),3.43(t,J=6Hz,2H),2.75(t,J=6Hz,4H),2.55(t,J=6Hz,4H),1.93-1.88(m,4H)。13C NMR(150MHz,DMSO-d6):δ=151.6,143.3,143.0,142.7,139.6,137.6,137.5,132.2,128.9,120.1,117.9,50.9,32.9,30.6,25.6,25.4LCMS(m/z):447[M+H]+

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -2-methoxybenzenesulphonamide

The title compound was obtained as a white solid (30mg, 48%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 2-methoxybenzenesulfonamide in general method C6. 1H NMR(400MHz,CD3OD)δ7.96(d,J=7.9Hz,1H),7.62(t,J=8.3Hz,1H),7.14–7.05(m,2H),6.97(s,1H),4.00(s,3H),2.83(t,J=7.3Hz,4H),2.56(t,J=7.3Hz,4H),2.09–1.90(m,4H)。13C NMR(101MHz,CD3OD)δ155.9,143.2,136.6,134.7,129.5,127.2,126.6,125.9,119.5,118.0,111.6,111.2,55.3,31.9,29.3,24.5.C20H23N2O4S[M+H]Hrms (esi) calculated value of (a): 387.1373, found: 387.1378.

n- (1,2,3,5,6, 7-hexahydro-indacen-4-ylcarbamoyl) -3- (trifluoromethyl) benzenesulfonamide

Using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 3- (trifluoromethyl) benzenesulfonamide in general method C4 to give the title compound (0.015g, 12%) as an off-white viscous solid;1H NMR(400MHz,DMSO-d6):δ=11.01(bs,1H),8.34(s,1H),8.25-8.23(m,2H),8.11(d,J=7.6Hz,1H),7.90(t, J ═ 8.0Hz,1H),6.93(s,1H),2.77(t, J ═ 7.2Hz,4H),2.50(m,4H),1.90(quin, J ═ 7.6Hz, 4H). LCMS, purity 96.69%, M/z 425.1(M + H)+)。C20H19F3N2O3S[M+H]+HRMS (FAB)+) Calculated values: 425.1068, found: 425.1009.

n- (1,2,3,5,6, 7-hexahydro-indacen-4-ylcarbamoyl) -3-methoxybenzenesulphonamide

The title compound was obtained as an off-white solid (0.025g, 23%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure a 1) and 3-methoxybenzenesulfonamide in general procedure C4.1H NMR(400MHz,DMSO-d6) δ is 10.77(bs,1H),8.15(s,1H),7.56-7.45(m,3H),7.27(d, J is 8.0Hz,1H),6.93(s,1H),3.82(s,3H),2.77(t, J is 7.2Hz,4H),2.53(t, J is 7.6Hz,4H),1.92(quin, J is 7.2Hz, 4H). LCMS, purity: 95.02%, tr 3.77min, M/z 387.28(M + H)+)。C20H22N2O4S[M+H]+HRMS (FAB) +) Calculated values: 387.1300, found: 387.1301.

n- (1,2,3,5,6, 7-hexahydro-indacen-4-ylcarbamoyl) -3- (trifluoromethoxy) benzenesulfonamide

4-Isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure A1) and 3- (trifluoromethoxy) benzenesulfonamide were used in general procedure C4 to give the title compound as an off-white solid (0.045g, 43%).1H NMR(400MHz,DMSO-d6):δ=8.04(d,J=8.0Hz,1H),7.95(s,1H),7.72(t,J=8.4HZ,1H),7.62(d,J=8.4Hz,1H),6.95(s,1H),2.82(t,J=7.6Hz,4H),2.59(t,J=7.6Hz,4H),1.99(quin,J=7.6Hz,4H)。13C NMR(100MHz,DMSO-d6):δ=149.2,147.9,143.1,142.2,137.3,131.5,128.5,126.4,125.9,121.2,119.7,118.6,118.1,32.5,29.4,25.0.19F NMR(233.33MHz,DMSO-d6) -57.10(s, 3F). LCMS, purity 95.56%, M/z 441.20(M + H)+)。C20H19F3N2O4S[M+H]+HRMS (FAB)+) Calculated values: 441.1018, found: 441.1015.

3- (difluoromethoxy) -N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) benzenesulfonamide

4-Isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure A2) and 3- (difluoromethoxy) benzenesulfonamide were used in general method C5 to give the title compound (0.056g, 50%) as a white solid.1H NMR (600MHz, acetonitrile-d)3)δ=7.85(d,J=8.0Hz,1H),7.75(t,J=2.1Hz,1H),7.60(t,J=8.0Hz,1H),7.53(s,1H),7.43(dd,J=8.0,2.1Hz,1H),6.95(s,1H),2.81(t,J=7.5Hz,4H),2.55(t,J=7.5Hz,4H),1.95(quin,J=7.5Hz,4H)。

N1- (1,2,3,5,6, 7-hexahydro-indacen-4-ylcarbamoyl) benzene-1, 3-disulfonamide

The title compound was obtained as a white solid (0.080g, 12%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and benzene-1, 3-disulfonamide in general method C4.1H NMR(400MHz,DMSO-d6):δ=11.02(bs,1H),8.36(s,1H),8.24(s,1H),8.15(d,J=7.8Hz,1H),8.11(d,J=8.0Hz,1H),7.84(t,J=8.0Hz,1H),7.63(s,2H),6.93(s,1H),2.77(t,J=7.2Hz,4H),2.54(t,J=7.6Hz,4H),1.92(quin,J=7.2Hz,4H)。13C NMR(100MHz,DMSO-d6) 98.6 purity of 149.0,144.9,143.1,140.9,137.3,130.4,130.2,128.5,124.4,118.1,32.4,30.0,25.0 LCMS 3%,m/z 436.03(M+H+)。C19H21N3O5S2[M+H]+HRMS (FAB)+) Calculated values: 436.0923, found: 436.0919.

N1- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -N3,N3-dimethylbenzene-1, 3-disulfonamides

In general procedure C1, 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general procedure A1) and N are used1,N1-dimethyl-1, 3-disulfonamide to give the title compound as a white solid (0.019g, 5%).1H NMR(400MHz,CD3OD)δ8.41(t,J=1.4Hz,1H),8.32(dt,J=7.9,1.4Hz,1H),8.08(dt,J=7.9,1.4Hz,1H),7.87(t,J=7.9Hz,1H),6.95(s,1H),2.84(t,J=7.4Hz,4H),2.73(s,6H),2.61(t,J=7.4Hz,4H),2.00(p,J=7.4Hz,4H)。

3- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) benzoic acid

Methyl 3- (N- (1,2,3,5,6, 7-hexahydro-s-indacen-4-ylcarbamoyl) sulfamoyl) benzoate (0.25g, 0.603mmol) was dissolved in tetrahydrofuran: methanol: water (9mL, 1:1:1) and the mixture was cooled to 0 ℃. Lithium hydroxide monohydrate (0.75g, 1.81mmol, 3 equiv.) was added and the mixture was stirred at ambient temperature for 3 hours. After completion, the reaction mixture was poured into cooling water and extracted with ethyl acetate. The combined organic extracts were washed with brine and dried (Na)2SO4) And concentrated in vacuo. The product was purified by reverse phase preparative HPLC to give the title compound as a white solid (0.017g, 3%).1H NMR(400MHz,DMSO-d6):δ=13.26(bs,1H),8.43(s,1H),8.13-8.08(m,2H),7.99(bs,1H) 7.67(t, J ═ 8.0Hz,1H),6.87(s,1H),6.52(s,1H),2.75(t, J ═ 7.2Hz,4H),2.55(t, J ═ 7.6Hz,4H),1.89(quin, J ═ 7.6Hz, 4H). LCMS, purity: 96%, M/z 400.98(M + H) +)。C20H20N2O5S[M+H]+HRMS (FAB)+) Calculated values: 401.1093, found: 401.4514.

3- (N- (1,2,3,5,6, 7-hexahydro-s-indacen-4-ylcarbamoyl) sulfamoyl) benzamide

3- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) benzoic acid (0.06g, 0.074mmol) was dissolved in anhydrous N, N-dimethylformamide (4mL) and the solution was cooled to 0 ℃. Diisopropylethylamine (3.0 eq) and HATU (2.0 eq) were added and the mixture was stirred at 0 ℃ for 15 min. Ammonium chloride (3.0 equivalents) was added and the mixture was stirred at ambient temperature for 5 hours. After completion, the reaction mixture was poured into brine (20mL) and extracted with ethyl acetate (2X 10 mL). The combined organic extracts were washed with brine (10mL) and dried (Na)2SO4) And concentrated in vacuo. The crude residue was purified by reverse phase preparative HPLC to give the title compound as a white solid (0.011g, 37%).1H NMR(400MHz,DMSO-d6) δ is 8.23(d, J is 9.2Hz,2H),8.02(s,1H),7.89(d, J is 7.6Hz,1H),7.84(d, J is 7.6Hz,1H),7.42(t, J is 8.0Hz,1H),7.38(s,1H),7.33(s, 1H). 6.74(s,1H),2.73(t, J ═ 6.8Hz,4H),2.62(t, J ═ 6.8Hz,4H),1.87(quin, J ═ 7.6Hz, 4H). LCMS, purity: 93%, M/z 400.05(M + H)+)。C20H21N3O4S[M+H]+HRMS (FAB)+) Calculated values: 400.1253, found: 400.1378.

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -3- (2-hydroxypropan-2-yl) benzenesulfonamide

4-Isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method A1) was added directly to methyl 3-sulfamoylbenzoate (0.447g, 2.07mmol, 1.20 equiv.) at ambient temperature and the mixture was stirred overnight. The reaction mixture was poured into cooling water, and extracted with ethyl acetate. The combined organic extracts were washed with brine and dried (Na)2SO4) And concentrated in vacuo. The resulting residue was purified by silica gel column chromatography using a 0-10% gradient of methanol in dichloromethane to give methyl 3- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) benzoate as a light brown solid (0.36g, 50%).

Methyl 3- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) benzoate (0.06g, 0.144mmol) was dissolved in anhydrous THF and the solution was cooled to 0 ℃. Methyl magnesium bromide (3M solution in diethyl ether, 0.14mL, 0.42mmol, 3.0 equiv.) was added and the mixture was stirred at ambient temperature for 4 hours. After completion, a saturated aqueous ammonium chloride solution was added to the reaction mixture, and extracted with ethyl acetate. The combined organic extracts were washed with brine and dried (Na) 2SO4) And concentrated in vacuo. The crude residue was purified by reverse phase preparative HPLC to give the title compound as an off white solid (0.015g, 25%).1H NMR(400MHz,DMSO-d6) δ is 8.16(s,1H),7.91(s,1H),7.62(d, J is 7.2Hz,1H),7.59-7.48(m,2H),7.32(t, J is 7.6Hz,1H),6.78(s,1H),5.10(s,1H),2.74(t, J is 7.2Hz,4H),2.60(t, J is 6.8Hz,4H),1.88(quin, J is 7.6Hz,4H),1.42(s, 6H). LCMS, purity: 91%, M/z 415.05(M + H)+)。

3-azido-N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) benzenesulfonamide

4-Isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general procedure A2) and 3-azidobenzenesulfonamide are used in general procedure C6 to give an off-white colorTitle compound as a solid (70mg, 50%).1H NMR(600MHz,DMSO-d6)δ=8.22(s,1H),7.72(m,J=5.2Hz,H),7.65(t,J=8.0Hz,1H),7.59(s,1H),7.46–7.42(m,1H),6.93(s,1H),2.77(t,J=7.4Hz,4H),2.53(t,J=7.4Hz,4H),1.92(m,4H)。13C NMR(101MHz,CD3OD)δ151.2,144.9,144.6,142.9,142.5,138.9,131.5,131.4,128.6,124.9,124.6,124.6,119.8,119.2,118.9,111.9,33.7,33.6,31.1,29.7,26.3.C19H20N5O3S[M+H]Hrms (esi) calculated value of (a): 398.1281, found: 398.1272.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -3- (4-phenyl-1H-1, 2, 3-triazol-1-yl) benzenesulfonamide

The title compound was obtained as a light yellow solid (10mg, 49%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure a 2) and 3- (4-phenyl-1H-1, 2, 3-triazol-1-yl) benzenesulfonamide in general procedure C6.1H NMR(400MHz,CD3OD)δ=8.89(s,1H),8.55(s,1H),8.21(d,J=8.2Hz,1H),8.13(d,J=7.8Hz,1H),7.92(d,J=7.6Hz,2H),7.79(t,J=8.0Hz,1H),7.68(s,1H),7.48(t,J=7.6Hz,2H),7.39(t,J=7.4Hz,1H),6.92(s,1H),2.82(t,J=7.4Hz,4H),2.70–2.63(m,4H),1.98(m,4H)。13C NMR(151MHz,CD3OD)δ=148.8,143.9,143.6,137.7,137.2,137.0,130.6,130.3,129.7,129.6,128.8,128.5,127.6,127.4,126.7,125.6,124.5,124.0,119.3,118.7,110.8,32.7,32.6,30.2,28.7,25.3.C27H26N5O3S[M+H]Hrms (esi) calculated value of (a): 500.1751, found: 500.1735.

N- (3- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) phenyl) pent-4-ynylamide

On-lineThe title compound was obtained as a white solid (116mg, 61%) by using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and N- (3-sulfamoylphenyl) pent-4-ynamide in method C6.1H NMR(400MHz,CD3OD)δ=8.18(s,1H),7.81(d,J=8.3Hz,1H),7.68(d,J=8.3Hz,1H),7.43(dd,J=8.3,7.8Hz,1H),6.87(s,1H),2.79(t,J=7.2Hz,4H),2.67–2.60(m,4H),2.60–2.48(m,4H),2.28–2.22(m,1H),2.04–1.89(m,4H)。13C NMR(101MHz,CD3OD)δ=170.9,143.3,143.0,138.8,137.7,128.7,128.3,126.4,122.8,122.0,117.9,117.7,82.0,68.9,35.4,32.4,29.9,25.1,13.9.C24H26N3O4S[M+H]Hrms (esi) calculated value of (a): 452.1639, found: 452.1658.

3- (1- (3-aminopropyl) -1H-1,2, 3-triazol-4-yl) -N- (3- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) phenyl) propionamide

N- (3- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) phenyl) pent-4-ynylamide and 3-azidopropan-1-amine were used in general procedure F to give the title compound as a white solid (6mg, 43%).1H NMR(600MHz,CD3OD)δ=7.85(s,1H),7.55(t,J=3.8Hz,2H),7.50(d,J=8.0Hz,2H),7.29(t,J=7.9Hz,1H),6.78(s,1H),4.26(t,J=6.4Hz,2H),3.00(t,J=6.6Hz,2H),2.71(t,J=7.3Hz,4H),2.64–2.50(m,8H),1.94-2.02(m,2H),1.92–1.83(m,4H)。13C NMR(151MHz,CD3OD)δ=173.0,147.4,146.8,144.7,144.6,139.5,139.2,131.6,130.0,129.8,124.2,123.9,123.2,119.5,118.6,48.3,37.7,34.0,31.6,26.7,26.6,22.9.C27H34N7O4S[M+H]Hrms (esi) calculated value of (a): 552.2387, found: 552.2368.

n- (3- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) phenyl) -3- (1- (3- ((7-nitrobenzo [ c ] [1,2,5] oxadiazol-4-yl) amino) propyl) -1H-1,2, 3-triazol-4-yl) propionamide

Synthesis of N- (2-azidopropyl) -7-nitrobenzo [ c ] via the method contained in Chun Li, Etienne Henry, Naresh Kumar Mani, Jie Tang, Jean-Claude Brochon, Eric Deprez and Juan Xie Eur.J.org.chem.2010, 2395-2405 ][1,2,5]Oxadiazol-4-amines. To 4-chloro-7-nitrobenzo [ c][1,2,5]To a solution of oxadiazole (300mg, 1.5mmol) in THF (10mL) was added 3-azidopropylamine (160mg, 1.65mmol) and Cs2CO3(480mg, 1.5 mmol). The reaction mixture was stirred at 50 ℃ for 4 hours. The reaction mixture was partitioned between EtOAc (50mL) and concentrated in vacuo. The residue was purified by silica gel column chromatography using 30% EtOAc-petroleum ether eluent to give N- (2-azidopropyl) -7-nitrobenzo [ c][1,2,5]Oxadiazol-4-amine (240mg, 76%).1H NMR(400MHz,CDCl3):δ=8.50(d,J=8.8Hz,1H),6.57(s,1H,NH),6.23(d,J=8.8Hz,1H),3.66(q,J=6.8Hz,2H),3.59(J=6.0Hz,2H),2.00-2.16(m,2H)。13C NMR(101MHz,CDCl3)δ144.2,144.0,143.8,136.7,123.7,98.8,49.1,41.6,27.6.C9H10N7O3Hrms (esi) calculated value of (a): 264.0840, respectively; measured value: 264.0711.

n- (3- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) phenyl) pent-4-ynylamide (10mg, 0.022mmol) and N- (2-azidopropyl) -7-nitrobenzo [ c-azidopropyl ] -amide][1,2,5]Oxadiazol-4-amine (7.0mg, 0.026mmol), 10 mol% THPTA, 5 mol% CuSO410 mol% sodium ascorbate in DMSO (500uL) was stirred at room temperature for 12 hours. The reaction mixture was subjected to reverse phase (Reveleria flash column chromatography, 4g, 18mL/min, mobile phase; 10mmol NH)4CO3Aqueous solution, MeCN) and freeze-dried to give the product as a white solid (7.0mg, 44%).1H NMR(600MHz,CD3OD)δ=8.46(d,J=8.7Hz,1H),8.18(s,1H),7.79(d,J=8.4Hz,1H),7.67(d,J=7.9Hz,1H),7.61(s,1H),6.94(s,1H),6.15(d,J=9.0Hz,1H),4.46(t,J=6.7Hz,2H),3.09(t,J=7.0Hz,2H),2.82(t,J=7.4Hz,4H),2.77(t,J=7.0Hz,2H),2.70–2.56(m,6H),2.37–2.26(m,2H),1.99(q,J=7.3Hz,4H)。13C NMR(151MHz,CD3OD)δ=172.9,147.9,145.4,140.5,139.0,138.4,130.6,129.1,128.0,125.6,124.2,123.6,120.3,119.6,112.4,70.6,48.9,37.2,34.3,34.2,31.7,30.2,26.8,22.3;C33H34N10O7S[M-H]Hrms (esi) calculated value of (a): 713.2260, found: 713.2290.

N- (3- (4- (3- ((3- (N- ((1,2,3,5,6, 7-hexahydro-S-indacen-4-yl) carbamoyl) sulfamoyl) phenyl) amino) -3-oxopropyl) -1H-1,2, 3-triazol-1-yl) propyl) -5- ((3aS,4S,6aR) -2-oxohexahydro-1H-thieno [3,4-d ] imidazol-4-yl) pentanamide

To a solution of biotin (0.4g, 1.63mmol) and 3-azidopropylamine (0.2g, 1.96mmol) in anhydrous DMF (10.0ml) was added HBTU (0.93g, 2.45mmol) followed by DIPEA (428uL, 2.45 mmol). The reaction mixture was stirred at room temperature for 12 hours. The reaction was monitored by LCMS and, after completion, it was diluted with EtOAc (50mL) with H2O (25mL), brine (25 mL). Separating the organic layer; drying (MgSO)4) And evaporated to give the crude product. The crude product was purified by silica gel column chromatography using 50% EtOAc-hexanes eluent to isolate N- (3-azidopropyl) -5- ((3aS,4S,6aR) -2-oxohexahydro-1H-thieno [3, 4-d) aS a white solid]Imidazol-4-yl) pentanamide (0.13g, 24%). H NMR (400MHz, CD)3OD)δ=4.52(dd,J=7.9,5.0Hz,1H),4.32(dd,J=7.9,4.5Hz,1H),3.36(t,J=6.7Hz,2H),3.28(d,J=6.8Hz,2H),3.21–3.14(m,1H),2.93(dd,J=12.8,5.0Hz,1H),2.75(d,J=12.8Hz,1H),2.20(t,J=7.3Hz,2H),1.78(q,J=6.8Hz,2H),1.74–1.57(m,4H),1.45(q,J=7.5Hz,2H)。13C NMR(101MHz,CD3OD)δ=173.5,163.4,61.0,59.3,54.7,48.2,39.4,35.8,34.8,27.7,27.5,27.2,24.6。

Coupling N- (3- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) phenyl) pent-4-ynylamide (1.0)mmol) and N- (3-azidopropyl) -5- ((3aS,4S,6aR) -2-oxohexahydro-1H-thieno [3, 4-d)]Imidazol-4-yl) pentanamide (2.0mmol), 10 mol% THPTA, 5 mol% CuSO 410 mol% sodium ascorbate in DMSO was stirred at room temperature for 12 hours. The reaction mixture was purified by reverse phase column chromatography to give the title compound as a white solid (8.0mg, 31%);1H NMR(600MHz,CD3OD)δ=8.26(s,1H),7.83–7.68(m,3H),7.50–7.43(m,1H),6.92(s,1H),4.48(dd,J=8.0,4.8Hz,1H),4.41–4.22(m,3H),3.18(dd,J=6.9,3.5Hz,1H),3.14(td,J=6.7,1.7Hz,2H),3.12–3.06(m,2H),2.90(dd,J=12.8,4.9Hz,1H),2.81(t,J=7.7Hz,4H),2.77(d,J=7.1Hz,1H),2.71(s,1H),2.62(t,J=7.3Hz,4H),2.19(td,J=7.4,1.7Hz,2H),2.05–2.01(m,2H),2.00–1.95(m,4H),1.76–1.57(m,4H),1.43(q,J=7.6,7.1Hz,2H)。13C NMR(151MHz,CD3OD)δ=174.8,174.8,171.6,171.5,164.5,146.2,143.6,139.1,137.7,129.1,129.1,128.7,128.1,126.5,123.7,122.9,122.4,122.2,120.9,118.4,118.4,118.3,118.2,118.2,117.2,110.5,69.0,61.9,60.2,55.6,39.8,36.1,36.0,35.8,35.4,35.4,32.6,32.6,30.0,29.7,29.7,28.6,28.3,28.0,25.3,25.2,25.2,20.9.C37H48N9O6S2[M+H]hrms (esi) calculated value of (a): 778.3163, found: 778.3145.

n- ((1- (3- (N- ((1,2,3,5,6, 7-hexahydro-S-indacen-4-yl) carbamoyl) sulfamoyl) phenyl) -1H-1,2, 3-triazol-4-yl) methyl) -5- ((3aS,4S,6aR) -2-oxohexahydro-1H-thieno [3,4-d ] imidazol-4-yl) pentanamide

Using 5- ((3aS,4S,6aR) -2-oxohexahydro-1H-thieno [3,4-d ]]Imidazol-4-yl) -N- (prop-2-yn-1-yl) pentanamide (1.0mmol) and 3-azidobenzenesulfonamide (2.0mmol)10 mol% THPTA, 5 mol% CuSO4Synthesis of 5- ((3aS,4S,6aR) -2-oxohexahydro-1H-thieno [3, 4-d) -by stirring 10 mol% NaAsc in DMSO at room temperature for 12 hours]Imidazol-4-yl) -N- ((1- (3-sulfamoylphenyl) -1H-1,2, 3-triazol-4-yl) methyl) pentanamide. Product observed in LCMSIs performed. After completion of the reaction, the reaction mixture was subjected to HPLC purification (Reveleria flash column chromatography, 4g, 18mL/min, mobile phase; 10mmol NH)4CO3Aqueous solution, MeCN) to isolate 5- ((3aS,4S,6aR) -2-oxohexahydro-1H-thieno [3, 4-d) aS a white solid ]Imidazol-4-yl) -N- ((1- (3-sulfamoylphenyl) -1H-1,2, 3-triazol-4-yl) methyl) pentanamide (24mg, 47%) which was used as is.

To 5- ((3aS,4S,6aR) -2-oxohexahydro-1H-thieno [3,4-d ] under a nitrogen atmosphere]To a solution of imidazol-4-yl) -N- ((1- (3-sulfamoylphenyl) -1H-1,2, 3-triazol-4-yl) methyl) pentanamide (15mg, 0.031mmol) in THF (5.0mL) was added DIPEA (605. mu.L, 0.037 mmol). The mixture was stirred at room temperature for 15 minutes. A solution of 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general method A2) (705mg, 0.037mmol) in THF was added dropwise. The reaction mixture was stirred at room temperature overnight, then the solvent was removed in vacuo to give crude compound, which was purified by reverse phase column chromatography using 10mM (NH)4)2CO3Aqueous solution and MeCN mobile phase this was purified to isolate the title compound as a white solid (5.2mg, 24%).1H NMR(600MHz,CD3OD)δ=8.53(d,J=2.4Hz,1H),8.50(d,J=7.5Hz,1H),8.22–8.11(m,2H),7.86–7.78(m,1H),6.99(s,1H),4.62(s,2H),4.57–4.49(m,1H),4.38–4.31(m,1H),3.27–3.20(m,1H),3.00–2.84(m,4H),2.78–2.70(m,4H),2.36(t,J=7.2Hz,2H),2.06(q,J=7.4Hz,4H),1.83–1.73(m,3H),1.71–1.63(m,1H),1.54–1.43(m,3H)。13C NMR(151MHz,CD3OD)δ=174.6,164.5,146.1,145.4,143.6,137.7,137.2,130.5,125.9,123.4,121.1,117.8,110.6,61.8,60.2,55.5,47.7,47.6,39.8,3.2,34.3,32.6,30.14,28.2,27.9,25.3,25.2.C32H39N8O5S2[M+H]Hrms (esi) calculated value of (a): 679.2479, found: 679.2456.

n- (quinolin-6-ylcarbamoyl) -3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide

Phenyl chloroformate (1 equivalent) was added to a solution of quinolin-6-amine (0.1g, 0.69mmol) in THF (50mL) and triethylamine (1.5 equivalents) to 0 deg.C. The solution was diluted with water, extracted with ethyl acetate (× 2), washed with water, brine and dried (Na) 2SO4) And concentrated in vacuo. The crude product was triturated with pentane to give quinolin-6-yl carbamic acid phenyl ester as an off-white solid, which was used directly in the next reaction step.

3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide (0.185g, 0.69mmol) in THF (30mL) was treated portionwise with sodium hydride (3 equiv.) at 0 deg.C and the suspension was stirred for 30 min (until effervescence ceased). The crude quinolin-6-yl phenyl carbamate was dissolved in THF (20mL) and then added slowly to the reaction and stirring was continued at ambient temperature until completion, typically for 2 hours. The reaction mixture is saturated with NH4Aqueous Cl was quenched, extracted with ethyl acetate (. times.2), washed with water (100mL), brine (100mL), and dried (Na)2SO4) And concentrated in vacuo. The crude product was triturated with diethyl ether then pentane to give the title compound N- (quinolin-6-ylcarbamoyl) -3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide as a white solid (10mg, 3%).1H NMR(400MHz,DMSO-d6)δ=8.88(s,1H),8.59(d,J=3.5Hz,1H),8.11(s,1H),8.04(d,J=8.3Hz,1H),7.96(d,J=7.9Hz,1H),7.74(d,J=9.1Hz,1H),7.70–7.60(m,2H),7.56(t,J=7.8Hz,1H),7.34(dd,J=8.3,4.2Hz,1H),7.30(d,J=8.0Hz,1H)。19F NMR(376MHz,DMSO-d6)δ-64.49。

N- (quinolin-5-ylcarbamoyl) -3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide

Synthesis of N- (quinolin-5-ylcarbamoyl) -3- (3- (trifluoromethyl) amino) benzenesulfonamide with a modification of the procedure used to prepare N- (quinolin-6-ylcarbamoyl) -3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide but using quinolin-5-amine in place of quinolin-6-amine Yl) -3H-bisaziridin-3-yl) benzenesulfonamide. The title compound was obtained as an off-white solid (10mg, 3%).1H NMR(400MHz,DMSO-d6)δ=8.79(d,J=4.1Hz,1H),8.61(s,1H),8.57(d,J=8.7Hz,1H),7.95(d,J=7.8Hz,1H),7.88(d,J=7.2Hz,1H),7.70(s,1H),7.59–7.50(m,2H),7.40(dd,J=8.7,4.1Hz,1H),7.29(d,J=7.9Hz,1H)。19F NMR(376MHz,DMSO-d6)δ-64.51。

N- ((6-methoxyquinolin-8-yl) carbamoyl) -3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide

N- ((6-Methoxyquinolin-8-yl) carbamoyl) -3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide was synthesized using a modification of the procedure used to prepare N- (quinolin-6-ylcarbamoyl) -3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide but using 6-methoxyquinolin-8-amine instead of quinolin-6-amine. The title compound was obtained as an off-white solid (35mg, 20%).1H NMR(400MHz,CD3OD)δ8.64(dd,J=4.2,1.6Hz,1H),8.18–8.02(m,3H),7.88(s,1H),7.60(t,J=7.9Hz,1H),7.50–7.36(m,2H),6.79(d,J=2.6Hz,1H),3.88(s,3H)。19F NMR(376MHz,CD3OD)δ-67.04。

N- (quinolin-8-ylcarbamoyl) -3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide

N- (quinolin-8-ylcarbamoyl) -3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide was synthesized with a modification of the procedure used to prepare N- (quinolin-6-ylcarbamoyl) -3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide, but using quinolin-8-amine in place of quinolin-6-amine. The title compound was obtained as a white solid (20mg, 16%).1H NMR(400MHz,CD3OD)δ=8.82(dd,J=4.3,1.6Hz,1H),8.35(dd,J=7.4,1.8Hz,1H),8.21(dd,J=8.3,1.7Hz,1H),8.12(d,J=7.9Hz,1H),7.88(s,1H),7.59(t,J=7.9Hz,1H),7.52–7.38(m,4H)。

N- ((2,3,6, 7-tetrahydrobenzo [1,2-b:4,5-b' ] difuran-4-yl) carbamoyl) -3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide

Use of 4-isocyanato-2, 3,6, 7-tetrahydrobenzo [1,2-b:4,5-b 'in general method C1']Difuran (prepared using general method a 1) and 3- (3- (trifluoromethyl) -3H-bisaziridin-3-yl) benzenesulfonamide to give the title compound as a white solid (0.01g, 2%).1H NMR(400MHz,CD3OD)δ=8.06(dt,J=7.9,1.3Hz,1H),7.80(s,1H),7.57(t,J=7.9Hz,1H),7.44(d,J=7.9Hz,1H),6.39(s,1H),4.49(t,J=8.6Hz,2H),4.42(t,J=8.6Hz,2H),3.09(t,J=8.4Hz,2H),3.02(t,J=8.6Hz,2H)。19F NMR(376MHz,CD3OD)δ-67.06。

4-chloro-N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) benzenesulfonamide

The title compound was obtained as a white solid (48mg, 43%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 4-chlorobenzenesulfonamide in general method C2.1H NMR(400MHz,DMSO-d6):δ=8.13(s,1H),7.93(d,J=8.0Hz,2H),7.68(d,J=12Hz,2H),6.92(s,1H),2.77(t,J=8.0Hz,4H),2.54(t,J=8.0Hz,4H),1.95-1.88(m,4H);13C NMR(100MHz,DMSO-d6) δ 150.1,143.4,139.9,138.1,137.6,129.6,129.4,129.2,32.8,30.5, 25.9; LCMS purity:>95%;LCMS(m/z):391[M+H]+;C19H19ClN2O3S[M+H]+HRMS calculated of (a): 391.0878, found: 391.0895.

n- (1,2,3,5,6, 7-hexahydro-s-indacen-4-ylcarbamoyl) -4-methylbenzenesulfonamide

4-Isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general method A1) and 4-methylbenzenesulfonamide were used in general method C4 to give the title compound as a white solid (0.045g, 27%).1H NMR(400MHz,DMSO-d6):δ=10.70(br.s,1H),8.08(s,1H),7.82(d,J=8Hz,2H),7.41(d,J=8.0Hz,2H),6.92(s,1H),2.79-2.68(m,4H),2.58-2.50(m,4H),2.39(s,3H),1.97-1.87(m,4H)。13C NMR(100MHz,DMSO-d6) Delta 149.0,143.6,143.0,137.1,129.4,128.6,127.3,117.9,32.4,30.0,25.0,21.0.LCMS, purity 95.08%, M/z 371.07(M + H)+)。C20H22N2O3S[M+H]+HRMS (FAB)+) Calculated values: 371.1351, found: 371.1419.

5-chloro-N- (4- (N- (1,2,3,5,6, 7-hexahydro-s-indacen-4-ylcarbamoyl) sulfamoyl) phenethyl) -2-methoxybenzamide

Using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 5-chloro-2-methoxy-N- (4-sulfamoylphenethyl) benzamide in general method C4 to give the title compound as a white solid (45mg, 10%).1H NMR(400MHz,DMSO-d6):δ=10.73(s,1H),8.27(t,J=5.2Hz,1H),8.09(s,1H),7.89(d,J=8.4Hz,2H),7.65(d,J=2.4Hz,1H),7.50(d,J=8.4Hz,2H),7.49(d,J=2.4Hz,1H),7.13(d,J=9.2Hz,1H),6.92(s,1H),3.78(s,3H),3.54(q,J=6.4Hz,2H),2.94(t,J=6.8Hz,2H),2.75(t,J=7.2Hz,4H),2.50(m,4H),1.89(quin,J=7.6Hz,4H)。13C NMR(100MHz,DMSO-d6):δ=163.6,155.7,145.3,143.6,143.0,142.4,142.1,139.6,137.1,131.5,129.5,129.2,127.4,125.7,124.8,124.3,1179,114.1,108.3,56.2,34.7,32.6,32.4,30.0,28.9,24.9 LCMS, purity 90.06%, tr 3.38min, M/z 566.37 (M-H)+)。C29H30ClN3O5S[M+H]+HRMS (FAB)+) Calculated values: 568.1595, found: 568.1589.

n- (4- (N- (1,2,3,5,6, 7-hexahydro-indacen-4-ylcarbamoyl) sulfamoyl) phenethyl) -5-methylpyrazine-2-carboxamide

The title compound was obtained as an off-white solid (0.02g, 4%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure a 1) and 5-methyl-N- (4-sulfamoylphenethyl) pyrazine-2-carboxamide in general procedure C4.1H NMR(400MHz,DMSO-d6) δ is 10.71(s,1H),9.02(s,1H),8.96(t, J is 6Hz,1H),8.59(s,1H),8.07(s,1H),7.85(d, J is 8.4Hz,2H),7.47(d, J is 8.0Hz,2H),6.92(s,1H),3.57(q, J is 6.8Hz,2H),2.97(t, J is 7.4Hz,2H),2.82-2.73(m,4H),2.53(s,3H),2.57-2.50(m,4H),1.97-1.84(m, 4H). LCMS, purity 88.15%, M/z 520.28(M + H) +)。C27H29N5O4S[M+H]+HRMS (FAB)+) Calculated values: 520.1940, found: 520.1977.

3- (4- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) phenyl) -N- (prop-2-yn-1-yl) propionamide

The title compound was obtained as a white solid (120mg, 68%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and N- (prop-2-yn-1-yl) -3- (4-sulfamoylphenyl) propionamide in general method C6.1H NMR(400MHz,CD3OD)δ=7.91(d,J=7.8Hz,2H),7.39(d,J=7.8Hz,2H),6.98(s,1H),3.95(d,J=2.9Hz,2H),3.03(t,J=7.7Hz,2H),2.85(t,J=7.4Hz,4H),2.62(t,J=6.9Hz,4H),2.55–2.46(m,2H),2.25(t,J=2.6Hz,1H),2.02(m,4H)。13C NMR(101MHz,CD3OD)δ=172.0,147.2,144.1,143.8,137.5,129.0,128.8,128.1,127.4,126.5,118.9,79.2,71.0,36.8,32.8,32.8,31.2,30.7,28.8,28.7,25.4,25.3.C25H28N3O4S[M+H]Hrms (esi) calculated value of (a): 466.1795, found: 466.1794.

n- (4- (N- (1,2,3,5,6, 7-hexahydro-indacen-4-ylcarbamoyl) sulfamoyl) phenethyl) -2- (methyl (7-nitrobenzo [ c ] [1,2,5] oxadiazol-4-yl) amino) acetamide

In general procedure C4, 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general procedure A1) and 2- (methyl (7-nitrobenzo [ C ] is used][1,2,5]Oxadiazol-4-yl) amino) -N- (4-sulfamoylphenethyl) acetamide to give the title compound as an orange solid (0.003g, 1%).1H NMR(400MHz,DMSO-d6) δ is 10.74(s,1H),8.51(d, J is 8.8Hz,1H),8.31(t, J is 7.6Hz,1H),8.09-7.96(m,1H),7.82(d, J is 8.4Hz,2H),7.41(d, J is 7.6Hz,2H),6.89(s,1H),6.42-6.32(m,1H),4.74(bs,2H),3.44-3.30(m,5H),2.80(t, J is 7.6Hz,2H),2.73-2.69(m,4H),2.61-2.50(m,4H),1.92-1.88(m, 4H). LCMS, purity 92.20%, M/z 632.35 (M-H) +)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2- ((7-nitrobenzo [ c ] [1,2,5] oxadiazol-4-yl) amino) ethyl) benzenesulfonamide

In general procedure C4, 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure A1) and 4- (2- ((7-nitrobenzo [ C ] are used][1,2,5]Oxadiazol-4-yl) amino) ethyl) benzenesulfonamide to give the title compound as a yellow solid (0.047g, 15%).1H NMR(400MHz,DMSO-d6):δ=10.69(bs,1H),9.55(s,1H),8.50(d,J=8.8Hz,1H),8.09(s,1H),7.87(d,J=8.0Hz,2H),7.56(d,J=8.0Hz,2H),6.92(s,1H),6.50(d,J=8.8Hz,1H),3.76(bs,2H),3.11(t,J=6.8Hz,2H),2.76(t,J=7.6Hz,4H),2.53(t,J=6.8Hz,4H),1.90(quin,J=7.6Hz,4H)。13C NMR(100MHz,DMSO-d6) δ 149.1,144.8,144.3,142.4,138.3,137.8,137.1,129.3,128.6,127.3,125.7,121.0,117.9,108.3,99.5,44.1,33.2,32.5,30.1,28.9,25.0.LCMS, purity: 96.50%, tr 2.29min, M/z 563.20(M + H)+)。C27H26N6O6S[M+H]+HRMS (FAB)+) Calculated values: 563.1635, and 563.1641.

2- (7- (dimethylamino) -2-oxo-2H-chroman-4-yl) -N- (4- (N- (1,2,3,5,6, 7-hexahydro-s-indacen-4-ylcarbamoyl) sulfamoyl) phenethyl) acetamide

The title compound was obtained as a light yellow solid in general procedure C4 using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure a 1) and 2- (7- (dimethylamino) -2-oxo-2H-chroman-4-yl) -N- (4-sulfamoylphenethyl) acetamide.1H NMR(400MHz,DMSO-d6) δ is 9.53(s,1H),8.29(t, J is 4.8Hz,1H),8.22(s,1H),7.68(d, J is 8.0Hz,2H),7.46(d, J is 8.8Hz,1H),8.42(s,1H),7.16(d, J is 8.0Hz,2H),6.74-6.70(m,2H),6.54(d J is 2.4Hz,1H),5.99(s,1H),3.56-3.52(m,2H),3.48(t, J is 6.0Hz,2H),3.31-3.24(m,2H),2.76-2.70(m,4H),3.02(s,6H),2.63(t, 7.88H), 7.88 (t, 4H), 7.88H, 7H). LCMS, purity 92.26%, m/z629.40 (MH) +)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) benzo [ d ] [1,3] dioxole-5-sulfonamide

In general useMethod C2 used 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general method A2) and benzo [ d][1,3]Dioxol-5-sulfonamide to give the title compound as a white solid (28mg, 27%).1H NMR(400MHz,DMSO-d6) δ 8.04(br.s.,1H),7.47(d, J ═ 8.0Hz,1H),7.38(s,1H),7.09(d, J ═ 4.0Hz,1H),6.91(s,1H),6.16(s,2H)2.77(t, J ═ 8.0Hz,4H),2.56(t, J ═ 8.0Hz,4H),1.96-1.89(m, 4H); LCMS purity:>95%;LCMS(m/z):401[M+H]+;C20H20N2O5S[M+H]+HRMS calculated of (a): 401.1166, found: 401.1182.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -6, 7-dihydro-5H-pyrrolo [1,2-a ] imidazole-2-sulfonamide

1,2,3,5,6, 7-hexahydro-s-indacen-4-amine (70mg, 0.40mmol) was dissolved in anhydrous THF (5mL) and treated with Et at room temperature3N (49mg, 0.49 mmol). The solution was treated with triphosgene (48mg, 0.161mmol) and the resulting mixture was stirred at 70 ℃ for 2 h. The reaction mixture was concentrated in vacuo. The resulting residue was stirred with 5% EtOAc-hexanes (20mL) for 10 min, filtered through a pad of celite and concentrated in vacuo to give the corresponding isocyanate as a white solid. In a separate flask, 6, 7-dihydro-5H-pyrrolo [1,2-a ] was placed ]Imidazole-2-sulfonamide (115mg, 0.61mmol) was dissolved in anhydrous THF (5mL) and carefully treated with NaH (25mg, 0.61mmol) under nitrogen at 0 deg.C and stirred for 20 min. The above isocyanate in THF was added to the reaction mixture under nitrogen atmosphere. The reaction mixture was warmed to room temperature, stirred for 4 hours, and then concentrated in vacuo. The resulting residue was diluted with 10mM ammonium bicarbonate in water (20mL), acetonitrile (20mL), ethyl acetate (10mL) and the solid formed was removed by filtration and washed with diethyl ether to give N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -6, 7-dihydro-5H-pyrrolo [1,2-a ] as a white solid]Imidazole-2-sulfonamide (50mg, 32%).1H NMR(400MHz,CD3OD):δ=7.29(s,1H),6.85(s,1H),4.23(t,J=7.2Hz,1H),2.86-2.79(m,6H),2.72(t,J=7.2Hz,4H),2.65-2.60(m,2H),2.02-1.95(m,4H)。LCMS(m/z):387.10[M+H]+;95.53%(210nm)。HPLC:94.43%(210nm)。C19H21N4O3S1[M-H]-HRMS calculated of (a): 385.1340, found: 385.1331.

4-acetyl-N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) benzenesulfonamide

The title compound was obtained as a white solid (31mg, 16%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 4-acetylbenzenesulfonamide in general method C2.1H NMR(600MHz,DMSO-d6)δ=11.03(bs,1H)8.08(d,J=8.5Hz,2H),7.99(d,J=8.5Hz,2H),7.03(bs,1H),6.87(s,1H),2.75(t,J=7.4Hz,4H),2.62(s,3H),2.56(t,J=7.4Hz,4H),1.90(p,J=7.4Hz,4H)。C21H21N2O4S1[M-H]-HRMS calculated of (a): 397.1128, found: 397.1225.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4-nitrobenzenesulfonamide

The title compound was obtained as a light yellow solid (148mg, 60%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 4-nitrobenzenesulfonamide in general method C2.1H NMR(600MHz,DMSO-d6)δ=10.00(bs,1H),8.21(d,J=9.0Hz,,2H),7.97(d,J=9.0Hz,2H),7.45(s,1H),6.75(s,1H),2.73(t,J=7.4Hz,4H),2.61(t,J=7.4Hz,4H),1.87(p,J=7.4Hz,4H)。C19H18N3O5S1[M-H]-HRMS calculated of (a): 400.0973, found: 400.0979.

4-amino-N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) benzenesulfonamide

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4-nitrobenzenesulfonamide) dissolved in a solution of ethyl acetate/DMF (4:1,25mL/mmol) was stirred with catalytic amount of Pd/C (0.1 mol%) for 1 hour at room temperature under hydrogen atmosphere to give the title compound as a white solid (16mg, 43%).1H NMR(600MHz,DMSO-d6)δ=7.95(s,1H),7.54(d,J=8.8Hz,2H),6.91(s,1H),6.59(d,J=8.8Hz,2H),6.05(s,2H),2.77(t,J=7.4Hz,4H),2.55(t,J=7.4Hz,4H),1.93(q,J=7.4Hz,4H)。C19H20N3O3S1[M-H]-HRMS calculated of (a): 370.1231, found: 370.1225.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -2, 3-dihydro-1H-indene-5-sulphonamide

The title compound was obtained as a white solid (48mg, 12%) using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 2, 3-dihydro-1H-indene-5-sulfonamide in general method C2.1H NMR(600MHz,DMSO-d6)δ=10.68(bs,1H),8.02(s,1H),7.75(d,J=1.7Hz,1H),7.68(dd,J=7.9,1.7Hz,1H),7.41(d,J=7.9Hz,1H),6.90(s,1H),2.91(t,J=7.5Hz,4H),2.76(t,J=7.4Hz,4H),2.53(t,J=7.4Hz,4H),2.05(p,J=7.5Hz,2H),1.91(p,J=7.4Hz,4H)。C22H23N2O3S1[M-H]-HRMS calculated of (a): 395.1435, found: 395.1430.

n- ((4-chlorophenyl) carbamoyl) -2, 3-dihydro-1H-indene-5-sulfonamide

1-chloro-4-isocyanatobenzene (prepared using general method B1) and 2, 3-dihydro-1H-indene-5-sulfonamide are used in general method C2 to give the title compound as a white solid (60mg, 32%).1H NMR(600MHz,DMSO-d6)δ10.90(bs,1H),8.90(s,1H),7.74(d,J=1.8Hz,1H)7.68(dd,J=7.9,1.7Hz,1H),7.41–7.35(m,3H),7.26(dt,2H),2.91(m,4H),2.05(p,J=7.5Hz,2H)。C16H14Cl1N2O3S1[M-H]-HRMS calculated of (a): 349.0419, found: HRMS 349.0418.

N- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) quinoline-8-sulfonamide

5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure A2) and quinoline-8-sulfonamide were used in general procedure C3 to give the title compound as a white solid (75mg, 71%).1H NMR(600MHz,CD3OD):δ=9.13(dd,J=4.2,1.6Hz,1H),8.57-8.49(m,2H),8.26(d,J=8.2Hz,1H),7.77-7.67(m,2H),6.99(s,2H),2.65-2.60,(m,2H),0.85(d,12H);13C NMR(150MHz,CD3OD) δ 151.2,149.0,143.3,136.8,136.7,133.8,133.5,132.3,129.4,129.1,125.3,123.0,122.1,109.1,28.3, 22.5; LCMS purity:>95%;LCMS(m/z):446[M+H]+;C22H25Cl1N3O3S1[M+H]+HRMS calculated of (a): 446.1300, found: 446.1314.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) isoquinoline-5-sulfonamide

The title compound was obtained as a white solid (70mg, 67%) using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and isoquinoline-5-sulfonamide (prepared using general procedure E3) in general procedure C3.1H NMR(600MHz,CD3OD):δ=9.41(s,1H),8.82(s,1H),8.59(d,J=7.3Hz,2H),8.35(d,J=8.2Hz,1H),7.79(t,J=7.6Hz,1H),6.96(s,2H),2.74-2.70(m,2H),0.96(s,6H),0.85(d,12H);13C NMR(150MHz,CD3OD) δ 156.3,152.5,149.1,143.8,137.2,133.9,133.1,132.6,131.5,130.4,126.3,124.8,122.8,122.1,28.3, 22.4; LCMS purity: >95%;LCMS(m/z):446[M+H]+;C22H25Cl1N3O3S1[M+H]+HRMS calculated of (a): 446.1300, found: 446.1319.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) quinoline-5-sulfonamide

The title compound was obtained as a white solid (31mg, 60%) using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and quinoline-5-sulfonamide (prepared using general procedure E3) in general procedure C3.1H NMR(600MHz,CD3OD):δ=9.53(d,J=8.9Hz,1H),8.94(d,J=3.8Hz,1H),8.35(dd,J=7.3,1.2Hz,1H),8.15(d,J=8.5Hz,1H),7.79(dd,J=8.5,7.3Hz,1H),7.69(dd,J=8.7,4.3Hz,1H),2.81-2.76(m,2H),0.85(d,12H);13C NMR(150MHz,CD3OD) δ 161.4,151.3,150.7,149.1,142.2,137.4,134.0,133.0,132.8,129.9,129.4,126.0,124.1,122.9,29.6, 24.0; LCMS purity:>95%;LCMS(m/z):446[M+H]+;C22H25Cl1N3O3S1[M+H]+HRMS calculated of (a): 446.1300, found: 446.1317.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) quinoline-8-sulfonamide

Using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 4H-quinoline-8-sulfonamide in general method C3 to give the title compound as a white solid (60mg, 51%);1H NMR(600MHz,DMSO-d6)δ=9.11(d,J=2.7Hz,1H),8.56(d,J=8.3Hz,1H),8.40(d,J=7.4Hz,1H),8.32(d,J=8.2Hz,1H),8.17(s,1H),7.76(t,J=7.7Hz,1H),7.73(dd,J=8.4,4.2Hz,1H),6.82(s,1H),2.67(t,J=7.4Hz,4H),2.26(t,J=7.4Hz,4H),1.79(p,J=7.5Hz,4H);13C NMR(150MHz,DMSO-d6):δ=151.8,151.7,143.3,143.2,137.5,137.1,134.5,133.4,132.8,129.9,126.0,122.8,118.0,108.7,32.7,30.2,25.3.LCMS(m/z):408[M+H]+.C22H22N3O3S1[M+H]+HRMS calculated of (a): 408.1376, found: 408.1371.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) quinoline-3-sulfonamide

The title compound was obtained as a white solid (30mg, 57%) using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and quinoline-3-sulfonamide (prepared using general procedure E3) in general procedure C3. 1H NMR(600MHz,DMSO-d6):δ1H NMR(600MHz,DMSO-d6)δ=9.24(s,1H),8.92(s,1H),8.20(d,J=8.3Hz,1H),8.12(d,J=8.5Hz,1H),7.98(s,1H),7.93(t,J=7.7Hz,1H),7.74(t,J=7.6Hz,1H),7.02(s,2H),2.81-2.78(m,2H),0.84(d,12H);13C NMR(150MHz,DMSO-d6) δ 153.7,149.3,148.7,147.6,141.5,136.8,132.6,132.4,131.5,129.9,129.2,128.4,126.4,123.3,28.5, 23.5; LCMS purity:>95%;LCMS(m/z):446[M+H]+;C22H25Cl1N3O3S1[M+H]+HRMS calculated of (a): 446.1300,measured value: 446.1315.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) quinoxaline-5-sulfonamide

Using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and quinoxaline-5-sulfonamide (prepared using general procedure E3) in general procedure C3 to give the title compound as a white solid (39mg, 75%);1H NMR(600MHz,DMSO-d6)δ=9.18(d,J=3.7Hz,2H),8.46(d,J=7.3Hz,1H),8.38(dd,J=8.1,2.7Hz,1H),8.04-7.95(m,1H),7.83(s,1H),6.99(s,2H),2.55-2.49(m,2H),0.74(d,12H);13C NMR(150MHz,DMSO-d6) 149.2,147.1,146.8,146.2,142.7,140.5,138.5,138.2,134.2,133.4,132.6,129.6,123.4,28.4, 22.7; LCMS purity:>95%;LCMS(m/z):447[M+H]+;C21H24Cl1N4O3S1[M+H]+HRMS calculated of (a): 447.1252, found: 447.1266.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) naphthalene-2-sulfonamide

The title compound was obtained as a white solid in general procedure C3 using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and naphthalene-2-sulfonamide (prepared using general procedure E3).1H NMR(600MHz,CD3OD):δ=8.55(s,1H),8.05-7.92(m,4H),7.64-7.58(m 2H),6.99(s,2H),2.94-2.89(m,2H),0.94(bs,12H);13C NMR(150MHz,CD3OD) δ 159.9,150.6,142.2,135.9,134.0,133.7,132.6,130.2,129.5,129.1,128.8,128.6,128.1,124.3,124.2,29.7, 24.0; LCMS purity:>95%;LCMS(m/z):445[M+H]+;C23H26Cl1N2O3S1[M+H]+HRMS calculated of (a): 445.1347, found: 445.1349.

N- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) -6-methoxynaphthalene-2-sulfonamide

Using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and 6-methoxynaphthalene-2-sulfonamide (prepared using general procedure E3) in general procedure C3 to give the title compound as a white solid (39mg, 70%);1H NMR(600MHz,DMSO-d6)δ=8.23(s,1H),7.86(d,J=9.0Hz,1H),7.78(dd,J=9.1,6.4Hz,2H),7.49(s,1H),7.35(d,J=2.6Hz,1H),7.20(dd,J=8.9,2.6Hz,1H),6.96(s,2H),3.08-2.98(m,2H),0.93(bs,12H);13C NMR(150MHz,DMSO-d6) δ 158.0,149.6,135.2,134.4,131.0,130.5,128.1,127.5,127.3,126.5,124.7,124.6,122.7,119.4,106.2,55.7,28.3, 23.4; LCMS purity:>95%;LCMS(m/z):475[M+H]+;C24H28Cl1N2O4S1[M+H]+HRMS calculated of (a): 475.1453, found: 475.1474.

6-chloro-N- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) naphthalene-2-sulfonamide

Using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and 6-chloronaphthalene-2-sulfonamide (prepared using general procedure E3) in general procedure C3 to give the title compound as a white solid (34mg, 61%);1H NMR(600MHz,DMSO-d6)δ=8.30(s,1H),8.06(d,J=2.2Hz,1H),8.01(d,J=8.8Hz,1H),7.93-7.85(m,2H),7.55(dd,J=8.7,2.2Hz,1H),7.40(s,1H),6.95(s,2H),3.09-2.97(m,2H),0.92(bs,12H);13C NMR(150MHz,DMSO-d6):δ=160.2,149.6,145.8,134.8,134.2,131.7,131.1,130.8,130.7,127.2,126.8,126.6,125.8,125.7,122.7,28.3, 23.6; LCMS purity:>95%;LCMS(m/z):479[M+H]+;C23H25Cl2N2O3S1[M+H]+HRMS calculated of (a): 479.0957, found: 479.0937.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) -5,6,7, 8-tetrahydronaphthalene-2-sulfonamide

Using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and 5,6,7, 8-tetrahydronaphthalene-2-sulfonamide (prepared using general procedure E3) in general procedure C1 to give the title compound as a white solid (8mg, 38%); 1H NMR(600MHz,DMSO-d6)δ=7.85(s,1H),7.58(s,1H),7.57(d,J=8.0Hz,1H),7.22(d,J=7.2Hz,1H),7.08(s,2H),2.85-2.81(m,2H),2.78-2.74(m,4H),1.74(t,J=3.3Hz,4H),0.98(bs,12H);13C NMR(150MHz,DMSO-d6) δ 149.3,137.7,137.5,132.4,129.8,129.5,129.7,126.3,124.3,123.4,122.9,29.3,29.2,28.5,23.4,22.8, 22.7; LCMS purity:>95%;LCMS(m/z):449[M+H]+;C23H30Cl1N2O3S1[M+H]+HRMS calculated of (a): 449.1660, found: 449.1664.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) thieno [3,2-b ] pyridine-6-sulfonamide

In general procedure C3, 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure A2) and thieno [3,2-b ]]Pyridine-6-sulfonamide to give the title compound as a white solid (35mg, 66%);1H NMR(600MHz,DMSO-d6)δ=9.07(d,J=2.0Hz,1H),9.05(s,1H),8.45(d,J=5.5Hz,1H),8.01(s,1H),7.70(d,J=5.5Hz,1H),7.05(s,2H),2.79-2.75(m,2H),0.87(d,12H);13C NMR(150MHz,DMSO-d6) δ 158.2,152.7,149.3,145.8,138.2,132.7,132.3,132.2,131.6,131.1,124.7,123.4,28.5, 22.9; LCMS purity:>95%;LCMS(m/z):452[M+H]+;C20H23Cl1N3O3S2[M+H]+HRMS calculated of (a): 452.0864, found: 452.0884.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) -3-ethylisoxazolo [5,4-b ] pyridine-5-sulfonamide

In general procedure C3, 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure A2) and thieno [3,2-b ]]Pyridine-6-sulfonamide to give the title compound as a white solid (38mg, 64%);1H NMR(600MHz,DMSO-d6)δ=9.11(s,1H),8.98(s,1H),8.14(s,1H),7.08(s,2H),3.09(q,J=7.5Hz,2H),2.82-2.77(m,2H),1.34(t,J=7.5Hz,3H),1.02-0.90(d,12H)。13C NMR(150MHz,DMSO-d6) δ 170.2,161.9,150.5,149.3,134.3,134.2,132.8,131.0,123.5,112.7,109.9,28.5,23.0,19.2, 11.9; LCMS purity:>95%;LCMS(m/z):465[M+H]+;C21H26Cl1N4O4S1[M+H]+HRMS calculated of (a): 465.1358, found: 465.1354.

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) benzofuran-2-sulfonamide

Using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and benzofuran-2-sulfonamide in general method C3 to give the title compound as a white solid (60mg, 52%);1H NMR(400MHz,DMSO-d6):δ=8.00(bs,1H),7.77(d,J=8Hz,1H),7.69(d,J=8Hz,1H),7.51(s,1H),7.49(d,J=8Hz,1H),7.36(t,J=8Hz,1H),7.08(bs,1H),6.87(s,1H),2.75(t,J=8Hz,4H),2.59(t,J=8Hz,4H),1.92-1.85(m,4H)。13C NMR(150MHz,DMSO-d6):δ=154.9,143.2,137.6,130.3,127.5,126.7,124.4,123.3,117.7,112.3,110.0,109.4,107.4,32.9,30.6,25.5.LCMS(m/z):397[M+H]+.C21H21N2O4S1[M+H]+HRMS calculated of (a): 397.1217, found: 397.1215.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) benzofuran-2-sulfonamide

The title compound was obtained as a white solid in general procedure C3 using 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared using general procedure a 2) and benzofuran-2-sulfonamide (prepared using general procedure E3).1H NMR(600MHz,DMSO-d6)δ=7.85(s,1H),7.73(d,J=7.8Hz,1H),7.66(d,J=8.4Hz,1H),7.47(s,1H),7.34(t,J=7.5Hz,1H),7.17(s,1H),7.04(s,2H),2.99-2.95(m,2H),0.94(bs,12H);13C NMR(150MHz,DMSO-d6) δ 154.9,149.5,132.6,132.3,132.0,127.2,126.9,126.8,123.3,123.2,112.1,112.0,109.8,28.5, 23.3; LCMS purity:>95%;LCMS(m/z):435[M+H]+;C21H24Cl1N2O4S1[M+H]+HRMS calculated of (a): 435.1140, found: 435.1140.

n- ((4-chloro-2, 6-diisopropylphenyl) carbamoyl) benzo [ b ] thiophene-2-sulfonamide

In general procedure C3, 5-chloro-2-isocyanato-1, 3-diisopropylbenzene (prepared by general procedure A2) and benzo [ b ] are used]Thiophene-2-sulfonamide (prepared using general procedure E3) to give the title compound as a white solid (38mg,72%);1H NMR(600MHz,DMSO-d6)δ8.03(d,J=7.9Hz,1H),7.93(d,J=8.0Hz,2H),7.80(s,1H),7.46(dt,J=15.4,7.0Hz,2H),7.04(s,2H),3.05-2.83(m,2H),0.94(bs,12H);13C NMR(150MHz,DMSO-d6) Delta 155.3,149.5,141.1,138.0,132.4,132.0,126.8,125.7,125.4,123.1,123.0,122.9,109.7,28.5, 23.3; LCMS purity:>95%;LCMS(m/z):451[M+H]+;C21H24Cl1N2O3S2[M+H]+HRMS calculated of (a): 451.0911, found: 451.0900.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2- (7-methoxy-4, 4-dimethyl-1, 3-dioxo-3, 4-dihydroisoquinolin-2 (1H) -yl) ethyl) benzenesulfonamide

In general procedure C2, 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure a 2) and 4- (2- (7-methoxy-4, 4-dimethyl-1, 3-dioxo-3, 4-dihydroisoquinolin-2 (1H) -yl) ethyl) benzenesulfonamide were used to give the title compound as a white solid (85mg, 52%).1H NMR(600MHz,DMSO-d6)δ=10.72(bs,1H),7.91(s,1H),7.80(d,J=8.1Hz,2H),7.58(d,J=8.7Hz,1H),7.53(d,J=2.9Hz,1H),7.39(d,J=7.9Hz,2H),7.29(dd,J=8.7,2.9Hz,1H),6.88(s,1H),4.13(t,J=7.5Hz,2H),3.83(s,3H),2.93(t,J=7.5Hz,2H),2.76(t,J=7.4Hz,4H),2.55(t,J=7.4Hz,4H),1.90(p,J=7.4Hz,4H),1.42(s,6H)。C33H34N3O6S1[M-H]-HRMS calculated of (a): 600.2174, found: 600.2183.

n- (4- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) phenethyl) -5-methylisoxazole-3-carboxamide

In general useMethod C2 used 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 5-methyl-N- (4-sulfamoylphenethyl) isoxazole-3-carboxamide to give the title compound as a white solid (14mg, 62%).1H NMR(600MHz,DMSO-d6)δ8.78(t,J=5.8Hz,1H),7.80(s,1H),7.74(d,J=8.3Hz,2H),7.41(d,J=8.3Hz,2H),6.85(s,1H),6.50(q,J=1.0Hz,1H),3.49(m,2H),2.91(t,J=7.0Hz,2H),2.75(t,J=7.4Hz,4H),2.56(t,J=7.4Hz,4H),2.45(d,J=0.9Hz,3H),1.89(p,J=7.4Hz,4H)。C26H27N4O5S1[M-H]-HRMS calculated of (a): 507.1708, found: 507.1709.

3-ethyl-N- (4- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) phenethyl) -4-methyl-2-oxo-2, 5-dihydro-1H-pyrrole-1-carboxamide

The title compound was obtained as a white solid in general procedure C2 using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general procedure a 2) and 3-ethyl-4-methyl-2-oxo-N- (4-sulfamoylphenethyl) -2, 5-dihydro-1H-pyrrole-1-carboxamide.1H NMR(600MHz,DMSO-d6)δ=10.78(bs,1H),8.38(t,J=7.6Hz,1H),7.97(s,1H),7.82(d,J=8.2Hz,2H),7.42(d,J=8.2Hz,2H),6.88(s,1H),4.16(s,2H),3.48(q,J=6.7Hz,2H),2.88(t,J=7.2Hz,2H),2.75(t,J=7.4Hz,4H),2.53(t,J=7.4Hz,4H),2.18(q,J=7.5Hz,2H),2.01(s,3H),1.90(p,J=7.4Hz,4H),0.97(t,J=7.5Hz,3H)。C29H33N4O5S1[M-H]-HRMS calculated of (a): 549.2177, found: 549.2169.

5-chloro-N- (4- (N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) sulfamoyl) phenethyl) -2-methoxybenzamide

Using 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 2) and 5-chloro-2-methoxy-N- (4-sulfamoylphenethyl) benzamide in general method C2 to give the title compound as a white solid (325mg, 70%).1H NMR(600MHz,DMSO-d6)δ=10.83(bs,1H),8.27(t,J=6.0Hz,1H),7.96(s,1H),7.84(d,J=8.0Hz,2H),7.66(d,J=2.8Hz,1H),7.50(dd,J=8.9Hz,2.8Hz,1H,7.44(t,J=8.0Hz,2H),7.13(d,J=8.9Hz,1H),6.87(s,1H),3.78(s,3H),3.53(q,J=6.6Hz,2H),2.91(t,J=7.2Hz,2H),2.74(t,J=7.4Hz,4H),2.53(t,J=7.4Hz,4H),1.88(p,J=7.3Hz,4H)。C29H29Cl1N3O5S1[M-H]-HRMS calculated of (a): 566.1522, found: 566.1543.

13C NMR(100MHz,DMSO-d6) 163.6,155.7,145.3,143.6,143.0,142.4,142.1,139.6,137.1,131.5,129.5,129.2,127.4,125.7,124.8,124.3,117.9,114.1,108.3%, 56.2,34.7,32.6,32.4,30.0,28.9 and 24.9 LCMS, purity 90.06%, tr 3.38min, M/z 566.37 (M-H)+)。C29H30ClN3O5S[M+H]+HRMS (FAB)+) Calculated values: 568.1595, found: 568.1589.

pyridine compound

N- (1,2,3,5,6, 7-hexahydro-s-indacen-4-ylcarbamoyl) -4- (2-hydroxypropan-2-yl) pyridine-2-sulfonamide

To a solution of 1,2,3,5,6, 7-hexahydro-indacen-4-amine (0.20g, 1.15mmol) in anhydrous THF (5mL) at 0 ℃ was added triethylamine (0.35g, 3.47mmol, 3.0 equiv), followed by triphosgene (0.265g, 0.86mmol, 0.5 equiv) and the mixture was stirred at ambient temperature for 3 hours. The mixture was cooled to 0 ℃, methyl 2-sulfamoylisonicotinate (0.27g, 1.27mmol, 1.1 equiv.) was added and stirring was continued at ambient temperature overnight. After completion, the reaction was mixedThe mixture was poured into brine and extracted with ethyl acetate. The combined organic extracts were washed with brine and dried (Na)2SO4) And concentrated in vacuo. The crude product was purified by silica gel column chromatography using a gradient of 20-50% EtOAc-hexanes eluent to give methyl 2- (N- (1,2,3,5,6, 7-hexahydro-s-indacen-4-ylcarbamoyl) sulfamoyl) isonicotinate as a light brown solid (0.31g, 65%).

Methyl 2- (N- (1,2,3,5,6, 7-hexahydro-s-indacen-4-ylcarbamoyl) sulfamoyl) isonicotinite (0.30g, 0.72mmol) was dissolved in anhydrous THF (8mL) and the solution was cooled to 0 ℃. Methyl magnesium bromide (3M solution in diethyl ether, 0.96mL, 2.88mmol, 4.0 equiv.) is added at 0 ℃ under nitrogen and stirring is continued for 3 hours at ambient temperature. After completion, the reaction mixture was poured into a saturated aqueous ammonium chloride solution and extracted with ethyl acetate. The combined organic extracts were washed with brine and dried (Na) 2SO4) And concentrated in vacuo. The crude residue was purified by reverse phase preparative HPLC to give the title compound as a white solid (0.016g, 5%).1H NMR(400MHz,CD3OD ═ 8.45(br.s,1H),8.16(s,1H),7.55(br.s,1H),6.87(s,1H),2.80(t, J ═ 7,2Hz,4H),2.66(t, J ═ 7.2Hz,4H),1.96(quin, J ═ 7.6Hz,4H),1.53(s, 6H). LCMS, purity 98%, M/z 416.09(M + H)+)。C21H25N3O4S[M+H]+HRMS (FAB)+) Calculated values: 416.1566, found: 416.1556.

n- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -6, 7-dihydro-5H-cyclopenta [ b ] pyridine-3-sulfonamide

In general procedure C2, 4-isocyanato-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared by general procedure A2) and 6, 7-dihydro-5H-cyclopenta [ b ] are used]Pyridine-3-sulfonamide to give the title compound as a white solid (12mg, 27%).1H NMR(600MHz,DMSO-d6):δ=8.71(s,1H),8.01(s,1H),7.96(bs,1H),6.87(s,1H),2.97-2.93(m,4H),2.75(t,J=6Hz,4H),2.55(t,J=6Hz,4H),2.11-2.07(m,2H),1.93-1.88(m,4H)。13C NMR(150MHz,DMSO-d6):δ169.6,146.4,144.9,143.2,137.4,137.2,131.0,129.6,117.7,108.7,34.0,32.9,30.6,30.3,25.4,23.2.LCMS(m/z):398[M+H]+

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) pyridine-2-sulfonamide

The title compound was obtained as a white solid (40mg, 10%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and pyridine-2-sulfonamide in general method C1.1H NMR(400MHz,DMSO-d6):δ=8.5(d,J=4.0Hz,1H),7.88(t,J=7.6Hz,1H),7.81(t,J=7.6Hz,1H),7.59(s,1H),7.4(t,J=5.8Hz,1H),6.76(s,1H),2.73(t,J=7.2Hz,4H),2.61(t,J=7.2Hz,4H),1.88(quin,J=7.2Hz,4H)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) pyridine-3-sulfonamide

The title compound was obtained as a white solid (12mg, 3%) using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and pyridine-3-sulfonamide in general method C1. 1H NMR(400MHz,CD3OD):δ=9.08(s,1H),8.65(d,J=4.4Hz,1H),8.36(d,J=8.0Hz,1H),7.56(dd,J=8.0,4.8Hz,1H),6.88(s,1H),2.82(t,J=7.2Hz,4H),2.69(t,J=7.2Hz,4H),2.0(quin,J=7.2Hz,4H)。

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (trifluoromethyl) pyridine-2-sulfonamide

Using 4-isocyanato-8-methyl-1, 2,3,5,6, 7-hexahydro-s-indacene (prepared using general method a 1) and 4- (trifluoromethyl) pyridine-2-sulfonamide in general method C1 to give the title compound as a white solid (16mg, 3%).1H NMR(400MHz,CD3OD):δ=8.47(s,1H),8.22(s,1H),7.42(s,1H),6.98(s,1H),2.82(t,J=7.2Hz,4H),2.66(t,J=7.2Hz,4H),1.95(quin,J=7.2Hz,4H);19F NMR(233.33MHz,DMSO-d6):-63.48(s,3F)。

Joint

N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbothioformyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide

To a solution of 1,2,3,5,6, 7-hexahydro-s-indacen-4-amine (0.10g, 0.58mmol) in anhydrous DCM (2.0mL) was added 1,1' -thiocarbonyldiimidazole (1.1 eq) and the reaction was stirred at ambient temperature for 4 h. The solvent was removed in vacuo, then the residue dissolved in acetone (2.0mL) and potassium carbonate (2.5 equiv.) was added followed by 4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (1.2 equiv.). The reaction mixture was heated at reflux overnight, concentrated in vacuo, then neutralized with 10% citric acid (10mL) and immediately extracted with ethyl acetate (2 × 10mL), dried (MgSO4) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using MeOH/DCM eluent followed by HPLC to give the title compound as an off-white solid (13mg, 4%). 1H NMR(400MHz,DMSO-d6):δ=12.0(bs,1H),9.72(s,1H),7.88(s,1H),7.43(s,1H),7.01(s,1H),5.15(br.s.,1H),2.81(t,J=6.8Hz,4H),2.59(t,J=6.8Hz,4H),1.95(quin,J=7.6Hz,4H),1.39(s,6H)。13C NMR(100MHz,DMSO-d6) Purity 95.08%, tr 3.45min, M/z 421.30(M + H) 176.9,143.1,142.7,138.7,137.1,130.4,119.3,117.7,66.6,32.4,30.9,29.9,24.9 LCMS+)。

Biological test method

NLRP3 inhibition assay

The inhibitory activity of test compounds against the inflammasome of NLRP3 can be determined using the following assay (using common stimulants such as adenosine triphosphate, nigericin, LeuLe u-OMe or monosodium urate crystals (MSU)).

Cell culture

To produce HMDM, human monocytes were separated from buffy coat blood using Ficoll-Plaque Plus (GE Healthcare) and density centrifugation. CD14 Using MACS magnetic beads (Miltenyl Biotec)+And (4) selecting cells. Isolated CD14+Monocytes were differentiated for 7 days in cultures of L-glutamine containing il-koff modified Dulbecco's medium (IMDM) supplemented with 10% FBS and 1% penicillin/streptomycin (Life Technologies) with 10ng/ml human CSF-1(Miltenyl Biotec), as described by Croker et al, 2013Immunol Cell Biol 91: 625, respectively.

Mouse bone marrow-derived macrophages (BMDM) were derived from bone marrow progenitor cells isolated from the femur and tibia of C57BL/6 mice. Bone was washed with medium and bone marrow cells were cultured for 7 days in RPMI 1640 medium supplemented with 10% heat-inactivated FCS, 2mM GlutaMAX (Life Technologies), 50U/ml penicillin-streptomycin (Life Technologies), and 150ng/ml recombinant human M-CSF (endotoxin free, expressed and purified by university of Queensland protein expression equipment).

NLRP3 inflammatory body activation assay

HMDM at 1 × 105Inoculation is carried out in a/ml mode. The next day the overnight medium was changed and the cells were incubated with E.coli (Escherichia coli) serotype 0111: b4(Sigma Aldrich) stimulated the cells for 3 hours. Media was removed 30 minutes prior to NLRP3 stimulation and replaced with Serum Free Media (SFM) containing test compounds. Cells were then stimulated with adenosine 5' -triphosphate disodium salt hydrate (5mM 1 hr), nigericin (10. mu.M 1 hr), Leu-OMe (1mM 2 hr) or MSU (200. mu.g/ml 15 hr). ATP may be derived from Sigma Aldrich, nigericin and MSU from Invivogen, and Leu-Ome from Chem-Impex International.

BMDM at 1X 105Inoculation is carried out in a/ml mode. The next day the overnight medium was changed and the cells were stimulated with Ultrapure lipopolysaccharide from the e.coli K12 strain (InvivoGen) for 3 hours. Media was removed 30 minutes prior to NLRP3 stimulation and replaced with Serum Free Media (SFM) containing test compounds. Cells were then stimulated with adenosine 5' -triphosphate disodium salt hydrate (1.25-5mM 1 hr), nigericin (5. mu.M 1 hr), Leu-OMe (1mM 2 hr) or MSU (200. mu.g/ml 15 hr). ATP may be derived from Sigma Aldrich, nigericin and MSU from Invivogen, and Leu-Ome from Chem-Impex International.

Measurement of IL-1 beta, IL-18, TNF alpha and cell death

For ELISA and cell death assays, cells were seeded in 96-well plates. According to the manufacturer's instructions (R&D Systems,ReadySetGo!eBioscience,BD OptEIATMOr Perkin Elmer) The supernatant was removed and analyzed using an ELISA kit. By usingCell death was assessed relative to measurement of LDH release for 100% cell lysis control by a non-radioactive cytotoxicity assay (Promega).

Murine studies on compound levels in plasma and brain

General experiments: carbutamide was purchased from Sigma Aldrich (Cat. No. 381578). The acetonitrile isHPLC grade (Sigma Aldrich, Sydney, Australia), formic acid 99% -100% Normapur (VWR Internati) for AR gradeAnal Pty Ltd, Brisbane, Australia), DMSO isGrade (D5879, Sigma Aldrich, Sydney, Australia), and H2O Milli-Q was filtered. HPLC vials and polypropylene inserts were from Agilent Technologies (Melbourne, Australia), while 1.5mL Eppendorf Tubes Protein Lobind Tubes were from VWR International Pty Ltd (Brisbane, Australia).

Preparation of a precipitation solution: 100mL of ACN and 5. mu.L of 10mM sulfabutamide in DMSO (ACN, 135ng/mL internal sulfabutamide MS standard).

Preparation of plasma standard curve: preparation of 10mM NH4HCO31mg/mL of test compound in (1), and diluted 10-fold to give 100,000ng/mL of stock solution. With 10mM NH 4HCO3A series of 10-fold dilutions of the 100,000ng/mL stock solution were performed to give concentrations of 10,000, 1,000, 100 and 10 ng/mL. With 10mM NH4HCO3100,000ng/mL stock solutions were diluted to 3:7 to give a concentration of 30,000ng/mL, and a series of 10-fold dilutions gave concentrations of 3,000, 300, 30, and 3 ng/mL.

mu.L of the test compound-containing solution and 160. mu.L of the precipitation solution were added to 20. mu.L of mouse plasma in a low-binding Eppendorf tube. The sample was vortexed, allowed to stand at 4 ℃ for 10 minutes, and centrifuged at 14,000 Xg for 8 minutes. Transfer 150 μ L of supernatant to HPLC vial insert. The samples were stored at 4 ℃ until analysis.

Preparing a brain homogenate standard curve: the sample solution prepared for the plasma standard curve was used for the brain homogenate standard curve.

Mouse brain homogenates from saline controls were thawed and vortexed for 3 minutes or until homogeneous, and sonicated for 1 minute. When the foam settled, 50. mu.L of mouse brain homogenate was transferred to Eppendorf tubes, followed by 10mM NH4HCO350 μ L of test Compound (II), 150 μ L H2O and 500 μ L ice-cold precipitation solution, vortexed after each addition. The standards were allowed to stand at 4 ℃ for 10 minutes and then centrifuged at 14,000 Xg for 8 minutes. Transfer 200 μ L of supernatant to HPLC vial insert, ensure absence of air bubbles And the samples were stored at 4 ℃ until analysis.

Mouse administration and Heart infusion

Administration: oral tube feeding at 20mg/kg

Time points are as follows: 2 hours

Stock compounds were prepared in sterile PBS for administration at 4 mg/ml. Each mouse was weighed and dosed with 20mg/kg oral gavage for each compound. After 2 hours, the mice were anesthetized with a combination of suetadine (50mg/kg) and xylazine (10mg/kg) and blood was collected by cardiac puncture into tubes containing 20 μ L of 100 mM EDTA. Blood was centrifuged at 2000 Xg for 15 minutes at 4 ℃ to collect plasma.

Plasma samples prepared for analysis: adding 20 mu L of NH4HCO3And 160. mu.L of the precipitation solution was added to 20. mu.L of mouse plasma in a low-binding Eppendorf tube. The sample was vortexed, allowed to stand at 4 ℃ for 10 minutes, and centrifuged at 14,000 Xg for 8 minutes. Transfer 150 μ L of supernatant to HPLC vial insert to ensure no air bubbles are present. The samples were stored at 4 ℃ until analysis.

Preparing a brain homogenate: the brains of the mice were perfused with PBS, maintained for 5 minutes, then dissected and weighed. Brain homogenates were prepared by homogenizing total brain (0.5g) with 4 volumes (2ml) of deionized water and stored at-20 ℃ prior to analysis. The homogenate was thawed, vortexed for 3 minutes or until homogeneous, and sonicated for 1 minute. When the foam settled, 50. mu.L of mouse brain homogenate was transferred to Eppendorf tubes, followed by the addition of 50. mu.L of 10mM NH 4HCO3,150μLH2O and 500 μ L ice-cold precipitation solution, vortexed after each addition. Transfer 200 μ L of supernatant to HPLC vial insert, ensure no air bubbles are present, and store sample at 4 ℃ until analysis.

Brain samples prepared for analysis: 50 μ L of mouse brain was transferred to Eppendorf tubes, followed by the addition of 50 μ L of 10mM NH4HCO3,150μL H2O and 500 μ L ice-cold precipitation solution, vortexed after each addition. The solution was allowed to stand at 4 ℃ for 10 minutes, and then centrifuged at 14,000 Xg for 8 minutes. Transfer 200 μ L of supernatant to HPLC vial insert, ensure no air bubbles are present, and store sample at 4 ℃ until analysis.

LC-MS/MS: a sample was taken using a sample analyzer having 2 Shimadzu Nexera LC-30AD Solvent Delivery Units (Solvent Delivery Units), Shimadzu Nexera SIL-30AC Auto-Sampler (Auto-Sampler), Shimadzu project DGU-20A5Samples were analyzed on AB Sciex 4000QTrap MS from the Degasser (Degasser), Shimadzu development CBM-20A System Controller (System Controller) and Shimadzu development CTO-20A Column Oven (Column Oven). The column oven was set to 40 ℃ and the autosampler to 15 ℃. mu.L of injection was prepared and MS analysis was performed using Turbo Spray (-) -ESI with low resolution Q1 and low resolution Q3 in a Selected Reaction Monitoring (SRM) mode. MS parameters: and (4) CUR: 30.00, IS: -4300.00, TEM: 500.00, GS 1: 50.00, GS 2: 50.00, ihe: ON, CAD: high, DP-60.00, EP-10.00, CXP-15.00. MCC950 SRM: q1403.2 to Q3204.3Da, held for 150 milliseconds, CE-27 and carbamide (IS) SRM: q1270.0 to Q3171.0 Da, held for 100 milliseconds, CE-25. HPLC column: T35 μm 2.1X 50mm, havingT35 μm 2.1X 10mm guard bar. Flow rate and solvent: 0.35ml/min, solvent a: 0.1% formic acid in H2In the solvent O, the solvent B: 0.1% formic acid in ACN; isocratic 2% B from 0 → 2 min, gradient 2% → 100% B from 2 → 5 min, isocratic 100% from 5 → 9 min, gradient 100% → 2% B from 9 → 9.1 min and isocratic 2% B from 9.1 → 13 min. The peak areas of SRM data for sulfadimidine and test compounds were analyzed using AB Sciex's Analyst software using quantitative Wizard (quantification Wizard). The peak areas are plotted against ng/mL concentration in 20 μ L of 3 to 30,000ng/mL test compound solution and the lower and upper limits of the linear response are determined. These data were then plotted in Microsoft Excel and the linear response equation was used to determine the concentration of test compound in 20 μ L of plasma solution. Similarly, for brain homogenate samples, the peak area of 50 μ L of a 3 to 3,000ng/mL test compound solution was used to determine the assay in 50 μ L of brain homogenate solutionCompound concentration.

Results

The full range of tpsas and biological results are provided in the table below, but selected data for certain compounds of the invention are given below.

Table 1: topological polar surface area (tPSA) and molecular weight of the selected compound.

Table 3: plasma levels of selected test compounds at a 2 hour time point after oral gavage at 20mg/Kg

Table 4: the properties of sulfonylureas, including increased BBB penetration, the presence and absence of hydroxyalkyl groups on the furan ring.

Plasma concentration after administration

Single dose pharmacokinetic studies of N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide (MCC7840, and being a compound of the first aspect) compared to (N) - ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (MCC950) using an intravenous dose of 4mg/Kg and a po dose of 20mg/Kg clearly show a prolonged half-life of the pyrazole derivative compared to furan, an increased maximum concentration (C.sub.C7840) of the pyrazole derivativemax) And area under the curve (AUC). This is advantageous, resulting in relatively lower doses or less frequent administration.

The following procedure was followed: male C57BL/6 mice, 7-9 weeks old, were used, 3 animals per group. The test compounds were administered to mice using a single bolus intravenous injection or oral feeding. Blood samples were collected via the inframandibular or saphenous vein to analyze plasma concentrations of compounds by LC-MS/MS at the following time points: IV (3 mice): PO (3 mice) 0.083, 0.25, 0.5, 1,2, 4, 8 and 24 hours post-dose: 0.25, 0.5, 1,2, 4, 8 and 24 hours post-dose. An LC-MS/MS method for the quantitative determination of test compounds in the corresponding biological matrices was developed. PK parameters were calculated using Phoenix WinNonlin 6.3. The results are shown schematically in fig. 1A to 1C (MCC950) and fig. 2A to 2C (MCC 7840).

The structures of the related compounds are shown below, and tables 5-8 contain the related data:

tables 5 and 6: PK and bioavailability data for N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4- (2-hydroxypropan-2-yl) furan-2-sulfonamide (MCC950)

Tables 7 and 8: PK and bioavailability data for N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide (MCC7840)

234页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:一种合成On-DNA N,N-单取代吲唑酮类化合物的方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!