Nothing Special   »   [go: up one dir, main page]

CN113412261A - Benzimidazole compounds, preparation method and use thereof - Google Patents

Benzimidazole compounds, preparation method and use thereof Download PDF

Info

Publication number
CN113412261A
CN113412261A CN202080013096.9A CN202080013096A CN113412261A CN 113412261 A CN113412261 A CN 113412261A CN 202080013096 A CN202080013096 A CN 202080013096A CN 113412261 A CN113412261 A CN 113412261A
Authority
CN
China
Prior art keywords
compound
alkyl
independently selected
added
alkoxy
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202080013096.9A
Other languages
Chinese (zh)
Inventor
王超磊
吴勇勇
吴政
陈文亮
刘金明
田强
宋宏梅
薛彤彤
王晶翼
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.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical 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 Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Publication of CN113412261A publication Critical patent/CN113412261A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Description

Benzimidazole compounds, preparation method and use thereof Technical Field
The present invention relates to benzimidazole compounds for the treatment of CCR2/CCR5 mediated diseases or disorders, pharmaceutical compositions comprising the same, kits comprising the same, methods for the preparation thereof and use thereof in the preparation of medicaments for the treatment of CCR2/CCR5 mediated diseases or disorders.
Background
Non-alcoholic fatty liver disease is a clinical pathological syndrome without history of excessive drinking, characterized by steatosis and fat storage of liver parenchymal cells, and is divided into three pathological stages according to liver histological changes: simple non-alcoholic fatty liver, non-alcoholic steatohepatitis (NASH), steatocirrhosis, and liver cancer. More seriously, NASH is also closely related to metabolic syndromes such as obesity, type 2 diabetes, dyslipidemia, etc., and NASH is associated with obesity of about 2/3 and patients with type 2 diabetes more than 1/3.
The exact pathogenic mechanism of NASH has not been elucidated and is different in almost every patient. Although morbidity is most associated with insulin resistance, obesity, and metabolic syndrome, not all patients in these states have NAFLD and/or NASH, nor do all patients with NAFLD and/or NASH experience these states. If NAFLD progresses to NASH, the risk of developing liver cirrhosis, liver failure and liver cancer will increase greatly.
At present, drugs developed aiming at NASH are developed around the key link of the pathogenesis of NASH, and the main action links of the research targets are fat transportation, free fatty acid formation, oxidative stress, insulin resistance, inflammation and the like. The main targets for NASH are: peroxisome Proliferator Activated Receptor Alpha (PPARA) and receptor delta (PPARD) agonists, Farnesoid X Receptor (FXR) agonists, monoclonal antibodies inhibiting lysyl oxidase 2(LOXL2) like proteins, apoptosis signal-regulating protein kinase (ASK1) inhibitors, acetyl-coa carboxylase (ACC) protein allosteric inhibitors, chemokine receptor (CCR2/CCR5) antagonists, and the like.
Ceniviroc, a potential new drug for the treatment of adult nonalcoholic steatohepatitis (NASH) and liver fibrosis, has been demonstrated to be a dual inhibitor of CCR2 and CCR5, can block chemokine receptors that cause liver damage and lead to liver disease, and phase 2b clinical trials have been completed. CCR2 and CCR5 are key regulators of the signaling pathway in NASH leading to liver injury and disease. NASH is a potentially life-threatening liver disease and there is currently no approved treatment, with clinical studies in stage 2b of ceriviroc having milestone significance.
Despite the existence of known CCR2 and CCR5 inhibitors, these inhibitors still have deficiencies in terms of in vivo efficacy, safety, and the like. Therefore, the development of a novel dual CCR2 and CCR5 inhibitor which has stronger in-vivo inhibition effect, improved properties such as drug metabolism stability and the like and smaller toxic and side effects is urgently desired.
Summary of The Invention
In one aspect, the invention provides a benzimidazole compound for use in treating a CCR2/CCR5 mediated disease or disorder. The compound is a compound shown as a general formula I or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (such as hydrate), metabolite, prodrug or a mixture thereof,
Figure PCTCN2020081558-APPB-000001
wherein,
R 1selected from hydrogen and C1-6Alkyl radical, said C1-6Alkyl is optionally substituted with one or more substituents independently selected from: hydrogen, hydroxy, carboxy, - (C)1-6Alkoxy group)m1-(C 1-6Alkoxy group)m2、NR aR bAnd a 5-6 membered heterocyclyl containing one or more heteroatoms independently selected from N, O and S;
R 2is selected from C1-6Alkyl and 4-6 membered heterocyclyl containing one or more heteroatoms independently selected from N, O and S, said C1-6Alkyl and 4-6 membered heterocyclyl are optionally substituted with 1-3 substituents independently selected from: hydrogen, deuterium, halogen, hydroxy, carboxy, - (C)1-6Alkoxy group)m1-(C 1-6Alkoxy group)m2And NRaR b
R 9、R 10Is absent or is each independently selected from halogen, hydroxy, carboxy, cyano, NRaR b、C 1-6Alkoxy radical, C1-6Alkyl radical, C1-6Haloalkyl and-COOC1-6An alkyl group;
m 1、m 2each independently selected from 0, 1,2,3 or 4;
R a、R beach independently selected from hydrogen and C1-6An alkyl group;
y is selected from N or CH;
a is selected from 4-7 membered heterocyclyl and 5-10 membered heteroaryl, said 4-7 membered heterocyclyl or 5-10 membered heteroaryl being optionally substituted with one or more substituents independently selected from: halogen, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-6Cycloalkyl radical, C3-6Halogenocycloalkyl, C3-6Cycloalkoxy and C1-6A haloalkyloxy group; and is
n is selected from 0, 1 or 2.
In another aspect, the present invention provides a pharmaceutical composition comprising an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, and one or more pharmaceutically acceptable carriers.
In yet another aspect, the invention provides a kit comprising a compound of the invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of the invention.
In a further aspect, the present invention provides a compound of the invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g. hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of the invention, or a kit of the invention, for use in the prevention or treatment of a disease or condition mediated by CCR2 and/or CCR5, particularly non-alcoholic fatty liver disease (NAFLD).
In a further aspect, the present invention provides the use of a compound of the invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g. hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of the invention, or a kit of the invention, in the manufacture of a medicament for the prevention or treatment of a disease or condition mediated by CCR2 and/or CCR5, in particular non-alcoholic fatty liver disease (NAFLD).
In a further aspect, the present invention provides a method of preventing or treating a disease or disorder mediated by CCR2 and/or CCR5, in particular nonalcoholic fatty liver disease (NAFLD), comprising administering to a subject in need thereof an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of the present invention, or a kit of the present invention.
In yet another aspect, the present invention provides a method of preparing a compound of the present invention, comprising the steps of:
Figure PCTCN2020081558-APPB-000002
wherein R is1、R 2、R 9、R 10A, Y and n are as defined above; and LG is a leaving group.
Detailed Description
Definition of
Unless defined otherwise below, all technical and scientific terms used herein are intended to have the same meaning as commonly understood by one of ordinary skill in the art. Reference to the techniques used herein is intended to refer to those techniques commonly understood in the art, including those variations of or alternatives to those techniques that would be apparent to those skilled in the art. While the following terms are believed to be well understood by those skilled in the art, the following definitions are set forth to better explain the present invention.
As used herein, the terms "comprises," "comprising," "has," "containing," or "involving," and other variations thereof herein, are inclusive or open-ended and do not exclude additional unrecited elements or method steps.
The term "alkyl" as used herein is defined as a straight or branched chain saturated aliphatic hydrocarbon group. Alkyl groups have, for example, 1 to 8, 1 to 4, or 1 to 3 carbon atoms. For example, as used herein, the term "C1-6Alkyl "refers to a straight or branched chain group having 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, or n-hexyl), which is optionally substituted with one or more (such as 1 to 3) suitable substituents such as halogen (when the group is referred to as" haloalkyl ", e.g., -CF)3、-C 2F 5、-CHF 2、-CH 2F、-CH 2CF 3、-CH 2Cl or-CH2CH 2CF 3Etc.).
As used herein, the term "cycloalkyl" refers to a saturated or unsaturated non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (e.g., monocyclic, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, or bicyclic ring, including spiro, fused or bridged systems (such as bicyclo [ 1.1.1)]Pentyl, bicyclo [2.2.1]Heptyl, etc.), optionally substituted with one or more (such as 1 to 3) suitable substituents. The cycloalkyl group has, for example, 3 to 15, 3 to 6 carbon atoms. For example, as used herein, the term "C3-6Cycloalkyl "refers to a saturated or unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl) having 3 to 6 ring carbon atoms, optionally substituted with one or more (such as 1 to 3) suitable substituents, for example, methyl-substituted cyclopropyl.
As used herein, the term "alkoxy" refers to a straight, branched or cyclic saturated monovalent hydrocarbon radical of the formula-O-alkyl, wherein the term "alkyl" is as defined above, e.g., C1-6Alkyl radical, C1-4Alkyl groups, and the like. C1-6Representative examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, t-butoxy, sec-butoxy, n-pentoxy, isopentoxy, n-hexoxy, and the like.
As used herein, the term "cycloalkoxy" refers to a straight, branched, or cyclic saturated monovalent hydrocarbon radical of the formula-O-cycloalkyl, wherein the term "cycloalkyl" is as defined above, e.g., C3-6A cycloalkyl group. C3-6Representative examples of cycloalkoxy groups include, but are not limited to, cyclopropoxy, cyclobutoxy, cyclopentoxy, cyclohexyloxy, and the like.
As used herein, the term "heterocyclyl" refers to a saturated or unsaturated monocyclic or bicyclic group having 2,3,4, 5, 6, 7, 8, or 9 carbon atoms and one or more (e.g., one, two, three, or four) heteroatoms independently selected from N, O and S in the ring, e.g., a 5-6 membered heterocyclyl, a 4-6 membered heterocyclyl, and the like. Representative examples of heterocyclyl groups include, but are not limited to, oxiranyl, aziridinyl, azetidinyl, oxetanyl, tetrahydrofuranyl, dioxolyl, pyrrolidinyl, pyrrolidinonyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl, tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, trithianyl, and the like.
As used herein, the term "heteroaryl" refers to a monocyclic, bicyclic or tricyclic aromatic ring system having 5, 6,8, 9, 10, 11, 12, 13 or 14 ring atoms, for example having 5 to 10 ring atoms, in particular 1,2,3,4, 5, 6, 9, 10 carbon atoms, and which contains at least one heteroatom (e.g. oxygen, nitrogen or sulfur) which may be the same or different and which, in addition, may be benzo-fused in each case. For example, as used herein, the term "5-10 membered heteroaryl" means a heteroaryl group containing 5 to 10 ring atoms. In particular, heteroaryl is selected from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl and the like, and benzo derivatives thereof; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, and the like, and benzo derivatives thereof.
As used herein, the term "halo" or "halogen" group is defined to include F, Cl, Br, or I.
As used herein, formula NRaR bRepresentative examples of (b) include amino (-NH)2)、-NHCH 3、-N(CH 3) 2And the like. Unless otherwise indicated, compounds of the present invention containing an amino moiety may include protected derivatives thereof. Suitable protecting groups for the amino moiety include acetyl, t-butyloxycarbonyl, benzyloxycarbonyl and the like.
As used herein, the term "carboxyl group," also known as a "carboxylic acid group," refers to a functional group of the formula-COOH. In the sense of the present invention, the term "carboxyl" also includes the relevant anion-COO-(carboxylate ion). The functional group may be-COOH or-COO depending on the environmental conditions, such as the pH of the environment-The form exists.
As used herein, the term "C1-6Haloalkyl "means C as described hereinbefore1-6Groups obtained by substituting one or more (e.g. 1,2,3 or 4) hydrogen atoms in an alkyl group with a halogen (e.g. fluorine, chlorine, bromine or iodine). C1-6Representative examples of haloalkyl groups include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, trifluoroethyl, chloroethyl, and the like.
The term "substituted" means that one or more (e.g., one, two, three, or four) hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the current circumstances is not exceeded and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
If a substituent is described as "optionally substituted," the substituent may be (1) unsubstituted or (2) substituted. If a carbon of a substituent is described as being optionally substituted with one or more of the list of substituents, one or more hydrogens on the carbon (to the extent of any hydrogens present) may be replaced individually and/or together with an independently selected optional substituent. If the nitrogen of a substituent is described as being optionally substituted with one or more of the list of substituents, then one or more hydrogens on the nitrogen (to the extent any hydrogen is present) may each be replaced with an independently selected optional substituent.
If a substituent is described as being "independently selected from" a group, each substituent is selected independently of the other. Thus, each substituent may be the same as or different from another (other) substituent.
When a bond of a substituent is shown through a bond connecting two atoms in a ring, then such substituent may be bonded to any ring atom in the substitutable ring.
As used herein, the term "one or more" means 1 or more than 1, such as 2,3,4, 5 or 10, under reasonable conditions.
Unless indicated, as used herein, the point of attachment of a substituent may be from any suitable position of the substituent.
The compounds of the invention may also contain one or more (e.g. one, two, three or four) isotopic substitutions. For example, in the compounds, H may be in any isotopic form, including1H、 2H (D or deuterium) and3h (T or tritium); c may be in any isotopic form, including12C、 13C and14c; o may be in any isotopic form, including16O and18o, and the like.
The term "stereoisomer" denotes an isomer formed as a result of at least one asymmetric center. In compounds having one or more (e.g., one, two, three, or four) asymmetric centers, they can give rise to racemates, racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Certain individual molecules may also exist as geometric isomers (cis/trans). Similarly, the compounds of the invention may exist as mixtures of two or more structurally different forms (commonly referred to as tautomers) in rapid equilibrium. Representative examples of tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers, and the like. It is understood that the scope of this application encompasses all such isomers or mixtures thereof in any ratio (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%).
Unless otherwise indicated, the compounds of the present invention are intended to exist as stereoisomers, including cis and trans isomers, optical isomers (e.g., R and S enantiomers), diastereomers, geometric isomers, rotamers, conformers, atropisomers, and mixtures thereof. The compounds of the present invention may exhibit more than one type of isomerization and consist of mixtures thereof (e.g., racemic mixtures and diastereomeric pairs).
The present invention encompasses all possible crystalline forms or polymorphs of the compounds of the present invention, which may be single polymorphs or mixtures of more than one polymorph in any ratio.
The present invention further includes within its scope prodrugs of the compounds of the present invention which are certain derivatives of the compounds of the present invention which may themselves have little or no pharmacological activity which, when administered into or onto the body, may be converted to the compounds of the present invention having the desired activity by, for example, hydrolytic cleavage. Typically such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the desired therapeutically active compound. Further information on the use of prodrugs can be found in "Pro-drugs as Novel Delivery Systems", volume 14, ACS Symposium Series (T.Higuchi and V.Stella) and "Bioreversible Carriers in Drug Design," Pergamon Press,1987(E.B.Roche editions, American Pharmaceutical Association). Prodrugs of the invention may be prepared, for example, by substituting certain moieties known to those skilled in the art as "pro-moieties" (e.g., "Design of Prodrugs", described in h. bundgaard (Elsevier, 1985)) for appropriate functional groups present in compounds of the invention.
The invention also encompasses compounds of the invention containing a protecting group. In any process for preparing the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned, thereby forming a chemically protected form of the compounds of the present invention. This can be achieved by conventional protecting Groups, for example, as described in Protective Groups in Organic Chemistry, ed.j.f.w.mcomie, Plenum Press, 1973; and T.W.Greene & P.G.M.Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons,1991, which are incorporated herein by reference. The protecting group may be removed at a suitable subsequent stage using methods known in the art.
The term "pharmaceutically acceptable" as used herein means that the substance or composition must be chemically and/or toxicologically compatible with the other components comprising the formulation and/or the mammal being treated therewith.
It will also be appreciated that certain compounds of the invention may be present in free form for use in therapy or, where appropriate, in the form of a pharmaceutically acceptable derivative thereof. According to the present invention, pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable salts, solvates, metabolites or prodrugs, which upon administration to a patient in need thereof are capable of providing, directly or indirectly, a compound of the present invention or a metabolite or residue thereof.
Pharmaceutically acceptable salts of the compounds of the present invention include acid addition salts and base addition salts thereof. Suitable acid addition salts are formed from acids which form non-toxic salts. Acceptable inorganic acids such as nitric acid, phosphoric acid, and the like, and acceptable organic acids such as 4-methylbicyclo [2.2.2] oct-2-ene-l-carboxylic acid, glucoheptonic acid, 4' -methylenebis (3-hydroxy-2-ene-l-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tert-butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, muconic acid, and the like. Suitable base addition salts are formed from bases which form non-toxic salts. Acceptable inorganic bases include aluminum hydroxide and calcium hydroxide, and acceptable organic bases include tromethamine, N-methylglucamine, and the like.
For a review of suitable Salts, see Stahl and Wermuth, "Handbook of Pharmaceutical Salts: Properties, Selection, and Use" (Wiley-VCH, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds of the present invention are known to those skilled in the art.
The compounds of the invention may be present in the form of solvates, preferably hydrates, wherein the compounds of the invention comprise as structural element of the crystal lattice of the compound a polar solvent, such as in particular water, methanol or ethanol. The amount of polar solvent, particularly water, may be present in stoichiometric or non-stoichiometric proportions.
Also included within the scope of the present invention are metabolites of the compounds of the present invention, i.e., compounds that are formed in vivo upon administration of a drug.
The term "pharmaceutical composition" as used herein includes products comprising an effective amount of a compound of the invention, as well as any product which results, directly or indirectly, from combination of compounds of the invention.
The term "effective amount" as used herein refers to an amount sufficient to achieve a desired therapeutic effect, e.g., to achieve alleviation of symptoms associated with the disease being treated.
The term "treatment" as used herein is intended to reduce or eliminate the disease state or condition for which it is directed. A subject is successfully "treated" if the subject receives a therapeutic amount of a compound, an optical isomer thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof according to the methods described herein, and the subject exhibits an observable and/or detectable decrease or improvement in one or more of the subject's indications and symptoms. It is also understood that treatment of the disease state or condition described includes not only complete treatment, but also less than complete treatment, but achieves some biologically or medically relevant result.
By "treatment" is meant any administration of a compound of the invention, including:
(1) preventing disease development in animals that may be predisposed to disease but have not yet experienced or exhibited disease pathology or symptomology;
(2) inhibiting disease (i.e., arresting further development of pathology and/or symptomatology) in an animal experiencing or exhibiting disease pathology or symptomatology; or
(3) Ameliorating the disease (i.e., reversing the pathology and/or symptomatology) in an animal experiencing or exhibiting disease pathology or symptomatology.
Compound (I)
An object of the present invention is to provide a compound represented by the general formula I or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof,
Figure PCTCN2020081558-APPB-000003
wherein,
R 1selected from hydrogen and C1-6Alkyl radical, said C1-6Alkyl is optionally substituted with one or more substituents independently selected from: hydrogen, hydroxy, carboxy, - (C)1-6Alkoxy group)m1-(C 1-6Alkoxy group)m2、NR aR bAnd a 5-6 membered heterocyclyl containing one or more heteroatoms independently selected from N, O and S;
R 2is selected from C1-6Alkyl and 4-6 membered heterocyclyl containing one or more heteroatoms independently selected from N, O and S, said C1-6Alkyl and 4-6 membered heterocyclyl are optionally substituted with 1-3 substituents independently selected from: hydrogen, deuterium, halogen, hydroxy, carboxy, - (C)1-6Alkoxy group)m1-(C 1-6Alkoxy group)m2And NRaR b
R 9、R 10Is absent or is independently selected from halogen, hydroxy, carboxy, cyano,NR aR b、C 1-6Alkoxy radical, C1-6Alkyl radical, C1-6Haloalkyl and-COOC1-6An alkyl group;
m 1、m 2each independently selected from 0, 1,2,3 or 4;
R a、R beach independently selected from hydrogen and C1-6An alkyl group;
y is selected from CH or N;
a is selected from 4-7 membered heterocyclyl and 5-10 membered heteroaryl, said 4-7 membered heterocyclyl or 5-10 membered heteroaryl being optionally substituted with one or more substituents independently selected from: halogen, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-6Cycloalkyl radical, C3-6Halogenocycloalkyl, C3-6Cycloalkoxy and C1-6A haloalkyloxy group; and is
n is selected from 0, 1 or 2.
According to some embodiments of the invention, Y is CH and the compound is of formula II,
Figure PCTCN2020081558-APPB-000004
wherein,
R 1selected from hydrogen and C1-6Alkyl radical, said C1-6Alkyl is optionally substituted with one or more substituents independently selected from: hydrogen, hydroxy, carboxy, - (C)1-6Alkoxy group)m1-(C 1-6Alkoxy group)m2、NR aR bAnd a 5-6 membered heterocyclyl containing one or more heteroatoms independently selected from N, O and S;
R 2is selected from C1-6Alkyl and 4-containing one or more heteroatoms independently selected from N, O and S6-membered heterocyclic group, said C1-6Alkyl and 4-6 membered heterocyclyl are optionally substituted with 1-3 substituents independently selected from: hydrogen, deuterium, halogen, hydroxy, carboxy, - (C)1-6Alkoxy group)m1-(C 1-6Alkoxy group)m2And NRaR b
R 9、R 10Absent, or each independently selected from halogen, hydroxy, carboxy, cyano, NRaR b、C 1-6Alkoxy radical, C1-6Alkyl radical, C1-6Haloalkyl and-COOC1-6An alkyl group;
m 1、m 2each independently selected from 0, 1,2,3 or 4;
R a、R beach independently selected from hydrogen and C1-6An alkyl group;
a is selected from 4-7 membered heterocyclyl and 5-10 membered heteroaryl, said 4-7 membered heterocyclyl or 5-10 membered heteroaryl being optionally substituted with one or more substituents independently selected from: halogen, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-6Cycloalkyl radical, C3-6Halogenocycloalkyl, C3-6Cycloalkoxy and C1-6A haloalkyloxy group; and is
n is selected from 0, 1 or 2.
According to some embodiments of the invention, R1Is C1-4Alkyl radical, said C1-4Alkyl is optionally substituted with one or more substituents independently selected from: hydroxy, C1-6Alkoxy and a 5-6 membered heterocyclyl containing one or more heteroatoms independently selected from N, O and S. In a preferred embodiment, R1Is optionally substituted by one or more C1-4Alkoxy-substituted C1-4An alkyl group. In a particularly preferred embodiment, R1Selected from 2-butoxyethyl, 2-methoxyethylA radical and a butyl radical.
According to some embodiments of the invention, R2Is selected from C1-3Alkyl and 4-6 membered heterocyclyl containing N and/or O atoms, said C1-3Alkyl is optionally substituted with 1-3 substituents independently selected from: hydrogen, halogen, hydroxy, carboxy, C1-4Alkoxy and NRaR b. In a preferred embodiment, R2Selected from the group consisting of isopropyl, fluoromethyl, methoxymethyl, hydroxymethyl, 1-hydroxyisopropyl, N-dimethylaminomethyl, 1-carboxyisopropyl, tetrahydropyranyl, oxetanyl, morpholinyl, and 1, 4-dioxanyl. In a particularly preferred embodiment, R2Is tetrahydropyranyl.
According to some embodiments of the invention, R9Is absent.
According to some embodiments of the invention, R10Is absent.
According to some embodiments of the invention, n is 1.
According to some embodiments of the invention, a is selected from a 4-7 membered nitrogen-containing heterocyclyl and a 5-10 membered nitrogen-containing heteroaryl, said 4-7 membered nitrogen-containing heterocyclyl and 5-10 membered nitrogen-containing heteroaryl being optionally substituted with 1-3 substituents independently selected from: halogen, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy and C1-6A haloalkoxy group.
In a preferred embodiment, A is
Figure PCTCN2020081558-APPB-000005
Wherein,
x is selected from N and C-R5
R 3Selected from hydrogen, deuterium, C1-6Alkyl and C3-6Cycloalkyl radical, said C1-6Alkyl or C3-6Cycloalkyl is optionally substituted with one or more substituents independently selected fromAnd (3) substituent: hydrogen, deuterium, hydroxy, cyano, C1-4Alkoxy and C3-6A cycloalkoxy group; and is
R 4、R 5Each independently selected from hydrogen, deuterium, halogen, cyano, C1-6Alkyl radical, C3-6Cycloalkyl, 3-6 membered heterocyclyl, C6-10Aryl and 5-12 membered heteroaryl, wherein said C1-6Alkyl radical, C3-6Cycloalkyl, 3-6 membered heterocyclyl, C6-10Aryl or 5-12 membered heteroaryl are each optionally substituted with one or more substituents independently selected from: hydrogen, deuterium, halogen, cyano, hydroxy and C1-6An alkoxy group.
In a preferred embodiment, R3Is C1-6Alkyl radical, said C1-6Alkyl is optionally substituted with one or more substituents independently selected from: hydrogen, deuterium, hydroxy, cyano and C1-4An alkoxy group. In a more preferred embodiment, R3Is C1-4Alkyl radical, said C1-4Alkyl is optionally substituted with one or more substituents independently selected from: hydrogen, hydroxy and C1-4An alkoxy group. In a more preferred embodiment, R3Is C1-4Alkyl radical, said C1-4Alkyl is optionally substituted with one or more substituents independently selected from: hydrogen, hydroxy, methoxy and ethoxy. In a particularly preferred embodiment, R3Selected from n-propyl and CH3OCH 2CH 2-。
In a preferred embodiment, R4、R 5Each independently selected from hydrogen, deuterium, C1-6Alkyl radical, C3-6Cycloalkyl and 3-6 membered heterocyclyl, wherein said C is1-6Alkyl is optionally substituted with one or more substituents independently selected from: hydrogen, deuterium, halogen, hydroxy and C1-6An alkoxy group. In a more preferred embodiment, R4、R 5Each independently selected from hydrogen,Deuterium, halogen, C1-6Alkyl, halo C1-6Alkyl and C3-6A cycloalkyl group. In a more preferred embodiment, R4、R 5Each independently selected from hydrogen, fluorine, C1-6Alkyl and fluoro C1-6An alkyl group. In a particularly preferred embodiment, R4、R 5Each independently selected from hydrogen, fluoro, methyl, trifluoromethyl and cyclopropyl.
In a preferred embodiment, A is
Figure PCTCN2020081558-APPB-000006
In a more preferred embodiment, A is
Figure PCTCN2020081558-APPB-000007
In a particularly preferred embodiment, a is selected from:
Figure PCTCN2020081558-APPB-000008
according to some embodiments of the invention, the compound is a compound of formula III,
Figure PCTCN2020081558-APPB-000009
wherein R is1、R 2A and n are as defined above.
According to some embodiments of the invention, the compound is a compound of formula IV,
Figure PCTCN2020081558-APPB-000010
wherein R is1、R 2、R 3、R 4X and n are as defined above.
According to some embodiments of the invention, the compound is a compound of formula IV-A or IV-B,
Figure PCTCN2020081558-APPB-000011
wherein R is1、R 2、R 3、R 4X and n are as defined above.
According to some embodiments of the invention, the compound of the invention is selected from:
Figure PCTCN2020081558-APPB-000012
Figure PCTCN2020081558-APPB-000013
Figure PCTCN2020081558-APPB-000014
according to some embodiments of the invention, the compound of the invention is selected from:
Figure PCTCN2020081558-APPB-000015
Figure PCTCN2020081558-APPB-000016
Figure PCTCN2020081558-APPB-000017
Figure PCTCN2020081558-APPB-000018
Figure PCTCN2020081558-APPB-000019
Figure PCTCN2020081558-APPB-000020
preparation method
It is a further object of the present invention to provide a process for the preparation of the compounds of the present invention, which process is carried out according to the following synthetic route:
Figure PCTCN2020081558-APPB-000021
wherein R is1、R 2、R 9、R 10A, Y and n are as defined above; and LG is a leaving group.
According to some embodiments of the invention, the method is performed according to the following synthetic route:
Figure PCTCN2020081558-APPB-000022
wherein R is1、R 2、R 9、R 10A and n are as defined above; and LG is a leaving group.
According to some embodiments of the invention, LG is halogen, methanesulfonyl or p-toluenesulfonyl. In a preferred embodiment, LG is bromo.
The method comprises the following steps:
the method comprises the following steps:
reacting a compound of formula a with a compound of formula b in a suitable organic solvent at a suitable temperature in the presence of a reducing agent under acidic conditions for a time to provide a compound of formula c.
The reducing agent may be selected from sodium borohydride, potassium borohydride, sodium cyanoborohydride and sodium triacetoxyborohydride. Preferably, the reducing agent is sodium cyanoborohydride or sodium triacetoxyborohydride.
The acid used to form the acidic conditions may be selected from hydrochloric acid, acetic acid and trifluoroacetic acid. Preferably, the acid is trifluoroacetic acid.
The reaction temperature is preferably-50 to 100 deg.C, more preferably-20 to 70 deg.C.
The reaction time is preferably 1 to 24 hours, for example 1 to 12 hours.
The organic solvent may be selected from linear or cyclic ethers (e.g., tetrahydrofuran, diethyl ether, etc.), halogenated hydrocarbons (e.g., dichloromethane, chloroform, 1, 2-dichloroethane, etc.), nitriles (e.g., acetonitrile, etc.), N-methylpyrrolidone, dimethylformamide, dimethylacetamide, 1, 4-dioxane, dimethylsulfoxide, or any combination thereof. Preferably, the organic solvent is dichloromethane, 1, 4-dioxane or 1, 2-dichloroethane.
Step two:
reacting a compound of formula c with a compound of formula d in a suitable solvent at a suitable temperature in the presence of a suitable base, a phosphorus ligand and a suitable catalyst for a time to give a compound of formula e.
The base can be selected from organic bases such as triethylamine, DIPEA, pyridine, NMM, etc., or Na2CO 3、K 2CO 3、NaHCO 3、Cs 2CO 3And inorganic bases such as potassium phosphate. Preferably, the base is potassium phosphate.
The phosphorus ligand may be selected from RuPhos, XPhos, SPhos, CPhos and tri-tert-butylphosphine tetrafluoroborate. Preferably, the phosphorus ligand is tri-tert-butylphosphine tetrafluoroborate.
The catalyst is a palladium catalyst, such as tetrakis (triphenylphosphine) palladium, [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex, bis (triphenylphosphine) palladium dichloride or palladium acetate. Preferably, the catalyst is palladium acetate.
The reaction temperature is preferably 20 to 150 ℃, more preferably 70 to 100 ℃.
The reaction time is preferably 1 to 24 hours, for example 1 to 3 hours.
The solvent may be selected from benzene, toluene, xylene, 1, 4-dioxane, N-dimethylformamide, or a mixed solvent of the above organic solvents and water, for example, a mixed solvent of toluene and water. Preferably, the solvent is a mixed solvent of toluene and water.
Step three:
reacting the compound of formula e with a reducing agent in a suitable organic solvent at a suitable temperature for a time to provide the compound of formula f.
The reducing agent may be selected from lithium aluminum hydride, aluminum isobutyl alkoxide, sodium borohydride, potassium borohydride, and sodium borohydride-zinc chloride. Preferably, the reducing agent is lithium aluminum hydride.
The reaction temperature is preferably-70 ℃ to 80 ℃, more preferably 0 ℃ to 25 ℃.
The reaction time is preferably 1 to 24 hours, for example 1 to 4 hours.
The organic solvent may be selected from alcohols (e.g., methanol, ethanol, etc.), ethers (e.g., tetrahydrofuran, diethyl ether, t-butyl methyl ether, 1, 4-dioxane), halogenated hydrocarbons (e.g., dichloromethane, chloroform, 1, 2-dichloroethane, etc.), and the like. Preferably, the organic solvent is tetrahydrofuran.
Step four:
reacting the compound of formula f with an oxidizing agent in a suitable organic solvent at a suitable temperature for a time to provide the compound of formula g.
The oxidant may be selected from manganese dioxide, pyridinium chlorochromate, chromium trioxide, Dess-Martin oxidant (periodinane 1,1, 1-triacetoxy-1, 1-dihydro-1, 2-phenyliodoyl-3- (1H) -one), IBX oxidant (2-iodoxybenzoic acid) and Swern oxidant (dimethyl sulfoxide). Preferably, the oxidant is manganese dioxide or IBX oxidant.
The reaction temperature is preferably 0 to 100 deg.C, more preferably 20 to 40 deg.C.
The reaction time is preferably 1 to 24 hours, for example 12 to 24 hours.
The organic solvent may be selected from the group consisting of linear or cyclic ethers (e.g., tetrahydrofuran, diethyl ether, etc.), halogenated hydrocarbons (e.g., dichloromethane, chloroform, 1, 2-dichloroethane, etc.), nitriles (e.g., acetonitrile, etc.), N-methylpyrrolidone, dimethylformamide, dimethylacetamide, 1, 4-dioxane, dimethylsulfoxide, and any combination thereof. Preferably, the organic solvent is dichloromethane, chloroform or dimethyl sulfoxide.
Step five:
reacting a compound of formula g with a compound of formula h in a suitable organic solvent at a suitable temperature and in the presence of an oxidizing agent under acidic conditions for a time to provide a compound of formula I.
The oxidant may be selected from H2O 2Sodium hypochlorite, t-butanol peroxide, peracetic acid, perbenzoic acid, m-CPBA (m-chloroperoxybenzoic acid), paranitroperoxybenzoic acid, and peroxytrifluoroacetic acid. Preferably, the oxidant is H2O 2
The acid used to form the acidic conditions may be selected from the group consisting of hydrochloric acid, acetic acid and trifluoroacetic acid. Preferably, the acid is hydrochloric acid.
The reaction temperature is preferably 0 to 100 deg.C, more preferably 20 to 40 deg.C.
The reaction time is preferably 1 to 24 hours, for example 1 to 2 hours.
The organic solvent may be selected from the group consisting of linear or cyclic ethers (e.g., tetrahydrofuran, diethyl ether, etc.), halogenated hydrocarbons (e.g., dichloromethane, chloroform, 1, 2-dichloroethane, etc.), nitriles (e.g., acetonitrile, etc.), N-methylpyrrolidone, dimethylformamide, dimethylacetamide, 1, 4-dioxane, dimethylsulfoxide, and any combination thereof. Preferably, the organic solvent is dioxane, tetrahydrofuran or acetonitrile.
Pharmaceutical composition and kit
It is another object of the present invention to provide a pharmaceutical composition comprising an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, and one or more pharmaceutically acceptable carriers. In the present invention, the purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of an active ingredient, and exert biological activity.
By "pharmaceutically acceptable carrier" in the context of the present invention is meant a diluent, adjuvant, excipient, or vehicle that is administered together with a therapeutic agent and which is, within the scope of sound medical judgment, suitable for contact with the tissues of humans and/or other animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio.
Pharmaceutically acceptable carriers that may be used in the pharmaceutical compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (e.g., human serum albumin), buffer substances (e.g., phosphates), glycerol, sorbic acid, 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, zinc salts), colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulosic materials, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, beeswax, lanolin, and the like.
The pharmaceutical compositions of the present invention may act systemically and/or locally. For this purpose, they can be administered by a suitable route, for example by the oral or parenteral route. The parenteral routes include, but are not limited to, topical, intravenous, subcutaneous, intraarterial, intradermal, transdermal, rectal, intracranial, intraperitoneal, intranasal, intramuscular routes, or as inhalants. The pharmaceutical compositions may optionally be administered in combination with other agents that have at least some effect in the treatment of various diseases.
For these routes of administration, the pharmaceutical compositions of the present invention may be administered in suitable dosage forms. Such dosage forms include, but are not limited to, tablets, capsules, lozenges, hard candies, powders, sprays, creams, ointments, suppositories, gels, pastes, lotions, ointments, aqueous suspensions, injectable solutions, elixirs, syrups, and the like.
When administered orally, the pharmaceutical compositions of the present invention may be formulated in any orally acceptable dosage form, including, but not limited to, tablets, capsules, aqueous solutions, or aqueous suspensions. Among them, carriers which can be used for tablets generally include lactose and corn starch, and additionally, a lubricant such as magnesium stearate can be added. Carriers that can be used in capsules generally include lactose and dried corn starch. Aqueous suspensions, in turn, typically include emulsifying and suspending agents. Optionally, some sweetener, aromatic or colorant may be added into the above oral preparation.
When administered topically, the pharmaceutical compositions of the present invention may be formulated in a suitable ointment, lotion, or cream form, wherein the active ingredient is added to one or more carriers. Carriers useful in ointments include, but are not limited to: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyethylene oxide, polypropylene oxide, emulsifying wax and water. Carriers that may be used in lotions or creams include, but are not limited to: mineral oil, sorbitan monostearate (Tween 60), cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
The pharmaceutical compositions of the present invention may also be administered in the form of sterile injectable preparations, including sterile injectable aqueous or oleaginous suspensions or sterile injectable solutions. Carriers and/or solvents that can be used for sterile injectable preparations include water, ringer's solution, isotonic sodium chloride solution and the like. In addition, the sterilized fixed oil may also be employed as a solvent or suspending medium, such as a monoglyceride or diglyceride.
The compound of the invention may be present in the pharmaceutical composition in an amount or dose of from about 0.01mg to about 1000mg, suitably 0.1-800mg, preferably 0.5-500mg, more preferably 1-350mg, especially 5-250 mg.
According to one embodiment of the invention, the pharmaceutical composition may further comprise one or more other agents useful for the prevention or treatment of diseases or conditions mediated by CCR2 and/or CCR5, in particular non-alcoholic fatty liver disease (NAFLD).
It is another object of the present invention to provide a pharmaceutical kit comprising a compound of the present invention or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g. hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of the present invention.
In another aspect, the invention provides a kit comprising:
a) a first container comprising at least one compound of the invention or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, as a first therapeutic agent, or a pharmaceutical composition of the invention as a first pharmaceutical composition;
b) optionally a second container comprising at least one further therapeutic agent as a second therapeutic agent, or a pharmaceutical composition comprising said further therapeutic agent as a second pharmaceutical composition; and
c) optionally product instructions.
Methods of treatment and uses
It is another object of the present invention to provide a compound of the present invention or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g. hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of the present invention, or a kit of the present invention, for use in the prevention or treatment of a disease or condition mediated by CCR2 and/or CCR5, in particular non-alcoholic fatty liver disease (NAFLD).
It is another object of the present invention to provide the use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g. hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of the present invention, or a kit of the present invention, in the manufacture of a medicament for the prevention or treatment of a disease or condition mediated by CCR2 and/or CCR5, in particular non-alcoholic fatty liver disease (NAFLD).
It is another object of the present invention to provide a method for the prevention or treatment of diseases or disorders mediated by CCR2 and/or CCR5, in particular nonalcoholic fatty liver disease (NAFLD), comprising administering to a subject in need thereof an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of the present invention, or a kit of the present invention.
It is another object of the present invention to provide a method for preventing or treating a disease or disorder mediated by CCR2 and/or CCR5, in particular nonalcoholic fatty liver disease (NAFLD), comprising administering to a subject in need thereof an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of the present invention, or a kit of the present invention, and one or more other agents useful for preventing or treating a disease or disorder mediated by CCR2 and/or CCR5, in particular nonalcoholic fatty liver disease (NAFLD).
The dosing regimen may be adjusted to provide the best desired response. For example, a single bolus may be administered, several divided doses may be administered over time, or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is noted that dosage values may vary with the type and severity of the condition being alleviated, and may include single or multiple doses. It is further understood that for any particular individual, the specific dosage regimen will be adjusted over time according to the individual need and the professional judgment of the person administering the composition or supervising the administration of the composition.
In some embodiments of the invention, suitable in vitro or in vivo assays are performed to determine the effectiveness of the pharmaceutical compositions of the invention and whether administration is suitable for treating a disease or medical condition in an individual. Typically, an effective amount of a compound of the invention sufficient to achieve a prophylactic or therapeutic effect is from about 0.001 mg/kg body weight/day to about 10000 mg/kg body weight/day. Suitably, the dose is from about 0.01 mg/kg body weight/day to about 1000mg/kg body weight/day. The dosage range may be about 0.01 to 1000mg/kg of subject body weight per day, every second day, or every third day, more usually 0.1 to 500mg/kg of subject body weight. Exemplary treatment regimens are once every two days or once a week or once a month. The pharmaceutical composition is typically administered multiple times, and the interval between single doses may be daily, weekly, monthly or yearly. Alternatively, the pharmaceutical composition may be administered in the form of a sustained release formulation, in which case less frequency of administration is required. The dose and frequency will vary depending on the half-life of the formulation in the subject. It may also vary depending on whether prophylactic or therapeutic treatment is carried out. In prophylactic applications, relatively low doses are administered chronically at relatively infrequent intervals. In therapeutic applications, it is sometimes desirable to administer relatively high doses at relatively short intervals until the progression of the disease is delayed or halted, and preferably until the individual exhibits a partial or complete improvement in the symptoms of the disease, after which a prophylactic regimen may be administered to the patient.
As used herein, "individual" includes a human or non-human animal. Exemplary human individuals include human individuals (referred to as patients) having a disease (e.g., a disease described herein) or normal individuals. "non-human animals" in the context of the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
Examples
In order to make the objects and technical solutions of the present invention clearer, embodiments of the present invention will be described in detail below with reference to examples. It will be understood by those skilled in the art that the following examples are illustrative of the present invention only and should not be taken as limiting the scope of the invention. The examples, in which the specific conditions are not specified, were conducted under the conventional conditions or conditions recommended by the manufacturer. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products commercially available.
In the conventional syntheses as well as in the examples and intermediate syntheses, the meanings of the abbreviations are shown in the following table.
Figure PCTCN2020081558-APPB-000023
The structures of the compounds described in the following examples were determined by NMR spectroscopy (1HNMR) or Mass Spectrometry (MS).
Nuclear magnetic resonance (1HNMR) is a Bruker 400MHz nuclear magnetic resonance instrument; the solvent was determined to be deuterated methanol (CD)3OD), deuterated chloroform (CDCl)3) Or hexadeutero dimethyl sulfoxide (DMSO-d)6) (ii) a The internal standard substance is Tetramethylsilane (TMS).
Abbreviations in Nuclear Magnetic Resonance (NMR) spectra mentioned in the examples are as follows:
s: unimodal (singlet), d: doublet (doublt), t: triplet (triplet), q: quartet (quartz), dd: doublet (doubledoubledoublet), qd: quartet doubtet, ddd: double doublet (double doublet), ddt: double double triplet (double double triple), dddd: double double doublet (double double doublet), m: multiplet (multiplex), br: broad (broad), J: coupling constant, Hz: hertz, DMSO-d6: deuterated dimethyl sulfoxide.
Chemical shifts (δ) are given in units of parts per million (ppm).
The Mass Spectrometer (MS) was an Agilent (ESI) mass spectrometer model Agilent 6120B.
EXAMPLE 1 Synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -1- ((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde
Figure PCTCN2020081558-APPB-000024
The method comprises the following steps: (E) synthesis of methyl (E) -8-bromo-1- ((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carboxylate (Compound 1-2)
Methyl 8-bromo-1, 2,3, 4-tetrahydrobenzo [ b ] azepino (m) tetraene-5-carboxylate (compound 1-1, 300mg, 1mmol), tetrahydropyran-4-carbaldehyde (560mg, 5mmol) were added to dichloromethane (20 mL). Trifluoroacetic acid was added dropwise thereto until the reaction system was acidic (pH 1.0). The reaction was incubated at 25 ℃ for 30 min. Sodium triacetoxyborohydride (1.8g, 8.5mmol) was added to the reaction system, and the reaction was maintained at 25 ℃ for 12 hours. Distilled water (100mL) was added to the reaction mixture, and the mixture was extracted with methylene chloride (30 mL. times.3). The organic phases were combined, washed with saturated sodium bicarbonate (100mL × 1), saturated brine (100mL × 1) and water (100mL × 1) in this order, dried over anhydrous sodium sulfate, and purified by column chromatography (PE: EA ═ 4:1) to give the title compound (310 mg).
ESI-MS(m/z):394.2[M+H] +
Step two: (E) synthesis of methyl [8- (4- (2-butoxyethoxy) phenyl ] -1- ((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacycloocta (m) tetraene-5-carboxylate (Compound 1-3)
Compound 1-2(280mg, 0.71mmol), 2- (4- (2-butoxyethoxy) phenyl) -4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan (228mg, 0.71mmol), palladium acetate (16mg, 0.071mmol), potassium phosphate (452mg, 2.13mmol), tri-tert-butylphosphine tetrafluoroborate (103mg, 0.355mmol) were added to a toluene/water (10mL/1mL) mixed solvent and reacted with a microwave at 100 ℃ for 3.0 hours under nitrogen protection. After cooling to room temperature, water (100mL) was added and extracted with ethyl acetate (30 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography (PE: EA ═ 4:1) to give the title compound (320 mg).
ESI-MS(m/z):508.3[M+H] +
Step three: (E) synthesis of [8- (4- (2-butoxyethoxy) phenyl) -1- ((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclooct (m) tetraen-5-yl ] methanol (Compound 1-4)
Under the protection of nitrogen, LiAlH4(45mg,1.2mmol) was added to dry tetrahydrofuran (10 mL). A solution of compound 1-3(200mg, 0.4mmol) in anhydrous tetrahydrofuran (10mL) was added dropwise and reacted at 25 ℃ for 4 hours. The reaction was terminated by dropwise addition of a saturated ammonium chloride solution, and water (20mL) was added to conduct extraction with ethyl acetate (30 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give the title compound (180 mg). It was used directly in the next reaction without purification.
ESI-MS(m/z):480.3[M+H] +
Step four: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -1- ((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacycloocta (m) tetraene-5-carbaldehyde (Compound 1-5)
Compounds 1-4(120mg, 0.25mmol) were added to dimethyl sulfoxide (10 mL). 2-iodoxybenzoic acid (70mg,0.25mmol) was added to the reaction system and reacted at 25 ℃ for 12 h. Saturated sodium sulfite solution is added dropwise until starch-potassium iodide paper does not turn blue. Water (50mL) was added, and the mixture was extracted with ethyl acetate (50 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography (PE: EA ═ 3:1) to give the title compound (100 mg).
ESI-MS(m/z):478.3[M+H] +
EXAMPLE 2 (E) -8- (4- (2-butoxyethoxy) phenyl) -5- (((1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- ((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocine (m) tetraene (Compound 1)
Figure PCTCN2020081558-APPB-000025
The method comprises the following steps: synthesis of 2-nitro-4- ((1-propyl-1H-imidazol-5-yl) methylthio) aniline (Compound 2-2)
4-amino-3-nitrothiophenol (Compound 2-1, 3.11g, 18.28mmol) was added to a mixture of triethylamine (5.1mL), isopropanol (56mL) and water (3mL) and cooled to-15 ℃. An aqueous solution (18mL) of 5- (chloromethyl) -1-propyl-1H-imidazole hydrochloride (2.97g,15.23mmol) was slowly added dropwise, and the reaction was incubated for 2H after completion of the addition, and then the completion of the reaction was detected by LC-MS. The isopropanol was removed by rotary evaporation, water (100mL) was added thereto to terminate the reaction, and the mixture was extracted with dichloromethane (100 mL. times.2). The organic phase was washed with an aqueous potassium carbonate solution (100mL × 2) and a saturated brine (100mL × 2), respectively, dried over anhydrous sodium sulfate, the drying agent was filtered off, the solvent was evaporated under reduced pressure, and flash column chromatography (DCM: MeOH ═ 10:1) was performed to obtain the title compound (3.31 g).
ESI-MS(m/z):293.2[M+H] +
Step two: synthesis of 4- (1-propyl-1H-imidazol-5-yl) methylthio-1, 2-phenylenediamine (Compound 2-3)
Compound 2-2(2.92g, 10mmoL) was added to ethanol (50mL), and sodium dithionite (3.48g, 20mmoL) was added thereto, followed by reflux reaction for 2 hours. It was cooled to dryness, water (100mL) was added, and the mixture was extracted with ethyl acetate (100 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated and purified by column chromatography (DCM: MeOH ═ 10:1) to give the title compound (2.1 g).
ESI-MS(m/z):263.2[M+H] +
Step three: synthesis of (4- ((1-propyl-1H-imidazol-5-yl) methyl) sulfinyl-1, 2-phenylenediamine (Compound 2-4)
Compound 2-3(263mg, 1mmol) was added to dichloromethane (10mL), cooled to-10 deg.C, and a solution of m-chloroperoxybenzoic acid (189.8mg, 1.1mmol) in dichloromethane (40mL) was slowly added dropwise thereto, after dropping, the reaction was incubated for 10 minutes and checked for completion by LC-MS. Saturated aqueous sodium carbonate (5mL) was added to the system to terminate the reaction, and the mixture was stirred for 20 minutes and warmed to 10 ℃. A saturated sodium sulfite solution (10mL) was added to the system, and the mixture was stirred for 10 minutes. The layers were separated and the aqueous layer was extracted with dichloromethane (10 mL. times.3). The organic phases were combined, washed with saturated brine (10mL × 2), dried over anhydrous sodium sulfate, filtered off the drying agent, the solvent was evaporated under reduced pressure, and purified by column chromatography (DCM: MeOH ═ 10:1) to give the title compound (213 mg).
ESI-MS(m/z):279.2[M+H] +
Step four: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -5- (((1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- ((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocin (m) tetraene (Compound 1)
Compound 2-4(278mg, 1mmol) and compound 1-5(478mg, 1mmol) prepared in example 1 were added to acetonitrile (5mL), followed by 30% H2O 2Aqueous (0.7. mu.L, 7mmol) and 37% aqueous HCl (0.3. mu.L, 3.5mmol) were reacted at room temperature for 2 hours. A saturated solution of sodium sulfite (10mL) was added to the reaction mixture, followed by extraction with ethyl acetate (20 mL. times.3). The combined organic phases were dried over anhydrous sodium sulfate, concentrated and purified by TLC prep. (DCM: MeOH ═ 20: 1-10: 1, R)f0.3-0.6) to give the title compound (110 mg).
The structure is characterized as follows:
ESI-MS(m/z):736.4[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.88(s,1H),7.82(s,1H),7.73(d,J=5.2Hz,1H),7.56(dd,J=34.2,7.1Hz,4H),7.44(d,J=9.1Hz,2H),7.37–7.29(m,1H),7.00(d,J=8.3Hz,2H),6.91(d,J=8.7Hz,1H),6.45(s,1H),4.36(t,J=13.5Hz,1H),4.19(d,J=14.0Hz,1H),4.15–4.07(m,2H),3.88(d,J=8.1Hz,2H),3.82–3.67(m,4H),3.47(dd,J=16.8,10.4Hz,4H),3.29(t,J=11.6Hz,2H),3.20(d,J=6.6Hz,2H),2.78(s,2H),2.14–1.97(m,1H),1.63(d,J=9.4Hz,4H),1.57–1.47(m,4H),1.34(dt,J=15.0,7.6Hz,4H),0.89(t,J=7.3Hz,3H),0.79(t,J=6.1Hz,3H)。
EXAMPLE 3 (E) -8- (4- (2-butoxyethoxy) phenyl) -5- ((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- ((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocine (m) tetraene (Compound 2)
Figure PCTCN2020081558-APPB-000026
The method comprises the following steps: synthesis of 2-nitro-4- (((4-methyl-1-propyl-1H-imidazol-5-yl) methylthio) aniline (Compound 3-1)
4-amino-3-nitrothiophenol (Compound 2-1, 3.11g, 18.28mmol) was added to a mixture of triethylamine (5.1mL), isopropanol (56mL) and water (3mL) and cooled to-15 ℃. Slowly adding an aqueous solution (18mL) of 5- (chloromethyl) -4-methyl-1-propyl-1H-imidazole hydrochloride (3.18g,15.23mmol) dropwise, preserving the temperature for 2H after finishing the dropwise addition, and detecting the completion of the reaction by LC-MS. The isopropanol was removed by rotary evaporation, water (100mL) was added thereto to terminate the reaction, dichloromethane (100mL × 2) was used for extraction, the organic phase was washed with an aqueous potassium carbonate solution (100mL × 2) and a saturated brine (100mL × 2), respectively, dried over anhydrous sodium sulfate, the drying agent was filtered off, the solvent was evaporated under reduced pressure, and flash column chromatography (DCM: MeOH ═ 10:1) was performed to obtain the title compound (3.34 g).
ESI-MS(m/z):306.2[M+H] +
Step two: synthesis of 4- ((4-methyl-1-propyl-1H-imidazol-5-yl) methylthio) -1, 2-phenylenediamine (Compound 3-2)
Compound 3-1(3.06g, 10mmol) was added to ethanol (50mL), to which was added sodium dithionite (3.48g, 20mmol), and the reaction was refluxed for 2 hours. It was cooled to dryness, water (100mL) was added, and the mixture was extracted with ethyl acetate (100 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated and purified by column chromatography (DCM: MeOH ═ 10:1) to give the title compound (2.2 g).
ESI-MS(m/z):277.2[M+H] +
Step three: synthesis of 4- (((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1, 2-phenylenediamine (Compound 3-3)
Compound 3-2(278mg, 1mmoL) was added to dichloromethane (10mL), cooled to-10 deg.C, and a solution of m-chloroperoxybenzoic acid (189.8mg, 1.1mmoL) in dichloromethane (40mL) was slowly added dropwise. After the dripping is finished, the reaction is kept for 10 minutes, and the reaction is finished through LC-MS detection. Saturated aqueous sodium carbonate (5mL) was added to the system to terminate the reaction, and the mixture was stirred for 20 minutes and warmed to 10 ℃. A saturated sodium sulfite solution (10mL) was added to the system, and the mixture was stirred for 10 minutes. The layers were separated and the aqueous layer was extracted with dichloromethane (10 mL. times.3). The organic phases were combined, washed with saturated brine (10mL × 2), dried over anhydrous sodium sulfate, filtered off the drying agent, the solvent was evaporated under reduced pressure, and purified by column chromatography (DCM: MeOH ═ 10:1) to give the title compound (213 mg).
ESI-MS(m/z):293.2[M+H] +
Step four: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -5- ((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- ((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocine (m) tetraene (Compound 2)
Compounds 3-3(278mg, 1mmol) and compounds 1-5(478mg, 1mmol) were added to acetonitrile (10mL), followed by 30% H2O 2Aqueous (0.7. mu.L, 7mmol) and 37% aqueous HCl (0.3. mu.L, 3.5mmol) were reacted at room temperature for 2 hours. A saturated solution of sodium sulfite (10mL) was added to the reaction mixture, followed by extraction with ethyl acetate (20 mL. times.3). The combined organic phases were dried over anhydrous sodium sulfate, concentrated and purified by TLC prep. (DCM: MeOH ═ 20: 1-10: 1, R)f0.3-0.6) to give the title compound (110 mg).
The structure is characterized as follows:
ESI-MS(m/z):750.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.80(d,J=22.8Hz,1H),7.80(s,1H),7.71–7.68(m,1H),7.60–7.40(m,6H),7.26(t,J=9.8Hz,1H),6.98(d,J=8.0Hz,2H),6.90(d,J=8.4Hz,1H),4.32–4.14(m,2H),4.09(s,2H),3.90–3.80(m,2H),3.82–3.73(m,2H),3.69(s,2H),3.50(s,2H),3.45(t,J=6.4Hz,2H),3.27(t,J=11.6Hz,2H),3.18(d,J=4.8Hz,2H),2.81–2.69(m,2H),2.10-2.00(m,1H),1.68–1.57(m,4H),1.56–1.41(m,7H),1.38–1.25(m,4H),0.88(t,J=7.2Hz,3H),0.80(t,J=7.2Hz,3H)。
Figure PCTCN2020081558-APPB-000027
and (3) separating chiral isomers of the prepared compound 2 by using an HPLC method through a chiral chromatographic column, and collecting corresponding components. The solvent was removed by rotary evaporation to give optically pure compounds 2-A (retention time 7.272min) and 2-B (retention time 10.109min), which are the optical isomers of Compound 2. The preparation conditions are as follows: chiral chromatographic column: CHIRALCEL OZ-H (OZH0CD-VF 004); chiral chromatographic column size: 0.46cm I.D. x 15cm L; mobile phase: MeOH/ACN 90/10 (V/V); flow rate: 1.0 mL/min; wavelength: UV 254 nm.
The structural characterization is as follows:
compound 2-a:
ESI-MS(m/z):750.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.80(d,J=22.8Hz,1H),7.80(s,1H),7.71–7.68(m,1H),7.60–7.40(m,6H),7.26(t,J=9.8Hz,1H),6.98(d,J=8.0Hz,2H),6.90(d,J=8.4Hz,1H),4.32–4.14(m,2H),4.09(s,2H),3.90–3.80(m,2H),3.82–3.73(m,2H),3.69(s,2H),3.50(s,2H),3.45(t,J=6.4Hz,2H),3.27(t,J=11.6Hz,2H),3.18(d,J=4.8Hz,2H),2.81–2.69(m,2H),2.10-2.00(m,1H),1.68–1.57(m,4H),1.56–1.41(m,7H),1.38–1.25(m,4H),0.88(t,J=7.2Hz,3H),0.80(t,J=7.2Hz,3H)。
compound 2-B:
ESI-MS(m/z):750.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.80(d,J=22.8Hz,1H),7.80(s,1H),7.71–7.68(m,1H),7.60–7.40(m,6H),7.26(t,J=9.8Hz,1H),6.98(d,J=8.0Hz,2H),6.90(d,J=8.4Hz,1H),4.32–4.14(m,2H),4.09(s,2H),3.90–3.80(m,2H),3.82–3.73(m,2H),3.69(s,2H),3.50(s,2H),3.45(t,J=6.4Hz,2H),3.27(t,J=11.6Hz,2H),3.18(d,J=4.8Hz,2H),2.81–2.69(m,2H),2.10-2.00(m,1H),1.68–1.57(m,4H),1.56–1.41(m,7H),1.38–1.25(m,4H),0.88(t,J=7.2Hz,3H),0.80(t,J=7.2Hz,3H)。
EXAMPLE 4 Synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -1-isobutyl-1, 2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde
Figure PCTCN2020081558-APPB-000028
The method comprises the following steps: (E) synthesis of methyl (4-1) -8-bromo-1-isobutyl-1, 2,3, 4-tetrahydrobenzo [ b ] azacyclo-tetraene-5-carboxylate
Methyl 8-bromo-1, 2,3, 4-tetrahydrobenzo [ b ] azepino (m) tetraene-5-carboxylate (compound 1-1, 300mg, 1mmol), isobutyraldehyde (360mg, 5mmol) were added to dichloromethane (20 mL). Trifluoroacetic acid was added dropwise thereto until the reaction system was acidic (pH 1.0). The reaction was incubated at 25 ℃ for 30 min. Sodium triacetoxyborohydride (1.8g, 8.5mmol) was added to the reaction system, and the reaction was maintained at 25 ℃ for 12 hours. Distilled water (100mL) was added to the reaction mixture, and the mixture was extracted with methylene chloride (30 mL. times.3). The organic phases were combined, washed with saturated sodium bicarbonate (100mL × 1), saturated brine (100mL × 1) and water (100mL × 1), dried over anhydrous sodium sulfate, and purified by column chromatography (PE: EA ═ 4:1) to give the title compound (300 mg).
ESI-MS(m/z):352.2[M+H] +
Step two: (E) synthesis of methyl [8- (4- (2-butoxyethoxy) phenyl ] -1-isobutyl-1, 2,3, 4-tetrahydrobenzo [ b ] azocyclocin (m) tetraene-5-carboxylate (Compound 4-2)
Compound 4-1(250mg, 0.71mmol), 2- (4- (2-butoxyethoxy) phenyl) -4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan (228mg, 0.71mmol), palladium acetate (16mg, 0.071mmol), potassium phosphate (452mg, 2.13mmol), tri-tert-butylphosphine tetrafluoroborate (103mg, 0.355mmol) were added to a toluene/water (10mL/1mL) mixed solvent and reacted with a microwave at 100 ℃ for 3.0 hours under nitrogen protection. After cooling to room temperature, water (100mL) was added and extracted with ethyl acetate (30 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography (PE: EA ═ 4:1) to give the title compound (290 mg).
ESI-MS(m/z):466.3[M+H] +
Step three: (E) synthesis of [8- (4- (2-butoxyethoxy) phenyl) -1-isobutyl-1, 2,3, 4-tetrahydrobenzo [ b ] azocyclooct (m) tetraen-5-yl ] methanol (Compound 4-3)
Under the protection of nitrogen, LiAlH4(45mg,1.2mmol) was added to dry tetrahydrofuran (10 mL). A solution of Compound 4-2(186mg, 0.4mmol) in anhydrous tetrahydrofuran (10mL) was added dropwise and reacted at 25 ℃ for 4 hours. The reaction was terminated by dropwise addition of a saturated ammonium chloride solution, and water (20mL) was added to conduct extraction with ethyl acetate (30 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give the title compound (160 mg). It was used directly in the next reaction without purification.
ESI-MS(m/z):438.3[M+H] +
Step four: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -1-isobutyl-1, 2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde (Compound 4-4)
Compound 4-3(110mg, 0.25mmol) was added to dimethyl sulfoxide (10 mL). 2-iodoxybenzoic acid (70mg,0.25mmol) was added to the reaction system and reacted at 25 ℃ for 12 h. Saturated sodium sulfite solution is added dropwise until starch-potassium iodide paper does not turn blue. Then, water (50mL) was added, and the mixture was extracted with ethyl acetate (50 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography (PE: EA ═ 3:1) to give the title compound (100 mg).
ESI-MS(m/z):436.3[M+H] +
EXAMPLE 5 (E) -8- (4- (2-butoxyethoxy) phenyl) -1-isobutyl-5- (6- (((1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ b ] imidazol-2-yl) -1,2,3, 4-tetrahydrobenzo [ b ] azocin (m) tetraene (Compound 3)
Figure PCTCN2020081558-APPB-000029
The compound 2-4(278mg, 1mmol) prepared in example 2 and (E) -8- (4- (2-butoxyethoxy) phenyl) -1-isobutyl-1, 2,3, 4-tetrahydrobenzo [ b ] prepared in example 4 were mixed]Azacyclooctan (m) tetraene-5-carboxaldehyde (compound 4-4, 435mg, 1mmol) is added to acetonitrile (10mL), to which 30% H is added in sequence2O 2Aqueous (0.7. mu.L, 7mmol) and 37% aqueous HCl (0.3. mu.L, 3.5mmol) were reacted at room temperature for 2 hours. A saturated solution of sodium sulfite (10mL) was added to the reaction mixture, followed by extraction with ethyl acetate (20 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by preparative liquid chromatography to give the title compound (107 mg).
The structure is characterized as follows:
ESI-MS(m/z):694.2[M+H] +
1H NMR(400MHz,CDCl 3)δ8.16(s,1H),7.90(d,J=8.7Hz,2H),7.62(s,2H),7.58–7.50(m,4H),7.44(dd,J=16.8,8.7Hz,3H),7.00(dd,J=14.7,8.9Hz,4H),6.48(s,1H),4.35(d,J=4.32Hz,1H),4.13–4.07(m,3H),3.87–3.78(m,2H),3.71(d,J=4.6Hz,2H),3.54(d,J=8.8Hz,3H),3.46(t,J=6.5Hz,4H),3.07–3.01(m,1H),2.67(s,1H),2.39(s,1H),1.91(s,1H),1.77(s,1H),1.65(d,J=7.3Hz,2H),1.49(d,J=7.9Hz,4H),1.45(s,3H),1.39(s,3H),1.36–1.30(m,3H),1.00(dd,J=7.1,4.2Hz,5H),0.88(t,J=7.4Hz,3H),0.81(t,J=7.3Hz,3H)。
EXAMPLE 6 Synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -1- (2-fluoroethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde
Figure PCTCN2020081558-APPB-000030
The method comprises the following steps: (E) synthesis of methyl (E) -8-bromo-1- (2-fluoroethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carboxylate (Compound 6-1)
Methyl 8-bromo-1, 2,3, 4-tetrahydrobenzo [ b ] azepino (m) tetraene-5-carboxylate (compound 1-1, 300mg, 1mmol), fluoroacetaldehyde (310mg, 5mmol) were added to dichloromethane (20 mL). Trifluoroacetic acid was added dropwise thereto until the reaction system was acidic (pH 1.0). The reaction was incubated at 25 ℃ for 30 min. Sodium triacetoxyborohydride (1.8g, 8.5mmol) was added to the reaction system, and the reaction was maintained at 25 ℃ for 12 hours. Distilled water (100mL) was added to the reaction mixture, and the mixture was extracted with methylene chloride (30 mL. times.3). The organic phases were combined, washed with saturated sodium bicarbonate (100mL × 1), saturated brine (100mL × 1) and water (100mL × 1), dried over anhydrous sodium sulfate, and purified by column chromatography (PE: EA ═ 4:1) to give the title compound (290 mg).
ESI-MS(m/z):342.2[M+H] +
Step two: (E) synthesis of methyl [8- (4- (2-butoxyethoxy) phenyl ] -1- (2-fluoroethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carboxylate (Compound 6-2)
Compound 6-1(242mg, 0.71mmol), 2- (4- (2-butoxyethoxy) phenyl) -4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan (228mg, 0.71mmol), palladium acetate (16mg, 0.071mmol), potassium phosphate (452mg, 2.13mmol), tri-tert-butylphosphine tetrafluoroborate (103mg, 0.355mmol) were added to a toluene/water (10mL/1mL) mixed solvent and reacted with a microwave at 100 ℃ for 3.0 hours under nitrogen protection. After cooling to room temperature, water (100mL) was added and extracted with ethyl acetate (30 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography (PE: EA ═ 4:1) to give the title compound (260 mg).
ESI-MS(m/z):456.3[M+H] +
Step three: (E) synthesis of- [8- (4- (2-butoxyethoxy) phenyl) -1- (2-fluoroethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azepin (m) tetraen-5-yl ] methanol (Compound 6-3)
Under the protection of nitrogen, LiAlH4(45mg,1.2mmol) was added to dry tetrahydrofuran (10 mL). A solution of Compound 6-2(182mg, 0.4mmol) in anhydrous tetrahydrofuran (10mL) was added dropwise thereto, and the reaction was carried out at 25 ℃ for 4 hours. The reaction was terminated by dropwise addition of a saturated ammonium chloride solution, and water (20mL) was added to conduct extraction with ethyl acetate (30 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give the title compound (140 mg). It was used directly in the next reaction without purification.
ESI-MS(m/z):428.3[M+H] +
Step four: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -1-isobutyl-1, 2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde (Compound 6-4)
Compound 6-3(107mg, 0.25mmol) was added to dimethyl sulfoxide (10 mL). 2-iodoxybenzoic acid (70mg,0.25mmol) was added to the reaction system and reacted at 25 ℃ for 12 h. Saturated sodium sulfite solution is added dropwise until starch-potassium iodide paper does not turn blue. Then, water (50mL) was added, and the mixture was extracted with ethyl acetate (50 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography (PE: EA ═ 3:1) to give the title compound (100 mg).
ESI-MS(m/z):426.3[M+H] +
EXAMPLE 7 (E) -8- (4- (2-Butoxyethoxy) phenyl) -1- (2-fluoroethyl) -5- (6- (((1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ b ] imidazol-2-yl) -1,2,3, 4-tetrahydrobenzo [ b ] azocine (m) tetraene (Compound 4)
Figure PCTCN2020081558-APPB-000031
(4- ((1-propyl-1H-imidazol-5-yl) methyl) sulfinyl-1, 2-phenylenediamine (compound 2-4, 278mg, 1mmol) and (E) -8- (4- (2-butoxyethoxy) phenyl) -1- (2-fluoroethyl) -1,2,3, 4-tetrahydrobenzo [ b ] b]Azacyclo (m) tetraene-5-carboxaldehyde (425mg, 1mmol) is added to acetonitrile (10mL), followed by 30% H2O 2Aqueous (0.7. mu.L, 7mmol) and 37% aqueous HCl (0.3. mu.L, 3.5mmol) were reacted at room temperature for 2 hours. A saturated solution of sodium sulfite (10mL) was added to the reaction mixture, followed by extraction with ethyl acetate (20 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by preparative liquid chromatography to give the title compound (91 mg).
The structure is characterized as follows:
ESI-MS(m/z):684.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ7.80(s,1H),7.73(s,1H),7.62–7.58(m,1H),7.53(d,J=8.7Hz,2H),7.45–7.41(m,1H),7.37–7.29(m,2H),7.00(d,J=8.7Hz,2H),6.95(d,J=9.9Hz,1H),6.43(s,1H),4.81–4.76(m,1H),4.69–4.64(m,1H),4.41–4.32(m,2H),4.22–4.16(m,2H),4.14–4.08(m,2H),3.82–3.63(m,6H),3.58(s,2H),3.46(t,J=6.5Hz,2H),2.79(s,2H),1.69–1.43(m,8H),1.37–1.30(m,2H),0.91–0.83(m,3H),0.82–0.75(m,3H)。
EXAMPLE 8 Synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -1- (2-methoxyethyl) -5- (6- ((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1,2,3, 4-tetrahydrobenzo [ b ] azocine (m) tetraene (Compound 5)
Figure PCTCN2020081558-APPB-000032
The method comprises the following steps: synthesis of 2-methoxyacetaldehyde (Compound 8-2)
The compound 1,1, 2-trimethoxyethane (1.03g,8.32mmol) was weighed, dissolved in trifluoroacetic acid (2mL) and water (2mL), and stirred at 50 ℃ for 0.5 hour. Cooled to room temperature and the product was used directly in the next reaction.
Step two: (E) synthesis of methyl (E) -8- (4- (2-butoxyethoxy) phenyl) -1- (2-methoxyethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carboxylate (Compound 8-4)
The reaction mixture of 8-2 and acetic acid (318.77mg,5.20mmol) was slowly added dropwise to dichloroethane (6mL) containing compound 8-3(946.80mg,2.08mmol) at 0 ℃. To the reaction solution was added sodium borohydride acetate (3.24g,4.16mmol), and the mixture was stirred at 25 ℃ for 16 hours. Water (20mL) was added to the reaction mixture to terminate the reaction. Extraction was carried out with methylene chloride (15 mL. times.3), and the organic phases were combined and dried over anhydrous sodium sulfate. Concentrate to give the title compound (864.80 mg).
ESI-MS(m/z):468.2[M+H] +
Step three: (E) synthesis of (8- (4- (2-butoxyethoxy) phenyl) -1- (2-methoxyethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azepin (m) tetraen-5-yl) methanol (Compound 8-5)
Compound 8-4(578.95mg,1.18mmol) was weighed out, added to tetrahydrofuran (18mL), and lithium aluminum hydride (1M,2.35mL) was added thereto at 0 ℃ and stirred for 1 hour. To the reaction mixture was added a saturated sodium sulfate solution (1mL), followed by addition of a little anhydrous sodium sulfate, drying and stirring for 0.5 hour. Filtration and concentration gave the title compound (515 mg).
ESI-MS(m/z):440.3[M+H] +
Step four: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -1- (2-methoxyethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde (Compound 8-6)
Compound 8-5(242.11mg,523.22umol) and 2-iodoxybenzoic acid (226.56mg,784.83umol) were weighed, dissolved in dimethyl sulfoxide (5mL), and stirred at 15 ℃ for 16 hours. To the reaction mixture was added 50mL of water, followed by extraction with ethyl acetate (10 mL. times.3), and the organic phases were combined, washed with saturated brine (10mL), dried over anhydrous sodium sulfate, and concentrated to give the title compound (210.31mg)
ESI-MS(m/z):438.2[M+H] +
Step five: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -1- (2-methoxyethyl) -5- (6- ((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1,2,3, 4-tetrahydrobenzo [ b ] azacycloocta (m) tetraene (Compound 5)
Compound 8-6(60mg,123.41umol) and Compound 3-3(60.14mg,185.11umol) prepared in example 3 were weighed, added to EtOH (4mL), and sodium bisulfite (65.52mg,617.04umol) was added and stirred at 70 ℃ for 16 hours. Cooling, dripping 3-5 drops of triethylamine into the reaction solution, and concentrating. The title compound (31mg) was isolated by preparative chromatography under neutral conditions.
The structure is characterized as follows:
ESI-MS(m/z):710.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.80(d,J=16.0Hz,1H),7.82(s,1H),7.71-7.75(m,1H),7.60-7.62(m,0.5H),7.54-7.56(m,3H),7.45-7.47(m,3H),7.27-7.32(m,1H),7.01(d,J=8.8Hz,2H),6.96(d,J=8.8Hz,1H),4.17-4.34(m,2H),4.14(t,J=6.8Hz,2H),3.78-3.84(m,2H),3.74(t,J=4.8Hz, 2H),3.65(t,J=6.0Hz,2H),3.56-3.60(m,2H),3.48-3.53(m,5H),2.81(s,2H),1.67-1.69(m,1H),1.50-1.56(m,8H),1.37(q,J=7.2Hz,2H),0.89-0.93(m,2H),0.84(t,J=7.6Hz,3H)。
EXAMPLE 9 Synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -5- (5- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- (2-hydroxy-2-methylpropyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocycloxin (m) tetraene (Compound 7)
Figure PCTCN2020081558-APPB-000033
The method comprises the following steps: synthesis of 1, 1-diethoxy-2-methylpropan-2-ol (Compound 9-2)
Compound 9-1(2g,11mmol) was added to anhydrous tetrahydrofuran (20mL) under nitrogen, a solution of methylmagnesium bromide in tetrahydrofuran (1M,28.38mL) was added dropwise to the solution at 0 deg.C, and the reaction was stirred at 25 deg.C for 2 hours. After completion of the reaction, a saturated ammonium chloride solution was added to the reaction mixture to terminate the reaction, followed by extraction with ethyl acetate (30 mL. times.3), and the organic phases were combined, washed with a saturated brine (30 mL. times.2), dried over anhydrous sodium sulfate (10g), and concentrated to obtain the title compound (1.6 g).
Step two: (E) synthesis of methyl (E) -8- (4- (2-butoxyethoxy) phenyl) -1- (2-hydroxy-2-methylpropyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-tetraene-5-carboxylate (Compound 9-3)
Compound 9-2(1.6g,9.86mmol) was added to 1, 4-dioxane (10mL), and trifluoroacetic acid (1.14g,9.86mmol) was added to react at 50 ℃ for 30 minutes. After completion of the reaction, a solution of methyl (5E) -8- [4- (2-butoxyethoxy) phenyl ] -1,2,3, 4-tetrahydrobenzo [ b ] azepino (m) tetraene-5-carboxylate (500mg,1.16mmol) in methylene chloride (10mL) was added, and after 30 minutes at 25 ℃, sodium borohydride acetate (2.59g,11.60mmol) was added and the reaction was continued with stirring for 12 hours. After completion of the reaction, water (30mL) was added to terminate the reaction, and the reaction mixture was extracted with methylene chloride (30 mL. times.3), and the organic phases were combined, washed with saturated brine (30 mL. times.2), dried over anhydrous sodium sulfate (10g), and concentrated to give the title compound (420 mg).
ESI-MS(m/z):482.15[M+H] +
Step three: synthesis of ((E) - (8- (4- (2-butoxyethoxy) phenyl) -1- (2-hydroxy-2-methylpropyl) -1,2,3, 4-tetrahydrobenzo [ b ] azepin (m) tetraen-5-yl) methanol (Compound 9-4)
Compound 9-3(350mg, 640. mu. mol) was weighed into tetrahydrofuran (4mL), and lithium aluminum hydride (1M,804uL) was added at 0 ℃ and stirred at 0 ℃ for 2 hours under nitrogen. To the reaction solution was added dropwise a saturated aqueous sodium sulfate solution (0.1 mL). Then, anhydrous sodium sulfate was added thereto, and the mixture was dried, filtered and concentrated to obtain the title compound (200 mg). The crude product was not purified and used directly in the next step.
ESI-MS(m/z):454.25[M+H] +
Step four: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -1- (2-hydroxy-2-methylpropyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde (Compound 9-5)
Compound 9-4(200mg, 418. mu. mol) was weighed into dimethyl sulfoxide (5mL), 2-iodoxybenzoic acid (145mg, 500. mu. mol) was added at 0 ℃ and stirred at 25 ℃ for 16 hours. Water (20mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The organic phase was washed three times with saturated brine, dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography (PE: EA ═ 1:1) to give the title compound (160 mg).
ESI-MS(m/z):452.3[M+H] +
Step five: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -5- (5- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- (2-hydroxy-2-methylpropyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclooct (m) tetraene (Compound 7)
Compound 9-5(60mg,130umol) and compound 3-3(57mg,190umol) prepared in example 3 were added to ethanol (4mL), sodium bisulfite (68mg,650umol) was added, and stirred at 70 ℃ for 16 hours. Two drops of triethylamine were added to the reaction solution, which was concentrated and purified by preparative liquid chromatography to give the title compound (30 mg).
The structure is characterized as follows:
ESI-MS(m/z):724.25[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.80(d,J=21.2Hz,1H),7.81(s,1H),7.70(m,1H),7.61–7.45(m,4H),7.40(s,2H),7.27(m,1H),7.15(m,1H),6.98(d,J=8.0Hz,2H),4.54(s,1H),4.25(t,2H),4.10(s,2H),3.71(m,6H),3.45(t,J=5.8Hz,3H),3.28(s,2H),2.70(s,2H),1.64(s,2H),1.51(m,6H),1.32(m,2H),1.20(s,6H),0.88(t,J=7.0Hz,3H),0.80(t,J=7.0Hz,3H)。
EXAMPLE 10 Synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -5- (6- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- (2-carboxy-2-methylpropyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocycloxin (m) tetraene (Compound 9)
Figure PCTCN2020081558-APPB-000034
The method comprises the following steps: synthesis of methyl 2, 2-dimethyl-3-hydroxypropionate (Compound 10-2)
Compound 10-1(25g,190mmol) was added to anhydrous dichloromethane (600mL), dimethyl sulfoxide (147.80g,1.89mol) and triethylamine (57.43g,567mmol) were added at 0 deg.C, the dropwise addition was completed, sulfur trioxide pyridine (90.32g,567mmol) was slowly added in portions, and the reaction was stirred at 25 deg.C for 16 hours. After completion of the reaction, 1M hydrochloric acid (500mL) was added to the reaction mixture to terminate the reaction, and the reaction mixture was extracted with methylene chloride (500 mL. times.3), and the organic phases were combined, washed with saturated brine (500 mL. times.2), dried over anhydrous sodium sulfate, spin-dried, and distilled under reduced pressure at 0.1atm to collect a fraction at 90 ℃ to obtain the title compound (16 g).
Step two: synthesis of ((5E) -8- (4- (2-butoxyethoxy) phenyl) -1,2,3, 4-tetrahydrobenzo [ b ] azepin (m) tetraen-5-yl) methanol (Compound 10-3)
Compound 8-3(1g,1.95mmol) was weighed into tetrahydrofuran (4mL), lithium aluminum hydride (1M,5.86mL) was added at 0 deg.C, and stirred at 0 deg.C for 2 hours under nitrogen. To the reaction solution was added dropwise a saturated aqueous sodium sulfate solution (1 mL). Then, anhydrous sodium sulfate was added thereto, and the mixture was dried, filtered and concentrated to obtain the title compound (600 mg). The crude product was not purified and used directly in the next step.
ESI-MS(m/z):382.25[M+H] +
Step three: synthesis of methyl 3- [ (5E) -8- [4- (2-butoxyethoxy) phenyl ] -5-hydroxymethyl-1, 2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraen-1-yl ] -2, 2-dimethylpropionate (Compound 10-4)
10-2(195mg,1.42mmol), compound 10-3(300mg, 710. mu. mol) were weighed into acetic acid (1mL) and dichloromethane (5mL), sodium borohydride acetate (1.09g,1.42mmol) was added, and the mixture was stirred at 15 ℃ for 16 hours. The reaction was added dropwise to a saturated sodium bicarbonate solution (50mL) and extracted with dichloromethane (20 mL. times.3). The organic phase was washed with saturated brine (20 mL. times.3), dried over anhydrous sodium sulfate, and concentrated to give the title compound (250 mg).
ESI-MS(m/z):496.25[M+H] +
Step four: synthesis of methyl 3- [ (5E) -8- [4- (2-butoxyethoxy) phenyl ] -5-formyl-1, 2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraen-1-yl ] -2, 2-dimethylpropionate (Compound 10-5)
Compound 10-4(250mg,410umol) was weighed into dimethyl sulfoxide (5mL), 2-iodoxybenzoic acid (145mg,500umol) was added at 0 ℃ and stirred at 25 ℃ for 16 hours. Water (20mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (20 mL. times.3). The organic phase was washed three times with saturated brine (20mL × 3), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography (PE: EA ═ 1:1) to give the title compound (150 mg).
ESI-MS(m/z):494.15[M+H] +
Step five: synthesis of methyl 3- [ ((5E) -8- [4- (2-butoxyethoxy) phenyl ] -5- [6- [ (5-methyl-3-propyl-1H-imidazol-4-yl) methylsulfinyl ] -1H-benzimidazol-2-yl ] -1,2,3, 4-tetrahydrobenzo [ b ] azocino (m) tetraen-1-yl ] -2, 2-dimethylpropionate (Compound 10-6)
Compound 10-5(120mg, 220. mu. mol) and compound 3-3(74mg, 240. mu. mol) prepared in example 3 were added to ethanol (4mL), sodium bisulfite (116mg,1.09mmol) was added, and the mixture was stirred at 70 ℃ for 16 hours. After completion of the reaction, the solvent was dried by evaporation, and the residue was extracted with methylene chloride (20 mL. times.3), washed with saturated brine (20 mL. times.3), dried over anhydrous sodium sulfate, and then dried by evaporation to give the title compound (140mg) as a yellow solid product.
ESI-MS(m/z):766.25[M+H] +
Step six: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -5- (6- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- (2-carboxy-2-methylpropyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocin (m) tetraene (Compound 9)
Compound 10-6(140mg, 170. mu. mol) was added to a mixed solvent of methanol (2mL) and tetrahydrofuran (2mL), and an aqueous solution (1mL) of LiOH (43.55mg,1.78mmol) was added to the mixture to react at 55 ℃ for 2 hr. After completion of the reaction, the solvent was dried by spinning, and the residue was extracted with methylene chloride (20 mL. times.3), washed with saturated brine (10mL), dried over anhydrous sodium sulfate, and then spun to obtain a yellow solid product. After addition of 5mL of methanol, 1N hydrochloric acid solution was added dropwise to make the solution pH 2-3, and the title compound (40mg) was obtained by preparative liquid chromatography.
The structure is characterized as follows:
ESI-MS(m/z):752.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ9.15(s,1H),8.33(s,1H),7.91(d,J=8.4Hz,1H),7.86(s,1H),7.64(d,J=1.6Hz,1H),7.59(m,4H),7.21(d,J=8.8Hz,1H),7.05(d,J=8.8Hz,2H),4.87(d,J=15.2Hz,1H),4.48(d,J=15.2Hz,1H),4.15(t,J=8.4Hz,2H),4.11–4.04(m,2H),3.74(t,J=4.4Hz,2H),3.58(s,2H),3.50(t,J=6.6Hz,2H),3.39(3,2H),2.79(s,2H),1.78-1.84(m,2H),1.68(s,3H),1.67–1.61(m,2H),1.57–1.51(m,2H),1.36(m,2H),1.22(s,6H),0.91(m,6H)。
EXAMPLE 11 Synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -5- (6- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- (oxetan-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocycloxin (m) tetraene (Compound 10)
Figure PCTCN2020081558-APPB-000035
The method comprises the following steps: synthesis of Oxetadin-3-yl-Formaldehyde (Compound 11-2)
Oxetadin-3-ylcarbinol (1.01g,11.35mmol) was dissolved in methylene chloride (40mL), and dessimutant reagent (5.89g,13.62mmol) was added to the reaction mixture twice at 25 ℃ and stirred for 2 hours. After filtration with suction, a saturated sodium bicarbonate solution and a saturated sodium bisulfite solution were added to the filtrate to terminate the reaction. Suction filtration was again performed, and to the filtrate was added anhydrous sodium sulfate, dried, stirred for 10 minutes, suction filtered, and the filtrate was concentrated to give the title compound (977.12 mg).
Step two: (E) synthesis of methyl (E) -8- (4- (2-butoxyethoxy) phenyl) -1- (oxetan-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-tetraene-5-carboxylate (Compound 11-3)
Compound 11-2(977.12mg,11.35mmol) was weighed, dissolved in dichloromethane (40mL), trifluoroacetic acid (165.07mg,1.42mmol) was added, and compound 8-3(645.56mg,1.42mmol) and sodium borohydride acetate (2.45g,11.35mmol) were added to the reaction solution under stirring at 25 ℃ and stirred for 12 hours. The reaction solution was filtered with suction, the filtrate was concentrated, water (15mL) was added, a saturated sodium bicarbonate solution was added dropwise to the solution at 0 ℃ to adjust the pH to 7-8, extraction was performed with dichloromethane (30 mL. times.3), the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give the title compound (621.70 mg).
ESI-MS(m/z):490.3[M+H] +
Step three: (E) synthesis of (8- (4- (2-butoxyethoxy) phenyl) -1- (oxetan-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclooct (m) tetraen-5-yl) methanol (Compound 11-4)
Compound 11-3(222.22mg,417.01umol) was weighed into tetrahydrofuran (5mL), lithium aluminum hydride (1M, 834.02. mu.L) was added under nitrogen, and stirred at 0 ℃ for 1 hour. To the reaction mixture was added a saturated sodium sulfate solution (0.5mL), followed by addition of anhydrous sodium sulfate, drying and stirring for 0.5 hour. Filtration and concentration gave the title compound (167 mg).
ESI-MS(m/z):382.1[M+H] +
Step four: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -1- (oxetan-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde (Compound 11-5)
Compound 11-4(160mg,283.44umol) and 2-iodoxybenzoic acid (122.73mg,425.16umol) were weighed out and dissolved in dimethyl sulfoxide (2mL) and stirred at 15 ℃ for 16 hours. To the reaction mixture was added water (10mL), extracted with ethyl acetate (10 mL. times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give the title compound (62.5 mg).
ESI-MS(m/z):450.2[M+H] +
Step five: (E) synthesis of (E) -8- (4- (2-butoxyethoxy) phenyl) -5- (6- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- (oxetan-3-ylmethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocin (m) tetraene (Compound 10)
Compound 11-5(60mg,120.11umol) and Compound 3-3(58.53mg,180.17umol) prepared in example 3 were weighed, added to ethanol (3mL), and sodium bisulfite (63.77mg,600.56umol) was added and stirred at 70 ℃ for 16 hours. The reaction solution is cooled, 3 to 5 drops of triethylamine are added, and the mixture is concentrated. Isolation by preparative chromatography under neutral conditions gave the title compound (30 mg).
The structure is characterized as follows:
ESI-MS(m/z):722.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.77(d,J=21.2Hz,1H),7.76(s,1H),7.74-7.70(m,1H),7.61-7.59(m,0.5H),7.55-7.53(m,3H),7.49-7.44(m,2.5H),7.31-7.26(m,1H),7.01(d,J=8.8Hz,2H),6.90(d,J=8.8Hz,1H),4.72-4.69(m,2H),4.48(t,J=6.0Hz,2H),4.33-4.16(m,1H),4.13(t,J=4.8Hz,2H),3.83-3.71(m,4H),3.65(d,J=6.8Hz,2H),3.50-3.45(m,5H),2.80(s,2H),1.68-1.66(m,2H),1.54-1.49(m,8H),1.40-1.31(m,2H),0.90(t,J=7.2Hz,3H),0.83(t,J=7.2Hz,3H).
EXAMPLE 12 Synthesis of (E) -8- (4-butoxyphenyl) -5- (6- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- (((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocycloxin (m) tetraene (Compound 17)
Figure PCTCN2020081558-APPB-000036
The method comprises the following steps: synthesis of 1-bromo-4-butoxybenzene (Compound 12-2)
1-bromobutane (7.92g,54.9mmol) and compound 12-1(5g,27.5mmol) were dissolved in N, N-dimethylformamide (50mL), potassium iodide (456mg,2.75mmol) and potassium carbonate (11.38g,82.4mmol) were added, followed by warming to 80 ℃ and reaction at this temperature for 16 hours. The temperature was reduced to room temperature, water (200mL) was added, and extraction was performed with ethyl acetate (150 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated and purified by column chromatography (PE: EA ═ 5:1) to afford the title compound (3 g).
Step two: synthesis of 2- (4-butoxyphenyl) -4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan (Compound 12-3)
Compound 12-2(1g,4.2mmol), pinacol diboron (1.66g,6.2mmol), Pd (dppf) Cl2(639mg,829.3umol) and potassium acetate (857mg,8.3mmol) were dissolved in 1, 4-dioxane (10 mL). Stirring was carried out at 100 ℃ for 16 hours under nitrogen. The reaction was directly filtered, concentrated, and purified by column chromatography (PE: EA ═ 10:1) to give the title compound (1 g).
Step three: (E) synthesis of methyl 8- (4-butoxyphenyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-tetraene-5-carboxylate (Compound 12-4)
Compound 1-1(300mg,962umol), compound 12-3(643mg,2.2mmol) were weighed out and added to tetrahydrofuran (10mL) and water (2mL), and palladium acetate (21.60mg,96.2umol), tris (o-methylphenyl) phosphine (60mg,192umol) and potassium phosphate (1.23g,5.8mmol) were added and reacted at 80 ℃ for 16 hours under nitrogen atmosphere. Water (60mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (60 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography (PE: EA ═ 5:1) to give the title compound (300 mg).
ESI-MS(m/z):366.2[M+H] +
Step four: (E) synthesis of methyl 8- (4-butoxyphenyl) -1- (((tetrahydro-2H-pyran-4-yl) methyl) -) methyl-1, 2,3, 4-tetrahydrobenzo [ b ] azacyclo-tetraene-5-carboxylate (Compound 12-5)
Compound 12-4(200mg,459umol) and tetrahydropyran-4-carbaldehyde (221mg,1.8mmol) were weighed out, added to acetic acid (2mL) and dichloromethane (4mL), added sodium borohydride acetate (292mg,1.4mmol), and stirred at 15 ℃ for 16 hours. Water (20mL) was added to the reaction solution, and the mixture was extracted with methylene chloride (20 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography (dichloromethane: methanol ═ 10:1) to give the title compound (250 mg).
ESI-MS(m/z):464.2[M+H] +
Step five: (E) synthesis of (8- (4-butoxyphenyl) -1- ((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclooct (m) tetraen-5-yl) methanol (Compound 12-6)
Compound 12-5(500mg,917umol) was weighed into tetrahydrofuran (10mL), lithium aluminum hydride (1M,1.83mL) was added at 0 ℃ and stirred at 0 ℃ for 1 hour. To the reaction mixture was added a saturated aqueous solution of anhydrous sodium sulfate (1mL), dried over anhydrous sodium sulfate, and stirred for half an hour. Filtration and concentration gave the title compound (400 mg). It was used directly in the next reaction without purification.
ESI-MS(m/z):436.3[M+H] +
Step six: (E) synthesis of (E) -8- (4-butoxyphenyl) -1- (((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacycloocta (m) tetraene-5-carbaldehyde (Compound 12-7)
Compound 12-6(400mg,826.45umol) was added to dimethyl sulfoxide (3mL), and 2-iodoxybenzoic acid (347.13mg,1.24mmol) was added and reacted at 15 ℃ for 16 hours. Water (50mL) was added to terminate the reaction, and extraction was performed with ethyl acetate (20mL × 3), and the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography (PE: EA ═ 3:1) to obtain the title compound (300 mg).
ESI-MS(m/z):434.2[M+H] +
Step seven: (E) -8- (4-butoxyphenyl) -5- (6- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- (((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclooct (m) tetraene (Compound 17)
The compounds 3-3(57mg, 189umol) prepared in example 3 and (E) -8- (4-butoxyphenyl) -1- (((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde (compound 12-7,60mg,126umol) were added to ethanol (4mL), sodium hydrogensulfite (66mg,630umol) was added, stirred at 70 ℃ for 16 hours, two drops of triethylamine were added to the reaction solution, concentrated, and purified by preparative liquid chromatography to give the title compound (19 mg).
The structure is characterized as follows:
ESI-MS(m/z):706.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.80(d,J=22.0Hz,1H),7.82(s,1H),7.74-7.70(m,1H),7.61-7.60(m,0.5H),7.55-7.52(m,3H),7.46-7.40(m,2.5H),7.31-7.26(m,1H),7.01(d,J=8.8Hz,2H),6.93(d,J=8.8Hz,1H),4.33-4.17(m,2H),4.02(t,J=6.8Hz,2H),3.92–3.89(m,2H),3.83-3.73(m,2H),3.60-3.50(m,2H),3.31-3.28(m,2H),3.22(d,J=6.4Hz,2H),2.85-2.75(m,2H),2.15-2.10(m,1H),1.75-1.65(m,4H),1.55-1.54(m,4H),1.48(q,J=7.2Hz,2H),1.40-1.30(m,2H),0.97(t,J=7.6Hz,3H),0.83(t,J=7.6Hz,3H)。
EXAMPLE 13 Synthesis of (E) -8- (4- ((2-butoxyethoxy) pyridin-3-yl)) -5- (6- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- ((1-methyl-1H-pyrazol-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo (m) tetraene (Compound 18)
Figure PCTCN2020081558-APPB-000037
The method comprises the following steps: synthesis of 5-bromo-2- (2-butoxyethoxy) pyridine (Compound 13-2)
Sodium hydride (1.23g,31mmol) was added to a solution of 2-butoxyethanol (3.68g,31mmol) in N, N-dimethylformamide (30mL) at 0 ℃ and after stirring for 30 minutes, compound 18-1(3g,15.43mmol) was added and the reaction mixture was reacted at 25 ℃ for 4 hours. After completion of the reaction, a saturated ammonium chloride solution (30mL) was added dropwise to terminate the reaction, followed by extraction with ethyl acetate (30 mL. times.3). The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and concentrated to give the title compound (3.6 g).
ESI-MS(m/z):273.95[M+H] +
Step two: synthesis of 2- (2-butoxyethoxy) -5- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) pyridine (Compound 13-3)
Compound 18-2(1.7g,6.0mmol), pinacol diboron (1.66g,6.2mmol), Pd (dppf) Cl2(639mg,829.3umol) and potassium acetate (857mg,8.3mmol) were dissolved in 1, 4-dioxane (10 mL). Stirring was carried out at 100 ℃ for 16 hours under nitrogen. The reaction was directly filtered, concentrated, and purified by column chromatography (PE: EA ═ 10:1) to give the title compound (1.2 g).
ESI-MS(m/z):322.25[M+H] +
Step three: synthesis of ((5E) -8-bromo-1- (tetrahydropyran-4-ylmethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azepin (m) tetraen-5-yl) methanol (Compound 13-4)
Compound 1-2(1.2g,1.95mmol) was weighed into tetrahydrofuran (15mL), lithium aluminum hydride (1M,3.58mL) was added at 0 deg.C, and stirred at 0 deg.C under nitrogen for 2 hours. To the reaction solution was added dropwise a saturated aqueous sodium sulfate solution (0.5 mL). Then, anhydrous sodium sulfate was added thereto, and the mixture was dried, filtered and concentrated to obtain the title compound (1 g). The crude product was not purified and used directly in the next step.
ESI-MS(m/z):366.05[M+H] +
Step four: (5E) synthesis of (E) -8-bromo-1- (tetrahydropyran-4-ylmethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde (compound 13-5)
Compound 13-4(1g,2.65mmol) was weighed into dimethyl sulfoxide (10mL), 2-iodoxybenzoic acid (1.5g,5.3mmol) was added at 0 deg.C, and the mixture was stirred at 25 deg.C for 16 hours. Water (20mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (30 mL. times.3). The organic phase was washed three times with saturated brine (30mL × 3), dried over anhydrous sodium sulfate, filtered, concentrated, and purified by column chromatography (PE: EA ═ 5:1) to give the title compound (910 mg).
ESI-MS(m/z):364.05[M+H] +
Step five: (5E) synthesis of (E) -8-bromo-5- [6- [ (5-methyl-3-propyl-imidazol-4-yl) methylsulfinyl ] -1H-benzimidazol-2-yl ] -1- (tetrahydropyran-4-ylmethyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocycloxin (m) tetraene (Compound 13-6)
Compound 13-5(200mg,538umol) and compound 2-4(190mg,590umol) prepared in example 2 were added to ethanol (4mL), sodium bisulfite (141mg,1.35mmol) was added, and the mixture was stirred at 70 ℃ for 16 hours. After the completion of the reaction, the solvent was dried by evaporation, and the residue was extracted with dichloromethane (20mL × 3), washed with saturated brine (20mL × 3), dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography (dichloromethane: methanol ═ 10:1) to obtain the title compound (310 mg).
ESI-MS(m/z):636.05[M+H] +
Step six: (E) synthesis of (E) -8- (4- ((2-butoxyethoxy) pyridin-3-yl)) -5- (6- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- ((1-methyl-1H-pyrazol-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocine (m) tetraene (Compound 18)
Compounds 13-6(70mg,105umol), 13-3(71mg,210umol), tetrakis (triphenylphosphine) palladium (12mg,10.5umol) and sodium carbonate (23mg,210umol) were added to 1, 4-dioxane (2mL) and water (0.4 mL). The reaction is carried out for 1 hour under the protection of nitrogen and the microwave of 150W at 100 ℃. After completion of the reaction, the reaction solution was directly filtered, concentrated and purified by preparative liquid chromatography to give the title compound (10 mg).
The structure is characterized as follows:
ESI-MS(m/z):751.25[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.80(d,J=20.0Hz,1H),8.38(d,J=2.4Hz,1H),7.93(dd,J=8.6,2.4Hz,1H),7.81(s,1H),7.71-7.47(m,2H),7.51(s,1H),7.45(m,2H),7.26(t,J=9.2Hz,1H),6.93(d,J=9.2Hz,1H),6.88(d,J=8.8Hz,1H),4.39(t,J=4.8Hz,1H),4.14-4.32(m,2H),3.88(m,2H),3.83–3.72(m,2H),3.72–3.69(m,2H),3.53(s,2H),3.45(t,J=6.6Hz,2H),3.27(m,2H),3.21(m,2H),2.78(s,2H),2.10–1.95(m,1H),1.64(m,4H),1.58–1.43(m,7H),1.42–1.26(m,4H),0.88(t,J=7.4Hz,3H),0.81(t,J=7.4Hz,3H)。
EXAMPLE 14 Synthesis of (E) -8- (4- (2-methoxyethoxy) phenyl) -5- (6- ((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- (((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene (Compound 19)
Figure PCTCN2020081558-APPB-000038
The method comprises the following steps: synthesis of 1-bromo-4- (2-methoxyethoxy) benzene (Compound 14-1)
1-chloro-2-methoxyethane (5.46g,57.80mmol) and the compound 12-1(5g,28.90mmol) were dissolved in N, N-dimethylformamide (50mL), and potassium iodide (480mg,2.89mmol) and potassium carbonate (10g,72mmol) were added, followed by warming to 80 ℃ and reaction at this temperature for 16 hours. The temperature was reduced to room temperature, water (200mL) was added, and extraction was performed with ethyl acetate (150 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate and concentrated to give the title compound (6.68 g). The crude product was not purified and used directly in the next step.
Step two: synthesis of 2- (4- (2-methoxyethoxy) phenyl) -4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan (Compound 14-2)
Compound 14-1(6.68g,28.9mmol) was dissolved in 1, 4-dioxane (70mL), and pinacol diboron (11.01g,43.36mmol), Pd (dppf) Cl were added2(4.23g,5.78mmol) and potassium acetate (5.67g,57.8mmol), followed by stirring at 100 ℃ for 16 hours under nitrogen. Adding water (150mL) to the reaction solution, extracting with ethyl acetate (150 mL. times.3), washing the organic phase with saturated saline water for three times, drying over anhydrous sodium sulfate, concentrating, and performing column chromatographyPurification (PE: EA ═ 10:1) afforded the title compound (8 g).
Step three: (E) synthesis of methyl (E) -8- (4- (2- (methoxyethoxy) phenyl) methyl-1, 2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carboxylate (Compound 14-3)
Compound 1-1(3g,9.62mmol) was weighed out and added to tetrahydrofuran (30mL) and water (3mL), compound 14-2(4.23g,14.4mmol), palladium acetate (432mg,1.92mmol), potassium phosphate (10g,48.1mmol) and tris (o-methylphenyl) phosphine (1.2g,3.9mmol) were added and reacted at 90 ℃ for 16 hours under nitrogen protection. Water (100mL) was added to the reaction mixture, and the mixture was extracted with ethyl acetate (100 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, concentrated and purified by column chromatography (PE: EA ═ 5:1) to afford the title compound (3 g).
ESI-MS(m/z):368.2[M+H] +
Step four: (E) synthesis of methyl (E) -8- (4- (2-methoxyethoxy) phenyl) -1- (((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carboxylate (Compound 14-4)
Tetrahydropyran-4-carbaldehyde (350mg,3.06mmol) and compound 14-3(500mg,1.22mmol) were weighed out, added to acetic acid (5mL) and dichloromethane (5mL), added sodium borohydride acetate (779mg,3.67mmol), and stirred at 15 ℃ for 16 hours. Water (20mL) was added to the reaction mixture, and the mixture was extracted with methylene chloride (20 mL. times.3). The organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give the title compound (550 mg).
ESI-MS(m/z):466.2[M+H] +
Step five: (E) synthesis of (8- (4- (2-methoxyethoxy) phenyl) -1- ((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclooct (m) tetraen-5-yl) methanol (Compound 14-5)
Compound 14-4(550mg,1.06mmol) was weighed into tetrahydrofuran (10mL), lithium aluminum hydride (121mg,3.2mmol) was added at 0 deg.C, and stirred at 0 deg.C for 2 hours under nitrogen. To the reaction mixture was added a saturated aqueous solution of anhydrous sodium sulfate (1mL), dried over anhydrous sodium sulfate, and stirred for half an hour. Filtration and concentration gave the title compound (460 mg). It was used directly in the next reaction without purification.
ESI-MS(m/z):438.3[M+H] +
Step six: (E) synthesis of (E) -8- (4- (2-methoxyethoxy) phenyl) -1- (((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde (Compound 14-6)
Compound 14-5(460mg,893.57umol) was added to dimethyl sulfoxide (4mL), and 2-iodoxybenzoic acid (375mg,1.34mmol) was added and reacted at 20 ℃ for 16 hours. Water (50mL) was added for quenching, extraction was performed with ethyl acetate (20mL × 3), and the organic phases were combined, dried over anhydrous sodium sulfate, concentrated, and purified by column chromatography (PE: EA ═ 1:1) to obtain the title compound (300 mg).
ESI-MS(m/z):436.2[M+H] +
Step seven: (E) -8- (4- (2-methoxyethoxy) phenyl) -5- (6- ((((4-methyl-1-propyl-1H-imidazol-5-yl) methyl) sulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1- (((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azocycloxin (m) tetraene (Compound 19)
The compounds 3-3(59mg, 192umol) and (E) -8- (4- (2-methoxyethoxy) phenyl) -1- (((tetrahydro-2H-pyran-4-yl) methyl) -1,2,3, 4-tetrahydrobenzo [ b ] azepino (m) tetraene-5-carbaldehyde prepared in example 3 (compound 14-6,80mg,175umol) were added to ethanol (3mL), sodium bisulfite (91mg,872umol) was added, stirred at 70 ℃ for 16 hours, two drops of triethylamine were added to the reaction solution, concentrated, and purified by preparative liquid chromatography to give the title compound (16 mg).
The structure is characterized as follows:
ESI-MS(m/z):708.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.80(d,J=22.0Hz,1H),7.82(s,1H),7.74-7.70(m,1H),7.61-7.59(m,0.5H),7.55-7.52(m,3H),7.46-7.40(m,2.5H),7.31-7.26(m,1H),7.01(d,J=8.8Hz,2H),6.93(d,J=8.8Hz,1H),4.33-4.26(m,1H),4.21-4.13(m,3H),3.92-3.89(m,2H),3.83-3.73(m,2H),3.70-3.67(m,2H),3.58-3.50(m,2H),3.22(d,J=6.4Hz,2H),2.85-2.75(m,2H),2.15-2.10(m,1H),1.75-1.65(m,4H),1.55-1.54(m,5H),1.40-1.32(m,2H),0.83(t,J=7.6Hz,3H)。
EXAMPLE 15 Synthesis of (E) -1- (((1, 4-dioxan-2-yl) methyl) -8- (4- (2-butoxyethoxy) phenyl) -5- (6- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methylsulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1,2,3, 4-tetrahydrobenzo [ b ] azocine (m) tetraene (Compound 20)
Figure PCTCN2020081558-APPB-000039
The method comprises the following steps: synthesis of 1, 4-dioxane-2-carbaldehyde (Compound 15-2)
A solution of oxalyl chloride (2.08g,16.08mmol) in dichloromethane (10mL) was cooled to-78 deg.C, a solution of dimethyl sulfoxide (1.52g,19.30mmol) in dichloromethane (10mL) was slowly added, the reaction was held for 30 minutes, a solution of compound 15-1(800mg,6.43mmol) in dichloromethane (10mL) was added dropwise, the reaction was continued for 1-2 hours, N-diisopropylethylamine (4.58g,35.06mmol) was added dropwise, the reaction was allowed to rise to 0 deg.C after 30 minutes, and the reaction was allowed to react for 30 minutes. The product is used for the next reaction without treatment.
Step two: (E) synthesis of methyl (E) -1- (((1, 4-dioxan-2-yl) methyl) -8- (4- (2-butoxyethoxy) phenyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carboxylate (Compound 15-3)
Trifluoroacetic acid (7.70g,66.18mmol) was added dropwise to a reaction solution of compound 15-2(747.03mg,6.43mmol), compound 8-3(292.74mg,643.35umol) was added, and stirring was carried out at 25 ℃ for 30 minutes, sodium borohydride acetate (1.42g,6.44mmol) was added, and stirring was carried out at 25 ℃ for 16 hours. To the reaction solution was added a saturated sodium bicarbonate solution to adjust the pH to 6-7, followed by extraction with dichloromethane (10 mL. times.3), and the organic phase was dried over anhydrous sodium sulfate and concentrated to give the title compound (310mg,547.45 umol).
ESI-MS(m/z):510.2[M+H] +
Step three: (E) synthesis of (1- (((1, 4-dioxa-2-yl) methyl) -8) - (4- (2-butoxyethoxy) phenyl) -1,2,3, 4-tetrahydrobenzo [ b ] azepin (m) tetraen-5-yl) methanol (Compound 15-4)
Compound 15-3(177.78mg,313.95umol) was weighed into tetrahydrofuran (5mL), and lithium aluminum hydride (1M,627.90uL) was added under nitrogen at 0 ℃ and stirred at 0 ℃ for 1 hour. To the reaction mixture was added a saturated sodium sulfate solution (0.5mL), followed by addition of anhydrous sodium sulfate, drying and stirring for 0.5 hour. Filtration and concentration gave the title compound (142 mg).
ESI-MS(m/z):482.1[M+H] +
Step four: (E) synthesis of (E) -1- (((1, 4-dioxan-2-yl) methyl) -8- (4- (2-butoxyethoxy) phenyl) -1,2,3, 4-tetrahydrobenzo [ b ] azacyclo-oct (m) tetraene-5-carbaldehyde (Compound 15-5)
Compound 15-4(150mg,280.30umol) and 2-iodoxybenzoic acid (121.38mg,420.45umol) were weighed out and dissolved in dimethyl sulfoxide (3mL) and stirred at 25 ℃ for 16 hours. To the reaction mixture was added water (10mL), extracted with ethyl acetate (10 mL. times.3), and the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated to give the title compound (108.53 mg).
ESI-MS(m/z):480.2[M+H] +
Step five: (E) synthesis of (E) -1- (((1, 4-dioxan-2-yl) methyl) -8- (4- (2-butoxyethoxy) phenyl) -5- (6- (((((4-methyl-1-propyl-1H-imidazol-5-yl) methylsulfinyl) -1H-benzo [ d ] imidazol-2-yl) -1,2,3, 4-tetrahydrobenzo [ b ] azocin (m) tetraene (Compound 20)
Compound 15-5(55.56mg,104.25umol) and Compound 3-3(50.81mg,156.38umol) prepared in example 3 were added to EtOH (4mL) and sodium bisulfite (55.35mg,521.26umol) was added and stirred at 70 ℃ for 16 hours. Two drops of triethylamine were added to the reaction solution, which was concentrated and purified by preparative liquid chromatography to give the title compound (30 mg).
The structure is characterized as follows:
ESI-MS(m/z):752.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.79(d,J=21.4Hz,1H),7.80(s,1H),7.72-7.66(m,1H),7.57-7.37(m,6H),7.25(t,J=11.6Hz,2H),6.98(d,J=8.4Hz,2H),4.31-4.07(m,4H),3.87(d,J=6.5Hz,4H),3.70(s,3H),3.60(s,2H),3.45(s,4H),2.67(s,2H),1.72-1.44(m,10H),1.32(d,J=8.4Hz,5H),0.87(t,J=7.3Hz,3H),0.80(t,J=7.2Hz,3H)。
the separation method comprises the following steps:
the compounds 3-20 of the invention are separated and purified by using an active 1260 type HPLC, the column temperature is 25 ℃, and other separation conditions are shown in the following table:
Figure PCTCN2020081558-APPB-000040
pharmacological Activity test
Experimental example 1 inhibition assay for cellular CCR2/CCR5 Activity
Adding 2X 10 of each well in 384-well plate4HEK293-CCR2 stable transfected cells or HEK293-CCR5 stable transfected cells were cultured overnight. After labeling with fluorescent probes, different concentrations of MCP-1 (ligand of CCR2, PeproTech, 300-04) and RANTES (ligand of CCR5, PeproTech, 300-06) were added and the fluorescent signal was immediately read (instrument: FLIPR Tetra). The fluorescence signal was plotted against ligand concentration and EC was calculated according to a four parameter model80Values were taken as the concentration of MCP-1 and RANTES at the time of subsequent compound detection.
Adding 2X 10 of each well in 384-well plate4HEK293-CCR2 stable transfected cells or HEK293-CCR5 stable transfected cells were cultured overnight. After labeling the cells with fluorescent probes, compounds (0.019nM,0.076nM,0.305nM,1.220nM,4.882nM,20nM,78nM,313nM,1250nM,5000nM) were added at different concentrations, together with the above EC80MCP-1 and RANTES at concentrations, and then the fluorescence signal was immediately read (instrument: FLIPR Tetra). And the fluorescence signals of each group were read with the vehicle group (DMSO without compound) as a negative control and the Buffer group (FLIPR Assay Buffer without ligand) as a blank control, and the inhibitory activity of each concentration group was calculated according to the following formula:
relative inhibition activity percentage is 1- (each concentration group fluorescence signal-blank group fluorescence signal)/(vehicle group fluorescence signal-blank group fluorescence signal) × 100%
Plotting the percentage of inhibitory activity of each concentration group against the concentration of the compound according to the four ginsengD/A fitting curve, calculating IC50The value:
y=min+(max-min)/(1+(x/IC 50)^(-Hillslope))
wherein y is the relative percent inhibitory activity; max and min are respectively the maximum value and the minimum value of the fitting curve; x is the compound concentration; hillslope is the slope of the curve.
Test results
The inhibitory activity of the compounds on CCR2/CCR5 was determined as described above and the results are shown in Table 1:
TABLE 1 inhibition of cellular CCR2/CCR5 by the compounds of the present invention
Figure PCTCN2020081558-APPB-000041
As can be seen from Table 1, the compounds of the present invention (e.g., compounds 1 and 2) have a very strong inhibitory effect on CCR2/CCR 5.
In the following experimental examples 2 and 3, compound 11 of example 8 (reference compound) in the specification of WO 03/014105a1 was compared with the example compounds of the present application to fully demonstrate the advantages of the example compounds of the present application.
The structure of the reference compound is:
Figure PCTCN2020081558-APPB-000042
experimental example 2: CYP enzyme inhibition assay (chemiluminescence method)
An experimental system:
P450-Glo TMCYP1a2 screening system, manufacturer: promega
P450-Glo TMCYP2D6 screening system, manufacturer: promega
Experimental parameters:
BMG PHERAstar FS Luminescent
the experimental steps are as follows:
inhibition of CYP1a 2: are respectively provided withCompound concentration group, blank group and negative control group. In the compound concentration group, compounds to be detected with different concentrations are added into a micropore plate, and Luciferin-ME EGE (100 mu M) and KPO are added4(100mM) and CYP1A2(0.01 pmol/. mu.L); in the blank, Membrance (0.01 pmol/. mu.L) was used in place of CYP1A2, and the other reagents added to the compound concentration group were added; in the negative control group, DMSO solvent without the test compound is used to replace the test compound solution, other reagents added in the blank group are added, each group is pre-incubated for 10min at room temperature, then substrate and NADP + are added, reaction is carried out for 30min at room temperature, finally, detection buffer with the same volume is added, and chemiluminescence detection is carried out after incubation for 20min at room temperature.
Inhibition of CYP2D 6: a compound concentration group, a blank group and a negative control group were set, respectively. In the compound concentration group, compounds to be detected with different concentrations are added into a micropore plate, and Luciferin-ME EGE (30 mu M) and KPO are added4(100mM) and CYP2D6(5 nM); in the blank, CYP2D6 was replaced with membrane (5nM) and the other reagents added in the compound concentration group were added; in the negative control group, the test compound solution was replaced with DMSO solvent containing no test compound, and the other reagents added to the blank group were added. Preincubation is carried out for 10min at room temperature, then substrate and NADP + are added for reaction for 30min at 37 ℃, finally an equal volume of detection buffer is added, and chemiluminescence detection is carried out after incubation for 20min at 25 ℃.
Data processing:
and calculating the inhibition rate according to the chemiluminescence detection signal values of the compound concentration group, the blank group and the negative control group. Percent inhibition (%) (1- (each compound concentration group signal-blank signal)/(negative control signal-blank signal)). 100. Half maximal Inhibitory Concentration (IC) of compounds was estimated based on their inhibition of P450 enzyme at different concentrations50) Or a range. IC (integrated circuit)50X (1-percent inhibition (%)/percent inhibition (%), where X is the compound concentration tested.
The experimental results are as follows:
the inhibition of both CYPs by the compounds was determined as described above and the results are shown in table 2.
TABLE 2 inhibition of CYP1A2 and CYP2D6 by compounds
Figure PCTCN2020081558-APPB-000043
The results show that the compound of the invention has no obvious inhibition effect on two main CYP subtypes, and the potential drug interaction possibility is relatively low, so that the compound has better drug property.
Experimental example 3: CYP3A4 enzyme inhibition assay (Mass Spectrometry)
Reagents and controls:
probe substrate: testosterone, midazolam,
Positive control: ketoconazole
Mixed Human Liver Microsome (HLM)
The experimental method comprises the following steps:
after 5min of pre-incubation (150. mu.l) of the probe substrate (50. mu.l), PBS (49. mu.l), test substance or positive control (1. mu.l) with the mixture (150. mu.l) of HLM (50. mu.l), NADPH (50. mu.l) was added and incubation was carried out for 30min, wherein the incubation concentration of HLM was 0.1 mg/mL. And adding 600 mu l of glacial acetonitrile containing rivaroxaban (internal standard) into a sample with testosterone as a substrate to terminate the reaction, and adding 800 mu l of glacial acetonitrile containing rivaroxaban (internal standard) into a sample with midazolam as a substrate to terminate the reaction. The reaction solution was vortexed and centrifuged to take the supernatant, and the concentration of the major metabolite produced by the probe substrate was measured.
The detection method comprises the following steps:
LC-MS/MS, Mass Spectrum API 5500, liquid phase Waters ACQUITY UPLC I-CLASS system. The chromatographic column is Hypersil GOLD C18Particle size 1.9 μm, 50X 2.1 mm; the mobile phase A is water and 0.1 percent formic acid, and the phase B is acetonitrile; the flow rate was 0.4mL/min and the column temperature was 40 ℃. The ion source is used as an ESI source positive ion mode, and the scanning mode is Multiple Reaction Monitoring (MRM).
Using a solvent group (DMSO) as a negative control, measuring the concentration of main metabolites generated by the probe substrate under different concentrations of the compound, and calculating the half number of the compoundInhibitory Concentration (IC)50)。
TABLE 3 inhibition of CYP3A4 by compounds
Compound (I) 3A4-M IC 50(μM) 3A4-T IC 50(μM)
Reference compound 2.21 10.7
Compound 9 28.3 >50
Compound 17 40.8 >50
Compound 18 7.48 11.8
Compound 20 6.66 24.0
Compound 2-A 17.9 21.2
Compound 2-B 68.9 >50
Note: m represents midazolam and T represents testosterone.
The results show that the tested compounds have no obvious inhibition effect or weak inhibition effect on CYP3A4 subtype, and the potential drug interaction possibility is relatively low, so that the compound has good drug property.
EXAMPLE 4 rat Pharmacokinetic (PK) study
The compounds of the invention were administered to male SD rats by Intravenous (IV) and intragastric (PO) administration, respectively, and pharmacokinetic profiles were examined. The dosages of IV and PO were 1mg/kg and 5mg/kg, respectively, the vehicle system was 5% DMSO, 5% Solutol, 90% physiological saline, and the vehicle of PO was 0.5% MC. IV and PO were administered and blood was collected at different time points and anticoagulated with EDTA. K2, centrifuged to obtain plasma samples, stored at-80 ℃. Plasma samples were processed for precipitated protein and analyzed by LC-MS/MS.
Pharmacokinetic parameters were calculated using the WinNonlin 6.3 software using a non-compartmental model, and the results are shown in tables 4 and 5.
TABLE 4 pharmacokinetic parameters of IV administered Compounds in rats
Figure PCTCN2020081558-APPB-000044
The data in Table 4 show that the compounds of the invention administered IV at a dose of 1mg/kg have excellent drug exposure in rats.
TABLE 5 pharmacokinetic parameters of PO dosed Compounds in rats
Figure PCTCN2020081558-APPB-000045
The data in table 5 show that the compounds of the invention administered with PO at a dose of 5mg/kg have excellent drug exposure and certain bioavailability in rats.
Combining tables 4 and 5, the compounds of the present invention have excellent plasma drug exposure and certain oral bioavailability in rats by IV and PO administration.
Other compounds of the invention also have better AUClastValue, CmaxThe value and bioavailability have better pharmacokinetic property in rats.
Various modifications of the invention in addition to those described herein will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference, including all patents, patent applications, journal articles, books, and any other publications, cited in this application is hereby incorporated by reference in its entirety.

Claims (16)

  1. A compound of formula I or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g. hydrate), metabolite, prodrug, or mixture thereof,
    Figure PCTCN2020081558-APPB-100001
    wherein,
    R 1selected from hydrogen and C1-6Alkyl radical, said C1-6Alkyl is optionally substituted with one or more substituents independently selected from: hydrogen, hydroxy, carboxy, - (C)1-6Alkoxy group)m1-(C 1-6Alkoxy group)m2、NR aR bAnd 5-6 membered hetero containing one or more heteroatoms independently selected from N, O and SCyclic radical, preferably R1Is C1-4Alkyl radical, said C1-4Alkyl is optionally substituted with one or more substituents independently selected from: hydroxy, C1-6Alkoxy and a 5-6 membered heterocyclyl containing one or more heteroatoms independently selected from N, O and S, more preferably, R1Is optionally substituted by one or more C1-4Alkoxy-substituted C1-4Alkyl, particularly preferably, R1Is 2-butoxyethyl;
    R 2is selected from C1-6Alkyl and 4-6 membered heterocyclyl containing one or more heteroatoms independently selected from N, O and S, said C1-6Alkyl and 4-6 membered heterocyclyl are optionally substituted with 1-3 substituents independently selected from: hydrogen, deuterium, halogen, hydroxy, carboxy, - (C)1-6Alkoxy group)m1-(C 1-6Alkoxy group)m2And NRaR bPreferably, R2Is selected from C1-3Alkyl and 4-6 membered heterocyclyl containing N and/or O atoms, said C1-3Alkyl is optionally substituted with 1-3 substituents independently selected from: hydrogen, halogen, hydroxy, carboxy, C1-4Alkoxy and NRaR bParticularly preferably, R2Is isopropyl, fluoromethyl, methoxymethyl, hydroxymethyl, 1-hydroxyisopropyl, N-dimethylaminomethyl, 1-carboxyisopropyl, tetrahydropyranyl, oxetanyl or morpholinyl;
    R 9、R 10absent, or each independently selected from halogen, hydroxy, carboxy, cyano, NRaR b、C 1-6Alkoxy radical, C1-6Alkyl radical, C1-6Haloalkyl and-COOC1-6Alkyl, preferably, R9、R 10Are not present;
    m 1、m 2each independentlySelected from 0, 1,2,3 or 4;
    R a、R beach independently selected from hydrogen and C1-6An alkyl group;
    y is selected from N or CH;
    a is selected from 4-7 membered heterocyclyl and 5-10 membered heteroaryl, said 4-7 membered heterocyclyl or 5-10 membered heteroaryl being optionally substituted with one or more substituents independently selected from: halogen, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-6Cycloalkyl radical, C3-6Halogenocycloalkyl, C3-6Cycloalkoxy and C1-6Haloalkyloxy, preferably, a is selected from a 4-7 membered nitrogen-containing heterocyclyl and a 5-10 membered nitrogen-containing heteroaryl, said 4-7 membered nitrogen-containing heterocyclyl and 5-10 membered nitrogen-containing heteroaryl being optionally substituted with 1-3 substituents independently selected from: halogen, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy and C1-6A haloalkoxy group; and is
    n is selected from 0, 1 or 2, preferably n is 1.
  2. The compound of claim 1, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, wherein the compound is of formula II:
    Figure PCTCN2020081558-APPB-100002
    wherein,
    R 1selected from hydrogen and C1-6Alkyl radical, said C1-6Alkyl is optionally substituted with one or more substituents independently selected from: hydrogen, hydroxy, carboxy, - (C)1-6Alkoxy group)m1-(C 1-6Alkoxy group)m2、NR aR bAnd 5-6 membered heterocyclyl containing one or more heteroatoms independently selected from N, O and S, preferably, R1Is C1-4Alkyl radical, said C1-4Alkyl is optionally substituted with one or more substituents independently selected from: hydroxy, C1-6Alkoxy and a 5-6 membered heterocyclyl containing one or more heteroatoms independently selected from N, O and S, more preferably, R1Is optionally substituted by one or more C1-4Alkoxy-substituted C1-4Alkyl, particularly preferably, R1Is 2-butoxyethyl;
    R 2is selected from C1-6Alkyl and 4-6 membered heterocyclyl containing one or more heteroatoms independently selected from N, O and S, said C1-6Alkyl and 4-6 membered heterocyclyl are optionally substituted with 1-3 substituents independently selected from: hydrogen, deuterium, halogen, hydroxy, carboxy, - (C)1-6Alkoxy group)m1-(C 1-6Alkoxy group)m2And NRaR bPreferably, R2Is selected from C1-3Alkyl and 4-6 membered heterocyclyl containing N and/or O atoms, said C1-3Alkyl is optionally substituted with 1-3 substituents independently selected from: hydrogen, halogen, hydroxy, carboxy, C1-4Alkoxy and NRaR bParticularly preferably, R2Is isopropyl, fluoromethyl, methoxymethyl, hydroxymethyl, 1-hydroxyisopropyl, N-dimethylaminomethyl, 1-carboxyisopropyl, tetrahydropyranyl, oxetanyl or morpholinyl;
    R 9、R 10absent, or each independently selected from halogen, hydroxy, carboxy, cyano, NRaR b、C 1-6Alkoxy radical, C1-6Alkyl radical, C1-6Haloalkyl and-COOC1-6Alkyl, preferably, R 9、R 10Are not present;
    m 1、m 2each independently selected from 0, 1,2,3 or 4;
    R a、R beach independently selected from hydrogen and C1-6An alkyl group;
    a is selected from 4-7 membered heterocyclyl and 5-10 membered heteroaryl, said 4-7 membered heterocyclyl or 5-10 membered heteroaryl being optionally substituted with one or more substituents independently selected from: halogen, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Haloalkoxy, C3-6Cycloalkyl radical, C3-6Halogenocycloalkyl, C3-6Cycloalkoxy and C1-6Haloalkyloxy, preferably, a is selected from a 4-7 membered nitrogen-containing heterocyclyl and a 5-10 membered nitrogen-containing heteroaryl, said 4-7 membered nitrogen-containing heterocyclyl and 5-10 membered nitrogen-containing heteroaryl being optionally substituted with 1-3 substituents independently selected from: halogen, C1-6Alkyl radical, C1-6Haloalkyl, C1-6Alkoxy and C1-6A haloalkoxy group; and is
    n is selected from 0, 1 or 2, preferably n is 1.
  3. The compound of claim 2, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, wherein the compound is of formula III,
    Figure PCTCN2020081558-APPB-100003
  4. a compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g. hydrate) thereof) A metabolite, prodrug, or mixture thereof, wherein A is
    Figure PCTCN2020081558-APPB-100004
    Figure PCTCN2020081558-APPB-100005
    Wherein,
    x is selected from N and C-R5
    R 3Selected from hydrogen, deuterium, C1-6Alkyl and C3-6Cycloalkyl radical, said C1-6Alkyl or C3-6Cycloalkyl is optionally substituted with one or more substituents independently selected from: hydrogen, deuterium, hydroxy, cyano, C1-4Alkoxy and C3-6Cycloalkoxy, preferably R3Is C1-4Alkyl radical, said C1-4Alkyl is optionally substituted with one or more substituents independently selected from: hydrogen, hydroxy and C1-4Alkoxy, particularly preferably, R3Selected from n-propyl and CH3OCH 2CH 2-; and is
    R 4、R 5Each independently selected from hydrogen, deuterium, halogen, cyano, C1-6Alkyl radical, C3-6Cycloalkyl, 3-6 membered heterocyclyl, C6-10Aryl and 5-12 membered heteroaryl, wherein said C1-6Alkyl radical, C3-6Cycloalkyl, 3-6 membered heterocyclyl, C6-10Aryl or 5-12 membered heteroaryl are each optionally substituted with one or more substituents independently selected from: hydrogen, deuterium, halogen, cyano, hydroxy and C1-6Alkoxy, preferably, R4、R 5Each independently selected from hydrogen, deuterium, halogen, C1-6Alkyl, halo C1-6Alkyl and C3-6A cycloalkyl group,particularly preferably, R4、R 5Each independently selected from hydrogen, fluoro, methyl, trifluoromethyl and cyclopropyl.
  5. The compound of claim 4, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, wherein the compound is of formula IV,
    Figure PCTCN2020081558-APPB-100006
  6. the compound of any one of claims 1-5, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, wherein
    R 1Selected from the group consisting of 2-butoxyethyl, 2-methoxyethyl, and butyl.
  7. The compound of any one of claims 1-5, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, wherein
    R 2Selected from the group consisting of isopropyl, fluoromethyl, methoxymethyl, hydroxymethyl, 1-hydroxyisopropyl, N-dimethylaminomethyl, 1-carboxyisopropyl, tetrahydropyranyl, oxetanyl, morpholinyl, and 1, 4-dioxanyl.
  8. The compound of claim 1, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, wherein said compound is selected from the group consisting of:
    Figure PCTCN2020081558-APPB-100007
    Figure PCTCN2020081558-APPB-100008
    Figure PCTCN2020081558-APPB-100009
  9. the compound of claim 8, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, wherein said compound is selected from the group consisting of:
    Figure PCTCN2020081558-APPB-100010
    Figure PCTCN2020081558-APPB-100011
    Figure PCTCN2020081558-APPB-100012
    Figure PCTCN2020081558-APPB-100013
    Figure PCTCN2020081558-APPB-100014
  10. a pharmaceutical composition comprising an effective amount of a compound of any one of claims 1-9, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, and one or more pharmaceutically acceptable carriers.
  11. The pharmaceutical composition of claim 10, further comprising one or more additional agents for the prevention or treatment of a disease or condition mediated by CCR2 and/or CCR5, in particular non-alcoholic fatty liver disease (NAFLD).
  12. A kit comprising a compound of any one of claims 1-9, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of claim 10 or 11.
  13. The kit of claim 12, comprising:
    a) a first container comprising as a first therapeutic agent at least one compound of any one of claims 1-9 or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g., hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of claim 10 or 11 as a first pharmaceutical composition;
    b) optionally a second container comprising at least one further therapeutic agent as a second therapeutic agent, or a pharmaceutical composition comprising said further therapeutic agent as a second pharmaceutical composition; and
    c) optionally product instructions.
  14. Use of a compound of any one of claims 1 to 9, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate (e.g. hydrate), metabolite, prodrug, or mixture thereof, or a pharmaceutical composition of claim 10 or 11, or a kit of claim 12 or 13, for the manufacture of a medicament for the prevention or treatment of a disease or condition mediated by CCR2 and/or CCR5, in particular non-alcoholic fatty liver disease (NAFLD).
  15. A process for preparing a compound of claim 1, comprising the steps of:
    Figure PCTCN2020081558-APPB-100015
    wherein R is1、R 2、R 9、R 10A, Y and n are as defined in claim 1; and LG is a leaving group.
  16. A process for preparing a compound of claim 2, comprising the steps of:
    Figure PCTCN2020081558-APPB-100016
    wherein R is1、R 2、R 9、R 10A and n are as defined in claim 2; and LG is a leaving group.
CN202080013096.9A 2019-04-08 2020-03-27 Benzimidazole compounds, preparation method and use thereof Pending CN113412261A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN201910275494 2019-04-08
CN2019102754947 2019-04-08
PCT/CN2020/081558 WO2020207263A1 (en) 2019-04-08 2020-03-27 Benzimidazole compound, preparation method therefor and use thereof

Publications (1)

Publication Number Publication Date
CN113412261A true CN113412261A (en) 2021-09-17

Family

ID=72750884

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202080013096.9A Pending CN113412261A (en) 2019-04-08 2020-03-27 Benzimidazole compounds, preparation method and use thereof

Country Status (2)

Country Link
CN (1) CN113412261A (en)
WO (1) WO2020207263A1 (en)

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2380860A1 (en) * 1999-06-16 2000-12-21 Takeda Chemical Industries, Ltd. Benzazepine derivative, production and use thereof
WO2002022599A2 (en) * 2000-09-15 2002-03-21 Anormed Inc. Chemokine receptor binding heterocyclic compounds
WO2003014105A1 (en) * 2001-08-08 2003-02-20 Takeda Chemical Industries, Ltd. Bicyclic compound, production and use as hiv inhibitors
JP2004256531A (en) * 2003-02-07 2004-09-16 Takeda Chem Ind Ltd Tricyclic ring compound, method for producing the same and use of the same
CN1956980A (en) * 2004-05-26 2007-05-02 詹森药业有限公司 Mercaptoimidazoles as CCR2 receptor antagonists
WO2012051361A1 (en) * 2010-10-13 2012-04-19 Abbott Laboratories Anti-viral compounds
CN106518946A (en) * 2015-09-10 2017-03-22 上海迪诺医药科技有限公司 Sulfonylurea derivative and pharmaceutical composition and application thereof
CN107001279A (en) * 2014-12-01 2017-08-01 埃科特莱茵药品有限公司 Cxcr7 receptor modulators
CN109071468A (en) * 2017-01-20 2018-12-21 四川科伦博泰生物医药股份有限公司 A kind of heterocyclic compound and its preparation method and application
CN111263759A (en) * 2017-12-22 2020-06-09 四川科伦博泰生物医药股份有限公司 Isoxazole derivative and preparation method and application thereof
CN111630047A (en) * 2018-02-02 2020-09-04 四川科伦博泰生物医药股份有限公司 Benzoazaheterocycle compound containing carboxylic acid group and preparation method and application thereof
CN114096245A (en) * 2019-06-24 2022-02-25 深圳市瓴方生物医药科技有限公司 Heterocycloalkyl compounds as CCR2/CCR5 antagonists
CN115667247A (en) * 2020-05-22 2023-01-31 南京明德新药研发有限公司 Pyridine derivatives and application thereof

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2380860A1 (en) * 1999-06-16 2000-12-21 Takeda Chemical Industries, Ltd. Benzazepine derivative, production and use thereof
WO2002022599A2 (en) * 2000-09-15 2002-03-21 Anormed Inc. Chemokine receptor binding heterocyclic compounds
WO2003014105A1 (en) * 2001-08-08 2003-02-20 Takeda Chemical Industries, Ltd. Bicyclic compound, production and use as hiv inhibitors
JP2004256531A (en) * 2003-02-07 2004-09-16 Takeda Chem Ind Ltd Tricyclic ring compound, method for producing the same and use of the same
CN1956980A (en) * 2004-05-26 2007-05-02 詹森药业有限公司 Mercaptoimidazoles as CCR2 receptor antagonists
CN103153988A (en) * 2010-10-13 2013-06-12 Abbvie公司 Anti-viral compounds
WO2012051361A1 (en) * 2010-10-13 2012-04-19 Abbott Laboratories Anti-viral compounds
CN107001279A (en) * 2014-12-01 2017-08-01 埃科特莱茵药品有限公司 Cxcr7 receptor modulators
CN106518946A (en) * 2015-09-10 2017-03-22 上海迪诺医药科技有限公司 Sulfonylurea derivative and pharmaceutical composition and application thereof
CN109071468A (en) * 2017-01-20 2018-12-21 四川科伦博泰生物医药股份有限公司 A kind of heterocyclic compound and its preparation method and application
CN111263759A (en) * 2017-12-22 2020-06-09 四川科伦博泰生物医药股份有限公司 Isoxazole derivative and preparation method and application thereof
CN111630047A (en) * 2018-02-02 2020-09-04 四川科伦博泰生物医药股份有限公司 Benzoazaheterocycle compound containing carboxylic acid group and preparation method and application thereof
CN114096245A (en) * 2019-06-24 2022-02-25 深圳市瓴方生物医药科技有限公司 Heterocycloalkyl compounds as CCR2/CCR5 antagonists
CN115667247A (en) * 2020-05-22 2023-01-31 南京明德新药研发有限公司 Pyridine derivatives and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MASAKI SETO ET AL.: "Highly Potent and Orally Active CCR5 Antagonists as Anti-HIV-1 Agents: Synthesis and Biological Activities of 1-Benzazocine Derivatives Containing a Sulfoxide Moiety", 《JOURNAL OF MEDICINAL CHEMISTRY》 *
姚虎等: "非肽类小分子趋化因子受体5拮抗剂的研究进展", 《中国新药杂志》 *

Also Published As

Publication number Publication date
WO2020207263A1 (en) 2020-10-15

Similar Documents

Publication Publication Date Title
CN109071498B (en) Kinase inhibitor and preparation method and application thereof
TWI711619B (en) Aminopyridazinone compounds as protein kinase inhibitors
WO2019196812A1 (en) Protein degradation targeting compound, anti-tumor application, intermediate thereof and use of intermediate
WO2023011513A1 (en) Shp2 inhibitor, pharmaceutical composition comprising same, and application thereof
WO2019134539A1 (en) Dihydropyrazolone and pyrimidine compound, preparation method and use therefor
TW201443023A (en) Phthalazinones and isoquinolinones as ROCK inhibitors
WO2020011246A1 (en) Benzene ring-containing compound, preparation method therefor and application thereof
CN112424200B (en) Iminosulfones compound as bromodomain protein inhibitor, pharmaceutical composition and medical application thereof
JP7207634B2 (en) P2X3 and/or P2X2/3 receptor antagonists, pharmaceutical compositions containing same and uses thereof
WO2019101086A1 (en) Halo-allylamine ssao/vap-1 inhibitor and use thereof
US20240166645A1 (en) Pyridopyrimidinone derivative, preparation method therefor, and use thereof
TWI833819B (en) Compounds and compositions for treating conditions associated with apj receptor activity
WO2022121813A1 (en) Sos1 inhibitor, pharmaceutical composition comprising same, and use thereof
TW202214639A (en) Pyridonelopyrimidine derivative, preparation method thereof, and medical use thereof
WO2022028506A1 (en) Sos1 inhibitor, pharmaceutical composition containing same, and use therefor
WO2023016484A1 (en) Sulfonamide derivative, preparation method therefor and medical use thereof
JP2023533003A (en) heterocyclic immunomodulators
TW202128686A (en) A fused heteroaryl derivative, a preparation method and medical use thereof
CN113412261A (en) Benzimidazole compounds, preparation method and use thereof
EP4219453A1 (en) Pyrazole compound and preparation method therefor and use thereof
WO2020151232A9 (en) 1,2,3,4-tetrahydroquinoxaline derivative, preparation method therefor and application thereof
CN113924293B (en) P2X3 and/or P2X2/3 receptor antagonists, pharmaceutical compositions containing them and their use
CN112119065B (en) Benzodiazepine compound, preparation method and application thereof
JP2024516860A (en) 2,8-DIHYDROPYRAZOLO[3,4-B]INDOLE DERIVATIVES FOR USE IN THE TREATMENT OF CANCER - Patent application
WO2023061263A1 (en) Shp2 inhibitor, pharmaceutical composition comprising same, and use 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
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20210917

WD01 Invention patent application deemed withdrawn after publication