CN115244058A - Pyrimidobicyclo derivatives, preparation method thereof and application thereof in medicines - Google Patents

Pyrimidobicyclo derivatives, preparation method thereof and application thereof in medicines Download PDF

Info

Publication number
CN115244058A
CN115244058A CN202180019688.6A CN202180019688A CN115244058A CN 115244058 A CN115244058 A CN 115244058A CN 202180019688 A CN202180019688 A CN 202180019688A CN 115244058 A CN115244058 A CN 115244058A
Authority
CN
China
Prior art keywords
compound
cancer
alkyl
pharmaceutically acceptable
racemate
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
CN202180019688.6A
Other languages
Chinese (zh)
Inventor
李心
蔡国栋
冯斌强
张喆
白昌
贺峰
陶维康
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 Hengrui Medicine Co Ltd
Shanghai Hengrui Pharmaceutical Co Ltd
Original Assignee
Jiangsu Hengrui Medicine Co Ltd
Shanghai Hengrui Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jiangsu Hengrui Medicine Co Ltd, Shanghai Hengrui Pharmaceutical Co Ltd filed Critical Jiangsu Hengrui Medicine Co Ltd
Publication of CN115244058A publication Critical patent/CN115244058A/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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4741Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having oxygen as a ring hetero atom, e.g. tubocuraran derivatives, noscapine, bicuculline
    • 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
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • 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
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/056Ortho-condensed systems with two or more oxygen atoms as ring hetero atoms in the oxygen-containing ring

Abstract

The present disclosure relates to pyrimido-bicyclic derivatives, methods for their preparation, and their use in medicine. In particular, the disclosure relates to pyrimido-bicyclic derivatives represented by general formula (I), a preparation method thereof, a pharmaceutical composition containing the derivatives, and a use thereof as a therapeutic agent, in particular a use thereof as an SOS1 inhibitor and a use thereof in preparing a medicament for treating a condition or disorder ameliorated by the inhibition of SOS 1.

Description

Pyrimidobicyclo derivatives, preparation method thereof and application thereof in medicines Technical Field
The disclosure belongs to the field of medicine, and relates to a pyrimido-bicyclic derivative shown as a general formula (I), a preparation method thereof, a pharmaceutical composition containing the derivative, and an application of the derivative as a therapeutic agent, in particular to an application of the derivative as an SOS1 inhibitor and an application of the derivative in preparation of a medicament for treating a condition or disorder improved by inhibition of SOS1.
Background
RAS is one of the most mutation-rate oncogenes in tumors, and about 30% of human malignancies are associated with mutations in the RAS gene. The RAS family includes KRAS, NRAS and HRAS, with KRAS mutations being most common, accounting for approximately 85%. After KRAS is activated, it regulates and controls multiple functions such as cell proliferation, survival, migration and metabolism through a plurality of downstream signaling pathways represented by RAF-MEK-ERK, PI3K-AKT-mTOR and TIAM 1-RAc. After KRAS gene mutation, the protein is continuously activated, resulting in continuous activation of downstream signaling pathways to promote tumorigenesis.
KRAS protein is considered as an unforgeable drug target for a long time because the surface of the KRAS protein lacks small molecule binding sites in the traditional sense and has ultrahigh affinity with guanylic acid and is extremely difficult to inhibit. However, based on the importance and prevalence of abnormal activation of KRAS in cancer progression, KRAS has been and remains a very interesting target for drug development. The current drug development thinking aiming at inhibiting the KRAS pathway mainly has the following aspects:
1) The small molecule covalent inhibitor developed for KRAS G12C can irreversibly lock the G12C mutant in an inactivated state, and clinical phase I data of advanced and mirati companies show unusual effects at present. However, the mutation of KRAS G12C is only one of many mutations, and other important mutants such as G12V, G12D, G12S, G12A, G13V/D and the like still lack effective drugs.
2) Finding other sites on KRAS that can target more mutants, mainly for binding to downstream effector molecules/protein molecule activation-related sites, is currently in preclinical stage, IC for activity inhibition 50 Commonly in micromolar grades.
3) For the inhibition of KRAS downstream signal protein, for example, for the development of inhibitors such as RAF, MEK, ERK, etc., the single use and multiple effects are not good clinically at present.
4) And (3) inhibiting KRAS upstream pathway, such as SHP2 inhibitor.
5) Aiming at the modification and positioning of KRAS, the membrane positioning of KRAS is blocked by farnesyl transferase and the like so as to achieve the effect of inhibiting the KRAS.
6) The expression of KRAS was knocked down by the method of RNAi.
In general, there is currently a lack of broad spectrum KRAS inhibitors other than KRAS G12C inhibitors that are effective against a variety of mutations. And the small-molecule inhibitor for selectively inhibiting SOS1, namely guanine nucleotide exchange factor (GEF) can block the activation of KRAS by interfering the RAS-SOS1 interaction, so that the aim of inhibiting the activity of KRAS in a broad spectrum can be fulfilled.
The KARS protein is a small GTPase (small GTPase) that switches intracellularly between an inactive state (bound to Guanosine Diphosphate (GDP)) and an active state (bound to Guanosine Triphosphate (GTP)). This conversion is regulated by guanine nucleotide exchange factor (GEF) and Gtpase Activator Protein (GAP). There are three major groups of GEFs of KRAS, namely SOS (seven less son) 1&2, ras-GRF and Ras-GRP, of which the latter two are expressed only in neurons and leukocytes, and only SOS is widely expressed in various tissues and is considered to play a dominant role in RAS activation. Since SOS1 is expressed in a higher amount than SOS2 and has a higher activity than SOS2, SOS1 is mainly studied at present. The specific activation pathways of SOS1 for KRAS protein are as follows: after upstream signals (such as growth factors) activate membrane surface receptors, SOS1 is activated through SHP2-Grb2, and SOS1 is combined with KRAS, and through causing a series of conformational changes, the KRAS is catalyzed to be dissociated from GDP and then combined with GTP to form active KRAS-GTP.
Relevant patents currently disclosed are WO2003082830A1, WO2018115380A1, WO2019201848A1, WO 20120069660 A1, WO1997003069A1, WO2019052046A1, CN105384745A, WO2019067543A1, US20010031760A1, WO9802434A1 and the like.
Disclosure of Invention
The purpose of the present disclosure is to provide a compound represented by the general formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure PCTCN2021000067-APPB-000001
wherein:
y is- (OCH) 2 CH 2 ) y -, where O side is attached to Z and C side is attached to O;
z is- (CR) m R n ) z -or- (CH) 2 ) v C(O)NR 0 (CH 2 ) w -;
T is- (OCH) 2 CH 2 ) t -, where O side is attached to Z and C side is attached to O;
R m and R n The same or different, each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, and hydroxyalkyl; or
R m 、R n Together with the carbon atom to which they are attached form a cycloalkyl or heterocyclyl group, which may optionally be substituted by one or more R 5 Substitution;
ring a is aryl or heteroaryl;
G 1 and G 2 Selected from nitrogen atoms or carbon atoms;
R 0 selected from the group consisting of hydrogen, alkyl, haloalkyl or hydroxyalkyl, wherein said alkyl, haloalkyl or hydroxyalkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, hydroxy, alkoxy, haloalkoxy, amino, nitro and cyano;
R 1 selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxyalkyl, cyano, and cycloalkyl;
R 2 selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, hydroxyalkyl, hydroxy, cyano, cycloalkyl, and heterocyclyl, wherein said alkyl, cycloalkyl, and heterocyclyl are optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, haloalkyl, hydroxy, hydroxyalkyl, alkoxy, haloalkoxy, amino, nitro, and cyano;
R 3 selected from the group consisting of hydrogen, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R 4 the same or different, each independently selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, hydroxyalkyl and-NR 6 R 7
R 5 The same or different, each is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkyloxy, heterocyclyloxy, aryloxy, heteroaryloxy, -NR 6 R 7 Cyano and nitro, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of alkyl, haloalkyl, alkoxy, haloalkoxy, halogen, cyano, nitro and-NR 6 R 7 Is substituted with one or more substituents of (1);
R 8 identical or different, each independently selected from halogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxy, haloalkoxy, cyano, -NR 6 R 7 Nitro, hydroxy, hydroxyalkyl, -S (O) 2 Alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of hydroxy, halogen, haloalkyl, alkoxy, haloalkoxy, cyano, nitro, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R 6 and R 7 The same or different, each independently selected from hydrogen, halogen, alkylAlkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
y is 0, 1,2,3, 4 or 5;
z is 2,3, 4 or 5;
v is 0, 1,2,3, 4 or 5;
w is 0, 1,2,3, 4 or 5;
t is 0, 1,2,3, 4 or 5; and the sum of y and t is not more than 5;
p is 0, 1,2,3, 4 or 5;
q is 0, 1 or 2;
n is 0, 1,2,3, 4 or 5.
The present disclosure provides a compound represented by general formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
Figure PCTCN2021000067-APPB-000002
wherein:
y is- (OCH) 2 CH 2 ) y -, where O side is attached to Z and C side is attached to O;
z is- (CH) 2 ) z -or- (CH) 2 ) v C(O)NR 0 (CH 2 ) w -;
T is- (OCH) 2 CH 2 ) t -, where O side is attached to Z and C side is attached to O;
ring a is aryl or heteroaryl;
G 1 and G 2 Selected from nitrogen atoms or carbon atoms;
R 0 selected from the group consisting of hydrogen, alkyl, haloalkyl or hydroxyalkyl, wherein said alkyl, haloalkyl or hydroxyalkyl is optionally substituted with a substituent selected from the group consisting of halogen, hydroxy, alkoxy,One or more substituents of haloalkoxy, amino, nitro and cyano;
R 1 selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxyalkyl, cyano, and cycloalkyl;
R 2 selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, hydroxyalkyl, hydroxy, cyano, cycloalkyl and heterocyclyl, wherein said alkyl, cycloalkyl and heterocyclyl are optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, haloalkyl, hydroxy, hydroxyalkyl, alkoxy, haloalkoxy, amino, nitro and cyano;
R 3 selected from the group consisting of hydrogen, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R 4 the same or different, each independently selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, hydroxyalkyl and-NR 6 R 7
R 5 The same or different, each is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkyloxy, heterocyclyloxy, aryloxy, heteroaryloxy, -NR 6 R 7 Cyano and nitro, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of alkyl, haloalkyl, alkoxy, haloalkoxy, halogen, cyano, nitro and-NR 6 R 7 Is substituted with one or more substituents of (a);
R 8 are the same or different and are each independently selected from halogenElements, alkyl, alkenyl, alkynyl, haloalkyl, alkoxy, haloalkoxy, cyano, -NR 6 R 7 Nitro, hydroxy, hydroxyalkyl, -S (O) 2 Alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of hydroxy, halogen, haloalkyl, alkoxy, haloalkoxy, cyano, nitro, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R 6 and R 7 The same or different, each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl;
y is 0, 1,2,3, 4 or 5;
z is 2,3, 4 or 5;
v is 0, 1,2,3, 4 or 5;
w is 0, 1,2,3, 4 or 5;
t is 0, 1,2,3, 4 or 5; and the sum of y and t is not more than 5;
p is 0, 1,2,3, 4 or 5;
q is 0, 1 or 2;
n is 0, 1,2,3, 4 or 5.
In some preferred embodiments of the present disclosure, a compound of formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is m And R n Together with the carbon atom to which they are attached form a 3-6 membered heterocyclic group, preferably a 5 membered heterocyclic group.
In some preferred embodiments of the present disclosure, a compound of formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein Z is- (CH) 2 ) z -or (a)CH 2 ) v C(O)NR 0 (CH 2 ) w -;R 0 Z, v and w are as defined in formula (I); preferably, Z is- (CH) 2 ) 2 -, and z is 2 or 4.
In some preferred embodiments of the present disclosure, a compound of formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein
Figure PCTCN2021000067-APPB-000003
Is selected from
Figure PCTCN2021000067-APPB-000004
Figure PCTCN2021000067-APPB-000005
Wherein m is 1,2,3, 4 or 5, j is 0, 1,2 or 3, k is 0, 1,2,3 or 4 0 As defined in formula (I).
In some preferred embodiments of the present disclosure, a compound of formula (I) or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, is a compound of formula (II) or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure PCTCN2021000067-APPB-000006
wherein m is 1,2,3, 4 or 5;
rings A, G 1 、G 2 、R 1 -R 5 、R 8 P, q and n are as defined in formula (I).
In some preferred embodiments of the present disclosure, a compound of formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein G 1 Is a carbon atom.
In some preferred embodiments of the present disclosure, a compound of formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein G 2 Is a carbon atom.
In some preferred embodiments of the present disclosure, a compound of formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein G 1 And G 2 Is a carbon atom.
In some preferred embodiments of the present disclosure, a compound of formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring a is C 6-10 Aryl, preferably phenyl or
Figure PCTCN2021000067-APPB-000007
More preferably phenyl.
In some preferred embodiments of the present disclosure, a compound represented by formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, which is a compound represented by formula (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure PCTCN2021000067-APPB-000008
wherein Z is- (CH) 2 ) z -;
z is 2 or 4;
y is- (OCH) 2 CH 2 ) y -, where O side is attached to Z and C side is attached to O;
t is- (OCH) 2 CH 2 ) t -, where O side is attached to Z and C side is attached to O;
y is 0, 1,2 or 3;
t is 0, 1,2 or 3; and the sum of y and t is not more than 3;
R 1 、R 2 、R 8 and n is as defined in formula (I) or (II).
In some preferred embodiments of the present disclosure, a compound of formula (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein
Figure PCTCN2021000067-APPB-000009
Is selected from
Figure PCTCN2021000067-APPB-000010
Figure PCTCN2021000067-APPB-000011
Wherein m is 1,2,3, 4 or 5, j is 0, 1,2 or 3, k is 0, 1,2,3 or 4 0 As defined in general formula (III) or (III-1).
In some preferred embodiments of the present disclosure, a compound of formula (I), (II), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, or derivative thereofIn the form of a isomer, diastereoisomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R 8 Are the same or different and are each independently selected from halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Alkoxy radical, C 1-6 Haloalkoxy, cyano, -NR 6 R 7 Hydroxy and C 1-6 Hydroxyalkyl radical, wherein said C 1-6 Alkyl and C 1-6 Haloalkyl being optionally selected from hydroxy, C 1-6 Alkoxy radical, C 1-6 Haloalkoxy, cyano and C 1-6 Substituted by one or more substituents of hydroxyalkyl, R 6 And R 7 Are the same or different and are each independently selected from hydrogen, C 1-6 Alkyl and C 1-6 A haloalkyl group;
preferably, R 8 Are the same or different and are each independently selected from halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -NR 6 R 7 And C 1-6 Hydroxyalkyl, wherein said C 1-6 Haloalkyl is optionally substituted with one or more hydroxy groups; r is 6 And R 7 Selected from hydrogen and C 1-6 An alkyl group.
In some preferred embodiments of the present disclosure, a compound of formula (I), (II), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is 8 Are the same or different and are each independently selected from halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Alkoxy radical, C 1-6 Haloalkoxy, cyano, -NR 6 R 7 Hydroxy and C 1-6 Hydroxyalkyl radical, wherein said C 1-6 Alkyl is optionally selected from hydroxy, halogen, C 1-6 Alkoxy radical, C 1-6 Haloalkoxy, cyano and C 1-6 One or more substitutions in hydroxyalkylIs substituted by radicals R 6 And R 7 Are the same or different and are each independently selected from hydrogen, C 1-6 Alkyl and C 1-6 A haloalkyl group.
In some preferred embodiments of the present disclosure, a compound of formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein
Figure PCTCN2021000067-APPB-000012
Is composed of
Figure PCTCN2021000067-APPB-000013
R 8 Are the same or different and are each independently selected from halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -NR 6 R 7 And C 1-6 Hydroxyalkyl radical, wherein said C 1-6 Haloalkyl is optionally substituted with one or more hydroxy; r is 6 And R 7 Selected from hydrogen and C 1-6 An alkyl group; preferably, R 8 Are the same or different and are each independently selected from amino, halogen and C 1-6 Haloalkyl, said C 1-6 Haloalkyl may be optionally substituted with hydroxy.
In some preferred embodiments of the present disclosure, a compound of formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein
Figure PCTCN2021000067-APPB-000014
Is composed of
Figure PCTCN2021000067-APPB-000015
R 8 Are the same or different and are each independently selected from halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -NR 6 R 7 And C 1-6 Hydroxyalkyl, wherein said C 1-6 Alkyl optionally substituted with one or more hydroxy and halogen; r 6 And R 7 Selected from hydrogen and C 1-6 An alkyl group.
In some preferred embodiments of the present disclosure, a compound of formula (I), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is 0 Selected from hydrogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; hydrogen is preferred.
In some preferred embodiments of the present disclosure, a compound of formula (I), (II), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is 1 Selected from hydrogen, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, C 1-6 Alkoxy radical, C 1-6 Haloalkoxy, C 1-6 Hydroxyalkyl and cyano; preferably, R 1 Selected from hydrogen, C 1-6 Alkyl and halogen; more preferably methyl.
In some preferred embodiments of the present disclosure, a compound of formula (I), (II), (III), or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is 2 Selected from hydrogen, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl and cyano; preferably, R 2 Selected from hydrogen and C 1-6 An alkyl group; more preferably methyl.
In some preferred embodiments of the present disclosure, a compound of formula (I) or (II) or a tautomer, mesoIn the form of a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R 3 Selected from hydrogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; preferably, R 3 Selected from hydrogen and C 1-6 An alkyl group; more preferably hydrogen.
In some preferred embodiments of the present disclosure, a compound of formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is 4 Are the same or different and are each independently selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; preferably, R 4 Is hydrogen.
In some preferred embodiments of the present disclosure, a compound of formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is 5 Are the same or different and are each independently selected from hydrogen, halogen, C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, amino and cyano; preferably, R 5 Are the same or different and are each independently selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; more preferably, R 5 Is hydrogen.
In some preferred embodiments of the present disclosure, a compound of formula (I), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein y is 0, 1,2 or 3, preferably 1.
In some preferred embodiments of the present disclosure, a compound of formula (I), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein z is 2,4 or 5, preferably 2.
In some preferred embodiments of the present disclosure, a compound of formula (I), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein v is 1.
In some preferred embodiments of the present disclosure, a compound of formula (I), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein w is 2.
In some preferred embodiments of the present disclosure, a compound of formula (I), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein t is 0, 1,2 or 3, preferably 1.
In some preferred embodiments of the present disclosure, a compound of formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein p is 0 or 1, preferably 0.
In some preferred embodiments of the present disclosure, a compound of formula (I) or (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein q is 0 or 1, preferably 0.
In some preferred embodiments of the present disclosure, a compound of formula (I), (II), (III), or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein n is 2.
In some preferred embodiments of the present disclosure, a compound of formula (I), (II), (III), or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein m is 1,2,3, or 4, preferably 3 or 4, more preferably 3.
In some preferred embodiments of the present disclosure, a compound of formula (I), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein j is 1.
In some preferred embodiments of the present disclosure, a compound of formula (I), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein k is 1.
In some preferred embodiments of the present disclosure, a compound of formula (I), (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein Z is- (CH) 2 ) i -(CR m R n )-(CH 2 ) r -,R m 、R n Together with the carbon atom to which they are attached form a cycloalkyl or heterocyclyl group, which may optionally be substituted by one or more R 5 Substitution; i is 0, 1,2 or 3, R is 0, 1,2 or 3, R5 is as defined in formula (I), (III) or (III-1); preferably, R m 、R n Together with the carbon atom to which they are attached form a 3-8 membered heterocyclyl, optionally substituted by halogen, C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, -NR 6 R 7 And one or more radicals of cyano, R 6 And R 7 Same or different, each independently selected from hydrogen, C 1-6 Alkyl and C 1-6 Haloalkyl, i is 1 or2, r is 1 or 2.
In some preferred embodiments of the present disclosure, a compound of formula (II)Or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein ring a is phenyl or
Figure PCTCN2021000067-APPB-000016
m is 1,2,3 or 4; r 8 Are the same or different and are each independently selected from halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -NR 6 R 7 And C 1-6 Hydroxyalkyl radical, wherein said C 1-6 Alkyl optionally substituted with one or more hydroxy and halogen; r is 6 And R 7 Selected from hydrogen and C 1-6 An alkyl group; r 1 Selected from hydrogen, C 1-6 Alkyl and halogen; r is 2 Selected from hydrogen, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl and cyano; r 3 Selected from hydrogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; r is 4 Are the same or different and are each independently selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; r is 5 Are the same or different and are each independently selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; g 1 And G 2 Is a carbon atom; p is 0 or 1; q is 0 or I; n is 2.
In some preferred embodiments of the present disclosure, a compound of formula (III) or (III-1) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein
Figure PCTCN2021000067-APPB-000017
Is selected from
Figure PCTCN2021000067-APPB-000018
Figure PCTCN2021000067-APPB-000019
Wherein m is 1,2,3 or 4, j is 1 or2, k is 0, 1 or 2; r is 0 Selected from hydrogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; r 8 Are the same or different and are each independently selected from halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -NR 6 R 7 And C 1-6 Hydroxyalkyl, wherein said C 1-6 Alkyl optionally substituted with one or more hydroxy and halogen; r is 6 And R 7 Selected from hydrogen or C 1-6 An alkyl group; r is 1 Selected from hydrogen, C 1-6 Alkyl and halogen; r is 2 Selected from hydrogen, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl and cyano; n is 2.
In some preferred embodiments of the present disclosure, a compound of formula (I) or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, is a compound of formula (IV) or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure PCTCN2021000067-APPB-000020
wherein
n is 1,2,3, 4 or 5;
ring A, Y, Z, T, G 1 、G 2 、R 1 -R 5 、R 8 P and q are as defined in formula (I).
Table a typical compounds of the present disclosure include, but are not limited to;
Figure PCTCN2021000067-APPB-000021
Figure PCTCN2021000067-APPB-000022
Figure PCTCN2021000067-APPB-000023
Figure PCTCN2021000067-APPB-000024
another aspect of the present disclosure relates to a compound of formula (IA) or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure PCTCN2021000067-APPB-000025
wherein:
R w is hydroxy or halogen, preferably hydroxy or chloro;
R 1 selected from the group consisting of halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxyalkyl, cyano, and cycloalkyl;
y is- (OCH) 2 CH 2 ) y -, where O side is attached to Z and C side is attached to O;
z is- (CH) 2 ) z -or- (CH) 2 ) v C(O)NR 0 (CH 2 ) w -;
T is- (OCH) 2 CH 2 ) t -, where O side is attached to Z and C side is attached to O;
y is 0, 1,2,3, 4 or 5;
t is 0, 1,2,3, 4 or 5; and the sum of y and t is not 0;
G 1 、G 2 、R 4 -R 5 、R 0 z, v, w, p and q are as defined in formula (I).
Another aspect of the present disclosure relates to a compound of formula (IA) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
Figure PCTCN2021000067-APPB-000026
wherein:
R w is hydroxy or halogen, preferably hydroxy or chloro;
R 1 selected from the group consisting of halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxyalkyl, cyano, and cycloalkyl;
y is- (OCH) 2 CH 2 ) y -, where O side is attached to Z and C side is attached to O;
z is- (CR) m R n ) z -or- (CH) 2 ) v C(O)NR 0 (CH 2 ) w -;
T is- (OCH) 2 CH 2 ) t -, where O side is attached to Z and C side is attached to O;
y is 0, 1,2,3, 4 or 5;
t is 0, 1,2,3, 4 or 5; and the sum of y and t is not 0;
G 1 、G 2 、R 4 -R 5 、R 0 、R m 、R n z, v, w, p and qAs defined in formula (I).
Another aspect of the present disclosure relates to a compound of formula (IVA) or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
Figure PCTCN2021000067-APPB-000027
wherein;
n is 1,2,3, 4 or 5; rings A, Y, Z, T, G 1 、G 2 、R 1 -R 5 、R 8 P and q are as defined in formula (I).
Typical intermediate compounds of the present disclosure include, but are not limited to:
Figure PCTCN2021000067-APPB-000028
Figure PCTCN2021000067-APPB-000029
Figure PCTCN2021000067-APPB-000030
another aspect of the present disclosure relates to a process for preparing a compound of formula (I) or (IV) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
Figure PCTCN2021000067-APPB-000031
Figure PCTCN2021000067-APPB-000032
reacting the general formula (IA) with the general formula (IB) or a salt thereof (preferably hydrochloride) to obtain a compound of the general formula (I), or subjecting the compound of the general formula (IVA) to a reduction reaction to obtain a compound of the general formula (IV);
wherein:
R w is hydroxy or halogen, preferably hydroxy or chlorine; in the general formula (I), n is 0, 1,2,3, 4 or 5;
in the general formula (IV), n is 1,2,3, 4 or 5;
ring A, Y, Z, T, G 1 、G 2 、R 1 -R 5 、R 8 P and q are as defined in formula (I).
Another aspect of the present disclosure relates to a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (I), (II), (III-1) or table a of the present disclosure, or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
The present disclosure further relates to the use of a compound of formula (I), (II), (III-1) or table a, or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for the manufacture of a medicament for the inhibition of SOS1.
The present disclosure further relates to the use of a compound of general formula (I), (II), (III-1) or table a or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for the manufacture of a medicament for the treatment and/or prevention of SOS 1-mediated diseases, preferably selected from the group consisting of cancer, inflammation, RAS disease, noonan Syndrome (NS), noonan syndrome with erythema (NSML), capillary malformation-arteriovenous malformation syndrome (CM-AVM), costello Syndrome (CS), cardiac-facial-skin syndrome (CFC), louse syndrome, hereditary gingival fibromatosis, or other proliferative diseases.
The present disclosure further relates to the use of a compound of general formula (I), (II), (III-1) or table a or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for the manufacture of a medicament for the treatment and/or prevention of cancer, inflammation, RAS disease, noonan Syndrome (NS), noonan syndrome with erythema (NSML), capillary malformation-arteriovenous malformation syndrome (CM-AVM), costello Syndrome (CS), heart-plane-skin syndrome (CFC), lounge syndrome, hereditary gingival fibromatosis, or other proliferative diseases, preferably cancer; the cancer is preferably selected from melanoma, skin cancer, liver cancer, kidney cancer, lung cancer, nasopharyngeal cancer, gastric cancer, esophageal cancer, colorectal cancer, colon cancer, rectal cancer, gallbladder cancer, cholangiocarcinoma, chorioepithelioma, pancreatic cancer, polycythemia vera, pediatric tumors, cervical cancer, ovarian cancer, breast cancer, bladder cancer, urothelial cancer, ureteral tumor, prostate cancer, seminoma, testicular tumor, leukemia, head and neck tumor, head and neck squamous cell carcinoma, uterine cancer, endometrial cancer, thyroid cancer, lymphoma, sarcoma, osteoma, osteosarcoma, neuroblastoma, brain tumor, myeloma, astrocytoma, glioblastoma and glioma; the RAS disease is preferably neurofibromatosis type 1 (NF 1); the lung cancer is preferably non-small cell lung cancer, and is further preferably metastatic non-small cell lung cancer; the leukemia is preferably chronic lymphocytic leukemia or acute myelogenous leukemia; the lymphoma is preferably diffuse large B cell lymphoma; the myeloma is preferably multiple myeloma; the liver cancer is preferably hepatocellular carcinoma; the osteoma is preferably osteochondroma.
The present disclosure also relates to a method of inhibiting SOS1 comprising administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I), (II), (III-1), or Table A, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
The present disclosure also relates to a method of treating and/or preventing SOS1 mediated diseases, comprising administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I), (II), (III-1), or table a, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, preferably selected from the group consisting of cancer, inflammation, RAS disease, noonan Syndrome (NS), noonan syndrome with erythema (NSML), capillary malformation-arteriovenous malformation syndrome (CM-AVM), costello Syndrome (CS), heart-face-skin syndrome (CFC), regis syndrome, hereditary gingival fibromatosis, or other proliferative diseases.
The present disclosure also relates to a method of treating and/or preventing cancer, inflammation, PAS disease, noonan Syndrome (NS), noonan syndrome with erythema (NSML), capillary malformation-arteriovenous malformation syndrome (CM-AVM), costerlos Syndrome (CS), cardiac-facial-skin syndrome (CFC), louses syndrome, hereditary gingival fibromatosis, or other proliferative diseases, preferably cancer, comprising administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I), (II), (III-1) or table a, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same; wherein said cancer is preferably selected from the group consisting of melanoma, skin cancer, liver cancer, kidney cancer, lung cancer, nasopharyngeal cancer, gastric cancer, esophageal cancer, colorectal cancer, colon cancer, rectal cancer, gallbladder cancer, cholangiocarcinoma, chorioepithelioma, pancreatic cancer, polycythemia vera, pediatric tumors, cervical cancer, ovarian cancer, breast cancer, bladder cancer, urothelial cancer, ureteral tumor, prostate cancer, seminoma, testicular tumor, leukemia, head and neck tumor, head and neck squamous cell carcinoma, uterine cancer, endometrial cancer, thyroid cancer, lymphoma, sarcoma, osteoma, osteosarcoma, neuroblastoma, brain tumor, myeloma, astrocytoma, glioblastoma and glioma; the PAS disease is preferably neurofibromatosis type 1 (NF 1); the lung cancer is preferably non-small cell lung cancer, and is further preferably metastatic non-small cell lung cancer; the leukemia is preferably chronic lymphocytic leukemia or acute myelogenous leukemia; the lymphoma is preferably diffuse large B cell lymphoma; the myeloma is preferably multiple myeloma; the liver cancer is preferably hepatocellular carcinoma; the osteoma is preferably osteochondroma.
The present disclosure further relates to a compound of general formula (I), (II), (III-1) or table a or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same, for use as a medicament.
The present disclosure also relates to compounds of general formula (I), (II), (III-1) or table a, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for use as an SOS1 inhibitor.
The present disclosure also relates to compounds of general formula (I), (II), (III-1) or table a or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for use in the treatment and/or prevention of SOS 1-mediated diseases, preferably selected from the group consisting of cancer, inflammation, RAS disease, noonan Syndrome (NS), noonan syndrome with erythema (NSML), capillary malformation-arteriovenous malformation syndrome (CM-AVM), costello Syndrome (CS), cardiac-facial-skin syndrome (CFC), regus syndrome, hereditary gingival fibromatosis, or other proliferative diseases.
The present disclosure also relates to compounds of general formula (I), (II), (III-1) or table a or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for use in the treatment and/or prevention of cancer, inflammation, RAS disease, noonan Syndrome (NS), noonan syndrome with erythema (NSML), capillary malformation-arteriovenous malformation syndrome (CM-AVM), costello Syndrome (CS), heart-plane-skin syndrome (CFC), louses syndrome, hereditary gingival fibromatosis, or other proliferative diseases, preferably for use in the treatment and/or prevention of cancer; wherein said cancer is preferably selected from the group consisting of melanoma, skin cancer, liver cancer, kidney cancer, lung cancer, nasopharyngeal cancer, stomach cancer, esophageal cancer, colorectal cancer, colon cancer, rectal cancer, gallbladder cancer, bile duct cancer, chorioepithelial cancer, pancreatic cancer, polycythemia vera, pediatric tumors, cervical cancer, ovarian cancer, breast cancer, bladder cancer, urothelial cancer, ureteral tumor, prostate cancer, seminoma, testicular tumor, leukemia, head and neck tumor, head and neck squamous cell carcinoma, uterine cancer, endometrial cancer, thyroid cancer, lymphoma, sarcoma, osteoma, osteosarcoma, neuroblastoma, brain tumor, myeloma, astrocytoma, glioblastoma and glioma; the RAS disease is preferably neurofibromatosis type 1 (NF 1); the lung cancer is preferably non-small cell lung cancer, and is further preferably metastatic non-small cell lung cancer; the leukemia is preferably chronic lymphocytic leukemia or acute myelogenous leukemia; the lymphoma is preferably diffuse large B cell lymphoma; the myeloma is preferably multiple myeloma; the liver cancer is preferably hepatocellular carcinoma; the osteoma is preferably osteochondroma.
The cancer described in the present disclosure is preferably selected from melanoma, skin cancer, liver cancer, kidney cancer, lung cancer, nasopharyngeal cancer, gastric cancer, esophageal cancer, colorectal cancer, colon cancer, rectal cancer, gallbladder cancer, bile duct cancer, chorioepithelial cancer, pancreatic cancer, polycythemia vera, pediatric tumors, cervical cancer, ovarian cancer, breast cancer, bladder cancer, urothelial cancer, ureteral tumor, prostate cancer, seminoma, testicular tumor, leukemia, head and neck tumor, head and neck squamous cell cancer, uterine cancer, endometrial cancer, thyroid cancer, lymphoma, sarcoma, osteoma bone tumor, osteosarcoma, neuroblastoma, brain tumor, myeloma, astrocytoma, glioblastoma and glioma; the RAS disease is preferably neurofibromatosis type 1 (NF 1); the lung cancer is preferably non-small cell lung cancer, and is further preferably metastatic non-small cell lung cancer; the leukemia is preferably chronic lymphocytic leukemia or acute myelogenous leukemia; the lymphoma is preferably diffuse large B cell lymphoma; the myeloma is preferably multiple myeloma; the liver cancer is preferably hepatocellular carcinoma; the osteoma is preferably osteochondroma.
The active compounds may be formulated in a form suitable for administration by any suitable route, using one or more pharmaceutically acceptable carriers, to formulate the compositions of the disclosure by conventional means. Thus, the active compounds of the present disclosure may be formulated in a variety of dosage forms for oral administration, injection (e.g., intravenous, intramuscular, or subcutaneous), inhalation, or insufflation. The compounds of the present disclosure may also be formulated in sustained release dosage forms, such as tablets, hard or soft capsules, aqueous or oily suspensions, emulsions, injections, dispersible powders or granules, suppositories, lozenges, or syrups.
As a general guide, the active compound is preferably administered in a unit dose, or in a manner such that the patient can self-administer the compound in a single dose. The unit dose of a compound or composition of the present disclosure may be expressed in the form of a tablet, capsule, cachet, bottled liquid, powder, granule, lozenge, suppository, reconstituted powder, or liquid. Suitable unit doses may be from 0.1 to 1000mg.
The pharmaceutical compositions of the present disclosure may contain, in addition to the active compound, one or more excipients selected from the following: fillers (diluents), binders, wetting agents, disintegrants, excipients, and the like. Depending on the method of administration, the compositions may contain from 0.1 to 99% by weight of active compound.
Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be inert excipients, granulating agents, disintegrating agents, binding agents and lubricating agents. These tablets may be uncoated or they may be coated by known techniques which mask the taste of the drug or delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
Oral formulations may also be provided in soft gelatin capsules wherein the active ingredient is mixed with an inert solid diluent or wherein the active ingredient is mixed with a water soluble carrier or an oil vehicle.
Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending, dispersing or wetting agents. Aqueous suspensions may also contain one or more preservatives, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents.
Oil suspensions may be formulated by suspending the active ingredient in a vegetable or mineral oil. The oil suspension may contain a thickener. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable preparation. These compositions can be preserved by the addition of an antioxidant.
The pharmaceutical compositions of the present disclosure may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, or a mineral oil, or a mixture thereof. Suitable emulsifiers may be naturally occurring phospholipids, and the emulsions may also contain sweetening, flavoring, preservative and antioxidant agents. Such formulations may also contain a demulcent, a preservative, a colorant and an antioxidant.
The pharmaceutical compositions of the present disclosure may be in the form of a sterile injectable aqueous solution. Among the acceptable vehicles or solvents that may be employed are water, ringer's solution and isotonic sodium chloride solution. The sterile injectable preparation may be a sterile injectable oil-in-water microemulsion, in which the active ingredient is dissolved in the oil phase, the injection solution or microemulsion being injectable in the bloodstream of a patient by local bolus injection. Alternatively, it may be desirable to administer the solutions and microemulsions in a manner that maintains a constant circulating concentration of the disclosed compounds. To maintain such a constant concentration, a continuous intravenous delivery device may be used. An example of such a device is an intravenous pump of the model Deltec CADD-PLUS. TM. 5400.
The pharmaceutical compositions of the present disclosure may be in the form of sterile injectable aqueous or oleaginous suspensions for intramuscular and subcutaneous administration. The suspensions may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a parenterally-acceptable, non-toxic diluent or solvent. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. Any blend fixed oil may be used for this purpose. In addition, fatty acids can also be used to prepare injections.
The compounds of the present disclosure may be administered in the form of suppositories for rectal administration. These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid in the rectum and therefore will melt in the rectum to release the drug.
The compounds of the present disclosure can be administered by the addition of water to prepare water-suspended dispersible powders and granules. These pharmaceutical compositions may be prepared by mixing the active ingredient with dispersing or wetting agents, suspending agents, or one or more preservatives.
As is well known to those skilled in the art, the dosage of the drug administered depends on a variety of factors, including but not limited to the following: the activity of the particular compound employed, the age of the patient, the weight of the patient, the health condition of the patient, the behavior of the patient, the diet of the patient, the time of administration, the mode of administration, the rate of excretion, the combination of drugs, the severity of the disease, and the like; in addition, the optimal treatment regimen, such as mode of treatment, daily amount of compound or type of pharmaceutically acceptable salt, can be verified according to conventional treatment protocols.
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 1 to 20 carbon atoms, preferably an alkyl group containing 1 to 12 (e.g., 1,2,3, 4, 5,6, 7,8, 9, 10, 11, and 12) carbon atoms, more preferably an alkyl group containing 1 to 6 carbon atoms. <xnotran> , , , , , , , , ,1,1- ,1,2- ,2,2- ,1- ,2- ,3- , ,1- -2- ,1,1,2- ,1,1- ,1,2- ,2,2- ,1,3- ,2- ,2- ,3- ,4- ,2,3- , ,2- ,3- ,4- ,5- ,2,3- ,2,4- ,2,2- ,3,3- ,2- ,3- , ,2,3- ,2,4- ,2,5- ,2,2- ,3,3- ,4,4- ,2- ,3- ,4- ,2- -2- ,2- -3- , ,2- -2- ,2- -3- , </xnotran> 2, 2-diethylpentyl, n-decyl, 3-diethylhexyl, 2-diethylhexyl, and various branched isomers thereof, and the like. More preferred are lower alkyl groups having 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. Alkyl groups may be substituted or unsubstituted and when substituted, the substituents may be substituted at any available point of attachment, preferably the substituents are independently optionally one or more substituents selected from the group consisting of D atom, halogen, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl.
The term "alkylene" refers to a saturated straight or branched chain aliphatic hydrocarbon radical, which is a residue derived from the parent alkane by removal of two hydrogen atoms from the same carbon atom or two different carbon atoms, and is a straight or branched chain radical containing from 1 to 20 carbon atoms, preferably from 1 to 12 (e.g., 1,2,3, 4, 5,6, 7,8, 9, 10, 11, and 12) carbon atoms, more preferably an alkylene radical containing from 1 to 6 carbon atoms. Non-limiting examples of alkylene groups include, but are not limited to, methylene (-CH) 2 -), 1-ethylene (-CH (CH) 3 ) -), 1, 2-ethylene (-CH) 2 CH 2 ) -, 1-propylene (-CH (CH) 2 CH 3 ) -), 1, 2-propylene (-CH) 2 CH(CH 3 ) -), 1, 3-propylene (-CH) 2 CH 2 CH 2 -) 1, 4-butylene (-CH 2 CH 2 CH 2 CH 2 -) and the like. The alkylene groups may be substituted or unsubstituted, and when substituted, the substituents may be substituted at any available point of attachment, preferably independently with one or more substituents optionally selected from alkenyl, alkynyl, alkoxy, haloalkoxy, cycloalkyloxy, heterocyclyloxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocyclyloxy, cycloalkylthio, heterocyclylthio and oxo.
The term "alkenyl" refers to an alkyl compound containing a carbon-carbon double bond in the molecule, wherein alkyl is as defined above. The alkenyl group may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more substituents independently selected from the group consisting of alkoxy, halogen, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
The term "alkynyl" refers to an alkyl compound containing a carbon-carbon triple bond in the molecule, wherein alkyl is as defined above. Alkynyl groups may be substituted or unsubstituted and when substituted, the substituents are preferably one or more substituents independently selected from alkoxy, halogen, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
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, preferably from 3 to 8 (e.g., 3, 4, 5,6, 7, and 8) carbon atoms, 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.
The term "spirocycloalkyl" refers to a 5 to 20 membered polycyclic group sharing one carbon atom (referred to as a spiro atom) between single rings, which may contain one or more double bonds. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8,9 or 10). Spirocycloalkyl groups are classified into a single spirocycloalkyl group, a double spirocycloalkyl group or a multi spirocycloalkyl group, according to the number of spiro atoms shared between rings, and preferably a single spirocycloalkyl group and a double spirocycloalkyl group. More preferably 3-membered/5-membered, 3-membered/6-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered, spirocycloalkyl. Non-limiting examples of spirocycloalkyl groups include:
Figure PCTCN2021000067-APPB-000033
the term "fused ring alkyl" refers to a 5 to 20 membered all carbon polycyclic group in which each ring in the system shares an adjacent pair of carbon atoms with other rings in the system, wherein one or more of the rings may contain one or more double bonds. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8,9 or 10). Bicyclic, tricyclic, tetracyclic, or polycyclic fused ring alkyls may be classified according to the number of constituent rings, preferably bicyclic or tricyclic, more preferably 3-membered/4-membered, 3-membered/5-membered, 3-membered/6-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/4-membered, 5-membered/5-membered, 5-membered/6-membered, 6-membered/3-membered, 6-membered/4-membered, 6-membered/5-membered, and 6-membered/6-membered bicycloalkyl groups. Non-limiting examples of fused ring alkyl groups include:
Figure PCTCN2021000067-APPB-000034
the term "bridged cycloalkyl" refers to a 5 to 20 membered all carbon polycyclic group in which any two rings share two carbon atoms not directly attached, which may contain one or more double bonds. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8,9 or 10). They may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl groups, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic, according to the number of constituent rings. Non-limiting examples of bridged cycloalkyl groups include:
Figure PCTCN2021000067-APPB-000035
the cycloalkyl ring includes a cycloalkyl ring (including monocyclic, spiro, fused and bridged rings) fused to an aryl, heteroaryl or heterocycloalkyl ring as described above, wherein the rings attached to the parent structure are cycloalkyl, non-limiting examples of which include indanyl, tetrahydronaphthyl, benzocycloheptanyl, and the like; indanyl, tetrahydronaphthyl are preferred.
Cycloalkyl groups may be substituted or unsubstituted, and when substituted, substituents may be substituted at any available point of attachment, preferably independently with one or more substituents optionally selected from halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl.
The term "alkoxy" refers to-O- (alkyl), wherein alkyl is as defined above. Non-limiting examples of alkoxy groups include: methoxy, ethoxy, propoxy and butoxy. Alkoxy groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more groups independently selected from D atoms, halogen, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
The term "heterocyclyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising 3 to 20 ring atoms wherein one or more of the ring atoms is selected from nitrogen, oxygen, sulfur, S (O) or S (O) 2 <xnotran> , -O-O-, -O-S- -S-S- , . </xnotran> Preferably 3 to 12 (e.g., 3, 4, 5,6, 7,8, 9, 10, 11 and 12) ring atoms, of which 1 to 4 (e.g., 1,2,3 and 4) are heteroatoms; more preferably 3 to 8 (e.g., 3, 4, 5,6, 7 and 8) ring atoms, of which 1-3 (e.g., 1,2 and 3) are heteroatoms; more preferably 3 to 6 ring atoms, of which 1-3 are heteroatoms; most preferably 5 or 6 ring atoms, of which 1 to 3 are heteroatoms. Non-limiting examples of monocyclic heterocyclyl groups include pyrrolidinyl, tetrahydropyranyl, 1,2,3, 6-tetrahydropyridinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like. Polycyclic heterocyclic groups include spiro, fused, and bridged heterocyclic groups.
The term "spiroheterocyclyl" refers to a 5 to 20 membered polycyclic heterocyclic group having a single ring atom (S) common between the rings (referred to as spiro atoms), wherein one or more of the ring atoms is selected from nitrogen, oxygen, sulfur, S (O) or S (O) 2 The remaining ring atoms are carbon. It may contain one or more double bonds. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8,9 or 10). The spiro heterocyclic group is classified into a mono-spiro heterocyclic group, a di-spiro heterocyclic group or a multi-spiro heterocyclic group, preferably a mono-spiro heterocyclic group and a di-spiro heterocyclic group, according to the number of spiro atoms shared between rings. More preferably a 3-membered/5-membered, 3-membered/6-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered monospiroheterocyclyl group. Non-limiting examples of spiro heterocyclyl groups include:
Figure PCTCN2021000067-APPB-000036
the term "fused heterocyclyl" refers to a 5 to 20 membered polycyclic heterocyclic group in which each ring in the system shares an adjacent pair of atoms with the other rings in the system, and one or more of the rings may contain one or more double bonds in which one or more of the ring atoms is selected from nitrogen, oxygen, sulfur, S (O) or S (O) 2 The remaining ring atoms are carbon. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8,9 or 10). They may be classified into bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic groups according to the number of constituting rings, preferably bicyclic or tricyclic, more preferably 3-membered/4-membered, 3-membered/5-membered, 3-membered/6-membered, 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/4-membered, 5-membered/5-membered, 5-membered/6-membered, 6-membered/3-membered, 6-membered/4-membered, 6-membered/5-membered and 6-membered/6-membered bicyclic fused heterocyclic groups. Non-limiting examples of fused heterocyclic groups include:
Figure PCTCN2021000067-APPB-000037
Figure PCTCN2021000067-APPB-000038
the term "bridged heterocyclyl" refers to 5 to 14 membered polycyclic heterocyclic group wherein any two rings share two atoms not directly attached, which may contain one or more double bonds wherein one or more of the ring atoms is selected from nitrogen, oxygen, sulfur, S (O) or S (O) 2 The remaining ring atoms are carbon. Preferably 6 to 14, more preferably 7 to 10 (e.g.7, 8,9 or 10). They may be classified into bicyclic, tricyclic, tetracyclic or polycyclic bridged heterocyclic groups according to the number of constituent rings, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of bridged heterocyclic groups include:
Figure PCTCN2021000067-APPB-000039
the heterocyclyl ring includes heterocyclyl (including monocyclic, spiroheterocyclic, fused heterocyclic, and bridged heterocyclic) fused to an aryl, heteroaryl, or cycloalkyl ring as described above, wherein the ring to which the parent structure is attached is heterocyclyl, non-limiting examples of which include:
Figure PCTCN2021000067-APPB-000040
and so on.
The heterocyclyl group may be substituted or unsubstituted and when substituted, the substituents may be substituted at any available point of attachment, preferably the substituents are independently optionally one or more substituents selected from halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl.
The term "aryl" refers to a 6 to 14 membered, all carbon monocyclic or fused polycyclic (fused polycyclic is a ring sharing adjacent pairs of carbon atoms) group having a conjugated pi-electron system, preferably 6 to 10 membered, such as phenyl and naphthyl. Such aryl rings include those wherein the aryl ring as described above is fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the ring attached to the parent structure is an aryl ring, non-limiting examples of which include:
Figure PCTCN2021000067-APPB-000041
Figure PCTCN2021000067-APPB-000042
aryl groups may be substituted or unsubstituted and when substituted, the substituents may be substituted at any available point of attachment, preferably the substituents are independently optionally one or more substituents selected from halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl.
The term "heteroaryl" refers to a heteroaromatic system containing 1 to 4 (e.g., 1,2,3, and 4) heteroatoms, 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur, and nitrogen. Heteroaryl is preferably 5 to 10 membered (e.g. 5,6, 7,8, 9 or 10 membered), more preferably 5 or 6 membered, e.g. furyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl and the like. The heteroaryl ring includes a heteroaryl fused to an aryl, heterocyclyl or cycloalkyl ring as described above, wherein the ring joined together with the parent structure is a heteroaryl ring, non-limiting examples of which include:
Figure PCTCN2021000067-APPB-000043
heteroaryl groups may be substituted or unsubstituted, and when substituted, substituents may be substituted at any available point of attachment, preferably the substituents are independently optionally one or more substituents selected from halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl, heteroaryl.
The above-mentioned cycloalkyl, heterocyclyl, aryl and heteroaryl groups include residues derived from the parent ring atom by removal of one hydrogen atom, or residues derived from the parent ring atom by removal of two hydrogen atoms from the same or two different ring atoms, i.e., "divalent cycloalkyl", "divalent heterocyclyl", "arylene", "heteroarylene".
The term "cycloalkyloxy" refers to cycloalkyl-O-wherein cycloalkyl is as defined above.
The term "heterocyclyloxy" refers to heterocyclyl-O-, wherein heterocyclyl is as defined above.
The term "aryloxy" refers to aryl-O-, wherein aryl is as defined above.
The term "heteroaryloxy" refers to heteroaryl-O-wherein heteroaryl is as defined above.
The term "alkylthio" refers to alkyl-S-, wherein alkyl is as defined above.
The term "haloalkyl" refers to an alkyl group substituted with one or more halogens, wherein alkyl is as defined above.
The term "haloalkoxy" refers to an alkoxy group substituted with one or more halogens, wherein the alkoxy group is as defined above.
The term "deuterated alkyl" refers to an alkyl group substituted with one or more deuterium atoms, wherein alkyl is as defined above.
The term "hydroxyalkyl" refers to an alkyl group substituted with one or more hydroxyl groups, wherein alkyl is as defined above.
The term "halogen" refers to fluorine, chlorine, bromine or iodine.
The term "hydroxy" refers to-OH.
The term "mercapto" refers to-SH.
The term "amino" refers to-NH 2
The term "cyano" refers to — CN.
The term "nitro" means-NO 2
The term "oxo" or "oxo" means = O.
The term "carbonyl" refers to C = O.
The term "carboxy" refers to-C (O) OH.
The term "carboxylate" refers to-C (O) O (alkyl), -C (O) O (cycloalkyl), (alkyl) C (O) O-or (cycloalkyl) C (O) O-, wherein alkyl and cycloalkyl are as defined above.
The compounds of the present disclosure may also comprise isotopic derivatives thereof. The term "isotopic derivative" refers to a compound that differs in structure only in the presence of one or more isotopically enriched atoms. For example, with the structure of the present disclosure,except for replacing hydrogen by "deuterium" or "tritium", or by 18 F-fluorine labeling: ( 18 Isotope of F) instead of fluorine, or with 11 C-、 13 C-, or 14 C-enriched carbon (C) 11 C-、 13 C-, or 14 C-carbon labeling; 11 C-、 13 c-, or 14 C-isotopes) instead of carbon atoms are within the scope of the present disclosure. Such compounds are useful as analytical tools or probes in, for example, biological assays, or as tracers for in vivo diagnostic imaging of disease, or as tracers for pharmacodynamic, pharmacokinetic or receptor studies.
The disclosure also includes various deuterated forms of the compounds of formula (I). Each available hydrogen atom attached to a carbon atom may be independently replaced with a deuterium atom. The person skilled in the art is able to synthesize the compounds of formula (I) in deuterated form with reference to the relevant literature. Commercially available deuterated starting materials can be used in preparing the deuterated forms of the compounds of formula (I), or they can be synthesized using conventional techniques using deuterated reagents including, but not limited to, deuterated boranes, trideuteroborane tetrahydrofuran solutions, deuterated lithium aluminum hydrides, deuterated iodoethanes, deuterated iodomethanes, and the like. Deuterations can generally retain comparable activity to non-deuterated compounds and can achieve better metabolic stability when deuterated at certain specific sites, thereby achieving certain therapeutic advantages.
"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 the alkyl group may, but need not, be present, and the description includes the case where the heterocyclic group is substituted with an alkyl group and the case where the heterocyclic group is not substituted with an alkyl group.
"substituted" means that one or more hydrogen atoms, preferably 1 to 5, more preferably 1 to 3, of the hydrogen atoms in the group are independently substituted with a corresponding number of substituents. Those skilled in the art are able to ascertain (by experiment or theory) without undue effort, substitutions that are possible or impossible. For example, amino or hydroxyl groups having free hydrogen may be unstable in combination with carbon atoms having unsaturated (e.g., olefinic) bonds.
"pharmaceutical composition" means a mixture containing one or more compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, as well as other components such as physiologically/pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of the active ingredient and exert biological activity.
By "pharmaceutically acceptable salt" is meant a salt of a compound of the present disclosure which is safe and effective for use in the body of a mammal and which has the requisite biological activity. Salts may be prepared separately during the final isolation and purification of the compounds, or by reacting the appropriate group with an appropriate base or acid. Bases commonly used to form pharmaceutically acceptable salts include inorganic bases such as sodium hydroxide and potassium hydroxide, and organic bases such as ammonia. Acids commonly used to form pharmaceutically acceptable salts include inorganic acids as well as organic acids.
The term "therapeutically effective amount" with respect to a drug or pharmacologically active agent refers to a sufficient amount of the drug or agent that is non-toxic and achieves the desired effect. The determination of an effective amount varies from person to person, depending on the age and general condition of the recipient and also on the particular active substance, and an appropriate effective amount in an individual case can be determined by a person skilled in the art according to routine tests.
The term "pharmaceutically acceptable" as used herein means that the compounds, materials, compositions, and/or dosage forms are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio, and effective for the intended use.
As used herein, the singular forms "a", "an" and "the" include plural references and vice versa unless the context clearly dictates otherwise.
When the term "about" is applied to a parameter such as pH, concentration, temperature, etc., it is meant that the parameter may vary by ± 10%, and sometimes more preferably within ± 5%. As will be appreciated by those skilled in the art, when the parameters are not critical, the numbers are generally given for illustrative purposes only and are not limiting.
Synthesis of the Compounds of the disclosure
In order to achieve the purpose of the present disclosure, the following technical solutions are adopted in the present disclosure.
Scheme one
The preparation method of the compound shown in the general formula (I) or the tautomer, the mesomer, the racemate, the enantiomer, the diastereomer or the mixture form or the pharmaceutically acceptable salt form comprises the following steps:
Figure PCTCN2021000067-APPB-000044
reacting general formula (IA) with general formula (IB) or a salt thereof (preferably a hydrochloride salt) under basic conditions, optionally in the presence of a condensing agent, to give a compound of general formula (I);
wherein: r w Is hydroxy or halogen, preferably hydroxy or chlorine;
rings A, Y, Z, T, G 1 、G 2 、R 1 -R 5 、R 8 P, q and n are as defined in formula (I).
Scheme two
The preparation method of the compound shown in the general formula (II) or the tautomer, the mesomer, the racemate, the enantiomer, the diastereomer or the mixture form thereof, or the pharmaceutically acceptable salt form thereof comprises the following steps:
Figure PCTCN2021000067-APPB-000045
reacting formula (IIA) with formula (IB) or a salt thereof (preferably the hydrochloride salt) under basic conditions, optionally in the presence of a condensing agent, to give a compound of formula (II);
wherein: r w Is hydroxy or halogen, preferably hydroxy or chlorine;
rings A, G 1 、G 2 、R 1 -R 5 、R 8 P, q and n are as defined in formula (II).
Scheme three
The preparation method of the compound shown in the general formula (III) or (III-1) or the tautomer, the mesomer, the racemate, the enantiomer, the diastereomer or the mixture form thereof, or the pharmaceutically acceptable salt form thereof comprises the following steps:
Figure PCTCN2021000067-APPB-000046
reacting general formula (IIIA) with general formula (IIIB) or a salt thereof (preferably hydrochloride) under basic conditions, optionally in the presence of a condensing agent, to give a compound of general formula (III); or
Reacting the general formula (IIIA-1) with the general formula (IIIB-1) or a salt thereof (preferably hydrochloride) under alkaline conditions, optionally in the presence of a condensing agent, to obtain a compound of the general formula (III-1);
wherein: r is w Is hydroxy or halogen, preferably hydroxy or chlorine;
Y、Z、T、R 1 -R 2 、R 8 and n is as defined in formula (III) or (III-1).
Scheme four
The preparation method of the compound shown in the general formula (IV) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereoisomer or a mixture form thereof, or a pharmaceutically acceptable salt thereof comprises the following steps:
Figure PCTCN2021000067-APPB-000047
carrying out a reduction reaction on the compound of the general formula (IVA) in the presence of a catalyst to obtain a compound of a general formula (IV);
n is 1,2,3, 4 or 5;
ring A, Y, Z, T, G 1 、G 2 、R 1 -R 5 、R 8 P and q are as defined in formula (IV).
The reagents in schemes one through three that provide basic conditions include organic bases including, but not limited to, triethylamine, N-diisopropylethylamine, N-butyllithium, lithium diisopropylamide, potassium acetate, sodium tert-butoxide, potassium tert-butoxide, or 1, 8-diazabicycloundecene-7-ene, and inorganic bases including, but not limited to, sodium hydride, potassium phosphate, sodium carbonate, sodium acetate, potassium carbonate, or cesium carbonate, sodium hydroxide, lithium hydroxide, and potassium hydroxide; n, N-diisopropylethylamine or 1, 8-diazabicycloundecen-7-ene is preferred.
The condensing agent described in the first to third embodiments includes, but is not limited to, 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride, N, N ' -dicyclohexylcarbodiimide, N, N ' -diisopropylcarbodiimide, O-benzotriazol-N, N, N ', N ' -tetramethyluronium tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy-7-azobenzotriazol, O-benzotriazol-N, N, N ' -tetramethyluronium hexafluorophosphate, 2- (7-azobenzotriazol) -N, N ' -tetramethyluronium hexafluorophosphate, 2- (7-oxybenzotriazolium) -N, N ' -tetramethyluronium hexafluorophosphate, benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate or benzotriazol-1-yl-oxytripyrrolidinylphosphine hexafluorophosphate; benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate is preferred.
Catalysts used in scheme four include, but are not limited to, palladium on carbon, iron powder, raney nickel, zinc powder, tetrakis-triphenylphosphine palladium, palladium dichloride, palladium acetate, 1' -bis (dibenzylphosphine) dichloropentairon palladium, tris (dibenzylideneacetone) dipalladium, and the like, preferably palladium on carbon. The reducing agent used includes, but is not limited to, hydrogen, dilute hydrochloric acid, acetic acid or dilute sulfuric acid, preferably hydrogen.
The above reaction is preferably carried out in a solvent including, but not limited to: acetic acid, methanol, ethanol, acetonitrile, N-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, N-hexane, dimethyl sulfoxide, 1, 4-dioxane, ethylene glycol dimethyl ether, water, N-dimethylacetamide or N, N-dimethylformamide, and a mixture thereof.
Detailed Description
The present disclosure is further described below with reference to examples, but these examples do not limit the scope of the present disclosure.
Examples
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) or/and Mass Spectrometry (MS). NMR shift (. Delta.) at 10 -6 The units in (ppm) are given. NMR was measured by Bruker AVANCE-400 NMR spectrometer using deuterated dimethyl sulfoxide (DMSO-d) 6 ) Deuterated chloroform (CDCl) 3 ) Deuterated methanol (CD) 3 OD), internal standard Tetramethylsilane (TMS).
MS was determined using a FINNIGAN LCQAD (ESI) mass spectrometer (manufacturer: thermo, model: finnigan LCQ advantage MAX).
High Performance Liquid Chromatography (HPLC) analysis was performed using Agilent HPLC 1200DAD, agilent HPLC 1200VWD and Waters HPLC e2695-2489 liquid chromatographs.
Chiral HPLC analytical determination Agilent 1260 DAD HPLC was used.
High performance liquid preparative chromatography using Waters 2767, waters 2767-SQ Detecor2, shimadzu LC-20AP and Gilson-281 preparative chromatographs.
Chiral preparation was performed using Shimadzu LC-20AP preparative chromatograph.
CombiFlash flash rapid preparation instrument uses CombiFlash Rf200 (TELEDYNE ISCO).
The thin layer chromatography silica gel plate adopts HSGF254 of tobacco yellow sea or GF254 of Qingdao, the specification of the silica gel plate used by Thin Layer Chromatography (TLC) is 0.15 mm-0.2 mm, and the specification of the thin layer chromatography separation and purification product is 0.4 mm-0.5 mm.
Silica gel column chromatography generally uses 200-300 mesh silica gel from Futai Huanghai silica gel as a carrier.
Average inhibition rate of kinase and IC 50 The values were determined with a NovoStar microplate reader (BMG, germany).
Known starting materials of the present disclosure may be synthesized using or according to methods known in the art, or may be purchased from companies such as ABCR GmbH & co.kg, acros Organics, aldrich Chemical Company, nephelo Chemical science and technology (Accela ChemBio Inc), dare chemicals, and the like.
In the examples, the reaction can be carried out in an argon atmosphere or a nitrogen atmosphere, unless otherwise specified.
An argon atmosphere or nitrogen atmosphere means that the reaction flask is connected to a balloon of argon or nitrogen with a volume of about 1L.
The hydrogen atmosphere refers to a reaction flask connected with a hydrogen balloon with a volume of about 1L.
The pressure hydrogenation reaction used a Parr 3916EKX type hydrogenator and a Qinglan QL-500 type hydrogen generator or HC2-SS type hydrogenator.
The hydrogenation reaction was usually evacuated and charged with hydrogen and repeated 3 times.
A CEM Discover-S908860 type microwave reactor was used for the microwave reaction.
In the examples, the solution means an aqueous solution without specific indication.
In the examples, the reaction temperature is, unless otherwise specified, room temperature and is 20 ℃ to 30 ℃.
The monitoring of the progress of the reaction in the examples employed Thin Layer Chromatography (TLC), a developing solvent used for the reaction, a system of eluents for column chromatography used for purifying compounds and a developing solvent system for thin layer chromatography including: a: dichloromethane/methanol system, B: n-hexane/ethyl acetate system, C: in the petroleum ether/ethyl acetate system, the volume ratio of the solvent is adjusted according to different polarities of the compounds, and a small amount of basic or acidic reagents such as triethylamine, acetic acid and the like can be added for adjustment.
Example 1
(R) -N- (1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) -2-methyl-7, 8, 10, 11, 13, 14, 16, 17-octahydro- [1,4,7, 10, 13] pentaoxacyclopenta [2,3-g ] quinazolin-4-amine 1
Figure PCTCN2021000067-APPB-000048
First step of
16-Nitro-2, 3,5,6,8,9, 11, 12-octahydrobenzo [ b ] [1,4,7, 10, 13] pentaoxacyclopentadecane-15-carboxylic acid methyl ester 1b
Methyl 2,3,5,6,8,9, 11, 12-octahydrobenzo [ b ] [1,4,7, 10, 13] pentaoxacyclopentadecano-15-carboxylate 1a (1.2 g,3.67mmol, prepared by the well-known method "Journal of the American Chemical Society,2017, 139, (36), 12338-1234" was dissolved in 4mL of acetic acid, cooled to 0 deg.C, 3mL of concentrated nitric acid and 2mL of concentrated sulfuric acid were added, and the reaction was stirred for 3 hours. The reaction was poured into ice water, filtered, and concentrated under reduced pressure to give the title compound 1b (1 g), yield: 73 percent.
MS m/z(ESI):372.1[M+1]。
Second step of
16-amino-2, 3,5,6,8,9, 11, 12-octahydrobenzo [ b ] [1,4,7, 10, 13] pentaoxacyclopentadecane-15-carboxylic acid methyl ester 1c
Compound 1b (1g, 2.69mmol) was dissolved in 50mL of methanol, 10% wet palladium on carbon (350 mg) was added, and after the addition, hydrogen was substituted three times, stirred for 14 hours, filtered under reduced pressure, and the filtrate was concentrated under reduced pressure to give crude title compound 1c (919 mg), which was used directly in the next reaction.
MS m/z(ESI):342.1[M+1]。
The third step
2-methyl-7, 8, 10, 11, 13, 14, 16, 17-octahydro- [1,4,7, 10, 13] pentaoxacyclopenta [2,3-g ] quinazolin-4-ol 1d
Compound 1c (500mg, 1.46mmol) was dissolved in 2mL of acetonitrile, 0.5mL of methanesulfonic acid was added, and the reaction was stirred at 100 ℃ for 14 hours. The reaction solution was concentrated under reduced pressure, and the resulting residue was purified by thin layer chromatography with developer system a to give the title compound 1d (130 mg), yield: 25 percent.
MS m/z(ESI):351.1[M+1]。
The fourth step
4-chloro-2-methyl-7, 8, 10, 11, 13, 14, 16, 17-octahydro- [1,4,7, 10, 13] pentaoxacyclopenta [2,3-g ] quinazoline 1e
The crude compound 1d (200mg, 0.57mmol) was dissolved in 3mL of phosphorus oxychloride and the reaction was stirred at 100 ℃ for 3 hours after the addition. The reaction solution was concentrated under reduced pressure, and the resulting residue was purified by thin layer chromatography with developer system a to give the title compound 1e (130 mg), yield: 61 percent.
MS m/z(ESI):369.1[M+1]。
The fifth step
(R) -N- (1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) -2-methyl-7, 8, 10, 11, 13, 14, 16, 17-octahydro- [1,4,7, 10, 13] pentaoxacyclopenta [2,3-g ] quinazolin-4-amine 1
Compound 1e (60mg, 0.16mmol), compound (R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethylamine hydrochloride 1f (55mg, 0.16mmol, prepared by the method disclosed in example B-5 on page 141 of the specification of patent application "EP 2018086197") was dissolved in 2mL of DMF, N-diisopropylethylamine (84mg, 0.65mmol) was added, and after the addition, the reaction was stirred at 90 ℃ for 14 hours. The reaction solution was concentrated under reduced pressure, and the obtained title compound 1 (1 mg) was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm: mobile phase: water (10 mmol/L ammonium hydrogencarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min), yield: 1 percent.
MS m/z(ESI):522.2[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.73(s,1H),7.61(t,1H),7.48(t,1H),7.24(t,1H),7.16-6.89(m,2H),5.87(q,1H),4.26(ddd,4H),3.96(ddd,4H),3.81-3.71(m,8H),2.42(s,3H),1.70(d,3H)。
Example 2
(R) -N- (1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) -2-methyl-7, 8, 10, 11, 13, 14-hexahydro- [1,4,7, 10] tetraoxacyclododeca [2,3-g ] quinazolin-4-amine 2
Figure PCTCN2021000067-APPB-000049
Figure PCTCN2021000067-APPB-000050
First step of
2-Methylquinazoline-4, 6, 7-trisphenol hydrobromide 2b
6, 7-dimethoxy-2-methylquinazoline-4-ol 2a (700mg, 3.18mmol, jiangsu Aikang biomedical research and development Co., ltd.) was dissolved in 10mL of an aqueous hydrobromic acid solution (40%) and reacted at 120 ℃ with stirring for 14 hours. Cooled to room temperature, aqueous ammonia was added dropwise to the reaction mixture to pH7, filtered, and the filtrate was concentrated under reduced pressure to give the title crude compound 2b (1.1 g).
MS m/z(ESI):193.2[M+1]。
Second step of
2-methyl-7, 8, 10, 11, 13, 14-hexahydro- [1,4,7, 10] tetraoxacyclododeca [2,3-g ] quinazolin-4-ol 2d
Compound 2b (400mg, 1.13mmol), compound triethylene glycol di (p-toluenesulfonate) 2c (518mg, 1.13mmol, shanghai bei pharma science co., ltd.), anhydrous potassium carbonate (936mg, 6.77mmol) were dissolved in 4ml of n, n-dimethylformamide, stirred at 90 ℃ for 14 hours, the reaction solution was concentrated under reduced pressure, and the resulting residue was purified by thin layer chromatography using developer system a to give the title compound 2d (30 mg), yield: 8 percent.
MS m/z(ESI):307.1[M+1]。
The third step
(R) -N- (1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) -2-methyl-7, 8, 10, 11, 13, 14-hexahydro- [1,4,7, 10] tetraoxacyclododeca [2,3-g ] quinazolin-4-amine 2
Compound 2d (30mg, 0.09mmol), compound 1f (26mg, 0.11mmol), benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate (43mg, 0.09mmol), 1, 8-diazabicycloundec-7-ene (14mg, 0.09mmol) were dissolved in 2mL of N, N-dimethylformamide, stirred at room temperature for 10 minutes, and then stirred at 60 ℃ for 2 hours. The reaction solution was concentrated under reduced pressure, and the obtained title compound 2 (2 mg) was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm; mobile phase: water (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min), yield: 4 percent.
MS m/z(ESI):478.2[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.92(s,1H),7.58(t,1H),7.46(t,1H),7.21(t,1H),7.16-6.86(m,2H),5.82(q,1H),4.28(dt,4H),3.87(dt,4H),3.77(d,4H),2.39(s, 3H),1.66(d,3H)。
Example 3
(R) -N- (1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) -2-methyl-7, 8, 10, 11, 13, 14-hexahydro- [1,4,7, 10] tetraoxacyclododeca [2,3-g ] quinazolin-4-amine 3
Figure PCTCN2021000067-APPB-000051
First step of
(R) -6, 7-dimethoxy-2-methyl-N- (1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) quinazolin-4-amine 3c
The compound 4-chloro-6,7-dimethoxy-2-methyl quinazoline 3a (50mg, 0.21mmol, shanghaibei pharmaceutical science Co., ltd.) and the compound (R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethylamine hydrochloride 3B (68mg, 0.25mmol, prepared by the method disclosed in example B-6a on page 89 of the specification in patent application "CN 110167928A") were dissolved in 2mL of DMF, N-diisopropylethylamine (63mg, 0.62mmol) was added thereto, and the reaction was stirred at 90 ℃ for 14 hours after completion of the addition. The reaction solution was concentrated under reduced pressure, and the resulting residue was purified by thin layer chromatography with developer system a to give the title compound 3c (50 mg), yield: 50 percent.
MS m/z(ESI):437.3[M+1]。
Second step of
(R) -2-methyl-4- ((1- (3-nitro-5-trifluoromethyl) phenyl) ethyl) amino) quinazoline-6, 7-diol 3d
Compound 3c (300mg, 0.68mmol) was dissolved in 6mL dichloromethane, boron tribromide (5 mL,17% in dichloromethane, echiei (shanghai) chemical development limited) was added under ice-bath, the reaction was stirred for 14 hours, quenched with methanol, washed with water (3 mL × 2), concentrated under reduced pressure by organic phase, and purified by column chromatography with developer system a to give the title compound 3d (130 mg), yield: 46 percent.
MS m/z(ESI):409.1[M+1]。
The third step
(R) -2-methyl-N- (1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) -7,8, 10, 11, 13, 14-hexahydro- [1,4,7, 10] tetraoxacyclododeca [2,3-g ] quinazolin-4-amine 3e
Compound 3d (20mg, 0.05mmol), compound 2c (24mg, 0.05mmol, from Shanghai Biao pharmaceutical science Co., ltd.), and anhydrous potassium carbonate (20mg, 0.14mmol) were dissolved in 2mL of N, N-dimethylformamide, and reacted for 14 hours with stirring. The reaction solution was concentrated under reduced pressure, and the resulting residue was purified by thin layer chromatography with developer system a to give the title compound 3e (10 mg), yield: 39 percent.
The fourth step
(R) -N- (1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) -2-methyl-7, 8, 10, 11, 13, 14-hexa-oxo- [1,4,7, 10] tetraoxacyclododeca [2,3-g ] quinazolin-4-amine 3
Crude compound 3e (10mg, 0.02mmol) was dissolved in 5mL of methanol, 10% wet palladium on charcoal (10 mg) was added, and after the addition, hydrogen substitution was performed three times, stirring was performed for 14 hours, filtration was performed under reduced pressure, the filtrate was concentrated under reduced pressure, and the residue was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm; mobile phase: water (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min) to give title compound 3 (5 mg), yield: and 53 percent.
MS m/z(ESI):493.2[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.89(d,1H),7.10(s,1H),6.97(d,2H),6.80(s,1H),5.60(q,1H),4.27(dt,4H),4.00-3.81(m,4H),3.77(s,4H),2.45(d,3H),1.62(d,3H)。
Example 4
(R) -N- (1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) -2-methyl-7, 8, 10, 11, 13, 14, 16, 17-octahydro- [1,4,7, 10, 13] pentaoxacyclopenta [2,3-g ] quinazolin-4-amine 4
Figure PCTCN2021000067-APPB-000052
First step of
(R) -2-methyl-N- (1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) -7,8, 10, 11, 13, 14, 16, 17-octahydro- [1,4,7, 10, 13] pentaoxacyclopenta [2,3-g ] quinazolin-4-amine 4b
Compound 3d (30mg, 0.07mmol), compound tetraethyleneglycol di-p-toluenesulfonate 4a (55 mg,0.11mmol, prepared by a known method "Bioorganic and Medicinal Chemistry,2007, 15 (14), 4841-4856"), and anhydrous potassium carbonate (50mg, 0.36mmol) were dissolved in 4mL of N, N-dimethylformamide and reacted with stirring for 14 hours. The reaction mixture was concentrated under reduced pressure, 2mL of water was added, extraction was performed with ethyl acetate (3 mL. Times.3), the organic phase was dried over anhydrous sodium sulfate, filtration was performed, and the filtrate was concentrated under reduced pressure to give the crude title compound 4b (40 mg) which was used directly in the next reaction.
Second step of
(R) -N- (1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) -2-methyl-7, 8, 10, 11, 13, 14, 16, 17-octahydro- [1,4,7, 10, 13] pentaoxacyclopenta [2,3-g ] quinazolin-4-amine 4
The crude compound 4b (40mg, 0.07mmol) was dissolved in 10mL of methanol, 10% wet palladium on charcoal (40 mg) was added, after which hydrogen was substituted three times, stirred for 14 hours, filtered under reduced pressure, the filtrate was concentrated under reduced pressure, and the residue was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm; mobile phase: water (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min) to give the title compound 4 (12 mg), yield: 31%.
MS m/z(ESI):537.2[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.64(d,1H),6.98(d,3H),6.80(d,1H),5.61(q,1H),4.22(dt,4H),3.98-3.88(m,4H),3.74(dd,8H),2.45(d,3H),1.63(dd,3H)。
Example 5
(R) -2, 2-difluoro-2- (2-fluoro-3- (1- ((2-methyl-7, 8, 10, 11, 13, 14-hexahydro- [1,4,7, 10] tetraoxacyclododeca [2,3-g ] quinazolin-4-yl) amino) ethyl) phenyl) ethan-1-ol 5
Figure PCTCN2021000067-APPB-000053
First step of
(R) -2, 2-difluoro-2- (2-fluoro-3- (1- ((2-methyl-7, 8, 10, 11, 13, 14-hexahydro- [1,4,7, 10] tetraoxacyclododeca [2,3-g ] quinazolin-4-yl) amino) ethyl) phenyl) ethanol 5
Compound 2d (50mg, 0.11mmol), compound (R) -2- (3- (1-ethylamine) -2-fluorophenyl) -2, 2-difluoroethanolate hydrochloride 5a (42mg, 0.14mmol, prepared using the method disclosed in example B-5 on page 105 of the specification in the patent application "US 2019194192"), benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate (93mg, 0.21mmol), 1, 8-diazabicycloundecan-7-ene (37mg, 0.24mmol) were dissolved in 2mL of N, N-dimethylformamide, stirred at room temperature for 10 minutes, and then stirred at 60 ℃ for 2 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm; mobile phase: water (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min) to give title compound 5 (7 mg), yield: 8 percent.
MS m/z(ESI):508.3[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.95(s,1H),7.63-7.54(m,1H),7.49-7.40(m,1H),7.20(t,1H),7.09(s,1H),5.86(q,1H),4.28(dt,4H),4.03(td,2H),3.91(t,2H),3.87-3.82(m,2H),3.81-3.73(m,4H),2.43(s,3H),1.67(d,3H)。
Example 6
(R) -N- (1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) -2-methyl-7, 8, 10, 11, 12, 13, 15, 16-octahydro- [1,4,7, 10] tetraoxacyclotetradeca [5,6-g ] quinazolin-4-amine 6
Figure PCTCN2021000067-APPB-000054
First step of
(butane-1, 4-diylbis (oxy)) bis (ethane-2, 1-diyl) bis (4-methylbenzenesulfonic acid) 6b
2,2' - (butane-1, 4-diylbis (oxy)) diethanol 6a (2.5 g,14.02mmol, prepared by the well-known method "Angewandte Chemie-International Edition,2015, 54, (12), 3763-3767 Angewaw.chem., 2015, 127 (12), 3834-3838") was dissolved in 50mL of dichloromethane, and potassium hydroxide (8g, 142.58mmol) was added in an ice bath and stirred for 14 hours. Filtration, addition of 20mL of water to the filtrate, concentration of the organic phase under reduced pressure, and purification of the resulting residue by thin layer chromatography using developer system C gave the title compound 6b (2 g), yield: 29 percent.
MS m/z(ESI):487.0[M+1]。
Second step of
2-methyl-7, 8, 10, 11, 12, 13, 15, 16-octahydro- [1,4,7, 10] tetraoxacyclotetradeco [5,6-g ] quinazolin-4-ol 6c
Compound 2b (150mg, 0.78mmol), compound 6b (455mg, 0.94mmol), anhydrous potassium carbonate (431mg, 3.12mmol) were dissolved in 6ml of n, n-dimethylformamide, stirred at 90 ℃ for 2 hours, the reaction solution was concentrated under reduced pressure, and the resulting residue was purified by thin layer chromatography using developer system a to give the title compound 6c (80 mg), yield: 30 percent.
MS m/z(ESI):335.0[M+1]。
The third step
(R) -N- (1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) -2-methyl-7, 8, 10, 11, 12, 13, 15, 16-octahydro- [1,4,7, 10] tetraoxacyclotetradeca [5,6-g ] quinazolin-4-amine 6
Compound 6c (50mg, 0.15mmol), compound (R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethylamine hydrochloride 1f (40mg, 0.18mmol), benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate (85mg, 0.19mmol), and 1, 8-diazabicycloundec-7-ene (45mg, 0.29mmol) were dissolved in 2mL of N, N-dimethylformamide, stirred at room temperature for 10 minutes, and then stirred at 60 ℃ for 14 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm; mobile phase: water (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min) to give title compound 6 (15 mg), yield: 20 percent.
MS m/z(ESI):506.1[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.70(s,1H),7.59(t,1H),7.46(t,1H),7.21(t,1H),7.16-6.86(m,2H),5.83(q,1H),4.34-4.17(m,4H),3.94-3.83(m,4H),3.78(dt,4H),2.38(s,3H),1.72(q,4H),1.68(d,3H)。
Example 7
(R) -2, 2-difluoro-2- (2-fluoro-3- (1- ((2-methyl-7, 8, 10, 11, 13, 14, 16, 17-octahydro- [1,4,7, 10, 13] pentaoxacyclopenta [2,3-g ] quinazolin-4-yl) amino) ethyl) phenyl) ethan-1-ol 7
Figure PCTCN2021000067-APPB-000055
First step of
(R) -2, 2-difluoro-2- (2-fluoro-3- (1- ((2-methyl-7, 8, 10, 11, 13, 14, 16, 17-octahydro- [1,4,7, 10, 13] pentaoxacyclopenta [2,3-g ] quinazolin-4-yl) amino) ethyl) phenyl) ethan-1-ol 7
Compound 1d (80mg, 0.29mmol), compound 5a (65mg, 0.25mmol), benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate (161mg, 0.36mmol), 1, 8-diazabicycloundecen-7-ene (69mg, 0.45mmol) were dissolved in 2mL of N, N-dimethylformamide, stirred at room temperature for 10 minutes, and then stirred at 60 ℃ for reaction for 14 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm: mobile phase: water (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min) to give title compound 7 (17 mg), yield: 13 percent.
MS m/z(ESI):552.1[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.67(s,1H),7.57(t,1H),7.49-7.39(m,1H),7.18(t,1H),6.98(s,1H),5.84(q,1H),4.23(ddd,4H),4.03(td,2H),3.98-3.87(m,4H),3.75(dq,8H),2.39(s,3H),1.67(d,3H)。
Example 8
(R) -N- (1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) -2-methyl-7, 8, 10, 11-tetrahydro- [1,4,7] trioxabicyclo [2,3-g ] quinazolin-4-amine 8
Figure PCTCN2021000067-APPB-000056
First step of
(R) -2-methyl-N- (1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) -7,8, 10, 11-tetrahydro- [1,4,7] trioxabicyclo [2,3-g ] quinazolin-4-amine 8b
Compound 3d (65mg, 0.16mmol), compound diethylene glycol bis-p-benzenesulfonate 8a (68mg, 0.16mmol, han hong Kogyo technology and technology Co., ltd., shanghai), and anhydrous potassium carbonate (110mg, 0.8mmol) were dissolved in 4mL of N, N-dimethylformamide and reacted for 14 hours with stirring. The reaction mixture was concentrated under reduced pressure, 2mL of water was added, extracted with ethyl acetate (3 mL × 3), the organic phase was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the crude title compound 8b (70 mg), yield: 91 percent.
MS m/z(ESI):479.1[M+1]。
Second step of
(R) -N- (1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) -2-methyl-7, 8, 10, 11-tetrahydro- [1,4,7] trioxabicyclo [2,3-g ] quinazolin-4-amine 8
The crude compound 8b (70mg, 0.15mmol) was dissolved in 10mL of methanol, 10% wet palladium on charcoal (40 mg) was added, after which hydrogen was substituted three times, stirred for 14 hours, filtered under reduced pressure, the filtrate was concentrated under reduced pressure, and the residue was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm; mobile phase: water (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min) to give the title compound 8 (5 mg), yield: 7.6 percent.
MS m/z(ESI):449.1[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.93(d,1H),7.14(d,1H),6.99(s,1H),6.95(s,1H),6.80(s,1H),5.58(d,1H),4.70-4.49(m,2H),4.36(t,2H),3.93(d,4H),2.43(d,3H),1.61(dd,3H)。
Example 9
((R) -N- (1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) -2-methyl-7, 8, 10, 11-tetrahydro- [1,4,7] trioxabicyclo [2,3-g ] quinazolin-4-amine 9
Figure PCTCN2021000067-APPB-000057
First step of
2-methyl-7, 8, 10, 11-tetrahydro- [1,4,7] trioxabicyclo [2,3-g ] quinazolin-4-ol 9a
The compound 2b (100mg, 0.52mmol), the compound diethylene glycol bis-p-benzenesulfonate 8a (259mg, 0.62mmol, han hong Kong Biotechnology medicine, inc., shanghai), and anhydrous potassium carbonate (216mg, 1.6 mmol) were dissolved in 10mL NN-dimethylformamide and stirred at 80 deg.C for 2 hours. The reaction solution was concentrated under reduced pressure, 2mL of water was added, extracted with ethyl acetate (3 mL × 3), concentrated under reduced pressure of the organic phase, and the resulting residue was purified by thin layer chromatography with developer system C to give the title compound 9a (40 mg), yield: 29.3 percent.
MS m/z(ESI):263.1[M+1]。
Second step of
((R) -N- (1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) -2-methyl-7, 8, 10, 11-tetrahydro- [1,4,7] trioxabicyclo [2,3-g ] quinazolin-4-amine 9
Compound 9a (40mg, 0.15mmol), compound 1f (40mg, 0.18mmol), benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate (81mg, 0.18mmol), and 1, 8-diazabicycloundecen-7-ene (35mg, 0.23mmol) were dissolved in 2mL of N, N-dimethylformamide, stirred at room temperature for 10 minutes, and then stirred at 60 ℃ for 14 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm; mobile phase: water (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min) to give title compound 9 (5 mg), yield: 7.6 percent.
MS m/z(ESI):434.1[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.96(s,1H),7.57(t,1H),7.45(t,1H),7.21(t,1H),7,7.19-6.89(m,2H),5.83-5.79(m,1H),4.59-4.57(m,2H),4.38-4.37(m,2H),3.93(d,4H),2.36(d,3H),1.61(dd,3H)。
Example 10
(R) -2, 2-difluoro-2- (2-fluoro-3- (1- ((2-methyl-7, 8, 10, 11-tetrahydro- [1,4,7] trioxabicyclo [2,3-g ] quinazolin-4-yl) amino) ethyl) phenyl) ethan-1-ol 10
Figure PCTCN2021000067-APPB-000058
First step of
(R) -2, 2-difluoro-2- (2-fluoro-3- (1- ((2-methyl-7, 8, 10, 11-tetrahydro- [1,4,7] trioxabicyclo [2,3-g ] quinazolin-4-yl) amino) ethyl) phenyl) ethan-1-ol 10
Compound 9a (30mg, 0.11mmol), compound 5a (30mg, 0.12mmol), benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate (61mg, 0.14mmol), 1, 8-diazabicycloundecen-7-ene (58mg, 0.23mmol) were dissolved in 2mL of N, N-dimethylformamide, stirred at room temperature for 10 minutes, and then stirred at 60 ℃ for 2 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm; mobile phase: water (10 mmol/L ammonium hydrogencarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min) to give the title compound 10 (11 mg), yield: 20.7 percent.
MS m/z(ESI):464.0[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.98(s,1H),7.59-7.56(m,1H),7.48-7.45(m, 1H),7.22-7.19(m,1H),7.16(s,1H),5.87(q,1H),4.88-4.60(m,2H),4.41-4.39(m,2H),4.10-3.96(m,3H),3.94(s,3H),2.41(s,3H),1.68(d,3H)。
Example 11
(R) -N- (1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) -2-methyl-7, 8-dihydro- [1,4] dioxa-do [2,3-g ] quinazolin-4-amine 11
Figure PCTCN2021000067-APPB-000059
First step of
(R) -2-methyl-N- (1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) -7, 8-dihydro- [1,4] dioxa-do [2,3-g ] quinazolin-4-amine 11b
Compound 3d (350mg, 0.86mmol), compound 1, 2-dibromoethane 11a (68mg, 0.16mmol, han hong Kong Biotechnology medicine, inc., shanghai), and cesium carbonate (838mg, 2.57mmol) were dissolved in 10mL of N, N-dimethylformamide and reacted at 80 ℃ with stirring for 16 hours. The reaction mixture was concentrated under reduced pressure, 2mL of water was added, extracted with ethyl acetate (3 mL. Times.3), the organic phase was concentrated under reduced pressure, and the residue was purified by preparative thin layer chromatography using eluent system A to give the title compound 11b (60 mg) in 16% yield.
MS m/z(ESI):435.1[M+1]。
Second step of
(R) -N- (1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) -2-methyl-7, 8-dihydro- [1,4] dioxa-do [2,3-g ] quinazolin-4-amine 11
Compound 11b (60mg, 0.14mmol) was dissolved in 5mL of methanol, 10% palladium on charcoal (10 mg) was added, after addition, hydrogen was replaced three times, stirring was carried out for 16 hours, filtration was carried out under reduced pressure, the filtrate was concentrated under reduced pressure, and the residue was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm: mobile phase: water (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min) to give title compound 11 (55 mg), yield: and 90 percent.
MS m/z(ESI):405.1[M+1]。
1 H NMR(400MHz,DMSO-d 6 ):δ7.90(d,1H),7.81(s,1H),6.97(s,1H),6.87(s,1H),6.83(s,1H),6.67(s,1H),5.53(s,3H),4.34(s,4H),2.33(s,3H),1.51(d,3H)。
Example 12
(R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2-methyl-10, 11-dihydro-7H- [1,4,7] dioxaazanonacyclo [2,3-g ] quinazolin-8 (9H) -one 12
Figure PCTCN2021000067-APPB-000060
First step of
(R) - (2- ((6-hydroxy-2-methyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) quinazolin-7-yl) oxy) ethyl) carbamic acid tert-butyl ester 12a
Compound 3d (308mg, 754.3. Mu. Mol), (2-bromoethyl) carbamic acid tert-butyl ester (1699 mg, 754.3. Mu. Mol, shanghai Biao) was dissolved in 2mL of N, N-dimethylformamide, anhydrous potassium carbonate (312.2mg, 2.26mmol) was added, and the reaction was stirred at 50 ℃ for 14 hours with a tube sealed. The reaction solution was concentrated under reduced pressure, and the residue was purified by column chromatography with eluent system a to give the title compound 12a (80 mg), yield: 19.2 percent.
MS m/z(ESI):552.3[M+1]。
Second step of
(R) -tert-butyl 2- ((7- (2- ((tert-butoxycarbonyl) amino) ethoxy) -2-methyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) quinazolin-6-yl) oxy) acetate 12b
Compound 12a (200mg, 326.6. Mu. Mol) and t-butyl bromoacetate (63.7 mg, 326.6. Mu. Mol, shanghaitan) were dissolved in 5mL of N, N-dimethylformamide, anhydrous potassium carbonate (180mg, 1.3 mmol) was added, and the reaction was stirred at 35 ℃ for 1.5 hours. The reaction solution was concentrated under reduced pressure, and the residue was purified by column chromatography with eluent system a to give the title compound 12b (200 mg), yield: 91.9 percent.
MS m/z(ESI):666.2[M+1]。
The third step
(R) -2- ((7- (2-aminoethoxy) -2-methyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) quinazolin-6-yl) oxy) acetic acid trifluoroacetate 12c
Compound 12b (20mg, 30. Mu. Mol) was dissolved in 2mL of methylene chloride, and 1mL of trifluoroacetic acid was added to stir the mixture for 1 hour. The reaction solution was concentrated under reduced pressure to give crude product 12c (20 mg), which was used in the next reaction without purification.
MS m/z(ESI):510.1[M+1]。
The fourth step
(R) -2-methyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -10, 11-dihydro-7H- [1,4,7] dioxaazanonacyclo [2,3-g ] quinazolin-8 (9H) -one 12d
Compound 12c (20mg, 39.2. Mu. Mol) was dissolved in 2mL of N, N-dimethylformamide, and N, N-diisopropylethylamine (15.2mg, 117.7. Mu. Mol) and 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (44.7mg, 117.7. Mu. Mol) were added thereto under ice-cooling, and the mixture was allowed to naturally warm to room temperature and reacted for 1 hour. After the reaction solution was concentrated under reduced pressure, the residue was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm,5 μm; mobile phase: water (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min) to give the title compound 12d (19 mg), yield: 98 percent.
MS m/z(ESI):492.2[M+1]。
The fifth step
(R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2-methyl-10, 11-dihydro-7H- [1,4,7] dioxaazanonacyclo [2,3-g ] quinazolin-8 (9H) -one 12
Compound 12d (19mg, 38.6.6. Mu. Mol) was dissolved in 5mL of methanol, 10% wet palladium on charcoal (40 mg) was added, and after the addition, hydrogen was substituted three times, stirring was carried out for 14 hours, filtration was carried out under reduced pressure, the filtrate was concentrated under reduced pressure, and the residue was purified by high performance liquid preparative chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm, 5. Mu.m: mobile phase: water (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -50%, flow rate: 30 mL/min) to give title compound 12 (2 mg), yield: 1.1 percent.
MS m/z(ESI):462.1[M+1]。
1 H NMR(500MHz,DMSO-d 6 ):δ8.18(d,1H),7.68(q,1H),7.28(d,1H),6.89(s,1H),6.84(s,1H),6.70(s,1H),5.54(s,1H),5.51(d,1H),4.91-4.75(m,2H),4.22-4.09(m,2H),2.37(s,3H),1.53(d,3H)。
Example 13
(R) -N- (1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) -2' -methyl-7 ',8',10',11' -tetrahydrospiro [ [1,3] dioxolane-2, 9' - [1,4] dioxanonacyclo [2,3-g ] quinazolin-4 ' -amine 13
Figure PCTCN2021000067-APPB-000061
Using the synthetic route in example 6, the first step starting material compound 6a was replaced with the compound 1, 3-dioxolane-2, 2-diethanol to give the title compound 13 (3.8 mg), yield: 1.4 percent.
MS m/z(ESI):490.2[M+1]。
1 H NMR(500MHz,CDCl 3 ):δ7.50(dq,2H),7.37(d,2H),7.20(t,1H),6.92(t,1H),5.85-5.71(m,1H),5.34(s,1H),4.40(dtd,4H),3.93(s,4H),2.53(s,3H),2.23(d,2H),2.09(t,2H),1.69(d,3H)。
Example 14
(R) -2-methyl-N- (1- (2-methyl-3- (trifluoromethyl) phenyl) ethyl) -7,8, 10, 11, 13, 14-hexahydro- [1,4,7, 10] tetraoxacyclododeca [2,3-g ] quinazolin-4-amine 14
Figure PCTCN2021000067-APPB-000062
Using the synthetic route in example 2, the starting compound 1f was replaced with the compound (R) -1- (2-methyl-3- (trifluoromethyl) phenyl) ethan-1-amine hydrochloride (prepared using the method disclosed in example B-6i on page 106 of the specification in the patent application "WO 2018/115380") in the third step to give the title compound 14 (10 mg), yield: 20.1 percent.
MS m/z(ESI):492.2[M+1]。
1 H NMR(500MHz,CDCl 3 ):δ7.61-7.57(m,2H),7.27-7.19(m,2H),5.85-5.83(m,1H),5.47-5.46(m,1H),4.29-4.25(m,4H),3.99-3.97(m,2H),3.88-3.85(m,2H),3.85-3.82(m,4H),2.63(s,3H),2.53(s,3H),1.68-1.64(d,3H)。
Example 15
(R) -N- (1- (3- (difluoromethyl) -2-methylphenyl) ethyl) -2-methyl-7, 8, 10, 11, 13, 14-hexahydro- [1,4,7, 10] tetraoxacyclododeca [2,3-g ] quinazolin-4-amine 15
Figure PCTCN2021000067-APPB-000063
Using the synthetic route in example 2, the starting compound 1f was replaced with the compound (R) -1- (3- (difluoromethyl) -2-methylphenyl) ethan-1-amine hydrochloride (prepared using the method disclosed in example B-6i on page 106 of the specification in the patent application "WO 2018/115380") in the third step to give the title compound 15 (15 mg), yield: 48.5 percent.
MS m/z(ESI):474.2[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.92(s,1H),7.62-7.60(m,1H),7.40-7.39(m,1H), 7.28-7.25(m,1H),7.10(s,1H),7.08-6.86(t,1H),5.87-5.83(m,1H),4.31-4.26(m,4H),3.93-3.85(m,4H),3.80-3.77(m,4H),2.60(s,3H),2.43(s,3H),1.63-1.61(d,3H)。
Example 16
(R) -N- (1- (3, 3-difluoro-2, 3-dihydrobenzofuran-7-yl) ethyl) -2-methyl-7, 8, 10, 11, 13, 14-hexahydro- [1,4,7, 10] tetraoxacyclododeca [2,3-g ] quinazolin-4-amine 16
Figure PCTCN2021000067-APPB-000064
Using the synthetic route in example 2, the third step starting compound 1f was replaced with the compound (R) -I- (3,3-difluoro-2,3-dihydrobenzofuran-7-yl) ethane-1-amine hydrochloride (prepared using the method disclosed in example B-5 on page 105 of the specification in patent application "US 2019194192") to give the title compound 16 (80 mg), yield: 45.6 percent.
MS m/z(ESI):488.2[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.94(s,1H),7.52(d,1H),7.43(dq,1H),7.12(s,1H),7.06(t,1H),5.79(q,1H),4.74(t,2H),4.34-4.30(m,2H),4.30-4.26(m,2H),3.96-3.90(m,2H),3.89-3.84(m,2H),3.82-3.76(m,4H),2.43(s,3H),1.68(d,3H)。
Biological evaluation
Test example 1 ability of Compounds of the present disclosure to inhibit the interaction between each subtype of KRAS protein, G12D, G12C or G12V, and SOS1 protein
The following methods were used to determine the ability of the compounds of the present disclosure to inhibit the interaction between each of the subtypes G12D, G12C or G12V of KRAS protein and SOS1 protein.
The experimental method is as follows:
1. experimental materials and instruments
1. Biotin labeling kit (Dojindo, LK 03)
2、GDP(SIGMA,G7127)
3. AlphaLISA Glutathione Acceptor Beads (AlphaLISA Glutathione acceptors) (PerkinElmer, AL 109C)
4. AlphaScreen Streptavidin Donor beads (AlphaScreen Streptavidin Donor beads) (PerkinElmer, 6760002S)
5. 1/2 AreaPlate-96 well plate (PerkinElmer, 6002290)
6. GST-TEV-SOS1 (564-1049) (Viya Biotechnology, SOS 1-191010)
7. KRasG12D, krasG12C, krasG12V (provided by Shanghai Pan Biotech Co., ltd.)
8. Phosphate Buffered Saline (PBS) PH7.4 (Shanghai Yangbei Biotech, B320)
9. Multifunctional enzyme mark instrument (PerkinElmer, envision)
2. Experimental procedure
Preparation of the experiment:
1. the experiment buffer was prepared prior to the start of the experiment: 1x PBS +0.1% Tween 20 after being subjected to BSA + 0.05%.
2. The KRAS G12D, KRAS-G12C and KRAS-G12V proteins are labeled with biotin using a biotin labeling kit.
The experimental steps are as follows:
1. KRAS G12C or KRAS G12V or KRAS G12D protein is mixed with SOS1 protein and GDP respectively and incubated for later use.
2. AlphaLISA glutathione acceptor beads and AlphaScreen streptavidin donor beads were mixed 1: 1 to 40ug/mL before use for future use.
Compounds were formulated in assay buffer at an initial concentration of 40 μ M, 5-fold gradient dilutions, 10 gradient series concentration points.
mu.L of a mixture of KRAS G12C or KRAS G12V or KRAS G12D protein with SOS1 and GDP and 5. Mu.L of the diluted compound were added to each well of a 1/2 area plate-96 well plate, and incubated at room temperature for 30 minutes in the absence of light.
3. Then 5. Mu.L of the bead mixture was added per well and incubated for 60 minutes at room temperature in the absence of light.
4. The fluorescence values were read on a multifunctional microplate reader.
5. The IC of the compound was calculated using Graphpad Prism 50 The value is obtained.
3. Experimental data
The ability of the disclosed compounds to inhibit the interaction between the various subtypes G12D, G12C or G12V of KRAS protein and SOS1 protein, measured as IC 50 The values are shown in Table 1.
TABLE 1 IC inhibiting the ability of compounds of the present disclosure to interact with SOS1 protein and each of the subtypes G12D, G12C or G12V of KRAS protein 50 Value of
Figure PCTCN2021000067-APPB-000065
Figure PCTCN2021000067-APPB-000066
And (4) conclusion: the disclosed compound can well inhibit the interaction between various subtypes G12D, G12C or G12V of KRAS protein and SOS1 protein.

Claims (22)

  1. A compound of formula (I) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2021000067-APPB-100001
    wherein:
    y is- (OCH) 2 CH 2 ) y -, where O side is attached to Z and C side is attached to O;
    z is- (CR) m R n ) z -or- (CH) 2 ) v C(O)NR 0 (CH 2 ) w -:
    T is- (OCH) 2 CH 2 ) t -, where O side is attached to Z and C side is attached to O;
    R m and R n The same or different, each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, and hydroxyalkyl; or
    R m 、R n Together with the carbon atom to which they are attached form a cycloalkyl or heterocyclyl group, which may optionally be substituted by one or more R 5 Substitution;
    ring a is aryl or heteroaryl;
    G 1 and G 2 Selected from nitrogen or carbon atoms;
    R 0 selected from the group consisting of hydrogen, alkyl, haloalkyl or hydroxyalkyl, wherein said alkyl, haloalkyl or hydroxyalkyl is optionally substituted with one or more substituents selected from the group consisting of halogen, hydroxy, alkoxy, haloalkoxy, amino, nitro and cyano;
    R 1 selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxyalkyl, cyano, and cycloalkyl;
    R 2 selected from oxygen, halogenAn alkyl, a haloalkyl, a hydroxyalkyl, a hydroxyl, a cyano, a cycloalkyl and a heterocyclyl, wherein the alkyl, cycloalkyl and heterocyclyl are optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, haloalkyl, hydroxyl, hydroxyalkyl, alkoxy, haloalkoxy, amino, nitro and cyano;
    R 3 selected from the group consisting of hydrogen, alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, haloalkyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
    R 4 are the same or different and are each independently selected from the group consisting of hydrogen, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxy, hydroxyalkyl, and-NR 6 R 7
    R 5 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkyloxy, heterocyclyloxy, aryloxy, heteroaryloxy, -NR 6 R 7 Cyano and nitro, wherein said alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted with one or more substituents selected from the group consisting of alkyl, haloalkyl, alkoxy, haloalkoxy, halogen, cyano, nitro and-NR 6 R 7 Is substituted with one or more substituents of (a);
    R 8 identical or different and are each independently selected from the group consisting of halogen, alkyl, alkenyl, alkynyl, haloalkyl, alkoxy, haloalkoxy, cyano, -NR 6 R 7 Nitro, hydroxy, hydroxyalkyl, -S (O) 2 Alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein said alkyl, haloalkyl, hydroxyalkylCycloalkyl, heterocyclyl, aryl and heteroaryl optionally substituted with one or more substituents selected from hydroxy, halogen, haloalkyl, alkoxy, haloalkoxy, cyano, nitro, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
    R 6 and R 7 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
    y is 0, 1,2,3, 4 or 5;
    z is 2,3, 4 or 5;
    v is 0, 1,2,3, 4 or 5;
    w is 0, 1,2,3, 4 or 5;
    t is 0, 1,2,3, 4 or 5; and the sum of y and t is not more than 5;
    p is 0, 1,2,3, 4 or 5;
    q is 0, 1 or 2;
    n is 0, 1,2,3, 4 or 5.
  2. The compound of formula (I) according to claim 1, wherein Z is- (CH), or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof 2 ) z -or- (CH) 2 ) v C(O)NR 0 (CH 2 ) w -;
    R 0 Z, v and w are as defined in claim 1.
  3. The compound of formula (I) according to claim 1 or2, wherein Z is- (CH) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof 2 ) z -, and z is 2 or 4.
  4. A compound of formula (I) according to any one of claims 1 to 3, which is a compound of formula (II) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2021000067-APPB-100002
    wherein m is 1,2,3, 4 or 5;
    rings A, G 1 、G 2 、R 1 -R 5 、R 8 P, q and n are as defined in claim 1 or 2.
  5. The compound of formula (I) according to any one of claims 1 to 4, wherein G is G or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof 1 And G 2 Is a carbon atom.
  6. The compound of the general formula (I) according to any one of claims 1 to 5, wherein ring a is C, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof 6-10 Aryl, preferably phenyl or
    Figure PCTCN2021000067-APPB-100003
    More preferably phenyl.
  7. The compound of the general formula (I) according to any one of claims 1 to 3,5 to 6, or a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, which is a compound of the general formula (III) or (III-1), or a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2021000067-APPB-100004
    wherein Z is- (CH) 2 ) z -;
    z is 2 or 4; y is 0, 1,2 or 3;
    t is 0, 1,2 or 3; and the sum of y and t is not more than 3;
    Y、T、R 1 、R 2 、R 8 and n is as defined in claim 1 or 2.
  8. The compound of formula (I) according to any one of claims 1 to 7, wherein R is R in the form of its tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof 8 Selected from halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, -NR 6 R 7 And C 1-6 Hydroxyalkyl of wherein said C 1-6 Haloalkyl is optionally substituted with one or more hydroxy; r is 6 And R 7 Selected from hydrogen and C 1-6 An alkyl group.
  9. The compound of formula (I) according to any one of claims 1 to 8, wherein R is in the form of its tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof 1 Selected from hydrogen, C 1-6 Alkyl and halogen.
  10. The compound of general formula (I) according to any one of claims 1 to 9, wherein R is a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof 2 Selected from hydrogen and C 1-6 An alkyl group.
  11. The compound of general formula (I) according to any one of claims 1 to 6,8 to 10, wherein R is a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof 3 Selected from hydrogen and C 1-6 An alkyl group.
  12. The compound of general formula (I) according to any one of claims 1 to 6,8 to 11, wherein R is a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof 4 Is hydrogen.
  13. The compound of formula (I) according to any one of claims 1 to 6,8 to 12, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, wherein R is 5 Is hydrogen.
  14. The compound of the general formula (I) according to any one of claims 1 to 3,5 to 6,8 to 13, or a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof, which is a compound of the general formula (IV) or a tautomer, a meso form, a racemate, an enantiomer, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2021000067-APPB-100005
    wherein ring A, Y, Z, T, G 1 、G 2 、R 1 -R 5 、R 8 P, q and n are as defined in claim 1 or 2.
  15. A compound of general formula (I) according to any one of claims 1 to 14, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, selected from any one of the following compounds:
    Figure PCTCN2021000067-APPB-100006
    Figure PCTCN2021000067-APPB-100007
  16. a compound of formula (IA) or a tautomer, mesomer, racemate, enantiomer, or diastereomer thereof, or a mixture thereof, or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2021000067-APPB-100008
    wherein:
    R w is hydroxy or halogen, preferably hydroxy or chlorine;
    R 1 selected from the group consisting of halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxyalkyl, cyano, and cycloalkyl;
    the sum of y and t is not 0;
    Y、Z、T、G 1 、G 2 、R 4 、R 5 p and q are as defined in claim 1 or 2.
  17. A compound of formula (IVA) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof;
    Figure PCTCN2021000067-APPB-100009
    wherein:
    rings A, Y, Z, T, G 1 、G 2 、R 1 -R 5 、R 8 P, q and n are as defined in claim 1 or 2.
  18. The compound of formula (IA) or (IVA) according to claim 16 or 17, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, selected from any one of the following compounds:
    Figure PCTCN2021000067-APPB-100010
  19. a process for preparing a compound of formula (I) or formula (IV) or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
    Figure PCTCN2021000067-APPB-100011
    reaction of formula (IA) with formula (IB) or a salt thereof to give a compound of formula (I), or
    Carrying out reduction reaction on the compound of the general formula (IVA) to obtain a compound of a general formula (IV);
    wherein:
    R w is hydroxy or halogen, preferably hydroxy or chloro;
    ring A, Y, Z, T, G 1 、G 2 、R 1 -R 5 、R 8 P, q and n are as defined in claim 1 or 2.
  20. A pharmaceutical composition comprising a therapeutically effective amount of a compound of general formula (I) according to any one of claims 1 to 15, or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
  21. Use of a compound of general formula (I) according to any one of claims 1 to 15 or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 20, for the preparation of a medicament for the inhibition of SOS1.
  22. Use of a compound of general formula (I) according to any one of claims 1 to 15 or a tautomer, mesomer, racemate, enantiomer, diastereomer, or mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 20, for the preparation of a medicament for the treatment and/or prevention of cancer, inflammation, RAS disease, noonan Syndrome (NS), noonan syndrome with erythema (NSML), capillary malformation-arteriovenous malformation (CM-AVM), costello Syndrome (CS), heart-plane-skin syndrome (CFC), louses syndrome, hereditary gingival fibromatosis, or other proliferative diseases, preferably cancer; preferably, the cancer is selected from melanoma, skin cancer, liver cancer, kidney cancer, lung cancer, nasopharyngeal cancer, gastric cancer, esophageal cancer, colorectal cancer, colon cancer, rectal cancer, gallbladder cancer, cholangiocarcinoma, chorioepithelioma, pancreatic cancer, polycythemia vera, pediatric tumors, cervical cancer, ovarian cancer, breast cancer, bladder cancer, urothelial cancer, ureteral tumor, prostate cancer, seminoma, testicular tumor, leukemia, head and neck tumor, head and neck squamous cell carcinoma, uterine cancer, endometrial cancer, thyroid cancer, lymphoma, sarcoma, osteoma, osteosarcoma, neuroblastoma, brain tumor, myeloma, astrocytoma, glioblastoma and glioma; the RAS disease is preferably neurofibromatosis type 1 (NF 1); the lung cancer is preferably non-small cell lung cancer, and is further preferably metastatic non-small cell lung cancer; the leukemia is preferably chronic lymphocytic leukemia or acute myelogenous leukemia; the lymphoma is preferably diffuse large B cell lymphoma; the myeloma is preferably multiple myeloma; the osteoma is preferably osteochondroma.
CN202180019688.6A 2020-04-08 2021-04-08 Pyrimidobicyclo derivatives, preparation method thereof and application thereof in medicines Pending CN115244058A (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CN202010269258 2020-04-08
CN2020102692587 2020-04-08
CN202011481898 2020-12-15
CN2020114818980 2020-12-15
PCT/CN2021/000067 WO2021203768A1 (en) 2020-04-08 2021-04-08 Pyrimido dicyclo derivative, preparation method therefor and use thereof in medicine

Publications (1)

Publication Number Publication Date
CN115244058A true CN115244058A (en) 2022-10-25

Family

ID=78022796

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180019688.6A Pending CN115244058A (en) 2020-04-08 2021-04-08 Pyrimidobicyclo derivatives, preparation method thereof and application thereof in medicines

Country Status (3)

Country Link
CN (1) CN115244058A (en)
TW (1) TW202144338A (en)
WO (1) WO2021203768A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115433196A (en) * 2021-06-01 2022-12-06 昆药集团股份有限公司 Crown ether quinazoline derivative, preparation method and application thereof

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11932633B2 (en) 2018-05-07 2024-03-19 Mirati Therapeutics, Inc. KRas G12C inhibitors
EP3908283A4 (en) 2019-01-10 2022-10-12 Mirati Therapeutics, Inc. Kras g12c inhibitors
EP4021444A4 (en) 2019-08-29 2023-01-04 Mirati Therapeutics, Inc. Kras g12d inhibitors
CA3152025A1 (en) 2019-09-24 2021-04-01 David BRIERE Combination therapies
US11702418B2 (en) 2019-12-20 2023-07-18 Mirati Therapeutics, Inc. SOS1 inhibitors
TW202229291A (en) * 2020-12-07 2022-08-01 大陸商北京泰德製藥股份有限公司 Sos1 inhibitor, pharmaceutical composition comprising same, and use thereof
CN116249529A (en) * 2020-12-30 2023-06-09 成都百裕制药股份有限公司 Quinazoline derivative and application thereof in medicine
CN114835719A (en) * 2021-02-01 2022-08-02 苏州泽璟生物制药股份有限公司 Substituted bicyclic aromatic heterocyclic amine inhibitor and preparation method and application thereof
WO2022171018A1 (en) * 2021-02-09 2022-08-18 苏州泽璟生物制药股份有限公司 Substituted benzopyrimidine or pyridopyrimidine amine inhibitor, and preparation method therefor and use thereof
CN117479942A (en) 2021-04-09 2024-01-30 勃林格殷格翰国际有限公司 Anticancer therapy
TWI807787B (en) * 2021-04-19 2023-07-01 大陸商昆藥集團股份有限公司 Benzopyrimidine tricyclic derivative and preparation method and application thereof
WO2022258057A1 (en) * 2021-06-11 2022-12-15 Jingrui Biopharma Co., Ltd. Compounds as anticancer agents
CN117677398A (en) 2021-07-27 2024-03-08 东丽株式会社 Medicine for treating and/or preventing cancer
KR20230121208A (en) * 2022-02-10 2023-08-18 (주)파로스아이바이오 SOS1 inhibitors and derivatives thereof
WO2023207991A1 (en) * 2022-04-29 2023-11-02 南京明德新药研发有限公司 Fused quinazoline compound and use thereof
WO2024022507A1 (en) * 2022-07-29 2024-02-01 江苏恒瑞医药股份有限公司 Pharmaceutical composition comprising kras g12d inhibitor

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1534026A (en) * 2002-03-28 2004-10-06 �Ϳ���ҽҩ��˾ Condensed quinazoline derirative used as tyrosine kinase inhibitor
WO2019122129A1 (en) * 2017-12-21 2019-06-27 Boehringer Ingelheim International Gmbh Novel benzylamino substituted pyridopyrimidinones and derivatives as sos1 inhibitors
CN110167928A (en) * 2016-12-22 2019-08-23 勃林格殷格翰国际有限公司 The quinazoline and derivative that Novel warp benzylamino as SOS1 inhibitor replaces

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1534026A (en) * 2002-03-28 2004-10-06 �Ϳ���ҽҩ��˾ Condensed quinazoline derirative used as tyrosine kinase inhibitor
CN110167928A (en) * 2016-12-22 2019-08-23 勃林格殷格翰国际有限公司 The quinazoline and derivative that Novel warp benzylamino as SOS1 inhibitor replaces
WO2019122129A1 (en) * 2017-12-21 2019-06-27 Boehringer Ingelheim International Gmbh Novel benzylamino substituted pyridopyrimidinones and derivatives as sos1 inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ROMAN C. HILLIG,等: "Discovery of potent SOS1 inhibitors that block RAS activation via disruption of the RAS–SOS1 interaction", 《PNAS》, vol. 116, no. 7, pages 2551, XP055841142, DOI: 10.1073/pnas.1812963116 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115433196A (en) * 2021-06-01 2022-12-06 昆药集团股份有限公司 Crown ether quinazoline derivative, preparation method and application thereof

Also Published As

Publication number Publication date
WO2021203768A1 (en) 2021-10-14
TW202144338A (en) 2021-12-01

Similar Documents

Publication Publication Date Title
CN115244058A (en) Pyrimidobicyclo derivatives, preparation method thereof and application thereof in medicines
CN113801114B (en) Fused bicyclic heteroaryl derivative, preparation method and application thereof in medicines
JP2023519815A (en) Spiro ring-containing quinazoline compounds
KR20240024903A (en) Fused tetracyclic compounds, methods for their preparation and their applications in medicine
CN113980032B (en) Fused tetracyclic derivative, preparation method thereof and application thereof in medicines
CN115605479A (en) Pyridoneopyrimidine derivative, preparation method thereof and application thereof in medicine
CN115697994A (en) Fused quinazoline derivative, preparation method and medical application thereof
WO2018084321A1 (en) Novel compound useful for both egfr inhibition and tumor therapy
CN113825757B (en) Substituted fused bicyclic derivatives, preparation method thereof and application thereof in medicine
CN115385938A (en) Benzopyrimidine compounds, preparation method thereof and application thereof in medicines
CN114907377A (en) Condensed tetracyclic compound, preparation method and medical application thereof
CA3184711A1 (en) Sulfur-containing isoindoline derivative, and preparation method therefor and medical use thereof
CN115772161A (en) Nitrogen-containing heteroaryl compound, preparation method and medical application thereof
CN115368383A (en) Condensed nitrogen-containing heterocyclic compound, preparation method and medical application thereof
CN112574212B (en) Pyrimido five-membered nitrogen heterocyclic derivative, preparation method and medical application thereof
WO2022174765A1 (en) Fused ring compound as wee-1 inhibitor
CN112996783B (en) 2-aminopyrimidine derivatives, preparation method and application thereof in medicines
CN113912608B (en) Pyrimidopyrimidinone derivatives, preparation method thereof and application thereof in medicines
CN115867552A (en) Imidazolopyrimidine derivative, preparation method thereof and application thereof in medicines
CN113754678B (en) Dihydrothiochromene pyrazole derivative, preparation method and medical application thereof
CN114456173B (en) Condensed ring group substituted cyclohexanediimide derivative, preparation method and medical application thereof
CN113754685B (en) Dihydrothiochromene pyrazole derivative, preparation method and application thereof in medicine
CN115385937A (en) Pyrimido-cycloalkyl compounds, preparation method and medical application thereof
CN115836077A (en) Oxaazaspiro derivatives, preparation method and medical application thereof
WO2020187291A1 (en) Pyrazolopyrimidine compound, pharmaceutical composition, and application therefor

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