CN115368383A - Condensed nitrogen-containing heterocyclic compound, preparation method and medical application thereof - Google Patents

Condensed nitrogen-containing heterocyclic compound, preparation method and medical application thereof Download PDF

Info

Publication number
CN115368383A
CN115368383A CN202210558739.9A CN202210558739A CN115368383A CN 115368383 A CN115368383 A CN 115368383A CN 202210558739 A CN202210558739 A CN 202210558739A CN 115368383 A CN115368383 A CN 115368383A
Authority
CN
China
Prior art keywords
group
cancer
membered
pharmaceutically acceptable
compound
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
CN202210558739.9A
Other languages
Chinese (zh)
Inventor
李心
沈峰
董怀德
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 CN115368383A publication Critical patent/CN115368383A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic System
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0803Compounds with Si-C or Si-Si linkages
    • C07F7/081Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te
    • C07F7/0812Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te comprising a heterocyclic ring

Abstract

The present disclosure relates to fused nitrogen-containing heterocycles, processes for their preparation and their use in medicine. Specifically, the disclosure relates to a condensed nitrogen-containing heterocyclic compound shown in a general formula (I), a preparation method thereof and a compound containing the compoundPharmaceutical compositions of the compounds and their use as therapeutic agents, in particular their use in the manufacture of medicaments for inhibiting KRAS G12D. Wherein each group in the general formula (I) is defined in the specification.

Description

Condensed nitrogen-containing heterocyclic compound, preparation method and medical application thereof
Technical Field
The disclosure belongs to the field of medicines, and relates to a condensed nitrogen-containing heterocyclic compound, a preparation method thereof and application thereof in medicines. In particular, the disclosure relates to a condensed nitrogen-containing heterocyclic compound shown in a general formula (I), a preparation method thereof, a pharmaceutical composition containing the compound, and an application thereof in preparing a medicament for inhibiting KRAS G12D.
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%. KRAS mutations are common in solid tumors, with high frequency mutations in all three human fatal cancers, lung cancer (17%), colorectal cancer (33%), and pancreatic cancer (61%). In KRAS gene mutation, 97% of amino acid residues are mutated at amino acid residue number 12 or 13, wherein G12D is an important mutation. Data analysis on the European and American population shows that: in pancreatic, colorectal and non-small cell lung cancers, G12D mutations account for 36%, 12% and 4% of patients, respectively.
After KRAS is activated, the KRAS regulates and controls multiple functions such as cell proliferation, survival, migration and metabolism through a plurality of downstream signal pathways represented by RAF-MEK-ERK, PI3K-AKT-mTOR and TIAM 1-RAc. After mutation of KRAS gene, the protein is continuously activated, resulting in continuous activation of downstream signaling pathways to promote tumorigenesis.
KRAS protein is considered as an unpageable drug target for a long time because of the surface lack of conventional small molecule binding sites and the ultrahigh affinity with guanylic acid, so that the KRAS protein 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. Currently, there is a lack of KRAS inhibitors other than KRAS G12C inhibitors that are effective against other mutations, such that a large proportion of KRAS mutated patients remain non-medicated. G12D, as a mutant with wide high expression in various tumors, has important clinical significance for developing an inhibitor against the same.
Related patents which have been disclosed so far are WO2021041671A1, WO2020146613A1, WO2017172979A1, WO2020238791A1, WO2021000885A1 and the like.
Disclosure of Invention
The object of the present disclosure is to provide a compound represented by the general formula (I):
Figure BDA0003653591470000021
wherein:
G 2 is NR d
Ring a is aryl or heteroaryl;
ring B is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
l is selected from the group consisting of a single bond, O and NR e
Each R is 1 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, - (CH) 2 ) u -NR f R g Hydroxyl and hydroxyalkyl groups;
R 2 、R 2c 、R 2a 、R 2b 、R 4a and R 4b Are the same or different and are each independently selected from the group consisting of a hydrogen atom, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, - (CH) 2 ) v -NR h R i Hydroxyl, hydroxyalkyl and cycloalkyl groups; or
R 2 、R 2c To the carbon atom to which it is attached, R 2a 、R 2b To the carbon atom to which it is attached, or R 4a 、R 4b Together with the carbon atom to which they are attached form a cycloalkyl or heterocyclyl group;
each R is 3 And R 6 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, - (CH) 2 ) w -NR j R k Nitro, hydroxyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R 5 are the same or different and are each independently selected from the group consisting of hydrogen atom, halogen, alkyl group, haloalkyl group, cyano group, hydroxy group and hydroxyalkyl group;
R d 、R e 、R f 、R g 、R h 、R i 、R j and R k Are the same or different and are each independently selected from the group consisting of hydrogen atoms, alkyl groups, alkenyl groups, alkynyl groups, haloalkyl groups, hydroxyalkyl groups, cycloalkyl groups, heterocyclyl groups, aryl groups, and heteroaryl groups;
u, v and w are the same or different and are each independently selected from 0, 1,2 and 3;
m is 0, 1 or 2;
n is 0 or 1;
r is 0, 1 or 2;
p is 0, 1,2,3, 4 or 5;
q is 0, 1,2,3, 4 or 5; and is provided with
t is 0, 1,2,3, 4 or 5.
Another aspect of the present disclosure relates to a compound represented by the general formula (I-1):
Figure BDA0003653591470000031
wherein
q is 0, 1,2,3 or 4;
rings A, G 2 、L、R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, r and t are as defined in formula (I).
In some preferred embodiments of the present disclosure, the compound of formula (I) or (I-1), or a pharmaceutically acceptable salt thereof, wherein ring A is 6-to 10-membered aryl or 5-to 10-membered heteroaryl; preferably, ring a is phenyl or naphthyl; further preferred is naphthyl.
In some preferred embodiments of the present disclosure, the compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein
Figure BDA0003653591470000032
Is composed of
Figure BDA0003653591470000033
R 3 The definition is defined in the general formula (I); preferably, each R 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkynyl, C 1-6 Haloalkyl, hydroxy, C 1-6 Hydroxyalkyl and 3 to 8 membered cycloalkyl; more preferably, each R 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkynyl, C 1-6 Haloalkyl, hydroxy and cyclopropyl; most preferably, each R 3 Are the same or different and are each independently selected from the group consisting of a hydrogen atom, a halogen, an ethyl group, an ethynyl group, and a hydroxyl group.
In some embodiments of the disclosure, the compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein ring A is phenyl and each R is 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl, aryl, heteroaryl, and heteroaryl,C 2-6 Alkynyl, C 1-6 Haloalkyl, hydroxy, C 1-6 Hydroxyalkyl and 3 to 8 membered cycloalkyl; preferably, ring A is phenyl, each R 3 Are the same or different and are each independently selected from the group consisting of a hydrogen atom, a halogen, a hydroxyl group, and a 3-to 8-membered cycloalkyl group; more preferably, ring A is phenyl, each R 3 Are the same or different and are each independently selected from the group consisting of a hydrogen atom, a halogen, a hydroxyl group and a cyclopropyl group.
In some preferred embodiments of the present disclosure, the compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein ring B is a 7-to 10-membered fused heterocyclyl and R is 6 Can be substituted at any position on the ring B; preferably, ring B is
Figure BDA0003653591470000041
R 6 May be substituted at any position on said ring B; it is further preferred that the first and second liquid crystal compositions,
Figure BDA0003653591470000042
is composed of
Figure BDA0003653591470000043
R 6 As defined in formula (I); more preferably still, the first and second liquid crystal compositions are,
Figure BDA0003653591470000044
is composed of
Figure BDA0003653591470000045
Figure BDA0003653591470000046
R 6 Is halogen; most preferably, the first and second liquid crystal display panels are,
Figure BDA0003653591470000047
is composed of
Figure BDA0003653591470000048
R 6 Is halogen (preferably F).
In some preferred embodiments of the present disclosure, the compound of formula (I) or a pharmaceutically acceptable salt thereofRing a is phenyl or naphthyl; and/or ring B is
Figure BDA0003653591470000049
R 6 Can be substituted at any position of the ring B.
Another aspect of the present disclosure relates to a compound represented by the general formula (II):
Figure BDA00036535914700000410
wherein
G 2 、L、R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, q, r and t are as defined in formula (I).
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein R d Is a hydrogen atom or C 1-6 An alkyl group; preferably, R d Is a hydrogen atom.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein n is 0.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein m is 1 or 2; preferably, m is 1.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein R 2 And R 2c Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 Haloalkyl, or R 2 、R 2c Together with the carbon atom to which they are attached form a 3-to 8-membered cycloalkyl group or a 3-to 8-membered heterocyclyl group; preferably, R 2 And R 2c Is a hydrogen atom, or R 2 、R 2c Together with the carbon atom to which they are attached form a cyclopropyl group; more preferably, R 2 And R 2c Is a hydrogen atom.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein R 2a And R 2b Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 Haloalkyl, or R 2a 、R 2b Together with the carbon atom to which they are attached form a 3-to 8-membered cycloalkyl or 3-to 8-membered heterocyclyl group; preferably, R 2a And R 2b Is a hydrogen atom, or R 2a 、R 2b Together with the carbon atom to which they are attached form a cyclopropyl group; more preferably, R 2a And R 2b Is a hydrogen atom.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein R 4a And R 4b Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 Haloalkyl, or R 4a 、R 4b Together with the carbon atom to which they are attached form a 3-to 8-membered cycloalkyl group or a 3-to 8-membered heterocyclyl group; preferably, R 4a And R 4b Is a hydrogen atom, or R 4a 、R 4b Together with the carbon atom to which they are attached form a cyclopropyl group; more preferably, R 4a And R 4b Is a hydrogen atom.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein R 2 And R 2c Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 Haloalkyl, or R 2 、R 2c Together with the carbon atom to which they are attached form a 3-to 8-membered cycloalkyl group or a 3-to 8-membered heterocyclyl group; and/or R 2a And R 2b Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 Haloalkyl, or R 2a 、R 2b Together with the carbon atom to which they are attached form a 3-to 8-membered cycloalkyl group or a 3-to 8-membered heterocyclyl group; and/or R 4a And R 4b Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 Haloalkyl, or R 4a 、R 4b Together with the carbon atom to which they are attached form a 3-to 8-membered cycloalkyl group or a 3-to 8-membered heterocyclyl group.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein G 2 Is NH.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein L is O.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein R e Is a hydrogen atom or C 1-6 An alkyl group; preferably, R e Is a hydrogen atom.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein each R is 1 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, cyano, amino, - (CH) 2 ) u -NR f R g Hydroxy and C 1-6 Hydroxyalkyl radical, R f And R g Are the same or different and are each independently a hydrogen atom or C 1-6 Alkyl, u is 0 or 1; preferably, each R 1 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; further preferably, R 1 Is a hydrogen atom.
In some preferred embodiments of the present disclosure, the compound of formula (I) or (II) or a pharmaceutically acceptable salt thereof, wherein each R is 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkenyl radical, C 2-6 Alkynyl, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, cyano, amino, - (CH) 2 ) w -NR j R k Hydroxy and C 1-6 Hydroxyalkyl radical, R j And R k Are the same or different and eachIndependently is a hydrogen atom or C 1-6 Alkyl, w is 0 or 1.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein each R is 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkynyl, C 1-6 Haloalkyl, hydroxy, C 1-6 Hydroxyalkyl and 3 to 8 membered cycloalkyl; preferably, each R 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkynyl, hydroxy and 3 to 8 membered cycloalkyl; more preferably, each R 3 Identical or different and are each independently selected from the group consisting of a hydrogen atom, a halogen, an ethyl group, an ethynyl group, a hydroxyl group and a cyclopropyl group.
In some preferred embodiments of the present disclosure, the compound of formula (I-1) or a pharmaceutically acceptable salt thereof, wherein each R is 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkynyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl and 3 to 8 membered cycloalkyl; preferably, each R 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkynyl and 3 to 8 membered cycloalkyl; more preferably, each R 3 Identical or different and are each independently selected from the group consisting of a hydrogen atom, a halogen, an ethyl group, an ethynyl group and a cyclopropyl group.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein R 5 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 1-6 Haloalkyl, hydroxy and C 1-6 A hydroxyalkyl group; preferably, R 5 Are the same or different and are each independently selected from the group consisting of a hydrogen atom, C 1-6 Alkyl, hydroxy and C 1-6 A hydroxyalkyl group; further preferably, R 5 Is a hydrogen atom.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a combination thereofA pharmaceutically acceptable salt, wherein each R 6 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 1-6 Alkoxy radical, C 1-6 Haloalkyl, C 1-6 Haloalkoxy, cyano, amino, - (CH) 2 ) w -NR j R k Hydroxy and C 1-6 Hydroxyalkyl radical, R j And R k Are the same or different and are each independently a hydrogen atom or C 1-6 Alkyl, w is 0 or 1; preferably, each R 6 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; more preferably a halogen; most preferably F.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein r is 0 or 1; preferably, r is 1.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein p is 0 or 1, preferably 1.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein p is 0.
In some preferred embodiments of the present disclosure, the compound of formula (I) or (II) or a pharmaceutically acceptable salt thereof, wherein q is 2 or 3, preferably 2.
In some preferred embodiments of the present disclosure, the compound represented by the general formula (I-1) or a pharmaceutically acceptable salt thereof, wherein q is 1 or 2.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein t is 1 or2, preferably 1.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein u is 0 or 1.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein v is 0 or 1.
In some preferred embodiments of the present disclosure, the compound of formula (I), (I-1) or (II) or a pharmaceutically acceptable salt thereof, wherein w is 0 or 1.
In some preferred embodiments of the present disclosure, the compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein ring a is 6-to 10-membered aryl or 5-to 10-membered heteroaryl; ring B is
Figure BDA0003653591470000071
R 6 May be substituted at any position on said ring B; g 2 Is NH; l is O; p is 1; r is 1 Selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; r 2 、R 2c 、R 2a 、R 2b 、R 4a And R 4b Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; or R 2 、R 2c To the carbon atom to which it is attached, or R 2a 、R 2b To the carbon atom to which it is attached, or R 4a 、R 4b Together with the carbon atom to which they are attached form a cyclopropyl group; q is 2 or 3; each R is 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkynyl, C 1-6 Haloalkyl, hydroxy, C 1-6 Hydroxyalkyl and 3 to 8 membered cycloalkyl; r is 0 or 1; r is 5 Selected from hydrogen atoms, C 1-6 Alkyl, hydroxy and C 1-6 A hydroxyalkyl group; t is 1; r 6 Selected from hydrogen atoms, halogens, C 1-6 Alkyl and C 1-6 A haloalkyl group; m is 1 or 2; n is 0 or 1.
In some preferred embodiments of the present disclosure, the compound of formula (I-1) or a pharmaceutically acceptable salt thereof, wherein ring A is a 6-to 10-membered aryl or 5-to 10-membered heteroaryl; g 2 Is NH; l is O; p is 1; r 1 Selected from hydrogen atoms, halogens, C 1-6 Alkyl and C 1-6 A haloalkyl group; r is 2 、R 2c 、R 2a 、R 2b 、R 4a And R 4b Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; or R 2 、R 2c To the carbon atom to which it is attached, or R 2a 、R 2b To the carbon atom to which it is attached, or R 4a 、R 4b Together with the carbon atom to which they are attached form a cyclopropyl group; q is 1 or 2; each R is 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkynyl, C 1-6 Haloalkyl, C 1-6 Hydroxyalkyl and 3 to 8 membered cycloalkyl; r is 0 or 1; r 5 Selected from hydrogen atoms, C 1-6 Alkyl, hydroxy and C 1-6 A hydroxyalkyl group; t is 1; r 6 Selected from hydrogen atoms, halogens, C 1-6 Alkyl and C 1-6 A haloalkyl group; m is 1 or 2; n is 0 or 1.
In some preferred embodiments of the present disclosure, the compound of formula (I-1) or a pharmaceutically acceptable salt thereof, wherein ring A is phenyl or naphthyl; g 2 Is NH; l is O; p is 0; r is 2 、R 2c 、R 2a 、R 2b 、R 4a And R 4b Are each a hydrogen atom; or R 2 、R 2c To the carbon atom to which it is attached, or R 2a 、R 2b To the carbon atom to which it is attached, or R 4a 、R 4b Together with the carbon atom to which they are attached form a cyclopropyl group; q is 1 or 2; each R is 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkynyl and 3 to 8 membered cycloalkyl; r is 1; r 5 Is a hydrogen atom; t is 1; r is 6 Is halogen; m is 1 or 2; n is 0 or 1.
In some preferred embodiments of the present disclosure, the compound of formula (II) or a pharmaceutically acceptable salt thereof, wherein G 2 Is NH; l is O; p is 1; r is 1 Selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; r 2 、R 2c 、R 2a 、R 2b 、R 4a And R 4b Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; or R 2 、R 2c To the carbon atom to which it is attached, or R 2a 、R 2b To the carbon atom to which it is attached, or R 4a 、R 4b Together with the carbon atom to which they are attached form a cyclopropyl group; q is 1 or 2; each R is 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkynyl, C 1-6 Haloalkyl, hydroxy and C 1-6 A hydroxyalkyl group; r is 0 or 1; r 5 Selected from hydrogen atoms, C 1-6 Alkyl, hydroxy and C 1-6 A hydroxyalkyl group; t is 1; r 6 Selected from hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group; m is 1 or 2; n is 0.
Table a typical compounds of the present disclosure include, but are not limited to:
Figure BDA0003653591470000091
Figure BDA0003653591470000101
Figure BDA0003653591470000111
Figure BDA0003653591470000121
Figure BDA0003653591470000131
Figure BDA0003653591470000141
Figure BDA0003653591470000151
Figure BDA0003653591470000161
Figure BDA0003653591470000171
Figure BDA0003653591470000181
Figure BDA0003653591470000191
Figure BDA0003653591470000201
another aspect of the present disclosure relates to a compound represented by the general formula (IA) or a salt thereof,
Figure BDA0003653591470000202
wherein, the first and the second end of the pipe are connected with each other,
r is an amino protecting group; preferably Boc;
ring A, ring B, L, R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, q, r and t are as defined in formula (I).
Another aspect of the present disclosure relates to a compound represented by the general formula (I-1A) or a salt thereof,
Figure BDA0003653591470000203
wherein the content of the first and second substances,
q is 0, 1,2,3 or 4;
R 0 is a hydrogen atom or a hydroxyl protecting group; the above-mentionedThe hydroxyl protecting group of (2) is preferably MOM;
r is an amino protecting group; preferably Boc;
ring A, L, R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, r and t are as defined in the general formula (I-1).
In some preferred embodiments of the present disclosure, the compound represented by the general formula (I-1A) or a pharmaceutically acceptable salt thereof, wherein the hydroxyl protecting group is preferably MOM or Bn, more preferably MOM.
Another aspect of the present disclosure relates to a compound of formula (IIA) or a salt thereof,
Figure BDA0003653591470000211
wherein the content of the first and second substances,
r is an amino protecting group; preferably Boc;
L、R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 m, n, p, q, r and t are as defined in formula (II).
Typical intermediate compounds of the present disclosure include, but are not limited to:
Figure BDA0003653591470000212
Figure BDA0003653591470000221
Figure BDA0003653591470000231
Figure BDA0003653591470000241
Figure BDA0003653591470000251
Figure BDA0003653591470000261
Figure BDA0003653591470000271
Figure BDA0003653591470000281
Figure BDA0003653591470000291
Figure BDA0003653591470000301
Figure BDA0003653591470000311
another aspect of the present disclosure relates to a method of preparing a compound of formula (I), or a pharmaceutically acceptable salt thereof, comprising:
Figure BDA0003653591470000312
carrying out deprotection reaction on the compound of the general formula (IA) or salt thereof to obtain the compound of the general formula (I) or pharmaceutically acceptable salt thereof; optionally, when R 3 And/or R 6 When the group contains a protecting group, the deprotection reaction may further comprise removing R before, simultaneously with or after the deprotection reaction 3 And/or R 6 A step of protecting groups on the group; preferably, the protecting group is a hydroxyl protecting group MOM;
wherein the content of the first and second substances,
r is an amino protecting group; preferably Boc;
G 2 is NH;
ring A, ring B, L, R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, q, r and t are as defined in formula (I).
Another aspect of the present disclosure relates to a method for preparing a compound represented by the general formula (I-1) or a pharmaceutically acceptable salt thereof, which comprises:
Figure BDA0003653591470000321
R 0 is a hydrogen atom or a hydroxyl protecting group; the hydroxyl protecting group is preferably MOM;
when R is 0 When the hydrogen atom is a hydrogen atom, removing the amino protecting group R from the compound of the general formula (I-1A) or a salt thereof to obtain a compound of the general formula (I-1) or a pharmaceutically acceptable salt thereof;
when R is 0 When the protecting group is a hydroxyl group, the compound of the formula (I-1A) or a salt thereof is freed from the protecting group R 0 And an amino protecting group R to give a compound of the general formula (I-1) or a pharmaceutically acceptable salt thereof;
wherein, the first and the second end of the pipe are connected with each other,
r is an amino protecting group; preferably Boc;
G 2 is NH;
ring A, L, R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, q, r and t are as defined in the general formula (I-1).
Another aspect of the present disclosure relates to a method of preparing a compound represented by the general formula (II) or a pharmaceutically acceptable salt thereof, the method comprising:
Figure BDA0003653591470000322
carrying out deprotection reaction on the compound of the general formula (IIA) or salt thereof to obtain a compound of a general formula (II) or pharmaceutically acceptable salt thereof; optionally, when R 3 And/or R 6 When the group contains a protecting group, the deprotection reaction may further comprise removing R before, simultaneously with or after the deprotection reaction 3 And/or R 6 A step of protecting groups on the group; preferably, the protecting group is a hydroxyl protecting group MOM;
wherein R is an amino protecting group; preferably Boc;
G 2 is NH;
L、R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 m, n, p, q, r and t are as defined in formula (II).
Another aspect of the present disclosure relates to a pharmaceutical composition comprising a compound of formula (I), (I-1), (II) or table a of the present disclosure, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients.
The disclosure further relates to the use of a compound of general formula (I), (I-1), (II) or shown in table a, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, in the preparation of a medicament for inhibiting KRAS G12D.
The present disclosure further relates to the use of a compound of formula (I), (I-1), (II) or a compound shown in table a, 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 a disease or disorder which is cancer; the disease or condition is preferably selected from the group consisting of brain cancer, thyroid cancer, head and neck cancer, nasopharyngeal cancer, laryngeal cancer, oral cancer, salivary gland cancer, esophageal cancer, gastric cancer, lung cancer, liver cancer, kidney cancer, pleural cancer, peritoneal cancer, pancreatic cancer, gall bladder cancer, bile duct cancer, colorectal cancer, small intestine cancer, gastrointestinal stromal tumors, urothelial cancer, urinary tract cancer, bladder cancer, anal cancer, joint cancer, breast cancer, vaginal cancer, ovarian cancer, endometrial cancer, cervical cancer, fallopian tube cancer, testicular cancer, prostate cancer, hemangioma, leukemia, lymphoma, myeloma, skin cancer, melanoma, lipoma, bone cancer, soft tissue sarcoma, neurofibroma, glioma, neuroblastoma, and glioblastoma; further preferred is a cancer selected from pancreatic cancer, colorectal cancer and non-small cell lung cancer.
The present disclosure further relates to a method of inhibiting KRAS G12D comprising administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I), (I-1), (II), or table a, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
The present disclosure further relates to a method of treating and/or preventing a disease or disorder, comprising administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I), (I-1), (II), or table a, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, wherein the disease or disorder is cancer; the disease or condition is preferably selected from the group consisting of brain cancer, thyroid cancer, head and neck cancer, nasopharyngeal cancer, laryngeal cancer, oral cancer, salivary gland cancer, esophageal cancer, gastric cancer, lung cancer, liver cancer, kidney cancer, pleural cancer, peritoneal cancer, pancreatic cancer, gall bladder cancer, bile duct cancer, colorectal cancer, small intestine cancer, gastrointestinal stromal tumors, urothelial cancer, urinary tract cancer, bladder cancer, anal cancer, joint cancer, breast cancer, vaginal cancer, ovarian cancer, endometrial cancer, cervical cancer, fallopian tube cancer, testicular cancer, prostate cancer, hemangioma, leukemia, lymphoma, myeloma, skin cancer, melanoma, lipoma, bone cancer, soft tissue sarcoma, neurofibroma, glioma, neuroblastoma, and glioblastoma; further preferably selected from pancreatic cancer, colorectal cancer and non-small cell lung cancer.
The present disclosure further relates to a compound of general formula (I), (I-1), (II) or shown in table a, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for use as a medicament.
The disclosure further relates to a compound of general formula (I), (I-1), (II) or table a or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for use as a medicament for inhibiting KRAS G12D.
The present disclosure further relates to a compound of general formula (I), (I-1), (II) or table a, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same, for use as a medicament for the treatment and/or prevention of a disease or disorder, wherein said disease or disorder is cancer; the disease or condition is preferably selected from the group consisting of brain cancer, thyroid cancer, head and neck cancer, nasopharyngeal cancer, laryngeal cancer, oral cancer, salivary gland cancer, esophageal cancer, gastric cancer, lung cancer, liver cancer, kidney cancer, pleural cancer, peritoneal cancer, pancreatic cancer, gall bladder cancer, bile duct cancer, colorectal cancer, small intestine cancer, gastrointestinal stromal tumors, urothelial cancer, urinary tract cancer, bladder cancer, anal cancer, joint cancer, breast cancer, vaginal cancer, ovarian cancer, endometrial cancer, cervical cancer, fallopian tube cancer, testicular cancer, prostate cancer, hemangioma, leukemia, lymphoma, myeloma, skin cancer, melanoma, lipoma, bone cancer, soft tissue sarcoma, neurofibroma, glioma, neuroblastoma, and glioblastoma; further preferably selected from pancreatic cancer, colorectal cancer and non-small cell lung cancer.
The diseases or disorders described in this disclosure are diseases or disorders that are treated and/or prevented by inhibiting KRAS G12D.
The colorectal cancer described in the present disclosure is preferably colon cancer or rectal cancer.
Preferably, the brain cancer described in the present disclosure is selected from glioblastoma multiforme or neuroblastoma; soft tissue cancer selected from fibrosarcoma, gastrointestinal sarcoma, rhabdomyoma, leiomyosarcoma, dedifferentiated liposarcoma, liposarcoma polymorpha, malignant fibrous histiocytoma, round cell sarcoma, and synovial sarcoma; the lymphoma is selected from hodgkin's disease and non-hodgkin's lymphoma (e.g., mantle cell lymphoma, diffuse large B-cell lymphoma, follicular center lymphoma, marginal zone B-cell lymphoma, lymphoplasmacytic lymphoma, and peripheral T-cell lymphoma); the liver cancer is preferably hepatocellular carcinoma; lung cancer (also known as bronchogenic lung cancer) is selected from non-small cell lung cancer (NSCLC) (e.g., squamous cell lung carcinoma (also known as squamous cell lung carcinoma)) and Small Cell Lung Cancer (SCLC); kidney cancer selected from renal cell carcinoma, clear cell and renal eosinophilic tumor; the leukemia is selected from Chronic Lymphocytic Leukemia (CLL), chronic myelogenous leukemia, acute Lymphoblastic Leukemia (ALL), T-cell acute lymphoblastic leukemia (T-ALL), chronic Myelogenous Leukemia (CML) and Acute Myelogenous Leukemia (AML); the skin cancer is selected from malignant melanoma, squamous cell carcinoma, basal cell carcinoma and angiosarcoma; the myeloma is preferably multiple myeloma.
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 compositions of the disclosure by conventional methods. 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 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. A suitable unit dose 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 or 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 oil, or in a 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 an oil phase, and the injection or microemulsion may be injected into the bloodstream of a patient by local mass injection. Alternatively, it may be desirable to administer the solution and microemulsion 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 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 prepared into 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 a drug administered depends on a variety of factors, including, but not limited to: the activity of the particular compound employed, the severity of the disease, 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, 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.
Description of the terms
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 (i.e., C) group having 1 to 12 (e.g., 1,2,3, 4,5, 6,7,8,9, 10, 11, and 12) carbon atoms 1-12 Alkyl), more preferably an alkyl group having 1 to 6 carbon atoms (i.e., C) 1-6 Alkyl groups). 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,<xnotran> 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- ,2,2- , ,3,3- ,2,2- , . </xnotran> The alkyl group may be substituted or unsubstituted, and when substituted, may be substituted at any available point of attachment, the substituents preferably being selected from one or more of a D atom, halogen, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
The term "alkylene" refers to a saturated straight or branched aliphatic hydrocarbon group, 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 group containing 1 to 20 carbon atoms, preferably having 1 to 12 (e.g., 1,2,3, 4,5, 6,7,8,9, 10, 11, and 12) carbon atoms (i.e., C) 1-12 Alkylene), more preferably alkylene having 1 to 6 carbon atoms (i.e., C) 1-6 Alkylene). 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 group may be substituted or unsubstituted, and when substituted, itMay be substituted at any available point of attachment, the substituents preferably being selected from one or more of 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 group containing at least one carbon-carbon double bond in the molecule, wherein alkyl is as defined above. Alkenyl groups (i.e., C) having 2 to 12 (e.g., 2,3, 4,5, 6,7,8,9, 10, 11, and 12) carbon atoms are preferred 2-12 Alkenyl), more preferably alkenyl having 2 to 6 carbon atoms (i.e., C) 2-6 Alkenyl). Non-limiting examples include: ethenyl, propenyl, isopropenyl, butenyl, and the like. The alkenyl group may be substituted or unsubstituted, and when substituted, the substituent is preferably selected from one or more of alkoxy, halogen, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
The term "alkynyl" refers to an alkyl group containing at least one carbon-carbon triple bond in the molecule, wherein alkyl is as defined above. Alkynyl groups (i.e., C) having 2 to 12 (e.g., 2,3, 4,5, 6,7,8,9, 10, 11, and 12) carbon atoms are preferred 2-12 Alkynyl), more preferably alkynyl having 2 to 6 carbon atoms (i.e., C) 2-6 Alkynyl). Non-limiting examples include: ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Alkynyl groups may be substituted or unsubstituted and when substituted, the substituents are preferably selected from one or more of 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, and the cycloalkyl ring contains 3 to 20 carbon atoms (i.e., 3 to 20-membered cycloalkyl groups), preferably 3 to 14 (e.g., 3,4, 5,6,7,8, 9, 10, 11, 12, 13, and 14) carbon atoms (i.e., 3 to 14-membered cycloalkyl groups), further preferably 3 to 8 (e.g., 3,4, 5,6,7, and 8) carbon atoms (i.e., 3 to 8-membered cycloalkyl groups), and more preferably 3 to 6 carbon atoms (i.e., 3 to 6-membered cycloalkyl groups). 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 spirocycloalkyl, fused ring alkyl, and bridged ring alkyl groups.
The term "spirocycloalkyl" refers to a 5 to 20 membered polycyclic group (i.e., a 5 to 20 membered spirocycloalkyl group) having a single ring with one carbon atom (referred to as a spiro atom) in common between the rings, which may contain one or more double bonds. Preferably 6 to 14 membered (i.e. 6 to 14 membered spirocycloalkyl), more preferably 7 to 10 membered (e.g. 7,8,9 or 10 membered) (i.e. 7 to 10 membered spirocycloalkyl). Spirocycloalkyl groups are classified into mono-spirocycloalkyl groups and multi-spirocycloalkyl groups (e.g., bis-spirocycloalkyl groups, etc.), preferably mono-spirocycloalkyl groups and bis-spirocycloalkyl groups, according to the number of spiro atoms shared between rings. 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/3-membered, 5-membered/4-membered, 5-membered/5-membered, 5-membered/6-membered, 5-membered/7-membered, 6-membered/3-membered, 6-membered/4-membered, 6-membered/5-membered, 6-membered/6-membered, 6-membered/7-membered, 7-membered/5-membered or 7-membered/6-membered spirocycloalkyl. Non-limiting examples of spirocycloalkyl groups include:
Figure BDA0003653591470000381
the term "fused cycloalkyl" refers to a 5 to 20 membered (i.e., 5 to 20 membered fused cycloalkyl) 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 (i.e. 6 to 14 fused cycloalkyl) members, more preferably 7 to 10 (e.g. 7,8,9 or 10) (i.e. 7 to 10 fused cycloalkyl) members. They can be divided into bicyclic and polycyclic (e.g., tricyclic, tetracyclic, etc.) fused cycloalkyl groups 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/3-membered, 5-membered/4-membered, 5-membered/5-membered, 5-membered/7-membered, 6-membered/3-membered, 6-membered/4-membered, 6-membered/5-membered, 6-membered/6-membered, 6-membered/7-membered, 7-membered/5-membered or 7-membered/6-membered bicycloalkyl groups. Non-limiting examples of fused ring alkyl groups include:
Figure BDA0003653591470000382
the term "bridged cycloalkyl" refers to a 5 to 20 membered (i.e., 5 to 20 membered bridged cycloalkyl), 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 (i.e. 6 to 14 bridged cycloalkyl), more preferably 7 to 10 (e.g. 7,8,9 or 10) (i.e. 7 to 10 bridged cycloalkyl). They may be classified into bicyclic and polycyclic (e.g., tricyclic, tetracyclic, etc.) bridged cycloalkyl groups according to the number of constituent rings, and are preferably bicyclic, tricyclic or tetracyclic, and more preferably bicyclic or tricyclic. Non-limiting examples of bridged cycloalkyl groups include:
Figure BDA0003653591470000383
the cycloalkyl ring includes a cycloalkyl (including monocyclic, spiro, fused, and bridged rings) fused to an aryl, heteroaryl, or heterocycloalkyl ring as described above, where the rings joined together to the parent structure are cycloalkyl, non-limiting examples of which include
Figure BDA0003653591470000384
Etc.; preference is given to
Figure BDA0003653591470000385
Figure BDA0003653591470000386
Cycloalkyl groups may be substituted or unsubstituted, and when substituted, may be substituted at any available point of attachment, with the substituents preferably being selected from one or more of halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and 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 selected from the group consisting of 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 mono-or polycyclic cyclic substituent comprising from 3 to 20 (e.g., 3,4, 5,6,7,8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20) ring atoms (i.e., a 3-to 20-membered heterocyclyl group) wherein one or more ring atoms is a heteroatom selected from nitrogen, oxygen, and sulfur, which may optionally be oxo (i.e., to form a sulfoxide or sulfone), but does not include a ring moiety of-O-, -O-S-, or-S-, with the remaining ring atoms being carbon. Preferably 3 to 14 (e.g., 3,4, 5,6,7,8, 9, 10, 11, 12, 13 and 14) ring atoms (i.e., 3 to 14 membered heterocyclyl), of which 1 to 4 (e.g., 1,2,3 and 4) are heteroatoms; more preferably from 3 to 8 ring atoms (e.g., 3,4, 5,6,7 and 8) or from 6 to 14 ring atoms (e.g., 6,7,8,9, 10, 11, 12, 13 and 14), of which 1-3 are heteroatoms (e.g., 1,2 and 3); more preferably 3 to 8 ring atoms (i.e. 3 to 8 membered heterocyclyl), of which 1-3 (e.g. 1,2 and 3) are heteroatoms; most preferably 5 or 6 ring atoms (i.e. a 5 or 6 membered heterocyclyl group) are included, 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 heterocyclic groups, fused heterocyclic groups, and bridged heterocyclic groups.
The term "spiroheterocyclyl" refers to a 5 to 20 membered polycyclic heterocyclic group (i.e., a 5 to 20 membered spiroheterocyclyl group) in which one atom (referred to as a spiro atom) is shared between single rings, wherein one or more of the ring atoms is a heteroatom selected from nitrogen, oxygen, and sulfur, which may optionally be oxo (i.e., form a sulfoxide or sulfone), with the remaining ring atoms being carbon. It may contain one or more double bonds. Preferably 6 to 14 (e.g. 6,7,8,9, 10, 11, 12, 13 and 14) membered (i.e. 6 to 14 membered spiroheterocyclyl), more preferably 7 to 10 (e.g. 7,8,9 or 10) membered (i.e. 7 to 10 membered spiroheterocyclyl). Spiro heterocyclic groups are classified into a mono-spiro heterocyclic group and a multi-spiro heterocyclic group (e.g., a bis-spiro heterocyclic group, etc.) according to the number of spiro atoms shared between rings, and preferably into a mono-spiro heterocyclic group and a bis-spiro heterocyclic group. 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/3-membered, 5-membered/4-membered, 5-membered/5-membered, 5-membered/6-membered, 5-membered/7-membered, 6-membered/3-membered, 6-membered/5-membered, 6-membered/6-membered, 6-membered/7-membered, 7-membered/5-membered or 7-membered/6-membered spiroheterocyclic group. Non-limiting examples of spiro heterocyclyl groups include:
Figure BDA0003653591470000401
the term "fused heterocyclyl" refers to 5 to 20 membered (i.e. 5 to 20 membered fused heterocyclyl) polycyclic heterocyclic groups in which each ring in the system shares an adjacent pair of atoms with the other rings in the system, one or more of the rings may contain one or more double bonds in which one or more of the ring atoms is a heteroatom selected from nitrogen, oxygen and sulphur, which may optionally be oxo (i.e. form a sulfoxide or sulfone), the remaining ring atoms being carbon. Preferably 6 to 14 (e.g. 6,7,8,9, 10, 11, 12, 13 and 14) membered (i.e. 6 to 14 fused heterocyclyl), more preferably 7 to 10 (e.g. 7,8,9 or 10) membered (i.e. 7 to 10 fused heterocyclyl). They can be divided into bicyclic and polycyclic (e.g., tricyclic, tetracyclic, etc.) fused heterocyclic groups according to the number of constituting rings, and are 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/3-membered, 5-membered/5-membered, 5-membered/6-membered, 5-membered/7-membered, 6-membered/3-membered, 6-membered/4-membered, 6-membered/5-membered, 6-membered/6-membered, 6-membered/7-membered, 7-membered/5-membered or 7-membered/6-membered bicyclic fused heterocyclic groups. Non-limiting examples of fused heterocyclic groups include:
Figure BDA0003653591470000402
the term "bridged heterocyclyl" refers to a 5 to 20 membered (i.e., 5 to 20 membered bridged heterocyclyl) polycyclic heterocyclic group in which 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 a heteroatom selected from nitrogen, oxygen, and sulfur, which may optionally be oxo (i.e., to form a sulfoxide or sulfone), and the remaining ring atoms are carbon. Preferably 6 to 14 (e.g. 6,7,8,9, 10, 11, 12, 13 and 14) membered (i.e. 6 to 14 bridged heterocyclyl), more preferably 7 to 10 (e.g. 7,8,9 or 10) membered (i.e. 7 to 10 bridged heterocyclyl). They may be classified into bicyclic and polycyclic (e.g., tricyclic, tetracyclic, etc.) bridged heterocyclic groups according to the number of constituent rings, and are preferably bicyclic, tricyclic or tetracyclic, and more preferably bicyclic or tricyclic. Non-limiting examples of bridged heterocyclic groups include:
Figure BDA0003653591470000411
the heterocyclyl ring includes a 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 a heterocyclyl, non-limiting examples of which include:
Figure BDA0003653591470000412
and the like.
The heterocyclyl group may be substituted or unsubstituted and, when substituted, may be substituted at any available point of attachment, the substituents preferably being selected from one or more of halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and 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 (i.e., a 6 to 14 membered aryl group) having a conjugated pi-electron system, preferably a 6 to 10 membered (i.e., 6 to 10 membered aryl group), such as phenyl and naphthyl. Such aryl rings include those wherein the aryl ring is fused to a heteroaryl, heterocyclyl or cycloalkyl ring as described above, wherein the ring to which the parent structure is attached is an aryl ring, non-limiting examples of which include:
Figure BDA0003653591470000413
aryl groups may be substituted or unsubstituted and, when substituted, may be substituted at any available point of attachment, the substituents preferably being selected from one or more of halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
The term "heteroaryl" refers to a heteroaromatic system (i.e., 5-to 14-membered heteroaryl) 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) (i.e., 5 to 10-membered heteroaryl), further preferably 8 to 10-membered (e.g., 8,9 or 10-membered), more preferably 5-or 6-membered (i.e., 5-or 6-membered heteroaryl), such as furyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, etc. 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 BDA0003653591470000421
heteroaryl groups may be substituted or unsubstituted, and when substituted, may be substituted at any available point of attachment, with the substituents preferably being selected from one or more of halogen, alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyloxy, heterocyclyloxy, hydroxy, hydroxyalkyl, cyano, amino, nitro, cycloalkyl, heterocyclyl, aryl and heteroaryl.
The above-mentioned cycloalkyl, heterocyclyl, aryl and heteroaryl groups include those derived by removal of one hydrogen atom from the parent ring atom, or those derived by removal of two hydrogen atoms from the parent ring atom or two different ring atoms, i.e., "divalent cycloalkyl", "divalent heterocyclyl", "arylene" and "heteroarylene".
The term "amino protecting group" is intended to protect an amino group with a group that can be easily removed in order to keep the amino group unchanged when the reaction is carried out elsewhere in the molecule. Non-limiting examples include (trimethylsilyl) ethoxymethyl, tetrahydropyranyl, t-butyloxycarbonyl (Boc), acetyl, benzyl, allyl, and p-methoxybenzyl, and the like. These groups may be optionally substituted with 1 to 3 substituents selected from halogen, alkoxy and nitro. Preferably, the amino protecting agent is Boc.
The term "hydroxyl protecting group" refers to an easily removable group introduced at a hydroxyl group for blocking or protecting the hydroxyl group while performing a reaction on other functional groups of a compound. Non-limiting examples include: trimethylsilyl (TMS), triethylsilyl (TES), triisopropylsilyl (TIPS), tert-butyldimethylsilyl (TBS), tert-butyldiphenylsilyl (TBDPS), methyl, tert-butyl, allyl, benzyl, methoxymethyl (MOM), ethoxyethyl, 2-Tetrahydropyranyl (THP), formyl, acetyl, benzoyl, p-nitrobenzoyl and the like. Preferably, the hydroxyl protecting group is MOM.
The term "cycloalkyloxy" refers to cycloalkyl-O-, wherein cycloalkyl is as defined above.
The term "heterocyclyloxy" refers to the 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.
Bn is benzyl.
MOM refers to oxymethyl.
Boc means t-butyloxycarbonyl.
TIPS refers to triisopropylsilyl groups.
The compounds and intermediates of the present disclosure may also exist in different tautomeric forms, and all such forms are included within the scope of the present disclosure. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies that can interconvert via a low energy barrier. For example, proton tautomers (also known as proton transfer tautomers) include interconversion via proton migration, such as keto-enol and imine-enamine, lactam-lactim isomerization. An example of a keto-enol equilibrium is between A and B as shown below.
Figure BDA0003653591470000441
All tautomeric forms are within the scope of the disclosure. The naming of the compounds does not exclude any tautomers.
The disclosed compounds may exist in specific stereoisomeric forms. The term "stereoisomers" refers to isomers that are identical in structure but differ in the arrangement of the atoms in space. It includes cis and trans (or Z and E) isomers, (-) -and (+) -isomers, (R) -and (S) -enantiomers, diastereomers, (D) -and (L) -isomers, tautomers, atropisomers, conformers, and mixtures thereof (e.g., racemates, mixtures of diastereomers). Additional asymmetric atoms may be present in substituents in the compounds of the present disclosure. All such stereoisomers, as well as mixtures thereof, are included within the scope of the present disclosure. Optically active (-) -and (+) -isomers, (R) -and (S) -enantiomers, and (D) -and (L) -isomers can be prepared by chiral synthesis, chiral reagents, or other conventional techniques. One isomer of a compound of the present disclosure may be prepared by asymmetric synthesis or chiral auxiliary, or, when a basic functional group (e.g., amino) or an acidic functional group (e.g., carboxyl) is contained in the molecule, a diastereoisomeric salt is formed with an appropriate optically active acid or base, and then diastereoisomeric resolution is performed by a conventional method known in the art to obtain pure isomers. Furthermore, separation of enantiomers and diastereomers is typically accomplished by chromatography.
In the chemical structure of the compounds described in the present disclosure, a bond
Figure BDA0003653591470000442
Denotes an unspecified configuration, i.e. a bond if a chiral isomer is present in the chemical structure
Figure BDA0003653591470000443
Can be that
Figure BDA0003653591470000444
Or
Figure BDA0003653591470000445
Or at the same time contain
Figure BDA0003653591470000446
And
Figure BDA0003653591470000447
two configurations. For all carbon-carbon double bonds, both Z-and E-forms are included, even if only one configuration is named.
The compounds of the present disclosure include all suitable isotopic derivatives of the compounds thereof. The term "isotopic derivative" refers to a compound in which at least one atom is replaced by an atom having the same atomic number but a different atomic mass. Examples of isotopes that can be incorporated into compounds of the present disclosure include stable and radioactive isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, bromine, and iodine, and the like, for example, respectively 2 H (deuterium, D), 3 H (tritium, T), 11 C、 13 C、 14 C、 15 N、 17 O、 18 O、 32 p、 33 p、 33 S、 34 S、 35 S、 36 S、 18 F、 36 Cl、 82 Br、 123 I、 124 I、 125 I、 129 I and 131 i and the like, preferably deuterium.
Compared with the non-deuterated drugs, the deuterated drugs have the advantages of reducing toxic and side effects, increasing the stability of the drugs, enhancing the curative effect, prolonging the biological half-life of the drugs and the like. All isotopic variations of the compounds of the present disclosure, whether radioactive or not, are intended to be encompassed within the scope of the present disclosure. Each available hydrogen atom attached to a carbon atom may be independently replaced by a deuterium atom, where replacement of deuterium may be partial or complete, and replacement of partial deuterium means replacement of at least one hydrogen by at least one deuterium.
When a position is specifically designated as deuterium D, that position is understood to have a natural meaning greater than deuteriumDeuterium at an abundance of at least 1000 times its abundance (i.e. at least 15% deuterium incorporation) at 0.015%. The compounds of examples may have a natural abundance of deuterium greater than at least 1000 times the abundance of deuterium (i.e., at least 15% deuterium incorporation), at least 2000 times the abundance of deuterium (i.e., at least 30% deuterium incorporation), at least 3000 times the abundance of deuterium (i.e., at least 45% deuterium incorporation), at least 3340 times the abundance of deuterium (i.e., at least 50.1% deuterium incorporation), at least 3500 times the abundance of deuterium (i.e., at least 52.5% deuterium incorporation), at least 4000 times the abundance of deuterium (i.e., at least 60% deuterium incorporation), at least 4500 times the abundance of deuterium (i.e., at least 67.5% deuterium incorporation), at least 5000 times the abundance of deuterium (i.e., at least 75% deuterium incorporation), at least 5500 times the abundance of deuterium (i.e., at least 82.5% deuterium incorporation), at least 6000 times the abundance of deuterium (i.e., at least 90% deuterium incorporation), at least 6333.3 times the abundance of deuterium (i.e., at least 95% deuterium incorporation), at least 6466.7 times the abundance of deuterium (i.e., at least 97% deuterium incorporation), at least 99.e., at least 99.99% deuterium incorporation of deuterium, or more than 99.99.99.99.3.99% deuterium incorporation of deuterium. "optionally" 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 "optionally halogen-or cyano-substituted C 1-6 Alkyl "means that halogen or cyano may, but need not, be present, and the description includes the case where alkyl is substituted with halogen or cyano and the case where alkyl is not substituted with halogen or cyano.
"substituted" means that one or more, preferably 1 to 6, more preferably 1 to 3, hydrogen atoms in a group are independently substituted with a corresponding number of substituents. Those skilled in the art are able to determine (experimentally or theoretically) possible or impossible substitutions without undue effort. 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 pharmaceutically acceptable salt or prodrug thereof, in admixture with other chemical components, as well as other components such as 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.
"pharmaceutically acceptable salt" refers to a salt of a compound of the disclosure, which may be selected from inorganic or organic salts. The salt has safety and effectiveness when being used in the body of a mammal, and has due biological activity. Can be prepared separately during the final isolation and purification of the compound, 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 an amount of drug or agent sufficient to achieve, or at least partially achieve, the desired effect. The determination of a therapeutically 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 therapeutically 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 refers to those compounds, materials, compositions, and/or dosage forms which 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 present disclosure
In order to achieve the purpose of the present disclosure, the present disclosure adopts the following technical solutions:
scheme one
The invention discloses a preparation method of a compound shown in a general formula (I) or a pharmaceutically acceptable salt thereof, which comprises the following steps:
Figure BDA0003653591470000461
carrying out deprotection reaction on the compound of the general formula (IA) or salt thereof under an acidic condition to obtain a compound of the general formula (I) or pharmaceutically acceptable salt thereof; optionally, when R 3 And/or R 6 When a protecting group is present on the group, the deprotection reaction may further comprise removing R under acidic or basic conditions before, simultaneously with or after the deprotection reaction 3 And/or R 6 A step of protecting groups on the group; preferably, the protecting group is a hydroxyl protecting group MOM;
wherein R is an amino protecting group; preferably Boc;
G 2 is NH;
ring A, ring B, L, R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, q, r and t are as defined in formula (I).
Scheme two
A process for producing a compound represented by the general formula (I-1) or a pharmaceutically acceptable salt thereof, which comprises:
Figure BDA0003653591470000471
R 0 is a hydrogen atom or a hydroxyl protecting group; the hydroxyl protecting group is preferably MOM;
when R is 0 When the hydrogen atom is a hydrogen atom, removing the amino protecting group R from the compound of the general formula (I-1A) or a salt thereof under acidic conditions to obtain a compound of the general formula (I-1) or a pharmaceutically acceptable salt thereof;
when R is 0 In the case of a hydroxyl-protecting group, the compound of the formula (I-1A) or a salt thereof is freed from the hydroxyl-protecting group R under acidic conditions 0 And an amino protecting group R to give a compound of the general formula (I-1) or a pharmaceutically acceptable salt thereof;
wherein the content of the first and second substances,
r is an amino protecting group; preferably Boc;
G 2 is NH;
ring A, L, R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, q, r and t are as defined in the general formula (I-1).
Scheme three
A process for preparing a compound of the general formula (II) or a pharmaceutically acceptable salt thereof, which comprises:
Figure BDA0003653591470000472
carrying out deprotection reaction on the compound of the general formula (IIA) or salt thereof under an acidic condition to obtain a compound of a general formula (II) or pharmaceutically acceptable salt thereof; optionally, when R 3 And/or R 6 When the group contains a protecting group, the deprotection reaction may be preceded, accompanied or followed by the removal of R under acidic or basic conditions 3 And/or R 6 A step of protecting groups on the group; preferably, the protecting group is a hydroxyl protecting group MOM;
wherein R is an amino protecting group; preferably Boc;
G 2 is NH;
L、R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 m, n, p, q, r and t are as defined in formula (II).
Reagents that provide acidic conditions in the above synthetic schemes include organic and inorganic acids including, but not limited to, trifluoroacetic acid, formic acid, acetic acid, methanesulfonic acid, p-toluenesulfonic acid, me 3 SiCl and TMSOTf; the inorganic acids include, but are not limited to, hydrogen chloride, hydrochloric acid dioxane solution, hydrochloric acid, sulfuric acid, nitric acid, and phosphoric acid; preferably dioxane hydrochloride solution.
The reagents in the above synthesis schemes 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, tetrabutylammonium fluoride, tetrahydrofuran solution of tetrabutylammonium fluoride or 1, 8-diazabicycloundec-7-ene, and inorganic bases including but not limited to sodium hydride, potassium phosphate, sodium carbonate, sodium acetate, potassium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, cesium fluoride and potassium hydroxide.
In the above synthetic scheme, when R 3 And/or R 6 When the terminal alkyne group is contained, the terminal alkyne can be protected by TIPS, and the reagent for removing the TIPS is preferably tetrabutylammonium fluoride in tetrahydrofuran or cesium fluoride.
The reaction of the above step is preferably carried out in a solvent including, but not limited to: pyridine, ethylene glycol dimethyl ether, acetic acid, methanol, ethanol, acetonitrile, N-butanol, toluene, tetrahydrofuran, dichloromethane, petroleum ether, ethyl acetate, N-hexane, dimethyl sulfoxide, 1, 4-dioxane, water, N-dimethylformamide, N-dimethylacetamide, 1, 2-dibromoethane, and mixtures 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 using Bruker AVANCE-400 NMR spectrometer or Bruker AVANCE NEO 500M in deuterated dimethyl sulfoxide (DMSO-d) 6 ) Deuterated chloroform (CDCl) 3 ) Deuterated methanol (CD) 3 OD), internal standard Tetramethylsilane (TMS).
MS was measured using an Agilent 1200/1290DAD-6110/6120Quadrupole MS liquid chromatograph-Mass spectrometer (manufacturer: agilent, MS model: 6110/6120Quadrupole MS).
waters ACQuity UPLC-QD/SQD (manufacturer: waters, MS model: waters ACQuity Qda Detector/waters SQ Detector)
THERMO Ultimate 3000-Q active (manufacturer: THERMO, MS model: THERMO QOxctive)
High Performance Liquid Chromatography (HPLC) analysis was performed using Agilent HPLC1200 DAD, agilent HPLC1200VWD and Waters HPLC e2695-2489 liquid chromatographs.
Chiral HPLC analytical determination Agilent 1260DAD HPLC was used.
High Performance liquid preparation preparative chromatographs were prepared using Waters 2545-2767, waters 2767-SQ Detector 2, shimadzu LC-20AP and Gilson GX-281.
Chiral preparation a Shimadzu LC-20AP preparative chromatograph was used.
The CombiFlash 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 of the Litsea crassirhizomes 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 for the present invention can be synthesized using or according to methods known in the art, or can be purchased from companies such as ABCR GmbH & Co. KG, acros Organics, aldrich Chemical Company, shaoyuan ChemBiotech (Accela ChemBio Inc), darril Chemicals, and the like.
In the examples, the reaction can be carried out under 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 is usually carried out by vacuum pumping, hydrogen filling and repeated operation for 3 times.
A CEM Discover-S908860 type microwave reactor was used for the microwave reaction.
In the examples, the solution means an aqueous solution unless otherwise specified.
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: in a dichloromethane/methanol 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 also be added for adjustment.
Example 1
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-fluoronaphthalen-2-ol 1
Figure BDA0003653591470000501
First step of
3- (benzyloxy) -1-bromo-8-fluoronaphthalene 1b
After the compound 4-bromo-5-fluoronaphthalene-2-ol 1a (180mg, 746.7. Mu. Mol, from Shanghai), benzyl bromide (127mg, 742.5. Mu. Mol) was dissolved in 10mL of acetonitrile, anhydrous potassium carbonate (310mg, 2.24mmol) was added, and the reaction mixture was stirred at 55 ℃ for 1 hour, the reaction mixture was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system A to give the title compound 1b (182 mg, yield 73.5%).
MS m/z(ESI):331.2[M+1]。
Second step of
(1R, 5S) -3- (7-benzyl-2-chloro-4a, 5,6,7,8, 8a-hexahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 1d
The compound 7-benzyl-2, 4-dichloro-5, 6,7, 8-tetrahydropyrido [3,4-d ] pyrimidine 1c (1g, 3.4mmol, prepared by the method disclosed in j.med.chem.2020,63, 6679-6693), N-diisopropylethylamine (1.1g, 8.5 mmol) was dissolved in 25mL of dimethyl sulfoxide, tert-butyl (1r, 5s) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate (760mg, 3.58mmol, jiangsu aikang) was added thereto, after stirring at 55 ℃ for 10 hours, 100mL of water was added to the reaction solution, ethyl acetate was extracted (30 mL × 3), the organic phases were combined, anhydrous sodium sulfate was dried, the filtrate was concentrated under reduced pressure after removing the drying agent by filtration, and the residue was purified by silica gel column chromatography to obtain the title compound 1d (1.28 g, yield 80.1%) using silica eluent system a.
MS m/z(ESI):472.2[M+1]。
The third step
(1R, 5S) -3- (7-benzyl-2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 1f
Compound 1d (200mg, 425.5. Mu. Mol), compound ((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methanol 1e (86.3mg, 542.6. Mu. Mol, phentermine) were dissolved in toluene (5 mL), palladium acetate (111mg, 48.9. Mu. Mol), 1 '-binaphthyl-2, 2' -bisdiphenylphosphine (54mg, 86.7. Mu. Mol), cesium carbonate (400mg, 1.22mmol) were added, heated to 110 ℃ for 6 hours, the reaction was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system A to give the title compound 1f (220 mg, yield: 87.2%).
MS m/z(ESI):593.2[M+1]。
The fourth step
(1R, 5S) -3- (2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 1g
Compound 1f (268mg, 370.7. Mu. Mol) was dissolved in 10mL of methanol, and ammonium acetate (142mg, 1.84mmol), 10% palladium hydroxide (wet) (260 mg) were added, and the mixture was replaced with hydrogen gas three times, followed by stirring at 70 ℃ for 2 hours. The reaction solution was cooled to room temperature, filtered through Celite, and the filtrate was concentrated to give the title compound 1g (160 mg, yield: 85.6%) which was used in the next reaction without purification.
MS m/z(ESI):503.2[M+1]
The fifth step
(1R, 5S) -3- (7- (3- (benzyloxy) -8-fluoronaphthalen-1-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 1H
1g (83mg, 132.1. Mu. Mol) of the crude compound, 1b (40mg, 120.7. Mu. Mol), methanesulfonic acid (2-dicyclohexylphosphino-2 ',6' -diisopropoxy-1, 1 '-biphenyl) (2-amino-1, 1' -biphenyl-2-yl) palladium (II) (20mg, 23.9. Mu. Mol, shanghai Shao), and anhydrous cesium carbonate (118mg, 362.1. Mu. Mol) were dissolved in 3mL of 1, 4-dioxane. The reaction was carried out at 110 ℃ for 14 hours under a nitrogen atmosphere, the reaction mixture was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system A to give the title compound 1h (89 mg, yield: 97.8%). MS m/z (ESI) 753.2[ 2 ], [ M +1].
The sixth step
(1R, 5S) -3- (7- (8-fluoro-3-hydroxynaphthalen-1-yl) -2- ((((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 1i
Compound 1h (89mg, 118.2. Mu. Mol) was dissolved in 3mL of methanol, 10% palladium hydroxide (wet) (80 mg) was added, hydrogen gas was replaced three times, and the mixture was stirred at 40 ℃ for 14 hours. After cooling, it was filtered through Celite, and the filtrate was concentrated to give the title compound 1i (78 mg, yield: 99.5%).
MS m/z(ESI):663.2[M+1]。
Seventh step
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-fluoronaphthalen-2-ol 1
Compound 1i (78mg, 117.8. Mu. Mol) was dissolved in acetonitrile (1 mL), 1mL of 4M dioxane hydrochloride was added, the reaction was stirred for 2 hours, and the reaction mixture was concentrated under reduced pressure and purified by high performance liquid chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm, 5. Mu.m; mobile phase: aqueous phase (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient: acetonitrile 35% -45%, flow rate: 30 mL/min) to give the title compound 1 (3 mg, yield: 4.5%).
MS m/z(ESI):563.2[M+1]。
1 H NMR(500MHz,CDCl 3 ):δ7.42(d,1H),7.31(d,1H),6.91(dd,1H),6.86(s,1H),6.73(s,1H),4.26-3.33(m,12H),3.32-2.98(m,5H),2.39-2.14(m,4H),2.10-1.73(m,8H)。
Example 2
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-ethylnaphthalene-2-ol
Figure BDA0003653591470000531
First step of
(1R, 5S) -3- (7- (8-Ethyl-3- (methoxymethyloxy) naphthalen-1-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 2b
1g (110mg, 197.6. Mu. Mol), the compound 8-ethyl-3- (methoxymethoxy) naphthalen-1-yl trifluoromethanesulfonate 2a (60mg, 164.6. Mu. Mol, prepared by the method disclosed in patent application "WO2021/041671A1" on page 111, intermediate 21), methanesulfonic acid (2-dicyclohexylphosphino-2 ',6' -diisopropoxy-1, 1 '-biphenyl) (2-amino-1, 1' -biphenyl-2-yl) palladium (II) (27.5mg, 32.9. Mu. Mol, shanghai Shao), cesium carbonate anhydrous (161mg, 494. Mu. Mol) was dissolved in 3mL of 1, 4-dioxane. The reaction was carried out at 110 ℃ for 14 hours under a nitrogen atmosphere, the reaction mixture was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system A to give the title compound 2b (36 mg, yield: 30.5%).
MS m/z(ESI):717.2[M+1]。
Second step of
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-ethylnaphthalene-2-ol
Compound 2b (19mg, 26.5. Mu. Mol) was dissolved in acetonitrile (1 mL), 0.5mL of 4M dioxane hydrochloride solution was added, the reaction was stirred for 2 hours, and the reaction mixture was concentrated under reduced pressure and purified by high performance liquid chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm, 5. Mu.m; mobile phase: aqueous phase (10 mmol/L ammonium hydrogencarbonate) and acetonitrile, gradient ratio: acetonitrile 35% -45%, flow rate: 30 mL/min) to give the title compound 2 (1.18 mg, yield: 7.7%).
MS m/z(ESI):573.2[M+1]。
1 H NMR(500MHz,CDCl 3 ):δ7.47-7.45(m,1H),7.32(t,1H),7.18(dd,1H),6.93(t,1H),6.46(d,1H),4.87-4.75(m,1H),4.67(d,2H),4.50-4.43(m,1H),4.35-4.29(m,1H),3.80(dt,2H),3.73(dd,2H),3.64(dd,2H),3.29-3.10(m,6H),2.96(qd,2H),2.92-2.86(m,1H),2.86-2.77(m,1H),2.16-2.04(m,1H),2.02-1.64(m,9H),1.33(t,3H)。
Example 3
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-ethynylnaphthalen-2-ol 3
Figure BDA0003653591470000541
Using the synthetic route described in example 2, the starting compound 2a was replaced with the compound 3- (methoxymethoxy) -8- (triisopropylsilyl) ethynyl) naphthalen-1-yl trifluoromethanesulfonate (prepared using the method disclosed in patent application "WO2021041671A1" at page 103 intermedate 17) to give the title compound 3.
Example 4
4- (4 ' - ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2' - ((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5',8' -dihydro-7 ' H-spiro [ cyclopropane-1, 6' -pyrido [3,4-d ] pyrimidin-7 ' -yl) -5-fluoronaphthalene-2-ol 4-ol
Figure BDA0003653591470000542
Example 5
4- (4 ' - ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2' - (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5',6' -dihydro-7 ' H-spiro [ cyclopropane-1, 8' -pyrido [3,4-d ] pyrimidin-7 ' -yl) -5-fluoronaphthalene-2-ol 5-ol
Figure BDA0003653591470000551
Example 6
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 9-tetrahydro-8H-pyrimido [4,5-c ] azepin-8-yl) -5-fluoronaphthalen-2-ol 6
Figure BDA0003653591470000552
First step of
5- (benzyl (2-ethoxy-2-oxoethyl) amino) pentanoic acid ethyl ester 6b
The compound ethyl N-benzylglycine 6a (23g, 119.02mmol, obtained from shanghai, pee), the compound ethyl 5-bromovalerate (25g, 119.57mmol), triethylamine (18.06g, 178.47mmol) were dissolved in acetonitrile (200 mL), refluxed at 80 ℃ overnight, the reaction was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system B to give the title compound 6B (24 g), yield: 62.7 percent.
MS m/z(ESI):322.2[M+1]。
Second step of
1-benzyl-3-oxoazepane-4-carboxylic acid ethyl ester 6c
Compound 6b (17g, 52.89mmol) was dissolved in tetrahydrofuran (200 mL), sodium hydrogen (3.59g, 89.9mmol,60% pure) was added under ice bath, heated to 70 ℃ and stirred for 14 hours, the reaction solution was cooled to room temperature, quenched by addition of saturated aqueous ammonium chloride solution, extracted with ethyl acetate (50 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, the filtrate was concentrated under reduced pressure after filtration to remove the drying agent, and the residue was purified by silica gel column chromatography with eluent System A to give the title compound 6c (5 g), yield: 34.3 percent.
MS m/z(ESI):276.2[M+1]。
The third step
8-benzyl-6, 7,8, 9-tetrahydro-5H-pyrimido [4,5-c ] azepin-2, 4-diol 6d
Compound 6c (1g, 3.63mmol), urea (654.33mg, 10.89mmol), and sodium methoxide (588.61mg, 10.89mmol) were dissolved in methanol (10 mL), and the mixture was refluxed for 16 hours. The reaction was cooled to room temperature and concentrated under reduced pressure to give crude title compound 6d (2.5 g), which was used in the next reaction without purification.
MS m/z(ESI):272.2[M+1]。
The fourth step
8-benzyl-2, 4-dichloro-6, 7,8, 9-tetrahydro-5H-pyrimido [4,5-c ] azepin 6e
Crude compound 6d (1g, 3.68mmol), N-diisopropylethylamine (1.90g, 14.70mmol), phosphorus oxychloride (15 mL) were mixed, heated to 90 ℃ for 6 hours, the reaction solution was concentrated under reduced pressure, diluted with dichloromethane, adjusted to pH 7-8 with saturated sodium bicarbonate solution, dried over anhydrous sodium sulfate, the filtrate was concentrated under reduced pressure after removal of the drying agent by filtration, and the residue was purified by silica gel column chromatography with eluent system a to give the title compound 6e (0.3 g), yield: 26.4 percent.
MS m/z(ESI):308.1[M+1]。
The fifth step
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 9-tetrahydro-8H-pyrimido [4,5-c ] azepin-8-yl) -5-fluoronaphthalen-2-ol 6
Using the synthetic route in example 1, the second step starting material compound 1c was replaced with compound 6e to give the title compound 6 (2 mg, yield: 23.4%).
MS m/z(ESI):577.2[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.40(d,1H),7.27-7.24(m,1H),6.81(dd,2H),6.77-6.73(m,1H),4.27-4.25(m,1H),4.22-4.07(m,2H),3.76(s,2H),3.44(t,2H),3.33-3.05(m,2H),3.03-3.02(m,1H),2.41-1.75(m,12H),1.66-1.60(m,1H),1.37-1.3(m,8H)。
Example 7
4- (4- (3, 9-diazabicyclo [3.3.1] non-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-ethylnaphthalene-2-ol 7
Figure BDA0003653591470000571
First step of
3- (2- (((2R, 7aS) -2-Fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 9-diazabicyclo [3.3.1] nonane-9-carboxylic acid tert-butyl ester 7a
Using the second to fourth steps of the synthetic route in example 1, the starting material compound (1R, 5S) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester in the second step was replaced with the compound 3, 9-diazabicyclo [3.3.1] nonane-9-carboxylic acid tert-butyl ester to give the title compound 7a (100 mg, yield: 30%)
MS m/z(ESI):517.2[M+1]。
Second step of
3- (7- (8-Ethyl-3- (methoxymethyloxy) naphthalen-1-yl) -2- (((2R, 7aS) -2-Fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 9-diazabicyclo [3.3.1] nonane-9-carboxylic acid tert-butyl ester 7b
Compound 7a (100mg, 174.1. Mu. Mol), compound 2a (63.46mg, 174.1. Mu. Mol) were dissolved in 3mL of dioxane, sulfonic acid (2-dicyclohexylphosphino-2 ',6' -diisopropoxy-1, 1 '-biphenyl) (2-amino-1, 1' -biphenyl-2-yl) palladium (II) (29mg, 34.8. Mu. Mol, shanghaitan), cesium carbonate (170mg, 522.5. Mu. Mol) was added, replaced with nitrogen, and the reaction was stirred at 100 ℃ for 5 hours. The reaction solution was cooled to room temperature and then filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system A to give the title compound 7b (40 mg, yield: 31.4%).
MS m/z(ESI):731.2[M+1]。
The third step
4- (4- (3, 9-diazabicyclo [3.3.1] non-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-ethylnaphthalene-2-ol 7
Compound 7b (40mg, 54.7. Mu. Mol) was dissolved in 1mL acetonitrile, 1mL of 4M dioxane hydrochloride solution was added, the reaction was stirred for 2 hours and concentrated under reduced pressure, and the residue was purified by high performance liquid chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm, 5. Mu.m; mobile phase: aqueous phase (10 mmol/L ammonium bicarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -45%, flow rate: 30 mL/min) to give the title compound 7 (1.3 mg, yield: 4%).
MS m/z(ESI):587.3[M+1]。
1 H NMR(500MHz,CDCl 3 ):δ7.54-7.48(m,1H),7.36(d,1H),7.11(d,1H),6.98(s,1H),6.85(d,1H),4.29(dd,2H),4.20(dd,2H),4.16-4.05(m,2H),3.97(d,2H),3.85(d,2H),3.50(d,2H),3.39-3.14(m,6H),3.00(s,3H),2.17(d,2H),2.11-1.86(m,10H),1.15(t,3H)。
Example 8
3- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-chloro-4-cyclopropylphenol 8
Figure BDA0003653591470000581
Figure BDA0003653591470000591
First step of
(1R, 5S) -3- (7- (3-chloro-2-cyclopropyl-5- (methoxymethyloxy) phenyl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 8b
The compound 1g (110mg, 218.8. Mu. Mol), 1-bromo-3-chloro-2-cyclopropyl-5- (methoxymethoxy) benzene 8a (95.7mg, 328.2. Mu. Mol, prepared by the method disclosed in Exampe 283 on page 526 of the specification of patent application "WO 2021/041671") was dissolved in 5mL dioxane, sulfonic acid (2-dicyclohexylphosphino-2 ',6' -diisopropoxy-1, 1 '-biphenyl) (2-amino-1, 1' -biphenyl-2-yl) palladium (II) (91.5mg, 109. Mu. Mol), cesium carbonate (142mg, 437mol) were added, replaced with nitrogen, and the reaction was stirred at 100 ℃ for 5 hours. The reaction solution was cooled to room temperature and then filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system A to give the title compound 8b (20 mg, yield: 12%).
MS m/z(ESI):713.2[M+1]。
Second step of
3- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-chloro-4-cyclopropylphenol 8
Compound 8b (20mg, 28. Mu. Mol) was dissolved in 2mL of methylene chloride, and 1mL of 4M dioxane hydrochloride solution was added, and after stirring to react for 1 hour, concentration was performed under reduced pressure, and the residue was purified by high performance liquid chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm, 5. Mu.m; mobile phase: aqueous phase (10 mmol/L ammonium hydrogencarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -45%, flow rate: 30 mL/min) to give the title compound 8 (15 mg, yield: 93%).
MS m/z(ESI):569.2[M+1]。
1 H NMR(500MHz,CD 3 OD):δ6.55(d,1H),6.41(d,1H),5.30(dd,1H),4.17(d,1H),4.11-3.98(m,5H),3.61(s,2H),3.31-3.18(m,7H),3.02(td,1H),2.83(t,2H),2.34-2.24(m,1H),2.23-2.04(m,3H),2.00(dtt,2H),1.91-1.84(m,4H),1.67(td,1H),1.05-0.98(m,2H),0.69(dt,2H)。
Example 9
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-ethynyl-6-fluoronaphthalen-2-ol 9-ol
Figure BDA0003653591470000601
Using the synthetic route described in example 2, the starting compound 2a was replaced with the compound 7-fluoro-3- (methoxymethyloxy) -8- (triisopropylsilyl) ethynyl) naphthalen-1-yl trifluoromethanesulfonate (prepared by the method disclosed in the patent application "WO2021041671A1" at page 96, intermedate 15) to give the title compound 9.
Example 10
4- (4 '- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2' - (((2R, 7As) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -6 'H-spiro [ cyclopropane-1, 5' -pyrido [3,4-d ] pyrimidine ] -7 '(8' H) -yl) -5-fluoronaphthalene
-2-phenol 10
Figure BDA0003653591470000602
Example 11
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5, 6-difluoronaphthalene-2-ol 11
Figure BDA0003653591470000611
First step of
(1R, 5S) -3- (7, 8-difluoro-3- (methoxymethyloxy) naphthalen-1-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methyloxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 11b
The compound 1g (200mg,537.2. Mu. Mol), 7,8-difluoro-3- (methoxymethyloxy) naphthalen-1-yl trifluoromethanesulfonate 11a (200mg,537.2. Mu. Mol, prepared by the method disclosed in Example 246 on page 437 of the specification of the patent application "WO 2021/041671") was dissolved in 5mL of dioxane, sulfonic acid (2-dicyclohexylphosphino-2 ',6' -diisopropoxy-1, 1 '-biphenyl) (2-amino-1, 1' -biphenyl-2-yl) palladium (II) (89.9mg,107.4. Mu. Mol, shanghaitan), cesium carbonate (525mg,1.6 mol) was added, replaced with nitrogen, and the reaction was stirred at 100 ℃ for 5 hours. The reaction solution was cooled to room temperature and then filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system A to give the title compound 11b (180 mg, yield: 46.2%).
MS m/z(ESI):725.2[M+1]。
Second step of
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5, 6-difluoronaphthalene-2-ol 11
Compound 11b (56mg, 77.3. Mu. Mol) was dissolved in 2mL of acetonitrile, 1mL of 4M dioxane hydrochloride solution was added, the reaction was stirred for 2 hours and then concentrated under reduced pressure, and the residue was purified by high performance liquid chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm, 5. Mu.m; mobile phase: aqueous phase (10 mmol/L ammonium hydrogencarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -45%, flow rate: 30 mL/min) to give the title compound 11 (12 mg, yield: 26.7%).
MS m/z(ESI):581.2[M+1]。
1 H NMR(500MHz,CD 3 OD):δ7.46(dd,1H),7.33(q,1H),6.92-6.80(m,2H),4.21(s,2H),4.15(d,2H),3.99(d,2H),3.83(s,2H),3.66(d,1H),3.56-3.42(m,2H),3.21-3.11(m,2H),3.11-3.00(m,2H),2.92(s,2H),2.64(s,2H),2.39-2.09(m,6H),2.06-1.96(m,4H)。
Example 12
5- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -4-bromonaphthalene-2-ol 12-p1
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-bromonaphthalene-2-ol 12-p 2-yl
Figure BDA0003653591470000621
First step of
(1R, 5S) -3- (7- (8-bromo-6- (methoxymethyloxy) naphthalen-1-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methyloxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 12b-p1
(1R, 5S) -3- (7- (8-bromo-3- (methoxymethyloxy) naphthalen-1-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methyloxy) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 12b-p2
Compound 1g (600mg, 1.07mmol), 1, 8-dibromo-3- (methoxymethoxy) naphthalene 12a (1.12g, 3.23mmol, prepared by the method disclosed in preparation 4 on page 82 of the specification of "WO202110623A 1") was dissolved in 10mL of toluene, tris (dibenzylideneacetone) dipalladium (197mg, 215. Mu. Mol), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (250mg, 432. Mu. Mol), cesium carbonate (1.75g, 5.37mmol) was added, nitrogen was substituted, and the reaction was stirred at 110 ℃ for 24 hours. The reaction solution was cooled to room temperature and then filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography with eluent system A to give a mixture of the title compounds 12b-p1 and 12b-p2 (600 mg, yield: 72.4%).
MS m/z(ESI):767.2[M+1]。
Second step of
5- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -4-bromonaphthalene-2-ol 12-p1
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-bromonaphthalene-2-ol 12-p2
A mixture of compounds 12b-p1 and 12b-p2 (50mg, 65.1. Mu. Mol) was dissolved in 2mL of acetonitrile and 2mL of methanol, 2mL of 4M dioxane hydrochloride solution was added, the reaction was stirred for 1 hour and then concentrated under reduced pressure, and the residue was purified by high performance liquid chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm, 5. Mu.m; mobile phase: aqueous phase (10 mmol/L ammonium hydrogencarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -45%, flow rate: 30 mL/min) to give the title compounds 12-p1 (3 mg, yield: 7.3%) and 12-p2 (2.5 mg, yield: 6.1%). Single configuration Compound (shorter Retention time) 12-p1 (3 mg, yield: 7.3%)
MS m/z(ESI):623.2[M+1]。
HPLC analysis: retention time 2.45 min, purity: 90% (chromatographic column: ACQUITY)
Figure BDA0003653591470000631
BEH, C18,1.7 μm,2.1 × 50mm; mobile phase: water (10 mM ammonium bicarbonate), acetonitrile, gradient: acetonitrile 10% -95%). 1 H NMR(500MHz,CD 3 OD):δ7.46(d,1H),7.41(d,1H),7.38(t,1H),7.13(d,1H),7.11(dd,1H),5.43-5.28(m,1H),4.27(t,2H),4.16(dd,1H),4.07(dd,1H),3.78(d,1H),3.63(s,2H),3.61-3.55(m,1H),3.52(d,1H),3.41(d,1H),3.25-3.16(m,4H),3.06(d,1H),3.01(dd,1H),2.61(d,1H),2.23-2.17(m,2H),2.13-2.02(m,4H),2.02-1.95(m,2H),1.90(d,3H)。
Single configuration Compound (longer Retention time) 12-p2 (2.5 mg, yield: 6.1%)
MS m/z(ESI):623.2[M+1]。
HPLC analysis: retention time 2.50 min, purity: 90% (chromatographic column: ACQUITY)
Figure BDA0003653591470000632
BEH, C18,1.7 μm,2.1 × 50mm; mobile phase: water (10 mM ammonium bicarbonate), acetonitrile, gradient: acetonitrile 10% -95%). 1 H NMR(500MHz,CD3OD):δ7.63(d,1H),7.55(d,1H),7.15(t,1H),7.00-6.89(m,2H),5.36-5.34(m,1H),4.33-4.22(m,2H),4.16(dd,1H),4.07(dd,1H),3.79(d,1H),3.63(s,2H),3.59(d,1H),3.54(d,1H),3.41(s,1H),3.24(d,2H),3.20-3.15(m,2H),3.07(d,1H),3.00(q,1H),2.62(d,1H),2.22-2.18(m,2H),2.09-2.02(m,4H),1.98(s,2H),1.88(s,3H)。
Example 13
5- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -4-iodonaphthalene-2-ol 13-p1
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-iodonaphthalene-2-ol 13-p2
Figure BDA0003653591470000641
First step of
(1R, 5S) -3- (2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -7- (8-iodo-6- (methoxymethoxy) naphthalen-1-yl) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 13a-p1
(1R, 5S) -3- (2- (((2R, 7aS) -2-Fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -7- (8-iodo-3- (methoxymethoxy) naphthalen-1-yl) -5,6,7, 8-tetrahydropyrido [3,4-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester 13a-p2
A mixture of compounds 12b-p1 and 12b-p2 (300mg, 390.7. Mu. Mol) was dissolved in 1, 4-dioxane, and cuprous iodide (60mg, 315. Mu. Mol), sodium iodide (176mg, 1.17mmol), N, N-diisopropylethylamine (60mg, 680. Mu. Mol) were added, under nitrogen, and the reaction was stirred while heating to 110 ℃ for 24 hours. After the reaction mixture was cooled to room temperature, cuprous iodide (120mg, 630. Mu. Mol), sodium iodide (352mg, 2.34mmol), N, N-diisopropylethylamine (120mg, 1.36mmol) were added thereto, and the mixture was stirred to react for 16 hours, the reaction mixture was brought over, water was added to the filtrate, extraction was performed with ethyl acetate (30 mL. Times.3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered to remove the drying agent, and concentrated under reduced pressure to obtain a mixture (318 mg) of the crude title compounds 13a-p1 and 13a-p2, which was used in the next reaction without purification.
MS m/z(ESI):815.2[M+1]。
Second step of
5- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -4-iodonaphthalene-2-ol 13-p1
4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R, 7aS) -2-fluorotetrahydro-1H-pyrrolizin-7 a (5H) -yl) methoxy) -5, 8-dihydropyrido [3,4-d ] pyrimidin-7 (6H) -yl) -5-iodonaphthalene-2-ol 13-p2
A mixture (50mg, 61.3. Mu. Mol) of crude compounds 13a-p1 and 13a-p2 was dissolved in 2mL of acetonitrile and 2mL of methanol, 2mL of 4M dioxane hydrochloride solution was added, the reaction was stirred for 1 hour and then concentrated under reduced pressure, and the residue was purified by high performance liquid chromatography (Waters-2545, column: sharpSil-T C18, 30X 150mm, 5. Mu.m; mobile phase: aqueous phase (10 mmol/L ammonium hydrogencarbonate) and acetonitrile, gradient ratio: acetonitrile 30% -45%, flow rate: 30 mL/min) to obtain the title compounds 13-p1 (1.6 mg, yield: 3.8%) and 13-p2 (2.2 mg, yield: 5.3%).
Mono-configuration compound (shorter retention time) 13-p1 (1.6 mg, yield: 3.8%)
MS m/z(ESI):671.2[M+1]。
HPLC analysis: retention time 2.42 minutes, purity: 94% (chromatographic column: ACQUITY)
Figure BDA0003653591470000651
BEH, C18,1.7 μm,2.1 × 50mm; mobile phase: water (10 mM ammonium bicarbonate), acetonitrile, gradient: acetonitrile 10% -95%). 1 H NMR(500MHz,CD 3 OD):δ7.84(dd,1H),7.46(d,1H),7.40(t,1H),7.17-7.12(m,2H),5.35(d,1H),4.28(d,1H),4.22-4.14(m,2H),4.13-4.04(m,1H),3.82(d,1H),3.76(s,2H),3.54(d,1H),3.44(d,2H),3.29-3.15(m,4H),3.11(d,1H),3.03(td,1H),2.60(d,1H),2.23-2.17(m,2H),2.14-2.10(m,2H),2.03(d,2H),1.99(d,2H),1.93(d,3H)。
Single configuration Compound (longer Retention time) 13-p2 (2.2 mg, yield: 5.3%)
MS m/z(ESI):671.2[M+1]。
HPLC analysis: retention time 2.50 min, purity: 90% (chromatographic column: ACQUITY)
Figure BDA0003653591470000652
BEH, C18,1.7 μm,2.1 × 50mm; mobile phase: water (10 mM carbon)Ammonium hydrogen acid), acetonitrile, gradient ratio: acetonitrile 10% -95%). 1 H NMR(500MHz,CD 3 OD):δ7.98(dt,1H),7.65(dd,1H),6.99–6.94(m,2H),6.93(d,1H),5.34(dq,1H),4.26(d,1H),4.23-4.13(m,2H),4.08(dd,1H),3.84-3.78(m,1H),3.70(s,2H),3.53(d,1H),3.48(d,1H),3.43-3.39(m,1H),3.28-3.23(m,2H),3.18(ddd,2H),3.10(d,1H),3.01(td,1H),2.60(d,1H),2.24-2.17(m,2H),2.13-2.07(m,2H),2.03(d,1H),1.99-1.97(m,1H),1.92(d,5H)。
Biological evaluation
Test example 1: AGS cell ERK phosphorylation inhibition Experimental biological evaluation (HTRF method)
1. Purpose of testing
This experiment was performed by examining the inhibitory effect of compounds on cell ERK phosphorylation, based on IC 50 The size of the compound disclosed in the disclosure is evaluated on the inhibition effect of the KRAS target.
2. Experimental method
AGS cells (nanjing kebai, CBP 60476) were cultured in RPMI1640 (Hyclone, SH 30809.01) complete medium containing 10% fetal bovine serum. The first day of experiment, AGS cells were seeded in 96-well plates at a density of 40000 cells/well using complete medium, 190. Mu.L cell suspension per well, left at 37 ℃,5% 2 The cell culture box was incubated overnight.
The following day, 10. Mu.L of test compound diluted in a gradient prepared with complete medium was added to each well, the final concentration of the compound was 9 concentration points at which 5-fold gradient dilution was performed from 10. Mu.M, a blank containing 0.5% DMSO was set, the well plate was placed at 37 ℃,5% CO 2 The cell culture chamber of (1) was incubated for 1 hour. After completion of incubation, the 96-well cell culture plate was removed, the medium was aspirated off, and 200. Mu.L of PBS (Shanghai culture Biotech Co., ltd., B320) was added to each well and washed once. PBS was aspirated, 50. Mu.L of lysis buffer (lysis buffer, cisbio,64KL1 FDF) containing blocking solution (blocking reagent, cisbio,64KB1 AAC) was added to each well, and the well plates were placed on a shaker and lysed for 40 minutes at room temperature with shaking. After lysis, the mixture was pipetted and mixed, 16. Mu.L of lysate was transferred to two HTRF 96-well assay plates (Cisbio, 66PL 96100) per well, and then 4. Mu.L of premixed phosphorylated ERK1/2 antibody solution (Cis) was added to each of the platesbio,64 aerreg) or 4 μ L of pre-mixed total ERK1/2 antibody solution (Cisbio, 64 NRKPEG). The plate was sealed with a sealing membrane, centrifuged for 1 min in a microplate centrifuge and incubated overnight at room temperature in the dark.
On the third day, fluorescence values of 337nm wavelength excitation, 665nm and 620nm wavelength emission were read using an ENVISION multifunctional microplate reader (PerkinElmer, ENVISION).
3. Data analysis
IC of inhibitory Activity of Compounds was calculated using Graphpad Prism software based on Compound concentration and ratio of phosphorylated ERK/Total ERK 50 See table 1 below for values, results.
TABLE 1 cellular ERK phosphorylation inhibitory Activity data
Figure BDA0003653591470000661
Figure BDA0003653591470000671
And (4) conclusion: the compound disclosed by the invention has a better inhibition effect on ERK phosphorylation of AGS cells.
Test example 2: SPR method for detecting affinity of disclosed compound and KRAS protein subtype G12D or WT
Biotinylated Avi-KRAS-WT or Avi-KRAS-G12D was first incubated with a medium containing 100mM MgCl 2 The 1 XHBS-P + (Cat. # BR 1006-71) buffer solution is diluted to 20 mu g/mL, then flows through SA ((Cat. # BR 1005-31) biosensing chip channel 2 for 420s to obtain a coupling level of about 5000-7000RU, samples of the small molecular compound are sequentially injected from low to high for 120s, and then are dissociated for 720s, a single-cycle kinetic mode is adopted in the test, a Biacore 8K instrument detects reaction signals in real time to obtain a binding dissociation curve, a Biacore 8K evaluation software is used for data analysis after the test is finished, and a model 1 is adopted for data fitting and affinity data are obtained.
TABLE 2 affinity data of the compounds for KRAS protein subtypes G12D or WT
Figure BDA0003653591470000672
And (4) conclusion: the compounds of the present disclosure have better affinity to KRAS protein subtype G12D or WT.
Test example 3: experimental biological evaluation of 3D proliferation inhibition of GP2D cells
1. Purpose of testing
The inhibition effect of the disclosed compound on the KRAS target spot is evaluated by testing the 3D proliferation inhibition effect of the disclosed compound on GP2D cells.
2. Experimental method
GP2d cells (CBP 60010, nanjing Kebai) were cultured in complete medium, DMEM/high sugar medium (Hyclone, SH 30243.01) containing 10% fetal bovine serum (Corning, 35-076-CV). On the first day of the experiment, GP2d cells were seeded at a density of 1000 cells/well on a 96-well low adsorption plate (Corning, CLS7007-24 EA) using complete medium, 90. Mu.L of cell suspension was centrifuged at 2000rpm for 5 minutes at room temperature and then placed at 37 ℃ for 5% CO 2 The cell culture box was cultured overnight.
The following day, 10 μ L of test compound diluted in a gradient of complete medium was added to each well, and the final concentration of compound in GP2d cells was 9 concentration points diluted in a 3-fold gradient starting from 1 μ M, setting a blank containing 0.5% DMSO. The well plate was left at 37 ℃ 5% CO 2 The cell culture chamber (2) was cultured for 120 hours. On the seventh day, 96 well cell culture plates were removed and 50. Mu.L of each well was added
Figure BDA0003653591470000681
After shaking the 3D reagent (Promega, G9682) at room temperature for 25 minutes, the mixture was aspirated and 50 μ L was transferred to a white opaque 96-well plate (PE, 6005290), and the luminescence signal value was read using a multi-functional microplate reader (PerkinElmer, ENVISION).
3. Data analysis
IC of compound inhibitory activity was calculated using Graphpad Prism software 50 See table 3 below for values, results.
TABLE 3 data on cell 3D proliferation inhibitory Activity
Example numbering GP2d/IC 50 (nM)
1 75.3
2 3.6
8 41.0
11 48.4
13-p2 44.3
And (4) conclusion: the compound disclosed by the invention has a better inhibiting effect on the 3D proliferation of GP2D cells.

Claims (22)

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure FDA0003653591460000011
wherein:
G 2 is NR d
Ring a is aryl or heteroaryl;
ring B is selected from cycloalkyl, heterocyclyl, aryl and heteroaryl;
l is selected from the group consisting of a single bond, O andNR e
each R is 1 Are the same or different and are each independently selected from the group consisting of a hydrogen atom, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, - (CH) 2 ) u -NR f R g Hydroxyl and hydroxyalkyl;
R 2 、R 2c 、R 2a 、R 2b 、R 4a and R 4b Are the same or different and are each independently selected from the group consisting of a hydrogen atom, halogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkyl, haloalkoxy, cyano, amino, - (CH) 2 ) v -NR h R i Hydroxyl, hydroxyalkyl and cycloalkyl; or
R 2 、R 2c To the carbon atom to which it is attached, or R 2a 、R 2b To the carbon atom to which it is attached, or R 4a 、R 4b Together with the carbon atom to which they are attached form a cycloalkyl or heterocyclyl group;
each R is 3 And R 6 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, - (CH) 2 ) w -NR j R k Nitro, hydroxyl, hydroxyalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl;
R 5 are the same or different and are each independently selected from the group consisting of hydrogen atom, halogen, alkyl group, haloalkyl group, cyano group, hydroxy group and hydroxyalkyl group;
R d 、R e 、R f 、R g 、R h 、R i 、R j and R k Are the same or different and are each independently selected from the group consisting of hydrogen atoms, alkyl groups, alkenyl groups, alkynyl groups, haloalkyl groups, hydroxyalkyl groups, cycloalkyl groups, heterocyclyl groups, aryl groups, and heteroaryl groups;
u, v and w are the same or different and are each independently selected from 0, 1,2 and 3;
m is 0, 1 or 2;
n is 0 or 1;
r is 0, 1 or 2;
p is 0, 1,2,3, 4 or 5;
q is 0, 1,2,3, 4 or 5; and is
t is 0, 1,2,3, 4 or 5.
2. The compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein ring a is phenyl or naphthyl; and/or ring B is
Figure FDA0003653591460000021
R 6 Can be substituted at any position of the ring B.
3. The compound according to claim 1, which is a compound represented by the general formula (I-1):
Figure FDA0003653591460000022
wherein the content of the first and second substances,
q is 0, 1,2,3 or 4;
rings A, G 2 、L、R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, r and t are as defined in claim 1.
4. The compound according to claim 1 or2, which is a compound represented by the general formula (II):
Figure FDA0003653591460000023
wherein G is 2 、L、R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, q, r and t are as defined in claim 1.
5. The compound according to any one of claims 1 to 4, or a pharmaceutically acceptable salt thereof, wherein n is 0.
6. The compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, wherein m is 1 or 2.
7. A compound according to any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, wherein R 2 And R 2c Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 Haloalkyl, or R 2 、R 2c Together with the carbon atom to which they are attached form a 3-to 8-membered cycloalkyl group or a 3-to 8-membered heterocyclyl group; and/or R 2a And R 2b Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 Haloalkyl, or R 2a 、R 2b Together with the carbon atom to which they are attached form a 3-to 8-membered cycloalkyl or 3-to 8-membered heterocyclyl group; and/or R 4a And R 4b Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 Haloalkyl, or R 4a 、R 4b Together with the carbon atom to which they are attached form a 3-to 8-membered cycloalkyl group or a 3-to 8-membered heterocyclyl group.
8. The compound according to any one of claims 1 to 7, or a pharmaceutically acceptable salt thereof, wherein G is 2 Is NH.
9. The compound according to any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, wherein L is O.
10. The compound according to any one of claims 1 to 9, or a pharmaceutically acceptable salt thereof, wherein each R 1 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group.
11. A compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, wherein q is 2 or 3.
12. The compound according to any one of claims 1 to 11, or a pharmaceutically acceptable salt thereof, wherein each R 3 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl radical, C 2-6 Alkynyl, C 1-6 Haloalkyl, hydroxy, C 1-6 Hydroxyalkyl and 3 to 8 membered cycloalkyl.
13. The compound according to any one of claims 1 to 12, or a pharmaceutically acceptable salt thereof, wherein R 5 Are the same or different and are each independently selected from the group consisting of a hydrogen atom, C 1-6 Alkyl, hydroxy and C 1-6 A hydroxyalkyl group; preferably, R 5 Is a hydrogen atom.
14. The compound according to any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, wherein each R 6 Are the same or different and are each independently selected from the group consisting of hydrogen, halogen, C 1-6 Alkyl and C 1-6 A haloalkyl group.
15. The compound according to any one of claims 1 to 4, 6 to 14, or a pharmaceutically acceptable salt thereof, which is a compound of:
Figure FDA0003653591460000041
Figure FDA0003653591460000051
Figure FDA0003653591460000061
Figure FDA0003653591460000071
Figure FDA0003653591460000081
Figure FDA0003653591460000091
16. a compound represented by the general formula (I-1A):
Figure FDA0003653591460000092
wherein:
q is 0, 1,2,3 or 4;
R 0 is a hydrogen atom or a hydroxyl protecting group;
r is an amino protecting group; preferably Boc;
ring A, L, R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, r and t are as defined in claim 3.
17. A compound having the structure:
Figure FDA0003653591460000093
Figure FDA0003653591460000101
Figure FDA0003653591460000111
Figure FDA0003653591460000121
Figure FDA0003653591460000131
Figure FDA0003653591460000141
Figure FDA0003653591460000151
18. a process for producing a compound represented by the general formula (I-1) or a pharmaceutically acceptable salt thereof according to claim 3, which comprises:
Figure FDA0003653591460000152
R 0 is a hydrogen atom or a hydroxyl protecting group;
when R is 0 When the hydrogen atom is a hydrogen atom, removing the amino protecting group R from the compound of the general formula (I-1A) or a salt thereof to obtain a compound of the general formula (I-1) or a pharmaceutically acceptable salt thereof;
when R is 0 When the protecting group is a hydroxyl group, the compound of the formula (I-1A) or a salt thereof is freed from the protecting group R 0 And an amino protecting group R to give a compound of the general formula (I-1) or a pharmaceutically acceptable salt thereof;
wherein the content of the first and second substances,
r is an amino protecting group; preferably Boc;
G 2 is NH;
ring A, L, R 1 、R 2 、R 2c 、R 2a 、R 2b 、R 3 、R 4a 、R 4b 、R 5 、R 6 M, n, p, q, r and t are as defined in claim 3.
19. A pharmaceutical composition comprising a compound according to any one of claims 1 to 15, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents or excipients.
20. Use of a compound according to any one of claims 1 to 15, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 19, in the manufacture of a medicament for inhibiting KRAS G12D.
21. Use of a compound according to any one of claims 1 to 15 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 19, in the manufacture of a medicament for the treatment and/or prophylaxis of a disease or condition which is cancer.
22. Use of a compound according to any one of claims 1 to 15 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 19, in the manufacture of a medicament for the treatment and/or prevention of a disease or condition selected from brain cancer, thyroid cancer, head and neck cancer, nasopharyngeal cancer, cancer of the throat, oral cancer, salivary gland cancer, esophageal cancer, gastric cancer, lung cancer, liver cancer, kidney cancer, pancreatic cancer, gall bladder cancer, bile duct cancer, colorectal cancer, small intestine cancer, gastrointestinal stromal tumor, urothelial cancer, urinary tract cancer, bladder cancer, breast cancer, vaginal cancer, ovarian cancer, endometrial cancer, cervical cancer, fallopian tube cancer, testicular cancer, prostate cancer, hemangioma, leukemia, lymphoma, myeloma, skin cancer, lipoma, bone cancer, soft tissue sarcoma, neurofibroma, glioma, neuroblastoma, and glioblastoma; preferably selected from pancreatic cancer, colorectal cancer and non-small cell lung cancer.
CN202210558739.9A 2021-05-21 2022-05-20 Condensed nitrogen-containing heterocyclic compound, preparation method and medical application thereof Pending CN115368383A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN2021105593874 2021-05-21
CN202110559387 2021-05-21
CN2021115052668 2021-12-10
CN202111505266 2021-12-10

Publications (1)

Publication Number Publication Date
CN115368383A true CN115368383A (en) 2022-11-22

Family

ID=84062276

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210558739.9A Pending CN115368383A (en) 2021-05-21 2022-05-20 Condensed nitrogen-containing heterocyclic compound, preparation method and medical application thereof

Country Status (1)

Country Link
CN (1) CN115368383A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023198191A1 (en) * 2022-04-15 2023-10-19 杭州多域生物技术有限公司 Six- and six-membered compound, preparation method, pharmaceutical composition, and application

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023198191A1 (en) * 2022-04-15 2023-10-19 杭州多域生物技术有限公司 Six- and six-membered compound, preparation method, pharmaceutical composition, and application

Similar Documents

Publication Publication Date Title
CN104884458B (en) fused heterocyclic compounds as protein kinase inhibitors
CN113801114B (en) Fused bicyclic heteroaryl derivative, preparation method and application thereof in medicines
CN115244058A (en) Pyrimidobicyclo derivatives, preparation method thereof and application thereof in medicines
CN113795483A (en) Carboxamide-pyrimidine derivatives as SHP2 antagonists
CN117255793A (en) Fused tetracyclic compound, preparation method thereof and application thereof in medicines
CN117203207A (en) Bridged ring compound, preparation method thereof and application thereof in medicine
AU2022280845A1 (en) Nitrogen-containing heterocyclic compound, preparation method therefor, and application thereof in medicines
CN115403595A (en) Nitrogen-containing heterocyclic compound, preparation method and medical application thereof
CN115385938A (en) Benzopyrimidine compounds, preparation method thereof and application thereof in medicines
CN112574236A (en) RET inhibitor, pharmaceutical composition and application thereof
CN112442050A (en) RET inhibitor, pharmaceutical composition and application thereof
CN114907377A (en) Condensed tetracyclic compound, preparation method and medical application thereof
CN115368383A (en) Condensed nitrogen-containing heterocyclic compound, preparation method and medical application thereof
WO2023041049A1 (en) Heterocyclic compound as sos1 inhibitor and uses thereof
TW202227452A (en) Fused azaheterocyclic compounds, preparation methods, methods of inhibiting tyrosine kinase, pharmaceutical composition and uses thereof
WO2021185238A1 (en) Fused bicyclic derivative, preparation method therefor, and pharmaceutical use thereof
CN115867552A (en) Imidazolopyrimidine derivative, preparation method thereof and application thereof in medicines
CN115385937A (en) Pyrimido-cycloalkyl compounds, preparation method and medical application thereof
CN113135942B (en) Condensed pyrimidine derivative, preparation method and medical application thereof
CN114456173B (en) Condensed ring group substituted cyclohexanediimide derivative, preparation method and medical application thereof
CN113912608B (en) Pyrimidopyrimidinone derivatives, preparation method thereof and application thereof in medicines
TW202409051A (en) Fused ring compounds, preparation method and medical use thereof
CN113444110A (en) Tetrahydropyrrolopyrazole derivative, preparation method and medical application thereof
WO2024022444A1 (en) Fused ring compound, preparation method therefor and medicinal application thereof
TWI602818B (en) Fused heterocyclic compounds as protein kinase inhibitors

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication