CN109422753B - Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity - Google Patents

Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity Download PDF

Info

Publication number
CN109422753B
CN109422753B CN201710783082.5A CN201710783082A CN109422753B CN 109422753 B CN109422753 B CN 109422753B CN 201710783082 A CN201710783082 A CN 201710783082A CN 109422753 B CN109422753 B CN 109422753B
Authority
CN
China
Prior art keywords
integer
compound
substituents
independently selected
absent
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.)
Active
Application number
CN201710783082.5A
Other languages
Chinese (zh)
Other versions
CN109422753A (en
Inventor
舒永志
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 Meizer Pharmaceuticals Co ltd
Original Assignee
Shanghai Meizer Pharmaceuticals 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 Meizer Pharmaceuticals Co ltd filed Critical Shanghai Meizer Pharmaceuticals Co ltd
Priority to CN201710783082.5A priority Critical patent/CN109422753B/en
Priority to CN201880004074.9A priority patent/CN109937203B/en
Priority to PCT/CN2018/103707 priority patent/WO2019042442A1/en
Publication of CN109422753A publication Critical patent/CN109422753A/en
Application granted granted Critical
Publication of CN109422753B publication Critical patent/CN109422753B/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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Abstract

The invention provides a compound capable of inhibiting and degrading the activity of tyrosine protein kinase JAK1 or JAK2, in particular to a compound shown as the following formula I; wherein, the definition of each group is described in the specification. The compound has good JAK1 or JAK2 inhibition and degradation activities, and can be used for preparing medicines for treating JAK1 or JAK2 related diseases.

Description

Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity
Technical Field
The invention belongs to the field of medicines, and particularly relates to a compound capable of inhibiting and degrading the activity of tyrosine protein kinase JAK1 or JAK2, and preparation and application thereof.
Background
Janus kinases (JAKs) are non-receptor tyrosine protein kinases existing in cells, are very important for multiple functions such as growth, activation and homeostatic regulation of T cells, and play a key role in regulating the cell functions of the lymphohematopoietic system. In mammals, this family contains 4 major family members, namely JAK1, JAK2, JAK3 and Tyk 2. Among them, JAK1, JAK2 and Tyk2 are widely expressed in various histiocytes, and JAK3 is mainly expressed in hematopoietic tissues at high efficiency, such as activated B lymphocytes, T lymphocytes, bone marrow cells and thymocytes.
JAK1 binds to IL-10, IL-19, IL-20, IL-22, IL-26, IL-28, IFN- α, IFN- γ, IL-6 in the gp130 family, and other receptors containing yc. JAK1 gene knockout experiments in mouse models have shown that this enzyme plays a key role in regulating the biological effects of the various cytokine receptors mentioned above. JAK1 has become a novel target in the disease areas of immunity, inflammation and cancer.
JAK2 plays an important role in the regulation of a variety of receptor signals including EPO, GH, PRL, IFN- γ, and IL-3, IL-5, GM-CSF in the β c family. Knockout of JAK2 in mouse models can lead to anemia-induced death of the animal. One base mutation JAK2V617F on JAK2 gene in human body, which is closely related to the occurrence of Polycythemia Vera (PV), Essential Thrombocythemia (ET), Idiopathic Myelofibrosis (IMF), Chronic Myelogenous Leukemia (CML), and the like in myeloproliferative diseases. Therefore, JAK2 has become a definite target for the treatment and prevention of this disease.
Due to the wide regulation of the JAK-STAT signal pathway, organ transplant rejection, xenotransplantation, lupus erythematosus, multiple sclerosis, rheumatoid arthritis, psoriasis (psoriasis), cancer, asthma, atopic dermatitis, type I diabetes and diabetic complications, autoimmune thyroid disorders, ulcerative colitis, Crohn's disease, Alzheimer's disease, leukemia, lymphoma, multiple myeloma, alopecia areata, vitiligo and other diseases also relate to JAK1/2, JAK1/2 plays a key role in the disease occurrence as a signal transmission member, and becomes a drug target for developing and treating the diseases in medicine development.
Accordingly, those skilled in the art are working to develop compounds capable of inhibiting the activity of JAK1 and/or JAK 2.
Disclosure of Invention
The invention aims to provide a compound capable of inhibiting and degrading JAK1 and/or JAK2, and preparation and application thereof.
In a first aspect of the invention, there is provided a compound of formula I, or a pharmaceutically acceptable salt thereof:
Figure BDA0001397384370000021
wherein:
Figure BDA0001397384370000022
represents a single bond;
Figure BDA0001397384370000023
represents a single bond or a double bond;
a is absent or selected from C (═ O), C (═ O) X1, SOX1, SO2X1, C with or without substituents1-8Hydrocarbyl, C with or without substituents1-8Cycloalkyl group, and C with or without substituent1-8A heterocyclic hydrocarbon group; wherein X1 is absent or selected from (CR)12R13)fO、(CR12R13)fS and NR14(ii) a Wherein R is12、R13、R14Each independently is H, C with or without substituents1-8With or without substituents C1-8And C with or without substituents1-8F is an integer between 0 and 3 (e.g., 1, 2, or 3);
w is absent or selected from O, NR17、-X2C(=O)X3、-X2S(=O)gX3; wherein R is17Is H, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8The heterocyclic hydrocarbon group of (a); wherein X2 and X3 are independently deleted or selected from O, S, NR18(ii) a Wherein g is an integer from 0 to 2; wherein R is18Is H, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8The heterocyclic hydrocarbon group of (a);
y is (CR)22R23)h、CHX4(CR22R23)h、CX4=CH(CR22R23)hOr (CR)22R23)h(ii) a Wherein h is an integer between 0 and 30; wherein R is22、R23Each independently selected from H, cyano, hydroxy, amino, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8A heterocyclic hydrocarbon group of (A), C with or without substituents1-8Hydrocarbyloxy groups of (a); wherein X4 is selected from H, halogen, cyano, nitro, hydroxy, with or without substituent C1-8Hydrocarbyloxy, with or without substituents C1-8Alkoxycarbonyl, with or without substituents C1-8Amino, with or without substituents C1-8Ester group, with or without substituents C1-8Aminocarbonyl, with or without substituents C1-8Hydrocarbyl, with or without substituents C1-8Cycloalkyl, with or without substituents C1-8A heterocyclic hydrocarbon group;
z is (CR)24R25)i、CHX5(CR24R25)i、CX5=CH(CR24R25)iOr C ≡ C (CR)24R25)i(ii) a Wherein i is an integer between 0 and 30; wherein R is24、R25Each independently selected from H, cyano, hydroxy, amino, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8Heterocycloalkyl radicals, with or without substituents C1-8A hydrocarbyloxy group; wherein X5 is selected from H, halogen, cyano, nitro, hydroxy, with or without substituent C1-8With or without substituents C1-8Alkoxycarbonyl, with or without substituents C1-8Amino, with or without substituents C1-8Ester group, with or without substituents C1-8Aminocarbonyl, with or without substituents C1-8Hydrocarbyl, with or without substituents C1-8Cycloalkyl, with or without substituents C1-8Heterocyclic ringsA hydrocarbyl group;
b is absent or selected from O, C ═ O, S, NR15、-NR15C(=O)-、-C(=O)NR15-、-C(=O)O-、OC(=O)O-、-NR15C(=O)O-、-OC(=O)NR15-、-NR15C(=O)NR16-, C with or without substituents1-12With or without substituents C1-12And C with or without substituents1-12The heterocyclic hydrocarbon group of (a); wherein R is15、R16Each independently selected from H, C with or without substituents1-8With or without substituents C1-8And C with or without substituents1-8The heterocyclic hydrocarbon group of (a);
x is selected from CR19R20、C(=O)、S(=O)、SO2、NR21(ii) a Wherein R is19、R20Each independently selected from H, cyano, hydroxy, amino, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8A heterocyclic hydrocarbon group of (A), C with or without substituents1-8Hydrocarbyloxy groups of (a); wherein R is21Selected from H, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8The heterocyclic hydrocarbon group of (a);
R1、R8each independently selected from H, C with or without substituents1-8Hydrocarbon group, cycloalkyl group with or without substituent, heterocycloalkyl group with or without substituent, C with or without substituent1-6An acyl group;
R2、R5each independently selected from: hydrogen, OR33、NR34R35Cyano, halogen, C with or without substituents1~8Hydrocarbon group, cycloalkyl group with or without substituent, heterocycloalkyl group with or without substituent, C with or without substituent1-6Acyl, with or without substituentsAn amide group; wherein R is33、R34、R35Each independently selected from H, C with or without substituents1~8A cycloalkyl group with or without substituents, a heterocycloalkyl group with or without substituents;
R3、R6、R7、R9、R10each independently selected from: H. OR (OR)27、NR28R29Cyano, halogen, nitro, C with or without substituents1-8A hydrocarbon group, a cycloalkyl group with or without a substituent, a heterocycloalkyl group with or without a substituent, X6S (═ O)kR30、X6C(=O)R31(ii) a Wherein k is an integer between 0 and 2; wherein R is27,R28,R29,R30,R31Each independently selected from H, C with or without substituents1~8Hydrocarbyl, cycloalkyl, heterocycloalkyi; wherein X6 is absent or selected from O, S, NR32(ii) a Wherein R is32Is H, C with or without substituents1-8A cycloalkyl group with or without substituents, and a heterocycloalkyl group with or without substituents;
R4selected from H, cyano, carboxyl, C with or without substituents1-8A hydrocarbon group of (a), a hydrocarbyloxycarbonyl group with or without a substituent group;
a is an integer between 0 and 5 (e.g., 1, 2, 3, 4, 5);
b is an integer between 0 and 3 (e.g., 1, 2, 3);
c is an integer between 0 and 30 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9);
d is an integer between 0 and 5 (e.g., 1, 2, 3, 4, 5);
e is an integer between 0 and 2 (e.g., 0, 1, 2).
In another preferred embodiment, a is absent; w is X2C (═ O) X3 where X2 is NR18And X3 is absent, or X3 is NR18And X2 is absent; y is (CR)22R23)hWherein R is22、R23Each independently selected from H, hydroxy, with or without substituentsC of (A)1-4H is an integer between 1 and 6; z is (CR)24R25)iWherein R is24、R25Each independently selected from H, hydroxy, C with or without substituents1-4I is an integer between 1 and 6; c is 0.
In another preferred embodiment, a is absent; w is absent or is O; y is (CR)22R23)hWherein R is22、R23Each independently selected from H, hydroxy, C with or without substituents1-4H is an integer between 0 and 3; b is O; z is (CR)24R25)iWherein R is24、R25Each independently selected from H, hydroxy, C with or without substituents1-4I is an integer between 0 and 3; c is an integer between 1 and 6.
In another preferred embodiment, a is C (═ O) X1, where X1 is absent or selected from (CR)12R13)fO, and (CR)12R13)fS, wherein f is an integer between 0 and 2, R12、R13、R14Each independently is hydrogen or C1-4Alkyl groups of (a); w is NR17Wherein R is17Is H, or C with or without substituents1-4A hydrocarbon group of (a); y is (CR)22R23)hWherein R is22、R23Each independently selected from H, hydroxy, C with or without substituents1-4H is an integer between 0 and 3; z is (CR)24R25)iWherein R is24、R25Each independently selected from H, hydroxy, C with or without substituents1-4I is an integer between 0 and 3; b is O; c is an integer between 1 and 4.
In another preferred embodiment, A is SO2X1, wherein X1 is absent or selected from O, and S; w is O; y is (CR)22R23)hWherein R is22、R23Each independently selected from H, hydroxy, C with or without substituents1-4H is an integer between 1 and 6; z is (CR)24R25)iWherein R is24、R25Each independently selected from H, hydroxy, C with or without substituents1-4I is an integer between 0 and 3; c is 0.
In another preferred embodiment, a is absent; w is NR17Wherein R is17Is H, or C with or without substituents1-4A hydrocarbon group of (a); y is (CR)22R23)hWherein R is22、R23Each independently selected from H, hydroxy, C with or without substituents1-4H is an integer between 0 and 3; z is (CR)24R25)iWherein R is24、R25Each independently selected from H, hydroxy, C with or without substituents1-4I is an integer between 0 and 4; b is O; c is an integer between 1 and 6.
In another preferred embodiment, a is absent; deletion of W; y is (CR)22R23)hWherein R is22、R23Each independently selected from H, hydroxy, C with or without substituents1-4H is an integer between 0 and 3; z is (CR)24R25)iWherein R is24、R25Each independently selected from H, hydroxy, C with or without substituents1-4I is an integer between 0 and 3; b is O; c is an integer between 1 and 10 (preferably between 1 and 6).
In another preferred embodiment, any of the substituents is selected from the group consisting of: halogen, unsubstituted or halogenated C1-C6 alkyl, unsubstituted or halogenated C1-C6 alkoxy, unsubstituted or halogenated C2-C6 alkoxyalkyl, unsubstituted or halogenated C3-C8 cycloalkyl, unsubstituted or halogenated C2-C6 alkylcarbonyl, unsubstituted or halogenated C1-C6 alkylene-hydroxy, unsubstituted or C1-C6 alkyl-substituted amine.
In another preferred embodiment, in formula I,
Figure BDA0001397384370000051
represents a single bond.
In another preferred example, X is C (═ O).
In another preferred embodiment, R1Selected from: H. and C with or without substituents1-4An alkyl group.
In another preferred embodiment, R2、R5Each independently selected from: hydrogen, and C with or without substituents1-4An alkyl group.
In another preferred embodiment, R3Selected from: hydrogen, and C with or without substituents1-4An alkyl group.
In another preferred embodiment, R6Selected from: hydrogen, C with or without substituents1-4Alkyl, OR29Wherein R is29Selected from H, C with or without substituents1-6An alkyl group.
In another preferred embodiment, R7Selected from: hydrogen, C with or without substituents1-4Alkyl, and NR28R29Wherein R is28,R29Each independently selected from: H. or C with or without substituents1-4A hydrocarbon group of (1).
In another preferred embodiment, R8Selected from: hydrogen, halogen, and C with or without substituents1-4An alkyl group.
In another preferred embodiment, R9Selected from: hydrogen, halogen, and C with or without substituents1-4An alkyl group.
In another preferred embodiment, R10Selected from: hydrogen, halogen, cyano, nitro, and C with or without substituents1-4An alkyl group.
In another preferred embodiment, R4Selected from H, cyano, C with or without substituents1-6An alkyl group.
In a second aspect of the present invention, there is provided a pharmaceutical composition comprising a compound of the first aspect, or a pharmaceutically acceptable salt, prodrug thereof, and a pharmaceutically acceptable carrier.
In another preferred embodiment, the effective amount refers to a therapeutically effective amount or an inhibitory effective amount, preferably 0.01 to 99.99%.
In another preferred embodiment, the pharmaceutical composition further comprises one or more additional antineoplastic agents.
In another preferred embodiment, the pharmaceutical composition is for inhibiting or degrading JAK1 and/or JAK 2.
In another preferred embodiment, the pharmaceutical composition is used for treating a disease associated with JAK1 and/or JAK2 activity or expression.
In a third aspect of the invention, there is provided the use of a compound according to the first aspect of the invention for:
(a) preparing a medicament for treating diseases related to the activity or expression amount of JAK1 and/or JAK 2;
(b) preparing a JAK1 and/or JAK2 targeted inhibitor or degrader;
(c) non-therapeutically inhibiting or degrading JAK1 and/or JAK2 in vitro;
(d) non-therapeutically inhibiting tumor cell proliferation in vitro; and/or
(e) Treating diseases associated with JAK1 and/or JAK2 activity or expression levels.
In another preferred embodiment, the disease associated with JAK1 and/or JAK2 activity or expression is a neoplastic or autoimmune disease.
In another preferred embodiment, the disease associated with JAK1 and/or JAK2 activity or expression is selected from the group consisting of: organ transplant rejection, xenograft rejection, lupus erythematosus, multiple sclerosis, rheumatoid arthritis, psoriasis, cancer, asthma, atopic dermatitis, type I diabetes and diabetic complications, autoimmune thyroid disorders, ulcerative colitis, crohn's disease, alzheimer's disease, leukemia, lymphoma, multiple myeloma, alopecia areata, vitiligo, and the like.
In a fourth aspect of the invention, there is provided a process for the preparation of a compound of formula I as described in the first aspect of the invention, comprising the steps of:
Figure BDA0001397384370000061
(a) reacting a compound shown in a formula IV with a compound shown in a formula II in an inert solvent to obtain a compound shown in a formula I;
in the above formulae, each group is as defined above, M1Is a leaving group.
In another preferred example, the method further comprises the steps of:
Figure BDA0001397384370000071
(b) reacting a compound of formula III with a compound of formula V in an inert solvent to give a compound of formula II, M1、M2Is a leaving group.
In a fifth aspect of the invention there is provided a method of inhibiting or degrading JAK1 and/or JAK2 comprising the steps of: administering to a subject an effective amount of a compound of formula I as described in the first aspect of the invention or a pharmaceutically acceptable salt thereof, or administering to a subject an inhibitory effective amount of a pharmaceutical composition as described in the fourth aspect of the invention.
In another preferred embodiment, said inhibition or degradation is non-therapeutic in vitro.
In another preferred embodiment, when an effective amount of a compound of formula I according to the first aspect of the invention or a pharmaceutically acceptable salt thereof is administered to a subject, the inhibitory effective amount is 0.001-500nmol/L, preferably 0.01-200 nmol/L.
In a sixth aspect of the present invention, there is provided a method of treating a disease associated with JAK1 and/or JAK2 activity or expression comprising: administering to a subject a therapeutically effective amount of a compound of formula I according to the first aspect of the invention, or a pharmaceutical composition according to the fourth aspect of the invention.
In another preferred embodiment, the subject is a mammal; preferably, the mammal is a human.
In another preferred embodiment, the disease associated with JAK1 and/or JAK2 activity or expression is a neoplastic or autoimmune disease.
In another preferred embodiment, the disease associated with JAK1 and/or JAK2 activity or expression is selected from the group consisting of: organ transplant rejection, xenograft rejection, lupus erythematosus, multiple sclerosis, rheumatoid arthritis, psoriasis, cancer, asthma, atopic dermatitis, type I diabetes and diabetic complications, autoimmune thyroid disorders, ulcerative colitis, crohn's disease, alzheimer's disease, leukemia, lymphoma, multiple myeloma, alopecia areata, vitiligo, and the like.
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. Not to be reiterated herein, but to the extent of space.
Detailed Description
The inventor of the invention has conducted extensive and intensive research to prepare a compound having a structure shown in formula I, and finds that the compound has JAK1 and/or JAK2 inhibition and degradation activities. And the compound can inhibit and degrade JAK1 and/or JAK2 at an extremely low concentration, and has extremely excellent activity, so that the compound can be used for treating diseases related to the activity or expression amount of JAK1 and/or JAK2, such as tumors. The present invention has been completed based on this finding.
The invention provides a novel compound and application thereof in degrading tyrosine protein kinase JAK1 and/or JAK 2. The compounds of the present invention can inhibit or degrade JAK1 and/or JAK2, and are useful as JAK1 and/or JAK2 inhibitors or degradants to exert immunomodulatory effects, and for the treatment of diseases in mammals such as rheumatoid arthritis, psoriasis, psoriatic arthritis, ankylosing spondylitis, atopic dermatitis, keratoconjunctivitis sicca, and crohn's disease.
Term(s) for
In the present invention, the term "C1-8The hydrocarbyl group means a functional group containing only carbon and hydrogen atoms, wherein the number of carbon atoms is 1 to 8. The hydrocarbyl group can be regarded as a radical remaining after the corresponding hydrocarbon loses one hydrogen atom, and can be an alkyl group, a cycloalkyl group, an alkenyl group, an alkynyl group or the like; the structure can be straight chain, branched chain or cyclic; may be aliphatic or aromatic.
The term "C1-6The "alkyl group" means a straight or branched alkyl group having 1 to 6 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, or the like.
The term "alkoxy" as used herein includes O-alkyl, wherein "alkyl" is as defined above.
The term "halo" as used herein, unless otherwise indicated, includes fluoro, chloro, bromo or iodo.
The compounds of the present invention may contain double bonds. When containing such double bonds, the compounds of the present invention exist in cis, trans, or mixtures thereof.
Halogen as used herein includes fluorine, chlorine, bromine and iodine.
Unless otherwise indicated, alkyl groups and the alkyl portion of alkoxy groups referred to herein may be straight chain, branched chain or cyclic.
In the present invention, the term "cycloalkyl group" refers to a functional group containing both carbon and hydrogen atoms. Including cycloalkyl, cycloalkenyl (containing at least one carbon-carbon double bond), and aryl. They may be monocyclic, bicyclic and polycyclic. They may be spiro or fused rings.
In the present invention, the term "heterocyclic hydrocarbon group" means a functional group containing carbon, hydrogen and at least one heteroatom other than carbon and hydrogen. Including heterocycloalkyl, heterocycloalkenyl (containing at least one carbon-carbon double bond), and heteroaryl. One or more of the ring-forming atoms in the ring is a heteroatom. The heteroatoms may be O, N and S atoms, and various combinations thereof. They may be monocyclic, bicyclic and polycyclic. They may be spiro or fused rings.
In the present invention, the term "substituent" includes, but is not limited to, fluorine, chlorine, bromine, cyano, hydroxyl, amino, C1-6Hydrocarbyloxy, C1-6Halogenated hydrocarbon group, C1-6Acyl radical, C1-6A sulfonyl group.
The term "hydrocarbyloxy" as used herein refers to an O-hydrocarbyl group, wherein "hydrocarbyl" is as defined above.
The term "alkoxycarbonyl" as used herein refers to a C (═ O) O-hydrocarbyl group, where "hydrocarbyl" is as defined above.
The term "amine" as used herein refers to N (H or hydrocarbyl 1) (H or hydrocarbyl 2), wherein "hydrocarbyl" is as defined above.
The term "aminocarbonyl" as used herein refers to C (═ O) -amine groups, where "amine groups" are as defined above.
The term "amido", as used herein, refers to an N (H or hydrocarbyl) -C (═ O) -hydrocarbyl group, where the "hydrocarbyl group" is as defined above.
As used herein, the terms "comprising," "including," or "including" mean that the various ingredients may be used together in a mixture or composition of the invention. Thus, the terms "consisting essentially of and" consisting of are encompassed by the term "comprising.
In the present invention, the term "pharmaceutically acceptable" ingredient refers to a substance that is suitable for use in humans and/or animals without undue adverse side effects (such as toxicity, irritation, and allergic response), i.e., at a reasonable benefit/risk ratio.
In the present invention, the term "effective amount" refers to an amount of a therapeutic agent that treats, alleviates, or prevents a target disease or condition, or an amount that exhibits a detectable therapeutic or prophylactic effect. The precise effective amount for a subject will depend upon the size and health of the subject, the nature and extent of the disorder, and the therapeutic agent and/or combination of therapeutic agents selected for administration. Therefore, it is not useful to specify an exact effective amount in advance. However, for a given condition, the effective amount can be determined by routine experimentation and can be determined by a clinician.
Herein, unless otherwise specified, the term "substituted" means that one or more hydrogen atoms on a group are replaced with a substituent selected from the group consisting of: halogen, unsubstituted or halogenated C1-6Alkyl, unsubstituted or halogenated C2-6Acyl, unsubstituted or halogenated C1-6Alkyl-hydroxy.
Unless otherwise specified, all occurrences of a compound in the present invention are intended to include all possible optical isomers, such as a single chiral compound, or a mixture of various chiral compounds (i.e., a racemate). In all compounds of the present invention, each chiral carbon atom may optionally be in the R configuration or the S configuration, or a mixture of the R configuration and the S configuration.
As used herein, the term "compounds of the invention" refers to compounds of formula I. The term also includes various crystalline forms, pharmaceutically acceptable salts, hydrates or solvates of the compounds of formula I.
As used herein, the term "pharmaceutically acceptable salt" refers to a salt of a compound of the present invention with an acid or base that is suitable for use as a pharmaceutical. Pharmaceutically acceptable salts include inorganic and organic salts. One preferred class of salts is that formed by reacting a compound of the present invention with an acid. Suitable acids for forming the salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, etc., organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, methanesulfonic acid, phenylmethanesulfonic acid, benzenesulfonic acid, etc.; and acidic amino acids such as aspartic acid and glutamic acid.
Compounds and pharmaceutically acceptable salts thereof
The present invention relates to compounds of formula I or a pharmaceutically acceptable salt thereof;
Figure BDA0001397384370000103
wherein:
Figure BDA0001397384370000101
represents a single bond;
Figure BDA0001397384370000102
represents a single bond or a double bond;
a is absent or selected from C (═ O), C (═ O) X1, SOX1, SO2X1, C with or without substituents1-8Hydrocarbyl, C with or without substituents1-8Cycloalkyl radicals, and with or without substitutionC of radical1-8A heterocyclic hydrocarbon group; wherein X1 is absent or selected from (CR)12R13)fO、(CR12R13)fS and NR14(ii) a Wherein R is12、R13、R14Each independently is H, C with or without substituents1-8With or without substituents C1-8And C with or without substituents1-8F is an integer between 0 and 3 (e.g., 1, 2, or 3);
w is absent or selected from O, NR17, -X2C (═ O) X3, -X2S (═ O)gX3; wherein R17 is H, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8The heterocyclic hydrocarbon group of (a); wherein X2 and X3 are independently deleted or selected from O, S, NR18(ii) a Wherein g is an integer from 0 to 2; wherein R is18Is H, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8The heterocyclic hydrocarbon group of (a);
y is (CR)22R23)h、CHX4(CR22R23)h、CX4=CH(CR22R23)hOr (CR)22R23)h(ii) a Wherein h is an integer between 0 and 30; wherein R is22、R23Each independently selected from H, cyano, hydroxy, amino, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8A heterocyclic hydrocarbon group of (A), C with or without substituents1-8Hydrocarbyloxy groups of (a); wherein X4 is selected from H, halogen, cyano, nitro, hydroxy, with or without substituent C1-8Hydrocarbyloxy, with or without substituents C1-8Alkoxycarbonyl, with or without substituents C1-8Amino, with or without substituents C1-8Ester group, with or without substituents C1-8Aminocarbonyl, with or without substituents C1-8Hydrocarbyl radicalWith or without substituents C1-8Cycloalkyl, with or without substituents C1-8A heterocyclic hydrocarbon group;
z is (CR)24R25)i、CHX5(CR24R25)i、CX5=CH(CR24R25)iOr C ≡ C (CR)24R25)i(ii) a Wherein i is an integer between 0 and 30; wherein R is24、R25Each independently selected from H, cyano, hydroxy, amino, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8Heterocycloalkyl radicals, with or without substituents C1-8A hydrocarbyloxy group; wherein X5 is selected from H, halogen, cyano, nitro, hydroxy, with or without substituent C1-8With or without substituents C1-8Alkoxycarbonyl, with or without substituents C1-8Amino, with or without substituents C1-8Ester group, with or without substituents C1-8Aminocarbonyl, with or without substituents C1-8Hydrocarbyl, with or without substituents C1-8Cycloalkyl, with or without substituents C1-8A heterocyclic hydrocarbon group;
b is absent or selected from O, C ═ O, S, NR15、-NR15C(=O)-、-C(=O)NR15-、-C(=O)O-、OC(=O)O-、-NR15C(=O)O-、-OC(=O)NR15-、-NR15C(=O)NR16-, C with or without substituents1-12With or without substituents C1-12And C with or without substituents1-12The heterocyclic hydrocarbon group of (a); wherein R is15、R16Each independently selected from H, C with or without substituents1-8With or without substituents C1-8And C with or without substituents1-8The heterocyclic hydrocarbon group of (a);
x is selected from CR19R20、C(=O)、S(=O)、SO2、NR21(ii) a Wherein R is19、R20Each independently selected from H, cyano, hydroxy, amino, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8A heterocyclic hydrocarbon group of (A), C with or without substituents1-8Hydrocarbyloxy groups of (a); wherein R is21Selected from H, C with or without substituents1-8With or without substituents C1-8Cycloalkyl, with or without substituents C1-8The heterocyclic hydrocarbon group of (a);
R1、R8each independently selected from H, C with or without substituents1-8Hydrocarbon group, cycloalkyl group with or without substituent, heterocycloalkyl group with or without substituent, C with or without substituent1-6An acyl group;
R2、R5each independently selected from: hydrogen, OR33、NR34R35Cyano, halogen, C with or without substituents1~8Hydrocarbon group, cycloalkyl group with or without substituent, heterocycloalkyl group with or without substituent, C with or without substituent1-6Acyl, amido with or without substituents; wherein R is33、R34、R35Each independently selected from H, C with or without substituents1~8A cycloalkyl group with or without substituents, a heterocycloalkyl group with or without substituents;
R3、R6、R7、R9、R10each independently selected from: H. OR (OR)27、NR28R29Cyano, halogen, nitro, C with or without substituents1-8A hydrocarbon group, a cycloalkyl group with or without a substituent, a heterocycloalkyl group with or without a substituent, X6S (═ O)kR30、X6C(=O)R31(ii) a Wherein k is an integer between 0 and 2; wherein R is27,R28,R29,R30,R31Each independently selected from H, C with or without substituents1~8Hydrocarbyl, cyclic hydrocarbonA phenyl, a heterocyclic hydrocarbon; wherein X6 is absent or selected from O, S, NR32(ii) a Wherein R is32Is H, C with or without substituents1-8A cycloalkyl group with or without substituents, and a heterocycloalkyl group with or without substituents;
R4selected from H, cyano, carboxyl, C with or without substituents1-8A hydrocarbon group of (a), a hydrocarbyloxycarbonyl group with or without a substituent group;
a is an integer between 0 and 5 (e.g., 1, 2, 3, 4, 5);
b is an integer between 0 and 3 (e.g., 1, 2, 3);
c is an integer between 0 and 30 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9);
d is an integer of 0 to 5 (e.g. 1, 2, 3, 4, 5)
e is an integer between 0 and 2 (e.g., 0, 1, 2).
The integer herein is 0, 1, 2, 3, 4, 5, 6, 7, 8, or 9, unless otherwise specified.
In a preferred embodiment of the invention, the compound is selected from the group consisting of:
Figure BDA0001397384370000121
the compound package of the present invention may form pharmaceutically acceptable salts with inorganic acids, organic acids or bases. The inorganic acid includes but is not limited to hydrochloric acid, hydrobromic acid, nitric acid, perchloric acid, sulfuric acid or phosphoric acid, etc.; such organic acids include, but are not limited to, methanesulfonic acid, trifluoromethanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, fumaric acid, oxalic acid, acetic acid, maleic acid, ascorbic acid, lactic acid, tartaric acid, malonic acid, glycolic acid, succinic acid, propionic acid, and the like; the base includes, but is not limited to, inorganic salts and amines.
The term pharmaceutically acceptable salts refers to those salts which are, according to medical judgment, suitable for use in contact with the tissues of humans and mammals without excessive toxicity, irritation, allergic response, and the like. Pharmaceutically acceptable salts are well known in the art.
The invention also encompasses pharmaceutical compositions containing prodrugs of compounds of formula I. Prodrugs include compounds wherein the precursor molecule is covalently bonded to a free carboxyl, hydroxyl, amino or amine group of the compound of formula I via a carbonate, carbamate, amide, alkyl ester, phosphate, phosphoramidate linkage.
Preparation of the Compounds
Preparation method
The process for the preparation of the compounds of formula I according to the invention is described in more detail below, but these particular processes do not limit the invention in any way. The compounds of the present invention may also be conveniently prepared by optionally combining various synthetic methods described in the present specification or known in the art, and such combinations may be readily carried out by those skilled in the art to which the present invention pertains.
The following reaction scheme illustrates the preparation of the compounds of the present invention. Unless otherwise indicated, A, B, W, Y, Z, X, a, B, c, d, e, R in the reaction schemes and discussion that follows1、R2、R3、R4、R5、R6、R7、R8、R9、R10As defined above.
In general, compounds of formula I can be obtained from formula III as described in the following scheme:
Figure BDA0001397384370000131
in compound II, when W is an ether, it can be prepared by direct nucleophilic substitution of formula III (W1 ═ OH) with an intermediate containing a leaving group under the action of a base, or by reaction with an alcohol using a mitsunobu reaction; when W is ester, carbamate (NHCO)2) When the compound is prepared, the compound can be prepared by reacting formula III (W1 ═ OH) with acyl chloride, activated ester (amide), carboxylic acid and isocyanate under the action of alkali; when W is an amine, formula III (W1 ═ NH) can be used2) Direct nucleophilic substitution with an intermediate containing a leaving group under the action of a base can also be prepared by using a compound of formula III (W1 ═ NH)2) Reductive amination reaction with aldehyde/ketonePreparing; when W is an amide, alkoxycarbonylamine (OCONH), or urea, formula III (W1 ═ NH) can be used2) Reacting with corresponding acyl chloride, activated ester (amide), carboxylic acid and isocyanate under the action of alkali.
In compound I, when A is linked to the nitrogen atom by a C-N bond, it can be prepared by direct substitution or reductive amination; when A is attached to the nitrogen atom by means of an amide, urea, carbamate, sulfonamide, thioamide, it can be prepared from the corresponding acid chlorides, activated esters (amides), carboxylic acids, isocyanates, sulfonyl chlorides, sulfuryl chlorides.
In general, depending on the nature of the linkages A and W, compounds I may also be obtained by linking formula IV to an intermediate chain and then reacting with formula III. The chemical synthesis method is the same as that described above.
The compounds of formula III, IV can be obtained by known synthetic methods or readily obtained by commercial purchase.
Use of compounds of formula I
The compounds of formula I may be used for one or more of the following uses:
(a) preparing a medicament for treating diseases related to the activity or expression amount of JAK1 and/or JAK 2;
(b) preparing a JAK1 and/or JAK2 targeted inhibitor or degrader;
(c) non-therapeutically inhibiting or degrading JAK1 and/or JAK2 in vitro;
(d) non-therapeutically inhibiting tumor cell proliferation in vitro; and/or
(e) Treating diseases associated with the activity or expression of JAK1 and/or JAK2 (e.g., autoimmune diseases).
In another preferred example, the disease associated with JAK1 and/or JAK2 activity or expression is organ transplant rejection, xenotransplantation, lupus erythematosus, multiple sclerosis, rheumatoid arthritis, psoriasis (psoriasis), cancer, asthma, atopic dermatitis, type I diabetes and diabetic complications, autoimmune thyroid disorders, ulcerative colitis, crohn's disease, alzheimer's disease, leukemia, lymphoma, multiple myeloma, alopecia areata, vitiligo, and the like.
The compounds of formula I of the present invention are useful for preparing a pharmaceutical composition comprising: (i) an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof; and (ii) a pharmaceutically acceptable carrier.
In another preferred embodiment, the effective amount refers to a therapeutically effective amount or an inhibitory effective amount.
The compounds of formula I of the present invention may also be used in methods of inhibiting or degrading bujak 1 and/or JAK2, either in vitro non-therapeutic or therapeutic inhibition.
In another preferred embodiment, when an inhibitory effective amount of a compound of formula I of the present invention or a pharmaceutically acceptable salt thereof is administered to a subject, said inhibitory effective amount is 0.001-500nmol/L, preferably 0.01-200 nmol/L.
In particular, the present invention also provides a method of treating a disease associated with JAK1 and/or JAK2 activity or expression comprising: administering to the subject a therapeutically effective amount of a compound of formula I, or said pharmaceutical composition comprising a compound of formula I as an active ingredient.
Pharmaceutical compositions and methods of administration
The compound can obviously degrade JAK1 and/or JAK2 so as to play an inhibitory activity on JAK1 and/or JAK2, so that the compound and various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates thereof, and a pharmaceutical composition containing the compound as a main active ingredient can be used for treating, preventing and relieving diseases related to the activity or expression amount of JAK1 and/or JAK 2. According to the prior art, the compounds of the present invention are useful in the treatment of diseases including tumors and the like.
The pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof in a safe and effective amount range and a pharmacologically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical composition contains 1-2000mg of a compound of the invention per dose, more preferably, 5-200mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.
"pharmaceutically acceptable carrier" refers to: one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. By "compatible" is meant herein that the components of the composition are capable of intermixing with and with the compounds of the present invention without significantly diminishing the efficacy of the compounds. Examples of pharmaceutically acceptable carrier moieties are cellulose and its derivatives (e.g. sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (e.g. stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g. soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (e.g. propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers
Figure BDA0001397384370000151
Wetting agents (such as sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of such materials and the like.
In addition to these inert diluents, the compositions may also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
Dosage forms for topical administration of the compounds of the present invention include ointments, powders, patches, sprays, and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
The compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
When the pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is suitable for mammals (such as human beings) to be treated, wherein the administration dose is a pharmaceutically-considered effective administration dose, and for a human body with a weight of 60kg, the daily administration dose is usually 1 to 2000mg, preferably 5 to 500 mg. Of course, the particular dosage will depend upon such factors as the route of administration, the health of the patient, and the like, and is within the skill of the skilled practitioner.
The main advantages of the invention include:
1. provides a compound shown as a formula I.
2. The JAK1 and/or JAK2 inhibitor with a novel structure and a degrading agent, and preparation and application thereof are provided, and the inhibitor can inhibit and degrade JAK1 and/or JAK2 at an extremely low concentration.
3. Pharmaceutical compositions for the treatment of diseases associated with JAK1 and/or JAK2 activity are provided.
The invention will be further illustrated with reference to the following 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. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or conditions recommended by the manufacturers. Unless otherwise indicated, percentages and parts are percentages and parts by weight.
EXAMPLE 1 preparation of Compound 3
Figure BDA0001397384370000171
The first step is as follows:
2.14g of 6-bromohexanoyl chloride and 1.37g of pomalidomide were dissolved in waterIn 50ml THF (tetrahydrofuran), stirring and refluxing were carried out for 8 hours, and after cooling to room temperature and concentration, vacuum drying was carried out at 40 ℃ to obtain 2.19g of compound (1). MS (ESI) 450[ M + H]+
The second step is that:
dissolving 1g of compound (1), 1.28g of compound (2) and 2.6g of DIPEA (N, N-diisopropylethylamine) in 50ml of DMF (N, N-dimethylformamide), stirring at 80 ℃ for 6h, cooling to room temperature, concentrating, and performing column chromatography to obtain 810mg of compound (3), yield: 65.6 percent. MS (ESI) 649[ M + H [ ]]+1H NMR(400MHz,CDCl3)δ10.62(br,1H),10.15(br,1H),9.02(s,1H),8.46(s,1H),8.24(s,1H),7.45(t,J=7.6Hz,1H),7.35(dd,J=3.2,2.0Hz,1H),7.11(d,J=7.6Hz,1H),6.88(d,J=7.6Hz,1H),6.70(dd,J=3.6,1.6Hz,1H),6.45(t,J=4.0Hz,1H),4.94(dd,J=12.0,5.6Hz,1H),3.65-3.85(m,4H),3.40(s,2H),2.71-2.88(m,4H),1.83-2.52(m,6H),1.60(m,2H),1.41(m,2H)。
EXAMPLE 2 preparation of Compound 7
Figure BDA0001397384370000181
The first step is as follows:
100mg of 4-hydroxythalidomide, 96mg of triethylene glycol monobenzyl ether and 100mg of triphenylphosphine were dissolved in 10ml of anhydrous THF, and 95mg of DIAD (diisopropyl azodicarboxylate) was added dropwise thereto and reacted at room temperature for 2 hours. THF was removed under reduced pressure, and column chromatography was performed to purify the residue to give 110mg of the compound (4). MS (ESI) 497[ M + H]+
The second step is that:
100mg of compound (4) and 100mg of 10% Pd-C were added to 10ml of methanol, and the mixture was hydrogenated at room temperature overnight. Filtration, concentration of the filtrate and purification of the residue by column chromatography gave 40mg of compound (5). MS (ESI) 407[ M + H]+
The third step:
30mg of the compound (5) was dissolved in 5ml of methylene chloride, and 47mg of Dess-martin oxidant was added thereto to conduct a reaction at room temperature for 3 hours. Adding NaHCO3Saturated aqueous solution and Na2S2O3Saturated aqueous solution, stirringStirring for 5 min. Separating the organic layer, passing through anhydrous Na2S2O3Drying and concentrating to dryness to obtain the compound (6) which is directly used for the next reaction.
After dissolving Compound (6) in 7ml of methylene chloride, 45mg of the starting Material (2) and NaBH (OAc) were added323mg, room temperature overnight. Distilling off dichloromethane under reduced pressure, and purifying by column chromatography to obtain compound (7), 25mg. MS (ESI):668[ M + H)]+1H NMR(400MHz,CDCl3)δ10.69(br,1H),10.26(br,1H),8.78(s,1H),8.44(s,1H),8.28(s,1H),7.52(t,J=8.0Hz,1H),7.30(m,2H),7.10(d,J=8.4Hz,1H),6.71(d,J=3.2Hz,1H),5.06(m,1H),4.31(m,2H),3.88-4.04(m,2H),3.73-3.84(m,6H),3.67(br,2H),3.58(t,J=4.8Hz,2H),3.41(s,2H),2.75-2.89(m,4H),2.06-2.35(m,2H)。
EXAMPLE 3 preparation of Compound 10
Figure BDA0001397384370000191
The first step is as follows:
benzyl 2- (2- (2-bromoethoxy) ethoxy) acetate 174mg, potassium carbonate 100mg, potassium iodide 20mg and pomalidomide 100mg were added to 20ml of DMF (N, N-dimethylformamide) and reacted at 80 ℃ overnight. The reaction solution was purified by column chromatography to give 113mg of the compound (8). MS (ESI) 510[ M + H]+
The second step is that:
100mg of Compound (8) and 10% Pd-C100mg were added to 10ml of methanol, and the mixture was hydrogenated at room temperature overnight. Filtration, concentration of the filtrate and purification of the residue by column chromatography gave 73mg of compound (9). MS (ESI negative ion) 418[ M-H]-
The third step:
50mg of the compound (9) and 50mg of the starting material (2) were dissolved in 5ml of methylene chloride, and 20mg of HOBt (1-hydroxybenzotriazole) and 40mg of EDC (1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride) were added to the solution to react at room temperature overnight. Distilling off dichloromethane under reduced pressure, and purifying by column chromatography to obtain compound (10), 23mg. MS (ESI):681[ M + H ]]+1H NMR(400MHz,CDCl3)δ10.56(br,1H),10.08(br,1H),8.81(s,1H),8.46(s,1H),8.28(s,1H),7.42(dd,J=8.0,7.2Hz,1H),7.33(dd,J=3.2,2.0Hz,1H),7.03(d,J=7.2Hz,1H),6.84(d,J=8.8Hz,1H),6.71(dd,J=3.6,1.6Hz,1H),6.46(t,J=5.2Hz,1H),4.94(dd,J=12.0,5.2Hz,1H),4.51(s,2H),3.65-3.75(m,4H),3.57(t,J=4.8Hz,2H),3.45(m,4H),3.40(s,2H),2.71-2.88(m,4H),2.1(m,2H)。
EXAMPLE 4 preparation of Compound 13
Figure BDA0001397384370000201
The first step is as follows:
200mg of the raw material (2) was dissolved in 10ml of dichloromethane, 100mg of triethylamine and 200mg of 5-benzyloxypentanesulfonyl chloride were added to the solution, the mixture was reacted overnight at room temperature, dichloromethane was distilled off under reduced pressure, and the product was purified by column chromatography to give 312mg of the compound (11). MS (ESI) 520[ M + H]+
The second step is that:
300mg of Compound (11) and 300mg of 10% Pd-C were added to 20ml of methanol, and the reaction was hydrogenated at room temperature overnight. Filtering, concentrating the filtrate, and purifying by column chromatography to obtain compound (12)211mg, MS (ESI):430[ M + H [)]+
The third step:
dissolving 4-hydroxy thalidomide 100mg, triphenylphosphine 100mg and compound (12)150mg in 30ml anhydrous tetrahydrofuran, adding DIAD (diisopropyl azodicarboxylate) 100mg dropwise, reacting at room temperature for 3h, removing tetrahydrofuran under reduced pressure, and purifying by column chromatography to obtain compound (13)97 mg. MS (ESI) 686[ M + H]+1H NMR(400MHz,CDCl3)δ10.55(br,1H),10.17(br,1H),8.80(s,1H),8.41(s,1H),8.29(s,1H),7.53(t,J=7.6Hz,1H),7.31(m,2H),7.08(d,J=8.0Hz,1H),6.75(d,J=3.6Hz,1H),5.01(m,1H),3.75-3.95(m,6H),3.53(m,2H),3.38(s,2H),2.58-2.77(m,2H),2.06-2.35(m,2H),1.63-1.95(m,4H),1.40(m,2H)。
The compounds prepared by a method analogous to examples 2, 3 are shown in the following table:
Figure BDA0001397384370000202
the compounds prepared by a method analogous to that of example 2 are shown in the following table:
Figure BDA0001397384370000203
Figure BDA0001397384370000211
example 5 test of the inhibitory Activity of Compounds on JAK1/2 Using Caliperassay method
The experimental procedure was as follows:
1 Xkinase reaction buffer (50mM HEPES, pH 7.5; 0.0015% Brij-35) and kinase reaction stop solution (100mM HEPES, pH 7.5; 0.0015% Brij-35; 0.2% Coating Reagent; 50mM EDTA) were prepared;
preparing an experimental sample: to a 96-well plate, 100. mu.L of 5. mu.M sample solution (dissolved in 100% DMSO) was added to obtain 50X sample solution. Two wells containing 100 μ L of 100% DMSO alone were placed on the same plate as controls. One was used as no-load control and the other as no-load control. mu.L of the sample and 90. mu.L of 1 Xkinase reaction buffer were added to a 96-well plate as a transfer plate. The plates were shaken for 10 minutes.
Preparing a test plate: samples prepared in 96-well transfer plates were taken 5. mu.L each to 384-well plates.
Kinase reaction: mu.L of a 2.5 XJAK 1/2 kinase solution (kinase diluted with 1 Xkinase reaction buffer) was added to 5. mu.L of a 5 Xcompound solution (dissolved in DMSO, diluted 10-fold with water), incubated at room temperature for 10min, and then 10. mu.L of a 2.5 Xsubstrate peptide solution (FAM-labeled peptide and ATP diluted in 1 Xkinase reaction buffer) was added.
Termination of the kinase reaction: after a reaction time at 28 ℃ 25. mu.L of a kinase reaction stop solution was added.
Fluorescence (F) was tested on Caliper and data was collected.
Calculating the inhibition rate of the kinase activity: percent inhibition of kinase activity(FDMSO control-FSample (I))/(FDMSO control-FNegative control) X 100, DMSO as a solution control, no kinase as a negative control.
The results show that the inhibitory activity of the compound (concentration 100nM) against JAK1/2 is shown in the following Table:
Figure BDA0001397384370000212
Figure BDA0001397384370000221
example 6 Western blot to detect the activity of the compounds to degrade JAK1 and JAK2 proteins
Cell lines: jurkat cell line was cultured in RPMI1640 containing 10% calf serum at 37 ℃ in 5% CO2And culturing in an incubator with saturated humidity.
A DMSO control group and a compound intervention group (10 mu M) are arranged, cells are collected after 6 hours of treatment, 100 mu L of precooled cell lysate is added, the cells are lysed for 30min on ice, total cell protein is extracted, and the protein concentration is determined and quantified by a bicinchoninic acid (BCA) method. Preparing gel conventionally, loading, performing electrophoresis, then transferring a membrane, sealing, adding rabbit anti-human JAK1 (1: 500) and rabbit anti-human JAK2 (1: 500) respectively, incubating overnight at 4 ℃, adding horseradish peroxidase-labeled goat anti-rabbit IgG (1: 5000) after rinsing, developing with ECL developer after rinsing, scanning and imaging by a Bio-Rad gel imaging system, and performing computer software processing and analysis. The control was referenced to glyceraldehyde phosphate dehydrogenase (GAPDH).
Grey scale analysis was performed on each band using Image J software to calculate the degradation rate of compounds degrading JAK1 or JAK2 proteins. The calculation formula is as follows:
Figure BDA0001397384370000222
the results show that the degradation rate of compound (10. mu.M) for JAK1/2 protein in Jurkat cells is shown in the following table:
Figure BDA0001397384370000223
Figure BDA0001397384370000231
example 7 CCK8 assay for inhibition of Jurkat cell proliferation (human leukemia T lymphocytes)
The inhibitory effect of the compounds on the proliferation of Jurkat cells was tested in vitro using the CCK8 assay. The method comprises the following specific steps:
culturing Jurkat cells in 1640 culture medium containing 10% calf serum, inoculating to 96-well plate, and culturing at 2 × 105Cells/well, 5% CO at 37 ℃2In an incubator. The compound was dissolved in Dimethylsulfoxide (DMSO) to give a solution having a concentration of 10mM, and then diluted with phosphate buffer to the desired concentration, and the diluted solution was added to the above 96-well plate at 2 wells per concentration and 10. mu.l per well, and each concentration was subjected to two parallel tests. DMSO was diluted accordingly in a gradient and added to the plate as a control.
Subjecting the 96-well plate to 5% CO at 37 deg.C2After culturing for 48 hours in the cell culture box, 10 μ l of CCK8 solution is added to each well, and the incubation is continued for 1-4 hours in the culture box. The 465nm light absorption value was measured.
The relative viability of the cells after compound treatment was calculated from the light absorption values. The calculation formula is as follows:
Figure BDA0001397384370000232
IC of compound on Jurkat cells was calculated by software50
The results show that the in vitro proliferation inhibitory effect of the synthesized compounds on Jurkat cells is shown in the following table:
Figure BDA0001397384370000233
Figure BDA0001397384370000241
the results show that the synthesized compound has good activity of inhibiting Jurkat cells.
Example 8 testing of in vivo anti-rheumatoid arthritis Activity
The anti-rheumatoid arthritis effect of the compound (3) is determined by taking Wistar rat rheumatoid arthritis induced by Freund's complete adjuvant as a model. The method comprises the following specific steps:
1) test sample
Sample preparation: compound (3).
2) Preparation method
Sample preparation: compound (3) dissolved in a physiological saline injection solution at the time of preparation
3) Animals and adjuvants
Wistar rats 18, male, weighing 170-200 g.
Adjuvant: complete Freund's Adjuvant (CFA), Sigma.
4) Test method
Wistar rats were randomly divided into 3 groups of 6 rats each. The rats were treated with administration of 0.1mL of CFA injected intradermally into the plantar layer of the left hind paw of rats, once a day on day 11 after the model creation of the rats, and after the treatment on day 22, the volume of the left hind paw was measured, and the rate of inhibition of foot swelling was calculated.
Figure BDA0001397384370000242
5) Test results
The foot swelling inhibition rate of the compound (3) on rheumatoid arthritis induced by complete Freund's adjuvant is shown in the following table:
group of Foot swelling inhibition ratio (%)
Blank control group -
Model set -
Compound (3) 85
The results of the in vivo experiments show that the compound (3) has good anti-rheumatoid arthritis activity in vivo.
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. A compound of formula I, or a pharmaceutically acceptable salt thereof:
Figure FDA0003266915810000011
wherein:
Figure FDA0003266915810000012
represents a single bond;
Figure FDA0003266915810000013
represents a single bond or a double bond;
a is absent or selected from C (═ O) and C (═ O) X1, SO2X1; wherein X1 is absent or is (CR)12R13)fO; wherein R is12、R13Each independently is H or C1-8F is an integer between 0 and 3;
w is absent or selected from O, NR17X2C (═ O) X3; wherein R is17Is H or C1-8A hydrocarbon group of (a); wherein X2 and X3 are each independently absent or NR18(ii) a Wherein R is18Is H or C1-8A hydrocarbon group of (a);
y is (CR)22R23)h(ii) a Wherein h is an integer between 0 and 30; wherein R is22、R23Each independently selected from H, cyano, hydroxy, amino, C1-8A hydrocarbon group of (a);
z is (CR)24R25)i(ii) a Wherein i is an integer between 0 and 30; wherein R is24、R25Each independently selected from H, cyano, hydroxy, amino, and C1-8A hydrocarbon group of (a);
b is absent or is O;
x is C (═ O);
R1、R8each independently selected from H and C1-8A hydrocarbyl group;
R2、R5each independently selected from: hydrogen and cyano-substituted C1~8A hydrocarbyl group;
R3、R6、R7、R9、R10each independently selected from: h and C1-8A hydrocarbyl group;
R4selected from H and C1-8A hydrocarbon group of (a);
a is an integer between 0 and 5;
b is an integer between 0 and 3;
c is an integer between 0 and 30;
d is an integer between 0 and 5;
e is an integer between 0 and 2.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:
in said formula I, A is absent; w is X2C (═ O) X3 where X3 is NR18And X2 is absent, or X2 is NR18And X3 is absent; y is (CR)22R23)hWherein R is22、R23Each independently selected from H and C1-4H is an integer between 1 and 6; z is (CR)24R25)iWherein R is24、R25Each independently selected from H and C1-4I is an integer between 1 and 6; c is 0; or
In said formula I, A is absent; w is absent or is O; y is (CR)22R23)hWherein R is22、R23Each independently selected from H and C1-4H is an integer between 0 and 3; b is O; z is (CR)24R25)iWherein R is24、R25Each independently selected from H and C1-4I is an integer between 0 and 3; c is an integer between 1 and 6; or
Wherein a is C (═ O) X1, where X1 is absent or is (CR)12R13)fO, where f is an integer between 0 and 2, R12、R13Each independently is hydrogen or C1-4Alkyl groups of (a); w is NR17Wherein R is17Is H or C1-4A hydrocarbon group of (a); y is (CR)22R23)hWherein R is22、R23Each independently selected from H and C1-4H is an integer between 0 and 3; z is (CR)24R25)iWherein R is24、R25Each independently selected from H and C1-4I is an integer between 0 and 3; b is O; c is an integer between 1 and 4; or
In the formula I, A is SO2X1, wherein X1 is absent; w is O; y is (CR)22R23)hWherein R is22、R23Each independently selected from H and C1-4H is a whole number between 1 and 6Counting; z is (CR)24R25)iWherein R is24、R25Each independently selected from H and C1-4I is an integer between 0 and 3; c is 0; or
In said formula I, A is absent; w is NR17Wherein R is17Is H or C1-4A hydrocarbon group of (a); y is (CR)22R23)hWherein R is22、R23Each independently selected from H and C1-4H is an integer between 0 and 3; z is (CR)24R25)iWherein R is24、R25Each independently selected from H and C1-4I is an integer between 0 and 4; b is O; c is an integer between 1 and 6; or
In said formula I, A is absent; deletion of W; y is (CR)22R23)hWherein R is22、R23Each independently selected from H and C1-4H is an integer between 0 and 3; z is (CR)24R25)iWherein R is24、R25Each independently selected from H and C1-4I is an integer between 0 and 3; b is O; c is an integer between 1 and 10.
3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein in formula I, X is C (═ O).
4. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R is1Is H.
5. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R is4Is H.
6. The compound of claim 1, or a pharmaceutically acceptable salt thereof, selected from the group consisting of:
Figure FDA0003266915810000031
7. a pharmaceutical composition comprising a compound of claim 1, or a pharmaceutically acceptable salt, prodrug thereof, and a pharmaceutically acceptable carrier.
8. Use of a compound of claim 1, or a pharmaceutically acceptable salt thereof, for:
(a) preparing a medicament for treating diseases related to the activity or expression amount of JAK1 and/or JAK 2;
(b) preparing a JAK1 and/or JAK2 targeted inhibitor or degrader;
(c) non-therapeutically inhibiting or degrading JAK1 and/or JAK2 in vitro; and/or
(d) Non-therapeutically inhibiting tumor cell proliferation in vitro.
9. The use according to claim 8, wherein the disease associated with JAK1 and/or JAK2 activity or expression is a neoplastic or autoimmune disease.
10. An in vitro non-therapeutic method of inhibiting or degrading JAK1 and/or JAK2 comprising the steps of: administering to a subject an effective amount of a compound of formula I as described in claim 1 or a pharmaceutically acceptable salt thereof, or administering to a subject an inhibitory effective amount of a pharmaceutical composition as described in claim 7.
CN201710783082.5A 2017-09-03 2017-09-03 Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity Active CN109422753B (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN201710783082.5A CN109422753B (en) 2017-09-03 2017-09-03 Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity
CN201880004074.9A CN109937203B (en) 2017-09-03 2018-09-02 Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity
PCT/CN2018/103707 WO2019042442A1 (en) 2017-09-03 2018-09-02 Compound having tyrosine protein kinase jak1- or jak2-inhibittion and degradation activity

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201710783082.5A CN109422753B (en) 2017-09-03 2017-09-03 Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity

Publications (2)

Publication Number Publication Date
CN109422753A CN109422753A (en) 2019-03-05
CN109422753B true CN109422753B (en) 2021-12-31

Family

ID=65512924

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201710783082.5A Active CN109422753B (en) 2017-09-03 2017-09-03 Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity
CN201880004074.9A Active CN109937203B (en) 2017-09-03 2018-09-02 Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN201880004074.9A Active CN109937203B (en) 2017-09-03 2018-09-02 Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity

Country Status (2)

Country Link
CN (2) CN109422753B (en)
WO (1) WO2019042442A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116583509A (en) * 2020-10-07 2023-08-11 上海睿跃生物科技有限公司 Compounds and methods for treating cancer
CN114478532A (en) * 2020-10-26 2022-05-13 上海美志医药科技有限公司 Targeted Btk degradation compound and anti-tumor application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2017200685A1 (en) * 2005-12-13 2017-02-23 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
CN107074867A (en) * 2014-08-21 2017-08-18 辉瑞公司 It is used as the aminopyrimidine based compound of JAK inhibitor

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007130712A1 (en) * 2006-01-31 2007-11-15 Janssen Pharmaceutica, Nv Substituted dipiperidine as ccr2 antagonists for the treatment of inflammatory diseases
SG183551A1 (en) * 2010-03-10 2012-10-30 Incyte Corp Piperidin-4-yl azetidine derivatives as jak1 inhibitors
CA2923829A1 (en) * 2013-09-10 2015-03-19 Synta Pharmaceuticals Corp. Targeted therapeutics
CN106336413B (en) * 2015-07-09 2021-04-20 广东东阳光药业有限公司 Compounds as JAK inhibitors and uses thereof
CN106432246B (en) * 2015-08-05 2020-07-07 广东东阳光药业有限公司 Heteroaromatic compounds and their use in medicine

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2017200685A1 (en) * 2005-12-13 2017-02-23 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
CN107074867A (en) * 2014-08-21 2017-08-18 辉瑞公司 It is used as the aminopyrimidine based compound of JAK inhibitor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Chemically induced degradation of CDK9 by a proteolysis targeting chimera (PROTAC);Caroline M. Robb et al.;《Chem. Commun.》;20170614;第53卷;第7577-7580页 *

Also Published As

Publication number Publication date
WO2019042442A1 (en) 2019-03-07
CN109937203A (en) 2019-06-25
CN109422753A (en) 2019-03-05
CN109937203B (en) 2021-07-23

Similar Documents

Publication Publication Date Title
CN110461846B (en) Compound capable of inhibiting and degrading Bruton tyrosine protein kinase Btk activity
CN109963844B (en) Compound for inhibiting and degrading tyrosine protein kinase ALK
KR102396717B1 (en) Treatment of b-cell malignancies by a combination jak and pi3k inhibitor
CN109952304B (en) CDK (CDK kinase) inhibiting and degrading compound
ES2461967T3 (en) Pyrrolo [2,3-d] pyrimidine compounds
CN111662294A (en) Compound with activity of degrading Btk
CA3184924A1 (en) Immunomodulator
EP4032890A1 (en) Heterocyclic amide compound, pharmaceutically acceptable salt thereof, and preparation method therefor and use thereof
CN109937203B (en) Compounds capable of inhibiting and degrading tyrosine protein kinase JAK1 or JAK2 activity
CN112979656A (en) Compound for targeted degradation of BTK protein
CN109963853B (en) Compound with activity of degrading tyrosine protein kinase JAK3
CN113490669B (en) Compound with activity of degrading Btk
KR20240054339A (en) Compounds as TYK2/JAK1 pseudokinase domain inhibitors and methods of synthesis and use
EP3618831A1 (en) An anti-cancer stemness drug
KR102628246B1 (en) Selective A2A receptor antagonist
CN114149435A (en) Targeted Btk degradation compound and application thereof
RU2782469C2 (en) Apoptosis-inducing agents
WO2023201044A2 (en) Jak inhibitor analogs, formulations, and uses thereof
CN116120327A (en) Beta-elemene 13, 14-position symmetrical disubstituted derivative and preparation method and application thereof
CN114761011A (en) Substituted (7H-pyrrolo [2,3-D ] pyrimidin-4-yl) amino compounds useful as JAK1 inhibitors

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