CN108602799B - Kinase inhibitor - Google Patents

Kinase inhibitor Download PDF

Info

Publication number
CN108602799B
CN108602799B CN201780010075.XA CN201780010075A CN108602799B CN 108602799 B CN108602799 B CN 108602799B CN 201780010075 A CN201780010075 A CN 201780010075A CN 108602799 B CN108602799 B CN 108602799B
Authority
CN
China
Prior art keywords
methyl
pyridin
amine
alkyl
isopropyl
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
CN201780010075.XA
Other languages
Chinese (zh)
Other versions
CN108602799A (en
Inventor
赵兴东
李同双
周祖文
王宪龙
陈岭
容悦
刘启洪
陈志方
张华杰
谭锐
谭浩瀚
李志福
张卫鹏
姜立花
刘研新
令狐莉
林敏�
孙婧
王为波
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.)
Chongqing Fushang Yuanchuang Pharmaceutical Technology Co.,Ltd.
Original Assignee
Shanghai Fochon Pharmaceutical Co Ltd
Fochon Pharmaceuticals 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 Fochon Pharmaceutical Co Ltd, Fochon Pharmaceuticals Ltd filed Critical Shanghai Fochon Pharmaceutical Co Ltd
Publication of CN108602799A publication Critical patent/CN108602799A/en
Application granted granted Critical
Publication of CN108602799B publication Critical patent/CN108602799B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • 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
    • C07D487/08Bridged systems
    • 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
    • C07D487/10Spiro-condensed systems

Abstract

The present invention relates to a class of CDK4/6 inhibitors, as well as pharmaceutical compositions and methods of use thereof.

Description

Kinase inhibitor
This application claims priority to U.S. provisional application 62/292,259, which is incorporated herein by reference in its entirety.
Technical Field
The present invention relates to a class of compounds which inhibit the activity of CDK4/6 kinase, or a pharmaceutically acceptable salt thereof, and as medicaments for the treatment of hyperproliferative diseases, such as cancer and inflammation.
Background
Hyperproliferative diseases such as cancer and inflammation have attracted the academic community to provide effective treatments for them. And efforts have been made in this regard to identify and target specific mechanisms that play a role in proliferative diseases.
The development of tumors is closely related to the genetic variation and abnormal regulation of cyclin-dependent kinases (CDKs) and their regulatory proteins, suggesting that CDK inhibitors may be effective anticancer therapies.
CDKs are serine/threonine protein kinases that are the prime movers for cell cycle and cell proliferation. CDKs regulate the initiation, progression and completion of the mammalian cell cycle and are critical for cell growth. Most known CDKs, including CDK1 through CDK9, are involved directly or indirectly in cell cycle progression. CDKs directly involved in cell cycle progression, such as CDK1-4 and CDK6, are classified as G1, S or G2M phase enzymes. Abnormal proliferation is a characteristic of cancer cells, and CDK dysfunction occurs at high frequency in many solid tumors.
Such therapies that target general CDKs or specific CDKs for the treatment of dysplastic diseases, such as cancer treatment, have great potential. CDK inhibitors may also be used to treat other diseases such as viral infections, autoimmune diseases and neurodegenerative diseases. CDK targeted therapies may also be used in combination with other therapeutic agents for the treatment of the above-mentioned disorders.
Therefore, compounds having CDK inhibitory activity are of great interest for the prevention and treatment of cancer. Although inhibitors of CDK4/6 have been reported in the literature, e.g., WO 2010075074, many have short half-lives or are toxic. Thus, there is an increasing need for new CDK4/6 inhibitors that have advantages in at least one of therapeutic efficacy, stability, selectivity, safety, pharmacodynamic and pharmacokinetic properties. The present invention relates to a novel class of CDK4/6 inhibitors.
Disclosure of Invention
The invention relates to a novel 6-5-membered fused azole ring derivative, a pharmaceutical composition thereof and application thereof as a medicament.
In one aspect, the invention provides a compound of formula (I) or (II):
Figure BDA0001755256780000021
or a pharmaceutically acceptable salt thereof, wherein,
q is selected from aryl and heteroaryl;
each R1Independently selected from hydrogen, halogen, hydroxy, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylamino, di (alkyl) amino, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from RxSubstituted with the substituent(s);
each R2Independently selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA1RB1、-ORA1、-S(O)rRA1、-S(O)2ORA1、-OS(O)2RA1、-P(O)RA1RB1、-P(O)(ORA1)(ORB1)、-C(O)RA1、-C(O)ORA1、-OC(O)RA1、-C(O)NRA1RB1、-NRA1C(O)RB1、-OC(O)NRA1RB1、-NRA1C(O)ORB1、-NRA1C(O)NRA1RB1、-NRA1C(S)NRA1RB1、-S(O)rNRA1RB1、-NRA1S(O)rRB1、-NRA1S(O)2NRA1RB1、-S(O)(=NRE1)RB1、-N=S(O)RA1RB1、-NRA1S(O)(=NRE1)RB1、-S(O)(=NRE1)NRA1RB1、-NRA1S(O)(=NRE1)NRA1RB1、-C(=NRE1)RA1、-C(=N-ORB1)RA1、-C(=NRE1)NRA1RB1、-NRA1C(=NRE1)RB1and-NRA1C(=NRE1)NRA1RB1Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RxSubstituted with the substituent(s);
each R3Independently selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA2RB2、-ORA2、-S(O)rRA2、-S(O)2ORA2、-OS(O)2RA2、-P(O)RA2RB2、-P(O)(ORA2)(ORB2)、-C(O)RA2、-C(O)ORA2、-OC(O)RA2、-C(O)NRA2RB2、-NRA2C(O)RB2、-OC(O)NRA2RB2、-NRA2C(O)ORB2、-NRA2C(O)NRA2RB2、-NRA2C(S)NRA2RB2、-S(O)rNRA2RB2、-NRA2S(O)rRB2、-NRA2S(O)2NRA2RB2、-S(O)(=NRE2)RB2、-N=S(O)RA2RB2、-NRA2S(O)(=NRE2)RB2、-S(O)(=NRE2)NRA2RB2、-NRA2S(O)(=NRE2)NRA2RB2、-C(=NRE2)RA2、-C(=N-ORB2)RA2、-C(=NRE2)NRA2RB2、-NRA2C(=NRE2)RB2and-NRA2C(=NRE2)NRA2RB2Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXSubstituted with the substituent(s);
each R4Selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA3RB3、-ORA3、-S(O)rRA3、-S(O)2ORA3、-OS(O)2RA3、-P(O)RA3RB3、-P(O)(ORA3)(ORB3)、-C(O)RA3、-C(O)ORA3、-OC(O)RA3、-C(O)NRA3RB3、-NRA3C(O)RB3、-OC(O)NRA3RB3、-NRA3C(O)ORB3、-NRA3C(O)NRA3RB3、-NRA3C(S)NRA3RB3、-S(O)rNRA3RB3、-NRA3S(O)rRB3、-NRA3S(O)2NRA3RB3、-S(O)(=NRE3)RB3、-N=S(O)RA3RB3、-NRA3S(O)(=NRE3)RB3、-S(O)(=NRE3)NRA3RB3、-NRA3S(O)(=NRE3)NRA3RB3、-C(=NRE3)RA3、-C(=N-ORB3)RA3、-C(=NRE3)NRA3RB3、-NRA3C(=NRE3)RB3and-NRA3C(=NRE3)NRA3RB3Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl andheteroaryl is unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXSubstituted with the substituent(s);
each R5Independently selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA4RB4、-(CH2)tNRA4RB4、-ORA4、-S(O)rRA4、-S(O)2ORA4、-OS(O)2RA4、-P(O)RA4RB4、-P(O)(ORA4)(ORB4)、-C(O)RA4、-C(O)ORA4、-OC(O)RA4、-C(O)NRA4RB4、-NRA4C(O)RB4、-OC(O)NRA4RB4、-NRA4C(O)ORB4、-NRA4C(O)NRA4RB4、-NRA4C(S)NRA4RB4、-S(O)rNRA4RB4、-NRA4S(O)rRB4、-NRA4S(O)2NRA4RB4、-S(O)(=NRE4)RB4、-N=S(O)RA4RB4、-NRA4S(O)(=NRE4)RB4、-S(O)(=NRE4)NRA4RB4、-NRA4S(O)(=NRE4)NRA4RB4、-C(=NRE4)RA4、-C(=N-ORB4)RA4、-C(=NRE4)NRA4RB4、-NRA4C(=NRE4)RB4and-NRA4C(=NRE4)NRA4RB4Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXSubstituted with the substituent(s);
each RA1、RA2、RA3、RA4、RB1、RB2、RB3And RB4Independently selected from hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from RXSubstituted with the substituent(s);
or each "RA1And RB1”、“RA2And RB2”、“RA3And RB3"or" RA4And RB4Taken together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0,1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be interrupted by 1,2 or 3RXSubstituted by groups;
each RE1、RE2、RE3And RE4Independently selected from hydrogen, C1-10Alkyl, CN, NO2、-ORa1、-SRa1、-S(O)rRa1、-C(O)Ra1、-C(O)ORa1、-C(O)NRa1Rb1and-S (O)rNRa1Rb1
Each RXIndependently selected from C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, halogen, CN, NO2、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tS(O)rRb1、-(CRc1Rd1)tS(O)2ORb1、-(CRc1Rd1)tOS(O)2Rb1、-(CRc1Rd1)tP(O)Ra1Rb1、-(CRc1Rd1)tP(O)(ORa1)(ORb1)、-(CRc1Rd1)tC(O)Ra1、-(CRc1Rd1)tC(O)ORb1、-(CRc1Rd1)tOC(O)Rb1、-(CRc1Rd1)tC(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(O)Rb1、-(CRc1Rd1)tOC(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(O)ORb1、-(CRc1Rd1)tNRa1C(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(S)NRa1Rb1、-(CRc1Rd1)tS(O)rNRa1Rb1、-(CRc1Rd1)tNRa1S(O)rRb1、-(CRc1Rd1)tNRa1S(O)2NRa1Rb1、-(CRc1Rd1)tS(O)(=NRe1)Rb1、-(CRc1Rd1)tN=S(O)Ra1Rb1、-(CRc1Rd1)tNRa1S(O)(=NRe1)Rb1、-(CRc1Rd1)tS(O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1S(O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tC(=NRe1)Ra1、-(CRc1Rd1)tC(=N-ORb1)Ra1、-(CRc1Rd1)tC(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)Rb1And- (CR)c1Rd1)tNRa1C(=NRe1)NRa1Rb1Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RYSubstituted with the substituent(s);
each Ra1And Rb1Independently selected from hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from RYSubstituted with the substituent(s);
or Ra1And Rb1Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0,1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be interrupted by 1,2 or 3RYSubstituted by groups;
each Rc1And Rd1Independently selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from RYSubstituted with the substituent(s);
or Rc1And Rd1Together with the carbon atom or atoms to which they are attached, form a 3-12 membered ring containing 0,1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may beOptionally substituted by 1,2 or 3RYSubstituted by groups;
each Re1Independently selected from hydrogen, C1-10Alkyl radical, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, CN, NO2、-ORa2、-SRa2、-S(O)rRa2、-C(O)Ra2、-C(O)ORa2、-S(O)rNRa2Rb2and-C (O) NRa2Rb2
Each RYIndependently selected from C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, halogen, CN, NO2、-(CRc2Rd2)tNRa2Rb2、-(CRc2Rd2)tORb2、-(CRc2Rd2)tS(O)rRb2、-(CRc2Rd2)tS(O)2ORb2、-(CRc2Rd2)tOS(O)2Rb2、-(CRc2Rd2)tP(O)Ra2Rb2、-(CRc2Rd2)tP(O)(ORa2)(ORb2)、-(CRc2Rd2)tC(O)Ra2、-(CRc2Rd2)tC(O)ORb2、-(CRc2Rd2)tOC(O)Rb2、-(CRc2Rd2)tC(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(O)Rb2、-(CRc2Rd2)tOC(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(O)ORb2、-(CRc2Rd2)tNRa2C(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(S)NRa2Rb2、-(CRc2Rd2)tS(O)rNRa2Rb2、-(CRc2Rd2)tNRa2S(O)rRb2、-(CRc2Rd2)tNRa2S(O)2NRa2Rb2、-(CRc2Rd2)tS(O)(=NRe2)Rb2、-(CRc2Rd2)tN=S(O)Ra2Rb2、-(CRc2Rd2)tNRa2S(O)(=NRe2)Rb2、-(CRc2Rd2)tS(O)(=NRe2)NRa2Rb2、-(CRc2Rd2)tNRa2S(O)(=NRe2)NRa2Rb2、-(CRc2Rd2)tC(=NRe2)Ra2,-(CRc2Rd2)tC(=N-ORb2)Ra2、-(CRc2Rd2)tC(=NRe2)NRa2Rb2、-(CRc2Rd2)tNRa2C(=NRe2)Rb2And- (CR)c2Rd2)tNRa2C(=NRe2)NRa2Rb2Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from OH, CN, amino, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Ra2And Rb2Independently selected from hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylamino, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
or Ra2And Rb2Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0,1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be substituted with 1-2 heteroatoms independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Rc2And Rd2Independently selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkyl radicalsAmino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylamino, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
or Rc2And Rd2Together with the carbon atom or atoms to which they are attached form a 3-12 membered ring containing 0,1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may optionally be substituted by 1 or 2 heteroatoms independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Re2Independently selected from hydrogen, CN, NO2、C1-10Alkyl radical, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, -C (O) C1-4Alkyl, -C (O) C3-10Cycloalkyl, -C (O) OC1-4Alkyl, -C (O) OC3-10Cycloalkyl, -C (O) N (C)1-4Alkyl radical)2、-C(O)N(C3-10Cycloalkyl radicals2、-S(O)2C1-4Alkyl, -S (O)2C3-10Cycloalkyl, -S (O)2N(C1-4Alkyl radical)2and-S (O)2N(C3-10Cycloalkyl radicals2
m is selected from 0,1, 2,3 and 4;
n is selected from 0,1 and 2;
p is selected from 0,1 and 2;
each r is independently selected from 0,1 and 2;
each t is independently selected from 0,1, 2,3, and 4.
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
In another aspect, the invention provides a method for modulating CDK4/6, comprising administering to a system or subject in need thereof a therapeutically effective amount of a compound of formula (I) or (II) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof, thereby modulating CDK 4/6.
In another aspect, the invention also provides a method of treating, ameliorating or preventing a disorder responsive to inhibition of CDK4/6 comprising administering to a system or subject in need thereof an effective amount of a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, and optionally in combination with a second therapeutic agent, for treating the disorder described above.
Alternatively, the invention provides the use of a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disorder mediated by CDK 4/6. In certain embodiments, the compounds may be used alone or in combination with a second therapeutic agent in the treatment of a CDK4/6 mediated disorder.
Alternatively, the present invention provides a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, for use in the treatment of a CDK4/6 mediated disorder.
In particular, wherein the condition includes, but is not limited to, an autoimmune disease, a transplantation disease, an infectious disease, or a cell proliferative disorder disease.
In addition, the present invention provides methods of treating a cell proliferative disorder comprising administering to a system or subject in need thereof an effective amount of a compound of formula (I) or (II), or a pharmaceutically acceptable salt or pharmaceutical composition thereof, optionally in combination with a second therapeutic agent, to treat the disorder.
Alternatively, the present invention provides the use of a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a cell proliferative disorder. In particular embodiments, the compounds can be used alone or in combination with a second therapeutic agent to treat a cell proliferative disorder.
In particular, wherein the cell proliferative disorder includes, but is not limited to, lymphoma, osteosarcoma, melanoma, or a tumor of the breast, kidney, prostate, colorectal, thyroid, ovarian, pancreatic, neuronal, lung, uterine or gastrointestinal tract.
In the above methods of using the compounds of the present invention, a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, can be administered to a system comprising cells or tissues, or to an individual including a mammalian individual, such as a human or animal individual.
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 this patent belongs. All patents, patent applications, published publications, etc. referred to herein are incorporated by reference in their entirety unless otherwise indicated. As used in this patent, the same terms are defined differently than the definitions in this section.
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 any claims. In this application, the use of the singular includes the plural unless otherwise indicated. It is noted that, in the specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. It is also noted that "or" represents "and/or" unless stated otherwise. Furthermore, "comprising," "including," and like terms are not intended to be limiting.
Standard definitions of chemical terms are referred to in the reference books, including Carey and Sundberg "ADVANCED ORGANIC CHEMISTRY, 4 th edition. Volumes "A (2000) and B (2001), Plenum Press, New York. Unless otherwise indicated, the conventional techniques of mass spectrometry, nuclear magnetic resonance, high performance liquid chromatography, infrared and ultraviolet spectroscopy, and pharmacology used in this patent are prior art. Unless specifically defined, the nomenclature, protocols, and techniques involved in analytical chemistry, synthetic organic chemistry, pharmaceutical and pharmaceutical chemistry are those known in the art. Standard techniques are available for chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and administration, and treatment of patients. The reaction and purification techniques may be carried out with reference to the manufacturer's instructions, or with reference to known, commonly used techniques, or with reference to the methods described in this patent. The techniques and procedures described above can be performed using methods that are conventional and well known in the literature cited in this specification. In the specification, groups and substituents may be selected by one skilled in the art to form stable structures and compounds.
When a substituent is referred to by a formula, the substituents in the formula are written from left to right as they are from right to left. As a non-limiting example, CH2O and OCH2The same is true.
As used herein, "optionally substituted" includes unsubstituted or substituted. "substituted" means that the hydrogen atom is replaced with a substituent. It is noted that substituents on a particular atom are constrained by their valency. In the definition section, "Ci-j"refers to a range including a start point and an end point, where i and j are both integers indicating the number of carbon atoms. E.g. C1-4,C1-10,C3-10And the like.
"alkyl", alone or in combination with other terms, refers to branched and straight chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. Unless otherwise indicated, "alkyl" means C1-10An alkyl group. For example, "C1-6C in alkyl1-C6"refers to a linear or branched arrangement of groups having 1,2,3,4, 5 or 6 carbon atoms. For example, "C1-8Alkyl "includes, but is not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, pentyl, hexyl, heptyl, and octyl.
"cycloalkyl" whether used alone or in combination with other terms, refers to a monocyclic or bridged hydrocarbon system. Monocyclic cycloalkyl groups contain 3 to 10 carbon atoms, no heteroatoms, no double bonds. Examples of monocyclic systems include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Monocyclic cycloalkyl contains one or two alkylene bridges, each of which contains 1,2 or 3 carbon atoms, which are bonded to two non-adjacent carbon atoms in the ring system. Representative examples of bridged ring hydrocarbon systems include, but are not limited to, bicyclo [3.1.1] heptane, bicyclo [2.2.1] heptane, bicyclo [2.2.2] octane, bicyclo [3.2.2] nonane, bicyclo [3.3.1] nonane, bicyclo [4.2.1] nonane, tricyclo [3.3.1.03,7] nonane, and tricyclo [3.3.1.13,7] decane (adamantane). The monocyclic and bridged hydrocarbon rings can be attached to the parent ring through any suitable atom in the ring system.
"alkenyl", alone or in combination with other terms, refers to a nonaromatic, straight chain, branched or cyclic hydrocarbon radical containing from 2 to 10 carbon atoms and having at least one carbon-carbon double bond. In some embodiments, 1 carbon-carbon double bond is present, and up to 4 non-aromatic carbon-carbon double bonds may be present. Thus, "C2-6Alkenyl "means alkenyl containing 2 to 6 carbon atoms. Alkenyl groups include, but are not limited to, ethenyl, propenyl, butenyl, 2-methylbutenyl, and cyclohexenyl. The straight, branched or cyclic portion of the alkenyl group may contain a double bond, and substituted alkenyl groups, if indicated, may be substituted.
"alkynyl", whether used alone or in combination with other terms, refers to a straight, branched or cyclic hydrocarbon radical containing from 2 to 10 carbon atoms and at least one carbon-carbon triple bond. In some embodiments, there may be 3 carbon-carbon triple bonds. Thus, "C2-6Alkynyl "refers to alkynyl groups containing 2-6 carbon atoms. Alkynyl groups include, but are not limited to, ethynyl, propynyl, butynyl, 3-methylbutynyl, and the like. The straight, branched or cyclic portion of the alkynyl group may contain a triple bond, and a substituted alkynyl group, if indicated, may be substituted.
"halogen" means fluorine, chlorine, bromine, iodine.
"alkoxy", used alone or in combination with other terms, refers to an alkyl group attached to an oxygen atom by a single bond. The alkoxy group is attached to the molecule through an oxygen atom. Alkoxy groups may be represented as-O-alkyl. "C1-10Alkoxy "means containing 1 to 10 carbon atomsAlkoxy, which may be linear or branched. Alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy, hexyloxy, and the like.
"Cycloalkoxy", used alone or in combination with other terms, means a cycloalkyl group attached by a single bond to an oxygen atom. The cycloalkoxy group is attached to the molecule through an oxygen atom. Cycloalkoxy can be represented as-O-cycloalkyl. "C3-10Cycloalkoxy "means a cycloalkoxy group containing 3 to 10 carbon atoms. Cycloalkoxy groups include, but are not limited to, cyclopropoxy, cyclobutoxy, cyclopentoxy, cyclohexyloxy, and the like.
"alkylthio", used alone or in combination with other terms, refers to an alkyl group attached by a single bond to a sulfur atom. Alkylthio groups are attached to the molecule through a sulfur atom. Alkylthio groups may be represented by-S-alkyl. "C1-10Alkylthio "refers to an alkylthio group containing 1 to 10 carbon atoms and can be a straight chain or branched structure. Alkylthio includes, but is not limited to, methylthio, ethylthio, propylthio, isopropylthio, butylthio, hexylthio, and the like.
"Cycloalkylsulfanyl", used alone or in combination with other terms, means a cycloalkyl group attached by a single bond to a sulfur atom. The cycloalkylthio group is bonded to the molecule through a sulfur atom. The cycloalkylthio group may be represented as-S-cycloalkyl. "C3-10Cycloalkylthio "means a cycloalkylthio group containing 3 to 10 carbon atoms. Cycloalkylthio groups include, but are not limited to, cyclopropylthio, cyclobutylthio, cyclohexylthio, and the like.
"alkylamino", used alone or in combination with other terms, refers to an alkyl group attached to a nitrogen atom by a single bond. The alkylamino group is attached to the molecule through a nitrogen atom. Alkylamino can be represented as-NH (alkyl). "C1-10Alkylamino "refers to alkylamino groups containing 1 to 10 carbon atoms, which may be straight chain or branched. Alkylamino groups include, but are not limited to, methylamino, ethylamino, propylamino, isopropylamino, butylamino, hexylamino and the like.
"Cycloalkylamino", used alone or in combination with other terms, refers to a cycloalkyl group attached to a nitrogen atom by a single bond. The cycloalkylamino group is attached to the molecule through a nitrogen atom. The cycloalkylamino group can representis-NH (cycloalkyl). "C3-10Cycloalkylamino "refers to cycloalkylamino groups containing 3 to 10 carbon atoms. Cycloalkylamino groups include, but are not limited to, cyclopropylamino, cyclobutylamino, cyclohexylamino and the like.
"Di (alkyl) amino", used alone or in combination with other terms, refers to two alkyl groups attached to a nitrogen atom by a single bond. The di (alkyl) amino group is attached to the molecule through a nitrogen atom. The di (alkyl) amino group may be represented by-N (alkyl)2. ' two (C)1-10Alkyl) amino "means a di (C) group in which the two alkyl moieties each contain 1 to 10 carbon atoms1-10Alkyl) amino, which may be linear or branched.
"aryl" includes: 5-and 6-membered aromatic carbocyclic rings, such as phenyl; bicyclic rings having at least one aromatic carbon ring, such as naphthyl, indane and 1,2,3, 4-tetrahydroquinoline, and tricyclic rings having at least one aromatic carbon ring, such as fluorene. Aryl substituents are considered to be linked through an aromatic ring if they are bicyclic or tricyclic and at least one of the rings is non-aromatic.
For example, aryl includes 5-and 6-membered aromatic carbocyclic rings fused to a 5-to 7-membered heterocyclic ring containing one or more heteroatoms selected from N, O and S, provided that the site of attachment is an aromatic carbocyclic ring. Divalent radicals, which are formed from substituted benzene derivatives and have free valence electrons present at the ring atoms, are designated as substituted phenylene radicals. Divalent radicals derived from monovalent polycyclic hydrocarbon radicals whose name ends with "-yl", which are obtained by removing one more hydrogen atom from a carbon atom containing a free valence electron, are named after the name of the monovalent radical plus "-idene (-idene)", for example, naphthyl, which has two attachment sites, is called naphthylidene. The definition of aryl, however, does not include, nor overlap with, heteroaryl, and is defined individually as follows. Thus, if one or more aromatic carbocyclic rings are fused to an aromatic ring of a heterocyclic ring, the ring system formed should be considered heteroaryl as defined herein rather than aryl.
"heteroaryl" means
A 5-to 8-membered aromatic monocyclic ring, which contains 1 to 4, and in certain embodiments 1 to 3, heteroatoms selected from N, O and S, the remainder being carbon atoms;
an 8-to 12-membered bicyclic ring containing 1 to 4, and in certain embodiments 1 to 3, heteroatoms selected from N, O and S, the remainder being carbon atoms, and wherein at least one heteroatom is present in the aromatic ring; and
11-to 14-membered tricyclic rings. The ring contains from 1 to 4, and in certain embodiments from 1 to 3, heteroatoms selected from N, O and S, the remainder being carbon atoms, and at least one of the heteroatoms is present in the aromatic ring.
When the total number of S and O atoms in the heteroaryl group is greater than 1, these heteroatoms are not adjacent to each other. In some embodiments, the total number of S and O atoms in the heteroaryl group is no greater than 2. In some embodiments, the total number of S and O atoms in the heteroaryl group is no greater than 1.
Examples of heteroaryl groups include, but are not limited to (the numbering of the attachment site is preferred, as in position 1), 2-pyridyl, 3-pyridyl, 4-pyridyl, 2, 3-pyrazinyl, 3, 4-pyrazinyl, 2, 4-pyrimidinyl, 3, 5-pyrimidinyl, 1-pyrazolyl, 2, 3-pyrazolyl, 2, 4-imidazolinyl, isoxazolyl, oxazolyl, thiazolyl, thiadiazolyl, tetrazolyl, thienyl, benzothienyl, furyl, benzofuranyl, benzimidazolyl, indolinyl, pyridazinyl (pyridizinyl), triazolyl, quinolinyl, pyrazolyl, and 5,6,7, 8-tetrahydroisoquinolinyl.
Further, heteroaryl groups include, but are not limited to, pyrrolyl, isothiazolyl, triazinyl, pyrazinyl, pyridazinyl, indolyl, benzotriazolyl, quinolyl, and isoquinolyl groups. As defined below for heterocyclic groups, "heteroaryl" includes N-oxide derivatives of nitrogen-containing heteroaryl groups.
The nomenclature of a monovalent heteroaryl group ends with the "radical", the divalent group derived by removing a hydrogen atom from a carbon atom containing a free valence electron, is that name given to the monovalent group plus a "ene" (for example: the pyridyl group having two attachment sites is called a pyridylidene. The definition of heteroaryl does not include, nor overlap with, aryl as defined above.
If the heteroaryl substituent is a bicyclo or tricyclic ring, and at least one of the rings is non-aromatic or contains no heteroatoms, it is generally considered to be linked via an aromatic ring or a heteroatom-containing ring, respectively.
"heterocycle" (and derivatives thereof such as "heterocyclic" or "heterocyclyl") refers broadly to a single cyclic aliphatic hydrocarbon, typically having from 3 to 12 ring atoms, containing at least 2 carbon atoms, and further containing from 1 to 3 heteroatoms independently selected from oxygen, sulfur, and nitrogen, and also to combinations comprising at least one of the foregoing heteroatoms. Alternatively, the heterocyclic ring as defined above may be a polycyclic ring system (e.g. bicyclic) wherein two or more rings are present in the form of a parallel or bridged ring or spiro ring wherein at least one ring contains one or more heteroatoms independently selected from oxygen, sulfur, nitrogen. "heterocycle" also refers to a 5-to 7-membered heterocyclic ring fused to 5-and 6-membered aromatic carbocyclic rings containing one or more heteroatoms selected from nitrogen, oxygen, and sulfur, provided that the site of attachment is on the heterocyclic ring. Heterocycles may be saturated or contain one to more double bonds (i.e., partially unsaturated). The heterocyclic ring may be substituted with oxo (oxo). Either the carbon atom or the heteroatom of the heterocycle may be the attachment site, provided that a stable structure is formed. When a substituent is present on the heterocycle, the substituent may be attached to any heteroatom or carbon atom on the heterocycle, provided that a stable chemical structure is formed. The heterocyclic and heteroaryl definitions described herein do not overlap.
Suitable heterocycles include, for example (attachment site is preferably 1) 1-pyrrolidinyl, 2,4- (imidazolinyl), 2, 3-pyrazolidinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl and 2, 5-piperazinyl, 1, 4-piperazinyl, 2, 3-pyridazinyl. Also contemplated are morpholinyl groups, including 2-morpholinyl and 3-morpholinyl (oxygen atom position numbering is preferably 1). Substituted heterocycles also include ring systems substituted with one or more oxo groups, such as piperidinyl-N-oxide, morpholinyl-N-oxide, 1-oxo-1-thiomorpholinyl, and 1, 1-dioxo-1-thiomorpholinyl. The bis-heterocyclic compounds include, for example:
Figure BDA0001755256780000111
as used herein, "aryl-alkyl" refers to an aryl-substituted alkyl group. Exemplary aralkyl groups include benzyl, phenethyl, naphthylmethyl, and the like. In some embodiments, aralkyl groups contain 7 to 20 or 7 to 11 carbon atoms. When using "aryl radicals C1-4Alkyl "in which" C1-4"refers to the number of carbon atoms in the alkyl portion rather than the aryl portion.
As used herein, "heterocyclyl-alkyl" refers to a heterocyclyl-substituted alkyl group. When using "heterocyclyl-C1-4Alkyl "in which" C1-4"refers to the number of carbon atoms in the alkyl moiety rather than in the heterocyclyl moiety.
As used herein, "cycloalkyl-alkyl" refers to a cycloalkyl-substituted alkyl group. When using "C3-10cycloalkyl-C1-4Alkyl "in which" C3-10"refers to the number of carbon atoms in the cycloalkyl moiety rather than the alkyl moiety. Wherein "C1-4"refers to the number of carbon atoms in the alkyl moiety rather than the cycloalkyl moiety.
As used herein, "heteroaryl-alkyl" refers to heteroaryl-substituted alkyl. When using "heteroaryl-C1-4Alkyl "in which" C1-4"refers to the number of carbon atoms in the alkyl moiety rather than in the heteroaryl moiety.
To avoid ambiguity, for example: when alkyl, cycloalkyl, heterocyclyl, aryl and/or heteroaryl substituents thereof are mentioned, it is meant that each of these groups is substituted individually or that these groups are mixed. That is: if R is1Is aryl-C1-4The alkyl, aryl moieties may be unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from RxMay also be unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RxA substituent of (1).
"pharmaceutically acceptable salt" refers to salts with pharmaceutically acceptable non-toxic bases or acids, including inorganic or organic bases and inorganic or organic acids. The salt of an inorganic base may be selected, for example, from: aluminum, ammonium, calcium, copper, iron, ferrous, lithium, magnesium, manganese, manganous, potassium, sodium, and zinc salts. Further, the salt of a pharmaceutically acceptable inorganic base may be selected from ammonium, calcium, magnesium, potassium and sodium salts. One or more crystal structures may be present in the solid salt, as well as in the form of hydrates. The pharmaceutically acceptable salts of organic non-toxic bases may be selected, for example, from: primary, secondary and tertiary amine salts, the substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins such as arginine, betaine, caffeine, choline, N-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine and tripropylamine, tromethamine.
When the compound referred to in this patent is a base, it is necessary to prepare a salt thereof with at least one pharmaceutically acceptable non-toxic acid selected from inorganic and organic acids. For example, selected from the group consisting of acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, fumaric acid, gluconic acid, glutamic acid, hydrobromic acid, hydrochloric acid, isethionic acid, lactic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, mucic acid, nitric acid, pamoic acid, pantothenic acid, phosphoric acid, succinic acid, sulfuric acid, tartaric acid, and p-toluenesulfonic acid. In some embodiments, these acids may be selected, for example: citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, fumaric and tartaric acids.
By "administering" or "administration" of a compound or a pharmaceutically acceptable salt thereof is meant providing a compound of the invention or a pharmaceutically acceptable salt thereof to a subject in need of treatment.
An "effective amount" is an amount of a compound or a pharmaceutically acceptable salt thereof that is capable of eliciting a biological or medical response in a tissue, system, animal or human that is observable by a researcher, veterinarian, clinician or other clinician.
The "composition" includes: the invention may take the form of a kit, article of manufacture, or any combination thereof. The pharmaceutical composition comprises: products comprising the active ingredient and an inert ingredient as a carrier, as well as products produced by any two or more of the ingredients, directly or indirectly, by combination, complexation or aggregation, or by dissociation of one or more of the ingredients, or by other types of reactions or interactions of one or more of the ingredients.
By "pharmaceutically acceptable" is meant compatible with the other ingredients of the formulation and not unacceptably toxic to the subject.
"subject" refers to a subject having a disease, disorder, or the like, and includes mammals and non-mammals. Mammals include, but are not limited to, any member of the mammalian family: humans, non-human primates such as chimpanzees, and other apes and monkeys; farm animals such as cattle, horses, sheep's yang, goats, pigs; domestic animals such as rabbits, dogs, and cats; the experimental animals include rodents such as rats, mice, guinea pigs, and the like. Non-mammalian animals include, but are not limited to, birds, fish, and the like. In one embodiment of the invention, the mammal is a human.
"treating" includes alleviating, alleviating or ameliorating a disease or condition, preventing other conditions, ameliorating or preventing a metabolic factor underlying a condition, inhibiting a disease or condition, e.g., arresting the development of a disease or condition, alleviating a disease or condition, promoting remission of a disease or condition, or arresting the signs of a disease or condition, and extends to include prevention. "treating" also includes achieving a therapeutic benefit and/or a prophylactic benefit. Therapeutic benefit refers to eradication or amelioration of the condition being treated. In addition, therapeutic benefit is achieved by eradicating or ameliorating one or more physiological signs associated with the underlying disease, and amelioration of the disease in the patient is observed, although the patient may still be suffering from the underlying disease. Prophylactic benefit refers to the use of a composition by a patient to prevent the risk of a disease, or the use of a patient presenting with one or more physiological conditions of a disease, although the disease has not yet been diagnosed.
"protecting group" (Pg) refers to a class of substituents used to block or protect a particular functional group by reacting with other functional groups on a compound. For example, "amino protecting group" refers to an amino group attached to a compound that blocks or protects the compoundA substituent of a radical functional group. Suitable amino protecting groups include, but are not limited to, acetyl, trifluoroacetyl, t-Butyloxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and-9-fluorenylmethyloxycarbonyl (Fmoc). Similarly, "hydroxy protecting group" refers to a class of hydroxy substituents that are effective in blocking or protecting the hydroxy function. Suitable protecting groups include, but are not limited to, acetyl and silyl groups. "carboxy protecting group" refers to a class of carboxy substituents that function effectively to block or protect a carboxy group. Common carboxyl protecting groups include-CH2CH2SO2Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrobenzenesulfonyl) ethyl, 2- (diphenylphosphino) -ethyl, nitroethyl and the like. For general description and instructions for use of protecting groups, see references: greene, Protective Groups in Organic Synthesis, John Wiley&Sons,New York,1991。
"NH protecting group" includes, but is not limited to, trichloroethoxycarbonyl, tribromoethoxycarbonyl, benzyloxycarbonyl, p-nitrobenzoyl, o-bromobenzyloxycarbonyl, chloroacetyl, dichloroacetyl, trichloroacetyl, trifluoroacetyl, phenylacetyl, formyl, acetyl, benzoyl, t-pentyloxycarbonyl, t-butyloxycarbonyl, p-methoxybenzyloxycarbonyl, 3, 4-dimethoxybenzyloxycarbonyl, 4- (phenylazo) benzyloxycarbonyl, 2-furfuryloxycarbonyl, diphenylmethoxycarbonyl, 1-dimethylpropoxycarbonyl, isopropyloxycarbonyl, phthaloyl, succinyl, alanyl, leucyl, 1-adamantyloxycarbonyl, 8-quinolinyloxycarbonyl, benzyl, benzhydryl, trityl, 2-nitrophenylthio, methanesulfonyl, p-toluenesulfonyl, N, n-dimethylaminomethylene, benzylidene, 2-hydroxybenzylidene, 2-hydroxy-5-chlorobenzylidene, 2-hydroxy-l-naphthylmethylene, 3-hydroxy-4-pyridylmethylene, cyclohexylidene, 2-ethoxycarbonylcyclohexylidene, 2-ethoxycarbonylcyclopentylidene, 2-acetylcyclohexylidene, 3-dimethyl-5-oxocyclohexylidene, diphenylphosphoryl, dibenzylphosphono, 5-methyl-2-oxo-2H-l, 3-dioxy-4-ylmethyl, trimethylsilyl, triethylsilyl and triphenylsilyl.
"C (O) OH" protecting groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, 1-dimethylpropyl, n-butyl, t-butyl, phenyl, naphthyl, benzyl, benzhydryl, trityl, p-nitrobenzyl, p-methoxybenzyl, bis (p-methoxyphenyl) methyl, acetylmethyl, phenacyl, p-nitrobenzoylmethyl, p-bromobenzoylmethyl, p-methanesulfonylbenzoylmethyl, 2-tetrahydropyranyl, 2-tetrahydrofuranyl, 2,2, 2-trichloroethyl, 2- (trimethylsilyl) ethyl, acetoxymethyl, propionyloxymethyl, pivaloyloxymethyl, phthalimidomethyl, succinimidylmethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methoxymethyl, methoxyethoxymethyl, 2- (trimethylsilyl) ethoxymethyl, Benzyloxymethyl, methylthiomethyl, 2-methylthioethyl, phenylthiomethyl, 1-dimethyl-2-propenyl, 3-methyl-3-butenyl, allyl, trimethylsilyl, triethylsilyl, triisopropylsilyl, diethylisopropylsilyl, tert-butyldimethylsilyl, tert-butyldiphenylsilyl, diphenylmethylsilyl and tert-butylmethoxyphenylsilyl.
"OH or SH" protecting groups include, but are not limited to, benzyloxycarbonyl, 4-nitrobenzyloxycarbonyl, 4-bromobenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 3, 4-dimethoxybenzyloxycarbonyl, methoxycarbonyl, ethoxycarbonyl, t-butoxycarbonyl, 1-dimethylpropoxycarbonyl, isopropyloxycarbonyl, isobutyloxycarbonyl, diphenylmethoxycarbonyl, 2,2, 2-trichloroethoxycarbonyl, 2,2, 2-tribromoethoxycarbonyl, 2- (trimethylsilane) ethoxycarbonyl, 2- (phenylsulfonyl) ethoxycarbonyl, 2- (triphenylphosphonio) ethoxycarbonyl, 2-furfuryloxycarbonyl, 1-adamantyloxycarbonyl, vinyloxycarbonyl, allyloxycarbonyl, 4-ethoxy-1-naphthyloxycarbonyl, N-phenyloxycarbonyl, N-O-carbonyl, N-phenyloxycarbonyl, N-O-carbonyl, N-O-C, O-C-O, 8-quinolyloxycarbonyl, acetyl, formyl, chloroacetyl, dichloroacetyl, trichloroacetyl, trifluoroacetyl, methoxyacetyl, phenoxyacetyl, pivaloyl, benzoyl, methyl, tert-butyl, 2,2, 2-trichloroethyl, 2-trimethylsilylethyl, 1-dimethyl-2-propenyl, 3-methyl-3-butenyl, allyl, benzyl (phenylmethyl), p-methoxybenzyl, 3, 4-dimethoxybenzyl, diphenylmethyl, triphenylmethyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiopyranyl, methoxymethyl, methylthiomethyl, benzyloxymethyl, 2-methoxyethoxymethyl, 2,2, 2-trichloro-ethoxymethyl, 2- (trimethylsilyl) ethoxymethyl, trichloroacetyl, trifluoroacetyl, methoxyacetyl, phenoxyacetyl, pivaloyl, benzoyl, methyl, tert-butyl, 2, 2-trichloroethyl, 2-trimethylsilylethyl, 1-dimethyl-2-propenyl, 3-methyl, benzyl (phenylmethyl), p-methoxybenzyl, 3, 4-dimethoxybenzyl, diphenylmethyl, triphenylmethyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiopyranyl, methoxymethyl, 2- (trimethylsilyl) ethoxymethyl, 2-ethoxymethyl, 2, 2-methyl, 2-ethoxymethyl, 2-methyl, and a, 1-ethoxyethyl group, methanesulfonyl group, p-toluenesulfonyl group, trimethylsilyl group, triethylsilyl group, triisopropylsilyl group, diethylisopropylsilyl group, tert-butyldimethylsilyl group, tert-butyldiphenylsilyl group, diphenylmethylsilyl group and tert-butylmethoxyphenylsilyl group.
Geometric isomers may exist in the compounds of the present invention. Compounds of the present invention may have carbon-carbon double bonds or carbon-nitrogen double bonds in either the E or Z configuration, where "E" represents the preferred substituent on the opposite side of the carbon-carbon double bond or carbon-nitrogen double bond and "Z" represents the preferred substituent on the same side of the carbon-carbon double bond or carbon-nitrogen double bond, as defined by Cahn-Ingold-Prelog preference. The compounds of the invention may also exist as mixtures of "E" and "Z" isomers. The substituents around the cycloalkyl or heterocyclyl group may be in either the cis or trans configuration. In addition, the present invention includes different isomers and mixtures thereof formed by different arrangements of substituents around the adamantane ring system. Two substituents around a single ring in an adamantane ring system are designated in either the Z or E relative configuration. See, for example, C.D.Jones, M.Kaselj, R.N.Salvatore, W.J.le Noble J.org.chem.1998,63, 2758-.
Compounds of the invention may contain asymmetrically substituted carbon atoms of R or S configuration, "R" and "S" are defined in IUPAC 1974 Recommendations for Section E, functional Stereochemistry, Pure appl. chem. (1976)45, 13-10. Compounds containing asymmetrically substituted carbon atoms are racemates if the amounts of R and S configuration are the same. If one of the configurations is present in a greater amount than the other configuration, the configuration of the chiral carbon atom is represented by the more abundant configuration, preferably with an enantiomeric excess of about 85-90%, more preferably about 95-99%, and even more preferably 99% or more. Thus, the present invention encompasses racemic mixtures, relative and absolute stereoisomers, and mixtures of relative and absolute stereoisomers.
Isotopically enriched or labelled compounds
The compounds of the invention may exist in isotopically-labelled or enriched forms containing one or more atoms of different mass and mass numbers from the atom mass and mass number most prevalent in nature. The isotope may be a radioactive or non-radioactive isotope. Isotopes of atoms such as hydrogen, carbon, nitrogen, phosphorus, sulfur, fluorine, chlorine, and iodine include, but are not limited to,2H、3H、13C、14C、15N、18O、32P、35S、18F、36cl and125I. other isotopes and/or other atoms containing these atoms are also within the scope of the present invention.
In another embodiment, the isotopically labeled compound comprises deuterium (A), (B), (C) and D) and (C)2H) Tritium (a)3H) Or14Isotope of C. Isotopically-labeled compounds of the present invention can be obtained by employing procedures well known to those skilled in the art. These isotopically labeled compounds can be obtained by substituting a non-labeling reagent with an isotopically labeled reagent by referring to the examples and reaction schemes of the present invention. In certain examples, compounds can be treated with isotopic labeling agents to replace atoms with isotopic atoms, e.g., replacement of hydrogen with deuterium can be accomplished by deuterated acids such as D2SO4/D2And exchanging the action of O. In addition, relevant synthetic steps and intermediates can be found, for example, in Lizondo, J et al, Drugs Fut,21(11),1116 (1996); brickner, S J et al, J Med Chem,39(3),673 (1996); mallesham, B et al, Org Lett,5(7),963 (2003); PCT publication nos. WO1997010223, WO2005099353, WO1995007271, WO 2006008754; U.S. patent nos. 7538189, 7534814, 7531685, 7528131, 7521421, 7514068, 7511013; and U.S. patent application publication nos. 20090137457, 20090131485, 20090131363, 20090118238, 20090111840, 20090105338, 20090105307, 20090105147, 20090093422, 20090088416, and 20090082471, for specific methods see references.
Isotopically-labelled compounds of the present invention are useful as standards for binding assays for determining the effectiveness of CDK4/6 inhibitors. Isotopically-containing compounds are useful in pharmaceutical research, evaluating the mechanism of action and metabolic pathways of non-isotopically-labeled parent compounds, and investigating the in vivo metabolic turnover of compounds (Blake et al.J.pharm.Sci.64,3,367-391 (1975)). Such metabolic studies are important for the design of safe and effective therapeutic agents, and can be judged to be toxic or carcinogenic to the in vivo active compound administered to the patient or to the metabolite of the parent compound (Foster et al, Advances in Drug Research Vol.14, pp.2-36, Academic press, London, 1985; Kato et al, J.Labelled Comp.Radiopharmaceut.,36(10):927-932 (1995); Kushner et al, Can.J.Physiol.Pharmacol,77,79-88 (1999)).
In addition, drugs containing non-radioactive isotopes, such as deuterated drugs, known as "heavy drugs," are useful for treating diseases and disorders associated with CDK4/6 activity. The proportion of a certain isotope in a compound that exceeds its natural abundance is called enrichment. The amount of enrichment is, for example, from about 0.5, 1,2,3,4, 5,6,7,8, 9, 10, 12, 16, 21, 25, 29, 33, 37, 42, 46, 50, 54, 58, 63, 67, 71, 75, 79, 84, 88, 92, 96 to 100 mol%. In mammals, the replacement of 15% of the common atoms with heavy isotopes is effective and can last from days to weeks, including rodents and dogs, with fewer adverse effects (zajka D M and Finkel a J, ann.n.y.acad.sci.196084: 770; Thomson J F, ann.new York acad.sci 196084: 736; Czakja D M et al, am.j.physiol.1961201: 357). Replacing up to 15-23% of the body fluids in humans with deuteration does not cause toxicity (Blagojevic N et al in "Dosimetry & Treatment Planning for Neutron Capture Therapy", Zamenhof R, Solares G and Harling O eds.1994.advanced Medical Publishing, Madison Wis. pp. 125-134; Diabetes Metab.23:251(1997))
Suitable stable isotopic labels for drugs can alter the physicochemical properties of the drug, such as pKa and liquid solubility. If isotopic substitution affects the region associated with ligand-receptor interaction, then these effects and changes may affect the pharmacodynamic response of the drug molecule. Certain physical properties of stable isotope-labeled molecules differ from unlabeled molecules, while chemical and biological properties are the same, but only one important difference: any chemical bond containing a heavy isotope and another atom is stronger than a light isotope due to the increased mass of the heavy isotope. Accordingly, the presence of isotopes at the metabolic or enzymatic conversion sites slows the reaction and may alter its pharmacokinetic or pharmacodynamic properties compared to non-isotopically labeled compounds.
In embodiment (1), the present invention provides a compound represented by formula (I) or (II):
Figure BDA0001755256780000161
or a pharmaceutically acceptable salt thereof, wherein,
q is selected from aryl and heteroaryl;
each R1Independently selected from hydrogen, halogen, hydroxy, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylamino, di (alkyl) amino, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from RxSubstituted with the substituent(s);
each R2Independently selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA1RB1、-ORA1、-S(O)rRA1、-S(O)2ORA1、-OS(O)2RA1、-P(O)RA1RB1、-P(O)(ORA1)(ORB1)、-C(O)RA1、-C(O)ORA1、-OC(O)RA1、-C(O)NRA1RB1、-NRA1C(O)RB1、-OC(O)NRA1RB1、-NRA1C(O)ORB1、-NRA1C(O)NRA1RB1、-NRA1C(S)NRA1RB1、-S(O)rNRA1RB1、-NRA1S(O)rRB1、-NRA1S(O)2NRA1RB1、-S(O)(=NRE1)RB1、-N=S(O)RA1RB1、-NRA1S(O)(=NRE1)RB1、-S(O)(=NRE1)NRA1RB1、-NRA1S(O)(=NRE1)NRA1RB1、-C(=NRE1)RA1、-C(=N-ORB1)RA1、-C(=NRE1)NRA1RB1、-NRA1C(=NRE1)RB1and-NRA1C(=NRE1)NRA1RB1Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RxSubstituted with the substituent(s);
each R3Independently selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA2RB2、-ORA2、-S(O)rRA2、-S(O)2ORA2、-OS(O)2RA2、-P(O)RA2RB2、-P(O)(ORA2)(ORB2)、-C(O)RA2、-C(O)ORA2、-OC(O)RA2、-C(O)NRA2RB2、-NRA2C(O)RB2、-OC(O)NRA2RB2、-NRA2C(O)ORB2、-NRA2C(O)NRA2RB2、-NRA2C(S)NRA2RB2、-S(O)rNRA2RB2、-NRA2S(O)rRB2、-NRA2S(O)2NRA2RB2、-S(O)(=NRE2)RB2、-N=S(O)RA2RB2、-NRA2S(O)(=NRE2)RB2、-S(O)(=NRE2)NRA2RB2、-NRA2S(O)(=NRE2)NRA2RB2、-C(=NRE2)RA2、-C(=N-ORB2)RA2、-C(=NRE2)NRA2RB2、-NRA2C(=NRE2)RB2and-NRA2C(=NRE2)NRA2RB2Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXSubstituted with the substituent(s);
each R4Selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA3RB3、-ORA3、-S(O)rRA3、-S(O)2ORA3、-OS(O)2RA3、-P(O)RA3RB3、-P(O)(ORA3)(ORB3)、-C(O)RA3、-C(O)ORA3、-OC(O)RA3、-C(O)NRA3RB3、-NRA3C(O)RB3、-OC(O)NRA3RB3、-NRA3C(O)ORB3、-NRA3C(O)NRA3RB3、-NRA3C(S)NRA3RB3、-S(O)rNRA3RB3、-NRA3S(O)rRB3、-NRA3S(O)2NRA3RB3、-S(O)(=NRE3)RB3、-N=S(O)RA3RB3、-NRA3S(O)(=NRE3)RB3、-S(O)(=NRE3)NRA3RB3、-NRA3S(O)(=NRE3)NRA3RB3、-C(=NRE3)RA3、-C(=N-ORB3)RA3、-C(=NRE3)NRA3RB3、-NRA3C(=NRE3)RB3and-NRA3C(=NRE3)NRA3RB3Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXSubstituted with the substituent(s);
each R5Independently selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, CN, NO2、-NRA4RB4、-(CH2)tNRA4RB4、-ORA4、-S(O)rRA4、-S(O)2ORA4、-OS(O)2RA4、-P(O)RA4RB4、-P(O)(ORA4)(ORB4)、-C(O)RA4、-C(O)ORA4、-OC(O)RA4、-C(O)NRA4RB4、-NRA4C(O)RB4、-OC(O)NRA4RB4、-NRA4C(O)ORB4、-NRA4C(O)NRA4RB4、-NRA4C(S)NRA4RB4、-S(O)rNRA4RB4、-NRA4S(O)rRB4、-NRA4S(O)2NRA4RB4、-S(O)(=NRE4)RB4、-N=S(O)RA4RB4、-NRA4S(O)(=NRE4)RB4、-S(O)(=NRE4)NRA4RB4、-NRA4S(O)(=NRE4)NRA4RB4、-C(=NRE4)RA4、-C(=N-ORB4)RA4、-C(=NRE4)NRA4RB4、-NRA4C(=NRE4)RB4and-NRA4C(=NRE4)NRA4RB4Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXSubstituted with the substituent(s);
each RA1、RA2、RA3、RA4、RB1、RB2、RB3And RB4Independently selected from hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from RXSubstituted with the substituent(s);
or each "RA1And RB1”、“RA2And RB2”、“RA3And RB3"or" RA4And RB4Taken together with the atom or atoms to which they are attached, form a 4-12 membered heterocyclic ring containing 0,1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be interrupted by 1,2 or 3RXSubstituted by groups;
each RE1、RE2、RE3And RE4Independently selected from hydrogen, C1-10Alkyl, CN, NO2、-ORa1、-SRa1、-S(O)rRa1、-C(O)Ra1、-C(O)ORa1、-C(O)NRa1Rb1and-S (O)rNRa1Rb1
Each RXIndependently selected from C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, halogen, CN, NO2、-(CRc1Rd1)tNRa1Rb1、-(CRc1Rd1)tORb1、-(CRc1Rd1)tS(O)rRb1、-(CRc1Rd1)tS(O)2ORb1、-(CRc1Rd1)tOS(O)2Rb1、-(CRc1Rd1)tP(O)Ra1Rb1、-(CRc1Rd1)tP(O)(ORa1)(ORb1)、-(CRc1Rd1)tC(O)Ra1、-(CRc1Rd1)tC(O)ORb1、-(CRc1Rd1)tOC(O)Rb1、-(CRc1Rd1)tC(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(O)Rb1、-(CRc1Rd1)tOC(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(O)ORb1、-(CRc1Rd1)tNRa1C(O)NRa1Rb1、-(CRc1Rd1)tNRa1C(S)NRa1Rb1、-(CRc1Rd1)tS(O)rNRa1Rb1、-(CRc1Rd1)tNRa1S(O)rRb1、-(CRc1Rd1)tNRa1S(O)2NRa1Rb1、-(CRc1Rd1)tS(O)(=NRe1)Rb1、-(CRc1Rd1)tN=S(O)Ra1Rb1、-(CRc1Rd1)tNRa1S(O)(=NRe1)Rb1、-(CRc1Rd1)tS(O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1S(O)(=NRe1)NRa1Rb1、-(CRc1Rd1)tC(=NRe1)Ra1、-(CRc1Rd1)tC(=N-ORb1)Ra1、-(CRc1Rd1)tC(=NRe1)NRa1Rb1、-(CRc1Rd1)tNRa1C(=NRe1)Rb1And- (CR)c1Rd1)tNRa1C(=NRe1)NRa1Rb1Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RYSubstituted with the substituent(s);
each Ra1And Rb1Independently selected from hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from RYSubstituted with the substituent(s);
or Ra1And Rb1Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0,1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be substituted with 1,2 or 3 heteroatoms selected from RYSubstituent group(s) takeGeneration;
each Rc1And Rd1Independently selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from RYSubstituted with the substituent(s);
or Rc1And Rd1Together with the carbon atom or atoms to which they are attached form a 3-12 membered ring containing 0,1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may optionally be interrupted by 1,2 or 3RYSubstituted by groups;
each Re1Independently selected from hydrogen, C1-10Alkyl radical, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, CN, NO2、-ORa2、-SRa2、-S(O)rRa2、-C(O)Ra2、-C(O)ORa2、-S(O)rNRa2Rb2and-C (O) NRa2Rb2
Each RYIndependently selected from C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl-C1-4Alkyl, halogen, CN, NO2、-(CRc2Rd2)tNRa2Rb2、-(CRc2Rd2)tORb2、-(CRc2Rd2)tS(O)rRb2、-(CRc2Rd2)tS(O)2ORb2、-(CRc2Rd2)tOS(O)2Rb2、-(CRc2Rd2)tP(O)Ra2Rb2、-(CRc2Rd2)tP(O)(ORa2)(ORb2)、-(CRc2Rd2)tC(O)Ra2、-(CRc2Rd2)tC(O)ORb2、-(CRc2Rd2)tOC(O)Rb2、-(CRc2Rd2)tC(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(O)Rb2、-(CRc2Rd2)tOC(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(O)ORb2、-(CRc2Rd2)tNRa2C(O)NRa2Rb2、-(CRc2Rd2)tNRa2C(S)NRa2Rb2、-(CRc2Rd2)tS(O)rNRa2Rb2、-(CRc2Rd2)tNRa2S(O)rRb2、-(CRc2Rd2)tNRa2S(O)2NRa2Rb2、-(CRc2Rd2)tS(O)(=NRe2)Rb2、-(CRc2Rd2)tN=S(O)Ra2Rb2、-(CRc2Rd2)tNRa2S(O)(=NRe2)Rb2、-(CRc2Rd2)tS(O)(=NRe2)NRa2Rb2、-(CRc2Rd2)tNRa2S(O)(=NRe2)NRa2Rb2、-(CRc2Rd2)tC(=NRe2)Ra2,-(CRc2Rd2)tC(=N-ORb2)Ra2、-(CRc2Rd2)tC(=NRe2)NRa2Rb2、-(CRc2Rd2)tNRa2C(=NRe2)Rb2And- (CR)c2Rd2)tNRa2C(=NRe2)NRa2Rb2Wherein each alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from OH, CN, amino, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Ra2And Rb2Independently selected from hydrogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylamino, heterocyclyl, aryl and heteroaryl is independently unsubstituted or substituted with at least one, such as 1,2,3 or 4, independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
or Ra2And Rb2Together with the atom or atoms to which they are attached form a 4-12 membered heterocyclic ring containing 0,1 or 2 additional heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorus, which ring may optionally be substituted with 1 or 2 heteroatoms independently selected from oxygen, sulfur, nitrogen and phosphorusHalogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Rc2And Rd2Independently selected from hydrogen, halogen, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio radical, C1-10Alkylamino radical, C3-10Cycloalkylamino, di (C)1-10Alkyl) amino, heterocyclyl-C1-4Alkyl, aryl-C1-4Alkyl, heteroaryl and heteroaryl-C1-4Alkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, cycloalkoxy, alkylthio, cycloalkylthio, alkylamino, cycloalkylamino, heterocyclyl, aryl and heteroaryl is unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
or Rc2And Rd2Together with the carbon atom or atoms to which they are attached, form a 3-12 membered ring containing 0,1 or 2 heteroatoms independently selected from oxygen, sulfur and nitrogen, which ring may optionally be substituted with 1 or 2 heteroatoms independently selected from halogen, CN, C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Cycloalkyl, hydroxy, C1-10Alkoxy radical, C3-10Cycloalkoxy, C1-10Alkylthio radical, C3-10Cycloalkylthio, amino, C1-10Alkylamino radical, C3-10Cycloalkylamino and di (C)1-10Alkyl) amino;
each Re2Independently selected from hydrogen, CN, NO2、C1-10Alkyl radical, C3-10Cycloalkyl radical, C3-10cycloalkyl-C1-4Alkyl radical, C1-10Alkoxy radical, C3-10Cycloalkoxy, -C (O) C1-4Alkyl, -C (O) C3-10Cycloalkyl, -C (O) OC1-4Alkyl, -C (O) OC3-10Cycloalkyl, -C (O) N (C)1-4Alkyl radical)2、-C(O)N(C3-10Cycloalkyl radicals2、-S(O)2C1-4Alkyl, -S (O)2C3-10Cycloalkyl, -S (O)2N(C1-4Alkyl radical)2and-S (O)2N(C3-10Cycloalkyl radicals2
m is selected from 0,1, 2,3 and 4;
n is selected from 0,1 and 2;
p is selected from 0,1 and 2;
each r is independently selected from 0,1 and 2;
each t is independently selected from 0,1, 2,3, and 4.
In another embodiment (2), the present invention provides a compound of embodiment (1), or a pharmaceutically acceptable salt thereof, having the formula
Figure BDA0001755256780000211
In another embodiment (3), the present invention provides a compound of embodiment (1), or a pharmaceutically acceptable salt thereof, having the formula
Figure BDA0001755256780000212
In another embodiment (4), the present invention provides a compound of any one of embodiments (1) - (3), or a pharmaceutically acceptable salt thereof, wherein Q is heteroaryl.
In another embodiment (5), the present invention provides a compound of embodiment (4) or a medicament thereofA pharmaceutically acceptable salt wherein Q is independently selected from
Figure BDA0001755256780000213
In another embodiment (6), the present invention provides a compound of any one of embodiments (1) to (5), or a pharmaceutically acceptable salt thereof, wherein each R is5Is independently selected from C1-10Alkyl, heterocyclyl-C1-4Alkyl, -S (O)rRA4、-C(O)RA4and-CH2NRA4RB4Wherein alkyl and heterocyclyl are unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXIs substituted with the substituent(s).
In another embodiment (7), the present invention provides a compound of embodiment (6), or a pharmaceutically acceptable salt thereof, wherein each R is5Independently selected from methyl, ethyl,
Figure BDA0001755256780000214
Figure BDA0001755256780000215
Which is unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXIs substituted with the substituent(s).
In another embodiment (8), the present invention provides a compound of embodiment (6), or a pharmaceutically acceptable salt thereof, wherein each R is5Is independently selected from S (O)2RA4and-C (O) RA4Wherein each R isA4Independently selected from heterocyclyl and heterocyclyl-C1-4Alkyl, wherein each alkyl and heterocyclyl is unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXAnd each R is substitutedB4Is independently selected from C1-10An alkyl group.
In another embodiment (9), the present invention provides a compound of embodiment (8), or a pharmaceutically acceptable salt thereof, wherein each R is5Is independently selected from S (O)2RA4and-C (O) RA4Wherein each R is4Is independently selected from
Figure BDA0001755256780000221
Figure BDA0001755256780000222
Which is unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXAnd each R is substitutedB4Is methyl.
In another embodiment (10), the present invention provides a compound of embodiment (6), or a pharmaceutically acceptable salt thereof, wherein each R is5Is independently selected from-CH2NRA4RB4Wherein each R isA4Is composed of
Figure BDA0001755256780000223
Which is unsubstituted or substituted by at least one, such as 1,2,3 or 4, independently selected from RXAnd each R is substitutedB4Is methyl.
In another embodiment (11), the present invention provides a compound of any one of embodiments (6) to (10), or a pharmaceutically acceptable salt thereof, wherein each R isXIndependently selected from methyl, ethyl, methoxymethyl, cyanoethyl and oxetan-3-yl.
In another embodiment (12), the present invention provides a compound of any one of embodiments (1) - (11), or a pharmaceutically acceptable salt thereof, wherein m is 1.
In another embodiment (13), the present invention provides a compound of any one of embodiments (1) - (12), or a pharmaceutically acceptable salt thereof, wherein R is1Is hydrogen.
In another embodiment (14), the present invention provides a compound of any one of embodiments (1) - (13), or a pharmaceutically acceptable salt thereof, wherein R2Is hydrogen.
In another embodiment (15), the present invention provides a compound of any one of embodiments (1) - (14), or a pharmaceutically acceptable salt thereof, wherein R3Is independently selected from C1-10An alkyl group.
In another embodiment (16), the present invention providesA compound of embodiment (15) or a pharmaceutically acceptable salt thereof, wherein R3Independently selected from methyl, isopropyl and tert-butyl.
In another embodiment (17), the present invention provides a compound of any one of embodiments (15) to (16), or a pharmaceutically acceptable salt thereof, wherein p is 2.
In another embodiment (18), the present invention provides a compound of any one of embodiments (1) - (17), or a pharmaceutically acceptable salt thereof, wherein R4Is a halogen.
In another embodiment (19), the present invention provides a compound of embodiment (18), or a pharmaceutically acceptable salt thereof, wherein each R is4Independently selected from fluorine and chlorine.
In another embodiment (20), the compounds provided herein are selected from
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- (piperazin-1-ylmethyl) pyridin-2-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
(S) -5-fluoro-N- (5- ((hexahydropyrrolo [1,2-a ] pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
(R) -5-fluoro-N- (5- ((hexahydropyrrolo [1,2-a ] pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
N- (5- (((1S,4S) -2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- (((1S,4S) -5-methyl-2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- (((1S,4S) -5-ethyl-2, 5-diazabicyclo [2.2.1] heptan-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
N- (5- ((3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((3-methyl-3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((3-ethyl-3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
N- (5- ((3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((8-methyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((8-ethyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-N- (5- ((hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) methyl) pyridin-2-yl) -4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((5-methylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((5-ethylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
N- (5- ((2, 7-diazaspiro [3.5] nonan-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((7-methyl-2, 7-diazaspiro [3.5] nonan-2-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((7-ethyl-2, 7-diazaspiro [3.5] nonan-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
N- (5- ((2, 6-diazaspiro [3.3] heptan-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((6-methyl-2, 6-diazaspiro [3.3] heptan-2-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((6-ethyl-2, 6-diazaspiro [3.3] heptan-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
N- (5- ((3, 9-diazaspiro [5.5] undecan-3-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((9-methyl-3, 9-diazaspiro [5.5] undecan-3-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((9-ethyl-3, 9-diazaspiro [5.5] undecan-3-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
N- (5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) -5,6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
N- (5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) -6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
6-ethyl-N- (5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) -5,6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
(6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (piperazin-1-yl) methanone,
(6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (4-methylpiperazin-1-yl) methanone,
(4-ethylpiperazin-1-yl) (6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
(3, 8-diazabicyclo [3.2.1] octan-3-yl) (6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
(6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (8-methyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) methanone,
(8-ethyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) (6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
(6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (2, 7-azaspiro [3.5] nonan-2-yl) methanone,
(6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (7-methyl-2, 7-diazaspiro [3.5] nonan-2-yl) methanone,
(7-ethyl-2, 7-diazaspiro [3.5] nonan-2-yl) (6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
(6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) methanone,
(6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (5-methylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) methanone,
(5-Ethylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) (6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- (piperazin-1-ylsulfonyl) pyridin-2-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((4-methylpiperazin-1-yl) sulfonyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((4-ethylpiperazin-1-yl) sulfonyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
N- (5- ((3, 8-diazabicyclo [3.2.1] octan-3-yl) sulfonyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((8-methyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) sulfonyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((8-ethyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) sulfonyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-N- (5- ((hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) sulfonyl) pyridin-2-yl) -4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((5-methylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) sulfonyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((5-ethylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) sulfonyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
N- (5- ((2, 7-diazaspiro [3.5] nonan-2-yl) sulfonyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((7-methyl-2, 7-diazaspiro [3.5] nonan-2-yl) sulfonyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((7-ethyl-2, 7-diazaspiro [3.5] nonan-2-yl) sulfonyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(S) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((2-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(R) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((2-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(R) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((3-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(S) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((3-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- ((4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(S) -4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- ((2-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(R) -4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- ((2-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(R) -4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- ((3-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(S) -4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- ((3-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- (piperazin-1-ylmethyl) pyridin-2-yl) pyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
N- (5- ((3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) -4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoropyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- ((3-methyl-3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((3-ethyl-3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
(S) -4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- ((3- (methoxymethyl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(S) -4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- ((3- (methoxymethyl) -4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(S) -4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((4-ethyl-3- (methoxymethyl) piperazin-1-yl) methyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
N- (5- ((3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) -4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoropyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- ((8-methyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((8-ethyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- (1- (piperazin-1-yl) ethyl) pyridin-2-yl) pyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-fluoro-N- (5- (1- (4-methylpiperazin-1-yl) ethyl) pyridin-2-yl) pyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- (1- (4-ethylpiperazin-1-yl) ethyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-chloro-N- (5- (piperazin-1-ylmethyl) pyridin-2-yl) pyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-chloro-N- (5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -5-chloro-N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- (piperazin-1-ylmethyl) pyridin-2-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-1-methyl-1H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-1-methyl-1H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
N- (5- ((3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((3-methyl-3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((3-ethyl-3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
N- (5- ((3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((8-methyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((8-ethyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
(R) -5-fluoro-N- (5- ((hexahydropyrrolo [1,2-a ] pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
N- (5- ((2, 7-diazaspiro [3.5] nonan-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((7-methyl-2, 7-diazaspiro [3.5] nonan-2-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((7-ethyl-2, 7-diazaspiro [3.5] nonan-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-N- (5- ((hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) methyl) pyridin-2-yl) -4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((5-methylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((5-ethylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
N- (5- ((2, 7-diazaspiro [3.5] nonan-7-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((2-methyl-2, 7-diazaspiro [3.5] nonan-7-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((2-ethyl-2, 7-diazaspiro [3.5] nonan-7-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
N- (5- ((3, 9-diazaspiro [5.5] undecan-3-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((9-methyl-3, 9-diazaspiro [5.5] undecan-3-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((9-ethyl-3, 9-diazaspiro [5.5] undecan-3-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(S) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((2-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(R) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((2-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(R) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((3-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(S) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((3-methyl-4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (piperazin-1-yl) methanone,
(6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (4-methylpiperazin-1-yl) methanone,
(4-ethylpiperazin-1-yl) (6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
(3, 8-diazabicyclo [3.2.1] octan-8-yl) (6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
(6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (3-methyl-3, 8-diazabicyclo [3.2.1] octan-8-yl) methanone,
(3-ethyl-3, 8-diazabicyclo [3.2.1] octan-8-yl) (6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
(3, 8-diazabicyclo [3.2.1] octan-3-yl) (6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
(6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (8-methyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) methanone,
(8-ethyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) (6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
(6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (2, 7-diazaspiro [3.5] nonan-2-yl) methanone,
(6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (7-methyl-2, 7-diazaspiro [3.5] nonan-2-yl) methanone,
(7-ethyl-2, 7-diazaspiro [3.5] nonan-2-yl) (6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
(6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) methanone,
(6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) (5-methylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) methanone,
(5-Ethylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) (6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methanone,
N- (5- ((3, 8-diazabicyclo [3.2.1] octan-3-yl) sulfonyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((8-methyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) sulfonyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((8-ethyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) sulfonyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
N- (5- ((2, 7-diazaspiro [3.5] nonan-2-yl) sulfonyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((7-methyl-2, 7-diazaspiro [3.5] nonan-2-yl) sulfonyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((7-ethyl-2, 7-diazaspiro [3.5] nonan-2-yl) sulfonyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-N- (5- ((hexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) sulfonyl) pyridin-2-yl) -4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((5-methylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) sulfonyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((5-ethylhexahydropyrrolo [3,4-c ] pyrrol-2 (1H) -yl) sulfonyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- ((4- (oxetan-3-yl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
(R) -4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- ((methyl ((1-methylpyrol-2-yl) methyl) amino) methyl) pyridin-2-yl) pyrimidin-2-amine,
(R) -4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- ((hexahydropyrrolo [1,2-a ] pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
N- (5- ((3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) -4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- ((3-methyl-3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((3-ethyl-3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
(S) -4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- ((3- (methoxymethyl) piperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(S) -4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- ((3- (methoxymethyl) -4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
(S) -4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((4-ethyl-3- (methoxymethyl) piperazin-1-yl) methyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
N- (5- (((1R,5S) -3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) -4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- (((1R,5S) -8-methyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- (((1R,5S) -8-ethyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- (1- (piperazin-1-yl) ethyl) pyridin-2-yl) pyrimidin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- (1- (4-methylpiperazin-1-yl) ethyl) pyridin-2-yl) pyrimidin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- (1- (4-ethylpiperazin-1-yl) ethyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
N- (4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-yl) -5,6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
N- (4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-yl) -6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
N- (4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-yl) -6-ethyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- (morpholinomethyl) pyridin-2-yl) pyrimidin-2-amine,
3- (4- ((6- ((4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-yl) amino) pyridin-3-yl) methyl) piperazin-1-yl) propionitrile,
N- (4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-yl) -6- (piperidin-4-yl) -5,6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
N- (4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-yl) -6- (1-methylpiperidin-4-yl) -5,6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
N- (4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-yl) -6- (1-ethylpiperidin-4-yl) -5,6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
or a pharmaceutically acceptable salt thereof.
In another embodiment (21), the invention provides a pharmaceutical composition comprising a compound of any one of embodiments (1) - (20), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
In another embodiment (22), the invention provides a method of treating, ameliorating or preventing a disorder responsive to inhibition of cyclin dependent kinase 4/6, comprising administering to a subject in need thereof an effective amount of a compound of any one of embodiments (1) - (20), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, optionally in combination with a second therapeutic agent.
In another embodiment (23), the invention provides the use of a compound of any one of embodiments (1) to (20), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a cell proliferative disorder.
In another aspect, the invention provides a kit comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof; and instructions including one or more of the following: information on what state the composition is applied to, information stored on the composition, dosage information, and instructions on how to use the composition. In one particular variant, the kit comprises the compound in a multiple dose form.
In another aspect, the present invention provides an article of manufacture comprising a compound disclosed herein, or a pharmaceutically acceptable salt thereof; and a packaging material. In one variation, the packaging material includes a container containing the compound. In one particular variation, the container includes a label that identifies one or more of the following: instructions for what disease the compound applies to, stored information, dosage information, and/or how to use the compound. In another variation, the article of manufacture comprises the compound in a multiple dose form.
In another aspect, the invention provides a method of treatment comprising administering to a subject a compound disclosed herein, or a pharmaceutically acceptable salt thereof.
In another aspect, the invention provides a method of inhibiting CDK4/6 kinase comprising contacting a compound disclosed herein, or a pharmaceutically acceptable salt thereof, with CDK 4/6.
In another aspect, the invention provides a method of inhibiting CDK4/6, comprising causing a compound disclosed herein, or a pharmaceutically acceptable salt thereof, to be present in a subject to inhibit CDK4/6 activity in vivo.
In another aspect, the invention provides a method of inhibiting CDK4/6 comprising administering to a subject a first compound which converts in vivo to a second compound, wherein the second compound inhibits CDK4/6 activity in vivo and the second compound is a compound of any one of the above embodiments and variations thereof.
In another aspect, the invention provides a method of treating a disease state in which CDK4/6 activity contributes to the pathology and/or symptomology of the disease state, comprising causing a therapeutically effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, to be present in a subject.
In another aspect, the invention provides a method of treating a disease state in which CDK4/6 activity contributes to the pathology and/or symptomology of the disease state, the method comprising administering to a subject a first compound which converts in vivo to a second compound, wherein the second compound inhibits CDK4/6 activity in vivo. It is noted that the compound of the present invention may be the first or second compound.
In variations of each of the above methods, the disease state is selected from: cancerous proliferative diseases (e.g., cancers of the brain, lung, squamous cell, bladder, stomach, pancreas, breast, head, neck, kidney, ovary, prostate, colon, epidermis, esophagus, testis, gynecology, or thyroid); non-cancerous proliferative diseases (e.g., benign skin hyperplasia (e.g., psoriasis), restenosis, and Benign Prostatic Hypertrophy (BPH)); pancreatitis; kidney disease; pain; preventing implantation of blastocysts; treating diseases associated with angiogenesis or vasculogenesis (e.g., tumor angiogenesis, acute and chronic inflammatory diseases such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, kaposi's sarcoma and ovarian cancer, breast cancer, lung cancer, pancreatic cancer, prostate cancer, colon cancer, and epidermoid cancer); asthma; neutrophil chemotaxis (e.g., reperfusion injury from myocardial infarction and stroke and inflammatory arthritis); septic shock; t cell mediated diseases in which immunosuppression is of value (e.g. prevention of organ transplant rejection, graft versus host disease, lupus erythematosus, multiple sclerosis and rheumatoid arthritis); atherosclerosis; inhibiting keratinocytes responsive to the growth factor mixture; chronic Obstructive Pulmonary Disease (COPD) and other diseases.
In another aspect, the invention provides a method of treating a disease state in which mutations in the CDK4/6 gene contribute to the pathology and/or symptomology of the disease, such as melanoma, lung, colon and other types of tumors.
In another aspect, the present invention relates to the use of a compound of any one of the above embodiments and variations thereof as a medicament. In another aspect, the invention relates to the use of a compound of any one of the above embodiments and variations thereof as a medicament for inhibiting CDK 4/6.
In another aspect, the invention relates to the manufacture of a compound of any one of the above embodiments and variations thereof as a medicament for the treatment of pathological and/or symptomatic disease states caused by CDK4/6 activity.
Administration and pharmaceutical compositions
Generally, the compounds of the present invention will be administered in a therapeutically effective amount, either alone or in combination with one or more therapeutic agents, by any of the usual and acceptable means known in the art. The therapeutically effective amount may vary widely depending on the severity of the disease, age and relative health of the subject, the potency of the compound used and other factors known in the art. For example, for the treatment of neoplastic diseases and immune system diseases, the required dosage will vary depending upon the mode of administration, the particular condition being treated and the desired effect.
In general, satisfactory results are achieved at daily dosages of from 0.001 to 100mg/kg body weight, in particular from about 0.03 to 2.5mg/kg body weight. Daily doses for larger mammals, such as humans, may be administered in a convenient form, for example in divided doses up to four times a day or in sustained release form, from about 0.5mg to about 2000mg, or more specifically, from 0.5mg to 1000 mg. Suitable unit dosage forms for oral administration contain from about 1 to 50mg of the active ingredient.
The compounds of the present invention may be administered in the form of pharmaceutical compositions, by any conventional route; e.g., enterally, e.g., orally, e.g., in the form of tablets or capsules, parenterally, e.g., in the form of injectable solutions or suspensions; or topically, e.g., in the form of a lotion, gel, ointment, or cream, or in the form of a nasal or suppository.
Pharmaceutical compositions containing a compound of the invention in free base or pharmaceutically acceptable salt form in association with at least one pharmaceutically acceptable carrier or diluent may be manufactured in conventional manner by means of mixing, granulating, coating, dissolving or lyophilizing processes. For example, pharmaceutical compositions comprising a compound of the invention in combination with at least one pharmaceutically acceptable carrier or diluent may be formulated in conventional manner by admixture with a pharmaceutically acceptable carrier or diluent. Unit dosage forms for oral administration contain, for example, from about 0.1mg to about 500mg of active substance.
In one embodiment, the pharmaceutical composition is a solution, including a suspension or dispersion, such as an isotonic aqueous solution, of the active ingredient. In the case of lyophilized compositions comprising the active ingredient alone or in admixture with a carrier such as mannitol, dispersions or suspensions may be prepared prior to use. The pharmaceutical compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. Suitable preservatives include, but are not limited to, antioxidants such as ascorbic acid, microbicides such as sorbic acid or benzoic acid. The solution or suspension may also contain a thickening agent including, but not limited to, sodium carboxymethylcellulose, dextran, polyvinylpyrrolidone, gelatin, or a solubilizing agent such as tween 80 (polyoxyethylene (20) sorbitan monooleate).
Suspensions in oils may contain, as oily component, vegetable oils, synthetic or semi-synthetic oils, commonly used for injection purposes. Examples include liquid fatty acid esters containing as the acid component a long chain fatty acid having from 8 to 22 carbon atoms, or in some embodiments, from 12 to 22 carbon atoms. Suitable liquid fatty acid esters include, but are not limited to, lauric acid, tridecylic acid, myristic acid, pentadecylic acid, palmitic acid, margaric acid, stearic acid, arachidic acid, behenic acid or corresponding unsaturated acids, such as oleic acid, elaidic acid, erucic acid, brassidic acid and linoleic acid, if desired, with antioxidants such as vitamin E, 3-carotene or 3, 5-di-tert-butylhydroxytoluene. The alcohol component of these fatty acid esters may have six carbon atoms and may be monovalent or polyvalent, such as mono-, di-or trivalent alcohols. Suitable alcohol components include, but are not limited to, methanol, ethanol, propanol, butanol or pentanol or isomers thereof, ethylene glycol and glycerol.
Other suitable fatty acid esters include, but are not limited to, ethyl oleate, isopropyl myristate, isopropyl palmitate, isopropyl myristate, isopropyl palmitate, isopropyl myristate, isopropyl palmitate, and isopropyl palmitate,
Figure BDA0001755256780000351
M2375 (polyoxyethylene glycerol),
Figure BDA0001755256780000352
M1944CS (unsaturated polyglycolyzed glycerides and mixtures containing glycerides and polyglycolyzed esters of almond oil by alcoholysis), LABRASOLTM(saturated polyglycolized glycerides prepared by alcoholysis of TCM and containing glycerides and polyethylene glycol esters; both available from GaKefosse, France), and/or
Figure BDA0001755256780000353
812 (triglycerides of saturated fatty acids with chain lengths of C8 to C12, from huls AG, germany), and vegetable oils such as cottonseed oil, almond oil, olive oil, castor oil, sesame oil, soybean oil or peanut oil.
Pharmaceutical compositions for oral administration may be obtained, for example, by mixing the active ingredient with one or more solid carriers, if desired granulating a resulting mixture, and processing the mixture or granules by adding further excipients, in the form of tablets or tablet cores.
Suitable carriers include, but are not limited to, fillers, for example sugars, such as lactose, sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and also binders, for example starches, such as corn, wheat, rice or potato starch, methylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone, and/or, if desired, disintegrants, such as the above-mentioned starches, carboxymethyl starch, crosslinked polyvinylpyrrolidone, alginic acid or a salt thereof, such as sodium alginate. Additional excipients include flow-regulating agents and lubricants, for example silicic acid, talc, stearic acid or salts thereof, such as magnesium or calcium stearate, and/or polyethylene glycol, or derivatives thereof.
The tablet cores may be provided with a suitable, optionally enteric, coating by using, inter alia, a concentrated sugar solution, which may comprise gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or a coating solution in a suitable organic solvent or solvent mixture, or, for enteric coatings, a solution of a suitable cellulose preparation, such as an acetylcellulose phthalate or hydroxypropylmethylcellulose phthalate solution. Dyes or pigments may be added to the tablets or tablet coatings, for example for identification purposes or to indicate different doses of the active ingredient.
Pharmaceutical compositions for oral administration may also include hard capsules, including gelatin or soft, sealed capsules containing gelatin and a plasticizer, such as glycerol or sorbitol. Hard capsules may contain the active ingredients in the form of granules, for example in admixture with fillers such as corn starch, binders and/or glidants such as talc or magnesium stearate, and optionally stabilizers. In soft capsules, the active ingredient may be dissolved or suspended in suitable liquid excipients, such as fatty oils, paraffin oils or liquid polyethylene glycols or fatty acid esters of ethylene glycol or propylene glycol, to which stabilizers and detergents, for example of the fatty acid ester type of polyoxyethylene sorbitol, are added.
Pharmaceutical compositions suitable for rectal administration, for example suppositories, comprise a combination of the active ingredient and a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, paraffin hydrocarbons, polyethylene glycols or higher alkanols.
Pharmaceutical compositions suitable for parenteral administration may comprise the active ingredient in water-soluble form, for example as a water-soluble salt or as an aqueous injection suspension comprising a viscosity-increasing substance, for example sodium carboxymethylcellulose, an aqueous solution of sorbitol and/or dextran, if desired, and a stabilizer. The active ingredient, optionally together with excipients, may also be in a lyophilized form and may be prepared as a solution by addition of a suitable solvent prior to parenteral administration. The solutions used, for example for parenteral administration, can also be used as infusion solutions. Injectable preparations are generally prepared under sterile conditions, and filled, for example, in ampoules or vials, and in sealed containers.
The invention also provides a pharmaceutical combination, e.g. a kit, comprising a) a compound disclosed herein, in free form or in pharmaceutically acceptable salt form, and b) at least one auxiliary agent. The kit may contain instructions for its use.
Combination therapy
The compounds or pharmaceutically acceptable salts described in this disclosure may be used alone or in combination with other therapeutic agents.
For example, the therapeutic benefit of a compound of the invention may be enhanced by the use of an adjuvant drug (e.g., the therapeutic benefit of the adjuvant drug alone may be minimal, but the therapeutic benefit of the subject may be enhanced when used in combination with another drug), or, for example, the therapeutic benefit of the subject may be enhanced by the use of a compound of the invention in combination with another therapeutic agent that is also therapeutically effective. For example, in the treatment of gout, the compound of the invention may be used in combination with another drug for gout therapy to enhance clinical benefit. Alternatively, for example, if the adverse effect of using the compounds of the present invention is nausea, then an anti-nausea agent may be used in combination. Alternatively, combination therapies may include, but are not limited to, physical therapy, psychotherapy, radiation therapy, compression therapy of diseased areas, rest, dietary improvement, and the like. Regardless of the disease, disorder, or condition being treated, both therapies should have additive or synergistic effects to benefit the treatment of an individual.
Where the compounds described herein are used in combination with other therapeutic agents, the route of administration of the pharmaceutical compositions of the compounds described herein may be the same as the other drugs, or the route of administration may be different due to differences in physical and chemical properties. For example, oral administration of a compound described herein may produce and maintain good blood levels, while intravenous administration of another therapeutic agent may be required. Thus, the compounds described herein and another therapeutic agent may be administered simultaneously, sequentially or separately.
The compounds of formula (I) or (II) are expected to be effective in combination with one or more of the following: alkylating agents, angiogenesis inhibitors, antibodies, antimetabolites, antimitotics, antiproliferatives, antivirals, aurora kinase inhibitors, promoters of other apoptosis (e.g., Bcl-xL, Bcl-W, and Bfl-1) inhibitors, death receptor pathway activators, Bcr-Abl kinase inhibitors, antibodies to BiTE (bispecific T-cell engagers), antibody drug conjugates, biological response modifiers, cyclin-dependent kinase inhibitors, cell cycle inhibitors, cyclooxygenase-2 ErbB inhibitors, DVDs, leukemia virus oncogene homolog (2) receptor inhibitors, growth factor inhibitors, Heat Shock Protein (HSP) -90 inhibitors, histone acetylases (HDAC) inhibitors, hormonal therapies, immunological agents, inhibitors of apoptosis protein Inhibitors (IAPs), intercalating antibiotics, inhibitors of apoptosis proteins, and the like, Kinase inhibitors, kinesin inhibitors, JAK2 inhibitors, rapamycin inhibitors for mammals, micrornas, mitogen-activated extracellular signal-regulated kinase inhibitors, multivalent binding proteins, nonsteroidal anti-inflammatory drugs (NSAIDs), poly ADP (adenosine diphosphate) -ribose polymerase (PARP) inhibitors, platinum-based chemotherapeutic drugs, polo-like kinase (PLK) inhibitors, phosphoinositide 3 kinase (PI3K) inhibitors, proteasome inhibitors, purine analogs, pyrimidine analogs, receptor tyrosine kinase inhibitors, retinoid/deltoid plant alkaloids, small interfering RNA (sirnas) inhibitors, topoisomerase inhibitors, ubiquitin ligase inhibitors, and the like.
Examples
There are various methods for synthesizing the compound of formula (I) or (II) or a pharmaceutically acceptable salt thereof, and representative methods are listed in this example. However, it is to be noted that the compounds of formula (I) or (II) or pharmaceutically acceptable salts thereof may also be obtained by synthesis by other synthetic schemes.
In certain compounds of formula (I) or (II), the attachment of an atom to another atom may result in the presence of a particular stereoisomer (e.g., a chiral center). The synthesis of a compound of formula (I) or (II) or a pharmaceutically acceptable salt thereof may result in a mixture of different isomers (enantiomers, diastereomers). Unless a particular configuration is specified, all recited compounds include different stereoisomers that may exist.
The compounds of formula (I) or (II) may also be prepared as pharmaceutically acceptable acid addition salts, for example by reacting the free base form of the compounds of the invention with a pharmaceutically acceptable inorganic or organic acid. Or a compound of formula (I) or (II) in free acid form with a pharmaceutically acceptable inorganic or organic base, to form a pharmaceutically acceptable base addition salt. Inorganic and organic acids and bases suitable for preparing pharmaceutically acceptable salts of compounds of formula (I) or (II) are described in the definitions section herein. Furthermore, salt forms of the compounds of formula (I) or (II) can also be prepared by using salts of the starting materials or intermediates.
The free acid or free base of the compounds of formula (I) or (II) may be prepared from the corresponding base addition salt or acid addition salt thereof. The acid addition salt forms of the compounds of formula (I) or (II) may be converted to the corresponding free base, for example by treatment with a suitable base such as ammonium hydroxide solution, sodium hydroxide and the like. The base addition salt forms of the compounds of formula (I) or (II) may be converted to the corresponding free acids, for example by treatment with a suitable acid, such as hydrochloric acid and the like.
An N-oxide of a compound of formula (I) or (II) or a pharmaceutically acceptable salt thereof may be prepared by methods known in the art. For example, the N-oxide may be obtained by reacting a non-oxidized form of the compound of formula (I) with an oxidizing agent (e.g., trifluoroperacetic acid, peroxymaleic acid, perbenzoic acid, peracetic acid, m-chloroperoxybenzoic acid, and the like) in an inert organic solvent (e.g., a halogenated hydrocarbon such as dichloromethane) at a temperature near 0 ℃. Alternatively, the N-oxides of the compounds of formula (I) or (II) may also be prepared from the N-oxides of the starting materials.
The non-oxidized compound of formula (I) or (II) can be prepared by reacting an N-oxide thereof with a reducing agent (such as sulfur, sulfur dioxide, triphenylphosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, phosphorus tribromide and the like) at 0-80 ℃ in a corresponding inert organic solvent (such as acetonitrile, ethanol, hydrated dioxane and the like).
Protected derivatives of compounds of formula (I) or (II) may be prepared by methods well known to those skilled in the art. For a detailed technical description of the addition and removal of protecting groups see: greene, Protecting Groups in Organic Synthesis,3rd edition, John Wiley & Sons, Inc.1999.
The labels and common sense, charts and examples used in these reactions are consistent with the current scientific literature, e.g., the journal of the American chemical Association or the journal of biochemistry. Standard single or three letter abbreviations, unless otherwise indicatedReferences generally refer to L-amino acid residues. All starting materials used were purchased from commercial suppliers and used without further purification unless otherwise indicated. For example, the following abbreviations are used in the examples and throughout the specification: g (g), mg (mg), L (L), mL (mL), μ L (μ L), psi (pounds per square inch), M (mol), mM (mmol), i.v. (i.v.), Hz (Hz), MHz (megahertz), mol (mol), mmol (mmol), RT (ambient temperature), min (min), h (h), mp (melting point), TLC (thin layer chromatography), Rr (retention time), RP (reversed phase), MeOH (methanol), i-PrOH (isopropanol), TEA (triethylamine), TFA (trifluoroacetic acid), TFAA (trifluoroacetic anhydride), THF (tetrahydrofuran), DMSO (dimethyl sulfoxide), EtOAc (ethyl acetate), DME (1, 2-dimethyl ether), DCM (dichloromethane), DCE (dichloroethane), DMF (N, N-dimethylformamide), DMPU (N, N-dimethylpropylurea), CDI (1, 1-carbonyldiimidazole), IBCF (isobutyl chloroformate), HOAc (acetic acid), HOSu (N-hydroxysuccinimide), HOBT (1-hydroxybenzotriazole), Et2O (diethyl ether), EDCI (1- (3-dimethylaminopropyl) 3-ethylcarbodiimide hydrochloride), BOC (t-butyloxycarbonyl), FMOC (9-fluorenylmethoxycarbonyl), DCC (dicyclohexylcarbodiimide), CBZ (benzyloxycarbonyl), Ac (acetyl), atm (atmospheric pressure), TMSE (2- (trimethylsilyl) ethyl), TMS (trimethylsilyl), TIPS (triisopropylsilyl), TBS (t-butyldimethylsilyl), DMAP (dimethylaminopyridine), Me (methyl), OMe (methoxy), Et (ethyl), tBu (t-butyl), HPLC (high performance liquid chromatography), BOP (bis (2-oxo-3-oxazolidinyl) hypophosphoryl chloride), AF (tetrabutylammonium fluoride), TBmC (m-chloroperbenzoic acid).
Ether or Et2O is diethyl ether; brine is then a saturated aqueous NaCl solution. Unless otherwise indicated, all temperatures refer to degrees Celsius (degrees Celsius) and all reactions are carried out in an inert atmosphere at room temperature.
1H NMR spectra were recorded using a Varian Mercury Plus 400 NMR spectrometer. Chemical shifts are expressed in ppm. The coupling constants are all in hertz (Hz). Apparent diversity is described in the split mode and is assigned as s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet) andbr (broad peak)
Low resolution Mass Spectrometry (MS) and compound purity data were from a single-pole system of Shimadzu mass spectrometry equipped with an electrospray ion detector (ESI), ultraviolet detectors (220 and 254nm) and an Evaporative Light Scattering Detector (ELSD). Thin layer chromatography was performed using 0.25mm Asahi-poise silica gel plate (60F-254), 5% ethanol phosphomolybdate solution, ninhydrin or p-methoxybenzaldehyde solution and observing under an ultraviolet lamp. Silica gel (200-300 mesh, Qingdao ocean chemical Co., Ltd.) was used for flash column chromatography.
Synthetic schemes
The compounds of formula (I) or (II) or pharmaceutically acceptable salts thereof may be synthesized by various methods, some exemplary methods are provided below and in the examples. Other synthetic methods can be readily suggested by those skilled in the art based on the information disclosed herein.
It may be necessary to protect reactive groups in the reactions described below in order to prevent these reactive groups from participating in other undesired reactions: such as hydroxyl, amino, imino, mercapto or carboxyl groups, which are contained in the final product. Commonly used protecting Groups are referred to T.W.Greene and P.G.M.Wuts in "Protective Groups in Organic Chemistry" John Wiley and Sons, 1991.
The synthetic schemes for all compounds of the present invention are illustrated by the following schemes and examples. The starting materials used are commercially available or may be prepared according to established procedures or by methods exemplified herein.
The intermediates listed in the following synthetic schemes are either obtained from the literature or are synthesized according to established analogous synthetic methods.
As illustrated below, one synthetic route for the compounds of formula I or II of the present invention is shown in FIG. 1. The compounds of formula I or II can be synthesized from intermediates III to VIII shown in the synthetic schemes, which can be synthesized according to literature or methods well known to those skilled in the art. The pyrimidine intermediates of formula IV or V are prepared from pyrimidine intermediates of formula VI and heteroarene intermediates of formula VII or VIII by palladium-catalyzed coupling reactions, such as Suzuki reactions or according to other coupling conditions reported in the literature. The compounds of formula I or II are prepared by coupling of formula IV or V with an aminoarene of formula III, such as Buchwald amination or according to other coupling conditions reported in the literature.
Figure BDA0001755256780000401
As an example of the preparation of intermediate IV, one synthetic route for the compound of formula IVa is shown in scheme 2. Starting from commercially available diaminopyridine A. The intermediate IVa is prepared by subsequent reductive amination, imidazole formation and conversion of the halogen group to a group such as B or Sn followed by a palladium catalyzed coupling reaction.
Figure BDA0001755256780000402
One synthetic route for the intermediate Vb compounds is shown in scheme 3. Reacting ketone N with hydrazine to obtain an intermediate O, alkylating the intermediate O to obtain an intermediate P, converting a halogen group in the intermediate P into a group such as B or Sn, and then carrying out palladium-catalyzed coupling reaction to obtain an intermediate Vb.
Figure BDA0001755256780000411
In some cases, the above synthetic schemes may be ordered as appropriate in order to facilitate the reaction or to avoid the production of unnecessary reaction products. In order that the invention may be more fully understood, the following examples are set forth. These examples are only examples and should not be construed as limiting the invention.
Preparation of intermediates
Intermediate A
6- (2-chloro-5-fluoropyrimidin-4-yl) -1-isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridine (intermediate A)
Figure BDA0001755256780000412
35-bromo-N-isopropylpyridine-2, 3-diamine (A-1)
To a solution of commercial 2, 3-diamino-5-bromopyridine (1.88g,10mmol, in plain translation 5-bromo-2, 3-diaminopyridine) (1.7 g,12mmol) and acetone (0.7g,12mmol) in isopropyl acetate (15mL) was added TFA (2.51g,22mmol) followed by NaBH (OAc) at 0 deg.C3(3.18g,15 mmol). The mixture was stirred at room temperature for 2 hours. The reaction was quenched with EtOAc (50 mL). With saturated NaHCO3Washed with aqueous solution (30mL) and saturated brine (30mL), and then with Na2SO4And (5) drying. Filtering and rotary evaporating, purifying the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate ═ 9: 1-2: 1) to obtain 5-bromo-N as yellow solid3Isopropylpyridine-2, 3-diamine (A-1). MS-ESI (M/z) 230,232[ M + 1]]+
6-bromo-1-isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridine (A-2)
5-bromo-N3Isopropylpyridine-2, 3-diamine (A-1) (1.2g,5.2mmol) and Ac2A mixture of O (2.6g,26mmol) in HOAc (10mL) was stirred at 90 deg.C overnight. The solvent was spun down in vacuo and the residue was diluted with DCM (50 mL). Adjusting the pH value to 8-9 by NaOH (1N). The organic layer was separated. The extract was washed with saturated brine (50mL), Na2SO4And (5) drying. Filtering and concentrating, purifying the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate ═ 5: 1-1: 2) to obtain 6-bromo-1-isopropyl-2-methyl-1H-imidazo [4,5-b ] as a yellow solid]Pyridine (A-2). MS-ESI (M/z) 254,256[ M + 1]]+
6- (tri-n-butylstannyl) -1-isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridine (A-3)
to-bromo-1-isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridine (A-2) (0.5g,2mmol) in THF (10mL) was added (Bu)3Sn)2(2.32g,4mmol),Pd(PPh3)(OAc)2(0.08g,0.1mmol) and TBAF (1.044g,8 mmol). Stirring at 70 ℃ for 4h under nitrogen protection. The mixture was concentrated and the residue was diluted with ethyl acetate (50 mL). Extraction ofThe material was washed with water (50mL), MgSO4And (5) drying. Filtered and concentrated, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate ═ 9:1 to 1:1) to give 6- (tri-n-butylstannyl) -1-isopropyl-2-methyl-1H-imidazo [4,5-b ] as a yellow oil]Pyridine (A-3). MS-ESI (M/z) 466[ M + 1]]+
6- (2-chloro-5-fluoropyrimidin-4-yl) -1-isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridine (intermediate A)
To 6- (tri-n-butylstannyl) -1-isopropyl-2-methyl-1H-imidazo [4,5-b]To a solution of pyridine (A-3) (0.455g,0.97mmol) and 2, 4-dichloro-5-fluoropyrimidine (0.25g,1.49mmol) in toluene (8mL) was added Pd (PPh)3)4(0.06g,0.05 mmol). Stirring at 110 ℃ for 4h under nitrogen protection. The mixture was concentrated. The residue was purified by flash column chromatography on silica gel (eluent: ethyl acetate) to give 6- (2-chloro-5-fluoropyrimidin-4-yl) -1-isopropyl-2-methyl-1H-imidazo [4,5-b ] as a yellow solid]Pyridine (intermediate a). MS-ESI (M/z) 306[ M + 1]]+
Intermediate B
5- (2-chloro-5-fluoropyrimidin-4-yl) -7-fluoro-3-isopropyl-1-methyl-3 a,7 a-dihydro-1H-indazole (intermediate) B)
Figure BDA0001755256780000421
5-bromo-2-chloronicotinic acid (isobutyl carbonate) anhydride (B-1)
To a solution of 5-bromo-2-chloronicotinic acid (10.0g,42.3mmol) in anhydrous THF (440mL) at 0 deg.C was added TEA (7.06mL,50.8mmol) followed by isobutyl chloroformate (6.64mL,50.76mmol) and a white precipitate formed rapidly. The mixture was stirred at 0 ℃ for 75 minutes. After filtration the solid was washed with THF (30 mL). The solution was concentrated to obtain 200mL of a solution containing 5-bromo-2-chloronicotinic acid (isobutylcarbonic acid) anhydride (B-1) which was used directly in the next reaction.
(5-bromo-2-chloropyridin-3-yl) methanol (B-2)
Water (10mL) was added to the above solution.After the mixture was cooled to 0 deg.C, NaBH was added4(3.38g,89mmol), the mixture was stirred at 0 ℃ for 1 hour and then warmed to room temperature for more than 2 hours. Ethyl acetate was added to the reaction mixture, which was washed with water and saturated brine, dried over anhydrous potassium carbonate and sodium sulfate, and concentrated to give a crude product of (5-bromo-2-chloropyridin-3-yl) methanol (B-2) which was used directly in the next reaction.
5-bromo-2-chloro-3-pyridinecarboxaldehyde (B-3)
At-78 deg.C, to (COCl)2To a solution of (2.95mL,34.84mmol) in anhydrous DCM (26mL) was added DMSO (4.94mL,69.7mmol) in DCM (45mL) dropwise and stirred at-78 deg.C for 10 min. A solution of (5-bromo-2-chloropyridin-3-yl) methanol (B-2) in DCM (45mL) was added dropwise. The mixture was stirred at-78 ℃ for 10 minutes. TEA (18.2mL,131mmol) was added dropwise and stirring continued at-78 deg.C for 1 hour. The mixture was allowed to warm to room temperature and stirred for 1 hour. After dilution with water (100mL), the organic layer was washed with water and saturated brine, dried over anhydrous magnesium sulfate, and concentrated. The residue was purified by silica gel column chromatography eluting with 3-5% ethyl acetate in hexane to give 5-bromo-2-chloro-3-pyridinecarboxaldehyde (B-3) as a white solid. MS-ESI (M/z) 220[ M + 1]]+
1- (5-bromo-2-chloropyridin-3-yl) -2-methylpropan-1-ol (B-4)
In N2To a solution of 5-bromo-2-chloro-3-pyridinecarboxaldehyde (B-3) (5.63g,25.53mmol) in dry THF (70mL) at-78 deg.C was added isopropyl magnesium chloride (15.3 mL of 2.0M solution in THF) dropwise at ambient temperature. The mixture was warmed to room temperature and stirred overnight. After water was added, the mixture was separated, the aqueous layer was extracted with ethyl acetate, and the organic layer was washed with water and saturated brine, dried over anhydrous magnesium sulfate, and concentrated. The residue was purified by silica gel column chromatography to give 1- (5-bromo-2-chloropyridin-3-yl) -2-methylpropan-1-ol (B-4). MS-ESI (M/z):264[ M +1 [)]+
1- (5-bromo-2-chloropyridin-3-yl) -2-methylpropan-1-one (B-5)
B-4(265mg,1mmol), PCC (431mg,2mmol) and SiO2A mixture (0.5g) in DCM (8mL) was stirred at room temperature for 5 h. The mixture was filtered through celite and the residual solid was washed with DCM. Concentrating the filtrate, and separating the residue with silica gel columnPurifying by chromatography to obtain 1- (5-bromo-2-chloropyridin-3-yl) -2-methylpropane-1-one (B-5). MS-ESI (M/z):262[ M +1 [)]+
5-bromo-3-isopropyl-1H-pyrazolo [3,4-b]Pyridine (B-6)
To a solution of 1- (5-bromo-2-chloropyridin-3-yl) -2-methylpropan-1-one (B-5) (177mg,0.674mmol) in isopropanol (2mL) was added hydrazine hydrate (0.5mL) at room temperature. The mixture was warmed to 80 ℃ and stirred overnight. The mixture was cooled to room temperature, diluted with water and extracted with ethyl acetate. The extract was washed with saturated brine and Na2SO4Dried and concentrated. The residue was purified by silica gel column chromatography (eluent: n-hexane/ethyl acetate ═ 1:1) to give 5-bromo-3-isopropyl-1H-pyrazolo [3, 4-b)]Pyridine (B-6). MS-ESI (M/z) 240[ M + 1]]+
5-bromo-3-isopropyl-2-methyl-2H-pyrazolo [3,4-b]Pyridine (B-7)
To 5-bromo-3-isopropyl-1H-pyrazolo [3,4-b ] at 0 DEG C]Pyridine (B-6) (48mg,0.2mmol) in THF was added NaH (10mg, 60% in oil), stirred for 5 min and then MeI (25. mu.L, 0.4mmol) was added dropwise. The mixture was stirred at 0 ℃ for 20 minutes. The reaction was quenched with water. The mixture was extracted with ethyl acetate, washed with saturated brine and Na2SO4Drying and concentrating. Purifying the residue by silica gel column chromatography, eluting with 10-100% ethyl acetate hexane solution to obtain 5-bromo-3-isopropyl-2-methyl-2H-pyrazolo [3, 4-b)]Pyridine (B-7), MS-ESI (M/z):254[ M + 1]]+And 5-bromo-3-isopropyl-1-methyl-1H-pyrazolo [3,4-b]Pyridine (B-7B). MS-ESI (M/z) 254[ M + 1]]+
(3-isopropyl-1-methyl-1H-pyrazolo [3, 4-b)]Pyridin-5-yl) boronic acid (B-8)
5-bromo-3-isopropyl-2-methyl-2H-pyrazolo [3,4-b]Pyridine (B-7) (724mg,2.85mmol), Biboronic acid pinacol ester (1.09g,4.27mmol), Pd (OAc)2(64mg,0.28mmol),Cy3A mixture of P (160mg,0.57mmol) and KOAc (838mg,8.55mmol) in DMSO (10mL) was stirred under nitrogen at 100 ℃ for 2h in a sealed tube. The reaction was quenched with water. The mixture was extracted with DCM and the extract Na2SO4Dried and concentrated to give the title compound (3-isopropyl-1-methyl-1H-pyrazolo [3, 4-b)]Pyridin-5-yl) boronic acid (B-8). MS-ESI (M/z) 220[ M + 1]]+
5- (2-chloro-5-fluoropyrimidin-4-yl) -7-fluoro-3-isopropyl-1-methyl-3 a,7 a-dihydro-1H-indazole (intermediate) B)
(3-isopropyl-1-methyl-1H-pyrazolo [3, 4-b)]Pyridin-5-yl) boronic acid (B-8) (624mg,2.85mmol), 2, 4-dichloro-5-fluoropyrimidine (952mg,5.7mmol), Pd (PPh)3)2Cl2(200mg,0.28mmol) and Na2CO3(906mg,8.55mmol) in DME/H2The mixture of O (24ml/10ml) solution was stirred under nitrogen at 80 ℃ for 2 hours in a sealed tube. The reaction was quenched with water. The mixture was extracted with DCM and the combined organic layers were washed with Na2SO4Drying and concentrating. The residue was purified by silica gel column chromatography (eluent: n-hexane/ethyl acetate ═ 2:1) to give the title compound 5- (2-chloro-5-fluoropyrimidin-4-yl) -7-fluoro-3-isopropyl-1-methyl-3 a,7 a-dihydro-1H-indazole (intermediate B). MS-ESI (M/z) 306[ M + 1]]+
Intermediate C
5- (2-chloro-5-fluoropyrimidin-4-yl) -3-isopropyl-1-methyl-1H-pyrazolo [3,4-b]Pyridine (intermediate C)
Figure BDA0001755256780000441
5-bromo-3-isopropyl-1-methyl-1H-pyrazolo [3,4-b]Pyridine (C-1)
To a solution of 1- (5-bromo-2-chloropyridin-3-yl) -2-methylpropan-1-one (B-5) (38mg,0.15mmol) and methylhydrazine sulfate (25mg,0.2mmol) in EtOH (2mL) was added TEA (50mg,5 mmol). The mixture was stirred at room temperature for 0.5 hour and then refluxed overnight. The mixture was cooled to room temperature and concentrated. The residue was diluted with DCM (20mL), washed successively with water and saturated brine (20mL), Na2SO4Drying and rotary steaming. Purifying the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate: 100: 1-50: 1) to obtain5-bromo-3-isopropyl-1-methyl-1H-pyrazolo [3,4-b]Pyridine (C-1). MS-ESI (M/z) 254[ M + 1]]+
(3-isopropyl-1H-pyrazolo [3, 4-b)]Pyridin-5-yl) boronic acid (C-2)
According to the synthesis method of B-8, 5-bromo-3-isopropyl-2-methyl-2H-pyrazolo [3, 4-B)]Replacement of pyridine (B-7) with 5-bromo-3-isopropyl-1-methyl-1H-pyrazolo [3,4-B]Pyridine (C-1) to prepare the title compound (C-2). MS-ESI (M/z) 220[ M + 1]]+
5- (2-chloro-5-fluoropyrimidin-4-yl) -3-isopropyl-1-methyl-1H-pyrazolo [3,4-b]Pyridine (intermediate J)
(3-isopropyl-1-methyl-1H-pyrazolo [3, 4-B) according to the synthesis method of intermediate B]Replacement of pyridin-5-yl) boronic acid (B-8) with (3-isopropyl-1H-pyrazolo [3, 4-B)]Pyridin-5-yl) boronic acid (C-2) gave the title compound (intermediate C). MS-ESI (M/z) 306[ M + 1]]+
Intermediate D
6-bromo-1-tert-butyl-2-methyl-1H-imidazo [4,5-b]Pyridine (intermediate D)
Figure BDA0001755256780000451
5-bromo-3-fluoropyridin-2-amine (D-1)
NBS (8.72g,49.0mmol) was added to a solution of 3-fluoropyridin-2-amine (11.0g,98.0mmol) in ACN (300mL) at 0 ℃ and stirred at ambient temperature for 1 hour. NBS (8.72g,49.0mmol) was added dropwise. The reaction mixture was concentrated and diluted with water (100 mL). Filtered through celite. The filter cake was washed with water and the filtrate was concentrated to give the title compound, 5-bromo-3-fluoropyridin-2-amine (D-1), as a crude product. MS-ESI (M/z):191[ M + 1]]+
5-bromo-3-fluoro-2-nitropyridine (D-2)
To a mixture of 5-bromo-3-fluoropyridin-2-amine (D-1) (4.0g,21.0mmol) in concentrated sulfuric acid (20mL) at 0 deg.C was added H2O2(33mL,29.1mmol) in concentrated sulfuric acid (66 mL).The mixture was allowed to warm to room temperature and stirred overnight. Basified with dilute sodium carbonate solution at 0 ℃ to pH 7 and extracted with ethyl acetate. The organic layer was successively washed with Na2S2O3Washing with saturated brine and Na2SO4And (5) drying. Filtered and concentrated, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate 40:1) to give the title compound 5-bromo-3-fluoro-2-nitropyridine (D-2). MS-ESI (M/z) 220[ M + 1]]+
5-bromo-N-tert-butyl-2-nitropyridin-3-amine (D-3)
A mixture of 5-bromo-3-fluoro-2-nitropyridine (D-2) (1.63g,7.38mmol), tert-butylamine (1.08g,14.8mmol) and TEA (1.49g,14.8mmol) in THF (30mL) was stirred at 45 deg.C overnight. Water and ethyl acetate were added. The aqueous layer was extracted with EtOAc (3X 30 mL). The organic layer was washed with saturated brine and Na2SO4Drying, concentration and purification of the residue by silica gel column chromatography (eluent: petroleum ether/ethyl acetate ═ 30:1) gave the title compound 5-bromo-N-tert-butyl-2-nitropyridin-3-amine (D-3). MS-ESI (M/z):274[ M + 1]]+
35-bromo-N-tert-butylpyridine-2, 3-diamine (D-4)
A mixture of 5-bromo-N-tert-butyl-2-nitropyridin-3-amine (D-3) (1.3g,4.7mmol), aqueous NH4Cl (16mL) and Fe (2.6g,46mmol) in EtOH (16mL) was stirred at 75 ℃ for 40 min. The mixture was filtered and the filtrate extracted with DCM (3X 30 mL). The organic layer was washed with saturated brine and Na2SO4Drying and concentrating to obtain the title compound 5-bromo-N3-tert-butylpyridine-2, 3-diamine (D-4). MS-ESI (M/z) 244[ M + 1]]+
6-bromo-1-tert-butyl-2-methyl-1H-imidazo [4,5-b]Pyridine (intermediate D)
5-bromo-N3-tert-butylpyridine-2, 3-diamine (D-4) (50mg,0.205mmol) and Ac2A mixture of O (21. mu.l, 0.225mmol) in AcOH (1mL) was stirred at 90 ℃ overnight. The mixture was concentrated and the residue was washed with sodium bicarbonate solution (50mL) and then extracted with EtOAc (3 × 30 mL). The extract was extracted with water (2X 30mL) and saturated salt in that orderWashed with water (30mL), dried and concentrated to give the title compound 6-bromo-1-tert-butyl-2-methyl-1H-imidazo [4,5-b]Pyridine (intermediate D). MS-ESI (M/z) 268[ M + 1]]+
Intermediate E
3-tert-butyl-2-methyl-5- (tributyltin) -2H-pyrazolo [3,4-b]Pyridine (intermediate E)
Figure BDA0001755256780000461
5-bromo-2-chloro-N-methoxy-N-methylnicotinamide (E-1)
The title compound 5-bromo-2-chloro-N-methoxy-N-methylnicotinamine (E-1) was prepared by replacing isobutyl chloroformate with dimethylhydroxylamine hydrochloride (translated as N-methoxymethylaminochloride, but not commonly) according to the synthetic method of B-1. MS-ESI (M/z):279[ M +1]+
5-bromo-2-chloro-3-pyridinecarboxaldehyde (E-2)
DIBAL-H (70mL,105mmol) was added to a CDM (250mL) solution of 5-bromo-2-chloro-N-methoxy-N-methylnicotinamine (E-1) (11.8g,42mmol) at-78 deg.C, and the mixture was stirred at 0 deg.C for 0.5H. The mixture was concentrated, and the residue was purified by silica gel column chromatography (eluent: petroleum ether/ethyl acetate ═ 10:1) to give the title compound 5-bromo-2-chloro-3-pyridinecarboxaldehyde (E-2). MS-ESI (M/z) 220[ M + 1]]+
1- (5-bromo-2-chloropyridin-3-yl) -2, 2-dimethylpropan-1-ol (E-3)
To a solution of 5-bromo-2-chloro-3-pyridinecarboxaldehyde (E-2) (2.1g,10mmol) in THF (60mL) at 0 deg.C was added tert-butylmagnesium chloride (50mL,50 mmol). The mixture was stirred at 0-10 ℃ for 2h, and the reaction was quenched with MeOH (10 mL). The mixture was concentrated to give the title compound 1- (5-bromo-2-chloropyridin-3-yl) -2, 2-dimethylpropan-1-ol (E-3). MS-ESI (M/z):278[ M +1]+
1- (5-bromo-2-chloropyridin-3-yl) -2, 2-dimethylpropan-1-one (E-4)
According toSynthesis of B-5 substituting 1- (5-bromo-2-chloropyridin-3-yl) -2-methylpropan-1-ol (B-4) with 1- (5-bromo-2-chloropyridin-3-yl) -2, 2-dimethylpropan-1-ol (E-3) produced the title compound 1- (5-bromo-2-chloropyridin-3-yl) -2, 2-dimethylpropan-1-one (E-4). MS-ESI (M/z):276[ M +1 [)]+
5-bromo-3-tert-butyl-1H-pyrazolo [3,4-b]Pyridine (E-5)
The title compound 5-bromo-3-tert-butyl-1H-pyrazolo [3,4-B ] was prepared according to the synthetic method of B-6 by replacing 1- (5-bromo-2-chloropyridin-3-yl) -2-methylpropan-1-one (B-5) with 1- (5-bromo-2-chloropyridin-3-yl) -2, 2-dimethylpropan-1-one (E-4)]Pyridine (E-5). MS-ESI (M/z) 254[ M + 1]]+
5-bromo-3-tert-butyl-2-methyl-2H-pyrazolo [3,4-b]Pyridine (E-6)
To 5-bromo-3-tert-butyl-1H-pyrazolo [3,4-b ] at 0 DEG C]Pyridine (E-5) (320mg,1.3mmol) in THF (10ml) was added NaH (80mg, 60% in oil), stirred for 5 min and then MeI (370mg,2.6mmol) was added dropwise. The mixture was stirred at 0 ℃ for 20 minutes. The reaction was quenched with water. The mixture was extracted with ethyl acetate, washed with saturated brine and Na2SO4Drying and concentrating. Purifying the residue by silica gel column chromatography, eluting with 10-100% ethyl acetate in hexane to obtain 5-bromo-3-tert-butyl-2-methyl-2H-pyrazolo [3, 4-b)]Pyridine (E-6), MS-ESI (M/z):268[ M +1]+And 5-bromo-3-tert-butyl-1-methyl-1H-pyrazolo [3,4-b]Pyridine (E-6 b). MS-ESI (M/z) 268[ M + 1]]+
3-tert-butyl-2-methyl-5- (tributyltin) -2H-pyrazolo [3,4-b]Pyridine (intermediate E)
5-bromo-3-tert-butyl-2-methyl-2H-pyrazolo [3,4-b]Pyridine (E-6) (134mg,0.5mmol), (Bu)3Sn)2(870mg,1.5mmol),Pd(PPh3)4A mixture of (22mg,0.02mmol) and TEA (150mg,1.5mmol) in 1, 4-dioxane (5mL) was stirred at 80 deg.C overnight. The mixture was concentrated, and the residue was purified by silica gel column chromatography (eluent: ethyl acetate) to give the title compound 3-tert-butyl-2-methyl-5- (tributyltin) -2H-pyrazolo[3,4-b]Pyridine (intermediate E). MS-ESI (M/z) 480[ M + 1]]+
Example 1
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) -N- (5- (piperazin-1-ylmethyl) Yl) pyridin-2-yl) pyrimidin-2-amine (1)
Figure BDA0001755256780000471
Tert-butyl 4- ((6-bromopyridin-3-yl) methyl) piperazine-1-carboxylic acid ester (1a)
To a solution of 6-bromonicotinaldehyde (11.0g,59.1mmol) and 1-Boc-piperazine (10.0g,53.7mmol) in DCM (100mL) at 0 deg.C was added NaBH (OAc) in portions3(13.6g,64.1 mmol). The mixture was allowed to warm to room temperature and stirred overnight. DCM (100mL) was added followed by aqueous NaOH (2N,100mL) at 0 ℃. The organic layer was separated and the aqueous layer was extracted with DCM (100 mL). The extract was washed with saturated brine (100mL) and Na2SO4Drying and concentrating to obtain a yellow oily crude product of tert-butyl 4- ((6-bromopyridine-3-yl) methyl) piperazine-1-carboxylic ester (1a), which is directly used for the next reaction. MS-ESI (M/z):356, 358[ M + 1]]+
Tert-butyl 4- ((6-aminopyridin-3-yl) methyl) piperazine-1-carboxylate (1b)
Under nitrogen protection, tert-butyl 4- ((6-bromopyridin-3-yl) methyl) piperazine-1-carboxylate (1a) (3.56g,10mmol), Cu2O (0.50g,0.3mmol) in NH3.H2A mixture of O (20mL) and MeOH (20mL) was stirred in a sealed tube at 70 ℃ overnight. The mixture was cooled to room temperature, filtered and the filtrate was concentrated. Diluted with aqueous NaOH (2N,50mL) and extracted with DCM (2X 100 mL). The extract was washed with saturated brine (100mL) and Na2SO4Drying, filtration and concentration gave a crude product as a yellow oil which was purified by recrystallization from MTBE to give tert-butyl 4- ((6-aminopyridin-3-yl) methyl) piperazine-1-carboxylate (1 b). MS-ESI (M/z):293[ M + 1]]+
Tert-butyl 4- ((6- ((5-fluoro-4- (1-isopropyl-2)-methyl-1H-imidazo [4,5-b]Pyridin-6-yl) pyrimidine- 2-yl) amino) pyridin-3-yl) methyl) piperazine-1-carboxylic acid ester (1c)
6- (2-chloro-5-fluoropyrimidin-4-yl) -1-isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridine (intermediate A) (0.045g,0.15mmol), tert-butyl 4- ((6-aminopyridin-3-yl) methyl) piperazine-1-carboxylate (1b) (0.055g,0.18mmol), Pd2(dba)3(0.008g,0.015mmol), Xantphos (0.01g,0.015mmol) and Cs2CO3(0.103g,0.3mmol) in 1, 4-dioxane (3mL) under nitrogen stirring at 90 ℃ for 1 h. The mixture was cooled to room temperature and diluted with DCM (10 mL). Filtered through celite. The filtrate was concentrated and the residue was purified by preparative thin layer chromatography (eluent: DCM/MeOH ═ 20:1) to give tert-butyl 4- ((6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methyl) piperazine-1-carboxylic acid ester (1 c). MS-ESI (M/z):562[ M + 1]]+
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) -N- (5- (piperazin-1-ylmethyl) Yl) pyridin-2-yl) pyrimidin-2-amine (1)
Tert-butyl 4- ((6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]A mixture of pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methyl) piperazine-1-carboxylate (1c) (0.03g,0.05mmol) in TFA (2mL) and DCM (4mL) was stirred at room temperature for 2 hours. The solvent was evaporated under vacuum and the residue was diluted with water (10mL) and washed with DCM (10 mL). The pH of the water layer is adjusted to 11-12 by NaOH (2N). Extract with IPA/DCM (25%, 4X 20 mL). The extract was washed with saturated brine (40mL), Na2SO4Dried and concentrated. The residue was purified by preparative thin layer chromatography (eluent: DCM/MeOH ═ 10:1) to give 5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) -N- (5- (piperazin-1-ylmethyl) pyridin-2-yl) pyrimidin-2-amine (1). MS-ESI (M/z):462[ M + 1[ ]]+
Example 2
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) -N- (5- ((4-methylpiperazine- 1-yl) methyl) pyridine-2-yl) pyrimidin-2-amine (2)
Figure BDA0001755256780000491
5- ((4-Methylpiperazin-1-yl) methyl) pyridin-2-amine (2a)
The title compound 5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-amine (2a) was prepared according to the synthesis of 1b substituting 1-Boc-piperazine with 1-methylpiperazine. MS-ESI (M/z):207[ M + 1]]+
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) -N- (5- ((4-methylpiperazine- 1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine (2)
The title compound 5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b) was prepared according to the synthesis of 1c, substituting tert-butyl 4- ((6-aminopyridin-3-yl) methyl) piperazine-1-carboxylate (1b) with 5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-amine (2a)]Pyridin-6-yl) -N- (5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine (2). MS-ESI (M/z):476[ M +1 [)]+
Example 3
N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazole Azolo [4,5-b ] s]Pyridin-6-yl) pyrimidin-2-amine (3)
Figure BDA0001755256780000492
5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-amine (3a)
The title compound 5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-amine (3a) was prepared according to the synthetic procedure of 1b substituting 1-Boc-piperazine with 1-ethylpiperazine. MS-ESI (M/z) 221[ M + 1]]+
N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H' Mimi Azolo [4,5-b ] s]Pyridin-6-yl) pyrimidin-2-amine (3)
The title compound N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b) was prepared according to the synthesis of 1c by replacing tert-butyl 4- ((6-aminopyridin-3-yl) methyl) piperazine-1-carboxylate (1b) with 5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-amine (3a)]Pyridin-6-yl) pyrimidin-2-amine (3). MS-ESI (M/z) 490[ M + 1]]+
Example 4
(S) -5-fluoro-N- (5- ((hexahydropyrrolo [1, 2-a)]Pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (1-iso-butyl) pyridine propyl-2-methyl-1H-imidazo [4,5-b]Pyridin-6-yl) pyrimidin-2-amine (4)
Figure BDA0001755256780000501
(tert-Butoxycarbonyl) -L-proline (4a)
(tert-Butoxycarbonyl) -L-proline (4a) was prepared as described in tetrahedron.2013,10,156.
Tert-butyl (S) -2- ((2-ethoxy-2-oxoethyl) carbamoyl) pyrrolidine-1-carboxylic acid ester (4b)
To a solution of (tert-butoxycarbonyl) -L-proline (4a) (21.5g,100mmol) in THF (200mL) at 0 deg.C was added CDI (17.8g,110mmol) in portions. The mixture was stirred at room temperature for 0.5 hour. Ethyl glycinate hydrochloride (15.3g,110mmol) was added at 0 ℃ followed by TEA (15.0g,150mmol) and the mixture was stirred at room temperature overnight. The solvent was spun dry in vacuo and the residue was diluted with ethyl acetate (250mL), followed by saturated NaHCO3The solution (200mL), 10% citric acid (2X 200mL) and saturated brine (200mL) were washed with Na2SO4Drying and concentration gave the title compound tert-butyl (S) -2- ((2-ethoxy-2-oxoethyl) carbamoyl) pyrrolidine-1-carboxylate (4 b).
(S) -prolyl glycine ethyl ester trifluoroacetate salt (4c)
A mixture of tert-butyl (S) -2- ((2-ethoxy-2-oxoethyl) carbamoyl) pyrrolidine-1-carboxylate (4b) (24.8g,82.6mmol) and TFA (100mL) in DCM (125mL) was stirred at room temperature for 2h and concentrated to give the crude (S) -prolylglycine ethyl trifluoroacetate (4c) which was used directly in the next reaction. MS-ESI (M/z) 201[ M + 1]]+
(S) -hexahydropyrrolo [1,2-a ] compounds]Pyrazine-1, 4-diones (4d)
A mixture of (S) -prolylglycine ethyl trifluoroacetate (4c) (40g,127mmol) in TEA (150mL) and MeOH (800mL) was refluxed overnight. The solvent was spun dry in vacuo and the residue was diluted with IPA (100 mL). Filtration and washing of the filter cake with n-hexane. Vacuum drying the solid to obtain (S) -hexahydropyrrolo [1,2-a]Pyrazine-1, 4-dione (4 d). MS-ESI (M/z) 154[ M + 1]]+
Benzyl (S) -hexahydropyrrolo [1,2-a]Pyrazine-2 (1H) -carboxylic acid esters (4e)
To a suspension of LAH (5.9g,156mmol) in THF (60mL) was added dropwise (S) -hexahydropyrrolo [1,2-a ] solution]A solution of pyrazine-1, 4-dione (4d) (4.0g,26mmol) in THF (40mL) was refluxed for 16 hours. The reaction mixture was cooled to 0 ℃ and saturated NaHCO was added dropwise3(100mL) and benzyl chloroformate (6.67g,39mmol) was added. The mixture was stirred at 0 ℃ for 1 hour and at room temperature for a further 2 hours. The reaction solution was extracted with chloroform (3X 200mL), and the extract was washed with saturated brine (400mL), Mg2SO4And (5) drying. Concentrated and the residue purified by column chromatography over silica gel (eluent: DCM/MeOH 25:1) to give benzyl (S) -hexahydropyrrolo [1,2-a ] benzene]Pyrazine-2 (1H) -carboxylate (4 e). MS-ESI (M/z):261[ M + 1]]+
(S) -octahydropyrrolo [1, 2-a)]Pyrazine (4f)
Benzyl (S) -hexahydropyrrolo [1,2-a]A mixture of pyrazine-2 (1H) -carboxylate (4e) (5.0g,19mmol) and Pd/C (10%, 1.0g) in MeOH (50mL) was stirred under hydrogen at room temperature for 2H. The mixture was filtered through celite, and the filtrate was concentrated to give (S) -octahydropyrrolo [1,2-a ]]Crude pyrazine (4 f). MS-ESI (M/z) 127[ M + 1]]+
(S) -2- ((6-bromopyridin-3-yl) methyl) octahydropyrrolo [1, 2-a)]Pyrazine (4g)
Following the synthesis of 1a, 1-Boc-piperazine was replaced with (S) -octahydropyrrolo [1,2-a ]]Pyrazine (4f) preparation of the title compound (S) -2- ((6-bromopyridin-3-yl) methyl) octahydropyrrolo [1, 2-a)]Pyrazine (4 g). MS-ESI (M/z):296,298[ M + 1]]+
(S) -5- ((hexahydropyrrolo [1, 2-a)]Pyrazin-2 (1H) -yl) methyl) pyridin-2-amine (4H)
Replacement of tert-butyl 4- ((6-bromopyridin-3-yl) methyl) piperazine-1-carboxylate (1a) with (S) -2- ((6-bromopyridin-3-yl) methyl) octahydropyrrolo [1,2-a ] according to the Synthesis procedure of 1b]Pyrazine (4g) gave the title compound (S) -5- ((hexahydropyrrolo [1, 2-a)]Pyrazin-2 (1H) -yl) methyl) pyridin-2-amine (4H). MS-ESI (M/z):233[ M +1 [)]+
(S) -5-fluoro-N- (5- ((hexahydropyrrolo [1, 2-a)]Pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (1-iso-butyl) pyridine propyl-2-methyl-1H-imidazo [4,5-b]Pyridin-6-yl) pyrimidin-2-amine (4)
Tert-butyl 4- ((6-aminopyridin-3-yl) methyl) piperazine-1-carboxylate (1b) was replaced with (S) -5- ((hexahydropyrrolo [1, 2-a) according to the synthetic method of 1c]Pyrazin-2 (1H) -yl) methyl) pyridin-2-amine (4H) and the title compound (S) -5-fluoro-N- (5- ((hexahydropyrrolo [1, 2-a)]Pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridin-6-yl) pyrimidin-2-amine (4). MS-ESI (M/z):502[ M +1 [)]+
Example 5
(R) -5-fluoro-N- (5- ((hexahydropyrrolo [1, 2-a)]Pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (1- isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridin-6-yl) pyrimidin-2-amine (5)
Figure BDA0001755256780000511
(R) -5- ((hexahydropyrrolo [1, 2-a)]Pyrazin-2 (1H) -yl) carboxamidesYl) pyridin-2-Amines (5a)
The title compound (R) -5- ((hexahydropyrrolo [1, 2-a) was prepared according to the 4h synthesis substituting the starting L-proline for D-proline]Pyrazin-2 (1H) -yl) methyl) pyridin-2-amine (5 a). MS-ESI (M/z):233[ M +1 [)]+
(R) -5-fluoro-N- (5- ((hexahydropyrrolo [1, 2-a)]Pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (1- isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridin-6-yl) pyrimidin-2-amine (5)
Tert-butyl 4- ((6-aminopyridin-3-yl) methyl) piperazine-1-carboxylate (1b) was replaced with (R) -5- ((hexahydropyrrolo [1, 2-a) according to the synthetic method of 1c]Pyrazin-2 (1H) -yl) methyl) pyridin-2-amine (5a), the title compound (R) -5-fluoro-N- (5- ((hexahydropyrrolo [1, 2-a)]Pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridin-6-yl) pyrimidin-2-amine (5). MS-ESI (M/z):502[ M +1 [)]+
Example 6
N- (5- (((1S,4S) -2, 5-diazabicyclo [ 2.2.1)]Heptane-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) pyrimidin-2-amine (6)
Figure BDA0001755256780000521
Tert-butyl (1S,4S) -5- ((6-bromopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Heptane-2-carboxylic acid Acid ester (6a)
To tert-butyl (1S,4S) -2, 5-diazabicyclo [2.2.1]To a solution of heptane-2-carboxylate (0.099g,0.5mmol), 5-bromo-2-pyridinecarboxaldehyde (0.093g,0.5mmol) and AcOH (0.03g,0.5mmol) in THF (1mL) was added sodium triacetoxyborohydride (0.159g,0.75 mmol). The reaction mixture was stirred at room temperature overnight. The mixture was saturated NaHCO3The solution was quenched, extracted with EtOAc, and the organic layer was washed with saturated brine (20mL), Na2SO4And (5) drying. Concentrating to obtain tert-butyl(1S,4S) -5- ((6-Bromopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Heptane-2-carboxylic acid ester (6 a). MS-ESI (M/z) 368,390[ M + 1]]+
Tert-butyl (1S,4S) -5- ((6-aminopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Heptane-2- Carboxylic acid ester (6b)
Tert-butyl (1S,4S) -5- ((6-bromopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Heptane-2-carboxylic acid ester (6a) (180mg,0.49mmol), NH3·H2O(4.5mL),Cu2A mixture of O (80.0mg,0.56mmol) in MeOH (1.5mL) was stirred in a sealed tube at 65 deg.C overnight. After cooling to room temperature, the mixture was filtered, and the filtrate was concentrated under reduced pressure. The residue was differentiated between DCM and NaOH (2N). The organic layer was separated, washed with saturated brine and Na2SO4Dried and concentrated, and the residue purified by column chromatography on silica gel (eluent: DCM: MeOH ═ 10:1) to give tert-butyl (1S,4S) -5- ((6-aminopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Heptane-2-carboxylic acid ester (6 b). MS-ESI (M/z):305[ M +1 [)]+
Tert-butyl (1S,4S) -5- ((6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridine-6- Yl) pyrimidin-2-yl) amino) pyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Heptane-2-carboxylate (6c)
Replacement of tert-butyl 4- ((6-aminopyridin-3-yl) methyl) piperazine-1-carboxylate (1b) with tert-butyl (1S,4S) -5- ((6-aminopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1] according to the Synthesis method of 1c]Heptane-2-carboxylic acid ester (6b), the title compound tert-butyl (1S,4S) -5- ((6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b) was prepared]Pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Heptane-2-carboxylic acid ester (6 c). MS-ESI (M/z):574[ M +1 [)]+
N- (5- (((1S,4S) -2, 5-diazabicyclo [ 2.2.1)]Heptane-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) pyrimidin-2-amine (6)
According to the synthesis method of 1, tert-butyl 4-((6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b))]Pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methyl) piperazine-1-carboxylate (1c) was replaced with tert-butyl (1S,4S) -5- ((6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Heptane-2-carboxylic acid ester (6c), the title compound N- (5- (((1S,4S) -2, 5-diazabicyclo [ 2.2.1) was prepared]Heptane-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridin-6-yl) pyrimidin-2-amine (6). MS-ESI (M/z):474[ M +1]+
Example 7
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) -N- (5- (((1S,4S) -5-) Methyl-2, 5-diazabicyclo [2.2.1]Heptane-2-yl) methyl) pyridin-2-yl) pyrimidin-2-amine (7)
Figure BDA0001755256780000531
(1S,4S) -2- ((6-Bromopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Heptane (7a)
To tert-butyl (1S,4S) -5- ((6-bromopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1 at room temperature]To a solution of heptane-2-carboxylate (6a) (0.81g,2.2mmol) in DCM (6mL) was added TFA (4 mL). The mixture was stirred for 2 hours. With Na2CO3The solution was adjusted to pH 8. The mixture was extracted with DCM (3X 6 mL). The extract was washed with water and saturated brine (5mL) in this order, and Na2SO4Drying and concentrating to give (1S,4S) -2- ((6-bromopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Heptane (7a) (0.534 g). MS-ESI (M/z) 268[ M + 1]]+
(1S,4S) -2- ((6-Bromopyridin-3-yl) methyl) -5-methyl-2, 5-diazabicyclo [2.2.1]Heptane (7b)
To (1S,4S) -2- ((6-bromopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1 at room temperature]To a solution of heptane (7a) (0.15g,0.56mmol) in DCM (2mL) was added formaldehyde (37%, 63 μ L) followed by NaBH (OAc)3(0.273g,1.12 mmol). The mixture was stirred vigorously for 30 minutes. The mixture was saturated with Na2CO3The solution was quenched, extracted with DCM (2X 5mL), and the extract was washed successively with water and saturated brine (5mL), Na2SO4And (5) drying. Filtered and concentrated. The residue was purified by silica gel column chromatography (eluent: DCM: MeOH ═ 10:1) to give (1S,4S) -2- ((6-bromopyridin-3-yl) methyl) -5-methyl-2, 5-diazabicyclo [2.2.1]Heptane (7 b). MS-ESI (M/z):282,284[ M + 1]]+
5- (((1S,4S) -5-methyl-2, 5-diazabicyclo [ 2.2.1)]Heptane-2-yl) methyl) pyridin-2-amine (7c)
According to the synthesis of 6b, tert-butyl (1S,4S) -5- ((6-bromopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Replacement of Heptane-2-carboxylate (6a) with (1S,4S) -2- ((6-bromopyridin-3-yl) methyl) -5-methyl-2, 5-diazabicyclo [2.2.1]Heptane (7b) to prepare the title compound 5- (((1S,4S) -5-methyl-2, 5-diazabicyclo [ 2.2.1)]Heptan-2-yl) methyl) pyridin-2-amine (7 c). MS-ESI (M/z) 219[ M + 1]]+
5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) -N- (5- (((1S,4S) -5-) Methyl-2, 5-diazabicyclo [2.2.1]Heptane-2-yl) methyl) pyridin-2-yl) pyrimidin-2-amine (7)
Replacement of tert-butyl 4- ((6-aminopyridin-3-yl) methyl) piperazine-1-carboxylate (1b) with 5- (((1S,4S) -5-methyl-2, 5-diazabicyclo [ 2.2.1) according to the Synthesis method of 1c]Heptane-2-yl) methyl) pyridin-2-amine (7c) the title compound 5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b) was prepared]Pyridin-6-yl) -N- (5- (((1S,4S) -5-methyl-2, 5-diazabicyclo [ 2.2.1)]Heptane-2-yl) methyl) pyridin-2-yl) pyrimidin-2-amine (7). MS-ESI (M/z):488[ M + 1]]+
Example 8
N- (5- (((1S,4S) -5-ethyl-2, 5-diazabicyclo [ 2.2.1)]Heptane-2-yl) methyl) pyridin-2-yl) - 5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) pyrimidin-2-amine (8)
Figure BDA0001755256780000541
(1S,4S) -2- ((6-Bromopyridin-3-yl) methyl) -5-ethyl-2, 5-diazabicyclo [2.2.1]Heptane (8a)
To (1S,4S) -2- ((6-bromopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1 at room temperature]To a solution of heptane (7a) (0.10g,0.37mmol) and TEA (60. mu.L, 0.41mmol) in DMF (1mL) was added dropwise ethyl bromide (27. mu.L, 0.33 mol). The mixture was stirred for 3.5 hours. Extract with water (5mL) and DCM (5 mL). The extract was washed with water (2mL) and saturated brine (2mL) in this order, and then with Na2SO4Dried and concentrated. The residue was purified by silica gel column chromatography (eluent: DCM: MeOH ═ 10:1) to give (1S,4S) -2- ((6-bromopyridin-3-yl) methyl) -5-ethyl-2, 5-diazabicyclo [2.2.1]Heptane (8 a). MS-ESI (M/z):296,298[ M + 1]]+
5- (((1S,4S) -5-Ethyl-2, 5-diazabicyclo [ 2.2.1)]Heptane-2-yl) methyl) pyridin-2-amine (8b)
According to the synthesis of 6b, tert-butyl (1S,4S) -5- ((6-bromopyridin-3-yl) methyl) -2, 5-diazabicyclo [2.2.1]Replacement of Heptane-2-carboxylate (6a) with (1S,4S) -2- ((6-bromopyridin-3-yl) methyl) -5-ethyl-2, 5-diazabicyclo [2.2.1]Heptane (8a) to prepare the title compound 5- (((1S,4S) -5-ethyl-2, 5-diazabicyclo [ 2.2.1)]Heptane-2-yl) methyl) pyridin-2-amine (8 b). MS-ESI (M/z):233[ M +1 [)]+
N- (5- (((1S,4S) -5-ethyl-2, 5-diazabicyclo [ 2.2.1)]Heptane-2-yl) methyl) pyridin-2-yl) - 5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b)]Pyridin-6-yl) pyrimidin-2-amine (8)
Replacement of tert-butyl 4- ((6-aminopyridin-3-yl) methyl) piperazine-1-carboxylate (1b) with 5- (((1S,4S) -5-ethyl-2, 5-diazabicyclo [ 2.2.1) according to the Synthesis method of 1c]Heptane-2-yl) methyl) pyridin-2-amine (8b) to prepare the title compound N- (5- (((1S,4S) -5-ethyl-2, 5-diazabicyclo [ 2.2.1)]Heptane-2-yl) methyl) pyridin-2-yl) -5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b]Pyridine compound-6-yl) pyrimidin-2-amine (8). MS-ESI (M/z):488[ M + 1]]+
Examples 9-81 listed in Table 1 are prepared essentially according to the same procedures as examples 1-8, or using similar synthetic strategies or procedures, using starting materials that are either commercially available or are prepared according to literature procedures and mutatis mutandis. The names and structures of examples 9-81 are given in Table 1.
TABLE 1
Figure BDA0001755256780000551
Figure BDA0001755256780000561
Figure BDA0001755256780000571
Figure BDA0001755256780000581
Figure BDA0001755256780000591
Figure BDA0001755256780000601
Figure BDA0001755256780000611
Figure BDA0001755256780000621
Figure BDA0001755256780000631
Figure BDA0001755256780000641
Example 83
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3, 4-b)]Pyridin-5-yl) -N- (5- (piperazin-1-ylmethyl) Yl) pyridin-2-yl) pyrimidin-2-amine (83)
Figure BDA0001755256780000642
Tert-butyl 4- ((6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3, 4-b)]Pyridin-5-yl) pyrimidine- 2-yl) amino) pyridin-3-yl) methyl) piperazine-1-carboxylic acid ester (83a)
The title compound tert-butyl 4- ((6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3, 4-B) was prepared according to the synthesis of 1c, reactants tert-butyl 4- ((6-aminopyridin-3-yl) methyl) piperazine-1-carboxylate (1B) and 5- (2-chloro-5-fluoropyrimidin-4-yl) -7-fluoro-3-isopropyl-1-methyl-3 a,7 a-dihydro-1H-indazole (intermediate B)]Pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methyl) piperazine-1-carboxylic acid ester (83 a). MS-ESI (M/z):562[ M + 1]]+
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3, 4-b)]Pyridin-5-yl) -N- (5- (piperazin-1-ylmethyl) Yl) pyridin-2-yl) pyrimidin-2-amine (83)
According to the synthesis method of 1, tert-butyl 4- ((6- ((5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4, 5-b))]Pyridin-6-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methyl) piperazine-1-carboxylate (1c) was replaced with tert-butyl 4- ((6- ((5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3, 4-b)]Pyridin-5-yl) pyrimidin-2-yl) amino) pyridin-3-yl) methyl) piperazine-1-carboxylic acid ester (83a) the title compound 5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3, 4-b) was prepared]Pyridin-5-yl) -N- (5- (piperazin-1-ylmethyl)) Pyridin-2-yl) pyrimidin-2-amine (83). MS-ESI (M/z):462[ M + 1[ ]]+
Example 84
5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3, 4-b)]Pyridin-5-yl) -N- (5- ((4-methylpiperazine- 1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine (84)
Figure BDA0001755256780000651
To 5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3, 4-b) at room temperature]Pyridin-5-yl) -N- (5- (piperazin-1-ylmethyl) pyridin-2-yl) pyrimidin-2-amine (83) (17.8mg,0.0386mmol), NaBH (OAc)3To a solution of (12.3mg,0.0579mmol) in DCE (1mL) was added dropwise HCHO (37% aqueous solution) (2 drops), and the mixture was stirred at room temperature for 4 hours. NaHCO for the mixture3The solution was diluted and extracted with DCM. Extract Na2SO4Dried and concentrated. Purifying the residue by preparative thin layer chromatography to obtain 5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3, 4-b)]Pyridin-5-yl) -N- (5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine (84). MS-ESI (M/z):476[ M +1 [)]+
Example 85
N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyri-dine Azolo [3,4-b]Pyridin-5-yl) pyrimidin-2-amine (85)
Figure BDA0001755256780000652
To 5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3, 4-b) at room temperature]Pyridin-5-yl) -N- (5- (piperazin-1-ylmethyl) pyridin-2-yl) pyrimidin-2-amine (83) (17.8mg,0.0386mmol), NaBH (OAc)3To a solution of (12.3mg,0.0579mmol) in DCE (1mL) was added dropwise MeCHO (40% aqueous solution) (2 drops), and the mixture was stirred at room temperature for 4 hours. NaHCO for the mixture3The solution was diluted and extracted with DCM. Extract Na2SO4Dried and concentrated. Disabled personThe retentate was purified by preparative thin layer chromatography to give N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3, 4-b)]Pyridin-5-yl) pyrimidin-2-amine (85). MS-ESI (M/z) 490[ M + 1]]+
Example 86
5-fluoro-4- (3-isopropyl-1-methyl-1H-pyrazolo [3, 4-b)]Pyridin-5-yl) -N- (5- ((4-methylpiperazine- 1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine (86)
Figure BDA0001755256780000661
According to the synthesis of 1c, the reactant is 5- (2-chloro-5-fluoropyrimidin-4-yl) -3-isopropyl-1-methyl-1H-pyrazolo [3,4-b]Pyridine (intermediate C) and 5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-amine (2a) to prepare the title compound 5-fluoro-4- (3-isopropyl-1-methyl-1H-pyrazolo [3, 4-b)]Pyridin-5-yl) -N- (5- ((4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine (86). MS-ESI (M/z):476[ M +1 [)]+
Example 87
N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-1-methyl-1H-pyri-dine Azolo [3,4-b]Pyridin-5-yl) pyrimidin-2-amine (87)
Figure BDA0001755256780000662
According to the synthesis of 1c, the reactant is 5- (2-chloro-5-fluoropyrimidin-4-yl) -3-isopropyl-1-methyl-1H-pyrazolo [3,4-b]Pyridine (intermediate C) and 5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-amine (3a) to prepare the title compound N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-1-methyl-1H-pyrazolo [3, 4-b)]Pyridin-5-yl) pyrimidin-2-amine (87). MS-ESI (M/z):476[ M +1 [)]+
Examples 88-156 listed in Table 2 were prepared using the methods described in examples 83-87, by substituting the corresponding aminopyridine and making the necessary modifications, or using similar synthetic strategies or methods.
TABLE 2
Figure BDA0001755256780000671
Figure BDA0001755256780000681
Figure BDA0001755256780000691
Figure BDA0001755256780000701
Figure BDA0001755256780000711
Figure BDA0001755256780000721
Figure BDA0001755256780000731
Figure BDA0001755256780000741
Figure BDA0001755256780000751
Cell proliferation assay
To test the in vitro inhibitory effect of compounds on CDK4/6 kinase, a BE (2) -C cell-based assay was established. In the present method, the inhibitory activity of a compound against CDK4/6 is determined by measuring the inhibitory effect on the proliferation of BE (2) -C cells. BE (2) -C cells were seeded into 96-well culture plates at 5000/well, and 150. mu.l of the medium was added per well. Compound dilution: each compound was diluted to 20mM with DMSO as a stock solution, and on the day of addition, a working solution of 4-fold final concentration was prepared on the medium. After 24 hours of BE (2) -C cell inoculation, 50. mu.l of compound working solution (duplicate wells for each compound) was added to each well. After 72 hours of incubation, the cell proliferation activity was measured by the MTS method.
The prepared compounds were tested according to the biological activity test method described above. The results are shown in Table 3.
TABLE 3
Examples IC50(nM) Examples IC50(nM) Examples IC50(nM) Examples IC50(nM)
1 857 49 728 91 574 126 192
2 343 54 58 92 124 129 464
3 204 56 740 93 96 134 323
4 19 57 456 94 68 135 418
5 49 58 279 95 768 136 46
7 583 59 37 96 51 137 28
8 811 60 435 97 55 138 18
10 82 61 217 99 176 139 26
11 60 62 251 100 74 140 38
13 546 63 91 102 74 141 102
14 392 65 58 103 99 142 26
16 751 66 51 105 640 143 20
19 396 68 58 106 177 144 31
20 272 69 31 107 178 145 16
22 840 70 148 108 456 146 37
23 514 71 38 109 297 147 535
27 91 72 25 110 165 148 51
28 15 73 901 111 253 149 27
29 85 74 182 112 954 151 121
31 364 75 22 113 243 152 99
32 273 77 624 114 61 153 51
34 473 78 59 116 228 154 13
35 53 80 911 117 157 155 22
40 460 81 365 118 463 156 35
41 332 84 64 119 42 157 30
43 662 85 19 120 46 159 44
44 440 88 994 121 489 160 22
46 420 88 85 122 433
47 152 89 28 125 309

Claims (20)

1. A compound of formula (I) or (II):
Figure FDA0003057138630000011
or a pharmaceutically acceptable salt thereof, wherein,
q is pyridine;
each R1Is hydrogen;
each R2Is hydrogen;
each R3Is independently selected from C1-10An alkyl group;
each R4Is halogen;
each R5Independently selected from methyl, ethyl,
Figure FDA0003057138630000012
Which is unsubstituted or substituted by at least one member independently selected from RXSubstituted with the substituent(s);
each RXIndependently selected from C1-10Alkyl, wherein each alkyl is independently unsubstituted or substituted with 1 independently selected from RYSubstituted with the substituent(s);
each RYIndependently selected from- (CR)c2Rd2)tORb2
Each Rb2Independently selected from C1-10An alkyl group;
m is 1;
n is selected from 0,1 and 2;
p is 2;
t is 0;
with the proviso that the compound of the formula (I) is not
Figure FDA0003057138630000021
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein at least one means 1,2,3 or 4.
3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, having the formula
Figure FDA0003057138630000022
(I)。
4. The compound of claim 1, or a pharmaceutically acceptable salt thereof, having the formula
Figure FDA0003057138630000023
(II)。
5. The compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof, wherein Q is
Figure FDA0003057138630000024
6. The compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof, wherein each R5Is independently selected from
Figure FDA0003057138630000025
Which is unsubstituted or substituted with at least one, independently selected from RXIs substituted with the substituent(s).
7. The compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof, wherein R5Is that
Figure FDA0003057138630000026
Which is unsubstituted or substituted by at least one member independently selected from RXIs substituted with the substituent(s).
8. A compound of formula (I) or (II):
Figure FDA0003057138630000027
Figure FDA0003057138630000031
or a pharmaceutically acceptable salt thereof, wherein,
q is
Figure FDA0003057138630000032
Each R1Is hydrogen;
each R2Is hydrogen;
each R3Is independently selected from C1-10An alkyl group;
each R4Is halogen;
each R5Independently selected from methyl, ethyl and
Figure FDA0003057138630000033
which is unsubstituted or substituted by at least one member independently selected from RXSubstituted with the substituent(s);
each RXIndependently selected from C1-10Alkyl, wherein each alkyl is independently unsubstituted or substituted with 1 independently selected from RYSubstituted with the substituent(s);
each RYIndependently selected from- (CR)c2Rd2)tORb2
Each Rb2Independently selected from C1-10An alkyl group;
m is 1;
n is selected from 0,1 and 2;
p is 2;
t is 0.
9. The compound of claim 8, or a pharmaceutically acceptable salt thereof, wherein at least one means 1,2,3 or 4.
10. The compound of claim 8, or a pharmaceutically acceptable salt thereof, having the formula
Figure FDA0003057138630000034
(I)。
11. The compound of claim 8, or a pharmaceutically acceptable salt thereof, having the formula
Figure FDA0003057138630000041
(II)。
12. The compound of any one of claims 1-4 or 8-11, or a pharmaceutically acceptable salt thereof, wherein each R3Independently selected from methyl, isopropyl and tert-butyl.
13. The compound of any one of claims 1-4 or 8-11, or a pharmaceutically acceptable salt thereof, wherein each R4Independently selected from fluorine.
14. The compound of any one of claims 1-4 or 8-11, or a pharmaceutically acceptable salt thereof, wherein each RXIs independently selected from C1-10Alkyl and methoxymethyl.
15. The compound of claim 14, or a pharmaceutically acceptable salt thereof, wherein RXIs independently selected from C1-10An alkyl group.
16. The compound of claim 14, or a pharmaceutically acceptable salt thereof, wherein each RXIndependently selected from methyl, ethyl and methoxymethyl.
17. A compound selected from
(S) -5-fluoro-N- (5- ((hexahydropyrrolo [1,2-a ] pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
(R) -5-fluoro-N- (5- ((hexahydropyrrolo [1,2-a ] pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-amine,
N- (5-fluoro-4- (1-isopropyl-2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) pyrimidin-2-yl) -6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
(S) -4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((4-ethyl-3- (methoxymethyl) piperazin-1-yl) methyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
4- (1- (tert-butyl) -2-methyl-1H-imidazo [4,5-b ] pyridin-6-yl) -N- (5- ((8-ethyl-3, 8-diazabicyclo [3.2.1] octan-3-yl) methyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
N- (5- ((4-ethylpiperazin-1-yl) methyl) pyridin-2-yl) -5-fluoro-4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
(R) -5-fluoro-N- (5- ((hexahydropyrrolo [1,2-a ] pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) -4- (3-isopropyl-2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) pyrimidin-2-amine,
(R) -4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- ((hexahydropyrrolo [1,2-a ] pyrazin-2 (1H) -yl) methyl) pyridin-2-yl) pyrimidin-2-amine,
4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -N- (5- ((3-ethyl-3, 8-diazabicyclo [3.2.1] octan-8-yl) methyl) pyridin-2-yl) -5-fluoropyrimidin-2-amine,
(S) -4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoro-N- (5- ((3- (methoxymethyl) -4-methylpiperazin-1-yl) methyl) pyridin-2-yl) pyrimidin-2-amine
N- (4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-yl) -6-methyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
N- (4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-yl) -6-ethyl-5, 6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine,
N- (4- (3- (tert-butyl) -2-methyl-2H-pyrazolo [3,4-b ] pyridin-5-yl) -5-fluoropyrimidin-2-yl) -6- (1-ethylpiperidin-4-yl) -5,6,7, 8-tetrahydro-1, 6-naphthyridin-2-amine
Or a pharmaceutically acceptable salt thereof.
18. A pharmaceutical composition comprising a compound of any one of claims 1-17, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
19. Use of a compound of any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 18, in the manufacture of a medicament for the treatment, amelioration or prevention of a condition that is responsive to inhibition of CDK4/6, wherein the compound or pharmaceutically acceptable salt thereof or pharmaceutical composition is optionally in combination with a second therapeutic agent.
20. Use of a compound of any one of claims 1-17, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 18, in the manufacture of a medicament for treating a cell proliferation disorder.
CN201780010075.XA 2016-02-06 2017-02-06 Kinase inhibitor Active CN108602799B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662292259P 2016-02-06 2016-02-06
US62/292,259 2016-02-06
PCT/CN2017/072958 WO2017133701A1 (en) 2016-02-06 2017-02-06 Certain protein kinase inhibitors

Publications (2)

Publication Number Publication Date
CN108602799A CN108602799A (en) 2018-09-28
CN108602799B true CN108602799B (en) 2021-08-03

Family

ID=59500239

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201780010075.XA Active CN108602799B (en) 2016-02-06 2017-02-06 Kinase inhibitor

Country Status (3)

Country Link
CN (1) CN108602799B (en)
TW (1) TWI736578B (en)
WO (1) WO2017133701A1 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107337634B (en) * 2017-08-28 2019-07-09 新发药业有限公司 A kind of preparation method of Abbe Seeley midbody compound
KR20200131246A (en) 2018-02-15 2020-11-23 누베이션 바이오 인크. Heterocyclic compounds as kinase inhibitors
UA126177C2 (en) 2018-04-26 2022-08-25 Пфайзер Інк. 2-amino-pyridine or 2-amino-pyrimidine derivatives as cyclin dependent kinase inhibitors
CA3138973A1 (en) * 2019-05-05 2020-11-12 Qilu Regor Therapeutics Inc. Cdk inhibitors
CN111303128B (en) * 2020-04-07 2020-10-27 苏州信诺维医药科技有限公司 Polycyclic compound, preparation method and application thereof
EP4214213A1 (en) * 2020-09-21 2023-07-26 Prelude Therapeutics, Incorporated Cdk inhibitors and their use as pharmaceuticals
TW202237585A (en) 2020-11-27 2022-10-01 瑞士商瑞森製藥公司 Cdk inhibitors
WO2022149057A1 (en) 2021-01-05 2022-07-14 Rhizen Pharmaceuticals Ag Cdk inhibitors
WO2023178547A1 (en) * 2022-03-23 2023-09-28 Prelude Therapeutics, Incorporated Polymorphic compounds and uses thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102264725A (en) * 2008-12-22 2011-11-30 伊莱利利公司 Protein kinase inhibitors
WO2016014904A1 (en) * 2014-07-24 2016-01-28 Beta Pharma, Inc. 2-h-indazole derivatives as cyclin-dependent kinase (cdk) inhibitors and therapeutic uses thereof
CN105294683A (en) * 2014-07-26 2016-02-03 广东东阳光药业有限公司 CDK small-molecule inhibitor compounds and application thereof
CN105732615A (en) * 2014-12-31 2016-07-06 山东轩竹医药科技有限公司 CDK kinase inhibitor

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2041132A2 (en) * 2006-07-06 2009-04-01 Boehringer Ingelheim International GmbH New compounds
WO2015101293A1 (en) * 2013-12-31 2015-07-09 山东轩竹医药科技有限公司 Kinase inhibitor and use thereof
CN105111191B (en) * 2015-07-21 2018-06-08 上海皓元生物医药科技有限公司 It is a kind of to be used to synthesize key intermediate of CDK9 inhibitor and its preparation method and application

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102264725A (en) * 2008-12-22 2011-11-30 伊莱利利公司 Protein kinase inhibitors
WO2016014904A1 (en) * 2014-07-24 2016-01-28 Beta Pharma, Inc. 2-h-indazole derivatives as cyclin-dependent kinase (cdk) inhibitors and therapeutic uses thereof
CN105294683A (en) * 2014-07-26 2016-02-03 广东东阳光药业有限公司 CDK small-molecule inhibitor compounds and application thereof
CN105732615A (en) * 2014-12-31 2016-07-06 山东轩竹医药科技有限公司 CDK kinase inhibitor

Also Published As

Publication number Publication date
TWI736578B (en) 2021-08-21
WO2017133701A1 (en) 2017-08-10
TW201801729A (en) 2018-01-16
CN108602799A (en) 2018-09-28

Similar Documents

Publication Publication Date Title
CN108602799B (en) Kinase inhibitor
CN110177788B (en) Compounds as BCL-2 selective apoptosis inducers
JP7416365B2 (en) Apoptosis inducer
KR101864589B1 (en) Certain protein kinase inhibitors
TW202309027A (en) Compound as a parp inhibitor
JP7294677B2 (en) Substituted (2-azabicyclo[3.1.0]hexane-2-yl)pyrazolo[1,5-a]pyrimidine compounds and substituted (2-azabicyclo[3.1.0]hexane-2 as TRK kinase inhibitors -yl)imidazo[1,2-b]pyridazine compounds
CN114008042B (en) Substituted pyrrolo [2,3-b ] pyridine and pyrazolo [3,4-b ] pyridine derivatives as protein kinase inhibitors
AU2019241260B2 (en) Macrocyclic compounds as TRK kinases inhibitors
CN115551856A (en) Compounds as CDK2/4/6 inhibitors
WO2021180107A1 (en) Compounds useful as kinase inhibitors
CN109153686B (en) Protein kinase inhibitor
CN112771047A (en) Substituted imidazo [1,2-a ] pyridine and [1,2,4] triazolo [1,5-a ] pyridine compounds as RET kinase inhibitors
CN109476666B (en) Compound as kinase inhibitor
TW202017927A (en) Substituted [1, 2, 4] triazolo [1, 5-a] pyridine compounds as ret kinase inhibitors
WO2021047584A1 (en) SUBSTITUTED (2-AZABICYCLO [3.1.0] HEXAN-2-YL) PYRAZOLO [1, 5-a] PYRIMIDINE AND IMIDAZO [1, 2-b] PYRIDAZINE COMPOUNDS AS TRK KINASES INHIBITORS
CN112771042A (en) Nalidinone and pyridylpyrimidinone compounds as protein kinase inhibitors
CN117321051A (en) Compounds as PARP inhibitors
CN115605481A (en) Compounds as kinase inhibitors
CN116568686A (en) Substituted pyrrolo [2,3-b ] pyridine and pyrazolo [3,4-b ] pyridine derivatives as protein kinase 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
TR01 Transfer of patent right

Effective date of registration: 20220822

Address after: 401121 building F, No. 2, Yangliu Road, Yubei District, Chongqing

Patentee after: Chongqing Fushang Yuanchuang Pharmaceutical Technology Co.,Ltd.

Address before: Room 512, A Building, 1289 Yishan Road, Xuhui District, Shanghai, 2003

Patentee before: CHINA SHANGHAI FOCHON PHARMACEUTICAL Co.,Ltd.

Patentee before: FOCHON PHARMACEUTICALS, LTD.

TR01 Transfer of patent right