CN114805361A - Amino-substituted aromatic heterocyclic pyrazole compounds, preparation method and application - Google Patents

Amino-substituted aromatic heterocyclic pyrazole compounds, preparation method and application Download PDF

Info

Publication number
CN114805361A
CN114805361A CN202210051634.4A CN202210051634A CN114805361A CN 114805361 A CN114805361 A CN 114805361A CN 202210051634 A CN202210051634 A CN 202210051634A CN 114805361 A CN114805361 A CN 114805361A
Authority
CN
China
Prior art keywords
substituted
membered
unsubstituted
alkyl
heterocycloalkyl
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.)
Granted
Application number
CN202210051634.4A
Other languages
Chinese (zh)
Other versions
CN114805361B (en
Inventor
万惠新
王亚周
马金贵
王亚辉
查传涛
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.)
Shanghai Lingda Biomedical Co ltd
Original Assignee
Shanghai Lingda Biomedical 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 Shanghai Lingda Biomedical Co ltd filed Critical Shanghai Lingda Biomedical Co ltd
Publication of CN114805361A publication Critical patent/CN114805361A/en
Application granted granted Critical
Publication of CN114805361B publication Critical patent/CN114805361B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/06Anti-spasmodics, e.g. drugs for colics, esophagic dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • C07F9/65616Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings containing the ring system having three or more than three double bonds between ring members or between ring members and non-ring members, e.g. purine or analogs

Abstract

The invention discloses an amino-substituted aromatic heterocyclic pyrazole compound, a preparation method and application thereof, in particular to an amino-substituted aromatic heterocyclic pyrazole compound shown as a general formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, a preparation method and a pharmaceutical application thereof, wherein the definition of each group is described in the specification.

Description

Amino-substituted aromatic heterocyclic pyrazole compounds, preparation method and application
Technical Field
The invention belongs to the field of medicinal chemistry, and discloses an amino-substituted aromatic heterocyclic pyrazole compound with CDK kinase inhibitory activity, a medicinal composition and application thereof, in particular to a compound shown as a general formula I. The compounds of the present invention and pharmaceutical compositions comprising these compounds are useful in the prevention or treatment of related diseases, particularly those mediated by the aberrant activity of CDK kinases, particularly CDK7 kinase.
Background
Cell cycle abnormalities are a hallmark feature of cancer, and cyclin-dependent kinases (CDKs) are a class of serine/threonine kinases that play a central role in the cell cycle, leading to the initiation, progression, and termination of the cell cycle. The CDK family are important signal transduction molecules within cells, which form CDK-cyclin complexes with cyclins, and are involved in the growth, proliferation, dormancy and apoptosis of cells.
Over the past 20 years, drug development targeting CDK kinases for tumor therapy has received extensive attention, as flavopiridol (alvocidib), Seliciclib (CYC202), Dinaciclib (SCH727965) and micciclib (PHA-848125) have all entered into different stages of clinical studies. However, the clinical application is limited due to the fact that the CDK inhibitor discovered early has low inhibitory activity on each CDK family subtype, or lacks certain selectivity, or is poorly absorbed in vivo. In recent years, drug development in this area has again been a focus due to the discovery of selective inhibitors that either increase the selectivity of CDK inhibitors for each CDK family subtype or increase the inhibitory activity of CDK kinases, particularly targeting CDK 4/6.
Recent studies have found that CDK7 kinase of the CDK family has dual functions of regulating both kinase and transcription: 1) in the cytoplasm, CDK7 exists as a heterotrimeric complex and acts as an activated kinase (CAK) of CDK1/2, whereby phosphorylation of conserved residues in CDK1/2 by CDK7 is necessary for full catalysis of CDK activity and cell cycle progression; 2) in the nucleus, CDK7 forms the kinase core of the RNA polymerase (RNAP) II universal transcription factor complex and is responsible for phosphorylating the C-terminal domain (CTD) of rnapi, an essential step in gene transcription initiation. Two functions of CDK7, CAK and CTD phosphorylation, support key aspects of cell proliferation, cell cycle, and transcription. Research shows that CDK7 kinase has a very important effect on the regulation of triple negative breast cancer, and the inhibition of CDK7 kinase has a significant killing effect on the growth of triple negative breast cancer cells.
In addition, the CDK9 kinases of the CDK family play a role primarily in the regulation of transcriptional elongation without affecting cell cycle processes. The CDK9 inhibitor can prevent the phosphorylation of positive transcription elongation factor P-TEFb (positive transcription elongation factor b) on the RNA Poly-II C terminal region by degrading and inhibiting CDK9, inhibit transcription, and rapidly reduce the level of intracellular mRNA, thereby causing tumor cell apoptosis.
Although great progress has been made in the development of CDK kinase inhibitor drugs, some unsolved problems such as drug resistance of existing CDK kinase inhibitor drugs, subtype selectivity to CDK kinase family targets and the like exist, so that research and development of novel CDK kinase inhibitors with high efficiency, low toxicity, resistance and clinical application value, such as specific CDK7 kinase inhibitors, are urgently needed in the art.
Disclosure of Invention
One of the technical problems to be solved by the present invention is to provide a novel CDK7 inhibitor for preparing tumor treatment drugs.
The scheme for solving the technical problems is as follows:
an amino-substituted aromatic heterocyclic pyrazole compound shown as a general formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof,
Figure BDA0003474551550000021
in the formula:
w is selected from CR w Or N; rw is independently selected from H, halogen, cyano, C1-C6 alkyl or haloalkyl;
R 1 independently selected from C1-C10 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl, C1-C10 alkoxy or haloalkoxy, 3-10 membered cycloalkyl ether or heterocycloalkyl ether, C1-C10 alkyl or haloalkyl substituted amino, 3-10 membered cycloalkyl or heterocycloalkyl substituted amino(ii) a And the above alkyl, cycloalkyl, heterocycloalkyl may be substituted with one or several substituents selected from the group consisting of: halogen, deuterium, hydroxyl, substituted or unsubstituted amino, C1-C6 alkyl, hydroxyl-substituted C1-C6 alkyl, amino-substituted C1-C6 alkyl, C1-C6 alkoxy, 3-10 membered cycloalkyl or heterocycloalkyl-substituted C1-C6 alkyl;
R 2 independently selected from C1-C10 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl;
ra is selected from hydrogen, deuterium, halogen, C1-C6 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl;
rb, Rc, Rd, Re are each independently selected from hydrogen, deuterium, halogen, C1-C6 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl, or Rb and Rc, Rd and Re each form a C ═ O bond or a 3-6 membered carbocyclic or heterocyclic ring;
m and n are respectively and independently selected from integers of 0-3;
cy is selected from 3-10 membered cycloalkyl or heterocycloalkyl, 5-12 membered monocyclic or fused aromatic or heteroaromatic group;
r3 is selected from hydrogen, deuterium, halogen, C1-C10 alkyl or haloalkyl, 3-to 10-membered cycloalkyl or heterocycloalkyl, 5-to 12-membered aryl or heteroaryl, substituted or unsubstituted amino, hydroxyl, C1-C10 alkoxy or haloalkoxy, cyano, substituted or unsubstituted amide, substituted or unsubstituted sulfonamide, substituted or unsubstituted ureido, substituted or unsubstituted sulfonylureido, substituted or unsubstituted carbamoyl, substituted or unsubstituted phosphoryl, substituted or unsubstituted alkylphosphite, substituted or unsubstituted alkylsilyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted acryloyl, substituted or unsubstituted propioloyl, haloacetyl or haloethylsulfonyl;
y is selected from N or CRy, Ry is independently selected from hydrogen, deuterium, halogen, hydroxy, amino, cyano, C1-C6 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl;
r4 is independently selected from the group consisting of hydrogen, deuterium, halogen, C1-C10 alkyl or haloalkyl, 3-to 10-membered cycloalkyl or heterocycloalkyl, 5-to 12-membered aryl or heteroaryl, substituted or unsubstituted amino, hydroxyl, C1-C10 alkoxy or haloalkoxy, cyano, substituted or unsubstituted amido, substituted or unsubstituted sulfonamide, substituted or unsubstituted ureido, substituted or unsubstituted sulfonylureido, substituted or unsubstituted carbamoyl, substituted or unsubstituted phosphoryl, substituted or unsubstituted alkylphosphite, substituted or unsubstituted alkylsilyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted acryloyl, substituted or unsubstituted propioloyl, haloacetyl or haloethylsulfonyl
The above-mentioned substituted or unsubstituted substituents are each independently selected from the group consisting of: including but not limited to deuterium, halogen, hydroxy, monoalkylamino, dialkylamino, C1-C6 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl, cyano, C1-C6 alkoxy or haloalkoxy; wherein said heteroaryl group contains 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, the heterocycloalkyl group containing 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, said ring system including spiro, bridged, fused, etc. saturated or partially unsaturated ring systems.
In some of the preferred embodiments, the first and second,
R 1 independently selected from C1-C6 alkyl, C1-C6 haloalkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, 5-8 membered heterocycloalkyl-O-, C1-C6 alkyl substituted amino, C1-C6 haloalkyl substituted amino, 3-8 membered cycloalkyl substituted amino, 3-8 membered heterocycloalkyl substituted amino; the R is 1 1 or more hydrogen atoms above may be optionally substituted with: halogen, hydroxyl, amino, cyano, mono C1-C3 alkylamino, di C1-C3 alkylamino, C1-C3 alkyl, hydroxyl-substituted C1-C3 alkyl, amino-substituted C1-C3 alkyl, C1-C3 alkoxy, 3-8 membered cycloalkyl C1-C3 alkyl-, 3-8 membered heterocycloalkyl-C1-C3 alkyl-, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl;
R 2 independently selected from C1-C6 alkyl, C1-C6 haloalkyl, 3-8 membered cycloalkyl or 3-8 membered heterocycloalkyl;
cy is selected from 5-8 membered cycloalkyl, 5-8 heterocycloalkyl, 6-10 membered monocyclic or fused aromatic group, 5-10 membered monocyclic or fused heteroaromatic group;
R 3 selected from hydrogen, deuterium, halogen, hydroxy, cyano, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, 3-6 membered cycloalkyl or heterocycloalkyl, 6-10 membered aryl, 5-8 membered heteroaryl, substituted or unsubstituted amino, substituted or unsubstituted amido;
R 4 independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, 3-6 membered cycloalkyl, 3-6 membered heterocycloalkyl, 6-10 membered aryl, 5-8 membered heteroaryl, C1-C6 alkoxy, C1-C6 haloalkoxy, 3-6 membered cycloalkyl-C (O) -, 3-6 membered heterocycloalkyl-C (O) -, 3-6 membered cycloalkyl-O-C (O) -, 3-6 membered heterocycloalkyl-O-C (O) -, 6-10 membered aryl-C (O) -, 5-8 membered heteroaryl-C (O) -; the R is 4 Optionally substituted by R 41 Substituted, said R 41 Independently selected from: halogen, hydroxy, cyano, mono-C1-C6 alkylamino, di-C1-C6 alkylamino, C1-C6 alkyl, C1-C6 haloalkyl, 3-6 membered cycloalkyl or heterocycloalkyl, C1-C6 alkoxy or haloalkoxy, C2-C6 alkenyl-C (O) -; the R is 41 May be further substituted with: halogen, hydroxy, cyano, mono-C1-C6 alkylamino, di-C1-C6 alkylamino.
In some preferred embodiments, R 1 Preferably, it is
Figure BDA0003474551550000041
Or, R 2 Preferably methyl, isopropyl;
or, R 3 Is H, F, Cl, CN, CH 3 、CH 3 O-、CF 3 、、
Figure BDA0003474551550000042
Figure BDA0003474551550000043
Alternatively, the first and second electrodes may be,R 4 is H, CH 3 An isopropyl group,
Figure BDA0003474551550000051
Figure BDA0003474551550000052
In some preferred embodiments, the compound having the general formula (I), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsomer, solvate, polymorph or prodrug thereof, preferably is a compound represented by the general formula (II), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsomer, solvate, polymorph or prodrug thereof:
Figure BDA0003474551550000053
all radicals i.e. R 1 、R 2 、R 3 、R 4 、R a 、R b 、R c 、R d 、R e Cy, W, m, n ranges are as defined above.
In some preferred embodiments, it is preferably a compound of formula (III), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph or prodrug thereof:
Figure BDA0003474551550000061
all radicals i.e. R 1 、R 2 、R 3 、R 4 、R a 、R b 、R c 、R d 、R e Cy, W, m, n ranges are as defined above.
In some preferred embodiments, it is preferably a compound of formula (IV), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph or prodrug thereof:
Figure BDA0003474551550000062
wherein Cy1 is selected from 5-10 membered aryl or heteroaryl, Cy2 is selected from 3-12 membered saturated carbocyclic or heterocyclic groups, and the range of other groups is as defined above.
In some embodiments, it is preferably a compound of formula (V), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph or prodrug thereof:
Figure BDA0003474551550000063
r1 is preferably selected from C1-C6 alkyl, such as methyl; substituted or unsubstituted 5-6 membered saturated ring such as cyclohexyl, piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl and the like; substituted or unsubstituted 5-6 membered saturated cyclic group ethers such as cyclohexyl ether, piperidinyl ether, tetrahydropyranyl ether; substituted or unsubstituted 5-6 membered saturated cyclic amines, such as cyclohexylamino, piperidinamino, tetrahydropyrylamino; substituted or unsubstituted 5-6 membered saturated cyclic alkylene ethers such as piperidylmethylene ether; substituted or unsubstituted 5-6 membered saturated cyclylalkyleneamine, such as piperidinylidenemethylamine; the substituent is preferably selected from deuterium, halogen, amino, hydroxyl, dimethylamino, methoxy, hydroxymethyl, aminomethyl and the like; the ranges for the other groups are as defined above.
A method for preparing the compound of the formula I is characterized in that the compound of the general formula (A) and an amino compound (B) are subjected to nucleophilic substitution reaction under the catalysis of alkali or acid to generate the compound of the general formula (I);
Figure BDA0003474551550000071
the definition of each group is as described above;
preferably, the reaction is carried out in a solvent, and the solvent is selected from the group consisting of: water, methanol, ethanol, isopropanol, butanol, ethylene glycol methyl ether, N-methyl pyrrolidone, dimethyl sulfoxide, tetrahydrofuran, toluene, dichloromethane, 1, 2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, or a combination thereof.
Preferably, the inorganic base is selected from the group consisting of: sodium hydride, potassium hydroxide, sodium acetate, potassium tert-butoxide, sodium tert-butoxide, potassium fluoride, cesium fluoride, potassium phosphate, potassium carbonate, potassium bicarbonate, sodium carbonate, sodium bicarbonate, or combinations thereof; the organic base is selected from the group consisting of: pyridine, triethylamine, N, N-diisopropylethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), lithium hexamethyldisilazide, sodium hexamethyldisilazide, lutidine, or a combination thereof.
Preferably, the acid is selected from the group consisting of: hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, toluenesulfonic acid, trifluoroacetic acid, formic acid, acetic acid, trifluoromethanesulfonic acid, or combinations thereof.
The invention provides a class of preferred compounds of formula (I) including, but not limited to, the following structures:
structural formula of example
Figure BDA0003474551550000081
Figure BDA0003474551550000091
Figure BDA0003474551550000101
Another objective of the invention is to provide a medicament for treating or preventing tumor or autoimmune diseases and a composition thereof. The technical scheme for realizing the purpose is as follows:
in one aspect, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula I, a stereoisomer, geometric isomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, and a pharmaceutically acceptable carrier. In certain embodiments of the pharmaceutical composition, the pharmaceutical composition is formulated for intravenous administration, intramuscular administration, oral administration, rectal administration, inhalation administration, nasal administration, topical administration, ocular administration, or otic administration. In other embodiments of the pharmaceutical composition, the pharmaceutical composition is a tablet, pill, capsule, liquid, inhalant, nasal spray solution, suppository, solution, emulsion, ointment, eye drop, or ear drop. In other embodiments of the pharmaceutical composition, it further comprises one or more additional therapeutic agents.
In another aspect, the present invention provides the use of a compound of formula I, a stereoisomer, geometric isomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof in the manufacture of a medicament for the prevention, treatment, or alleviation of a disorder or disease mediated by abnormal activity of a CDK kinase, particularly CDK7 kinase.
In another aspect, the present invention provides an amino-substituted aromatic heterocyclic pyrazole compound represented by the general formula (I), or a pharmaceutically acceptable salt, enantiomer, diastereomer, tautomer, solvate, polymorph or prodrug thereof, for use in the preparation of a method for treating or preventing a proliferative disease (e.g., cancer (e.g., leukemia, melanoma, multiple myeloma), benign neoplasm, angiogenesis, inflammatory disease, autoinflammatory disease and autoimmune disease) in a subject, wherein the tumor is independently selected from the group consisting of non-small cell lung cancer, lung adenocarcinoma, squamous cell carcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, stomach cancer, intestinal cancer, cholangiocarcinoma, brain cancer, leukemia, lymph cancer, fibroma sarcoma, basal cell carcinoma, colon cancer, and prostate cancer, Glioma, renal cancer, melanoma, osteocarcinoma, thyroid cancer, nasopharyngeal carcinoma, pancreatic cancer, etc.; the autoimmune disease is independently selected from rheumatoid arthritis, systemic lupus erythematosus, idiopathic thrombocytopenic purpura, hemolytic anemia, or psoriasis; the inflammatory disease is independently selected from osteoarthritis, gouty arthritis, ulcerative colitis, inflammatory bowel disease and/or the like; the infectious disease is independently selected from sepsis, septic shock, endotoxic shock, gram-negative sepsis, and/or toxic shock syndrome.
The invention relates to a novel compound with the structural characteristics of a general formula (I), which selectively inhibits the enzyme activity of CDK7, obviously inhibits the growth of various tumor cells, and is a treatment drug with a brand-new action mechanism.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. The space is not described herein in a repeated fashion.
Detailed Description
The inventors have long and intensively studied to prepare a class of amino-substituted heteroaromatic pyrazole compounds having a novel structure shown in formula I, and found that the compounds have a good activity of inhibiting cyclin-dependent kinase 7(CDK7), and the compounds have a specific inhibitory effect on cyclin-dependent kinase 7(CDK7) at a very low concentration (which can be as low as less than 10nM), and are excellent in a cell proliferation inhibitory activity related to CDK7, and thus can be used for treating diseases such as tumors related to the abnormal activity of cyclin-dependent kinase 7(CDK 7). Based on the above findings, the inventors have completed the present invention.
Term(s) for
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the claimed subject matter belongs. All patents, patent applications, and publications cited herein are incorporated by reference in their entirety unless otherwise indicated.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the subject matter claimed. In this application, the use of the singular also includes the plural unless specifically stated otherwise. It must be noted that, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. It should also be noted that the use of "or", "or" means "and/or" unless stated otherwise. Furthermore, the term "comprising" as well as other forms, such as "includes," "including," and "containing," are not limiting.
Definitions for the terms of the standardization sector can be found in the literature references including Carey and Sundberg "ADVANCED ORGANIC CHEMISTRY 4TH ED." Vols.A (2000) and B (2001), Plenum Press, New York. Unless otherwise indicated, conventional methods within the skill of the art are employed, such as mass spectrometry, NMR, IR and UV/VIS spectroscopy, and pharmacological methods. Unless a specific definition is set forth, the terms used herein in the pertinent description of analytical chemistry, organic synthetic chemistry, and pharmaceutical chemistry are known in the art. Standard techniques can be used in chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and delivery, and treatment of patients. For example, the reaction and purification can be carried out using the instructions of the kit from the manufacturer, or according to the methods known in the art or the instructions of the present invention. The techniques and methods described above can generally be practiced according to conventional methods well known in the art, as described in various general and more specific documents referred to and discussed in this specification. In the present specification, groups and substituents thereof may be selected by one skilled in the art to provide stable moieties and compounds.
When a substituent is described by a general formula written from left to right, the substituent also includes chemically equivalent substituents obtained when the formula is written from right to left. For example, -CH 2O-is equivalent to-OCH 2-.
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including but not limited to patents, patent applications, articles, books, operating manuals, and treatises, are hereby incorporated by reference in their entirety.
Certain chemical groups defined herein are preceded by a shorthand notation to indicate the total number of carbon atoms present in the group. For example, C1-6 alkyl refers to an alkyl group as defined below having a total of 1 to 6 carbon atoms. The total number of carbon atoms in the shorthand notation excludes carbons that may be present in a substituent of the group.
In addition to the foregoing, the following terms, when used in the specification and claims of this application, have the meanings indicated below, unless otherwise specifically indicated.
In the present application, the term "halogen" means fluorine, chlorine, bromine or iodine; "hydroxy" means an-OH group; "hydroxyalkyl" refers to an alkyl group as defined below substituted with a hydroxyl (-OH) group; "carbonyl" refers to a-C (═ O) -group; "Nitro" means-NO 2 (ii) a "cyano" means-CN; "amino" means-NH 2 (ii) a "substituted amino" refers to an amino group substituted with one or two alkyl, alkylcarbonyl, aralkyl, heteroaralkyl groups as defined below, e.g., monoalkylamino, dialkylamino, alkylamido, aralkylamino, heteroaralkylamino; "carboxyl" means-COOH.
In the present application, the term "alkyl", as a group or as part of another group (e.g. as used in groups such as halogen-substituted alkyl), means a straight or branched hydrocarbon chain group consisting only of carbon and hydrogen atoms, containing no unsaturated bonds, having, for example, from 1 to 12 (preferably from 1 to 8, more preferably from 1 to 6) carbon atoms and being attached to the rest of the molecule by single bonds. Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2-dimethylpropyl, n-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl, decyl, and the like.
In the present application, the term "alkenyl" as a group or part of another group means a straight or branched hydrocarbon chain group consisting of only carbon atoms and hydrogen atoms, containing at least one double bond, having, for example, 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms, and attached to the rest of the molecule by a single bond, such as, but not limited to, vinyl, propenyl, allyl, but-1-enyl, but-2-enyl, pent-1, 4-dienyl, and the like.
In the present application, the term "alkynyl" as a group or part of another group means a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one triple bond and optionally one or more double bonds, having for example 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms and being connected to the rest of the molecule by single bonds, such as but not limited to ethynyl, prop-1-ynyl, but-1-ynyl, pent-1-en-4-ynyl and the like.
In the present application, the term "cycloalkyl" as a group or part of another group means a stable non-aromatic monocyclic or polycyclic hydrocarbon group consisting of only carbon atoms and hydrogen atoms, which may include fused, bridged or spiro ring systems, having 3 to 15 carbon atoms, preferably having 3 to 10 carbon atoms, more preferably having 3 to 8 carbon atoms, and which is saturated or unsaturated and may be attached to the rest of the molecule by a single bond via any suitable carbon atom. Unless otherwise specifically indicated in the specification, carbon atoms in cycloalkyl groups may be optionally oxidized. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, 1H-indenyl, 2, 3-indanyl, 1,2,3, 4-tetrahydro-naphthyl, 5,6,7, 8-tetrahydro-naphthyl, 8, 9-dihydro-7H-benzocyclohepten-6-yl, 6,7,8, 9-tetrahydro-5H-benzocycloheptenyl, 5,6,7,8,9, 10-hexahydro-benzocyclooctenyl, fluorenyl, bicyclo [2.2.1] heptyl, 7-dimethyl-bicyclo [2.2.1] heptyl, bicyclo [2.2.1] heptenyl, bicyclo [2.2.2] octyl, bicyclo [3.1.1] heptyl, bicyclo [3.2.1] octyl, bicyclo [2.2.2] octenyl, Bicyclo [3.2.1] octenyl, adamantyl, octahydro-4, 7-methylene-1H-indenyl, octahydro-2, 5-methylene-pentalenyl and the like.
In this application, the term "heterocyclyl" as a group or part of another group means a stable 3-to 20-membered non-aromatic cyclic group consisting of 2 to 14 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, phosphorus, oxygen, and sulfur. Unless otherwise specifically indicated in the specification, a heterocyclic group may be a monocyclic, bicyclic, tricyclic or higher ring system, which may include fused ring systems, bridged ring systems or spiro ring systems; wherein the nitrogen, carbon or sulfur atom in the heterocyclic group thereof may be optionally oxidized; the nitrogen atoms may optionally be quaternized; and the heterocyclic group may be partially or fully saturated. The heterocyclic group may be attached to the rest of the molecule via a carbon atom or a heteroatom and by a single bond. In heterocyclic groups containing fused rings, one or more of the rings may be aryl or heteroaryl as defined below, provided that the point of attachment to the rest of the molecule is a non-aromatic ring atom. For the purposes of the present invention, heterocyclyl is preferably a stable 4-to 11-membered non-aromatic monocyclic, bicyclic, bridged or spiro group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 4-to 8-membered non-aromatic monocyclic, bicyclic, bridged or spiro group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heterocyclyl groups include, but are not limited to: pyrrolidinyl, morpholinyl, piperazinyl, homopiperazinyl, piperidinyl, thiomorpholinyl, 2, 7-diaza-spiro [3.5] nonan-7-yl, 2-oxa-6-aza-spiro [3.3] heptan-6-yl, 2, 5-diaza-bicyclo [2.2.1] heptan-2-yl, azetidinyl, pyranyl, tetrahydropyranyl, thiopyranyl, tetrahydrofuranyl, oxazinyl, dioxolanyl, tetrahydroisoquinolinyl, decahydroisoquinolinyl, imidazolinyl, imidazolidinyl, quinolizinyl, thiazolidinyl, isothiazolidinyl, isoxazolidinyl, dihydroindolyl, octahydroindolyl, octahydroisoindolyl, pyrrolidinyl, pyrazolidinyl, phthalimido, and the like.
In this application, the term "aryl" as a group or as part of another group means a conjugated hydrocarbon ring system group having 6 to 18 carbon atoms, preferably having 6 to 10 carbon atoms. For the purposes of the present invention, an aryl group may be a monocyclic, bicyclic, tricyclic or higher polycyclic ring system and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the aryl group is attached to the remainder of the molecule by a single bond via an atom on the aromatic ring. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, phenanthrenyl, fluorenyl, 2, 3-dihydro-1H-isoindolyl, 2-benzoxazolinone, 2H-1, 4-benzoxazin-3 (4H) -one-7-yl, and the like.
In the present application, the term "arylalkyl" refers to an alkyl group as defined above substituted with an aryl group as defined above.
In this application, the term "heteroaryl" as a group or part of another group means a 5-to 16-membered conjugated ring system group having 1 to 15 carbon atoms (preferably having 1 to 10 carbon atoms) and 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur in the ring. Unless otherwise specifically indicated in the specification, a heteroaryl group may be a monocyclic, bicyclic, tricyclic or higher ring system, and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the heteroaryl group is attached to the rest of the molecule by a single bond via an atom on the aromatic ring. The nitrogen, carbon or sulfur atoms in the heteroaryl group may be optionally oxidized; the nitrogen atoms may optionally be quaternized. For the purposes of the present invention, heteroaryl is preferably a stable 5-to 12-membered aromatic group containing 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 5-to 10-membered aromatic group containing 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur or a 5-to 6-membered aromatic group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heteroaryl groups include, but are not limited to, thienyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, benzopyrazolyl, indolyl, furyl, pyrrolyl, triazolyl, tetrazolyl, triazinyl, indolizinyl, isoindolyl, indazolyl, isoindolyl, purinyl, quinolyl, isoquinolyl, diazonaphthyl, naphthyridinyl, quinoxalinyl, pteridinyl, carbazolyl, carbolinyl, phenanthridinyl, phenanthrolinyl, acridinyl, phenazinyl, isothiazolyl, benzothiazolyl, benzothienyl, oxazolyl, cinnolinyl, quinazolinyl, thiophenyl, indolizinyl, orthophenanthrolidinyl, isoxazolyl, phenoxazinyl, phenothiazinyl, 4,5,6, 7-tetrahydrobenzo [ b ] thienyl, naphthopyridyl, pyridinyl, and the like, [1,2,4] triazolo [4,3-b ] pyridazine, [1,2,4] triazolo [4,3-a ] pyrazine, [1,2,4] triazolo [4,3-c ] pyrimidine, [1,2,4] triazolo [4,3-a ] pyridine, imidazo [1,2-b ] pyridazine, imidazo [1,2-a ] pyrazine and the like.
In the present application, the term "heteroarylalkyl" refers to an alkyl group as defined above substituted with a heteroaryl group as defined above.
In this application, "optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "optionally substituted aryl" means that the aryl group is substituted or unsubstituted, and the description includes both substituted and unsubstituted aryl groups.
The terms "moiety," "structural moiety," "chemical moiety," "group," "chemical group" as used herein refer to a specific fragment or functional group in a molecule. Chemical moieties are generally considered to be chemical entities that are embedded in or attached to a molecule.
"stereoisomers" refers to compounds that consist of the same atoms, are bonded by the same bonds, but have different three-dimensional structures. The present invention is intended to cover various stereoisomers and mixtures thereof.
When the compounds of the present invention contain olefinic double bonds, the compounds of the present invention are intended to include both E-and Z-geometric isomers unless otherwise specified.
"tautomer" refers to an isomer formed by the transfer of a proton from one atom of a molecule to another atom of the same molecule. All tautomeric forms of the compounds of the invention are also intended to be included within the scope of the invention.
The compounds of the present invention or pharmaceutically acceptable salts thereof may contain one or more chiral carbon atoms and may therefore give rise to enantiomers, diastereomers and other stereoisomeric forms. Each chiral carbon atom may be defined as (R) -or (S) -, based on stereochemistry. The present invention is intended to include all possible isomers, as well as racemates and optically pure forms thereof. The compounds of the invention may be prepared by selecting as starting materials or intermediates racemates, diastereomers or enantiomers. Optically active isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, e.g., crystallization and chiral chromatography.
Conventional techniques for the preparation/separation of individual isomers include Chiral synthesis from suitable optically pure precursors, or resolution of racemates (or racemates of salts or derivatives) using, for example, Chiral high performance liquid chromatography, as described, for example, in Gerald Gubitz and Martin G.Schmid (Eds.), Chiral Separations, Methods and Protocols, Methods in Molecular Biology, Vol.243, 2004; m. Stalcup, Chiral Separations, Annu. Rev. anal. chem.3:341-63, 2010; fumiss et al (eds.), VOGEL' S ENCYCOPEDIA OF PRACTICAL ORGANIC CHEMISTRY 5. TH ED., Longman Scientific and Technical Ltd., Essex,1991, 809-816; heller, acc, chem, res, 1990,23,128.
In the present application, the term "pharmaceutically acceptable salts" includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
"pharmaceutically acceptable acid addition salts" refers to salts with inorganic or organic acids which retain the biological effectiveness of the free base without other side effects. Inorganic acid salts include, but are not limited to, hydrochloride, hydrobromide, sulfate, nitrate, phosphate, and the like; organic acid salts include, but are not limited to, formates, acetates, 2-dichloroacetates, trifluoroacetates, propionates, caproates, caprylates, caprates, undecylenates, glycolates, gluconates, lactates, sebacates, adipates, glutarates, malonates, oxalates, maleates, succinates, fumarates, tartrates, citrates, palmitates, stearates, oleates, cinnamates, laurates, malates, glutamates, pyroglutamates, aspartates, benzoates, methanesulfonates, benzenesulfonates, p-toluenesulfonates, alginates, ascorbates, salicylates, 4-aminosalicylates, napadisylates, and the like. These salts can be prepared by methods known in the art.
"pharmaceutically acceptable base addition salts" refers to salts with inorganic or organic bases which maintain the biological effectiveness of the free acid without other side effects. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Preferred inorganic salts are ammonium, sodium, potassium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, the following: primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, triethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purine, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like. Preferred organic bases include isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine. These salts can be prepared by methods known in the art.
"polymorph" refers to different solid crystalline phases of certain compounds of the present invention in the solid state due to the presence of two or more different molecular arrangements. Certain compounds of the present invention may exist in more than one crystalline form and the present invention is intended to include the various crystalline forms and mixtures thereof.
Typically, crystallization will result in solvates of the compounds of the invention. The term "solvate" as used herein refers to an aggregate comprising one or more molecules of the compound of the present invention and one or more solvent molecules. The solvent may be water, in which case the solvate is a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present invention may exist as hydrates, including monohydrates, dihydrate, hemihydrate, sesquihydrates, trihydrate, tetrahydrate, and the like, as well as the corresponding solvated forms. The compounds of the invention may form true solvates, but in some cases it is also possible to retain only adventitious water or a mixture of water plus a portion of adventitious solvent. The compounds of the invention may be reacted in a solvent or precipitated or crystallized from a solvent. Solvates of the compounds of the invention are also included within the scope of the invention.
The invention also includes prodrugs of the above compounds. In the present application, the term "prodrug" denotes a compound that can be converted under physiological conditions or by solvolysis to the biologically active compound of the invention. Thus, the term "prodrug" refers to a pharmaceutically acceptable metabolic precursor of a compound of the invention. Prodrugs may not be active when administered to a subject in need thereof, but are converted in vivo to the active compounds of the invention. Prodrugs are generally rapidly converted in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. Prodrug compounds generally provide the advantages of solubility, histocompatibility, or sustained release in mammalian organisms. Prodrugs include known amino protecting groups and carboxyl protecting groups. Specific methods for preparing prodrugs can be found in Saulnier, M.G., et al, bioorg.Med.chem.Lett.1994,4, 1985-1990; greenwald, r.b., et al, j.med.chem.2000,43,475.
In the present application, a "pharmaceutical composition" refers to a formulation of a compound of the present invention with a vehicle generally accepted in the art for delivery of biologically active compounds to a mammal (e.g., a human). The medium includes a pharmaceutically acceptable carrier. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of active ingredients and exert biological activity.
The term "pharmaceutically acceptable" as used herein refers to a substance (e.g., carrier or diluent) that does not affect the biological activity or properties of the compounds of the present invention and is relatively non-toxic, i.e., the substance can be administered to an individual without causing an adverse biological response or interacting in an adverse manner with any of the components contained in the composition.
As used herein, a "pharmaceutically acceptable carrier" includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersing agent, suspending agent, stabilizing agent, isotonic agent, solvent, or emulsifying agent that is approved by the relevant governmental regulatory agency for human or livestock use.
The "tumor" and "diseases related to abnormal cell proliferation" include, but are not limited to, leukemia, gastrointestinal stromal tumor, histiocytic lymphoma, non-small cell lung cancer, pancreatic cancer, squamous cell lung cancer, lung adenocarcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, cervical cancer, ovarian cancer, intestinal cancer, nasopharyngeal cancer, brain cancer, bone cancer, esophageal cancer, melanoma, renal cancer, oral cancer, and the like.
The terms "preventing," "prevention," and "prevention" as used herein include reducing the likelihood of occurrence or worsening of a disease or disorder in a patient.
As used herein, the term "treatment" and other similar synonyms include the following meanings:
(i) preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is susceptible to the disease or condition, but has not been diagnosed as having the disease or condition;
(ii) inhibiting the disease or disorder, i.e., arresting its development;
(iii) alleviating the disease or condition, i.e., causing regression of the state of the disease or condition; or
(iv) Alleviating the symptoms caused by the disease or disorder.
The terms "effective amount," "therapeutically effective amount," or "pharmaceutically effective amount" as used herein, refer to an amount of at least one agent or compound that is sufficient to alleviate one or more symptoms of the disease or disorder being treated to some extent after administration. The result may be a reduction and/or alleviation of signs, symptoms, or causes, or any other desired change in a biological system. For example, an "effective amount" for treatment is the amount of a composition comprising a compound disclosed herein that is clinically necessary to provide a significant remission effect of the condition. An effective amount suitable in any individual case can be determined using techniques such as a dose escalation assay.
The terms "administering," "administration," "administering," and the like as used herein refer to a method capable of delivering a compound or composition to a desired site for biological action. These methods include, but are not limited to, oral routes, via the duodenal route, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. Administration techniques useful for The compounds and methods described herein are well known to those skilled in The art, for example, in Goodman and Gilman, The pharmaceutical Basis of Therapeutics, current ed.; pergamon; and Remington's, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa. In preferred embodiments, the compounds and compositions discussed herein are administered orally.
The terms "drug combination", "administering other treatment", "administering other therapeutic agent" and the like as used herein refer to a drug treatment obtained by mixing or combining more than one active ingredient, including fixed and unfixed combinations of active ingredients. The term "fixed combination" refers to the simultaneous administration of at least one compound described herein and at least one co-agent to a patient in the form of a single entity or a single dosage form. The term "non-fixed combination" refers to the simultaneous administration, concomitant administration, or sequential administration at variable intervals of at least one compound described herein and at least one synergistic formulation to a patient as separate entities. These also apply to cocktail therapy, for example the administration of three or more active ingredients.
It will also be appreciated by those skilled in the art that in the processes described below, the functional groups of the intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxyl, amino, mercapto and carboxylic acid. Suitable hydroxy protecting groups include trialkylsilyl or diarylalkylsilyl groups (e.g.tert-butyldimethylsilyl, tert-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butyloxycarbonyl, benzyloxycarbonyl and the like. Suitable thiol protecting groups include-C (O) -R "(where R" is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like. Suitable carboxyl protecting groups include alkyl, aryl or aralkyl esters.
Protecting groups may be introduced and removed according to standard techniques known to those skilled in the art and as described herein. The use of protecting Groups is described in detail in Greene, T.W. and P.G.M.Wuts, Protective Groups in Organic Synthesis, (1999),4th Ed., Wiley. The protecting group may also be a polymeric resin.
The invention will be further illustrated with reference to specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, generally according to conventional conditions, or according to conditions recommended by the manufacturer. Percentages and parts are by weight unless otherwise indicated.
In each example, the laboratory instruments are described (e.g. 1 H NMR was recorded by a nuclear magnetic resonance apparatus of the Varian Mercury-300 or Varian Mercury-400 type, 13 c NMR is recorded by a nuclear magnetic resonance apparatus of the Varian Mercury-400 or Varian Mercury-500 or Varian Mercury-600 type, chemical shifts are expressed in delta (ppm); mass spectra were recorded on a Finnigan/MAT-95(EI) and Finnigan LCQ/DECA and Micromass Ultra Q-TOF (ESI) type mass spectrometer; silica gel for reverse phase preparative HPLC separation 200-300 mesh).
Wherein, the chemical formula or the Chinese name table of the reagent represented by the English letter abbreviation is as follows:
iPrOH: isopropyl alcohol; EtOH: ethanol; DCM: dichloromethane; TFA: trifluoroacetic acid; MeOH: methanol; NaOH: sodium hydroxide; HCl: hydrogen chloride; TEA: triethylamine; raney Ni: raney nickel; 1, 4-dioxane: 1, 4-dioxane; NaH: sodium hydride; h 2 O: water; Pd/C: palladium on carbon; h 2 : hydrogen gas; HATU: 2- (7-benzotriazole oxide) -N, N, N ', N' -tetramethyluronium hexafluorophosphate; DMF: n, N-dimethylformamide; THF: tetrahydrofuran; boc 2 O: di-tert-butyl dicarbonate; NBS: n-bromosuccinimide; NCS: n-chlorosuccinimide; NIS: n-iodosuccinimide; MeCN: acetonitrile; DIPEA/DIEA: n, N-diisopropylethylamine; NaBH 4 : sodium borohydride; AcOH: acetic acid; ethyl acetate: ethyl acetate; NaBH 3 CN: sodium cyanoborohydride; k 2 CO 3 : potassium carbonate; cs 2 CO 3 : cesium carbonate; nBuLi:n-butyl lithium; LiAlH 4 : lithium aluminum hydride; pd (dppf) Cl 2 : [1, 1' -bis (diphenylphosphino) ferrocene]Palladium dichloride; KOAc (Koac): and (4) potassium acetate. Fumaronitril: nitrile fumarate; p (nBu) 3 : tri-n-butylphosphine; LDA: lithium diisopropylamide; LiOH: lithium hydroxide; MeI: methyl iodide; EtI: ethyl iodide; (CH) 2 O) n : paraformaldehyde; HCO 2 H: formic acid; CH (CH) 3 COCl: acetyl chloride; LCMS: liquid chromatography-mass spectrometry; xantphos: 4, 5-bis diphenylphosphino-9, 9-dimethylxanthene; TLC: thin layer chromatography; eq.: equivalent weight; DCE: 1, 2-dichloroethane; HEPES (high efficiency particulate air): 4-hydroxyethylpiperazine ethanesulfonic acid; EGTA: ethylene glycol bis (2-aminoethyl ether) tetraacetic acid; DTT: dithiothreitol
Examples general preparation method
Figure BDA0003474551550000191
The pyrazolocyclochloride intermediate (1eq.) is dissolved in acetonitrile, DIPEA (1.6eq.) and benzylamine intermediate (1.0eq.) are added in sequence, and the mixture is heated to 70 ℃ under the protection of nitrogen and stirred for 12 hours. TLC monitoring reaction is completed, cooling to room temperature, decompression concentration, silica gel column chromatography separation and purification of the remainder to obtain target product, and nuclear magnetic and mass spectrum are adopted to confirm the structure.
Or dissolving the product (1eq.) in methanol, adding hydrochloric acid aqueous solution/hydrochloric acid alcoholic solution (20eq.) or hydrobromic acid/acetic acid (10eq.), stirring at room temperature or heating for 3-10 hours to remove Boc and/or Cbz protecting groups, performing column chromatography separation and purification after conventional treatment to obtain a target product, and confirming the structure by nuclear magnetism and mass spectrometry;
or dissolving the product (1eq.) in methanol, and removing the protecting groups such as Cbz/benzyl under the catalysis of palladium carbon in hydrogen atmosphere. TLC monitoring reaction is complete, decompression concentration is carried out, the remainder is separated and purified by silica gel column chromatography to obtain a target product, and nuclear magnetism and mass spectrum are adopted to confirm the structure;
or dissolving the product (1eq.) in a proper solvent, performing proper conversion through conventional functional group conversion, such as carbonyl reagent acylation, acid-amine condensation, reductive amination, halide alkylation, metal reagent cross-coupling and other reactions to obtain a target product, and confirming the structure by nuclear magnetism and mass spectrometry.
Preparation of key intermediate:
Figure BDA0003474551550000201
examples preparation
Example 1: n- (2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) -1,2,3, 4-tetrahydroisoquinolin-4-amine
Figure BDA0003474551550000202
The first step is as follows: intermediate A (30mg,0.1mmol), tert-butyl 4-amino-3, 4-dihydroisoquinoline-2 (1H) carbonate (49mg,0.2mmol), DIEA (78mg,0.6mmol) were dissolved in acetonitrile (5mL) under nitrogen and reacted at 70 ℃ for 4H. The reaction mixture was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (PE: EA ═ 2:1) to give intermediate product B (70mg) as a yellow oil. LC-MS [ M + H ]] + :m/z 641.3。
The second step is that: the above intermediate product B (70mg) was dissolved in 6N HCl aqueous solution (5mL), heated to 70 ℃ and reacted with stirring for 4 hours. The reaction solution was cooled to room temperature, neutralized to pH 8-9 by adding aqueous ammonia, and extracted three times with dichloromethane (50 mL). The separated organic phase was over MgSO 4 Drying, filtering and concentrating. The crude product was purified by HPLC prep. to give the desired product as a yellow solid (37 mg). LC-MS [ M + H ]] + :m/z 407.2。 1 H-NMR(400MHz,MeOD-d 4 ):δ7.73(s,1H),7.49-7.32(m,4H),5.83(t,J=6.0Hz,1H),4.91-4.83(m,2H),4.53-4.43(m,2H),3.81-3.71(m,2H),3.41-3.37(m,1H),3.08-2.99(m,3H),2.07-2.04(m,2H),1.61-1.52(m,2H),1.29(d,J=6.8Hz,6H)。
Chiral preparative separation of the A1 intermediate gave intermediates A1-1 and A1-2, respectively, which were deprotected in accordance with the procedure described above for the second reaction to give examples 1-1 and 1-2. The preparation conditions are as follows: separation column (SunFire Prep C18 OBDTM,10um, 19 x 250 mm); gradient (5% -95% acetonitrile/0.1% formic acid/water, 16min, flow 20 mL/min). Analysis conditions were as follows: analytical column (Waters SunFire C18, 4.6 x 50mm, 5 um); gradient (5% -95% acetonitrile/0.1% formic acid/water, 3.0min, flow rate 2.0mL/min, 2.6 min); column temperature: 40 ℃; detection wavelength: 254 nM.
Figure BDA0003474551550000211
Examples 1-1 and 1-2.
Starting from intermediate a, and various aromatic and saturated ring-substituted amines, the syntheses described in example 1 gave examples 2-36;
Figure BDA0003474551550000212
Figure BDA0003474551550000221
Figure BDA0003474551550000231
Figure BDA0003474551550000241
example 17: 1- (8-isopropyl-4- ((-1,2,3, 4-tetrahydroisoquinolin-4-yl) amine) pyrazolo [1,5, -a ] [1,3,5] triazin-2-yl) piperidin-4-ol
Figure BDA0003474551550000242
The first step is as follows: intermediate B (73mg,0.3mmol), tert-butyl 4-amino-3, 4-dihydroisoquinoline-2 (1H) carbonate (74mg,0.32mmol), DIEA (78mg,0.6mmol) was dissolved in acetonitrile (10mL) under nitrogen and reacted at 70 ℃ for 4H. Concentrating the reaction solution under reduced pressure, and purifying the crude product by silica gel column chromatography (PE: EA ═ 2:1) to obtain a yellow oily intermediateProduct (80 mg). LC-MS [ M + H ]] + :m/z 455.6。
The second step is that: to a solution of the above intermediate compound (80mg,0.17mmol) in dichloromethane (10mL) was added m-chloroperoxybenzoic acid (75mg, 0.34mmol, 80% purity) with ice bath cooling. After the reaction temperature was slowly raised to room temperature, the reaction was stirred for further 3 hours. After the LC-MS detection reaction is completed, adding saturated NaSO into the reaction solution 3 Aqueous solution (10mL) and dichloromethane (20mL) and the organic phase was separated. The aqueous phase was washed twice more with dichloromethane (20 mL). The organic phases are combined and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (PE: EA ═ 2:1) to give the product as a yellow solid (50 mg). LC-MS [ M + H ]] + :m/z 487.6。
The third step: the above intermediate compound (50.0mg, 0.10mmol) and tetrahydroxypiperidine (61mg, 0.6mmol) were dissolved in NMP (1mL), DIEA (68mg, 0.62mmol) was added, and the mixture was heated to 115 ℃ and reacted overnight. After completion of the TLC detection, water (40mL) and ethyl acetate (40mL) were added. The separated aqueous phase was extracted twice with ethyl acetate (40 mL). The combined organic phases were concentrated under reduced pressure and dried to give an intermediate (30mg) as a white solid. LC-MS [ M + H ]] + :m/z 508.6.
The fourth step: trifluoroacetic acid (3mL) was added to the above intermediate compound (30mg,0.06mmol) in dichloromethane (10mL), and the reaction was stirred at room temperature for 4 hours. Ammonia was added to neutralize to pH 8-9 and extracted three times with dichloromethane (30 mL). The combined organic phases were concentrated under reduced pressure. The crude product was purified by HPLC prep. to give the desired product as a white solid (21 mg). LC-MS [ M + H ]] + :m/z 408.1。 1 H NMR(400MHz,MeOD-d 4 ):δ7.75(s,1H),7.50-7.52(m,1H),7.40-7.42(m,2H),7.32-7.34(m,1H),5.81(m,1H),4.47-4.52(m,2H),4.37-4.39(m,2H),3.87-3.91(m,1H),3.75-3.79(m,2H),3.47-3.40(m,2H),3.05-3.09(m,1H),1.89-1.93(m,2H),1.51(m,2H),1.24-1.31(d,J=7.2Hz,6H).
Intermediate B and different aromatic and saturated ring-substituted amines and heterocycloalkyl alcohols or heterocycloalkylamines as starting materials are synthesized according to the method of example 17 to give examples 18-26;
Figure BDA0003474551550000251
Figure BDA0003474551550000261
Figure BDA0003474551550000271
example 27: n- (5- (4- (dimethylamino) piperidin-1-yl) -3-isopropylpyrazolo [1, 5-a ] pyridin-7-yl) -1,2,3, 4-tetrahydroisoquinolin-4-amine
Figure BDA0003474551550000272
The first step is as follows: intermediate C (60.0mg, 0.26mmol) was dissolved in ethanol (5mL), DIEA (1mL) and tert-butyl 4-amino-3, 4-dihydroisoquinoline-2 (1H) carbonate (74mg,0.32mmol) were added, and the mixture was stirred under reflux for 8 hours. After completion of the reaction was detected by LC-MS, water (50mL) and ethyl acetate (50mL) were added to the reaction mixture, followed by extraction twice with ethyl acetate (50mL), and the separated organic phase was concentrated under reduced pressure and dried to give a yellow solid intermediate product (92 mg). LC-MS [ M + H ]] + :m/z 443.1。
The second step is that: to a solution of the above intermediate (92mg,0.21mmol) in toluene (10mL) was added 4-N, N-dimethylaminopiperidine (54mg,0.42mmol), Pd 2 (dba) 3 (3mg,0.003mmol), potassium tert-butoxide (36mg, 0.32mmol) and BINAP (2mg, 0.003mmol) were heated to 100 ℃ and the reaction was stirred overnight. After the reaction was checked by LC-MS to be substantially complete, water (50mL) and methylene chloride (50mL) were added to the reaction mixture. The organic phase was separated and the aqueous phase was extracted once more with dichloromethane (50 mL). The organic phases were combined and concentrated under reduced pressure and the crude product was purified by silica gel column chromatography (PE: EA ═ 2:1) to give the compound (65mg) as a yellow solid. MS M/z 664.8[ M + H ]] +
The third step: to the intermediate (65mg, 0.1mmol) in dichloromethane (10mL) was added trifluoroacetic acid (3mL), and the reaction was stirred at room temperature for 4 hours. Adding ammonia water to neutralize until pH is 8-9, and using dichloroMethane (30mL) was extracted three times. The combined organic phases were concentrated under reduced pressure. The crude product was purified by HPLC prep. to give the desired product as a white solid (18 mg). MS M/z 434.6[ M + H ]] + . 1 H NMR(400MHz,CD 3 OD):δ7.36(s,1H),7.50-7.42(m,3H),7.36-7.34(m,1H),5.46(m,1H),4.61-4.50(m,2H),4.53-4.35(m,2H),3.97-3.78(m,2H),3.48-3.41(m,1H),3.16-3.07(m,3H),2.92(s,6H),2.12-2.07(m,2H),1.73-1.62(m,2H),1.32-1.28(d,J=6.8Hz,6H).
Synthesized according to the procedure for example 27, starting from intermediates C or E, and different aromatic and saturated ring-substituted amines and heterocycloalkyl-substituted amines or alcohols, to give examples 28-39;
Figure BDA0003474551550000281
Figure BDA0003474551550000291
Figure BDA0003474551550000301
example 40: 2- (4-aminophenyl) -N- (2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) -1,2,3, 4-tetrahydroisoquinolin-4-amine
Figure BDA0003474551550000302
The first step is as follows: intermediate A1(60.0mg, 0.093mmol) was dissolved in methanol (10mL), 4M HCl/MeOH (1mL) was added, and the mixture was stirred at room temperature for 2 h. After completion of the reaction was detected by LC-MS, water (40mL) and ethyl acetate (40mL) were added to the reaction mixture, followed by extraction with ethyl acetate (40mL) twice, and the separated organic phase was concentrated under reduced pressure and dried to give a yellow solid intermediate product (40.0 mg). LC-MS [ M + H ]] + :m/z 541.7。
The second step is that: to a solution of the above intermediate (40mg,0.074mmol) in DMF (5mL) was addedAnhydrous K 2 CO 3 (21mg,0.15mmol) and p-fluoronitrobenzene (13mg,0.092mmol), the reaction was heated to 130 ℃ overnight. After the reaction was checked by LC-MS to be substantially complete, water (30mL) and ethyl acetate (30mL) were added to the reaction mixture. The organic phase was separated and the aqueous phase was extracted once more with ethyl acetate (30 mL). The organic phases were combined and concentrated under reduced pressure and the crude product was purified by silica gel column chromatography (PE: EA ═ 2:1) to give the compound as a yellow oil (24 mg). LC-MS [ M + H ]] + :m/z 662.8。
The third step: Pd/C (10% wt,5mg) was added to a methanol solution (10mL) of the above intermediate (24mg, 0.037mmol), and the reaction mixture was stirred at room temperature for 3 hours under a hydrogen atmosphere of 1 atm. LC-MS detects the reaction is finished, and the reaction liquid is filtered by diatomite and then is concentrated under reduced pressure. After drying, the product was obtained as a white solid (14 mg). LC-MS [ M + H ]] + :m/z 498.7。 1 H NMR(400MHz,MeOD-d 4 ):δ7.66(s,1H),7.46(d,J=7.6Hz,2H),7.26-7.33(m,2H),7.02-6.88(m,3H),6.69(d,J=7.6Hz,1H),5.61-5.63(m,1H),4.97-4.92(m,2H),4.62-4.45(m,2H),3.89-3.75(m,2H),3.48-3.38(m,1H),3.07-3.00(m,3H),2.08-2.04(m,2H),1.63-1.56(m,2H),1.28(d,J=6.8Hz,6H)。
Synthesized according to the procedure of example 40, intermediate a was reacted with 2-chloro-4-trifluoromethylpyridine to give example 41;
Figure BDA0003474551550000311
m/z:552.3[M+H] + . 1 H NMR(400MHz,CD 3 OD):δ8.27(m,1H),7.75(s,1H),7.34(s,1H),7.50-7.34(m,5H),5.46(m,1H),4.61-4.53(m,2H),4.53-4.43(m,2H),3.97-3.78(m,2H),3.48-3.40(m,1H),3.16-3.06(m,3H),2.12-2.09(m,2H),1.73-1.63(m,2H),1.32-1.29(d,J=7.2Hz,6H).
example 42: 2- (3-aminophenyl) -N- (2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) -1,2,3, 4-tetrahydroisoquinolin-4-amine
Figure BDA0003474551550000312
The first step is as follows: n- (1,2,3, 4-tetrahydroisoquinolin-4-yl) formyl tert-butyl ester (405mg,1.63mol), 3-nitrobromobenzene (300mg,1.48mol), Pd2(dba)3(67.8mg,0.074mmol), Xantphos (70mg,0.148mmol) and cesium carbonate (1.4g,4.44mol) were dissolved in 1,4-dioxane (10mL) under nitrogen. The reaction was heated to 110 ℃ overnight. The reaction mixture was filtered through celite, the filtrate was concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 20:1) to give an intermediate compound (322mg) as a yellow oil. LC-MS [ M + H ] +: M/z 370.1.
The second step is that: the intermediate compound (322mg) was dissolved in dichloromethane (15mL), and trifluoroacetic acid (8mL) was added thereto, followed by stirring at room temperature for 3 hours. The reaction solution was concentrated under reduced pressure to give a crude intermediate compound (250mg) as a pale black oil. LC-MS [ M + H ]] + :m/z 270.1。
The third step: the above intermediate compound (65.5mg,0.24mmol) and intermediate A (80mg,0.19mmol) were dissolved in acetonitrile (5mL) and N, N-diisopropylethylamine (72.4mg,0.56mmol) was added. The reaction was stirred at room temperature for 16 hours. The reaction solution was concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate ═ 5:1) to give a yellow solid intermediate compound (102 mg). LC-MS [ M + H ]] + :m/z 662.2。
The fourth step: the above intermediate compound (92mg,0.13mmol) was dissolved in MeOH/THF/H 2 To a mixed solution of O (4mL/4mL/4mL), ammonium chloride (46mg,0.08mmol) and zinc powder (28.5mg,0.08mmol) were added in this order, and the mixture was stirred at room temperature for 16 hours. The reaction was checked by LC-MS to be substantially complete, and the reaction solution was poured into water (20mL) and extracted twice with ethyl acetate (30 mL). The combined organic phases were dried and concentrated under reduced pressure and the crude product purified by Prep-TLC (eluent: petroleum ether/ethyl acetate ═ 2:1) to give the intermediate compound (78mg) as a yellow solid. LC-MS [ M + H ]] + :m/z 632.3。
The fifth step: the intermediate compound (10.3mg,0.016mmol) was dissolved in ethyl acetate (1mL), Pd/C (5mg) was added, and the mixture was purged with hydrogen balloon 3 times, and stirred at room temperature overnight for 3 days. Celite filtration and washing of the filtrate with methanol, concentration of the filtrate under reduced pressure and preparative HPLC separation of the resulting crude gave example 42 as a grey solid (1.04mg)] + :m/z 498.3。 1 H NMR(400MHz,MeOD-d 4 ):δ7.66(s,1H),7.46(d,J=7.6Hz,1H),7.26-7.33(m,4H),7.02(d,J=9.6Hz,1H),6.88(s,1H),6.69(d,J=7.6Hz,1H),5.61-5.63(m,1H),4.92-4.97(m,2H),4.62(m,1H),4.45(m,1H),3.94-3.79(m,2H),3.38-3.48(m,1H),3.00-3.07(m,3H),2.04-2.08(m,2H),1.56-1.62(m,2H),1.28(d,J=6.8Hz,6H)。
Synthesized according to the procedure of example 40, intermediate a was reacted with 2-chloro-4-cyanopyrimidine to provide example 43;
Figure BDA0003474551550000321
m/z:510.3[M+H] +1 H NMR(400MHz,CD 3 OD):δ8.15(s,2H),7.34(s,1H),7.50-7.40(m,3H),7.36-7.34(m,1H),5.46(m,1H),4.61-4.53(m,2H),4.53-4.43(m,2H),3.97-3.78(m,2H),3.48-3.40(m,1H),3.16-3.06(m,3H),2.12-2.09(m,2H),1.73-1.63(m,2H),1.32-1.29(d,J=7.2Hz,6H).
prepared according to the method of example 42, intermediate obtained by reacting tert-butyl-N- (1,2,3, 4-tetrahydroquinolin-4-yl) formyl tert-butyl ester with 3-nitrobromobenzene was reacted with intermediate A to obtain example 44;
Figure BDA0003474551550000322
m/z:498.3[M+H] +1 H NMR(400MHz,MeOD-d 4 ):δ7.68(s,1H),7.46(m,1H),7.26-7.02(m,5H),6.88-6.69(m,2H),5.61-5.63(m,1H),4.92-4.97(m,2H),4.62(m,1H),4.45(m,1H),3.94-3.79(m,2H),3.38-3.48(m,1H),3.00-3.07(m,3H),2.04-2.08(m,2H),1.56-1.62(m,2H),1.30(d,J=6.8Hz,6H)。
example 45: n- (2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) -2- (piperidin-3-yl) -1,2,3, 4-tetrahydroisoquinolin-4-amine
Figure BDA0003474551550000331
The first step is as follows: to a solution of intermediate A2(60mg, 0.11mmol) in 1, 2-dichloroethane (10mL) was added N-tert-butoxycarbonyl-3-piperidone (22mg,0.11mmol) and acetic acid (0.5 mL). After the reaction mixture was stirred at room temperature for 30 minutes, sodium cyanoborohydride (15mg,0.24mmol) was added to the reaction. The reaction was stirred at room temperature overnight. After completion of the reaction was checked by LC-MS, water (30mL) and methylene chloride (50mL) were added to the reaction mixture. After separation of the organic phase, the aqueous phase was extracted once more with dichloromethane (50 mL). The combined organic phases were concentrated under reduced pressure and dried to give an oily intermediate compound (51 mg).
The third step: the above compound (51mg, 0.07mmol) was dissolved in HBr/AcOH (2mL), reacted overnight, and NaOH solution (10mL) and dichloromethane (30mL) were added. The organic phase was separated and the aqueous phase was extracted twice more with dichloromethane (30 mL). The combined organic phases were concentrated under reduced pressure and the crude product isolated by HPLC prep to give a pale yellow solid (15 mg). LC-MS [ M + H ]] + :m/z 490.3。 1 H NMR(400MHz,MeOD-d 4 ):7.72(s,1H),7.49-7.44(m,1H),7.36-7.21(m,4H),5.69-5.67(m,1H),4.25-4.15(m,1H),4.06-3.95(m,1H),3.53-3.34(m,4H),3.26-3.02(m,7H),2.09-2.97(m,4H),1.96-1.92(m,1H),1.89-1.57(m,3H),1.28(d,J=6.8Hz,6H)。
Synthesized according to the procedure of example 45, intermediate a2 or an analog thereof was reacted with various commercially available aldehydes or ketones to afford examples 46-55;
Figure BDA0003474551550000332
Figure BDA0003474551550000341
Figure BDA0003474551550000351
example 56: (E) -N- (4- (4- ((2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) amino) -3, 4-dihydroisoquinolin-2- (1H) -yl) phenyl) -4- (dimethylamino) but-2-enoyl
Figure BDA0003474551550000352
The first step is as follows: to a solution of intermediate A3(60mg, 0.09mmol) in ethanol (10mL) and water (5mL) were added iron powder (30mg,0.54mmol) and ammonium chloride (30mg,0.56 mmol). The reaction solution was heated to 80 ℃ and stirred for 5 hours. Detecting the reaction completion by LC-MS, filtering the reaction solution, and concentrating the filtrate under reduced pressure. The crude product was dried to give an off-white solid (51.2 mg).
The second step is that: to a solution of the above intermediate (51mg, 0.08mmol) in dichloromethane (10mL) was added (E) -3-N, N-dimethylaminomethylacryloyl chloride hydrochloride (17mg,0.09mmol) and DIEA (40mg,0.31mmol). The reaction was stirred at room temperature overnight. After completion of the reaction was detected by LC-MS, water (30mL) and ethyl diacetate (50mL) were added to the reaction mixture. After separation of the organic phase, the aqueous phase was extracted twice more with ethyl acetate (50 mL). The combined organic phases were concentrated under reduced pressure and dried to give an oily intermediate compound (52 mg).
The third step: the above compound (52mg, 0.07mmol) was dissolved in HBr/AcOH (2mL), reacted overnight, and NaOH solution (10mL) and dichloromethane (30mL) were added. The organic phase was separated and the aqueous phase was extracted twice more with dichloromethane (30 mL). The combined organic phases were concentrated under reduced pressure and the crude product isolated by HPLC prep to give a pale yellow solid (15 mg). LC-MS [ M + H ]] + :m/z 690.8。1H NMR(400MHz,MeOD-d4):δ7.67(s,1H),7.48-7.43(m,2H),7.30-7.25(m,4H),7.10(m,1H),6.81-6.89(m,2H),6.53(m,1H),5.64(m,1H),4.90-4.95(m,2H),4.57(m,1H),4.43(m,1H),3.97-3.99(m,2H),3.72-3.85(m,2H),3.36-3.49(m,2H),3.01-3.06(m,3H),2.92(s,6H),2.06-2.09(m,2H),1.54-1.66(m,2H),1.29(d,J=6.8Hz,3H).
Synthesized according to the procedure for example 56, intermediate a2 or an analog thereof was reacted with various commercially available carboxylic acids or acid chlorides to afford examples 57-63;
Figure BDA0003474551550000361
Figure BDA0003474551550000371
synthesis according to the procedures of examples 27 and 56 using intermediate D with various commercially available carboxylic acids or acid chlorides gave examples 64-66;
Figure BDA0003474551550000372
Figure BDA0003474551550000381
example 67: n- (2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) -2- (4-aminopyrimidin-2-yl) -1,2,3, 4-tetrahydroisoquinolin-4-amine
Figure BDA0003474551550000382
The first step is as follows: under the protection of nitrogen, the compound N- (1,2,3, 4-tetrahydroisoquinolin-4-yl) formyl tert-butyl ester (400mg,1.6mol), 2-chloropyrimidin-4-amine (418mg,3.2mol), N, N-diisopropylethylamine (619.2mg,4.8mmol) was dissolved in DMF (8mL) and heated to 80 ℃ for reaction overnight. After filtration through celite, the filtrate was concentrated under reduced pressure and the resulting crude product was purified by column chromatography (eluent: petroleum ether/ethyl acetate 10:1) to give a grey solid (96 mg). LC-MS [ M + H ] +: M/z 342.1.
The second step is that: the above intermediate compound (76mg) was dissolved in methylene chloride (1.5mL), and trifluoroacetic acid (0.8mL) was added thereto and the mixture was stirred at room temperature for 3 hours. The reaction mixture was concentrated under reduced pressure to give a crude intermediate compound (65mg) as a pale black oil. LC-MS [ M + H ]] + :m/z 242.2。
The third step: the above intermediate compound (45mg,0.18mmol) and intermediate A (65mg,0.27mmol) were dissolved in acetonitrile (5mL), and N, N-diisopropylethylamine (104mg,0.81mmol) was added, and the reaction was stirred at room temperature for 16 hours. The reaction mixture is concentrated under reduced pressure and the crude product obtained is purified by column chromatography (eluent: petroleum ether/ethyl acetate 5:1) to give greySolid intermediate compound (110 mg). LC-MS [ M + H ]] + :m/z 663.2。
The fourth step: the above intermediate compound (21mg,0.03mmol) was dissolved in acetonitrile (5mL), and trimethyliodosilane (0.5mL) was added, and the mixture was stirred at room temperature overnight. Water (30mL) was added to the reaction mixture, which was then extracted twice with ethyl acetate (30 mL). The combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure and the crude product was purified by HPLC prep. to give example 67 compound as an off-white solid (5.74mg)] + :m/z 499.3。1H NMR(400MHz,MeOD-d 4 ):δ7.67(s,1H),7.65(d,J=7.2Hz,1H),7.45(d,J=7.6Hz,1H),7.40-7.37(m,1H),7.33-7.29(m,2H),6.14(d,J=7.2Hz,1H),5.69(m,1H),5.11(m,1H),4.95-4.94(m,1H),4.81(m,1H),4.57-4.54(m,1H),4.02(m,1H),3.45-3.39(m,1H),3.09-3.00(m,3H),2.11-2.05(m,2H),1.64-1.58(m,2H),1.28(d,J=6.8Hz,6H)。
Example 68: n- (2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) -2- (4-aminopyridin-2-yl) -1,2,3, 4-tetrahydroisoquinolin-4-amine
Figure BDA0003474551550000391
The first step is as follows: under the protection of nitrogen, the compounds N- (1,2,3, 4-tetrahydroisoquinolin-4-yl) formyl tert-butyl ester (400mg,1.6mol), 2-chloro-4-nitropyridine (201.4mg,1.2mol), Pd2(dba)3(55.4mg,0.06mmol), Xantphos (57.6mg,0.12mmol) and cesium carbonate (1.2g,3.63mol) were dissolved in 1,4-dioxane (10 mL). The reaction solution was heated to 110 ℃ and reacted overnight. The reaction solution was filtered through celite, and then concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 10:1) to give the intermediate product (26.5mg) as a yellow solid. LC-MS [ M + H ] +: M/z 371.1.
The second step: the above intermediate compound (26.5mg) was dissolved in dichloromethane (2mL), followed by addition of TFA (1mL), stirring at room temperature for 3 hours, and concentration under reduced pressure to give the intermediate product as a dark gray oil (21 mg). LC-MS [ M + H ]] + :m/z 271.1
The third step: the intermediate compound (21mg,0.08mmol) and intermediate A were added(33.4mg,0.08mmol) was dissolved in acetonitrile (2mL) and N, N-diisopropylethylamine (30.2mg,0.23mmol) was added and the reaction stirred at room temperature for 16 h. The reaction solution was concentrated under reduced pressure, and the resulting crude product was purified by Prep-TLC (eluent: petroleum ether/ethyl acetate 4:1) to give a yellow solid intermediate product (29 mg). LC-MS [ M + H ]] + :m/z 663.2。
The fourth step: the above intermediate compound (29mg,0.04mmol) was dissolved in MeOH/THF/H 2 To O (2mL/2mL/2mL), ammonium chloride (46mg,0.08mmol) and zinc powder (28.5mg,0.08mmol) were added in this order, and the mixture was stirred at room temperature for 16 hours. Water (8mL) was added to the reaction solution, and the mixture was extracted twice with ethyl acetate (20 mL). The combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure and the crude product purified by Prep-TLC (eluent: dichloromethane/methanol-50: 1) to give the intermediate product as a white solid (20 mg). LC-MS [ M + H ]] + :m/z 633.3。
The fifth step: the above intermediate compound (5mg) was dissolved in acetonitrile (1mL), and trimethyliodosilane (0.3mL) was added thereto, followed by stirring at room temperature overnight. Water (20mL) was added to the reaction mixture, which was then extracted twice with ethyl acetate (20 mL). The combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure and the crude product was purified by HPLC preparative to give the intermediate product as an off-white solid (2.02mg)]+:m/z 499.3。 1 H NMR(400MHz,MeOD-d 4 ):δ7.68(s,1H),7.52(d,J=6.8Hz,1H),7.45(d,J=7.6Hz,1H),7.40-7.30(m,3H),6.32(m,1H),6.16(m,1H),5.72-5.70(m,1H),4.82(s,1H),4.68(m,1H),4.09-4.04(m,1H),3.93-3.88(m,1H),3.43-3.37(m,1H),3.06-2.98(m,3H),2.09-2.06(m,2H),1.61-1.55(m,2H),1.33-1.27(m,8H)。
Example 69: (E) -N- (3- (4- ((2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) amino) -3, 4-dihydroisoquinolin-2 (1H) -yl) phenyl) -4- (dimethylamino) but-2-enoyl
Figure BDA0003474551550000401
The first step is as follows: the compound (1- (4- ((2- (3-aminophenyl) -1,2,3, 4-tetrahydroisoquinolin-4-yl) amino) -8-isopropylpyrazolo [1,5, -a)][1,3,5]Triazin-2-yl) piperidin-4-yl) carbonoyl benzyl ester (64mg,0.1mmol) was dissolved in pyridine/dichloromethane (2mL/1mL) and (E) -4- (dimethylamino) but-2-enoic acid (20.2mg,0.12mmol), one drop of POCl was added 3 Stirred at room temperature for 1.5 hours, and saturated NaHCO was poured in 3 To a solution of water (20mL), ethyl acetate (20mL) was extracted three times. The combined organic phases were dried over anhydrous sodium sulfate and the filtrate was concentrated under reduced pressure. The crude product was slurried with methanol, the filter cake washed with methanol, and the combined filtrates concentrated to give a crude intermediate product (34mg) as a yellow solid. LC-MS [ M + H ]] + :m/z 743.7。
The second step is that: the above intermediate compound (29mg,0.04mmol) was dissolved in acetonitrile (1mL), and trimethyliodosilane (42.2mg,0.19mmol) was added, and the mixture was stirred at room temperature overnight. The reaction was quenched by addition of methanol, the reaction was concentrated under reduced pressure, and the crude product was purified by HPLC prep. to give example 69 (5.1mg) as a pale yellow solid. LC-MS [ M + H ] +: M/z 609.4. 1H NMR (400MHz, MeOD-d4): δ 7.67(s,1H),7.45-7.48(m,2H),7.30-7.32(m,2H),7.21-7.27(m,2H),7.10(d, J ═ 8.4Hz,1H),6.81-6.89(m,2H),6.53(m,1H),5.61-5.64(m,1H),4.90-4.95(m,2H),4.57(m,1H),4.43(m,1H),3.97-3.99(m,2H),3.72-3.85(m,2H),3.36-3.49(m,2H),3.01-3.06(m,3H),2.92(s,6H),2.06-2.09(m,2H), 1.54-6H), 1.29.54 (m, 6H), 1H, 6H).
Example 70: (E) -1- (3- (4- ((2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) amino) -3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-1-yl) -4- (dimethylamino) but-2-enoyl
Figure BDA0003474551550000411
The first step is as follows: the intermediate compound 3- (4- ((2- (4- (((benzyloxy) carbonyl) amino) piperidin-1-yl) -8-isopropylpyrazolo [1,5, -a)][1,3,5]Triazin-4-yl) amino) -3, 4-dihydroisoquinolin-2 (1H) -yl) piperidine-1-carboxylic acid tert-butyl ester (30mg,0.04mmol) was dissolved in dichloromethane (2mL), followed by addition of TFA (1mL) and stirring at room temperature for 3 hours. The reaction mixture was concentrated under reduced pressure to give a crude intermediate compound (20mg) as a pale black oil. LC-MS [ M + H ]] + :m/z 624.3。
Second step of: the above intermediate compound (34mg,0.055mmol) was dissolved in DMF (2mL), followed by addition of (E) -4- (dimethylamino) but-2-enoic acid (9.94mg,0.06mmol), HATU (27mg,0.07mmol), DIEA (21mg,0.16mmol), and stirring at room temperature for 5 hours. Water (8mL) was added to the reaction mixture, and a solid precipitated. The precipitated solid was dried to give a crude intermediate compound (39mg) as a white solid. LC-MS [ M + H ]] + :m/z 735.5。
The third step: the above intermediate compound (39mg, 0.05mmol) was dissolved in acetonitrile (1mL), and trimethyliodosilane (0.3mL) was added thereto, followed by stirring at room temperature for 5 hours. Water (20mL) was added to the reaction mixture, which was then extracted twice with ethyl acetate (20 mL). The combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure and the crude product was purified by HPLC prep. to give example 70 as a grey solid (12.9mg)] + :m/z 601.5。 1 H NMR(400MHz,MeOD-d 4 ):δ7.73(s,1H),7.46-7.34(m,4H),6.98-6.93(m,1H),6.72-6.66(m,1H),5.97-5.92(m,1H),4.90-4.91(m,1H),4.78-4.66(m,2H),4.61-4.54(m,3H),4.05-3.92(m,4H),3.76-3.63(m,1H),3.51-3.33(m,4H),3.13-2.98(m,3H),2.90(s,6H),2.35-2.31(m,1H),2.04-1.98(m,4H),1.67-1.54(m,3H),1.29(d,J=6.8Hz,6H)。
Synthesized according to the procedures of examples 69 and 70 using intermediate a3 or an analog thereof reacted with various commercially available carboxylic acids or acid chlorides to afford examples 71-73;
Figure BDA0003474551550000421
example 74: n- (2- (4- ((2- (4-aminopyridin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) amino) -3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-4-yl) acrylamide
Figure BDA0003474551550000422
The first step is as follows: the intermediate compound (1- (4- ((2- (4-aminopyridin-2-yl) -1,2,3, 4-tetrahydroisoquinolin-4-yl) amino) -8-isopropylpyrazolo [1,5, -a)][1,3,5]Triazine-2-yl)Piperidin-4-yl) formylbenzyl ester (15mg,0.02mmol), potassium phosphate (20mg,0.08mmol) in THF/H 2 To the mixed solution of O (6mL/3mL), 3-chloropropionyl chloride (4mg,0.02mmol) was added, and the reaction was carried out at room temperature for 2 hours. After dilution with water (30mL), the mixture was extracted three times with dichloromethane (30mL), and the combined organic phases were dried over anhydrous sodium sulfate. The filtrate was concentrated under reduced pressure to give the crude intermediate product (18mg) as a grey solid.
LC-MS[M-H] - :m/z 721.4。
The second step is that: the above intermediate compound (18mg,0.02mmol) was dissolved in trifluoroacetic acid (3mL) and reacted at room temperature for 5 hours. After the reaction mixture was diluted with 30mL of water, it was extracted three times with methylene chloride (30 mL). The combined organic phases are dried over anhydrous sodium sulfate, and the filtrate is concentrated under reduced pressure to obtain a crude intermediate. The crude intermediate was dissolved in THF/H 2 To a mixed solvent of O (6mL/3mL) was added NaOH (2mg,0.04mmol), and the reaction mixture was stirred overnight at room temperature. To the above reaction solution was added water (50mL) for dilution, and extracted three times with dichloromethane (50mL), the combined organic phases were dried over anhydrous sodium sulfate, the filtrate was concentrated under reduced pressure, and the resulting crude product was prepared by HPLC to give example 74 compound (4.35mg) as a yellow solid. LC-MS [ M + H ]] + :m/z 553.3。 1 H NMR(400MHz,DMSO-d 6 ):δ8.67(s,1H),8.02(d,J=6.0Hz,1H),7.86-7.82(m,2H),7.75(s,1H),7.36-7.30(m,3H),7.26-7.22(m,1H),6.98(d,J=6.0Hz,1H),6.47-6.40(m,1H),6.33(dd,J=17.0,2.0Hz,1H),5.87(d,J=11.6Hz,1H),5.63-5.59(m,1H),4.84-4.56(m,5H),4.17-4.11(m,2H),4.03-3.92(m,2H),3.31-3.25(m,1H),3.01-2.84(m,3H),1.95-1.91(m,2H),1.55-1.35(m,2H),1.25(d,J=6.8Hz,6H)。
Example 75: n- (2- (4- ((2- (4-aminopyrimidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) amino) -3, 4-dihydroisoquinolin-2 (1H) -yl) piperidin-4-yl) acrylamide
Figure BDA0003474551550000431
The first step is as follows: the intermediate compound (1- (4- ((2- (4-aminopyrimidin-2-yl) -1,2,3, 4-tetrahydroisoquinolin-4-yl) amino) -8-isopropylpyrazolo [1,5, -a)][1,3,5]Triazine-2-yl) piperidin-4-yl) carbonoyl benzyl ester (60mg,0.09mmol), triethylamine (67mg,0.66mmol) were dissolved in DCM (6mL), and 3-chloropropionyl chloride (60mg,0.47mmol) was added under cooling on an ice bath for reaction at room temperature for 2 hours. After dilution with water (20mL), the mixture was extracted three times with dichloromethane (20mL), and the combined organic phases were dried over anhydrous sodium sulfate. The filtrate was concentrated under reduced pressure to give the crude intermediate product (68mg) as a grey solid. LC-MS [ M-H ]] - :m/z 722.5。
The second step is that: the intermediate compound (68mg,0.09mmol) was dissolved in acetonitrile (3mL), and TMSI (3drops) was added dropwise to the reaction mixture, followed by reaction at room temperature for 1 hour. The reaction was quenched by addition of methanol (3 mL). Concentrating under reduced pressure, and dissolving the crude product in THF/H 2 To a mixed solution of O (6mL/3mL), NaOH (38mg,0.94mmol) was added, and the reaction was carried out at room temperature for 5 hours. Dilute with water (50mL), extract three times with dichloromethane (50mL), dry the combined organic phases over anhydrous sodium sulfate, concentrate the filtrate under reduced pressure, and prepare the crude product by HPLC to give example 75 as a grey solid (1.6 mg). LC-MS [ M + H ]] + :m/z 554.3。 1 H NMR(400MHz,CD 3 OD):δ8.19(d,J=5.6Hz,1H),7.64(s,1H),7.48(d,J=5.6Hz,1H),7.43(d,J=7.6Hz,1H),7.34-7.25(m,3H),6.46-6.42(m,2H),5.82(dd,J=9.2,2.8Hz,1H),5.57-5.55(m,1H),5.11(d,J=17.2Hz,1H),4.97-4.86(m,3H),4.48-4.40(m,1H),4.28-4.24(m,1H),3.41-3.36(m,1H),3.06-3.00(m,3H),2.07-2.03(m,2H),1.62-1.58(m,2H),1.28(d,J=6.8Hz,6H).
Synthesized according to the procedure for example 56, intermediate a2 or an analog thereof was reacted with various commercially available carboxylic acids or acid chlorides to afford examples 76-77;
Figure BDA0003474551550000441
example 78: r-1- ((E) -4- (dimethylamino) but-2-enyl) piperidin-3-yl-4- ((8-isopropyl-2- (((R) -piperidin-3-yl) -oxy) pyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) amino) -3, 4-dihydroisoquinoline-2 (1H) -carbonyl
Figure BDA0003474551550000451
The first step is as follows: to a solution of 4-amino-3, 4-dihydroisoquinoline-2 (1H) -formylbenzyl ester (500mg,1.77mmol) and triethylamine (335mg,3.3mmol) in methylene chloride (10mL) was added intermediate B (436mg,1.8 mmol). The reaction mixture was reacted at 60 ℃ for 2 hours. After dilution with water (20mL), extraction was performed three times with dichloromethane (20mL), and the combined organic phases were dried over anhydrous sodium sulfate. The filtrate was concentrated under reduced pressure to give the crude intermediate product (630mg) as a grey solid. LC-MS [ M-H ]] - :m/z 489.2。
The second step is that: to a solution of the above intermediate (630mg,1.29mmol) in methylene chloride (10mL) was added m-chloroperoxybenzoic acid (225mg,1.30 mmol). The reaction mixture was reacted at room temperature for 2 hours. After dilution with aqueous sodium thiosulfate (20mL), the mixture was extracted three times with dichloromethane (20mL), and the combined organic phases were dried over anhydrous sodium sulfate. The filtrate was concentrated under reduced pressure to give the crude intermediate product (330mg) as a grey solid. LC-MS [ M-H ]] - :m/z 521.2。
The third step: to the above intermediate compound (101mg,0.19mmol) in N, N-dimethylformamide (1mL) under a nitrogen blanket was added NaH (7mg,0.28 mmol). The reaction mixture was stirred at room temperature for 30 minutes. Then, (S) -3-hydroxypiperidine-1-formyl tert-butyl ester (200mg,1.0mmol) was added thereto, and the mixture was heated to 80 ℃ to react overnight. After the reaction was cooled to room temperature, water (10mL) was added and extracted twice with ethyl acetate (30 mL). The combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure and the resulting crude product was isolated by HPLC prep. to give a white solid (12.4 mg). LC-MS [ M + H ]] + :m/z 642.2。
The fourth step: to a solution of the above intermediate compound (12.4mg,0.02mmol) in ethyl acetate (2mL) was added Pd/C (10mg), and after 3 balloon replacements, the mixture was stirred at room temperature overnight for 2 days. After filtration through celite, concentration under reduced pressure gave crude pale yellow solid (13 mg). LC-MS [ M + H ]] + :m/z 508.3。
The fifth step: to a solution of (S) -3-hydroxypiperidine-1-formylbenzyl ester (100mg,0.43mmol) in tetrahydrofuran (5mL) under nitrogen blanket was added triphosgene (148mg,0.50 mmol). The reaction mixture was stirred at room temperature for 30 minutes. Then adding the above intermediate compound (50mg,0.1mmol), and reacted at room temperature overnight. After water (30mL) was added, the mixture was extracted with ethyl acetate (50 mL). The separated organic phase was dried over anhydrous sodium sulfate, concentrated under reduced pressure and the resulting crude product was isolated by HPLC prep. to give a white solid (40 mg). LC-MS [ M + H ]] + :m/z 769.4。
And a sixth step: to a solution of the above intermediate compound (40mg,0.05mmol) in ethyl acetate (5mL) was added Pd/C (10mg), and the mixture was purged with hydrogen balloon 3 times, followed by stirring overnight at room temperature for 2 days. After filtration through celite, concentration under reduced pressure gave crude product as a pale yellow solid (20 mg). LC-MS [ M + H ]] + :m/z 635.4。
The seventh step: the above intermediate compound (20mg,0.03mmol) was dissolved in DMF (2mL), and (E) -4- (dimethylamino) but-2-enoic acid (6mg,0.04mmol), HATU (16mg,0.04mmol) and DIEA (21mg,0.16mmol) were added in this order and stirred at room temperature for 5 hours. Water (8mL) was added to the reaction mixture, and a solid precipitated. The precipitated solid was dried to give a crude intermediate compound (20mg) as a white solid. LC-MS [ M + H ]] + :m/z 746.4。
Eighth step: the above intermediate compound (20mg,0.03mmol) was dissolved in acetonitrile (1mL), and trimethyliodosilane (0.3mL) was added thereto, followed by stirring at room temperature for 5 hours. After water (20mL) was added to the reaction mixture, the mixture was extracted twice with ethyl acetate (20 mL). The combined organic phases were dried over anhydrous sodium sulfate, concentrated under reduced pressure and the crude product was purified by HPLC prep. to give example 78 compound (5mg) as a grey solid] + :m/z 646.2。 1 H NMR(400MHz,MeOD-d 4 ):δ7.81-7.85(m,1H),7.28-7.39(m,4H),6.59-6.79(m,2H),5.47-5.56(m,2H),3.43-3.87(m,12H),3.05-3.29(m,3H),2.83-2.87(m,6H),1.86-2.18(m,6H),1.30-1.31(m,2H),1.20-1.29(m,7H)。
Synthesized according to the procedure for example 78, intermediate a2 or an analog thereof was reacted with various commercially available carboxylic acids or acid chlorides to afford examples 79-63;
Figure BDA0003474551550000461
Figure BDA0003474551550000471
examples 82 and 83: (R, E) -1- (4- ((2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) amino) -3, 4-dihydroisoquinolin-2 (1H) -yl) -4- (dimethylamino) but-2-enoyl and (S, E) -1- (4- ((2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) amino) -3, 4-dihydroisoquinolin-2 (1H) -yl) -4- (dimethylamino) but-2-enoyl.
Figure BDA0003474551550000472
The first step is as follows: intermediate A2(130mg) was isolated by SFC chiral preparation to give intermediates P1(61mg) and P2(52 mg).
The preparation conditions are as follows: a separation column (
Figure BDA0003474551550000473
OD,250 × 25mm10 μm); gradient (mobile phase: A: SupercriticalcO) 2 B EtOH (+ 0.1% 7.0mol/l AmmoniinMeOH), A: B ═ 60:40, 5min, flow 100 g/min). The retention times (Rt) of P1 and P2 on the preparative column were (Rt ═ 7.551min, P1) and (Rt ═ 6.741min, P2), respectively.
The analysis conditions are as follows: analytical column (
Figure BDA0003474551550000474
OD-H, 250 × 4.6mm,5 μm); gradient (mobile phase: A: SupercriticalcO) 2 B is EtOH (0.1% DEA), A is 60:40, 10min, flow rate 3.0 mL/min); column temperature: 35 ℃; detection wavelength: 214 nM.
In the second step, intermediate compound P1(61.0mg,0.11mol) and (E) -4- (dimethylamino) but-2-enoic acid (20.0mg,0.12mol) were dissolved in anhydrous DMF (3.0mL) and HATU (54.0mg,0.14mol) and DIPEA (43.0mg,0.33mol) were added. The reaction was stirred at room temperature for 4 hours under nitrogen. After completion of the reaction was checked by LCMS, water (8mL) and saturated aqueous sodium carbonate (2mL) were added and a white solid precipitated which was filtered off and washed with water and the filter cake was dried to give the white intermediate product P1-1(56 mg). LC-MS [ M + H ]] + :m/z 652.4。
Referring to the above synthetic procedure for P1-1, the intermediate product P2-1(50mg) was obtained as a white solid.
In the third step, compound P1-1(56.0mg,0.09mmol) was dissolved in acetonitrile (3mL), and trimethylsilyl iodide (0.3mL) was added and the mixture was stirred at room temperature for 2 hours under nitrogen. After the reaction was completed, the reaction was concentrated by LCMS, and then concentrated by adding 10mL of methanol, and this was repeated three times. The concentrated crude product was purified by HPLC prep to give example 82 compound (17.8mg) as a white solid. LC-MS [ M + H ]] + :m/z 518.3。 1 H NMR(400MHz,MeOD-d 4 ):7.66(d,J=6.4Hz,1H),7.45-7.26(m,4H),7.07(d,J=14.8Hz,0.5H),6.84(d,J=15.2Hz,0.5H),6.70-6.57(m,1H),5.52-5.47(m,1H),5.33(d,J=17.6Hz,0.5H),5.07(d,J=16.4Hz,0.5H),4.95-4.88(m,2H),4.83-4.82(m,0.5H),4.63-4.53(m,1H),4.40(dd,J=14.0Hz,4.0Hz,0.5H),3.97-3.90(m,1.5H),3.82-3.73(m,1H),3.55-3.39(m,0.5H),3.38-3.31(m,1H),3.08-3.00(m,3H),2.90(s,3H),2.72(s,3H),2.09-2.07(m,2H),1.66-1.54(m,2H),1.30-1.27(m,6H)。
Referring to the synthesis of the compound of example 82, a white solid, the compound of example 83 (15.6mg) was obtained. LC-MS [ M + H ]] + :m/z 518.3。 1 H NMR(400MHz,MeOD-d 4 ):7.66(d,J=4.8Hz,1H),7.44-7.25(m,4H),7.08(d,J=15.2Hz,0.5H),6.87(d,J=15.2Hz,0.5H),6.71-6.57(m,1H),5.52-5.47(m,1H),5.33(d,J=17.6Hz,0.5H),5.07(d,J=17.8Hz,0.5H),4.94-4.80(m,2H),4.85-4.83(m,0.5H),4.60-4.53(m,1H),4.41(dd,J=14.0Hz,3.6Hz,0.5H),3.97-3.94(m,1.5H),3.85-3.74(m,1H),3.56-3.39(m,0.5H),3.38-3.31(m,1H),3.09-2.91(m,3H),2.90(s,3H),2.72(s,3H),2.10-2.07(m,2H),1.68-1.56(m,2H),1.30-1.27(m,6H)。
By following the procedure of examples 74, 78, 82 and 83, intermediate a2 or an analog thereof was reacted with various commercially available carboxylic acids or acid chlorides to afford examples 84-93;
Figure BDA0003474551550000481
Figure BDA0003474551550000491
Figure BDA0003474551550000501
example 94: 1- ((2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) amino) -2, 3-dihydro-1H-indene-5-carbonitrile
Figure BDA0003474551550000502
The first step is as follows: intermediate A (50mg,0.11mmol), 5-bromo-2, 3-dihydro-1H-inden-1-amine (42mg,0.2mmol), DIEA (78mg,0.6mmol) was dissolved in acetonitrile (5mL) under nitrogen and reacted at 70 ℃ for 4H. The reaction mixture was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (PE: EA ═ 2:1) to give a yellow solid intermediate product (84 mg). LC-MS [ M + H ]] + :m/z 604.2。
The second step is that: to the above intermediate (84mg,0.14mmol) and zinc cyanide (25mg,0.21mmol) in N, N-dimethylformamide (10mL) was added tetrakistriphenylphosphine palladium (24mg,0.02mmol) under nitrogen. The reaction mixture was heated to 110 ℃ for 10 hours. The reaction mixture was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (PE: EA ═ 2:1) to give a white solid intermediate product (50 mg). LC-MS [ M + H ]] + :m/z 551.2。
The third step: to a solution of the above intermediate compound (50mg,0.09mmol) in acetonitrile (3mL) under nitrogen, was added trimethyliodosilane (0.5 mL). Stirred at room temperature for 2 hours. After the reaction was completed by LCMS detection, it was concentrated under reduced pressure, then concentrated again by adding 10mL of methanol, and this was repeated three times. Purification of the concentrated crude by HPLC prep. gave example 94 as a white solid (24 mg). LC-MS [ M + H ]] + :m/z 417.6. 1 H NMR(400MHz,MeOD-d 4 ):8.54(s,1H),7.68(s,1H),7.65(s,1H),7.54(d,J=7.6Hz,1H),7.45(d,J=8.0Hz,1H),5.77(t,J=8.4Hz,1H),4.78-4.72(m,2H),3.31-3.30(m,1H),3.24-3.19(m,1H),3.12-3.05(m,4H),2.90-2.65(m,1H),2.30-2.20(m,1H),1.98-1.94(m,2H),1.54-1.41(m,2H),1.29(d,J=6.8Hz,6H).
Example 95: (3- ((2- (4-aminopiperidin-1-yl) -8-isopropylpyrazolo [1,5, -a ] [1,3,5] triazin-4-yl) amino) -2, 3-dihydro-1H-inden-4-yl) dimethylphosphoryl oxide
Figure BDA0003474551550000511
The first step is as follows: intermediate A (30mg,0.10mmol), 5-bromo-2, 3-dihydro-1H-inden-1-amine (42mg,0.20mmol), DIEA (78mg,0.6mmol) were dissolved in acetonitrile (5mL) under nitrogen and reacted at 70 ℃ for 4H. The reaction mixture was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (PE: EA ═ 2:1) to give a yellow solid intermediate product (34 mg). LC-MS [ M + H ]] + :m/z 602.2。
The second step is that: to a solution of the above intermediate compound (34mg,0.06mmol) in acetonitrile (3mL) under nitrogen, was added trimethyliodosilane (0.5 mL). Stirred at room temperature for 2 hours. Upon completion of the LCMS detection reaction, concentrated under reduced pressure and the crude product purified by HPLC prep. to give example 95 compound (5mg) as a white solid. LC-MS [ M + H ]] + :m/z 468.5. 1 H NMR(400MHz,CDCl 3 ):7.50(s,1H),7.41(m,1H),7.35-7.32(m,2H),6.38(m,1H),5.96(m,1H),4.81-4.72(m,2H),3.26-2.97(m,4H),2.84-2.75(m,4H),2.39(m,2H),2.30(m,2H),2.12(m,2H),1.76-1.68(m,6H),1.20(m,6H).
Synthesized according to the methods of examples 94, 95 using intermediate a2 or an analog thereof reacted with various commercially available carboxylic acids or acid chlorides to afford examples 96-98;
Figure BDA0003474551550000521
test example 1 enzyme Activity test
The compounds of the examples were tested for their inhibition on CDK7/CycH/MAT1(Carna) and CDK9/CycT1(Carna) kinases, with PHA-793887 and Dinaciciclib as positive control compounds. The activity test of the compounds of the examples was performed on 2 kinases using the method of Mobility shift assay.
The specific operation flow is as follows: (1) preparing 1 XKinase buffer; (2) preparation of compound concentration gradient: test compounds were tested starting at 10000nM concentration, diluted 100-fold final concentration in 100% DMSO solution in 384source plates, 3-fold compound dilution, 10 concentrations. Using a dispenser Echo 550 to the target plate 384-well-plate to transfer 250nL 100 times the final concentration of the compound. A2.5 fold final concentration of Kinase solution was prepared using a1 XKinase buffer. (3) Add 10. mu.L of 2.5 fold final concentration kinase solution to the compound well and positive control well, respectively; mu.L of 1 × Kinase buffer was added to the negative control wells. (4) Centrifuge at 1000rpm for 30 seconds, shake the plate and incubate at room temperature for 10 minutes. (5) A mixture of ATP and Kinase substrate at 5/3 fold final concentration was made up using 1 XKinase buffer. (6) The reaction was initiated by adding 15. mu.l of a mixed solution of ATP and substrate at 5/3 fold final concentration. (7) The 384 well plates were centrifuged at 1000rpm for 30 seconds, shaken and mixed and incubated at room temperature for the appropriate time. (8) Add 30. mu.l of termination detection solution to stop the kinase reaction, centrifuge at 1000rpm for 30 seconds, and mix by shaking. (9) The conversion was read using a Caliper EZ Reader. (10) The formula% Inhibition ═ Conversion% max -Conversion%_ sample/ Conversion%_ max -Conversion%_ min X 100, wherein: conversion% \ u sample Is the conversion reading for the sample; conversion% \ u min Is the mean of the negative control wells, representing conversion readings without enzyme live wells; conversion% \ u max Is the mean of the positive control wells and represents the conversion reading for wells without compound inhibition. The dose-response curves were fitted using the log values of the concentrations as the X-axis and the percent inhibition as the Y-axis, using the analysis software GraphPad Prism 5 log (inhibitor) vs. response-Variable slope, to obtain the IC50 values of each compound for enzyme activity. The calculation formula is Y ═ Bottom + (Top-Bottom)/(1+10^ ((LogIC) 50 -X)*Hill Slope))。
As a result: most of the compounds of the examples of the invention have high CDK7 kinase inhibitory activity, and still show high inhibitory activity (the inhibitory rate is more than 60%) at the concentration as low as 100 nM; and most of the compounds of the examples had weak inhibitory activity against CDK9, and still showed a better inhibition at 1000nM concentrationLow inhibitory activity (inhibition less than 50%). Most of the example compounds showed higher CDK7/CDK9 kinase selectivity. (wherein the inhibition rate is ++++ > or more than 80%; 80%<+++≤60%;60%<++≤30%;+<30%;IC 50 Represents A is less than or equal to 20 nM; 20nM<B≤100nM;100nM<C≤1000nM;D>1000nM。)
Figure BDA0003474551550000531
Figure BDA0003474551550000541
Figure BDA0003474551550000551
Figure BDA0003474551550000561
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (10)

1. An amino-substituted aromatic heterocyclic pyrazole compound shown as a general formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof,
Figure FDA0003474551540000011
in the formula:
w is selected from CR w Or N; r w Independently selected from H, halogen, cyano, C1-C6 alkyl or haloalkyl;
R 1 independently selected from C1-C10 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl, C1-C10 alkoxy or haloalkoxy, 3-10 membered cycloalkyl ether or heterocycloalkyl ether, C1-C10 alkyl or haloalkyl substituted amino, 3-10 membered cycloalkyl or heterocycloalkyl substituted amino; and the above alkyl, cycloalkyl, heterocycloalkyl may be substituted with one or several substituents selected from the group consisting of: halogen, deuterium, hydroxyl, substituted or unsubstituted amino, C1-C6 alkyl, hydroxyl-substituted C1-C6 alkyl, amino-substituted C1-C6 alkyl, C1-C6 alkoxy, 3-10 membered cycloalkyl or heterocycloalkyl-substituted C1-C6 alkyl;
R 2 independently selected from C1-C10 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl;
R a selected from hydrogen, deuterium, halogen, C1-C6 alkyl or haloalkyl, 3-to 10-membered cycloalkyl or heterocycloalkyl;
R b 、R c 、R d 、R e each independently selected from hydrogen, deuterium, halogen, C1-C6 alkyl or haloalkyl, 3-to 10-membered cycloalkyl or heterocycloalkyl, or R b And R c 、R d And R e Form a C ═ O bond or a 3-6 membered carbocyclic or heterocyclic ring, respectively;
m and n are respectively and independently selected from integers of 0-3;
cy is selected from 3-10 membered cycloalkyl or heterocycloalkyl, 5-12 membered monocyclic or fused aromatic or heteroaromatic group;
R 3 selected from the group consisting of hydrogen, deuterium, halogen, C1-C10 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl, 5-12 membered aryl or heteroaryl, substituted or unsubstituted amino, hydroxyl, C1-C10 alkoxy or haloalkoxy, cyano, substituted or unsubstituted amido, substituted or unsubstituted sulfonamide, substituted or unsubstituted ureido, substituted or unsubstituted sulfonylureido, substituted or unsubstituted carbamoyl, substituted or unsubstituted phosphoryl, substituted or unsubstituted alkylphosphite, substituted or unsubstitutedSubstituted alkylsilyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted acryloyl, substituted or unsubstituted propioylyl, haloacetyl or haloethylsulfonyl;
y is selected from N or CRy, Ry is independently selected from hydrogen, deuterium, halogen, hydroxy, amino, cyano, C1-C6 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl;
R 4 independently selected from hydrogen, deuterium, halogen, C1-C10 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl, 5-12 membered aryl or heteroaryl, substituted or unsubstituted amino, hydroxyl, C1-C10 alkoxy or haloalkoxy, cyano, substituted or unsubstituted amido, substituted or unsubstituted sulfonamide, substituted or unsubstituted ureido, substituted or unsubstituted sulfonylureido, substituted or unsubstituted carbamoyl, substituted or unsubstituted phosphoryl, substituted or unsubstituted alkylphosphioxy, substituted or unsubstituted alkylsilyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted acryloyl, substituted or unsubstituted propioyl, haloacetyl or haloethylsulfonyl
The above-mentioned substituted or unsubstituted substituents are each independently selected from the group consisting of: including but not limited to deuterium, halogen, hydroxy, monoalkylamino, dialkylamino, C1-C6 alkyl or haloalkyl, 3-10 membered cycloalkyl or heterocycloalkyl, cyano, C1-C6 alkoxy or haloalkoxy; wherein said heteroaryl group contains 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, the heterocycloalkyl group containing 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, said ring system including spiro, bridged, fused, etc. saturated or partially unsaturated ring systems.
2. The compound according to claim 1, which is preferably a compound of formula (II), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph or prodrug thereof:
Figure FDA0003474551540000021
wherein R is 1 、R 2 、R 3 、R 4 、R a 、R b 、R c 、R d 、R e Cy, W, m, n are as defined in claim 1.
3. The compound of claim 1, which is preferably a compound of formula (III), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph or prodrug thereof:
Figure FDA0003474551540000022
wherein R is 1 、R 2 、R 3 、R 4 、R a 、R b 、R c 、R d 、R e Cy, W, m and n are defined as in claim 1.
4. A compound as claimed in claim 1, which is preferably a compound of general formula (IV), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsionade, solvate, polymorph or prodrug thereof:
Figure FDA0003474551540000031
wherein Cy1 is selected from 5-10 membered aryl or heteroaryl, Cy2 is selected from 3-12 membered saturated carbocyclic or heterocyclic groups, and the range of other groups is as defined in claim 1.
5. The compound according to any one of claims 1 to 4, which is preferably a compound of general formula V, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph or prodrug thereof:
Figure FDA0003474551540000032
R 1 preferably C1-C6 alkyl, such as methyl; substituted or unsubstituted 5-6 membered saturated ring such as cyclohexyl, piperidinyl, piperazinyl, morpholinyl, tetrahydropyranyl and the like; substituted or unsubstituted 5-6 membered saturated cyclic group ethers such as cyclohexyl ether, piperidinyl ether, tetrahydropyranyl ether; substituted or unsubstituted 5-6 membered saturated cyclic amines, such as cyclohexylamino, piperidinamino, tetrahydropyrylamino; substituted or unsubstituted 5-6 membered saturated cyclic alkylene ethers such as piperidylmethylene ether; substituted or unsubstituted 5-6 membered saturated cyclylalkyleneamine, such as piperidinylidenemethylamine; the substituent is preferably selected from deuterium, halogen, amino, hydroxyl, dimethylamino, methoxy, hydroxymethyl, aminomethyl and the like; r 3 、R 4 The ranges W, m and n are as defined in any one of claims 1-4.
6. The compound of claim 1, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph, or prodrug thereof, wherein:
R 1 independently selected from C1-C6 alkyl, C1-C6 haloalkyl, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, 5-8 membered heterocycloalkyl-O-, C1-C6 alkyl substituted amino, C1-C6 haloalkyl substituted amino, 3-8 membered cycloalkyl substituted amino, 3-8 membered heterocycloalkyl substituted amino; the R is 1 1 or more hydrogen atoms above may be optionally substituted with: halogen, hydroxyl, amino, cyano, single C1-C3 alkylamino, double C1-C3 alkylamino, C1-C3 alkyl, hydroxyl-substituted C1-C3 alkyl, amino-substituted C1-C3 alkyl, C1-C3 alkoxy, 3-8 membered cycloalkyl C1-C3 alkyl-, 3-8 membered heterocycloalkyl-C1-C3 alkyl-, 3-8 membered cycloalkyl, 3-8 membered heterocychcA cycloalkyl group;
or, R 2 Independently selected from C1-C6 alkyl, C1-C6 haloalkyl, 3-8 membered cycloalkyl or 3-8 membered heterocycloalkyl;
or Cy is selected from 5-8 membered cycloalkyl, 5-8 membered heterocycloalkyl, 6-10 membered monocyclic or fused aromatic group, 5-10 membered monocyclic or fused heteroaromatic group;
or, R 3 Selected from hydrogen, deuterium, halogen, hydroxyl, cyano, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 alkyl, C1-C6 haloalkyl, 3-6 membered cycloalkyl or heterocycloalkyl, 6-10 membered aryl, 5-8 membered heteroaryl, substituted or unsubstituted amino, substituted or unsubstituted amido, substituted or unsubstituted phosphoryl;
or, R 4 Independently selected from hydrogen, deuterium, halogen, hydroxy, cyano, C1-C6 alkyl, C1-C6 haloalkyl, 3-6 membered cycloalkyl, 3-6 membered heterocycloalkyl, 6-10 membered aryl, 5-8 membered heteroaryl, C1-C6 alkoxy, C1-C6 haloalkoxy, 3-6 membered cycloalkyl-C (O) -, 3-6 membered heterocycloalkyl-C (O) -, 3-6 membered cycloalkyl-O-C (O) -, 3-6 membered heterocycloalkyl-O-C (O) -, 6-10 membered aryl-C (O) -, 5-8 membered heteroaryl-C (O) -; said R is 4 Optionally substituted by R 41 Substituted, said R 41 Independently selected from: halogen, hydroxy, cyano, mono-C1-C6 alkylamino, di-C1-C6 alkylamino, C1-C6 alkyl, C1-C6 haloalkyl, 3-6 membered cycloalkyl or heterocycloalkyl, C1-C6 alkoxy or haloalkoxy, C2-C6 alkenyl-C (O) -; the R is 41 May be further substituted with: halogen, hydroxy, cyano, mono-C1-C6 alkylamino, di-C1-C6 alkylamino.
7. The compound of claim 1, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph, or prodrug thereof, wherein:
R 1 is composed of
Figure FDA0003474551540000041
Or, R 2 Is methyl or isopropyl;
or, R 3 Is H, F, Cl, CN, CH 3 、CH 3 O-、CF 3
Figure FDA0003474551540000051
Figure FDA0003474551540000052
Or, R 4 Is H, CH 3 An isopropyl group,
Figure FDA0003474551540000053
Figure FDA0003474551540000054
8. The compound of claim 1, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph, or prodrug thereof, wherein the compound has the structure:
Figure FDA0003474551540000061
Figure FDA0003474551540000071
Figure FDA0003474551540000081
9. use of a compound according to any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsomer, solvate, polymorph or prodrug thereof, for the manufacture of a medicament for the prevention, treatment, or alleviation of a disorder or a disease mediated by abnormal activity of a CDK kinase, particularly CDK7 kinase, including cancer (e.g., leukemia, melanoma, multiple myeloma), benign neoplasms, inflammatory diseases, autoinflammatory diseases, autoimmune diseases, or infectious diseases; the autoimmune disease is independently selected from rheumatoid arthritis, systemic lupus erythematosus, idiopathic thrombocytopenic purpura, hemolytic anemia, or psoriasis; the inflammatory disease is independently selected from osteoarthritis, gouty arthritis, ulcerative colitis, inflammatory bowel disease and/or the like; the infectious disease is independently selected from sepsis, septic shock, endotoxic shock, gram-negative sepsis, and/or toxic shock syndrome.
10. A pharmaceutical composition comprising a compound according to any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph or prodrug thereof, wherein the pharmaceutical composition comprises:
(i) an effective amount of a compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph, or prodrug thereof; and
(ii) a pharmaceutically acceptable carrier.
CN202210051634.4A 2021-01-17 2022-01-17 Amino substituted aromatic heterocyclic pyrazole compound, preparation method and application Active CN114805361B (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN2021100589779 2021-01-17
CN202110058977 2021-01-17
CN202110185488 2021-02-10
CN202110185488X 2021-02-10

Publications (2)

Publication Number Publication Date
CN114805361A true CN114805361A (en) 2022-07-29
CN114805361B CN114805361B (en) 2024-02-20

Family

ID=82527479

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210051634.4A Active CN114805361B (en) 2021-01-17 2022-01-17 Amino substituted aromatic heterocyclic pyrazole compound, preparation method and application

Country Status (1)

Country Link
CN (1) CN114805361B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114213417A (en) * 2021-11-16 2022-03-22 郑州大学 Pyrazolo six-membered nitrogen heterocyclic compound and synthesis method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008130570A1 (en) * 2007-04-20 2008-10-30 Schering Corporation Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
CN110831600A (en) * 2017-04-21 2020-02-21 金恩医疗公司 Indole AHR inhibitors and uses thereof
WO2020081636A1 (en) * 2018-10-16 2020-04-23 Ikena Oncology, Inc. Indole ahr inhibitors and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008130570A1 (en) * 2007-04-20 2008-10-30 Schering Corporation Novel pyrazolopyrimidines as cyclin dependent kinase inhibitors
CN110831600A (en) * 2017-04-21 2020-02-21 金恩医疗公司 Indole AHR inhibitors and uses thereof
WO2020081636A1 (en) * 2018-10-16 2020-04-23 Ikena Oncology, Inc. Indole ahr inhibitors and uses thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114213417A (en) * 2021-11-16 2022-03-22 郑州大学 Pyrazolo six-membered nitrogen heterocyclic compound and synthesis method and application thereof

Also Published As

Publication number Publication date
CN114805361B (en) 2024-02-20

Similar Documents

Publication Publication Date Title
CN110156786B (en) Pyrimido-cyclic compounds, process for their preparation and their use
CN112300194B (en) Condensed ring pyridone compounds, preparation method and application
ES2866152T3 (en) Tyrosine amide derivatives as Rho-kinase inhibitors
CN112142735B (en) Condensed cyanopyridine compound, preparation method and application
CN111153901A (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
CN112135612A (en) Amino acid compounds and methods of use
CN114173803A (en) Dosage forms and regimens for amino acid compounds
WO2020035049A1 (en) Degradation agents for cyclin dependent kinase, preparation method therefor, pharmaceutical composition thereof and use thereof
CN111936144B (en) JAK inhibitors
WO2016192630A1 (en) Compound having kinase inhibiting activity, method of preparing same, and use of same
BR112021011147A2 (en) PYRAZOLYL-AMINO-PYRIMIDINIL DERIVATIVES BENZAMIDES AND COMPOSITIONS AND METHODS THEREOF
CN112457326B (en) Aromatic heterocyclic lactam compound, preparation method and application
CN114249712A (en) Pyrimidinyl derivatives, method for the production thereof and use thereof
CN110655520A (en) Pyrimido-cyclic compounds, process for their preparation and their use
CN113527299A (en) Nitrogen-containing condensed ring compounds, preparation method and application
CN112300196A (en) Piperidine condensed ring compound, preparation method and application
CN112094269B (en) Saturated six-membered ring heterocyclic compound, preparation method and application
CN114524810A (en) Pyrimidine heterocyclic compound, preparation method and application
CN109790160B (en) Pyrido five-membered aromatic ring compound, preparation method and application thereof
CN112778336B (en) Nitrogen-containing condensed ring STING regulator compound, preparation method and application
CN112300173B (en) Nitrogen-containing polycyclic compounds, preparation method and application
CN114805361A (en) Amino-substituted aromatic heterocyclic pyrazole compounds, preparation method and application
CN114728932A (en) Polyarylates as EGFR kinase inhibitors
CN111057048B (en) Aminopyrazine/pyridine compound, preparation method and application
CN114746089A (en) Thiazolecarboxamide compounds and their use for the treatment of mycobacterial infections

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
GR01 Patent grant
GR01 Patent grant