CN116134029A - Oxo-nitrogen ring derivative regulator, preparation method and application thereof - Google Patents

Oxo-nitrogen ring derivative regulator, preparation method and application thereof Download PDF

Info

Publication number
CN116134029A
CN116134029A CN202180055582.1A CN202180055582A CN116134029A CN 116134029 A CN116134029 A CN 116134029A CN 202180055582 A CN202180055582 A CN 202180055582A CN 116134029 A CN116134029 A CN 116134029A
Authority
CN
China
Prior art keywords
alkyl
mmol
cycloalkyl
alkoxy
aryl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202180055582.1A
Other languages
Chinese (zh)
Inventor
董加强
邓欣贤
龚珍
肖华玲
包如迪
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.)
Jiangsu Hansoh Pharmaceutical Group Co Ltd
Shanghai Hansoh Biomedical Co Ltd
Original Assignee
Jiangsu Hansoh Pharmaceutical Group Co Ltd
Shanghai Hansoh Biomedical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jiangsu Hansoh Pharmaceutical Group Co Ltd, Shanghai Hansoh Biomedical Co Ltd filed Critical Jiangsu Hansoh Pharmaceutical Group Co Ltd
Publication of CN116134029A publication Critical patent/CN116134029A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • 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
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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

Abstract

An oxo-nitrogen ring derivative regulator, a preparation method and application thereof, and relates to an oxo-nitrogen ring derivative regulator, a preparation method and application thereof. In particular to a compound shown in a general formula (I), a preparation method thereof, a pharmaceutical composition containing the compound and application of the compound as a regulator in preparing medicines related to cancer, wherein each substituent in the general formula (I) is defined as in the specification.

Description

Oxo-nitrogen ring derivative regulator, preparation method and application thereof Technical Field
The invention belongs to the field of biological medicine, and in particular relates to an oxo-nitrogen ring derivative regulator, a preparation method and application thereof.
Background
CD73 is a class of cell surface glycosyl phosphatidylinositol-immobilized glycoproteins encoded by the NT5E gene, also known as extracellular-5' -nucleotidase, expressed on the surface of multiple cell types, including endothelial cells, lymphocytes, stromal cells, and multiple tumor cells. CD73 plays an important role in the conversion of intracellular AMP to adenosine. ATP (adenosine triphosphate) released by stressed or dead cells can provide an inflammatory signal, activating immunity. While extracellular ATP reacts via CD39 to form ADP (adenosine diphosphate) or AMP (adenosine monophosphate), which is converted from CD73 to adenosine, which passes through receptors expressed on different cell surfaces (A1, A2) A ,A2 B ,A3) The combination plays the physiological effects of immunosuppression, angiogenesis induction, mucosal hydration and the like.
Various researches show that CD73 is expressed on the surfaces of tumor cells such as rectal cancer, pancreatic cancer, non-small cell lung cancer, triple negative breast cancer, acute myelogenous leukemia and the like, and is also expressed on the surfaces of regulatory T cells, macrophages, dendritic cells and the like in the tumor microenvironment, so that the increase of the concentration of adenosine in the tumor microenvironment is promoted, and the adenosine passes through A2 on the surfaces of immune cells such as T cells A Or A2 B Binding, inhibiting proliferation and activation of T cells, and exerting immunosuppressive effects. Hypoxia and TGF-beta factors in the tumor environment can improve the expression level of CD73, TP53, KRAS, BRAF, EGFR and other gene mutations are also related to the high expression of CD73 in the tumor, and in addition, chemotherapeutic drugs, tyrosine kinase inhibitors, PD-1/PD-L1 inhibitors can up-regulate the expression of CD39 and CD 73. CD73 overexpression is thought to be associated with poor prognosis of cancer and drug resistance of certain drugs. Targeting CD73 can reduce adenosine formation in tumor microenvironment, thereby achieving immune activation effect, and can be combined with PD-1/PD-L1 inhibitor or other target inhibitors of adenosine pathway such as A2 A The combination of the inhibitors shows better synergistic effect of immune activation before clinic.
At present, a plurality of macromolecular and micromolecular CD73 inhibitors enter clinical researches, CD73 antibody medicaments such as BMS-986179 of BMS company and MEDI-9447 of MedImmune/AstraZeneca company enter clinical second phase, and single medicament or combined medicament is used for treating a plurality of solid tumors, so that certain curative effects are shown in early clinical results, and safety is good. The CD73 small molecule inhibitor has fast progress in AB-680 of Arcus company and LY-3475070 of Eli Lilly company, and has been developed in clinical stage one, and has not been published with curative effect data. There are also several companies developing preclinical studies.
In conclusion, the CD73 target has no available targeting drugs, and clinical single drugs or combined drugs have the potential of treating various solid tumors and have wide market prospect.
Disclosure of Invention
The invention aims to provide a compound shown in a general formula (I), a stereoisomer or a pharmaceutically acceptable salt thereof:
Figure PCTCN2021136249-APPB-000001
wherein:
ring a is selected from cycloalkyl, heterocyclyl, aryl or heteroaryl;
ring B is selected from cycloalkyl, heterocyclyl, aryl or heteroaryl;
R 1 Each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, - (CH) 2 ) n1 NR AA R BB 、-CR AA R BB (CH 2 ) n1 NR CC R DD 、-(CH 2 ) n1 R AA 、-CR AA R BB R CC 、-(CH 2 ) n1 OR AA 、-(CH 2 ) n1 C(O)OR AA 、-(CH 2 ) n1 OR AA 、-(CH 2 ) n1 SR AA 、-(CH 2 ) n1 NR AA C(O)(CH 2 ) n2 R BB 、-(CH 2 ) n1 NR AA C(O)OR BB 、-(CH 2 ) n1 NR AA C(O)NR BB R CC or-NR AA (CH 2 ) n1 R BB Said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl,Heterocyclyl, aryl, and heteroaryl, optionally further substituted;
R AA 、R BB 、R CC and R is DD Each independently selected from the group consisting of hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, optionally further substituted;
alternatively, R AA 、R BB 、R CC And R is DD And the nitrogen or carbon atoms to which they are attached are linked to form cycloalkyl, heterocyclyl, aryl or heteroaryl, which cycloalkyl, heterocyclyl, aryl and heteroaryl groups may optionally be further substituted;
R 2 Each independently selected from the group consisting of hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, alkyl, deuterated alkoxy, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, - (CH) 2 ) n3 NR A1 R B1 、-CR A1 R B1 (CH 2 ) n3 NR C1 R D1 、-(CH 2 ) n3 R A1 、-CR A1 R B1 R C1 、-(CH 2 ) n3 OR A1 、-(CH 2 ) n3 C(O)OR A1 、-(CH 2 ) n3 OR A1 、-(CH 2 ) n3 SR A1 、-(CH 2 ) n3 NR A1 C(O)(CH 2 ) n4 R B1 、-(CH 2 ) n3 NR A1 C(O)OR B1 、-(CH 2 ) n3 NR A1 C(O)NR B1 R C1 or-NR A1 (CH 2 ) n3 R B1 Said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl groups optionally being further substituted;
R A1 、R B1 、R C1 and R is D1 Each independently selected from the group consisting of hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, optionally further substituted;
alternatively, R A1 、R B1 、R C1 And R is D1 And the nitrogen or carbon atoms to which they are attached are linked to form cycloalkyl, heterocyclyl, aryl or heteroaryl, which cycloalkyl, heterocyclyl, aryl and heteroaryl groups may optionally be further substituted;
n1, n2, n3, n4 are each independently selected from 0, 1, 2, 3, 4, 5 or 6; and is also provided with
x, y are each independently selected from 0, 1, 2, 3 or 4. In a further preferred embodiment of the invention, ring A is selected from C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl;
preferably C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl;
more preferably, a six membered heterocyclic group containing nitrogen, oxygen or sulfur atoms;
further preferred, the oxo-nitrogen-containing six-membered heterocyclic group;
even more preferred
Figure PCTCN2021136249-APPB-000002
In a further preferred embodiment of the present invention, the compound is further represented by the general formula (I-A):
Figure PCTCN2021136249-APPB-000003
wherein:
ring C is selected from C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl;
preferably C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl;
more preferably C 3-6 Cycloalkyl;
further preferred are cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl or cyclohexenyl;
r is each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Deuterated alkoxy, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said amino, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in aryl and 5-14 membered heteroaryl;
preferably hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, said amino, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl;
y is selected from 0, 1, 2, 3 or 4;
z is selected from 0, 1, 2, 3, 4, 5 or 6; and is also provided with
m is selected from 0 or 1.
In a further preferred embodiment of the present invention, the compound is further represented by the general formula (II-A):
Figure PCTCN2021136249-APPB-000004
wherein:
-is a single bond or a double bond;
m1 is selected from 0, 1, 2, 3 or 4;
in a further preferred embodiment of the invention, ring B is selected from C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl;
preferably C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl;
more preferably, a 5-10 membered mono-heteroaryl or a 5-10 membered di-heteroaryl;
further preferred, a 5-10 membered mono-heteroaryl or a 5-10 membered di-heteroaryl containing nitrogen, oxygen or sulfur atoms;
even more preferably, a 5-6 membered nitrogen containing mono-heteroaryl or an 8-10 membered nitrogen containing di-heteroaryl;
still further preferably, the method comprises the steps of,
Ring B is selected from
Figure PCTCN2021136249-APPB-000005
X 1 Selected from-N-or-CH-, X 2 And X 3 Each independently selected from-N-or-C-, X 4 And X 5 Each independently selected from-N-, -NH-or-CH-,
Figure PCTCN2021136249-APPB-000006
is a single bond or a double bond;
alternatively, ring B is selected from
Figure PCTCN2021136249-APPB-000007
Y 1 、Y 2 、Y 5 、Y 6 Each independently selected from-N-or-CH-; y is Y 3 Selected from-N-, -NH-or-CH-; y is Y 4 Selected from-N-, -NH-, -CH-or-C (O) -,
Figure PCTCN2021136249-APPB-000008
is a single bond or a double bond;
most preferably, ring B is selected from
Figure PCTCN2021136249-APPB-000009
Figure PCTCN2021136249-APPB-000010
In a further preferred embodiment of the invention, the ring B is selected from
Figure PCTCN2021136249-APPB-000011
Figure PCTCN2021136249-APPB-000012
Figure PCTCN2021136249-APPB-000013
In a further preferred embodiment of the invention, R 1 Independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy groups、C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl, 5-14 membered heteroaryl, - (CH) 2 ) n1 NR AA R BB 、-CR AA R BB (CH 2 ) n1 NR CC R DD 、-(CH 2 ) n1 R AA 、-CR AA R BB R CC 、-(CH 2 ) n1 OR AA 、-(CH 2 ) n1 C(O)OR AA 、-(CH 2 ) n1 OR AA 、-(CH 2 ) n1 SR AA 、-(CH 2 ) n1 NR AA C(O)(CH 2 ) n2 R BB 、-(CH 2 ) n1 NR AA C(O)OR BB 、-(CH 2 ) n1 NR AA C(O)NR BB R CC or-NR AA (CH 2 ) n1 R BB The amino group, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heterologyOne or more substituents in the aryl group,
preferably, R 1 Independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl, 5-10 membered heteroaryl, - (CH) 2 ) n1 NR AA R BB 、-CR AA R BB (CH 2 ) n1 NR CC R DD 、-(CH 2 ) n1 R AA 、-CR AA R BB R CC 、-(CH 2 ) n1 OR AA 、-(CH 2 ) n1 C(O)OR AA 、-(CH 2 ) n1 OR AA 、-(CH 2 ) n1 SR AA 、-(CH 2 ) n1 NR AA C(O)(CH 2 ) n2 R BB 、-(CH 2 ) n1 NR AA C(O)OR BB 、-(CH 2 ) n1 NR AA C(O)NR BB R CC or-NR AA (CH 2 ) n1 R BB The amino group, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-10 membered heteroaryl, optionally further substituted with deuterium, halogen, amino,Nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl;
R AA 、R BB 、R CC and R is DD Each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said amino, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in the aryl and 5-14 membered heteroaryl groups,
preferably, R AA 、R BB 、R CC And R is DD Each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C 2-6 Alkyl, C 2-6 Deuterated alkyl, C 2-6 Haloalkyl, C 2-6 Hydroxyalkyl, C 2-6 Alkoxy, C 2-6 Haloalkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, said amino, C 2-6 Alkyl, C 2-6 Deuterated alkyl, C 2-6 Haloalkyl, C 2-6 Hydroxyalkyl, C 2-6 Alkoxy, C 2-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 2-6 Alkyl, C 2-6 Deuterated alkyl, C 2-6 Haloalkyl, C 2-6 Hydroxyalkyl, C 2-6 Alkoxy, C 2-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl;
alternatively, R AA 、R BB 、R CC And R is DD Any two of which are linked to the nitrogen or carbon atom to which they are attached to form C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy group,C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in the aryl and 5-14 membered heteroaryl groups,
preferably form C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl, said C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl.
In a further preferred embodiment of the invention, R 2 Each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Deuterated alkoxy, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl, 5-14 membered heteroaryl, - (CH) 2 ) n3 NR A1 R B1 、-CR A1 R B1 (CH 2 ) n3 NR C1 R D1 、-(CH 2 ) n3 R A1 、-CR A1 R B1 R C1 、-(CH 2 ) n3 OR A1 、-(CH 2 ) n3 C(O)OR A1 、-(CH 2 ) n3 OR A1 、-(CH 2 ) n3 SR A1 、-(CH 2 ) n3 NR A1 C(O)(CH 2 ) n4 R B1 、-(CH 2 ) n3 NR A1 C(O)OR B1 、-(CH 2 ) n3 NR A1 C(O)NR B1 R C1 or-NR A1 (CH 2 ) n3 R B1 Said amino, hydroxy, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Deuterated alkoxy, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Deuterated alkoxy, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl,
preferably, R 2 Independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl group、C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl, 5-10 membered heteroaryl, - (CH) 2 ) n3 NR A1 R B1 、-CR A1 R B1 (CH 2 ) n3 NR C1 R D1 、-(CH 2 ) n3 R A1 、-CR A1 R B1 R C1 、-(CH 2 ) n3 OR A1 、-(CH 2 ) n3 C(O)OR A1 、-(CH 2 ) n3 OR A1 、-(CH 2 ) n3 SR A1 、-(CH 2 ) n3 NR A1 C(O)(CH 2 ) n4 R B1 、-(CH 2 ) n3 NR A1 C(O)OR B1 、-(CH 2 ) n3 NR A1 C(O)NR B1 R C1 or-NR A1 (CH 2 ) n3 R B1 Said amino, hydroxy, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl or 5-10 membered heteroaryl;
R A1 、R B1 、R C1 and R is D1 Each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said amino, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl,
preferably, R A1 、R B1 、R C1 And R is D1 Each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or aryl radicals5-10 membered heteroaryl, said amino, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl or 5-10 membered heteroaryl;
alternatively, R A1 、R B1 、R C1 And R is D1 Any two of which are linked to the nitrogen or carbon atom to which they are attached to form C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl,
preferably form C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl, said C 3-8 Cycloalkyl, 3-8Membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in the aryl or 5-10 membered heteroaryl group.
In a further preferred embodiment of the present invention, the compound is further represented by the general formula (II):
Figure PCTCN2021136249-APPB-000014
wherein:
R 3 and R is 4 Each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said amino, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in aryl and 5-14 membered heteroaryl;
preferably hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, said amino, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl; more preferably hydrogen, deuterium, halogen, hydroxy, cyano, carboxy, oxo, thioxo, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl, said C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl;
more preferably hydrogen, deuterium, halogen, hydroxy, cyano, carboxy, oxo, thioxo, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl or 3-8 membered heterocyclyl, said C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl or 3-8 membered heterocyclyl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl;
further preferred are hydrogen, deuterium, halogen, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl or C 3-8 Cycloalkyl; the C is 1-6 Alkyl or C 3-8 Cycloalkyl optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Substituted by one or more substituents in aryl and 5-to 10-membered heteroaryl
More preferably hydrogen, halogen, C 1-3 Alkyl, C 1-3 Hydroxyalkyl, C 1-3 Deuterated alkyl, C 1-3 Haloalkyl, C 3-6 Cycloalkyl;
still more preferred are hydrogen, fluorine, chlorine, bromine, iodine, methyl, cyclopropyl, ethyl, propyl, isopropyl or trifluoromethyl.
In a further preferred embodiment of the invention, R 2 Independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl, C 1-3 Alkoxy, C 1-3 Haloalkoxy, C 3-6 Cycloalkyl, 3-6 membered heterocyclyl, C 6-10 Aryl or 5-6 membered heteroaryl;
preferably hydrogen, deuterium, halogen, oxo, thioxo, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C 1-3 Haloalkyl, C 1-3 Alkoxy, C 1-3 Haloalkoxy, C 3-6 Cycloalkyl, 3-6 membered heterocyclyl, C 6-10 Aryl or 5-6 membered heteroaryl;
more preferably hydrogen, halogen, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Haloalkyl, C 3-6 Cycloalkyl;
further preferred are hydrogen, fluorine, chlorine, bromine, -CD 3 Methyl, ethyl, cyclopropyl;
y is selected from 1, 2 or 3, preferably 1.
In a further preferred embodiment of the present invention, the compound is further represented by the general formula (II-1) or the general formula (II-2):
Figure PCTCN2021136249-APPB-000015
When ring B is selected from
Figure PCTCN2021136249-APPB-000016
And R is 2 When methyl, R 3 Is not isopropyl.
In a further preferred embodiment of the present invention, the compound is further represented by the general formula (IV), the general formula (VI-1) or the general formula (VI-2):
Figure PCTCN2021136249-APPB-000017
wherein:
ring B is selected from 5-10 membered mono-heteroaryl or 5-10 membered di-heteroaryl;
preferably a 5-6 membered nitrogen containing mono-heteroaryl or an 8-10 membered nitrogen containing di-heteroaryl;
more preferably, the method further comprises the steps of,
ring B is selected from
Figure PCTCN2021136249-APPB-000018
X 1 Selected from-N-or-CH-, X 2 And X 3 Each independently selected from-N-or-C-, X 4 And X 5 Each independently selected from-N-, -NH-or-CH-,
Figure PCTCN2021136249-APPB-000019
is a single bond or a double bond;
alternatively, ring B is selected from
Figure PCTCN2021136249-APPB-000020
Y 1 、Y 2 、Y 5 、Y 6 Each independently selected from-N-or-CH-; y is Y 3 Selected from-N-, -NH-, -CH-or-CH 2 -;Y 4 Selected from-N-, -NH-, -CH 2 -or-C (O) -,
Figure PCTCN2021136249-APPB-000021
is a single bond or a double bond;
most preferably
Figure PCTCN2021136249-APPB-000022
Figure PCTCN2021136249-APPB-000023
Ring D is selected from C 3-6 Cycloalkyl, 3-6 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl;
preferably, ring D is selected from cyclopropyl or cyclobutyl;
R 2 independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy or C 1-6 Haloalkoxy groups;
preferably hydrogen, deuterium, halogen, amino, oxo, thio, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C 1-3 Haloalkyl, C 1-3 Alkoxy or C 1-3 Haloalkoxy groups;
more preferably hydrogen, halogen, amino, oxo, thioxo, methyl, methoxy or-CD 3;
y is selected from 0, 1, 2 or 3.
In a further preferred embodiment of the invention, the compound is further represented by the general formula (III-A), (III-B), (III-C), (III-D) or (III-E):
Figure PCTCN2021136249-APPB-000024
wherein:
R 5 selected from C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl, said C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocycleRadical, C 6-10 One or more substituents in aryl or 5-10 membered heteroaryl;
preferably, R 5 Selected from C 3-6 Cycloalkyl or 3-6 membered heterocyclyl, said C 3-6 Cycloalkyl and 3-6 membered heterocyclyl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl, C 1-3 Alkoxy, C 1-3 Haloalkoxy and C 3-6 One or more substituents in cycloalkyl are substituted;
R 6 、R 7 R 8 each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl or C 2-6 Alkynyl; preferably, R 6 、R 7 R 8 Each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl, C 1-3 Alkoxy or C 1-3 Haloalkoxy groups.
In a further preferred embodiment of the present invention, the compound is further represented by the general formula (III-C):
Figure PCTCN2021136249-APPB-000025
R 5 selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl or cyclohexenyl;the cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl and cyclohexenyl radicals being optionally further substituted by halogen, methyl, ethyl, n-propyl, isopropyl, trifluoromethyl, cyclopropyl, cyclobutyl, cyclopropylmethyl or-CH 2 F is substituted by one or more substituents;
R 6 、R 7 each independently selected from hydrogen, deuterium, fluorine, chlorine, bromine, amino, methyl, ethyl or trifluoromethyl.
In Sup>A further preferred embodiment of the invention, the compound is further represented by the general formulSup>A (IV-A-1), (IV-A-2), (IV-B-1), (IV-B-2), (IV-C-1), (IV-C-2), (IV-D-1) or (IV-D-2):
Figure PCTCN2021136249-APPB-000026
wherein:
R 9 selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl, said C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl or 5-10 membered heteroaryl;
R 6 、R 7 and R is 8 As described above.
In a further preferred embodiment of the invention, the specific structure of the compound is as follows:
Figure PCTCN2021136249-APPB-000027
Figure PCTCN2021136249-APPB-000028
Figure PCTCN2021136249-APPB-000029
Figure PCTCN2021136249-APPB-000030
Figure PCTCN2021136249-APPB-000031
Figure PCTCN2021136249-APPB-000032
Figure PCTCN2021136249-APPB-000033
Figure PCTCN2021136249-APPB-000034
Figure PCTCN2021136249-APPB-000035
Figure PCTCN2021136249-APPB-000036
Figure PCTCN2021136249-APPB-000037
Figure PCTCN2021136249-APPB-000038
Figure PCTCN2021136249-APPB-000039
Figure PCTCN2021136249-APPB-000040
The invention further provides an intermediate for synthesizing the compound disclosed by the invention, wherein the intermediate is shown as a general formula (A):
Figure PCTCN2021136249-APPB-000041
wherein: ring B, ring C, R, R 1 、R 2 Y, z, m and z are as defined above.
The invention further provides an intermediate for synthesizing the compound disclosed by the invention, wherein the intermediate is shown as a general formula (B):
Figure PCTCN2021136249-APPB-000042
wherein: ring B, R, R 1 、R 2 M1 and z are as defined in the above general formula (II-A).
The invention further provides an intermediate for synthesizing the compound disclosed by the invention, wherein the intermediate is shown as a general formula (C), a general formula (C-1) or a general formula (C-2):
Figure PCTCN2021136249-APPB-000043
wherein: ring B, ring D, R 2 And y is as defined above for formula (IV), formula (IV-1) or formula (IV-2).
The invention further provides an intermediate for synthesizing the compound disclosed by the invention, wherein the intermediate is shown as a general formula (D):
Figure PCTCN2021136249-APPB-000044
wherein: r is R 5 、R 6 And R 7 All as defined above for general formula (III-C).
The invention further provides a process for preparing a compound of the invention comprising the steps of:
Figure PCTCN2021136249-APPB-000045
wherein: the acid is selected from an organic acid or an inorganic acid, preferably the inorganic acid is selected from hydrochloric acid, sulfuric acid, nitric acid, hydrobromic acid, hydrofluoric acid, hydroiodic acid or phosphoric acid; the organic acid is selected from 2, 5-dihydroxybenzoic acid, 1-hydroxy-2-naphthoic acid, acetic acid, dichloroacetic acid, trichloroacetic acid, acetohydroxamic acid, adipic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, 4-aminobenzoic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclohexane sulfamic acid, camphorsulfonic acid, aspartic acid, camphoric acid, gluconic acid, glucuronic acid, glutamic acid, isoascorbic acid, lactic acid, malic acid, mandelic acid, pyroglutamic acid, tartaric acid, dodecylsulfuric acid, dibenzoyltartaric acid, ethane-1, 2-disulfonic acid, ethanesulfonic acid, formic acid, fumaric acid, galactonic acid, gentisic acid, glutaric acid, 2-ketoglutaric acid, glycolic acid, hippuric acid, isethionic acid, lactonic acid, ascorbic acid, aspartic acid, lauric acid, camphoric acid, maleic acid, malonic acid, methanesulfonic acid, 1, 5-naphthalenedisulfonic acid, naphthalene-2-sulfonic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, thiocyanic acid, pamoic acid, formic acid, undecylenic acid, trifluoroacetic acid, benzenesulfonic acid, p-toluenesulfonic acid or L-malic acid;
Ring B, ring C, R, R 1 、R 2 Y, m and z are as defined above.
The invention further provides a process for preparing a compound of the invention comprising the steps of:
Figure PCTCN2021136249-APPB-000046
wherein:
acid, ring B, R, R 1 、R 2 Y, m1 and z are as defined above.
The invention further provides a process for preparing a compound of the invention comprising the steps of:
Figure PCTCN2021136249-APPB-000047
wherein:
acid, ring B, ring D, R 2 And y are as defined above.
The invention further provides a process for preparing a compound of the invention comprising the steps of:
Figure PCTCN2021136249-APPB-000048
wherein:
acid, R 5 、R 6 And R 7 As defined above.
The invention further relates to a pharmaceutical composition comprising a therapeutically effective dose of a compound of any one of claims, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers or excipients.
The invention further relates to the application of the compound of the general formula, the stereoisomer or the pharmaceutically acceptable salt thereof or the pharmaceutical composition in preparing CD73 inhibitor medicines.
The invention further relates to application of the compound shown in the general formula, the stereoisomer or the pharmaceutically acceptable salt thereof or the pharmaceutical composition thereof in preparing medicaments for treating cancers and related diseases.
The present invention also relates to a method for the treatment and prevention and/or treatment of cancer and related diseases, which comprises administering to a patient a therapeutically effective dose of a compound represented by the general formula, a stereoisomer thereof or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
The invention also provides methods of treating disease conditions, including but not limited to conditions associated with CD73 inhibitors, using the compounds or pharmaceutical compositions of the invention.
The invention also relates to methods of treating cancer and related diseases in a mammal comprising administering to the mammal a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
The cancer according to the invention is selected from solid tumors, gliomas or other tumors, preferably colorectal cancer, bladder cancer, gastric cancer, thyroid cancer, esophageal cancer, head and neck cancer, brain cancer, glioblastoma, hepatocellular carcinoma, lung cancer, melanoma, myeloma, pancreatic cancer, renal cell carcinoma, cervical cancer, urothelial cancer, prostate cancer, ovarian cancer, breast cancer, leukemia or lymphoma.
Detailed description of the invention
Unless stated to the contrary, the terms used in the specification and claims have the following meanings.
The term "alkyl" refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group containing from 1 to 8 carbon atoms, more preferably an alkyl group containing from 1 to 6 carbon atoms, still more preferably an alkyl group containing from 1 to 4 carbon atoms, and most preferably an alkyl group containing from 1 to 3 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, n-heptyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl 5-methylhexyl, 2, 3-dimethylpentyl, 2, 4-dimethylpentyl, 2-dimethylpentyl, 3-dimethylpentyl, 2-ethylpentyl, 3-ethylpentyl, n-octyl, 2, 3-dimethylhexyl, 2, 4-dimethylhexyl, 2, 5-dimethylhexyl, 2-dimethylhexyl, 3-dimethylhexyl 4, 4-dimethylhexyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, n-nonyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2-diethylpentyl, n-decyl, 3-diethylhexyl, 2-diethylhexyl, and various branched isomers thereof. More preferred are lower alkyl groups containing 1 to 6 carbon atoms, and non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, and the like. The alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxy or carboxylate, with methyl, ethyl, isopropyl, t-butyl, haloalkyl, deuteroalkyl, alkoxy-substituted alkyl and hydroxy-substituted alkyl being preferred.
The term "cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring containing from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably from 3 to 8 carbon atoms, and even more preferably from 3 to 6 carbon atoms. Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like; polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups, preferably cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
Cycloalkyl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxyl, or carboxylate groups.
The term "heterocyclyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms in which one or more ring atoms are selected from nitrogen, oxygen or S (O) m (wherein m is an integer from 0 to 2), but does not include a ring moiety of-O-O-, -O-S-, or-S-S-, and the remaining ring atoms are carbon. Preferably containing 3 to 12 ring atoms, of which 1 to 4 are heteroatoms; more preferably from 3 to 10 ring atoms; further preferred contain 3 to 8 ring atoms. Non-limiting examples of monocyclic heterocyclyl groups include pyrrolidinyl, azetidinyl, oxetanyl, oxahexanyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothienyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrrolyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl, and the like; preferably, pyrrolidinyl, azetidinyl, oxetanyl, tetrahydrofuranyl, pyrazolidinyl, morpholinyl, piperazinyl and pyranyl; more preferred are pyrrolidinyl, azetidinyl, oxetanyl, oxahexanyl, piperidinyl, piperazinyl and pyranyl. Polycyclic heterocyclyl groups include spiro, fused and bridged heterocyclic groups; the heterocyclic groups of the spiro ring, the condensed ring and the bridged ring are optionally connected with other groups through single bonds, or are further connected with other cycloalkyl groups, heterocyclic groups, aryl groups and heteroaryl groups through any two or more atoms on the ring in a parallel ring mode.
The heterocyclic group may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxyl, or carboxylate groups.
The term "aryl" refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic (i.e., rings sharing adjacent pairs of carbon atoms) group having a conjugated pi-electron system, preferably 6 to 10 membered, more preferably 6 to 8 membered, such as phenyl and naphthyl. More preferably phenyl.
Aryl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
The term "heteroaryl" refers to a heteroaromatic system containing from 1 to 4 heteroatoms, from 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen. Heteroaryl is preferably 5 to 10 membered, more preferably 5 to 8 membered, most preferably 5 or 6 membered, such as imidazolyl, furyl, thienyl, thiazolyl, pyrazolyl, oxazolyl, pyrrolyl, triazolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl, and the like, preferably triazolyl, thienyl, imidazolyl, pyrazolyl or pyrimidinyl, thiazolyl; more preferably triazolyl, pyrrolyl, thienyl, thiazolyl and pyrimidinyl. The heteroaryl ring may be fused to an aryl, heterocyclyl, or cycloalkyl ring, wherein the ring attached to the parent structure is a heteroaryl ring, non-limiting examples of which include:
Figure PCTCN2021136249-APPB-000049
Heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
The term "alkoxy" refers to-O- (alkyl) and-O- (unsubstituted cycloalkyl) wherein alkyl is as defined above, preferably alkyl having 1 to 8 carbon atoms, more preferably alkyl having 1 to 6 carbon atoms, and most preferably alkyl having 1 to 3 carbon atoms. Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentoxy, cyclohexyloxy. The alkoxy groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl, or carboxylate groups.
"haloalkyl" refers to an alkyl group substituted with one or more halogens, where alkyl is as defined above.
"haloalkoxy" refers to an alkoxy group substituted with one or more halogens, wherein the alkoxy group is as defined above.
"hydroxyalkyl" refers to an alkyl group substituted with a hydroxy group, wherein alkyl is as defined above.
"alkenyl" refers to alkenyl, also known as alkenyl, preferably alkyl having 2 to 8 carbon atoms, more preferably alkyl having 2 to 6 carbon atoms, and most preferably alkyl having 2 to 3 carbon atoms. Wherein said alkenyl group may be further substituted with other related groups, such as: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate groups.
"alkynyl" refers to (CH≡C-), preferably an alkyl group containing 2 to 8 carbon atoms, more preferably an alkyl group of 2 to 6 carbon atoms, and most preferably an alkyl group of 2 to 3 carbon atoms. Wherein said alkynyl group may be further substituted with other related groups such as: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate groups.
"fused ring group" means a polycyclic group formed by two or more carbocycles or heterocycles sharing a ring edge, the fused ring group comprising a fused ring alkyl group, a fused ring heteroaryl group, a fused ring aryl group and a fused ring heteroaryl group, wherein the fused ring alkyl group refers to a polycyclic group formed by a cyclic alkyl group and a heterocyclic group, an aryl group, a heteroaryl group sharing a ring edge; the condensed ring heterocyclic group refers to heterocyclic groups, cycloalkyl, aryl and heteroaryl groups which are formed by sharing a ring edge; the condensed ring aryl refers to aryl, cycloalkyl, heterocyclic group and polycyclic group formed by sharing ring edges by heteroaryl; the condensed ring heteroaryl refers to heteroaryl, cycloalkyl, heterocyclic and polycyclic groups formed by sharing a ring edge by the heterocyclic group; for example:
Figure PCTCN2021136249-APPB-000050
Figure PCTCN2021136249-APPB-000051
"deuterated alkyl" refers to an alkyl group substituted with one or more deuterium, wherein alkyl is as defined above.
"deuteroalkoxy" refers to an alkoxy group substituted with one or more deuterium, wherein alkoxy is as defined above.
"hydroxy" refers to an-OH group.
"halogen" means fluorine, chlorine, bromine or iodine.
"amino" means-NH 2
"cyano" refers to-CN.
"nitro" means-NO 2
"carboxy" means-C (O) OH.
"THF" refers to tetrahydrofuran.
"EtOAc" refers to ethyl acetate.
"MeOH" refers to methanol.
"DMF" refers to N, N-dimethylformamide.
"DEA" refers to diethylamine.
"TFA" refers to trifluoroacetic acid.
"MeCN" refers to acetonitrile.
"DMA" refers to N, N-dimethylacetamide.
“Et 2 O "refers to diethyl ether.
"DCE" refers to 1,2 dichloroethane.
"NBS" refers to N-bromosuccinimide.
"NIS" refers to N-iodosuccinimide.
"Cbz-Cl" refers to benzyl chloroformate.
“Pd 2 (dba) 3 "means tris (dibenzylideneacetone) dipalladium.
"Dppf" refers to 1,1' -bis-diphenylphosphino ferrocene.
"HATU" refers to 2- (7-oxo-benzotriazol) -N, N' -tetramethylurea hexafluorophosphate.
"KHMDS" refers to potassium hexamethyldisilazide.
"LiHMDS" refers to lithium bis (trimethylsilylamide).
"MeLi" refers to lithium-based.
"n-BuLi" refers to n-butyllithium.
“NaBH(OAc) 3 "means sodium triacetoxyborohydride.
The terms "X is selected from A, B or C", "X is selected from A, B and C", "X is A, B or C", "X is A, B and C", etc. all express the same meaning, that is, X may be any one or several of A, B, C.
The hydrogen atoms of the invention can be replaced by the isotope deuterium thereof, and any hydrogen atom in the compound of the embodiment of the invention can be replaced by deuterium atoms.
"optional" or "optionally" means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs or does not. For example, "a heterocyclic group optionally substituted with an alkyl group" means that an alkyl group may be, but is not necessarily, present, and the description includes cases where the heterocyclic group is substituted with an alkyl group and cases where the heterocyclic group is not substituted with an alkyl group.
"substituted" means that one or more hydrogen atoms, preferably up to 5, more preferably 1 to 3 hydrogen atoms in the group are independently substituted with a corresponding number of substituents. It goes without saying that substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable when bound to carbon atoms having unsaturated (e.g., olefinic) bonds.
"pharmaceutical composition" means a mixture comprising one or more of the compounds described herein or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to promote the administration to organisms, facilitate the absorption of active ingredients and thus exert biological activity.
By "pharmaceutically acceptable salts" is meant salts of the compounds of the invention which are safe and effective when used in a mammal and which have the desired biological activity.
Detailed Description
The invention is further described below in connection with examples, which are not intended to limit the scope of the invention.
Examples
The structure of the compounds of the present invention is determined by Nuclear Magnetic Resonance (NMR) or/and liquid chromatography-mass spectrometry (LC-MS). NMR chemical shifts (δ) are given in parts per million (ppm). NMR was performed using Bruker AVANCE-400 nuclear magnetic resonance apparatus with deuterated dimethyl sulfoxide (DMSO-d) 6 ) Deuterated methanol (CD) 3 OD)Deuterated chloroform (CDCl) 3 ) Or deuterium water (D) 2 O), internal standard is Tetramethylsilane (TMS).
An Agilent 1200 affinity Series mass spectrometer was used for LC-MS measurement. HPLC was performed using Agilent 1200DAD high pressure liquid chromatography (Sunfire C18X 4.6mm column) and Waters 2695-2996 high pressure liquid chromatography (Gimini C18X 4.6mm column).
The thin layer chromatography silica gel plate uses a smoke table yellow sea HSGF254 or Qingdao GF254 silica gel plate, the specification adopted by TLC is 0.15 mm-0.20 mm, and the specification adopted by the thin layer chromatography separation and purification product is 0.4 mm-0.5 mm. Column chromatography generally uses tobacco stand yellow sea silica gel 200-300 mesh silica gel as a carrier.
The starting materials in the examples of the present invention are known and commercially available or may be synthesized using or according to methods known in the art.
All reactions of the invention were carried out under continuous magnetic stirring under dry nitrogen or argon atmosphere, with the solvent being a dry solvent and the reaction temperature being in degrees celsius, without specific explanation.
The system of eluent for silica gel column chromatography and the developing agent system for thin layer chromatography used for the purification of the compound in the intermediate and examples include: a: dichloromethane and methanol system, B: n-hexane and ethyl acetate system, C: the volume ratio of the solvent in the methylene dichloride and acetone system is adjusted according to the polarity of the compound, and small amounts of alkaline or acidic reagents such as triethylamine and acetic acid can be added for adjustment.
Intermediate 1
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine
Figure PCTCN2021136249-APPB-000052
First step
3, 6-dichloropyridazine (1.0 g,6.71 mmol) was suspended in 30mL of deionized water, and (1R, 2R) -2-isopropylcyclopropane-1-carboxylic acid (866 mg,6.71mmol, prepared as described in WO2019168744A 1) and concentrated sulfuric acid (1 mL) were added and heated to 70℃under nitrogen. A solution of silver nitrate (228 mg,1.34 mmol) was added (1 mL), followed by dropwise addition of a solution of ammonium persulfate (4.5 g,20.1 mmol) in 15mL, over about 30 minutes, and the reaction was continued at 70℃for 1 hour. The reaction mixture was cooled to room temperature, neutralized to pH 8-9 with aqueous ammonia, and extracted with ethyl acetate (60 mL. Times.2). The organic phases were combined, washed successively with water (60 mL), saturated sodium chloride solution (60 mL), dried over anhydrous sodium sulfate, filtered, the filtrate concentrated under reduced pressure, and the resulting residue purified by silica gel column chromatography with eluent system B to give product 3, 6-dichloro-4- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazine 1a (1.0 g), yield: 64.5%.
MS m/z(ESI):230.8[M+H] + .
1 H NMR(400MHz,CDCl 3 ):δ6.87(s,1H),2.03-1.98(m,1H),1.32-1.29(m,1H),1.27-1.23(m,1H),1.18-1.13(m,1H),1.07-1.04(m,6H),1.02-0.98(m,1H).
Second step
3, 6-dichloro-4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine 1a (740 mg,3.20 mmol) was dissolved in a mixed solvent of 8mL of 1, 4-dioxane and 2mL of water, 2, 4-dimethoxypyrimidine-5-boronic acid (589 mg,3.20 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (61 mg,0.080 mmol) and cesium carbonate (1.25 g,3.84 mmol) were added, the mixture was replaced with nitrogen gas for 3 times, and the reaction was carried out by microwave at 70℃for 1 hour. The reaction mixture was cooled to room temperature, and 50mL of water was added thereto, followed by extraction with ethyl acetate (50 mL. Times.2). The organic phases were combined, washed successively with water (50 mL), saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the title product 3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazine intermediate 1 (455 mg), yield: 42.2%.
MS m/z(ESI):335.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ8.76(s,1H),7.58(s,1H),3.99(s,3H),3.98(s,3H),2.02-1.97(m,1H),1.29-1.23(m,2H),1.13-1.07(m,2H),1.03-1.10(m,6H).
Intermediate 2
4-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine
Figure PCTCN2021136249-APPB-000053
2, 4-dimethoxy-5- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) pyrimidine (390 mg,1.47 mmol), 4, 6-dichloro-3-methylpyridazine (200 mg,1.23 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (50 mg,0.06 mmol) and cesium carbonate (1.02 g,3.07 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 5 ml), and the reaction was stirred under nitrogen for 3 hours at 70 ℃. Saturated sodium chloride (10 mL) was added to the reaction, dichloromethane (10 mL x 3) was used to extract, the organic phases were combined, dried, and concentrated to give crude product, which was purified by silica gel column chromatography with eluent system B to give the title product 4-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine intermediate 2 (190 mg), yield: 58.1%.
MS m/z(ESI):267.1[M+1] + .
Example 1
5- (5- ((1S, 2R) -2-isopropylcyclopropyl) -6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000054
First step
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine intermediate 1 (80 mg,0.239 mmol) was dissolved in 3mL of acetic acid, sodium acetate (98 mg,1.19 mmol) was added thereto, and the mixture was subjected to microwave reaction at 120℃for 1 hour under nitrogen. The reaction mixture was cooled to room temperature, 30mL of water was added, and extraction was performed with ethyl acetate (30 mL. Times.2). The organic phases were combined, washed sequentially with water (30 mL), saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure to give the crude product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazin-3 (2H) -one 1a (70 mg) which was directly used in the next reaction.
MS m/z(ESI):317.0[M+H] + .
Second step
6- (2, 4-Dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3 (2H) -one 1a (30 mg,0.095 mmol) was dissolved in 3mL of methanol, and hydrochloric acid (2M, 1 mL) was added to react at 70℃for 3 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was subjected to reverse phase HPLC to give the title product 5- (5- ((1 s,2 r) -2-isopropylcyclopropyl) -6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 1 (16 mg), yield: 58.6%.
MS m/z(ESI):289.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ12.87(s,1H),11.31(br s,2H),7.71(s,1H),7.23(s,1H),1.92-1.87(m,1H),1.20-1.10(m,1H),0.99-0.94(m,7H),0.92-0.85(m,2H).
Example 2
5- (5- ((1S, 2R) -2-isopropylcyclopropyl) -1-methyl-6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000055
First step
6- (2, 4-Dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3 (2H) -one 1a (30 mg,0.095 mmol) was dissolved in 3mL of acetonitrile, and potassium carbonate (39 mg,0.284 mmol) and methyl iodide (40 mg,0.284 mmol) were added to react at room temperature for 6 hours. The reaction mixture was poured into 30mL of water, and extracted with ethyl acetate (30 mL. Times.2). The organic phases were combined, washed sequentially with water (30 mL), saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure to give the crude product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) -2-methylpyridazin-3 (2H) -one 2a (30 mg) which was used directly in the next reaction.
MS m/z(ESI):331.0[M+H] + .
Second step
6- (2, 4-Dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) -2-methylpyridazin-3 (2H) -one 2a (30 mg,0.091 mmol) was dissolved in 3mL of methanol, hydrochloric acid (2M, 1 mL) was added, and the mixture was reacted at 70℃for 3 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was subjected to reverse phase HPLC to give the title product 5- (5- ((1 s,2 r) -2-isopropylcyclopropyl) -1-methyl-6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 2 (22 mg), yield: 80.1%.
MS m/z(ESI):303.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ11.27(br s,2H),7.75(s,1H),7.27(s,1H),3.67(s,3H),1.96-1.92(m,1H),1.17-1.11(m,1H),1.00-0.94(m,7H),0.93-0.88(m,2H).
Example 3
5- (6- ((1S, 2R) -2-isopropylcyclopropyl) -5-methylimidazo [1,2-a ] pyridin-8-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000056
First step
2-bromo-1, 1-diethoxyethane (1.67 g,8.46 mmol) was dissolved in hydrobromic acid (48%, 3 mL) and heated to 100deg.C for 2 hours. The reaction solution was cooled to room temperature, 15mL of ethanol was added, cooled to 0℃and sodium bicarbonate was added to adjust the pH to alkaline. Filtration was performed, and 3, 5-dibromo-6-methylpyridin-2-amine (1.5 g,5.64 mmol) was added to the filtrate, and the reaction solution was refluxed for 5 hours. After cooling to room temperature, the reaction mixture was poured into 50mL of water and extracted with ethyl acetate (50 mL. Times.2). The organic phases were combined, washed successively with water (50 mL), saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered, the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the product 6, 8-dibromo-5-methylimidazole [1,2-a ] pyridine 3a (1.0 g), yield: 61.1%.
MS m/z(ESI):288.8[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ8.11(d,J=1.2Hz,1H),7.84(s,1H),7.73(d,J=1.2Hz,1H),2.71(s,3H).
Second step
6, 8-dibromo-5-methylimidazole [1,2-a ] pyridine 3a (300 mg,1.03 mmol) was dissolved in 6mL of 1, 4-dioxane and 1.5mL of water, 4, 6-dichloro-3-methylpyridazine (190 mg,1.03 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (38 mg,0.052 mmol) and cesium carbonate (506 mg,1.55 mmol) were added, nitrogen was replaced three times, and the reaction was carried out at 90℃for 1 hour under microwaves. The reaction mixture was cooled to room temperature, and 50mL of water was added thereto, followed by extraction with ethyl acetate (50 mL. Times.2). The organic phases were combined, washed successively with water (50 mL), saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give product 6-bromo-8- (2, 4-dimethoxypyrimidin-5-yl) -5-methylimidazole [1,2-a ] pyridine 3B (180 mg), yield: 49.8%.
MS m/z(ESI):348.8[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ8.73(s,1H),8.03(d,J=1.2Hz,1H),7.66(d,J=1.2Hz,1H),7.58(s,1H),3.98(s,3H),3.92(s,3H),2.78(s,3H).
Third step
6-bromo-8- (2, 4-dimethoxypyrimidin-5-yl) -5-methylimidazole [1,2-a ] pyridine 3b (30 mg,0.086 mmol) was dissolved in 1mL of 1, 4-dioxane and 0.3mL of water, [ (1S, 2S) -2-isopropylcyclopropyl ] -4, 5-tetramethyl-1, 3, 2-dioxolane (18 mg,0.086mmol, for preparation see WO2019168744A 1), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (6 mg,0.0086 mmol) and cesium carbonate (42 mg,0.129 mmol) were added, and the reaction was performed three times under microwaves at 100℃for 1 hour. The reaction mixture was cooled to room temperature, 30mL of water was added, and extraction was performed with ethyl acetate (30 mL. Times.2). The organic phases were combined, washed sequentially with water (30 mL), saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the product 8- (2, 4-dimethoxypyrimidin-5-yl) -6- ((1 s,2 r) -2-isopropylcyclopropyl) -5-methylimidazo [1,2-a ] pyridine 3c (25 mg), yield: 83.6%.
MS m/z(ESI):353.0[M+H] + .
Fourth step
8- (2, 4-Dimethoxypyrimidin-5-yl) -6- ((1S, 2R) -2-isopropylcyclopropyl) -5-methylimidazo [1,2-a ] pyridine 3c (25 mg,0.071 mmol) was dissolved in 3mL of methanol, hydrochloric acid (4M, 1 mL) was added, and the mixture was reacted at 70℃for 3 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by reverse phase HPLC to give the title product 5- (6- ((1 s,2 r) -2-isopropylcyclopropyl) -5-methylimidazo [1,2-a ] pyridin-8-yl) pyrimidine-2, 4 (1 h,3 h) -dione 3 (9.3 mg), yield: 40.4%.
MS m/z(ESI):325.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ11.27(s,2H),9.14(s,1H),7.93(s,1H),7.84(d,J=1.2Hz,1H),7.60(d,J=1.2Hz,1H),2.68(s,3H),1.88-1.83(m,1H),1.22-1.18(m,1H),1.08(d,J=6.8Hz,3H),1.02(d,J=6.8Hz,3H),0.87-0.81(m,2H), 0.80-0.75(m,1H).
Example 4
5- (5- ((1S, 2R) -2-isopropylcyclopropyl) -1-ethyl-6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000057
First step
6- (2, 4-Dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3 (2H) -one 1a (30 mg,0.0946 mmol) was dissolved in acetonitrile (3 mL) at room temperature, potassium carbonate (65 mg, 0.470 mmol) was added with stirring, and then iodoethane (74 mg, 0.470 mmol) was added dropwise. The reaction was stirred at room temperature for 20 hours. The reaction solution was poured into water (30 mL), and the aqueous phase was extracted with ethyl acetate (30 mL. Times.2). The organic phases were combined, washed sequentially with water (30 mL) and saturated sodium chloride (30 mL), dried, concentrated to give crude 6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) -2-ethylpyridazin-3 (2H) -one 4a (30 mg) which was used in the next reaction without purification.
MS m/z(ESI):345.0[M+H] + .
Second step
4a (30 mg,0.0906 mmol) was dissolved in methanol (3 mL) and stirred at room temperature, and 6M hydrochloric acid solution (2 mL) was added to the reaction solution. The reaction solution was stirred at 70℃for 3 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the crude product was separated by reverse phase HPLC to give the product 5- (5- ((1 s,2 r) -2-isopropylcyclopropyl) -1-ethyl-6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 4 (14.3 mg), yield: 51.5%.
MS m/z(ESI):317.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ7.77(s,1H),7.24(s,1H),4.15-4.05(m,2H),1.98-1.93(m,1H),1.28(t,J=7.2Hz,3H),1.19-1.10(m,1H),0.98-0.90(m,9H).
Example 5
5- (5- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000058
Figure PCTCN2021136249-APPB-000059
First step
To a mixture of 20% aqueous potassium hydroxide (2 mL) and diethyl ether (2 mL) was added N-methyl-N-nitrosourea (500 mg,4.8 mmol) under ice-bath conditions, and the mixture was stirred at this temperature for 1 hour. The organic phase of the above mixed solution was added to a solution of 5a (200 mg,1.0 mmol) in diethyl ether (2 mL) under ice-bath conditions, followed by adding palladium acetate (22 mg,0.01 mmol) to the reaction solution, and the reaction was stirred under ice-bath conditions for 30 minutes. The reaction solution was filtered, and the cake was washed with methylene chloride (5 mL. Times.3). The filtrate was concentrated and the resulting crude compound was separated by silica gel column chromatography (eluent system B) to give the product 2- ([ [1,1' -bis (cyclopropane) ] -2-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborane 5B (182 mg), yield: 84.8%.
1 H NMR(400MHz,CDCl 3 ):δ1.21(s,12H),1.02(ddd,1H),0.85–0.72(m,1H),0.57(ddd,1H),0.45–0.24(m,3H),0.16–0.01(m,2H),-0.36(dt,1H).
Second step
5b (92 mg,0.44 mmol), 4-bromo-3, 6-dichloropyridazine (100 mg,0.44 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (36 mg,0.044 mmol) and potassium carbonate (121 mg,0.88 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 ml). At N 2 The reaction solution was stirred under microwave conditions at 80℃for 1 hour under protection. Saturated sodium chloride (10 mL) was added to the reaction mixture, the aqueous phase was extracted with ethyl acetate (10 mL. Times.3), the organic phases were combined, dried, concentrated, and the crude compound was separated by silica gel column chromatography (eluent system B) to give the product 4- ([ 1,1' -bis (cyclopropane) ]-2-yl) -3, 6-dichloropyridazine 5c (79 mg), yield: 78%.
MS m/z(ESI):228.8[M+1] + .
Third step
5c (50 mg,0.22 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (44 mg,0.24 mmol), palladium tetraphenylphosphine (26 mg,0.022 mmol) and potassium carbonate (61 mg,0.44 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 ml). At N 2 The reaction solution was stirred under microwave conditions at 80℃for 1 hour under protection. Saturated sodium chloride (10 mL) was added to the reaction mixture, the aqueous phase was extracted with ethyl acetate (10 mL. Times.3), the organic phases were combined, dried, concentrated, and the crude compound was separated by silica gel column chromatography (eluent system B) to give the product 4- ([ 1,1' -bis (cyclopropane)]-2-yl) -3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine 5d (35 mg), yield: 48%.
MS m/z(ESI):333.1[M+1] + .
Fourth step
5d (100 mg,0.31 mmol) and sodium acetate (3831 mg,4.65 mmol) were dissolved in acetic acid (2 mL). The reaction was stirred for 1 hour at 120℃under microwave heating. The reaction solution was cooled to room temperature, concentrated, and the residue was neutralized to pH neutrality by adding saturated sodium bicarbonate solution. The aqueous phase was extracted with ethyl acetate (25 mL. Times.3). The organic phases were combined, washed sequentially with water (20 mL) and saturated sodium chloride (20 mL), dried, and concentrated to give crude 4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazin-3 (2H) -one 5e (87 mg), yield: 82%. The product was used in the next reaction without purification.
MS m/z(ESI):315.0[M+1] + .
Fifth step
5e (80 mg,0.25 mmol) was dissolved in 1M hydrochloric acid (4 mL) and the reaction was stirred at 70℃for 12 hours. The reaction solution was concentrated, and the crude compound was separated by reverse phase HPLC to give the title product 5- (5- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 5 (40.2 mg), yield: 57%.
MS m/z(ESI):287.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 ):δ12.90(s,1H),11.35(s,2H),7.70(s,1H),7.22(s,1H),1.92–1.81(m,1H),0.99–0.91(m,2H),0.89–0.78(m,2H),0.49–0.32(m,2H),0.22–0.08(m,2H).
Examples 5-1 and 5-2
5- (5- ((1 r,2 s) - [1,1 '-bis (cyclopropane) ] -2-yl) -6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione and 5- (5- ((1 s,2 r) - [1,1' -bis (cyclopropane) ] -2-yl) -6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000060
Resolution of example 5 (40 mg,0.14 mmol) by SFC gave example 5-1 (10.7 mg, r.t= 1.987min, yield: 26.8%) and example 5-2 (15 mg, r.t=1.601 min, yield: 37.5%).
SFC: chiral preparation conditions:
Figure PCTCN2021136249-APPB-000061
chiral analysis method:
Figure PCTCN2021136249-APPB-000062
Figure PCTCN2021136249-APPB-000063
example 5-1: MS m/z (ESI): 286.9[ M+1 ]] +
Example 5-2: MS m/z (ESI) 287.0[ M+1 ]] + .
Example 6
5- (5- ([ 1,1' -bis (cyclopropane) ] -2-yl) -1-methyl-6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000064
First step
5e (100 mg,0.32 mmol) and potassium carbonate (110 mg,0.8 mmol) were dissolved in anhydrous acetonitrile (8 mL) under ice-bath and N-rayed 2 And (5) protecting. At this temperature, methyl iodide (68.16 mg,0.48 mmol) was added dropwise with stirring. The reaction mixture was allowed to warm to room temperature and stirred for a further 12 hours. The reaction solution was filtered, and the cake was washed with ethyl acetate (3 mL. Times.3), and the filtrate was concentrated to give crude 4- ([ 1,1' -bis (cyclopropane)]-2-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) -2-methylpyridazin-3 (2H) -one 6a (83 mg) the product was used in the next reaction without purification.
MS m/z(ESI):329.0[M+1] + .
Second step
6a (80 mg,0.24 mmol) was dissolved in 1M hydrochloric acid (4 mL) and the reaction was stirred at 70℃for 12 hours. The reaction solution was concentrated, and the crude compound was separated by reverse phase HPLC to give the title product 5- (5- ([ 1,1' -bis (cyclopropane) ] -2-yl) -1-methyl-6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 6 (48 mg), yield: 65%.
MS m/z(ESI):300.8[M+1] + .
1 H NMR(400MHz,DMSO-d 6 ):δ11.31(brs,2H),7.76(s,1H),7.26(s,1H),3.66(s,3H),1.97–1.83(m,1H),1.30–1.13(m,1H),1.01–0.90(m,2H),0.89–0.80(m,1H),0.47–0.33(m,2H),0.22–0.09(m,2H).
Examples 6-1 and 6-2
5- (5- ((1 r,2 s) - [1,1 '-bis (cyclopropane) ] -2-yl) -6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione and 5- (5- ((1 s,2 r) - [1,1' -bis (cyclopropane) ] -2-yl) -6-carbonyl-1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000065
Example 6 (55 mg,0.18 mmol) was resolved by SFC to give example 6-1 (11.81 mg, R.T=1.381 min, yield: 21.5%) and example 6-2 (17.44 mg, R.T=1.222 min, yield: 31.7%),
Example 6-1: MS m/z (ESI) 300.8[ M+1 ]] +
Example 6-2: MS m/z (ESI) 300.8[ M+1 ]] + .
Example 7
5- (6-carbonyl-5- (2- (trifluoromethyl) cyclopropyl) -1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000066
First step
7a (1.5 g,11.11 mmol) was dissolved in a mixed solution of methyl tert-butyl ether (5 mL) and water (2 mL) and stirred. The reaction solution was cooled using an ice bath, and an aqueous solution (2 mL) of sodium nitrite (0.843 g,12.22 mmol) was added to the reaction solution. The reaction solution was warmed to room temperature and stirred for 3 hours. The aqueous phase of the reaction solution was separated off, and the remaining organic phase (5 mL, according to WO2015/52226, expected to contain 594mg of the product 2-diazonium-1, 1-trifluoroethane 7b, yield: 48.6%) was used directly in the next reaction.
Second step
Palladium acetate (120.9 mg,0.54 mmol) was added to a solution of 4, 5-tetramethyl-2-vinyl-1, 3, 2-dioxaborane (748.4 mg,4.86 mmol) in methyl tert-butyl ether (5 mL) at room temperature, and then a solution of 7b in methyl tert-butyl ether (5 mL,594mg,5.4 mmol) was slowly added to the reaction. The reaction solution was stirred at room temperature for 2 hours. The reaction solution was filtered, the filtrate was concentrated, and the crude compound was purified by silica gel column chromatography (eluent system B) to give product 4, 5-tetramethyl-2- (2- (trifluoromethyl) cyclopropyl) -1,3, 2-dioxaborane intermediate 7c (0.7 g), yield: 60.9%.
1 H NMR(400MHz,CDCl 3 )δ1.83–1.63(m,1H),1.24(d,12H),1.11–0.95(m,1H),0.85(dd,1H),0.41–0.26(m,1H).
Third step
4-bromo-6-chloropyridazin-3-amine (1.0 g,4.80 mmol), 7c (1.25 g,5.28 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (391.5 mg,0.48 mmol) and cesium carbonate (4.69 g14.39 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:15 ml) and stirred. N (N) 2 The reaction was heated to 110℃with protection and stirred for 16 hours, 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (391.5 mg,0.48 mmol) was added and stirring was continued at 110℃for 16 hours. To the reaction solution was added saturated sodium chloride (10 mL), the aqueous phase was extracted with ethyl acetate (10 mL. Times.3), and the organic phases were combined, dried, and concentrated. The crude compound was separated by silica gel column chromatography (eluent system B) to give the title product 6-chloro-4- (2- (trifluoromethyl) cyclopropyl) pyridazin-3-amine 7d (450 mg), yield: 39.5%.
MS m/z(ESI):238.1[M+1] + .
Fourth step
Sodium nitrite (156.8 mg,2.27 mmol) was dissolved in concentrated sulfuric acid (2.0 mL) and stirred. Compound 7d (450 mg,1.89 mmol) dissolved in acetic acid (5 mL) was slowly added to the reaction solution under ice bath, and the reaction solution was then warmed to room temperature and stirred for 1 hour. Water (5 mL) was added to the reaction solution, and the reaction solution was stirred at room temperature for 0.5 hours. To the reaction solution was added saturated NaCl (10 mL), the aqueous phase was extracted with ethyl acetate (10 mL. Times.3), the organic phases were combined, dried, and concentrated to give crude 6-chloro-4- (2- (trifluoromethyl) cyclopropyl) pyridazin-3 (2H) -one 7e (440 mg), which was used in the next reaction without purification.
MS m/z(ESI):239.1[M+1] + .
Fifth step
7e (230.0 mg,0.96 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (212.8 mg,1.16 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (78.7 mg,0.096 mmol) and cesium carbonate (942.8 mg,2.89 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2.5 ml) and stirred. N (N) 2 The reaction solution was heated to 100℃under protection with microwave stirring for 1 hour. To the reaction was also added saturated sodium chloride (10 mL), the aqueous phase was extracted with ethyl acetate (5 ml×3), the organic phases were combined, dried, concentrated, and the crude compound was separated by silica gel column chromatography (eluent system B) to give the product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- (2- (trifluoromethyl) cyclopropyl) pyridazin-3 (2H) -one 7f (260 mg), yield: 78.8%.
MS m/z(ESI):342.8[M+1] + .
Sixth step
7f (100 mg,0.29 mmol) was dissolved in 1M hydrochloric acid (2 mL) and the reaction was stirred at 70℃for 6 hours. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (6-carbonyl-5- (2- (trifluoromethyl) cyclopropyl) -1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 7 (52 mg), yield: 65.2%.
MS m/z(ESI):314.8[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ13.08(s,1H),11.32(s,2H),7.75(s,1H),7.57(s,1H),2.42(dd,J=9.4,5.8Hz,2H),1.53–1.41(m,1H),1.40–1.29(m,1H).
Examples 7-1 and 7-2
5- (6-carbonyl-5- ((1S, 2S) -2- (trifluoromethyl) cyclopropyl) -1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (6-carbonyl-5- ((1R, 2R) -2- (trifluoromethyl) cyclopropyl) -1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000067
Example 7 (52 mg,0.17 mmol) was resolved by SFC to give example 7-1 (19.2 mg, R.T=2.870 min, yield: 36.8%) and example 7-2 (15.6 mg, R.T=2.305 min, yield: 29.9%),
SFC: chiral preparation conditions:
instrument for measuring and controlling the intensity of light Waters SFC 150
Column type 250*25mm,10μm(REGIS(S,S)WHELK-O1)
Column press 100bar
Mobile phase Supercritical CO 2 /MeOH(0.1%7.0mol/L Ammonia in MeOH)
Flow rate 80g/min
Detection wavelength UV 214nm
Column temperature Room temperature
Chiral analysis method:
Figure PCTCN2021136249-APPB-000068
example 7-1: MS m/z (ESI) 314.8[ M+1 ]] +
Example 7-2: MS m/z (ESI) 314.8[ M+1 ]] + .
Example 8
5- (1-methyl-6-carbonyl-5- (2- (trifluoromethyl) cyclopropyl) -1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000069
First step
Compound 7f (120 mg,0.35 mmol) and potassium carbonate (238.41 mg,1.75 mmol) were dissolved in acetonitrile (5 mL) at room temperature and stirred. Methyl iodide (74.15 mg,0.526 mmol) was added to the reaction solution, and the reaction solution was stirred at room temperature for 2 hours. To the reaction mixture was added saturated sodium chloride (10 ml), and the aqueous phase was extracted with methylene chloride (10 ml. Times.3). The organic phases were combined, dried and concentrated to give the crude product 6- (2, 4-dimethoxypyrimidin-5-yl) -2-methyl-4- (2- (trifluoromethyl) cyclopropyl) pyridazin-3 (2H) -one 8a (120 mg) which was used in the next reaction without purification.
MS m/z(ESI):356.8[M+1] + .
Second step
8a (120 mg, crude product) was dissolved in 1M hydrochloric acid (2 mL), and the reaction solution was stirred at 70℃for 6 hours. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (1-methyl-6-carbonyl-5- (2- (trifluoromethyl) cyclopropyl) -1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione (44 mg), yield: 39.6%.
MS m/z(ESI):328.8[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.37(s,2H),7.79(s,1H),7.60(s,1H),3.68(s,3H),2.51–2.39(m,2H),1.60–1.29(m,2H).
Examples 8-1 and 8-2
5- (1-methyl-6-carbonyl-5- ((1 s,2 s) -2- (trifluoromethyl) cyclopropyl) -1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione and 5- (1-methyl-6-carbonyl-5- (((1 r,2 r) -2- (trifluoromethyl) cyclopropyl) -1, 6-dihydropyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000070
The resolution of example 8 (52 mg,0.17 mmol) by SFC gave example 8-1 (11.8 mg, R.T= 3.114min, yield: 22.7%) and example 8-2 (15.4 mg, R.T= 3.029min, yield: 29.5%),
example 8-1: MS m/z (ESI): 328.8[ M+1 ]] +
Example 8-2: MS m/z (ESI): 328.8[ M+1 ]] + .
Example 9
5- (8- ((1S, 2R) -2-isopropylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000071
First step
Intermediate 1 (40 mg,0.119 mmol) and tert-butyl carbamate (139.96 mg,1.19 mmol) were dissolved in 1, 4-dioxane (1 mL) and chloro (2-dicyclohexylphosphino-2 ',4',6 '-triisopropyl-1, 1' -biphenyl) [2- (2 '-amino-1, 1' -biphenyl) ] palladium (II) (9 mg,0.012 mmol) and cesium carbonate (78 mg,0.239 mmol) were added. The mixture was replaced with nitrogen for 3 times and reacted at 100℃for 1 hour with microwaves. The reaction solution was cooled to room temperature, water (20 mL) was added, and the aqueous phase was extracted with ethyl acetate (25 mL. Times.3). The organic phases were combined, washed sequentially with water (20 mL) and saturated sodium chloride (20 mL), dried, and concentrated. The resulting residue was purified by silica gel column chromatography with eluent system B to give tert-butyl (6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazin-3-yl) carbamate 9a (25 mg), yield: 50.4%.
MS m/z(ESI):416.2[M+H] + .
Second step
9a (25 mg,0.060 mmol) was dissolved in dichloromethane (2 mL) at room temperature and trifluoroacetic acid (1 mL) was added dropwise with stirring. The reaction was stirred at room temperature for 2 hours. To the reaction solution was added dichloromethane (20 mL), and the organic phase was washed with saturated sodium bicarbonate (10 mL) and saturated sodium chloride (5 mL), dried, and concentrated to give crude 6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazin-3-amine 9b (20 mg), which was used in the next reaction without purification.
MS m/z(ESI):316.2[M+1] + .
Third step
9b (20 mg,0.063 mmol), 2-chloro-1, 1-dimethoxy-ethane (79 mg,0.634 mmol) and p-toluenesulfonic acid (22 mg,0.127 mmol) were dissolved in isopropanol (3 mL). The reaction was heated to 100 ℃ and stirred overnight. The reaction solution was concentrated, and the crude product was separated using reverse phase HPLC (formic acid system) to give the title compound 5- (8- ((1 s,2 r) -2-isopropylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 9 (2.3 mg), yield: 11.2%.
MS m/z(ESI):311.9[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.16(s,1H),8.16(s,1H),8.03(s,1H),7.65(s,1H),7.32(s,1H),1.50–1.43(m,2H),1.41(s,2H),1.07(d,1H),1.00(dd,6H).
Example 10
5- (8- (2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000072
First step
Compound 7d (410 mg,1.73 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (317.42 mg,1.73 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (140.80 mg,0.17 mmol) and cesium carbonate (1.69 g,5.18 mmol) were dissolved in a 1, 4-dioxane/water (v/v=4:1, 5 mL) mixture. The reaction was heated to 100 ℃ with microwaves under nitrogen protection and stirred for 1 hour. To the reaction solution was added saturated sodium chloride (10 mL), and the aqueous phase was extracted with ethyl acetate (5 mL. Times.3). The organic phases were combined, dried, concentrated and the crude compound was separated by silica gel column chromatography (eluent system B) to give the product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- (2- (trifluoromethyl) cyclopropyl) pyridazin-3-amine 10a (320 mg), yield: 54.3%.
MS m/z(ESI):341.8[M+1] + .
Second step
Compound 10a (100 mg,0.29 mmol), 2-chloro-1, 1-dimethoxy-ethane (43.80 mg,0.35 mmol) was dissolved in isopropanol (2 mL), and p-toluenesulfonic acid (60.2 mg,0.35 mmol) was then added to the reaction solution. The reaction was heated to 100 ℃ under sealing and stirred for 16 hours. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (8- (2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 10 (35 mg), yield: 35.4%.
MS m/z(ESI):337.8[M+1] + .
1 H NMR(400MHz,DMSO-d 6 ))δ11.39(s,2H),8.24(s,1H),8.01(s,1H),7.71(s,1H),7.60(s,1H),3.01(s,1H),2.92–2.72(m,1H),1.88(dd,1H),1.67–1.47(m,1H).
Examples 10-1 and 10-2
5- (8- ((1S, 2S) -2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (8- ((1R, 2R) -2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000073
Resolution of example 10 (50 mg,0.18 mmol) by SFC gave example 10-1 (18 mg, RT=3.98 min, yield: 36%) and example 10-2 (15 mg, RT=4.08 min, yield: 30%),
example 10-1: MS m/z (ESI) 338.1[ M+1 ]] +
Example 10-2: MS m/z (ESI) 338.1[ M+1 ]] + .
Example 11
5- (2-methyl-8- (2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000074
Compound 10a (50 mg,0.15 mmol) and 1-bromo-2, 2-dimethoxypropane (32.18 mg,0.18 mmol) were dissolved in isopropanol (2 mL), and p-toluenesulfonic acid (30.9 mg,0.18 mmol) was then added to the reaction solution. The reaction solution was heated to 100℃under sealing and stirred for 16 hours. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (2-methyl-8- (2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 11 (18 mg) in 34.5% yield.
MS m/z(ESI):351.8[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.36(s,2H),7.98(d,J=12.5Hz,2H),7.51(s,1H),2.97(s,1H),2.87–2.67(m,1H),2.38(s,3H),1.83(s,1H),1.69–1.46(m,1H).
Example 12
5- (8- ([ 1,1' -bis (cyclopropane) ] -2-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000075
First step
12a (100 mg,0.48 mmol), 4-bromo-6-chloropyridazin-3-amine (99 mg,0.48 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (39 mg,0.048 mmol) and potassium carbonate (132 mg,0.96 mmol) were dissolved in a 1, 4-dioxane/water (v/v=4:1, 2 ml) mixture. The reaction was stirred under nitrogen at 80 ℃ under microwave conditions for 1 hour. To the reaction solution was added saturated sodium chloride (10 mL), the aqueous phase was extracted with ethyl acetate (10 ml×3), the organic phases were combined, dried, and concentrated, and the crude compound was separated by silica gel column chromatography (eluent system B) to give product 4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6-chloropyridazin-3-amine 12B (64 mg), yield: 63%.
MS m/z(ESI):210.1[M+1] + .
Second step
12b (50 mg,0.24 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (66 mg,0.36 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (19 mg,0.024 mmol) and cesium carbonate (156 mg,0.48 mmol) were dissolved in a 1, 4-dioxane/water (v/v=4:1, 2 ml) mixture. The reaction was stirred under nitrogen at 100 ℃ under microwave conditions for 1 hour. To the reaction solution was added saturated sodium chloride (10 mL), the aqueous phase was extracted with ethyl acetate (10 ml×3), the organic phases were combined, dried, and concentrated, and the crude compound was separated by silica gel column chromatography (eluent system B) to give the product 4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazin-3-amine 12c (46 mg), yield: 61%.
MS m/z(ESI):314.2[M+1] + .
Third step
12c (35 mg,0.11 mmol), p-toluenesulfonic acid (22 mg,0.13 mmol) and 2-chloro-1, 1-dimethoxyethane (16 mg,0.13 mmol) were dissolved in isopropanol (5 mL). The reaction solution was stirred at 100℃for 12 hours under nitrogen. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (8- ([ 1,1' -bis (cyclopropane) ] -2-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 12 (15 mg), yield: 42%.
MS m/z(ESI):309.9[M+1] + .
1 H NMR(400MHz,DMSO-d6):δ11.31(s,1H),8.17(d,1H),7.99(s,1H),7.66(d,1H),7.28(s,1H),2.28–2.18(m,1H),1.77–1.59(m,1H),1.44–1.32(m,1H),1.08–0.94(m,2H),0.50–0.36(m,2H),0.28–0.16(m,2H).
Examples 12-1 and 12-2
5- (8- ((1R, 2S) - [1,1 '-bis (cyclopropane) ] -2-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (8- ((1S, 2R) - [1,1' -bis (cyclopropane) ] -2-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000076
Chiral resolution of example 12 (50 mg,0.16 mmol) by SFC gave example 12-1 (12.87 mg, r.t= 3.917min, yield: 25.7%) and example 12-2 (16.64 mg, r.t= 3.483min, yield: 33.2%),
example 12-1: MS m/z (ESI) 310.1[ M+1 ]] +
Example 12-2: MS m/z (ESI) 310.1[ M+1 ]] + .
SFC chiral resolution conditions:
Figure PCTCN2021136249-APPB-000077
chiral analysis method:
Figure PCTCN2021136249-APPB-000078
example 13
5- (8- ((1R, 2S) - [1,1' -bis (cyclopropane) ] -2-yl) -3-chloroimidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000079
12-1 (10 mg,0.03 mmol) and N-chlorosuccinimide (5 mg,0.03 mmol) were dissolved in N, N-dimethylformamide (2 mL). The reaction was heated to 50℃and stirred for 4 hours. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (8- ((1 r,2 s) - [1,1' -bis (cyclopropane) ] -2-yl) -3-chloroimidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 13 (8 mg), yield: 74%.
MS m/z(ESI):344.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.44(s,2H),8.02(s,1H),7.80(s,1H),7.37(s,1H),2.25(m,1H),1.87–1.61(m,1H),1.39(m,1H),1.04(m,2H),0.61–0.40(m, 2H),0.28–0.18(m,2H).
Example 14
5- (8- ([ 1,1' -bis (cyclopropane) ] -2-yl) -3-fluoroimidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000080
First step
8-bromo-6-chloroimidazo [1,2-b ] pyridazine 14a (5 g,21.65 mmol) and N-fluoro-N' -chloromethyl-triethylenediamine bis (tetrafluoroborate) (9.2 g,25.97 mmol) were dissolved in ethyl (100 mL) under nitrogen and stirred. The reaction solution was heated to 50℃and stirred for 12 hours. The reaction solution was concentrated, and the resulting crude compound was separated by silica gel column chromatography (eluent system B) to give the product 8-bromo-6-chloro-3-fluoroimidazo [1,2-B ] pyridazine 14B (862 mg), yield: 16%.
MS m/z(ESI):249.9[M+1] + .
Second step
8-bromo-6-chloro-3-fluoroimidazo [1,2-b ] pyridazine 14b (800 mg,3.21 mmol), 2- ([ [1,1' -bis (cyclopropane) ] -2-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborane (1.33 g,6.42 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (261 mg,0.32 mmol) and cesium carbonate (2.09 g,6.42 mmol) were dissolved in 1, 4-dioxane/water (v/v=20:1, 1 ml) under nitrogen atmosphere with stirring. The reaction solution was heated to 120℃and stirred for 16 hours. To the reaction mixture was added saturated sodium chloride (30 mL), and the aqueous phase was extracted with ethyl acetate (30 mL. Times.3). The organic phases were combined, dried and concentrated to give crude product which was separated by silica gel column chromatography (eluent system B) to give the product 8- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6-chloro-3-fluoroimidazo [1,2-B ] pyridazine 14c (129 mg), yield: 16%.
MS m/z(ESI):252.0[M+1] + .
Third step
8- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6-chloro-3-fluoroimidazo [1,2-b ] pyridazine 14c (100 mg,0.40 mmol), (2, 4-dimethoxypyrimidin-5-yl) boronic acid (88 mg,0.48 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (33 mg,0.040 mmol) and cesium carbonate (196 mg, 0.60 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 ml) under nitrogen. The reaction solution was heated to 100℃with microwaves and stirred for 2 hours. To the reaction mixture was added saturated sodium chloride (10 mL), and the aqueous phase was extracted with ethyl acetate (10 mL. Times.3). The organic phases were combined, dried and concentrated to give crude product which was separated by silica gel column chromatography (eluent system B) to give product 8- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3-fluoroimidazo [1,2-B ] pyridazine 14d (92 mg), yield: 65%.
MS m/z(ESI):356.1[M+1] + .
Fourth step
8- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3-fluoroimidazo [1,2-b ] pyridazine 14d (50 mg,0.14 mmol) was dissolved in a mixture of hydrochloric acid (1M, 1 mL) and methanol (1 mL), and heated to 70℃and stirred for 3 hours. The reaction solution was concentrated, and the crude compound was separated by reverse phase HPLC to give the title product 5- (8- ([ 1,1' -bis (cyclopropane) ] -2-yl) -3-fluoroimidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 14 (24 mg), yield: 53%.
MS m/z(ESI):326.9[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.37(s,2H),8.02(s,1H),7.50(d,J=7.1Hz,1H),7.29(s,1H),2.30–2.15(m,1H),1.76–1.63(m,1H),1.45–1.34(m,1H),1.11–0.95(m,2H),0.51–0.35(m,2H),0.27–0.15(m,2H).
Example 15
5- (3-fluoro-8- ((1S, 2R) -2-isopropylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000081
First step
8-bromo-6-chloro-3-fluoroimidazo [1,2-b ] pyridazine 14b (800 mg,3.21 mmol) was added to (1S, 2R) -2-isopropylcyclopropane-1-carboxylic acid (87mg, 6.71mmol, see WO2019168744A 1) (1.33 g,6.42 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (261 mg,0.32 mmol) and cesium carbonate (2.09 g,6.42 mmol) under nitrogen atmosphere, and stirred in 1, 4-dioxane/water (v/v=20:1, 1 mL). The reaction solution was heated to 120℃and stirred for 16 hours. To the reaction solution was added saturated sodium chloride (30 mL), the aqueous phase was extracted with ethyl acetate (30 ml×3), the organic phases were combined, dried, and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system B) to give the product 6-chloro-3-fluoro-8- ((1 s,2 r) -2-isopropylcyclopropyl) imidazo [1,2-B ] pyridazine 15a (82 mg), yield: 10%.
MS m/z(ESI):254.0[M+1] + .
Second step
6-chloro-3-fluoro-8- ((1S, 2R) -2-isopropylcyclopropyl) imidazo [1,2-b ] pyridazine 15a (50 mg,0.20 mmol), (2, 4-dimethoxypyrimidin-5-yl) boronic acid (44 mg,0.24 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (17 mg,0.02 mmol) and cesium carbonate (98 mg,0.30 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 ml) under nitrogen. The reaction solution was heated to 100℃with microwaves and stirred for 2 hours. To the reaction solution was added saturated sodium chloride (10 mL), the aqueous phase was extracted with ethyl acetate (10 ml×3), the organic phases were combined, dried, and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system B) to give the product 6- (2, 4-dimethoxypyrimidin-5-yl) -3-fluoro-8- ((1 s,2 r) -2-isopropylcyclopropyl) imidazo [1,2-B ] pyridazine 15B (43 mg), yield: 60%.
MS m/z(ESI):358.1[M+1] + .
Third step
6- (2, 4-Dimethoxypyrimidin-5-yl) -3-fluoro-8- ((1S, 2R) -2-isopropylcyclopropyl) imidazo [1,2-b ] pyridazine 15b (40 mg,0.11 mmol) was dissolved in a mixture of hydrochloric acid (1M, 1 mL) and methanol (1 mL), and heated to 70℃and stirred for 3 hours. The reaction solution was concentrated, and the crude compound was separated by reverse phase HPLC preparation (formic acid system) to give the title product 5- (3-fluoro-8- ((1 s,2 r) -2-isopropylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 15 (17 mg), yield: 47%.
MS m/z(ESI):330.1[M+1] + .
1 H NMR(400MHz,DMSO-d6)δ11.49(s,2H),7.99(s,1H),7.52(d,1H),7.26(s,1H),2.29–2.22(m,1H),1.47–1.38(m,2H),1.32–1.22(m,1H),1.15–1.08(m,1H),0.99(dd,6H).
Example 16
5- (8- (2-isobutylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000082
First step
16a (1.5 g,11.72 mmol), 4-methyl-1-pentyne (1.06 g,12.89 mmol) and zirconocene hydrochloride (302 mg,1.17 mmol) were mixed. The reaction was heated to 60 ℃ in a closed tube and stirred overnight. The reaction solution was filtered, the filtrate was concentrated, and the crude product was separated by silica gel column chromatography (eluent system a) to give product (E) -4, 5-tetramethyl-2- (4-methylpent-1-en-1-yl) -1,3, 2-dioxaborolan 16b (1.75 g), yield: 72%.
1 H NMR(400MHz,CDCl 3 )δ6.68-6.55(m,1H),5.48-5.35(m,1H),2.09-2.01(m,2H),1.77-1.65(m,1H),1.27(s,12H),0.90(d,6H).
Second step
Potassium hydroxide (2.72 g,48.54 mmol) was dissolved in a mixture of methyl tert-butyl ether (10 mL) and water (10 mL) at 0deg.C. To the above lye was added 1-methyl-1-nitrosourea (4.17 g,40.45 mmol) in portions to give an ether solution of diazomethane after all solids have dissolved. 16b (340 mg,1.62 mmol) of palladium acetate (36 mg,0.16 mmol) was dissolved in methyl tert-butyl ether (5 mL) under nitrogen, and the reaction mixture was cooled to 0deg.C and stirred. The diazomethane ether solution was added dropwise to the system with stirring using a syringe. After the completion of the dropwise addition, the reaction was returned to room temperature and stirring was continued for 2 hours. The reaction solution was filtered, the filtrate was concentrated, and the crude product was separated by silica gel column chromatography (eluent system a) to give product 2- (2-isobutylcyclopropyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan 16c (257 mg), yield: 70%.
1 H NMR(400MHz,CDCl 3 )δ1.68(m,1H),1.22(s,12H),1.12-1.01(m,1H),0.96-0.84(m,8H),0.73-0.65(m,1H),0.43-0.34(m,1H),-0.39--0.49(m,1H).
Third step
16c (200 mg,0.89 mmol), 3-amino-4-bromo-6-chloropyridazine (223 mg,1.07 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride (65 mg,0.089 mmol) and potassium carbonate (308 mg,2.23 mmol) were dissolved in a mixture of 1, 4-dioxane (3 mL) and water (1 mL) under nitrogen. The reaction was heated to 90℃with microwaves and stirred for 2.5 hours. The organic phase was separated and the aqueous phase was extracted with ethyl acetate (5 mL x 2), the organic phases combined, dried, concentrated and the crude product separated by silica gel column chromatography (eluent system a) to give the product 6-chloro-4- (2-isobutylcyclopropyl) pyridazin-3-amine 16d (102 mg), yield: 40%.
MS m/z(ESI):226.1[M+1] + .
Fourth step
16d (102 mg,0.45 mmol), 2, 4-dimethoxypyrimidine-5-boronic acid (100 mg,0.54 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride (33 mg,0.045 mmol) and potassium carbonate (187 mg,1.36 mmol) were dissolved in a mixture of 1, 4-dioxane (2.1 mL) and water (0.7 mL) under nitrogen. The reaction was heated to 90℃with microwaves and stirred for 2 hours. The organic phase was separated and the aqueous phase was extracted with ethyl acetate (5 mL. Times.2). The organic phases were combined, dried and concentrated, and the crude product was separated by silica gel column chromatography (eluent system a) to give product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- (2-isobutylcyclopropyl) pyridazin-3-amine 16e (135 mg), yield: 90.7%.
MS m/z(ESI):330.0[M+1] + .
Fifth step
16e (135 mg,0.41 mmol), 2-chloroacetaldehyde dimethyl acetal (102 mg,0.82 mmol) and p-toluenesulfonic acid (85 mg,0.49 mmol) were dissolved in N, N-dimethylformamide (2 mL). The reaction was heated to 100deg.C and stirred for 16 hours. The reaction solution was filtered, and the filtrate was separated by reverse phase HPLC to give the title product 5- (8- (2-isobutylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 16 (31 mg), yield: 23.3%.
MS m/z(ESI):326.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.30(s,1H),8.42(s,1H),8.17(d,1H),8.00(s,1H),7.67(s,1H),7.30(s,1H),2.26-2.17(m,1H),1.79-1.67(m,1H),1.65-1.56(m,1H),1.51-1.26(m,3H),1.07-0.98(m,1H),0.95-0.87(m,6H).
Example 16-1 and example 16-2
5- (8- ((1S, 2S) -2-isobutylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (8- ((1R, 2R) -2-isobutylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000083
Example 16 (20 mg,0.061 mmol) was resolved by chiral column (AS column) to give the title product 16-1 (2.4 mg, r.t=1.419 min), yield: 12.5%;16-2 (4.9 mg, r.t=1.275 min), yield: 24.5%. AS column chiral resolution conditions:
Figure PCTCN2021136249-APPB-000084
chiral analysis method:
Figure PCTCN2021136249-APPB-000085
example 16-1, MS m/z (ESI): 326.2[ M+1 ]] +
Example 16-2, MS m/z (ESI): 326.1[ M+1 ]] +
Example 17
5- (8- ((1S, 2S) -2-ethylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000086
Figure PCTCN2021136249-APPB-000087
First step
17a (75 mg,0.36 mmol), 2- ((1S, 2S) -2-ethylcyclopropyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (78 mg,0.40mmol, for its preparation see WO2019168744A 1), 1-bis (diphenylphosphine) ferrocene palladium dichloride (26 mg,0.036 mmol) and cesium carbonate (234.45 mg,0.72 mmol) were dissolved in a mixed solvent of 1, 4-dioxane (1.5 mL) and water (0.5 mL) under nitrogen. The reaction was heated to 120℃and stirred for 16 hours. The reaction solution was concentrated, and the residue was separated by silica gel column chromatography (eluent system B) to give product 6-chloro-4- ((1 s,2 s) -2-ethylcyclopropyl) pyridazin-3-amine 17B (74 mg), yield: 55%.
MS m/z(ESI):198.1[M+1] + .
Second step
17b (74 mg,0.37 mmol), 2, 4-dimethoxypyrimidine-5-boronic acid (90 mg,0.49 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride (27 mg,0.037 mmol) and potassium carbonate (103 mg,0.75 mmol) were dissolved in a mixture of 1, 4-dioxane (1.5 mL) and water (0.5 mL) under nitrogen. The reaction was heated to 90℃and stirred for 2 hours. The organic phase was separated and the aqueous phase was extracted with ethyl acetate (2 mL. Times.2). The organic phases were combined, dried, concentrated and the residue was separated by silica gel column chromatography (eluent system B) to give the title product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 s) -2-ethylcyclopropyl) pyridazin-3-amine 17c (70 mg), yield: 62%.
MS m/z(ESI):302.0[M+1] + .
Third step
17c (70 mg,0.23 mmol), 2-chloroacetaldehyde dimethyl acetal (58 mg,0.46 mmol) and p-toluenesulfonic acid (100 mg,0.58 mmol) were dissolved in isopropanol (1 mL). The reaction was heated to 100deg.C and stirred for 16 hours. The reaction solution was filtered, and the filtrate was prepared and separated (formic acid system) by reverse phase HPLC to give the title product 5- (8- ((1 s,2 s) -2-ethylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 17 (35 mg), yield: 51%.
MS m/z(ESI):298.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,2H),8.18(d,1H),7.96(s,1H),7.68(d,1H),7.25(s,1H),2.28-2.19(m,1H),1.64-1.56(m,1H),1.51-1.40(m,3H),1.10-0.95(m,4H).
Example 18
5- (3-chloro-8- ((1S, 2S) -2-ethylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000088
Figure PCTCN2021136249-APPB-000089
First step
Example 17 (28 mg,0.094 mmol) N-chlorosuccinimide (12.58 mg,0.94 mmol) was dissolved in N, N-dimethylformamide (0.8 mL) and stirred at room temperature for 72 hours. The reaction solution was filtered, and the filtrate was separated by reverse phase HPLC to give the title product 5- (3-chloro-8- ((1 s,2 s) -2-ethylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 18 (16 mg) in 51.2% yield.
MS m/z(ESI):332.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.42(s,2H),8.03(s,1H),7.81(s,1H),7.39(s,1H),2.25(m,1H),1.58(m,1H),1.51-1.40(m,3H),1.13-1.06(m,1H),0.98(t,J=7.2Hz,3H).
Example 19
5- (8- (2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000090
First step
Example 10 (90 mg,0.27 mmol) and 1-chloropyrrolidine-2, 5-dione (36 mg,0.27 mmol) were dissolved in N, N-dimethylformamide (3 mL) and stirred. The reaction was stirred at 30℃for 48 hours. The reaction solution was filtered, and the filtrate was separated using reverse phase HPLC to give the title product 5- (8- (2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 19 (83 mg), yield: 83.7%.
MS m/z(ESI):371.7[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ8.19(s,1H),7.70(s,1H),7.68(s,1H),2.93–2.84(m,1H),2.79–2.70(m,1H),1.81–1.72(m,1H),1.66–1.59(m,1H).
Example 19-1 and example 19-2
5- (3-chloro-8- ((1S, 2S) -2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (3-chloro-8- ((1R, 2R) -2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000091
Example 19 (26 mg,0.07 mmol) was resolved by chiral column (GC 7 column) to give example 19-1 (10 mg, r.t=5.02 min, yield: 38.5%); and example 19-2 (11 mg, r.t=6.03 min, yield: 42.3%).
Chiral resolution conditions:
Figure PCTCN2021136249-APPB-000092
chiral analysis method:
Figure PCTCN2021136249-APPB-000093
example 19-1, MS m/z (ESI): 371.7[ M+1 ]] +
Example 19-2, MS m/z (ESI): 371.7[ M+1 ]] +
Example 20
5- (8- (cyclopent-1-en-1-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000094
First step
20a (100 mg,0.48 mmol), 2- (cyclopent-1-en-1-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (93 mg,0.48 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (39 mg,0.048 mmol) and cesium carbonate (313 mg,0.96 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 ml) under nitrogen. The reaction solution was heated to 100℃with microwaves and stirred for 2 hours. To the reaction mixture was added saturated sodium chloride (10 mL), and the aqueous phase was extracted with ethyl acetate (10 mL. Times.3). The organic phases were combined, dried and concentrated to give crude product which was separated by silica gel column chromatography (eluent system B) to give product 6-chloro-4- (cyclopent-1-en-1-yl) pyridazin-3-amine 20B (74 mg), yield: 78%.
MS m/z(ESI):196.0[M+1] + .
Second step
4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6-chloropyridazin-3-amine 20b (60 mg,0.31 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (68 mg,0.37 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (25 mg,0.031 mmol) and cesium carbonate (202 mg,0.62 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 ml) under nitrogen. The reaction solution was heated to 100℃with microwaves and stirred for 2 hours. To the reaction mixture was added saturated sodium chloride (10 mL), and the aqueous phase was extracted with ethyl acetate (10 mL. Times.3). The organic phases were combined, dried and concentrated to give crude product which was separated by silica gel column chromatography (eluent system B) to give product 4- (cyclopent-1-en-1-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazin-3-amine 20c (62 mg), yield: 67%.
MS m/z(ESI):300.1[M+1] + .
Third step
20c (50 mg,0.17 mmol), p-toluene sulfonic acid (29 mg,0.51 mmol) and 2-chloro-1, 1-dimethoxyethane (106 mg,0.85 mmol) were dissolved in isopropanol (5 mL) under nitrogen. The reaction solution was heated to 100℃and stirred for 12 hours. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (8- (cyclopent-1-en-1-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 20 (19 mg), yield: 38%.
MS m/z(ESI):296.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.50(s,2H),8.22(d,J=1.3Hz,1H),8.03(s,1H),7.78(t,J=2.3Hz,1H),7.73(d,J=1.3Hz,1H),7.56(s,1H),2.83–2.70(m,2H),2.71–2.63(m,2H),2.13–1.94(m,2H).
Example 21
5- (8- (3, 3-dimethylcyclopent-1-en-1-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000095
Figure PCTCN2021136249-APPB-000096
First step
21a (987 mg,8.80 mmol) was dissolved in tetrahydrofuran (20 mL) and cooled to-78deg.C and stirred for 5 min. Lithium bis (trimethylsilylamide) (1M, 8 mL) was added dropwise to the above system, and the reaction was stirred at-78℃for 30 minutes. A solution of N-phenylbis (trifluoromethanesulfonyl) imide (3.14 g,8.80 mmol) in tetrahydrofuran (10 mL) was then added dropwise to the reaction. The reaction was slowly returned to room temperature and stirred overnight. To the reaction mixture was added a saturated aqueous ammonium chloride solution (15 mL). The organic phase was separated and the aqueous phase was extracted with methyl tert-butyl ether (20 mL. Times.2). The organic phases were combined and washed successively with 5% aqueous sodium carbonate, saturated sodium chloride solution, dried and concentrated to give the crude product 3, 3-dimethylcyclopent-1-en-1-yl triflic acid 21b (1.9 g), which was used directly in the next reaction without purification.
1 H NMR(400MHz,CDCl 3 )δ5.47(s,1H),2.66-2.58(m,2H),1.81(t,J=7.8Hz,2H),1.12(s,6H).
Second step
21b (500 mg,2.05 mmol), pinacol biborate (624 mg,2.46 mmol), tris (dibenzylideneacetone) dipalladium (94 mg,0.102 mmol), 2-dicyclohexylphosphine-2 ',4',6' -triisopropylbiphenyl (195 mg, 0.09 mmol) and potassium acetate (402 mg,4.09 mmol) were dissolved in anhydrous 1, 4-dioxane (9 mL) under nitrogen. The reaction was heated to 100deg.C and stirred for 2 hours. The reaction solution was filtered, and the filtrate was concentrated to give the crude title product 2- (3, 3-dimethylcyclopent-1-en-1-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan 21c (454 mg), which was used in the next reaction without purification.
Third step
3-amino-4-bromo-6-chloropyridazine (511 mg,2.45 mmol), 21c (454 mg,2.04 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride (150 mg,0.204 mmol) and potassium carbonate (560 mg,4.09 mmol) were dissolved in a mixed solvent of 1, 4-dioxane (8 mL) and water (2 mL) under nitrogen, and the reaction solution was heated to 100℃and stirred for 2 hours. The reaction solution was concentrated, and the crude product was separated by silica gel column chromatography (eluent system a) to give 6-chloro-4- (3, 3-dimethylcyclopent-1-en-1-yl) pyridazin-3-amine 21d (1 g), yield: 90%.
MS m/z(ESI):224.1[M+1] + .
Fourth step
21d (620 mg,2.77 mmol), 2, 4-dimethoxypyrimidine-5-boronic acid (612 mg,3.33 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride (203 mg,0.277 mmol) and potassium carbonate (766 mg,5.54 mmol) were dissolved in a mixture of 1, 4-dioxane (10 mL) and water (3 mL) under nitrogen and the reaction was heated to 90℃with microwaves and stirred for 2 hours. The organic phase was separated and the aqueous phase was extracted with ethyl acetate (10 mL. Times.2). The organic phases were combined, dried and concentrated, and the crude product was separated by silica gel column chromatography (eluent system a) to give the product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- (3, 3-dimethylcyclopent-1-en-1-yl) pyridazin-3-amine 21e (107 mg) in 11.8% yield.
MS m/z(ESI):328.2[M+1] + .
Fifth step
21e (184 mg,0.562 mmol), 2-chloroacetaldehyde dimethyl acetal (280 mg,2.25 mmol) and p-methylbenzenesulfonic acid (387 mg,2.25 mmol) were dissolved in N, N-dimethylformamide (2.5 mL) and the reaction was heated to 100deg.C and stirred for 16 hours. The reaction solution was filtered, and the filtrate was separated by reverse phase HPLC to give the title product 5- (8- (3, 3-dimethylcyclopent-1-en-1-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione 21 (97 mg), yield 53.4%.
MS m/z(ESI):324.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.47(s,2H),8.21(s,1H),8.03(s,1H),7.72(s,1H),7.61-7.55(m,2H),2.83(t,2H),1.82(t,2H),1.18(s,6H).
Example 22
5- (8- (2- (cyclopropylmethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000097
First step
2, 6-tetramethylpiperidine lithium (525 mg,3.57 mmol) was dissolved in dry tetrahydrofuran (1.4 mL) under nitrogen and stirred at 0deg.C. To the reaction solution was added dropwise a solution of bis (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) methane (956 mg,3.57 mmol) in tetrahydrofuran (2.8 mL). The reaction was stirred at 0℃for 5 min. The reaction solution was cooled to-78℃and 2-cyclopropylacetaldehyde (250 mg,2.97 mmol) was added to the reaction solution. The reaction was stirred at-78℃for 4 hours. The reaction solution was concentrated, ethyl acetate (30 mL) was added to the residue, and the organic phase was washed with saturated ammonium chloride, dried, concentrated, and the crude compound was separated by silica gel column chromatography (eluent system a) to give the product (E) -2- (3-cyclopropylprop-1-en-1-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan 22b (210 mg), yield: 33.9%.
1 H NMR(400MHz,CDCl 3 )δ6.70(dt,1H),5.54(dt,1H),2.10–1.98(m,2H),1.27(s,12H),0.69–0.54(m,1H),0.45(dd,4H).
Second step
22b (210 mg,1.01 mmol) and palladium acetate (23 mg,0.1 mmol) were dissolved in methyl tert-butyl ether (5 mL) under nitrogen and stirred under an ice bath. Potassium hydroxide (1.13 g,20.18 mmol) was dissolved in a mixture of water (10 mL) and methyl tert-butyl ether (10 mL) under an ice bath. 1-methyl-1-nitroso-urea (1.04 g,10.09 mmol) was added in portions with stirring. The reaction was stirred for 30 minutes. The bright yellow supernatant was added dropwise to the solution of 22b under ice bath. After completion of the dropwise addition, the reaction solution was stirred under an ice bath for 2 hours. The reaction solution was filtered, the filtrate was concentrated, and the residue was separated by silica gel column chromatography (eluent system a) to give the product 2- [2- (cyclopropylmethyl) cyclopropyl ] -4, 5-tetramethyl-1, 3, 2-dioxaborolan 22c (120 mg), yield: 53.5%.
1 H NMR(400MHz,CDCl 3 )δ1.22(s,12H),1.05–0.91(m,2H),0.87–0.70(m,2H),0.70–0.58(m,2H),0.50–0.35(m,4H),-0.38(dt,1H).
Third step
22c (120 mg,0.540 mmol), 4-bromo-6-chloro-pyridazin-3-amine (124 mg,0.594 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (91 mg,0.054 mmol) and cesium carbonate (352 mg,1.08 mmol) were dissolved in a mixture of 1, 4-dioxane (2 mL) and water (0.5 mL) under nitrogen. The reaction solution was heated to 120℃and stirred for 12 hours. The reaction solution was filtered, the filter residue was washed with ethyl acetate, the organic phase was washed with water, saturated ammonium chloride, dried and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system a) to give the product 6-chloro-4- [2- (cyclopropylmethyl) cyclopropyl ] pyridazin-3-amine 22d (81.2 mg), yield: 67.2%.
MS m/z(ESI):224.1[M+1] + .
Fourth step
22d (80 mg, 0.258 mmol), 2, 4-dimethoxy-5- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyrimidine (105 mg,0.393 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (60 mg,0.036 mmol) and cesium carbonate (233 mg, 0.015 mmol) were dissolved in a mixture of 1, 4-dioxan (2 mL) and water (0.5 mL) under nitrogen. The reaction solution was heated to 100℃with microwaves and stirred for 1 hour. The reaction solution was concentrated, and the crude product was separated by silica gel column chromatography (eluent system a) to give product 4- [2- (cyclopropylmethyl) cyclopropyl ] -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazin-3-amine 22e (60.7 mg), yield: 51.9%.
MS m/z(ESI):328.1[M+1] + .
Fifth step
22e (61 mg,0.185 mmol), 2-chloro-1, 1-dimethoxy-ethane (115 mg,0.927 mmol) and p-toluene sulfonic acid (32 mg,0.185 mmol) were dissolved in DMF (3 mL). The reaction solution was heated to 100℃and stirred for 3 hours. The reaction solution was cooled and isolated (formic acid system) by reverse phase HPLC to give the title product 5- (8- (2- (cyclopropylmethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione (33 mg), 55.0% yield.
MS m/z(ESI):324.2[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,2H),8.18(d,1H),7.95(d,1H),7.68(s,1H),7.26(s,1H),2.34–2.24(m,1H),1.70(s,1H),1.51–1.42(m,1H),1.39(t,2H),1.15–1.04(m,1H),0.81(s,1H),0.38(d,2H),0.09(d,2H).
Example 22-1 and example 22-2
5- (8- ((1S, 2S) -2- (cyclopropylmethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (8- ((1R, 2R) -2- (cyclopropylmethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000098
Example 22 (33 mg) was resolved by chiral column (C4 AS column) to give example 22-1 (10.28 mg, r.t=3.739 min, yield: 31.2%); and example 22-2 (6.2 mg, r.t=3.422 min, yield: 18.8%).
Chiral resolution conditions:
Figure PCTCN2021136249-APPB-000099
chiral analysis method:
Figure PCTCN2021136249-APPB-000100
example 22-1, MS m/z (ESI): 324.2[ M+1 ]] +
Example 22-2, MS m/z (ESI): 324.2[ M+1 ]] +
Example 23
5- (8- (2-cyclobutylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000101
First step
2, 6-tetramethylpiperidine lithium (1.05 g,7.13 mmol) was dissolved in dry tetrahydrofuran (10 mL) at 0deg.C under nitrogen and stirred. To the reaction solution was added dropwise a solution of bis (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) methane (1.91 g,7.13 mmol) in tetrahydrofuran (3 mL). The reaction was stirred at 0℃for 5 min. The reaction solution was cooled to-78℃and ring Ding Jiaquan (500 mg,5.94 mmol) was added to the reaction solution. The reaction was stirred at-78℃for 4 hours. The reaction solution was concentrated, ethyl acetate (30 mL) was added to the residue, and the organic phase was washed with saturated ammonium chloride, dried, concentrated, and the crude compound was separated by silica gel column chromatography (eluent system a) to give the title product (E) -2- (2-cyclobutylvinyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolane 23b (710 mg), yield: 58%.
1 H NMR(400MHz,CDCl 3 )δ6.73(dd,1H),5.34(dd,1H),3.11–2.93(m,1H),2.21–2.02(m,2H),1.98–1.72(m,4H),1.26(s,12H).
Second step
23b (600 mg,2.88 mmol) and palladium acetate (65 mg,0.29 mmol) were dissolved in methyl tert-butyl ether (10 mL) under nitrogen and stirred. Potassium hydroxide (3.24 g,57.66 mmol) was dissolved in a mixture of water (10 mL) and methyl tert-butyl ether (10 mL) under an ice bath. 1-methyl-1-nitroso-urea (5.94 g,57.66 mmol) was added in portions with stirring. The reaction was stirred for 20 minutes. The bright yellow supernatant was added dropwise to the solution of 23b under ice bath. After completion of the dropwise addition, the reaction solution was stirred under an ice bath for 2 hours. The reaction solution was filtered, the filtrate was concentrated, and the residue was separated by silica gel column chromatography (eluent system a) to give the title product 2- (2-cyclobutylcyclopropyl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan 23c (410 mg), yield: 64%.
1 H NMR(400MHz,CDCl 3 )δ2.03–1.92(m,1H),1.92–1.79(m,2H),1.74–1.65(m,2H),1.64–1.53(m,2H),1.15(s,12H),1.05–0.94(m,1H),0.61–0.48(m,1H),0.44–0.30(m,1H),-0.36–-0.47(m,1H).
Third step
23c (384 mg,1.73 mmol), 4-bromo-6-chloro-pyridazin-3-amine (300 mg, 1.44 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (117 mg,0.14 mmol) and cesium carbonate (937 mg,2.88 mmol) were dissolved in a mixture of 1, 4-dioxane (20 mL) and water (2 mL) under nitrogen. The reaction solution was heated to 120℃and stirred for 12 hours. The reaction solution was filtered, the filter residue was washed with ethyl acetate, the organic phase was washed with water, saturated ammonium chloride, dried and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system a) to give the product 6-chloro-4- (2-cyclobutylcyclopropyl) pyridazin-3-amine 23d (322 mg), yield: 43%.
MS m/z(ESI):224.0[M+1] + .
Fourth step
23d (130 mg,0.58 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (160 mg,0.87 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (47 mg,0.058 mmol) and cesium carbonate (377 mg,1.16 mmol) were dissolved in a mixture of 1, 4-dioxane (2 mL) and water (0.5 mL) under nitrogen. The reaction solution was heated to 100℃with microwaves and stirred for 1 hour. The reaction solution was concentrated, and the crude product was separated by silica gel column chromatography (eluent system a) to give product 4- (2-cyclobutylcyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazin-3-amine 23e (150 mg), yield: 78%.
MS m/z(ESI):328.1[M+1] + .
Fifth step
23e (180 mg,0.55 mmol), 2-chloro-1, 1-dimethoxy-ethane (348 mg,2.75 mmol) and trifluoroacetic acid (940 mg,8.25 mmol) were dissolved in isopropanol (5 mL). The reaction solution was heated to 90℃and stirred for 10 hours. The reaction solution was cooled, and the product 5- (8- (2-cyclobutylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione 23 (120 mg) was obtained by preparative separation (formic acid system) by reverse phase HPLC, and the yield was 67%.
MS m/z(ESI):324.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,2H),8.18(s,1H),7.95(s,1H),7.67(s,1H),7.26(s,1H),2.40–2.22(m,2H),1.99(dq,2H),1.86–1.71(m,4H),1.73–1.62(m,1H),1.56–1.41(m,1H),1.25–1.00(m,1H).
Example 23-1 and example 23-2
5- (8- ((1S, 2R) -2-cyclobutylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (8- ((1R, 2S) -2-cyclobutylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000102
Example 23 (120 mg) was resolved by chiral column (C4 AS column) to give example 23-1 (17 mg, r.t= 3.767min, 15% yield); and example 23-2 (25 mg, r.t= 3.477min, yield: 21%). Chiral resolution conditions:
Figure PCTCN2021136249-APPB-000103
Figure PCTCN2021136249-APPB-000104
chiral analysis method:
Figure PCTCN2021136249-APPB-000105
example 23-1, MS m/z (ESI): 324.1[ M+1 ]] +
Example 23-2, MS m/z (ESI): 324.1[ M+1 ]] +
Example 24
5- (5-fluoro-4- (2- (trifluoromethyl) cyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000106
First step
Sodium hydride (183mg, 7.64mmo,60% in mineral oil) was dispersed in a mixed solution of tetrahydrofuran (20 mL) and N, N-dimethylformamide (20 mL) and stirred. 3-fluoro-1H-pyrrole-2-carboxylic acid ethyl ester (1.00 g,6.36 mmol) was added to the reaction solution under nitrogen in an ice bath. The reaction was stirred under ice bath for 20 minutes. O-diphenylphosphino hydroxylamine (1.93 g,8.27 mmol) was added to the reaction solution. The reaction was stirred at room temperature for 3 hours. To the reaction solution was added a saturated solution (50 mL) of sodium thiosulfate, and the reaction was heated to 30℃for 1 hour. The reaction mixture was diluted with ethyl acetate (120 mL), the organic phase was washed with water and saturated sodium chloride, dried, and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system a) to give the product, ethyl 1-amino-3-fluoro-1H-pyrrole-2-carboxylate 24b (1.10 g), yield: 100%.
MS m/z(ESI):173.1[M+1] + .
Second step
24b (4.25 g,24.69 mmol) and triethylamine (4.99 g,49.37 mmol) were dissolved in dichloromethane (50 mL) and stirred. 3-chloro-3-carbonyl-propionic acid methyl ester (3.37 g,24.69 mmol) was added dropwise to the reaction solution under an ice bath. The reaction solution was stirred at room temperature for 2 hours. The reaction solution was concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system a) to give the product, ethyl 3-fluoro-1- (3-methoxy-3-carbonylpropionylamino) -1H-pyrrole-2-carboxylate 24c (5.80 g), yield: 86.3%.
MS m/z(ESI):273.1[M+1] + .
Third step
Potassium tert-butoxide (4.23 g,37.74 mmol) was added to a solution of 24c (5.14 g,18.87 mmol) in tetrahydrofuran (200 mL) at room temperature. The reaction was stirred at room temperature for 3 hours. The reaction mixture was acidified with formic acid (5 mL), concentrated and the crude product was separated by silica gel column chromatography (eluent system B) to give methyl 5-fluoro-2, 4-dicarbonyl-1, 2,3, 4-tetrahydropyrrolo [1,2-B ] pyridazine-3-carboxylate 24d (3.80 g), yield: 89.0%.
MS m/z(ESI):227.0[M+1] + .
Fourth step
24d (2.00 g,8.84 mmol) was dissolved under nitrogen in a mixture of acetic acid (10 mL) and water (10 mL). The reaction was heated to 100℃with microwaves and stirred for 1 hour. The reaction solution was concentrated and the crude product was separated by silica gel column chromatography (tetrahydrofuran/petroleum ether, from 0% to 80% v/v) to give 5-fluoropyrrolo [1,2-b ] pyridazine-2, 4-diol 24e (1.10 g), yield: 74.0%.
MS m/z(ESI):169.0[M+1] + .
Fifth step
1, 1-trifluoro-N-phenyl-N- (trifluoromethanesulfonyl) methanesulfonamide (5.10 g,14.28 mmol) was added to a solution of 24e (2.00 g,11.90 mmol) and triethylamine (2.40 g,3.79 mmol) in tetrahydrofuran (36 mL). The reaction was stirred at room temperature for 16 hours. Formic acid (1 mL) was added to the reaction mixture, concentrated, and the crude product was separated by silica gel column chromatography (eluent system B) to give the product, 5-fluoro-4-hydroxypyrrolo [1,2-B ] pyridazin-2-yl trifluoromethanesulfonic acid 24f (2.60 g), yield: 72.8%.
MS m/z(ESI):301.0[M+1] + .
Sixth step
24f (600 mg,2.00 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (441 mg,2.4 mmol), cesium carbonate (974.39 mg,3 mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (293 mg,0.4 mmol) were dissolved in a mixture of dioxane (6 mL) and water (1 mL) under nitrogen. The reaction was heated to 100℃with microwaves and stirred for 1 hour. The reaction solution was acidified with formic acid (1 mL), concentrated and the crude product was separated by silica gel column chromatography (tetrahydrofuran/petroleum ether, from 0% to 40% v/v) to give the title product 2- (2, 4-dimethoxypyrimidin-5-yl) -5-fluoropyrrolo [1,2-b ] pyridazin-4-ol 24g (44 mg), yield: 82.0%.
MS m/z(ESI):291.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.80(s,1H),8.59(s,1H),7.73–7.47(m,1H),6.62(d,1H),6.30(s,1H),4.01(s,3H),3.98(s,3H).
Seventh step
1, 1-trifluoro-N-phenyl-N- (trifluoromethanesulfonyl) methanesulfonamide (2.22 g,6.20 mmol) was added to a solution of 24g (1.50 g,5.17 mmol) and triethylamine (1.04 g,10.34 mmol) in tetrahydrofuran (30 mL). The reaction was stirred at room temperature for 16 hours. The reaction solution was neutralized with formic acid (6 mL), concentrated, and the crude product was purified using a C18 reverse phase system (acetonitrile/water (containing 0.05% formic acid), from 0% to 90% v/v) to give the product 2- (2, 4-dimethoxypyrimidin-5-yl) -5-fluoropyrrolo [1,2-b ] pyridazin-4-yl trifluoromethanesulfonic acid for 24h (1.10 g), yield: 50.4%.
MS m/z(ESI):423.0[M+1] + .
Eighth step
4, 5-tetramethyl-2- [2- (trifluoromethyl) cyclopropyl ] -1,3, 2-dioxaborolan (252 mg,1.07 mmol), 24h (150 mg, 0.356 mmol), cesium carbonate (139 mg,0.426 mmol), and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (26 mg,0.036 mmol) were dissolved in a mixture of dioxane (3 mL) and water (0.2 mL) under nitrogen, and the reaction was heated to 100deg.C with microwaves and stirred for 1 hour. The reaction solution was concentrated and the crude product was separated by silica gel column chromatography (tetrahydrofuran/petroleum ether, from 0% to 40% v/v) to give the product 2- (2, 4-dimethoxypyrimidin-5-yl) -5-fluoro-4- (2- (trifluoromethyl) cyclopropyl) pyrrolo [1,2-b ] pyridazine 24i (110 mg), yield: 81.0%.
MS m/z(ESI):383.1[M+1] + .
Ninth step
24i (100 mg,0.262 mmol) was dissolved in a mixture of hydrochloric acid (4M, 2 mL) and methanol (2 mL), heated to 70℃and stirred for 8 hours. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 2- (2, 4-dimethoxypyrimidin-5-yl) -5-fluoro-4- (2- (trifluoromethyl) cyclopropyl) pyrrolo [1,2-b ] pyridazine 24 (78 mg), yield: 84.2%.
MS m/z(ESI):354.2[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.32(s,2H),7.92(s,1H),7.78–7.55(m,1H),6.82(s,1H),6.77(d,1H),2.79–2.66(m,1H),2.45–2.35(m,1H),1.56–1.39(m,2H).
Example 24-1 and example 24-2
5- (5-fluoro-4- ((1S, 2S) -2- (trifluoromethyl) cyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (5-fluoro-4- ((1R, 2R) -2- (trifluoromethyl) cyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000107
Resolution of example 24 (40 mg,0.11 mmol) via chiral column (C4 AS column) afforded example 24-1 (10 mg, r.t=2.68 min, yield: 25.0%) and example 24-2 (8 mg, r.t=2.90 min, yield: 20.0%). Chiral resolution conditions:
Figure PCTCN2021136249-APPB-000108
chiral analysis method:
Figure PCTCN2021136249-APPB-000109
example 24-1, MS m/z (ESI): 355.1[ M+1 ]] +
Example 24-2, MS m/z (ESI): 355.1[ M+1 ]] +
Example 25
5- (7-chloro-5-ethyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000110
Figure PCTCN2021136249-APPB-000111
First step
3, 6-dichloropyridazin-4-amine 25a (10 g,61.35 mmol), trimethylsilyyne (6.3 g,64.42 mmol), bis triphenylphosphine palladium dichloride (1.3 g,1.84 mmol), cuprous iodide (0.7 g,3.68 mmol) and triethylamine (31 g,306.75 mmol) were dissolved in acetonitrile (100 mL) under nitrogen. The reaction was heated to 60 ℃ and stirred for 5 hours. The reaction solution was concentrated, and the residue was separated by silica gel column chromatography (eluent system a) to give product 6-chloro-3- ((trimethylsilyl) ethynyl) pyridazin-4-amine 25b (11.3 g), yield: 81.9%.
MS m/z(ESI):226.1[M+1] + .
Second step
25b (5.9 g,26.22 mmol) and cuprous iodide (1 g,5.24 mmol) were dissolved in N, N-dimethylformamide (50 mL). The reaction was heated to 120 ℃ and stirred overnight. Cooled to room temperature, the reaction solution was poured into ice water, and filtered to give the crude compound 3-chloro-5H-pyrrolo [3,2-c ] pyridazine 25c (3.4 g), which was used in the next reaction without purification.
MS m/z(ESI):154.1[M+1] + .
Third step
25c (1 g,6.53 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (1.8 g,9.80 mmol), 1' -bis-diphenylphosphino ferrocene palladium dichloride (284 mg,0.65 mmol) and potassium carbonate (3.6 g,26.14 mmol) were dissolved in dioxane/water (v: v=8 mL/2 mL) under nitrogen. The reaction was heated to 120℃with microwaves and stirred for 1 hour. The reaction solution was filtered, the filtrate was concentrated, and the residue was separated by silica gel column chromatography (eluent system B) to give the product 3- (2, 4-dimethoxypyrimidin-5-yl) -5H-pyrrolo [3,2-c ] pyridazine 25d (500 mg), yield: 29.8%.
MS m/z(ESI):258.1[M+1] + .
Fourth step
25d (300 mg,1.17 mmol) was dissolved in N, N-dimethylformamide (5 mL) at room temperature, and N-chlorosuccinimide (156 mg,1.17 mmol) was added. The reaction was stirred at 50℃for 4 hours. The reaction mixture was concentrated, and methylene chloride (20 mL) was added to the residue to slurry, followed by filtration to give 7-chloro-3- (2, 4-dimethoxypyrimidin-5-yl) -5H-pyrrolo [3,2-c ] pyridazine 25e (210 mg), yield: 61.8%.
MS m/z(ESI):292.0[M+1] + .
Fifth step
25e (200 mg,0.68 mmol) was dissolved in N, N-dimethylformamide (5 mL). To the reaction solution were successively added ethyl iodide (319 mg,2.06 mmol) and potassium carbonate (284 mg,2.06 mmol). The reaction was stirred at 80℃overnight. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated to give the crude product 7-chloro-3- (2, 4-dimethoxypyrimidin-5-yl) -5-ethyl-5H-pyrrolo [3,2-c ] pyridazine 25f (230 mg), which was used in the next reaction without purification.
MS m/z(ESI):320.1[M+1] + .
Sixth step
25f (230 mg,0.72 mmol) was dissolved in hydrochloric acid (1M, 5 mL) and stirred overnight at room temperature. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (7-chloro-5-ethyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione 25 (12.48 mg). Yield: 5.9%.
MS m/z(ESI):292.1[M+1] + .
1 H NMR(400MHz,DMSO-d6)δ11.47(s,2H),8.45(s,1H),8.40(s,1H),8.12(s,1H),4.23(q,2H),1.38(t,3H).
Example 26
5- (7-bromo-5-ethyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000112
First step
25d (1.0 g,3.89 mmol) was dissolved in N, N-dimethylformamide (30 mL) at room temperature, and bromine (620 mg,3.89 mmol) was added dropwise. The reaction was stirred at room temperature for 4 hours. The reaction solution was concentrated, and methylene chloride (100 mL) was added to the residue and stirred. The reaction solution was filtered, and the obtained solid was the product 7-bromo-3- (2, 4-dimethoxypyrimidin-5-yl) -5H-pyrrolo [3,2-c ] pyridazine 26a (1.2 g), yield: 91.8%.
MS m/z(ESI):336.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ12.17(s,1H),8.93(s,1H),8.11(s,1H),8.05(s,1H),4.04(s,3H),4.00(s,3H).
Second step
26a (150 mg,0.45 mmol) was dissolved in N, N-dimethylformamide (2.5 mL) at room temperature, and iodoethane (209 mg,1.34 mmol) was added dropwise with stirring. The reaction was heated to 85 ℃ and stirred for 3 hours. To the reaction mixture was added water (20 mL), and the aqueous phase was extracted with ethyl acetate (30 mL. Times.2). The organic phase was washed with saturated sodium chloride solution, dried, concentrated and the residue was separated by silica gel column chromatography (eluent system a) to give the title product 7-bromo-3- (2, 4-dimethoxypyrimidin-5-yl) -5-ethyl-5H-pyrrolo [3,2-c ] pyridazine 26b (35 mg), yield: 21.5%.
MS m/z(ESI):366.0[M+1] + .
Third step
26b (35 mg,0.096 mmol) was dissolved in a mixture of methanol (0.5 mL) and hydrochloric acid (2M, 1 mL). The reaction was heated to 75 ℃ and stirred for 6 hours. The reaction solution was filtered, and the filtrate was separated by reverse phase HPLC to give the title product 5- (7-bromo-5-ethyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione 26 (11 mg), yield: 34%.
MS m/z(ESI):336.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.55-11.32(m,2H),8.43(s,1H),8.39(s,1H),8.14(s,1H),4.24(q,2H),1.38(t,3H).
Example 27
5- (7-chloro-5-isopropyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000113
First step
25e (100 mg,0.34 mmol) was dissolved in N, N-dimethylformamide (2 mL). To the reaction solution were added 2-iodopropane (175 mg,1.03 mmol) and potassium carbonate (142 mg,1.03 mmol) in this order. The reaction was stirred at 80℃overnight. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated to give the crude product 7-chloro-3- (2, 4-dimethoxypyrimidin-5-yl) -5-isopropyl-5H-pyrrolo [3,2-c ] pyridazine 27a (108 mg), which was used in the next reaction without purification.
MS m/z(ESI):334.0[M+1] + .
Second step
27a (108 mg,0.32 mmol) was dissolved in hydrochloric acid (1M, 5 mL) and stirred overnight at room temperature. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (7-chloro-5-isopropyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione 27 (17 mg). Yield: 17%
MS m/z(ESI):306.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.47(s,2H),8.47(s,1H),8.39(s,1H),8.25(s,1H),4.83–4.74(m,1H),1.47(d,6H).
Example 28
5- (3-bromo-8- ((1S, 2R) -2-isopropylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000114
First step
Example 9 (60 mg,0.19 mmol) and 1-bromopyrrolidine-2, 5-dione (34 mg,0.19 mmol) were dissolved in N, N-dimethylformamide (3 mL) and stirred. The reaction was stirred at 30℃for 48 hours. The reaction solution was filtered, and the filtrate was separated using reverse phase HPLC to prepare a separate (formic acid system) to give the title product 5- (3-bromo-8- ((1 s,2 r) -2-isopropylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 28 (55 mg), yield: 74.2%.
MS m/z(ESI):390.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.45(s,2H),8.02(s,1H),7.83(s,1H),7.39(s,1H),2.31-2.27(m,1H),1.43-1.39(m,2H),1.26-1.22(m,1H),1.13-1.10(m,1H),1.02-0.98(m,6H).
Example 29
5- (3-chloro-8- ((1S, 2R) -2-isopropylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000115
Figure PCTCN2021136249-APPB-000116
First step
Compound 9 (60 mg,0.19 mmol) and 1-chloropyrrolidine-2, 5-dione (25 mg,0.19 mmol) were dissolved in N, N-dimethylformamide (3 mL) and stirred. The reaction was stirred at 30℃for 48 hours. The reaction solution was filtered, and the filtrate was separated using reverse phase HPLC to prepare a separate (formic acid system) to give the title product 5- (3-chloro-8- ((1 s,2 r) -2-isopropylcyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 29 (50 mg), yield: 76.0%.
MS m/z(ESI):346.1[M+1] + .
1H NMR(400MHz,DMSO-d 6 )δ11.42(s,2H),8.03(s,1H),7.81(s,1H),7.39(s,1H),2.32–2.23(m,1H),1.48–1.37(m,2H),1.30–1.18(m,1H),1.16–1.07(m,1H),1.04–0.93(m,6H).
Example 30
5- (3-chloro-8- (3, 3-dimethylcyclopent-1-en-1-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000117
First step
21 (43 mg,0.13 mmol) N-chlorosuccinimide (18 mg,0.13 mmol) was dissolved in N, N-dimethylformamide (1 mL). The reaction was heated to 50℃and stirred for 3 hours. The reaction solution was filtered, and the filtrate was separated by reverse phase HPLC to give the product 5- (3-chloro-8- (3, 3-dimethylcyclopent-1-en-1-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 30 (27.9 mg), yield: 58.6%.
MS m/z(ESI):358.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.54(s,2H),8.07(s,1H),7.86(s,1H),7.68(s,1H),7.55(s,1H),2.84(t,2H),1.82(t,2H),1.18(s,6H).
Example 31
5- (4- ((1S, 2R) -2-isopropylcyclopropyl) pyrazolo [1,5-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000118
First step
methyl-1H-pyrazole-3-carboxylate 31a (5 g,39.65 mmol) was dissolved in dry tetrahydrofuran (200 mL) at 0deg.C under nitrogen and stirred. To the reaction solution was slowly added dropwise a solution of lithium bis (trimethylsilyl) amide (1 m,47.58 mmol). After the completion of the dropping, (aminooxy) diphenylphosphine oxide (11 g,47.58 mmol) was added to the reaction solution. The reaction was stirred at room temperature for a further 12 hours. Dichloromethane (50 mL) was added to the reaction solution, the reaction solution was filtered, the residue was washed with dichloromethane, the organic phase was concentrated to obtain a crude product, and the crude product was separated by silica gel column chromatography (eluent system a) to obtain methyl 1-amino-1H-pyrazole-5-carboxylate 31b (1.5 g), yield: 26%.
MS m/z(ESI):142.0[M+1] + .
Second step
31b (1.0 g,7.08 mmol) and methyl-3-chloro-3-carbonylpropionate (1.0 g,7.79 mmol) were dissolved in dichloromethane (10 mL) at room temperature and stirred. Triethylamine (2.2 g,21.26 mmol) was added to the reaction solution. The reaction was stirred at room temperature for 4 hours. Methanol (2 mL) was added to the reaction solution, the reaction solution was concentrated, and the crude product was separated by silica gel column chromatography (eluent system A) to give methyl 1- (3-methoxy-3-carbonylpropionylamino) -1H-pyrazole-5-carboxylate 31c (1.2 g), yield: 70%.
MS m/z(ESI):242.0[M+1] + .
Third step
31c (1.0 g,4.14 mmol) and potassium tert-butoxide (1.4 g,12.42 mmol) were dissolved in tetrahydrofuran (15 mL) and stirred at 0deg.C for 4 hours. The reaction mixture was concentrated, and water (15 mL) and formic acid (2 mL) were added to neutrality. The reaction solution was filtered to give methyl 4, 6-dicarbonyl-4, 5,6, 7-tetrahydropyrazolo [1,5-b ] pyridazine-5-carboxylate 31d (867 mg), yield: 74%
MS m/z(ESI):210.0[M+1] + .
Fourth step
31d (500 mg,2.38 mmol) was dissolved in a solution of sodium hydroxide (382 mg,9.56 mmol) in water (25 mL). The reaction solution was heated to 100℃and stirred for 12 hours. The reaction solution was cooled, and hydrochloric acid (1M) was added thereto to neutrality. The reaction solution was filtered to give pyrazolo [1,5-b ] pyridazine-4, 6-diol 31e (242 mg) in 67% yield.
MS m/z(ESI):152.0[M+1] + .
Fifth step
31e (200 mg,1.33 mmol), N-bis (trifluoromethanesulfonyl) aniline (520 mg,1.46 mmol) and triethylamine (401 mg,3.97 mmol) were dissolved in tetrahydrofuran (10 mL) at room temperature and stirred for 5 hours. Formic acid (1 mL) was added to the reaction solution, the reaction solution was concentrated, and the crude product was separated by silica gel column chromatography (eluent system a) to give the product 4-hydroxypyrazolo [1,5-b ] pyridazin-6-yl trifluoromethanesulfonic acid 31f (150 mg, white solid), yield: 40%.
MS m/z(ESI):283.9[M+1] + .
Sixth step
31f (100 mg,0.35 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (97 mg,0.53 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (29 mg,0.035 mmol) and cesium carbonate (228 mg,0.70 mmol) were dissolved in a mixture of 1, 4-dioxane (5 mL) and water (1 mL) under nitrogen. The reaction solution was heated to 100℃with microwaves and stirred for 1 hour. The reaction mixture was concentrated, and the crude product was separated by silica gel column chromatography (eluent system a) to give 31g (54 mg) of 6- (2, 4-dimethoxypyrimidin-5-yl) pyrazolo [1,5-b ] pyridazin-4-ol as a product, yield: 56%.
MS m/z(ESI):274.0[M+1] + .
Seventh step
31g (50 mg,0.18 mmol) of N-bis (trifluoromethanesulfonyl) aniline (72 mg,0.2 mmol) and triethylamine (56 mg,0.54 mmol) were dissolved in tetrahydrofuran (2 mL) and stirred at room temperature for 5 hours. Formic acid (0.1 mL) was added to the reaction solution, the reaction solution was concentrated, and the crude product was separated by silica gel column chromatography (eluent system a) to give the product 6- (2, 4-dimethoxypyrimidin-5-yl) pyrazolo [1,5-b ] pyridazin-4-yl trifluoromethanesulfonic acid 31h (50 mg), yield: 67%.
MS m/z(ESI):406.0[M+1] + .
Eighth step
31h (40 mg,0.1 mmol), (1S, 2R) -2-isopropylcyclopropane-1-carboxylic acid (62 mg,0.29 mmol), prepared as described in WO2019168744A1, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (8.16 mg,0.01 mmol) and cesium carbonate (65 mg,0.2 mmol) were dissolved in a mixture of 1, 4-dioxane (2 mL) and water (0.01 mL) under nitrogen. The reaction solution was heated to 100℃with microwaves and stirred for 1 hour. The reaction solution was filtered, the filter residue was washed with ethyl acetate, the organic phase was washed with water, saturated ammonium chloride, dried and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system a) to give the product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) pyrazolo [1,5-b ] pyridazine 31i (24 mg), yield: 72%.
MS m/z(ESI):340.1[M+1] + .
Ninth step
6- (2, 4-Dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyrazolo [1,5-b ] pyridazine 31i (20 mg,0.06 mmol) was dissolved in hydrochloric acid (1M, 2 mL) and stirred overnight at 70 ℃. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (4- ((1 s,2 r) -2-isopropylcyclopropyl) pyrazolo [1,5-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 31 (12 mg). Yield: 64 percent of
MS m/z(ESI):312.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.42(s,2H),8.02(d,2H),7.18(s,1H),6.98(d,1H),2.18–2.07(m,1H),1.31–1.19(m,2H),1.16–1.06(m,2H),1.01(t,6H).
Example 32
5- (4- (3, 3-dimethylcyclopent-1-en-1-yl) pyrazolo [1,5-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000119
First step
6- (2, 4-Dimethoxypyrimidin-5-yl) pyrazolo [1,5-b ] pyridazin-4-yl trifluoromethanesulfonic acid 31h (30 mg,0.07 mmol), 2- (3, 3-dimethylcyclopent-1-en-1-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (33 mg,0.15 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (6 mg, 0.0070 mmol) and cesium carbonate (46 mg,0.14 mmol) were dissolved in a mixture of 1, 4-dioxane (2 mL) and water (0.01 mL) under nitrogen. The reaction solution was heated to 100℃with microwaves and stirred for 1 hour. The reaction solution was filtered, the filter residue was washed with ethyl acetate, the organic phase was washed with water, saturated sodium chloride, dried and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system a) to give the product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- (3, 3-dimethylcyclopent-1-en-1-yl) pyrazolo [1,5-b ] pyridazine 32a (24 mg), yield: 21%.
MS m/z(ESI):352.1[M+1] + .
Second step
6- (2, 4-Dimethoxypyrimidin-5-yl) -4- (3, 3-dimethylcyclopent-1-en-1-yl) pyrazolo [1,5-b ] pyridazine 32a (21 mg,0.06 mmol) was dissolved in hydrochloric acid (1M, 2 mL) and stirred overnight at 70 ℃. The reaction solution was concentrated, and the residue was subjected to preparative separation (formic acid system) by reverse phase HPLC to give the title product 5- (4- (3, 3-dimethylcyclopent-1-en-1-yl) pyrazolo [1,5-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione 32 (13 mg). Yield: 67%.
MS m/z(ESI):324.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ8.08(s,2H),7.61(s,1H),7.06(d,1H),6.71(s,1H),2.84(t,2H),1.83(t,2H),1.18(d,6H).
Example 33
5- (5- (cyclopent-1-en-1-yl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000120
First step
Intermediate 2 (50 mg,0.19 mmol), 2- (cyclopent-1-en-1-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (73 mg,0.38 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (16 mg,0.019 mmol) and cesium carbonate (124 mg,0.38 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 ml) under nitrogen. The reaction mixture was heated to 100℃with microwaves and stirred for 1 hour. To the reaction was added saturated sodium chloride (10 mL), and the aqueous phase was extracted with ethyl acetate (10 mL. Times.3). The organic phases were combined, dried and concentrated to give crude product which was separated by silica gel column chromatography (eluent system B) to give product 4- (cyclopent-1-en-1-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine 19a (42 mg), yield: 74%.
MS m/z(ESI):299.1[M+1] + .
Second step
33a (30 mg,0.10 mmol) was dissolved in hydrochloric acid (1M, 2 mL). The reaction was heated to 70 ℃ and stirred overnight. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (5- (cyclopent-1-en-1-yl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 33 (18 mg). Yield: 67%.
MS m/z(ESI):271.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.41(s,2H),8.31(s,1H),8.07(s,1H),6.32(d,1H),2.75–2.65(m,5H),2.57(ddt,2H),1.97(p,2H).
Example 34
5- (5- (3, 3-dimethylcyclopent-1-en-1-yl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000121
First step
34a (987 mg,8.80 mmol) was dissolved in tetrahydrofuran (20 mL) and cooled to-78deg.C and stirred for 5 min. Lithium bis (trimethylsilylamide) (1M, 8 mL) was added dropwise to the above system, and the reaction was stirred at-78℃for 30 minutes. A solution of N-phenylbis (trifluoromethanesulfonyl) imide (3.14 g,8.80 mmol) in tetrahydrofuran (10 mL) was then added dropwise to the reaction. The reaction was slowly returned to room temperature and stirred overnight. To the reaction mixture was added a saturated aqueous ammonium chloride solution (15 mL). The organic phase was separated and the aqueous phase was extracted with methyl tert-butyl ether (20 mL. Times.2). The organic phases were combined and washed successively with 5% aqueous sodium carbonate, saturated sodium chloride solution, dried and concentrated to give the crude product 3, 3-dimethylcyclopent-1-en-1-yl triflic acid 34b (1.9 g), which was used directly in the next reaction without purification.
1 H NMR(400MHz,CDCl 3 )δ5.47(s,1H),2.66-2.58(m,2H),1.81(t,J=7.8Hz,2H),1.12(s,6H).
Second step
34b (500 mg,2.05 mmol), pinacol biborate (624 mg,2.46 mmol), tris (dibenzylideneacetone) dipalladium (93 mg,0.102 mmol), 2-dicyclohexylphosphine-2 ',4',6' -triisopropylbiphenyl (195 mg, 0.09 mmol) and potassium acetate (402 mg,4.09 mmol) were dissolved in anhydrous 1, 4-dioxane (9 mL) under nitrogen. The reaction was heated to 100deg.C and stirred for 2 hours. The reaction solution was filtered, and the filtrate was concentrated to give the crude product 2- (3, 3-dimethylcyclopent-1-en-1-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan 34c (454 mg), which was used in the next reaction without purification.
Third step
Intermediate 2 (127 mg,0.48 mmol), 34c (127 mg,0.57 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride (35 mg,0.048 mmol), cesium carbonate (310 mg,0.95 mmol) was dissolved in a mixed solvent of 1, 4-dioxane (2 mL) and water (0.5 mL) under nitrogen. The reaction was heated to 100deg.C and stirred for 3 hours. The organic phase was separated and the aqueous phase was extracted with ethyl acetate (2 mL. Times.2). The combined organic phases were dried, concentrated and the crude product was separated by silica gel column chromatography (eluent system a) to give the product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- (3, 3-dimethylcyclopent-1-en-1-yl) -3-methylpyridazine 34d (140 mg), yield: 90%.
MS m/z(ESI):327.2[M+1] + .
Fourth step
34d (140 mg,0.43 mmol) was dissolved in a mixed solvent of hydrochloric acid (2M, 2.5 mL), methanol (1 mL) and tetrahydrofuran (1 mL), and the reaction was heated to 68℃and stirred for 3 hours. The reaction solution was filtered, and the filtrate was separated by reverse phase HPLC to give the title product 5- (5- (3, 3-dimethylcyclopent-1-en-1-yl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 34 (50.8 mg), yield: 39.7%.
MS m/z(ESI):299.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,2H),8.29(s,1H),8.05(s,1H),6.11(s,1H),2.78-2.71(m,2H),2.68(s,3H),1.79(t,2H),1.15(s,6H).
Example 35
5- (8- (2- (fluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000122
First step
35a (2 g,11.74 mmol), 4, 5-tetramethyl-1, 3, 2-dioxaborolan (1.65 g,12.92 mmol), zirconocene chloride hydride (303 mg,1.77 mmol) and triethylamine (119 mg,1.17 mmol) were dosed into a reaction flask. The reaction was stirred at 60℃for 12 hours under nitrogen. The reaction solution was cooled to room temperature, filtered, and the cake was washed with methylene chloride (10 mL), and the filtrate was concentrated under reduced pressure, and the obtained crude compound was separated by silica gel column chromatography (eluent system a) to give the product tert-butyldimethyl ((3- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) allyl) oxo) silane 35b (2.2 g), yield: 63%.
1 H NMR(400MHz,CDCl 3 )δ6.67(dt,1H),5.74(dt,1H),4.24(dd,2H),1.26(s,12H),0.90(s,9H),0.05(s,6H).
Second step
To a mixture of 20% aqueous potassium hydroxide (2 mL) and diethyl ether (2 mL) was added N-methyl-N-nitrosourea (500 mg,4.8 mmol) under ice-bath conditions, and the mixture was stirred at this temperature for 1 hour. The organic phase of the above mixed solution was added to a solution of 35b (300 mg,1.0 mmol) in diethyl ether (2 mL) under ice-bath conditions, followed by addition of palladium acetate (22 mg,0.01 mmol) to the reaction solution, and the reaction was stirred under ice-bath conditions for 30 minutes. The reaction solution was filtered, and the cake was washed with methylene chloride (5 mL. Times.3). The filtrate was concentrated, and the resulting crude compound was separated by silica gel column chromatography (eluent system a) to give the product tert-butyldimethyl ((2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) cyclopropyl) methoxy) silane 35c (180 mg), yield: 57%.
1 H NMR(400MHz,CDCl 3 )δ3.59(dd,1H),3.45(dd,1H),1.25(m,1H),1.21(s,12H),0.88(s,9H),0.68(ddd,1H),0.54(ddd,1H),0.04(s,6H),-0.17–-0.30(m,1H).
Third step
35c (150 mg,0.48 mmol), 4-bromo-6-chloropyridazin-3-amine (99 mg,0.48 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (39 mg,0.048 mmol) and potassium carbonate (132 mg,0.96 mmol) were dissolved in 1, 4-dioxane/water (v: v=4:1, 2 ml) and the reaction was stirred under nitrogen at 80℃for 1 hour under microwave conditions. Saturated sodium chloride (10 mL) was added to the reaction solution, ethyl acetate (10 ml×3) was extracted, the organic phases were combined, dried, and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system a) to give the product 4- (2- (((tert-butyldimethylsilyl) oxo) methyl) cyclopropyl) -3, 6-dichloropyridazine 35d (91 mg), yield: 57%.
MS m/z(ESI):333.0[M+1] + .
Fourth step
35d (350 mg,1.05 mmol) was dissolved in tetrahydrofuran and tetra-n-butylammonium fluoride (1M in THF,1.58mmol) was added. The reaction solution was stirred at 25℃for 4 hours. The reaction solution was concentrated to obtain a crude product, which was separated by silica gel column chromatography (eluent system a) to obtain product (2- (3, 6-dichloropyridazin-4-yl) cyclopropyl) methanol 35e (210 mg), yield: 90%.
MS m/z(ESI):219.0[M+1] + .
Fifth step
35e (200 mg,0.91 mmol) was dissolved in dry dichloromethane (3 mL) and the reaction was stirred at-78deg.C under nitrogen and diethylaminosulfur trifluoride (221 mg,1.37 mmol) was added and the reaction was stirred at-78deg.C for a further 4 hours. The reaction solution was quenched with saturated sodium bicarbonate solution until no bubbles were generated, dichloromethane (10 ml×3) was extracted, the organic phases were combined, dried, and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system a) to give the product 3, 6-dichloro-4- (2- (fluoromethyl) cyclopropyl) pyridazine 35f (83 mg), yield: 41%.
MS m/z(ESI):220.9[M+1]+.
Sixth step
35f (53 mg,0.24 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (66 mg,0.36 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (19 mg,0.024 mmol) and cesium carbonate (156 mg,0.48 mol) were dissolved in 1, 4-dioxane/water (4:1, 2 mL) and the reaction was stirred under nitrogen at 100deg.C under microwave conditions for 1 hour. Saturated sodium chloride (10 mL) was added to the reaction, ethyl acetate (10 mL. Times.3) was used to extract, the organic phases were combined, dried, and concentrated to give crude product, which was separated by silica gel column chromatography (eluent system A) to give 35g (53 mg) of 3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- (2- (fluoromethyl) cyclopropyl) pyridazine as a product, yield: 68%.
MS m/z(ESI):325.0[M+1] + .
Seventh step
35g (198 mg,0.61 mmol), diphenylazomethine (331 mg,1.83 mmol), tris (dibenzylideneacetone) dipalladium (56 mg,0.06 mmol), 1 '-binaphthyl-2, 2' -bisdiphenylphosphine (76 mg,0.121 mmol) and cesium carbonate (390 mg,1.22 mmol) were dissolved in 1, 4-dioxane (10 mL). The reaction was stirred at 120℃for 12 hours under nitrogen. The reaction solution was filtered, the filter residue was washed with ethyl acetate, the organic phase was washed with water, saturated sodium chloride, dried and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system a) to give the product N- (6- (2, 4-dimethoxypyrimidin-5-yl) -4- (2- (fluoromethyl) cyclopropyl) pyridazin-3-yl) -1, 1-diphenylazonimine 35h (224 mg), yield: 78%.
MS m/z(ESI):470.1[M+1] + .
Eighth step
35h (198 mg,0.42 mmol) was dissolved in tetrahydrofuran (4 mL) and methanol (4 mL) and hydrochloric acid (1M, 3 mL) was added. The reaction solution was heated to 70℃and stirred for 5 hours. The reaction solution was concentrated, and the crude product was separated by silica gel column chromatography (eluent system B) to give the product 5- (6-amino-5- (2- (fluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 35i (72 mg). Yield: 60 percent of
MS m/z(ESI):278.0[M+1] + .
Ninth step
35i (44 mg,0.16 mmol), p-toluene sulfonic acid (32 mg,0.19 mmol) and 2-chloro-1, 1-dimethoxyethane (60 mg,0.48 mmol) were dissolved in isopropanol (2 mL). The reaction was stirred at 100℃for 10 hours under nitrogen. The reaction solution was concentrated, and reversed phase HPLC was performed to isolate (formic acid system) to give the product 5- (8- (2- (fluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 35 (32 mg), yield: 66%.
MS m/z(ESI):302.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ8.19(s,1H),8.02(s,1H),7.67(s,1H),7.43(s,1H),4.66–4.32(m,2H),2.05–1.93(m,1H),1.68–1.56(m,1H),1.28–1.20(m,2H).
Example 36
5- (8- (2, 2-difluorocyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000123
First step
3, 6-dichloropyridazine (1.0 g,6.76 mmol) was suspended in 30mL of deionized water, 2-difluorocyclopropane-1-carboxylic acid (284 mg,6.76 mmol) and concentrated sulfuric acid (1 mL) were added and heated to 70deg.C under nitrogen. A solution of silver nitrate (228 mg,1.34 mmol) in water (1 mL) was added followed by dropwise addition of a solution of ammonium persulfate (4.5 g,20.1 mmol) in water (15 mL) over about 15 minutes and the reaction was continued at 70℃for 1 hour. The reaction mixture was cooled to room temperature, neutralized to pH 8-9 with aqueous ammonia, and extracted with ethyl acetate (60 mL. Times.3). The organic phases were combined, washed successively with water (60 mL), saturated sodium chloride solution (60 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure, and the resulting crude compound was separated by silica gel column chromatography (eluent system B) to give the product 3, 6-dichloro-4- (2, 2-difluorocyclopropyl) pyridazine 36B (400 mg), yield: 26%.
MS m/z(ESI):224.9[M+H] + .
Second step
36b (400 mg,1.78 mmol), 2, 4-dimethoxypyrimidine-5-boronic acid (327 mg,1.78 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (145 mg,0.177 mmol) and cesium carbonate (1.16 g,3.56 mmol) were dissolved in 1, 4-dioxane/water (v: v=4:1, 2 ml). The reaction solution was stirred for 1 hour at 70℃under microwave conditions under nitrogen protection. The reaction mixture was cooled to room temperature, and 25mL of water and ethyl acetate (25 mL. Times.3) were added thereto for extraction. The organic phases were combined, washed successively with water (25 mL), saturated sodium chloride solution (25 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure, and the resulting crude compound was separated by silica gel column chromatography (eluent system a) to give the product 3-chloro-4- (2, 2-difluorocyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine 36c (300 mg), yield: 51%.
MS m/z(ESI):329.0[M+H] + .
Third step
36c (200 mg,0.61 mmol), diphenylazomethine (331 mg,1.83 mmol), tris (dibenzylideneacetone) dipalladium (56 mg,0.06 mmol), 1 '-binaphthyl-2, 2' -bisdiphenylphosphine (76 mg,0.121 mmol) and cesium carbonate (390 mg,1.22 mmol) were dissolved in 1, 4-dioxane (10 mL). The reaction was stirred at 120℃for 12 hours under nitrogen. The reaction solution was filtered, the filter residue was washed with ethyl acetate, the organic phase was washed with water, saturated sodium chloride, dried and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system a) to give the product N- (4- (2, 2-difluorocyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazin-3-yl) -1, 1-diphenylazonimine 36d (210 mg), yield: 72%.
MS m/z(ESI):474.1[M+1] + .
Fourth step
36d (200 mg,0.422 mmol) was dissolved in tetrahydrofuran (4 mL) and methanol (4 mL), and hydrochloric acid (1M, 3 mL) was added. The reaction solution was heated to 70℃and stirred for 5 hours. The reaction solution was concentrated, and the crude product was separated by silica gel column chromatography (eluent system B) to give the product 5- (6-amino-5- (2, 2-difluorocyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 36e (72 mg). Yield: 60 percent of
MS m/z(ESI):282.0[M+1] + .
Fifth step
36e (45 mg,0.16 mmol), p-toluenesulfonic acid (32 mg,0.19 mmol) and 2-chloro-1, 1-dimethoxyethane (60 mg,0.48 mmol) were dissolved in isopropanol (2 mL). The reaction was stirred at 100℃for 10 hours under nitrogen. The reaction solution was concentrated, and reversed phase HPLC was performed to isolate (formic acid system) to give the product 5- (8- (2, 2-difluorocyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 36 (30 mg), yield: 60%.
MS m/z(ESI):306.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.55(s,2H),8.38(s,1H),8.04(d,1H),7.90(s,1H),7.64(s,1H),2.37(m,3H).
Example 37
5- (8-cyclopentylimidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000124
The compound 5- (8-cyclopentylimidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione 37 was obtained according to the method of example 36.
MS m/z(ESI):298.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ8.19(s,1H),7.99(s,1H),7.69(s,1H),7.57(s,1H),3.61–3.48(m,1H),2.18–2.06(m,2H),1.87–1.70(m,6H).
Example 38
5- (8-cyclobutylimidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000125
The compound 5- (8-cyclobutylimidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione 38 was obtained according to the method of example 36.
MS m/z(ESI):284.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ8.40(d,1H),8.07(d,1H),7.97(s,1H),7.85(s,1H),4.00(t,1H),2.48–2.40(m,2H),2.36–2.29(m,2H),2.19–2.06(m,1H),2.00–1.84(m,1H).
Example 39
5- (8- (3, 3-dimethylcyclobutyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000126
The compound 5- (8- (3, 3-dimethylcyclobutyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione 39 was obtained according to the method of example 36.
MS m/z(ESI):312.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.37(s,2H),8.19(s,1H),8.02(s,1H),7.67(s,1H),7.59(s,1H),4.14–3.81(m,1H),2.26–2.17(m,2H),2.18–2.09(m,2H),1.30(s,3H),1.13(s,3H).
Example 40
5- (8- (3, 3-difluorocyclobutyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000127
The compound 5- (8- (3, 3-difluorocyclobutyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione 40 was obtained according to the method of example 36.
MS m/z(ESI):320.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.38(s,2H),8.25(s,1H),8.04(s,1H),7.73(s,1H),7.66(s,1H),3.96–3.76(m,1H),3.26–2.97(m,4H).
Example 41
5- (6-amino-5- (3, 3-difluorocyclobutyl) pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000128
The compound 5- (6-amino-5- (3, 3-difluorocyclobutyl) pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione 41 was obtained according to the method of example 40.
MS m/z(ESI):296.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ7.98(s,1H),7.76(s,1H),6.26(s,2H),3.42–3.23(m,1H),3.21–3.01(m,2H),2.73–2.53(m,2H).
Example 42
5- (8- (cyclohex-1-en-1-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000129
First step
42a (553mg, 2.64 mmol), 2- (cyclohexen-1-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborolan (500 mg,2.40 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (176 mg,0.24 mmol) and potassium carbonate (264 mg,4.81 mmol) were dissolved in a mixed solvent of 1, 4-dioxane (8 mL) and water (2 mL) and nitrogen was replaced for 2 minutes. The reaction was heated to 100deg.C and stirred for 2 hours. The reaction was cooled to room temperature, concentrated, and the residue was separated by silica gel column chromatography (eluent system a) to give product 6-chloro-4- (cyclohex-1-en-1-yl) pyridazin-3-amine 42b (490 mg), yield: 97%.
MS m/z(ESI):210.1[M+1] + .
Second step
42b (490 mg,2.34 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (516 mg,2.80 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (171 mg,0.23 mmol), potassium carbonate (640 mg,4.67 mmol) was dissolved in a mixed solvent of 1, 4-dioxane (8 mL) and water (2 mL), nitrogen was substituted for 2 minutes, and the reaction was heated to 100℃with microwaves and stirred for 1 hour. The reaction was cooled to room temperature and concentrated. The residue was separated by silica gel column chromatography (eluent system a) to give product 4- (cyclohex-1-en-1-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) pyridazin-3-amine 42c (597 mg), yield: 81.5%.
MS m/z(ESI):314.1[M+1] + .
Third step
42c (120 mg,0.38 mmol), 2-chloro-1, 1-dimethoxy-ethane (95 mg,0.77 mmol), p-toluenesulfonic acid (165 mg,0.96 mmol) was dissolved in a mixed solvent of isopropanol (1 mL) and N, N-dimethylformamide (0.5 mL). The reaction was heated to 100deg.C and stirred for 16 hours. The reaction was cooled to room temperature and filtered. The filter cake was prepared by reverse phase HPLC to isolate (formic acid system) to give the product 5- (8- (cyclohex-1-en-1-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 42 (20 mg), yield: 16.9%.
MS m/z(ESI):310.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.41(s,2H),8.20(d,1H),8.04(s,1H),7.82-7.76(m,1H),7.70(d,1H),7.65(s,1H),2.51-2.49(m,2H),2.37-2.30(m,2H),1.85-1.74(m,2H),1.70-1.61(m,2H).
Example 43
5- (7-chloro-5-isobutyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000130
The title product 5- (7-chloro-5-isobutyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione 43 was obtained by reference to the synthetic method of example 25.
MS m/z(ESI):320.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.52-11.48(m,2H),8.49(s,1H),8.37(d,1H),8.16(s,1H),4.04(d,2H),2.16-2.13(m,1H),0.85(d,6H).
Example 44
5- (5-ethyl-7-methyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000131
The title product 5- (5-ethyl-7-methyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione 44 was obtained by reference to the synthetic method of example 25.
MS m/z(ESI):272.1[M+1] + .
1 H NMR(400MHz,DMSO)δ12.06(s,1H),11.84(s,1H),8.69(s,1H),8.40(d,1H),8.19(s,1H),4.33(q,2H),2.40(s,3H),1.40(t,3H)。
Example 45
5- (4- ((1S, 2R) -2-isopropylcyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000132
The title product 5- (4- ((1 s,2 r) -2-isopropylcyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1 h,3 h) -dione 45 was obtained by the method of synthesis of example 24.
MS m/z(ESI):311.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.29(s,2H),7.90(s,1H),7.78(s,1H),6.83–6.79(m,1H),6.75–6.71(m,1H),6.67(s,1H),2.08–1.99(m,1H),1.30–1.16(m,1H),1.16–1.09(m,1H),1.09–0.95(m,8H).
Example 46
5- (4- ([ 1,1' -bis (cyclopropane) ] -2-yl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000133
The title product 5- (4- ([ 1,1' -bis (cyclopropane) ] -2-yl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1 h,3 h) -dione 46 was obtained by the method of synthesis of example 24.
MS m/z(ESI):309.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ7.88(s,1H),7.78(s,1H),6.81(dd,1H),6.77–6.60(m,2H),2.13–1.86(m,1H),1.38–1.22(m,1H),1.12–1.04(m,1H),1.03–0.92(m,2H),0.51–0.37(m,2H),0.29–0.12(m,2H).
Examples 46-1 and 46-2
5- (4- ((1R, 2S) - [1,1 '-bis (cyclopropane) ] -2-yl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (4- ((1S, 2R) - [1,1' -bis (cyclopropane) ] -2-yl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000134
The resolution procedure of example 24 was followed to give the title product 5- (4- ((1 r,2 s) - [1,1 '-bis (cyclopropane) ] -2-yl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1 h,3 h) -dione 46-1 and 5- (4- ((1 s,2 r) - [1,1' -bis (cyclopropane) ] -2-yl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1 h,3 h) -dione 46-2.
Example 46-1: MS m/z (ESI): 309.1[ M+1 ]] +
Example 46-2: MS m/z (ESI): 309.1[ M+1 ]] +
Example 47
5- (4- (2- (trifluoromethyl) cyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000135
The title product 5- (4- (2- (trifluoromethyl) cyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione 47 was obtained according to the method of example 24.
MS m/z(ESI):337.1[M+1] +
Examples 47-1 and 47-2
5- (4- ((1S, 2S) -2- (trifluoromethyl) cyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (4- ((1R, 2R) -2- (trifluoromethyl) cyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000136
Referring to the synthetic method of example 24, the title product 5- (4- ((1 s,2 s) -2- (trifluoromethyl) cyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1 h,3 h) -dione 47-1 and 5- (4- ((1 r,2 r) -2- (trifluoromethyl) cyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1 h,3 h) -dione 47-2 was obtained by chiral resolution.
Example 47-1: MS m/z (ESI): 337.1[ M+1 ]] +
1 H NMR(400MHz,CD 3 OD)δ8.00(s,1H),7.75(s,1H),6.94–6.90(m,1H),6.88–6.83(m,1H),6.72–6.66(m,1H),2.64–2.58(m,1H),2.22–2.17(m,1H),1.47(m,2H).
Example 47-2: MS m/z (ESI): 337.1[ M+1 ]] + .
1 H NMR(400MHz,CD 3 OD)δ8.02(s,1H),7.78(d,1H),6.94(s,1H),6.89(d,1H),6.71(d,1H),2.67–2.60(m,1H),2.26–2.19(s,1H),1.56–1.48(m,2H).
Example 48
5- (5-fluoro-4- ((1S, 2R) -2-isopropylcyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000137
The title product 5- (5-fluoro-4- ((1 s,2 r) -2-isopropylcyclopropyl) pyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1 h,3 h) -dione 48 was obtained according to the synthetic method of example 24.
MS m/z(ESI):329.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.37(s,2H),7.87(s,1H),7.69–7.52(m,1H),6.73(d,1H),6.56(s,1H),2.12(d,1H),1.29–1.11(m,3H),1.08–0.96(m,7H).
Example 49
5- (4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -5-fluoropyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000138
The title product 5- (4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -5-fluoropyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1 h,3 h) -dione 49 was obtained by the synthesis method of example 24.
MS m/z(ESI):327.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ7.94(s,1H),7.45(t,J=3.7Hz,1H),6.65–6.46(m,2H),2.22–2.12(m,1H),1.38–1.31(m,1H),1.17–1.08(m,1H),1.06–0.93(m,2H),0.54–0.39(m,2H),0.27–0.15(m,2H).
Examples 49-1 and 49-2
5- (4- ((1R, 2S) - [1,1 '-bis (cyclopropane) ] -2-yl) -5-fluoropyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (4- ((1S, 2R) - [1,1' -bis (cyclopropane) ] -2-yl) -5-fluoropyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000139
The resolution procedures of example 24-1 and example 24-2 were referenced to give the title product 5- (4- ((1R, 2S) - [1,1 '-bis (cyclopropane) ] -2-yl) -5-fluoropyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione 49-1 and 5- (4- ((1S, 2R) - [1,1' -bis (cyclopropane) ] -2-yl) -5-fluoropyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione 49-2.
Example 49-1: MS m/z (ESI): 327.1[ M+1 ]] +
Example 49-2, MS m/z (ESI): 327.1[ M+1 ]] +
Example 50
5- (4- ((1S, 2S) -2-ethylcyclopropyl) -5-fluoropyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000140
The title product 5- (4- ((1 s,2 s) -2-ethylcyclopropyl) -5-fluoropyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1 h,3 h) -dione 50 was obtained according to the synthetic method of example 24.
MS m/z(ESI):315.1[M+1] + .
1 H NMR(400MHz,CD 3 OD)δ7.95(s,1H),7.50–7.41(m,1H),6.63–6.53(m,2H),2.18–2.08(m,1H),1.55–1.44(m,2H),1.27–1.15(m,2H),1.05(t,3H),0.99–0.94(m,1H).
Example 51
5- (4- (3, 3-dimethylcyclopent-1-en-1-yl) -5-fluoropyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000141
The title product 5- (4- (3, 3-dimethylcyclopent-1-en-1-yl) -5-fluoropyrrolo [1,2-b ] pyridazin-2-yl) pyrimidine-2, 4 (1H, 3H) -dione 51 was obtained by the synthetic method of example 24.
MS m/z(ESI):341.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.41(s,2H),7.93(s,1H),7.69(t,1H),7.02(s,1H),6.75(d,1H),6.31–6.25(m,1H),2.78(t,2H),1.81(t,2H),1.15(s,6H).
Example 52
5- (4- ((1R, 2S) - [1,1' -bis (cyclopropane) ] -2-yl) pyrazolo [1,5-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000142
The title product 5- (4- ((1 r,2 s) - [1,1' -bis (cyclopropane) ] -2-yl) pyrazolo [1,5-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 52 was obtained by the method of synthesis example 31.
MS m/z(ESI):310.1[M+1] +
1H NMR(400MHz,DMSO-d6)δ11.47(s,2H),8.02(d,1H),8.01(s,1H),7.19(s,1H),6.94(d,1H),2.12–2.03(m,1H),1.46–1.36(m,1H),1.20–1.12(m,1H),1.10–0.96(m,2H),0.56–0.42(m,2H),0.28–0.17(m,2H).
Example 53
5- (5- ([ [1,1' -bis (cyclopropane) ] -2-yl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000143
Figure PCTCN2021136249-APPB-000144
First step
To a mixture of 2mL of 20% aqueous potassium hydroxide and 2mL of diethyl ether was added N-methyl-N-nitrosourea (500 mg,4.8 mmol) in ice bath, and the mixture was stirred at this temperature for 1 hour. The organic phase of the above mixed solution was added to 53a (200 mg,1.0 mmol) dissolved in 2mL of diethyl ether under ice bath conditions, followed by addition of palladium acetate (22 mg,0.01 mmol) to the reaction solution, which was stirred under ice bath conditions for 30 minutes. The reaction solution was filtered, the filter cake was washed with dichloromethane (5 ml×3), the filtrate was concentrated, and the product 2- ([ [1,1' -bis (cyclopropane) ] -2-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborane 53B (182 mg) was isolated by silica gel column chromatography (eluent system B), yield: 84.8%.
1 H NMR(400MHz,CDCl 3 ):δ1.21(s,12H),1.02(ddd,J=10.5,8.6,6.0Hz,1H),0.85–0.72(m,1H),0.57(ddd,J=7.9,6.4,3.5Hz,1H),0.45–0.24(m,3H),0.16–0.01(m,2H),-0.36(dt,J=9.6,5.9Hz,1H).
Second step
53b (110 mg,0.53 mmol), intermediate 2 (100 mg,0.035 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (28 mg,35 umol) and cesium carbonate (284 mg,0.88 mmol) were dissolved in 1, 4-dioxane/water (v: v=4:1, 2 ml), N 2 Stirring the reaction solution at 100 ℃ under the protection of microwave condition for reactionAnd 1 hour. Saturated sodium chloride (10 mL) is added into the reaction solution, dichloromethane (10 mL multiplied by 3) is added for extraction, organic phases are combined, dried and concentrated to obtain crude products, and the crude products are separated by silica gel column chromatography (eluent system B) to obtain the product 4- ([ 1,1' -bis (cyclopropane)]-2-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine 53c (100 mg), yield: 85.3%.
MS m/z(ESI):313.3[M+1] + .
Third step
53c (100 mg,0.32 mmol) was dissolved in 2mL of 1M aqueous hydrochloric acid and the reaction was stirred at 70℃for 1 hour. The reaction solution was concentrated, and reversed phase HPLC was prepared to isolate (formic acid system) the title product 5- (5- ([ [1,1' -bis (cyclopropane) ] -2-yl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 53 (38 mg), yield: 32.5%.
MS m/z(ESI):285.3[M+1] + .
1 H NMR(400MHz,DMSO-d 6 ):δ8.03(s,1H),7.49(s,1H),2.40–2.10(m,3H),1.57(dd,J=12.0,6.4Hz,1H),0.87(dd,J=8.5,3.5Hz,1H),0.83–0.75(m,1H),0.72(t,J=7.1Hz,2H),0.35–0.16(m,2H),0.01(m,2H).
Resolution of example 53
5- (5- ((1 r,2 s) - [1,1 '-bis (cyclopropane) ] -2-yl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione and 5- (5- ((1 s,2 r) - [1,1' -bis (cyclopropane) ] -2-yl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000145
Resolution of example 53 (28 mg,0.089 mmol) via chiral column (OZ column) afforded the title product 53-1 (6.54 mg, r.t= 5.722 min), yield: 24.2%;53-2 (5.4 mg, R.T = 3.934 min), yield: 22.4%.
MS m/z(ESI):53-1,285.3[M+1] + ;53-2,285.3[M+1] + .
Example 54
5- (5- (2- (tert-butyl) cyclopropyl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000146
First step
To a mixture of 3, 3-dimethyl-1-butyne 54a (5 g,60.87 mmol) and triethylamine (616 mg,6.09 mmol) was added Cp 2 Zr. HCl (1.57 g,6.09 mmol) and pinacol borane (7.79 g,60.87 mmol), and the mixture was stirred under nitrogen at 60℃for 12 hours. After cooling, the reaction solution was concentrated, and the resulting residue was purified by silica gel column chromatography with eluent system B to give product (E) -2- (3, 3-dimethyl-1-en-1-yl) -4, 5-tetramethyl-1, 3, 2-dioxolane 54B (8 g), yield: 63.5%.
1 H NMR(400MHz,CDCl 3 ):δ6.64(d,1H),5.35(d,1H),1.27(s,12H),1.02(s,9H).
Second step
To a mixture of 20% aqueous potassium hydroxide (10 mL) and methylene chloride (10 mL) at 0deg.C was slowly added 1-methyl-1-nitrosourea (1.55 g,15 mmol), and after stirring for 30 minutes, the lower yellow solution was slowly added dropwise to a solution of (E) -2- (3, 3-dimethyl-1-en-1-yl) -4, 5-tetramethyl-1, 3, 2-dioxolane 54b (210 mg,1 mmol) in methylene chloride (5 mL) at 0deg.C, and stirring was continued for 30 minutes at 25deg.C. The reaction solution was filtered, dried and concentrated to give 2- (2- (tert-butyl) cyclopropyl) -4, 5-tetramethyl-1, 3, 2-dioxolane 54c (200 mg), yield: 80.1%.
1 H NMR(400MHz,CDCl 3 ):δ1.21(s,12H),0.93–0.90(m,1H),0.82(s,9H),0.52(dd,2H),-0.23–-0.30(m,1H).
Third step
To a system of 2- (2- (tert-butyl) cyclopropyl) -4, 5-tetramethyl-1, 3, 2-dioxolane 54c (200 mg,0.89 mmol) and 4-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine intermediate 2 (238 mg,0.89 mmol) in 1, 4-dioxane (3 mL) and water (0.6 mL) was added Pd (dppf) Cl 2 *DCM (73 mg,0.09 mmol) and cesium carbonate (552 mg,1.78 mmol). The mixture was reacted under nitrogen at 100℃for 1 hour in a microwave. After cooling the reaction, 10mL of water was added, extracted with ethyl acetate (15 ml×3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give product 4- (2- (tert-butyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine 54d (30 mg), yield: 10.2%.
MS m/z(ESI):329.0[M+1] + .
Fourth step
To a round bottom flask was added 4- (2- (tert-butyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine 54d (30 mg,0.09 mmol) and hydrochloric acid (1M, 2 mL) and the mixture was stirred at 70℃for 16 h. The reaction solution was cooled and concentrated, and the residue was purified by reverse phase preparative HPLC to give the title product 5- (5- (2- (tert-butyl) cyclopropyl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 2 (13.5 mg), yield: 48.7%.
MS m/z(ESI):301.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 ):δ11.74(s,1H),11.62(s,1H),8.34(d,1H),7.90(s,1H),2.76(s,3H),2.01(d,1H),1.19(d,2H),1.06(d,1H),0.92(s,9H).
Example 55
5- (6-methyl-5- (2- (trifluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000147
Figure PCTCN2021136249-APPB-000148
First step
55a (1.5 g,15.14 mmol) was dissolved in a mixed solution of 5mL of methyl tert-butyl ether and 2mL of water, and 2mL of an aqueous solution of sodium nitrite (1.15 g,16.66 mmol) was added to the reaction solution under ice-bath conditions. The reaction solution was stirred at room temperature for 3 hours. The reaction liquid and water phase are separated, and the organic phase is directly used for the next reaction. Product yield reference (WO 2015052226 A1) estimates, product 2-diazo-1, 1-trifluoroethane 55b (700 mg) yield: 48.6%.
Second step
Palladium acetate (139.93 mg,0.625 mmol) was added to a solution of 55b in methyl tert-butyl ether (10 mL,700mg,6.25 mmol), and then 4, 5-tetramethyl-2-vinyl-1, 3, 2-dioxaborane (865.91 mg,5.62 mmol) was added to the reaction solution, which was stirred at room temperature for 2 hours. The reaction solution was filtered, and the filtrate was concentrated to give a crude product, which was purified by silica gel column chromatography (eluent system B) to give the product 4, 5-tetramethyl-2- (2- (trifluoromethyl) cyclopropyl) -1,3, 2-dioxaborane 55c (0.7 g), yield: 47.5%. 1 H NMR(400MHz,CDCl 3 )δ1.83–1.63(m,1H),1.24(d,J=11.3Hz,12H),1.11–0.95(m,1H),0.85(dd,J=11.2,7.1Hz,1H),0.41–0.26(m,1H).
Third step
55c (133 mg,0.55 mmol), intermediate 2 (100 mg,0.37 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (28 mg,0.035 mmol), cesium carbonate (284 mg,0.88 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 mL) and the reaction stirred under nitrogen at 100deg.C under microwave conditions for 1 hour. Saturated sodium chloride (10 mL) was added to the reaction, extracted with dichloromethane (10 mL. Times.3), the organic phases were combined, dried, and concentrated to give crude product, which was purified by silica gel column chromatography (eluent system B) to give product 6- (2, 4-dimethoxypyrimidin-5-yl) -3-methyl-4- (2- (trifluoromethyl) cyclopropyl) pyridazine 55d (70 mg), yield: 55.6%.
MS m/z(ESI):342.8[M+1] + .
Fourth step
55d (70 mg,0.21 mmol) was dissolved in 2.0mL of 1M aqueous hydrochloric acid, and the reaction solution was stirred at 70℃for 1 hour. The reaction solution was concentrated, and the title product 5- (6-methyl-5- (2- (trifluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 55 (21 mg) was obtained by reverse phase HPLC preparation and isolation (formic acid system), yield: 55.6%.
MS m/z(ESI):313.3[M+1] + .
1 H NMR(400MHz,DMSO-d 6 ):δ8.26(s,1H),7.91(s,1H),2.70(s,3H),2.31(dd,J=14.7,6.3Hz,1H),2.00(dd,J=14.7,7.1Hz,1H),1.55–1.42(m,1H),1.35(dd,J=14.8,6.1Hz,1H).
Resolution of example 55
5- (6-methyl-5- ((1S, 2S) -2- (trifluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione and 5- (6-methyl-5- ((1R, 2R) -2- (trifluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000149
Resolution of example 55 (20 mg,0.064 mmol) via chiral column (MIC column) gave 55-1 (4.48 mg, r.t= 4.640 min), yield: 22.6%;55-2 (4.79 mg, r.t=4.965 min), yield: 23.9%. MS m/z (ESI) 55-1, 313.3[ M+1 ]] + ;55-2,313.3[M+1] + .
Example 56
5- (5- (2- (hydroxymethyl) cyclopropyl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000150
First step
To a mixture of propargyl tert-butyldimethylsilyl ether 56a (5.64 g,33.10 mmol) and triethylamine (352 mg,3.48 mmol) was added pinacol borane (4.45 g,34.80 mmol) and Cp 2 Zr.HCl (897 mg,3.48 mmol). The resulting solution was heated to 60 ℃ and stirred under nitrogen for 16 hours. The mixture was cooled to room temperature, concentrated, and the residue was purified by silica gel column chromatography (eluent system B) to give (E) -tert-butyldimethyl ((3- (4, 5-trimethyl-)) 1,3, 2-dioxaborolan-2-yl) allyl) oxy) silane 56b (9 g), yield 90%.
1 H NMR(CDCl 3 ):δ6.61(dt,J=18.0,3.5Hz,1H),5.68(dd,J=18.0,2.2Hz,1H),4.18(dd,J=3.6,2.4Hz,2H),1.19(s,12H),0.85(s,9H),0.00(s,6H).
Second step
56b (1 g,3.35 mmol) was dissolved in dichloromethane (20 mL) and a solution of diazomethane in diethyl ether (17 mL,33.5 mmol) was added at zero degrees followed by a catalytic amount of Pd (OAc) 2 (65 mg,0.30 mmol) was stirred at room temperature for 1 hour. The reaction solution was directly suspended, and the crude product was purified by silica gel column chromatography (eluent system B) to give tert-butyldimethyl ((2- (4, 5-tetramethyl-1, 3, 2-dioxaborane-2-yl) cyclopropyl) methoxy) silane 56c (0.8 g), yield: 80%.
1 H NMR(CDCl 3 ):δ3.55(dd,J=10.8,5.6Hz,1H),3.42–3.38(m,1H),1.17(d,J=1.2Hz,12H),0.84(s,9H),0.65(ddd,J=9.9,7.9,4.3Hz,1H),0.51(ddd,J=9.0,5.2,3.5Hz,1H),0.08(dt,J=9.6,5.8Hz,2H).
Third step
4-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine intermediate 2 (0.05 g,0.19 mmol) and 56c (0.12 g,0.38 mmol) were added to dioxane (5 mL) and water (0.5 mL), and potassium carbonate (52 mg,0.38 mmol) and Pd (dppf) Cl were added to the reaction mixture under nitrogen 2 (15 mg,0.019 mmol). The resulting mixture was stirred at reflux for 5 hours. Water (20 mL) was added, the organic phase was concentrated, and the resulting residue was purified by silica gel column chromatography with eluent system A to give the title compound 4- (2- ((((tert-butyldimethylsilyl) oxy) methyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine 56d (0.05 g) in 68% yield.
MS m/z(ESI):417.2[M+1] + .
Fourth step
56d (0.05 g,0.12 mmol) was dissolved in hydrochloric acid solution (1M, 10 mL) and reacted overnight at 70 ℃. The reaction solution was concentrated, and the residue was purified by reverse phase preparative HPLC (formic acid) to give 5- (5- (2- (hydroxymethyl) cyclopropyl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 56 (3 mg), yield: 10%.
MS m/z(ESI):275.1[M+1] + .
1 H NMR(400MHz,DMSO-d6):δ11.41(s,2H),8.24(s,1H),7.73(s,1H),3.58(dd,J=11.4,5.3Hz,2H),2.70(s,3H),2.02–1.94(m,1H),1.87(dt,J=8.7,4.9Hz,1H),0.99(ddt,J=31.5,9.3,4.9Hz,2H).
Example 57
5- (5- (2- (1, 3-difluoropropan-2-yl) cyclopropyl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000151
Figure PCTCN2021136249-APPB-000152
First step
Dimethyl sulfoxide (4.28 g,54.73 mmol) was slowly added dropwise to a solution of oxalyl chloride (3.48 g,27.36 mmol) in dichloromethane (30 mL) under nitrogen, stirred for 30 min, followed by a slow addition of a solution of (2, 2-dimethyl-1, 3-dioxan-5-yl) methanol 57a (2 g,13.68 mmol) in dichloromethane (10 mL). The reaction was stirred for two hours, followed by slow addition of triethylamine (8.29 g,82.09 mmol). After stirring was continued for 30 minutes, the temperature was slowly raised to 25℃under stirring, and stirring was continued for about 30 minutes. The reaction solution was quenched with 50mL of water, extracted with dichloromethane (50 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give 2, 2-dimethyl-1, 3-dioxane-5-acetaldehyde 57B (1.2 g), yield: 61.5%.
1 H NMR(400MHz,CDCl 3 ):9.89(s,1H),4.28-4.17(m,4H),2.42-2.32(m,1H),1.49(s,3H),1.39(s,3H).
Second step
To a solution of 2, 2-dimethyl-1, 3-dioxane-5-acetaldehyde 57b (1.2 g,8.32 mmol) in methanol (40 mL) and tert-butyl methyl ether (20 mL) was added dimethyl (1-diazo-2-oxopropyl) phosphonate (3.20 g,16.65 mmol) and potassium carbonate (4.59 g,33.29 mmol) at 25deg.C and the mixture was stirred overnight for 16 hours. The reaction solution was quenched with 30mL of water, extracted with t-butyl methyl ether (30 ml×3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the product 5-ethynyl-2, 2-dimethyl-1, 3-dioxane 57c (750 mg), yield: 64.3%. 1 H NMR(400MHz,CDCl 3 ):δ3.99–3.90(m,2H),3.87–3.81(m,2H),2.82(dddq,1H),2.10(d,1H),1.47(s,3H),1.38(s,3H).
Third step
To a mixture of 5-ethynyl-2, 2-dimethyl-1, 3-dioxane 57c (700 mg,4.99 mmol) and triethylamine (50 mg,0.50 mmol) was added Cp 2 Zr HCl (129 mg,0.50 mmol) and pinacol borane (639 mg,4.99 mmol), and the mixture was stirred overnight at 60 ℃ under nitrogen for 12 hours. After cooling, the reaction solution was concentrated, and the resulting residue was purified by silica gel column chromatography with eluent system B to give product (E) -2- (2, 2-dimethyl-1, 3-dioxan-5-yl) vinyl) -4, 5-tetramethyl-1, 3, 2-dioxapentane 57d (750 mg), yield: 45.8%.
1 H NMR(400MHz,CDCl 3 ):δ6.38(dd,1H),5.54(dd,1H),3.88–3.72(m,4H),2.72–2.56(m,1H),1.43(s,3H),1.39(s,3H),1.30–1.21(12H).
Fourth step
4- (2- (tert-butyl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine intermediate 2 (150 mg,0.56 mmol), (E) -2- (2, 2-dimethyl-1, 3-dioxan-5-yl) vinyl) -4, 5-tetramethyl-1, 3, 2-dioxapentane 57d (150 mg,0.56 mmol), pd (dppf) Cl 2 *CH 2 Cl 2 (46 mg,0.06 mmol) and cesium carbonate (365 mg,1.12 mmol) were added to 1, 4-dioxane (2 mL) and water (0.5 mL), the reaction solution was replaced with nitrogen 3 times, and then subjected to microwavesStirred at 100℃for 1 hour. To the reaction solution was added 5mL of water, extracted with dichloromethane (10 ml×3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give product (E) -6- (2, 4-dimethoxypyrimidin-5-yl) -4- (2, 2-dimethyl-1, 3-dioxan-5-yl) vinyl) -3-methylpyridazine 57E (175 mg), yield: 84.5%.
MS m/z(ESI):373.0[M+1] + .
1 H NMR(400MHz,CDCl 3 ):δ9.02(s,1H),7.90(s,1H),6.64(d,1H),6.43(dd,1H),4.10(s,3H),4.07(s,3H),4.06–4.01(m,2H),3.85(dd,2H),2.77(s,3H),2.74–2.68(m,1H),1.48(s,3H),1.47(s,3H).
Fifth step
To a solution of trimethylsulfoxide iodide (207 mg,0.94 mmol) in dimethyl sulfoxide (5 mL) at 0deg.C under nitrogen was added potassium tert-butoxide (105 mg,0.94 mmol), and after stirring the mixture at 25deg.C for 1 hour, (E) -6- (2, 4-dimethoxypyrimidin-5-yl) -4- (2, 2-dimethyl-1, 3-dioxan-5-yl) vinyl) -3-methylpyridazine 57E (175 mg,0.47 mmol) was added and stirring was continued for 2 hours. To the reaction solution was added 20mL of ice water, extracted with dichloromethane (15 ml×3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- (2, 2-dimethyl-1, 3-dioxan-5-yl) cyclopropyl) -3-methylpyridazine 57f (80 mg), yield: 44.1%.
MS m/z(ESI):387.0[M+1] + .
1 H NMR(400MHz,CDCl 3 ):δ9.00(s,1H),7.43(s,1H),4.08(s,3H),4.07(s,3H),4.04(dd,2H),3.83–3.77(m,2H),2.83(d,4H),1.79(s,1H),1.44(s,6H),1.08(t,2H),0.87(t,1H).
Sixth step
To a solution of 6- (2, 4-dimethoxypyrimidin-5-yl) -4- (2, 2-dimethyl-1, 3-dioxan-5-yl) cyclopropyl) -3-methylpyridazine 57f (55 mg,0.14 mmol) in methanol (5 mL) was slowly added dropwise hydrochloric acid (2M, 0.5 mL) and the mixture stirred at 25℃for 30 min. The reaction solution was adjusted to pH 7 to 8 with saturated sodium bicarbonate, extracted with dichloromethane (20 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give 57g (45 mg) of 2- (2- (6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazin-4-yl) cyclopropyl) propane-1, 3-diol as a product, yield: 91%.
MS m/z(ESI):347.0[M+1] + .
1 H NMR(400MHz,CDCl 3 ):δ8.96(s,1H),7.48(s,1H),4.07(s,3H),4.06(s,3H),3.97(d,2H),3.92–3.83(m,2H),2.89(s,3H),2.01(d,1H),1.90(s,1H),1.62(d,1H),0.87(d,2H).
Seventh step
To a solution of 57g (45 mg,0.13 mmol) of 2- (2- (6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazin-4-yl) cyclopropyl) propane-1, 3-diol in dichloromethane (6 mL) at-78deg.C under nitrogen was slowly added dropwise diethylaminosulfur trifluoride (63 mg,0.39 mmol), and the mixture was stirred overnight at 30deg.C for 12 hours. To the reaction solution was added 10mL of saturated sodium bicarbonate solution, extracted with dichloromethane (20 ml×3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the crude product 4- (2- (1, 3-difluoropropan-2-yl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine 57h (20 mg, yellow solid), which was directly subjected to the next reaction without purification.
MS m/z(ESI):351.0[M+1] + .
Eighth step
To a round bottom flask was added 4- (2- (1, 3-difluoropropan-2-yl) cyclopropyl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3-methylpyridazine 57h (20 mg,0.06 mmol) and hydrochloric acid (1M, 2 mL) and the mixture was stirred at 70℃for 2 h. The reaction solution was cooled and concentrated, and the residue was purified by preparative HPLC to give the title product 5- (5- (2- (1, 3-difluoropropan-2-yl) cyclopropyl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 57 (3 mg), yield: 15.2%.
MS m/z(ESI):323.0[M+1] + .
1 H NMR(400MHz,CD 3 OD):δ8.28(s,1H),7.83(s,1H),4.58–4.54(m,4H),2.78(s,3H),2.11–2.03(m,1H),1.60–1.56(m,1H),1.18–1.13(m,1H),0.93–0.85(m,2H).
Example 58
5- (5- (2- (1, 3-dihydroxypropyl-2-yl) cyclopropyl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000153
Figure PCTCN2021136249-APPB-000154
First step
To a round bottom flask was added 57g (15 mg,0.04 mmol) of the compound of example 57 and hydrochloric acid (1M, 2 mL) and the mixture was stirred at 70℃for 2 hours. The reaction solution was cooled and concentrated, and the residue was purified by preparative HPLC to give the title product 5- (5- (2- (1, 3-dihydroxypropyl-2-yl) cyclopropyl) -6-methylpyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 58 (3 mg), yield: 21.8%.
MS m/z(ESI):319.0[M+1] + .
1 H NMR(400MHz,CD 3 OD):δ8.13(s,1H),7.58(s,1H),3.68–3.58(m,4H),2.70(s,3H),1.92–1.88(m,1H),1.55–1.46(m,1H),1.15–1.11(m,1H),1.06–1.01(m,1H),0.81–0.76(m,1H).
Example 59
5- (6-ethyl-5- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000155
First step
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine intermediate 1 (60 mg, 0.178 mmol) was dissolved in a mixed solvent of 2mL of 1, 4-dioxane and 0.4mL of water, and ethylboronic acid (39 mg,0.537 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (15 mg,0.0179 mmol) and cesium carbonate (117 mg, 0.178 mmol) were added. The reaction mixture was replaced with nitrogen gas for 3 times and subjected to microwave reaction at 100℃for 1 hour. The reaction mixture was cooled to room temperature, 30mL of water was added, and extraction was performed with ethyl acetate (30 mL. Times.2). The organic phases were combined, washed successively with water (30 mL), saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the crude product 6- (2, 4-dimethoxypyrimidin-5-yl) -3-ethyl-4- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazine 59a (60 mg) which was directly used in the next reaction.
MS m/z(ESI):329.0[M+H] + .
Second step
6- (2, 4-Dimethoxypyrimidin-5-yl) -3-ethyl-4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine 59a (60 mg,0.182 mmol) was dissolved in 3mL of methanol, and hydrochloric acid (2M, 1 mL) was added to react at 70℃for 3 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by reverse phase HPLC to give the title product 5- (6-ethyl-5- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 59 (24 mg), yield: 43.8%.
MS m/z(ESI):301.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ11.42(s,2H),8.24(s,1H),7.64(s,1H),3.09(q,J=7.6Hz,2H),1.86-1.82(m,1H),1.32(t,J=7.6Hz,3H),1.27-1.24(m,1H),1.03-0.98(m,7H),0.97-0.90(m,2H).
Example 60
5- (6-cyclopropyl-5- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000156
First step
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine intermediate 1 (60 mg, 0.178 mmol) was dissolved in 2mL of 1, 4-dioxane and 0.4mL of water, and cyclopropylboronic acid (23 mg, 0.399 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (15 mg,0.0179 mmol) and cesium carbonate (86 mg, 0.399 mmol) were added. The reaction mixture was replaced with nitrogen gas for 3 times and subjected to microwave reaction at 100℃for 1 hour. The reaction mixture was cooled to room temperature, 30mL of water was added, and extraction was performed with ethyl acetate (30 mL. Times.2). The organic phases were combined, washed successively with water (30 mL), saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered, the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give the product 3-cyclopropyl-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazin 60a (60 mg), yield: 98.4%.
MS m/z(ESI):341.0[M+H] + .
Second step
3-cyclopropyl-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine 60a (60 mg,0.178 mmol) was dissolved in 3mL of methanol, and hydrochloric acid (2M, 1 mL) was added to react at 70℃for 3 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by reverse phase HPLC to give the title product 5- (6-cyclopropyl-5- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 60 (44 mg), yield: 80.0%.
MS m/z(ESI):313.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ11.41(s,2H),8.21(s,1H),7.65(s,1H),2.09-2.05(m,1H),1.30-1.24(m,1H),1.18-1.15(m,1H),1.10-1.06(m,3H),1.03-0.98(m,7H),0.97-0.91(m,3H).
Example 61
5- (6- (hydroxymethyl) -5- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000157
First step
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine intermediate 1 (80 mg,0.239 mmol) was dissolved in 3mL of 1, 4-dioxane, and (tributyltin) methanol (153 mg,0.178 mmol) and tetrakis (triphenylphosphine) palladium (28 mg,0.0239 mmol) were added, the reaction solution was replaced with nitrogen gas for 3 times and the reaction was subjected to microwave reaction at 110℃for 1 hour. The reaction solution was cooled to room temperature, 50mL of water and a small amount of potassium fluoride were added to precipitate a tin reagent, and the solution was filtered, and the cake was washed with ethyl acetate (40 mL). The mixture was separated and the aqueous phase was extracted with ethyl acetate (40 mL). The organic phases were combined, washed successively with water (40 mL), saturated sodium chloride solution (40 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography with eluent system B to give crude (6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazin-3-yl) methanol 61a (50 mg) which was directly used for the next reaction.
MS m/z(ESI):331.0[M+H] + .
Second step
(6- (2, 4-Dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3-yl) methanol 61a (30 mg,0.0908 mmol) was dissolved in 3mL of methanol, hydrochloric acid (2M, 1 mL) was added, and the mixture was reacted at 70℃for 3 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was subjected to reverse phase HPLC to give the title product 5- (6- (hydroxymethyl) -5- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 61 (9 mg), yield: 32.8%.
MS m/z(ESI):303.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ8.39(s,1H),7.76(s,1H),5.43-5.39(m,1H),4.86(d,J=4.8Hz,2H),2.06-2.03(m,1H),1.25-1.18(m,1H),1.02-0.98(m,6H),0.96-0.90(m,3H).
Example 62
5- (6- (fluoromethyl) -5- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3-yl) pyrimidin-2, 4 (1H,
3H) -diketones
Figure PCTCN2021136249-APPB-000158
First step
(6- (2, 4-Dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3-yl) methanol 61a (30 mg,0.091 mmol) was dissolved in 4mL of tetrahydrofuran, and triethylamine (44 mg,0.272 mmol), triethylamine trihydrofluoride (46 mg,0.454 mmol) and perfluorobutylsulfonyl fluoride (82 mg,0.272 mmol) were added to react at room temperature for 2 hours. Poured into 30mL of saturated sodium bicarbonate solution and extracted with ethyl acetate (30 mL. Times.2). The organic phases were combined, washed successively with water (30 mL), saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure to give the crude product 6- (2, 4-dimethoxypyrimidin-5-yl) -3- (fluoromethyl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazine 62a (20 mg) which was used directly in the next reaction.
MS m/z(ESI):333.0[M+H] + .
Second step
6- (2, 4-Dimethoxypyrimidin-5-yl) -3- (fluoromethyl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine 62a (20 mg,0.0601 mmol) was dissolved in 3mL of 1, 4-dioxane, 1mL of 2M hydrochloric acid was added, and the mixture was reacted at 70℃for 3 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was subjected to reverse phase HPLC to give the title product 5- (6- (fluoromethyl) -5- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 62 (2.0 mg), yield: 10.9%.
MS m/z(ESI):305.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ8.52(s,1H),7.90(s,1H),5.88(dd,J=37.2Hz,10.4Hz,1H),5.77(dd,J=37.2,10.4Hz,1H),1.96-1.96(m,1H),1.09-1.04(m,1H),1.03-0.96(m,9H).
Example 63
5- (6-amino-5- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000159
Figure PCTCN2021136249-APPB-000160
First step
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine intermediate 1 (60 mg, 0.178 mmol) was dissolved in 2mL of 1, 4-dioxane, tert-butyl carbamate (63 mg,0.537 mmol), tris (dibenzylideneacetone) dipalladium (0) (16 mg,0.0179 mmol), 2-dicyclohexylphosphorus-2, 4, 6-triisopropylbiphenyl (8.5 mg,0.0179 mmol) and cesium carbonate (88 mg,0.269 mmol) were added thereto, and the reaction mixture was subjected to microwave reaction at 110℃for 1 hour by replacing 3 times with nitrogen. The reaction mixture was cooled to room temperature, and 50mL of water was added thereto, followed by extraction with ethyl acetate (50 mL. Times.2). The organic phases were combined, washed successively with water (30 mL), saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure to give the crude tert-butyl (6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3-yl) carbamate 63a (40 mg) which was used directly in the next reaction.
MS m/z(ESI):416.0[M+H] + .
Second step
Tert-butyl (6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazin-3-yl) carbamate 63a (40 mg,0.096 mmol) was dissolved in 3mL of methanol, 1mL of 2M hydrochloric acid was added, and the mixture was reacted at 70℃for 3 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was subjected to reverse phase HPLC to give the title product 5- (6-amino-5- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 63 (9.1 mg), yield: 32.8%.
MS m/z(ESI):288.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ11.24(s,2H),7.94(s,1H),7.37(s,1H),6.25(s,2H),1.71-1.66(m,1H),1.21-1.13(m,1H),1.00(dd,J=8.4,6.8Hz,6H),0.89-0.83(m,2H),0.78-0.74(m,1H).
Example 64
5- (5- ((1S, 2R) -2-isopropylcyclopropyl) -6-methoxypyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000161
First step
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine intermediate 1 (60 mg, 0.178 mmol) was dissolved in 3mL of methanol, sodium methoxide (29 mg,0.537 mmol) was added thereto, and the mixture was heated to 70℃under nitrogen atmosphere to react for 2 hours. The reaction mixture was cooled to room temperature, 30mL of water was added, and extraction was performed with ethyl acetate (30 mL. Times.2). The organic phases were combined, washed sequentially with water (30 mL), saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure to give the crude product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) -3-methoxypyridazine 64a (55 mg) which was used directly in the next reaction.
MS m/z(ESI):331.0[M+H] + .
Second step
6- (2, 4-Dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) -3-methoxypyridazine 64a (55 mg,0.0949 mmol) was dissolved in 3mL of methanol, 1mL of 2M hydrochloric acid was added, and the mixture was reacted at 70℃for 3 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was subjected to reverse phase HPLC to give the title product 5- (5- ((1 s,2 r) -2-isopropylcyclopropyl) -6-methoxypyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 64 (37 mg), yield: 73.6%.
MS m/z(ESI):303.0[M+H] + .
1 H NMR(400MHz,DMSO-d 6 ):δ11.37(br s,2H),8.08(s,1H),7.61(s,1H),4.07(s,3H),1.85-1.81(m,1H),1.20-1.13(m,1H),1.02-0.89(m,9H).
Example 65
5- (5- ((1S, 2R) -2-isopropylcyclopropyl) -6-ethoxypyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000162
First step
3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine intermediate 1 (40 mg,0.12 mmol) was dissolved in 5mL of ethanol, sodium ethoxide (68 mg,1 mmol) was added, and the mixture was heated to 60℃under nitrogen protection and reacted for 4 hours. The reaction mixture was cooled to room temperature, and 20mL of water was added thereto, followed by extraction with methylene chloride (20 mL. Times.3). The organic phases were combined, washed sequentially with water (30 mL), saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated under reduced pressure to give the crude product 6- (2, 4-diethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) -3-ethoxypyridazine 65a (30 mg) as a product, which was used directly in the next reaction.
MS m/z(ESI):373.0[M+1] + .
Second step
6- (2, 4-diethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) -3-ethoxypyridazine 65a (30 mg,0.08 mmol) was dissolved in 1mL of methanol, 1mL of 2M hydrochloric acid was added, and the mixture was reacted at 70℃for 2 hours. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was subjected to reverse phase HPLC to give the title product 5- (5- ((1 s,2 r) -2-isopropylcyclopropyl) -6-ethoxypyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 65 (15 mg), yield: 55.3%.
MS m/z(ESI):317.0[M+1] + .
1 H NMR(400MHz,DMSO-d 6 ):δ11.34(s,2H),8.08(s,1H),7.61(s,1H),4.51(p,2H),1.81(dt,1H),1.40(t,3H),1.19–1.13(m,1H),1.05–1.01(m,1H),0.98(dd,6H),0.94–0.91(m,1H),0.88–0.84(m,1H).
Example 66
5- (8- ((1R, 2S) - (1, 1' -bis (cyclopropane) ] -2-yl) -3-methylimidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000163
The title product 5- (8- ((1 r,2 s) - (1, 1' -bis (cyclopropane) ] -2-yl) -3-methylimidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1 h,3 h) -dione 66 was obtained by reference to the synthetic method of example 13.
MS m/z(ESI):324.0[M+1] +
1 H NMR(400MHz,DMSO-d 6 )δ11.36(s,2H),8.03(s,1H),7.48(s,1H),7.24(s,1H),2.47(s,3H),2.25–2.20(m,1H),1.69–1.66(m,1H),1.40–1.35(m,1H),1.02
0.97 (m, 2H), 0.45-0.40 (m, 2H), 0.20 (s, 2H) example 675- (5- ((1S, 2R) -2-isopropylcyclopropyl) -6- (methoxy-d 3) pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000164
Figure PCTCN2021136249-APPB-000165
First step
3, 6-dichloropyridazine (1.0 g,6.71 mmol) was suspended in 30mL of deionized water under nitrogen, and (1R, 2R) -2-isopropylcyclopropane-1-carboxylic acid (866 mg,6.71mmol, preparation method for which was referred to WO2019168744A 1) and concentrated sulfuric acid (1 mL) was added. After the reaction was heated to 70 ℃, a solution of silver nitrate (228 mg,1.34 mmol) in water (1 mL) was added dropwise, followed by ammonium persulfate (4.5 g,20.1 mmol) in water (15 mL) for about 15 minutes. The reaction solution was stirred at 70℃for 1 hour. The reaction solution was cooled to room temperature and neutralized with ammonia to pH 8-9. The aqueous phase was extracted with ethyl acetate (60 mL. Times.3). The organic phases were combined, washed sequentially with water (60 mL), saturated sodium chloride solution (60 mL), dried, concentrated, and the resulting crude compound was separated by silica gel column chromatography (eluent system B) to give the product 3, 6-dichloro-4- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazine 67B (1.0 g), yield: 64.5%.
MS m/z(ESI):231.0[M+H] + .
Second step
3, 6-dichloro-4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine 67b (500 mg,2.17 mmol), 2, 4-dimethoxypyrimidine-5-boronic acid (400 mg,2.17 mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (178 mg,0.22 mmol) and cesium carbonate (1.41 g,4.34 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 mL) under nitrogen. The reaction solution was stirred for 1 hour at 70℃under microwave conditions. The reaction solution was cooled to room temperature, water (25 mL) was added, and the aqueous phase was extracted with ethyl acetate (25 mL. Times.3). The organic phases were combined, washed sequentially with water (25 mL), saturated sodium chloride solution (25 mL), dried, concentrated, and the resulting crude compound was separated by silica gel column chromatography (eluent system B) to give the product 3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) pyridazine 67c (306 mg), yield: 42.2%.
MS m/z(ESI):335.1[M+H] + .
Third step
Sodium hydride (288 mg,12 mmol) was dissolved in deuterated methanol (3 mL) at room temperature and stirred for 10 min. 3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) pyridazine 67c (200 mg,0.60 mmol) was added to the above mixed solution. The reaction solution was stirred at 65℃for 4 hours. The reaction solution was cooled to room temperature, water (5 mL) was added, and the aqueous phase was extracted with ethyl acetate (5 mL. Times.3). The organic phases were combined, washed successively with water (5 mL), saturated sodium chloride solution (5 mL), dried, concentrated, and the resulting crude compound was separated by silica gel column chromatography (eluent system B) to give the product 6- (2, 4-dimethoxypyrimidin-5-yl) -4- ((1 s,2 r) -2-isopropylcyclopropyl) -3- (methoxy-d 3) pyridazine 67d (156 mg), yield: 78%.
MS m/z(ESI):334.1[M+H] + .
Fourth step
6- (2, 4-Dimethoxypyrimidin-5-yl) -4- ((1S, 2R) -2-isopropylcyclopropyl) -3- (methoxy-d 3) pyridazine 67d (100 mg,0.30 mmol) was dissolved in a mixture of hydrochloric acid (1M, 2 mL) and methanol (2 mL). The reaction was heated to 70℃and stirred for 3 hours. The reaction solution was concentrated to give a crude product, which was isolated by reverse phase HPLC preparation (formic acid system) to give the title product 5- (5- ((1 s,2 r) -2-isopropylcyclopropyl) -6- (methoxy-d 3) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 67 (59 mg), yield: 65%.
MS m/z(ESI):306.1[M+H] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.37(br s,2H),8.08(s,1H),7.61(s,1H),1.85-1.81(m,1H),1.20-1.13(m,1H),1.02-0.89(m,9H).
Example 68
5- (5- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6-methoxypyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000166
First step
To a mixture of 20% aqueous potassium hydroxide (2 mL) and diethyl ether (2 mL) was added N-methyl-N-nitrosourea (500 mg,4.8 mmol) under ice-bath conditions, and the mixture was stirred at this temperature for 1 hour. The organic phase of the above mixed solution was added to a solution of 68a (200 mg,1.0 mmol) in diethyl ether (2 mL) under ice-bath conditions, followed by adding palladium acetate (22 mg,0.01 mmol) to the reaction solution, and the reaction was stirred under ice-bath conditions for 30 minutes. The reaction solution was filtered, and the cake was washed with methylene chloride (5 mL. Times.3). The filtrate was concentrated and the resulting crude compound was separated by silica gel column chromatography (eluent system B) to give the product 2- ([ [1,1' -bis (cyclopropane) ] -2-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborane 68B (182 mg), yield: 84.8%.
1 H NMR(400MHz,CDCl 3 ):δ1.21(s,12H),1.02(ddd,1H),0.85–0.72(m,1H),0.57(ddd,1H),0.45–0.24(m,3H),0.16–0.01(m,2H),-0.36(dt,1H).
Second step
68b (92 mg,0.44 mmol), 4-bromo-3, 6-dichloropyridazine (100 mg,0.44 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (36 mg,0.044 mmol) and potassium carbonate (121 mg, 0.88 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 ml). At N 2 The reaction solution was stirred under microwave conditions at 80℃for 1 hour under protection. Adding saturated sodium chloride into the reaction solution(10 mL) extraction of the aqueous phase with ethyl acetate (10 mL. Times.3), combining the organic phases, drying, concentrating, and separation of the crude compound by silica gel column chromatography (eluent system B) to give the product 4- ([ 1,1' -bis (cyclopropane)]-2-yl) -3, 6-dichloropyridazine 68c (79 mg), yield: 78%.
MS m/z(ESI):228.8[M+1] + .
Third step
68c (50 mg,0.22 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (44 mg,0.24 mmol), palladium tetraphenylphosphine (26 mg,0.022 mmol) and potassium carbonate (61 mg,0.44 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 ml). The reaction solution was stirred under nitrogen protection at 80℃for 1 hour under microwave conditions. To the reaction solution was added saturated sodium chloride (10 mL), the aqueous phase was extracted with ethyl acetate (10 ml×3), the organic phases were combined, dried, and concentrated, and the crude compound was separated by silica gel column chromatography (eluent system B) to give the product 4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine 68d (35 mg), yield: 48%.
MS m/z(ESI):333.1[M+1] + .
Fourth step
Metallic sodium (43 mg,1.9 mmol) was added portionwise to methanol (20 mL) under ice bath and stirred until the sodium cake was completely dissolved. 68d (120 mg,0.36 mmol) was added to the reaction solution. The reaction was heated to 65℃and stirred for 3 hours. To the reaction solution was added ethyl acetate (50 mL), and the organic phase was washed with saturated ammonium chloride and saturated sodium chloride, dried and concentrated to give a crude product of 4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3-methoxypyridazine 68e (118 mg), which was used in the next reaction without purification.
MS m/z(ESI):329.0[M+1] + .
Fifth step
68e (118 mg, 0.399 mmol) was dissolved in a mixture of hydrochloric acid (2M, 5 mL) and methanol (5 mL), heated to 65℃and stirred for 3 hours. The reaction solution was concentrated to give a crude product, which was isolated by reverse phase HPLC (formic acid system) to give the title product 5- (5- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6-methoxypyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 68 (45 mg), yield: 41.7%.
MS m/z(ESI):300.9[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.38(s,2H),8.08(s,1H),7.59(s,1H),4.06(s,3H),1.80(dd,1H),1.23–1.16(m,1H),1.02–0.95(m,1H),0.95–0.88(m,2H),0.49–0.34(m,2H),0.22–0.12(m,2H).
Example 68-1 and example 68-2
5- (5- ((1 r,2 s) - [1,1 '-bis (cyclopropane) ] -2-yl) -6-methoxypyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione and 5- (5- ((1 s,2 r) - [1,1' -bis (cyclopropane) ] -2-yl) -6-methoxypyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000167
Chiral resolution of example 68 (47 mg,0.16 mmol) by SFC gave example 68-1 (4.53 mg, R.T= 1.899min, yield: 9.6%) and example 68-2 (5.21 mg, R.T=1.676 min, yield: 11.1%),
Example 68-1: MS m/z (ESI) 300.9[ M+1 ]] +
Example 68-2: MS m/z (ESI) 300.9[ M+1 ]] + .
Chiral resolution conditions:
Figure PCTCN2021136249-APPB-000168
chiral analysis method:
Figure PCTCN2021136249-APPB-000169
example 69
5- (6- (methoxy-d) 3 ) -5- (2- (trifluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000170
Figure PCTCN2021136249-APPB-000171
First step
69a (1.5 g,11.11 mmol) was dissolved in a mixed solution of methyl tert-butyl ether (5 mL) and water (2 mL) and stirred. The reaction solution was cooled using an ice bath, and an aqueous solution (2 mL) of sodium nitrite (0.843 g,12.22 mmol) was added to the reaction solution. The reaction solution was warmed to room temperature and stirred for 3 hours. The aqueous phase of the reaction solution was separated, and the remaining organic phase (5 mL, according to WO2015/52226, expected to contain 594mg of the product 2-diazonium-1, 1-trifluoroethane 69b, yield: 48.6%) was directly used for the next reaction.
Second step
Palladium acetate (121 mg,0.54 mmol) was added to a solution of 4, 5-tetramethyl-2-vinyl-1, 3, 2-dioxaborane (748 mg,4.86 mmol) in methyl tert-butyl ether (5 mL) at room temperature, and then a solution of 69b in methyl tert-butyl ether (5 mL,594mg,5.4 mmol) was slowly added to the reaction solution. The reaction solution was stirred at room temperature for 2 hours. The reaction solution was filtered, the filtrate was concentrated, and the crude compound was purified by silica gel column chromatography (eluent system B) to give product 4, 5-tetramethyl-2- (2- (trifluoromethyl) cyclopropyl) -1,3, 2-dioxaborane intermediate 69c (0.7 g), yield: 60.9%.
1 H NMR(400MHz,CDCl 3 )δ1.83–1.63(m,1H),1.24(d,12H),1.11–0.95(m,1H),0.85(dd,1H),0.41–0.26(m,1H).
Third step
4-bromo-6-chloropyridazin-3-amine (1.0 g,4.80 mmol), 69c (1.25 g,5.28 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (390 mg,0.48 mmol) and cesium carbonate (4.69 g,14.39 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 5 ml) and stirred. The reaction was heated to 110℃under nitrogen and stirred for 16 hours, 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (399mg, 0.48 mmol) was added and stirring was continued at 110℃for 16 hours. To the reaction solution was added saturated sodium chloride (10 mL), the aqueous phase was extracted with ethyl acetate (10 mL. Times.3), and the organic phases were combined, dried, and concentrated. The crude compound was separated by silica gel column chromatography (eluent system B) to give product 6-chloro-4- (2- (trifluoromethyl) cyclopropyl) pyridazin-3-amine 69d (450 mg), yield: 39.5%.
MS m/z(ESI):238.1[M+1] + .
Fourth step
Sodium nitrite (157 mg,2.27 mmol) was dissolved in concentrated sulfuric acid (2.0 mL) and stirred. Compound 69d (450 mg,1.89 mmol) dissolved in acetic acid (5 mL) was slowly added to the reaction solution under ice bath, and the reaction solution was then warmed to room temperature and stirred for 1 hour. Water (5 mL) was added to the reaction solution, and the reaction solution was stirred at room temperature for 0.5 hours. To the reaction solution was added saturated sodium chloride (10 mL), the aqueous phase was extracted with ethyl acetate (10 mL. Times.3), the organic phases were combined, dried, and concentrated to give the crude product 6-chloro-4- (2- (trifluoromethyl) cyclopropyl) pyridazin-3 (2H) -one 69e (440 mg), which was used in the next reaction without purification.
MS m/z(ESI):239.1[M+1] + .
Fifth step
69e (100 mg,0.419 mmol) was dissolved in phosphorus oxychloride (64.26 mg,0.419 mmol), and the reaction solution was heated to 100℃and stirred for 1 hour. The reaction solution was concentrated, saturated sodium hydrogencarbonate (10 ml) was added to the residue, the aqueous phase was extracted with methylene chloride (10 ml. Times.3), the organic phases were combined, dried, and concentrated to give a crude product, which was separated by silica gel column chromatography (eluent system B) to give 3, 6-dichloro-4- (2- (trifluoromethyl) cyclopropyl) pyridazine 69f (100 mg), yield: 92.82%.
MS:m/z(ESI):258.3[M+1] +
Sixth step
69f (250 mg,0.98 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (180 mg,0.98 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (78 mg,0.098 mmol), cesium carbonate (480 mg,2.94 mmol) was dissolved in a 1, 4-dioxane/water (v/v=4:1, 2 ml) mixture and the reaction was heated to 100℃with microwaves and stirred for 1 hour. To the reaction solution was added saturated sodium chloride (10 ml), the aqueous phase was extracted with methylene chloride (10 ml. Times.3), the organic phases were combined, dried, and concentrated to give a crude product, which was purified by silica gel column chromatography (eluent system B) to give 69g (240 mg) of 3-chloro-6- (2, 4-dimethoxy-4, 5-dihydropyrimidin-5-yl) -4- (2- (trifluoromethyl) cyclopropyl) pyridazine as a product: 68.6%.
MS:m/z(ESI):361.3[M+1] +
Seventh step
Sodium hydride (166 mg,6.93 mmol) was dissolved in deuterated methanol (1 mL) at room temperature, and after the reaction solution was stirred at room temperature for 1 hour, 69g (250 mg,0.69 mmol) was added to the reaction solution. The reaction mixture was stirred at room temperature for 1 hour. Saturated sodium chloride (10 mL) was added to the reaction, the aqueous phase was extracted with dichloromethane (10 mL. Times.3), the organic phases were combined, dried, and concentrated to give the crude product 6- (2, 4-dimethoxypyrimidin-5-yl) -3- (methoxy-d) 3 ) 4- (2- (trifluoromethyl) cyclopropyl) pyridazine 69h (220 mg), the product was used in the next reaction without purification.
MS:m/z(ESI):360.3[M+1] +
Eighth step
69h (200 mg,0.55 mmol) were dissolved in hydrochloric acid solution (1M, 2 mL) and the reaction was heated to 70℃and stirred for 1 h. The reaction solution was concentrated, and the residue was separated by reverse phase HPLC to give the title product 5- (6- (methoxy-d) 3 ) -5- (2- (trifluoromethyl) cyclopropyl) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 69 (44 mg), yield: 23.6%.
MS:m/z(ESI):332.3[M+1] +
1 H NMR(400MHz,DMSO-d 6 )δ11.43(s,2H),8.08(s,1H),7.84(s,1H),2.41(dt,2H),1.62–1.21(m,2H).
Example 70
5- (5- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6- (methyl-d 3) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000172
First step
To a mixture of 20% aqueous potassium hydroxide (2 mL) and diethyl ether (2 mL) was added N-methyl-N-nitrosourea (500 mg,4.8 mmol) under ice-bath conditions, and the mixture was stirred at this temperature for 1 hour. The organic phase of the above mixed solution was added to a solution of 70a (200 mg,1.0 mmol) in diethyl ether (2 mL) under ice-bath conditions, followed by adding palladium acetate (22 mg,0.01 mmol) to the reaction solution, and the reaction was stirred under ice-bath conditions for 30 minutes. The reaction solution was filtered, and the cake was washed with methylene chloride (5 mL. Times.3). The filtrate was concentrated and the resulting crude compound was separated by silica gel column chromatography (eluent system B) to give the product 2- ([ [1,1' -bis (cyclopropane) ] -2-yl) -4, 5-tetramethyl-1, 3, 2-dioxaborane 70B (182 mg), yield: 84.8%.
1 H NMR(400MHz,CDCl 3 ):δ1.21(s,12H),1.02(ddd,1H),0.85–0.72(m,1H),0.57(ddd,1H),0.45–0.24(m,3H),0.16–0.01(m,2H),-0.36(dt,1H).
Second step
70b (92 mg,0.44 mmol), 4-bromo-3, 6-dichloropyridazine (100 mg,0.44 mmol), 1-bis (diphenylphosphine) ferrocene palladium dichloride dichloromethane complex (36 mg,0.044 mmol) and potassium carbonate (121 mg,0.88 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 ml). At N 2 The reaction solution was stirred under microwave conditions at 80℃for 1 hour under protection. Saturated sodium chloride (10 mL) was added to the reaction mixture, the aqueous phase was extracted with ethyl acetate (10 mL. Times.3), the organic phases were combined, dried, concentrated, and the crude compound was separated by silica gel column chromatography (eluent system B) to give the product 4- ([ 1,1' -bis (cyclopropane)]-2-yl) -3, 6-dichloropyridazine 70c (79 mg), yield: 78%.
MS m/z(ESI):228.8[M+1] + .
Third step
70c (50 mg,0.22 mmol), (2, 4-dimethoxypyrimidin-5-yl) boric acid (44 mg,0.24 mmol), palladium tetraphenylphosphine (26 mg,0.022 mmol) and potassium carbonate (61 mg,0.44 mmol) were dissolved in 1, 4-dioxane/water (v/v=4:1, 2 ml). The reaction solution was stirred under nitrogen protection at 80℃for 1 hour under microwave conditions. To the reaction solution was added saturated sodium chloride (10 mL), the aqueous phase was extracted with ethyl acetate (10 ml×3), the organic phases were combined, dried, and concentrated, and the crude compound was separated by silica gel column chromatography (eluent system B) to give the product 4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine 70d (35 mg), yield: 48%.
MS m/z(ESI):333.1[M+1] + .
Fourth step
4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -3-chloro-6- (2, 4-dimethoxypyrimidin-5-yl) pyridazine 70d (100 mg,0.30 mmol), iron triacetylacetonate (11 mg,0.03 mmol), N-methylpyrrolidone (one drop) were dissolved in tetrahydrofuran (5 mL) under nitrogen atmosphere and stirred. To the reaction solution was added dropwise methyl-d 3-magnesium iodide (1M, 0.6 mL). After completion of the dropwise addition, the reaction solution was stirred at room temperature for 4 hours. To the reaction solution was added saturated sodium chloride (10 mL), the aqueous phase was extracted with ethyl acetate (10 ml×3), the organic phases were combined, dried, and concentrated, and the crude compound was separated by silica gel column chromatography (eluent system B) to give the product 4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3- (methyl-d 3) pyridazine 70e (77 mg), yield: 82%.
MS m/z(ESI):316.1[M+1] + .
Fifth step
4- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6- (2, 4-dimethoxypyrimidin-5-yl) -3- (methyl-d 3) pyridazine 70e (77 mg,0.24 mmol) was dissolved in a mixture of hydrochloric acid (1M, 2 mL) and methanol (2 mL), heated to 70℃and stirred for 4 hours. The reaction solution was concentrated to give a crude product, which was isolated by reverse phase HPLC (formic acid system) to give the title product 5- (5- ([ 1,1' -bis (cyclopropane) ] -2-yl) -6- (methyl-d 3) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione 70 (33 mg), yield: 47%.
MS m/z(ESI):288.1[M+1] + .
1 H NMR(400MHz,DMSO-d 6 )δ11.42(s,2H),8.21(s,1H),7.68(s,1H),1.81–1.72(m,1H),1.12–1.04(m,1H),1.03–0.96(m,1H),0.95–0.86(m,2H),0.52–0.37(m,2H),0.24–0.19(m,2H).
Examples 70-1 and 70-2
5- (5- ((1 r,2 s) - [1,1 '-bis (cyclopropane) ] -2-yl) -6- (methyl-d 3) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione and 5- (5- ((1 s,2 r) - [1,1' -bis (cyclopropane) ] -2-yl) -6- (methyl-d 3) pyridazin-3-yl) pyrimidine-2, 4 (1 h,3 h) -dione
Figure PCTCN2021136249-APPB-000173
Chiral resolution of example 70 (33 mg,0.11 mmol) by SFC gave example 70-1 (6.32 mg, r.t= 1.653min, yield: 19.2%) and example 70-2 (8.80 mg, r.t=2.376 min, yield: 26.7%),
example 70-1: MS m/z (ESI) 288.1[ M+1 ]] +
1 H NMR(400MHz,DMSO-d6)δ12.00(s,1H),11.75(s,1H),8.41(s,1H),8.03(s,1H),1.95(s,1H),1.35(s,1H),1.22(s,1H),1.13(s,1H),1.03(td,1H),0.60–0.37(m,2H),0.28–0.12(m,2H)
Example 70-2: MS m/z (ESI) 288.1[ M+1 ]] + .
Chiral resolution conditions:
Figure PCTCN2021136249-APPB-000174
chiral analysis method:
Figure PCTCN2021136249-APPB-000175
example 71
5- (8- (5, 5-dimethylcyclohex-1-en-1-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000176
The title product 5- (8- (5, 5-dimethylcyclohex-1-en-1-yl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione 71 was obtained by the method of synthesis according to example 12.
MS m/z(ESI):338.0[M+1] +
1 H NMR(400MHz,DMSO-d 6 )δ11.46(s,2H),8.20(d,1H),8.013(s,1H),7.79(d,1H),7.71(d,1H),7.61(s,1H),2.36(t,2H),2.30(d,2H),1.43(t,2H),1.00(s,6H).
Example 72
5- (7-cyclopropyl-5-ethyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000177
The title product 5- (7-cyclopropyl-5-ethyl-5H-pyrrolo [3,2-c ] pyridazin-3-yl) pyrimidine-2, 4 (1H, 3H) -dione 72 was obtained by the method of example 25.
MS m/z(ESI):298.1[M+1] +
1 H NMR(400MHz,DMSO-d 6 )δ11.41(s,2H),8.36(s,1H),8.28(s,1H),7.58(s,1H),4.13(q,2H),2.22-2.11(m,1H),1.34(t,3H),1.01-0.91(m,4H).
Example 73
5- (8- (3-methylcyclobutyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000178
The title product 5- (8- (3-methylcyclobutyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione 73 is obtained according to the method of example 12.
MS m/z(ESI):298.1[M+1] +
1 H NMR(400MHz,DMSO-d 6 )δ11.52(s,2H),8.27(d,1H),8.02(dd,1H),7.79(s,1H),7.68(d,1H),3.87–3.72(m,1H),2.65–2.53(m,1H),2.50–2.38(m,2H),2.16–2.06(m,1H),1.95–1.80(m,1H),1.09(d,3H).
Example 74
5- (2, 3-dichloro-8- (2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione
Figure PCTCN2021136249-APPB-000179
The title product 5- (2, 3-dichloro-8- (2- (trifluoromethyl) cyclopropyl) imidazo [1,2-b ] pyridazin-6-yl) pyrimidine-2, 4 (1H, 3H) -dione 74 is obtained according to the method of example 19.
MS m/z(ESI):405.8[M+1] +
1 H NMR(400MHz,DMSO-d 6 )δ11.32(s,2H),8.11(s,1H),7.82(s,1H),2.88–2.81(m,2H),1.81–1.76(m,1H),1.63–1.58(m,1H).
Biological test evaluation
The invention is further illustrated below in conjunction with test examples, which are not meant to limit the scope of the invention.
Test example 1 inhibition of free human CD73 Activity by Compounds of the invention
1 purpose of experiment:
the inhibition of the conversion of AMP to adenosine by the free human CD73protein by the compounds of the present invention was measured.
2 laboratory instrument and reagents:
2.1 instrument:
microplate reader (BioTek Synergy H1);
pipettes (Eppendorf & Rainin);
centrifuge (Eppendorf).
2.2 reagents:
recombinant Human 5' -Nucleototase/CD 73Protein, CF Protein from R & D Systems, cat# 5795-EN-010;
AMP was purchased from Sigma under the product number a1752;
Phosphate Assay Kit-PiColorLock TM purchased from Abcam corporation under the trade designation ab270004;
UltraPure TM 1M Tris-HCI Buffer, pH 7.5, available from Invitrogen under the trade designation 15567027;
magnesium chloride was purchased from Sigma under the trade designation M1028-100ML;
384 well plates were purchased from Thermo corporation under the accession number 464718.
3, experimental method:
the present experiment is based on the principle that human CD73 protein converts AMP into adenosine and produces phosphoric acid, and the enzyme activity of CD73 is characterized by detecting the content of the generated phosphoric acid. The experiment was performed in 384 well plates using 25mM Tris-HCI (pH 7.5) buffer, 5mM MgCl in the experimental system 2 0.005% Tween 20. The CD73 protein is diluted to 150pM by using an experiment buffer, added into a 384-well plate, 10 mu L of each well is centrifuged at 1000rpm for 1 minute, then compound solutions with different concentrations prepared by the experiment buffer are added into the 384-well plate, 10 mu L of each well is centrifuged at 1000rpm for 1 minute and mixed evenly, 50 mu M of AMP solution is added into the experiment well, 20 mu L of each well is centrifuged at 1000rpm for 1 minute and mixed evenly, a sealing plate is closed, and the reaction is carried out at room temperature for 45 minutes. PiColorLock was added to the experimental wells TM And Accelerator (1:100), 10. Mu.L per well, centrifuged at 1000rpm for 1 min, mixed, shaken at room temperature for 5 min, then added with Stabiliser, centrifuged at 4. Mu.L per well, centrifuged at 1000rpm for 1 min, mixed, and shaken at room temperature for 5 min. The plate was read using the absorbance program in BioTek Synergy H1 and absorbance at 635nm was measured.
4, experimental data processing method:
calculating the inhibition rate of each concentration according to the absorbance of each hole, and performing nonlinear regression curve fitting on the concentration and the inhibition rate by using Graphpad Prism software to obtain the IC 50 Values.
5 experimental results:
the method is characterized by comprising the following stepsThe compounds of the present invention show about 0.01nM to 1000nM (IC) in the inhibition assay of free human CD73 enzyme activity 50 ) Is a biological activity of (a). The specific experimental results are shown in table 1:
TABLE 1
Figure PCTCN2021136249-APPB-000180
Figure PCTCN2021136249-APPB-000181
6 conclusion of experiment:
the data show that the compound of the embodiment of the invention has stronger inhibition activity on the activity of free human CD73 enzyme.
Test example 2 inhibition of CD73 Activity of Compounds of the invention on tumor cell surfaces assay
1. The purpose of the experiment is as follows:
the inhibition of the conversion of the CD73 protein on the surface of human breast cancer cells (MDA-MB-231, CD73 high expression) to AMP into adenosine function by the compounds of the present invention was measured.
2. Laboratory instruments and reagents:
2.1 instrument:
microplate reader (BioTek Synergy H1);
pipettes (Eppendorf & Rainin);
centrifuge (Eppendorf).
2.2 reagents:
MDA-MB-231 is purchased from ATCC;
Figure PCTCN2021136249-APPB-000182
luminescent Cell Viability Assay from Promega corporation under the designation G7573;
96 well plates were purchased from Corning corporation under the trade designation 3610;
RPMI 1640 medium was purchased from Gibco under the trade designation 22400089;
ATP solutions (10 mM) were purchased from Life Technologies under the designation PV3227;
AMP was purchased from Sigma under the accession number a1752;
FBS is available from Gibco under the accession number 10091148;
pancreatin was purchased from Gibco company under the accession number 25200056;
PBS was purchased from Gibco under the trade designation 10010023.
3. The experimental method comprises the following steps:
the experiment adopts a CTG method to detect the inhibition of the compound on the conversion of CD73 protein on the surface of CD73 high-expression human breast cancer cells MDA-MB-231 into adenosine.
When MDA-MB-231 cells were cultured to a suitable degree of fusion, the cells were collected, the cells were adjusted to a suitable density using complete medium (RPMI 1640 medium containing 10% FBS), the cell suspension was spread in 96-well plates at 100. Mu.L per well, and placed at 37℃in 5% CO 2 The incubator was incubated overnight, the supernatant was removed, washed 3 times with serum-free 1640 medium, 25. Mu.L of serum-free 1640 medium and 25. Mu.L of compound solutions of different concentrations prepared using serum-free 1640 medium were added, the vehicle control was DMSO-containing serum-free 1640 medium, centrifuged at 1000rpm for 1 min, mixed well, and incubated at 37℃for 15min. 1.2mM AMP solution was added to the wells, centrifuged at 1000rpm for 1 min and mixed well at 25. Mu.L per well, and incubated at 37℃for 2 hours. Transfer supernatant was added to a new 96-well plate, 25. Mu.L per well, 100. Mu.M ATP solution was added, 25. Mu.L per well, centrifuged at 1000rpm for 1 min, mixed well, cellTiter-Glo solution was added, mixed well by shaking, incubated in the dark for 10 min, and read with a BioTek Synergy H1 microplate reader to detect luminescence signal values.
4. The experimental data processing method comprises the following steps:
calculating inhibition rate by using the luminous signal value, and performing nonlinear regression curve fitting on the concentration and inhibition rate by using Graphpad Prism software to obtain the IC 50 Values.
5. Experimental results:
the above scheme shows that the compounds shown in the invention are on MDA-MB-231 cell surfaceThe inhibition of the surface CD73 enzyme activity was shown to be about 0.01nM to 1000nM (IC 50 ) Is a biological activity of (a). The specific experimental results are shown in table 2:
TABLE 2
Figure PCTCN2021136249-APPB-000183
6. Conclusion of experiment:
the data show that the compound of the embodiment of the invention has stronger inhibition activity on the CD73 enzyme activity on the cell surface of MDA-MB-231.
Test example 3, liver microsome stability test protocol
1. Purpose of experiment
The purpose of this experiment was to examine the stability of the example compounds in liver microsomes.
2. Experimental instrument and reagent
2.1 instruments
Liquid phase mass spectrometer (Shimadzu/AB SCIEX);
centrifuge (Eppendorf);
vortex finders (IKA);
pipettes (Eppendorf);
2.2 reagents
96-well plates (Axyben, P-DW-11-C);
methanol (Fisher, A452-4);
acetonitrile (Fisher, A955-4);
human liver particles (Xenotech, H0610);
mouse liver microparticles (Xenotech, M1000);
NADPH (pich, BD 116582);
UDPGA(Sigma,U6751-500MG);
Alamethicin(Abcam,ab141893);
phosphate buffer (Gibco, 10010-023);
DMSO(Sigma,34869);
A test compound.
3. Experimental procedure
3.1 preparing working solution of Compound
Working solution preparation of the compound: the compound stock solution was added to phosphate buffer at a final concentration of 20. Mu.M.
3.2 preparation of liver microsome working solution
Diluted with 100mM phosphate buffer to a final concentration of 0.625mg/mL.
3.3 preparation of NADPH and UDPGA
NADPH and UDPGA were weighed and added to 100mM phosphate buffer, at a final concentration of 20mM.
3.4 preparation of a perforating agent (Alamethicin)
1mg of Alamethacin was weighed and 200. Mu.L of DMSO was added to prepare a 5mg/mL solution. And diluted with phosphate buffer to a final concentration of 50. Mu.g/mL.
3.5 preparing a reaction termination solution
Stop solution, namely cold acetonitrile which contains 100ng/mL of labetalol hydrochloride and 400ng/mL of tolbutamide as internal standards.
3.6 incubation procedure
400. Mu.L of the prepared liver microsomes, 25. Mu.L of the compound working solution and 25. Mu.L of Alamethacin were sequentially added to a 96-well plate, and pre-incubated at 37℃for 10min. Then 50. Mu.L of the prepared NADPH/UDPGA was added to start the reaction, and incubated at 37℃with a total volume of 500. Mu.L, and the final contents of the respective components were as follows:
Figure PCTCN2021136249-APPB-000184
3.7 sample analysis
3.7.1 chromatographic conditions
Instrument: shimadzu LC-30AD
Chromatographic column:
Figure PCTCN2021136249-APPB-000185
c18 (50 x 4.6mm,5 μm particle size);
mobile phase: a:0.1% formic acid solution, B methanol
Flushing gradient: 0.2-1.6 min 5% A to 95% A, 3.0-3.1 min 95% A to 5%A
Run time: 4.0min
3.7.2 Mass Spectrometry conditions
The instrument is an API5500 type liquid chromatography-mass spectrometer and AB Sciex company;
ion source: electrospray ionization source (ESI);
drying gas: n (N) 2 The temperature is 500 ℃;
electrospray voltage: 5000V;
the detection mode is as follows: detecting positive ions;
scanning mode: reaction Monitoring (MRM) mode.
4. Experimental results:
the results of the test experiments of the compounds of the present invention in liver microsome stability are shown in the following table 3:
TABLE 3 Table 3
Figure PCTCN2021136249-APPB-000186
4. Conclusion of experiment:
the data show that the compound of the embodiment of the invention has better metabolic stability in liver microsome stability test experiments of human and mice.
Test example 4 mouse pharmacokinetic assay
1. Study purposes:
the compounds of the examples of the present invention were studied for their pharmacokinetic behavior in plasma in mice orally administered at a dose of 50mg/kg using Balb/C mice as the test animals.
2. Test protocol
2.1 test drug:
the compound of the embodiment of the invention is self-made;
preparing the medicine:
a solvent: 20% aqueous HP-beta-CD: 20g of HP-beta-CD is weighed and dissolved in 100mL of purified water, and the mixture is vortexed, mixed uniformly and sonicated to obtain a clear solution.
The compound of the embodiment of the invention is weighed, added into a 4-mL glass bottle, added with 2.4mL solvent and subjected to ultrasonic treatment for 10 minutes to obtain colorless clear solution with the concentration of 5mg/mL.
2.2 test animals:
Balb/C Mouse 3 (3/example), male.
2.3 administration:
3 Balb/C mice, male; after one night of fasting, the dosages were 50mg/kg, respectively, and the dosing volumes were 10mL/kg.
2.4 sample collection:
mice were bled with 0.04mL of orbital blood at 0.5, 1, 2, 4, 6, 8 and 24 hours before and after dosing, and placed in EDTA-K 2 In a test tube, plasma was separated by centrifugation at 6000rpm at 4℃for 6min and stored at-80 ℃.
2.5 sample treatment:
1) The plasma sample 40uL was precipitated by adding 160uL acetonitrile, and centrifuged at 3500 Xg for 5-20 minutes after mixing.
2) The concentration of the test compound was analyzed by LC/MS/MS by taking 100uL of the supernatant solution after the treatment.
2.6 liquid phase analysis
● Liquid phase conditions: shimadzu LC-20AD pump
● Mass spectrum conditions AB Sciex API 4000 mass spectrometer
● Chromatographic column: phenomenex Gemiu 5um C18.times.4.6 mm
● Mobile phase: solution A is 0.1% formic acid water solution, solution B is acetonitrile
● Flow rate: 0.8mL/min
● Elution time: 0-4.0 minutes, the eluent is as follows:
Figure PCTCN2021136249-APPB-000187
3. test results and analysis
The main pharmacokinetic parameters were calculated using WinNonlin 6.1 and the results of the mouse drug substitution experiments are shown in table 4 below:
Table 4: oral administration pharmacokinetic parameters of the Compounds of the invention in mice
Figure PCTCN2021136249-APPB-000188
Figure PCTCN2021136249-APPB-000189
4. Conclusion of experiment:
the data in the table show that the compounds of the examples of the present invention show higher exposure after oral administration in the pharmacokinetic evaluation experiments of oral administration in mice.
Test example 5: in vivo pharmacodynamics study of compound on mouse breast cancer cell EMT-6 transplantation tumor model
5.1 Experimental purposes:
the in vivo efficacy of the compounds on a mouse breast cancer cell EMT-6 transplantation tumor model was evaluated.
5.2 laboratory instruments and reagents:
5.2.1 instruments:
1. ultra clean bench (BSC-1300 II A2, shanghai Bo Xie medical equipment factory)
2. Ultra clean bench (CJ-2F, von Willebrand laboratory animal Co., ltd.)
3、CO 2 Incubator (Thermo-311, thermo)
4. Centrifuge (Centrifuge 5720R, eppendorf)
5. Full-automatic cell counter (Countess II, life Technologies)
6. Vernier caliper (CD-6' AX, sanfeng Japan)
7. Cell culture bottle (T75/T225, corning)
8. Electronic balance (CPA 2202S, sidoris)
9. Electronic balance (BSA 2202S-CW, sidoris)
5.2.2 reagents:
1. waymouth's MB 752/1 Medium (11220-035, gibco)
2. Fetal Bovine Serum (FBS) (10099-141C, gibco)
3. Phosphate Buffered Saline (PBS) (10010-023, gibco)
4. HP-beta-CD (128446-35-5, bohui Biotech Co., ltd.)
5.3 experimental operation and data processing:
5.3.1 animals
BALB/c mice, 6-8 weeks, females.
5.3.2 cell culture and preparation of cell suspension
a, taking out an EMT-6 cell from a cell bank, resuscitating the cell with Waymouth's MB 752/1 medium (Waymouth's MB 752/1+15% FBS) containing 2mM L-glutamine, placing the resuscitated cell in a cell culture flask (labeling cell type, date, name of cultured person, etc. on the flask wall), placing the resuscitated cell in CO 2 Culturing in incubator (temperature of incubator is 37deg.C, CO) 2 Concentration 5%).
b, after the cells are spread at 80-90% of the bottom of the culture flask, passaging, and placing the cells in CO continuously after passaging 2 Culturing in an incubator. This process is repeated until the number of cells meets the in vivo pharmacodynamic requirements.
c, collecting cultured cells, counting with a fully automatic cell counter, and re-suspending the cells with PBS according to the counting result to obtain a cell suspension (density 5×10) 6 /mL), placed in an ice bin for use.
5.3.3 cell seeding
a, marking nude mice with disposable universal ear tags for the rats and the mice before inoculation.
b, mixing the cell suspension during inoculation, extracting 0.1-1mL of the cell suspension by using a 1mL syringe, removing bubbles, and then placing the syringe on an ice bag for standby.
c, the left hand is kept to be a nude mouse, the right shoulder position (inoculation position) of the right back of the nude mouse is sterilized by 75% alcohol, and the inoculation is started after 30 seconds.
d, the test nude mice were inoculated sequentially (0.1 mL cell suspension was inoculated per mouse).
5.3.4 grouping, metering and administration of tumor-bearing mice
a, grouping on day 2 after inoculation according to the weight of the mice; and according to the grouping result, starting to administer the test drug (administration mode: oral administration; administration dose: 100mg/kg; administration volume: 10mL/kg; administration frequency: 2 times/day; administration period: 18 days; vehicle: 20% HP-beta-CD).
b, tumor is measured on the 4 th to 7 th days after inoculation according to the growth condition of the tumor, and the tumor size is calculated.
Tumor volume calculation: tumor volume (mm) 3 ) =length (mm) ×width (mm)/2
c, tumor is measured twice a week and weighed.
d, euthanized animals after the end of the experiment.
e, processing the data by Excel and other software. Calculation of compound tumor inhibition rate TGI (%): when the tumor did not regress, TGI (%) = [1- (mean tumor volume at the end of dosing of a treatment group-mean tumor volume at the beginning of dosing of a treatment group)/(mean tumor volume at the end of treatment of a solvent control group-mean tumor volume at the beginning of treatment of a solvent control group) ]x100%. When there was regression of the tumor, TGI (%) = [1- (mean tumor volume at the end of dosing of a treatment group-mean tumor volume at the beginning of dosing of the treatment group)/mean tumor volume at the beginning of dosing of the treatment group ] ×100%.
5.4 experimental conclusion:
the data in the table show that in the test of evaluating pharmacodynamics of oral administration of EMT-6 model mice, the compound of the present invention shows better tumor inhibiting effect after oral administration, the TGI of the compound of the present invention is more than 40%, preferably the TGI of the compound is more than 50%, more preferably the TGI of the compound is more than 60%, and even more preferably the TGI of the compound is more than 70%.

Claims (18)

  1. A compound of formula (I), a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2021136249-APPB-100001
    wherein:
    ring A is selected from
    Figure PCTCN2021136249-APPB-100002
    Ring B is selected from cycloalkyl, heterocyclyl, aryl or heteroaryl;
    R 1 independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, or thioxo, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, - (CH) 2 ) n1 NR AA R BB 、-CR AA R BB (CH 2 ) n1 NR CC R DD 、-(CH 2 ) n1 R AA 、-CR AA R BB R CC 、-(CH 2 ) n1 OR AA 、-(CH 2 ) n1 C(O)OR AA 、-(CH 2 ) n1 OR AA 、-(CH 2 ) n1 SR AA 、-(CH 2 ) n1 NR AA C(O)(CH 2 ) n2 R BB 、-(CH 2 ) n1 NR AA C(O)OR BB 、-(CH 2 ) n1 NR AA C(O)NR BB R CC or-NR AA (CH 2 ) n1 R BB Said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl groups optionally being further substituted;
    R AA 、R BB 、R CC and R is DD Each independently selected from the group consisting of hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, optionally further substituted;
    Alternatively, R AA 、R BB 、R CC And R is DD And the nitrogen or carbon atoms to which they are attached are linked to form cycloalkyl, heterocyclyl, aryl or heteroaryl, which cycloalkyl, heterocyclyl, aryl and heteroaryl groups may optionally be further substituted;
    R 2 independently selected from the group consisting of hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, alkyl, deuterated alkoxy, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, - (CH) 2 ) n3 NR A1 R B1 、-CR A1 R B1 (CH 2 ) n3 NR C1 R D1 、-(CH 2 ) n3 R A1 、-CR A1 R B1 R C1 、-(CH 2 ) n3 OR A1 、-(CH 2 ) n3 C(O)OR A1 、-(CH 2 ) n3 OR A1 、-(CH 2 ) n3 SR A1 、-(CH 2 ) n3 NR A1 C(O)(CH 2 ) n4 R B1 、-(CH 2 ) n3 NR A1 C(O)OR B1 、-(CH 2 ) n3 NR A1 C(O)NR B1 R C1 or-NR A1 (CH 2 ) n3 R B1 Said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl groups optionally being further substituted;
    R A1 、R B1 、R C1 and R is D1 Each independently selected from the group consisting of hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, or heteroaryl, said amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, optionally further substituted;
    Alternatively, R A1 、R B1 、R C1 And R is D1 And the nitrogen or carbon atoms to which they are attached are linked to form cycloalkyl, heterocyclyl, aryl or heteroaryl, which cycloalkyl, heterocyclyl, aryl and heteroaryl groups may optionally be further substituted;
    n1 is selected from 0, 1, 2, 3, 4, 5 or 6;
    n2 is selected from 0, 1, 2, 3, 4, 5 or 6;
    n3 is selected from 0, 1, 2, 3, 4, 5 or 6;
    n4 is selected from 0, 1, 2, 3, 4, 5 or 6;
    x is selected from 0, 1, 2, 3 or 4; and is also provided with
    y is selected from 0, 1, 2, 3 or 4.
  2. The compound of claim 1, a stereoisomer or pharmaceutically acceptable salt thereof, wherein the compound is further represented by the general formula (I-a);
    Figure PCTCN2021136249-APPB-100003
    wherein:
    ring C is independently selected from C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl;
    preferably C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl;
    more preferably C 3-6 Cycloalkyl;
    further preferred are cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl or cyclohexenyl; r is each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Deuterated alkoxy, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said amino, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocycleRadical, C 6-14 One or more substituents in aryl and 5-14 membered heteroaryl;
    preferably hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, said amino, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl; z is selected from 0, 1, 2, 3, 4, 5 or 6; and is also provided with
    m is selected from 0 or 1;
    when R is 1 Is hydrogen, ring B is
    Figure PCTCN2021136249-APPB-100004
    Ring C is C 3-6 Saturated cycloalkyl, z is 1, and R 2 When hydrogen, methyl, chlorine, cyano or methoxy, R is not hydrogen,Halogen, C 1-3 Alkyl, isobutyl, C substituted by 1 or 2 fluorine atoms 1-2 An alkyl group.
  3. The compound of claim 2, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein the compound is further represented by the general formula (II-a):
    Figure PCTCN2021136249-APPB-100005
    wherein:
    Figure PCTCN2021136249-APPB-100006
    is a single bond or a double bond;
    m 1 selected from 0, 1, 2, 3 or 4.
  4. A compound according to any one of claims 1 to 3, a stereoisomer or a pharmaceutically acceptable salt thereof, wherein ring B is selected from C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl;
    Preferably C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl;
    more preferably a 5-10 membered mono-heteroaryl or a 5-10 membered di-heteroaryl;
    further preferred are 5-6 membered nitrogen containing mono-heteroaryl or 8-10 membered nitrogen containing di-heteroaryl;
    it is still further preferred that the composition,
    ring B is selected from
    Figure PCTCN2021136249-APPB-100007
    X 1 Selected from-N-or-CH-, X 2 And X 3 Each independently selected from-N-or-C-, X 4 And X 5 Each independently selected from-N-, -NH-or-CH-,
    Figure PCTCN2021136249-APPB-100008
    is a single bond or a double bond;
    alternatively, ring B is selected from
    Figure PCTCN2021136249-APPB-100009
    Y 1 、Y 2 、Y 5 、Y 6 Each independently selected from-N-or-CH-; y is Y 3 Selected from-N-, -NH-, -CH-or-CH 2 -;Y 4 Selected from-N-, -NH-, -CH 2 -or-C (O) -,
    Figure PCTCN2021136249-APPB-100010
    is a single bond or a double bond;
    most preferably
    Figure PCTCN2021136249-APPB-100011
    Figure PCTCN2021136249-APPB-100012
  5. The compound, stereoisomer or pharmaceutically acceptable salt thereof according to any one of claims 1 to 4, wherein R 1 Independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, or thio.
  6. The compound, stereoisomer or pharmaceutically acceptable salt thereof according to any one of claims 1 to 4, wherein R 2 Independently selected from hydrogen, deuterium, halogen, amino, nitro,Hydroxy, cyano, carboxy, oxo, thioxo, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Deuterated alkoxy, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said amino, hydroxy, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Deuterated alkoxy, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Deuterated alkoxy, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in aryl or 5-14 membered heteroaryl;
    preferably, R 2 Independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl group,3-8 membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, said amino, hydroxy, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in the aryl or 5-10 membered heteroaryl group.
  7. The compound of claim 4, a stereoisomer or pharmaceutically acceptable salt thereof, wherein the compound is further represented by formula (II):
    Figure PCTCN2021136249-APPB-100013
    wherein:
    R 3 and R is 4 Each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl or 5-14 membered heteroaryl, said amino, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 Aryl and 5-14 membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-8 Alkyl, C 1-8 Deuterated alkyl, C 1-8 Haloalkyl, C 1-8 Hydroxyalkyl, C 1-8 Alkoxy, C 1-8 Haloalkoxy, C 2-8 Alkenyl, C 2-8 Alkynyl, C 3-12 Cycloalkyl, 3-12 membered heterocyclyl, C 6-14 One or more substituents in aryl and 5-14 membered heteroaryl;
    preferably hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxyl, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-to 10-membered heteroaryl, said amino, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-10 membered heteroaryl, optionally further substituted with deuterium, halogen, amino,Nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl;
    more preferably hydrogen, deuterium, halogen, hydroxy, cyano, carboxy, oxo, thioxo, C 1-6 Alkyl or C 3-8 Cycloalkyl; the C is 1-6 Alkyl or C 3-8 Cycloalkyl optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl and 5-10 membered heteroaryl;
    further preferably hydrogen, halogen, C 1-3 Alkyl, C 1-3 Hydroxyalkyl, C 1-3 Deuterated alkyl, C 1-3 Haloalkyl, C 3-6 Cycloalkyl;
    still more preferred is hydrogen, fluorine, chlorine, bromine, iodine, methyl, cyclopropyl, ethyl, propyl, isopropyl or trifluoromethyl.
  8. A compound, stereoisomer or pharmaceutically acceptable salt thereof according to claim 6 or 7,
    R 2 independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl, C 1-3 Alkoxy, C 1-3 Haloalkoxy, C 3-6 Cycloalkyl, 3-6 membered heterocyclyl, C 6-10 Aryl or 5-6 membered heteroaryl;
    preferably hydrogen, deuterium, halogen, oxo, thioxo, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C 1-3 Haloalkyl, C 1-3 Alkoxy, C 1-3 Haloalkoxy, C 3-6 Cycloalkyl, 3-6 membered heterocyclyl, C 6-10 Aryl or 5-6 membered heteroaryl;
    more preferably hydrogen, halogen, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Haloalkyl, C 3-6 Cycloalkyl;
    further preferred is hydrogen, methyl, ethyl or cyclopropyl;
    y is selected from 0, 1, 2 or 3.
  9. The compound of claim 7, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein the compound is further represented by formula (II-1) or formula (II-2):
    Figure PCTCN2021136249-APPB-100014
  10. the compound of claim 6, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, wherein the compound is further represented by formula (IV), formula (VI-1), or formula (VI-2):
    Figure PCTCN2021136249-APPB-100015
    Figure PCTCN2021136249-APPB-100016
    wherein:
    ring D is selected from C 3-6 Cycloalkyl, 3-6 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl;
    preferably, ring D is selected from cyclopropyl or cyclobutyl;
    R 2 independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, oxo, thio, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy or C 1-6 Haloalkoxy groups;
    preferably hydrogen, deuterium, halogen, amino, oxo, thio, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C 1-3 Haloalkyl, C 1-3 Alkoxy or C 1-3 Haloalkoxy groups;
    more preferably hydrogen, halogen, amino, oxo, thioxo, methyl, methoxy or-CD 3;
    y is selected from 0, 1, 2 or 3.
  11. The compound of claim 2, a stereoisomer or pharmaceutically acceptable salt thereof, wherein the compound is further represented by the general formula (III-a), (III-B), (III-C), (III-D) or (III-E):
    Figure PCTCN2021136249-APPB-100017
    wherein:
    R 5 selected from C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl or 5-10 membered heteroaryl, said C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 Aryl and 5-to 10-membered heteroaryl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl, C 2-6 Alkynyl, C 3-8 Cycloalkyl, 3-8 membered heterocyclyl, C 6-10 One or more substituents in aryl or 5-10 membered heteroaryl;
    preferably, R 5 Selected from C 3-6 Cycloalkyl or 3-6 membered heterocyclyl, said C 3-6 Cycloalkyl and 3-6 membered heterocyclyl, optionally further substituted with deuterium, halogen, amino, nitro, hydroxy, cyano, carboxy, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl, C 1-3 Alkoxy, C 1-3 Haloalkoxy and C 3-6 One or more substituents in cycloalkyl are substituted;
    R 6 、R 7 、R 8 each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C 1-6 Alkyl, C 1-6 Deuterated alkyl, C 1-6 Deuterated alkoxy, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl, C 1-6 Alkoxy, C 1-6 Haloalkoxy, C 2-6 Alkenyl or C 2-6 Alkynyl;
    preferably, R 6 、R 7 、R 8 Each independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, C 1-3 Alkyl, C 1-3 Deuterated alkyl, C 1-3 Deuterated alkoxy, C 1-3 Haloalkyl, C 1-3 Hydroxyalkyl, C 1-3 Alkoxy or C 1-3 Haloalkoxy groups.
  12. The compound of claim 11, a stereoisomer or pharmaceutically acceptable salt thereof, wherein the compound is further represented by the general formula (III-C):
    Figure PCTCN2021136249-APPB-100018
    wherein:
    R 5 selected from cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl or cyclohexenyl; the cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl and cyclohexenyl radicals being optionally further substituted by halogen, methyl, ethyl, n-propyl, isopropyl, trifluoromethyl, cyclopropyl, cyclobutyl, cyclopropylmethyl or-CH 2 F is substituted by one or more substituents;
    R 6 、R 7 each independently selected from hydrogen, deuterium, fluorine, chlorine, bromine, amino, methyl, ethyl or trifluoromethyl.
  13. The compound, stereoisomer or pharmaceutically acceptable salt thereof according to any one of claims 1 to 12, wherein the specific structure of the compound is as follows:
    Figure PCTCN2021136249-APPB-100019
    Figure PCTCN2021136249-APPB-100020
    Figure PCTCN2021136249-APPB-100021
    Figure PCTCN2021136249-APPB-100022
    Figure PCTCN2021136249-APPB-100023
    Figure PCTCN2021136249-APPB-100024
    Figure PCTCN2021136249-APPB-100025
    Figure PCTCN2021136249-APPB-100026
    Figure PCTCN2021136249-APPB-100027
    Figure PCTCN2021136249-APPB-100028
    Figure PCTCN2021136249-APPB-100029
    Figure PCTCN2021136249-APPB-100030
    Figure PCTCN2021136249-APPB-100031
    Figure PCTCN2021136249-APPB-100032
    Figure PCTCN2021136249-APPB-100033
    Figure PCTCN2021136249-APPB-100034
  14. a compound represented by the general formula (a), a stereoisomer thereof, or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2021136249-APPB-100035
    wherein:
    ring B, ring C, R, R 1 、R 2 Y, m and z are as defined in claim 2.
  15. A process for preparing a compound of claim 2, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
    Figure PCTCN2021136249-APPB-100036
    wherein:
    the acid is selected from organic or inorganic acids,
    ring B, ring C, R, R 1 、R 2 Y, m and z are as defined in claim 2.
  16. A pharmaceutical composition comprising a therapeutically effective amount of a compound of any one of claims 1 to 13, a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers or excipients.
  17. Use of a compound according to any one of claims 1 to 13, a stereoisomer thereof or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 16, for the preparation of a CD73 inhibitor medicament.
  18. Use of a compound according to any one of claims 1 to 13, a stereoisomer thereof or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 16, for the manufacture of a medicament for the treatment of cancer, preferably cancer selected from colorectal cancer, bladder cancer, gastric cancer, thyroid cancer, esophageal cancer, head and neck cancer, brain cancer, glioblastoma, hepatocellular carcinoma, lung cancer, melanoma, myeloma, pancreatic cancer, renal cell carcinoma, cervical cancer, urothelial cancer, prostate cancer, ovarian cancer, breast cancer, leukemia or lymphoma.
CN202180055582.1A 2020-12-10 2021-12-08 Oxo-nitrogen ring derivative regulator, preparation method and application thereof Pending CN116134029A (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
CN202011457659 2020-12-10
CN2020114576591 2020-12-10
CN2021108195065 2021-07-20
CN202110819506 2021-07-20
CN202110841215 2021-07-23
CN2021108412156 2021-07-23
PCT/CN2021/136249 WO2022121914A1 (en) 2020-12-10 2021-12-08 Oxo-nitrogen ring derivative regulator, preparation method therefor, and application thereof

Publications (1)

Publication Number Publication Date
CN116134029A true CN116134029A (en) 2023-05-16

Family

ID=81973081

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180055582.1A Pending CN116134029A (en) 2020-12-10 2021-12-08 Oxo-nitrogen ring derivative regulator, preparation method and application thereof

Country Status (3)

Country Link
CN (1) CN116134029A (en)
TW (1) TW202227413A (en)
WO (1) WO2022121914A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023169327A1 (en) * 2022-03-07 2023-09-14 贝达药业股份有限公司 Crystal form of pyridazine derivative, and preparation method therefor and use thereof
WO2023201267A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers
WO2023233130A1 (en) * 2022-05-30 2023-12-07 AdoRx Therapeutics Limited Cd73 inhibitor compounds
US20240116928A1 (en) * 2022-07-01 2024-04-11 Gilead Sciences, Inc. Cd73 compounds
WO2024033454A1 (en) * 2022-08-12 2024-02-15 F. Hoffmann-La Roche Ag Heteroaryl compounds for the treatment of cancer
CN116874405B (en) * 2023-06-27 2024-02-20 山东轩硕医药科技有限公司 Indole compound as immunomodulator and preparation method and application thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2008012875A (en) * 2006-04-03 2008-10-13 Glaxo Group Ltd Azabicyclo [3. 1. 0] hexyl derivatives as modulators of dopamine d3 receptors.
WO2017098421A1 (en) * 2015-12-08 2017-06-15 Glaxosmithkline Intellectual Property Development Limited Benzothiadiazine compounds
TWI702954B (en) * 2018-03-01 2020-09-01 美商美國禮來大藥廠 Cd73 inhibitors
EP4021902B1 (en) * 2019-08-29 2024-03-20 Eli Lilly and Company Crystalline forms of a cd73 inhibitor
CN115698009A (en) * 2020-05-01 2023-02-03 吉利德科学公司 CD73 inhibiting 2,4-dioxopyrimidine compounds

Also Published As

Publication number Publication date
WO2022121914A1 (en) 2022-06-16
TW202227413A (en) 2022-07-16

Similar Documents

Publication Publication Date Title
CN116134029A (en) Oxo-nitrogen ring derivative regulator, preparation method and application thereof
JP6431593B6 (en) Pyridinylaminopyrimidine derivative, method for producing the same, and use thereof
CN113166078A (en) 2-oxoquinazoline derivatives as methionine adenosyltransferase 2A inhibitors
JP6026427B2 (en) Substituted 6,6-fused nitrogen heterocyclic compounds and uses thereof
TWI751163B (en) Fgfr4 inhibitor, its preparation method and use thereof
JP4718637B2 (en) Pyrido (2,3-D) pyrimidinone compounds and their use as PI3 inhibitors
TW202128691A (en) Kras mutein inhibitors
CN111295384A (en) Bicyclic derivative inhibitor, preparation method and application thereof
CN111212834A (en) Pyridine, pyrazine and triazine compounds as allosteric SHP2 inhibitors
CN112552294B (en) Piperazine heterocyclic derivative-containing inhibitor, preparation method and application thereof
JP2010514676A (en) 5-Cyano-4- (pyrrolo [2,3B] pyridin-3-yl) -pyrimidine derivatives useful as protein kinase inhibitors
KR20140041583A (en) Inhibitors of lrrk2 kinase activity
CN113544131B (en) Pyrrolo-heterocyclic derivative, preparation method and medical application thereof
TW202128670A (en) Nitrogen-containing polycyclic derivative inhibitor, preparation method therefor and application thereof
JP2016501251A (en) Novel bicyclic phenyl-pyridine / pyrazine for the treatment of cancer
CN107312009B (en) Quinoline compound, preparation method, intermediate, pharmaceutical composition and application thereof
CA3138648A1 (en) Indole derivative-containing inhibitor, preparation method therefor and application thereof
CN117730075A (en) Pyrazole derivative, preparation method and pharmaceutical application thereof
WO2021228248A1 (en) Fused aza-heterocyclic amide compound and use thereof
JP7273030B2 (en) Cycloolefin-substituted heteroaromatic compounds and their uses
JP2023145548A (en) Cd73 inhibitor, preparation method therefor and application thereof
TWI820414B (en) Quinazoline compounds, preparation method and use thereof
WO2022194265A1 (en) Quinazoline-based compound, composition, and application of quinazoline-based compound
JP2023505550A (en) Fluorine-substituted double bond derivatives of biphenyl groups, their production methods and pharmaceutical applications
CN117466897A (en) Highly selective FGFR2 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