CN112898312B - Fused polycyclic pyridone derivative and application thereof - Google Patents

Fused polycyclic pyridone derivative and application thereof Download PDF

Info

Publication number
CN112898312B
CN112898312B CN202110108914.XA CN202110108914A CN112898312B CN 112898312 B CN112898312 B CN 112898312B CN 202110108914 A CN202110108914 A CN 202110108914A CN 112898312 B CN112898312 B CN 112898312B
Authority
CN
China
Prior art keywords
optionally substituted
compound
pharmaceutically acceptable
influenza
hydrogen
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
CN202110108914.XA
Other languages
Chinese (zh)
Other versions
CN112898312A (en
Inventor
王永钢
林寨伟
廖辉
胡双华
张世喜
杨文逊
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.)
Hunan Nanxin Pharmaceutical Co ltd
Original Assignee
Hunan Nanxin Pharmaceutical Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Hunan Nanxin Pharmaceutical Co ltd filed Critical Hunan Nanxin Pharmaceutical Co ltd
Priority to CN202110108914.XA priority Critical patent/CN112898312B/en
Publication of CN112898312A publication Critical patent/CN112898312A/en
Application granted granted Critical
Publication of CN112898312B publication Critical patent/CN112898312B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/22Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains four or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Health & Medical Sciences (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Molecular Biology (AREA)
  • Pulmonology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the field of medicines, and particularly relates to a condensed polycyclic pyridone derivative shown as a formula I, pharmaceutically acceptable salts and solvates thereof, including hydrates, polymorphs, prodrugs, co-crystals, tautomers, stereoisomers and uses thereof, and more particularly, the compound can be used as an anti-influenza medicine with a CEN (cytokine induced potentiator) inhibition effect.

Description

Fused polycyclic pyridone derivative and application thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to a fused polycyclic pyridone derivative and application thereof. More specifically, the compound of the present invention can be used as an anti-influenza drug having a CEN inhibitory effect.
Background
Influenza is a highly contagious respiratory disease caused by influenza virus, wherein influenza A virus has the widest host range, can infect both birds and mammals, and is very easy to cause pandemics worldwide. Influenza viruses have developed 4 major outbreaks of influenza in only a few hundred years (1918, 1957, 1968, 2009). Among them, the outbreak of "spanish influenza" (H1N1) in 1918 directly caused about 5000 thousands of deaths, and is a big outbreak of influenza with the largest number of deaths in human history. Whereas the last outbreak of "Mexico flu" (A/2009/H1N1) in 2009 spread rapidly to 214 countries worldwide, only 12 months of the outbreak led to approximately 20 million deaths. Therefore, it is very important to develop a novel broad-spectrum anti-influenza virus drug for the prevention and early treatment of influenza virus infection. Typically, these new strains result from the transmission of existing influenza viruses from other animal species to humans.
The influenza virus is an RNA virus of Orthomyxoviridae (Orthomyxoviridae) and belongs to the genus influenza virus. Influenza viruses are mainly classified into A, B, C types, also called type A, B and C, according to the antigenic and genetic properties of the viral particle Nucleoprotein (NP) and matrix protein (M). The three types of viruses have similar biochemical and biological characteristics. The virus particles are 80-120nm in diameter and are usually approximately spherical, but filamentous forms may occur. The virus is composed of three layers, the inner layer is the virus nucleocapsid, containing Nucleoprotein (NP), P protein and RNA. NP is a soluble antigen (S antigen), has type specificity, and is antigenically stable. The P protein (P1, P2, P3) may be a polymerase required for RNA transcription and replication. The middle layer is virus envelope composed of one lipoid and one Membrane Protein (MP), and the MP has stable antigenicity and type specificity. The outer layer is a radial protuberance made of two different glycoproteins, hemagglutinin (H) and neuraminidase (N). H can cause erythrocyte agglutination, is a tool for adsorbing viruses on the surfaces of sensitive cells, N can hydrolyze mucin, and N-acetylneuraminic acid at the tail end of a receptor specific glycoprotein on the cell surface is a tool for separating the viruses from the cell surface after the replication of the viruses is finished. H and N both have variation characteristics, so that only the specific antigenicity of the strain exists, and the antibody has a protection effect.
Unusual for viruses, nucleic acids whose genomes are not single fragments; in contrast, the genome contains 7 or 8 segments of negative-sense RNA. The influenza a genome encodes 11 proteins: hemagglutinin (H), neuraminidase (N), Nucleoprotein (NP), M1, M2, NS1, NS2(NEP), PA, PB1, PB1-F2, and PB 2. H and N are macromolecular glycoproteins outside the virion. HA is a lectin that mediates binding of the virus to the target cell and entry of the viral genome into the target cell, while NA is involved in the release of progeny virus from infected cells by cleaving sugars that bind to mature viral particles. Therefore, these proteins have been targets for antiviral drugs. Moreover, these proteins are antigens of antibodies that can be produced. Influenza a viruses are classified into subtypes based on antibody responses to H and N, forming the basis for the distinction between H and N in, for example, H5N 1.
Because the influenza virus genome is small, the synthesis of its desired proteins is dependent on the translation system of the host cell. Thus, the messenger rna (mrna) of influenza virus needs to have both a 5 'CAP (CAP) structure and a 3' -poly (a) tail structure that can be recognized by the translation system of the host cell. Wherein the 5 'cap is "captured" by cleavage of the PA subunit endonuclease activity of the influenza RNA polymerase complex from the 5' end of the host cell precursor mRNA. This so-called CAP-dependent mode, the CAP-like structure that captures host mRNA for viral self-mRNA transcription, is necessary for influenza virus transcription initiation.
Just because the cap-dependent endonuclease is a key link in the replication cycle of influenza virus, and because a similar mechanism and corresponding protease do not exist in host cells, the inhibitor aiming at the cap-dependent endonuclease can selectively block the transcription process of the influenza virus, and does not influence the host cells, namely has the effect of inhibiting the cap-dependent endonuclease (CEN). This mechanism then becomes a potential anti-influenza drug target.
The existing means for preventing and treating influenza virus infection mainly comprise influenza vaccine inoculation and influenza virus resisting medicine application. Vaccination with influenza is the primary prophylactic method and is highly effective, but it has the major disadvantages of having to be injected once a year and having low efficacy against adults and young children with low immunity and high risk of disease. Furthermore, if the predicted influenza vaccine species are incorrect, the efficacy of the vaccine will be reduced to 25%. Thus, anti-influenza virus drugs are the first line of treatment for influenza.
To date, FDA-approved anti-influenza virus drugs for clinical use fall broadly into two categories: 1) m2 ion channel blockers including amantadine and rimantadine; 2) neuraminidase inhibitors, mainly oseltamivir and zanamivir, and peramivir and ranimivir, which have been approved in several countries. In addition, the anti-influenza drugs currently approved only in japan on the market are the RNA inhibitors faviravir and xoflurza. Because the influenza viruses which are epidemic at present are resistant to amantadine drugs (S31N mutation appears in the NS gene of the virus), the amantadine drugs are no longer the first choice drugs recommended by WHO for preventing and treating influenza. Neuraminidase inhibitor drugs are the main choice for antiviral therapy of influenza patients, however, researchers have successively found drug-resistant strains of H1N1, H5N1, H3N2 and B that are resistant to oseltamivir, indicating that their resistance remains of great concern. Recently, the endonuclease inhibitors baloxavirmoxil (Xofluza) were approved in Japan (Hayden FG, et al, "Baloxavir Marboxil for functional Influenza in Adults and adolescents." N Engl J Med.2018; 379 (10): 913-. However, the drug resistance of influenza virus inevitably appears, and small molecules with different chemical structures aiming at the same target point can have different drug resistance, so that the development of new influenza virus nuclease endonuclease inhibitors is still necessary.
The cap-dependent endonuclease is a key link in the replication cycle of the influenza virus, and because a similar mechanism and corresponding protease do not exist in host cells, an inhibitor aiming at the cap-dependent endonuclease can selectively block the transcription process of the influenza virus without influencing the host cells. This mechanism becomes a potential anti-influenza drug target. The present invention fulfills these needs and provides other related advantages.
Disclosure of Invention
It is an object of the present invention to provide novel pyridone derivatives which are useful as cap-dependent endonuclease inhibitors and which are superior to existing pyridone derivatives in at least one of activity, pharmacokinetic properties such as bioavailability, cytotoxicity and the like.
It is another object of the present invention to provide pyridone derivatives which not only have excellent cap-dependent endonuclease inhibitory activity and low cytotoxicity, but also have significantly improved pharmacokinetic properties, particularly bioavailability.
The invention provides a novel compound serving as an influenza virus RNA polymerase inhibitor, and particularly provides a fused polycyclic pyridone derivative with a CEN (cytokine-like protein) inhibition effect as shown in a general formula I, and pharmaceutically acceptable salts, solvates, hydrates, N-oxides, polycrystals, cocrystals, tautomers, stereoisomers or mixtures or prodrugs thereof.
Figure BDA0002922605880000031
Wherein the content of the first and second substances,
R1、R2and R3Each independently selected from hydrogen or deuterium, and R1、R2And R3At least one of them is deuterium;
R4、R5、R6and R7Each independently selected from hydrogen or fluorine;
R8is hydrogen, an optionally substituted phosphate group, C1-4Alkyl radical, C2-4Alkenyl radical, C3-6Carbocyclyl group, (C)3-6Carbocyclyl) -C1-2Alkylene, heterocyclic group consisting of 3 to 6 atoms, (heterocyclic group consisting of 3 to 6 atoms) -C1-2Alkylene radical, C6-10Aryl radical, C6-10aryl-C1-2Alkylene, heteroaryl of 5 to 10 atoms, (heteroaryl of 5 to 10 atoms) -C1-2Alkylene, wherein said C1-4Alkyl radical, C2-4Alkenyl radical, C3-6Carbocyclyl, C3-6carbocyclyl-C1-2Alkylene, heterocyclic group consisting of 3 to 6 atoms, (heterocyclic group consisting of 3 to 6 atoms) -C1-2Alkylene radical, C6-10Aryl radical, C6-10aryl-C1-2Alkylene, heteroaryl of 5 to 10 atoms and (heteroaryl of 5 to 10 atoms) -C1-2Each alkylene is independently unsubstituted or substituted with 1, 2, 3 or 4 substituents, or-C (═ O) -Y1、-C(=O)-O-Y2、-(CH2)-O-(C=O)-Y1、-(CH2)-O-(C=O)-O-Y2、 -(CHCH3)-O-(C=O)-Y1And- (CHCH)3)-O-(C=O)-O-Y2
Y1Is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, mono-substituted amino group, di-substituted amino and-C (R)*)2NHR**(ii) a And each R*And R**Independently is hydrogen or optionally substituted C1-6An alkyl group;
Y2is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, monosubstituted amino groups.
Wherein R is1When is deuterium; preferred compounds are those having the structure shown in formula II below:
Figure BDA0002922605880000041
R2and R3Each independently selected from hydrogen or deuterium;
R4、R5、R6and R7Each independently selected from hydrogen or fluorine;
R8is hydrogen, an optionally substituted phosphate group, C1-4Alkyl radical, C2-4Alkenyl radical, C3-6Carbocyclyl group, (C)3-6Carbocyclyl) -C1-2Alkylene, heterocyclic group consisting of 3 to 6 atoms, (heterocyclic group consisting of 3 to 6 atoms) -C1-2Alkylene radical, C6-10Aryl radical, C6-10aryl-C1-2Alkylene, heteroaryl of 5 to 10 atoms, (heteroaryl of 5 to 10 atoms) -C1-2Alkylene, wherein said C1-4Alkyl radical, C2-4Alkenyl radical, C3-6Carbocyclyl, C3-6carbocyclyl-C1-2Alkylene, heterocyclic group consisting of 3 to 6 atoms, (heterocyclic group consisting of 3 to 6 atoms) -C1-2Alkylene radical, C6-10Aryl radical, C6-10aryl-C1-2Alkylene, heteroaryl of 5 to 10 atoms and (heteroaryl of 5 to 10 atoms) -C1-2Each alkylene is independently unsubstituted or substituted with 1, 2, 3 or 4 substituents, or-C (═ O) -Y1、-C(=O)-O-Y2、-(CH2)-O-(C=O)-Y1、-(CH2)-O-(C=O)-O-Y2、 -(CHCH2)-O-(C=O)-Y1And- (CHCH)3)-O-(C=O)-O-Y2
Y1Is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, mono-substituted amino group, di-substituted amino and-C (R)*)2NHR**(ii) a And each R*And R**Independently is hydrogen or optionally substituted C1-6An alkyl group;
Y2is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, monosubstituted amino groups.
Wherein R is1Selected from hydrogen, R2And R3When selected from deuterium, preferred structures are those of formula III below:
Figure BDA0002922605880000051
R4、R5、R6and R7Each independently selected from hydrogen or fluorine;
R8is hydrogen, an optionally substituted phosphate group, C1-4Alkyl radical, C2-4Alkenyl radical, C3-6Carbocyclyl group, (C)3-6Carbocyclyl) -C1-2Alkylene, heterocyclic group consisting of 3 to 6 atoms, (heterocyclic group consisting of 3 to 6 atoms) -C1-2Alkylene radical, C6-10Aryl radical, C6-10aryl-C1-2Alkylene, heteroaryl of 5 to 10 atoms, (heteroaryl of 5 to 10 atoms) -C1-2Alkylene, wherein said C1-4Alkyl radical, C2-4Alkenyl radical, C3-6Carbocyclyl, C3-6carbocyclyl-C1-2Alkylene, heterocyclic group consisting of 3 to 6 atoms, (heterocyclic group consisting of 3 to 6 atoms) -C1-2Alkylene radical, C6-10Aryl radical, C6-10aryl-C1-2Alkylene, heteroaryl of 5 to 10 atoms and (heteroaryl of 5 to 10 atoms) -C1-2Alkylene is each independently unsubstituted or substituted by 1, 2, 3 or 4Substituted by a substituent, or-C (═ O) -Y1、-C(=O)-O-Y2、-(CH2)-O-(C=O)-Y1、-(CH2)-O-(C=O)-O-Y2、 -(CHCH3)-O-(C=O)-Y1And- (CHCH)3)-O-(C=O)-O-Y2
Y1Is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, mono-substituted amino group, di-substituted amino and-C (R)*)2NHR**(ii) a And each R*And R**Independently is hydrogen or optionally substituted C1-6An alkyl group;
Y2is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclyl, monosubstituted amino groups.
The present invention provides a fused polycyclic pyridone derivative, and pharmaceutically acceptable salts, solvates, hydrates, N-oxides, polymorphs, co-crystals, tautomers, stereoisomers or mixtures or prodrugs thereof, pharmaceutical compositions and pharmaceutically acceptable carriers, diluents, excipients, or combinations thereof. The compounds according to the invention are preferably the following compounds:
Figure BDA0002922605880000061
Figure BDA0002922605880000071
in some embodiments of the invention, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier, adjuvant, vehicle, or combination thereof.
In some embodiments, the pharmaceutical compositions provided herein further comprise one or more additional therapeutic agents.
In still other embodiments, the additional therapeutic agent is selected from an anti-influenza virus agent or a vaccine.
In other embodiments, the pharmaceutical composition may be in a liquid, solid, semi-solid, gel or spray dosage form.
In still other embodiments, the pharmaceutical composition of the present invention may comprise one or more agents that inhibit replication of influenza virus infection selected from the group consisting of neuraminidase inhibitors, M2 protein inhibitors, polymerase inhibitors, PB2Inhibitors, immunomodulators.
In still other embodiments, the pharmaceutical composition of the invention, wherein the additional therapeutic agent is Amantadine (Amantadine), Rimantadine (Rimantadine), oseltamivir (0seltamivir), Zanamivir (Zanamivir), Peramivir (Peramivir), Laninamivir (Laninamivir), Laninamivir Octanoate (Laninamivir Octanoate), Favipiravir (Favipiravir), Arbidol (Arbidol), Ribavirin (ribarin), Beraprost (Beraprost), stafurelin, Ingavirin (Ingavirin), influenza enzyme (flase), drugs No. 1422050-75-6, JNJ-872, S-033188, influenza vaccines (e.g., FluMist, Quadrivalent), or combinations thereof.
In another aspect, the invention provides the use of the compound or the pharmaceutical composition for the manufacture of a medicament for the prevention, alleviation or treatment of a viral infectious disease in a patient.
In some embodiments, the viral infection is an influenza viral infection. The influenza virus is selected from influenza A, B, C or avian influenza (H5N1, H7N 9); the symptoms are selected from the group consisting of cold-like symptoms accompanied by fever, chill, headache, myalgia, general malaise, and the like, gastrointestinal symptoms accompanied by pharyngalgia, rhinorrhea, nasal obstruction, cough, respiratory inflammation with phlegm, abdominal pain, emesis, diarrhea, and complications accompanied by acute encephalopathy, pneumonia secondary infection, and combinations thereof.
In some further embodiments, the present invention provides the use of the compound or the pharmaceutical composition for the manufacture of a medicament for inhibiting RNA polymerase of influenza virus.
In some further embodiments, the present invention provides the use of the compound or the pharmaceutical composition in the manufacture of a medicament for the inhibition of cap-dependent endonuclease (CEN).
Unless otherwise indicated, the present invention includes all compounds of the present invention and pharmaceutically acceptable salts, solvates, including hydrates, polymorphs, prodrugs, co-crystals, tautomers, stereoisomers thereof.
In some embodiments, the salt refers to a pharmaceutically acceptable salt. The term "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal being treated therewith.
The compounds of the present invention also include the salt forms thereof, which are not necessarily pharmaceutically acceptable salts, but may be used for the preparation and/or purification of the compounds of the present invention and/or intermediates used in the isolation of isomers of the compounds of the present invention.
The compounds of the invention, including salts thereof, may also be obtained in the form of their hydrates or include other solvents used for their crystallization. The compounds of the present invention may form solvates, either inherently or by design, with pharmaceutically acceptable solvents (including water); thus, the invention also includes solvated and unsolvated forms thereof.
On the other hand, the compounds of the invention may contain several asymmetric centers or their racemic mixtures as generally described. The invention further comprises racemic mixtures, partial racemic mixtures and isolated enantiomers and diastereomers.
The compound of the present invention may exist in one of the possible isomers, rotamers, atropisomers, tautomers or mixtures thereof, and the present invention may further comprise the isomers, rotamers, atropisomers, tautomers or mixtures thereof, or partial mixtures or separated isomers, rotamers, atropisomers, tautomers of the compound of the present invention.
In another aspect, the invention relates to methods of preparation, isolation and purification of compounds encompassed by formula I.
The foregoing merely summarizes certain aspects of the invention and is not intended to be limiting. These and other aspects will be more fully described below.
Detailed Description
Definitions and general terms
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 this invention belongs and all patent publications cited throughout the disclosure of the present invention are hereby incorporated by reference in their entirety.
The articles "a," "an," and "the" as used herein are intended to include "at least one" or "one or more" unless otherwise indicated or clearly contradicted by context. Thus, as used herein, the articles refer to articles of one or more than one (i.e., at least one) object. For example, "a component" refers to one or more components, i.e., there may be more than one component contemplated for use or use in embodiments of the described embodiments.
The term "subject" as used herein refers to an animal. Typically the animal is a mammal. Subjects also refer to primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds, and the like. In certain embodiments, the subject is a primate. In still other embodiments, the subject is a human.
The terms "subject" and "patient" as used herein are used interchangeably. The terms "subject" and "patient" refer to animals (e.g., birds or mammals such as chickens, quails or turkeys), particularly "mammals" including non-primates (e.g., cows, pigs, horses, sheep, rabbits, guinea pigs, rats, cats, dogs, and mice) and primates (e.g., monkeys, chimpanzees, and humans), and more particularly humans. In one embodiment, the subject is a non-human animal, such as a farm animal (e.g., a horse, cow, pig, or sheep) or a pet (e.g., a dog, cat, guinea pig, or rabbit). In other embodiments, the "patient" refers to a human.
All stereoisomeric forms of the compounds of the present invention, including but not limited to diastereomers, enantiomers, and atropisomers (atropisomers) and mixtures thereof, such as racemic mixtures, are also included within the scope of the present invention. Many organic compounds exist in an optically active form, i.e., they have the ability to rotate the plane of plane polarized light. When describing optically active compounds, the prefixes D and L or R and S are used to denote the absolute configuration of the molecule with respect to the chiral center (or centers) in the molecule. The prefixes d and l or (+) and (-) are the symbols used to specify the rotation of plane polarized light by the compound, where (-) or l indicates that the compound is left-handed. Compounds prefixed with (+) or d are dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of each other. A particular stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often referred to as a mixture of enantiomers. A50: 50 mixture of enantiomers is referred to as a racemic mixture or racemate, which may occur when there is no stereoselectivity or stereospecificity in the chemical reaction or process.
Depending on the choice of starting materials and process, the compounds according to the invention may be present as one of the possible isomers or as a mixture thereof, for example as the pure optical isomer, or as a mixture of isomers, for example as a mixture of racemic and non-corresponding isomers, depending on the number of asymmetric carbon atoms. Optically active (R) -or (S) -isomers can be prepared using chiral synthons or chiral preparations, or resolved using conventional techniques. If the compound contains a double bond, the substituents may be in the E or Z configuration; if the compound contains a disubstituted cycloalkyl group, the substituents of the cycloalkyl group may be in the cis or trans (cis-or trans-) configuration.
The term "stereoisomers" refers to compounds having the same chemical structure, but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric isomers (cis/trans), atropisomers, and the like.
"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, aminoethanol. The term "hydrate" refers to an association of solvent molecules that is water.
As used herein, "pharmaceutically acceptable salts" refer to organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, salts of inorganic acids formed by reaction with amino groups such as hydrochlorides, hydrobromides, phosphates, sulfates, perchlorates, and salts of organic acids such as acetates, oxalates, maleates, tartrates, citrates, succinates, malonates, or those obtained by other methods described in the literature above, such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, cyclopentylpropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, palmitate, pamoate, pectate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, stearate, thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like. Salts obtained with a suitable base includeAlkali metal, alkaline earth metal, ammonium and N+(C1-4Alkyl radical)4A salt. The present invention also contemplates quaternary ammonium salts formed from compounds containing groups of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C1-8Sulfonates and aromatic sulfonates.
The term "prodrug", as used herein, represents a compound that is converted in vivo to a compound of formula (I). Such conversion is effected by hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue. The prodrug compound of the invention can be ester, and in the prior invention, the ester can be used as the prodrug and comprises phenyl ester and aliphatic (C)1-24) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, a compound of the present invention contains a hydroxy group, i.e., it can be acylated to provide the compound in prodrug form. Other prodrug forms include phosphate esters, such as those obtained by phosphorylation of a hydroxyl group on the parent.
Any asymmetric atom (e.g., carbon, etc.) of a compound of the invention can exist in racemic or enantiomerically enriched forms, such as the (R) -, (S) -, (R, R) -, (S, S) -, (S, R) -or (R, S) -configurations. In certain embodiments, each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R) -or (S) -configuration. The substituents on the atoms having an unsaturated double bond may be present in the- (Z) -or- (E) -form, if possible.
As described herein, the compounds of the present invention may be optionally substituted with one or more substituents, such asThe compounds of the above general formula, or as specified in the examples, subclasses, and classes of compounds encompassed by the present invention. It will be appreciated that the terms "optionally substituted" and "unsubstituted or substituted with … … substituents" are used interchangeably. The terms "optionally," "optional" or "optionally" mean that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. In general, the term "optionally" whether preceded by the term "substituted" or not, indicates that one or more hydrogen atoms in a given structure are unsubstituted or substituted with a particular substituent. Unless otherwise indicated, an optional substituent group may be substituted at each substitutable position of the group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, the substituents may be substituted at each position, identically or differently. Wherein the substituent can be, but is not limited to, F, Cl, Br, I, CN, N3、OH、NH2、NO2Oxo (═ O), C1-12Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Alkylamino radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-12Cycloalkyl radical, C3-12cycloalkyl-C1-4Alkylene radical, C3-12Carbocyclyl, C3-12carbocyclyl-C1-4Alkylene, heterocyclic group consisting of 3 to 12 atoms, (heterocyclic group consisting of 3 to 12 atoms) -C1-4Alkylene radical, C6-10Aryl radical, C6-10aryl-C1-4Alkylene, heteroaryl of 5 to 16 atoms or (heteroaryl of 5 to 16 atoms) -C1-4An alkylene group.
In the various parts of this specification, substituents of the disclosed compounds are disclosed in terms of group type or range. It is specifically intended that the invention includes each and every independent subcombination of the various members of these groups and ranges. For example, the term "C1-6Alkyl "in particular denotes independently disclosed methyl, ethyl, C3Alkyl radical, C4Alkyl radical, C5Alkyl and C6An alkyl group; the term "heteroaryl of 5 to 10 atoms" refers in particular to heteroaryl of 5 atoms, heteroaryl of 6 atoms, heteroaryl of 7 atoms, heteroaryl of 8 atoms, heteroaryl of 9 atoms and heteroaryl of 10 atoms, which are independently disclosed.
The term "alkyl" as used herein, denotes a saturated straight or branched chain monovalent hydrocarbon radical containing from 1 to 20 carbon atoms. 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-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, n-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2, 3-dimethyl-2-butyl, 3, 3-dimethyl-2-butyl, n-heptyl, n-octyl, and the like, wherein the alkyl groups can be independently unsubstituted or substituted with one or more substituents described herein.
The term "alkyl" and its prefix "alk", as used herein, are intended to encompass both straight and branched saturated carbon chains.
The term "cycloalkyl" refers to a monocyclic, bicyclic, or tricyclic ring system containing 3-6 ring carbon atoms that is saturated, having one or more points of attachment to the rest of the molecule. In some embodiments, cycloalkyl is a ring system containing 3 to 6 ring carbon atoms, e.g., C3-6A cycloalkyl group; in some embodiments, cycloalkyl is a ring system containing 5 to 6 ring carbon atoms, e.g., C5-6A cycloalkyl group; examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like, and the cycloalkyl groups can be independently unsubstituted or substituted with one or more substituents described herein.
The term "heterocyclyl" may be used alone or as a majority of "heterocyclylalkyl" or "heterocyclylalkoxy" and refers to a saturated or partially unsaturated, non-aromatic, monocyclic, bicyclic, or tricyclic ring system containing 3 to 12 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur, and oxygen, wherein the heterocyclyl is non-aromatic and does not contain any aromatic rings, and wherein the ring system has one or more attachment points to the rest of the molecule. The terms "heterocyclyl" and "heterocycle" are used interchangeably herein. The term "heterocyclyl" includes monocyclic, bicyclic or polycyclic fused, spiro or bridged heterocyclic ring systems. Bicyclic heterocyclic groups include bridged bicyclic heterocyclic groups, fused bicyclic heterocyclic groups, and spiro bicyclic heterocyclic groups. In some embodiments, heterocyclyl is a ring system of 3-8 ring atoms; in other embodiments, heterocyclyl is a ring system of 3-6 ring atoms; in other embodiments, heterocyclyl is a ring system of 5-7 ring atoms; in other embodiments, heterocyclyl is a ring system of 5-8 ring atoms; in other embodiments, heterocyclyl is a ring system of 6-8 ring atoms; in other embodiments, heterocyclyl is a ring system of 5-6 ring atoms; in other embodiments, heterocyclyl is a ring system of 4 ring atoms; in other embodiments, heterocyclyl is a ring system of 5 ring atoms; in other embodiments, heterocyclyl is a ring system of 6 ring atoms; in other embodiments, heterocyclyl is a ring system of 7 ring atoms; in other embodiments, heterocyclyl is a ring system of 8 ring atoms.
Examples of heterocyclyl groups include, but are not limited to: oxirane, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiocyclopentyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiaoxanyl, homopiperazinyl, homopiperidinyl, oxepanyl, thietanyl. In heterocyclic radicals of-CH2The radicals being substituted by-C (═ O) -Examples of generations include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidinonyl, 3, 5-dioxopiperidinyl, pyrimidinedione. Examples of heterocyclic sulfur atoms that are oxidized include, but are not limited to, sulfolane and 1, 1-dioxothiomorpholinyl. Bridging heterocyclyl groups include, but are not limited to, 2-oxabicyclo [2.2.2]Octyl, 1-azabicyclo [2.2.2]Octyl, 3-azabicyclo [3.2.1]Octyl, and the like. The heterocyclyl group may be optionally substituted with one or more substituents as described herein.
The term "aryl" may be used alone or as a majority of "arylalkyl" or "arylalkoxy" and refers to monocyclic, bicyclic, and tricyclic aromatic carbocyclic ring systems containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring system is aromatic, each ring contains 3 to 7 ring atoms, and one or more attachment points are attached to the rest of the molecule. The term "aryl" may be used interchangeably with the terms "aromatic ring" or "aromatic ring", e.g., aryl may include phenyl, naphthyl and anthracenyl. The aryl group can be independently unsubstituted or substituted with one or more substituents described herein.
The term "heteroaryl" may be used alone or as a majority of "heteroarylalkyl" or "heteroarylalkoxy" and denotes monocyclic, bicyclic and tricyclic aromatic systems containing 5 to 16 ring atoms, or 5 to 14 ring atoms, or 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 8 ring atoms, or 5 to 7 ring atoms, or 5 to 6 ring atoms, wherein at least one ring system is aromatic and at least one ring contains one or more heteroatoms, wherein each ring contains 5 to 7 ring atoms, and wherein the heteroaryl group has one or more attachment points to the rest of the molecule. when-CH is present in the heteroaryl group2When it is a group, -CH2-the group may optionally be replaced by-C (═ O) -. Unless otherwise indicated, the heteroaryl group may be attached to the rest of the molecule (e.g., C in CH, or N in NH) via any reasonable site (e.g., C in CH, or N in NH)Host structure as in formula (xxxvii). The term "heteroaryl" may be used interchangeably with the terms "heteroaromatic ring" or "heteroaromatic compound". In some embodiments, heteroaryl is 5-14 atom composed of 1, 2, 3, or 4 heteroatoms independently selected from O, S, and N. In other embodiments, heteroaryl is a heteroaryl consisting of 5 to 12 atoms containing 1, 2, 3, or 4 heteroatoms independently selected from O, S, and N; in other embodiments, heteroaryl is a heteroaryl consisting of 5 to 10 atoms containing 1, 2, 3, or 4 heteroatoms independently selected from O, S, and N; in other embodiments, heteroaryl is a heteroaryl consisting of 5 to 8 atoms containing 1, 2, 3, or 4 heteroatoms independently selected from O, S, and N; in other embodiments, heteroaryl is a heteroaryl consisting of 5 to 7 atoms containing 1, 2, 3, or 4 heteroatoms independently selected from O, S, and N; in other embodiments, heteroaryl is a heteroaryl consisting of 5 to 6 atoms containing 1, 2, 3, or 4 heteroatoms independently selected from O, S, and N; in other embodiments, heteroaryl is a heteroaryl consisting of 5 atoms containing 1, 2, 3, or 4 heteroatoms independently selected from O, S, and N; in other embodiments, heteroaryl is a heteroaryl consisting of 6 atoms containing 1, 2, 3, or 4 heteroatoms independently selected from O, S, and N.
In other embodiments, heteroaryl includes, but is not limited to, the following monocyclic groups: 2-furyl group, 3-furyl group, N-imidazolyl group, 2-imidazolyl group, 4-imidazolyl group, 5-imidazolyl group, 3-isoxazolyl group, 4-isoxazolyl group, 5-isoxazolyl group, 2-oxazolyl group, 4-oxazolyl group, 5-oxazolyl group, N-pyrrolyl group, 2-pyrrolyl group, 3-pyrrolyl group, 2-pyridyl group, 3-pyridyl group, 4-pyridyl group, 2-pyrimidinyl group, 4-pyrimidinyl group, 5-pyrimidinyl group, pyridazinyl group (e.g., 3-pyridazinyl group), 2-thiazolyl group, 4-thiazolyl group, 5-thiazolyl group, tetrazolyl group (e.g., 5H-tetrazolyl group, 2H-tetrazolyl group), triazolyl group (e.g., 2-triazolyl group, 5-triazolyl group, 4H-1, 2, 4-triazolyl, 1H-1, 2, 4-triazolyl, 1, 2, 3-triazolyl), 2-thienyl, 3-thienyl, pyrazolyl (e.g. 2-pyrazolyl and 3-pyrazolyl), isothiazolyl, 1, 2, 3-oxadiazolyl, 1, 2, 5-oxadiazolyl, 1, 2, 4-oxadiazolyl, 1, 3, 4-oxadiazolyl, 1, 2, 3-thiadiazolyl, 1, 3, 4-thiadiazolyl, 1, 2, 5-thiadiazolyl, pyrazinyl, 1, 3, 5-triazinyl; the following bi-or tricyclic groups are also included, but are in no way limited to these groups: indolinyl, 1, 2, 3, 4-tetrahydroisoquinolinyl, 4, 5, 6, 7-tetrahydrobenzofuranyl, benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl), purinyl, quinolinyl (e.g., 2-quinolinyl, 3-quinolinyl, 4-quinolinyl), isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl, or 4-isoquinolinyl), phenoxathiyl, dibenzoimidazolyl, dibenzofuranyl, dibenzothienyl, said heteroaryl group optionally being substituted with one or more substituents as described herein.
As used herein, the term "pharmaceutically acceptable carrier" includes any solvent, dispersion medium, coating, surfactant, antioxidant, preservative (e.g., antibacterial, antifungal), isotonic agent, salt, pharmaceutical stabilizer, binder, excipient, dispersant, lubricant, sweetener, flavoring agent, coloring agent, or combination thereof, which are known to those skilled in the art. Except insofar as any conventional carrier is incompatible with the active ingredient, its use in therapeutic or pharmaceutical compositions is contemplated.
The term "effective amount" of a compound of the invention refers to an amount that elicits the desired biological response. In the present invention, the biological response is expected to be inhibition of influenza virus replication, reduction in the amount of influenza virus or reduction or amelioration of the severity, duration, progression or onset of influenza virus infection, prevention of spread of influenza virus infection, prevention of recurrence, evolution, onset or progression of symptoms associated with influenza virus infection, or enhancement of the prophylactic or therapeutic effect of another anti-influenza infection therapy used. The exact amount of the compound to be administered to a subject will depend on the mode of administration, the type and severity of the infection and the characteristics of the subject, such as health, age, sex, weight and tolerance to drugs. The skilled artisan will be able to determine the appropriate dosage based on these and other factors. When administered in combination with other antiviral agents, such as anti-influenza drugs, the "effective amount" of the second agent will depend on the type of drug used. Suitable dosages of approved agents are known and can be adjusted by the skilled artisan according to the condition of the subject, the type of condition being treated and the amount of the compound of the invention being used. In the case where amounts are not explicitly specified, an effective amount should be taken. For example, a compound of the invention may be administered to a subject at a dosage in the range of about 0.01-100 mg/body weight/day for therapeutic or prophylactic treatment.
The term "treatment" as used herein refers to both therapeutic and prophylactic treatment. For example, therapeutic treatment includes reducing or ameliorating the progression, severity, and/or duration of an influenza virus-mediated condition, or ameliorating one or more symptoms (particularly, one or more discernible symptoms) of an influenza virus-mediated condition as a result of administration of one or more therapies (e.g., one or more therapeutic agents (e.g., compounds and compositions of the invention)) The number of days they were ill was reduced.
In other embodiments, the present invention relates to a compound, but is in no way limited to, one of the following, or a stereoisomer, tautomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof: (specific Compound)
In another aspect, the invention provides a pharmaceutical composition comprising an effective amount of a compound of the invention.
In some embodiments of the invention, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier, adjuvant, vehicle, or combination thereof.
In some embodiments, the pharmaceutical compositions provided herein further comprise a second agent.
In still other embodiments, the second agent is one or more agents that inhibit replication of influenza virus infection selected from the group consisting of neuraminidase inhibitors, M2 protein inhibitors, polymerase inhibitors, PB2 inhibitors, zanamivir, oseltamivir, peramivir, laninamivir octanoate, faviravir, immunomodulators, beraprost, ribavirin.
In another aspect, the invention provides the use of the compound or the pharmaceutical composition for the manufacture of a medicament for the prevention, treatment or alleviation of a viral infectious disease in a patient.
In some embodiments, the viral infection is an influenza viral infection.
In some further embodiments, the present invention provides the use of the compound or the pharmaceutical composition for the manufacture of a medicament for inhibiting RNA polymerase of influenza virus.
In some further embodiments, the present invention provides the use of the compound or the pharmaceutical composition in the manufacture of a medicament for the inhibition of cap-dependent endonuclease (CEN).
In some embodiments, the salt refers to a pharmaceutically acceptable salt. The term "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal being treated therewith.
The compounds of the present invention also include other salts of such compounds, which are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for the preparation and/or purification of the compounds of the present invention and/or for the isolation of enantiomers of the compounds of the present invention.
Pharmaceutically acceptable acid addition salts may be formed with inorganic and organic acids, for example, acetate, aspartate, benzoate, benzenesulfonate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlorotheophylline, citrate, edisylate, fumarate, glucoheptonate, gluconate, glucuronate, hippurate, hydroiodide, isethionate, lactate, lactobionate, lauryl sulfate, malate, maleate, malonate, mandelate, methanesulfonate, methylsulfate, naphthoate, naphthalenesulfonate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/biphosphate/dihydrogen phosphate, dihydrogenphosphate, and the like, Polysilonolactates, propionates, stearates, succinates, sulfosalicylates, tartrates, tosylates and trifluoroacetates.
Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, sulfosalicylic acid, and the like.
Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
Inorganic bases from which salts can be derived include, for example, ammonium salts and metals of groups I to XII of the periodic table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
Organic bases from which salts can be derived include primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Certain organic amines include, for example, isopropylamine, benzathine, choline salts, diethanolamine, diethylamine, lysine, meglumine, piperazine, and tromethamine.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, basic or acidic moiety, by conventional chemical methods. In general, such salts can be prepared by reacting the free acid forms of these compounds with a stoichiometric amount of the appropriate base (e.g., Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, etc.), or by reacting the free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are usually carried out in water or an organic solvent or a mixture of both. Generally, where appropriate, it is desirable to use a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile.
Furthermore, the compounds of the present invention, including salts thereof, may also be obtained in the form of their hydrates or include other solvents used for their crystallization. The compounds of the present invention may form solvates, either inherently or by design, with pharmaceutically acceptable solvents (including water); thus, the present invention is intended to include both solvated and unsolvated forms.
Compositions, formulations and administration of the compounds of the invention
The invention provides a pharmaceutical composition which comprises a compound shown as a formula (I), a formula (I-a) or a formula (I-b) or a stereoisomer, a racemic or non-racemic mixture of isomers or a pharmaceutically acceptable salt or solvate thereof. The pharmaceutical composition further comprises at least one pharmaceutically acceptable carrier, diluent, adjuvant or vehicle, and optionally, other therapeutic and/or prophylactic ingredients. In some embodiments, the pharmaceutical composition comprises an effective amount of at least one pharmaceutically acceptable carrier, diluent, adjuvant, or vehicle.
Pharmaceutically acceptable carriers may contain inert ingredients that do not unduly inhibit the biological activity of the compound. The pharmaceutically acceptable carrier should be biocompatible, e.g., non-toxic, non-inflammatory, non-immunogenic, or free of other adverse or side effects once administered to a patient. Standard pharmaceutical techniques may be employed.
The pharmaceutical composition or pharmaceutically acceptable composition of the present invention further comprises a pharmaceutically acceptable carrier, adjuvant or excipient, as described herein, including any solvent, diluent, liquid excipient, dispersant, suspending agent, surfactant, isotonic agent, thickener, emulsifier, preservative, solid binder or lubricant, and the like, as appropriate for the particular intended dosage form, as used herein. In addition to conventional carrier vehicles which are incompatible with the compounds of the present invention, e.g., may produce undesirable biological effects or may deleteriously interact with any other component of the pharmaceutically acceptable composition, any other conventional carrier vehicle and its use are contemplated by the present invention.
Some examples of substances that may be used as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (e.g., human serum albumin), buffer substances (e.g., tween 80, phosphate, glycine, sorbic acid, or potassium sorbate), partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (e.g., protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, or zinc salts), silica gel, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block copolymers, methylcellulose, hydroxypropylmethylcellulose, lanolin, sugars (e.g., lactose, glucose, and sucrose), starches (e.g., corn starch and potato starch), celluloses and derivatives thereof (e.g., sodium carboxymethylcellulose, ethylcellulose, and cellulose acetate), Powdered tragacanth, malt, gelatin, talc, excipients (such as cocoa butter and suppository waxes), oils (such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil), glycols (such as propylene glycol or polyethylene glycol), esters (such as ethyl oleate and ethyl dodecanoate), agar, buffers (such as magnesium hydroxide and aluminum hydroxide), alginic acid, pyrogen-free water, isotonic saline, Ringer's solution, ethanol and phosphate buffers, as well as other non-toxic compatible lubricants (such as sodium lauryl sulfate and magnesium stearate), as well as coloring agents, detackifiers, coating agents, sweetening and flavoring agents, preservatives and antioxidants, according to the judgment of the formulator, may also be present in the composition.
The compounds or compositions of the present invention may be administered by any suitable means, and the above-described compounds and pharmaceutically acceptable compositions may be administered to humans or other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally as an oral or nasal spray, and the like, depending on the severity of the infection being treated.
Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. In addition to inert diluents, oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Injectable preparations may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents, for example sterile injectable aqueous or oily suspensions. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, ringer's solution, u.s.p. and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. In addition, fatty acids, such as octadecenoic acid, are used in the preparation of injections.
For example, injectable formulations can be sterilized by filtration through a bacteria retaining filter or by the addition of a sterilizing agent in the form of a sterile solid composition which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
To prolong the effect of the compounds or compositions of the present invention, it is often desirable to slow the absorption of the compounds from subcutaneous or intramuscular injection. This can be achieved by using a liquid suspension of crystalline or amorphous material which is poorly water soluble. The rate of absorption of the compound then depends on its rate of dissolution, which in turn depends on crystal size and crystal form. Alternatively, delayed absorption of the parenterally administered compound is achieved by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming a microcapsule matrix of the compound in a biodegradable polymer such as polylactide-polyglycolic acid. Depending on the ratio of compound to polymer and the nature of the particular polymer employed, the rate of release of the compound can be controlled. Examples of other biodegradable polymers include polyorthoesters and polyanhydrides. Depot injectable formulations can also be prepared by entrapping the compound in liposomes or microemulsions which are compatible with body tissues.
Oral solid dosage forms include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders, such as carboxymethylcellulose, alginates, gels, polyvinylpyrrolidone, sucrose, and acacia, c) humectants, such as glycerol, d) disintegrating agents, such as agar- -agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents, such as paraffin, f) absorption accelerators, such as quaternary ammonium compounds, g) wetting agents, such as cetyl alcohol and glycerol monostearate, h) absorbents, such as kaolin and bentonite, and i) lubricants, such as talc, calcium stearate, sodium stearate, and sodium stearate, magnesium, Magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid compositions of a similar type may also be employed as fillers in soft and hard gelatin capsules using excipients such as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. Solid dosage forms of tablets, dragees, capsules, pills and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical art. They may optionally contain opacifying agents and may also have the properties of a composition such that the active ingredient is released only, optionally in a delayed manner, or preferably, in a certain part of the intestinal tract. Examples of embedding compositions that can be used include polymers and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard gelatin capsules using excipients such as lactose or milk sugar as well as high molecular weight polyethylene glycols.
The active compound may also be in the form of a microencapsulated form with one or more of the above-mentioned excipients. Solid dosage forms of tablets, dragees, capsules, pills and granules can be prepared with coatings and shells such as enteric coatings, controlled release coatings and other coatings well known in the pharmaceutical art. In such solid dosage forms, the active compound may be mixed with at least one inert diluent, for example sucrose, lactose or starch. In general, such dosage forms may also contain additional substances in addition to the inert diluents, such as tableting lubricants and other tableting aids, for example magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and may also have the properties of a composition such that the active ingredient is released only, optionally in a delayed manner, or preferably, in a certain part of the intestinal tract. Examples of embedding compositions that can be used include polymers and waxes.
Formulations for topical or transdermal administration of the compounds of the present invention include ointments, salves, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. Under sterile conditions, the active compound is combined with a pharmaceutically acceptable carrier and any required preservatives or buffers that may be required. Ophthalmic formulations, ear drops and eye drops are also contemplated within the scope of the present invention. In addition, the present invention contemplates the use of a dermal patch that has the added advantage of providing controlled delivery of the compound to the body. Such dosage forms may be made by dissolving or dispersing the compound in the appropriate medium. Absorption enhancers may also be used to increase the flux of the compound through the skin. The rate can be controlled by providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
The compositions of the present invention may also be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted kit. The term "parenteral" as used herein includes, but is not limited to, subcutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. In particular, the compositions are administered orally, intraperitoneally, or intravenously.
The sterile injectable form of the composition of the invention may be an aqueous or oily suspension. These suspensions may be prepared using suitable dispersing or wetting agents and suspending agents following techniques known in the art. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. In addition, as natural pharmaceutically acceptable oils, such as olive oil or castor oil, especially in polyoxyethylated form, fatty acids, such as octadecenoic acid and its glyceride derivatives are used for the preparation of injections. These oil solutions or suspensions may also contain a long chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents commonly used in formulating pharmaceutically acceptable dosage forms, including emulsions and suspensions. Other commonly used surfactants such as Tweens, Spans, and other emulsifiers or bioavailability enhancers commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for formulation purposes.
The pharmaceutical compositions of the present invention may be administered orally in any orally acceptable dosage form, including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral administration, carriers that are commonly used include, but are not limited to, lactose and starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are required for oral administration, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
For topical application, the pharmaceutical compositions may be formulated as a suitable ointment containing the active ingredient suspended or dissolved in one or more carriers. Suitable carriers for topical application of the compounds of the present invention include, but are not limited to, mineral oil, petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutical compositions may be formulated as a suitable lotion or cream containing the active ingredient suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
For ophthalmic use, the pharmaceutical compositions may be formulated as micronized suspensions in isotonic pH adjusted sterile saline, or solutions in isotonic pH adjusted sterile saline in particular, with or without preservatives such as benzalkonium chloride. Alternatively, for ophthalmic use, the pharmaceutical composition may be formulated as an ointment, such as petrolatum.
The pharmaceutical compositions may also be administered by nasal aerosol spray or inhalation. Such compositions are prepared according to techniques well known in the pharmaceutical art and are prepared as solutions in saline using benzyl alcohol and other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons and/or other conventional solubilizing or dispersing agents.
The compounds for use in the methods of the invention may be formulated in unit dosage form. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for subjects, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, optionally in association with a suitable pharmaceutical carrier. The unit dosage form can be administered in a single daily dose or in multiple daily doses (e.g., about 1-4 or more times per day). When multiple daily doses are used, the unit dosage form for each dose may be the same or different.
In summary, the present invention provides the use of said compound or said pharmaceutical composition for the manufacture of a medicament for the inhibition of cap-dependent endonuclease (CEN). The compound of the invention is suitable for being prepared into medicines with various dosage forms, and can be widely used for treating seasonal influenza, avian influenza, swine influenza and influenza virus mutant strains with drug resistance to tamiflu.
In addition to being beneficial for human therapy, the compounds and pharmaceutical compositions of the present invention may also find application in veterinary therapy for pets, animals of the introduced species and mammals in farm animals. Examples of other animals include horses, dogs, and cats. Herein, the compound of the present invention includes pharmaceutically acceptable derivatives thereof.
Advantages of the compounds of the invention: compared with the existing similar compounds, the compound of the invention has larger difference in structure, not only can well inhibit influenza virus, but also has lower cytotoxicity, and more excellent in vivo pharmacokinetic property and in vivo pharmacodynamic property. In addition, compared with the existing similar compounds, the compound provided by the invention has better drugability.
Detailed description of the preferred embodimentsthe following examples are set forth to illustrate the invention. It is to be understood that the invention is not limited to these embodiments, but is provided as a means of practicing the invention. Modifications, changes, variations, etc. made within the scope of the present invention are within the scope of the present invention.
In this specification, a structure is dominant if there is any difference between the chemical name and the chemical structure.
In general, the compounds of the present invention may be prepared by the methods described herein, wherein the substituents are as defined in formula I, unless otherwise specified. The following reaction schemes and examples serve to further illustrate the context of the invention. The following examples are intended to illustrate the invention but are not intended to limit the scope of the invention.
Preparation examples
The compounds provided herein can be prepared from readily available starting materials using the procedures of the particular synthetic schemes set forth below, which will be well known to those skilled in the art. Experimental procedures, under which specific conditions are not noted in the following examples, can be determined by those skilled in the art through routine optimization procedures, according to conventional methods and conditions. In the following preparation examples, the inventors described in detail the preparation of the compounds of the present invention by taking some of the compounds of the present invention as examples.
In the following examples, abbreviations are explained:
Boc2o: di-tert-butyl dicarbonate;
DIEA: n, N-diisopropylethylamine
Pd (OAc) 2: palladium acetate
Pd (PPh3) 4: tetrakis (triphenylphosphine) palladium
Pd (Pd (PPh3)2Cl 2: bis (phenylphosphoryl) dichloropalladium
Pd/C: palladium carbon catalyst
KOtBu: potassium tert-butoxide
NaOtBu: sodium tert-butoxide
SEMCl: 2- (trimethylsilyl) ethoxymethyl chloride
NCS: n-chlorosuccinamides
NBS: n-bromosuccinamide
NIS: n-iodosuccinamides
TMSOTf: trimethylsilane trifluoromethanesulfonate
TFA: trifluoroacetic acid
n-BuLi: butyl lithium
T3P: 1-propylphosphoric acid cyclic anhydride
SnCl 4: tin tetrachloride
LiCl: lithium chloride
PE: petroleum ether;
EtOAc: ethyl acetate;
DMF: n, N-dimethylformamide;
MeCN: acetonitrile
DCM: dichloromethane;
THF: tetrahydrofuran (THF)
MeOH: methanol
Na2CO 3: sodium carbonate
Prep-HPLC: high pressure preparative liquid chromatography
Rf: a ratio shift value;
g; keke (Chinese character of 'Keke')
mg: milligrams of
h: hour(s)
rt: at room temperature
mol: mole of
mmol: millimole
mL: milliliter (ml)
M: mole/liter
Synthesis of intermediates
10-fluoro-6, 11-dihydrodibenzo [ b, e ] thiazepin-11-deuterium-11-hydroxy (compounds 1-16)
The synthetic route is as follows:
Figure BDA0002922605880000221
step 1: compound 1-1' (1.82g, 10mmol) was dissolved in 1, 2-dichloroethane (25 mL). N-bromosuccinimide (1.96g, 11mmol) and azobisisobutyronitrile (164.2mg, 1mmol) were added in this order, and the mixture was heated and stirred in an oil bath at 90 ℃ for 6 hours. Cooling to room temperature, filtering, and evaporating the filtrate under reduced pressure. Purification by silica gel column chromatography (petroleum ether/ethyl acetate (V/V) ═ 20/1) gave compound 1-2' (2.13g, yield: 81.7%) as a colorless liquid. LCMS (ESI) M/z 247.05[ M + H ] +.
Step 2: compound 1-2 '((1.35 g, 5.17mmol) was dissolved in DMF (10mL), sodium thiophenolate (820 mg, 6.20mmol) and potassium carbonate (1.43g, 10.34mmol) were added, stirred at room temperature, reacted overnight, ethyl acetate 60mL and water 60mL were added, shaken well to separate the organic layer, the aqueous layer was extracted with ethyl acetate (30mL × 2), the combined organic phases were washed with saturated brine (60mL × 2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give a yellow liquid, which was purified by silica gel column chromatography (petroleum ether/ethyl acetate (V/V) ═ 40/1) to give compound 1-3' (1.13g, yield: 75.3%. lcms (esi) M/z 277.07[ M + H ] +.
And step 3: dissolving compound 1-3 '((1.13 g, 3.89mmol) in methanol (8mL), adding tetrahydrofuran (16mL), adding dropwise sodium hydroxide solution (prepared by dissolving sodium hydroxide (1.62g, 40.44mmol) in water (16 mL)), stirring at room temperature for 4 hours, evaporating the organic solvent under reduced pressure, adjusting the pH to 7 with 2N HCl solution, adjusting the pH to 3-4 with 2N HCl solution, extracting with ethyl acetate (30 mL. times.3), combining the organic phases, washing with saturated brine (50 mL. times.2), drying over anhydrous sodium sulfate, filtering, and concentrating the filtrate under reduced pressure to obtain compound 1-4' (1.0g, yield: 97.9%) as a yellow solid, LCMS (ESI) M/z 263.10M + H ] +.
mg, 2.14mmol) and polyphosphoric acid (18g) were mixed well and heated in a 120 ℃ oil bath for 4 hours. After cooling to room temperature, 100mL of ice-water was added, and the mixture was stirred to completely dissolve the viscous substance, followed by extraction with ethyl acetate (30 mL. times.3). The organic phases were combined, washed successively with a saturated sodium carbonate solution (50 mL. times.2) and a saturated brine (50 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate (V/V) ═ 20/1) to give 1-5' (nuclear magnetic confirmed structure) of the compound 1-5 mg as a white solid, in yield: 82.7 percent. LCMS (ESI) M/z 245.08[ M + H ]]+;1H NMR (500MHz,CDCl3)δ(ppm)8.24(dd,J=8.0,1.0Hz,1H),7.67(dd,J=8.0,6.0 Hz,1H),7.40-7.34(m,2H),7.30-7.27(m,1H),7.04(td,J=8.5,2.5Hz,1H), 6.92(dd,J=9.0,2.5Hz,1H),4.02(s,2H).
And 5: dissolving compound 1-5 ' (compound 1-10 ' (450mg, 1.65mmol) in methanol (5mL), stirring at-5 deg.C for 0.5 hr, adding sodium deuteroborohydride (0.15g, 3.80mmol) into the reaction in 3 times, continuing to stir at-5 deg.C for 0.5 hr, moving to room temperature, stirring for 2 hr, adding water (40mL), continuing to stir for 1 hr, filtering under reduced pressure, and vacuum drying the filter cake to obtain white solid 1-11 ' 420mg, 93% yield, LCMS (ESI) M/z 248.10[ M + H ], [ M]+;1H NMR(400MHz,CDCl3)δ(ppm)7.45-7.48(m,1H),7.00-7.20(m,6H),6.30(d,J=3.2Hz, 1H),4.20(d,J=3.6Hz,1H),4.12(d,J=3.6Hz,1H).
1-amino-3- (benzyloxy) -4-one-1, 4-dihydropyridine-2-carboxylic acid ethyl ester (Compound 1-12)
The synthetic route is as follows:
Figure BDA0002922605880000241
step 1: containing chemical compoundsA solution of substance 1-7' (200.0g), iodoethane (228.0g), DBU (185.5g) in DMF (1000mL) was stirred at 20-30 ℃ for 60min, TLC (PE: EA ═ 1: 2) showed completion of the reaction; the reaction mixture was poured into 4L of water and extracted with 3x1000mL ethyl acetate (1000mLX 3); the organic layer was washed with 2 × 1000mL brine (1000mLX2) and concentrated to give compound 1-8' (190.0g) as a yellow oil. LCMS (ESI) M/z 275.1[ M + H ]]+;1H NMR:(400MHz, CDCl3)δ7.74(d,J=5.6Hz,1H),7.51-7.44(m,2H),7.39-7.29(m,3H),6.46 (d,J=5.6Hz,1H),5.36-5.23(m,2H),4.33(q,J=7.2Hz,2H),1.31(t,J=7.1 Hz,3H)。
Step 2: a DMA (2200mL) solution of compound 1-8 '(190.0 g), tert-butylcarbazole (137.3g), PPTS (522.3g) was stirred at 50-60 ℃ for 60min, TLC (PE: EA ═ 1: 2) showed complete reaction, the reaction solution was poured into 10L of water, extracted with 3x1000mL ethyl acetate (1000mLX3), washed with 3x1000mL brine (500mLX3), concentrated, and the residue was purified by column chromatography to give compound 1-9' (120g, 44.6% yield) as a yellow oil. LCMS (ESI) M/z 389.2[ M + H ]] +;1H NMR:(400MHz,CDCl3)δ7.37-7.31(m,2H),7.29-7.17(m,4H),6.29(d, J=7.9Hz,1H),5.17(s,2H),4.18(q,J=7.1Hz,2H),1.37(s,9H),1.18-1.13 (m,3H)。
And step 3: to a solution of the compound 1-9' (120.0g) in ethyl acetate (200mL) was added dropwise a hydrogen chloride-ethyl acetate solution (4M, 500mL) at 20-30 ℃; stirring the reaction liquid at 20-30 ℃ for 2 h; TLC (PE: EA 1: 1) showed complete reaction; concentrating the reaction solution; na for concentrated solution2CO3The solution (200mL) was adjusted to pH 8, extracted with 3x200mL ethyl acetate (200mLX3), the organic layer collected and concentrated to give compound 1-12(108.0g, crop) as a yellow oil. LCMS (ESI) M/z 289.1[ M + H ]] +;1H NMR:(400MHz,DMSO-d6) δ 7.37-7.30(m, 2H), 7.29-7.15(m, 5H), 6.30(d, J ═ 7.9Hz, 1H), 5.18(s, 2H), 4.18(q, J ═ 7.1Hz, 2H), 1.18-1.13(m, 3H). 7, 8-difluoro-6, 11-dihydrodibenzo [ b, e ]]Thiazone-11-deuterium-11-hydroxy (compound 1-11')
The synthetic route is as follows:
Figure BDA0002922605880000242
dissolving compound 1-10 '(1.0 g, 3.81mmol) in methanol (10mL), stirring at-5 ℃ for 0.5 h, adding sodium deuteroborohydride (0.30g, 7.62mmol) in 5 times to the reaction, continuing to stir at-5 ℃ for 0.5 h after the addition is finished, moving to room temperature and stirring for 3h, adding water (40mL), continuing to stir for 1h, filtering under reduced pressure, and drying the filter cake in vacuum to obtain white solid 1-11' (nuclear magnetic structure confirmation) 0.92g, yield: 91.5 percent. LCMS (ESI) M/z 367.4[ M + H ]]+;1H NMR (400MHz,CDCl3)δ(ppm)7.45-7.48(m,1H),7.15-7.20(m,2H),7.10-7.16(m,2H), 7.00-7.05(m,1H),6.10(d,J=3.2Hz,1H),4.20(d,J=14.4Hz,1H),2.72(d,J= 3.6Hz,1H).
The synthetic route of the intermediate 3-2 is as follows:
Figure BDA0002922605880000251
intermediate 3-1 was obtained from Nanjing Leizhong pharmaceutical science and technology, Inc., and was also prepared by the method described in Rajsner et al, Collectionof Czechosvak Chemical Communications, 1987, 47, 65-71. LCMS (ESI) M/z 246.1[ M + H ] +
Dissolving the intermediate 3-1(2.0g, 7.6mmol) in methanol (20mL), stirring at-5 ℃ for 0.5 hour, adding sodium deuteroborohydride (0.60g, 15.2mmol) into the reaction by 5 times, continuing to stir at-5 ℃ for 0.5 hour after the addition is finished, moving to room temperature, stirring for 4 hours, adding water (40mL), continuing to stir for 1 hour, filtering under reduced pressure, and drying a filter cake in vacuum to obtain 1.9g of a white solid 3-2 (nuclear magnetic confirmed structure), wherein the yield is as follows: 90 percent. LCMS (ESI) M/z 248.1[ M + H ]]+;1H NMR(400MHz, CDCl3)δ(ppm)7.45-7.48(m,1H),7.15-7.20(m,2H),7.10-7.16(m,2H),7.00-7.05 (m,1H),6.10(d,J=3.2Hz,1H).
The synthetic route of the intermediate 5-1 is as follows:
Figure BDA0002922605880000252
dissolving the intermediate 3-1(2.0g, 7.6mmol) in methanol (20mL), stirring at-5 ℃ for 0.5 hour, adding sodium deuteroborohydride (0.60g, 15.2mmol) into the reaction by 5 times, continuing to stir at-5 ℃ for 0.5 hour after the addition is finished, moving to room temperature, stirring for 4 hours, adding water (40mL), continuing to stir for 1 hour, filtering under reduced pressure, and drying a filter cake in vacuum to obtain 1.85g of a white solid intermediate 5-1 (nuclear magnetic confirmed structure), wherein the yield is as follows: 88 percent. LCMS (ESI) M/z 250.1[ M + H ]]+;1H NMR (400MHz,CDCl3)δ(ppm)7.45-7.48(m,1H),7.15-7.20(m,2H),7.10-7.16(m,2H), 7.00-7.05(m,1H).
Examples 1 and 2:
(2S, 4aS, 14aR, 14bR) -14- ((S) -10-fluoro-6-hydro-11-deuterodibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ compound 1] and (2S, 4aS, 14aR, 14bR) -14- ((R) -10-fluoro-6-hydro-11-deuterium dibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ Compound 2]
The synthetic route is as follows:
Figure BDA0002922605880000261
step 1: a round bottom flask was charged with DMF (300mL) intermediate 1-1(50g, 521mmol) and nitromethane (32g, 521 mmol). NaOH (62.4g, 1.56mmol) was added at 0 ℃. The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was quenched with water and then concentrated in vacuo. The residue was diluted with water and extracted with ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated in vacuo to yield 57.7g (crude) of intermediate 1-2 as a dark yellow oil. LCMS (ESI) M/z 140.1[ M +1] +.
Step 2: a round bottom flask was charged with THF (300mL) intermediate 1-2(57g, 410mmol) and lithium aluminum hydride (15.1g, 410mmol) added portionwise at 0 deg.C. Stirring was continued for 2 hours at 0 ℃. The mixture was filtered and the filtrate was concentrated in vacuo to give 38g (84%) of intermediates 1-3 as a colorless oil. GCMS (ESI) M/z 112.1[ M +1] +.
And step 3: a round bottom flask was charged with MeOH (300mL) intermediate 1-3(36.5.0g, 328.4mmol) and 2, 4-dimethoxybenzaldehyde (54.5g, 328 mmol). The resulting solution was stirred at 0 ℃ for 30 minutes and NaBH4(12.5g, 331mmol) was added portionwise. The resulting mixture was stirred at room temperature for an additional 2 hours. The reaction was quenched with water and then concentrated in vacuo. The residue was diluted with water and extracted with ethyl acetate. The combined organic phases were dried over anhydrous sodium sulfate and concentrated in vacuo to yield 34.5g (crude) of intermediates 1-4 as a dark yellow oil. LCMS (ESI) M/z 262.1[ M +1] +.
And 4, step 4: a round bottom flask was charged with intermediates 1-4(32.5g, 124.3mmol), DCM (300mL) and TEA (26.0g, 257 mmol). Prop-2-enecarbonyl chloride (35.0g, 386.7mmol, 1.55 eq.) was added dropwise, cooling to 0 ℃. After stirring at 0 ℃ for 30 minutes, the reaction was quenched with water. The resulting mixture was extracted with DCM and the combined organic layers were washed with water and brine, dried over anhydrous sodium sulfate and concentrated to give 19.5g (yield 50%) of intermediates 1-5 as yellow oil. LCMS (ESI) M/z 316.2[ M +1] +.
And 5: a round bottom flask was charged with intermediates 1-5(19.5g, 62.4mmol) and toluene (600 mL). Stirred at 110 ℃ for 12 hours and then concentrated. The residue was applied to a silica gel column and eluted with ethyl acetate/petroleum ether to give 16g (yield 82%) of intermediates 1-6 as pale yellow oil. LCMS (ESI) M/z 316.2[ M +1 ].
Step 6: a round bottom flask was charged with intermediates 1-6(16g, 50.6mmol), MeOH (300mL), and Pd/C (1.6g, 10% on activated carbon). The mixture was stirred at room temperature for 1 hour under an atmosphere of H2 (2-3 atm). The mixture was filtered and the filtrate was concentrated in vacuo to give 15g (92%) of intermediates 1-7 as a colorless oil. LCMS (ESI) M/z 318.2[ M +1] +.
And 7: a round-bottom flask was charged with intermediates 1-7(0.8g, 2.4mmol) and TFA (10 mL). Stirred at 60 ℃ for 1 hour and then concentrated. The residue was diluted with water. The pH of the aqueous solution was adjusted to 7 with saturated sodium bicarbonate solution. The resulting solution was extracted with DCM. The combined organic layers were concentrated in vacuo to afford 0.65g (crude) of intermediates 1-8 as white solids. LCMS (ESI) M/z 168.2[ M +1] +.
And 8: a round-bottom flask was charged with intermediates 1-8(2.60g, 15.6mmol) and THF (40 mL). n-BuLi (6.2mL, 2.5M in hexane, 715.6mmol) was added dropwise at-78 deg.C, and after the addition was complete, stirring was carried out for 1 hour at-78 deg.C. Prop-2-en-1-yl chloroformate (1.88g, 15.6mmol) was added dropwise at-78 ℃ and stirred for 1 hour while maintaining the temperature. The reaction was quenched with water, and the mixture was extracted with ethyl acetate. The combined organic layers were washed with water and brine, dried over anhydrous sodium sulfate, and concentrated. The residue was purified by column on silica eluting with ethyl acetate/petroleum ether to give 1.51g (two steps 63.4%) of intermediates 1-9. LCMS (ESI) M/z 252.2[ M +1] +.
And step 9: a round bottom flask was charged with a solution of intermediate 1-9(2g, 7.95mmol) in THF (20mL) and cooled to-78 deg.C LiAlH4(226.56mg, 5.9mmol, 1.5 equiv) was added portionwise. The temperature was maintained with stirring for 1 hour, then warmed to 0 ℃ and quenched with aqueous NaHCO3(5.0 mL). The resulting mixture was extracted with EtOAc and the combined organic layers were dried over anhydrous Na2S 04. After filtration, the filtrate was concentrated under reduced pressure to give intermediates 1 to 10(1.63 g). The crude product was used directly in the next step without further purification. LCMS (ESI) M/z 254.2[ M +1] +.
Step 10: to a round bottom flask was added intermediate 1-10(1.6g, 6.68mmol) in MeOH (10mL) and a stirred solution was added methanesulfonic acid (64mg, 0.67 mmol). Stirred at room temperature for 20 hours and then quenched with aqueous NaHC03(510 mL). The aqueous layer was extracted with EtOAc and the combined organic layers were dried over anhydrous Na2S 04. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with PE/EA (1/1) to give intermediates 1-11(450mg, 25%) as a pale yellow oil.
LCMS(ESI)m/z 268.15[M+1]+。
Step 11: a round-bottomed flask was charged with a solution of intermediates 1-11(40mg, 0.150mmol, 1 eq.) and intermediates 1-12 (90mg, 0.32mmol) in MeCN (2mL) at-20 deg.C and SnCl4(160mg, 0.6mmol) was added dropwise. Stir at rt overnight, then quench with aqueous NaHCO3(5mL) at 0 ℃ and extract the resulting mixture with EtOAc. The combined organic layers were dried over anhydrous Na2SO 4. After filtration, the filtrate was concentrated under reduced pressure to give intermediates 1-13(85mg, 53%), which were used in the next step without further purification. LCMS (ESI) M/z 524.3[ M +1] +.
Step 12: a round-bottom flask was charged with intermediate 1-13(1.2g, 2.25mmol) and morpholine (401mg, 4.6mmol) in a stirred solution of THF (20mL) to which was added Pd (PPh3)4(260mg, 0.24mmol, 0.1 equiv) in portions. The resulting mixture was stirred at room temperature for 20 h and diluted with Et20(30 mL). The precipitated solid was collected by filtration and washed with Et20 to give 760mg of the product as a yellow solid. LCMS (ESI) M/z 394.2[ M +1] +. Chiral preparation (CHIRALPAKIA-3) purified solid to give two enantiomers. Intermediate 1-14, (2R, 4aR, 14aS, 14bS) -9- (benzyloxy) -1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione trifluoroacetate salt: rt 1.47min, (365mg, 33.0%), LCMS (ESI) M/z 394.2[ M +1] +; and intermediate 1-15 (2S, 4aS, 14aR, 14bR) -9- (benzyloxy) -1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione trifluoroacetate salt: RT 3.3min, (335mg, 31%), LCMS (ESI) M/z 394.2[ M +1] +.
Step 13: to a round bottom flask was added a stirred solution of intermediates 1-14(335mg, 0.69mmol) and intermediates 1-16(181mg, 0.70mmol of T3P (3mL) and EtOAc (3mL) was added methanesulfonic acid (65mg, 0.69 mmol). stirring at 50 deg.C for 24H the reaction was quenched with H20 the resulting mixture was extracted with EtOAc and the combined organic layers were dried over anhydrous Na2S04 after filtration the filtrate was concentrated under reduced pressure to give intermediates 1-17(590mg, crude). crude was used in the next step without further purification LCMS ESI (M/z 623.3[ M +1] +.
Step 14: a round-bottomed flask was charged with a solution of intermediates 1-17(580mg, 0.91mmol) and LiCl (390mg, 9.2mmol) in DMF (4.0mL), and stirred at 70 ℃ for 3 hours. The reaction was quenched with H20. The resulting mixture was extracted with EtOAc and the combined organic layers were dried over anhydrous Na2S 04. After filtration, the filtrate was concentrated under reduced pressure and purified by preparative purification to give (2S, 4aS, 14aR, 14bR) -14- ((S) -7, 8-difluoro-6, 11-dihydrodibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ compound 1]150mg and (2S, 4aS, 14aR, 14bR) -14- ((R) -7, 8-difluoro-6, 11-dihydrodibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino- [3, 4-a ] isoquinoline-8, 10-dione [ compound 2]165 mg. Compound 1: LCMS (ESI) M/z 533.2[ M +1] +. Compound 2: MS (ES, m/z): LCMS (ESI) M/z 533.2[ M +1] +.
Examples 3 and 4:
(2S, 4aS, 14aR, 14bR) -14- ((S) -10-fluoro-6, 6-didehydro-11-hydrodibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ compound 3] and (2S, 4aS, 14aR, 14bR) -14- ((R) -10-fluoro-6, 6-didehydro-11-hydrodibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ Compound 4]
The synthetic route is as follows:
Figure BDA0002922605880000291
step 1: a round bottom flask was charged with a stirred solution of intermediate 1-14(200mg, 0.41mmol) and intermediate 3-1(300mg, 0.41mmol of T3P (1.8mL) and EtOAc (2mL) to which was added methanesulfonic acid (39mg, 0.41 mmol). stirring at 50 deg.C for 24H the reaction was quenched with H20 the resulting mixture was extracted with EtOAc and the combined organic layers were dried over anhydrous Na2S04 after filtration the filtrate was concentrated under reduced pressure to give intermediate 3-2(360mg, crude). the crude was used in the next step without further purification LCMS (ESI) M/z 623.2[ M +1] +.
Step 2: a round-bottomed flask was charged with a solution of intermediate 3-2(300mg, 0.47mmol) and LiCl (203mg, 4.7mmol) in DMF (3.0mL), and stirred at 70 ℃ for 3 hours. The reaction was quenched with H20. The resulting mixture was extracted with EtOAc and the combined organic layers were dried over anhydrous Na2S 04. After filtration, the filtrate was concentrated under reduced pressure and purified by preparative-HPLC to give ((2S, 4aS, 14aR, 14bR) -14- ((S) -10-fluoro-6, 6-didehydro-11-dibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ compound 3]67mg and (2S, 4aS, 14aR, 14bR) -14- ((R) -10-fluoro-6, 6-didehydro-11-dibenzo [ b ] aS white solids, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ Compound 4]52 mg. Compound 3: LCMS (ESI) M/z 534.2[ M +1] +. Compound 4: MS (ES, m/z): LCMS (ESI) M/z 534.2[ M +1] +.
Examples 5 and 6:
(2S, 4aS, 14aR, 14bR) -14- ((S) -10-fluoro-6, 6-dideutero-11-deutero-dibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-oxidopyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ compound 5] and (2S, 4aS, 14aR, 14bR) -14- ((R) -10-fluoro-6, 6-dideutero-11-deuterodibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ Compound 6]
The synthetic route is as follows:
Figure BDA0002922605880000301
step 1: a round bottom flask was charged with a stirred solution of intermediate 1-14(150mg, 0.31mmol) and intermediate 5-1(225mg, 0.31mmol of T3P (1.35mL) and EtOAc (1.5mL) to which was added methanesulfonic acid (30mg, 0.31 mmol). stirring at 50 deg.C for 24H the reaction was quenched with H2O the resulting mixture was extracted with EtOAc and the combined organic layers were dried over anhydrous Na2SO4 after filtration the filtrate was concentrated under reduced pressure to give intermediate 5-2(250mg, crude). the crude was used in the next step without further purification LCMS (ESI) M/z 625.2[ M +1] +.
Step 2: a round-bottomed flask was charged with a solution of intermediate 5-2(200mg, 0.31mmol) and LiCl (136mg, 3.1mmol) in DMF (2mL), and stirred at 70 ℃ for 3 hours. The reaction was quenched with H20. The resulting mixture was extracted with EtOAc and the combined organic layers were dried over anhydrous Na2S 04. After filtration, the filtrate was concentrated under reduced pressure and purified by preparative purification to give ((2S, 4aS, 14aR, 14bR) -14- ((S) -10-fluoro-6, 6-didehydro-11-dibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ compound 5]42mg and (2S, 4aS, 14aR, 14bR) -14- ((R) -10-fluoro-6, 6-didehydro-11-dibenzo [ b ] aS white solids, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ Compound 6]35 mg. Compound 3: LCMS (ESI) M/z 535.2[ M +1] +. Compound 4: MS (ES, m/z): LCMS (ESI) M/z 535.2[ M +1] +.
Examples 7 and 8:
(2S, 4aS, 14aR, 14bR) -14- ((S) -7, 8-difluoro-6-deuterium-11-hydrodibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ compound 7] and (2S, 4aS, 14aR, 14bR) -14- ((R) -7, 8-difluoro-6-deuterium-11-hydrodibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ Compound 8]
The synthetic route is as follows:
Figure BDA0002922605880000311
step 1: to a round bottom flask was added a stirred solution of intermediates 1-14(96mg, 0.24mmol, 1 eq) and intermediates 7-16 (64mg, 0.24mmol) in T3P (1.2mL) and EtOAc (1.0mL) to which was added methanesulfonic acid (23.6mg, 0.24 mmol). The resulting mixture was stirred at 70 ℃ under an atmosphere of N2 for 16 hours and then quenched with H20. The resulting mixture was extracted with EtOAc. The combined organic layers were dried over anhydrous Na2S 04. After filtration, the filtrate was concentrated under reduced pressure to give intermediate 7-1(180mg, crude). It was used directly in the next step without further purification. LCMS (ESI) M/z 641.2[ M +1] +.
Step 2: a mixture of intermediate 7-1(180mg, crude, 0.25mmol) and LiCl (51mg, 1.2mmol) in DMA (1.0mL) was stirred for 4 hours. The reaction solution was purified by preparative method to give (2S, 4aS, 14aR, 14bR) -14- ((S) -7, 8-difluoro-6-deuterium-11-hydrodibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ compound 7] and (2S, 4aS, 14aR, 14bR) -14- ((R) -7, 8-difluoro-6-deuterium-11-hydrodibenzo [ b, e ] thiepin-11-yl) -9-hydroxy-1, 3, 4, 5, 6, 14, 14a, 14 b-octahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazino [3, 4-a ] isoquinoline-8, 10-dione [ compound 8 ]. Compound 7: LCMS (ESI) M/z 551.2[ M +1] +. Compound 8: LCMS (ESI) M/z 551.2[ M +1] +.
The synthesis methods of reference examples 1 to 8 were used to synthesize the following compounds:
Figure BDA0002922605880000321
example 9:
synthesis of ((2S, 4aS, 14aR, 14bR) -14- ((S) -10-fluoro-6-deuterium-11-hydrodibenzo [ b, e ] thiepin-11-yl) -8, 10-dioxo-1, 3, 4, 5, 6, 8, 10, 14, 14a, 14 b-decahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazin [3, 4-a ] isoquinolin-9-yl) oxy) methyl carbonate:
the synthetic route is as follows:
Figure BDA0002922605880000331
a round-bottom flask was charged with a stirred mixture of Compound 1(20mg, 0.038mmol), Ag2CO 3(42mg, 0.118mmol) and KI (19mg, 0.118mmol) in DMA (0.1mL) chloromethyl methyl carbonate (14mg, 0.118mmol) was added in one portion and the resulting mixture was stirred at 45 ℃ for 12 h. After filtration, the reaction solution was purified by preparation of the filtrate to obtain the title compound 13(3mg) as a white solid. LCMS (ESI) M/z 621.2[ M +1] +.
Example 10:
synthesis of ((2S, 4aS, 14aR, 14bR) -14- ((S) -7, 8-difluoro-6-hydro-11-deuterodibenzo [ b, e ] thiepin-11-yl) -8, 10-dioxo-1, 3, 4, 5, 6, 8, 10, 14, 14a, 14 b-decahydro-2H-2, 4 a-epoxypyrido [1 ', 2': 1, 6] [1, 2, 4] triazin [3, 4-a ] isoquinolin-9-yl) oxy) methyl carbonate [ Compound 14 ]:
the synthetic route is as follows:
Figure BDA0002922605880000332
a round-bottomed flask was charged with a stirred mixture of compound 7(10mg, 0.02mmol), Ag2CO3(15mg, 055mmol) and KI (9mg, 0.054mmol) in DMA (0.2mL) and chloromethyl methyl carbonate (7mg, 0.55mmol) was added in one portion and the resulting mixture was stirred at 45 ℃ for 16 hours. The reaction solution was purified by prep to give the title compound (compound 14) LCMS (ESI) M/z 639.2[ M +1] +as a white solid
The synthesis of reference examples 9, 10 was used to synthesize the following experimental examples:
Figure BDA0002922605880000341
Figure BDA0002922605880000351
Figure BDA0002922605880000361
Figure BDA0002922605880000371
Figure BDA0002922605880000381
Figure BDA0002922605880000391
Figure BDA0002922605880000401
example 11: in vitro cytopathic Effect (CPE) assay for evaluating the Activity of Compounds against influenza Virus
The antiviral activity of the compounds against influenza A/WSN/33(H1N1) and the toxicity against MDCK cells were examined by the following experiments of cytopathic effect (CPE).
The following acronyms are used throughout this embodiment
Figure BDA0002922605880000402
Figure BDA0002922605880000411
Test materials
(1) Compounds were made up in 100% DMSO solution in 20mM stock. Compounds were tested at 8 concentration points, 4-fold gradient dilution, duplicate wells.
(2) MDCK canine kidney cells were purchased from ATCC. Cells were cultured in EMEM medium (Sigma) supplemented with 10% fetal bovine serum (Hyclone), 2mM L-glutamine (Gibco), 1% non-essential amino acids (Gibco), 100U/ml penicillin and 100. mu.g/ml streptomycin (Hyclone). OptiPRO SFM medium (Gibco) supplemented with 2mM L-glutamine, 1% non-essential amino acids, 100U/ml penicillin and 100. mu.g/ml streptomycin was used as the test medium.
(3) Influenza A/WSN/33(H1N1) strains were purchased from Virapur.
(4) The main reagent used in the project is a cell viability detection kit CCK8 (Shanghai Liji biology)
(5) The main instrument used in the project is a microplate reader SpectraMax340PC384(Molecular Device).
Test method
CPE refers to the phenomenon of massive proliferation of the virus in the host cell, leading to cytopathic and even death. By measuring cell viability, the CPE assay is widely used to determine the inhibitory activity of compounds against viruses that cause cytopathic effects. The in vitro inhibitory activity of compounds against influenza A/WSN/33(H1N1) was tested using the CPE assay.
MDCK cells were plated at a density of 2,000 cells per well in 384-well cell culture plates and in 5% CO2And cultured overnight in an incubator at 37 ℃. The next day compounds (8 concentration points, 4-fold gradient dilution, duplicate wells) and virus (M0I ═ 0.04) were added separately. The final concentrations of DMSO and pancreatin in the cell culture broth were 0.5% and 2.5. mu.g/ml, respectively. Cells were incubated at 37 ℃ with 5% CO2The culture was continued for 5 days under the conditions until the cytopathic rate in the virus control wells (cells infected with virus, no compound treatment) reached 80-95%. The cytotoxicity test and the antiviral test are carried out simultaneously, the test conditions are consistent, but no virus infection exists.
Cell viability was measured using cell viability assay reagent CCK 8. The antiviral activity and cytotoxicity of the compound are represented by the inhibition rate (%) and cell viability (%) of the compound against the viral-induced cytopathic effect, respectively. The calculation formula is as follows:
Figure BDA0002922605880000421
Figure BDA0002922605880000422
note: sample well: compound treating the wells;
virus control wells: cells infected with virus, no compound treatment;
cell control wells: cells, no compound treatment or viral infection;
culture solution control wells: only the culture broth, without cells, viruses or compounds.
Nonlinear fitting analysis of the inhibition rate and cell viability rate of the compound was performed using GraphPad Prism (version 5) software to obtain the EC of the compound50And CC50The value is obtained.
Test results
The activity and cytotoxicity of the compounds against influenza virus A/WSN/33(H1N1) against MDCK are summarized in Table 1.
TABLE 1 anti-influenza Virus Activity test results
Compound (I) EC50(nM)# CC50(μM)*
Compound 1 A c
Compound 2 B b
Compound 3 A c
Compound 4 B c
Compound 5 A c
Compound 6 B b
Compound 7 A c
Compound 8 B c
Compound 9 A c
Compound 10 B c
Compound 11 A c
Compound 12 B c
#1nM<A<10nM;10nM<B<100nM;C>100nM
*1M<a<10M;10M<b<50M;c>50M
As can be seen from Table 1, the experimental results show that the compounds of the examples have better inhibitory activity and lower cytotoxicity against influenza A/WSN/33(H1N 1).
Example 12: hERG assay
To assess the risk of prolongation of the QT interval of the electrocardiogram, the effect on the delayed rectified K ten current (IKr), which plays an important role in ventricular repolarization, was studied using HEK293 cells expressing the human ether-a-go-go related gene (hERG) channel. After recording the membrane potential at-80 mV by the whole-cell patch clamp method using a full-automatic patch clamp system, the current generated by IKr induced by depolarization stimulation of +50mV for 2 seconds and repolarization stimulation of-50 mY for 2 seconds was stabilized, and the extracellular fluid (NaCl: 137mmol/L, KCl 4mmol/L, Ca Cl/L) in which the test substance was dissolved at the desired concentration was allowed to flow2:1.8mmol/L,MgCl2-6H2O: 1mmol/L glucose 10mmol/L, HEPES 10mmol/L, pH 7.4.4) at room temperature for 10 minutes. From the obtained IKr, the absolute value of the maximum tail current was measured using analytical software at a current value of the resting membrane potential of Haruki. Further, the inhibition rate of the maximum tail current before application to a thousand test substances was calculated, and the test substances were evaluated for I in comparison with the medium application group (0.1% DMSO solution)KrInfluence of (c).
TABLE 2 inhibition of the compounds of the examples at 0.3-10. mu. mol/L
Compound (I) Inhibition ratio%
Compound 1 2.5
Compound 3 4.2
Compound 5 3.1
Compound 7 4.8
Compound 9 5.2
Compound 11 5.8
S-033188A 8.4
As can be seen from Table 2, the results of the experiment show that the example compounds have lower hERG inhibitory activity.
Example 13: pharmacokinetic testing of rats
Test materials and methods
(1) Animals used were SD rats, male beagle dogs.
(2) Animals were fasted for at least 12 hours prior to dosing and fed 2 hours after dosing. Water was provided ad libitum during the study.
(3) In each study, three animals were included, rats were dosed with 5mg/kg and dogs were dosed with 2mg/kg of compound 13 or bacloxavir disoproxil, each suspended in 0.5% HPMC in water. Blood samples were taken at 0.25, 0.5, 1, 2, 4, 8 and 24 hours post-dose of P0. The collected blood samples were then centrifuged at 3500rpm for 10 minutes at 4 ℃ to obtain plasma. The plasma samples were transferred to polyethylene tubes and immediately stored at about-80 ℃ until analysis.
(4) Evaluation items blood was collected over time, and drug concentration in plasma was measured using LC/MS.
(5) Statistical analysis of concentration changes in thousand plasma, calculation of the area under the time curve (AUC) of the concentration in plasma using a non-linear least squares procedure, calculation of Bioavailability (BA) from the AUC of the oral administration group and the intravenous administration group, and statistical analysis of the elimination half-life T of the plasma concentration in intravenous administration1/2
The test results were as follows:
TABLE 3 pharmacokinetic test results of rat compounds of the examples, 5mg/kg, orally administered
Figure BDA0002922605880000441
After rat administration compound 1 had a Cmax at least 16-fold higher than baroxavir acid and compound 1 had an AUC at least 4-fold higher than baroxavir acid.
TABLE 3 determination of the Biggee pharmacokinetic test for the compound of the example 2mg/kg, orally administered
Figure BDA0002922605880000442
After administration to beagle dogs, compound 1 had a Cmax at least 7-fold higher than baroxavir acid and compound 1 had an AUC at least 3-fold higher than baroxavir acid.
In the description herein, references to the description of the term "one embodiment," "some embodiments," "an example," "a specific example," or "some examples," etc., mean that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the invention. In this specification, the schematic representations of the terms used above are not necessarily intended to refer to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, various embodiments or examples and features of different embodiments or examples described in this specification can be combined and combined by one skilled in the art without contradiction.
Although embodiments of the present invention have been shown and described above, it is understood that the above embodiments are exemplary and should not be construed as limiting the present invention, and that variations, modifications, substitutions and alterations can be made to the above embodiments by those of ordinary skill in the art within the scope of the present invention.

Claims (8)

1. A fused polycyclic pyridone derivative represented by the general formula I and pharmaceutically acceptable salts thereof,
Figure FDA0003267126260000011
wherein the content of the first and second substances,
R1、R2and R3Each independently selected from hydrogen or deuterium, and R1、R2And R3At least one of them is deuterium;
R4、R5、R6and R7Each independently selected from hydrogen or fluorine;
R8is hydrogen, an optionally substituted phosphate group, C2-4Alkenyl, -C (═ O) -Y1、-C(=O)-O-Y2、-(CH2)-O-(C=O)-Y1、-(CH2)-O-(C=O)-O-Y2、-(CHCH3)-O-(C=O)-Y1、-(CHCH3)-O-(C=O)-O-Y2
Y1Is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted heterocyclyl;
Y2is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted heterocyclyl.
2. A fused polycyclic pyridone derivative and a pharmaceutically acceptable salt thereof according to claim 1, wherein R is1When is deuterium; is a compound represented by the structure of formula II:
Figure FDA0003267126260000012
R2and R3Each independently selected from hydrogen or deuterium;
R4、R5、R6and R7Each independently selected from hydrogen or fluorine;
R8is hydrogen, an optionally substituted phosphate group, C2-4Alkenyl, -C (═ O) -Y1、-C(=O)-O-Y2、-(CH2)-O-(C=O)-Y1、-(CH2)-O-(C=O)-O-Y2、-(CHCH3)-O-(C=O)-Y1、-(CHCH3)-O-(C=O)-O-Y2
Y1Is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted heterocyclyl;
Y2is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted heterocyclyl.
3. A fused polycyclic pyridone derivative and a pharmaceutically acceptable salt thereof according to claim 1, wherein R is1Selected from hydrogen, R2And R3When selected from deuterium, is a compound having the structure shown in formula III below:
Figure FDA0003267126260000021
R4、R5、R6and R7Each independently selected from hydrogen or fluorine;
R8is hydrogen, an optionally substituted phosphate group, C2-4Alkenyl, -C (═ O) -Y1、-C(=O)-O-Y2、-(CH2)-O-(C=O)-Y1、-(CH2)-O-(C=O)-O-Y2、-(CHCH3)-O-(C=O)-Y1、-(CHCH3)-O-(C=O)-O-Y2
Y1Is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted heterocyclyl;
Y2is optionally substituted C1-10Alkyl, optionally substituted C3-8Cycloalkyl, optionally substituted heterocyclyl.
4. A fused polycyclic pyridone derivative and pharmaceutically acceptable salts thereof, wherein the fused polycyclic pyridone derivative has one of the following structures:
Figure FDA0003267126260000031
Figure FDA0003267126260000041
5. a pharmaceutical composition comprising the fused polycyclic pyridone derivative of any one of claims 1 to 4, and a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, excipient, or combination thereof.
6. The pharmaceutical composition of claim 5, further comprising one or more additional therapeutic agents, wherein the additional therapeutic agent is selected from an anti-influenza virus agent selected from amantadine, rimantadine, oseltamivir, zanamivir, peramivir, ranimivir, faviravir, abidol, ribavirin, or a combination thereof, or a vaccine.
7. Use of a fused polycyclic pyridone derivative according to any one of claims 1 to 4, and pharmaceutically acceptable salts thereof, and a pharmaceutical composition according to claim 5 for the preparation of a medicament for the prevention, alleviation or treatment of influenza virus infection.
8. The use of claim 7, wherein the influenza virus is selected from influenza a, influenza b, influenza c, avian influenza H5N1 or avian influenza H7N 9; the symptoms of influenza virus infection are selected from fever, chill, headache, muscle pain, general malaise, pharyngalgia, rhinorrhea, nasal obstruction, cough, expectoration, abdominal pain, emesis, diarrhea, complications with acute encephalopathy, pneumonia secondary infection or their combination.
CN202110108914.XA 2021-01-29 2021-01-29 Fused polycyclic pyridone derivative and application thereof Active CN112898312B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110108914.XA CN112898312B (en) 2021-01-29 2021-01-29 Fused polycyclic pyridone derivative and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110108914.XA CN112898312B (en) 2021-01-29 2021-01-29 Fused polycyclic pyridone derivative and application thereof

Publications (2)

Publication Number Publication Date
CN112898312A CN112898312A (en) 2021-06-04
CN112898312B true CN112898312B (en) 2021-11-12

Family

ID=76120573

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110108914.XA Active CN112898312B (en) 2021-01-29 2021-01-29 Fused polycyclic pyridone derivative and application thereof

Country Status (1)

Country Link
CN (1) CN112898312B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115925658B (en) * 2022-11-21 2024-02-27 常州佳德医药科技有限公司 Preparation method of 2-aminoethylfuran

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003087103A1 (en) * 2002-04-18 2003-10-23 Astrazeneca Ab Thienyl compounds
CN108440564A (en) * 2018-04-11 2018-08-24 朱孝云 Substituted polycyclic carbamoylpyridone derivative and its prodrug
CN109503625A (en) * 2018-01-19 2019-03-22 赵蕾 A kind of polycyclic pyridines ketone compound and its pharmaceutical composition and purposes
CN109721615A (en) * 2017-09-18 2019-05-07 广东东阳光药业有限公司 Inhibitors of influenza viruses replication and application thereof
CN110317211A (en) * 2018-07-27 2019-10-11 深圳市塔吉瑞生物医药有限公司 Substituted polycyclic pyridone compound and prodrug thereof
WO2020205835A1 (en) * 2019-04-01 2020-10-08 Nikang Therapeutics, Inc. Fused polycyclic pyridone compounds as influenza virus replication inhibitors

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20100061819A (en) * 2007-08-29 2010-06-09 쉐링 코포레이션 Tetracyclic indole derivatives and their use for treating or preventing viral infections

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003087103A1 (en) * 2002-04-18 2003-10-23 Astrazeneca Ab Thienyl compounds
CN109721615A (en) * 2017-09-18 2019-05-07 广东东阳光药业有限公司 Inhibitors of influenza viruses replication and application thereof
CN109503625A (en) * 2018-01-19 2019-03-22 赵蕾 A kind of polycyclic pyridines ketone compound and its pharmaceutical composition and purposes
CN108440564A (en) * 2018-04-11 2018-08-24 朱孝云 Substituted polycyclic carbamoylpyridone derivative and its prodrug
CN110317211A (en) * 2018-07-27 2019-10-11 深圳市塔吉瑞生物医药有限公司 Substituted polycyclic pyridone compound and prodrug thereof
WO2020205835A1 (en) * 2019-04-01 2020-10-08 Nikang Therapeutics, Inc. Fused polycyclic pyridone compounds as influenza virus replication inhibitors
CN112118891A (en) * 2019-04-01 2020-12-22 尼坎医疗公司 Fused polycyclic pyridone compounds as inhibitors of influenza virus replication

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
抗流感病毒药物研究进展;高岩等;《中国医药生物科技》;20151231;第10卷(第6期);第533-539页 *

Also Published As

Publication number Publication date
CN112898312A (en) 2021-06-04

Similar Documents

Publication Publication Date Title
CN109721615B (en) Inhibitors of influenza virus replication and uses thereof
CN109641868B (en) Inhibitors of influenza virus replication and methods of use and uses thereof
CN109748915B (en) Inhibitors of influenza virus replication and methods of use and uses thereof
CN107759571B (en) Inhibitors of influenza virus replication and methods of use and uses thereof
CN108218873B (en) Inhibitors of influenza virus replication and uses thereof
CN108276401B (en) Inhibitors of influenza virus replication and uses thereof
US20110076276A1 (en) Novel HIV reverse transcriptase inhibitors
US20090215778A1 (en) Alpha-substituted Arylmethyl Piperazine Pyrazolo [1,5-alpha]Pyrimidine Amide Derivatives
EA037949B1 (en) Aza-pyridone compounds and uses thereof
CN108976223B (en) Fused tricyclic compound and application thereof in medicines
CN111057074B (en) Inhibitors of influenza virus replication and uses thereof
WO2018202155A1 (en) Bicyclic nucleocapsid inhibitor and use of same as drug in treatment of hepatitis b
CN112521386B (en) Polycyclic pyridone compounds having antiviral action, pharmaceutical combinations and uses thereof
WO2009005693A1 (en) Novel hiv reverse transcriptase inhibitors
CN112174955B (en) 2-substituted-5-hexahydropyridazinone-4-carboxylic acid amine derivative and application thereof
CN113286793A (en) Pyrrolopyrazine and pyridotriazine influenza virus replication inhibitors
CN112898312B (en) Fused polycyclic pyridone derivative and application thereof
CN110446711B (en) Inhibitors of influenza virus replication and uses thereof
CN110117285B (en) Inhibitors of influenza virus replication and uses thereof
CN113614085B (en) Inhibitors of influenza virus replication and uses thereof
CN113748104A (en) Isoquinolinone compound for inhibiting SSAO/VAP-1 and application thereof
CN113880831A (en) Pyrazine dione derivative and application thereof
CN113416198A (en) Fused polycyclic pyridone derivative and application thereof
CN112174956B (en) 2-substituted-5-hexahydropyridazinone-4-carboxylic ester derivative and application thereof
CN114014857A (en) Fused polycyclic pyridone derivative 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