CN107880023B - Fluoroquinolone amino derivative and application thereof - Google Patents

Fluoroquinolone amino derivative and application thereof Download PDF

Info

Publication number
CN107880023B
CN107880023B CN201711102862.5A CN201711102862A CN107880023B CN 107880023 B CN107880023 B CN 107880023B CN 201711102862 A CN201711102862 A CN 201711102862A CN 107880023 B CN107880023 B CN 107880023B
Authority
CN
China
Prior art keywords
group
compound
acid
halogen
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201711102862.5A
Other languages
Chinese (zh)
Other versions
CN107880023A (en
Inventor
杨大成
冯计周
范莉
黄敏
谢建平
刘洁
胡军华
陈菲菲
牟希
徐兴然
郭思瑶
付彬
龙艳玲
杨真
王帆
张书虹
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.)
Southwest University
Original Assignee
Southwest University
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 Southwest University filed Critical Southwest University
Priority to CN201711102862.5A priority Critical patent/CN107880023B/en
Priority to CN201911252190.5A priority patent/CN111087390B/en
Publication of CN107880023A publication Critical patent/CN107880023A/en
Application granted granted Critical
Publication of CN107880023B publication Critical patent/CN107880023B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The invention belongs to the field of medicinal chemistry, and particularly relates to a fluoroquinolone derivative and application thereof. The compound shown in the formula I is obtained by carrying out structural modification on fluoroquinolone medicaments, and the compound not only can treat infection caused by mycobacterium tuberculosis and common bacteria, but also has inhibitory activity on bacterial persistent bacteria, citrus pathogenic bacteria, nicotinamide N-methyltransferase (NNMT) and interleukin IL-17 PPI. The preparation process of the compound is simple and convenient to operate, the conditions are mild, a plurality of compounds with enhanced antibacterial activity, improved water solubility and reduced toxic and side effects are obtained, the dosage of the compound is expected to be reduced, the treatment period is shortened, the compliance of patients is improved, and a new molecular type and a new research idea are provided for research of tuberculosis drugs and other diseases.

Description

Fluoroquinolone amino derivative and application thereof
Technical Field
The invention belongs to the field of medicinal chemistry, and particularly relates to a fluoroquinolone amino derivative and application thereof.
Background
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium Tuberculosis (MTB) that is mainly transmitted through the respiratory tract. MTB mainly causes tuberculosis, can affect all other organs of human body, is one of serious diseases seriously harming human health, and is a public health problem and a social problem which are commonly concerned for a long time in the world. In the process of using antitubercular drugs, drug-resistant tuberculosis (DR-TB), especially multidrug-resistant tuberculosis, is generated and gradually increased, and the suppression of drug-resistant tuberculosis has become a very urgent research subject.
The results and experience of tuberculosis treatment in the world show that no matter how strong the bactericidal activity of a single antitubercular drug is, no matter whether the drug is an old drug (such as streptomycin or isoniazid) or a new drug developed in recent 2 years (such as bedaquiline and delamanic), corresponding drug-resistant strains always appear as long as the drug is used singly. Therefore, the WHO recommended tuberculosis treatment regimens are all combinations of different types of clinical tuberculosis drugs. The recommended treatment scheme for drug-resistant tuberculosis contains 5 effective antitubercular drugs, wherein the prescription is made by WHO (2016 updated edition). Although the cure rate of tuberculosis can reach more than 90 percent at present, the use of the standard scheme faces a plurality of challenges due to the defects of long treatment course, large adverse drug reaction and the like, and the reasons of self-stopping of the drug, poor drug administration compliance, lack of necessary support measures and the like of patients. In addition, drug-resistant tuberculosis is emerging continuously, which makes the treatment of tuberculosis more difficult. Therefore, the research and development of novel antibacterial drugs, the optimization of treatment schemes, the improvement of curative effects, the shortening of treatment cycles, the improvement of treatment compliance, and the great significance in the defense and treatment of tuberculosis and other diseases are achieved.
The human body is a complex organism, and in multi-mechanism diseases, the single action on a certain target spot is used for treatment, the action effect is not quite satisfactory, target mutation or bypass regulation and the like can be caused, so that serious side effects are generated, the tolerance of the organism to the medicine is enhanced, and the treatment effect is difficult to ensure for a long time. To solve this problem, a multi-target drug concept rationally designed based on the overall objective of improving the therapeutic effect and/or improving safety has attracted attention. The multi-target medicine can act on a plurality of target points related to diseases, generate a plurality of pharmacological activities, effectively regulate a whole cell complex system, incompletely eliminate the relationship among all members in a signal transmission system and reduce side effects. According to the difference of composition and action targets, the multiple targets can be divided into three types of medicines, namely 'multi-medicine multi-target' medicine (medicine combination), 'multi-medicine single-target' medicine (multi-component medicine) and 'single-medicine multi-target' medicine, wherein the first two types of medicines relate to two or more medicines when in administration, and due to the complexity of interaction between the medicines, the two administration modes can have the defects of increased difficulty of dose control, increased medicine compatibility risk, poor compliance of patients and the like, so that the single-medicine multi-target compound design gradually becomes a hot point direction of new medicine design. In recent years, shuffling of molecules with biological activity (i.e., combining two or more different pharmacophores of different biologically active molecules to form new molecules) has become one of the research hotspots in the field of medicinal chemistry. The structural modification of the original drug or the candidate drug with better activity and the active fragment thereof is a strategy for developing novel anti-tuberculosis drugs.
Quinolone drugs are synthetic antibacterial drugs consisting of aromatic rings such as pyridone acid biphenyl or pyridine, have unique action mechanisms, can quickly kill bacteria, and have excellent pharmacokinetic properties, so that the quinolone drugs have been popular clinical treatment drugs. Fluoroquinolone (FQs) drugs are third and fourth generation quinolone drugs developed in recent years, have good early bactericidal activity, are one of the main drugs for treating drug-resistant tuberculosis at present, and are mostly used as second-line antitubercular drugs for adjuvant therapy of drug-resistant tuberculosis or tuberculosis patients intolerant to the first-line antitubercular drugs. FQs class of drugs not only have good antituberculosis activity, but also have good pharmacokinetic properties, such as good oral bioavailability, high blood concentration, wide tissue distribution, long drug half-life, less adverse reaction, and no cross-resistance with other types of anti-mycobacterial drugs.
Isoniazid (INH) is a classic antituberculotic drug, and the target of action is InhA. Since 1952 for tuberculosis treatment, INH remains one of the most effective first-line drugs for tuberculosis treatment. Recent research shows that the modification of terminal amino group of INH hydrazide can form hydrazone or heterocyclic compound or combine terminal amino group of hydrazide with other pharmacodynamic group to form new molecule by using heterocycle as Linker, and has better activity against mycobacterium tuberculosis. Therefore, the isoniazid structural modification has better research and development prospect. Considering that INH has small molecular weight, low price, high activity, heterocyclic ring-containing and special action mechanism, and can be conjugated with other molecules in various ways, INH and variant isonicotinic acid thereof are preferably taken as active fragments to be connected with fluoroquinolone active fragments, and a single-drug multi-target drug with the activity of treating drug-resistant tuberculosis is expected to be obtained.
Pyrazinamide (PZA) is a unique first-line antitubercular drug: the activity of the compound is not ideal, but the compound shows strong bactericidal activity and synergistic effect when being used in combination with other anti-tuberculosis drugs; most researchers believe that PZA and INH are also prodrugs, activated by a mycobacterium tuberculosis-specific PZA activating enzyme, producing pyrazinoic acid (POA). Therefore, PZA and POA can be used as active fragments to be connected with fluoroquinolone active fragments, and a new single-drug multi-target drug with the activity of treating drug-resistant tuberculosis is expected to be obtained.
Disclosure of Invention
The invention selects representative fluoroquinolone drugs to be connected with isoniazid/isonicotinic acid or pyrazinamide/pyrazinoic acid by chemical bonds through a connecting structure so as to obtain active molecules with enhanced antibacterial activity and reduced toxic and side effects, and can treat pulmonary tuberculosis and secondary common bacterial infection at the same time, thereby reducing the dosage and the drug types, shortening the treatment period and improving the compliance of patients. Surprisingly, in the process of multi-target screening of medicines for the compounds designed and prepared by the application, the compounds are unexpectedly found to have inhibitory effects on bacterial persisting bacteria, citrus pathogenic bacteria, nicotinamide N-methyltransferase (NNMT) and interleukin IL-17PPI, and have further research values.
The invention provides a compound shown as a formula I, a racemate, a stereoisomer, a tautomer, a nitrogen oxide or a pharmaceutically acceptable salt thereof:
Figure BDA0001463694600000031
wherein X is selected from: C1-C6 alkyl; C3-C6 cycloalkyl; a substituted or unsubstituted C6-C10 aryl group, the substituents on the aryl group being one or more independently selected from: halogen; an amino group; a hydroxyl group; C1-C6 alkyl; C3-C6 cycloalkyl;
z is selected from: n or C-R7;R7Selected from: h; C1-C6 alkoxy or halogen;
R1and R2Each independently selected from: h; C1-C6 alkyl; C1-C6 haloalkyl; C1-C6 alkoxy; halogen; a hydroxyl group; alkylamino of C1-C6; amino or cyano;
R3、R4together with the attached groups, form a substituted or unsubstituted 5-7 membered saturated heterocyclic ring containing at least 1N atom, the substituents on the saturated heterocyclic ring being one or more independently selected from: halogen; an amino group; a hydroxyl group; C1-C6 alkyl; C3-C6 cycloalkyl; r5Selected from H, C1-C6 alkyl;
or R3、R5Taken together with the attached group to form a 5-7 membered saturated heterocyclic ring containing 2N atoms; r4Selected from: h; halogen; an amino group; a hydroxyl group; C1-C6 alkyl;
a is selected from
Figure BDA0001463694600000032
Carboxy or halogen; het is a 5-7 membered heteroaromatic ring containing at least 1 nitrogen atom; r6 is selected from H, C1-C6 alkyl;
n, k are independently selected from integers of 0 to 3.
Preferably, the invention also provides a compound shown as a formula II and a formula III, a racemate, a stereoisomer, a tautomer, a nitrogen oxide or a pharmaceutically acceptable salt thereof:
Figure BDA0001463694600000033
Figure BDA0001463694600000041
wherein X is selected from: C1-C6 alkyl; C3-C6 cycloalkyl; a substituted or unsubstituted C6-C10 aryl group, the substituents on the aryl group being one or more independently selected from: halogen; an amino group; a hydroxyl group; C1-C6 alkyl; C3-C6 cycloalkyl;
z is selected from: n or C-R7;R7Selected from: h; C1-C6 alkoxy or halogen;
R1and R2Each independently selected from: h; C1-C6 alkyl; C1-C6 haloalkyl; C1-C6 alkoxy; halogen; a hydroxyl group; alkylamino of C1-C6; amino or cyano;
R4selected from: h; halogen; an amino group; a hydroxyl group; C1-C6 alkyl;
R5selected from H, C1-C6 alkyl;
a is selected from
Figure BDA0001463694600000042
Or halogen;
het is a 5-7 membered heteroaromatic ring containing at least 1 nitrogen atom; r6 is selected from H, C1-C6 alkyl;
n, k are independently selected from integers from 0 to 3;
m is 1 or 2.
As a further preference, a is selected from the following structures:
Figure BDA0001463694600000043
in certain classes of preparations/embodiments, the present invention also provides compounds of formula IV, formula V, formula VI, or formula VII, racemates, stereoisomers, tautomers, nitroxides thereof, or pharmaceutically acceptable salts thereof:
Figure BDA0001463694600000051
x is selected from: a methyl group; an ethyl group; propyl; a butyl group; a cyclopropyl group; halogen substituted or unsubstituted phenyl;
z is selected from: n or C-R7;R7Selected from: h; a methoxy group; an ethoxy group; fluorine or chlorine;
wherein R is1And R2Each independently selected from: h; a methyl group; an ethyl group; a trifluoromethyl group; a methoxy group; an ethoxy group; halogen; a hydroxyl group or an amino group;
r4 is selected from: h; halogen; an amino group; a hydroxyl group; methyl or ethyl;
r5 is selected from H; methyl or ethyl;
a is selected from
Figure BDA0001463694600000052
Figure BDA0001463694600000061
n is 1 or 2 or 3;
m is 1 or 2.
In certain preparations/embodiments, the present invention also provides compounds of formula VIII, formula IX, racemates, stereoisomers, tautomers, nitroxides or pharmaceutically acceptable salts thereof;
Figure BDA0001463694600000062
x is selected from: a methyl group; an ethyl group; propyl; a butyl group; a cyclopropyl group; halogen substituted or unsubstituted phenyl;
z is selected from: n or C-R7;R7Is selected from H; a methoxy group; an ethoxy group; fluorine or chlorine;
R4selected from: halogen; an amino group; a hydroxyl group; methyl or ethyl;
R5is selected from H; methyl or ethyl;
n is 1 or 2 or 3;
m is 1 or 2.
The present invention also provides the compound according to preparation/example, its racemate, stereoisomer, tautomer, nitroxide or pharmaceutically acceptable salt thereof.
The invention also claims a pharmaceutical composition comprising any of the above compounds, racemates, stereoisomers, tautomers, nitric oxides or pharmaceutically acceptable salts thereof.
Preferably, any compound, racemate, stereoisomer, tautomer, nitrogen oxide or pharmaceutically acceptable salt thereof can be prepared into any pharmaceutically acceptable dosage form, namely the pharmaceutical composition of the invention can also comprise pharmaceutically acceptable carriers and/or auxiliary agents.
Preferably, any of the compounds of the present invention, racemates, stereoisomers, tautomers, nitroxides or pharmaceutically acceptable salts thereof can be prepared into a clinical diagnostic reagent (kit).
Preferably, one or more other active ingredients and any compound, racemate, stereoisomer, tautomer, nitrogen oxide or pharmaceutically acceptable salt thereof can be prepared into a compound medicament according to the requirements.
The invention also claims the use of any of the above compounds, racemates, stereoisomers, tautomers, nitric oxides or pharmaceutically acceptable salts thereof in the manufacture of a medicament for the treatment of a disease selected from the group consisting of a disease caused by infection with a bacterium and a bacterium harboring bacterium, a disease caused by infection with a citrus bacterium, an inflammatory disease or a tumor. Such bacteria include, but are not limited to: mycobacterium tuberculosis, Staphylococcus aureus, Escherichia coli, Pseudomonas aeruginosa, or Salmonella. The citrus pathogenic bacteria are selected from the group consisting of: citrus canker pathogens; citrus brown rot or citrus green mold; the inflammatory disease is an IL-17 related inflammatory disease, preferably Rheumatoid Arthritis (RA), asthma, Multiple Sclerosis (MS), Chronic Obstructive Pulmonary Disease (COPD), Acute Respiratory Distress Syndrome (ARDS), Idiopathic Pulmonary Fibrosis (IPF), Inflammatory Bowel Disease (IBD), crohn's disease, uveitis, macular degeneration, colitis, psoriasis, wallerian degeneration, antiphospholipid antibody syndrome (APS), acute coronary syndrome, restenosis, atherosclerosis, Relapsing Polychondritis (RP), acute or chronic hepatitis, failed orthopedic implants, glomerulonephritis, lupus and autoimmune diseases. The tumor is selected from: lung cancer, kidney cancer, colorectal cancer, nasopharyngeal cancer, breast cancer, pancreatic cancer, liver cancer, gastric cancer, brain glioma, bladder cancer, and oral squamous cell carcinoma or medulloblastoma.
The invention also claims application of the compound, the stereoisomer or the pharmaceutically acceptable salt thereof in preparing a tumor diagnosis reagent.
Terms definition and interpretation:
unless otherwise indicated, the definitions of groups and terms described in the specification and claims of the present application, including definitions thereof as examples, exemplary definitions, preferred definitions, definitions described in tables, definitions of specific compounds in the examples, and the like, may be arbitrarily combined and coupled with each other. The definitions of the groups and the structures of the compounds in such combinations and after the combination are within the scope of the present specification.
When "the present compound" or "the present compound" is used herein, unless otherwise specified, at least the compound represented by the formula (I), its racemate, stereoisomer, tautomer, nitrogen oxide, or a pharmaceutically acceptable salt thereof is intended to be encompassed.
The term "tautomer" refers to an isomer of a functional group resulting from the rapid movement of an atom in two positions in a molecule. The compounds of the invention may exhibit tautomerism. Tautomeric compounds may exist in two or more interconvertible species. Prototropic tautomers result from the migration of a covalently bonded hydrogen atom between two atoms. Tautomers generally exist in equilibrium, and attempts to isolate a single tautomer often result in a mixture whose physicochemical properties are consistent with the mixture of compounds. The position of equilibrium depends on the chemical properties within the molecule. For example, in many aliphatic aldehydes and ketones such as acetaldehyde, the keto form predominates; whereas in phenol the enol type predominates. The present invention encompasses all tautomeric forms of the compounds.
The term "halogen" refers to F, Cl, Br and I. In other words, F, Cl, Br, and I may be described as "halogen" in the present specification.
The term "C1-C6" is understood to mean preferably a straight-chain or branched, saturated monovalent hydrocarbon radical having from 1 to 6 carbon atoms, for example, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, 2-methylbutyl, 1-ethylpropyl, 1, 2-dimethylpropyl, neopentyl, 1-dimethylpropyl, 4-methylpentyl, 3-methylpentyl, 2-methylpentyl, 1-methylpentyl, 2-ethylbutyl, 1-ethylbutyl, 3-dimethylbutyl, 2-dimethylbutyl, 1-dimethylbutyl, 2, 3-dimethylbutyl, 1, 3-dimethylbutyl, or 1, 2-dimethylbutyl, and the like, or isomers thereof.
The term "5-7 membered saturated heterocyclic group containing 1 or 2N atoms" may include, but is not limited to: 5-membered rings such as pyrrolidinyl, imidazolidinyl, pyrazolidinyl; or a 6-membered ring, such as piperidinyl, morpholinyl, thiomorpholinyl, or piperazinyl; or a 7-membered ring, such as azepanyl.
The term "C6-C10 aryl" is understood to mean preferably an aromatic or partially aromatic monocyclic, bicyclic or tricyclic hydrocarbon ring having 6 to 10 carbon atoms, in particular a ring having 6 carbon atoms ("C)6Aryl "), such as phenyl; or biphenyl, or is a compound havingA ring of 9 carbon atoms (' C)9Aryl), such as indanyl or indenyl, or a ring having 10 carbon atoms ("C10Aryl), such as tetralinyl, dihydronaphthyl, or naphthyl.
The term "5-7 membered heteroaromatic ring containing at least 1 nitrogen atom" is understood to have 5-7 ring atoms and comprises at least 1N atom, in particular the heteroaryl group is selected from pyrrolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl and the like.
Where a range of numerical values is recited in the specification and claims herein, and where the range of numerical values is defined as an "integer," it is understood that the two endpoints of the range are recited and each integer within the range is recited. For example, "any integer from 0 to 4" should be understood to recite each integer from 0, 1,2,3, 4.
The pharmaceutically acceptable salts of the compounds of the present invention may be acidic salts or basic salts. Pharmaceutically acceptable salts may be acid addition salts of the compounds of the invention having sufficient basicity, for example having a nitrogen atom in the chain or ring, for example with the following inorganic acids: for example hydrochloric, hydrofluoric, hydrobromic, hydroiodic, sulfuric, pyrosulfuric, phosphoric or nitric acid, or hydrogen sulfates, or the acid addition salts with the following organic acids: such as formic acid, acetic acid, acetoacetic acid, pyruvic acid, trifluoroacetic acid, propionic acid, butyric acid, caproic acid, heptanoic acid, undecanoic acid, lauric acid, benzoic acid, salicylic acid, 2- (4-hydroxybenzoyl) benzoic acid, camphoric acid, cinnamic acid, cyclopentanepropionic acid, 3-hydroxy-2-naphthoic acid, nicotinic acid, pamoic acid, pectinic acid, persulfuric acid, 3-phenylpropionic acid, picric acid, pivalic acid, 2-hydroxyethanesulfonic acid, itaconic acid, sulfamic acid, trifluoromethanesulfonic acid, dodecylsulfuric acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, methanesulfonic acid, 2-naphthalenesulfonic acid, naphthalenedisulfonic acid, camphorsulfonic acid, citric acid, tartaric acid, stearic acid, lactic acid, oxalic acid, malonic acid, succinic acid, malic acid, adipic acid, alginic acid, maleic acid, fumaric acid, D-gluconic acid, mandelic acid, malic acid, adipic acid, alginic acid, fumaric acid, ascorbic acid, glucoheptylic acid, glycerophosphate, aspartic acid, sulfosalicylic acid, hemisulfuric acid, or thiocyanic acid. In addition, another suitable pharmaceutically acceptable salt of a compound of the invention which is sufficiently acidic is an alkali metal salt (e.g., sodium or potassium salt), an alkaline earth metal salt (e.g., calcium or magnesium salt), an ammonium salt, or a salt with an organic base which affords a physiologically acceptable cation, such as a salt with: sodium ions, potassium ions, N-methylglucamine, dimethylglucamine, ethylglucamine, lysine, dicyclohexylamine, 1, 6-hexanediamine, ethanolamine, glucosamine, meglumine, sarcosine, serinol, trihydroxymethylaminomethane, aminopropanediol, 1-amino-2, 3, 4-butanetriol.
The compounds of the invention are preferably applied internally and externally in unmodified form or preferably together with adjuvants conventionally used in formulations in the art and can therefore be processed in known ways to give, for example, liquid formulations (sprays, emulsions, suspensions, solutions, emulsifiable concentrates, solution concentrates), semisolid formulations (e.g. creams, ointments, patches, gels, liposomal formulations) and solid formulations (e.g. powders, granules, tablets, etc.).
Examples of the term pharmaceutically acceptable carriers and/or adjuvants are: sugars such as lactose, sucrose, mannitol, and sorbitol; starches, such as corn starch, tapioca starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and methyl cellulose; calcium phosphates such as dicalcium phosphate and tricalcium phosphate; sodium sulfate; calcium sulfate; polyvinylpyrrolidone; polyvinyl alcohol; stearic acid; alkaline earth metal stearates, such as magnesium stearate and calcium stearate; vegetable oils such as peanut oil, cottonseed oil, sesame oil, olive oil and corn oil; nonionic, cationic and anionic surfactants; polyethylene glycol; fatty alcohols; and grain hydrolyzed solids and other nontoxic compatible excipients commonly used in pharmaceutical formulations, such as fillers, binders, disintegrants, buffers, preservatives, antioxidants, lubricants, colorants, and the like.
The invention has the beneficial effects that:
1) the invention carries out structural modification and improvement on fluoroquinolone medicaments to obtain a new active molecule with enhanced antibacterial activity and reduced toxic and side effects. The compound of the invention shows good in vitro antibacterial activity, and the antibacterial activity on mycobacterium tuberculosis, escherichia coli and staphylococcus aureus is equivalent to that of fluoroquinolone medicaments. The compound is expected to reduce the dosage and combination types of medicaments when used in medicaments, shorten the treatment period, improve the compliance of patients and provide new molecular types and research ideas for the medicament research of tuberculosis and other diseases.
2) The compound also has an inhibiting effect on bacterial persistent bacteria, citrus pathogenic bacteria, nicotinamide N-methyltransferase (NNMT) and interleukin IL-17PPI, and has further research value.
Detailed Description
Hereinafter, preferred embodiments of the present invention will be described in detail. The experimental methods of the preferred embodiments, which do not indicate specific conditions, are generally performed according to conventional conditions, and the examples are given for better illustration of the present invention, but the present invention is not limited to the examples. Therefore, those skilled in the art should make insubstantial modifications and adaptations to the embodiments of the present invention in light of the above teachings and remain within the scope of the invention.
Preparation examples of target Compounds
Synthesis of a series of compounds of formula XI:
Figure BDA0001463694600000101
preparation examples 1 to 24
Adding 1mmol of floxacin and 2mL of DCM into a 100mL round-bottom flask, cooling in an ice bath, magnetically stirring, and adding 3mmol of NaHCO3After 20min, 2.5mmol of chlorocarbonyl chloride (Cl- (CH) were added dropwise from a constant pressure dropping funnel2)nCOCl, n ═ 1,2,3) in DCM (2mL) solution (drop rate about 1d/2s), the reaction was continued in a dropwise ice bath (2-CC, shift to-5 ℃) and TLC tracking was monitored to the end of the reaction. Stopping stirring, adding H2O15 mL and DCM 20mL, pH adjusted to 3-4 with 1N HCl solution under stirring, ifStanding and filtering if solid exists, washing a filter cake for 3 times by using DCM, and reserving the filter cake for further purification; the filtrate was transferred to a separatory funnel, separated, the aqueous phase was extracted with DCM (15 mL. times.1), the organic phases were combined, washed with saturated NaCl solution (15 mL. times.1), the organic phase was collected, and anhydrous Na was added2SO4And (5) drying. And (3) carrying out spin drying on a rotary evaporator to obtain a crude product, carrying out column chromatography to obtain a pure product, and carrying out vacuum drying to obtain the intermediate compound X.
TABLE 1 intermediate type X Synthesis results
Figure BDA0001463694600000102
Figure BDA0001463694600000103
Figure BDA0001463694600000111
Examples 1 to 35
A100 mL round-bottom flask was charged with 1mmol of intermediate of formula X, 5mL of toluene, 2mmol of Et3And N, stirring for 20-30min, adding 2mmol of isonicotinic acid (INA) or pyrazinecarboxylic acid (POA), moving to an oil bath kettle at 110-120 ℃ for reflux reaction after 10min, and monitoring by TLC until the reaction is complete. Spin-drying the reaction solution with rotary evaporator, adding 30mL DCM, stirring for dissolving, vacuum filtering, washing the filter cake with DCM for 3 times, washing the filtrate with 10% citric acid solution (15mL × 1), collecting the organic phase, and collecting anhydrous Na2SO4And (5) drying. And (4) carrying out spin drying on a rotary evaporator to obtain a crude product, carrying out column chromatography to obtain a pure product, and carrying out vacuum drying to obtain a target product.
TABLE 2 Synthesis results of the object Compounds of formula XI
Figure BDA0001463694600000112
Figure BDA0001463694600000113
Figure BDA0001463694600000121
TABLE 3 Synthesis results of the object Compounds of formula XI
Figure BDA0001463694600000122
Figure BDA0001463694600000123
Figure BDA0001463694600000131
Synthesis of a series of compounds of formula XII:
Figure BDA0001463694600000132
examples 36 to 41
A100 mL round-bottom flask was sequentially added with 2mmol isoniazid and 4mmol NaHCO3And 5mL of DCM, stirring for 20min at-5 ℃, then dropwise adding 3mmol of chloroacetyl chloride solution by using a constant pressure dropping funnel (the dropping speed is about 1d/2s), continuously reacting at-5 ℃, and tracking and monitoring by TLC until the reaction is finished. With stirring, DCM-CH was added3OH solution (V)DCM:VCH3OH2: 1) standing until the solid in the reaction flask does not decrease, vacuum filtering, and adding DCM-CH to the filter cake3OH solution was washed 3 times, and the filtrate was washed with anhydrous Na2SO4Drying, spin-drying with a rotary evaporator to obtain a crude product, adding 5mL of DCM, stirring for 20min, carrying out suction filtration, washing a filter cake with DCM for 3 times, and carrying out vacuum drying to obtain an intermediate for later use.
1mmol of Saxaxin and 4mmol of Et are sequentially added into a 100mL round-bottom flask3N and 2mL DMF, stirring for 20min, adding the intermediate, 10min later, moving to 60 ℃ and stirringThe reaction was monitored by TLC to the end of the reaction. Adding 10mL of ice-cold saturated aqueous salt solution while stirring, adjusting pH to about 8 with 1N HCl solution, suction filtering, extracting the filtrate with DCM (10 mL. times.2), combining the organic phases, and adding anhydrous Na2SO4Drying, spin-drying by a rotary evaporator, and purifying by column chromatography together with the filter cake to obtain the target compound formula XII.
TABLE 4 Synthesis results of the object Compound formula XII
Figure BDA0001463694600000133
Figure BDA0001463694600000134
Figure BDA0001463694600000141
Synthesis of a series of compounds of formula XV:
Figure BDA0001463694600000142
preparation examples 25 to 40
Amino acid (50mmol) and 100mL of methanol are added into a reaction bottle, and SOCl is slowly dropped under ice bath2(125mmol) and after dropping, the reaction solution is transferred into a water bath at 60 ℃ and stirred for reflux reaction, and TLC monitoring is carried out until the reaction is finished. Reduced pressure rotary evaporation to remove methanol and most SOCl220mL of methanol was added and the mixture was re-rotary evaporated to remove SOCl as much as possible2Vacuum drying to obtain corresponding amino acid methyl ester hydrochloride.
Adding INA/POA (20mmol), HOBt (24mmol), DCC (24mmol) and DCM (20mL) in sequence into a reaction flask, stirring for 10min, and adding Et3N (60mmol, when the amino acid is alanine, DIPEA is added under ice bath), glycine/alanine methyl ester hydrochloride (22mmol) is added after 0.5-1 h, and TLC is used for monitoring until the reaction is finished. Freezing in refrigerator for 2 hr, filtering, washing filter cake with DCM, and filtering to obtain filtrate with saturated Na2CO3Aqueous (20 mL. times.2), aqueous phase extracted with DCM (30 mL. times.3), combined organic phases, anhydrous Na2SO4Drying for 1h, spin-drying on a rotary evaporator, and performing column chromatography to obtain the compound of formula XIII.
The intermediate (15mmol) prepared in preparation example 25-32 and CH were added to the reaction flask in sequence3OH-H2O(V CH3OH:V H2O(3: 1) solution (15mL), LiOH · H was added under ice-bath2O (45mmol), stirred and monitored by TLC to the end of the reaction. Rotating evaporator to remove CH3OH was removed, pH was adjusted to about 3 with 4N HCl solution in ice bath, a large amount of white solid precipitated, filtered, the filter cake was washed 2 times with filtrate, 2 times with acetone, and dried at 50 ℃ to give compound of formula XIV.
TABLE 5 intermediate of formula XIII XIV Synthesis results
Figure BDA0001463694600000143
Figure BDA0001463694600000151
Examples 42 to 105
HBTU/TBTU method: a reaction flask was charged with the compound of formula XIV (1mmol), HBTU/TBTU (1.2mmol), DCM (3mL), Et3N (3mmol, Ala as Linker) and DIPEA and fleroxacin (1mmol) are added under ice bath, the temperature is controlled and stirring is carried out, and TLC is used for monitoring until the reaction is finished. Add DCM 20mL, move to separatory funnel, saturate Na2CO3Washing with an aqueous solution (20 mL. times.1), washing with a 0.5N HCl solution (20 mL. times.1), washing with a saturated saline solution (20 mL. times.1), and anhydrous Na2SO4Drying for 1h, spin-drying by a rotary evaporator, and carrying out column chromatography to obtain the XV series compound.
EDCI-HOBt method: a reaction flask was charged with the compound of formula XIV (1mmol), HOBt (1.2mmol), EDCI (1.2mmol), DCM (3mL), Et3N (3mmol, Ala is added in the Linker in ice bath), stirring is carried out for 0.5-1 h, then Saixing (1mmol) is added, and TLC is used for monitoring until the reaction is finished. Add DCM 20mL, move to separatory funnel and saturate Na2CO3Dissolving in waterWashing with HCl (20 mL. times.1), washing with 0.5N HCl solution (20 mL. times.1), washing with saturated brine (20 mL. times.1), anhydrous Na2SO4Drying for 1h, spin-drying by a rotary evaporator, and carrying out column chromatography to obtain the XV series compound.
TABLE 6 Synthesis results of XV series of compounds of formula
Figure BDA0001463694600000161
Figure BDA0001463694600000162
Figure BDA0001463694600000171
TABLE 7 Synthesis results of XV series of compounds of formula
Figure BDA0001463694600000172
Figure BDA0001463694600000173
Figure BDA0001463694600000181
In vitro Activity Effect examples
1. Inhibitory Activity test for Mycobacterium tuberculosis
Mycobacterium tuberculosis (m.tuberculosis) is the causative pathogen of tuberculosis. Can invade all organs of the body, but pulmonary tuberculosis is the most common (because of aerobic nature). Tuberculosis remains an important infectious disease to date. 1/3 are estimated to be infected by Mycobacterium tuberculosis in the world population. It is reported by WHO that about 800 new cases occur each year, and at least 300 million people die from the disease.
Mycobacterium tuberculosis (H3) for target compound7Rv standard strain) was tested: a tuberculosis standard strain H37Rv is selected, a single-concentration whole cell activity test (Primary SP) is carried out, the inhibition rate of the series of compounds of the invention under the sample concentration of 20 mu M is determined, a test sample with higher activity is diluted (multi-concentration, CRC) and then is subjected to Secondary screening (Secondary assays), the lowest drug concentration (MIC) for inhibiting the growth of H37Rv and the cytotoxicity for HELA cells are determined, and the half Inhibition Concentration (IC) is used50) And (4) showing.
By passing
Figure BDA0001463694600000182
(homogeneous immunoassay kit based on chemiluminescence of beads, PerkinElmer) the Mycobacterium tuberculosis inhibitory activity of the target compounds was determined. And adopt
Figure BDA0001463694600000183
Luminescent Cell Viability Assay(
Figure BDA0001463694600000184
Luminescence method cell viability detection kit) test the cytotoxicity (half inhibitory concentration IC) of test samples on HELA cells50)。
The screening result of the anti-tuberculosis activity shows that the inhibition rate of the series of compounds of the invention is more than 80 percent under the sample concentration of 20 mu M. The test of multi-concentration inhibitory activity was carried out on compounds with partial inhibitory activity exceeding 90%, and the results showed that the MIC values of the high-activity compounds other than the compound of example 90 were less than 10 μ M, particularly, the MIC values of examples 33, 34, 17, and 4 were as low as 0.16, 0.23, 0.24, and 0.35 μ M. Cytotoxicity test (HELA Cytotox activity) shows that the relative inhibition rate Rel IC of the high-activity compounds on HELA cells50>20.0 μ M, with lower cytotoxicity. See table 8 for details.
Bacteriostatic Activity of the Compounds of Table 8 against Mycobacterium tuberculosis
Figure BDA0001463694600000191
Figure BDA0001463694600000201
2. Conventional bacterial and fungal bacteriostatic activity test
The bacterial infection is acute systemic infection caused by invasion of pathogenic bacteria or conditional pathogenic bacteria into blood circulation, growth and reproduction, and production of toxins and other metabolites, is clinically characterized by chills, hyperpyrexia, rash, arthralgia and hepatosplenomegaly, and can be partially infected with septic shock and migratory focus. Most patients with tuberculosis are malnutrition, and are affected by immunosuppressive treatment for a long time, the patients have weak constitution, the body immunity is reduced, and various conventional bacterial infectious diseases are easy to be complicated. In order to verify whether the compounds of the invention have multi-target activity, the invention performed bacteriostatic activity experiments.
Minimum Inhibitory Concentration (MIC) determination
Test strains: staphylococcus aureus (s.aureus) ATCC 29213; coli (e.coli) clinical isolates.
Sterilized 96-well plates were opened under sterile conditions, and 200. mu.L of M-H broth containing the drug was added to well 1 to a final concentration of 32. mu.g/mL. The remaining 11 wells were filled with 100. mu.L of blank medium. Accurately sucking 100 mu L from the 1 st hole by using a pipette gun, adding the pipette gun into the second hole, blowing and uniformly mixing, sucking 100 mu L from the 2 nd hole to the 3 rd hole, and repeating the steps until reaching the 10 th hole, sucking 100 mu L after uniform mixing, and discarding. The drug concentration per well at this time was 64, 32, 16, 8, 4, 2, 1, 0.5. mu.g/mL. The last 2 wells contained no drug, one as bacterial growth control and one as negative control.
In the above 96-well plate, 10 was added to each of the first 11 wells8CFU/mL of bacterial suspension 0.5. mu.L, the final inoculation amount is about 105CFU/mL. The bacterium adding speed is as fast as possible, so that the phenomenon that the result judgment is influenced due to the fact that the bacterium growth time of each hole is inconsistent because of too long time is avoided. Each strain was tested in 3 replicates simultaneously. The whole process is finished beside the sterilized alcohol lamp of the superclean bench. Carefully handling to prevent contamination. And (3) putting the inoculated 96-well plate into a constant-temperature incubator at 37 ℃ for culturing for 16-20h, and observing and recording the result.
After the completion of the culture, the 96-well plate was taken out from the incubator, and the growth of bacteria in the well was observed. Before the results are determined, the results are meaningful only when the bacteria in the growth control wells grow normally and the negative control wells do not grow. The concentration of the drug in the wells with no bacterial growth was visually observed as the MIC of the drug against the bacteria. If the hole jumping phenomenon occurs, repeated tests are needed for verification.
The results of the antimicrobial activity tests of the partial compounds of the series of the present invention are shown in Table 9. Overall, the compounds had better inhibitory activity on s.aureus than e.coli; for the s.aureus strain, the MIC of 34 compounds is less than 0.5 μ g/mL, and the MIC of 34 compounds is 1-8 μ g/mL; coli strain, MIC of 6 compounds <0.5 μ g/mL, and MIC of 16 compounds 1-8 μ g/mL.
Table 9 biological activity results of compounds on s
Figure BDA0001463694600000211
Figure BDA0001463694600000221
3. Determination of inhibitory Activity of Escherichia coli Retention bacteria
The persistent bacteria (Bacterial persaster) is a small subgroup with a certain proportion of phenotypic dissimilarity in a certain Bacterial population, is expressed in a slow growth state or a temporary dormancy state, and can tolerate the action of lethal concentration antibiotics. At present, no antibacterial drug and therapeutic means capable of completely eliminating the retained bacteria are found, and the appearance of the antibacterial drug and the therapeutic means brings great challenges to modern medical research. Studies have shown that persistants are associated with a number of microbial challenges, including bacterial infections, chronic infections, and multidrug resistance of bacterial biofilms, and thus effective clearance of persistants is a key tool in treating these microbial challenges. The invention performs the research on the activity of anti-retention bacteria by taking escherichia coli as a research object on part of target molecules so as to obtain the chemical micromolecules with the capability of killing the retention bacteria. The escherichia coli strains adopted by the invention are as follows: escherichia coli ATCC 25922 strain is a strain reserved by the modern biomedical research institute of the institute of Life sciences of southwest university.
Primary screening of compounds: the compounds were weighed out as needed and dissolved in DMSO. Inoculating the strain-preserved Escherichia coli into sterilized LB medium at a ratio of 1:100, and culturing in shaker at 37 deg.C to logarithmic phase (OD)6000.2), 100 μ L per well was added dropwise to a 96-well plate, ampicillin Amp was added per well at a final concentration of 100 μ g/mL at which a model of retained bacteria was formed according to the experimental design, and then a compound was added per well at a final concentration of 30 μ M, and a DMSO-only added bacterial solution was set as a negative control. After the 96-well plate is placed in an incubator at 37 ℃ for culturing for 9h, 10 mu L of bacterial liquid is taken out of each well, the solid LB plate is cultured overnight, and the growth condition of the bacterial colony is observed.
TABLE 10 Experimental results of the compounds for killing Escherichia coli persistant bacteria
Figure BDA0001463694600000222
Effect of compounds on bacteria in normal growth state: selecting 9 compounds which show 75-100% in the primary screening result to test the killing effect of the compounds on bacteria in a normal growth state: inoculating the strain-preserved Escherichia coli into sterilized LB medium at a ratio of 1:100, and culturing in shaker at 37 deg.C to logarithmic phase (OD)6000.2), 100 μ L per well was added dropwise to a 96-well plate, the above 9 compounds were added to a final concentration of 30 μ M, respectively, and a DMSO-only added bacterial suspension was set as a negative control. After the 96-well plate is placed in an incubator at 37 ℃ for culturing for 9h, 10 mu L of bacterial liquid is taken out of each well, the solid LB plate is cultured overnight, and the growth condition of the bacterial colony is observed. And the bacterial liquid is diluted in a gradient (the dilution gradient is 10)-1) And (5) dropping the plate, and further observing the growth condition of the colonies.
If the bacterial liquid is diluted 102After bacterial culture, the existence of bacteria can not be found, which shows that the bacterial liquid has strong sterilization capability corresponding to the added compound; on the contrary, if the bacterial liquid is diluted 106And (3) after culture, no obvious difference is observed between the culture solution and the control (DMSO) bacterial solutionIndicating that the corresponding compound has almost no bactericidal activity.
Experimental results show that the compounds of preparation examples 6, 22 and 24 have the capability of killing retained strains and stronger capability of killing normal Escherichia coli among the 9 compounds with better inhibitory activity to Escherichia coli retained bacteria; the compounds of example 4, preparation 4 and preparation 12 had the ability to kill the resident strain, but had the ability to kill normal E.coli comparable to the negative control, and had almost no bactericidal activity.
Results of experiments on the Effect of Compounds of Table 11 on bacteria in Normal growth conditions
Figure BDA0001463694600000231
4. Determination of bacteriostatic activity of citrus pathogenic bacteria
Citrus canker (citrus canker) is a major epidemic disease affecting the production of citrus in the world, and can damage dozens of rutaceae plants. The disease seriously damages citrus seedlings and young trees, causes fallen leaves and declines tree vigor; can also cause harm to adult fruit trees, can cause a large amount of fruit drop in serious conditions, and greatly reduces the economic value of citrus fruits due to scab.
The pathogen of the citrus brown spot is alternaria alternate (Alternaria alternate), which belongs to the genus Alternaria of the family Hymenochaetaceae of the order Hyphomycetales of the class Hyphomycetes of the subdivision Deuteromycotina, leaves of the plant are easy to fall off after being infected by diseases, and fallen leaves on the ground can be used as an initial infection source to cause the spread of the diseases. Conidia are mainly on the surface of a diseased mature leaf, spread by wind, and then stay on a sensitive young fruit or leaf. When the humidity conditions are met, the conidia germinate quickly and begin to produce toxins, which infect the leaves directly or through stomata.
And (3) determining the bacteriostatic activity of citrus canker germs: a1 mg sample of the compound was weighed out and dissolved in 50. mu.L DMSO, and the volume was adjusted to 550. mu.L with ultrapure water as a sample stock solution (1.82 mg/mL). mu.L of the mother liquor was taken as a sample solution a (0.0182mg/mL) in 1mL of ultrapure water (0.02% Tween), and then a sample solution b (0.0091mg/mL) was prepared in order by the double dilution method.
Washing the ulcer germs cultured for 3 days on the PDA culture medium with 5mL of LB liquid culture medium, adding into 195mL of LB liquid culture medium, and shaking and mixing for later use. Adding 450 mu L of the bacterial liquid of the citrus canker pathogen and 50 mu L of the sample solutions with different concentrations (a and B) into each 2mL centrifuge tube respectively to ensure that the final concentrations of the samples in each mixed bacterial liquid are A (0.00182mg/mL) and B (0.00091mg/mL), the temperature is 28 ℃, and the temperature is 200 r.min-1Determination of OD after constant temperature shaking culture for 14h600The OD value of each mixed bacterial solution was calculated and the inhibition ratio was calculated (inhibition ratio%Blank space-ODSample (I))/ODBlank spaceX 100%). Each treatment was replicated three times.
Experimental results show that the compound has certain bacteriostatic activity on citrus canker pathogenic bacteria, particularly the inhibition rate of the compounds of preparation examples 6, 22, 23 and 24 under low concentration (0.00091mg/mL) is still over 80%, and the compound shows good development prospect.
TABLE 12 results of anti-Cicer Activity of target Compounds
Figure BDA0001463694600000241
And (3) determining the bacteriostatic activity of the citrus brown spot germs: washing conidia of the brown spot germs cultured for 7 days on the PDA culture medium by using 0.05 percent Tween 80, and filtering four layers of sterile lens wiping paper for later use; a1 mg sample of the compound was weighed out and dissolved in 50. mu.L DMSO, and the volume was adjusted to 550. mu.L with ultrapure water as a sample stock solution (1.82 mg/mL). mu.L of the mother solution was taken as a sample solution a (0.00364mg/mL) in 1mL of PDA (0.02% Tween), and a sample solution b (0.00182mg/mL) was prepared in order by the double dilution method. After adding 0.5mL of the sample solution and 1.5mL of LPDA medium to each well of the 48-well plate to give final concentrations of A (0.000910mg/mL) and B (0.000455mg/mL), respectively, 2. mu.L of the conidia suspension was inoculated and cultured with light at 28 ℃ for 3 days to measure the colony diameter. Using the colony inoculated on the PDA culture medium without sample solution as a blank control, calculating the inhibition ratio (the inhibition ratio is%Blank spaceDiameter of coloniesSample (I)) Colony diameterBlank space×100%)。
The experimental result shows that the relative inhibition rate of 24 compounds is over 20 percent when the tested concentration is 0.000910 mg/mL; the tested concentration is reduced by one time, and the relative inhibition rate of 5 compounds exceeds 20 percent, thereby showing certain development potential.
TABLE 13 results of activity of target compounds against Curvularia citrifolia
Figure BDA0001463694600000251
5. Determination of tumor target point nicotinamide N-methyltransferase (hNNMT) inhibitory activity
Nicotinamide N-methyltransferase (NNMT) is a protease which is screened out in tumor tissues and abnormally expressed when different expression molecules between different cancer tissues and paracancer (or normal) tissues are compared by utilizing proteomics and gene chip technology in recent years. The NNMT is differentially expressed in various tumor tissues, and researches find that the NNMT is closely related to various characteristic biological functions of tumor cell proliferation, metastasis, chemoradiotherapy resistance and the like, and the NNMT can be used as a potential marker for tumor auxiliary diagnosis, individuation and targeted therapy.
Assay of inhibitory activity of hNNMT target: the inhibitory activity of the target compound on hNNMT target is shown by a 10 mu M single-concentration (SP) inhibitory activity test and a multiple-concentration (CRC) inhibitory activity test for detecting products MNAN (Methyl n-amyl nitrosamine) and SAH (S-adenosyl homocysteine) in hNNMT expression pathway.
By passing
Figure BDA0001463694600000261
Determining the inhibitory activity of the target compound by liquid chromatography/mass spectrometry (LC/MS) and
Figure BDA0001463694600000262
Figure BDA0001463694600000263
luminescent Cell Viability Assay test multiple concentrations of test sample versus HEK293 cellsHalf inhibitory concentration of cells IC50
The activity data show that 9 compounds with more than 10% inhibitory activity of hNNMT MNAN SP at the tested concentration of 10 μ M had the best inhibitory activity of the compound of example 66, with 74.2% and 71.2% inhibition of hNNMT MNAN SP and hNNMT SAH SP, respectively, and IC's of the compounds50The values were 90.0. mu.M and 118.9. mu.M, respectively.
TABLE 14 inhibitory Activity results (10. mu.M) for hNNMT target
Figure BDA0001463694600000264
TABLE 15 relative inhibition of hNNMT target (Rel IC) by the compound of example 6650)
Figure BDA0001463694600000265
6. Interleukin IL-17PPI inhibitory Activity assay
IL-17 is an inflammatory cytokine produced primarily by activated T cells, and can promote the activation of T cells and stimulate the production of various cytokines such as IL-6, IL-8, granulocyte-macrophage stimulating factor (GM-CSF), and chemo-activin and cell adhesion molecule 1 (CAM-1) by fibroblasts, endothelial cells, and fibroblasts, resulting in the production of inflammation. IL-17 is an early initiator of T cell-induced inflammatory responses and may amplify inflammatory responses by promoting the release of pro-inflammatory cytokines. After IL-17 binds to a receptor, it exerts its biological effects through the MAP kinase pathway and the Nuclear transcription factor kappa B (NF-. kappa.B) pathway. IL-17 produced by Th17 cells is effective in mediating the excitatory process of neutrophil mobilization and thus the inflammatory response of tissues.
Method for determining the IL-17 inhibitory activity of a target Compound: cultured cells stimulated with recombinant IL-17A were tested for inhibitory activity against 100. mu.M of the target compound in a single concentration (SP) and multiple concentrations (CRC), respectively
Figure BDA0001463694600000266
The IL-17 inhibitory activity of the target compound is measured.
The activity results show that: at a test concentration of 100. mu.M, 7 compounds with an inhibition rate of more than 20% were tested, of which the compound of example 66 had the best inhibitory activity, 52.4%.
TABLE 16 IL17PPI inhibitory Activity results (100. mu.M) for the target Compounds
Figure BDA0001463694600000271
Finally, the above embodiments are only for illustrating the technical solutions of the present invention and not for limiting, although the present invention has been described in detail with reference to the preferred embodiments, it should be understood by those skilled in the art that modifications or equivalent substitutions may be made to the technical solutions of the present invention without departing from the spirit and scope of the technical solutions of the present invention, and all of them should be covered in the claims of the present invention.

Claims (8)

1. A compound of formula II or a pharmaceutically acceptable salt thereof:
Figure FDA0002715725620000011
wherein X is selected from: C1-C6 alkyl; C3-C6 cycloalkyl; a substituted or unsubstituted C6-C10 aryl group, the substituents on the aryl group being one or more independently selected from: halogen; an amino group; a hydroxyl group; C1-C6 alkyl; C3-C6 cycloalkyl;
z is selected from: n or C-R7;R7Is selected from H; C1-C6 alkoxy or halogen;
R1and R2Each independently selected from: h; C1-C6 alkyl; C1-C6 haloalkyl; C1-C6 alkoxy; halogen; a hydroxyl group; alkylamino of C1-C6; amino or cyano;
R4selected from: h, halogen; an amino group; a hydroxyl group; C1-C6 alkyl;
a is selected from the following structures:
Figure FDA0002715725620000012
k is 0 or 1, and n is an integer of 0 to 3.
2. A compound of formula IV or formula VI:
Figure FDA0002715725620000013
x is selected from: a methyl group; an ethyl group; propyl; a butyl group; a cyclopropyl group; halogen substituted or unsubstituted phenyl;
z is selected from: n or C-R7;R7Is selected from H; a methoxy group; an ethoxy group; fluorine or chlorine;
wherein R is1And R2Each independently selected from: h; a methyl group; an ethyl group; a trifluoromethyl group; a methoxy group; an ethoxy group; halogen; a hydroxyl group or an amino group;
R4selected from: h, halogen; an amino group; a hydroxyl group; methyl or ethyl;
a is selected from
Figure FDA0002715725620000021
K is 0 or 1; n is 1 or 2 or 3.
3. A compound represented by:
Figure FDA0002715725620000022
Figure FDA0002715725620000031
Figure FDA0002715725620000041
4. a pharmaceutical composition comprising a compound according to any one of claims 1 to 3 or a pharmaceutically acceptable salt thereof.
5. The pharmaceutical composition of claim 4, wherein the pharmaceutical composition further comprises: a) pharmaceutically acceptable carriers and/or adjuvants; and/or b) one or more suitable further active ingredients.
6. Use of a compound according to any one of claims 1 to 3 or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of a disease selected from the group consisting of a disease caused by infection with a bacterium and its retentate, a disease caused by infection with a citrus-pathogenic bacterium, an inflammatory disease or a tumour.
7. The use of claim 6, wherein the bacteria comprise Mycobacterium tuberculosis, Staphylococcus aureus, Escherichia coli, Salmonella aeruginosa; the citrus pathogenic bacteria are selected from the group consisting of: citrus canker pathogens; citrus brown rot or citrus green mold; the inflammatory disease is an IL-17 related inflammatory disease, and the tumor is selected from the group consisting of: lung cancer, kidney cancer, colorectal cancer, nasopharyngeal cancer, breast cancer, pancreatic cancer, liver cancer, gastric cancer, brain glioma, bladder cancer, and oral squamous cell carcinoma or medulloblastoma.
8. Use according to claim 7, wherein the IL-17 related inflammatory disease is selected from Rheumatoid Arthritis (RA), asthma, Multiple Sclerosis (MS), Chronic Obstructive Pulmonary Disease (COPD), Acute Respiratory Distress Syndrome (ARDS), Idiopathic Pulmonary Fibrosis (IPF), Inflammatory Bowel Disease (IBD), Crohn's disease, uveitis, macular degeneration, colitis, psoriasis, Waller's degeneration, antiphospholipid antibody syndrome (APS), acute coronary syndrome, restenosis, atherosclerosis, Relapsing Polychondritis (RP), acute or chronic hepatitis, failed orthopedic implants, glomerulonephritis, lupus and autoimmune diseases.
CN201711102862.5A 2017-11-10 2017-11-10 Fluoroquinolone amino derivative and application thereof Active CN107880023B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201711102862.5A CN107880023B (en) 2017-11-10 2017-11-10 Fluoroquinolone amino derivative and application thereof
CN201911252190.5A CN111087390B (en) 2017-11-10 2017-11-10 Fluoroquinolone amino derivative and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201711102862.5A CN107880023B (en) 2017-11-10 2017-11-10 Fluoroquinolone amino derivative and application thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN201911252190.5A Division CN111087390B (en) 2017-11-10 2017-11-10 Fluoroquinolone amino derivative and application thereof

Publications (2)

Publication Number Publication Date
CN107880023A CN107880023A (en) 2018-04-06
CN107880023B true CN107880023B (en) 2021-02-19

Family

ID=61779800

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201911252190.5A Active CN111087390B (en) 2017-11-10 2017-11-10 Fluoroquinolone amino derivative and application thereof
CN201711102862.5A Active CN107880023B (en) 2017-11-10 2017-11-10 Fluoroquinolone amino derivative and application thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN201911252190.5A Active CN111087390B (en) 2017-11-10 2017-11-10 Fluoroquinolone amino derivative and application thereof

Country Status (1)

Country Link
CN (2) CN111087390B (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11202101335WA (en) * 2018-08-13 2021-03-30 Otsuka Pharma Co Ltd Novel medicament for treating inflammatory bowel disease
CN108864043A (en) * 2018-09-18 2018-11-23 中国医学科学院生物医学工程研究所 A kind of preparation method and purposes of novel quinolone compounds
CN109096278B (en) * 2018-09-26 2021-05-28 西南大学 Fluoroquinolone-azole hybrid derivative, preparation method and application thereof
CN112159390B (en) * 2020-09-25 2022-07-19 西南大学 Synephrine fluoroquinolone derivative and preparation method and application thereof
CN112159355B (en) * 2020-09-25 2022-06-24 西南大学 Fluoroquinolone p-aminosalicylate derivative and intermediate, preparation method and application thereof
CN112110898B (en) * 2020-09-25 2022-07-05 西南大学 Synephrine sulfonylation derivative and intermediate, preparation method and application thereof
CN113480519B (en) * 2021-07-23 2022-05-31 西南大学 Oxacycloxacin derivative and preparation method and application thereof
CN113907079A (en) * 2021-11-10 2022-01-11 西南大学 Application of chloralkanoyl fluoroquinolone in preparing medicine for preventing and treating citrus canker

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN100341855C (en) * 2005-02-22 2007-10-10 南京澳新医药科技有限公司 7-substituted-8-methoxy fluoroquinolone carboxylic derivatives, preparing process, preparation and use thereof
US7632944B2 (en) * 2007-01-24 2009-12-15 Bausch & Lomb Incorporated Quinolone carboxylic acids, derivatives thereof, and methods of making and using same
GB0705915D0 (en) * 2007-03-28 2007-05-09 Helperby Therapeutics Ltd New use
CN102827187B (en) * 2012-07-18 2014-05-14 河南大学 Fluoroquinolone acetal isoniazone, and preparation method and application thereof
CN102993170A (en) * 2012-11-23 2013-03-27 西南大学 Clinafloxacin derivative as well as preparation method and application thereof
CN103396396B (en) * 2013-08-12 2015-09-09 西南大学 Amido alkane acyl Clinafloxacin and application thereof
CN103405435B (en) * 2013-08-30 2014-12-10 西南大学 Application of clinafloxacin amino derivatives and medicinal salts thereof in preparing antitubercular medicaments
JP2018528260A (en) * 2015-07-28 2018-09-27 ビオーム バイオサイエンシズ プライベート リミティド Antibacterial therapeutics and preventives
CN107459507B (en) * 2016-06-03 2021-02-19 西南大学 Clinafloxacin oxazolidinyl derivative and preparation method and application thereof
CN107721924B (en) * 2017-11-10 2021-10-22 西南大学 Gatifloxacin derivative and preparation method and application thereof
CN107827815B (en) * 2017-11-10 2021-08-03 西南大学 Fluoroquinolone amino derivatives and application thereof in preventing and treating citrus diseases

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
fluorescent-labeled amino acid-antibiotic conjugates;ANAND D等;《synthesis》;20140624;第46卷;第2430-2435页 *

Also Published As

Publication number Publication date
CN111087390B (en) 2022-07-05
CN107880023A (en) 2018-04-06
CN111087390A (en) 2020-05-01

Similar Documents

Publication Publication Date Title
CN107880023B (en) Fluoroquinolone amino derivative and application thereof
CN110724174B (en) Pyrrolotriazine compound, composition and application thereof
CN109096278B (en) Fluoroquinolone-azole hybrid derivative, preparation method and application thereof
CN107827815B (en) Fluoroquinolone amino derivatives and application thereof in preventing and treating citrus diseases
CN107721924B (en) Gatifloxacin derivative and preparation method and application thereof
EP2678319A2 (en) Antibiotic tolerance inhibitors
RU2527769C2 (en) 5-hydroxymethyloxazolin-2-one derivatives for treating bacterial intestinal diseases
CN110818648A (en) Pleuromutilin derivative with triazole side chain as well as preparation method and application thereof
CN112159354A (en) Fluoroquinolone derivative of p-aminosalicylic acid and intermediate, preparation method and application thereof
US11731946B2 (en) Broad spectrum antivirulence, anti-persistence compounds
JP7371838B2 (en) Ring-fused thiazolino 2-pyridones, methods of their preparation and their use in the treatment and/or prevention of diseases involving Gram-positive bacteria
JP6793927B2 (en) Ciprofloxacin derivative antibacterial agent
JP6930060B2 (en) Novel dihydropyranopyrimidinone derivatives and their uses {Novell dihydropyranopylidinone derivatives, and use thetheof}
CN112159355B (en) Fluoroquinolone p-aminosalicylate derivative and intermediate, preparation method and application thereof
US20120225889A1 (en) Use of macrocyclic lactone derivatives for the treatment of inflammatory disorders
CN110981888B (en) N-aryl dithiopyrryl ketonuria and amino ester derivatives, preparation and application thereof
CN111527093B (en) Antibacterial heterocyclic compounds and synthesis thereof
CZ20013712A3 (en) Use of maduraphthalazine derivatives for treating diseases mediated by the interleukin-2 cytokines
EA007540B1 (en) Beta-lactamase inhibitor prodrug
RU2634122C1 (en) Fluoroquinolones based on 4-deoxypyridoxine
JP5756096B2 (en) Guanine riboswitch binding compounds and their use as antibiotics
CA2473724A1 (en) Purine and isosteric antibacterial compounds
US11773092B1 (en) Pyrido[3,4-b]indol-1-one compounds as antibacterial agents
KR101893005B1 (en) Novel luffariellolide pyridazinone derivatives and antibacterial composition comprising thereof
JP2016523230A (en) New pyrrole derivatives

Legal Events

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