CN114685489A - Polysubstituted nitrogen-containing heterocyclic compound and application thereof - Google Patents

Polysubstituted nitrogen-containing heterocyclic compound and application thereof Download PDF

Info

Publication number
CN114685489A
CN114685489A CN202111659383.XA CN202111659383A CN114685489A CN 114685489 A CN114685489 A CN 114685489A CN 202111659383 A CN202111659383 A CN 202111659383A CN 114685489 A CN114685489 A CN 114685489A
Authority
CN
China
Prior art keywords
compound
reaction
pharmaceutically acceptable
substituted
acceptable salt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202111659383.XA
Other languages
Chinese (zh)
Inventor
刘扬
刘磊
周峰
杨桂梅
唐任宏
任晋生
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nanjing Zaiming Pharmaceutical Co ltd
Original Assignee
Jiangsu Simcere Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jiangsu Simcere Pharmaceutical Co Ltd filed Critical Jiangsu Simcere Pharmaceutical Co Ltd
Publication of CN114685489A publication Critical patent/CN114685489A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides a compound shown as a formula (I) or pharmaceutically acceptable salt thereof, a pharmaceutical composition and a preparation method thereof, and application of the compound as an HPK1 inhibitor.

Description

Polysubstituted nitrogen-containing heterocyclic compound and application thereof
The invention requires the priority of the prior application with patent application number 202011637920.6 and invented name of 'polysubstituted nitrogen heterocyclic compound and application thereof' submitted to the intellectual property office of China at 31.12.2020. The entire contents of the above-mentioned prior application are incorporated herein by reference.
Technical Field
The invention relates to a novel polysubstituted nitrogen-containing heterocyclic compound or a pharmaceutically acceptable salt thereof, a pharmaceutical composition containing the polysubstituted nitrogen-containing heterocyclic compound or the pharmaceutically acceptable salt thereof and application of the polysubstituted nitrogen-containing heterocyclic compound or the pharmaceutically acceptable salt thereof as an HPK1 inhibitor in preventing or treating related diseases.
Background
One of the main features of cancer is the ability to evade immunity. Tumor cells inhibit the recognition and attack of the body's immune system through a variety of complex mechanisms. To address this immune suppression, various strategies for tumor immunotherapy have been devised, including mechanisms that interfere with negatively regulated effector T cell function, such as PD1/PDL1 immune checkpoint inhibitors, to address immune suppression of T lymphocytes in PDL 1-highly expressing cancer cells by blocking the PD1 and PDL1 interactions, and clinical benefits in a variety of cancer types have also been demonstrated for PD1 or PDL1 inhibitor antibody development. In addition, therapeutic antibodies that block the interaction between CD80/CD86 and the T cell co-inhibitory receptor (CTLA-4) can promote T cell expansion in lymphoid tissues at various levels. In addition to these cell surface-associated proteins, intracellular signals have been found to be involved in immune down-regulation, wherein HPK1 (hematopoietic progenitor kinase 1, also known as MAP4K1) specifically expressed in hematopoietic cells is a serine/threonine kinase mainly involved in immune down-regulation in cells.
The research finds that the inactivation of HPK1 in human cells and mouse cells is frequently accompanied with the occurrence of autoimmune diseases, and indicates that HPK1 regulates the immune tolerance of the body. For example, HPK1 expression was found to be down-regulated in peripheral mononuclear cells (PBMC) from psoriatic arthritis patients and T cells from systemic lupus erythematosus patients (J Autoimmun 2011,37(3), 180-9); mouse model experiments found that mice deficient in HPK1 were more susceptible to autoimmune meningitis (Nat Immunol 2007,8(1), 84-91). In vitro studies demonstrated that antigen stimulation of T and B lymphocytes derived from HPK1 depletion had a greater activating effect (Cancer immunol. 2010,59(3),419-429), indicating that HPK1 negatively regulates T and B lymphocyte function. In addition, HPK 1-depleted dendritic cells (DC cells) exhibited more efficient antigen presentation and T cell activation properties, indicating that HPK1 is also involved in immune regulation of DC cells.
After activation of T Cell Receptor (TCR) and B Cell Receptor (BCR), cytoplasmic HPK1 is recruited to the vicinity of cell membrane to be activated, and activated HPK1 phosphorylates adaptor protein SLP76 or LAT, so that activated SLP76 is used as a docking site of negative regulatory protein 14-3-3 pi, mediates SLP76 ubiquitination degradation, and finally leads to instability of TCR signal complex, thereby down-regulating TCR signal (J.cell biol.2011,195(5), 839-853). It was also found that HPK1 could be activated by PGE2 (prostaglandin E2) in a PKA-dependent manner and possibly even by immunosuppressive factors expressed by tumor cells (Blood 2003,101(9), 3687-3689).
HPK1 in comparison with the wild type-/-The mouse shows strong growth inhibition effect on the growth of the inoculated isogenic lung cancer tumor. Demonstration of HPK by anti-tumor immunoreaction studies in a T cell-transplanted mouse model-/-The strong anti-tumor effect of the knockdown is at least in part T-cell dependent. The dendritic cells also contribute to the antitumor activity by being derived from HPK1-/-DC cell transplantation experiments of defective mouse bone marrow were confirmed (j. immunol.2009,182(10), 6187-61). Recently, it was found that the HPK1 transgenic mice with inactivated catalytic enzyme were equally effective in inhibiting the growth of glioblastoma GL261 and in enhancing the efficacy of anti-PD 1 in treating MC38 tumors, as compared to wild-type HPK transgenic mice. Therefore, HPK1 is a potential anti-tumor therapeutic target, and the effect of developing small molecule inhibitors against HPK1 kinase on anti-tumor therapy as single drugs or combined with other immune regulation therapeutic strategies is expected.
Disclosure of Invention
The invention provides a compound shown as a formula (I) or a pharmaceutically acceptable salt thereof:
Figure BDA0003446981180000021
wherein,
ring Q is selected from phenyl, 5-6 membered heteroaryl or 4-7 membered heterocyclyl;
R1selected from H or optionally substituted by Ra1Substituted of the following groups: c1-C6Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl;
R2selected from the group consisting ofa2Substituted of the following groups: c1-C6Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl, or R1、R2Together with the atoms to which they are attached form a 5-7 membered heterocyclyl;
L1is absent, or L1Is selected from C1-C6Alkylene, (C)1-C3Alkylene) -O- (C)1-C3Alkylene) or (C)1-C3Alkylene) -NH- (C)1-C3Alkylene groups);
R3is absent, or, R3Selected from H or optionally substituted by Ra3Substituted of the following groups: OH, NH2、C1-C6Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl, or R1、R3Together with the atom to which they are attached form a 5-7 membered heterocyclic group, or R2、R3Together with the atoms to which they are attached form a 5-7 membered heterocyclyl;
R4selected from H, halogen, CN or optionally substituted by Ra4Substituted of the following groups: OH, NH2、C1-C6Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl;
R5、R6independently selected from H, halogen, CN, C1-C6Alkyl or C1-C6Alkoxy radical, said C1-C6Alkyl or C1-C6Alkoxy is optionally substituted by F, Cl, Br, I, CN;
ring A is selected from
Figure BDA0003446981180000031
R7Selected from the group consisting ofa7A substituted 4-14 membered heterocyclyl;
X1、X4independently selected from CR9Or N;
X2、X3independently selected from C (R)10)(R11) Or NR12
R8、R9、R10、R11Independently selected from H, halogen, CN or optionally Ra8Substituted of the following groups: OH, NH2、C1-C6Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl;
R12selected from H or optionally substituted by Ra12Substituted of the following groups: c1-C6Alkyl radical, C3-C6Cycloalkyl or 4-7 membered heterocyclyl;
p, q are independently selected from 0, 1,2,3 or 4;
m and n are independently selected from 0, 1 or 2;
each Ra1、Ra2、Ra3、Ra4、Ra7、Ra8、Ra12Independently selected from halogen, CN, ═ O or optionally RbSubstituted of the following groups: OH, NH2、C1-C3Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl;
each RbIndependently selected from halogen, CN, ═ O, C1-C3Alkyl, OH, O (C)1-C3Alkyl), NH2、NH(C1-C3Alkyl), N (C)1-C3Alkyl radical)2
In some embodiments, R1Selected from H or optionally substituted by Ra1Substituted of the following groups: c1-C3Alkyl radical, C3-C6A cycloalkyl group.
In some embodiments, R1Selected from H, C1-C3Alkyl or C3-C6A cycloalkyl group.
In some embodiments, R1Selected from H, methyl, cyclopropyl or cyclopentyl.
In some embodiments, R2Selected from the group consisting ofa2Substituted of the following groups: c1-C3Alkyl radical, C3-C6A cycloalkyl group.
In some embodiments, R2Is selected from C1-C3Alkyl or C3-C6A cycloalkyl group.
In some embodiments, R2Selected from methyl or cyclopropyl.
In some embodiments, R1、R2Together with the atoms to which they are attached form a 5-6 membered heterocyclic group.
In some embodiments, L is1Is absent, or L1Is selected from C1-C3An alkylene group.
In some embodiments, L1Is absent, or L1Is selected from CH2
In some embodiments, a building block
Figure BDA0003446981180000032
Is selected from
Figure BDA0003446981180000033
Figure BDA0003446981180000041
In some embodiments, ring Q is selected from phenyl or 5-6 membered heteroaryl.
In some embodiments, ring Q is selected from phenyl or 5-membered heteroaryl.
In some embodiments, ring Q is selected from phenyl, thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, or
Figure BDA0003446981180000042
In some embodiments, ring Q is selected fromFrom phenyl or
Figure BDA0003446981180000043
In some embodiments, R3Is absent, or R3Is selected from H.
In some embodiments, R4Is selected from H.
In some embodiments, p is selected from 0 or 1.
In some embodiments, R1、R3Together with the atoms to which they are attached form a 5-6 membered heterocyclic group.
In some embodiments, R2、R3Together with the atoms to which they are attached form a 5-6 membered heterocyclic group.
In some embodiments, R3And
Figure BDA0003446981180000044
are ortho-substituents to each other.
In some embodiments, Q is taken from phenyl and R is3And
Figure BDA0003446981180000045
are ortho-substituents to each other.
In some embodiments, a building block
Figure BDA0003446981180000046
Is selected from
Figure BDA0003446981180000047
In some embodiments, a building block
Figure BDA0003446981180000051
Is selected from
Figure BDA0003446981180000052
Figure BDA0003446981180000053
In some embodimentsIn a table, a structural unit
Figure BDA0003446981180000054
Is selected from
Figure BDA0003446981180000055
Figure BDA0003446981180000056
In some embodiments, R5、R6Independently selected from H, halogen, CN or C1-C3An alkyl group.
In some embodiments, R5、R6Independently selected from H.
In some embodiments, X1Selected from the group consisting of CR9
In some embodiments, X1Is selected from CH.
In some embodiments, R7Selected from the group consisting ofa7Substituted 6-8 membered heterocyclyl.
In some embodiments, R7Selected from the group consisting ofa7Substituted piperazinyl, morpholinyl, tetrahydropyranyl or
Figure BDA0003446981180000057
In some embodiments, Ra7Selected from the group consisting ofbSubstituted of the following groups: c1-C3Alkyl, 5-6 membered heterocyclyl.
In some embodiments, Ra7Selected from methyl or optionally substituted by RbSubstituted of the following groups: tetrahydropyranyl, piperidinyl.
In some embodiments, RbIs selected from C1-C3An alkyl group.
In some embodiments, RbSelected from methyl.
In some embodiments, R7Is selected from
Figure BDA0003446981180000061
Figure BDA0003446981180000062
In some embodiments, R8Selected from H or optionally substituted by Ra8Substituted of the following groups: c1-C3Alkyl, 5-membered heterocyclic group.
In some embodiments, Ra8Is selected from quilt C1-C3Alkyl substituted NH2
In some embodiments, Ra8Is selected from N (CH)3)2
In some embodiments, R8Selected from H, methyl, CH2N(CH3)2Or
Figure BDA0003446981180000063
In some embodiments, q is selected from 1 or 2.
In some embodiments, a building block
Figure BDA0003446981180000064
Is selected from
Figure BDA0003446981180000065
Figure BDA0003446981180000066
In some embodiments, X4Selected from the group consisting of CR9
In some embodiments, X4Is selected from CH.
In some embodiments, R10、R11Independently selected from H.
In some embodiments, R12Selected from methyl.
In some embodiments, X2、X3Independently selected from CH2Or NCH3
In some embodiments, m, n are independently selected from 1.
In some embodiments, a building block
Figure BDA0003446981180000067
Is selected from
Figure BDA0003446981180000068
In some embodiments, ring a is selected from
Figure BDA0003446981180000069
Figure BDA0003446981180000071
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is selected from the group consisting of compounds of formula (II) or a pharmaceutically acceptable salt thereof:
Figure BDA0003446981180000072
wherein, ring A, L1、R1、R2、R3、R4、R5、R6、X1P is as defined above.
In some embodiments, the compound of formula (I), or a pharmaceutically acceptable salt thereof, is selected from the following compounds, or a pharmaceutically acceptable salt thereof:
Figure BDA0003446981180000073
Figure BDA0003446981180000081
the invention also provides a pharmaceutical composition which comprises the compound shown in the formula (I) or pharmaceutically acceptable salt thereof and pharmaceutically acceptable auxiliary materials.
Further, the invention relates to application of the compound shown in the formula (I) or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof in preparing a medicament for preventing or treating HPK1 related diseases.
Further, the invention relates to an application of the compound shown in the formula (I) or the pharmaceutically acceptable salt thereof, or the pharmaceutical composition thereof in preparing a medicament for preventing or treating tumors.
Further, the invention relates to application of the compound shown in the formula (I) or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof in preventing or treating HPK1 related diseases.
Further, the invention relates to an application of the compound shown in the formula (I) or the pharmaceutically acceptable salt thereof or the pharmaceutical composition thereof in preventing or treating tumors.
Further, the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof for preventing or treating HPK 1-related diseases.
Further, the present invention relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof for preventing or treating tumors.
The present invention also relates to a method of treating HPK1 related diseases comprising administering to a patient a therapeutically effective dose of a pharmaceutical formulation comprising a compound of formula (I) as described herein or a pharmaceutically acceptable salt thereof.
The invention also relates to a method of treating neoplasms comprising administering to a patient a therapeutically effective amount of a pharmaceutical formulation comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, as described herein.
In some embodiments, the HPK 1-related disease is selected from a tumor.
Definition and description of terms
Unless otherwise indicated, the definitions of groups and terms described in the specification and claims of the present invention, including definitions thereof as examples, exemplary definitions, preferred definitions, definitions described in tables, definitions of specific compounds in the examples, and the like, may be arbitrarily combined and coupled with each other. The definitions of the groups and the structures of the compounds in such combinations and after the combination are within the scope of the present invention as defined in the specification.
In this context
Figure BDA0003446981180000091
Indicates the attachment site.
The term "pharmaceutically acceptable salts" refers to pharmaceutically acceptable salts of non-toxic acids or bases, including salts of inorganic acids and bases, organic acids and bases.
The term "stereoisomer" refers to isomers resulting from the different arrangement of atoms in a molecule, including cis-trans isomers, enantiomers, and diastereomers.
The compounds of the present invention may have an asymmetric atom such as a carbon atom, a sulfur atom, a nitrogen atom, a phosphorus atom (optical center) or an asymmetric double bond. Racemates, enantiomers, diastereomers, geometric isomers are included within the scope of the present invention.
When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, they include the E, Z geometric isomer unless otherwise specified. Likewise, all tautomeric forms are included within the scope of the invention.
The compounds of the present invention may exist in specific geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis and trans isomers, (-) -and (+) -enantiomers, (R) -and (S) -enantiomers, diastereomers, (D) -isomers, (L) -isomers, as well as racemic and other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, all of which are within the scope of the present invention. An additional asymmetric carbon atom, asymmetric sulfur atom, asymmetric nitrogen atom or asymmetric phosphorus atom may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are included within the scope of the present invention. The compounds of the present application containing asymmetric atoms can be isolated in optically active pure form or in racemic form. The optically active pure form can be resolved from a racemic mixture or synthesized by using chiral starting materials or chiral reagents.
The term "tautomer" refers to an isomer of a functional group resulting from the rapid movement of an atom in two positions in a molecule. The compounds of the invention may exhibit tautomerism. Tautomeric compounds may exist in two or more interconvertible species. Tautomers generally exist in equilibrium, and attempts to isolate a single tautomer often result in a mixture whose physicochemical properties are consistent with the mixture of compounds. The position of equilibrium depends on the chemical properties within the molecule. For example, in many aliphatic aldehydes and ketones such as acetaldehyde, the keto form predominates; whereas in phenol the enol type predominates. The present invention encompasses all tautomeric forms of the compounds.
The term "pharmaceutical composition" denotes a mixture of one or more compounds described herein or a physiologically/pharmaceutically acceptable salt or prodrug thereof with other chemical components, e.g. physiologically/pharmaceutically acceptable excipients. The purpose of the pharmaceutical composition is to facilitate the administration of the compound to an organism.
The term "substituted" means that any one or more hydrogen atoms on a particular atom is replaced with a substituent, so long as the valence of the particular atom is normal and the substituted compound is stable. When the substituent is oxo (i.e., ═ O), meaning that two hydrogen atoms are substituted, oxo does not occur on the aryl.
The terms "optionally" or "optionally" mean that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. For example, ethyl is "optionally" substituted with halo, meaning that ethyl may be unsubstituted (CH)2CH3) Monosubstituted (e.g. CH)2CH2F) Polysubstituted (e.g. CHFCH)2F、CH2CHF2Etc.) or completely substituted (CF2CF3). It will be appreciated by those skilled in the art that any group containing one or more substituents will not incorporate any substitution or substitution pattern which is sterically impossible and/or cannot be synthesized.
When a linking group is absent, it means that the two groups it is attached toDirectly connected, e.g. as building blocks
Figure BDA0003446981180000101
L in (1)1When not present, the structure represented by the structural unit is
Figure BDA0003446981180000102
The direction of attachment of the linking group referred to herein is arbitrary unless it is indicated. For example, when the structural unit
Figure BDA0003446981180000103
L in (1)1Is selected from "C1-C3alkylene-O', when L is1The rings Q and R may be connected in the same direction as the reading sequence from left to right1Form "Ring Q-C1-C3alkylene-O-R1", the rings Q and R may be connected in the opposite direction of the reading sequence from left to right1Form "Ring Q-O-C1-C3alkylene-R1”。
The term "halo" or "halogen" refers to fluorine, chlorine, bromine and iodine.
The term "C1-C6Alkyl "is understood to mean a straight-chain or branched, saturated monovalent hydrocarbon radical having 1,2,3,4, 5 or 6 carbon atoms. Specific examples of the alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-ethylpropyl, 1, 2-dimethylpropyl, neopentyl, 1-dimethylpropyl, 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3-dimethylbutyl, 2-dimethylbutyl, 1-dimethylbutyl, 2, 3-dimethylbutyl, 1, 2-dimethylbutyl, and the like; preferably, "C1-C6Alkyl groups "may contain" C1-C3Alkyl group and C1-C3Alkyl is understood to mean having 1,2,3A straight or branched chain saturated monovalent hydrocarbon group of carbon atoms.
The term "C1-C6Alkoxy "is understood to mean" C1-C6Alkyloxy "or" C1-C6alkyl-O', preferably, "C1-C6Alkoxy groups "may contain" C1-C3Alkoxy ".
The term "alkylene" is understood to mean a saturated divalent hydrocarbon radical having a straight or branched chain. "C1-C6Alkylene "is understood to mean a saturated divalent hydrocarbon radical having a linear or branched chain and having from 1 to 6 carbon atoms, preferably" C1-C6Alkylene groups "may comprise" C1-C3An alkylene group ".
The term "C3-C6Cycloalkyl "is understood to mean a saturated, monovalent monocyclic or bicyclic hydrocarbon ring radical having 3 to 6 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.
The term "4-14 membered heterocyclyl" means a saturated or partially saturated monovalent monocyclic, bicyclic, spiro or bridged ring group containing 4 to 14 ring atoms, and containing 1 to 5, preferably 1 to 3 ring atoms selected from N, O, S, S (O) or S (O)2A heteroatom or a heteroatom group of (a). The "4-to 14-membered heterocyclic group" of the present invention may include "4-to 7-membered heterocyclic group", "5-to 6-membered heterocyclic group", "6-to 8-membered heterocyclic group" and the like. Specific examples include, but are not limited to: 4-membered rings such as azetidinyl, oxetanyl; or a 5-membered ring such as tetrahydrofuranyl, dioxolyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl; or a 6-membered ring such as tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, or trithianyl; or a partially saturated 6-membered ring such as tetrahydropyridinyl; or a 7-membered ring, such as diazepanyl; or 5, 5-membered fused rings, e.g. hexahydrocyclopenta [ c]Pyrrol-2 (1H) -yl,
Figure BDA0003446981180000111
Or a 5,6 membered bicyclic ring, e.g. hexahydropyrrolo [1,2-a ]]Pyrazin-2 (1H) -yl, 5,6,7, 8-tetrahydro- [1,2,4]Triazole compoundsAnd [4,3-a ]]A pyrazinyl group; optionally, the "4-14 membered heterocyclic group" may also be a benzo-fused ring group of the above-mentioned 4-membered heterocyclic group, 5-membered heterocyclic group or 6-membered heterocyclic group.
The term "5-6 membered heteroaryl" is to be understood as a monovalent aromatic ring system having 5 or 6 ring atoms and which contains 1-3 heteroatoms independently selected from N, O and S. In particular from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl and the like.
In particular, the term "treating" means administering a compound or formulation described herein to prevent, ameliorate or eliminate a disease or one or more symptoms associated with the disease, and includes:
(i) preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is susceptible to the disease condition, but has not yet been diagnosed as having the disease condition;
(ii) inhibiting the disease or disease state, i.e., arresting its development;
(iii) alleviating the disease or condition, i.e., causing regression of the disease or condition.
The term "therapeutically effective amount" means an amount of a compound of the invention that (i) treats or prevents a particular disease, condition, or disorder, (ii) alleviates, ameliorates, or eliminates one or more symptoms of a particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of a particular disease, condition, or disorder described herein. The amount of a compound of the present invention that constitutes a "therapeutically effective amount" varies depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by those skilled in the art with their own knowledge and this disclosure.
The term "adjuvant" refers to a pharmaceutically acceptable inert ingredient. Examples of classes of the term "excipient" include, without limitation, binders, disintegrants, lubricants, glidants, stabilizers, fillers, diluents, and the like. Excipients enhance the handling characteristics of the pharmaceutical formulation, i.e., make the formulation more amenable to direct compression by increasing flowability and/or cohesiveness. Examples of typical "pharmaceutically acceptable carriers" suitable for use in the above formulations are: saccharides, starches, cellulose and its derivatives and the like are commonly used as excipients in pharmaceutical preparations.
The term "pharmaceutically acceptable adjuvants" refers to those adjuvants which do not have a significant irritating effect on the organism and do not impair the biological activity and properties of the active compound. Suitable adjuvants are well known to those skilled in the art, such as carbohydrates, waxes, water-soluble and/or water-swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water, and the like.
The words "comprise", "comprises", "comprising" or "including" and variations thereof such as "comprises" or "comprising" are to be interpreted in an open, non-exclusive sense, i.e., "including but not limited to".
The compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combinations thereof with other chemical synthetic methods, and equivalents thereof known to those skilled in the art, with preferred embodiments including, but not limited to, examples of the present invention.
The present application also includes isotopically-labeled compounds of the present application, which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the present application include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine and chlorine, such as respectively2H、3H、11C、13C、14C、13N、15N、15O、17O、18O、31P、32P、35S、18F、123I、125I and36cl, and the like.
Certain isotopically-labelled compounds of the present application (e.g. with3H and14c-labelledThese) can be used in compound and/or substrate tissue distribution assays. Tritiated (i.e. by tritiation)3H) And carbon-14 (i.e.14C) Isotopes are particularly preferred for their ease of preparation and detectability. Positron emitting isotopes, such as15O、13N、11C and18f can be used in Positron Emission Tomography (PET) studies to determine substrate occupancy. Isotopically labeled compounds of the present application can generally be prepared by following procedures analogous to those disclosed in the schemes and/or in the examples below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
In addition, heavier isotopes are used (such as deuterium (i.e., deuterium)2H) Substitution may provide certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements), and thus may be preferred in certain circumstances where deuterium substitution may be partial or complete, partial deuterium substitution meaning that at least one hydrogen is substituted with at least one deuterium.
The pharmaceutical compositions of the present application can be prepared by combining the compounds of the present application with suitable pharmaceutically acceptable excipients, for example, can be formulated into solid, semi-solid, liquid or gaseous formulations, such as tablets, pills, capsules, powders, granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres, aerosols, and the like.
Typical routes of administration of a compound of the present application or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof include, but are not limited to, oral, rectal, topical, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, intramuscular, subcutaneous, intravenous administration.
The pharmaceutical compositions of the present application can be manufactured by methods well known in the art, such as conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, lyophilizing, and the like.
In some embodiments, the pharmaceutical composition is in an oral form. For oral administration, the pharmaceutical compositions may be formulated by mixing the active compounds with pharmaceutically acceptable excipients well known in the art. These adjuvants enable the compounds of the present application to be formulated as tablets, pills, lozenges, dragees, capsules, liquids, gels, slurries, suspensions and the like, for oral administration to a patient.
Solid oral compositions may be prepared by conventional mixing, filling or tableting methods. For example, it can be obtained by the following method: the active compounds are mixed with solid adjuvants, optionally the mixture obtained is milled, if desired with further suitable adjuvants, and the mixture is then processed to granules, to give tablets or dragee cores. Suitable excipients include, but are not limited to: binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, and the like.
The pharmaceutical compositions may also be adapted for parenteral administration, as sterile solutions, suspensions or lyophilized products in suitable unit dosage forms.
In all methods of administration of the compounds of the general formula I described herein, the daily dose is from 0.01 to 100mg/kg body weight, preferably from 0.05 to 50mg/kg body weight, more preferably from 0.1 to 30mg/kg body weight, in single or divided doses.
The chemical reactions of the embodiments of the present invention are carried out in a suitable solvent that is compatible with the chemical changes of the present invention and the reagents and materials required therefor. In order to obtain the compounds of the present invention, it is sometimes necessary for a person skilled in the art to modify or select the synthesis steps or reaction schemes based on the existing embodiments.
Detailed Description
The invention is described in detail below by way of examples, but is not meant to be limited to any of the disadvantages of the present invention. Having described the invention in detail and having disclosed specific embodiments thereof, it will be apparent to one skilled in the art that various changes and modifications can be made therein without departing from the spirit and scope thereof. All reagents used in the present invention were commercially available and used without further purification.
Unless otherwise specified, the ratios expressed by the mixed solvents are volume mixing ratios.
Unless otherwise specified,% means weight percent wt%.
The compounds are prepared by hand or
Figure BDA0003446981180000131
The software names, and the commercial compounds used the supplier catalog names.
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) and/or Mass Spectrometry (MS). NMR shift in units of 10-6(ppm). Solvents for NMR determination are deuterated dimethyl sulfoxide, deuterated chloroform, deuterated methanol and the like, and an internal standard is Tetramethylsilane (TMS); ' IC50"half inhibitory concentration" means the concentration at which half of the maximum inhibitory effect is achieved.
Example 1: synthesis of 2-methyl-7- (3- (4- (S-methylsulfonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridin-5-yl) -1,2,3, 4-tetrahydroisoquinoline (Compound 1)
Figure BDA0003446981180000141
Compounds 1-2:
Figure BDA0003446981180000142
4-Bromobenzophenone sulfide (1.020g, 5.02mmol) was dissolved in ethanol (50mL), diacetoxyiodobenzene (4.85g, 15.07mmol) and ammonium acetate (1.55g, 20.09mmol) were added to the reaction system, and the reaction mixture was stirred at 25 ℃ for 13 hours. LCMS monitor reaction completion. The reaction mixture was filtered and concentrated, and the concentrated solution was purified by column chromatography (petroleum ether/ethyl acetate: 2/1) to give the product 1-bromo-4- (S-methylsulphonimidoyl) benzene (compound 1-2) (0.92 g).
LCMS m/z(ESI):233.7,235.7[M+H];
Compounds 1-3:
Figure BDA0003446981180000143
compound 1-2(400mg, 1.71mmol) was dissolved in dioxane (8mL), and pinacol ester diboronate (521.08mg, 2.05mmol), potassium acetate (503.05mg, 5.13mmol) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane (139.53mg, 171.00. mu. mol) were added to the reaction system under a nitrogen atmosphere, and replaced three times with nitrogen. The reaction mixture was stirred at 80 ℃ for 3 hours. LCMS monitor reaction completion. After the reaction solution was filtered, the filtrate was concentrated to dryness under reduced pressure, and then purified by column chromatography (dichloromethane/methanol ═ 10/1) to give 4,4,5, 5-tetramethyl-2- (4- (S-methylsulphonimidoyl) phenyl) -1,3, 2-dioxaborolan (compound 1-3) (480 mg).
1H NMR(400MHz,METHANOL-d4)δ8.01-7.94(m,4H),3.15(s,3H),1.36(s,12H).
Compounds 1-4:
Figure BDA0003446981180000151
5-bromo-1H-pyrrolo [2,3-b ] pyridine (5g, 25.38mmol), pinacol diboron (7.73g, 30.45mmol) and potassium acetate (7.98g, 50.75mmol) were dissolved in dioxane (100mL) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane (414.47mg, 507.53. mu. mol) was added under nitrogen. The reaction solution was stirred at 80 ℃ for 16 h. LCMS check reaction complete. The reaction solution was concentrated to dryness under reduced pressure. Water (200mL) was added, extraction was performed twice with ethyl acetate (300mL), the organic layer was dried over anhydrous magnesium sulfate, suction filtration was performed, and the filtrate was concentrated to dryness under reduced pressure. Purification by column chromatography (petroleum ether: ethyl acetate ═ 3: 1) gave compound 5- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrrolo [2,3-b ] pyridine (compound 1-4) (5.12 g).
LCMS m/z(ESI):245.2[M+H];
1H NMR(400MHz,DMSO-d6):δ11.75(br s,1H),8.46(d,J=1.5Hz,1H),8.27-8.16(m,1H),7.51-7.43(m,1H),6.52-6.39(m,1H),1.31(s,12H).
Compounds 1-5:
Figure BDA0003446981180000152
compounds 1-4(3.24g, 13.27mmol), 7-bromo-2-methyl-3, 4-dihydro-1H-isoquinoline (2g, 8.85mmol) and potassium phosphate (3.76g, 17.69mmol) were dissolved in anhydrous dioxane (80mL) and water (20mL) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane (722.33mg, 884.52. mu. mol) was added under nitrogen. The reaction mixture was stirred at 100 ℃ for 4 hours. LCMS check reaction complete. The reaction solution was concentrated to dryness under reduced pressure. Water (300mL) was added and the mixture was extracted three times with ethyl acetate (300 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered with suction, and the filtrate was concentrated to dryness under reduced pressure. Purification by column chromatography (petroleum ether/tetrahydrofuran ═ 1/2) gave the compound 2-methyl-7- (1H-pyrrolo [2,3-b ] pyridin-5-yl) -1,2,3, 4-tetrahydroisoquinoline (compound 1-5) (17 g).
LCMS m/z(ESI):264.1[M+H];
Compounds 1-6:
Figure BDA0003446981180000161
compound 1-5(1.4g, 5.32mmol) was dissolved in acetonitrile (30mL) and N-iodosuccinimide (1.2g, 5.32mmol) was added. The reaction mixture was stirred at 25 ℃ for 2 hours. LCMS check reaction complete. The reaction solution was concentrated to dryness under reduced pressure. Water (60mL) was added and the mixture was extracted three times with ethyl acetate (180 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered, and the filtrate was concentrated under reduced pressure to dryness to give crude 7- (3-iodo-1H-pyrrolo [2,3-b ] pyridin-5-yl) -2-methyl-1, 2,3, 4-tetrahydroisoquinoline (compound 1-6) (1.7 g). LCMS M/z (ESI) 390.1[ M + H ];
compounds 1-7:
Figure BDA0003446981180000162
compound 1-6(500mg, 1.28mmol) was dissolved in tetrahydrofuran (7mL), and sodium hydride (102.76mg, 2.57mmol, effective content 60%) was added in an ice-water bath at 0 ℃ and the reaction was stirred at 0 ℃ for 1 hour. Then, p-toluenesulfonyl chloride (367.35mg, 1.93mmol) was added to the reaction solution, and the reaction was stirred at room temperature for 5 hours. LCMS detected most of the starting material was completely reacted. The reaction was quenched at 0 ℃ with slowly dropping water (7mL), then the aqueous phase was extracted twice with ethyl acetate (20mL), and the organic phase was washed three times with saturated brine (15 mL). The collected organic phase was dried over anhydrous sodium sulfate and concentrated by filtration. The concentrated solution was purified by column chromatography (petroleum ether/tetrahydrofuran: 100/40, 0.5% methanol amide) to give 7- (3-iodo-1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridin-5-yl) -2-methyl-1, 2,3, 4-tetrahydroisoquinoline (compound 1-7) (270.00 mg).
LCMS m/z(ESI):543.9[M+H];
1H NMR(400MHz,METHANOL-d4)δ8.55(d,J=2.0Hz,1H),8.07-8.00(m,3H),7.81(d,J=2.3Hz,1H),7.43-7.31(m,4H),7.24(d,J=8.0Hz,1H),3.68(s,2H),2.98(t,J=5.9Hz,2H),2.81-2.73(m,2H),2.47(s,3H),2.36(s,3H).
Compounds 1-8:
Figure BDA0003446981180000171
7- (3-iodo-1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridin-5-yl) -2-methyl-1, 2,3, 4-tetrahydroisoquinoline (compound 1-7) (80mg, 147.22. mu. mol) was dissolved in water (0.8mL) and N, N-dimethylformamide (3.2mL), and compounds 1-3(58.60mg, 208.54. mu. mol), cesium carbonate (143.90mg, 441.65. mu. mol) and 1, 1-bis (di-tert-butylphosphine) ferrocene dichloropalladium (9.59mg, 14.72. mu. mol) were added to the reaction system under a nitrogen atmosphere and replaced with nitrogen three times. The reaction mixture was stirred at 100 ℃ for 13 hours. LCMS detects that the reaction is finished, the reaction solution is cooled to 25 ℃, the reaction solution is filtered, the filtrate is decompressed and concentrated to be dry, and crude compounds 1-8 are obtained and are directly used for the next reaction.
Compound 1:
Figure BDA0003446981180000172
the crude compounds 1 to 8 obtained in the above reaction were dissolved in N, N-dimethylformamide (2mL), and sodium hydroxide solution (4M, 2mL) was added to the reaction system. The reaction mixture was stirred at 80 ℃ for 2 hours. After the reaction solution was cooled to room temperature, ice water (10mL) was added and stirred for 10 minutes, extracted twice with ethyl acetate (20mL), the organic phase was washed twice with saturated sodium chloride solution (20mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography (Phenomenex Gemini-NX column, 3 μm silica, 40mm diameter, 80mm length, using a mixture of decreasing polarity of water (containing 0.225% formic acid) and acetonitrile as an eluent) to give 2-methyl-7- (3- (4- (S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridin-5-yl) -1,2,3, 4-tetrahydroisoquinoline (Compound 1) (33.45 mg).
LCMS m/z(ESI):417.0[M+H];
1H NMR(400MHz,METHANOL-d4)δ8.49-8.44(m,2H),8.06(d,J=8.4Hz,2H),7.95(d,J=8.6Hz,2H),7.89(s,1H),7.61(dd,J=1.6,7.9Hz,1H),7.50(s,1H),7.36(d,J=8.1Hz,1H),4.34(s,2H),3.45(t,J=6.2Hz,2H),3.23-3.20(m,5H),2.96(s,3H).
Example 2: synthesis of 5- (3, 5-dimethyl-4- (4- (tetrahydro-2H-pyran-4-yl) piperazin-1-yl) phenyl) -3- (4- (S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (Compound 2)
Figure BDA0003446981180000181
Compound 2-2:
Figure BDA0003446981180000182
5-bromo-3-iodo-1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine (2g, 4.19mmol) and compound 1-3(1.34g, 4.77mmol) were dissolved in acetonitrile (20mL) and aqueous sodium carbonate (1M, 20mL), dichlorobis (triphenylphosphine) palladium (II) (294.23mg, 419.19. mu. mol) was added at 25 ℃ and the reaction stirred at 60 ℃ for 3 hours. LCMS detected complete reaction of starting material. The reaction solution was added dropwise to water (40mL) at 25 ℃ and the aqueous phase was extracted three times with ethyl acetate (30 mL). The collected organic phase was dried over anhydrous magnesium sulfate, filtered and concentrated. The concentrated solution was purified by column chromatography (petroleum ether/tetrahydrofuran ═ 1/2) to give 5-bromo-3- (4- (S-methylsulphonimidoyl) phenyl) -1-toluenesulphonyl-1H-pyrrolo [2,3-b ] pyridine (compound 2-2) (800 mg).
LCMS m/z(ESI):504.1,506.1[M+H];
1H NMR(400MHz,METHANOL-d4)δ8.47-8.44(m,2H),8.28(s,1H),8.12-8.07(m,4H),7.94-7.92(m,2H),7.40-7.38(m,2H),3.19(s,3H),2.38(s,3H)
Compounds 2-3:
Figure BDA0003446981180000183
4-bromo-2, 6-dimethylaniline (2g, 10.00mmol) and bis (2-chloroethyl) amine hydrochloride (2.68g, 14.99mmol) were dissolved in xylene (11mL), p-toluenesulfonic acid (27.54mg, 159.94. mu. mol) was added to the reaction system, and the reaction mixture was stirred at 140 ℃ for 16 hours. LCMS monitored most of the reaction completion of the starting material. The reaction was cooled and concentrated, the concentrate was poured into ice water and adjusted to pH 8 with saturated sodium bicarbonate solution, extracted twice with ethyl acetate (50mL), the organic phase was washed twice with saturated sodium chloride solution (50mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure, and the residue was purified by column chromatography to give the product 1- (4-bromo-2, 6-dimethylphenyl) piperazine (compound 2-3) (0.67 g).
LCMS m/z(ESI):268.9,270.9[M+H];
Compounds 2-4:
Figure BDA0003446981180000191
1- (4-bromo-2, 6-dimethylphenyl) piperazine (2-3) (335mg, 1.24mmol) and tetrahydropyran-4-one (249.19mg, 2.49mmol) were dissolved in methanol (1mL), acetic acid (14.95mg, 248.90. mu. mol) and 2-methylpyridine borane complex (266.23mg, 2.49mmol) were added to the reaction system, and the reaction solution was stirred at 65 ℃ for 2 hours. LCMS monitored completion of the starting reaction. The reaction solution was cooled and concentrated, and the residue was purified by column chromatography (petroleum ether/tetrahydrofuran ═ 1/2) to give the product 1- (4-bromo-2, 6-dimethylphenyl) -4- (tetrahydro-2H-pyran-4-yl) piperazine (compound 2-4) (0.35 g).
LCMS m/z(ESI):353.2,355.2[M+H];
Compounds 2-5:
Figure BDA0003446981180000192
compounds 2-4(350mg, 990.67. mu. mol) were dissolved in dioxane (8mL), and pinacol ester diborate (503.14mg, 1.98mmol), potassium acetate (291.67mg, 2.97mmol) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane (40.45mg, 49.53. mu. mol) were added to the reaction system under nitrogen atmosphere, and replaced three times with nitrogen. The reaction mixture was stirred at 80 ℃ for 13 hours. LCMS monitor reaction completion. The reaction mixture was filtered, and the filtrate was concentrated to dryness under reduced pressure, followed by purification by column chromatography (petroleum ether/tetrahydrofuran ═ 1/2) to give the product 1- (2, 6-dimethyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -4- (tetrahydro-2H-pyran-4-yl) piperazine (compound 2-5) (300 mg).
LCMS m/z(ESI):401.4[M+H];
Compound 2:
Figure BDA0003446981180000201
5-bromo-3- (4- (S-methylsulphonimidoyl) phenyl) -1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine (2-2) (60mg, 118.95. mu. mol) and 1- (2, 6-dimethyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -4- (tetrahydro-2H-pyran-4-yl) piperazine (2-5) (52.38mg, 130.84. mu. mol) were dissolved in water (1mL) and 1, 4-dioxane (4mL), potassium phosphate (50.50mg, 237.90. mu. mol) and [1, 1-bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane (4.86 mg) were dissolved under a nitrogen atmosphere, 5.95. mu. mol) was added to the reaction system, and replaced with nitrogen three times. The reaction mixture was stirred at 100 ℃ for 10 hours. After completion of the reaction by LCMS detection, it was cooled to 25 ℃ and sodium hydroxide (95.16mg, 2.38mmol) and N, N-dimethylformamide (1mL) were added to the reaction. The reaction mixture was stirred at 100 ℃ for 16 hours. After the reaction solution was cooled to room temperature, ice water (3mL) was added and stirred for 10 minutes, extracted twice with ethyl acetate (5mL), the organic phase was washed twice with saturated sodium chloride solution (5mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography (YMC Triart C18 column, 7 μm silica, 50mm diameter, 250mm length; using a mixture of decreasing polarity of water (containing 0.05% aqueous ammonia) and acetonitrile as an eluent) to give 5- (3, 5-dimethyl-4- (4- (tetrahydro-2H-pyran-4-yl) piperazin-1-yl) phenyl) -3- (4- (S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (Compound 2) (6.04 mg). LCMS M/z (ESI) 544.2[ M + H ];
1H NMR(400MHz,DMSO-d6)δ8.54(d,J=1.8Hz,1H),8.44(d,J=2.0Hz,1H),8.09(s,1H),8.03-7.96(m,4H),7.38(s,2H),4.17(s,1H),3.92-3.90(m,2H),3.30-3.27(m,2H),3.10-3.06(m,7H),2.61(s,4H),2.45-2.42(m,1H),2.37(s,6H),1.76-1.73(m,2H),1.49-1.41(m,2H).
example 3: synthesis of 5- (3, 5-dimethyl-4- (4- (1-methylpiperidin-4-yl) piperazin-1-yl) phenyl) -3- (4- (S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (Compound 3)
Figure BDA0003446981180000202
Compound 3-2:
Figure BDA0003446981180000211
1- (4-bromo-2, 6-dimethylphenyl) piperazine (2-3) (335mg, 1.24mmol) and 1-methyl-4-piperidone (281.65mg, 2.49mmol) were dissolved in methanol (1mL), acetic acid (14.95mg, 248.90. mu. mol) and 2-methylpyridine borane complex (266.23mg, 2.49mmol) were added to the reaction system, and the reaction solution was stirred at 65 ℃ for 2 hours. LCMS monitored completion of the starting reaction. The reaction solution was cooled and concentrated, and the residue was purified by column chromatography (petroleum ether/tetrahydrofuran ═ 1/1) to give the product 1- (4-bromo-2, 6-dimethylphenyl) -4- (1-methylpiperidin-4-yl) piperazine (compound 3-2) (0.35 g).
LCMS m/z(ESI):366.2,368.2[M+H];
Compound 3-3:
Figure BDA0003446981180000212
compound 3-2(350mg, 955.40. mu. mol) was dissolved in dioxane (8mL), and pinacol ester diborate (485.23mg, 1.91mmol), potassium acetate (281.29mg, 2.87mmol) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane (39.01mg, 47.77. mu. mol) were added to the reaction system under a nitrogen atmosphere, and replaced three times with nitrogen. The reaction mixture was stirred at 80 ℃ for 13 hours. LCMS monitor reaction completion. The reaction mixture was filtered, and the filtrate was concentrated to dryness under reduced pressure, followed by purification by column chromatography (dichloromethane/methanol ═ 10/1) to give 1- (2, 6-dimethyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -4- (1-methylpiperidin-4-yl) piperazine (compound 3-3) (300 mg).
LCMS m/z(ESI):414.4[M+H];
Compound 3:
Figure BDA0003446981180000221
dissolving 5-bromo-3- (4- (S-methylsulphonimidoyl) phenyl) -1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine (2-2) (100mg, 198.25. mu. mol) and 1- (2, 6-dimethyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -4- (1-methylpiperidin-4-yl) piperazine (3-3) (90.15mg, 218.07. mu. mol) in water (1mL) and 1, 4-dioxane (4mL), under a nitrogen atmosphere, potassium phosphate (84.16mg, 396.50. mu. mol) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane (16.19mg, 19.82. mu. mol) was added to the reaction system, and replaced with nitrogen three times. The reaction mixture was stirred at 100 ℃ for 10 hours. After completion of the reaction by LCMS detection, it was cooled to 25 ℃ and sodium hydroxide (158.59mg, 3.96mmol) and N, N-dimethylformamide (2mL) were added to the reaction. The reaction was stirred at 100 ℃ for 16 hours. After the reaction solution was cooled to room temperature, ice water (3mL) was added and stirred for 10 minutes, extracted twice with ethyl acetate (10mL), the organic phase was washed twice with saturated sodium chloride solution (10mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography (YMC Triart C18 column, 7 μm silica, 50mm diameter, 250mm length; using a decreasing polarity mixture of water (containing 0.225% formic acid) and acetonitrile as an eluent) to give 5- (3, 5-dimethyl-4- (4- (1-methylpiperidin-4-yl) piperazin-1-yl) phenyl) -3- (4- (S-methylsulfonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (Compound 3) (5.34 mg).
LCMS m/z(ESI):557.5[M+H];
1H NMR(400MHz,DMSO-d6)δ12.20(s,1H),8.54(d,J=2.0Hz,1H),8.44(d,J=1.9Hz,1H),8.23(s,1H),8.10(d,J=1.5Hz,1H),8.03-7.96(m,4H),7.38(s,2H),3.10(s,3H),3.06(s,4H),2.92(br d,J=10.9Hz,2H),2.61(s,4H),2.37(s,6H),2.26-2.24(m,4H),2.08-2.02(m,2H),1.80-1.77(m,2H),1.52-1.50(m,2H).
Example 4: synthesis of 4- (2, 6-dimethyl-4- (3- (4- (S-methylsulfonylimino) phenyl) -1H-pyrrolo [2,3-b ] pyridin-5-yl) phenyl) morpholine (Compound 4)
Figure BDA0003446981180000231
Compound 4-2:
Figure BDA0003446981180000232
4-bromo-2, 6-dimethylaniline (1g, 5.00mmol) was dissolved in N, N-dimethylamide (15mL) at 20 deg.C, and potassium carbonate (3.45g,24.99mmol) and 1-chloro-2- (2-chloroethoxy) ethane (4.64g,19.99mmol) were added to the mixture. The reaction was stirred at 60 ℃ for 16 hours under nitrogen. LCMS showed the starting material had reacted to completion. After completion of the reaction, water (200mL) and ethyl acetate (50mL) were added to the reaction mixture, and after separation, the organic phase was washed 3 times with a 10% lithium chloride solution (300mL), and dried over anhydrous sodium sulfate. After filtration, the organic phase was concentrated under reduced pressure to remove the solvent, whereby 4- (4-bromo-2, 6-dimethylphenyl) morpholine (compound 4-2) (1.3g) was obtained.
LCMS m/z(ESI):270.2,272.0[M+H];
1HNMR(400MHz,CDCl3) δ 7.14(s,2H),3.75(d, J ═ 3.5Hz,4H),3.08-3.05(m,4H),2.31(s,6H).
Figure BDA0003446981180000233
Compound 4-2(500mg, 1.85mmol) was dissolved in dioxane (8mL) at 25 deg.C, and potassium acetate (544.90mg, 5.55mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (67.71mg, 92.54. mu. mol) and bis (pinacolato) borate (704.95mg,2.78mmol) were added to the mixture and reacted at 90 deg.C for 0.5 hour under nitrogen. LCMS monitor reaction completion. After completion of the reaction, water (100mL) and ethyl acetate (50mL) were added to the reaction mixture, and the mixture was extracted 2 times with water (100mL), and the organic phases were combined and dried over anhydrous sodium sulfate. After filtration, the organic phase is concentrated under reduced pressure to remove the solvent. The residue was purified by column chromatography (petroleum ether: ethyl acetate 1:0 to 5:1) to give 4- (2, 6-dimethyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) morpholine, compound 4-3 (270 mg).
LCMS m/z(ESI):318.1[M+H];
1H NMR(400MHz,METHANOL-d4)δ7.38(s,2H),3.82-3.78(m,4H),3.13-3.10(m,4H),2.36(s,6H),1.34(s,12H).
Compounds 4-4:
Figure BDA0003446981180000241
compound 4-3(45.28mg, 142.74. mu. mol) was dissolved in dioxane (2mL) and water (0.5mL) at 25 ℃, and potassium phosphate (37.87mg, 178.42. mu. mol), Pd (dtbpf) Cl, was added to the mixture2(2.91mg, 4.46. mu. mol) and Compound 2-2(45mg, 89.21. mu. mol), under NitrogenThe reaction is carried out for 16 hours at 100 ℃ under the protection of gas. TLC showed the reaction was complete. After completion of the reaction, water (100mL) and ethyl acetate (50mL) were added to the reaction mixture, and the mixture was extracted 2 times with water (100mL), and the organic phases were combined and dried over anhydrous sodium sulfate. After filtration, the organic phase is concentrated under reduced pressure to remove the solvent. The residue was purified by thin layer chromatography (dichloromethane: methanol ═ 10:1) to give the compound 4- (2, 6-dimethyl-4- (3- (4- (S-methylsulfonimido) phenyl) -1-toluenesulfonyl-1H-pyrrolo [2, 3-b)]Pyridin-5-yl) phenyl) morpholine (compound 4-4) (50 mg).
LCMS m/z(ESI):615.5[M+H];
1H NMR(400MHz,METHANOL-d4)δ8.39-8.35(m,1H),8.30(d,J=2.8Hz,1H),8.08-7.98(m,3H),7.71-7.67(m,4H),7.62-7.55(m,4H),3.86-3.79(m,4H),3.13(d,J=13.6Hz,4H),2.65-2.65(m,3H),2.45(s,3H),2.41(d,J=2.5Hz,3H),2.37(d,J=3.5Hz,3H).
Compound 4:
Figure BDA0003446981180000242
compound 4-4(50mg, 81.33. mu. mol) was dissolved in N, N-dimethylformamide (4.5mL) at room temperature, and 2M sodium hydroxide (16.26mg, 406.65. mu. mol) was added to the mixture and reacted at 80 ℃ for 4 hours under nitrogen. LCMS showed reaction complete. After completion of the reaction, the mixture was adjusted to pH 8 with 2N hydrochloric acid, water (100mL) and ethyl acetate (50mL) were added, and the organic phases were washed 2 times with water (100mL), combined and dried over anhydrous sodium sulfate. After filtration, the organic phase is concentrated under reduced pressure to remove the solvent. The residue was purified by preparative liquid chromatography (Phenomenex Gemini-NX C18 column: 5 μm silica, 30mm diameter, 150mm length, using a mixture of water (containing 0.05% ammonia) and acetonitrile of decreasing polarity (acetonitrile ratio: 38% -58%) as eluent) to give the compound 4- (2, 6-dimethyl-4- (3- (4- (S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridin-5-yl) phenyl) morpholine (compound 4) (0.9 mg).
LCMS m/z(ESI):461.1[M+H];
1H NMR(400MHz,METHANOL-d4)δ8.53-8.46(m,2H),8.14-8.09(m,2H),8.05-7.98(m,2H),7.93(s,1H),7.35(s,2H),3.89-3.80(m,4H),3.22(s,3H),3.19-3.15(m,4H),2.47(s,6H).
Example 5: synthesis of 3- (4- (cyclopropylsulfoximinoyl) phenyl) -5- (3, 5-dimethyl-4- (4-methylpiperazin-1-yl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (Compound 5)
Figure BDA0003446981180000251
Compound 5-2:
Figure BDA0003446981180000252
4-bromobenzenethiol (1g, 5.29mmol) and bromocyclopropane (959.76mg, 24.69. mu. mol) were dissolved in N, N-dimethylformamide (20mL), potassium carbonate (1.46g, 10.58mmol) was added thereto at 25 ℃ and the reaction mixture was stirred at 120 ℃ for 17 hours after three nitrogen replacements. LCMS monitor reaction completion. The reaction mixture was filtered, the filtrate was diluted with ethyl acetate (30mL) and water (30mL), the aqueous phase was extracted three times with ethyl acetate (60mL), the collected organic phases were dried over sodium sulfate and concentrated to dryness under reduced pressure, and purified by column chromatography (petroleum ether/ethyl acetate. 1/0 to 1/1) to give (4-bromophenyl) (cyclopropyl) sulfane (compound 5-2) (390 mg).
1H NMR(400MHz,CDCl3)δ7.43-7.40(m,1H),7.40-7.38(m,1H),7.26-7.23(m,1H),7.23-7.21(m,1H),2.20-2.14(m,1H),1.12-1.05(m,2H),0.72-0.67(m,2H).
Compound 5-3:
Figure BDA0003446981180000261
(4-bromophenyl) (cyclopropyl) sulfane (5-2) (390mg, 1.70mmol) was dissolved in ethanol (5mL), and ammonium acetate (524.77mg, 6.81mmol) and diacetoxyiodobenzene (1.64g, 5.11mmol) were added. After the nitrogen substitution was carried out three times, the reaction mixture was stirred at 25 ℃ for 5 hours. LCMS monitored reaction completion. The reaction mixture was filtered, the filtrate was diluted with ethyl acetate (30mL) and water (30mL), the aqueous phase was extracted three times with ethyl acetate (60mL), the collected organic phases were dried over sodium sulfate and concentrated to dryness under reduced pressure, and purified by column chromatography (petroleum ether/tetrahydrofuran ═ 1/1) to give 1-bromo-4- (cyclopropyliminoacyl) benzene (compound 5-3) (385 mg).
LCMS m/z(ESI):260.0,262.0[M+H];
1H NMR(400MHz,CDCl3)δ7.92-7.79(m,2H),7.74-7.61(m,2H),2.58-2.52(m,1H),1.48-1.35(m,2H),1.28-1.03(m,2H).
Compounds 5-4:
Figure BDA0003446981180000262
compound 5-3(375mg, 1.44mmol) and pinacol diboron (549.07mg, 2.16mmol) were dissolved in dioxane (5mL), 1-bis (diphenylphosphino) ferrocene palladium chloride (105.47mg, 144.15. mu. mol) and potassium acetate (282.93mg, 2.88mmol) were added, and the reaction mixture was stirred at 100 ℃ for 16 hours after nitrogen substitution three times. LCMS monitored reaction completion. The reaction solution was filtered, the filtrate was diluted with ethyl acetate (30mL) and water (30mL), the aqueous phase was extracted 3 times with ethyl acetate (60mL), the organic phase was dried over anhydrous magnesium sulfate, filtered, concentrated to dryness under reduced pressure, and purified by column chromatography (petroleum ether/ethyl acetate. RTM. 1/0 to 1/1 to 0/1) to give the product (4- (cyclopropylsulfoxido) phenyl) boronic acid pinacol ester (compound 5-4) (86 mg).
LCMS m/z(ESI):308.1[M+H];
Compounds 5-5:
Figure BDA0003446981180000271
compound 5-4(80mg, 260.40. mu. mol) and 5-bromo-3-iodo-1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine (124.24mg, 260.40. mu. mol) were dissolved in acetonitrile (0.8mL) and water (0.2mL), bis (triphenylphosphine) palladium dichloride (18.28mg, 26.04. mu. mol) and sodium carbonate (82.80mg, 781.21. mu. mol) were added thereto, the reaction mixture was replaced with nitrogen three times, and the reaction mixture was stirred at 60 ℃ for 3 hours. LCMS monitored reaction completion. The reaction solution was filtered, the filtrate was diluted with dichloromethane (30mL) and water (30mL), the aqueous phase was extracted 3 times with dichloromethane (60mL), the organic phase was dried over anhydrous sodium sulfate, filtered, concentrated to dryness under reduced pressure, and purified by prep-TLC (dichloromethane/methanol ═ 10/1) to give the product 5-bromo-3- (4- (cyclopropylsulfoximinato) phenyl) -1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine (compound 5-5) (50 mg).
LCMS m/z(ESI):530.0,532.0[M+H];
1H NMR(400MHz,CDCl3)δ8.53(d,J=2.1Hz,1H),8.22(d,J=2.1Hz,1H),8.10(dd,J=8.4,16.8Hz,4H),8.00(s,1H),7.72(d,J=8.4Hz,2H),7.33(d,J=8.3Hz,2H),2.41(s,3H),1.45(dd,J=6.8,10.5Hz,1H),1.30-1.20(m,2H),1.13-0.97(m,2H).
Compounds 5-6:
Figure BDA0003446981180000272
5-bromo-3- (4- (cyclopropylsulfoximidoyl) phenyl) -1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine (5-5) (40mg) and 1- (2, 6-dimethyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -4-methylpiperazine (37.36mg, 113.12. mu. mol) were dissolved in dioxane (0.4mL) and water (0.1mL), potassium phosphate (48.02mg, 226.23. mu. mol) and 1, 1-bis (diphenylphosphino) ferrocenylpalladium dichloride (5.52mg, 7.54. mu. mol) were added, and the reaction mixture was stirred at 100 ℃ for 16 hours after three nitrogen substitution. LCMS monitor reaction complete. The reaction solution was filtered, concentrated to dryness under reduced pressure and used directly in the next step.
LCMS m/z(ESI):654.3[M+H];
Compound 5:
Figure BDA0003446981180000281
the crude compound 5-6(40mg) obtained in the above reaction was dissolved in N, N-dimethylformamide (0.2mL) and water (0.2mL), and sodium hydroxide (122.35mg, 3.06mmol) was added thereto, and the reaction mixture was stirred at 80 ℃ for 3 hours after nitrogen substitution was carried out three times. LCMS monitored reaction completion. The reaction was filtered, the filtrate diluted with dichloromethane (20mL) and water (20mL), the aqueous phase extracted 3 times with dichloromethane (30mL), the organic phase dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the residue purified by preparative liquid chromatography (Phenomenex Gemini-NX column, 3 μm silica, 40mm diameter, 80mm length, using a decreasing polarity mixture of water (containing 0.05% ammonia) and acetonitrile as eluent) to give 3- (4- (cyclopropyliminoacyl) phenyl) -5- (3, 5-dimethyl-4- (4-methylpiperazin-1-yl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (compound 5) (9.6 mg).
LCMS m/z(ESI):500.2[M+H];
1H NMR(400MHz,DMSO-d6)δ12.18(s,1H),8.54(d,J=2.1Hz,1H),8.45(d,J=2.1Hz,1H),8.09(d,J=2.2Hz,1H),8.04-7.98(m,2H),7.97-7.89(m,2H),7.39(s,2H),4.16(s,1H),3.06(d,J=4.5Hz,4H),2.73-2.64(m,1H),2.48-2.42(m,4H),2.37(s,6H),2.30-2.25(m,3H),1.12(t,J=4.0,10.6Hz,1H),1.06-0.88(m,3H).
Example 6: synthesis of 2-methyl-7- (3- (4- (N, S-dimethylsulfonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridin-5-yl) -1,2,3, 4-tetrahydroisoquinoline (Compound 6)
Figure BDA0003446981180000282
Compound 6-2:
Figure BDA0003446981180000291
compound 1-2(200mg, 854.29. mu. mol) was dissolved in 1, 4-dioxane (7mL), and copper acetate (232.75mg, 1.28mmol) and pyridine (162.18mg,2.05mmol) were added to the reaction system. The reaction was stirred at 25 ℃ for 0.2 h. To the reaction system was added methylboronic acid (102.28mg, 1.71mmol), and the reaction was stirred at 100 ℃ for 1 hour. Upon completion of the reaction by LCMS, the reaction was cooled and concentrated, and the residue was purified by column chromatography (petroleum ether/ethyl acetate: 2/1) to give the product 1-bromo-4- (N, S-dimethylsulfonimidoyl) benzene (compound 6-2) (0.2 g).
LCMS m/z(ESI):248.0,250.0[M+H];
Compound 6-3:
Figure BDA0003446981180000292
compound 6-2(200mg, 806.00. mu. mol) was dissolved in dioxane (3mL), and pinacol ester diborate (307.01mg,1.21mmol), potassium acetate (237.30mg, 2.42mmol) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane (32.91mg, 40.30. mu. mol) were added to the reaction system under a nitrogen atmosphere, and replaced three times with nitrogen. The reaction mixture was stirred at 80 ℃ for 10 hours. LCMS monitor reaction completion. The reaction mixture was filtered, and the filtrate was concentrated to dryness under reduced pressure, followed by purification by column chromatography (dichloromethane/methanol-10/1) to give 4,4,5, 5-tetramethyl-2- (4- (N, S-dimethylsulfonimidoyl) phenyl) -1,3, 2-dioxaborolan (compound 6-3) (200 mg).
LCMS m/z(ESI):296.2[M+H];
Compound 6:
Figure BDA0003446981180000301
7- (3-iodo-1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridin-5-yl) -2-methyl-1, 2,3, 4-tetrahydroisoquinoline (compound 1-7) (100mg, 184.02. mu. mol) was dissolved in water (0.8mL) and N, N-dimethylformamide (3.2mL), and compounds 6-3(78.41mg, 265.60. mu. mol), cesium carbonate (179.87mg, 552.06. mu. mol) and 1, 1-bis (di-tert-butylphosphine) ferrocene dichloropalladium (11.99mg, 18.40. mu. mol) were added to the reaction system under a nitrogen atmosphere and replaced three times with nitrogen. The reaction mixture was stirred at 100 ℃ for 10 hours. After completion of the reaction by LCMS detection, it was cooled to 25 ℃ and sodium hydroxide solution (4M, 920.10. mu.L) was added to the reaction. The reaction mixture was stirred at 80 ℃ for 2 hours. After the reaction solution was cooled to room temperature, ice water (10mL) was added and stirred for 10 minutes, extracted twice with ethyl acetate (20mL), the organic phase was washed twice with saturated sodium chloride solution (20mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure, and the residue was purified by preparative liquid chromatography (Agela DuraShell C18 column, 5 μm silica, 25mm diameter, 150mm length; using a mixture of decreasing polarity of water (containing 0.05% ammonia) and acetonitrile as an eluent) to give 2-methyl-7- (3- (4- (N, S-dimethylsulfoximinato) phenyl) -1H-pyrrolo [2,3-b ] pyridin-5-yl) -1,2,3, 4-tetrahydroisoquinoline (Compound 6) (36.5 mg). LCMS M/z (ESI) 431.1[ M + H ];
1H NMR(400MHz,METHANOL-d4)δ8.53-8.51(m,2H),8.05-8.03(m,2H),7.98-7.96(m,2H),7.94(s,1H),7.53-7.50(m,1H),7.44(s,1H),7.28(d,J=8.0Hz,1H),3.74(s,2H),3.21(s,3H),3.04–3.01(m,2H),2.83-2.80(m,2H),2.66(s,3H),2.51(s,3H).
example 7: synthesis of 2-methyl-7- (3- (4- (N-cyclopropyl-S-methylsulfonylimino) phenyl) -1H-pyrrolo [2,3-b ] pyridin-5-yl) -1,2,3, 4-tetrahydroisoquinoline (Compound 7)
Figure BDA0003446981180000302
Compound 7:
Figure BDA0003446981180000311
2-methyl-7- (3- (4- (S-methylsulfonimidoyl) phenyl) -1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridin-5-yl) -1,2,3, 4-tetrahydroisoquinoline (Compound 1-8) (60mg, 105.13. mu. mol) was dissolved in 1, 4-dioxane (5mL), and copper acetate (38.19mg, 210.26. mu. mol) and pyridine (19.96mg, 252.31. mu. mol) were added to the reaction system. The reaction was stirred at 25 ℃ for 0.2 h. To the reaction system was added cyclopropylboronic acid (18.06mg, 210.26. mu. mol), and the reaction was stirred at 100 ℃ for 1 hour. After completion of the LCMS detection reaction, cooled to 25 ℃ and concentrated, to the concentrate was added sodium hydroxide (84.10mg, 2.10mmol), water (2mL) and N, N-dimethylformamide (2 mL). The reaction solution was stirred at 100 ℃ for 13 hours. After LCMS detection reaction is completed, the reaction solution is cooled to room temperature, then ice water (10mL) is added and stirred for 10 min, extracted twice with ethyl acetate (10mL), the organic phase is washed twice with saturated sodium chloride solution (10mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure, and the residue is purified by preparative liquid chromatography (Agela DuraShell C18 column, 5 μm silica, 25mm diameter, 150mm length; using a mixture of decreasing polarity of water (containing 0.05% ammonia) and acetonitrile as eluent) to give 2-methyl-7- (3- (4- (N-cyclopropyl-S-methylsulfonimido) phenyl) -1H-pyrrolo [2,3-b ] pyridin-5-yl) -1,2,3, 4-tetrahydroisoquinoline (Compound 7) (3.04 mg).
LCMS m/z(ESI):457.2[M+H];
1H NMR(400MHz,METHANOL-d4)δ8.52-8.51(m,2H),8.04(d,J=1.3Hz,4H),7.94(s,1H),7.53-7.51(m,1H),7.44(s,1H),7.28(d,J=7.8Hz,1H),3.75(s,2H),3.22(s,3H),3.04-3.01(m,2H),2.84-2.81(m,2H),2.52(s,3H),2.47-2.43(m,1H),0.59-0.44(m,4H).
Example 8: synthesis of 5- (3, 5-dimethyl-4- (4- (tetrahydro-2H-pyran-4-yl) piperazin-1-yl) phenyl) -3- (4- (N-cyclopropyl-S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (Compound 8)
Figure BDA0003446981180000312
Compound 8-2:
Figure BDA0003446981180000321
compound 1-2(200mg, 854.29. mu. mol) was dissolved in dioxane (7mL), copper acetate (232.75mg, 1.28mmol) was added to the reaction, after stirring open to the atmosphere for half an hour at 25 ℃, cyclopropylboronic acid (146.76mg,1.71mmol) and pyridine (162.18mg,2.05mmol) were added, and the temperature was raised to 100 ℃ for 1 hour. After completion of the reaction was monitored by LCMS, the reaction solution was filtered and concentrated, and the concentrate was purified by column chromatography (petroleum ether/tetrahydrofuran ═ 1/1) to give the product 1-bromo-4- (N-cyclopropyl-S-methylsulphonimidoyl) benzene (compound 8-2) (230 mg).
LCMS m/z(ESI):273.9,275.9[M+H];
1H NMR(400MHz,METHANOL-d4)δ8.05-7.58(m,4H),3.20-3.12(m,3H),2.39-2.31(m,1H),0.65-0.21(m,4H).
Compound 8-3:
Figure BDA0003446981180000322
compound 8-2(230mg, 838.87. mu. mol) was dissolved in dioxane (4mL), and pinacol ester diboron (426.04mg,1.68mmol) and potassium acetate (246.99mg,2.52mmol), 1, 1-bis (diphenylphosphino) ferrocene palladium chloride dichloromethane complex (34.25mg, 41.94. mu. mol) were added to the reaction system under nitrogen atmosphere, and replaced three times with nitrogen. The reaction mixture was stirred at 80 ℃ for 1 hour. LCMS monitor reaction completion. The reaction mixture was concentrated by filtration and purified by column chromatography (petroleum ether/tetrahydrofuran: 1/1) to give 4,4,5, 5-tetramethyl-2- (4- (N-cyclopropyl-S-methylsulphonimidoyl) phenyl) -1,3, 2-dioxaborolan (compound 8-3) (167 mg).
LCMS m/z(ESI):322.1[M+H];
1H NMR(400MHz,METHANOL-d4)δ8.01-7.91(m,4H),3.18-3.14(m,3H),1.41-1.35(m,12H),0.60-0.24(m,5H).
Compounds 8-4:
Figure BDA0003446981180000331
compound 8-3(153.51mg) was dissolved in acetonitrile (2mL) and 1M sodium carbonate solution (2mL), and 5-bromo-3-iodo-1- (p-toluenesulfonyl) pyrrolo [2,3-b ] pyridine (200mg, 419.19. mu. mol) and dichlorobis (triphenylphosphine) palladium (29.42mg, 41.92. mu. mol) were added to the reaction system under a nitrogen atmosphere, and replaced with nitrogen three times. The reaction was stirred at 60 ℃ for 15 hours. LCMS check reaction completion. The reaction mixture was concentrated by filtration and purified by column chromatography (petroleum ether/tetrahydrofuran: 1/1) to give 5-bromo-3- (4- (N-cyclopropyl-S-methylsulphonimidoyl) phenyl) -1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine (compound 8-4) (120 mg).
LCMS m/z(ESI):544.2,546.0[M+H];
1H NMR(400MHz,METHANOL-d4)δ8.49-8.46(m,1H),8.32(s,1H),8.11-8.04(m,3H),8.00-7.93(m,2H),7.75-7.60(m,1H),7.75-7.60(m,1H),7.40(d,J=8.0Hz,2H),3.21(s,2H),3.16(s,1H),2.45-2.32(m,3H),0.60-0.35(m,1H),0.60-0.35(m,3H).
Compounds 8-5:
Figure BDA0003446981180000332
compound 8-4(60mg, 110.20. mu. mol) was dissolved in dioxane (3.2mL) and water (0.8mL), and compound 2-5(57.35mg, 143.26. mu. mol), potassium phosphate (46.78mg, 220.39. mu. mol) and 1, 1-bis (diphenylphosphino) ferrocene palladium dichloride dichloromethane complex (9.00mg, 11.02. mu. mol) were added to the reaction system under a nitrogen atmosphere, and replaced with nitrogen three times. The reaction was stirred at 100 ℃ for 15 hours. LCMS check reaction completed. The reaction mixture was concentrated by filtration to give 5- (3, 5-dimethyl-4- (4- (tetrahydro-2H-pyran-4-yl) piperazin-1-yl) phenyl) -3- (4- (N-cyclopropyl-S-methylsulphonimidoyl) phenyl) -1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine (compound 8-5) (60 mg).
LCMS m/z(ESI):370.0[M/2+H];
Compound 8:
Figure BDA0003446981180000341
compound 8-5(60mg, 81.30. mu. mol) was dissolved in N, N-dimethylformamide (2mL) and water (0.5mL), and sodium hydroxide (65.04mg,1.63mmol) was added. The reaction was stirred at 80 ℃ for 16 hours. After completion of the reaction by LCMS, water (10mL) and methylene chloride (40mL) were added to the system, and extracted 3 times with dichloromethane (5mL), dried over anhydrous magnesium sulfate, filtered and concentrated, and the residue purified by preparative liquid chromatography (Phenomenex Gemini-NX column, 3 μm silica, 30mm diameter, 80mm length, using a decreasing polarity mixture of water (containing 0.225% formic acid) and acetonitrile as eluent) to give the compound 5- (3, 5-dimethyl-4- (4- (tetrahydro-2H-pyran-4-yl) piperazin-1-yl) phenyl) -3- (4- (N-cyclopropyl-S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (compound 8) (27.49 mg).
LCMS m/z(ESI):584.5[M+H];
1H NMR(400MHz,DMSO-d6)δ12.19(d,J=2.2Hz,1H),8.53(d,J=2.1Hz,1H),8.45(d,J=2.0Hz,1H),8.17(s,1H),8.12(d,J=2.6Hz,1H),8.05(d,J=8.4Hz,2H),7.92(d,J=8.6Hz,2H),7.38(s,2H),3.91(dd,J=3.7,10.6Hz,2H),3.27(br s,2H),3.14(s,3H),3.05(d,J=4.5Hz,4H),2.68-2.55(m,4H),2.48-2.42(m,1H),2.37(s,5H),2.34-2.27(m,1H),1.74(d,J=13.9Hz,2H),1.47-1.43(m,2H),0.44-0.21(m,4H).
Example 9: synthesis of 5- (3, 5-dimethyl-4- (4- (1-methylpiperidin-4-yl) piperazin-1-yl) phenyl) -3- (4- (N-cyclopropyl-S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (Compound 9)
Figure BDA0003446981180000342
Compound 9-2:
Figure BDA0003446981180000351
compounds 8-4(240mg, 440.79 μmol) and 1- (2, 6-dimethyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -4- (1-methylpiperidin-4-yl) piperazine (compound 3-3) (364.44mg, 881.57 μmol) were dissolved in water (0.4mL) and dioxane (1.6mL), and potassium phosphate (280.70mg, 1.32mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (32.25mg, 44.08 μmol) were added. After the nitrogen gas was replaced three times, the reaction mixture was stirred at 100 ℃ for 16 hours. LCMS check reaction complete. The reaction mixture was concentrated to dryness under reduced pressure to give 5- (3, 5-dimethyl-4- (4- (1-methylpiperidin-4-yl) piperazin-1-yl) phenyl) -3- (4- (N-cyclopropyl-S-methylsulphonimidoyl) phenyl) -1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine (compound 9-2) (360 mg).
LCMS m/z(ESI):751.6[M+H];
1H NMR(400MHz,CDCl3)δ8.68(d,J=1.8Hz,1H),8.17(d,J=7.4Hz,2H),8.06(d,J=8.4Hz,1H),8.01(s,1H),7.82(d,J=8.3Hz,2H),7.64-7.41(m,2H),7.39-7.29(m,2H),7.25-7.09(m,2H),3.17(s,3H),3.14(s,3H),3.03(s,2H),2.80-2.59(m,4H),2.49-2.26(m,12H),2.20-2.01(m,2H),2.00-1.84(m,2H),1.84-1.63(m,2H),1.35-1.31(m,1H),1.26(s,1H),1.25-1.21(m,1H),0.97-0.57(m,2H),0.55-0.39(m,2H).
Compound 9:
Figure BDA0003446981180000352
compound 9-2(100mg, 133.15. mu. mol) was dissolved in water (2mL) and N, N-dimethylformamide (2mL), and sodium hydroxide (106.52mg, 2.66mmol) was added. After the nitrogen substitution was carried out three times, the reaction mixture was stirred at 80 ℃ for 16 hours. LCMS check reaction complete. To the reaction mixture were added water (30mL) and dichloromethane (15 mL). Then extracted three times with dichloromethane (30mL), the organic phase dried over anhydrous sodium sulfate and concentrated to dryness under reduced pressure, then purified by preparative liquid chromatography (Phenomenex Gemini-NX C18 column: 3 μm silica, 40mm diameter, 80mm length; using as eluent a mixture of water (containing 0.225% formic acid) and decreasing polarity of acetonitrile (acetonitrile ratio: 5% -45%), to give 5- (3, 5-dimethyl-4- (4- (1-methylpiperidin-4-yl) piperazin-1-yl) phenyl) -3- (4- (N-cyclopropyl-S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (Compound 9) (10 mg).
LCMS m/z(ESI):597.5[M+H];
1H NMR(400MHz,METHANOL-d4)δ12.22(s,1H),8.54(d,J=2.0Hz,1H),8.46(d,J=2.0Hz,1H),8.24(s,1H,FA),8.13(s,1H),8.06(d,J=8.5Hz,2H),7.93(d,J=8.5Hz,2H),7.38(s,2H),3.15(s,3H),3.11-3.04(m,4H),2.91(d,J=10.8Hz,2H),2.65-2.54(m,4H),2.37(s,6H),2.34-2.26(m,2H),2.24(s,3H),2.08-1.99(m,2H),1.82-1.72(m,2H),1.56-1.39(m,1H),0.45-0.22(m,4H).
Example 10: synthesis of 5- (3, 5-dimethyl-4- (4- (tetrahydro-2H-pyran-4-yl) piperazin-1-yl) phenyl) -3- (4- (N-cyclopentyl-S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (Compound 10)
Figure BDA0003446981180000361
Compound 10-2:
Figure BDA0003446981180000362
compound 1-2(12.33g,52.65mmol) was dissolved in dimethyl sulfoxide (30mL), potassium hydroxide (232.75mg, 1.28mmol) and bromocyclopentane (3g, 20.13mmol) were added to the reaction, and the mixture was left to stir at 25 ℃ for 16 hours. LCMS monitor reaction completion. To the reaction mixture were added water (30mL) and ethyl acetate (25mL), and the mixture was extracted three times with ethyl acetate (20mL), and the organic phases were combined, dried over anhydrous magnesium sulfate, filtered, and concentrated to give a concentrate. The concentrated solution was purified by column chromatography (petroleum ether/ethyl acetate: 1/1) to give the product 1-bromo-4- (N-cyclopentyl-S-methylsulphonimidoyl) benzene (compound 10-2) (795 mg).
LCMS m/z(ESI):302.0,304.0[M+H];
1H NMR(400MHz,METHANOL-d4)δ7.85-7.77(m,4H),3.14(s,3H),1.84-1.29(m,9H).
The remaining procedures referring to the synthesis of example 8 (compound 8), 1-bromo-4- (N-cyclopropyl-S-methylsulphonimidoyl) benzene (compound 8-2) was replaced with 1-bromo-4- (N-cyclopentyl-S-methylsulphonimidoyl) benzene (compound 10-2), the target product, 5- (3, 5-dimethyl-4- (4- (tetrahydro-2H-pyran-4-yl) piperazin-1-yl) phenyl) -3- (4- (N-cyclopentyl-S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (compound 10) (7.33mg) was obtained.
LCMS m/z(ESI):612.6[M+H];
1H NMR(400MHz,DMSO-d6)δ12.19(d,J=2.3Hz,1H),8.53(d,J=2.0Hz,1H),8.45(d,J=2.0Hz,1H),8.12(d,J=2.6Hz,1H),8.04(d,J=8.4Hz,2H),7.88(d,J=8.6Hz,2H),7.38(s,2H),3.91(dd,J=3.7,10.3Hz,2H),3.30-3.26(m,2H),3.11(s,3H),3.05(br s,4H),2.61(br s,4H),2.48-2.46(m,1H),2.37(s,6H),1.78-1.58(m,6H),1.52-1.31(m,7H).
Example 11: synthesis of 5- (3, 5-dimethyl-4- (4-methylpiperazin-1-yl) phenyl) -3- (4- (N-cyclopropyl-S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (Compound 11)
Figure BDA0003446981180000371
Compound 11-2:
Figure BDA0003446981180000372
compounds 8 to 4(60mg,110.20 μmol) were dissolved in dioxane (3.2mL) and water (0.8mL), and 1- (2, 6-dimethyl-4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -4-methylpiperazine (47.31mg,143.26 μmol), potassium phosphate (46.78mg,220.40 μmol) and [1, 1-bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane (9.00mg,11.02 μmol) were added to the reaction system under a nitrogen atmosphere, and replaced three times with nitrogen. The reaction was stirred at 100 ℃ for 15 h and LCMS checked for completion. The reaction mixture was concentrated by filtration to give a crude product, 5- (3, 5-dimethyl-4- (4-methylpiperazin-1-yl) phenyl) -3- (4- (N-cyclopropyl-S-methylsulphonimidoyl) phenyl) -1-toluenesulfonyl-1H-pyrrolo [2,3-b ] pyridine (compound 11-2) (60 mg).
LCMS m/z(ESI):668.5[M+H];
Compound 11:
Figure BDA0003446981180000381
crude compound 11-2(60mg, 89.84. mu. mol) was dissolved in N, N-dimethylformamide (2mL) and water (0.5mL), and sodium hydroxide (71.86mg,1.80mmol) was added. The reaction was stirred at 80 ℃ for 16 hours. After completion of the reaction by LCMS, water (10mL) and dichloromethane (40mL) were added to the system and extracted 3 times with dichloromethane (5 mL). The organic phases were combined and dried over anhydrous magnesium sulphate, filtered and concentrated, and the residue was purified by preparative liquid chromatography (Phenomenex Gemini-NX column, 3 μm silica, 30mm diameter, 80mm length, using a decreasing polarity mixture of water (containing 0.225% formic acid) and acetonitrile as eluent) to give the product 5- (3, 5-dimethyl-4- (4-methylpiperazin-1-yl) phenyl) -3- (4- (N-cyclopropyl-S-methylsulphonimidoyl) phenyl) -1H-pyrrolo [2,3-b ] pyridine (compound 11) (12.12 mg).
LCMS m/z(ESI):514.4[M+H];
1H NMR(400MHz,DMSO-d6)δ12.19(br s,1H),8.53(d,J=2.0Hz,1H),8.45(d,J=2.0Hz,1H),8.17(s,1H),8.11(d,J=2.7Hz,1H),8.04(d,J=8.6Hz,2H),7.91(d,J=8.4Hz,2H),7.38(s,2H),3.13(s,3H),3.08-3.03(m,4H),2.46(br d,J=3.8Hz,4H),2.36(s,6H),2.33-2.28(m,1H),2.26(s,3H),0.44-0.24(m,4H).
With reference to the synthetic methods of the above examples, the following compounds were also synthesized, the structures and characterization data of which are:
Figure BDA0003446981180000382
Figure BDA0003446981180000391
examples of biological Activity and related Properties
The compounds in the following test examples were prepared according to the methods of the above examples of the present invention.
Test example 1: assay for HPK1 kinase Activity
Experimental materials:
HPK1(MAP4K1)35948 was purchased from Signalchem, # M23-11G
MBP35951 is available from Signalchem, # M42-51N
ADP-GLO from Promega, # V9102
DMSO was purchased from Sigma
384-well assay plate available from Perkin Elmer #6007290
384-well assay plate available from LABCYTE
MgCl2, MnCl2, DTT, Tween-20, HEPES, BSA were purchased from Sigma
An experimental instrument:
nano-scale acoustic pipetting systems:
Figure BDA0003446981180000402
LIQUID HANDLERS(LABCYTE,USA)
multi-label detection analyzer: envision Multilabel Reader (PerkinElmer, USA)
The experimental method comprises the following steps:
the test adopts a kinase detection mode (ADP-Glo) developed by Promega corporationTM) The synthesized compound was tested for its inhibitory activity against HPK1 kinase. The specific method comprises the following steps: compounds were diluted with ECHO650 gradients and transferred to reaction plates (384-well white plate, Perkin Elmer #6007290) at 50 nL/well with a final compound starting concentration of 100nM, 3-fold gradient dilution, 10 concentration points; HPK1 kinase reaction buffer (50mM HEPES (pH 7.5), 0.01% Tween-20,5mM MgCl20.01% BSA and 0.05mM DTT) to an appropriate concentration, 3. mu.l of enzyme (final concentration 50nM) or enzyme reaction buffer was added to each reaction well, the reaction plate was placed in a centrifuge, centrifuged at 1000 rpm for 30 seconds, and incubated on ice for 30 minutes. 2. mu.l/well 2.5 XATP (62.5. mu.M)/substrate mix (250. mu.g/mL) was added, centrifuged at 1000 rpm for 30 seconds, and incubated at room temperature for 60 minutes. Add 5. mu.L/well ADP-Glo and mix well, react for 40 minutes at room temperature. Assay substrate was added, 10. mu.L/well, and incubated for 30 minutes at room temperature. The chemiluminescent signal was read in a microplate reader (Envision, Perkin Elmer). The inhibition rate of the test compound was calculated according to the formula (n ═ 2): the inhibition ratio [% ], [% ] is the maximum signal value. The maximum signal value is the reading value of the enzyme reaction activity strongest hole only containing DMSO; the minimum signal value is the reading of the reaction well without enzyme. Data were imported into MS Excel and curve-fitted using XLFit Excel add-in version 5.4.0.8: y ═ Bottom + (Top-Bottom)/(1+ (IC50/X) ^ HillSlope), and IC is calculated according to a fitted curve50
The test results are shown in Table 1.
TABLE 1 enzymatic inhibitory Activity of HPK1
Figure BDA0003446981180000401
Figure BDA0003446981180000411
Test example 2: t cell activation assay
Experimental materials:
jurkat T cells purchased from ATCC
RPMI1640 ex Gibco (ThermoFisher, USA)
FBS is available from Gibco (ThermoFisher, USA)
DMSO was purchased from Sigma
anti-CD 3 monoclonal antibody (OKT3) was purchased from BD Biosciences #566685
anti-CD 28 monoclonal antibody (CD28.2) was purchased from BD Biosciences #555725
Human IL-2ELISA detection kit purchased from BD Biosciences #555190
96-well culture plate from Corning
An experimental instrument:
CO2a cell culture box: ThermoFisher (USA)
Multi-label detection analyzer: envision Multilabel Reader (PerkinElmer, USA)
Cell counting instrument: Vi-CELL (Beckman, USA)
The experimental method comprises the following steps:
coating an anti-CD 3 antibody on a 96-well cell culture plate for pretreatment, diluting an anti-CD 3 antibody to 2 mu g/mL by using PBS, adding 100 mu L of the anti-CD 3 antibody into each well, incubating for 4 hours at 37 ℃, washing for 1-2 times by using the PBS, and spin-drying for later use; jurkat T cells were collected, counted using a cytometer, and cell density adjusted to 1 × 10 per well5Cell counting plates. Dissolving the synthesized compound by DMSO, performing gradient dilution, adding into each corresponding hole of a cell culture plate, controlling the final concentration of the DMSO to be below 0.1%, and controlling the initial treatment concentration of the final compound to be 10 mu M, performing 3-fold gradient dilution, and obtaining 8 concentration points; t cells were preincubated with compound at 37 ℃ for 1 hour. The incubated T cells were then transferred to a CD3 antibody coated cell culture plate at 100. mu.L/well for cell number1x105(ii) a Adding anti-CD 28 antibody to the solution to a final concentration of 1 μ g/mL; the cell culture plate was placed in a carbon dioxide incubator for 48 hours. And taking cell culture supernatant, diluting the cell culture supernatant in a proper amount, and detecting the content of the human IL-2 in the cell culture supernatant by adopting an ELISA method. And carrying out quantitative conversion according to the amount of the standard substance. Fold count of IL-2 production: fold-IL-2 production/minimum IL-2 production, DMSO blank treated wells. Maximum effect refers to the highest fold production of IL-2 under drug treatment. The test results are shown in Table 2.
TABLE 2 results of T cell activation experiments
Figure BDA0003446981180000421

Claims (12)

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure FDA0003446981170000011
wherein,
ring Q is selected from phenyl, 5-6 membered heteroaryl or 4-7 membered heterocyclyl;
R1selected from H or optionally substituted by Ra1Substituted of the following groups: c1-C6Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl;
R2selected from the group consisting ofa2Substituted of the following groups: c1-C6Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl, or R1、R2Together with the atoms to which they are attached form a 5-7 membered heterocyclyl;
L1is absent, or L1Is selected from C1-C6Alkylene, (C)1-C3Alkylene) -O- (C)1-C3Alkylene) or (C)1-C3Alkylene) -NH- (C)1-C3Alkylene groups);
R3is absent, or, R3Selected from H or optionally substituted by Ra3Substituted of the following groups: OH, NH2、C1-C6Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl, or R1、R3Together with the atom to which they are attached form a 5-7 membered heterocyclic group, or R2、R3Together with the atoms to which they are attached form a 5-7 membered heterocyclyl;
R4selected from H, halogen, CN or optionally substituted by Ra4Substituted of the following groups: OH, NH2、C1-C6Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl;
R5、R6independently selected from H, halogen, CN, C1-C6Alkyl or C1-C6Alkoxy radical, said C1-C6Alkyl or C1-C6Alkoxy is optionally substituted by F, Cl, Br, I, CN;
ring A is selected from
Figure FDA0003446981170000012
R7Selected from the group consisting ofa7A substituted 4-14 membered heterocyclyl;
X1、X4independently selected from CR9Or N;
X2、X3independently selected from C (R)10)(R11) Or NR12
R8、R9、R10、R11Independently selected from H, halogen, CN or optionally Ra8Substituted of the following groups: OH, NH2、C1-C6Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl;
R12selected from H or optionally substituted by Ra12Substituted of the following groups: c1-C6Alkyl radical, C3-C6Cycloalkyl or 4-7 membered heterocyclyl;
p, q are independently selected from 0, 1,2,3 or 4;
m and n are independently selected from 0, 1 or 2;
each Ra1、Ra2、Ra3、Ra4、Ra7、Ra8、Ra12Independently selected from halogen, CN, ═ O or optionally RbSubstituted of the following groups: OH, NH2、C1-C3Alkyl radical, C3-C6Cycloalkyl, 4-7 membered heterocyclyl;
each RbIndependently selected from halogen, CN, ═ O, C1-C3Alkyl, OH, O (C)1-C3Alkyl), NH2、NH(C1-C3Alkyl group), N (C)1-C3Alkyl radical)2
2. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein R is1Selected from H or optionally substituted by Ra1Substituted of the following groups: c1-C3Alkyl radical, C3-C6A cycloalkyl group.
3. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein R is2Selected from the group consisting ofa2Substituted of the following groups: c1-C3Alkyl radical, C3-C6Cycloalkyl, or R1、R2Together with the atoms to which they are attached form a 5-6 membered heterocyclic group.
4. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein R is3Is absent, or R3Selected from H, or R1、R3Together with the atom to which they are attached form a 5-6 membered heterocyclic group, or R2、R3Together with the atoms to which they are attached form a 5-6 membered heterocyclic group.
5. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein the structural unit is a moiety
Figure FDA0003446981170000021
Is selected from
Figure FDA0003446981170000022
6. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein the structural unit is a moiety
Figure FDA0003446981170000023
Is selected from
Figure FDA0003446981170000024
Figure FDA0003446981170000025
7. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein R is5、R6Independently selected from H, halogen, CN or C1-C3An alkyl group.
8. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein R is7Selected from the group consisting ofa7Substituted 6-8 membered heterocyclyl, Ra7Selected from the group consisting ofbSubstituted of the following groups: c1-C3Alkyl, 5-6 membered heterocyclyl.
9. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein R is8Selected from H or optionally substituted by Ra8Substituted of the following groups: c1-C3Alkyl, 5-membered heterocyclic group.
10. The compound of formula (I) or a pharmaceutically acceptable salt thereof according to claim 1, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is selected from the following compounds or pharmaceutically acceptable salts thereof:
Figure FDA0003446981170000031
Figure FDA0003446981170000041
11. a pharmaceutical composition comprising a compound of any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable adjuvant.
12. Use of a compound of any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 11, for the manufacture of a medicament for the prevention or treatment of a HPK 1-related disease.
CN202111659383.XA 2020-12-31 2021-12-30 Polysubstituted nitrogen-containing heterocyclic compound and application thereof Pending CN114685489A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011637920 2020-12-31
CN2020116379206 2020-12-31

Publications (1)

Publication Number Publication Date
CN114685489A true CN114685489A (en) 2022-07-01

Family

ID=82137262

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111659383.XA Pending CN114685489A (en) 2020-12-31 2021-12-30 Polysubstituted nitrogen-containing heterocyclic compound and application thereof

Country Status (1)

Country Link
CN (1) CN114685489A (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11897878B2 (en) 2018-10-31 2024-02-13 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds
US11925631B2 (en) 2018-10-31 2024-03-12 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds
US12037342B2 (en) 2019-05-23 2024-07-16 Gilead Sciences, Inc. Substituted eneoxindoles and uses thereof

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110396087A (en) * 2018-04-25 2019-11-01 珠海宇繁生物科技有限责任公司 HPK1 kinase inhibitor, preparation method and applications
CN110402248A (en) * 2017-03-15 2019-11-01 豪夫迈·罗氏有限公司 Azaindoles as HPK1 inhibitor
WO2019238067A1 (en) * 2018-06-13 2019-12-19 Beigene, Ltd. Pyrrolo [2, 3-b] pyridines or pyrrolo [2, 3-b] pyrazines as hpk1 inhibitor and the use thereof
US20200108075A1 (en) * 2018-10-03 2020-04-09 Genentech, Inc. 8-aminoisoquinoline compounds and uses thereof
WO2020103896A1 (en) * 2018-11-22 2020-05-28 Beigene, Ltd. Pyrrolo[2,3-b]pyridines as hpk1 inhibitor and uses thereof
CN115433184A (en) * 2021-06-04 2022-12-06 武汉人福创新药物研发中心有限公司 HPK1 inhibitor and application thereof
CN116162087A (en) * 2023-02-20 2023-05-26 杭州医学院 HPK 1 and/or LCK kinase regulator, preparation method and application thereof
CN116848113A (en) * 2021-02-10 2023-10-03 南京再明医药有限公司 Bicyclic compounds as HPK1 inhibitors and application thereof

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110402248A (en) * 2017-03-15 2019-11-01 豪夫迈·罗氏有限公司 Azaindoles as HPK1 inhibitor
CN110396087A (en) * 2018-04-25 2019-11-01 珠海宇繁生物科技有限责任公司 HPK1 kinase inhibitor, preparation method and applications
WO2019238067A1 (en) * 2018-06-13 2019-12-19 Beigene, Ltd. Pyrrolo [2, 3-b] pyridines or pyrrolo [2, 3-b] pyrazines as hpk1 inhibitor and the use thereof
US20200108075A1 (en) * 2018-10-03 2020-04-09 Genentech, Inc. 8-aminoisoquinoline compounds and uses thereof
WO2020103896A1 (en) * 2018-11-22 2020-05-28 Beigene, Ltd. Pyrrolo[2,3-b]pyridines as hpk1 inhibitor and uses thereof
CN113166139A (en) * 2018-11-22 2021-07-23 百济神州有限公司 Pyrrolo [2,3-b ] pyridines as HPK1 inhibitors and uses thereof
CN116848113A (en) * 2021-02-10 2023-10-03 南京再明医药有限公司 Bicyclic compounds as HPK1 inhibitors and application thereof
CN115433184A (en) * 2021-06-04 2022-12-06 武汉人福创新药物研发中心有限公司 HPK1 inhibitor and application thereof
CN116162087A (en) * 2023-02-20 2023-05-26 杭州医学院 HPK 1 and/or LCK kinase regulator, preparation method and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DILLON H. MILES ET AL.: ""Discovery of Potent and Selective 7‑Azaindole Isoindolinone-Based PI3Kγ Inhibitors"", 《ACS MED. CHEM. LETT.》, vol. 11, 24 September 2020 (2020-09-24), pages 2244 - 2252, XP055954608, DOI: 10.1021/acsmedchemlett.0c00387 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11897878B2 (en) 2018-10-31 2024-02-13 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds
US11925631B2 (en) 2018-10-31 2024-03-12 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds
US12037342B2 (en) 2019-05-23 2024-07-16 Gilead Sciences, Inc. Substituted eneoxindoles and uses thereof

Similar Documents

Publication Publication Date Title
TWI765908B (en) Benzimidazole compounds as kinase inhibitors, and preparation methods and applications thereof
CN109879859B (en) COT modulators and methods of use thereof
TWI704152B (en) Naphthyridine compounds as jak kinase inhibitors
AU2014324595B2 (en) Substituted nicotinimide inhibitors of BTK and their preparation and use in the treatment of cancer, inflammation and autoimmune disease
JP5808869B2 (en) Bicyclic piperazine compounds
CN114685489A (en) Polysubstituted nitrogen-containing heterocyclic compound and application thereof
AU2012282229B2 (en) Novel pyrrolo pyrimidine derivatives
JP6507234B2 (en) Pyrazole carboxamide compounds for use in the treatment of disorders mediated by bruton tyrosine kinase (BTK)
BR102018002879A2 (en) compound, and method for treating an inflammation-related disease or disorder in a patient in need thereof.
TW201900640A (en) Thienopyridine and benzothiophene as IRAK4 inhibitors
TWI580679B (en) Heteroaryl-pyrimidine derivatives, preparation process and pharmaceutical use thereof
JP2023511337A (en) Pyrimidin-4(3H)-one heterocyclic compounds, processes for their preparation, and pharmaceutical uses thereof
BR112014030655B1 (en) pyrimidinyl tyrosine kinase inhibitor compounds, and pharmaceutical composition comprising them
WO2022171034A1 (en) Bicyclic compound as hpk1 inhibitor and application thereof
CN114685490A (en) Pyrrolopyridine compound and application thereof
CN117062818A (en) Novel SOS1 inhibitor and preparation method and application thereof
JPWO2018097234A1 (en) Novel oxoisoquinoline derivatives
CN116023378A (en) Lactam compounds as HPK1 inhibitors and uses thereof
EP4065230A1 (en) Therapeutic compounds
WO2022166793A1 (en) Cdk inhibitor
TW202220998A (en) Heteroaryl heterocyclic compounds and uses thereof
CN115023428A (en) Pyrimidopyrroles
CN117050095A (en) Tricyclic compounds as HPK1 inhibitors and application thereof
CN105985354B (en) Pyrimidine derivatives, cytotoxic agents, pharmaceutical compositions and uses thereof
CN115867552A (en) Imidazolopyrimidine derivative, preparation method thereof and application thereof in medicines

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20230807

Address after: 210032 No.99, Huakang Road, Jiangbei new district, Nanjing City, Jiangsu Province

Applicant after: Nanjing Zaiming Pharmaceutical Co.,Ltd.

Address before: 210042 699 Xuanwu Road, Xuanwu District, Nanjing, Jiangsu -18

Applicant before: JIANGSU SIMCERE PHARMACEUTICAL Co.,Ltd.