CN112142735B - Condensed cyanopyridine compound, preparation method and application - Google Patents

Condensed cyanopyridine compound, preparation method and application Download PDF

Info

Publication number
CN112142735B
CN112142735B CN202011126559.0A CN202011126559A CN112142735B CN 112142735 B CN112142735 B CN 112142735B CN 202011126559 A CN202011126559 A CN 202011126559A CN 112142735 B CN112142735 B CN 112142735B
Authority
CN
China
Prior art keywords
compound
group
hydrogen
pharmaceutically acceptable
independently selected
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202011126559.0A
Other languages
Chinese (zh)
Other versions
CN112142735A (en
Inventor
万惠新
查传涛
马金贵
潘建峰
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.)
Shanghai Lingji Biotechnology Co Ltd
Shanghai Lingda Biomedical Co Ltd
Original Assignee
Shanghai Lingji Biotechnology Co Ltd
Shanghai Lingda Biomedical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Lingji Biotechnology Co Ltd, Shanghai Lingda Biomedical Co Ltd filed Critical Shanghai Lingji Biotechnology Co Ltd
Publication of CN112142735A publication Critical patent/CN112142735A/en
Application granted granted Critical
Publication of CN112142735B publication Critical patent/CN112142735B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

Abstract

The invention discloses a condensed cyanopyridine compound shown as a general formula I-1 or I-2, or a pharmaceutically acceptable salt thereof, or an enantiomer, a diastereoisomer, a tautomer, a torsional isomer, a solvate, a polymorph or a prodrug thereof, a preparation method and a pharmaceutical application thereof, wherein the definition of each group is described in the specification.

Description

Condensed cyanopyridine compound, preparation method and application
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and particularly relates to a condensed cyanopyridine compound, a compound with Ras mutein inhibition activity, a preparation method and application.
Background
Ras is the first oncogene identified in human tumors and was first found in two murine sarcoma viruses. There are three members of the RAS gene family, H-RAS, K-RAS, and N-RAS. In human tumors, K-Ras mutations are most common, accounting for approximately 85%. Previous studies have shown that K-Ras mutations are oncogenic because missense mutations occur at codon 12, altering the structure of the K-Ras protein and leaving it activated at all times. Ras plays a major role in signal pathway transmission, mainly activating kinases controlling gene transcription, thereby regulating cell differentiation and proliferation, and is closely related to survival, proliferation, migration, metastasis and angiogenesis of tumor cells. Statistically, K-Ras is present in 11-16% of lung adenocarcinoma casesG12CSome of the mutations, pancreatic cancer, colorectal cancer, ovarian cancer and cholangiocarcinoma, are caused by the-Rras mutation. However, since thirty years have passed since the first discovery of K-Ras oncogenes, targeting drugs for EGFR, BCL and other common protooncogenes have been carried out for several generations, and targeting drugs for K-Ras have not been successfully developed. Historically, targeted drugs against K-Ras pathway mutant tumors have focused primarily on farnesyl transferase inhibitors and Raf-MEK pathway inhibitors, but with little success. In recent years, the development of inhibitors against specific gene mutations of K-Ras has been a hot spot, and some inhibitors are gradually moving from preclinical hatch to clinical research, such as K-RasG12CInhibitors AMG510, MRTX1257 and the like, and shows certain curative effect in early clinical experiments. First global cost KrasG12CThe first clinical data for inhibitor AMG510 was formally published by the american clinical oncology institute held 6 months 2019, in which the installed drug AMG510 was shown to prevent tumor growth in most non-small cell lung and colorectal cancer patients with K-Ras mutations. Therefore, the discovery and search of a targeted drug against a specific mutated K-Ras gene with high specificity and excellent drug availability has become a great hotspot in the industry.
Disclosure of Invention
One of the technical problems to be solved by the invention is to provide a novel K-RasG12CThe inhibitor is used for preparing a medicament for treating tumors.
The scheme for solving the technical problems is as follows:
in a first aspect of the invention, a condensed cyanopyridine compound shown in general formula I-1 or I-2, or a pharmaceutically acceptable salt thereof, or an enantiomer, a diastereomer, a tautomer, a torsional isomer, a solvate, a polymorph or a prodrug thereof is provided,
Figure BDA0002733791230000021
in the formula:
r1 is independently selected from hydrogen, halogen, cyano, nitro, C1-C6Alkyl radical, C1-C6alkyl-SO2-、C1-C6alkyl-SO-, or C1-C6A haloalkyl group; r2 and R3 are independently selected from hydrogen, halogen, cyano, nitro and C1-C6Alkyl radical, C1-C6alkyl-SO2-、C1-C6alkyl-SO-, N (R)2a)(R2b)-(CH2) x-; wherein R is2aAnd R2bEach independently selected from hydrogen or C1-C6Alkyl, x is selected from any integer from 0 to 5 (i.e., 0, 1,2,3,4, or 5);
r4 is independently selected from substituted or unsubstituted C1-C12 alkyl, substituted or unsubstituted 3-12 membered cycloalkyl or heterocycloalkyl;
r5 is independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 haloalkyl, alkoxy, haloalkoxy, oxo, etc., m is independently selected from an integer of 0-6;
m is independently selected from NR6, O, S, etc.; r6 is selected from H or C1-C6 alkyl, or R6 may form a ring system with R4;
ra, Rb, Rc, Rd, Re, Rf, Rg and Rh are respectively and independently selected from hydrogen, C1-C6 alkyl, alkoxy, haloalkyl, carboxyl and the like, or Ra, Rb, Rc, Rd, Re, Rf, Rg and Rh form a 3-8-membered saturated or partially unsaturated ring system between every two of them; or Rg and R6 form a 3-8 membered saturated or partially unsaturated ring system;
ar is independently selected from a 5-12 membered aromatic ring or aromatic condensed ring, a 5-12 membered aromatic heterocycle or aromatic condensed heterocycle; and the Ar ring may be substituted with one or more of the following groups: hydrogen, halogen, C1-C6 alkyl, alkoxy, 3-8 membered cycloalkyl or heterocycloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, substituted or unsubstituted amino, amido, sulfonamido, and the like;
one or more hydrogen atoms on any of the above groups may be substituted with a substituent selected from the group consisting of: including but not limited to deuterium, halogen, hydroxy, amino or cyclic amino, cyano, nitro, sulfone or sulfoxide, C1-C8 alkyl, 3-8 membered cycloalkyl or heterocycloalkyl, C1-C8 alkoxy, C1-C8 alkylamino, alkenyl, alkynyl, acyl or sulfonyl, urea or sulfonylurea, 5-to 8-membered aryl or heteroaryl; wherein said heteroaryl group contains 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, the heterocycloalkyl group containing 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, said ring system including spiro, bridged, fused, etc. saturated or partially unsaturated ring systems.
In another preferred embodiment, the moiety-M-R4 is
Figure BDA0002733791230000022
Wherein ring C is a substituted or unsubstituted 3-8 membered cycloalkyl or heterocycloalkyl group, n is 1,2,3,4 or 5, and said substitution means that one or more hydrogen atoms on the ring may be substituted with a substituent selected from the group consisting of: deuterium, halogen, hydroxyl, amino or cyclic amino, cyano, nitro, sulfone or sulfoxide, C1-C8 alkyl, 3-8 membered cycloalkyl or heterocycloalkyl, C1-C8 alkoxy, C1-C8 alkylamino, alkenyl, alkynyl, acyl or sulfonyl, urea or sulfonylurea, 5-to 8-membered aryl or heteroaryl.
In another preferred embodiment, said compound of formula I-1 or said compound of formula I-2 has the structure shown in formula II-1 or II-2:
Figure BDA0002733791230000031
in the formula (I), the compound is shown in the specification,
ring C is a substituted or unsubstituted 3-8 membered cycloalkyl or heterocycloalkyl, which substitution means that one or more hydrogen atoms on the ring may be substituted with a substituent selected from the group consisting of: deuterium, halogen, hydroxyl, amino or cyclic amino, cyano, nitro, sulfonyl or sulfoxide, C1-C8 alkyl, 3-8 membered cycloalkyl or heterocycloalkyl, C1-C8 alkoxy, C1-C8 alkylamino, alkenyl, alkynyl, acyl or sulfonyl, urea or sulfonylurea, 5-to 8-membered aryl or heteroaryl;
r1, R2, R3, R5, Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh, Ar and m are as defined above.
In another preferred embodiment, ring C is a substituted or unsubstituted 3-8 membered cycloalkyl or heterocycloalkyl, said substituents being selected from deuterium, halogen, hydroxy, amino, cyano, amido, sulfonamido, monoalkylamino, dialkylamino, alkoxy, C1-C6Alkyl, 3-8 membered cycloalkyl or heterocycloalkyl.
In another preferred embodiment, Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh are each independently selected from: hydrogen, C1-C6 alkyl, carboxy, wherein one or more hydrogen atoms of the C1-C6 alkyl may be substituted by a substituent selected from the group consisting of: deuterium, halogen, hydroxyl, amino or cyclic amino, cyano, nitro, sulfone or sulfoxide, C1-C8 alkyl, 3-8 membered cycloalkyl or heterocycloalkyl, C1-C8 alkoxy, C1-C8 alkylamino, alkenyl, alkynyl, acyl or sulfonyl, urea or sulfonylurea, 5-to 8-membered aryl or heteroaryl.
In another preferred embodiment, Ar is independently selected from substituted or unsubstituted phenyl and pyridyl, or substituted or unsubstituted naphthyl, naphthyridinyl, indazolyl, benzimidazolyl, benzothiazolyl; the substitution means substitution with one or more substituents selected from the group consisting of: hydrogen, halogen, C1-C4 alkyl, hydroxyl, amino, cyano, C1-C4 alkoxy, C1-C4 haloalkyl, C1-C4 haloalkoxy.
In another preferred embodiment, Ar is independently selected from
Figure BDA0002733791230000032
Wherein Rp is selected from: halogen, C1-C4 alkyl, hydroxyl, amino, cyano, C1-C4 alkoxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, and q is 1,2,3,4 or 5.
In another preferred embodiment, R4 is independently selected from the group consisting of substituted or unsubstituted C4-C12 alkyl or cycloalkyl or heterocycloalkyl, 3-8 membered cycloalkyl or heterocycloalkyl substituted alkylene; preferably C1-C6 alkoxyalkyl, monoalkylamino, dialkylamino, 5-6 membered heterocycloalkyl substituted alkyl, wherein one or more hydrogen atoms of said alkoxy, alkyl, heterocycloalkyl may be substituted by a substituent selected from the group consisting of: deuterium, halogen, C1-C8 alkyl; more preferably CH3OCH2-、CH3CH2OCH2-、CH3NHCH2-、CH3CH2NHCH2-、(CH3)2NCH2-、(CH3CH2)2NCH2-、
Figure BDA0002733791230000041
Wherein p is 1 or 2.
In another preferred embodiment, R1, R2, R3 are each independently preferably selected from hydrogen, fluoro, methyl, cyano.
In another preferred embodiment, Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh are each independently selected from hydrogen, fluoro, methyl, hydroxymethyl, cyanomethylene.
In another preferred embodiment, R5 is independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy.
In another preferred embodiment, m is 0, 1 or 2.
In another preferred embodiment, the compound of formula (1), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph or prodrug thereof, wherein,
r1, R2, R3 are each independently preferably selected from hydrogen, fluoro, methyl, cyano, etc.;
ra, Rb, Rc, Rd, Re, Rf, Rg, Rh are each independently preferably selected from hydrogen, fluorine, methyl, hydroxymethyl, cyanomethyl;
m is independently preferably selected from NH or O, etc.;
r4 is independently preferably selected from C4-C12 alkyl or cycloalkyl or heterocycloalkyl, 3-to 8-membered cycloalkyl or heterocycloalkyl substituted alkylene, and the like;
r5 is independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, and the like; m is preferably selected from 0, 1, 2;
ar is independently preferably a monocyclic aromatic group such as a substituted or unsubstituted phenyl group or pyridyl group, or a substituted or unsubstituted bicyclic aromatic group such as a naphthyl group, a naphthyridinyl group, an indazolyl group, a benzimidazolyl group, or a benzothiazolyl group; said one or more substituents are preferably selected from the group consisting of: hydrogen, halogen, C1-C4 alkyl, hydroxy, amino, cyano, C1-C4 alkoxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, and the like.
In another preferred embodiment, R1, R2, R3, R4, R5, Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh, Ar, M and M are the groups corresponding to the specific compounds of the examples.
In another preferred embodiment, the compound of formula I-1 or formula I-2 has the structure:
Figure BDA0002733791230000051
in another preferred embodiment, the compound of formula I-1 or I-2 is selected from the compounds shown in the examples.
In a second aspect of the invention, there is provided a process for the preparation of a compound of formula I-1 or I-2, said process comprising steps a-j:
a) converting the compound of the general formula (A) and a cyanoacetate compound into a compound of a general formula (B) through various functional group conversion reactions;
b) reacting the compound of the general formula (B) with a piperazine derivative to produce a compound of the general formula (C);
c) carrying out substitution reaction on the general formula compound (C) and a halide or carrying out Mitsunobu reaction on an alcohol compound to obtain a general formula compound D;
d) removing the protecting group of the compound (D) with the general formula, and reacting with acrylic acid/acryloyl chloride/chloropropionyl chloride and the like to generate a compound (Ia) with the general formula;
e) converting the compound of the general formula (E) and a cyanoacetate compound into a compound of the general formula (F) through various functional group conversion reactions;
f) reacting the compound (F) with a piperazine derivative to produce a compound (G);
g) converting compound G of formula (la) with an arylboronic acid or arylboronic ester or arylmetal reagent by a transition metal catalysed coupling reaction to compound (C);
h) converting the compound (C) of the general formula into a compound (H) of the general formula under the condition of a chlorinated reagent;
i) carrying out substitution or catalytic coupling reaction on the general formula compound (H) and different amine or alcohol compounds to obtain a general formula compound (I);
j) removing a protecting group of the compound (I) with the general formula, and reacting with acrylic acid/acryloyl chloride/chloropropionyl chloride and the like to generate a compound (Ib) with the general formula;
Figure BDA0002733791230000061
in the formula (I), the compound is shown in the specification,
Figure BDA0002733791230000062
is composed of
Figure BDA0002733791230000063
LG is a leaving group such as I, -OTs;
PG is selected from: benzyloxycarbonyl, t-butoxycarbonyl, phthaloyl, benzyl, p-toluenesulfonyl, trifluoroacetyl, fluorenylmethyloxycarbonyl, allyloxycarbonyl, o- (p) -nitrobenzenesulfonyl, trityl.
The definition of each group is as described above.
Preferably, said steps a), b), c), d), e), f), g), h), i), j) are each carried out in a solvent, and said solvent is selected from the group consisting of: water, methanol, ethanol, isopropanol, butanol, ethylene glycol methyl ether, N-methyl pyrrolidone, dimethyl sulfoxide, tetrahydrofuran, toluene, dichloromethane, 1, 2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, or a combination thereof.
Preferably, the transition metal catalyst is selected from the group consisting of: tris (dibenzylideneacetone) dipalladium (Pd)2(dba)3) Tetrakis (triphenylphosphine) palladium (Pd (PPh)3)4) Palladium acetate, palladium chloride, dichlorobis (triphenylphosphine) palladium, palladium trifluoroacetate, triphenylphosphine palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride, bis (tri-o-phenylphosphino) palladium dichloride, 1, 2-bis (diphenylphosphino) ethane palladium dichloride, or a combination thereof; the catalyst ligand is selected from the group consisting of: tri-tert-butylphosphine, tri-tert-butylphosphine tetrafluoroborate, tri-n-butylphosphine, triphenylphosphine, tri-p-benzylphosphine, tricyclohexylphosphine, tri-o-phenylphosphine, or a combination thereof.
Preferably, the condensing agent is selected from the group consisting of: dicyclohexylcarbodiimide DCC, diisopropylcarbodiimide DIC, N, N ' -carbonyldiimidazole CDI, 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride EDCI, N-hydroxy-7-azobenzotriazol HOAt, 1-hydroxybenzotriazol HOBt, benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate BOP, benzotriazol-1-yl-oxytriazolylphosphonium hexafluorophosphate PyBOP, 2- (7-azabenzotriazolyl) -N, N, N ', N ' -tetramethylurea hexafluorophosphate HATU, O-benzotriazol-N, N, N ', N ' -tetramethylurea tetrafluoroborate TBTU, etc., or a combination thereof.
Preferably, the inorganic base is selected from the group consisting of: sodium hydride, potassium hydroxide, sodium acetate, potassium tert-butoxide, sodium tert-butoxide, potassium fluoride, cesium fluoride, potassium phosphate, potassium carbonate, potassium bicarbonate, sodium carbonate, sodium bicarbonate, or combinations thereof; the organic base is selected from the group consisting of: pyridine, triethylamine, N, N-diisopropylethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), lithium hexamethyldisilazide, sodium hexamethyldisilazide, lutidine, or a combination thereof.
Preferably, the acid is selected from the group consisting of: hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, toluenesulfonic acid, trifluoroacetic acid, formic acid, acetic acid, trifluoromethanesulfonic acid, or combinations thereof.
In another preferred embodiment, the compound is selected from the following structures:
Figure BDA0002733791230000071
in a third aspect of the invention, there is provided a pharmaceutical composition comprising (I) a fused cyanopyridine compound of formula I-1 or I-2, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph or prodrug thereof, and (ii) a pharmaceutically acceptable carrier.
In another preferred embodiment, the pharmaceutical composition further comprises a drug selected from the group consisting of:
PD-1 inhibitors (e.g., nivolumab, pembrolizumab, pidilizumab, etc.), PD-L1 inhibitors (e.g., durvalumab, atezolizumab, avelumab, etc.), CD20 antibodies (e.g., rituximab, obinutuzumab), ALK inhibitors (e.g., Ceritinib, ocatinib), PI3K inhibitors (e.g., Idelalisib, Duvelisib, etc.), BTK inhibitors (e.g., Ibrutinib), EGFR inhibitors (e.g., Afatinib, Gefitinib, etc.), or combinations thereof.
In another preferred embodiment, there is provided a method for preparing a pharmaceutical composition, comprising the steps of: mixing a pharmaceutically acceptable carrier with a compound of the first aspect of the invention, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph or prodrug thereof, to form a pharmaceutical composition.
In a fourth aspect of the present invention, there is provided a use of the compound of the first aspect or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph or prodrug thereof, for the manufacture of a medicament for the treatment of a disease associated with Ras mutein activity or expression level, in particular, a tumor. The tumor is independently selected from non-small cell lung cancer, lung adenocarcinoma, lung squamous carcinoma, breast cancer, prostatic cancer, liver cancer, skin cancer, gastric cancer, intestinal cancer, cholangiocarcinoma, brain cancer, leukemia, lymph cancer, fibroma, sarcoma, basal cell carcinoma, glioma, renal cancer, melanoma, bone cancer, thyroid cancer, nasopharyngeal cancer, pancreatic cancer, etc.
The invention relates to a compound with the structural characteristics of general formula (I-1 or I-2), which can inhibit various tumor cells, in particular can kill K-Ras with high efficiencyG12CThe tumor related to the abnormal signal path of the mutant protein is a treatment medicine with a brand new action mechanism.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. The space is not described herein in a repeated fashion.
Detailed Description
Through long-term and intensive research, the inventor prepares a compound with a novel structure shown in formula I and finds that the compound has better inhibition effect on K-RasG12CProtein inhibitory activity and said compounds are at very low concentrations (as low as less than 100nM), i.e., against K-RasG12CSpecific inhibition of protein production and of K-RasG12CThe relevant cell proliferation inhibitory activity is quite excellent and the compounds are at very low concentrations (as low as less than 10nM), i.e., against K-RasG12CThe positive tumor cells have strong killing effect, so that the composition can be used for treating K-RasG12CRelated diseases caused by mutation or abnormal expression amount such as tumor. Based on the above findings, the inventors have completed the present invention.
Term(s) for
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the claimed subject matter belongs. All patents, patent applications, and publications cited herein are incorporated by reference in their entirety unless otherwise indicated.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the subject matter claimed. In this application, the use of the singular also includes the plural unless specifically stated otherwise. It must be noted that, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. It should also be noted that the use of "or", "or" means "and/or" unless stated otherwise. Furthermore, the term "comprising" as well as other forms, such as "includes," "including," and "containing," are not limiting.
Definitions for the terms of the standardization sector can be found in the literature references including Carey and Sundberg "ADVANCED ORGANIC CHEMISTRY 4TH ED." Vols.A (2000) and B (2001), Plenum Press, New York. Unless otherwise indicated, conventional methods within the skill of the art are employed, such as mass spectrometry, NMR, IR and UV/VIS spectroscopy, and pharmacological methods. Unless a specific definition is set forth, the terms used herein in the pertinent description of analytical chemistry, organic synthetic chemistry, and pharmaceutical chemistry are known in the art. Standard techniques can be used in chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and delivery, and treatment of patients. For example, the reaction and purification can be carried out using the instructions of the kit from the manufacturer, or according to the methods known in the art or the instructions of the present invention. The techniques and methods described above can generally be practiced according to conventional methods well known in the art, as described in various general and more specific documents referred to and discussed in this specification. In the present specification, groups and substituents thereof may be selected by one skilled in the art to provide stable moieties and compounds.
When a substituent is described by a general formula written from left to right, the substituent also includes chemically equivalent substituents obtained when the formula is written from right to left. For example, -CH 2O-is equivalent to-OCH 2-.
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including but not limited to patents, patent applications, articles, books, operating manuals, and treatises, are hereby incorporated by reference in their entirety.
Certain chemical groups defined herein are preceded by a shorthand notation to indicate the total number of carbon atoms present in the group. For example, C1-6 alkyl refers to an alkyl group as defined below having a total of 1 to 6 carbon atoms. The total number of carbon atoms in the shorthand notation excludes carbons that may be present in a substituent of the group.
In addition to the foregoing, the following terms, when used in the specification and claims of this application, have the meanings indicated below, unless otherwise specifically indicated.
In the present application, the term "halogen" means fluorine, chlorine, bromine or iodine; "hydroxy" means an-OH group; "hydroxyalkyl" refers to an alkyl group as defined below substituted with a hydroxyl (-OH) group; "carbonyl" refers to a-C (═ O) -group; "nitro" means-NO2(ii) a "cyano" means-CN; "amino" means-NH2(ii) a "substituted amino" refers to an amino group substituted with one or two alkyl, alkylcarbonyl, aralkyl, heteroaralkyl groups as defined below, e.g., monoalkylamino (e.g., CH)3-NH-、CH3CH2-NH-、CH3CH2CH2-NH-), dialkylamino (e.g. (CH3)2-N-、(CH3CH2)2-N-、(CH3CH2CH2)2-N-), alkylamido, aralkylamino, heteroaralkylamino; "carboxyl" means-COOH.
In the present application, the term "alkyl", as a group or as part of another group (e.g. as used in groups such as halogen-substituted alkyl), means a straight or branched hydrocarbon chain group consisting only of carbon and hydrogen atoms, containing no unsaturated bonds, having, for example, from 1 to 12 (preferably from 1 to 8, more preferably from 1 to 6) carbon atoms and being attached to the rest of the molecule by single bonds. Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2-dimethylpropyl, n-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl, decyl, and the like.
In the present application, the term "alkenyl" as a group or part of another group means a straight or branched hydrocarbon chain group consisting of only carbon atoms and hydrogen atoms, containing at least one double bond, having, for example, 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms, and being connected to the rest of the molecule by a single bond, such as, but not limited to, vinyl, propenyl, allyl, but-1-enyl, but-2-enyl, pent-1, 4-dienyl, and the like.
In the present application, the term "alkynyl" as a group or part of another group means a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one triple bond and optionally one or more double bonds, having for example 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms and being connected to the rest of the molecule by single bonds, such as but not limited to ethynyl, prop-1-ynyl, but-1-ynyl, pent-1-en-4-ynyl and the like.
In the present application, the term "cycloalkyl" as a group or part of another group means a stable non-aromatic monocyclic or polycyclic hydrocarbon group consisting of only carbon atoms and hydrogen atoms, which may include fused, bridged or spiro ring systems, having 3 to 15 carbon atoms, preferably having 3 to 10 carbon atoms, more preferably having 3 to 8 carbon atoms, and which is saturated or unsaturated and may be attached to the rest of the molecule by a single bond via any suitable carbon atom. Unless otherwise specifically indicated in the specification, carbon atoms in cycloalkyl groups may be optionally oxidized. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, 1H-indenyl, 2, 3-indanyl, 1,2,3, 4-tetrahydro-naphthyl, 5,6,7, 8-tetrahydro-naphthyl, 8, 9-dihydro-7H-benzocyclohepten-6-yl, 6,7,8, 9-tetrahydro-5H-benzocycloheptenyl, 5,6,7,8,9, 10-hexahydro-benzocyclooctenyl, fluorenyl, bicyclo [2.2.1] heptyl, 7-dimethyl-bicyclo [2.2.1] heptyl, bicyclo [2.2.1] heptenyl, bicyclo [2.2.2] octyl, bicyclo [3.1.1] heptyl, bicyclo [3.2.1] octyl, bicyclo [2.2.2] octenyl, Bicyclo [3.2.1] octenyl, adamantyl, octahydro-4, 7-methylene-1H-indenyl, octahydro-2, 5-methylene-pentalenyl and the like.
In this application, the term "heterocyclyl" as a group or part of another group means a stable 3-to 20-membered non-aromatic cyclic group consisting of 2 to 14 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, phosphorus, oxygen, and sulfur. Unless otherwise specifically indicated in the specification, a heterocyclic group may be a monocyclic, bicyclic, tricyclic or higher ring system, which may include fused ring systems, bridged ring systems or spiro ring systems; wherein the nitrogen, carbon or sulfur atom in the heterocyclic group may be optionally oxidized; the nitrogen atoms may optionally be quaternized; and the heterocyclic group may be partially or fully saturated. The heterocyclic group may be attached to the rest of the molecule via a carbon atom or a heteroatom and by a single bond. In heterocyclic groups containing fused rings, one or more of the rings may be aryl or heteroaryl as defined below, provided that the point of attachment to the rest of the molecule is a non-aromatic ring atom. For the purposes of the present invention, heterocyclyl is preferably a stable 4-to 11-membered non-aromatic monocyclic, bicyclic, bridged or spiro group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 4-to 8-membered non-aromatic monocyclic, bicyclic, bridged or spiro group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heterocyclyl groups include, but are not limited to: pyrrolidinyl, morpholinyl, piperazinyl, homopiperazinyl, piperidinyl, thiomorpholinyl, 2, 7-diaza-spiro [3.5] nonan-7-yl, 2-oxa-6-aza-spiro [3.3] heptan-6-yl, 2, 5-diaza-bicyclo [2.2.1] heptan-2-yl, azetidinyl, pyranyl, tetrahydropyranyl, thiopyranyl, tetrahydrofuranyl, oxazinyl, dioxolanyl, tetrahydroisoquinolinyl, decahydroisoquinolinyl, imidazolinyl, imidazolidinyl, quinolizinyl, thiazolidinyl, isothiazolidinyl, isoxazolidinyl, indolinyl, octahydroindolyl, octahydroisoindolyl, pyrrolidinyl, pyrazolidinyl, phthalimidyl, and the like.
In this application, the term "aryl" as a group or as part of another group means a conjugated hydrocarbon ring system group having 6 to 18 carbon atoms, preferably having 6 to 10 carbon atoms. For the purposes of the present invention, an aryl group may be a monocyclic, bicyclic, tricyclic or higher polycyclic ring system and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the aryl group is attached to the remainder of the molecule by a single bond via an atom on the aromatic ring. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, phenanthrenyl, fluorenyl, 2, 3-dihydro-1H-isoindolyl, 2-benzoxazolinone, 2H-1, 4-benzoxazin-3 (4H) -one-7-yl, and the like.
In the present application, the term "arylalkyl" refers to an alkyl group as defined above substituted with an aryl group as defined above.
In this application, the term "heteroaryl" as a group or part of another group means a 5-to 16-membered conjugated ring system group having 1 to 15 carbon atoms (preferably having 1 to 10 carbon atoms) and 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur in the ring. Unless otherwise specifically indicated in the specification, a heteroaryl group may be a monocyclic, bicyclic, tricyclic or higher ring system, and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the heteroaryl group is attached to the rest of the molecule by a single bond via an atom on the aromatic ring. The nitrogen, carbon or sulfur atoms in the heteroaryl group may be optionally oxidized; the nitrogen atoms may optionally be quaternized. For the purposes of the present invention, heteroaryl is preferably a stable 5-to 12-membered aromatic group containing 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 5-to 10-membered aromatic group containing 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur or a 5-to 6-membered aromatic group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heteroaryl groups include, but are not limited to, thienyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, benzopyrazolyl, indolyl, furyl, pyrrolyl, triazolyl, tetrazolyl, triazinyl, indolizinyl, isoindolyl, indazolyl, isoindolyl, purinyl, quinolyl, isoquinolyl, diazonaphthyl, naphthyridinyl, quinoxalinyl, pteridinyl, carbazolyl, carbolinyl, phenanthridinyl, phenanthrolinyl, acridinyl, phenazinyl, isothiazolyl, benzothiazolyl, benzothienyl, oxazolyl, cinnolinyl, quinazolinyl, thiophenyl, indolizinyl, orthophenanthrolidinyl, isoxazolyl, phenoxazinyl, phenothiazinyl, 4,5,6, 7-tetrahydrobenzo [ b ] thienyl, naphthopyridyl, pyridinyl, and the like, [1,2,4] triazolo [4,3-b ] pyridazine, [1,2,4] triazolo [4,3-a ] pyrazine, [1,2,4] triazolo [4,3-c ] pyrimidine, [1,2,4] triazolo [4,3-a ] pyridine, imidazo [1,2-b ] pyridazine, imidazo [1,2-a ] pyrazine and the like.
In the present application, the term "heteroarylalkyl" refers to an alkyl group as defined above substituted with a heteroaryl group as defined above.
In the present application, "saturated or partially unsaturated ring" means a cycloalkyl group or a heterocyclic group as defined above.
In this application, "optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "optionally substituted aryl" means that the aryl group is substituted or unsubstituted, and the description includes both substituted and unsubstituted aryl groups.
The terms "moiety," "structural moiety," "chemical moiety," "group," "chemical group" as used herein refer to a specific fragment or functional group in a molecule. Chemical moieties are generally considered to be chemical entities that are embedded in or attached to a molecule.
"stereoisomers" refers to compounds that consist of the same atoms, are bonded by the same bonds, but have different three-dimensional structures. The present invention is intended to cover various stereoisomers and mixtures thereof.
When the compounds of the present invention contain olefinic double bonds, the compounds of the present invention are intended to include both E-and Z-geometric isomers unless otherwise specified.
"tautomer" refers to an isomer formed by the transfer of a proton from one atom of a molecule to another atom of the same molecule. All tautomeric forms of the compounds of the invention are also intended to be included within the scope of the invention.
In the present invention, each of the above-mentioned alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclic, aryl, heteroaryl, arylalkyl, heteroarylalkyl, etc. may be substituted or unsubstituted, and the "substitution" means substitution with one or more groups selected from the group consisting of: deuterium, halogen, hydroxyl, amino or cyclic amino, cyano, nitro, sulphonyl or sulphoxy, C1-C8 alkyl, 3-8 membered cycloalkyl or heterocycloalkyl, C1-C8 alkoxy, C1-C8 alkylamino, alkenyl, alkynyl, acyl or sulphonyl, urea or sulphonyl urea, 5-to 8-membered aryl or heteroaryl, preferably selected from: deuterium, halogen, C1-C8 alkyl, 3-8 membered cycloalkyl or heterocycloalkyl.
Active ingredient
As used herein, the terms "compound of the invention" or "active ingredient of the invention" are used interchangeably to refer to a compound of formula I-1 or I-2, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph or prodrug thereof.
In the present invention, the compound of formula I-1 or I-2 has the following structure:
Figure BDA0002733791230000121
r1, R2, R3, R4, R5, Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh, Ar, M and M are as defined above.
Preferably, R4 is independently selected from substituted or unsubstituted C4-C12 alkyl or cycloalkyl or heterocycloalkyl, 3-8 membered cycloalkyl or heterocycloalkyl substituted alkylene; preferably C1-C6 alkoxyalkyl, monoalkylamino, dialkylamino, 5-6 membered heterocycloalkyl substituted alkyl, wherein one or more hydrogen atoms of said alkoxy, alkyl, heterocycloalkyl may be substituted by a substituent selected from the group consisting of: deuterium, halogen, C1-C8 alkyl; more preferably CH3OCH2-、CH3CH2OCH2-、CH3NHCH2-、CH3CH2NHCH2-、(CH3)2NCH2-、(CH3CH2)2NCH2-、
Figure BDA0002733791230000122
Wherein p is 1 or 2.
Preferably, in formula I-1 or I-2 the moiety-M-R4 is
Figure BDA0002733791230000123
Wherein ring C is a substituted or unsubstituted 3-8 membered cycloalkyl or heterocycloalkyl group, n is 1,2,3,4 or 5, and said substitution means that one or more hydrogen atoms on the ring may be substituted with a substituent selected from the group consisting of: deuterium, halogen, hydroxyl, amino or cyclic amino, cyano, nitro, sulfone or sulfoxide, C1-C8 alkyl, 3-8 membered cycloalkyl or heterocycloalkyl, C1-C8 alkoxy, C1-C8 alkylamino, alkenyl, alkynyl, acyl or sulfonyl, urea or sulfonylurea, 5-to 8-membered aryl or heteroaryl.
Preferably, said compound of formula I-1 or said compound of formula I-2 has the structure shown in formula II-1 or II-2:
Figure BDA0002733791230000131
in the formula (I), the compound is shown in the specification,
ring C is a substituted or unsubstituted 3-8 membered cycloalkyl or heterocycloalkyl, which substitution means that one or more hydrogen atoms on the ring may be substituted with a substituent selected from the group consisting of: deuterium, halogen, hydroxyl, amino or cyclic amino, cyano, nitro, sulfonyl or sulfoxide, C1-C8 alkyl, 3-8 membered cycloalkyl or heterocycloalkyl, C1-C8 alkoxy, C1-C8 alkylamino, alkenyl, alkynyl, acyl or sulfonyl, urea or sulfonylurea, 5-to 8-membered aryl or heteroaryl; preferably, ring C is a substituted or unsubstituted 3-8 membered cycloalkyl or heterocycloalkyl group, said substituents being selected from deuterium, halogen, hydroxy, amino, cyano, amido, sulfonamido, monoalkylamino, dialkylamino, alkoxy, C1-C6Alkyl, 3-8 membered cycloalkyl or heterocycloalkyl,
r1, R2, R3, R5, Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh, Ar and m are as defined above.
Preferably, in the above formulae, Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh are each independently selected from: hydrogen, C1-C6 alkyl, carboxy, wherein one or more hydrogen atoms of the C1-C6 alkyl may be substituted by a substituent selected from the group consisting of: deuterium, halogen, hydroxyl, amino or cyclic amino, cyano, nitro, sulfone or sulfoxide, C1-C8 alkyl, 3-8 membered cycloalkyl or heterocycloalkyl, C1-C8 alkoxy, C1-C8 alkylamino, alkenyl, alkynyl, acyl or sulfonyl, urea or sulfonylurea, 5-to 8-membered aryl or heteroaryl.
Preferably, in each of the above formulae, Ar is independently selected from substituted or unsubstituted phenyl and pyridyl, or substituted or unsubstituted naphthyl, naphthyridinyl, indazolyl, benzimidazolyl, benzothiazolyl; the substitution means substitution with one or more substituents selected from the group consisting of: hydrogen, halogen, C1-C4 alkyl, hydroxy, amino, cyano, C1-C4 alkoxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, more preferably, Ar is independently selected from the group consisting of
Figure BDA0002733791230000132
Wherein Rp is selected from: halogen, C1-C4 alkyl, hydroxyl, amino, cyano, C1-C4 alkoxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, and q is 1,2,3,4 or 5.
The compounds of the present invention or pharmaceutically acceptable salts thereof may contain one or more chiral carbon atoms and may therefore give rise to enantiomers, diastereomers and other stereoisomeric forms. Each chiral carbon atom may be defined as (R) -or (S) -, based on stereochemistry. The present invention is intended to include all possible isomers, as well as racemates and optically pure forms thereof. The compounds of the invention may be prepared by selecting as starting materials or intermediates racemates, diastereomers or enantiomers. Optically active isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, e.g., crystallization and chiral chromatography.
Conventional techniques for the preparation/separation of individual isomers include Chiral synthesis from suitable optically pure precursors, or resolution of racemates (or racemates of salts or derivatives) using, for example, Chiral high performance liquid chromatography, as described, for example, in Gerald Gubitz and Martin G.Schmid (Eds.), Chiral Separations, Methods and Protocols, Methods in Molecular Biology, Vol.243, 2004; m. Stalcup, Chiral Separations, Annu. Rev. anal. chem.3:341-63, 2010; fumiss et al (eds.), VOGEL' S ENCYCOPEDIA OF PRACTICAL ORGANIC CHEMISTRY 5. TH ED., Longman Scientific and Technical Ltd., Essex,1991, 809-816; heller, acc, chem, res, 1990,23,128.
In the present application, the term "pharmaceutically acceptable salts" includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
"pharmaceutically acceptable acid addition salts" refers to salts with inorganic or organic acids which retain the biological effectiveness of the free base without other side effects. Inorganic acid salts include, but are not limited to, hydrochloride, hydrobromide, sulfate, nitrate, phosphate, and the like; organic acid salts include, but are not limited to, formates, acetates, 2-dichloroacetates, trifluoroacetates, propionates, caproates, caprylates, caprates, undecylenates, glycolates, gluconates, lactates, sebacates, adipates, glutarates, malonates, oxalates, maleates, succinates, fumarates, tartrates, citrates, palmitates, stearates, oleates, cinnamates, laurates, malates, glutamates, pyroglutamates, aspartates, benzoates, methanesulfonates, benzenesulfonates, p-toluenesulfonates, alginates, ascorbates, salicylates, 4-aminosalicylates, napadisylates, and the like. These salts can be prepared by methods known in the art.
"pharmaceutically acceptable base addition salts" refers to salts with inorganic or organic bases which maintain the biological effectiveness of the free acid without other side effects. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Preferred inorganic salts are ammonium, sodium, potassium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, the following: primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, triethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purine, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like. Preferred organic bases include isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine. These salts can be prepared by methods known in the art.
"polymorph" refers to different solid crystalline phases of certain compounds of the present invention in the solid state due to the presence of two or more different molecular arrangements. Certain compounds of the present invention may exist in more than one crystalline form and the present invention is intended to include the various crystalline forms and mixtures thereof.
Typically, crystallization will result in solvates of the compounds of the invention. The term "solvate" as used herein refers to an aggregate comprising one or more molecules of the compound of the present invention and one or more solvent molecules. The solvent may be water, in which case the solvate is a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present invention may exist as hydrates, including monohydrates, dihydrate, hemihydrate, sesquihydrates, trihydrate, tetrahydrate, and the like, as well as the corresponding solvated forms. The compounds of the invention may form true solvates, but in some cases it is also possible to retain only adventitious water or a mixture of water plus a portion of adventitious solvent. The compounds of the invention may be reacted in a solvent or precipitated or crystallized from a solvent. Solvates of the compounds of the invention are also included within the scope of the invention.
The invention also includes prodrugs of the above compounds. In the present application, the term "prodrug" denotes a compound that can be converted under physiological conditions or by solvolysis to the biologically active compound of the invention. Thus, the term "prodrug" refers to a pharmaceutically acceptable metabolic precursor of a compound of the invention. Prodrugs may not be active when administered to a subject in need thereof, but are converted in vivo to the active compounds of the invention. Prodrugs are generally rapidly converted in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. Prodrug compounds generally provide solubility, histocompatibility, or sustained release advantages in mammalian organisms. Prodrugs include known amino protecting groups and carboxyl protecting groups. Specific methods for preparing prodrugs can be found in Saulnier, M.G., et al, bioorg.Med.chem.Lett.1994,4, 1985-1990; greenwald, r.b., et al, j.med.chem.2000,43,475.
In the present application, a "pharmaceutical composition" refers to a formulation of a compound of the present invention with a vehicle generally accepted in the art for delivery of biologically active compounds to a mammal (e.g., a human). The medium includes a pharmaceutically acceptable carrier. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of active ingredients and exert biological activity.
The term "pharmaceutically acceptable" as used herein refers to a substance (e.g., carrier or diluent) that does not affect the biological activity or properties of the compounds of the present invention and is relatively non-toxic, i.e., the substance can be administered to an individual without causing an adverse biological response or interacting in an adverse manner with any of the components contained in the composition.
As used herein, a "pharmaceutically acceptable carrier" includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersing agent, suspending agent, stabilizing agent, isotonic agent, solvent, or emulsifying agent that is approved by the relevant governmental regulatory agency for human or livestock use.
The "tumor" and "diseases related to abnormal cell proliferation" include, but are not limited to, leukemia, gastrointestinal stromal tumor, histiocytic lymphoma, non-small cell lung cancer, pancreatic cancer, squamous cell lung cancer, lung adenocarcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, cervical cancer, ovarian cancer, intestinal cancer, nasopharyngeal cancer, brain cancer, bone cancer, esophageal cancer, melanoma, renal cancer, oral cancer, and the like.
The terms "preventing," "prevention," and "prevention" as used herein include reducing the likelihood of occurrence or worsening of a disease or disorder in a patient.
As used herein, the term "treatment" and other similar synonyms include the following meanings:
(i) preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is susceptible to the disease or condition, but has not been diagnosed as having the disease or condition;
(ii) inhibiting the disease or disorder, i.e., arresting its development;
(iii) alleviating the disease or condition, i.e., causing regression of the state of the disease or condition; or
(iv) Alleviating the symptoms caused by the disease or disorder.
The terms "effective amount," "therapeutically effective amount," or "pharmaceutically effective amount" as used herein, refer to an amount of at least one agent or compound that is sufficient to alleviate one or more symptoms of the disease or disorder being treated to some extent after administration. The result may be a reduction and/or alleviation of signs, symptoms, or causes, or any other desired change in a biological system. For example, an "effective amount" for treatment is the amount of a composition comprising a compound disclosed herein that is clinically necessary to provide a significant remission effect of the condition. An effective amount suitable in any individual case can be determined using techniques such as a dose escalation assay.
The terms "administering," "administration," "administering," and the like as used herein refer to a method capable of delivering a compound or composition to a desired site for biological action. These methods include, but are not limited to, oral routes, via the duodenal route, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. Administration techniques useful for The compounds and methods described herein are well known to those skilled in The art, for example, in Goodman and Gilman, The pharmaceutical Basis of Therapeutics, current ed.; pergamon; and Remington's, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa. In preferred embodiments, the compounds and compositions discussed herein are administered orally.
The terms "drug combination", "administering other treatment", "administering other therapeutic agent" and the like as used herein refer to a drug treatment obtained by mixing or combining more than one active ingredient, including fixed and unfixed combinations of active ingredients. The term "fixed combination" refers to the simultaneous administration of at least one compound described herein and at least one co-agent to a patient in the form of a single entity or a single dosage form. The term "non-fixed combination" refers to the simultaneous administration, concomitant administration, or sequential administration at variable intervals of at least one compound described herein and at least one synergistic formulation to a patient as separate entities. These also apply to cocktail therapy, for example the administration of three or more active ingredients.
It will also be appreciated by those skilled in the art that in the processes described below, the functional groups of the intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxyl, amino, mercapto and carboxylic acid. Suitable hydroxy protecting groups include trialkylsilyl or diarylalkylsilyl groups (e.g.tert-butyldimethylsilyl, tert-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butyloxycarbonyl, benzyloxycarbonyl and the like. Suitable thiol protecting groups include-C (O) -R "(where R" is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like. Suitable carboxyl protecting groups include alkyl, aryl or aralkyl esters.
Protecting groups may be introduced and removed according to standard techniques known to those skilled in the art and as described herein. The use of protecting Groups is described in detail in Greene, T.W. and P.G.M.Wuts, Protective Groups in Organic Synthesis, (1999),4th Ed., Wiley. The protecting group may also be a polymeric resin.
The invention has the following main advantages:
(1) said combination ofObject pair K-RASG12CHas good selective inhibition effect;
(2) the compound has better pharmacodynamics and pharmacokinetic performance and lower toxic and side effects.
The invention will be further illustrated with reference to specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, generally according to conventional conditions, or according to conditions recommended by the manufacturer. Percentages and parts are by weight unless otherwise indicated.
Intermediate 1: 7-benzyl-4-chloro-2-hydroxy-5, 6,7, 8-tetrahydro-1, 7-naphthyridine-3-carbonitrile
Figure BDA0002733791230000171
The first step is as follows: 1-benzyl-3-oxopiperidine-4-carboxylic acid ethyl ester hydrochloride (40.0g,134.7mmol) is dissolved in ethanol (EtOH) (600mL) and ammonium acetate (NH)4OAc) (103.4g,1.34mol), room temperature for 3 hours, LC-MS detection showed reaction completion. 1M aqueous sodium hydroxide (NaOH) was added to adjust pH to 9, the solid was filtered off, the aqueous phase was extracted with DCM and concentrated, and combined column chromatography purification gave ethyl 5-amino-1-benzyl-1, 2,3, 6-tetrahydropyridine-4-carboxylate (29.2g, white solid). LC-MS ESI [ M + H ]]+=261.4。
The second step is that: sodium Na particles (5.3g, 0.23mol) were added in portions slowly to EtOH (200mL), stirred at room temperature until the Na particles disappeared completely, ethyl 5-amino-1-benzyl-1, 2,3, 6-tetrahydropyridine-4-carboxylate (15.0g,57.7mmol) and ethyl 2-cyanoacetate (13.0g,115.0mmol) were added, and the mixture was heated to 120 ℃ in a closed pot for four days. After cooling, filtration was carried out and the filter cake was washed twice with ethanol. The filtrate and washings were combined, spun dry, diluted with water (300mL), and stripped with Dichloromethane (DCM) (200mL) three times. The pH of the aqueous phase was adjusted to 5 with dilute hydrochloric acid (3M) and solids precipitated and filtered. The filter cake was purified by slurrying with ethanol (60mL) to give 7-benzyl-2, 4-dihydroxy-5, 6,7, 8-tetrahydro-1, 7-naphthyridine-3-carbonitrile (9.0g, as a white solid). LC-MS ESI [ M + H ]]+=282.4。
The third step: 7-benzyl-2, 4-dihydroxy-5, 6,7, 8-tetrahydro-1, 7-naphthyridine-3-carbonitrile (10.0g,35.6mmol) was dissolved in POCl3(60mL), the mixture was stirred at 55 ℃ for 6 hours. The reaction was cooled to room temperature, concentrated under reduced pressure, added with 20mL of water, and then diluted with saturated sodium bicarbonate NaHCO3The pH was adjusted to about 9 and stirred at room temperature overnight. Filtration, pH adjustment of the filtrate to 6-7 with dilute hydrochloric acid, precipitation of solids, filtration, beating with ethyl acetate, and drying to give intermediate 1(9.5g, grey solid). LC-MS ESI [ M + H ]]+=300.1。
Intermediate 2A: 2, 4-dichloro-6-fluoro-7- (2-fluoro-6-methoxyphenyl) -1, 8-naphthyridine-3-carbonitrile
Figure BDA0002733791230000172
The first step is as follows: 2, 6-dichloro-5-fluoronicotinic acid (100g, 478.5mmol) was dissolved in methanol (1.0L) and thionyl chloride (SOCl) was added dropwise at 0 deg.C2) (69mL,949.8mmol) was refluxed for 4 hours under nitrogen. The reaction was cooled to room temperature, concentrated under reduced pressure, and the residue was dissolved in DCM and NaHCO3The saturated solution was washed twice with water and once with saturated brine, dried and concentrated under reduced pressure to give methyl 2, 6-dichloro-5-fluoronicotinate (106g, yellow oil). LC-MS ESI [ M + H ]]+=224.0;1H NMR(400MHz,CDCl3):δ8.01(d,J=7.6MHz,1H),3.98(s,3H)。
The second step is that: methyl 2, 6-dichloro-5-fluoronicotinate (20g, 89.7mmol), (2-fluoro-6-methoxyphenyl) boronic acid (19.4g, 114.1mmol) and potassium phosphate (K)3PO4) (24.3g, 114.6mmol) in dioxane/water (1, 4-dioxane/H)2O) (200mL/20mL), Pd-Xphos-G3(3.75G,4.76mmol) and Ru-phos (4.44G, 9.52mmol) were added and reacted overnight at 60 ℃ under nitrogen. The reaction mixture was cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure and purified by column chromatography to give methyl 2-chloro-5-fluoro-6- (2-fluoro-6-methoxyphenyl) nicotinate (8.92g, yellow oil). LC-MS ESI [ M + H ]]+=314.3;1H NMR(400MHz,DMSO-d6):δ8.39(d,J=8.4MHz,1H),7.57-7.59(m,1H),6.99-7.08(m,2H),3.93(s,3H),3.79(s,3H)。
The third step: methyl 2-chloro-5-fluoro-6- (2-fluoro-6-methoxyphenyl) nicotinate (8.92g, 28.5mmol) was dissolved in tetrahydrofuran/water THF/H2O (90mL/45mL), lithium hydroxide monohydrate LiOH.H was added2O (3.58g,85.2mmol), and reacted at room temperature for 5 hours. The reaction solution was concentrated under reduced pressure to remove most of the organic solvent, the pH was adjusted to 3-4 with 3N HCl, and then extracted with DCM, and the organic phases were combined, washed with saturated brine, dried, and concentrated under reduced pressure to obtain 2-chloro-5-fluoro-6- (2-fluoro-6-methoxyphenyl) nicotinic acid (8.33g, yellow solid). LC-MS ESI [ M + H ]]+=300.0。
The fourth step: 2-chloro-5-fluoro-6- (2-fluoro-6-methoxyphenyl) nicotinic acid (8.33g, 27.9mmol) was dissolved in N-methylpyrrolidone NMP (25mL), p-methoxybenzylamine PMB-NH was added2(11.44g, 83.5mmol) and diisopropylethylamine DIEA (10.77g,83.5mmol), under nitrogen protection at 180 ℃ for 16 h. The reaction solution is cooled to room temperature, and saturated ammonium chloride NH is slowly poured into the reaction solution4Filtering in Cl water solution, dissolving filter cake with methanol, drying, concentrating under reduced pressure, and purifying by column chromatography to obtain 5-fluoro-6- (2-fluoro-6-methoxyphenyl-2- ((4-methoxybenzyl) amino) nicotinic acid (13.6g, tan oily substance, crude product)]+=401.1。
The fifth step: dissolve 5-fluoro-6- (2-fluoro-6-methoxyphenyl-2- ((4-methoxybenzyl) amino) nicotinic acid (9.6g, crude) in methanol MeOH (100mL) and add TMSCHN2(2.0M in hexane, 18mL, 36mmol), and reacted at room temperature for 3 hours. Concentrating the reaction solution under reduced pressure, and purifying by column chromatography to obtain 5-fluoro-6- (2-fluoro-6-methoxyphenyl-2- ((4-methoxybenzyl) amino) methyl nicotinate (5.4g, yellow oily substance)]+=415.4。
And a sixth step: methyl 5-fluoro-6- (2-fluoro-6-methoxyphenyl-2- ((4-methoxybenzyl) amino) nicotinate (5.4g,13.0mmol) was dissolved in TFA (14mL) and DCM (30mL), heated to 40 ℃ and reacted for 3 hours, the reaction solution was concentrated under reduced pressure, dissolved in 100mL ethyl acetate, and saturated Na was added2CO3The aqueous solution was washed twice, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give methyl 2-amino-5-fluoro-6- (2-fluoro-6-methoxyphenyl) nicotinate (3.7g, yellow solid). LC-MS ESI [ M + H ]]+=295.3。
The seventh step: na (2.7g,117.4mmol) was slowly added to anhydrous EtOH (50mL), stirred at room temperature until Na particles disappeared completely, added with methyl 2-amino-5-fluoro-6- (2-fluoro-6-methoxyphenyl) nicotinate (3.7g,12.6mmol) and diethyl malonate (4.0g,25.0mmol), and heated in a jar to 120 ℃ for three days. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and purified by column chromatography to give ethyl 6-fluoro-7- (2-fluoro-6-methoxyphenyl) -4-hydroxy-2-oxo-1, 2-dihydro-1, 8-naphthyridine-3-carboxylate (1.1g, yellow solid). LC-MS ESI [ M + H ]]+=377.4。
Eighth step: ethyl 6-fluoro-7- (2-fluoro-6-methoxyphenyl) -4-hydroxy-2-oxo-1, 2-dihydro-1, 8-naphthyridine-3-carboxylate (700mg, 1.9mmol) was dissolved in EtOH (70mL) and concentrated ammonia (3.5mL), and the mixture was heated to 120 ℃ in a jar closed to react for three days. The reaction solution was cooled to room temperature, and concentrated under reduced pressure to give 6-fluoro-7- (2-fluoro-6-methoxyphenyl) -4-hydroxy-2-oxo-1, 2-dihydro-1, 8-naphthyridine-3-carboxamide (680mg, pale yellow solid, crude product). LC-MS ESI [ M + H ]]+=348.6。
The ninth step: 6-fluoro-7- (2-fluoro-6-methoxyphenyl) -4-hydroxy-2-oxo-1, 2-dihydro-1, 8-naphthyridine-3-carboxamide (680mg, crude) was dissolved in phosphorus oxychloride (POCl)3) (10mL), the mixture was heated to 110 ℃ and reacted for 3 hours. The reaction solution is cooled to room temperature, concentrated under reduced pressure and dissolved in 10mL ethyl acetate, saturated NaHCO is used3The aqueous solution was made alkaline, the organic layer was separated, dried and concentrated under reduced pressure to give intermediate 2A (670mg, red solid). LC-MS ESI [ M + H ]]+=366.3。
Intermediate 2B: 2, 4-dichloro-6-fluoro-7- (2-fluoro-6-methoxyphenyl) -1, 8-naphthyridine-3-carbonitrile
Figure BDA0002733791230000191
Intermediate 2B, MS (M + H):354.3, was prepared using commercial reagents as starting materials, according to the synthetic procedure for intermediate 2A.
The intermediate 2C is 7- (2-amino-7-fluoro-benzo [ d ] thiazole-4-yl) -2, 4-dichloro-6-fluoro-1, 8-naphthyridine-3-methyl cyanide
Figure BDA0002733791230000192
Intermediate 2B was prepared using commercial reagents as starting materials, as described for the synthesis of intermediate 2A, and MS (M + H): 407.1/409.1.
Examples
Example 1: (S) -4- (4-acryloylpiperazin-1-yl) -7- (5-methyl-1H-indazol-4-yl) -2- ((1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydro-1, 7-naphthyridine-3-carbonitrile
Figure BDA0002733791230000193
The first step is as follows: intermediate 1(9.5g, 31.7mmol) and N-Boc-piperazine (8.81g, 47.4mmol) were dissolved in 1,4-dioxane (100mL), Diisopropylethylamine (DIEA) (8.18g, 63.4mmol) was added, and the temperature was raised to 90 ℃ for reaction overnight. The reaction solution is cooled to room temperature, concentrated under reduced pressure and concentrated with saturated sodium bicarbonate NaHCO3The aqueous solution was slurried, filtered, and the filter cake was dried and then slurried with ethyl acetate to purify it to obtain tert-butyl 4- (7-benzyl-3-cyano-2-hydroxy-5, 6,7, 8-tetrahydro-1, 7-naphthyridin-4-yl) piperazine-1-carboxylate (3.5g, dark green solid). LC-MS ESI [ M + H ]]+=450.4;1H NMR(400MHz,DMSO_d6):δ11.53(brs,1H),7.28-7.34(m,5H),3.62(s,2H),3.23-3.42(m,10H),2.40-2.56(m,4H),1.42(s,9H)。
The second step is that: tert-butyl 4- (7-benzyl-3-cyano-2-hydroxy-5, 6,7, 8-tetrahydro-1, 7-naphthyridin-4-yl) piperazine-1-carboxylate (1.0g, 2.23mmol) and (S) - (1-methylpyrrolidin-2-yl) methanol (384mg,3.34mmol) were dissolved in tetrahydrofuran THF (20mL) and triphenylphosphine PPh was added under nitrogen protection3(1.17g,4.57mmol) and diisopropyl azodicarboxylate DIAD (900mg,4.46mmol) were stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure and purified by column chromatography to give tert-butyl (S) -4- (7-benzyl-3-cyano-2- ((1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydro-1, 7-naphthyridin-4-yl) piperazine-1-carboxylate (750mg, pale yellow solid). LC-MS ESI [ M + H ]]+=547.8;1H NMR(400MHz,CD3Cl):δ7.26-7.36(m,5H),4.24-4.26(m,2H),3.68(s,2H),3.53-3.58(m,6H),3.31(brs,4H),3.06-3.10(m,1H),2.64(brs,4H),2.50(s,3H),2.25-2.29(m,1H),1.98-2.03(m,1H),1.64-1.84(m,4H),1.47(s,9H)。
The third step: tert-butyl (S) -4- (7-benzyl-3-cyano-2- ((1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydro-1, 7-naphthyridin-4-yl) piperazine-1-carboxylate (350mg,0.64mmol) was dissolved in THF (15mL), AcOH acetate (2mL) and 10% Pd/C (100mg) were added, and stirring was performed at room temperature for 3 days under a hydrogen balloon atmosphere. Filtering the reaction solution, concentrating the filtrate under reduced pressure, dissolving the residue in ethyl acetate, and adding saturated sodium carbonate Na2CO3The aqueous solution was washed, dried over anhydrous magnesium sulfate MgSO4, and concentrated under reduced pressure to give tert-butyl (S) -4- (3-cyano-2- ((1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydro-1, 7-naphthyridin-4-yl) piperazine-1-carboxylate (260mg, pale yellow solid). LC-MS ESI [ M + H ]]+=457.6。
The fourth step: (S) -4- (3-cyano-2- ((1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydro-1, 7-naphthyridin-4-yl) piperazine-1-carboxylic acid tert-butyl ester (260mg, 0.57mmol) and 4-bromo-5-methyl-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-indazole (290mg,0.85mmol) were dissolved in 1,4-dioxane (10mL) and cesium carbonate Cs, respectively, was added under argon2CO3(372mg,1.14mmol), 2-dicyclohexylphosphine-2 ',6' -diisopropoxybiphenyl Ru-phos (51mg,0.11mmol, 0.2) and Pd-Ruphos-G3(24mg,0.028mmol), after addition, it was replaced with argon three times and heated to 90 ℃ for overnight reaction. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and purified by column chromatography to give tert-butyl (S) -4- (3-cyano-7- (5-methyl-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-indazol-4-yl) -2- ((1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydro-1, 7-naphthyridin-4-yl) piperazine-1-carboxylate (70mg, white solid). LC-MS ESI [ M + H ]]+=717.9;1H NMR(400MHz,CDCl3):δ7.99(s,1H),7.29(s,2H),5.70(s,2H),4.31(s,2H),3.49-3.57(m,8H),3.39(brs,4H),3.13(brs,1H),2.73(brs,2H),2.53(brs,2H),1.62-2.07(m,4H),1.70(brs,3H),1.48(s,9H),0.86-0.92(m,2H),-0.07(s,9H)。
The fifth step: mixing (S) -4- (3-cyano-7- (5-methyl-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-indazol-4-yl) -2- ((1-methylpyrrolidin-2-yl) methoxy) -5,6,7, 8-tetrahydro-1, 7-naphthalenePyridin-4-yl) piperazine-1-carboxylic acid tert-butyl ester (70mg, 0.10mmol) was dissolved in trifluoroacetic acid/dichloromethane TFA/DCM (5mL/5mL) and stirred at room temperature overnight. The reaction solution is decompressed and concentrated, and saturated NaHCO is added3The aqueous phase was extracted three times with dichloromethane, the organic phases were combined, dried over anhydrous MgSO4, filtered, and concentrated to give (S) -7- (5-methyl-1H-indazol-4-yl) -2- ((1-methylpyrrolidin-2-yl) methoxy) -4- (piperazin-1-yl) -5,6,7, 8-tetrahydro-1, 7-naphthyridine-3-carbonitrile (20mg, yellow solid). LC-MS ESI [ M + H ]]+=487.3。
And a sixth step: (S) -7- (5-methyl-1H-indazol-4-yl) -2- ((1-methylpyrrolidin-2-yl) methoxy) -4- (piperazin-1-yl) -5,6,7, 8-tetrahydro-1, 7-naphthyridine-3-carbonitrile (20mg, 0.041mmol) was dissolved in dichloromethane (5mL), diisopropylethylamine DIPEA (16mg, 0.12mmol) and acryloyl chloride (4mg, 0.044mmol) were added sequentially at 0 degrees, and stirred at room temperature for 2 hours. The reaction mixture was washed with a saturated ammonium chloride solution and a saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the residue was purified by preparative chromatography to give the objective compound (yellow solid). LC-MS ESI [ M + H ]]+=541.3;1H NMR(400MHz,CD3OD):δ8.07(s,1H),7.23-7.28(m,2H),6.78-6.85(m,1H),6.25(dd,J=16.8,2.0Hz,1H),5.78(dd,J=10.8,2.0Hz,1H),4.43-4.47(m,1H),4.24-4.31(m,3H),3.82(brs,4H),3.52-3.55(m,2H),3.48(brs,4H),3.06-3.10(m,1H),2.87-2.89(m,2H),2.76-2.80(m,1H),2.54(s,3H),2.34-2.41(m,4H),2.06-2.11(m,1H),1.78-1.84(m,2H),1.65-1.70(m,1H)。
Example 2: 4- (4-acryloylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -1, 8-naphthyridine-3-carbonitrile
Figure BDA0002733791230000211
The first step is as follows: 2, 4-dichloro-6-fluoro-7- (2-fluoro-6-methoxyphenyl) -1, 8-naphthyridine-3-carbonitrile (670mg,1.8mmol), 1-Cbz-piperazine (306mg, 1.4mmol), and DIEA (1.2g, 9.3mmol) were dissolved in 1.4-dioxane (20mL) and reacted at room temperature overnight. Diluting the reaction solution with 100mL ethyl acetate, washing twice with water, drying, and concentrating under reduced pressure to obtain 4-, (Benzyl 2-chloro-3-cyano-6-fluoro-7- (2-fluoro-6-methoxyphenyl) -1, 8-naphthyridin-4-yl) piperazine-1-carboxylate (605mg, red solid). LC-MS ESI [ M + H ]]+=550.6。
The second step is that: benzyl 4- (2-chloro-3-cyano-6-fluoro-7- (2-fluoro-6-6-methoxyphenyl) -1, 8-naphthyridin-4-yl) piperazine-1-carboxylate (605mg, 1.1mmol) was dissolved in TFA (2.5mL) and water (0.5mL) and heated to 120 degrees for reaction overnight. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was added to ethyl acetate and concentrated under reduced pressure twice to give 6-fluoro-7- (2-fluoro-6-methoxyphenyl) -2-oxo-4- (piperazin-1-yl) -1, 2-dihydro-1, 8-naphthyridine-3-carbonitrile (690mg, red solid). LC-MS ESI [ M + H ]]+=398.5。
The third step: 6-fluoro-7- (2-fluoro-6-methoxyphenyl) -2-oxo-4- (piperazin-1-yl) -1, 2-dihydro-1, 8-naphthyridine-3-carbonitrile (690mg, crude) was dissolved in DCM (20mL) and triethylamine TEA (1.9g, 19mmol) and (Boc) were added2O (828mg, 3.8mmol), and reacted at room temperature for 3 hours. The reaction solution was concentrated under reduced pressure and purified by column chromatography to give tert-butyl 4- (3-cyano-6-fluoro-7- (2-fluoro-6-methoxyphenyl) -2-oxo-1, 2-dihydro-1, 8-naphthyridin-4-yl) piperazine-1-carboxylate (280mg, yellow solid). LC-MS ESI [ M + H ]]+=498.5。
The fourth step: tert-butyl 4- (3-cyano-6-fluoro-7- (2-fluoro-6-methoxyphenyl) -2-oxo-1, 2-dihydro-1, 8-naphthyridin-4-yl) piperazine-1-carboxylate (280mg, 0.56mmol) and (S) - (1-methylpyrrolidin-2-yl) methanol (96mg,0.83mol) were dissolved in THF (10mL) and triphenylphosphine PPh was added under nitrogen protection3(287mg,1.12mmol) and diisopropyl azodicarboxylate DIAD (226mg,1.12mmol) were stirred at room temperature overnight. The reaction solution was concentrated under reduced pressure and purified by column chromatography to give tert-butyl 4- (3-cyano-6-fluoro-7- (2-fluoro-6-methoxyphenyl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -1, 8-naphthyridin-4-yl) piperazine-1-carboxylate (180mg, pale yellow solid). LC-MS ESI [ M + H ]]+=595.3。
The fifth step: tert-butyl 4- (3-cyano-6-fluoro-7- (2-fluoro-6-methoxyphenyl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -1, 8-naphthyridin-4-yl) piperazine-1-carboxylate (180mg, 0.30mmol) was dissolved in DCM (5mL) and boron tribromide BBr was added dropwise at-78 degrees3(1.0M in DCM, 6mL) and slowly warmed to room temperature for reactionOvernight. The reaction was quenched by addition of MeOH (10mL) and Na2CO3The solids were neutralized, filtered, and the filtrate was concentrated under reduced pressure to give 6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -4- (piperazin-1-yl) -1, 8-naphthyridine-3-carbonitrile (200mg, yellow solid, crude). LC-MS ESI [ M + H ]]+=481.4。
And a sixth step: 6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -4- (piperazin-1-yl) -1, 8-naphthyridine-3-carbonitrile (200mg, crude) and TEA (0.5mL) were dissolved in DCM (1mL), and acryloyl chloride (2 drops) was added at room temperature and reacted at room temperature for 2 hours. The reaction solution was filtered, and the filtrate was concentrated under reduced pressure and purified by preparative chromatography to give 4- (4-acryloylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -2- (((S) -1-methylpyrrolidin-2-yl) methoxy) -1, 8-naphthyridine-3-carbonitrile (17mg, yellow solid). LC-MS ESI [ M + H ]]+=535.5。1H NMR(400MHz,CD3OD):δ8.20(d,1H),7.20-7.26(m,1H),6.62-6.89(m,1H),6.69(d,1H),6.53(t,1H),6.28(dd,1H),5.81(dd,1H),4.68-4.72(m,1H),4.40-4.45(m,1H),3.97(brs,4H),3.73(brs,4H),3.07-3.10(m,1H),2.81-2.87(m,1H),2.56(s,3H),2.35-2.41(m,1H),2.08-2.15(m,1H),1.76-1.90(m,3H)。
The following compounds of examples were prepared according to the synthetic method of example 1 using commercial reagents as starting materials.
Figure BDA0002733791230000221
Figure BDA0002733791230000231
Figure BDA0002733791230000241
The following compounds of examples were prepared according to the synthetic method of example 2 using commercial reagents as starting materials.
Figure BDA0002733791230000242
Figure BDA0002733791230000251
Test example 1 KrasG12CFunctional analysis
KRAS Using CisBioG12CSOS1 kit for testing compound inhibition SOS1 and KRAS by using Binding assay methodG12CThe efficacy of protein-protein interactions between, the results are in IC50The values are represented.
The test method comprises the following steps: (1) test compounds were tested at 1000nM concentration, compounds were diluted 3-fold in a 384-well plate in 100% DMSO at 200-fold final concentration, 10 concentrations. A50 nL 200-fold final concentration of compound was transferred to the 384well plates of interest using the knockout Echo 550. Respectively adding 50nL of 100% DMSO into the negative control well and the positive control well; (2) preparing a Tag1 SOS1 solution with 4 times of final concentration by using a Diluent buffer; (3) add 2.5. mu.L of a 4-fold final concentration solution of Tag1 SOS1 to a 384-well plate; (4) 4-fold final concentration of Tag2KRAS was made up using Diluent bufferG12CA solution; (5) add 2.5. mu.L of Tag2KRAS at 4-fold final concentration to the compound wells and positive control wells, respectivelyG12CA solution; add 2.5. mu.L of differential buffer to the negative control wells; (6) centrifuging a 384-hole plate at 1000rpm for 30 seconds, shaking and uniformly mixing, and incubating at room temperature for 15 min; (7) preparing a solution of Anti Tag1 TB3+ with the final concentration of 1 time and a solution of Anti Tag2 XL665 with the final concentration of 1 time by using a Detection buffer, mixing the two solutions uniformly, and adding 5 mu L of mixed solution into each hole; (8) centrifuging a 384-well plate at 1000rpm for 30 seconds, shaking and uniformly mixing, and incubating for 120 minutes at room temperature; (9) reading Em665/620 by an Envision microplate reader; (10) data analysis, calculation formula
Figure BDA0002733791230000261
Wherein Min signal negative control well mean Max signal positive control well mean. The dose-effect curve was fitted with the log of the concentration as the X-axis and the percent inhibition as the Y-axis using the analysis software GraphPad Prism 5 log (inhibitor) vResponse Variable slope was fitted to the dose-response curves to derive the IC of each compound for enzyme activity50The value is obtained. The fitting formula is: y ═ Bottom + (Top Bottom)/(1+10^ ((LogIC)50X)*HillSlope))。
As a result: example Compounds of the invention vs KRasG12CBinding to SOS1 showed significant inhibitory effect, IC of most of the example compounds50Less than 1000nM, IC of some compounds as examples 7, 1350Less than 100 nM.
Test example 2: test of Effect of Compounds of the present invention on the cell proliferation of NCI-H358, MiaPaca-2 and phosphorylation of downstream Signal ERK
Test method one (2D) NCI-H358 (lung cancer) and MiaPaca-2 cells (pancreatic cancer) cells (100. mu.L/well, 20000 cells/mL) were seeded into 96-well culture plates and supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin sulfate, respectively. Cells were treated with a starting 10. mu.M solution of test compound diluted three times in eight gradients using 0.5% dimethylsulfoxide as a blank and 5% CO2Incubate in incubator for a certain period of time (5-7 days). At the end of the incubation, 10. mu.L of MTT stock solution (5mg/mL) was added to each well. The plates were incubated at 37 ℃ for 4 hours and then the medium was removed. Dimethylsulfoxide (100 μ L) was added to each well, followed by sufficient shaking. The absorbance of the formazan product was measured at 570nm on a Thermo Scientific Varioskan Flash multimodal reader. IC was obtained by fitting dose-response data to a three-parameter nonlinear regression model using GraphPad Prism 6.0 software50The values, test results are shown in Table 1, where A < 500nM, 500 nM. ltoreq.B < 5000, 5000nM < C.
TABLE 1
Figure BDA0002733791230000262
Figure BDA0002733791230000271
As a result, the invention provides the most partProliferation inhibitory Activity of the Compounds of the examples on NCI-H358 and MiaPaca-2 cells, IC50The values were all less than 5000nM, and the cell proliferation inhibitory activity of some examples was less than 500nM, as in examples 7, 13, 15.
Test method two (3D) tumor cells in logarithmic growth phase were diluted to a certain concentration with culture medium and seeded in 96-well plates with ultra-low attachment surface at 80. mu.L/well. Cells were incubated overnight at 37 ℃ in a humidity chamber. The next day the plate was added serial dilutions of test compound (10 concentrations, 3-fold dilution), 20 μ L/well and incubated in incubator for 96 h. Taking out the plate, standing at room temperature, and adding the same volume
Figure BDA0002733791230000272
Incubation of reagents for 1h, En VisionTMThe plate reader detects the signal. The signal was converted to percent inhibition using the following equation: % inhibition 100- [ (test compound signal-median minimum signal)/(median maximum signal-median minimum signal) x 100]. The maximum signal is the signal value for wells without inhibitor and the minimum signal is the signal value for wells containing a reference inhibitor sufficient to completely inhibit cell proliferation, a four-parameter non-linear regression fit curve was performed on the percent inhibition for each concentration of compound and the IC50 was calculated. The test results are shown in Table 2, wherein A is less than 200nM, B is more than or equal to 200nM and less than 1000nM, and C is more than 1000nM
TABLE 2
Compound (I) NCI-H358 cell Activity MiaPaca-2 cell Activity
Example 1 B B
Example 2 B B
Example 3 A A
Example 4 A A
Example 5 B B
Example 6 A A
Example 7 A A
Example 8 B B
Example 9 A A
Example 10 A A
Example 11 B B
Example 12 B B
Example 13 A A
Example 14 A A
Example 15 A A
Example 16 B B
As a result: the majority of the example compounds provided by the present invention have proliferation-inhibiting activity, IC, on NCI-H358 and MiaPaca-2 cells50Less than 1000nM, and in some examples, less than 200nM IC50 for cell proliferation inhibitory activity of example 3,4, 5,6,7, 9,10, 12, 13, 14, 15 on NCI-H358 and MiaPaca-2.
Test method three (ERK phosphorylation): miapaca-2 or H358 cells were seeded at a certain concentration in 96-well plates and placed at 37 ℃ in 5% CO2The next day, the plate was incubated overnight with serial dilutions of test compounds (5 concentrations, 3 fold dilutions) for 24H (Miapaca-2) or 3H (H358), followed by lysis of the cells with lysis solutions containing protease and phosphatase inhibitors to extract the protein, and western blot to detect the level of p-ERK, as shown in Table 3, where A < 500nM,500nM≤B<1000,1000nM≤C<5000nM,
TABLE 3
Compound (I) Inhibition of ERK phosphorylation level
Example 1 B
Example 2 B
Example 3 A
Example 4 A
Example 5 A
Example 6 A
Example 7 A
Example 8 B
Example 9 A
Example 10 A
Example 11 B
Example 12 A
Example 13 A
Example 14 A
Example 15 A
Example 16 C
As a result: most of the example compounds provided by the invention have obvious inhibition effect on the level of phosphorylation ERK of NCI-H358 and MiaPaca-2, the IC50 is less than 500nM, and the IC50 of some example compounds such as examples 7 and 13 for the inhibition of phosphorylation ERK is less than 200 nM.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (13)

1. A condensed cyanopyridine compound shown in general formula I-1 or I-2, or its pharmaceutically acceptable salt,
Figure FDA0003205533900000011
in the formula:
r1 is independently selected from hydrogen, halogen;
r2, R3 are independently hydrogen;
r4 is
Figure FDA0003205533900000012
Wherein p is 1 or 2;
r5 is independently selected from hydrogen, halogen, m is independently selected from an integer from 0 to 6;
m is O; ra, Rb, Rc, Rd, Re, Rf, Rg, Rh are independently selected from hydrogen, C1-C6 alkyl; wherein one or more hydrogen atoms of the C1-C6 alkyl group may be substituted with a substituent selected from the group consisting of: deuterium, cyano;
ar is independently selected from substituted or unsubstituted phenyl and pyridyl, or substituted or unsubstituted naphthyl, naphthyridinyl, indazolyl, benzimidazolyl, benzothiazolyl; the substitution means substitution with one or more substituents selected from the group consisting of: halogen, C1-C4 alkyl, hydroxyl, amino, cyano, C1-C4 alkoxy, C1-C4 haloalkyl, C1-C4 haloalkoxy.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein m is 0, 1, or 2.
3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein p is 1.
4. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein Ar is independently selected from substituted or unsubstituted phenyl, or substituted or unsubstituted naphthyl, indazolyl, benzothiazolyl; the one or more substituents are selected from the group consisting of: hydrogen, halogen, C1-C4 alkyl, hydroxyl and amino.
5. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein Ra, Rb, Rc, Rd, Re, Rf, Rg, Rh are each independently selected from: hydrogen, methyl, cyanomethylene.
6. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein Ar is independently selected from
Figure FDA0003205533900000021
Wherein Rp is selected from: halogen, C1-C4 alkyl, hydroxyl and amino, and q is 1,2,3 or 4.
7. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R1 is independently selected from the group consisting of hydrogen, fluoro; r2, R3 are independently hydrogen.
8. The compound of claim 1, or a pharmaceutically acceptable salt thereof,
r1 is selected from hydrogen, fluoro;
r2, R3 are independently hydrogen;
ra, Rb, Rc, Rd, Re, Rf, Rg, Rh are each independently selected from hydrogen, methyl, cyanomethylene;
m is O;
r4 is
Figure FDA0003205533900000022
Wherein p is 1;
r5 is independently selected from hydrogen, halogen;
m is selected from 0, 1 or 2;
ar is independently selected from substituted or unsubstituted phenyl, or substituted or unsubstituted naphthyl, indazolyl, benzothiazolyl; the one or more substituents are selected from the group consisting of: hydrogen, halogen, C1-C4 alkyl, hydroxyl and amino.
9. The compound of any one of claims 1-8, or a pharmaceutically acceptable salt thereof, wherein the compound has the structure:
Figure FDA0003205533900000023
Figure FDA0003205533900000031
10. the use of a compound of any one of claims 1-9, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a disease associated with a Ras protein mutation.
11. The use according to claim 10, wherein the medicament is a therapeutic agent for tumors.
12. The use of claim 11, wherein the tumor is independently selected from lung cancer, pancreatic cancer, liver cancer, colorectal cancer, bile duct cancer, brain cancer, leukemia, lymphoma, melanoma, thyroid cancer, nasopharyngeal cancer.
13. A pharmaceutical composition, comprising:
(i) an effective amount of a compound of any one of claims 1-9, or a pharmaceutically acceptable salt thereof; and
(ii) a pharmaceutically acceptable carrier.
CN202011126559.0A 2020-04-09 2020-10-20 Condensed cyanopyridine compound, preparation method and application Active CN112142735B (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN2020102764335 2020-04-09
CN202010276433 2020-04-09
CN202010359411 2020-04-29
CN2020103594115 2020-04-29

Publications (2)

Publication Number Publication Date
CN112142735A CN112142735A (en) 2020-12-29
CN112142735B true CN112142735B (en) 2021-09-17

Family

ID=73954185

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011126559.0A Active CN112142735B (en) 2020-04-09 2020-10-20 Condensed cyanopyridine compound, preparation method and application

Country Status (1)

Country Link
CN (1) CN112142735B (en)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021057832A1 (en) * 2019-09-25 2021-04-01 Jacobio Pharmaceuticals Co., Ltd. Kras mutant protein inhibitor
WO2022095960A1 (en) * 2020-11-06 2022-05-12 泰励生物科技(上海)有限公司 Kras inhibitors for treatment of cancers
CN116600808A (en) * 2021-02-09 2023-08-15 苏州阿尔脉生物科技有限公司 Tetrahydronaphthyridine derivative serving as KRAS mutant G12C inhibitor, and preparation method and application thereof
CN115073450A (en) * 2021-03-15 2022-09-20 药雅科技(上海)有限公司 KRAS G12C Preparation and application of mutant protein inhibitor
CN115073451A (en) * 2021-03-15 2022-09-20 药雅科技(上海)有限公司 KRAS G12D Preparation and application of mutant protein inhibitor
TWI814234B (en) * 2021-03-15 2023-09-01 大陸商藥雅科技(上海)有限公司 Preparation and Application of Mutant Protein Inhibitors
TWI810803B (en) * 2021-03-15 2023-08-01 大陸商藥雅科技(上海)有限公司 Preparation and Application of Mutant Protein Inhibitors
CN115160309B (en) * 2021-04-07 2024-04-09 药雅科技(上海)有限公司 KRAS G12C Preparation and application of mutant protein heterocyclic inhibitor
WO2022217042A1 (en) * 2021-04-09 2022-10-13 Ikena Oncology, Inc. Naphthyl-substituted quinoline-4(1h)-ones and related compounds and their use in treating medical conditions
US20230106174A1 (en) 2021-05-05 2023-04-06 Revolution Medicines, Inc. Ras inhibitors
KR20240017811A (en) 2021-05-05 2024-02-08 레볼루션 메디슨즈, 인크. RAS inhibitors for the treatment of cancer
WO2023125627A1 (en) * 2021-12-28 2023-07-06 Lynk Pharmaceuticals Co., Ltd. Nitrogen-containing heterocyclic compound and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016168540A1 (en) * 2015-04-15 2016-10-20 Araxes Pharma Llc Fused-tricyclic inhibitors of kras and methods of use thereof
WO2018140600A1 (en) * 2017-01-26 2018-08-02 Araxes Pharma Llc Fused hetero-hetero bicyclic compounds and methods of use thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016168540A1 (en) * 2015-04-15 2016-10-20 Araxes Pharma Llc Fused-tricyclic inhibitors of kras and methods of use thereof
WO2018140600A1 (en) * 2017-01-26 2018-08-02 Araxes Pharma Llc Fused hetero-hetero bicyclic compounds and methods of use thereof

Also Published As

Publication number Publication date
CN112142735A (en) 2020-12-29

Similar Documents

Publication Publication Date Title
CN112142735B (en) Condensed cyanopyridine compound, preparation method and application
CN112300194B (en) Condensed ring pyridone compounds, preparation method and application
EP3712151B1 (en) (s)-1&#39;-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4&#39;-piperidine]-5-amine as shp2 inhibitor for the treatment of cancer
CN111704611B (en) Aryl spiro SHP2 inhibitor compound, preparation method and application
EA035499B1 (en) Novel glutaminase inhibitors
CN113061132B (en) Condensed ring lactam compound, preparation method and application
CN109721599B (en) Amino-substituted nitrogen-containing fused ring compound and preparation method and application thereof
CN109721600B (en) Nitrogen-containing fused ring compounds and preparation method and application thereof
CN110950876B (en) Furanolactam compounds, preparation method and application
CN112457326B (en) Aromatic heterocyclic lactam compound, preparation method and application
CN115697986A (en) Isotopically substituted spiroaromatic ring compounds and their use
CN113527299B (en) Nitrogen-containing condensed ring compound, preparation method and application
CN111518100A (en) Cyclopropenoarylbenzofuran substituted nitrogen heteroaryl compound and application thereof
CN112300196A (en) Piperidine condensed ring compound, preparation method and application
CN110857300B (en) Condensed ring triazole compounds, preparation method and application
CN112094269B (en) Saturated six-membered ring heterocyclic compound, preparation method and application
CN112300173B (en) Nitrogen-containing polycyclic compounds, preparation method and application
CN114524810A (en) Pyrimidine heterocyclic compound, preparation method and application
CN112778336B (en) Nitrogen-containing condensed ring STING regulator compound, preparation method and application
CN111704610B (en) Pyrrolylamido pyridone compounds, preparation method and application
CN111057048B (en) Aminopyrazine/pyridine compound, preparation method and application
CN114907350A (en) Nitrogen-containing condensed ring compounds, preparation method and application
CN111763217B (en) Thieno-nitrogen heterocyclic compounds, preparation method and application
CN114685487A (en) Pyrimidine heterocyclic compound, preparation method and application
CN114075196A (en) Aromatic ring-lactam compound, preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant