CN116635371A - Polycyclic pyridazinone derivative serving as SOS1 inhibitor, and preparation method and application thereof - Google Patents

Polycyclic pyridazinone derivative serving as SOS1 inhibitor, and preparation method and application thereof Download PDF

Info

Publication number
CN116635371A
CN116635371A CN202280007414.XA CN202280007414A CN116635371A CN 116635371 A CN116635371 A CN 116635371A CN 202280007414 A CN202280007414 A CN 202280007414A CN 116635371 A CN116635371 A CN 116635371A
Authority
CN
China
Prior art keywords
membered
compound
heterocyclic group
general formula
mmol
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
CN202280007414.XA
Other languages
Chinese (zh)
Inventor
郑苏欣
谢成英
陆晓杰
郑明月
乔刚
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.)
Suzhou Almai Biotechnology Co ltd
Original Assignee
Suzhou Almai Biotechnology 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 Suzhou Almai Biotechnology Co ltd filed Critical Suzhou Almai Biotechnology Co ltd
Publication of CN116635371A publication Critical patent/CN116635371A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/26Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Abstract

Polycyclic pyridazinone derivatives as SOS1 inhibitors, methods of preparation and uses thereof are provided. The structure of the polycyclic pyridazinone derivative is shown as a formula (I), and the polycyclic pyridazinone derivative has remarkable effects of inhibiting RAS signal channels and treating cancers, wherein the cancers are pancreatic cancer, colorectal cancer, lung cancer, hepatocellular carcinoma, renal cancer, gastric cancer, cholangiocarcinoma and the like.

Description

Polycyclic pyridazinone derivative serving as SOS1 inhibitor, and preparation method and application thereof Technical Field
The invention belongs to the technical field of polycyclic pyridazinone derivatives, and particularly relates to a polycyclic pyridazinone derivative serving as an SOS1 inhibitor, and a preparation method and application thereof.
Background
RAS family proteins belong to a small GTPase comprising three subfamilies of KRAS, NRAS and HRAS. Mutated RAS gene is an important oncogene, and the discovery of RAS gene mutation is found in 20-30% of human tumors, particularly pancreatic, colorectal and lung cancers. The various isoforms of RAS proteins have a balance of GTP-bound active and GDP-bound inactive states, GTPase-activated proteins (GTPase-activating proteins, GAPs) may facilitate the conversion of GTP to GDP, thereby converting RAS proteins to inactive states, and guanine nucleotide exchange factors (guanine nucleotide exchange factor, GEFs) may facilitate the release of GDP and the binding of GTP, thereby converting RAS proteins to active states. Activation of RAS proteins promotes proliferation of cells, apoptosis escape, metabolic recombination and the like through signal pathways of RAS-RAF-MEK-ERK and RAS-PI3K-PDK1-AKT, thereby promoting occurrence and development of tumors.
SOS1 (son of seveless 1) is a key guanine nucleotide exchange factor (GEF) capable of binding to RAS proteins, promoting the binding of RAS proteins to GTP, and turning RAS proteins to an active state. Recent studies have found SOS1 inhibitionThe preparation not only can inhibit RAS mutant cell growth, but also can produce synergistic effect with MEK inhibitor, and has remarkable inhibition effect on KRAS-driven tumor 1-2 . The development of SOS1 inhibitors has become a focus of research and SOS1 inhibitors of different structural types have been reported in several patents, such as WO2018172250, WO2019201848, WO2018115380, WO2019122129, WO2020173935, WO2020180768 and WO2020180770, etc.
However, there is still uncertainty in the effectiveness, safety or selectivity of these compounds and test drugs disclosed in the prior art, and thus there is a need to study and develop new selective SOS1 inhibitors.
Reference is made to:
1、Hillig et al.Discovery of poetent SOS1inhibitors that block RAS activation via disruption of the RAS-SOS1interaction.PNAS.116,2251-2560(2019).
2、Hofmann et al.BI-3406,a potent and selective SOS1::KRAS interaction inhibitor,is effective in KRAS-driven cancers through combined MEK inhibition.Cancer Discov.CD-20-0142(2020).
disclosure of Invention
In order to solve the above problems of the prior art, an object of the present invention is to provide a polycyclic pyridazinone derivative, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof, for screening out a compound useful as an SOS1 inhibitor having excellent properties in terms of performance such as effectiveness, safety and selectivity.
It is another object of the present invention to provide a process for the preparation of said derivatives, pharmaceutically acceptable salts thereof, tautomers thereof or stereoisomers thereof.
In order to achieve the aim of the invention, the invention adopts the following technical scheme:
in a first aspect, the present invention provides a polycyclic pyridazinone derivative, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof, wherein the structure of the polycyclic pyridazinone derivative is shown as formula (I):
wherein: r is R 1 Selected from hydrogen or methyl;
R 2 selected from C 1 -C 3 Alkyl, -OR 21 Halogen, 3-7 membered cycloalkyl, 5-7 membered cycloalkenyl, 6-10 membered fused ring alkyl, 7-10 membered bridged cycloalkyl, 7-10 membered spirocycloalkyl, 4-7 membered heterocyclyl, 5-7 membered heterocyclenyl, 6-10 membered fused heterocyclyl, 7-10 membered bridged heterocyclyl, 7-10 membered spiroheterocyclyl, wherein 3-7 membered cycloalkyl, 5-7 membered cycloalkenyl, 6-10 membered fused ring alkyl, 7-10 membered bridged cycloalkyl, 7-10 membered spirocycloalkyl, 4-7 membered heterocyclyl, 5-7 membered heterocyclenyl, 6-10 membered fused heterocyclyl, 7-10 membered bridged heterocyclyl, 7-10 membered spiroheterocyclyl is optionally substituted with 1-3R 22 Substituted;
R 21 selected from H, C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl, wherein C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl are optionally substituted with 1-3R 22 Substituted;
R 22 selected from C 1 -C 3 Alkyl, hydroxy, halogen, cyano, -NR a R b 、C 1 -C 3 Alkoxy, -C (O) R a 、-C(O)OR a 、-OC(O)R a 、-NR b C(O)R a 、-NR b C(O)OR a 、-C(O)NR a R b Phenyl, 5-6 membered heteroaryl and =o, wherein alkyl, alkoxy, phenyl, 5-6 membered heteroaryl are optionally further substituted with 1-3 halogen, C 1 -C 3 Alkyl, hydroxy, cyano, amino and C 1 -C 3 Alkoxy substituted;
R a and R is b Independently selected from H, substituted or unsubstituted C 1 -C 3 Substituted or unsubstituted 3-6 membered cycloalkyl or substituted or unsubstituted 4-7 membered heterocyclyl; "substituted" herein means optionally substituted with 1 to 3 substituents selected from C 1 -C 3 Is substituted by alkyl, hydroxy, halogen, cyano, amino or alkoxy;
q is selected from N or-CR 3
R 3 Selected from H, C 1 -C 3 Alkyl, halogen, cyano OR-OR 21
AR is selected from 6-10 membered aryl or 5-10 membered heteroaryl, wherein aryl or heteroaryl is optionally substituted with 1-4R c Substituted;
R c selected from H, halogen, C 1 -C 4 Alkyl, C 1 -C 4 Haloalkyl, hydroxy-C 1 -C 4 Alkyl, hydroxy-C 1 -C 4 Haloalkyl, 3-6 membered cycloalkyl, 4-7 membered heterocyclyl, -OR 21 、-NR a R b 、NR a R b -C 1 -C 4 Alkyl, NR a R b -C 1 -C 4 Haloalkyl, 6-10 membered aryl or 5-10 membered heteroaryl, wherein 6-10 membered aryl or 5-10 membered heteroaryl is optionally substituted with 1-4R d Substituted;
R d selected from H, halogen, C 1 -C 4 Alkyl, C 1 -C 4 Haloalkyl, hydroxy-C 1 -C 4 Alkyl, hydroxy-C 1 -C 4 Haloalkyl, 3-6 membered cycloalkyl, 4-7 membered heterocyclyl, -OR 21 、-NR a R b 、NR a R b -C 1 -C 4 Alkyl, NR a R b -C 1 -C 4 A haloalkyl group;
the hetero atoms in the heterocyclic group, the heteroaryl, the heterocyclenyl, the fused heterocyclic group, the bridged heterocyclic group and the spiro heterocyclic group in the formula (I) are 1-7 and are selected from one or more of oxygen, nitrogen, sulfur and S (O) m, and m is 1 or 2.
Preferably, the structure of the polycyclic pyridazinone derivative is shown as a formula (II):
wherein R is 1 、R 2 Q and R c Has the same limit range as the above; n=1-4 (e.g., n=1, n=2, n=3, n=4).
Preferably, for compounds of formula (II), the phenyl group is optionally substituted with 1 to 3R c Substituted when said R c When the number of R is 2-3, R is c May be the same or different;
and/or when said R c Is C 1 -C 4 When halogenoalkyl, the halogen atom is fluorine;
and/or when said R c When halogen, the halogen atom is fluorine;
and/or when said R c is-NR a R b When said R is a And R is b May be the same or different.
Preferably, the structure of the polycyclic pyridazinone derivative is shown as a formula (III):
wherein R is 1 、R 2 And R is c Has the same limit range as the above; n=1-4 (e.g., n=1, n=2, n=3, n=4).
Preferably, for the compound of formula (III), the R 2 Is optionalGround cover 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 When the number of the components is 2-3, R 22 The same or different;
and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclyl, the heterocyclyl contains 1-2 heteroatoms;
and/or, the R 2 Is optionally covered with 1-3R 22 When the substituted 4-7 membered heterocyclic group is, the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
and/or, the R 2 Is optionally covered with 1-3R 22 When the number of the hetero atoms of the heterocyclic group is two, the two hetero atoms are the same or different;
and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 Selected from C 1 -C 3 Alkyl, -NR a R b 、-C(O)R a 、-NR b C(O)R a 、-NR b C(O)OR a 、-C(O)NR a R b And = O.
Preferably, the structure of the polycyclic pyridazinone derivative is shown as a formula (IV):
wherein R is 1 、R 2 、R 3 And R is c Has the same limit range as the above; n=1-4 (e.g., n=1, n=2, n=3, n=4).
Preferably, for the compound of formula (IV), the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 When the number of the components is 2-3, R 22 The same or different;
and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclyl, the heterocyclyl contains 1-2 heteroatoms;
And/or, the R 2 Is optionally covered with 1-3R 22 When the substituted 4-7 membered heterocyclic group is, the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
and/or, the R 2 Is optionally covered with 1-3R 22 When the number of the hetero atoms of the heterocyclic group is two, the two hetero atoms are the same or different;
and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 Selected from C 1 -C 3 Alkyl, -NR a R b 、-C(O)R a 、-NR b C(O)R a 、-NR b C(O)OR a 、-C(O)NR a R b And = O;
and/or, the R 3 is-OR 21 When R is 21 Selected from unsubstituted C 1 -C 3 Alkyl or unsubstituted 3-7 membered cycloalkyl.
Preferably, the structure of the polycyclic pyridazinone derivative is shown as a formula (V):
wherein R is 1 、R 2 Q and R d Has the same limit range as the above; n=1-4 (e.g., n=1, n=2, n=3, n=4).
Preferably, said R d Is 1-NR a R b When said R is a And R is b Independently selected from H, substituted or unsubstituted C 1 -C 3 Alkyl of (a);
and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 When the number of the components is 2-3, R 22 The same or different;
and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclyl, the heterocyclyl contains 1-2 heteroatoms;
and/or, the R 2 Is optionally covered with 1-3R 22 When the substituted 4-7 membered heterocyclic group is, the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
and/or, the R 2 Is optionally covered with 1-3R 22 When the number of the hetero atoms of the heterocyclic group is two, the two hetero atoms are the same or different;
and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 Selected from C 1 -C 3 Alkyl, -NR a R b 、-C(O)R a 、-NR b C(O)R a 、-NR b C(O)OR a 、-C(O)NR a R b And = O;
and/or, the R 2 is-OR 21 When R is 21 Is a 4-7 membered heterocyclic group;
and/or, the R 21 In the case of 4-7 membered heterocyclic group, the 4-7 membered heterocyclic group is 5-6 membered heterocyclic group;
and/or, the R 21 In the case of 4-7 membered heterocyclic groups, the heteroatoms of the heterocyclic groups are nitrogen and/or oxygen;
and/or, the R 21 In the case of a 4-7 membered heterocyclic group, the heterocyclic group contains 1-2 heteroatoms;
and/or, the R 21 In the case of 4-7 membered heterocyclic groups, when the number of heteroatoms in the heterocyclic group is two, the two heteroatoms may be the same or different.
Preferably, the structure of the polycyclic pyridazinone derivative is shown as a formula (VI):
wherein R is 1 、R 2 And R is d Has the same limit range as the above; n=1-4 (e.g., n=1, n=2, n=3, n=4).
Preferably, when said R 2 is-OR 21 The R is 21 Is a 4-7 membered heterocyclic group;
and/or when said R 21 In the case of 4-7 membered heterocyclic group, the 4-7 membered heterocyclic group is 5-7 membered heterocyclic group;
and/or when said R 21 In the case of 4-7 membered heterocyclic groups, the heteroatoms of the heterocyclic groups are nitrogen and/or oxygen;
and/or when said R 21 In the case of a 4-7 membered heterocyclic group, the heterocyclic group contains two heteroatoms;
and/or when said R 21 In the case of a 4-7 membered heterocyclyl, the two heteroatoms may be the same or different;
and/or, the R d Selected from halogen, C 1 -C 4 Alkyl, -NR a R b 、-OR 21 、NR a R b -C 1 -C 4 An alkyl group;
and/or, the R d Is 1-NR a R b -C 1 -C 4 In the case of alkyl, said R a And R is b Independently selected from H, substituted or unsubstituted C 1 -C 3 Is a hydrocarbon group.
Preferably, the structure of the polycyclic pyridazinone derivative is shown as a formula (VII):
wherein R is 1 、R 2 、R 3 And R is d Has the same limit range as the above; n=1-4 (e.g., n=1, n=2, n=3, n=4).
Preferably, when said R 2 is-OR 21 The R is 21 Is a 4-7 membered heterocyclic group;
and/or when said R 21 In the case of 4-7 membered heterocyclic group, the 4-7 membered heterocyclic group is 5-7 membered heterocyclic group;
and/or when said R 21 In the case of 4-7 membered heterocyclic groups, the heteroatoms of the heterocyclic groups are nitrogen and/or oxygen;
and/or when said R 21 In the case of a 4-7 membered heterocyclic group, the heterocyclic group contains two heteroatoms;
and/or when said R 21 In the case of a 4-7 membered heterocyclyl, the two heteroatoms may be the same or different;
and/or, the R d Selected from halogen, C 1 -C 4 Alkyl, -NR a R b 、-OR 21 、NR a R b -C 1 -C 4 An alkyl group;
and/or, the R d Is 1 NR a R b -C 1 -C 4 In the case of alkyl, said R a And R is b Independently selected from H, substituted or unsubstituted C 1 -C 3 Is a hydrocarbon group.
Further preferably, the polycyclic pyridazinone derivative is selected from any one of the following structures:
typical compounds of the invention include, but are not limited to, compounds in the following table:
in a second aspect, the present invention provides a process for the preparation of a polycyclic pyridazinone derivative, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof according to the first aspect, selected from one of the following two schemes:
scheme one
The preparation method of the compound shown in the general formula (I) or the stereoisomer, the tautomer or the medicinal salt thereof comprises the following steps:
preparation of key intermediate (I-A):
step one, performing metal catalytic cross coupling on an aromatic compound of a general formula (I-1) to obtain a compound of a general formula (I-2);
secondly, reacting the compound shown in the general formula (I-2) under the condition of a catalyst to obtain a chiral sulfimide compound shown in the general formula (I-3);
thirdly, reducing the compound of the general formula (I-3) by a metal reducing agent to obtain a chiral compound of the general formula (I-4);
Fourthly, the compound of the general formula (I-4) is cracked into chiral benzylamine compound of the general formula (I-A) under the acid condition;
among them, X halogen is preferably bromine.
Preparation of key intermediate (I-B), method one:
step one, diazotizing a compound of a general formula (I-5) to obtain a compound of a general formula (I-6);
secondly, carrying out substitution reaction on the compound of the general formula (I-6) and cuprous cyanide to obtain a compound of the general formula (I-7);
thirdly, cyano groups in the compound shown in the general formula (I-7) form amidines, and then the amidines and ester groups are subjected to transesterification reaction to obtain the compound shown in the general formula (I-B);
therein, X, X 1 Is halogen, X is preferably bromine, X 1 Preferably iodine;
preparation of key intermediate (I-B), method II:
firstly, carrying out halogenation reaction on a compound with a general formula (I-12) to obtain a compound with a general formula (I-13);
secondly, carrying out esterification reaction on the compound shown in the general formula (I-13) to obtain a compound shown in the general formula (I-6);
thirdly, carrying out substitution reaction on the compound of the general formula (I-6) to obtain a compound of the general formula (I-14);
fourthly, carrying out oxidation reaction on the compound shown in the general formula (I-14) to obtain a compound shown in the general formula (I-15);
fifthly, reacting the compound of the general formula (I-15) with hydroxylamine sulfonic acid to obtain a compound of the general formula (I-16);
Sixthly, carrying out Abnormal Beckmann rearrangement reaction on the compound of the general formula (I-16) to obtain a compound of the general formula (I-7);
seventhly, forming amidine by cyano in the compound shown in the general formula (I-7), and carrying out transesterification reaction on the amidine and ester group to obtain the compound shown in the general formula (I-B);
therein, X, X 1 Is halogen, X is preferably chlorine, X 1 Preferably iodine;
preparation of general formula (I):
the first step, the compound of the general formula (I-A) and the compound of the general formula (I-B) are subjected to imine addition reaction to obtain the compound of the general formula (I-8);
secondly, performing imine addition and ring expansion reaction on the compound shown in the general formula (I-8) and hydrazine hydrate to obtain a compound shown in the general formula (I-9);
in the third step, the compound of the general formula (I-9) and the compound of the general formula (I-10) are subjected to substitution reaction to obtain the compound of the general formula (I-11) (such as R 1 This step is omitted, =h);
fourthly, carrying out Buchwald/Suzuki reaction on the compound shown in the general formula (I-11) and the compound shown in the general formula (I-25) under alkaline conditions in the presence of a metal catalyst and a ligand to obtain the compound shown in the general formula (I);
therein, X, X 1 Is halogen, X is preferably bromine or chlorine, X 1 Preferably iodine; w is H,Q、AR、R 1 And R is 2 Has the same limiting scope as described above.
Scheme II
The preparation method of the compound shown in the general formula (I) or the stereoisomer, the tautomer or the medicinal salt thereof comprises the following steps:
Step one, carrying out substitution reaction on a compound of a general formula (I-17) to obtain a compound of a general formula (I-18);
secondly, removing protecting groups from the compound of the general formula (I-18) under acidic conditions to obtain a compound of the general formula (I-19);
thirdly, the compound of the general formula (I-19) and the compound of the general formula (I-26) are subjected to alkaline conditions to obtain the compound of the general formula (I-20);
fourth, the compound of the general formula (I-20) is subjected to oxidation reaction to obtain a compound of the general formula (I-21);
fifthly, carrying out Bouveault aldehyde synthesis reaction on the compound shown in the general formula (I-21) to obtain a compound shown in the general formula (I-22);
step six, the compound of the general formula (I-22) and hydrazine hydrate undergo an addition cyclization reaction to obtain a compound of the general formula (I-23);
seventhly, carrying out substitution reaction on the compound of the general formula (I-23) to obtain a compound of the general formula (I-24);
eighth step, the compound of the general formula (I-24) and the compound of the general formula (I-10) are subjected to substitution reaction to obtain the general formulaCompounds of formula (I-Bb) (e.g. R 1 This step is omitted, =h);
ninth, the compound of the general formula (I-Bb) and the compound of the general formula (I-A) are subjected to Buchwald reaction under alkaline conditions in the presence of a metal catalyst and a ligand to obtain the compound of the general formula (I);
wherein X is 2 、X 3 、X 4 Is halogen, X 2 、X 3 Preferably bromine, X 4 Preferably iodine; q is selected from N or CR 3 ;R 3 Selected from H, C 1 -C 3 Alkyl, halogen, cyano OR-OR 21 ;R 1 Selected from hydrogen or methyl; AR and R 2 Has the same limiting scope as described above.
In the preparation method, the reagent providing alkaline conditions is selected from organic base or inorganic base, wherein the organic base is one or more of triethylamine, N-diisopropylethylamine, N-butyllithium, lithium diisopropylamide, lithium bistrimethylsilylamino, sodium tert-butoxide, sodium methoxide and potassium tert-butoxide, and the inorganic base is one or more of sodium hydride, potassium phosphate, sodium carbonate, potassium acetate, cesium carbonate, sodium hydroxide, potassium hydroxide, sodium bicarbonate and lithium hydroxide;
the reagent providing the acidic condition is one or more of hydrogen chloride, 1, 4-dioxane solution of hydrogen chloride, methanol solution of hydrogen chloride, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid, methanesulfonic acid, nitric acid and phosphoric acid;
the metal catalyst is palladium/carbon, raney nickel, tetra-triphenylphosphine palladium, palladium dichloride, palladium acetate, and [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride (Pd (dppf) Cl) 2 ) [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride dichloromethane complex, ditriphenylphosphine palladium dichloride (Pd (PPh) 3 )Cl 2 ) And tris (dibenzylideneacetone) dipalladium (Pd) 2 (dba) 3 ) One or more of the following;
the ligand is one or more of 2-dicyclohexylphosphine-2, 6' -dimethoxybiphenyl (SPhos), 4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene (Xantphos), 2-dicyclohexylphosphine-2, 4, 6-triisopropylbiphenyl (XPhos), 2-dicyclohexylphosphino-2 ' - (N, N-dimethylamine) -biphenyl (DavePhos), 1' -bis (diphenylphosphine) ferrocene (Dppf) and 1,1' -binaphthyl-2, 2' -bis-diphenylphosphine (BINAP), preferably 1,1' -binaphthyl-2, 2' -bis-diphenylphosphine (BINAP);
the reducing agent is one or more of sodium borohydride, potassium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride and lithium aluminum hydride;
the oxidant is one or more of potassium permanganate, manganese dioxide, potassium dichromate, sodium dichromate and potassium osmium;
the above reaction is preferably carried out in a solvent selected from one or more of N, N-dimethylformamide, N-methylpyrrolidone, dimethylsulfoxide, 1, 4-dioxane, water, tetrahydrofuran, methylene chloride, 1, 2-dichloroethane, methanol, ethanol, toluene, petroleum ether, ethyl acetate, N-hexane and acetone.
In a third aspect, the present invention provides a pharmaceutical composition comprising a polycyclic pyridazinone derivative according to the first aspect, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof;
Preferably, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier and/or excipient.
In a fourth aspect, the present invention provides the use of a polycyclic pyridazinone derivative according to the first aspect, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof or a pharmaceutical composition according to the third aspect for the manufacture of a medicament for the treatment of cancer or for the manufacture of an SOS1 inhibitor;
preferably, the cancer is pancreatic cancer, colorectal cancer, lung cancer, hepatocellular cancer, renal cancer, gastric cancer, or cholangiocarcinoma.
In a fifth aspect, the present invention provides a method for preventing and/or treating cancer comprising administering to a human a therapeutically effective amount of a polycyclic pyridazinone derivative according to the first aspect, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof or a pharmaceutical composition according to the third aspect.
Interpretation of the terms
Unless stated to the contrary, some of the terms used in the specification and claims of the present invention are defined as follows:
"alkyl" refers to a saturated aliphatic hydrocarbon group comprising 1 to 20 carbon atoms, or 1 to 10 carbon atoms, or 1 to 6 carbon atoms, or 1 to 4 carbon atoms, or 1 to 3 carbon atoms, or 1 to 2 carbon atoms, saturated straight or branched chain monovalent hydrocarbon groups, wherein the alkyl groups may be independently optionally substituted with one or more substituents described herein. Examples of alkyl groups further include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2, 3-dimethylbutyl, and the like. The alkyl group may be optionally substituted or unsubstituted.
"alkenyl" refers to a straight or branched monovalent hydrocarbon radical of 2 to 12 carbon atoms, or 2 to 8 carbon atoms, or 2 to 6 carbon atoms, or 2 to 4 carbon atoms, wherein at least one C-C is sp 2 Double bonds, wherein the alkenyl groups may be independently optionally substituted with 1 or more substituents as described herein, specific examples of which include, but are not limited to, vinyl, allyl, and allyl groups, and the like. Alkenyl groups may be optionally substituted or unsubstituted.
"cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring comprising 3 to 20 carbon atoms, preferably comprising 3 to 12 carbon atoms, more preferably comprising 3 to 6 carbon atoms. Non-limiting examples of monocyclic cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like; polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups. Cycloalkyl groups may be optionally substituted or unsubstituted.
"spirocycloalkyl" refers to a 5 to 18 membered, two or more cyclic structure, and monocyclic polycyclic groups sharing one carbon atom (called spiro atom) with each other, containing 1 or more double bonds within the ring, but no ring has a completely conjugated pi-electron aromatic system. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The spirocycloalkyl group is classified into a single spiro group, a double spiro group or a multiple spirocycloalkyl group according to the number of common spiro atoms between rings, preferably single spiro group and double spirocycloalkyl group, preferably 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered. Non-limiting examples of "spirocycloalkyl" include, but are not limited to:
"fused ring alkyl" refers to an all-carbon polycyclic group containing two or more cyclic structures sharing a pair of carbon atoms with each other, one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron aromatic system, preferably 6 to 12 members, more preferably 7 to 10 members. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused ring alkyl group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicycloalkyl group. Non-limiting examples of "fused ring alkyl" include, but are not limited to:
"bridged cycloalkyl" means an aromatic system having 5 to 18 members, containing two or more cyclic structures, sharing two all-carbon polycyclic groups with one another that are not directly attached to a carbon atom, one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi electron, preferably 6 to 12 members, more preferably 7 to 10 members. Cycloalkyl groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of "bridged cycloalkyl" include, but are not limited to:
The cycloalkyl ring may be fused to an aryl, heteroaryl or heterocyclyl ring, wherein the ring attached to the parent structure is cycloalkyl, non-limiting examples include indanyl, tetrahydronaphthyl, benzocycloheptyl, and the like.
"heterocyclyl", "heterocycle" or "heterocyclic" are used interchangeably herein and refer to a saturated or partially unsaturated, monocyclic, bicyclic or tricyclic, non-aromatic heterocyclic group containing 3 to 12 ring atoms, at least one of which is a heteroatom such as oxygen, nitrogen, sulfur, and the like. Preferably having a 5 to 7 membered mono-or 7 to 10 membered bi-or tri-ring, which may contain 1,2 or 3 atoms selected from nitrogen, oxygen and/or sulphur. Examples of "heterocyclyl" include, but are not limited to, morpholinyl, oxetanyl, thiomorpholinyl, tetrahydropyranyl, 1-dioxo-thiomorpholinyl, piperidinyl, 2-oxo-piperidinyl, pyrrolidinyl, 2-oxo-pyrrolidinyl, piperazin-2-one, 8-oxa-3-aza-bicyclo [3.2.1] octyl, and piperazinyl. The heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring attached to the parent structure is heterocyclyl. The heterocyclyl group may be optionally substituted or unsubstituted.
"spiroheterocyclyl" refers to a 5-to 18-membered, two or more cyclic structure, polycyclic group having single rings sharing one atom with each other, containing 1 or more double bonds within the ring, but no ring having a fully conjugated pi-electron aromatic system in which one or more ring atoms are selected from nitrogen, oxygen, sulfur or S (O) m The remaining ring atoms are carbon, m=1 or 2. Preferably 6 to 14 membered, more preferably 7 to 10 membered. Dividing spiroheterocyclic groups into mono-spiroheterocyclic groups according to the number of common spiro atoms between ringsThe group, a dispiro-heterocyclic group or a polysiro-heterocyclic group is preferably a mono-spiro-heterocyclic group or a dispiro-heterocyclic group. More preferably a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered single spiro heterocyclic group. Non-limiting examples of "spiroheterocyclyl" include, but are not limited to:
"fused heterocyclyl" refers to an all-carbon polycyclic group containing two or more cyclic structures sharing a common pair of atoms with each other, one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron aromatic system in which one or more of the ring atoms is selected from nitrogen, oxygen, sulfur or S (O) m The remaining ring atoms are carbon, m=1 or 2. Preferably 6 to 14 membered, more preferably 7 to 10 membered. The number of constituent rings may be classified as a bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic group, preferably a bicyclic or tricyclic, more preferably a 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic group. Non-limiting examples of "fused heterocyclyl" include, but are not limited to:
"bridged heterocyclyl" means a 5-to 18-membered, polycyclic group containing two or more cyclic structures sharing two atoms not directly attached to each other, one or more of the rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron aromatic system in which one or more of the ring atoms is selected from nitrogen, oxygen, sulfur or S (O) m The remaining ring atoms are carbon, m=1 or 2. Preferably 6 to 14 membered, more preferably 7 to 10 membered. Heterocyclic groups which may be classified as bicyclic, tricyclic, tetracyclic or polycyclic bridged according to the number of constituent rings are preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic. Non-limiting examples of "bridged heterocyclyl" include, but are not limited to:
"aryl" refers to a carbocyclic aromatic system containing one or two rings, wherein the rings may be linked together in a fused manner. The term "aryl" includes aromatic groups such as phenyl, naphthyl, tetrahydronaphthyl. Preferably aryl is C 6 -C 10 Aryl, more preferably aryl is phenyl and naphthyl, most preferably phenyl. Aryl groups may be substituted or unsubstituted. The "aryl" may be fused to a heteroaryl, heterocyclyl, or cycloalkyl group, wherein the aryl ring is attached to the parent structure, non-limiting examples include, but are not limited to:
"heteroaryl" refers to an aromatic 5-to 6-membered monocyclic or 9-to 10-membered bicyclic ring, which may contain 1 to 4 atoms selected from nitrogen, oxygen and/or sulfur. Examples of "heteroaryl" include, but are not limited to, furyl, pyridyl, 2-oxo-1, 2-dihydropyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, thienyl, isoxazolyl, oxazolyl, oxadiazolyl, imidazolyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, isothiazolyl, 1,2, 3-thiadiazolyl, benzodioxolyl, benzimidazolyl, indolyl, isoindolyl, 1, 3-dioxo-isoindolyl, quinolinyl, indazolyl, benzisothiazolyl, benzoxazolyl and benzisoxazolyl. Heteroaryl groups may be optionally substituted or unsubstituted. The heteroaryl ring may be fused to an aryl, heterocyclyl, or cycloalkyl ring, wherein the ring attached to the parent structure is a heteroaryl ring, non-limiting examples include, but are not limited to:
"alkoxy" refers to a group of (alkyl-O-). Wherein alkyl is as defined herein. C (C) 1 -C 6 Is preferably selected. Examples include, but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy and the like.
"haloalkyl" refers to an alkyl group having one or more halo substituents, wherein the alkyl group has the meaning as described herein. Examples of haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, perfluoroethyl, 1-dichloroethyl, 1, 2-dichloropropyl, and the like.
"hydroxy" refers to an-OH group.
"halogen" means fluorine, chlorine, bromine and iodine, preferably fluorine, chlorine and bromine.
"amino" means-NH 2
"cyano" refers to-CN.
"nitro" means-NO 2
"benzyl" means-CH 2 -phenyl.
"carboxy" means-C (O) OH.
"acetyl" means-C (O) CH 3 Or Ac.
"carboxylate" refers to-C (O) O (alkyl) or (cycloalkyl), wherein alkyl, cycloalkyl are as defined above.
"optional" means that the event described may, but need not, occur. For example, "AR is optionally 1 to multiple R c Substitution the description includes that the AR group may be substituted with 1 to a plurality of R c Substituted or not by R c Substitution case.
"substituted" means that one or more hydrogen atoms, preferably up to 5, more preferably 1 to 3 hydrogen atoms in the group are independently substituted with a corresponding number of substituents. It goes without saying that substituents are only in their possible chemical positions, and that the person skilled in the art is able to determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups having free hydrogen may be unstable when bound to carbon atoms having unsaturated (e.g., olefinic) bonds.
"substituted" or "substituted" as used herein, unless otherwise indicated, means that the group may be substituted with one or more groups selected from the group consisting of: alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, alkenyl, hydroxy, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, amino, haloalkyl, hydroxyalkyl, carboxyl, carboxylate, =o, -C (O) R b 、-OC(O)R b 、-NR b R b 、-C(O)NR b R b 、-NR b C(O)R b 、-S(O)NR b R b or-S (O) 2 NR b R b Wherein R is b The definition of (C) is as described in the general formula (I).
As used herein, the term "subject," "individual," or "patient" is used interchangeably to refer to any animal, including mice, rats, other rodents, rabbits, dogs, cats, pigs, cattle, sheep, horses, primates, and humans. In some embodiments, the patient is a human. In some embodiments, the subject has experienced and/or displayed a disease or at least one symptom of a disease to be treated and/or prevented. In some embodiments, the subject has been determined or diagnosed as a cancer having a KRAS G12 or G13 mutation (e.g., as determined by an FDA-approved regulatory agency, such as an FDA-approved, assay, or kit). In some embodiments, the subject has a tumor positive for a KRAS G12C mutation, a KRAS G12D mutation, a KRAS G12S mutation, a KRAS G12V mutation, a KRAS G12A mutation, a KRAS G13D mutation, or a KRAS G13C mutation (e.g., as determined by regulatory agency-approved assays or kits). The subject may be a tumor patient with a KRAS G12C mutation, a KRAS G12D mutation, a KRAS G12V mutation, a KRAS G12A mutation, a KRAS G13D mutation, or a KRAS G13C mutation (e.g., approved regulatory bodies-such as FDA approved, analytical, or kit). The subject may be a subject whose tumor has a KRAS G12C mutation, a KRAS G12D mutation, a KRAS G12V mutation, a KRAS G12A mutation, a KRAS G13D mutation, or a KRAS G13C mutation (e.g., the tumor is determined by an FDA-approved regulatory agency, kit, or assay). In some embodiments, the subject is suspected of having a KRAS G12 or G13 gene-related cancer. In some embodiments, the subject has a clinical record indicating that the subject has a tumor with a KRAS G12C mutation (and optionally a clinical record indicating that the subject should be treated with any of the compositions provided herein).
The term "pediatric patient" as used herein refers to a patient less than 16 years of age at the time of diagnosis or treatment. The term "child" can also be divided into the following subclasses: neonates (from birth to first month of birth); infants (1 month to two years); children (2 to 12 years); teenagers (12 to 21 years old (up to but not including 22 years of birth)). Berhman RE, kliegman R, arvin AM, nelson we. Nelson textbook, 15 th edition. Philadelphia: w.b. samanders company, 1996; rudolph AM, et al. Pediatric version 21 of rudoffer. New York: mcGrow-Hill, 2002; and Avery MD, first LR. Pediatric medicine, second edition. Balm: williams & Wilkins;1994.
as used herein, an "effective amount" of a compound refers to an amount sufficient to down-regulate or inhibit SOS 1 enzymatic activity.
As used herein, a "therapeutically effective dose" of a compound refers to an amount sufficient to ameliorate or somehow reduce symptoms, stop or reverse progression of a disease, or down regulate or inhibit SOS 1 activity. Such doses may be administered as a single dose or may be administered according to a regimen so as to be effective.
As used herein, "treating" refers to ameliorating or otherwise altering the condition, disorder, or symptom or pathology of a disease in a patient in any manner.
As used herein, "ameliorating a symptom of a particular disease by use of a particular compound or pharmaceutical composition" refers to any reduction, whether permanent or temporary, persistent or temporary, attributable to or associated with the use of the composition.
The definition and use of stereochemistry in the present invention is generally referred to in the following documents:
S.P. Parker, ed., mcGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hillbook Company, new York; and Eliel, e.and Wilen, s., "Stereochemistry of Organic Compounds", john Wiley & Sons, inc., new York,1994. The compounds of the invention may contain asymmetric or chiral centers and thus exist as different stereoisomers. All stereoisomeric forms of the compounds of the invention, including, but in no way limited to, diastereomers, enantiomers, atropisomers, and mixtures thereof, such as racemic mixtures, form part of the present invention. Diastereomers can be separated into the individual diastereomers by chromatography, crystallization, distillation, or sublimation, based on their physical-chemical differences. Enantiomers may be converted into diastereomeric mixtures by separation by reaction with an appropriate optically active compound (e.g., a chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers, and converting the individual diastereomers to the corresponding pure enantiomers. The intermediates and compounds of the invention may also exist in different tautomeric forms and all such forms are encompassed within the scope of the invention. Many organic compounds exist in optically active form, i.e. they have the ability to rotate the plane of plane polarized light. In describing optically active compounds, the prefix D, L or R, S is used to denote the absolute configuration of the chiral center of the molecule. The prefix d, l or (+), (-) is used to name the sign of the compound plane polarization rotation, where (-) or l means that the compound is left-handed and the prefix (+) or d means that the compound is right-handed. The atoms or groups of atoms of these stereoisomers are connected in the same order but in different steric structures. The particular stereoisomer may be an enantiomer, and the mixture of isomers is commonly referred to as an enantiomeric mixture. 50: mixtures of enantiomers of 50 are referred to as racemic mixtures or racemates, which may result in no stereoselectivity or stereospecificity during chemical reactions. The terms "racemic mixture" and "racemate" refer to a mixture of two enantiomers in equimolar amounts, lacking optical activity.
"tautomer" or "tautomeric form" refers to isomers of structures of different energies that can be interconverted by a low energy barrier. For example, proton tautomers (i.e., proton-shifted tautomers) include tautomerism by proton shift, such as keto-enol and imine-enamine isomerisation. Valency (valence) tautomers include tautomers that reorganize into bond electrons. Unless otherwise indicated, the structural formulae described herein include all isomeric forms (e.g., enantiomers, diastereomers, and geometric isomers): for example, R, S configuration containing asymmetric centers, the (Z), (E) isomers of double bonds, and the conformational isomers of (Z), (E). Thus, individual stereochemical isomers of the compounds of the invention, or enantiomers, diastereomers, or mixtures of geometric isomers thereof, are all within the scope of the invention.
By "pharmaceutically acceptable salts" is meant salts of the compounds of the present invention which are safe and effective when used in the human or animal body. Salts of the compounds may be obtained by dissolving the corresponding addition salts in pure solution or in a suitable inert solvent with sufficient amounts of base or acid. Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia, magnesium salts, and the like, and pharmaceutically acceptable acid addition salts include inorganic and organic acid salts including hydrochloric acid, hydrobromic acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, monohydrogen sulfate, acetic acid, maleic acid, malonic acid, succinic acid, fumaric acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, methanesulfonic acid, and the like (see Berge et al, "Pharmaceutical Salts", journal of Pharmaceutical Science 66:1-19 (1977)).
The invention provides an SOS1 inhibitor with a novel structure, and test results show that the polycyclic pyridazinone derivative has excellent SOS1 inhibition activity and excellent safety and selectivity, and can be used for preparing medicines for treating cancers, especially pancreatic cancer, colorectal cancer, lung cancer, hepatocellular carcinoma, renal cancer, gastric cancer, cholangiocarcinoma and other diseases.
Drawings
FIG. 1 is a graph showing the effect of compounds according to the present invention on KRAS/ERK1/2 signaling pathways of K-562 cells.
FIG. 2 is a graph showing the effect of compounds of the present invention on KRAS/ERK1/2 signaling pathways of K-562 cells.
Detailed Description
The method of the present invention will be described by way of specific examples, so that the technical solution of the present invention can be understood and grasped more easily, but the present invention is not limited thereto. In the following examples 1 The H NMR spectrum was determined with a Bruker instrument (400 MHz) and the chemical shifts were expressed in ppm. Tetramethylsilane internal standard (0.00 ppm) was used. 1 H NMR representation method: s=singlet, d=doublet, t=triplet, q=quartet, m=multiplet, br=broad, dd=doublet of doublet, dt=doublet of triplet. If coupling constants are provided, they are in Hz.
The mass spectrum is measured by an LC/MS instrument, and the ionization mode is ESI.
High performance liquid chromatograph model: agilent 1260, siemens flying U3000; chromatographic column model: waters xbridge C18 (4.6. Times.150 mm,3.5 μm); mobile phase: ACN, B Water (0.1% H) 3 PO 4 ) The method comprises the steps of carrying out a first treatment on the surface of the Flow rate: 1.0mL/min; gradient: 5%A for 1min,increase to 20%A within 4min,increase to 80%A within 8min,80%A for 2min,back to 5%A within 0.1min; wavelength: 220nm; column incubator: 35 ℃.
The thin layer chromatography silica gel plate uses a smoke table yellow sea HSGF254 or Qingdao GF254 silica gel plate, the specification of the silica gel plate used by the Thin Layer Chromatography (TLC) is 0.2mm-0.3mm, and the specification of the thin layer chromatography separation and purification product is 0.4mm-0.5mm.
Column chromatography generally uses tobacco stage yellow sea silica gel 200-300 mesh silica gel as carrier.
In the following examples, unless otherwise indicated, all temperatures are in degrees celsius and unless otherwise indicated, various starting materials and reagents are either commercially available or synthesized according to known methods, and are used without further purification, and unless otherwise indicated, commercially available manufacturers include, but are not limited to, the national pharmaceutical community, the carbofuran technologies, the tencel (Shanghai) chemical industry development limited, the Shanghai Pico pharmaceutical technologies limited, the Shanghai Michelson chemical technologies limited, and the like.
CD 3 OD: deuterated methanol
CDCl 3 : deuterated chloroform
DMSO-d 6 : deuterated dimethyl sulfoxide
Pd 2 (dba) 3 : tris (dibenzylideneacetone) dipalladium
Pd(dppf)Cl 2 : [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
XantPhos:4, 5-bis-diphenylphosphine-9, 9-dimethylxanthene
XPhos: 2-dicyclohexylphosphorus-2, 4, 6-triisopropylbiphenyl
HATU:2- (7-Oxybenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate
TLC: thin layer chromatography
HPLC: high performance liquid chromatography
purity: purity of
And (3) the following steps: and
the hydrogen atmosphere is defined as the reaction flask being connected to a hydrogen balloon of about 1L volume.
The examples are not particularly described, and the solution in the reaction is an aqueous solution.
The examples are not specifically described, and the reaction temperature is room temperature and is 20℃to 30 ℃.
The monitoring of the progress of the reaction in the examples employed Thin Layer Chromatography (TLC), the developing reagent used for the reaction, the system of eluent for column chromatography employed for purifying the compound or the developing reagent system of thin layer chromatography included: a: petroleum ether and ethyl acetate systems; b: methylene chloride and methanol systems; c: n-hexane: ethyl acetate; the volume ratio of the solvent is different according to the polarity of the compound, and can be adjusted by adding a small amount of acidic or alkaline reagent, such as acetic acid or triethylamine.
Preparation of intermediates
Intermediate 1
(R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethanamine IN-1
First step 1- (3-nitro-5- (trifluoromethyl) phenyl) ethan-1-one IN-1b
3-bromo-5-nitrobenzotrifluoride IN-1a (2.0 g,7.41 mmol), tributyl (1-ethoxyethylene) tin (3.5 g,9.69 mmol) and ditolylphosphoropalladium dichloride (520 mg,0.74 mmol) were added sequentially to toluene (25 mL), heated to 100deg.C under nitrogen protection overnight, and TLC showed complete reaction. The reaction solution was cooled to room temperature, hydrochloric acid (15 ml,3 n) was added, stirring was performed for 30 minutes, celite was added, the filtrate was separated, the aqueous phase was extracted with ethyl acetate, the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-1b (1.25 g, yield 72%) as a yellow oil.
1 H NMR(400MHz,CDCl 3 )δ8.94(s,1H),8.68(s,1H),8.53(s,1H),2.75(s,3H).
Second step (R, Z) -2-methyl-N- (1- (3-nitro-5- (trifluoromethyl) phenyl) ethylene) propane-2-sulfinamide IN-1c
A mixture of compound IN-1b (1.25 g,5.36 mmol), (R) - (+) -tert-butylsulfinamide (974 mg,8.04 mmol) and tetraethyl titanate (10 mL,47.70 mmol) was heated to 80℃for 3 hours and the TLC showed little starting material remaining. The reaction solution was cooled to room temperature, poured into ice water (60 mL), extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was chromatographed on silica gel to give the title compound IN-1c (1.01 g, yield 56%) as a yellow oil.
Third step (R) -2-methyl-N- ((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) propane-2-sulfinamide IN-1d
Compound IN-1c (260 mg,0.77 mmol) was dissolved IN tetrahydrofuran (2.5 mL) and water (0.05 mL), cooled to-60℃and sodium borohydride (74 mg,1.95 mmol) was added IN portions, and after addition, stirring was continued for 1 hour at-60℃until TLC showed complete reaction. The reaction solution was quenched with water, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-1d (150 mg, yield 58%) as a white solid.
1 H NMR(400MHz,CDCl 3 )δ8.43-8.42(m,2H),7.95(s,1H),4.75-4.69(m,1H),3.55(d,J=4.4Hz,1H),1.61(d,J=6.8Hz,3H),1.25(s,9H).
Fourth step (R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethanamine IN-1
Compound IN-1d (164 mg,0.48 mmol) was dissolved IN tetrahydrofuran (3 mL), concentrated hydrochloric acid (0.5 mL) was added dropwise, and after the addition, the reaction was completed by TLC at room temperature for 1 hour. The reaction was added dropwise to saturated aqueous sodium carbonate to adjust ph=8, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound IN-1 (105 mg, crude) as a yellow oil, which was used directly IN the next step.
LC-MS:m/z=235.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.56(s,1H),8.32(s,1H),8.24(s,1H),4.24(q,J=6.8Hz,1H),2.22(br,2H),1.30(d,J=2.8Hz,3H).
Intermediate 2
(R) -6-bromo-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) phthalazin-1 (2H) -one IN-2
First step 4-bromo-2-iodobenzoic acid methyl ester IN-2b
Methyl 2-amino-4-bromobenzoate IN-2a (2.0 g,8.70 mmol) was dispersed IN hydrochloric acid (20 mL,120mmol, 6M), cooled to about 0deg.C, dropwise added with a solution of sodium nitrite (360 mg,5.22 mmol) IN water (1 mL), reacted for 1 hour, dropwise added with potassium iodide (1.4 g,8.43 mmol), reacted for 2 hours at room temperature, and TLC detected the raw materials to be substantially complete. The reaction solution was poured into water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-2b (2.1 g, yield 70%) as yellow oil.
Second step 4-bromo-2-cyanobenzoic acid methyl ester IN-2c
Compound IN-2b (2.1 g,6.16 mmol) was dissolved IN N-methylpyrrolidone (10 mL), cuprous cyanide (284 mg,9.31 mmol) was added at room temperature, and the reaction was allowed to proceed overnight at 60℃with TLC to detect complete reaction of starting material. The reaction solution was cooled to room temperature, filtered through celite, washed with filter cake, poured into water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-2c (1.2 g, yield 80%) as a white solid.
Third step 5-bromo-3-aminoisoindol-1-one IN-2d
Compound IN-2c (1.2 g,5.00 mmol) was dissolved IN methanol (20 mL), cooled to 0deg.C under nitrogen protection, and after ammonia gas was introduced to saturation, the reaction was slowly returned to room temperature for 48 hours, and TLC detected that the starting material was substantially complete. The reaction solution was concentrated to give the title compound IN-2d (1.05 g, crude) as a yellow solid, which was used directly IN the next step.
LC-MS:m/z=225.0[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ10.27(br,1H),9.69(br,1H),8.30(s,1H),7.88(dd,J=8.0,1.6Hz,1H),7.64(d,J=8.0Hz,1H).
Fourth step (R) -5-bromo-3- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -1H-isoindol-1-one IN-2e
Compound IN-2d (1.05 g, crude) and intermediate IN-1 (1.6 g,6.83 mmol) were dissolved IN isopropanol (50 mL), heated to 90℃and reacted for 48 hours, and TLC detected some starting material remained. The reaction solution was concentrated to give the title compound IN-2e (2.3 g, crude) as a pale yellow solid, which was used directly IN the next step.
LC-MS:m/z=442.0[M+H] +
Fifth step (R) -6-bromo-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) phthalazin-1 (2H) -one IN-2
Compound IN-2e (2.25 g, crude) was dissolved IN ethanol (40 mL) and hydrazine hydrate (636 mg,10.16mmol, 80%) was added at room temperature and heated to 40℃for 2 hours, and TLC checked for complete reaction of starting materials. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-2 (1.1 g, three-step yield 48%) as a yellow solid.
LCMS:m/z=457.0[M+H] +
Intermediate 3
(R) -7-chloro-1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4- (3H) -one IN-3
First step 6-chloro-4-iodonicotinic acid IN-3b
2, 6-tetramethylpiperidine (11.2 g,79.29 mmol) was dissolved IN tetrahydrofuran (100 mL), cooled to about-60℃under nitrogen protection, n-butyllithium (31.7 mL,79.25mmol, 2.5M) was added dropwise, slowly returned to 0℃for half an hour, cooled to about-60℃again, 6-chloronicotinic acid IN-3a (5.0 g,31.74 mmol) was added IN portions, stirring was continued for 1 hour at-60℃after addition, iodine (9.7 g,38.22 mmol) was added dropwise, and the reaction was slowly returned to room temperature overnight, TLC indicated that most of the starting material had reacted. The reaction mixture was quenched with 1N hydrochloric acid, filtered, and the filter cake was washed and dried to give the title compound IN-3b (4.7 g, crude) as a tan solid, which was used directly IN the next step.
Second step 6-chloro-4-iodonicotinic acid methyl ester IN-3c
Compound IN-3b (4.7 g, crude) was dissolved IN N, N-dimethylformamide (30 mL), potassium carbonate (6.9 g,49.92 mmol) was added, stirred at room temperature for 10 minutes, methyl iodide (4.7 g,33.11 mmol) was added, and the reaction was carried out at room temperature overnight, and the completion of the reaction was detected by TLC. The reaction mixture was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-3c (2.0 g, 21% yield IN two steps) as a pale yellow solid.
Third step 6-chloro-4-vinylnicotinic acid methyl ester IN-3d
Compound IN-3c (3.0 g,10.08 mmol) and potassium vinyltrifluoroacetate (1.35 g,10.08 mmol) were dissolved IN 1, 4-dioxane (36 mL) and water (12 mL), triethylamine (3.06 g,30.24 mmol) and Pd (dppf) Cl were added at room temperature 2 Dichloromethane complex (200 mg,0.24 mmol), nitrogen displacement 3 times, heating to 80 ℃ for 5 hours, LCMS monitored reaction was complete. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-3d (1.9 g, yield 95%) as a pale yellow solid.
LC-MS:m/z=198.1[M+H] +
Fourth step 6-chloro-4-formylnicotinic acid methyl ester IN-3e
Compound IN-3d (1.8 g,9.11 mmol) was dissolved IN a mixed solvent of ethanol (20 mL), tetrahydrofuran (10 mL) and water (10 mL), sodium periodate (19.2 g,89.76 mmol) was added at room temperature, a water (small amount) solution of potassium osmium sulfate (catalytic amount) was added dropwise with stirring, and the reaction was continued at room temperature for 0.5 hour after the addition, and the reaction was completed by TLC. The reaction mixture was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by column chromatography to give the title compound IN-3e (1.3 g, yield 72%) as pale yellow oil (solidification on standing).
Fifth step (E) -6-chloro-4- ((hydroxyimino) methyl) nicotinic acid methyl ester IN-3f
Compound IN-3e (1.0 g,5.01 mmol) and hydroxylamine sulfonic acid (1.70 g,15.04 mmol) were dissolved IN water (15 mL) and reacted overnight at room temperature with complete TLC. The reaction mixture was extracted with water and ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound IN-3f (1.05 g, crude) as an off-white solid, which was used directly IN the next step.
LCMS:m/z=215.1[M+H] +
Sixth step 6-chloro-4-cyanonicotinic acid methyl ester IN-3g
Compound IN-3f (1.05 g, crude) was dissolved IN phosphorus oxychloride (10 mL), heated to 100deg.C and reacted for 2 hours, and TLC checked for completion of the reaction. The reaction solution was cooled to room temperature, concentrated, and the residue was poured into ice water, neutralized with aqueous sodium bicarbonate, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-3g (800 mg, two-step yield 81%) as a white solid.
LCMS:m/z=197.1[M+H] +
Seventh step 6-chloro-1-imino-1, 2-dihydro-3H-pyrrolo [3,4-c ] pyridin-3-one IN-3H
Compound IN-3g (800 mg,4.07 mmol) was dissolved IN methanol (60 mL), cooled to about 0deg.C, saturated with ammonia gas, and the reaction was slowly warmed to room temperature overnight, and TLC showed complete reaction of starting material. The reaction solution was concentrated to give the title compound IN-3h (766 mg, crude) as a pale yellow solid, which was used directly IN the next step.
LCMS:m/z=182.1[M+H] +
Eighth step (R) -6-chloro-1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -3H-pyrrolo [3,4-c ] pyridin-3-one IN-3i
Compound IN-3h (766 mg, crude) was dissolved IN isopropanol (30 mL), intermediate IN-1 (988 mg,4.22 mmol) was added at room temperature, heated to 85deg.C and reacted overnight, and TLC detected complete reaction of starting materials. The reaction solution was cooled to room temperature and concentrated to give the title compound IN-3i (1.75 g, crude) as a yellow solid, which was used directly IN the next step.
LCMS:m/z=399.1[M+H] +
Ninth step (R) -7-chloro-1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4- (3H) -one IN-3
Compound IN-3i (1.75 g, crude) was dissolved IN ethanol (15 mL) and hydrazine hydrate (579 mg,9.25mmol, 80%) was added at room temperature and allowed to react at 40℃for 1 hour, with TLC detecting complete reaction of starting material. The reaction solution was cooled to room temperature, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-3 (1.0 g, three-step yield 59%) as a yellow solid.
LCMS:m/z=414.4[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.86(s,1H),9.19(s,1H),8.54(s,1H),8.43(s,1H),8.34(s,1H),8.26(s,1H),7.41(d,J=2.8Hz,1H),5.18-5.08(m,1H),1.57(d,J=2.8Hz,3H).
Example 1
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7-methoxy-2-methyl-6- (((S) -tetrahydrofurane-3-yl) oxy) phthalazin-1 (2H) -one 1
First step 5-bromo-4-formyl-2-methoxyphenylacetic acid ester 1b
Acetylvanillin 1a (20.0 g,103.0 mmol) and potassium bromide (39.96 g,335.8 mmol) were dissolved in water (100 mL) and cooled to 0deg.C, and bromine (18.27 g,114.3 mmol) was added dropwise. After the addition was completed and stirred for 15 hours at room temperature, TLC showed little remaining starting material. The reaction solution was filtered, and the filter cake was washed with water and dried to give the title compound 1b (crude aqueous product) which was used directly in the next step.
Second step 2-bromo-4-hydroxy-5-methoxybenzaldehyde 1c
Compound 1b (crude) was dissolved in concentrated hydrochloric acid (400 mL, 6N), warmed to 90℃and reacted overnight, and TLC checked the starting material for substantially complete reaction. The reaction solution was cooled to room temperature, filtered, and the filter cake was washed and dried to give the title compound 1c (16.8 g, crude) which was used directly in the next step.
Third step (R) -3-hydroxytetrahydrofuran p-toluenesulfonate 1e
(R) -3-hydroxytetrahydrofuran 1d (1.0 g,11.35 mmol) was dissolved in dichloromethane (8 mL), cooled to 0deg.C, pyridine (2.5 g,31.61 mmol) was added, p-toluenesulfonyl chloride (3.03 g,15.89 mmol) was added in portions, and the mixture was allowed to react overnight at room temperature. The reaction solution was concentrated, diluted with water, the aqueous phase was adjusted to ph=1 with hydrochloric acid (3N), extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was chromatographed on silica gel to give the title compound 1e (2.57 g, three steps yield 93%) as a colorless oil.
1 H NMR(400MHz,CDCl 3 )δ7.78(d,J=8.4Hz,2H),7.35(d,J=8.4Hz,2H),5.11-5.09(m,1H),3.91-3.77(m,4H),2.45(s,3H),2.10-2.02(m,2H).
Fourth step (S) -2-bromo-5-methoxy-4- ((tetrahydrofuran-3-yl) oxy) benzaldehyde 1f
Compound 1c (3.7 g, crude), compound 1e (4.3 g,17.75 mmol) and cesium carbonate (7.8 g,24.0 mmol) were dispersed in N, N-dimethylformamide (20 mL), warmed to 100deg.C and reacted overnight, and LCMS detected complete reaction of starting materials (TLC indicated one point for starting materials and product). The reaction solution was cooled to room temperature, diluted with ethyl acetate, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to give the title compound 1f (4.7 g, crude product) which was used directly in the next step.
Fifth step (S) -2-bromo-5-methoxy-4- ((tetrahydrofuran-3-yl) oxy) benzoic acid 1g
Potassium permanganate (1.94 g,12.29 mmol) and compound 1f (3.7 g, crude) were dispersed in water (120 mL), warmed to 75deg.C and stirred for 5 hours, and TLC detection was complete. The reaction solution was cooled to room temperature, diluted with water, filtered, the filter cake was washed with water and ethyl acetate, the filtrate was separated, the organic phase was discarded, and the aqueous phase was extracted 1 time with ethyl acetate. The aqueous phases were combined, diluted hydrochloric acid (1N) was added to adjust acidity, a large amount of white solid was precipitated, filtered, and the filter cake was washed and dried to give 1g (2.0 g, crude product) of the title compound, which was used directly in the next step.
LC-MS:m/z=317.02[M+H] +
Sixth step (S) -2-formyl-5-methoxy-4- ((tetrahydrofuran-3-yl) oxy) benzoic acid for 1h
1g (2.4 g, crude) of the compound was dissolved in tetrahydrofuran (50 mL), cooled to-78℃under nitrogen protection, N-butyllithium (6.8 mL,17.0mmol,2.5M N-hexane solution) was added dropwise, stirred for half an hour at-78℃was completed, N-dimethylformamide (719 mg,9.84 mmol) was added dropwise, the reaction was slowly warmed to room temperature, and TLC detection was complete. The reaction mixture was diluted with hydrochloric acid (1N) to adjust to weak acidity, extracted with ethyl acetate, combined with organic phase, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and purified by silica gel column chromatography to give the title compound (1.4 g, three-step yield 21%) for 1 h.
Seventh step (S) -7-methoxy-6- ((tetrahydrofuran-3-yl) oxy) phthalazin-1 (2H) -one 1i
Compound 1h (1.0 g,3.76 mmol) and aqueous hydrazine hydrate (1.96 g,31.36mmol, 80%) were dissolved in isopropanol (10 mL), warmed to 80deg.C and stirred for 3 h, and TLC detected the starting material was essentially complete. The reaction solution was cooled to room temperature, diluted with water, concentrated to remove isopropanol, solid precipitated, filtered, and the cake was washed and dried to give the title compound 1i (800 mg, crude product) which was used directly in the next step.
LC-MS:m/z=263.1[M+H] +
Eighth step (S) -4-bromo-7-methoxy-6- ((tetrahydrofuran-3-yl) oxy) phthalazin-1 (2H) -one 1j
Compound 1i (500 mg, crude) benzyl trimethyl ammonium tribromide (1.49 g,3.82 mmol) and potassium carbonate (528 mg,3.82 mmol) were dispersed in N, N-dimethylformamide (10 mL), warmed to 40℃and stirred for 5 hours, and TLC detected that the starting material was substantially completely reacted. The reaction mixture was diluted with ethyl acetate, washed with saturated brine, concentrated, and purified by Prep-TLC to give the title compound 1j (538 mg, 67% yield in two steps).
Ninth step (S) -4-bromo-7-methoxy-2-methyl-6- ((tetrahydrofuran-3-yl) oxy) phthalazin-1 (2H) -one 1k
Compound 1j (492 mg,1.44 mmol) was dissolved in N, N-dimethylformamide (5 mL), sodium hydride (699 mg,1.73mmol, 60%) was added, stirred at room temperature for 30 minutes, methyl iodide (409 mg,2.88 mmol) was added, and stirred at room temperature for 2 hours, and the TLC detection of the starting materials was essentially complete. The reaction mixture was added with water, and a white solid was precipitated, filtered, and the filter cake was washed and dried to give the title compound 1k (461 mg, crude product) which was used directly in the next step.
Tenth step 7-methoxy-2-methyl-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (((S) -tetrahydrofurane-3-yl) oxy) phthalazin-1 (2H) -one 1l
Compound 1k (161 mg, crude), intermediate IN-1 (130 mg,0.56 mmol), potassium tert-butoxide (101 mg,0.90 mmol), BINAP (13 mg,0.02 mmol) and Pd 2 (dba) 3 (21 mg,0.02 mmol) was dispersed in 1, 4-dioxane (5 mL), and the mixture was stirred overnight at 100℃under nitrogen. TLC detectionThe raw materials react completely. The reaction was cooled to room temperature, filtered with celite, washed with ethyl acetate, the filtrate concentrated and the crude Prep-TLC purified to give the title compound 1l (50 mg, 20% yield in two steps).
LC-MS:m/z=509.2[M+H] +
Eleventh step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7-methoxy-2-methyl-6- (((S) -tetrahydrofurane-3-yl) oxy) phthalazin-1 (2H) -one 1
Compound 1l (52 mg,0.10 mmol) and palladium on carbon (5 mg, 10%) were dispersed in methanol (5 mL) and reacted at room temperature under hydrogen atmosphere for 3 hours, and the reaction was essentially complete by TLC. The reaction was filtered through celite, the filter cake was washed with methanol, the filtrate was concentrated, and the crude Prep-TLC was purified to give the title compound 1 (22 mg, yield 46%).
LC-MS:m/z=479.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ7.70(s,1H),7.59(s,1H),6.98(s,2H),6.78(s,1H),5.32-5.29(m,1H),4.99(q,J=6.8Hz,1H),4.08-3.99(m,3H),3.98-3.89(m,4H),3.56(s,3H),2.40-2.31(m,1H),2.24-2.18(m,1H),1.58(d,J=6.8Hz,3H).(98.25%purity by HPLC)
Example 2
(R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2-methyl-6- (4-methylpiperazin-1-yl) phthalazin-1 (2H) -one 2
First step (R) -6-bromo-2-methyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) phthalazin-1 (2H) -one 2a
Intermediate IN-2 (300 mg,0.66 mmol) was dissolved IN N, N-dimethylformamide (5 mL), potassium carbonate (182 mg,1.32 mmol) was added, stirred at room temperature for 5 min, methyl iodide (280 mg,1.97 mmol) was added, the reaction was carried out overnight at room temperature, and TLC showed complete reaction of starting materials. The reaction solution was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 2a (280 mg, crude) as a yellow solid, which was used directly in the next step
Second step (R) -2-methyl-6- (4-methylpiperazin-1-yl) -4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) phthalazin-1 (2H) -one 2c
Compound 2a (280 mg, crude) and N-methylpiperazine 2b (298 mg,2.97 mmol) were dissolved in 1, 4-dioxane (30 mL), cesium carbonate (284 mg,1.18 mmol), BINAP (60 mg,0.10 mmol) and Pd were added at room temperature 2 (dba) 3 (60 mg,0.06 mmol) was replaced with nitrogen 3 times and heated to 100deg.C overnight, TLC showed complete reaction of starting materials. The reaction solution was cooled to room temperature, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 2c (240 mg, two-step yield 74%) as a tan solid.
LCMS:m/z=491.2[M+H] +
Third step (R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2-methyl-6- (4-methylpiperazin-1-yl) phthalazin-1 (2H) -one 2
Compound 2c (240 mg,0.49 mmol) was dissolved in ethyl acetate (10 mL) and methanol (3 mL), palladium on carbon (50 mg, 10%) was added, concentrated hydrochloric acid (3 drops) was added dropwise, and the reaction was allowed to proceed at room temperature under a hydrogen atmosphere for 2 hours, and TLC indicated complete reaction of the starting materials. The reaction was neutralized with methanolic ammonia, filtered through celite, and the filtrate concentrated, and the crude product was purified by Prep-TLC to give the title compound 2 (126 mg, yield 56%) as a pale yellow solid.
LCMS:m/z=461.3[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.01(d,J=9.2Hz,1H),7.48(d,J=2.0Hz,1H),7.40(dd,J=9.2,2.0Hz,1H),6.91(d,J=7.2Hz,1H),6.85(s,2H),6.67(s,1H),5.49(s,2H),4.96-4.89(m,1H),3.46(s,4H),3.39(s,3H),2.58(s,4H),2.31(s,3H),1.51(d,J=7.2Hz,3H).(99.72%purity by HPLC)
Example 3
(R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (4-methylpiperazin-1-yl) phthalazin-1 (2H) -one 3
First step (R) -6- (4-methylpiperazin-1-yl) -4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) phthalazin-1 (2H) -one 3a
Intermediate IN-2 (200 mg,0.44 mmol) and N-methylpiperidine 2b (219 mg,2.19 mmol) were dissolved IN 1, 4-dioxane (10 mL) and cesium carbonate (430 mg,1.32 mmol), BINAP (40 mg,0.06 mmol) and Pd were added at room temperature 2 (dba) 3 (40 mg,0.04 mmol) was replaced with nitrogen 3 times and heated to 100deg.C overnight, and TLC showed complete reaction of starting materials. The reaction solution was cooled to room temperature, concentrated, and purified by silica gel column chromatography to give the title compound 3a (48 mg, yield 23%) as a tan solid.
Second step (R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (4-methylpiperazin-1-yl) phthalazin-1 (2H) -one 3
Compound 3a (48 mg,0.10 mmol) was dissolved in ethyl acetate (10 mL) and methanol (3 mL), palladium on carbon (50 mg, 10%) was added, concentrated hydrochloric acid (2 drops) was added dropwise, and the reaction was allowed to proceed at room temperature under a hydrogen atmosphere for 2 hours, and TLC was used to detect completion of the starting material reaction. The reaction was neutralized with methanolic ammonia, filtered through celite, and the filtrate concentrated, and the crude product was purified by Prep-TLC to give the title compound 3 (20 mg, yield 44%) as a pale yellow solid.
LCMS:m/z=447.3[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.11(s,1H),7.99(d,J=9.2Hz,1H),7.50(s,1H),7.40(d,J=9.2Hz,1H),6.83-6.72(m,3H),6.66(s,1H),5.47(s,2H),4.95-4.83(m,1H),3.47(s,4H),2.58(s,3H),2.31(s,3H),1.49(d,J=6.8Hz,3H).(97.85%purity by HPLC)
Example 4
(R) -1- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -3-methyl-7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 4
First step (R) -7-chloro-3-methyl-1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4- (3H) -one 4a
Intermediate IN-3 (460 mg,1.11 mmol) was dissolved IN N, N-dimethylformamide (5 mL), potassium carbonate (268 mg,1.94 mmol) was added, and after stirring at room temperature for 5 minutes, methyl iodide (275 mg,1.94 mmol) was added and the reaction was stirred at room temperature for 2 hours and was essentially complete by TLC. The reaction solution was extracted with water and ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 4a (430 mg, yield 90%) as a yellow solid.
LCMS:m/z=428.2[M+H] +
Second step (R) -3-methyl-7- (4-methylpiperazin-1-yl) -1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4- (3H) -one 4b
Compound 4a (150 mg,0.35 mmol) was dissolved in dioxane (10 mL) and cesium carbonate (352 mg,1.08 mmol), N-methylpiperazine (180 mg,1.80 mmol), pd was added at room temperature 2 (dba) 3 (50 mg,0.05 mmol) and BINAP (50 mg,0.08 mmol), after 3 nitrogen substitutions, heat to 100deg.C for 5 hours, and TLC detects complete reaction of starting materials. The reaction solution was cooled to room temperature, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 4b (105 mg, yield 61%) as a yellow solid.
LCMS:m/z=492.2[M+H] +
Third step (R) -1- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -3-methyl-7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 4
Compound 4b (105 mg,0.21 mmol) was dissolved in ethyl acetate (10 mL) and methanol (2 mL), concentrated hydrochloric acid (3 drops) and 10% palladium on carbon (catalytic amount) were added, and the reaction was allowed to proceed at room temperature under a hydrogen atmosphere for 2 hours, and TLC was used to detect completion of the starting material reaction. The reaction was neutralized with methanolic ammonia, filtered through celite, the filter cake was washed, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 4 (70 mg, yield 70%) as a yellow solid.
LCMS:m/z=462.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.04(s,1H),7.29(s,1H),6.96(s,2H),6.78(s,1H),4.94(q,J=6.8Hz,1H),3.97(s,4H),3.49(s,3H),2.97(s,4H),2.64(s,3H),1.58(d,J=6.8Hz,3H).(98.96%purity by HPLC)
Example 5
(R) -1- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4- (3H) -one 5
First step (R) -7- (4-methylpiperazin-1-yl) -1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4 (3H) -one 5a
Intermediate IN-3 (150 mg,0.36 mmol) was dissolved IN 1, 4-dioxane (10 mL) and cesium carbonate (352 mg,1.08 mmol), N-methylpiperazine 2b (180 mg,1.80 mmol), pd was added at room temperature 2 (dba) 3 (50 mg,0.05 mmol) and BINAP (50 mg,0.08 mmol), after 3 nitrogen substitutions, heat to 100deg.C overnight and TLC checked the completion of the starting material. The reaction was cooled to room temperature, concentrated, and purified by column chromatography on crude silica gel to give the title compound 5a (40 mg, crude) (TLC purity about 60%) as a yellow solid, which was used directly in the next step.
Second step (R) -1- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4- (3H) -one 5
Compound 5a (40 mg, crude) was dissolved in ethyl acetate (5 mL) and methanol (1 mL), concentrated hydrochloric acid (2 drops) and 10% palladium on carbon (catalytic amount) were added, and the reaction was carried out at room temperature under a hydrogen atmosphere for 2 hours, and TLC detected that the starting material was complete. After the reaction solution was neutralized with aminomethylol, it was filtered through celite pad, the cake was washed, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 5 (12 mg, yield 7% in two steps) as a yellow solid.
LCMS:m/z=448.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.04(s,1H),7.27(s,1H),6.92(s,2H),6.77(s,1H),4.94(q,J=6.8Hz,1H),3.95(s,4H),2.90(s,4H),2.60(s,3H),1.57(d,J=7.2Hz,3H).(97.91%purity by HPLC)
Example 6
(R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6-morpholino phthalazin-1 (2H) -one 6
First step (R) -6-morpholino-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) phthalazin-1 (2H) -one 6b
Intermediate IN-2 (200 mg,0.44 mmol) and morpholine 6a (381 mg,4.37 mmol) were dissolved IN 1, 4-dioxane (20 mL) under nitrogen and Pd was added at room temperature 2 (dba) 3 (82 mg,0.09 mmol), BINAP (82 mg,0.13 mmol) and cesium carbonate (428 mg,1.31 mmol) were reacted overnight at 100℃and TLC detected complete reaction of starting materials. The reaction solution was concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 6b (55 mg, yield 27%).
LC-MS:m/z=464.2[M+H] +
Second step (R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6-morpholino-phthalazin-1 (2H) -one 6
Compound 6b (55 mg,0.12 mmol) was dissolved in methanol (10 mL), palladium on carbon (20 mg, 10%) was added and reacted at room temperature under a hydrogen atmosphere for 2 hours, and the reaction was complete by TLC. The reaction was filtered, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 6 (16 mg, yield 31%).
LC-MS:m/z=434.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.12(d,J=8.8Hz,1H),7.45(s,1H),7.39(d,J=9.2Hz,1H),6.98(s,2H),6.80(s,1H),4.98(q,J=6.8Hz,1H),3.91-3.83(m,4H),3.50-3.40(m,4H),1.57(d,J=6.8Hz,3H).(96.33%purity by HPLC)
Example 7
(R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (4-methyl-3-oxopiperazin-1-yl) phthalazin-1 (2H) -one 7
First step 3-Oxopiperazine-1-carboxylic acid benzyl ester 7b
2-piperazinone 7a (1.0 g,9.99 mmol) was dissolved in ethyl acetate (20 mL), water (20 mL) and sodium carbonate (3.2 g,30.19 mmol) were added, stirred at room temperature for 10 min, cooled to 0deg.C, benzyl chloroformate (2.1 g,12.31 mmol) was slowly added dropwise, stirred at room temperature after addition, and the reaction was complete by TLC. The reaction mixture was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography on crude silica gel to give the title compound 7b (2.3 g, yield 98%).
LC-MS:m/z=235.1[M+H] +
Second step 4-methyl-3-oxopiperazine-1-carboxylic acid benzyl ester 7c
Compound 7b (2.3 g,9.82 mmol) was dissolved in N, N-dimethylformamide (30 mL), cooled to 0deg.C, sodium hydride (780 mg,19.50mmol, 60%) was added in portions, stirring was continued for 20 minutes after the addition, methyl iodide (2.8 g,19.73 mmol) was added, and stirring was continued for 2 hours at room temperature. The reaction solution was quenched with water, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 7c (2.0 g, yield 82%).
LC-MS:m/z=249.1[M+H] +
Third step 1-methylpiperazin-2-one 7d
Compound 7c (2.0 g,8.06 mmol) was dissolved in ethyl acetate (30 mL) and palladium on carbon (1.0 g, 10%) was added and reacted overnight at room temperature under hydrogen atmosphere, and the reaction was complete by TLC. The reaction was filtered, the filter cake was rinsed 3 times with ethyl acetate, and the filtrate was concentrated to give the title compound 7d (950 mg, crude) which was used directly in the next step.
LC-MS:m/z=115.2[M+H] +
Fourth step (R) -6- (4-methyl-3-oxopiperazin-1-yl) -4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amine) phthalazin-1 (2H) -one 7e
Compound 7d (200 mg,0.44 mmol) and intermediate IN-2 (75 mg,0.66 mmol) were dissolved IN toluene (10 mL) and Pd was added at room temperature 2 (dba) 3 (80 mg,0.09 mmol), BINAP (82 mg,0.13 mmol) and cesium carbonate (328 mg,1.01 mmol) were heated to 100℃and reacted for 4 hours. The reaction solution was concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 7e (38 mg, 18% in two steps).
LC-MS:m/z=491.2[M+H] +
Fifth step (R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (4-methyl-3-oxopiperazin-1-yl) phthalazin-1 (2H) -one 7
Compound 7e (38 mg,0.08 mmol) was dissolved in ethyl acetate (15 mL), palladium on carbon (50 mg, 10%) was added, and the mixture was stirred at room temperature under a hydrogen atmosphere for 2 hours. The reaction was filtered, the filtrate was concentrated, and the crude Prep-TLC purified to give the title compound 7 (18 mg, 51% yield).
LC-MS:m/z=461.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.13(s,1H),8.01(d,J=8.8Hz,1H),7.56-7.35(m,2H),6.85(d,J=7.2Hz,1H),6.79(s,2H),6.66(s,1H),5.48(s,2H),5.10-4.78(m,1H),4.15-4.06(m,2H),3.82-3.69(m,2H),3.57-3.46(m,2H),2.95(s,3H),1.50(d,J=6.8Hz,3H).(98.33%purity by HPLC)
Example 8
(R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methylphthalazin-1 (2H) -one 8
First step (R) -6- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) phthalazin-1 (2H) -one 8b
Compound 2a (100 mg,0.21 mmol) and N, N, N' -trimethylethylenediamine 8a (109 mg,1.07 mmol) were dissolved in 1, 4-dioxane (10 mL), and Pd was added at room temperature 2 (dba) 3 (20 mg,0.04 mmol), BINAP (40 mg,0.06 mmol) and cesium carbonate (140 mg,0.43 mmol) were heated to 100℃for reaction for 4 hours. The reaction solution was concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 8b (50 mg, yield 48%).
LC-MS:m/z=493.3[M+H] +
Second step (R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- ((2- (dimethylamino) ethyl) (methyl) amino) -2-methylphthalazin-1 (2H) -one 8
Compound 8b (50 mg,0.10 mmol) was dissolved in ethyl acetate (10 mL), palladium on carbon (30 mg, 10%) was added, and the mixture was stirred overnight at room temperature under hydrogen atmosphere. The reaction was filtered, the filtrate was concentrated, and the crude Prep-TLC purified to give the title compound 8 (28 mg, 60% yield).
LC-MS:m/z=463.3[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ7.98(d,J=8.8Hz,1H),7.27-7.14(m,2H),6.93-6.84(m,3H),6.68(s,1H),5.47(s,2H),5.01-4.86(m,1H),3.72(t,J=6.4Hz,2H),3.39(s,3H),3.10(s,3H),2.63(s,2H),2.36(s,6H),1.52(d,J=6.8Hz,3H).(98.53%purity by HPLC)
Example 9
(R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- ((2- (dimethylamino) ethyl) (methyl) amino) phthalazin-1 (2H) -one 9
First step 6-bromo-4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -2- (tetrahydro-2H-pyran-2-yl) phthalazin-1 (2H) -one 9a
Intermediate IN-2 (500 mg,1.09 mmol) was dissolved IN tetrahydrofuran (20 mL), 3, 4-dihydro-2H-pyran (276 mg,3.28 mmol) and pyridinium p-toluenesulfonate (55 mg,0.22 mmol) were added and heated to reflux for 3 hours, leaving a major portion of the starting material as detected by TLC. Additional 3, 4-dihydro-2H-pyran (276 mg,3.28 mmol) was heated to reflux overnight leaving a major portion of the starting material unreacted by TLC. 3, 4-dihydro-2H-pyran (460 mg,5.47 mmol) was added and the reaction was heated at reflux overnight with complete TLC. The reaction solution was cooled to room temperature, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 9a (620 mg, yield 105%).
LC-MS:m/z=541.0[M+H] +
Second step 6- ((2- (dimethylamino) ethyl) (methyl) amino) -4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -2- (tetrahydro-2H-pyran-2-yl) phthalazin-1 (2H) -one 9b
Compound 9a (150 mg,0.28 mmol) and N, N, N' -trimethylethylenediamine 8a (142 mg,1.39 mmol) were dissolved in 1, 4-dioxane (10 mL), and Pd was added at room temperature 2 (dba) 3 (51 mg,0.06 mmol), BINAP (52 mg,0.08 mmol) and cesium carbonate (183 mg,0.56 mmol) were heated to 100℃and reacted for 3 hours. The reaction solution was concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 9b (90 mg, yield 58%).
LC-MS:m/z=563.3[M+H] +
Third step (R) -6- ((2- (dimethylamino) ethyl) (methyl) amino) -4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) phthalazin-1 (2H) -one 9c
Compound 9b (90 mg,0.16 mmol) was dissolved in hydrochloric acid/methanol (5 mL,20.0mmol, 4M) and stirred at room temperature for 1 h, and the reaction was complete by TLC. Reaction liquid ammonia methanol neutralization and concentration, crude purification by Prep-TLC gave the title compound 9c (70 mg, 82% yield).
LC-MS:m/z=479.2[M+H] +
Fourth step (R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- ((2- (dimethylamino) ethyl) (methyl) amino) phthalazin-1 (2H) -one 9
Compound 9c (70 mg,0.15 mmol) was dissolved in ethyl acetate (5 mL) and methanol (3 mL) and reacted at room temperature under a hydrogen atmosphere for 2 hours. The reaction solution was filtered, the filtrate was concentrated, and the crude Prep-TLC was purified to give the title compound 9 (18 mg, yield 28%).
LC-MS:m/z=449.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.13(d,J=9.2Hz,1H),7.28-7.23(m,2H),6.95(d,J=1.4Hz,2H),6.77(s,1H),5.00(q,J=6.8Hz,1H),3.85(t,J=7.2Hz,2H),3.20(s,3H),2.99(t,J=6.8Hz,2H),2.65(s,6H),1.58(d,J=6.8Hz,3H).(98.53%purity by HPLC)
Example 10
(R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (4- (dimethylamino) piperidin-1-yl) phthalazin-1 (2H) -one 10
First step 6- (4- (dimethylamino) piperidin-1-yl) -4- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -2- (tetrahydro-2H-pyran-2-yl) phthalazin-1 (2H) -one 10b
Compound 9a (150 mg,0.28 mmol) and 4-dimethylaminopiperidine 10a (178 mg,1.39 mmol) were dissolved in 1, 4-dioxane (10 mL) and Pd was added at room temperature 2 (dba) 3 (51 mg,0.06 mmol), BINAP (52 mg,0.08 mmol) and cesium carbonate (183 mg,0.56 mmol) were heated to 100℃and reacted overnight. The reaction was concentrated and the crude Prep-TLC purified to give the title compound 10b (90 mg, yield 55%).
LC-MS:m/z=589.3[M+H] +
Second step (R) -6- (4- (dimethylamino) piperidin-1-yl) -4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) phthalazin-1 (2H) -one 10c
Compound 10b (90 mg,0.15 mmol) was dissolved in hydrochloric acid/methanol (5 mL,20.0mmol, 4M) and stirred at room temperature for 2 h. The reaction was concentrated to give the title compound 10c (70 mg, crude) which was used directly in the next step.
LC-MS:m/z=505.3[M+H] +
Third step (R) -4- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (4- (dimethylamino) piperidin-1-yl) phthalazin-1 (2H) -one 10
Compound 10c (70 mg, crude) was dissolved in ethyl acetate (5 mL) and methanol (3 mL), and reacted at room temperature under a hydrogen atmosphere for 2 hours. The reaction was filtered, the filtrate was concentrated, and the crude Prep-TLC was purified to give the title compound 10 (36 mg, two-step yield 54%).
LC-MS:m/z=475.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.08(d,J=8.8Hz,1H),7.45(d,J=2.4Hz,1H),7.36(dd,J=9.2,2.4Hz,1H),6.95(s,2H),6.77(s,1H),4.98(q,J=6.8Hz,1H),4.23(d,J=13.2Hz,2H),3.35(s,1H),2.97(t,J=12.0Hz,2H),2.87-2.75(m,1H),2.53(s,6H),2.09(d,J=12.0Hz,2H),1.71-1.61(m,2H),1.58(d,J=6.8Hz,3H).(97.80%purity by HPLC)
Example 11
N- ((R) -1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -3-methyl-4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) pyrrolidin-3-yl) acetamide 11
First step ((R) -1- (3-methyl-1- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) pyrrolidin-3-yl) carbamic acid tert-butyl ester 11b
Compound 4a (130 mg,0.30 mmol) was dissolved in 1, 4-dioxane (8 mL), and (R) -3-t-butoxycarbonylaminopyrrolidine 11a (169 mg,0.91 mmol), pd was added at room temperature 2 (dba) 3 (40 mg,0.04 mmol), BINAP (40 mg,0.06 mmol) and cesium carbonate (198 mg,0.61 mmol), nitrogen substitution 3 times, heating to 100deg.C overnight, TLC indicating complete reaction of starting materials. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was subjected to silica gel column chromatography to give the title compound 11b (190 mg, crude product) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=578.3[M+H] +
Second step 7- ((R) -3-Aminopyrrolidin-1-yl) -3-methyl-1- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4- (3H) -one 11c
Compound 11b (190 mg, crude) was dissolved in ethanol hydrochloride (3 mL,12mmol, 4M) and reacted at room temperature for 1 hour, TLC showed complete reaction of starting material. The reaction was then added dropwise to an aqueous sodium bicarbonate solution to adjust to basicity, extracted with ethyl acetate, the combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the crude Prep-TLC purified to give the title compound 11c as a yellow oil (90 mg, 63% yield in two steps).
LC-MS:m/z=478.2[M+H] +
Step three N- ((R) -1- (3-methyl-1- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) pyrrolidin-3-yl) acetamide 11d
Compound 11c (50 mg,0.10 mmol) was dissolved in dichloromethane (3 mL), triethylamine (44 mg,0.43 mmol) and acetic anhydride (23 mg,0.22 mmol) were added and reacted at room temperature for 1 hour, TLC showed complete reaction of starting material. The reaction mixture was extracted with water and dichloromethane, the organic phases combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude Prep-TLC purified to give the title compound 11d (57 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=520.2[M+H] +
Fourth step N- ((R) -1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -3-methyl-4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) pyrrolidin-3-yl) acetamide 11
Compound 11d (57 mg, crude) was dissolved in ethanol (10 mL) and water (3 mL), reduced iron powder (30 mg,0.54 mmol) and ammonium chloride (27 mg,0.50 mmol) were added at room temperature, and the reaction was heated to 90℃for 2 hours, and TLC showed complete reaction of the starting material. The reaction solution was cooled to room temperature, filtered, the filtrate was added with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude Prep-TLC was purified to give the title compound 11 (25 mg, two-step yield 47%) as a pale yellow solid.
LC-MS:m/z=490.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.97(s,1H),6.96(s,2H),6.88(s,1H),6.78(s,1H),4.94(q,J=6.8Hz,1H),4.55-4.49(m,1H),3.85-3.80(m,1H),3.76-3.62(m,2H),3.53-3.43(m,4H),2.37-2.28(m,1H),2.12-2.03(m,1H),1.96(s,3H),1.58(d,J=6.8Hz,3H).(97.19%purity by HPLC)
Example 12
(R) -1- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (1-methylpiperidin-4-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 12
First step (R) -7- (1-methyl-1, 2,3, 6-tetrahydropyridin-4-yl) -1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4- (3H) -one 12b
Intermediate IN-3 (130 mg,0.31 mmol) and 1-methyl-1, 2,3, 6-tetrahydropyridine-4-boronic acid pinacol ester 12a (105 mg,0.47 mmol) were dissolved IN 1, 4-dioxane (4 mL) and water (1 mL), sodium carbonate (67 mg,0.63 mmol) and Pd (dppf) Cl were added sequentially at room temperature 2 (26 mg,0.03 mmol) was replaced with nitrogen multiple times and stirred for 3 hours at 100℃with TLC indicating complete reaction of starting material. The reaction solution was cooled to room temperature, diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 12b (130 mg, yield 87%) as a yellow solid.
LC-MS:m/z=475.2[M+H] +
Second step (R) -1- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (1-methylpiperidin-4-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 12
Compound 12b (120 mg,0.25 mmol) was dissolved in ethyl acetate (4 mL) and methanol (1 mL), concentrated hydrochloric acid (2 drops) and palladium on carbon (catalytic amount, 10%) were added and stirred at room temperature under hydrogen atmosphere for 5 hours, and LCMS showed the main product. The reaction mixture was adjusted to pH with methanolic ammonia, filtered through celite, the cake was washed with methanol, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 12 (31 mg, yield 27%) as a white solid.
LC-MS:m/z=447.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.69(s,1H),9.32(s,1H),8.18(s,1H),7.15(d,J=6.8Hz,1H),6.79(d,J=8.0Hz,2H),6.68(s,1H),5.50(s,2H),4.90-4.83(m,1H),3.19-3.15(m,2H),3.02-2.93(m,1H),2.47(s,3H),2.03(s,4H),1.50(d,J=6.8Hz,3H).(95.98%purity by HPLC)
Example 13
(R) -1- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (4- (dimethylamino) piperidin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 13
First step (R) -7- (4- (dimethylamino) piperidin-1-yl) -1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4 (3H) -one 13a
Intermediate IN-3 (100 mg,0.24 mmol) was dissolved IN 4-dimethylaminopiperidine 10a (310 mg,2.42 mmol) and dimethyl sulfoxide (3 drops) was added dropwise at room temperature, and the mixture was stirred for 3 hours at 80℃and TLC showed complete reaction of starting materials. The reaction solution was cooled to room temperature, diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 13a (140 mg, crude) which was used directly in the next step.
LC-MS:m/z=506.3[M+H] +
Second step (R) -1- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (4- (dimethylamino) piperidin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 13
Compound 13a (130 mg, crude) was dissolved in ethanol (4 mL) and water (2 mL), iron powder (72 mg,1.29 mmol) and ammonium chloride (70 mg,1.31 mmol) were added at room temperature, and stirred for 2 hours at 80℃and LCMS showed complete reaction. The reaction was filtered while hot, the filter cake was washed with ethanol several times, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 13 (7.7 mg, 6% yield in two steps) as a white solid.
LC-MS:m/z=476.3[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.18(s,1H),8.93(s,1H),7.41(s,1H),6.94(d,J=6.8Hz,1H),6.78(s,2H),6.67(s,1H),5.51(s,2H),4.90-4.83(m,1H),4.80-4.76(m,2H),3.51-3.46(m,1H),3.04-2.98(m,2H),2.74(s,6H),2.17(d,J=11.2Hz,2H),1.69-1.61(m,2H),1.50(d,J=6.8Hz,3H).(97.49%purity by HPLC)
Example 14
(R) -1- ((1- (2-fluoro-3- (trifluoromethyl) phenyl) ethyl) amino) -7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 14
First step 1- (2-fluoro-3- (trifluoromethyl) phenethyl alcohol 14b
2-fluoro-3-trifluoromethylbenzaldehyde 14a (10.0 g,52.05 mmol) was dissolved in tetrahydrofuran (200 mL), cooled to 0deg.C, and methylmagnesium bromide (9.3 g,78.00 mmol) was slowly added dropwise, and after the dropwise addition, the mixture was slowly warmed to room temperature and stirred for 1 hour, and TLC showed complete reaction of the starting material. The reaction solution was poured into a saturated aqueous ammonium chloride solution to quench, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 14b (11.1 g, crude product) which was used directly in the next step.
Second step 2-fluoro-3- (trifluoromethyl) acetophenone 14c
Compound 14b (11.1 g, crude) was dissolved in dichloromethane (200 mL), cooled to 0deg.C, nitrogen displaced multiple times, and dessmartin reagent (26.9 g,63.42 mmol) was added and stirred for 1 hour at room temperature, TLC indicated complete reaction of starting materials. The reaction was also quenched by pouring into saturated aqueous sodium carbonate, extracted with dichloromethane, the combined organic phases were washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 14c (7.3 g, 68% yield in two steps) as a yellow liquid.
1 H NMR(400MHz,CDCl 3 )δ8.10-8.06(m,1H),7.83-7.79(m,1H),7.35(t,J=8.0Hz,1H),2.69(d,J=5.2Hz,3H).
Third step (R, Z) -N- (1- (2-fluoro-3- (trifluoromethyl) phenyl) ethylene) -2-methylpropane-2-sulfinamide 14d
Compound 14c (7.3 g,35.41 mmol) was dissolved in tetraethyl titanate (80 mL), and (R) - (+) -tert-butylsulfinamide (12.9 g,106.4 mmol) was added at room temperature, and after the addition, the temperature was slowly raised to 100deg.C and stirred for 1 hour, TLC indicated complete reaction of the starting materials. The reaction solution was poured into vigorously stirred ice water, stirring was continued for 10 minutes, filtration, the cake was repeatedly washed with ethyl acetate, the filtrate was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 14d (11.0 g, crude product) which was used directly in the next step.
LC-MS:m/z=310.1[M+H] +
Fourth step (R) -N- ((R) -1- (2-fluoro-3- (trifluoromethyl) phenyl) ethyl) -2-methylpropane-2-sulfinamide 14e
Compound 14d (11.0 g, crude) was dissolved in tetrahydrofuran (120 mL) and water (2 mL), cooled to about-60℃and then sodium borohydride (4.0 g,105.7 mmol) was added in portions. After the addition, the reaction was continued for 2 hours, and TLC showed the starting material to be consumed. Pouring the reaction solution into ice water for quenching, extracting with ethyl acetate, washing an organic phase for a plurality of times, washing with saturated saline water, drying with sodium sulfate, and concentrating to obtain a crude product. The crude product was purified by column chromatography on silica gel to give the title compound 14e (3.4 g, 28% yield in two steps) as a yellow oil.
LC-MS:m/z=312.1[M+H] +
Fifth step (R) -1- (2-fluoro-3- (trifluoromethyl) phenethylamine 14f
Compound 14e (3.4 g,10.92 mmol) was dissolved in tetrahydrofuran (30 mL), concentrated hydrochloric acid (800 mg,21.8 mmol) was added, the temperature was raised to 80℃and stirred for 1 hour, TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, saturated aqueous sodium hydrogencarbonate solution was then added to adjust the pH to alkaline, extraction was performed with ethyl acetate, the organic phase was saturated brine washed, dried over sodium sulfate, and concentrated to give the title compound 14f (2.0 g, yield 87%) as a yellow liquid.
LC-MS:m/z=208.1[M+H] +
Sixth step (R) -6-chloro-1- ((1- (2-fluoro-3- (trifluoromethyl) phenyl) ethyl) amino) -3H-pyrrolo [3,4-c ] pyridin-3-one 14g
Compound IN-3h (400 mg,2.20 mmol) and compound 14f (411 mg,1.98 mmol) were dissolved IN isopropanol (15 mL), heated to 90℃and reacted overnight, and TLC detected the starting material was essentially complete. The reaction solution was cooled to room temperature and concentrated to give 14g (800 mg, crude) of the title compound as a yellow solid, which was used directly in the next step.
Seventh step (R) -7-chloro-1- ((1- (2-fluoro-3- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4- (3H) -one 14H
14g (800 mg, crude) of the compound was dissolved in ethanol (10 mL), hydrazine hydrate (300 mg,4.79mmol, 80%) was added at room temperature, and the reaction was heated to 40℃for 1 hour, and the TLC detection of the starting material was essentially complete. The reaction solution was cooled to room temperature, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound as a yellow solid for 14h (340 mg, two-step yield 44%).
Eighth step (R) -1- ((1- (2-fluoro-3- (trifluoromethyl) phenyl) ethyl) amino) -7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 14
Compound 14h (80 mg,0.21 mmol) and N-methylpiperazine (105 mg,1.05 mmol) were dissolved in toluene (10 mL), BINAP (30 mg,0.048 mmol), palladium acetate (30 mg,0.13 mmol) and sodium tert-butoxide (81 mg,0.84 mmol) were added sequentially at room temperature, nitrogen was replaced 3 times, and the reaction was heated to 100deg.C for 5 hours, and the TLC detection of the starting materials was complete. The reaction was cooled to room temperature, concentrated, and the crude product was purified by Prep-TLC to give the title compound 14 (20 mg, yield 21%) as an off-white solid.
LC-MS:m/z=451.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.18(s,1H),8.92(s,1H),7.71(t,J=7.2Hz,1H),7.63(t,J=7.2Hz,1H),7.42-7.30(m,2H),7.06(d,J=6.8Hz,1H),5.26-5.21(m,1H),3.80(s,4H),2.60(s,4H),2.35(s,3H),1.56(d,J=6.8Hz,3H).(99.25%purity by HPLC)
Example 15
(R) -1- ((1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4- (3H) -one 15
First step 1-bromo-3-difluoromethyl-2-fluorobenzene 15b
2-fluoro-3-bromobenzaldehyde 15a (10.0 g,49.26 mmol) was dissolved in dichloromethane (200 mL), cooled to 0deg.C, diethylaminosulfur trifluoride (15.9 g,98.64 mmol) was slowly added dropwise, and after the dropwise addition, the mixture was slowly warmed to room temperature and stirred for 1 hour, and TLC showed complete reaction of the starting materials. The reaction solution was quenched by pouring into saturated aqueous sodium bicarbonate, extracted with dichloromethane, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 15b (8.1 g, yield 73%) as a yellow liquid.
1 H NMR(400MHz,CDCl 3 )δ7.68(t,J=7.2Hz,1H),7.55(t,J=7.2Hz,1H),7.14(t,J=8.0Hz,1H),6.89(t,J=54.8Hz,1H).
Second step 1- (3- (difluoromethyl) -2-fluoroacetophenone 15c
Compound 15b (8.1 g,36.00 mmol) was dissolved in 1, 4-dioxane (80 mL), triethylamine (9.1 g,89.93 mmol) and tributyl (1-ethoxyethylene) tin (15.6 g,43.20 mmol) were added sequentially at room temperature, nitrogen was bubbled for 15 min, bis triphenylphosphine palladium dichloride (250 mg,0.36 mmol) was added, nitrogen was replaced multiple times, and stirring was carried out at 100℃for 1 hour, and TLC showed complete reaction. The reaction was cooled to room temperature, diluted hydrochloric acid (14.4 mL,72.0mmol, 5M) was added and stirred at room temperature for 1 hour, TLC indicated complete reaction of starting materials. The reaction solution was diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 15c (5.6 g, yield 83%) as a yellow liquid.
1 H NMR(400MHz,CDCl 3 )δ8.00(t,J=7.2Hz,1H),7.79(t,J=6.8Hz,1H),7.34(t,J=7.6Hz,1H),6.94(t,J=54.8Hz,1H),2.67(d,J=5.2Hz,3H).
Third step (R, Z) -N- (1- (3- (difluoromethyl) -2-fluorophenyl) ethylene) -2-methylpropane-2-sulfinamide 15d
Compound 15c (5.6 g,29.76 mmol) was dissolved in tetraethyltitanate (50 mL) and (R) - (+) -tert-butylsulfinamide (10.8 g,89.11 mmol) was added at room temperature and slowly warmed to 100deg.C and stirred for 1 hour, TLC indicated complete reaction of starting materials. The reaction solution was cooled to room temperature, poured into vigorously stirred ice water, stirred for 10 minutes, filtered, the filter cake was repeatedly washed with ethyl acetate, the filtrate extracted with ethyl acetate, the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 15d (9.5 g, crude) as a yellow oil, which was used directly in the next step.
LC-MS:m/z=292.1[M+H] +
Fourth step (R) -N- ((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) -2-methylpropane-2-sulfinamide 15e
Compound 15d (9.5 g, crude) was dissolved in tetrahydrofuran (100 mL) and water (2 mL), cooled to about-60℃and sodium borohydride (3.7 mg,97.80 mmol) was added in portions, and after addition, the reaction was continued at room temperature for 2 hours with slow warming to room temperature, and TLC indicated that the starting material was consumed. The reaction solution was poured into ice water to quench, extracted with ethyl acetate, and the organic phases were combined, washed with water, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 15e (2.9 g, two-step yield 33%) as a yellow oil.
LC-MS:m/z=294.1[M+H] +
Fifth step (R) -1- (3- (difluoromethyl) -2-fluorophenylethylamine 15f
Compound 15e (2.9 g,9.88 mmol) was dissolved in tetrahydrofuran (50 mL) and concentrated hydrochloric acid (720 mg,19.8 mmol) was added at room temperature and stirred for 1 hour at 80℃with TLC indicating depletion of the starting material. The reaction solution was cooled to room temperature, saturated aqueous sodium bicarbonate was then added to adjust the pH to alkaline, extraction was performed with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 15f (2.0 g, crude) as a brown liquid, which was used directly in the next step.
LC-MS:m/z=190.1[M+H] +
Sixth step (R) -6-chloro-1- ((1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -3H-pyrrolo [3,4-c ] pyridin-3-one 15g
Compound IN-3h (400 mg,2.20 mmol) and compound 15f (375 mg,1.98 mmol) were dissolved IN isopropanol (15 mL), heated to 90℃and reacted overnight, and TLC detected the starting material was essentially complete. The reaction solution was cooled to room temperature and concentrated to give 15g (826 mg, crude) of the title compound as a yellow solid, which was used directly in the next step.
Seventh step (R) -7-chloro-1- ((1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4- (3H) -one for 15H
15g (800 mg, crude) of the compound was dissolved in ethanol (10 mL), hydrazine hydrate (300 mg,4.79mmol, 80%) was added, the reaction mixture was heated to 40℃and reacted for 1 hour, and the TLC detection of the starting materials was essentially complete. The reaction solution was cooled to room temperature, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound as a yellow solid for 15h (360 mg, three-step yield 46%).
Eighth step (R) -1- ((1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4- (3H) -one 15
Compound 15h (80 mg,0.22 mmol) and N-methylpiperazine (220 mg,2.20 mmol) were dissolved in dimethyl sulfoxide (0.5 mL), heated to 100deg.C and reacted for 2 h, and TLC detected completion of the starting material reaction. The reaction was cooled to room temperature, water was added, solids precipitated, filtered, and the filter cake washed to give crude product, which was purified by Prep-TLC to give the title compound 15 (43 mg, yield 46%) as a yellow solid.
LC-MS:m/z=433.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.16(s,1H),8.91(s,1H),7.56(t,J=7.2Hz,1H),7.48(t,J=6.8Hz,1H),7.35(s,1H),7.27(t,J=7.6Hz,1H),7.22(t,J=54.4Hz,1H),7.00(d,J=6.8Hz,1H),5.24-5.19(m,1H), 3.77(s,4H),2.52(s,4H),2.30(s,3H),1.54(d,J=6.8Hz,3H).(97.61%purity by HPLC)
Example 16
(R) -1- ((1- (5-amino-3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4- (3H) -one
16
First step 1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethan-1-one 16a
Potassium nitrate (45.6 g,0.45 mmol) was dissolved in concentrated sulfuric acid (100 mL), stirred at room temperature for 30 min, cooled to about 0deg.C, compound 15c (8.5 g,45.18 mmol) was slowly added dropwise, and stirred at 0deg.C for 5 min, TLC indicated complete reaction. The reaction solution was slowly added to ice water, extracted with ethyl acetate, and the organic phases were combined, washed with saturated aqueous sodium bicarbonate, saturated brine, concentrated, and the crude product was purified by silica gel column to give the title compound 16a (9.0 g, yield 86%).
1 H NMR(400MHz,CDCl 3 )δ8.88-8.86(m,1H),8.66-8.64(m,1H),7.12-6.85(m,1H),2.73(d,J=4.8Hz,3H).
Second step (R, Z) -N- (1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethylene) -2-methylpropane-2-sulfinamide 16b
Compound 16a (9.0 g,38.60 mmol) was dispersed in tetraethyltitanate (90 mL) and (R) - (+) -tert-butylsulfinamide (7.0 g,57.76 mmol) was added at room temperature and slowly warmed to 100deg.C and stirred for 1 hour, TLC indicated complete reaction of starting materials. The reaction solution was cooled to room temperature, poured into vigorously stirred ice water, stirred for 10 minutes, filtered, the filter cake was repeatedly washed with ethyl acetate, the filtrate extracted with ethyl acetate, the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 16b (9.0 g, crude) as a yellow oil, which was used directly in the next step.
LC-MS:m/z=337.1[M+H] +
Third step (R) -N- ((R) -1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethyl) -2-methylpropane-2-sulfinamide 16c
Compound 16b (9.0 g, crude) was dissolved in tetrahydrofuran (150 mL) and water (2 mL), cooled to about-60℃and sodium borohydride (3.1 g,81.95 mmol) was added in portions, and after addition, the reaction was continued at room temperature for 2 hours with slow warming to room temperature, and TLC indicated that the starting material was consumed. The reaction solution was poured into ice water to quench, extracted with ethyl acetate, and the organic phases were combined, washed with water, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column to give the title compound 16c (2.8 g, two-step yield 21%) as a brown oil.
LC-MS:m/z=339.1[M+H] +
Fourth step (R) -1- (3- (difluoromethyl) -2-fluoro-5-nitrophenyl) ethan-1-amine 16d
Compound 16c (2.8 g,8.28 mmol) was dissolved in tetrahydrofuran (30 mL), concentrated hydrochloric acid (1.4 mL,16.52 mmol) was added and stirred at room temperature for 1 hour, and TLC showed the starting material to be consumed. The reaction solution was cooled to room temperature, saturated aqueous sodium hydrogencarbonate solution was then added to adjust the pH to alkaline, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and concentrated to give a brown liquid, which solidified upon standing to give the title compound 16d (1.8 g, yield 95%).
LC-MS:m/z=235.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ8.76-8.74(m,1H),8.41-8.39(m,1H),7.52-7.25(m,1H),4.42(q,J=6.4Hz,1H),2.48-2.24(m,2H),1.35(d,J=6.8Hz,3H).
Fifth step (R) -3- (1-aminoethyl) -5- (difluoromethyl) -4-fluoroaniline 16e
Compound 16d (300 mg,1.28 mmol) was dissolved in ethanol and water (6 mL/2 mL), reduced iron powder (356 mg,6.41 mmol) and ammonium chloride (348 mg,6.69 mmol) were added at room temperature, and the reaction was heated to 90℃for 2 hours, and TLC detection showed complete reaction. The reaction solution was cooled to room temperature, filtered through celite, and the filtrate was concentrated to give the title compound 16e (600 mg, crude) which was used directly in the next step.
Sixth step (R) -1- ((1- (5-amino-3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -6-chloro-3H-pyrrolo [3,4-c ] pyridin-3-one 16f
Compound 16e (200 mg, crude) and compound IN-3h (89 mg,0.45 mmol) were dispersed IN isopropanol (5 mL) and heated to 90℃for 12 hours, and TLC detection showed complete reaction. The reaction solution was cooled to room temperature and concentrated to give the title compound 16f (180 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=369.1[M+H] +
Seventh step (R) -1- ((1- (5-amino-3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -7-chloro-pyrido [3,4-d ] pyridazin-4- (3H) -one 16g
Compound 16f (180 mg, crude) was dissolved in methanol (10 mL), hydrazine hydrate (100 mg,1.60mmol, 80%) was added dropwise at room temperature, and the reaction was completed by TLC detection after heating to 45℃for 1.5 hours. The reaction was cooled to room temperature, filtered, and the filtrate was concentrated, and the crude product was purified by Prep-TLC to give 16g (44 mg, 27% yield in three steps) of the title compound as a yellow solid.
LC-MS:m/z=382.1[M-H] -
Eighth step (R) -1- ((1- (5-amino-3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4- (3H) -one 16
Compound 16g (20 mg,0.05 mmol) was dissolved in DMSO (0.5 mL) and N-methylpiperazine (52 mg,0.52 mmol) was added at room temperature and heated to 85℃for 12 hours, and TLC detection showed complete reaction. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 16 (7 mg, yield 30%) as a yellow solid.
LC-MS:m/z=448.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.04(s,1H),7.33(s,1H),6.95-6.82(m,2H),6.76-6.72(m,1H),5.20(q,J=6.8Hz,1H),3.99(s,4H),3.00(s,4H),2.66(s,3H),1.58(d,J=7.2Hz,3H).(97.85%purity by HPLC)
Example 17
1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 17
First step (R) -5- (1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 17b
Intermediate IN-3 (110 mg,0.27 mmol) and 1-tert-butoxycarbonyl-3, 6-dihydro-2H-pyridine-5-boronic acid pinacol ester 17a (123 mg,0.40 mmol) were dissolved IN 1, 4-dioxane (4 mL) and water (1 mL) and sodium carbonate (56 mg,0.53 mmol) and Pd (dppf) Cl were added sequentially at room temperature 2 (22 mg,0.023 mmol), nitrogen substitution was repeated after the addition, and the temperature was raised to 100℃and stirred Stirring was carried out for 3 hours and TLC showed complete reaction of starting materials. The reaction solution was cooled to room temperature, diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by a silica gel column to give the title compound 17b (120 mg, yield 81%) as a yellow solid.
LC-MS:m/z=561.3[M+H] +
Second step (R) -1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (1, 2,5, 6-tetrahydropyridin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 17c
Compound 17b (120 mg,0.21 mmol) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (1 mL) was added and stirred at room temperature for 1 h, and TLC showed the starting material to be consumed. The reaction solution was concentrated, the pH was adjusted to be alkaline with saturated aqueous sodium bicarbonate, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 17c (110 mg, crude) as a yellow oil, which was used directly in the next step.
LC-MS:m/z=461.2[M+H] +
Third step (R) -7- (1-methyl-1, 2,5, 6-tetrahydropyridin-3-yl) -1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4 (3H) -one 17d
Compound 17c (100 mg, crude) was dissolved in acetonitrile (2 mL), and potassium carbonate (60 mg,0.43 mmol) and methyl p-toluenesulfonate (32 mg,0.17 mmol) were added and stirred at room temperature overnight. The reaction solution was diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column to give the title compound 17d (25 mg, two-step yield 25%) as a yellow solid.
LC-MS:m/z=475.2[M+H] +
Fourth step 1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 17
Compound 17d (25 mg,0.053 mmol) was dissolved in a mixed solvent of ethyl acetate (1 mL) and tetrahydrofuran (1 mL), palladium on carbon (20 mg, 10%) was added at room temperature, and the mixture was stirred overnight at 50℃under a hydrogen atmosphere. The reaction solution was filtered through celite, the cake was washed with methanol several times, the filtrate was concentrated, and the crude Prep-TLC was purified to give the title compound 17 (10 mg, yield 42%) as a yellow solid.
LC-MS:m/z=447.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.41(s,1H),8.09(s,1H),6.92(s,2H),6.76(s,1H),4.96(q,J=6.8Hz,1H),3.38-3.32(m,2H),3.14-3.11(m,1H),2.82-2.70(m,1H),2.56(s,3H),2.50-2.37(m,1H),2.14-2.12(m,1H),1.95-1.78(m,3H),1.57(d,J=7.2Hz,3H).(94.55%purity by HPLC)
Example 18
1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (6-methyl-3, 6-diazabicyclo [3.1.1] hept-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 18
First step 3- (1- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) -3, 6-diazabicyclo [3.1.1] heptane-6-carboxylic acid tert-butyl ester 18b
Intermediate IN-3 (300 mg,0.73 mmol) and 6- (tert-butoxycarbonyl) -3, 6-diazabicyclo [3.1.1] heptane 18a (575 mg,2.90 mmol) were dissolved IN dimethyl sulfoxide (6 mL), warmed to 100deg.C and stirred for 2 hours, and TLC detection was complete. The reaction solution was cooled to room temperature, water was added, and a solid was precipitated, filtered, and the cake was washed and dried to give the title compound 18b (400 mg, crude product) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=576.3[M+H] +
Second step 7- (3, 6-diazabicyclo [3.1.1] hept-3-yl) -1- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4 (3H) -one 18c
Compound 18b (200 mg, crude) was dissolved in methanol (2 mL), and a methanol solution of hydrochloric acid (4 mL, 4M) was added, followed by stirring at room temperature for 3 hours, and TLC detection was complete. The reaction solution was neutralized with ammonia methanol, filtered, and the filtrate was concentrated to give the title compound 18c (170 mg, crude product) which was used directly in the next step.
Third step 7- (6-methyl-3, 6-diazabicyclo [3.1.1] hept-3-yl) -1- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4 (3H) -one 18d
Compound 18c (170 mg, crude) and methyl p-toluenesulfonate (65 mg,0.35 mmol) were dissolved in acetonitrile (15 mL), potassium carbonate (72 mg,0.52 mmol) was added, stirred overnight at room temperature, and TLC detected the large amount of starting material remaining. The reaction mixture was stirred for 8 hours at 80℃and the reaction was essentially complete as measured by TLC. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 18d (37 mg, three-step yield 21%).
LC-MS:m/z=490.2[M+H] +
Fourth step 1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (6-methyl-3, 6-diazabicyclo [3.1.1] hept-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 18
Compound 18d (37 mg,0.076 mmol) was dissolved in ethanol (10 mL) and water (2 mL), iron powder (21 mg,0.38 mmol) and ammonium chloride (20 mg,0.37 mmol) were added at room temperature, and the mixture was stirred for 2 hours at 90℃and the reaction was complete by TLC. The reaction was filtered while hot, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 18 (20 mg, yield 57%).
LC-MS:m/z=460.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 +D 2 O)δ9.03(s,1H),7.22(s,1H),6.97-6.73(m,3H),4.88(q,J=6.8Hz,1H),4.03-3.61(m,6H),2.57(s,3H),2.32-2.22(m,1H),1.83-1.68(m,1H),1.52(d,J=6.8Hz,3H).(98.46%purity by HPLC)
Example 19
1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- ((R) -2, 4-dimethylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 19
First step (R) -2, 4-dimethylpiperazine-1-carboxylic acid tert-butyl ester 19b
(R) -2-methylpiperazine-1-carboxylic acid tert-butyl ester 19a (12.0 g,59.92 mmol) was dissolved in methanol (50 mL), and aqueous formaldehyde (9.7 g,119.51mmol, 37%) and palladium on carbon (500 mg, 10%) were added and stirred at room temperature under a hydrogen atmosphere for 2.5 hours, and the reaction was complete by TLC. The reaction solution was filtered through celite, and the filtrate was concentrated to give the title compound 19b (12.2 g, crude) as a clear oil, which was used directly in the next step.
Second step (R) -1, 3-dimethylpiperazine dihydrochloride 19c
Compound 19b (12.2 g, crude) was dissolved in methanol hydrochloride (20 mL, 4M) and stirred at room temperature for 30 min, and TLC was complete. The reaction solution was concentrated to give the title compound 19c (12.0 g, crude) as a yellowish oil, which was used directly in the next step.
Third step (R) -2, 4-dimethylpiperazine-1-carboxylic acid benzyl ester 19d
Compound 19c (13.2 g, crude product) was dissolved in tetrahydrofuran (130 mL), water (50 mL) and sodium carbonate (37.4 g,352.86 mmol) were added at room temperature, the temperature was reduced to 0℃and benzyl chloroformate (20.9 g,122.52 mmol) was added dropwise, and after the addition, the mixture was stirred at room temperature for 1 hour, and TLC was found to be complete. The reaction mixture was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 19d (8.9 g, three-step yield 54%) as a yellow oil.
Fourth step (R) -1, 3-dimethylpiperazine 19e
Compound 19d (8.9 g,35.84 mmol) was dissolved in tetrahydrofuran (80 mL), palladium on carbon (2.0 g, 10%) was added and stirred at room temperature under hydrogen atmosphere for 1.5 hours, and the reaction was complete by TLC. The reaction solution was filtered through celite, and the filtrate was concentrated to give the title compound 19e (1.8 g, crude) as a yellow oil, which was used directly in the next step.
Fifth step 7- ((R) -2, 4-dimethylpiperazin-1-yl) -1- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4 (3H) -one 19f
Compound 19e (311 mg, crude) was dissolved IN dimethyl sulfoxide (1.5 mL), intermediate IN-3 (120 mg,0.29 mmol) was added at room temperature, and the reaction was allowed to warm to 120deg.C overnight, and the reaction was complete by TLC. The reaction was cooled to room temperature, water (3 mL) was added, the solid was precipitated, filtered, and the filter cake was washed and dried to give the title compound 19f (45 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=492.3[M+H] +
Sixth step 1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- ((R) -2, 4-dimethylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 19
Compound 19f (45 mg, crude) was dispersed in ethanol and water (3 mL/1 mL), reduced iron powder (25 mg,0.45 mmol) and ammonium chloride (24 mg,0.45 mmol) were added at room temperature, and the reaction was heated to 90℃for 3 hours, and the reaction was complete by TLC. The reaction solution was filtered hot through celite, the filter cake was washed, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 19 (28 mg, three step yield 21%) as a yellow solid.
LC-MS:m/z=462.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.05(s,1H),7.24(s,1H),6.99-6.87(m,2H),6.77(s,1H),5.05-4.91(m,2H),4.56(d,J=14.4Hz,1H),3.44-3.34(m,1H),3.30-3.25(m,1H),3.22(d,J=12.0Hz,1H),2.85-2.73(m,1H),2.64(s,4H),1.57(d,J=6.8Hz,3H),1.37(d,J=6.8Hz,3H).(96.26%purity by HPLC)
Example 20
1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- ((S) -2, 4-dimethylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 20
First step 1-benzyl 4- (tert-butyl) (S) -2-methylpiperazine-1, 4-dicarboxylic acid ester 20b
(S) -4-N-Boc-2-methylpiperazine 20a (3.0 g,14.98 mmol) was dissolved in ethyl acetate (30 mL) and water (30 mL), sodium hydrogencarbonate (4.0 g,47.61 mmol) was added at room temperature, cooled to about 0deg.C, benzyl chloroformate (3.8 g,22.28 mmol) was added dropwise, and the mixture was reacted at 0deg.C for 1 hour. The reaction mixture was extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column to give the title compound 20b (5.0 g, yield 99.8%) as a yellow oil.
Second step (S) -2-methylpiperazine-1-carboxylic acid benzyl ester 20c
Compound 20b (5.0 g,14.95 mmol) was dissolved in dichloromethane (50 mL), trifluoroacetic acid (10 mL) was added and stirred at room temperature for 1 h, and TLC showed completion of the reaction. The reaction solution was concentrated, saturated aqueous sodium hydrogencarbonate solution was added to adjust the pH to alkaline, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 20c (3.7 g, crude) as a yellow oil, which was used directly in the next step.
LC-MS:m/z=235.2[M+H] +
Third step (S) -2, 4-dimethylpiperazine-1-carboxylic acid benzyl ester 20d
Compound 20c (3.7 g, crude) was dissolved in formic acid (3.1 g,67.35 mmol), aqueous formaldehyde (2.4 g,29.57mmol, 37%) was added at room temperature, warmed to 80℃and stirred overnight, TLC indicated complete reaction. The reaction solution was cooled to room temperature, the reaction solution was concentrated, the pH was adjusted to be alkaline with saturated aqueous sodium hydrogencarbonate, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column to give the title compound 20d (3.1 g, two-step yield 84%) as a colorless liquid.
LC-MS:m/z=249.2[M+H] +
Fourth step (S) -1, 3-dimethylpiperazine 20e
Compound 20d (3.0 g,12.08 mmol) was dissolved in tetrahydrofuran (30 mL), palladium on carbon (300 mg, 10%) was added and stirred overnight at room temperature under hydrogen atmosphere, TLC indicated complete reaction of starting materials. The reaction solution was filtered through celite, and the filter cake was washed with ethyl acetate, and the filtrate was concentrated to give the title compound 20e (500 mg, crude) as a yellow oil, which was used directly in the next step.
Fifth step 7- ((S) -2, 4-dimethylpiperazin-1-yl) -1- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4 (3H) -one 20f
Intermediate IN-3 (80 mg,0.19 mmol) was dissolved IN dimethyl sulfoxide (0.5 mL) and compound 20e (230 mg, crude) was added at room temperature, warmed to 120deg.C and stirred overnight, TLC indicated that the starting material was consumed. The reaction solution was cooled to room temperature, diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column to give the title compound 20f (90 mg, yield 95%) as a brown solid.
LC-MS:m/z=492.3[M+1] +
Sixth step 1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- ((S) -2, 4-dimethylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 20
Compound 20f (90 mg,0.18 mmol) was dissolved in ethanol (4 mL) and water (2 mL), iron powder (57 mg,1.02 mmol) and ammonium chloride (55 mg,1.03 mmol) were added at room temperature, and after the addition, the temperature was raised to 80℃and stirred for 1 hour, TLC indicated complete reaction of the starting materials. The reaction solution was filtered while hot, the filter cake was washed with ethanol, the filtrate was concentrated, diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 20 (22 mg, yield 26%) as a yellow solid.
LC-MS:m/z=462.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.06(s,1H),7.20(s,1H),6.92(s,2H),6.77(s,1H),4.94(q,J=6.8Hz,2H),4.47(d,J=12.4Hz,1H),3.37-3.33(m,1H),3.13-3.02(m,2H),2.54-2.31(m,5H),1.56(d,J=6.8Hz,3H),1.35(d,J=6.8Hz,3H).(96.68%purity by HPLC)
Example 21
1- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -7- (1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4- (3H) -one 21
First step (R) -5- (1- ((1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 21a
Compound 15H (110 mg,0.30 mmol) and 1-tert-butoxycarbonyl-3, 6-dihydro-2H-pyridine-5-boronic acid pinacol ester 17a (113 mg,0.37 mmol) were dissolved in 1, 4-dioxane (4 mL) and water (1 mL), sodium carbonate (52 mg,0.49 mmol) and Pd (dppf) Cl were added sequentially at room temperature 2 (20 mg,0.027 mmol), nitrogen substitution multiple times, heating to 100deg.C and stirring for 3 hours, TLC showed complete reaction of starting materials. The reaction solution was cooled to room temperature, diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column to give the title compound 21a (120 mg, yield 78%) as a yellow solid.
LC-MS:m/z=516.3[M+H] +
Second step 3- (1- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) piperidine-1-carboxylic acid tert-butyl ester 21b
Compound 21a (120 mg,0.23 mmol) was dissolved in ethyl acetate (2 mL) and tetrahydrofuran (2 mL), palladium on carbon (40 mg, 10%) was added at room temperature, and the mixture was stirred overnight at 50℃under a hydrogen atmosphere. The reaction was cooled to room temperature, filtered through celite, and the filter cake was washed with methanol, and the filtrate was concentrated to give the title compound 21b (110 mg, crude) as a white solid, which was used directly in the next step.
LC-MS:m/z=518.3[M+H] +
Third step 1- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -7- (piperidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 21c
Compound 21b (50 mg, crude) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (1 mL) was added and stirred at room temperature for 1 hour, and TLC showed completion of the reaction. The reaction was concentrated, pH was adjusted to basic with an ammonia methanol solution (7M), and the crude product was purified by Prep-TLC to give the title compound 21c as a white solid (30 mg, 69% yield in two steps).
LC-MS:m/z=418.2[M+H] +
Fourth step 1- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -7- (1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4- (3H) -one 21
Compound 21c (30 mg,0.072 mmol) was dispersed in methanol (2 mL), aqueous formaldehyde (58 mg,0.71mmol, 37%) and palladium on carbon (20 mg, 10%) were added and stirred overnight at room temperature under hydrogen atmosphere. The reaction solution was filtered through celite, the cake was washed with methanol several times, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 21 (8 mg, yield 26%) as a white solid.
LC-MS:m/z=432.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.42(s,1H),8.13(s,1H),7.57(t,J=7.2Hz,1H),7.44(t,J=6.8Hz,1H),7.21(t,J=7.6Hz,1H),6.98(t,J=54.8Hz,1H),5.32(q,J=7.2Hz,1H),3.45-3.31(m,2H),3.20-3.12(m,1H),2.98-2.78(m,1H),2.62(s,3H),2.54-2.42(m,1H),2.17-2.14(m,1H),1.96-1.84(m,3H),1.63(d,J=6.8Hz,3H).(96.73%purity by HPLC)
Example 22
1- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -3-methyl-7- (1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4- (3H) -one 22
First step 3- (1- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -3-methyl-4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) piperidine-1-carboxylic acid tert-butyl ester 22a
Compound 21b (80 mg,0.15 mmol) was dissolved in N, N-dimethylformamide (1 mL), followed by addition of potassium carbonate (43 mg,0.31 mmol) and iodomethane (33 mg,0.23 mmol), stirring at room temperature for 4 hours, and TLC showed complete reaction of the starting materials. The reaction solution was diluted with water, extracted with ethyl acetate, and the organic phases were combined, washed with water, saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 22a (100 mg, crude) as a white solid, which was used directly in the next step.
Second step 1- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -3-methyl-7- (piperidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 22b
Compound 22a (100 mg, crude) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (1 mL) was added and stirred at room temperature for 1 hour, TLC showed completion of the reaction. The reaction was concentrated, pH was adjusted to basic with an ammonia methanol solution (7M), and the crude product was purified by Prep-TLC to give the title compound 22b (50 mg, 75% yield in two steps) as a white solid.
LC-MS:m/z=432.2[M+H] +
Third step 1- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -3-methyl-7- (1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4- (3H) -one 22
Compound 22b (50 mg,0.12 mmol) was dispersed in methanol (2 mL), aqueous formaldehyde (94 mg,1.16mmol, 37%) and palladium on carbon (30 mg, 10%) were added and stirred overnight at room temperature under a hydrogen atmosphere. The reaction solution was filtered through celite, the cake was washed with methanol several times, the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 22 (13 mg, yield 25%) as a pale yellow solid.
LC-MS:m/z=446.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.40(s,1H),8.07(s,1H),7.59(t,J=7.2Hz,1H),7.45(t,J=7.2Hz,1H),7.21(t,J=7.6Hz,1H),7.01(t,J=54.8Hz,1H),5.31(q,J=6.8Hz,1H),3.50(s,3H),3.28-3.18(m,2H),3.06-2.98(m,1H),2.52-2.47(m,1H),2.43(s,3H),2.29-2.15(m,1H),2.13-2.05(m,1H),1.93-1.71(m,3H),1.64(d,J=6.8Hz,3H).(99.14%purity by HPLC)
Example 23
(R) -1- (((1- (4- (2- (((methylamino) methyl) phenyl) thiophen-2-yl) ethyl) amino) -7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 23)
First step (2- ((methylamino) methyl) phenyl) boronic acid 23b
O-formylboronic acid 23a (5.0 g,33.35 mmol) was dissolved in methylamine ethanol (30 mL, 7M), palladium on carbon (200 mg, 10%) was added and reacted at room temperature under hydrogen atmosphere for 5 hours, and TLC detected complete reaction of starting materials. The reaction solution was filtered through celite, the cake was washed, and the filtrate was concentrated to give the title compound 23b (5.3 g, crude) as a pale yellow foamy solid, which was used directly in the next step.
Second step (2- (((tert-Butoxycarbonyl) (methyl) amino) methyl) phenyl) boronic acid 23c
Compound 23b (5.3 g, crude product) was dissolved in tetrahydrofuran (100 mL) and water (20 mL), sodium carbonate (10.3 g,97.18 mmol) was added at room temperature, cooled to 0deg.C, di-tert-butyl dicarbonate (8.5 g,38.95 mmol) was added dropwise, and after the addition, the reaction was resumed at room temperature overnight, and TLC detection was complete. The reaction mixture was extracted with water, ethyl acetate, dried over anhydrous sodium sulfate, concentrated, slurried with crude petroleum ether and ethyl acetate, filtered, and the filter cake was washed and dried to give the title compound 23c as a white solid (7.4 g, 84% yield in two steps).
1 H NMR(400MHz,DMSO-d 6 )δ8.14(s,2H),7.50(dd,J=7.2,0.8Hz,1H),7.38-7.30(m,1H),7.21(t,J=7.2Hz,1H),7.08(d,J=7.6Hz,1H),4.56(s,2H),2.74(s,3H),1.40(s,9H).
Third step (R) -1- (4-bromothiophen-2-yl) ethan-1-amine 23e
Compound 23d (1.1 g,3.59 mmol) was dissolved in methanol hydrochloride (10 mL,4 mol/L) and reacted at room temperature for 1 hour, and TLC showed complete reaction of the starting material. The reaction solution was neutralized with saturated aqueous sodium hydrogencarbonate, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 23e (1.0 g, crude product) as a yellow oil, which was used directly in the next step.
Fourth step (R) - (2- (5- (1-aminoethyl) thiophen-3-yl) benzyl) (methyl) carbamic acid tert-butyl ester 23f
Compound 23e (1.0 g, crude) and compound 23c (1.9 g,7.17 mmol) were dissolved in 1, 4-dioxane (20 mL) and water (5 mL), sodium carbonate (1.5 g,14.15 mmol) and Pd (dppf) Cl were added at room temperature 2 Dichloromethane complex (200 mg,0.24 mmol), nitrogen displacement 3 times, heating to 100 ℃ for 4 hours, TLC detects complete reaction of starting material. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by column chromatography to give the title compound 23f (800 mg, two-step yield 64%) as a red oil.
LC-MS:m/z=347.2[M+H] +
Fifth step (R) - (2- (5- (1- ((6-chloro-3-oxo-3H-pyrrolo [3,4-c ] pyridin-1-yl) amino) ethyl) thiophen-3-yl) benzyl) (methyl) carbamic acid tert-butyl ester 23g
Compound 23f (800 mg,2.31 mmol) and compound IN-3h (400 mg,2.20 mmol) were dissolved IN isopropanol (20 mL), heated to 90℃and stirred overnight, and the reaction was essentially complete as determined by TLC. The reaction solution was cooled to room temperature and concentrated to give 23g (1.1 g, crude) of the title compound as a brown foam, which was used directly in the next step.
Sixth step (R) - (2- (5- (1- ((7-chloro-4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-1-yl) amino) ethyl) thiophen-3-yl) benzyl) (methyl) carbamic acid tert-butyl ester 23h
23g (1.1 g, crude) of compound was dissolved in methanol (15 mL), hydrazine hydrate (279 mg,4.30mmol, 80%) was added at room temperature, heated to 40℃and reacted for 2 hours, and TLC detection was complete. The reaction solution was cooled to room temperature, filtered, and the filtrate was concentrated, and the crude product was purified by silica gel column chromatography to give the title compound as a yellowish brown oil for 23h (910 mg, 79% in two steps).
Seventh step (R) -methyl (2- (5- (1- ((7- (4-methylpiperazin-1-yl) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-1-yl) amino) ethyl) thiophen-3-yl) benzyl) carbamic acid tert-butyl ester 23i
Compound 23h (100 mg,0.17 mmol) and N-methylpiperazine 2b (95 mg,0.95 mmol) were dissolved in dimethyl sulfoxide (1 mL), heated to 80deg.C and reacted for 2 h, and TLC detected the reaction was complete. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 23i (75 mg, crude) as a yellow solid, which was used directly in the next step.
Eighth step (R) -1- (((1- (4- (2- (((methylamino) methyl) phenyl) thiophen-2-yl) ethyl) amino) -7- (4-methylpiperazin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 23
Compound 23i (75 mg, crude) was dissolved in hydrochloric acid/1, 4-dioxane solution (3 mL, 4M) and reacted at room temperature for 1 hour, and TLC detection was complete. The reaction solution was neutralized with saturated aqueous sodium carbonate, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 23 (17 mg, 20% yield in two steps) as a yellow solid.
LC-MS:m/z=490.3[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.31(s,1H),8.93(s,1H),7.50-7.46(m,1H),7.44(d,J=1.2Hz,1H),7.34-7.29(m,3H),7.23(d,J=8.0Hz,2H),7.00(d,J=7.6Hz,1H),5.41-5.25(m,1H),3.76-3.69(m,4H),3.68(s,2H),2.47-2.40(m,4H),2.28(s,3H),2.24(s,3H),1.67(d,J=6.8Hz,3H).(99.72%purity by HPLC)
Example 24
1- (((R) -1- (4- (2- ((methylamino) methyl) phenyl) thiophen-2-yl) ethyl) amino) -7- (((S) -tetrahydrofuran-3-yl) oxy) pyrido [3,4-d ] pyridazin-4 (3H) -one 24
First step methyl (2- (5- ((R) -1- ((4-oxo-7- (((S) -tetrahydrofurane-3-yl) oxy) -3, 4-dihydropyrido [3,4-d ] pyridazin-1-yl) amino) ethyl) thiophen-3-yl) benzyl) carbamic acid tert-butyl ester 24a
(S) - (+) -3-hydroxytetrahydrofuran (1 mL) was cooled to 0deg.C, sodium hydrogen (76 mg,1.90mmol, 60%) was added, and the mixture was stirred at room temperature for 1 hour, the reaction mixture was changed from emulsion to orange transparent, and compound 23h (100 mg,0.19 mmol) was added, and the temperature was raised to 80deg.C and stirred overnight. The reaction solution was quenched with water, extracted with ethyl acetate, and the organic phases were combined, washed with water, saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column to give the title compound 24a (100 mg, yield 91%)
LC-MS:m/z=578.3[M+H] +
Second step 1- (((R) -1- (4- (2- ((methylamino) methyl) phenyl) thiophen-2-yl) ethyl) amino) -7- (((S) -tetrahydrofurane-3-yl) oxy) pyrido [3,4-d ] pyridazin-4 (3H) -one 24
Compound 24a (100 mg,0.17 mmol) was dissolved in hydrochloric acid/1, 4-dioxane solution (1 mL, 4M) and stirred at room temperature for 1 hour, TLC showed the disappearance of starting material. The reaction solution was adjusted to pH with saturated aqueous sodium bicarbonate, extracted with ethyl acetate, the organic phases combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product purified by Prep-TLC to give the title compound 24 (7 mg, yield 8%) as a yellow solid.
LC-MS:m/z=478.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.12(s,1H),7.44-7.36(m,2H),7.34-7.25(m,3H),7.14(s,1H),7.11(s,1H),5.77-5.68(m,1H),5.39-5.29(m,1H),4.07-3.80(m,6H),2.39-2.30(m,1H),2.24(s,3H),2.20-2.14(m,1H),1.72(d,J=6.8Hz,3H).(88.28%purity by HPLC)
Example 25
1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (3- (dimethylamino) pyrrolidin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 25
First step 3- (dimethylamino) pyrrolidine-1-carboxylic acid tert-butyl ester 25b
3-aminopyrrolidine-1-carboxylic acid tert-butyl ester 25a (800 mg,4.30 mmol) was dispersed in methanol (10 mL), aqueous formaldehyde (3.5 g,43.12mmol, 37%) and palladium on carbon (200 mg, 10%) were added and stirred overnight at room temperature under hydrogen atmosphere. The reaction solution was filtered through celite, the cake was washed with methanol several times, and the filtrate was concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 25b (900 mg, yield 98%) as a yellow liquid.
LC-MS:m/z=215.2[M+H] +
1 H NMR(400MHz,CDCl 3 )δ3.52-3.44(m,1H),3.23-3.17(m,1H),3.04-2.96(m,1H),2.63-2.54(m,1H),2.19(s,6H),2.01-1.95(m,1H),1.74-1.55(m,2H),1.39(s,6H).
Second step N, N-dimethylpyrrolidin-3-amine 25c
Compound 25b (900 mg,4.20 mmol) was dissolved in hydrochloric acid/1, 4-dioxane solution (10 mL, 4M) and stirred at room temperature for 1 hour, TLC showed complete reaction of the starting material. The reaction solution was concentrated to give the title compound 25c (700 mg, crude) as a brown liquid, which was used directly in the next step.
LC-MS:m/z=115.2[M+H] +
Third step 7- (3- (dimethylamino) pyrrolidin-1-yl) -1- (((R) -1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) pyrido [3,4-d ] pyridazin-4 (3H) -one 25d
Intermediate IN-3 (70 mg,0.17 mmol) was dissolved IN dimethyl sulfoxide (0.5 mL), compound 25c (58 mg, crude) and N, N-diisopropylethylamine (87 mg,0.68 mmol) were added at room temperature, warmed to 80℃and stirred overnight, and TLC monitored for disappearance of the starting material. The reaction solution was extracted with water and ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column to give the title compound as a brown solid (25 d, 50mg, yield 60%).
LC-MS:m/z=492.2[M+H] +
Fourth step 1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (3- (dimethylamino) pyrrolidin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 25
Compound 25d (50 mg,0.10 mmol) was dissolved in ethanol (2 mL) and water (1 mL), iron powder (28 mg,0.50 mmol) and ammonium chloride (27 mg,0.50 mmol) were added at room temperature, and stirred for 1 hour at 80℃with TLC indicating complete reaction of starting materials. The reaction solution was filtered while hot, the filter cake was washed with ethanol, the filtrate was concentrated, diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude Prep-TLC purified to give the title compound 25 (30 mg, yield 64%) as a yellow solid.
LC-MS:m/z=462.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.98(s,1H),6.94(s,1H),6.92(s,1H),6.87(s,1H),6.77(s,1H),4.95(q,J=7.2Hz,1H),3.92-3.83(m,2H),3.57-3.48(m,1H),3.40-3.33(m,1H),3.10-3.05(m,1H),2.44(s,6H),2.41-2.34(m,1H),2.07-1.93(m,1H),1.57(d,J=7.2Hz,3H).(99.28%purity by HPLC)
Example 26
1- (((R) -1- (3- (difluoromethyl) -2-fluorophenyl) ethyl) amino) -7- (3- (dimethylamino) pyrrolidin-1-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 26
Compound 15h (60 mg,0.16 mmol) was dissolved in dimethyl sulfoxide (0.5 mL), compound 25c (56 mg,0.49 mmol) and N, N-diisopropylethylamine (84 mg,0.65 mmol) were added at room temperature, and the mixture was warmed to 80℃and stirred overnight, the TLC disappeared starting material disappeared. The reaction mixture was extracted with water and ethyl acetate, the organic phases combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the crude product as a gray solid, title compound 26 (25 mg, yield 34%) by Prep-TLC.
LC-MS:m/z=447.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.00(s,1H),7.56(t,J=7.6Hz,1H),7.43(t,J=6.8Hz,1H),7.20(t,J=8.0Hz,1H),7.11-6.84(m,2H),5.30(q,J=6.8Hz,1H),4.04-3.99(m,1H),3.91-3.87(m,1H),3.63-3.50(m,2H),3.41-3.38(m,1H),2.62(s,6H),2.52-2.45(m,1H),2.17-2.07(m,1H),1.62(d,J=7.2Hz,3H).(99.44%purity by HPLC)
Example 27
1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (1-methylpyrrolidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 27
First step (R) -3- (1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) -2, 5-dihydro-1H-pyrrole-1-carboxylic acid tert-butyl ester 27b
Intermediate IN-3 (100 mg,0.24 mmol) was dissolved IN 1, 4-dioxane (5 mL) and water (1 mL) and 1-tert-butoxycarbonyl-2, 5-dihydro-1H-pyrrole-3-boronic acid pinacol ester 27a (85 mg,0.29 mmol), sodium carbonate (76 mg,0.72 mmol) and Pd (dppf) Cl were added at room temperature 2 Dichloromethane complex (20 mg,0.024 mmol), nitrogenAfter 3 substitutions, the reaction was heated to 100℃for 3 hours, and the reaction was complete by TLC. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 27b (115 mg, yield 87%) as a yellow solid.
LC-MS:m/z=547.3[M+H] +
Second step 3- (1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 27c
Compound 27b (115 mg,0.21 mmol) was dissolved in ethyl acetate (10 mL), palladium on carbon (20 mg, 10%) was added at room temperature and reacted at 50℃for 8 hours under a hydrogen atmosphere, and LCMS showed complete reaction of the starting materials. The reaction was cooled to room temperature, filtered through celite, and the filter cake was washed, and the filtrate was concentrated to give the title compound 27c (100 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=519.3[M+H] +
Third step 3- (1- (((R) -1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) pyrrolidine-1-carboxylic acid tert-butyl ester 27d
Compound 27c (100 mg, crude) was dissolved in dichloromethane (10 mL), N-diisopropylethylamine (65 mg,0.50 mmol) and acetic anhydride (38 mg,0.37 mmol) were added at room temperature and heated to 30deg.C for 4 hours, and TLC detected the starting material was essentially complete. The reaction was cooled to room temperature, extracted with dichloromethane, the organic phases combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the crude product as a yellow solid, title compound 27d (50 mg, 42% yield in two steps) by Prep-TLC.
Fourth step N- (3- ((1R) -1- ((4-oxo-7- (pyrrolidin-3-yl) -3, 4-dihydropyrido [3,4-d ] pyridazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 27e
Compound 27d (50 mg,0.096 mmol) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (1 mL) was added and reacted at room temperature for 2 hours, and TLC was used to detect completion of the starting material reaction. The reaction solution was neutralized with saturated aqueous sodium bicarbonate, extracted with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 27e (38 mg, crude) as a yellow solid, which was used directly in the next step.
Fifth step N- (3- ((1R) -1- ((7- (1-methylpyrrolidin-3-yl) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 27f
Compound 27e (38 mg, crude) was dissolved in methanol (3 mL), aqueous formaldehyde (3 d) and palladium on carbon (15 mg, 10%) were added and reacted at room temperature under a hydrogen atmosphere for 2 hours, and the reaction was essentially complete by TLC. The reaction solution was filtered through celite, and the filtrate was concentrated to give the title compound 27f (54 mg, crude) which was used directly in the next step.
Sixth step 1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- (1-methylpyrrolidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 27
Compound 27f (54 mg, crude) was dissolved in ethanol (3 mL), aqueous sodium hydroxide (3 mL,12mmol, 4M) was added at room temperature, and the reaction was heated to 80℃overnight, and the basic reaction was complete as detected by TLC. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 27 (8 mg, three-step yield 19%) as a pale yellow solid.
LC-MS:m/z=433.2[M+H] +
1 HNMR(400MHz,CD 3 OD)δ9.43(s,1H),8.07(s,1H),6.93(s,2H),6.77(s,1H),4.95(q,J=6.8Hz,1H),4.05-3.75(m,1H),3.40-3.36(m,1H),3.12-3.06(m,3H),2.63(s,3H),2.59-2.42(m,1H),2.27-2.22(m,1H),1.58(d,J=6.8Hz,3H).(97.64%purity by HPLC)
Example 28
1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -3-methyl-7- (1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 28
First step (R) -5- (3-methyl-1- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 28a
Compound 4a (200 mg,0.47 mmol) and 1-tert-butoxycarbonyl-3, 6-dihydro-2H-pyridine-5-boronic acid pinacol ester 17a (217 mg,0.70 mmol) were dissolved in 1, 4-dioxane (15 mL) and water (3 mL) and Pd (dppf) Cl was added at room temperature 2 Dichloromethane complex (18 mg,0.022 mmol) and sodium carbonate (148 mg,1.40 mmol), and was stirred for 1 hour at 100℃until completion of the reaction by TLC. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column chromatography to give the title compound 28a (282 mg, crude product) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=575.3[M+H] +
Second step 3- (1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -3-methyl-4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) piperidine-1-carboxylic acid tert-butyl ester 28b
Compound 28a (282 mg, crude) was dissolved in ethyl acetate (10 mL), palladium on carbon (100 mg, 10%) was added and reacted overnight at 50℃under hydrogen atmosphere, and LCMS detected completion of the reaction of the starting materials. The reaction was cooled to room temperature, filtered through celite, and the filter cake was washed with methanol, and the filtrate was concentrated to give the title compound 28b (290 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=547.3[M+H] +
Third step 3- (1- (((R) -1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -3-methyl-4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) piperidine-1-carboxylic acid tert-butyl ester 28c
Compound 28b (290 mg, crude) was dissolved in dichloromethane (3 mL), acetic anhydride (81 mg,0.79 mmol) and triethylamine (107 mg,1.06 mmol) were added at room temperature, and the temperature was raised to 30℃for 4 hours, and TLC showed complete reaction of starting materials. The reaction was concentrated, and the crude product was purified by Prep-TLC to give the title compound 28c (152 mg, three steps yield 55%) as a pale yellow solid.
LC-MS:m/z=589.3[M+H] +
Fourth step N- (3- ((1R) -1- ((3-methyl-4-oxo-7- (piperidin-3-yl) -3, 4-dihydropyrido [3,4-d ] pyridazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 28d
Compound 28c (152 mg,0.26 mmol) was dissolved in dichloromethane (2 mL) and trifluoroacetic acid (2 mL) was added and reacted at room temperature for 2 hours, TLC indicated that the starting material was complete. The reaction solution was adjusted to be basic by adding saturated aqueous sodium bicarbonate, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 28d (143 mg, crude) as a yellow solid, which was used directly in the next step.
Fifth step N- (3- ((1R) -1- ((3-methyl-7- (1-methylpiperidin-3-yl) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 28e
Compound 28d (143 mg, crude) was dissolved in methanol (5 mL), aqueous formaldehyde (71 mg,0.87mmol, 37%) and palladium on carbon (70 mg, 10%) were added and reacted at room temperature under hydrogen atmosphere for 2 hours, TLC indicated complete reaction of starting materials. The reaction solution was filtered through celite, and the cake was washed with methanol, and the filtrate was concentrated to give the title compound 28e (190 mg, crude) as a yellow oil, which was used directly in the next step.
LC-MS:m/z=503.3[M+H] +
Sixth step 1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -3-methyl-7- (1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 28
Compound 28e (190 mg, crude) was dissolved in methanol (4 mL) and aqueous sodium hydroxide (1.89 mL,7.56mmol, 4N) was added at room temperature and heated to 80deg.C overnight, TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by Prep-TLC to give the title compound 28 (47 mg, 39% in three steps) as a white solid.
LC-MS:m/z=461.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.41(s,1H),8.07(s,1H),6.97(s,2H),6.78(s,1H),4.96(q,J=6.8Hz,1H),3.56(s,3H),3.38-3.26(m,2H),3.15-3.08(m,1H),2.82-2.65(m,1H),2.55(s,3H),2.50-2.36(m,1H),2.16-2.07(m,1H),1.99-1.70(m,3H),1.60(d,J=7.2Hz,3H).(99.78%purity by HPLC)
Example 29
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (1-methylpiperidin-3-yl) phthalazin-1 (2H) -one 29
First step (R) -5- (4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -1-oxo-1, 2-dihydro-phthalazin-6-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 29a
Intermediate IN-2 (300 mg,0.66 mmol) and 1-tert-butoxycarbonyl-3, 6-dihydro-2H-pyridine-5-boronic acid pinacol ester 17a (325 mg,1.05 mmol) were dissolved IN 1, 4-dioxane (6 mL) and water (2 mL) and sodium carbonate (150 mg,1.42 mmol) and Pd (dppf) Cl were added sequentially at room temperature 2 (58 mg,0.08 mmol), nitrogen substitution multiple times, heating to 100deg.C and stirring for 1 hr, TLC showed complete reaction of starting materials. The reaction solution was cooled to room temperature, diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated, and the crude product was purified by silica gel column to give the title compound 29a (260 mg, yield 70%) as a yellow solid.
LC-MS:m/z=560.3[M+H] +
Second step 3- (4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -1-oxo-1, 2-dihydro-phthalazin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 29b
Compound 29a (250 mg,0.45 mmol) was dissolved in ethyl acetate (8 mL), palladium on carbon (60 mg, 10%) was added, and the mixture was stirred overnight at 50℃under hydrogen. The reaction solution was filtered through celite, the cake was washed with methanol several times, the filtrate was concentrated, and the crude product was purified by silica gel column to give the title compound 29b (210 mg, yield 88%) as a white solid.
LC-MS:m/z=532.3[M+H] +
Third step 3- (4- (((R) -1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -1-oxo-1, 2-dihydro-phthalazin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 29c
Compound 29b (210 mg,0.40 mmol) was dissolved in dichloromethane (5 mL) and N, N-diisopropylethylamine (102 mg,0.79 mmol) and acetic anhydride (61 mg,0.60 mmol) were added sequentially at room temperature and stirred for 5 hours at 30℃and TLC showed substantially complete reaction. The reaction solution was diluted with water, extracted with dichloromethane, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 29c (250 mg, crude) as a white solid, which was used directly in the next step.
LC-MS:m/z=574.3[M+H] +
Fourth step N- (3- ((1R) -1- ((4-oxo-7- (piperidin-3-yl) -3, 4-dihydro-phthalazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 29d
Compound 29c (250 mg, crude) was dissolved in dichloromethane (5 mL), trifluoroacetic acid (2 mL) was added and stirred at room temperature for 1 hour, TLC showed completion of the reaction. The reaction solution was concentrated, the pH was adjusted to weakly alkaline with saturated aqueous sodium bicarbonate, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 29d (200 mg, crude) as a white solid, which was used directly in the next step.
LC-MS:m/z=474.3[M+H] +
Fifth step N- (3- ((1R) -1- ((7- (1-methylpiperidin-3-yl) -4-oxo-3, 4-dihydrophthalazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 29e
Compound 29d (200 mg, crude) was dissolved in methanol (4 mL), aqueous formaldehyde (127 mg,1.56mmol, 37%) and palladium on carbon (50 mg, 10%) were added sequentially and stirred overnight at room temperature under hydrogen atmosphere, TLC showed complete reaction. The reaction solution was filtered through celite, and the cake was washed with methanol, and the filtrate was concentrated to give the title compound 29e (200 mg, crude) as a white solid, which was used directly in the next step.
LC-MS:m/z=488.3[M+H] +
Sixth step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -6- (1-methylpiperidin-3-yl) phthalazin-1 (2H) -one 29
Compound 29e (200 mg, crude) was dissolved in ethanol (6 mL), aqueous sodium hydroxide (3 mL,15mmol, 5M) was added at room temperature, and heated under reflux overnight. The reaction solution was concentrated, diluted with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by Prep-TLC to give the title compound 29 (37 mg, four-step yield 21%) as a yellow solid.
LC-MS:m/z=446.3[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ11.43(s,1H),8.21(s,1H),8.13(d,J=8.4Hz,1H),7.73(dd,J=8.4,0.8Hz,1H),6.92(d,J=6.8Hz,1H),6.80(d,J=6.0Hz,2H),6.66(s,1H),5.50(s,2H),4.92-4.85(m,1H),3.03-2.85(m,3H),2.25(s,3H),2.13-1.84(m,3H),1.80-1.76(m,1H),1.71-1.58(m,2H),1.50(d,J=7.2Hz,3H).(98.90%purity by HPLC)
Example 30
4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2-methyl-6- (1-methylpiperidin-3-yl) phthalazin-1 (2H) -one 30
First step (R) -5- (2-methyl-4- ((1- (3-nitro-5- (trifluoromethyl) phenyl) ethyl) amino) -1-oxo-1, 2-dihydro-phthalazin-6-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 30a
Compound 2a (200 mg,0.42 mmol) and 1-tert-butoxycarbonyl-3, 6-dihydro-2H-pyridine-5-boronic acid pinacol ester 17a (197mg, 0.64 mmol) were dissolved in 1, 4-dioxane (15 mL) and water (3 mL) and Pd (dppf) Cl was added at room temperature 2 Dichloromethane complex (18 mg,0.022 mmol) and sodium carbonate (90 mg,0.85 mmol) were heated to 100 ℃ under nitrogen and stirred for 1 hour, and TLC detection was complete. The reaction solution was cooled to room temperature, water was then added thereto, extraction was performed with ethyl acetate, and the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the crude product, 30a (240 mg, yield 99%) by silica gel column chromatography purification.
LC-MS:m/z=574.3[M+H] +
Second step 3- (4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2-methyl-1-oxo-1, 2-dihydro-phthalazin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 30b
Compound 30a (240 mg,0.42 mmol) was dissolved in tetrahydrofuran (10 mL) and ethyl acetate (6 mL), palladium on carbon (catalytic amount) was added at room temperature, and the reaction was allowed to proceed overnight at 50℃under hydrogen atmosphere, and completion of the reaction was detected by TLC. The reaction solution was cooled to room temperature, filtered through celite, and the filtrate was concentrated to give the title compound 30b (240 mg, crude) which was used directly in the next step.
LC-MS:m/z=546.3[M+H] +
Third step 3- (4- (((R) -1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -2-methyl-1-oxo-1, 2-dihydro-phthalazin-6-yl) piperidine-1-carboxylic acid tert-butyl ester 30c
Compound 30b (240 mg, crude) and N, N-diisopropylethylamine (108 mg,0.84 mmol) were dissolved in 1, 4-dioxane (60 mL), acetic anhydride (64 mg,0.63 mmol) was added, stirred overnight at room temperature, and the reaction was complete by TLC. The reaction solution was extracted with water and dichloromethane, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 30c (270 mg, crude product) which was used directly in the next step.
Fourth step N- (3- ((1R) -1- ((3-methyl-4-oxo-7- (piperidin-3-yl) -3, 4-dihydro-phthalazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 30d
Compound 30c (270 mg, crude) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL) was added and stirred at room temperature for 2 hours, and TLC detection was complete. The reaction solution was concentrated, the residue was neutralized with saturated aqueous sodium bicarbonate, extracted with dichloromethane, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound 30d (220 mg, crude) which was used directly in the next step.
LC-MS:m/z=488.3[M+H] +
Fifth step N- (3- ((1R) -1- ((3-methyl-7- (1-methylpiperidin-3-yl) -4-oxo-3, 4-dihydro-phthalazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 30e
Compound 30d (125 mg, crude) and aqueous formaldehyde (62 mg,0.76mmol, 37%) were dissolved in methanol (15 mL), palladium on carbon (catalytic amount) was added and reacted at room temperature under hydrogen atmosphere for 2 hours, and TLC detection was complete. The reaction solution was filtered, and the filtrate was concentrated to give the title compound 30e (130 mg, crude product) which was used directly in the next step.
LC-MS:m/z=502.3[M+H] +
Sixth step 4- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -2-methyl-6- (1-methylpiperidin-3-yl) phthalazin-1 (2H) -one 30
Compound 30e (130 mg, crude) was dissolved in ethanol (6 mL), sodium hydroxide (103 mg,2.58 mmol) was added at room temperature, and the reaction was allowed to proceed overnight at 90℃with TLC detection of essentially complete starting material. The reaction was cooled to room temperature, filtered, and the filtrate was concentrated, and the crude product was purified by Prep-TLC to give the title compound 30 (40 mg, five step yield 36%).
LC-MS:m/z=460.3[M+H] +
1 H NMR(400MHz,CD 3 OD)δ8.24(d,J=8.4Hz,1H),8.11(d,J=1.2Hz,1H),7.72(dd,J=1.2,8.0Hz,1H),7.00-6.97(m,2H),6.78(s,1H),5.00(q,J=7.2Hz,1H),3.56(s,3H),3.14-3.03(m,3H),2.44(s,3H),2.41-2.33(m,1H),2.27-2.19(m,1H),2.02-1.99(m,1H),1.95-1.89(m,1H),1.88-1.76(m,1H),1.71-1.64(m,1H),1.60(d,J=6.8Hz,3H).(98.98%purity by HPLC)
Example 31
1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- ((R) -1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 31-1
1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- ((S) -1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 31-2
First step (R) -5- (1- ((1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester 31a
Compound 17b (260 mg,0.46 mmol) was dissolved in ethanol (10 mL) and water (3 mL), reduced iron powder (129 mg,2.31 mmol) and ammonium chloride (125 mg,2.31 mmol) were added at room temperature, and the reaction was heated to 90℃for 3 hours, and TLC showed complete reaction of the starting material. The reaction solution was filtered through celite, the filtrate was extracted with water (10 mL), ethyl acetate (10 mL), the organic phases were combined, washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, and concentrated to give the title compound 31a (296 mg, crude) as a yellow solid, which was used directly in the next step.
Second step (R) -3- (1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) piperidine-1-carboxylic acid tert-butyl ester 31b-1& (S) -3- (1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) piperidine-1-carboxylic acid tert-butyl ester 31b-2
Compound 31a (290 mg, crude) was dissolved in ethanol (5 mL), palladium on carbon (50 mg, 10%) was added and reacted at room temperature under hydrogen atmosphere for 5 hours, TLC showed complete reaction of starting material. The reaction solution was filtered through celite, and the filtrate was concentrated to give the title compound 31b (120 mg, 46% in two steps) as a yellow solid, and the title compound 31b was chiral-resolved (cellophane AD-H,30×250mm,5 μm,20mL/min, etOH: hexane=1:9) to give the title compound 31b-1 (peak No. 1, RT 15.577 min) (40 mg, 33% in yield) and the title compound 31b-2 (peak No. 2, RT 19.628 min) (41 mg, 34% in yield) as a pale yellow solid.
The configuration and the properties of the compound need to be further detected, and the tentative 31b-1 and 31b-2 are in the above configurations.
Third step (R) -3- (1- (((R) -1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) piperidine-1-carboxylic acid tert-butyl ester 31c-1& (S) -3- (1- (((R) -1- (3-acetamido-5- (trifluoromethyl) phenyl) ethyl) amino) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-7-yl) piperidine-1-carboxylic acid tert-butyl ester 31c-2
Compound 31b-1 (40 mg,0.075 mmol) was dissolved in tetrahydrofuran (6 mL) and N, N-diisopropylethylamine (58 mg,0.45 mmol) and acetic anhydride (30 mg,0.30 mmol) were added at room temperature and heated to 35℃for 16 hours, TLC indicated complete reaction of starting materials. The reaction solution was diluted with water (10 mL), extracted with ethyl acetate (10 mL), the organic phases combined, washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, and concentrated to give the title compound 31c-1 (75 mg, crude) as a yellow solid, which was used directly in the next step.
Compound 31b-2 (41 mg,0.076 mmol) was dissolved in tetrahydrofuran (6 mL), N-diisopropylethylamine (59 mg,0.46 mmol) and acetic anhydride (31 mg,0.31 mmol) were added at room temperature, and the reaction was heated to 35℃for 16 hours, and TLC showed complete reaction of the starting material. The reaction solution was diluted with water (10 mL), extracted with ethyl acetate (10 mL), the organic phases combined, washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, and concentrated to give the title compound 31c-2 (75 mg, crude) as a yellow solid, which was used directly in the next step.
LC-MS:m/z=575.3[M+H] +
The configuration and the properties of the compound need to be further detected, and the tentative 31c-1 and 31c-2 are in the above configurations.
Fourth step N- (3- ((R) -1- ((4-oxo-7- ((R) -piperidin-3-yl) -3, 4-dihydropyrido [3,4-d ] pyridazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 31d-1&N- (3- ((R) -1- ((4-oxo-7- ((S) -piperidin-3-yl) -3, 4-dihydropyrido [3,4-d ] pyridazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 31d-2
Compound 31c-1 (75 mg, crude) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL) was added and reacted at room temperature for 2 hours, and TLC showed complete reaction of the starting material. Saturated aqueous sodium bicarbonate (15 mL) was added dropwise to the reaction solution to adjust to ph=8-9, extracted with ethyl acetate (10 mL), the organic phases were combined, washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, and concentrated to give the title compound 31d-1 (67 mg, crude) as a yellow solid, which was used directly in the next step.
Compound 31c-2 (75 mg, crude) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (2 mL) was added and reacted at room temperature for 2 hours, and TLC showed complete reaction of the starting material. Saturated aqueous sodium bicarbonate (15 mL) was added dropwise to the reaction solution to adjust to ph=8-9, extracted with ethyl acetate (10 mL), the organic phases were combined, washed with saturated brine (10 mL), dried over anhydrous sodium sulfate, and concentrated to give the title compound 31d-2 (50 mg, crude) as a yellow solid, which was used directly in the next step.
The configuration and the properties of the compound need to be further detected, and the tentative 31d-1 and 31d-2 are the configurations.
Fifth step N- (3- ((R) -1- ((7- ((R) -1-methylpiperidin-3-yl) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 31e-1&N- (3- ((R) -1- ((7- ((S) -1-methylpiperidin-3-yl) -4-oxo-3, 4-dihydropyrido [3,4-d ] pyridazin-1-yl) amino) ethyl) -5- (trifluoromethyl) phenyl) acetamide 31e-2
Compound 31d-1 (67 mg, crude) was dissolved in ethanol (3 mL), aqueous formaldehyde (35 mg,0.43mmol, 30%) and palladium on carbon (20 mg, 10%) were added and reacted at room temperature under hydrogen atmosphere for 4 hours, and TLC showed complete reaction of the starting materials. The reaction solution was filtered through celite, and the filtrate was concentrated to give the title compound 31e-1 (70 mg, crude) as a yellow solid, which was used directly in the next step.
Compound 31d-2 (50 mg, crude) was dissolved in ethanol (3 mL), aqueous formaldehyde (35 mg,0.43mmol, 30%) and palladium on carbon (20 mg, 10%) were added and reacted at room temperature under hydrogen atmosphere for 4 hours, and TLC showed complete reaction of the starting materials. The reaction solution was filtered through celite, and the filtrate was concentrated to give the title compound 31e-2 (60 mg, crude) as a yellow solid, which was used directly in the next step.
The configuration and the properties of the compound need to be further detected, and the tentative 31e-1 and 31e-2 are in the above configurations.
Sixth step 1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- ((R) -1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 31-1&1- (((R) -1- (3-amino-5- (trifluoromethyl) phenyl) ethyl) amino) -7- ((S) -1-methylpiperidin-3-yl) pyrido [3,4-d ] pyridazin-4 (3H) -one 31-2
Compound 31e-1 (70 mg, crude) was dissolved in methanol (3 mL), aqueous sodium hydroxide (2 mL,8mmol, 4N) was added at room temperature, and the reaction was heated to 70℃for 16 hours, and TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, diluted with water (10 mL), extracted with ethyl acetate (10 mL), the combined organic phases, saturated brine (10 mL), dried over anhydrous sodium sulfate, and concentrated to give the crude Prep-TLC purified (dichloromethane/ammonia in methanol=10/1) as a pale yellow solid title compound 31-1 (15 mg, four-step yield 45%).
LC-MS:m/z=447.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.43(s,1H),8.13(s,1H),6.96-6.89(m,2H),6.79-6.75(m,1H),4.96(q,J=6.8Hz,1H),3.49-3.39(m,2H),3.28-3.20(m,1H),3.18-2.99(m,1H),2.83-2.62(m,4H),2.24-2.12(m,1H), 2.03-1.82(m,3H),1.58(d,J=6.8Hz,3H).(96.67%purity by HPLC)
Compound 31e-2 (60 mg, crude) was dissolved in methanol (3 mL), aqueous sodium hydroxide (2 mL,8mmol, 4N) was added at room temperature, and the reaction was heated to 70℃for 16 hours, and TLC showed complete reaction of starting material. The reaction solution was cooled to room temperature, diluted with water (10 mL), extracted with ethyl acetate (10 mL), the combined organic phases, saturated brine (10 mL), dried over anhydrous sodium sulfate, and concentrated to give the crude Prep-TLC purified (dichloromethane/ammonia in methanol=10/1) as a pale yellow solid title compound 31-2 (13 mg, four step yield 37%).
LC-MS:m/z=447.2[M+H] +
1 H NMR(400MHz,CD 3 OD)δ9.41(s,1H),8.09(s,1H),6.98-6.88(m,2H),6.81-6.73(m,1H),4.95(q,J=6.8Hz,1H),3.30-3.20(m,2H),3.13-3.01(m,1H),2.75-2.58(m,1H),2.51(s,3H),2.44-2.28(m,1H),2.16-2.05(m,1H),1.97-1.71(m,3H),1.58(d,J=6.8Hz,3H).(99.69%purity by HPLC)
The configuration and the property of the compound need to be further detected, and the tentative 31-1 and 31-2 are in the above configurations.
Test example 1 Compounds for inhibition of K-562 cell proliferation 50 Measurement
Human chronic myelogenous leukemia cell K-562 (CCL-243) used in the present invention was purchased from American Type Culture Collection (ATCC). Cells in RPMI 1640 medium with 10% Fetal Bovine Serum (FBS) and 1% diabody at 37 ℃,5% co 2 Is grown in the environment of (a).
The inhibition of proliferation of K-562 cells cultured in vitro by the compounds was determined by the following method:
1) Cell inoculation: inoculating K-562 cells in good logarithmic phase into 96-well plate at 20000 cells/well and 90 μl, inoculating at 37deg.C with 5% CO 2 Culturing for 24 hours under the condition.
2) Adding the medicine: the compound to be tested was diluted in gradient with complete medium and 10. Mu.L of diluted compound was added to 9In 0. Mu.L of cells, the final concentration of the compound was 10000, 3000, 1000, 300, 100, 30, 10, 3, 1nM, and the corresponding vehicle control was set. Placing at 37deg.C and 5% CO 2 The cells were cultured in a cell incubator for 96 hours.
3) And (3) detection: after adding 10. Mu.L of 5mg/mL MTT working solution (ABCONE, M9609) per well and allowing to act at 37℃for 4 hours, a triple solution (10% SDS,0.5% isopropyl alcohol, 0.1mol/L HCL) was added until the cell lysate was completely dissolved, and OD570 and OD690 values were read using a TECAN SPARK microplate reader.
4) And (3) calculating: the cell growth inhibition was calculated by the following formula:
inhibition = (control wells OD570nm-OD690nm -an administration orifice OD570nm-OD690nm ) Control well OD570nm-OD690nm ×100%
Calculation of IC from compound concentration and corresponding inhibition ratio using Graphpad prism 5.0 software 50 Values. The test results are shown in Table 1.
TABLE 1 IC for the proliferation inhibition of K-562 cells by the compounds of the present invention 50 (nM)
Numbering of compounds IC 50 (nM) Numbering of compounds IC 50 (nM) Numbering of compounds IC 50 (nM)
BI3406 35.2 11 1619 22 107.7
1 735.7 12 134 23 142.8
2 822.7 13 104 24 231.9
3 111.1 14 362.3 25 125.9
4 150.8 15 279.25 26 137.6
5 44.7 16 43.8 27 91.3
6 2489 17 33.1 28 66.65
7 5917 18 60.0 29 51.0
8 1599 19 63.4 30 143.9
9 988 20 39.3 31-1 43.4
10 121.4 21 97.7 31-2 13.1
Conclusion: as shown in Table 1, the compounds of the examples of the present invention have proliferation inhibitory effect on K-562 cells, and the activity of a plurality of compounds is equivalent to that of BI-3402.
Test example 2 Effect of Compounds on the level of ERK1/2 phosphorylation of the KRAS downstream Signal molecule in K-562 cells
The effect of the compounds of the invention on ERK1/2 phosphorylation levels in K-562 cells was detected by the following method:
1) Cell inoculation: taking K-562 cells with good logarithmic growth phase at 1×10 6 The cells/wells were inoculated into six well plates and incubated overnight at 37℃under 5% CO 2.
2) Adding the medicine: the compounds to be tested were added to the cells after gradient dilution in complete medium to give final concentrations of 1000, 100, 10, 1nM. Placing at 37deg.C and 5% CO 2 The cells were cultured in a cell incubator for 24 hours.
3) Protein sample preparation: cell suspensions were collected, centrifuged at 500g for 5 min, the supernatant was discarded, washed 3 times with PBS, and 100. Mu.L of cells were lysed with 1 XSDS gel loading buffer (50 mM Tris-HCl (pH 6.8), 100mM DTT,2%SDS,10% glycerol, 0.1% bromophenol blue). Cell lysates were denatured by heating at 100℃for 10 min.
4) Western blot: carrying out SDS-PAGE electrophoresis on a protein sample, transferring the protein to a PVDF membrane by a wet transfer system after the electrophoresis is finished, placing the PVDF membrane in a sealing solution (5% skimmed milk powder is diluted in TBS/T) for sealing for 1 hour at room temperature, and then resisting reaction I and II; after washing the membrane, color development was performed with Immobilon Western HRP Substrate luminal reagent reagent and a Western Blot imager (Tanon, 4600) was used to photograph. The following are the antibody information used: p-ERK1/2 (CST: 4370); ERK1/2 (CST: 9102); beta-tubulin (CST: 2146); GAPDH (CST: 5174). The effect of compounds on ERK1/2 phosphorylation in K-562 cells is shown in FIGS. 1 and 2.
Conclusion: the compounds 5, 20, 31-1 and 31-2 in the embodiment of the invention have obvious inhibition effect on the phosphorylation of ERK1/2 of K-562 cells; inhibitory activity was concentration gradient dependent; overall, the activity was approximately comparable to that of BI-3406.
Test example 3 Compounds for SOS1 inhibitory Activity
The experimental steps are as follows:
1. treatment of the compound: the compound was formulated at 400-fold final concentration, e.g., 5uM for detection, and at 400-fold concentration, i.e., 2mM. The compound was diluted in gradient to the set number of concentration spots with an automated microwell pipette.
2. Transfer compounds to 384 well plate reaction plates: the diluted compounds were transferred from Echo 384 well plates to 384 well reaction plates using an ultrasonic nanoliter liquid treatment system, and both negative and positive controls transferred 50nL of 100% dmso.
3. 4-fold Tag1-SOS1 solution was prepared and transferred: a4-fold Tag1-SOS1 solution was prepared with the reagent provided in the kit (KRAS-G12C/SOS 1BINGDING ASSAY KIT (Cisbio, cat. No.63 ADK000CB16PEG)), transferred into 5ul to 384 well reaction plates, and for negative control wells, transferred 5ul of reagent instead of enzyme solution, and centrifuged at 1000rpm for 1 min.
4. 4 times Tag2-KRAS G12C solution was prepared: a4-fold Tag2-KRAS G12C solution was prepared with the reagent provided in the kit, transferred to a 5ul 384 well reaction plate, and centrifuged at 1000rpm for 1 minute.
5. Transfer 2-fold detection solution: 2-fold Anti-Tag1-Tb3+ and Anti-Tag2-XL665 solutions were prepared using the Detection Buffer provided in the kit, transferred to 10ul to 384 well reaction plates, centrifuged at 1000rpm for 1 min and incubated at room temperature for 60 min.
6. Reading: the data fluorescence signal values were read with a microplate reader Envision (Ex 665/Em 615).
7. Inhibition calculation and IC50 fitting
Values were copied from the plate reader, where maximum refers to the reading of the positive control and minimum refers to the reading of the negative control. Inhibition ratio (%) = (maximum value-sample value)/(maximum value-minimum value) ×100%.
Data were imported into MS Excel and IC50 values were fitted with XLFIT Excel add-in version 5.4.0.8;
inhibitory Activity of Compounds of Table 2 on SOS1
Numbering of compounds IC 50 (nM) Numbering of compounds IC 50 (nM) Numbering of compounds IC 50 (nM)
3 15 4 13 5 7.6
10 16 12 9.7 13 7.8
14 18 15 14 16 8.0
17 7.7 18 12 19 6.8
20 6.0 21 20 22 29
23 5.4 24 18 25 11
26 30 28 12 29 14
30 23
Test example 4 Compounds for liver microsomal stability experiments on mice and humans
The experimental steps are as follows:
(1) Taking out liver microsomes (20 mg protein/mL) from a refrigerator at-80 ℃, placing the liver microsomes on a water bath constant temperature oscillator at 37 ℃ for pre-incubation for 3min, and melting for later use.
(2) According to the above "constitution of the experimental incubation system", a mixed solution of the incubation system (without beta-NADPH) was prepared.
(3) Preparing 100 mu M test compound working solution for later use.
(4) Control group (without β -NADPH): and (3) respectively taking 25 mu L of PB solution into 75 mu L of the mixed solution of the incubation system (2), swirling for 30s, uniformly mixing, and carrying out reaction with the total volume of 100 mu L, and repeating. Incubation was performed in a 37℃water bath thermostatted shaker and timing was started with sampling time points of 0min and 60min.
(5) Sample group: mu.L of beta-NADPH solution (4 mM) was added to 75. Mu.L of the reaction system (2), and the mixture was vortexed for 30s, mixed well, and the total volume of the reaction was 100. Mu.L, and the reaction was repeated. Incubation was performed in a 37℃water bath thermostated shaker and timing was started with sampling times of 0min,5min,15min,30min,60min.
(6) The sample tube was removed at each time point and 300. Mu.L of cold stop reagent (containing internal standard) was added to stop the reaction.
(7) Vortex and centrifuge.
(8) 150 μl of the supernatant was added with 150 μl of water, vortexed and mixed well, and analyzed by LC-MS/MS.
Data analysis: half-life (t) was calculated using the following first order kinetic equation 1/2 ) And Clearance (CL)
C t =C 0 *e -kt
C t =(1/2)*C 0
t 1/2 =ln2/k=0.693/k
CL=V d *k
Vd=1/protein content in liver microsomes
CL int(liver) =CL int(mic) X liver weight to weight ratio x liver microsomal protein concentration per gram of liver
The parameters in the formula are shown in Table 3:
TABLE 3 common parameters of liver and blood in mice, rats, humans
The experimental results are shown in table 4:
liver microsomal stability of the compounds of Table 4 in different species
In the human liver microsome stability experiment, the human microsome stability of a plurality of compounds is equivalent to that of BI-3406, and the mouse liver microsome stability is superior to that of BI-3406, which is more beneficial to the development of drug research.
Test of the mouse pharmacokinetic Properties of the Compounds of example 5
The compound of example 20 (10 mg/kg) was orally administered to overnight fasted ICR mice (female, n=3). Blood samples were collected before and 0.25, 0.5, 1, 2, 4 and 8 hours after the administration, respectively, and centrifuged (4500 rpm) at 4℃for 10min to obtain serum. mu.L of MeOH/ACN (1:1, v/v) was added to 10. Mu.L of serum to precipitate the mixture, which was then vortexed for 1min and centrifuged (11000 rpm) for 5min to obtain a supernatant. mu.L of the supernatant was dissolved in 20. Mu.L of ACN/H 2 O (1:1, v/v) and analyzed by ultra performance liquid chromatography. The results are shown in Table 5:
table 5 pharmacokinetic profile of compound 20 in mice
As can be seen from table 5, compound 20 had better plasma exposure in mice.
The applicant states that the present invention is illustrated by the above examples as a polycyclic pyridazinone derivative of the present invention as an SOS1 inhibitor, and a method for preparing the same and use thereof, but the present invention is not limited to the above examples, i.e., it is not meant that the present invention must be practiced depending on the above examples. It should be apparent to those skilled in the art that any modification of the present invention, equivalent substitution of raw materials for the product of the present invention, addition of auxiliary components, selection of specific modes, etc., falls within the scope of the present invention and the scope of disclosure.
The preferred embodiments of the present invention have been described in detail above, but the present invention is not limited to the specific details of the above embodiments, and various simple modifications can be made to the technical solution of the present invention within the scope of the technical concept of the present invention, and all the simple modifications belong to the protection scope of the present invention.
In addition, the specific features described in the above embodiments may be combined in any suitable manner, and in order to avoid unnecessary repetition, various possible combinations are not described further.

Claims (12)

  1. A polycyclic pyridazinone derivative, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof,
    the polycyclic pyridazinone derivative is characterized in that the structure of the polycyclic pyridazinone derivative is shown as a formula (I):
    wherein: r is R 1 Selected from hydrogen or methyl;
    R 2 selected from C 1 -C 3 Alkyl, -OR 21 Halogen, 3-7 membered cycloalkyl, 5-7 membered cycloalkenyl, 6-10 membered fused ring alkyl, 7-10 membered bridged cycloalkyl, 7-10 membered spirocycloalkyl, 4-7 membered heterocyclyl, 5-7 membered heterocyclenyl, 6-10 membered fused heterocyclyl, 7-10 membered bridged heterocyclyl, 7-10 membered spiroheterocyclyl, wherein 3-7 membered cycloalkyl, 5-7 membered cycloalkenyl, 6-10 membered fused ring alkyl, 7-10 membered bridged cycloalkyl, 7-10 membered spirocycloalkyl, 4-7 membered heterocyclyl, 5-7 membered heterocyclenyl, 6-10 membered fused heterocyclyl, 7-10 membered bridged heterocyclyl, 7-10 membered spiroheterocyclyl is optionally substituted with 1-3R 22 Substituted;
    R 21 selected from H, C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl, wherein C 1 -C 3 Alkyl, 3-7 membered cycloalkyl, 4-7 membered heterocyclyl are optionally substituted with 1-3R 22 Substituted;
    R 22 selected from C 1 -C 3 Alkyl, hydroxy, halogen, cyano, -NR a R b 、C 1 -C 3 Alkoxy, -C (O) R a 、-C(O)OR a 、-OC(O)R a 、-NR b C(O)R a 、-NR b C(O)OR a 、-C(O)NR a R b Phenyl, 5-6 membered heteroaryl and =o, wherein alkyl, alkoxy, phenyl, 5-6 membered heteroaryl are optionally further substituted with 1-3 halogen, C 1 -C 3 Alkyl, hydroxy, cyano, amino and C 1 -C 3 Alkoxy substituted;
    R a and R is b Independently selected from H, substituted or unsubstituted C 1 -C 3 Substituted or unsubstituted 3-6 membered cycloalkyl or substituted or unsubstituted 4-7 membered heterocyclyl; "substituted" herein means optionally substituted with 1 to 3 substituents selected from C 1 -C 3 Is substituted by alkyl, hydroxy, halogen, cyano, amino or alkoxy;
    q is selected from N or-CR 3
    R 3 Selected from H, C 1 -C 3 Alkyl, halogen, cyano OR-OR 21
    AR is selected from 6-10 membered aryl or 5-10 membered heteroaryl, wherein aryl or heteroaryl is optionally substituted with 1-4R c Substituted;
    R c selected from H, halogen, C 1 -C 4 Alkyl, C 1 -C 4 Haloalkyl, hydroxy-C 1 -C 4 Alkyl, hydroxy-C 1 -C 4 Haloalkyl, 3-6 membered cycloalkyl, 4-7 membered heterocyclyl, -OR 21 、-NR a R b 、NR a R b -C 1 -C 4 Alkyl, NR a R b -C 1 -C 4 Haloalkyl, 6-10 membered aryl or 5-10 membered heteroaryl, wherein 6-10 membered aryl or 5-10 membered heteroaryl is optionally substituted with 1-4R d Substituted;
    R d selected from H, halogen, C 1 -C 4 Alkyl, C 1 -C 4 Haloalkyl, hydroxy-C 1 -C 4 Alkyl, hydroxy-C 1 -C 4 Haloalkyl, 3-6 membered cycloalkyl, 4-7 membered heterocyclyl, -OR 21 、-NR a R b 、NR a R b -C 1 -C 4 Alkyl, NR a R b -C 1 -C 4 A haloalkyl group;
    the hetero atoms in the heterocyclic group, the heteroaryl, the heterocyclenyl, the fused heterocyclic group, the bridged heterocyclic group and the spiro heterocyclic group in the formula (I) are 1-7 and are selected from one or more of oxygen, nitrogen, sulfur and S (O) m, and m is 1 or 2.
  2. The polycyclic pyridazinone derivative, the pharmaceutically acceptable salt thereof, the tautomer thereof or the stereoisomer thereof according to claim 1,
    the structure of the polycyclic pyridazinone derivative is shown as a formula (II):
    wherein R is 1 、R 2 Q and R c Having the same limitations as claim 1; n=1 to 4;
    preferably, for compounds of formula (II), the phenyl group is optionally substituted with 1 to 3R c Substituted when said R c When the number of R is 2-3, R is c May be the same or different;
    and/or when said R c Is C 1 -C 4 When halogenoalkyl, the halogen atom is fluorine;
    and/or when said R c When halogen, the halogen atom is fluorine;
    and/or when said R c is-NR a R b When said R is a And R is b May be the same or different.
  3. The polycyclopyridazinone derivative, pharmaceutically acceptable salt thereof, tautomer thereof or stereoisomer thereof according to claim 1 or 2, wherein the structure of the polycyclopyridazinone derivative is as shown in formula (III):
    Wherein R is 1 、R 2 And R is c Having the same limitations as claim 1; n=1 to 4;
    preferably, for the compound of formula (III), the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 When the number of the components is 2-3, R 22 The same or different;
    and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclyl, the heterocyclyl contains 1-2 heteroatoms;
    and/or, the R 2 Is optionally covered with 1-3R 22 When the substituted 4-7 membered heterocyclic group is, the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
    and/or, the R 2 Is optionally covered with 1-3R 22 When the number of the hetero atoms of the heterocyclic group is two, the two hetero atoms are the same or different;
    and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 Selected from C 1 -C 3 Alkyl, -NR a R b 、-C(O)R a 、-NR b C(O)R a 、-NR b C(O)OR a 、-C(O)NR a R b And = O.
  4. The polycyclopyridazinone derivative, pharmaceutically acceptable salt thereof, tautomer thereof or stereoisomer thereof according to claim 1 or 2, wherein the structure of the polycyclopyridazinone derivative is as shown in formula (IV):
    wherein R is 1 、R 2 、R 3 And R is c Having the same limitations as claim 1; n=1 to 4;
    preferably, for the compound of formula (IV), the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 When the number of the components is 2-3, R 22 The same or different;
    and/or, the R 2 Is optionally covered with 1-3R 22 Substituted 4-7 membered heterocyclyl groups, when present, areHeterocyclyl contains 1-2 heteroatoms;
    and/or, the R 2 Is optionally covered with 1-3R 22 When the substituted 4-7 membered heterocyclic group is, the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
    and/or, the R 2 Is optionally covered with 1-3R 22 When the number of the hetero atoms of the heterocyclic group is two, the two hetero atoms are the same or different;
    and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 Selected from C 1 -C 3 Alkyl, -NR a R b 、-C(O)R a 、-NR b C(O)R a 、-NR b C(O)OR a 、-C(O)NR a R b And = O;
    and/or, the R 3 is-OR 21 When R is 21 Selected from unsubstituted C 1 -C 3 Alkyl or unsubstituted 3-7 membered cycloalkyl.
  5. The polycyclopyridazinone derivative, pharmaceutically acceptable salt thereof, tautomer thereof or stereoisomer thereof according to claim 1, wherein the structure of the polycyclopyridazinone derivative is as shown in formula (V):
    wherein R is 1 、R 2 Q and R d Having the same limitations as claim 1; n=1 to 4;
    preferably, for the compound of formula (V), the R d Is 1-NR a R b When said R is a And R is b Independently selected from H, substituted or unsubstituted C 1 -C 3 Alkyl of (a);
    and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 When the number of the components is 2-3, R 22 The same or different;
    and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclyl, the heterocyclyl contains 1-2 heteroatoms;
    and/or, the R 2 Is optionally covered with 1-3R 22 When the substituted 4-7 membered heterocyclic group is, the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
    and/or, the R 2 Is optionally covered with 1-3R 22 When the number of the hetero atoms of the heterocyclic group is two, the two hetero atoms are the same or different;
    and/or, the R 2 Is optionally covered with 1-3R 22 When substituted 4-7 membered heterocyclic group, the R 22 Selected from C 1 -C 3 Alkyl, -NR a R b 、-C(O)R a 、-NR b C(O)R a 、-NR b C(O)OR a 、-C(O)NR a R b And = O;
    and/or, the R 2 is-OR 21 When R is 21 Is a 4-7 membered heterocyclic group;
    and/or, the R 21 In the case of 4-7 membered heterocyclic group, the 4-7 membered heterocyclic group is 5-6 membered heterocyclic group;
    and/or, the R 21 In the case of 4-7 membered heterocyclic groups, the heteroatoms of the heterocyclic groups are nitrogen and/or oxygen;
    and/or, the R 21 In the case of a 4-7 membered heterocyclic group, the heterocyclic group contains 1-2 heteroatoms;
    and/or, the R 21 In the case of 4-7 membered heterocyclic groups, when the number of heteroatoms in the heterocyclic group is two, the two heteroatoms may be the same or different.
  6. The polycyclopyridazinone derivative, pharmaceutically acceptable salt thereof, tautomer thereof or stereoisomer thereof according to claim 1 or 5, wherein the structure of the polycyclopyridazinone derivative is as shown in formula (VI):
    wherein R is 1 、R 2 And R is d Having the same limitations as claim 1; n=1 to 4;
    preferably, when said R 2 is-OR 21 The R is 21 Is a 4-7 membered heterocyclic group;
    and/or when said R 21 In the case of 4-7 membered heterocyclic group, the 4-7 membered heterocyclic group is 5-7 membered heterocyclic group;
    and/or when said R 21 In the case of 4-7 membered heterocyclic groups, the heteroatoms of the heterocyclic groups are nitrogen and/or oxygen;
    and/or when said R 21 In the case of a 4-7 membered heterocyclic group, the heterocyclic group contains two heteroatoms;
    and/or when said R 21 In the case of a 4-7 membered heterocyclyl, the two heteroatoms may be the same or different;
    and/or, the R d Selected from halogen, C 1 -C 4 Alkyl, -NR a R b 、-OR 21 、-NR a R b -C 1 -C 4 An alkyl group;
    and/or, the R d Is 1-NR a R b -C 1 -C 4 In the case of alkyl, said R a And R is b Independently selected from H, substituted or unsubstituted C 1 -C 3 Is a hydrocarbon group.
  7. The polycyclopyridazinone derivative, pharmaceutically acceptable salt thereof, tautomer thereof or stereoisomer thereof according to claim 1 or 5, wherein the structure of the polycyclopyridazinone derivative is as shown in formula (VII):
    Wherein R is 1 、R 2 、R 3 And R is d Having the same limitations as claim 1; n=1 to 4;
    preferably, when said R 2 is-OR 21 The R is 21 Is a 4-7 membered heterocyclic group;
    and/or when said R 21 In the case of 4-7 membered heterocyclic group, the 4-7 membered heterocyclic group is 5-7 membered heterocyclic group;
    and/or when said R 21 In the case of 4-7 membered heterocyclic groups, the heteroatoms of the heterocyclic groups are nitrogen and/or oxygen;
    and/or when said R 21 In the case of a 4-7 membered heterocyclic group, the heterocyclic group contains two heteroatoms;
    and/or when said R 21 In the case of a 4-7 membered heterocyclyl, the two heteroatoms may be the same or different;
    and/or, the R d Selected from halogen, C 1 -C 4 Alkyl, -NR a R b 、-OR 21 、NR a R b -C 1 -C 4 An alkyl group;
    and/or, the R d Is 1 NR a R b -C 1 -C 4 In the case of alkyl, said R a And R is b Independently selected from H, substituted or unsubstituted C 1 -C 3 Is a hydrocarbon group.
  8. The polycyclopyridazinone derivative, pharmaceutically acceptable salt thereof, tautomer thereof or stereoisomer thereof according to claim 1, wherein the polycyclopyridazinone derivative is selected from any one of the following structures:
  9. a process for the preparation of a polycyclic pyridazinone derivative according to any one of claims 1-8, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof selected from one of two processes:
    A first method,
    The first step, the compound of the general formula (I-A) and the compound of the general formula (I-B) are subjected to imine addition reaction to obtain the compound of the general formula (I-8);
    secondly, performing imine addition and ring expansion reaction on the compound shown in the general formula (I-8) and hydrazine hydrate to obtain a compound shown in the general formula (I-9);
    thirdly, carrying out substitution reaction on the compound of the general formula (I-9) and the compound of the general formula (I-10) to obtain a compound of the general formula (I-11);
    fourthly, carrying out Buchwald/Suzuki reaction on the compound shown in the general formula (I-11) and the compound shown in the general formula (I-25) under alkaline conditions in the presence of a metal catalyst and a ligand to obtain the compound shown in the general formula (I);
    therein, X, X 1 Is halogen, X is preferably bromine or chlorine, X 1 Preferably iodine; w is H,Q、AR、R 1 And R is 2 Having the same limitations as claim 1; when R is 1 And when H is H, omitting the third step.
    A second method,
    Step one, carrying out substitution reaction on a compound of a general formula (I-17) to obtain a compound of a general formula (I-18);
    secondly, removing protecting groups from the compound of the general formula (I-18) under acidic conditions to obtain a compound of the general formula (I-19);
    thirdly, the compound of the general formula (I-19) and the compound of the general formula (I-26) are subjected to alkaline conditions to obtain the compound of the general formula (I-20);
    fourth, the compound of the general formula (I-20) is subjected to oxidation reaction to obtain a compound of the general formula (I-21);
    Fifthly, carrying out Bouveault aldehyde synthesis reaction on the compound shown in the general formula (I-21) to obtain a compound shown in the general formula (I-22);
    step six, the compound of the general formula (I-22) and hydrazine hydrate undergo an addition cyclization reaction to obtain a compound of the general formula (I-23);
    seventhly, carrying out substitution reaction on the compound of the general formula (I-23) to obtain a compound of the general formula (I-24);
    eighth, the compound of the general formula (I-24) and the compound of the general formula (I-10) are subjected to substitution reaction to obtain the compound of the general formula (I-Bb);
    ninth, the compound of the general formula (I-Bb) and the compound of the general formula (I-A) are subjected to Buchwald reaction under alkaline conditions in the presence of a metal catalyst and a ligand to obtain the compound of the general formula (I);
    wherein X is 2 、X 3 、X 4 Is halogen, X 2 、X 3 Preferably bromine, X 4 Preferably iodine; q is selected from N or CR 3 ;R 3 Selected from H, C1-C3 alkyl, halogen, cyano OR-OR 21 ;R 1 Selected from hydrogen or methyl; AR and R 2 Having the same limitations as claim 1; when R is 1 When H is H, the eighth step is omitted.
  10. A pharmaceutical composition comprising a polycyclic pyridazinone derivative according to any one of claims 1-8, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof; preferably, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier and/or excipient.
  11. Use of a polycyclic pyridazinone derivative according to any one of claims 1-8, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof, or a pharmaceutical composition according to claim 10 for the manufacture of a medicament for the treatment of cancer or for the manufacture of a SOS1 inhibitor;
    preferably, the cancer is pancreatic cancer, colorectal cancer, lung cancer, hepatocellular cancer, renal cancer, gastric cancer, or cholangiocarcinoma.
  12. A method for preventing and/or treating cancer comprising administering to a human a therapeutically effective amount of a polycyclic pyridazinone derivative according to any one of claims 1-8, a pharmaceutically acceptable salt thereof, a tautomer thereof or a stereoisomer thereof or a pharmaceutical composition according to claim 10.
CN202280007414.XA 2021-02-09 2022-01-24 Polycyclic pyridazinone derivative serving as SOS1 inhibitor, and preparation method and application thereof Pending CN116635371A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN202110178538 2021-02-09
CN2021101785381 2021-02-09
PCT/CN2022/073466 WO2022170952A1 (en) 2021-02-09 2022-01-24 Polycyclic pyridazinone derivative serving as sos1 inhibitor, preparation method therefor and use thereof

Publications (1)

Publication Number Publication Date
CN116635371A true CN116635371A (en) 2023-08-22

Family

ID=82837444

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280007414.XA Pending CN116635371A (en) 2021-02-09 2022-01-24 Polycyclic pyridazinone derivative serving as SOS1 inhibitor, and preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN116635371A (en)
WO (1) WO2022170952A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW202334095A (en) * 2021-12-23 2023-09-01 大陸商北京望實智慧科技有限公司 SOS1 inhibitor
WO2024074827A1 (en) 2022-10-05 2024-04-11 Sevenless Therapeutics Limited New treatments for pain
CN115819251B (en) * 2022-12-20 2024-04-09 沧州普瑞东方科技有限公司 Preparation method of (1R) -1- [3- (difluoromethyl) -2-fluorophenyl ] ethylamine

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL155645A0 (en) * 2000-10-30 2003-11-23 Kudos Pharm Ltd Phthalazinone derivatives
MXPA04001353A (en) * 2001-08-15 2004-10-27 Icos Corp 2h-phthalazin-1-ones and methods for use thereof.
AUPS019702A0 (en) * 2002-01-29 2002-02-21 Fujisawa Pharmaceutical Co., Ltd. Condensed heterocyclic compounds
EP1771175B1 (en) * 2004-06-30 2015-12-23 Janssen Pharmaceutica NV Phthalazine derivatives as parp inhibitors
CN107074824B (en) * 2014-09-05 2021-01-08 基因泰克公司 Phthalazine derivatives of formula (I) as PCAF and GCN5 inhibitors for the treatment of cancer
CN107417687A (en) * 2016-05-24 2017-12-01 中国科学院上海药物研究所 Five-ring heterocycles simultaneously [3,4 d] pyridazinone compound, its preparation method, pharmaceutical composition and its application
CN109280032B (en) * 2017-07-19 2023-05-12 中国科学院上海药物研究所 Pyridazinone mother nucleus structure histone deacetylase inhibitor and preparation method and application thereof
US20230312482A1 (en) * 2020-07-28 2023-10-05 Mirati Therapeutics, Inc. Sos1 inhibitors

Also Published As

Publication number Publication date
WO2022170952A1 (en) 2022-08-18

Similar Documents

Publication Publication Date Title
CN111704611B (en) Aryl spiro SHP2 inhibitor compound, preparation method and application
AU2016228660B2 (en) Substituted 2-hydrogen-pyrazole derivative serving as anticancer drug
AU2012323085B2 (en) PDE9i with imidazo pyrazinone backbone
CN115232154A (en) Heterocyclic derivative inhibitor, preparation method and application thereof
CN114456176A (en) Pyrrolotriazine compounds as TAM inhibitors
CN116635371A (en) Polycyclic pyridazinone derivative serving as SOS1 inhibitor, and preparation method and application thereof
AU2013343105B2 (en) Heteroaromatic compounds as dopamine D1 ligands
AU2018212593A1 (en) Tyrosine amide derivatives as Rho- kinase inhibitors
EP2178874A1 (en) Heterocyclic compounds useful as mk2 inhibitors
WO2022132914A1 (en) Prmts inhibitors
EP3414234A1 (en) Bruton's tyrosine kinase inhibitors
CA2624649A1 (en) Novel imidazo [4,5 -b] pyridine derivatives as inhibitors of glycogen synthase kinase 3 for use in the treatment of dementia and neurodegenerative disorders
CN113061132B (en) Condensed ring lactam compound, preparation method and application
EP3638680B1 (en) Heteroaromatic compounds as vanin inhibitors
CN114685487A (en) Pyrimidine heterocyclic compound, preparation method and application
JP2022517085A (en) Halogenated allylamine compounds and their applications
WO2016192630A1 (en) Compound having kinase inhibiting activity, method of preparing same, and use of same
CN115485278A (en) Degradation of Bruton's Tyrosine Kinase (BTK) by conjugation of BTK inhibitors with E3 ligase ligands and methods of use thereof
CN116669738A (en) Pyrimidopyridone derivative as SOS1 inhibitor, and preparation method and application thereof
JP2022530371A (en) Pyrimid 5-membered heterocyclic compound and its use as a mutant IDH2 inhibitor
CN116568689A (en) Polycyclic pyrimidine derivative as SOS1 inhibitor, and preparation method and application thereof
KR20210105375A (en) Macrocyclic compounds as CDK inhibitors, methods for their preparation, and their use in medicine
CN112300173B (en) Nitrogen-containing polycyclic compounds, preparation method and application
KR20160086930A (en) Pyrrolopyrrolone derivatives and their use as bet inhibitors
CN111057048B (en) Aminopyrazine/pyridine compound, preparation method and application

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination