WO2016062151A1 - 吡唑并[1,5-a]吡啶类化合物及其应用 - Google Patents

吡唑并[1,5-a]吡啶类化合物及其应用 Download PDF

Info

Publication number
WO2016062151A1
WO2016062151A1 PCT/CN2015/086852 CN2015086852W WO2016062151A1 WO 2016062151 A1 WO2016062151 A1 WO 2016062151A1 CN 2015086852 W CN2015086852 W CN 2015086852W WO 2016062151 A1 WO2016062151 A1 WO 2016062151A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyridine
phenyl
substituted
pyrazole
trifluoromethoxy
Prior art date
Application number
PCT/CN2015/086852
Other languages
English (en)
French (fr)
Inventor
陆小云
汤健
丁克
张天宇
涂正超
伍甜
万军庭
曹元元
Original Assignee
中国科学院广州生物医药与健康研究院
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 中国科学院广州生物医药与健康研究院 filed Critical 中国科学院广州生物医药与健康研究院
Priority to US15/520,765 priority Critical patent/US10155756B2/en
Publication of WO2016062151A1 publication Critical patent/WO2016062151A1/zh

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis

Definitions

  • the invention belongs to the field of chemical medicine, in particular to pyrazolo[1,5-a]pyridine compounds and applications thereof.
  • Tuberculosis is caused by infection with Mycobacterium tuberculosis (Mtb) and is one of the most prevalent diseases in the world.
  • Mtb Mycobacterium tuberculosis
  • tuberculosis is the most common, accounting for 80-90% of the total number of tuberculosis in various organs. Due to the emergence of drug-resistant tuberculosis and the parallel infection of AIDS, the therapeutic effect of existing clinical drugs is greatly reduced.
  • the current treatment regimen is the combination of first-line drugs isoniazid, rifampicin, pyrazinamide and ethambutol with second-line drugs for 6-9 months, and for drug-resistant tuberculosis and retention bacteria
  • the resistance mechanism of the first-line anti-tuberculosis drugs currently used has been relatively clear. The main reason is that due to the improper use of drugs, the corresponding target gene or drug activation-related enzyme gene or related regulatory gene is mutated, resulting in drug failure. Therefore, under the launch of the Global Alliance for TB Drug Development, people began to re-focus on the development of anti-tuberculosis drugs.
  • New drugs should have the following characteristics: 1 development of new drugs that can shorten the treatment cycle; 2 development of drugs that are effective against drug-resistant tuberculosis; 3 development of new drugs that can effectively treat latent bacterial infections.
  • anti-tuberculosis drugs there are three main aspects in the research and development of anti-tuberculosis drugs: natural material screening, design and synthesis of new structural type compounds, and re-modification of antibacterial drugs.
  • some newly synthesized anti-tuberculosis drugs at home and abroad include quinolines, nitroimidazopyrans, oxazolidinones, azoles, imidazopyridines and the like.
  • the bis-aryl quinoline drug bedaquiline was approved by the FDA to be marketed as one of the components of the adult multidrug-resistant tuberculosis combination treatment program.
  • This is the first new anti-tuberculosis drug since the introduction of rifampicin in 1970.
  • subsequent clinical trials have shown that bedaquiline also exhibits certain toxic side effects, and safety risks remain. Therefore, it is of great significance to further develop safe and effective anti-tuberculosis drugs to overcome the harm of clinical drug-resistant tuberculosis.
  • the present invention adopts the following technical solutions:
  • a pyrazolo[1,5-a]pyridine compound having the structural feature of formula (I) or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a prodrug molecule thereof:
  • R 1 is selected from the group consisting of: 1) H; 2) halogen; 3) O a C 1 - C 5 alkyl; 4) C 3 - C 6 cycloalkyl; 5) aryl; 6) amino, hydroxy, cyano a nitro group; 7) a heteroaryl group; the alkyl group, the aryl group, the cycloalkyl group and the heteroaryl group are each optionally substituted by 0, 1, 2 or 3 substituents selected from R 6 ;
  • R 2 is optionally selected from the group consisting of: 1) H; 2) O a C 1 - C 5 alkyl; 3) C 3 - C 6 cycloalkyl; 4) aryl; 5) heteroaryl;
  • alkyl, aryl, cycloalkyl and heteroaryl groups are each optionally substituted by 0, 1, 2 or 3 substituents selected from R 6 ;
  • the alkyl group, the aryl group, the cycloalkyl group, the heterocycloalkyl group and the heteroaryl group are each optionally substituted by 0, 1, 2 or 3 substituents selected from R 6 , wherein the a is 0 or 1; R 6 is optionally selected from the group consisting of: 1) H; 2) C 3 - C 6 cycloalkyl; 3) heterocyclic; 4) C 1 - C 3 alkyl; 5) C 1 - C 3 fluorine-containing alkyl ; 6) C 0 - C 3 alkylene-heterocyclic group; 7) halogen.
  • the R 1 is selected from the group consisting of: 1) H; 2) F, Cl, Br, I; 3) OH, OCH 3 , OEt, OCF 3 ; 4) methyl, ethyl, iso Propyl, tert-butyl; 5) cyclopropyl; 6) CF 3 ; 7) phenyl;
  • R 2 is selected from the group consisting of: 1) H; 2) methyl, ethyl, propyl, isopropyl, tert-butyl; 3) cyclopropyl; 4) phenyl;
  • the R 1 is a methyl group
  • R 2 is a methyl group
  • the R 1 is selected from the group consisting of: 1) H; 2) F, Cl, Br, I; 3) OH, OCH 3 , OEt, OCF 3 ; 4) methyl, ethyl, isopropyl, tert-butyl; 5) cyclopropyl; 6) CF 3 ; 7) phenyl;
  • R 2 is selected from the group consisting of: H; methyl, ethyl, propyl, isopropyl; cyclopropyl; phenyl;
  • the R 1 is selected from the group consisting of: 1) H; 2) F, Cl, Br; 3) OH, OCH 3 , OEt; 4) methyl, ethyl, isopropyl, t-butyl; 5) CF 3 ; 6) phenyl;
  • R 2 is selected from the group consisting of: 1) H; 2) methyl, ethyl, propyl; 3) cyclopropyl; 4) phenyl;
  • the R 9 is selected from the group consisting of: 1) F, Cl, Br; 2) OCH 3 , OCF 3 ;
  • the Q is selected from the group consisting of CH, N.
  • the R 1 is selected from the group consisting of: 1) 2-Cl, 2-Br; 2) 2-OCH 3 , 2-OEt; 3) 2-methyl, 2-ethyl, 3-methyl, 3-ethyl ;4)H;
  • R 2 is selected from the group consisting of: 1) methyl, ethyl, propyl; 2) cyclopropyl;
  • the R 9 is selected from the group consisting of: 1) F, Cl; 2) OCH 3 , OCF 3 ;
  • the Q is selected from the group consisting of CH, N.
  • the R 1 is selected from the group consisting of: 1) H; 2) F, Cl, Br; 3) OH, OCH 3 , OEt; 4) methyl, ethyl, isopropyl, t-butyl; 5) CF 3 ; 6) phenyl;
  • R 2 is selected from the group consisting of: 1) H; 2) methyl, ethyl, propyl; 3) cyclopropyl; 4) phenyl;
  • the R 10 is selected from the group consisting of: 1) H; 2) F, Cl, Br; 3) CF 3 ; 4)
  • the U is selected from the group consisting of: CH, N.
  • the R 1 is a methyl group
  • R 2 is a methyl group
  • the R 3 is selected from the group consisting of:
  • the compound is selected from the group consisting of
  • the present invention also provides the use of the pyrazolo[1,5-a]pyridine compound or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a prodrug molecule thereof for preparing a medicament having anti-tuberculosis effect .
  • the present invention also provides an anti-tuberculosis pharmaceutical composition, wherein the active ingredient comprises the pyrazolo[1,5-a]pyridine compound or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a prodrug thereof molecule.
  • any variable e.g., R 3 , R 4 , R 5 , R 6 , R 7 , etc.
  • its definition of each occurrence is independent of each other occurrence. definition.
  • combinations of substituents and variables are allowed as long as such combinations stabilize the compound.
  • a line drawn from a substituent into the ring system means that the bond referred to can be attached to any ring atom that can be substituted. If the ring system is polycyclic, it means that such a bond is only attached to any suitable carbon atom of the adjacent ring.
  • substituents and substitution patterns for the compounds of the present invention can select substituents and substitution patterns for the compounds of the present invention to provide compounds which are chemically stable and which are readily synthesized from readily available starting materials by techniques in the art and the methods set forth below. If the substituent itself is substituted by more than one group, it is understood that these groups may be on the same carbon atom or on different carbon atoms as long as the structure is stabilized.
  • the C 0 alkane in the "C 0 - C 5 linear or branched alkane" of the present invention is H.
  • Alkoxy as used in the present invention represents a cyclic or acyclic alkyl group of the indicated number of carbon atoms attached through an oxygen bridge.
  • alkoxy includes the definition of the alkyl and cycloalkyl.
  • aryl refers to any stable monocyclic ring of up to 7 atoms in the ring or up to 7 atomic bicyclic carbon rings in each ring, at least one of which is an aromatic ring.
  • aryl include, but are not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl.
  • the "aryl” substituent is bicyclic and one ring is non-aromatic, it is understood to be attached via an aromatic ring.
  • heteroaryl of the present invention represents a stable monocyclic ring of up to 7 atoms in the ring or up to 7 atomic bicyclic carbon rings in each ring, at least one of which is an aromatic ring and contains 1-4 selected Heteroatoms from O, N and S.
  • heteroaryl examples include, but are not limited to, acridinyl, oxazolyl, porphyrin, quinoxalinyl, pyrrazolyl, fluorenyl, benzotriazolyl, fluorene Base, thienyl, benzothienyl, benzofuranyl, quinolyl, isoquinolyl, oxazolyl, isoxazolyl, fluorenyl, pyrazinyl, pyridazinyl, pyridyl, pyrimidine Base, pyrrolyl, tetrahydroquinoline.
  • heteroaryl is also understood to include any N-oxide derivative of a nitrogen-containing heteroaryl group.
  • heteroaryl substituent is bicyclic and contains a ring which is non-aromatic or contains no heteroatoms, it is understood that each is attached via an aromatic ring or via a heteroatom-containing ring.
  • heterocyclic ring or “heterocyclic group” as used in the present invention means a 5- to 10-membered aromatic or non-aromatic heterocyclic ring containing from 1 to 4 hetero atoms selected from O, N and S, and includes a bicyclic group.
  • heterocyclyl includes the “heteroaryl” as well as the dihydrogenated and tetrahydrogenated analogs thereof.
  • heterocyclic groups include, but are not limited to, benzimidazolyl, benzofuranyl, benzopyranyl, benzopyrazolyl, benzotriazolyl, benzothienyl, benzoic Azyl, carbazolyl, porphyrinyl, porphyrinyl, furyl, imidazolyl, indanyl, fluorenyl, indolazinyl, carbazolyl, isobenzopyranyl, isodecyl, iso Quinolinyl, isothiazolyl, isoxazolyl, napyridyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyridyl Zinyl, pyrazolyl, pyridazinyl, pyridopyridyl, pyridazinyl, pyridyl, pyridyl,
  • the "heterocyclic group" of the present invention is 2-azadrosterone, benzimidazolyl, 2-diazepine, imidazolyl, 2-imidazolidinone, fluorenyl, isoquinoline. , morpholinyl, piperidinyl, piperazinyl, pyridyl, pyrrolidinyl, 2-piperidone, 2-pyrimidinone, 2-pyrrolidone, quinolyl, tetrahydrofuranyl, tetrahydroisoquinolinyl or thienyl.
  • halogen as used in the present invention means chlorine, fluorine, bromine and iodine.
  • alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl and heterocyclyl substituents of the present invention may be unsubstituted or substituted.
  • a C 3 -C 6 alkyl group may be selected from one, two or three selected from OH, oxo, halogen, alkoxy, dialkylamino or heterocyclic (eg morpholinyl or piperidinyl) and the like. Substituted for the substituent.
  • the invention includes free forms of the compounds of formula (I), including pharmaceutically acceptable salts or stereoisomers or prodrug molecules thereof.
  • Some specific exemplary compounds in the present invention are protonated salts of amine compounds.
  • the term "free form" refers to an amine compound in a non-salt form.
  • the pharmaceutically acceptable salts included include not only exemplary salts of the particular compounds described herein, but also all typical pharmaceutically acceptable salts of the free forms of the compounds of formula (I).
  • the free form of the particular salt of the compound can be isolated using techniques known in the art.
  • the free form can be regenerated by treating the salt with a suitable aqueous dilute aqueous solution, such as a dilute aqueous solution of NaOH, a dilute aqueous solution of potassium carbonate, dilute aqueous ammonia, and a dilute aqueous solution of sodium bicarbonate.
  • a suitable aqueous dilute aqueous solution such as a dilute aqueous solution of NaOH, a dilute aqueous solution of potassium carbonate, dilute aqueous ammonia, and a dilute aqueous solution of sodium bicarbonate.
  • the free form differs somewhat from its salt form in certain physical properties, such as solubility in polar solvents, but for purposes of the invention such acid and base salts are otherwise pharmaceutically equivalent to their free form.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the compounds of the present invention containing a basic moiety or an acidic moiety by conventional chemical methods.
  • the salt of the basic compound is prepared by ion exchange chromatography or by reaction of the free base with a stoichiometric or excess amount of the desired salt or mixture of the inorganic or organic acid in a suitable solvent or combination of solvents.
  • a salt of an acidic compound is formed by reaction with a suitable inorganic or organic base.
  • pharmaceutically acceptable salts of the compounds of the invention include the conventional non-toxic salts of the compounds of the invention which are formed by the reaction of a basic compound of the invention with an inorganic or organic acid.
  • conventional non-toxic salts include those prepared from inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, phosphoric acid, nitric acid, and the like, and also include organic acids such as acetic acid, propionic acid, succinic acid, glycolic acid, and hard acids.
  • Fatty acid lactic acid, malic acid, tartaric acid, citric acid, ascorbic acid, pamoic acid, maleic acid, hydroxymaleic acid, phenylacetic acid, glutamic acid, benzoic acid, salicylic acid, p-aminobenzenesulfonic acid, 2-acetyl A salt prepared from oxy-benzoic acid, fumaric acid, toluenesulfonic acid, methanesulfonic acid, ethanedisulfonic acid, oxalic acid, isethionic acid, trifluoroacetic acid or the like.
  • a suitable "pharmaceutically acceptable salt” refers to a salt prepared by pharmaceutically acceptable non-toxic bases including inorganic bases and organic bases.
  • conventional non-toxic salts include salts derived from inorganic bases including aluminum, ammonium, calcium, copper, iron, ferrous, lithium, magnesium, manganese, manganese, potassium, sodium. Salt, zinc salt, etc. Ammonium salts, calcium salts, magnesium salts, potassium salts and sodium salts are particularly preferred.
  • a salt derived from a pharmaceutically acceptable organic non-toxic base comprising a salt of a primary, secondary and tertiary amine, the substituted amine comprising a naturally occurring substituted amine, a cyclic amine and a basic ion exchange resin, for example Arginine, betaine, caffeine, choline, N,N'-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, aminoethanol, ethanolamine, Ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucosamine, glucosamine, histidine, hydroxylamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine Azine, piperidine, guanidine, polyamine resin, procaine, guanidine, theobromine, triethylamine, trimethylamine, tripropylamine,
  • the present invention has the following beneficial effects:
  • the pyrazolo[1,5-a]pyridine compound of the present invention has good anti-tuberculosis activity in vitro, and the minimum inhibitory concentration (MIC) of the compound is less than 0.1 ⁇ g/mL, and partially reaches 0.01 ⁇ g. /mL, has a strong inhibitory effect on clinically sorted multidrug-resistant tuberculosis (MDR-TB) strains.
  • MDR-TB multidrug-resistant tuberculosis
  • the pyrazolo[1,5-a]pyridine compound of the present invention can effectively eliminate the infection amount of H37Ra in mice at a dose of 20 mg/kg/d, and is a new type of anti-tuberculosis. Compound.
  • the pyrazolo[1,5-a]pyridine compound of the present invention has good pharmacokinetic properties and low toxic and side effects, has good drug-forming properties, and has obvious pharmacological effects in vivo. Very good development prospects.
  • Figure 1 is a test result of the anti-tuberculosis activity of the compound L298 in real time
  • Figure 3 is a test result of the anti-tuberculosis activity of the compound L372 in real time
  • Fig. 4 shows the results of measuring the anti-tuberculosis activity of the compound L298 in the spleen.
  • the compounds of the invention may be administered to a mammal, preferably a human, alone or in a pharmaceutical composition in combination with a pharmaceutically acceptable carrier, according to standard pharmaceutical techniques.
  • the compound can be administered orally or subcutaneously, intramuscularly, intraperitoneally, intravenously, rectally, or topically.
  • Orally administered for example, as a tablet, lozenge, lozenge, aqueous or oily suspension, dispersible powder or granule, emulsion, hard or soft capsule, or syrup or expectorant; , intramuscular injection, intraperitoneal, intravenous administration, can be made into a sterile injectable aqueous solution; the rectal administration can be made into a suppository form; the topical administration can be made into a cream, an ointment, a gel, a solution Agent or suspension, etc.
  • the pharmaceutically acceptable carrier can be, for example, an inert diluent such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents such as microcrystalline cellulose, crosslinked carboxymethyl fibers.
  • an inert diluent such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate
  • granulating and disintegrating agents such as microcrystalline cellulose, crosslinked carboxymethyl fibers.
  • the tablets may be uncoated or coated by known techniques to mask the unpleasant taste of the drug or to prolong disintegration and absorption in the gastrointestinal tract and thus provide a drug effect that lasts longer.
  • Step 1 4-(4-(Trifluoromethoxy)phenyl)-5,6-dihydropyridine-1(2H)-tert-butyl carbonate (A 1 )
  • N-phenylbis(trifluoromethanesulfonyl)imide 39.6 mmol, 14.15 g
  • THF dry THF
  • Step 2 4-(4-(Trifluoromethoxy)phenyl)piperidine (A 2 )
  • Step 3 4-(4-(4-(Trifluoromethoxy)phenyl)piperidin-1-substituted) benzonitrile (A 3 )
  • Step 4 (4-(4-(4-(Trifluoromethoxy)phenyl)piperidin-1-substituted)phenyl)methanamine hydrochloride A 4
  • Step 5 2-Ethyl-5-((tert-butoxycarbonyl)amine)-pyrazole [1,5-a]pyridine-3-ethyl carbonate B 1
  • Step 7 2-Ethyl-5-chloro-pyrazole [1,5-a]pyridin-3-ethyl carbonate B 3
  • Step 8 2-Ethyl-5-chloro-pyrazole [1,5-a]pyridine-3-carboxylic acid B 4
  • Step 9 5-Chloro-2-ethyl-N-(4-(4-(4-(trifluoromethoxy)phenyl)piperidin-1-substituted)benzyl)pyrazole [1,5- a]pyridine-3-carboxamide (TJ170298)
  • the synthesis method is as in steps 5-9 in Example 1.
  • Step 2 2-Methyl-N-(4-(4-(4-(trifluoromethoxy)phenyl)piperidin-1-substituted)benzyl)pyrazole [1,5-a]pyridine- 3-formamide (TJ170381)
  • the synthesis method is as in steps 8 and 9 in Example 1.
  • the synthesis method is as in Steps 5-9 of Example 1.
  • Step 1 4-(4-(4-(Trifluoromethoxy)phenyl)piperidine-1-substituted) Aniline
  • Step 2 5-Chloro-2-ethyl N-(4-(4-(4-(trifluoromethoxy)phenyl)piperidin-1-substituted)phenyl)pyrazole [1,5-a Pyridine-3-carboxamide (TJ064819)
  • Step 2 5-Chloro-2-ethyl N-((1-(4-(trifluoromethoxy)phenyl)-1H-1,2,3-triazole-4-substituted)methyl) Pyrazole [1,5-a]pyridine-3-carboxamide (TJ064983)
  • the synthesis method was as in Step 9 in Example 1, and the yield was 64.0%.
  • TJ170298 (0.2 g, 0.34 mmol) was dissolved in anhydrous DMF (7 mL), and 60% NaH (0.02 g, 0.51 mmol) was added under ice-cooling. After stirring for 0.5 h, then CH 3 I (0.063 g, 0.44) Mm). The reaction was allowed to move to room temperature and stirring was continued for 1 h. After extracting three times with ethyl acetate, the organic phase was collected, and the solvent was evaporated to dryness, and then purified by column chromatography to afford product TJ064985, 0.18 g (92.2%).
  • Step 1 2-(4-(4-(4-(Trifluoromethoxy)phenyl)piperidin-1-substituted)phenyl)propan-2-amine hydrochloride
  • Step 2 5-Chloro-2-ethyl-N-(2-(4-(4-(4-(trifluoromethoxy)phenyl)piperidin-1-substituted)phenyl)propane-2- Substituted) pyrazole [1,5-a]pyridine-3-carboxamide (TJ064987)
  • the synthesis method was as in Step 9 in Example 1, and the yield was 59.8%.
  • Step 2 5-Trifluoromethyl-2-methyl-N-(4-(4-(4-(trifluoromethoxy)phenyl)piperidin-1-substituted)benzyl)pyrazole [1 ,5-a]pyridine-3-carboxamide (TJ064995)
  • the synthesis method was as in steps 8 and 9 in Example 1, and the total yield in two steps was 85.1%.
  • Step 2 (6-(4-(Trifluoromethoxy)phenyl)pyridine-3 substituted) methylamine hydrochloride
  • 6-(4-(Trifluoromethoxy)phenyl)-3-cyanopyridine (2.0 g, 7.57 mmol) was dissolved in methanol (100 mL) and a catalytic amount of Raney nickel was added. After replacing the hydrogen balloon three times, it was stirred at room temperature for 3.5 h. After completion of the reaction, the mixture was filtered through a short pad of silica gel. The product was dissolved in ethanol, and an excess of HCl solution in HCl was added to make a salt. The solvent was evaporated to dryness to give (6-(4-(trifluoromethoxy)phenyl)pyridin-3-substituted)methylamine hydrochloride. , solid 1.5 g (58.1%).
  • Step 3 5-Chloro-2-ethyl-N-((6-(4-(trifluoromethoxy)phenyl)pyridine-3-substituted)methyl)pyrazole [1,5-a]pyridine -3-carboxamide (TJ830028)
  • the synthesis method was as in Step 9 in Example 1, and the yield was 57.9%.
  • Step 3 1-(4-(1-azidoethyl)phenyl-4-(4-(trifluoromethoxy)phenyl)piperidine
  • Step 5 5-Chloro-2-ethyl-N-(1-(4-(4-(4-(trifluoromethoxy)phenyl)piperidin-1-substituted)phenyl)ethanol)pyrazole [1,5-a]pyridine-3-carboxamide (TJ830070)
  • the synthesis method was as in Step 9 in Example 1, and the yield was 51.1%.
  • Step 2 2-(4-(4-(4-(Trifluoromethoxy)phenyl)piperidin-1-substituted)phenethyl)isoindoline-1,3-dione
  • Step 4 5-Chloro-2-ethyl N-(4-(4-(4-(trifluoromethoxy)phenyl)piperidin-1-substituted)phenethyl)pyrazole [1,5- a]pyridine-3-carboxamide (TJ830128)
  • the synthesis method was as in Step 9 in Example 1, and the yield was 63.8%.
  • the thiophene-2-substituted methylamine (1.4 g, 12.36 mmol) and sodium hydrogencarbonate (1.04 g, 12.36 mmol) were dissolved in anhydrous THF (20 mL), and Boc 2 O (2.76 g, 13.6 mmol) was slowly added dropwise. After reacting for 3 h at room temperature, the solvent was evaporated to dryness and evaporated. The organic phase was collected by column chromatography to give the product 2.61 g (99.1%).
  • tert-Butyl (thiophene-2 substituted methyl)carbamate (2.22 g, 10.41 mmol) was dissolved in DMF (11 mL) and then NBS (2.04 g, 11.45 mmol). After reacting for 5 hours at room temperature, a large amount of water was added, and after extracting three times with ethyl acetate, the organic phase was collected. Column chromatography gave the product 2.56 g (84.3%).
  • Step 3 tert-Butyl ((5-(4-(trifluoromethoxy)phenyl)thiophene-2-substituted)carbamate)
  • Step 4 (5-(4-(Trifluoromethoxy)phenyl)thiophene-2-substituted) methylamine
  • Step 5 2,5-Dimethyl-N-((5-(4-(trifluoromethoxy)phenyl)thiophene-2-substituted)methyl)pyrazole [1,5-a]pyridine- 3-formamide (TJ830132)
  • the synthesis method was as in Step 9 in Example 1, and the yield was 75.9%.
  • the synthesis method is as in Example 22.
  • Step 2 N-(4-(4-Fluorophenoxy)benzyl)-2,5-dimethylpyrazole[1,5-a]pyridine-3-carboxamide (TJ830147)
  • the synthesis method was as in steps 4 and 9 in Example 1, and the yield in two steps was 80.6%.
  • the synthesis method is as in steps 2, 3 and 4 in Example 26.
  • the synthesis method is as in step 9 of Example 1.
  • Step 2 2,5-Dimethyl-N-((4'-(trifluoromethoxy)-[1,1'-biphenyl]-4-substituted)methyl)pyrazole [1,5- a]pyridine-3-carboxamide (TJ830151)
  • the synthesis method was as in Step 9 in Example 1, and the yield was 67.3%.
  • the synthesis method is as in steps 2, 3 and 4 in Example 1.
  • Step 3 2,5-Dimethyl-N-(4-(4-(4-(trifluoromethoxy)phenoxy)piperidine-1-substituted)benzyl)pyrazole [1,5- a]pyridine-3-carboxamide (TJ830161)
  • the synthesis method was as in Step 9 in Example 1, and the yield was 68.0%.
  • Example 51 Inhibitory activity against Autonomous Light-emitting Tuberculosis.
  • a stable primary luminescent bacterium having no resistance screening marker frozen at -80 ° C was inoculated into a conical flask containing 50 mL of 7H9 (containing 0.1% soil temperature 80) medium in 2 mL, and cultured until the OD value reached 0.3-1.0. between.
  • the fluorescence value is detected in a 1.5 mL EP tube, and the fluorescence value is preferably about 10 to 1,000,000 / mL or more.
  • the bacterial solution was diluted 10, 100, and 1000 times with the stock solution (dilution method: 100 ⁇ L of the previous concentration was added to 900 ⁇ L of 7H9 medium for the next concentration, and the mixture was fully aspirated and then sequentially diluted).
  • a bacterial solution diluted with a dilution factor of 2000-10000/25 ⁇ L was used as a bacterial solution for detection.
  • the diluted bacterial solution was added to a 384-well plate with a lancet, 25 ⁇ L per well, and the 384-well plate was incubated in a 37 ° C incubator for about 1-2 hours, and the fluorescence value of each well was measured, and the fluorescence value was measured. The average value is recorded as the Day 0 fluorescence value.
  • test sample was uniformly dispensed into 10 mg/mL mother liquor in DMSO, and subjected to 3-fold gradient dilution.
  • the positive drugs rifampicin (RIF) and isoniazid (INH) were conditioned in two concentrations of DMSO, 2 mg/mL and 1 mg/mL. .
  • the compound was added to a 384 well plate. In a 384-well plate, 5% ⁇ L of the test sample per well was stored at -20 °C.
  • Dosing was performed using the Echo520 Ultrasonic Micropipetting System. The sample was sampled at 25 nL per well (operated in a total volume of 0.025 ml of 7H9 medium) to form a 3-fold gradient dilution starting at 10 ⁇ g/mL of the final concentration of the compound.
  • the concentrations were 10 mg/mL, 3 mg/mL, 1 mg/mL, 0.3 mg/mL, 0.1 mg/mL, 0.03 mg/mL, 0.01 mg/mL, 0.003 mg/mL, 0.001 mg/mL, 0.0003 mg/mL, 0.0001 mg/mL, 0.00003 mg/mL, 0.00001 mg/mL, 0.000003 mg/mL, 0.000001 mg/mL.
  • the positive drug was loaded from the mother solution of 2mg/mL to 125nL and 12.5nL, and from the mother solution of 1mg/mL to 2.5nL to the final volume of 0.025nL. The final concentration of the positive drug was 10 ⁇ g/mL, 1 ⁇ g/ mL, 0.1 ⁇ g/mL.
  • DMSO was used as a control. Stir on the shaker for 5 min and continue to culture.
  • the cells were incubated at 37 ° C in an incubator at 24 hours, 48 hours, and 72 hours, respectively.
  • a sample whose fluorescence value gradually decreases with time is active, wherein RLU is the relative light unit of the fluorescence value.
  • the results are shown in Table 1.
  • Activity range A indicates ⁇ 1 ⁇ g/mL, B indicates 1-10 ⁇ g/mL, and C indicates >10 ⁇ g/mL.
  • Example 52 Determination of MIC of MIC of Mycobacterium tuberculosis by the method of the present invention by plate method (minimum inhibitory concentration)
  • Liquid culture of Mycobacterium tuberculosis standard strain H37Rv cultured in 7H9 medium, and cultured at 37 ° C for a long time until the bacterial solution has an absorbance (OD 600 ) of 0.3-0.7 at 600 nm.
  • MIC detection dilute the M. tuberculosis standard strain H37Rv culture solution with sterile water to 50 times to obtain the mother liquid, and then dilute the mother liquid to a concentration of 10 -3 and 10 -5 , and then the two concentrations of the bacterial liquid 0.5 mL was placed on the above 7H11 plate, and each was repeated twice.
  • the positive drug isoniazid 0.1 ⁇ g/mL was used as a control.
  • Example 53 Sensitivity determination of the compound to clinically sorted multidrug-resistant tuberculosis (MDR-TB) strains
  • Mycobacterium tuberculosis standard strain H37Rv and clinically sorted MDR-TB bacteria P91, P105 and P103 liquid culture cultured in 7H9 medium, cultured at 37 ° C shaker to absorb absorbance at 600 nm (OD 600) ) is 0.7 or more.
  • MIC detection dilute the culture solution of each strain with sterile water to 50 times to obtain the mother liquid, and then dilute the mother liquid to a concentration of 10 -3 and 10 -5 , and then take 0.5mL of each of the two concentrations of the bacterial liquid. On the above 7H11 plates, each was repeated twice.
  • the positive drug RIF 10 ⁇ g/mL and 1 ⁇ g/mL were used as controls.
  • Example 54 Determination of anti-tuberculosis activity of compounds of the invention in vivo
  • Liquid culture of Mycobacterium tuberculosis autonomous luminescent bacteria H37Ra cultured in 7H9 medium, and cultured at 37 ° C for a long time until the luminescence value of the bacterial liquid is 22,000,000 RLU/mL.
  • Rat intravenous administration 3 SD rats, weighing 180-220 g. Fasting for 12 hours before administration, a small amount of food during the test, and free drinking water.
  • Compounds TJ170298 and TJ170385 were administered intravenously at a dose of 2 mg/kg. 2 min, 10 min, 30 min, 1.0 h, 2.0 h, 3.0 h, 4.0 h, 6.0 h, 8.0 h, 12.0 h, 21 h, 24.0 h, 30 h, 36 h, 48 h, 60 h, 72 h after administration, respectively.
  • Approximately 0.3 mL was placed in a heparinized test tube, centrifuged at 6000 rpm for 10 min, plasma was separated, and stored at 4 ° C for testing.
  • Rat oral administration 3 SD rats, weighing 180-220 g. Fasting for 12 hours before administration, a small amount of food during the test, free access to water.
  • Compounds TJ170298 and TJ170385 were administered orally at a dose of 10 mg/kg, respectively. 5 min, 10 min, 30 min, 1.0 h, 2.0 h, 3.0 h, 4.0 h, 6.0 h, 8.0 h, 12.0 h, 21 h, 24.0 h, 30 h, 36 h, 48 h, 60 h, 72 h after administration, respectively.
  • Approximately 0.3 mL of heparinized tubes were centrifuged at 6000 rpm for 10 min, plasma was separated, and stored at 4 ° C for testing.
  • the pharmacokinetic parameters of the rats after administration were calculated using DAS 2.0 software.
  • AUC Absolute Under the Curve: The area under the blood concentration-time curve, representing the bioavailability of the drug (the extent to which the drug is absorbed and utilized in the human body), and the AUC is higher in bioavailability, and vice versa. AUC is called area under concentration-time curve.
  • Peak Concentration refers to the maximum concentration of blood in the blood concentration-time curve, that is, the highest plasma drug concentration that can be achieved after administration.
  • the peak concentration is closely related to the clinical application of the drug.
  • the concentration of the drug peak can reach the effective concentration to be effective, and if it is higher than the safe range, the toxicity reaction can be indicated.
  • drug peak concentration is an important indicator of the absorption and safety of the formulation.
  • T 1/2 half life time. It refers to the time required for the concentration of the drug to decrease by half in the body, reflecting the speed at which the drug is eliminated from the body through biotransformation or excretion.
  • Tmax (Peak Time) is the time required to reach the highest concentration (drug peak concentration) on the human plasma drug concentration curve after administration.
  • the short peak time indicates that the drug is absorbed quickly and the effect is rapid, but it is also eliminated quickly.
  • the long peak time indicates that the drug absorption and onset are slow, and the duration of drug action is often prolonged. Peak time is an important indicator of applied drugs and research formulations.
  • Bioavailability Bioavailability. It refers to the speed and extent to which the drug is absorbed into the large circulation. Bioavailability is divided into absolute bioavailability and relative bioavailability. Absolute bioavailability refers to the percentage of the drug that is used 100% when administered intravenously, and the other dosage forms of the drug are absorbed and utilized by the body when the dosage is equal; the relative bioavailability is in any arbitrarily specified dosage form (eg, oral water preparation). ) was utilized at 100% and then the percent utilization of the other dosage forms of the drug under the same conditions was determined.
  • Absolute bioavailability refers to the percentage of the drug that is used 100% when administered intravenously, and the other dosage forms of the drug are absorbed and utilized by the body when the dosage is equal; the relative bioavailability is in any arbitrarily specified dosage form (eg, oral water preparation). ) was utilized at 100% and then the percent utilization of the other dosage forms of the drug under the same conditions was determined.

Abstract

本发明公开一种具有式(I)结构特征的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子及其应用。该类化合物具有良好的体外抗结核病菌活性,化合物的最小抑菌浓度(MIC)低于0.1μg/mL,部分达到0.01μg/mL,对临床分选的耐多药结核菌(MDR-TB)菌株具有很强的抑制作用。体内实验中,本发明所述的吡唑并[1,5-a]吡啶类化合物在20mg/kg/d的剂量下,可有效清除小鼠体内H37Ra的感染量,是一类新型的抗结核化合物。

Description

吡唑并[1,5-a]吡啶类化合物及其应用 技术领域
本发明属于化学医药领域,特别是涉及吡唑并[1,5-a]吡啶类化合物及其应用。
背景技术
结核病(Tuberculosis,TB)是由结核分枝杆菌(Mycobacterium tuberculosis,Mtb)感染引起的,是全球流行最广的疾病之一。在人类结核病中,最常见的是肺结核,占各器官结核病总数的80-90%。由于耐药结核菌的出现和艾滋病的并行感染,使得现有临床药物的治疗效果大大降低。
根据第18次WHO全球结核病报告显示,在2012年世界范围内约有860万人结核病新生患者,死亡人数达到130万人。在中国,结核病的发病率占世界发病率的12%(100万人)居世界第二,死亡人数4.4万居世界第五。而且耐多药结核菌在世界范围内进一步广泛传播,2012年约有45万的新生耐病例。而我国是全球27个耐多药结核病流行严重的国家之一,约有1.6万人被报道为耐多药结核病。
目前使用的治疗方案是一线药物异烟肼,利福平,吡嗪酰胺和乙胺丁醇等与二线药物的联合用药,治疗时间达6-9个月,且对耐药结核菌和持留菌没有较好的治疗效果,给患者带来很大的困扰和经济负担。目前使用的一线抗结核药物的耐药机理都已比较明确,主要原因是由于药物的不当使用,导致相应的作用靶点基因或药物活化相关酶基因或者相关调节基因发生突变,致使药物失效。因此,在全球抗结核药物开发联盟(the Global Alliance for TB Drug Development)的发动下,人们又开始重新关注抗结核药物的研发工作。新型的药物应当具有以下特征:①开发能将治疗周期缩短的新药;②开发对耐药结核菌有疗效的药物;③开发可有效治疗潜伏态细菌感染的新药。
抗结核药物的研究与开发主要有三个方面:天然物筛选、新结构类型化合物设计合成、抗菌药物的再修饰。近年来国内外一些新合成的抗结核药物有喹啉类、硝基咪唑吡喃类、恶唑烷酮类、吡咯类、咪唑并吡啶类等。2012年底,双芳基喹啉类药物bedaquiline被FDA加速批准上市,作为成人耐多药结核病联合治疗方案的组分之一,这也是自1970年引进利福平以来首个新型抗结核药物。但是后续的临床试验表明bedaquiline还表现出一定的毒副作用,安全隐患依然存在。所以进一步开发安全有效的抗结核药物以克服临床上耐药性结核病的危害具有重大意义。
发明内容
基于此,本发明的目的在于提供一种吡唑并[1,5-a]吡啶类化合物及其应用。
为实现上述目的,本发明采取以下技术方案:
具有式(I)结构特征的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子:
Figure PCTCN2015086852-appb-000001
m为0,1,2,3或4;n为0或1;V为CH或N;W为CH或N;X任选自:CH,O,N或S;Y任选自:CH,O,N或S;Z任选自:CH,O,N或S;L任选自:C0~C5直链或支链的烷烃;R8为H或甲基;
R1任选自:1)H;2)卤素;3)OaC1~C5烷基;4)C3~C6环烷基;5)芳基;6)氨基, 羟基,氰基,硝基;7)杂芳基;所述烷基、芳基、环烷基和杂芳基分别任选被0,1,2或3个选自R6的取代基取代;
R2任选自:1)H;2)OaC1~C5烷基;3)C3~C6环烷基;4)芳基;5)杂芳基;
所述烷基、芳基、环烷基和杂芳基分别任选被0,1,2或3个选自R6的取代基取代;
R3任选自:1)H;2)卤素;3)OaC1~C5烷基;4)C3~C6环烷基;5)
Figure PCTCN2015086852-appb-000002
6)
Figure PCTCN2015086852-appb-000003
b为0或1;U为CH或N;R4任选自:H;F,Cl,Br;C1~C3烷基;C1~C3烷氧基;苯氧基;7)
Figure PCTCN2015086852-appb-000004
R5任选自:H;F,Cl,Br;OaC1~C3烷基;8)
Figure PCTCN2015086852-appb-000005
9)(C=O)cOdR7,其中,c为0或1;d为0或1;R7任选自:a)CF3;b)C1~C5烷基;10)-N(CH3)2;11)
Figure PCTCN2015086852-appb-000006
12)
Figure PCTCN2015086852-appb-000007
所述烷基、芳基、环烷基、杂环烷基和杂芳基分别任选被0,1,2或3个选自R6取代基取代,其中,所述a为0或1;所述R6任选自:1)H;2)C3~C6环烷基;3)杂环基;4)C1~C3烷基;5)C1~C3含氟烷基;6)C0~C3亚烷基-杂环基;7)卤素。
在其中一个实施例中,所述R1任选自:1)H;2)F,Cl,Br,I;3)OH,OCH3,OEt,OCF3;4)甲基,乙基,异丙基,叔丁基;5)环丙基;6)CF3;7)苯基;
所述R2任选自:1)H;2)甲基,乙基,丙基,异丙基,叔丁基;3)环丙基;4)苯基;
所述R3任选自:1)
Figure PCTCN2015086852-appb-000008
2)
Figure PCTCN2015086852-appb-000009
Figure PCTCN2015086852-appb-000010
4)
Figure PCTCN2015086852-appb-000011
5)CF3;6)(C=O)cOdR7,其中,c为0或1;d为0或1;R7任选自:a)CF3;b)C1~C5烷基;7)H;8)卤素。
在其中一个实施例中,所述
Figure PCTCN2015086852-appb-000012
选自:
Figure PCTCN2015086852-appb-000013
Figure PCTCN2015086852-appb-000014
在其中一个实施例中,具有如式(II)所示结构特征:
Figure PCTCN2015086852-appb-000015
所述R1为甲基;R2为甲基;
所述R3任选自:1)-N(CH3)2;2)
Figure PCTCN2015086852-appb-000016
3)-C(CH3)3;4)
Figure PCTCN2015086852-appb-000017
5)
Figure PCTCN2015086852-appb-000018
6)
Figure PCTCN2015086852-appb-000019
n=1。
在其中一个实施例中,具有如式(II)所示结构特征:
Figure PCTCN2015086852-appb-000020
所述R1任选自:1)H;2)F,Cl,Br,I;3)OH,OCH3,OEt,OCF3;4)甲基,乙基,异丙基,叔丁基;5)环丙基;6)CF3;7)苯基;
所述R2任选自:H;甲基,乙基,丙基,异丙基;环丙基;苯基;
所述R3任选自:1)
Figure PCTCN2015086852-appb-000021
2)
Figure PCTCN2015086852-appb-000022
3)
Figure PCTCN2015086852-appb-000023
4)
Figure PCTCN2015086852-appb-000024
5)CF3;6)(C=O)OMe;7)H;8)卤素;
所述
Figure PCTCN2015086852-appb-000025
选自
Figure PCTCN2015086852-appb-000026
Figure PCTCN2015086852-appb-000027
在其中一个实施例中,具有如式(III)所示结构特征:
Figure PCTCN2015086852-appb-000028
所述R1任选自:1)H;2)F,Cl,Br;3)OH,OCH3,OEt;4)甲基,乙基,异丙基,叔丁基;5)CF3;6)苯基;
所述R2任选自:1)H;2)甲基,乙基,丙基;3)环丙基;4)苯基;
所述R9任选自:1)F,Cl,Br;2)OCH3,OCF3
所述Q任选自:CH,N。
在其中一个实施例中,
所述R1任选自:1)2-Cl,2-Br;2)2-OCH3,2-OEt;3)2-甲基,2-乙基,3-甲基,3-乙基;4)H;
所述R2任选自:1)甲基,乙基,丙基;2)环丙基;
所述R9任选自:1)F,Cl;2)OCH3,OCF3
所述Q任选自:CH,N。
在其中一个实施例中,具有如式(IV)所示结构特征:
Figure PCTCN2015086852-appb-000029
所述R1任选自:1)H;2)F,Cl,Br;3)OH,OCH3,OEt;4)甲基,乙基,异丙基,叔丁基;5)CF3;6)苯基;
所述R2任选自:1)H;2)甲基,乙基,丙基;3)环丙基;4)苯基;
所述R10任选自:1)H;2)F,Cl,Br;3)CF3;4)
Figure PCTCN2015086852-appb-000030
所述U任选自:CH,N。
在其中一个实施例中,具有如式(II)所示结构特征:
Figure PCTCN2015086852-appb-000031
所述R1为甲基;R2为甲基;
所述R3任选自:
1)
Figure PCTCN2015086852-appb-000032
在其中一个实施例中,所述化合物选自:
5-氯-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-2-乙基-N-(4-(三氟甲氧基)苯基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-2-环丙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-2-苯基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;4-((5-氯-2-乙基吡唑[1,5-a]吡啶-3-甲酰胺)甲基)苯甲酸甲酯;5-氯-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)吡唑[1,5-a]吡啶-3-甲酰胺;2,7-二甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;2,6-二甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;2,4-二甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-甲氧基-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-2-乙基-N-((1-(4-(三氟甲氧基)苯基)-1H-1,2,3-三氮唑-4-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-2-乙基-N-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-2-乙基-N-(2-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)丙烷-2-取代)吡唑[1,5-a]吡啶-3-甲酰胺;5-三氟甲基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-((1-(4-(三氟甲氧基)苯基)-1H-1,2,3-三氮唑-4-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺;5-溴-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-苯基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-2-乙基-N-((6-(4-(三氟甲氧基)苯基)吡啶-3-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-((6-(4-(三氟甲氧基)苯基)吡啶-3-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺;5-甲氧基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;2-甲基-N-(4-(三氟甲氧基)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-2-乙基-N-(1-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)乙醇)吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-(4-(三氟甲氧基)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-异丙基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-叔丁基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-乙基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-甲基-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-甲基-2-乙氧基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯乙基)吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-((5-(4-(三氟甲氧基)苯基)噻吩-2-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-((5-(4-(三氟甲氧基)苯基)吡啶-2-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-((5-(4-(三氟甲氧基)苯基)吡啶-2-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺;N-(4-(叔丁基)苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺;N-(4-(二甲氨基)苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺;N-(4-(4-(4-甲氧基苯基)哌啶-1-取代)苄基)-2,5-二甲基-吡唑[1,5-a]吡啶-3-甲酰胺;N-(4-(4-(4-氟苯基)哌啶-1-取代)苄基)-2,5-二甲基-吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-(4-(哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;N-(4-(4-氟苯氧基)苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-(4-(4-(三氟甲基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-((2-(4-(三氟甲氧基)苯基)嘧啶-5-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-((4’-(三氟甲氧基)-[1,1’-联苯]-4-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺;N-(4-(4-(4-氟苯基)哌嗪-1-取代)苄基)-2,5-二甲基-吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-(4-(4-(三氟甲氧基)苯基)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;2,5-二甲基-N-(4-(4-(4-(三氟甲氧基)苯氧基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺;5-氯-2-乙基-N-(4-氟苄基)吡唑[1,5-a]吡啶-3-甲酰胺;N-(4-氟苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺。
本发明还提供所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子在制备具有抗结核病作用的药物中的应用。
本发明还提供一种抗结核病药物组合物,其活性成分包括所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子。
本发明所述化合物,当任何变量(例如R3、R4、R5、R6、R7等)在任何组分中出现超过一次,则其每次出现的定义独立于其它每次出现的定义。同样,允许取代基及变量的组合,只要这种组合使化合物稳定。自取代基划入环系统的线表示所指的键可连接到任何能取代的环原子上。如果环系统为多环,其意味着这种键仅连接到邻近环的任何适当的碳原子上。要理解本领域普通技术人员可选择本发明化合物的取代基及取代型式而提供化学上稳定的并可通过本领域技术和下列提出的方法自可容易获得的原料容易的合成的化合物。如果取代基自身被超过一个基团取代,应理解这些基团可在相同碳原子上或不同碳原子上,只要使结构稳定。
本发明所述“C0~C5直链或支链的烷烃”中的C0烷烃即为H。
本发明所述“任选被一个或多个取代基取代”被认为与短语“任选被至少一个取代基取代”相当,且在此情况下优选的实施方案将具有0-3个取代基。
本发明所述“烷氧基”代表通过氧桥连接的指明数目的碳原子的环状的或非环状的烷基。因此“烷氧基”包括所述烷基和环烷基的定义。
本发明所述“芳基”是指环中多达7个原子的任何稳定的单环或每个环中多达7个原子双环碳环,其中至少一个环为芳香环。这种“芳基”的实例包括但不限于:苯基、萘基、四氢萘基、茚满基、联苯基。在“芳基”取代基是双环的且一个环为非芳香性的例子中,应理解为经芳香环连接。
本发明所述“杂芳基”代表环中多达7个原子的稳定的单环或每个环中多达7个原子双环碳环,其中至少一个环为芳香环且含有1-4个选自O、N和S的杂原子。这种“杂芳基”的实例包括但不限于:丫啶基、咔唑基、噌啉基、喹噁啉基、吡唑基(pyrrazolyl)、吲哚基、苯并三唑基、吠喃基、噻吩基、苯并噻吩基、苯并吠喃基、喹啉基、异喹啉基、噁唑基、异噁唑基、吲哚基、吡嗪基、哒嗪基、吡啶基、嘧啶基、吡咯基、四氢喹啉。所述“杂芳基”也理解为包括任何含有氮的杂芳基的N-氧化物衍生物。在“杂芳基”取代基是双环的且含有一个环为非芳香性或不含有杂原子的例子中,应理解各自经芳香环或经含杂原子环连接。
本发明所述“杂环”或“杂环基”是指含有1-4个选自O、N和S的杂原子的5-10元芳香性或非芳香性杂环,且包括双环基团。因此所述“杂环基”包括所述“杂芳基”,也包括其二氢化及四氢化类似物。这种“杂环基”的实例包括但不限于:苯并咪唑基、苯并呋喃基、苯并吡喃基、苯并吡唑基、苯并三唑基、苯并噻吩基、苯并噁唑基、咔唑基、咔啉基、噌啉基、呋喃基、咪唑基、二氢吲哚基、吲哚基,indolazinyl、吲唑基、异苯并吠喃基、异吲哚基、异喹啉基、异噻唑基、异噁唑基、奈吡啶基、噁二唑基、噁唑基、噁唑啉、异噁唑啉、氧杂环丁烷基(oxetanyl)、吡喃基、吡嗪基、吡唑基、哒嗪基、吡啶并吡啶基、哒嗪基、吡啶基、嘧啶基、吡咯基、喹唑啉基、喹啉基、喹噁啉基、四氢吡喃基、四唑基、四唑并吡啶基、噻二唑基、噻唑基、噻吩基、三唑基、氮杂环丁烷基、1,4-二噁烷基、氮杂环庚烷基(hexallydroazepinyl)、哌嗪基、哌啶基、吡啶-2-酮基、吡咯烷基、吗啉基、硫代吗啉基(thiomorpholinyl)、二氢苯并咪唑基、二氢苯并呋喃基、二氢苯并噻吩基、二氢苯并噁唑基、二氢呋喃基、二氢咪唑基、二氢吲哚基、二氢异噁唑基、二氢异噻唑基、二氢噁二唑基、二氢噁唑基、二氢吡嗪基、二氢吡唑基、二氢吡啶基、二氢嘧啶基、二氢吡咯基、二氢喹啉基、二氢四唑基、二氢噻二唑基、二氢噻唑基、二氢噻吩基、二氢三唑基、二氢氮杂环丁烷基、亚甲基二氧基苯甲酰基、四氢呋喃基和四氢噻吩基,及其N-氧化物。“杂环基”取代基的连接可通过碳原子或杂原子实现。
作为优选地,本发明所述“杂环基”为2-氮杂卓酮、苯并咪唑基、2-二氮杂卓酮、咪唑基、2-咪唑啉酮、吲哚基、异喹啉基、吗啉基、哌啶基、哌嗪基、吡啶基、吡咯烷基、2-哌啶酮、 2-嘧啶酮、2-吡咯烷酮、喹啉基、四氢呋喃基、四氢异喹啉基或噻吩基。
本发明所述“卤素”指氯、氟、溴和碘。
除非另有定义,本发明所述烷基、烯基、炔基、环烷基、芳基、杂芳基和杂环基取代基可为未被取代的或取代的。例如,C3-C6烷基可被一个、两个或三个选自OH、氧代、卤素、烷氧基、二烷基氨基或杂环基(例如吗啉基或哌啶基)等的取代基取代。在这种情况下,如果一个取代基为氧代且另一个为OH,则定义包括如下实例:-(C=O)C H2CH(OH)CH3、-(C=O)OH、-CH2(OH)CH2CH(O)等。
本发明包括式(I)化合物的游离形式,也包括其药学上可接受的盐或立体异构体或前药分子。本发明中一些特定的示例性化合物为胺类化合物的质子化了的盐。术语“游离形式”指以非盐形式的胺类化合物。包括在内的药学上可接受盐不仅包括本发明所述特定化合物的示例性盐,也包括所有式(I)化合物游离形式的典型的药学上可接受的盐。可使用本领域已知技术分离所述化合物特定盐的游离形式。例如,可通过用适当的碱稀水溶液,例如NaOH稀水溶液、碳酸钾稀水溶液、稀氨水及碳酸氢钠稀水溶液,处理该盐使游离形式再生。游离形式在某些物理性质,例如在极性溶剂中溶解度,与其盐形式多少有些区别,但是为发明的目的这种酸盐及碱盐在其它药学方面与其游离形式相当。
可通过常规化学方法自含有碱性部分或酸性部分的本发明化合物合成本发明的药学上可接受的盐。通常,通过离子交换色谱或通过游离碱和化学计算量或过量的所需盐形式的无机或有机酸在适当溶剂或多种溶剂的组合中反应制备碱性化合物的盐。类似的,通过和适当的无机或有机碱反应形成酸性化合物的盐。
因此,本发明化合物的药学上可接受的盐包括通过碱性的本发明化合物和无机或有机酸反应形成的本发明化合物的常规无毒盐。例如,常规无毒盐包括得自无机酸例如盐酸、氢溴酸、硫酸、氨基磺酸、磷酸、硝酸等制备的盐,也包括自有机酸例如乙酸、丙酸、琥珀酸、乙醇酸、硬脂酸、乳酸、苹果酸、酒石酸、柠檬酸、抗坏血酸、扑酸、马来酸、羟基马来酸、苯乙酸、谷氨酸、苯甲酸、水杨酸、对氨基苯磺酸、2-乙酰氧基-苯甲酸、富马酸、甲苯磺酸、甲磺酸、乙烷二磺酸、草酸、羟乙基磺酸、三氟乙酸等制备的盐。
如果本发明化合物为酸性,则适当的“药学上可接受的盐”指通过药学上可接受的无毒碱包括无机碱及有机碱制备的盐。例如,常规无毒盐包括得自无机碱的盐包括铝盐、铵盐、钙盐、铜盐、铁盐、亚铁盐、锂盐、镁盐、锰盐、亚锰盐、钾盐、钠盐、锌盐等。特别优选铵盐、钙盐、镁盐、钾盐和钠盐。得自药学上可接受的有机无毒碱的盐,所述碱包括伯胺、仲胺和叔胺的盐,取代的胺包括天然存在的取代胺、环状胺及碱性离子交换树脂,例如精氨酸、甜菜碱、咖啡因、胆碱、N,N′-二苄基乙二胺、二乙胺、2-二乙基氨基乙醇、2-二甲基氨基乙醇、氨基乙醇、乙醇胺、乙二胺、N-乙基吗啉、N-乙基哌啶、葡萄糖胺、氨基葡萄糖、组氨酸、羟钴胺、异丙基胺、赖氨酸、甲基葡萄糖胺、吗啉、哌嗪,哌啶、呱咤、多胺树脂、普鲁卡因、嘌呤、可可碱、三乙胺、三甲胺、三丙胺、氨基丁三醇等。
与现有技术相比,本发明具有以下有益效果:
(1)本发明所述的吡唑并[1,5-a]吡啶类化合物具有良好的体外抗结核病菌活性,化合物的最小抑菌浓度(MIC)低于0.1μg/mL,部分达到0.01μg/mL,对临床分选的耐多药结核菌(MDR-TB)菌株具有很强的抑制作用。体内实验中,本发明所述的吡唑并[1,5-a]吡啶类化合物在20mg/kg/d的剂量下,可有效清除小鼠体内H37Ra的感染量,是一类新型的抗结核化合物。
(2)本发明所述的吡唑并[1,5-a]吡啶类化合物具有良好的药代动力学性质和很低的毒副作用,具备很好的成药性,且体内药效明显,具有很好的开发前景。
附图说明
图1为所述化合物L298实时体内抗结核病菌活性测定结果;
图2为所述化合物L385实时体内抗结核病菌活性测定结果;
图3为所述化合物L372实时体内抗结核病菌活性测定结果;
图4为所述化合物L298脾脏内抗结核活病菌性测定结果。
具体实施方式
根据标准药学技术,本发明化合物可单独或在药用组合物中与药学上可接受的载体组合给予哺乳动物,优选人。可口服或皮下、肌注、腹膜内、静脉、直肠、局部给予化合物。
所述口服给予,例如片剂、含片、锭剂、水或油混悬液、可分散粉剂或颗粒剂、乳剂、硬胶囊剂或软胶囊剂、或糖浆剂或酏剂等;所述皮下、肌注、腹膜内、静脉给予,可制成无菌可注射的水溶液;所述直肠给予,可制成栓剂形式;所述局部给予,可制成乳膏、软膏剂、凝胶剂、溶液剂或混悬液等。
所述药学上可接受的载体可例如:惰性稀释剂例如碳酸钙、碳酸钠、乳糖、磷酸钙或磷酸钠;制粒剂(granulating)和崩解剂例如微晶纤维素、交联羧甲纤维素钠(sodium crosscarmellose)、玉米淀粉或海藻酸;粘合剂例如淀粉、明胶、聚乙烯吡咯烷酮或阿拉伯胶;及润滑剂例如硬脂酸镁、硬脂酸或滑石粉。片剂可不包衣或通过已知技术包衣从而掩盖药物的不良味道或延长在胃肠道中崩解和吸收且因而提供持续更长时间的药物效应。例如,可采用水溶性掩盖味道的原料例如羟丙基-甲基纤维素或羟丙纤维素,或采用延时原料例如乙基纤维素、醋酸丁酸纤维素。
以下结合附图和具体实施例对本发明所述吡唑并[1,5-a]吡啶类化合物及其应用作进一步的阐述。
实施例1:5-氯-2-乙基N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ170298)
Figure PCTCN2015086852-appb-000033
Figure PCTCN2015086852-appb-000034
步骤1:4-(4-(三氟甲氧基)苯基)-5,6-二氢吡啶-1(2H)-碳酸叔丁酯(A1)
Figure PCTCN2015086852-appb-000035
在干燥的500mL的圆底烧瓶中加入重蒸THF(100mL)和二异丙基胺(5mL,36mmol),在Ar保护和-78℃的条件下,滴加2.4M正丁基锂环己烷溶液(36mmol,15mL)。搅拌1h后,将溶于25mL无水THF的1-Boc-4-哌啶酮(36mmol,7.2g)滴加到反应体系中。反应1.5h后,将溶于5mL无水THF的N-苯基双(三氟甲烷磺酰)亚胺(39.6mmol,14.15g)滴加到烧瓶中,再将反应升至室温搅拌过夜。减压蒸除溶剂,得粗产品,将粗产品直接投下一步,无需进一步纯化。
将所述粗产品和4-三氟甲氧基苯硼酸(50mmol,10.3g)、四三苯基磷钯(0.21g,0.5%)、无水LiCl(108mmol,5.1g)、2N碳酸钠溶液(48ml)混合溶于125mlDME中,加热至85℃反应9小时。反应结束后,经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂。经柱层析得到A1,黄色油状物9.60g(77.64%)。
1H NMR(400MHz,CDCl3):δ7.38(d,J=7.6Hz,2H),7.17(d,J=8.0Hz,2H),6.03(s,1H),4.08(s,2H),3.64(t,J=5.2Hz,2H),2.50(m,2H),1.49(s,9H)。
MS(ESI),m/z:344(M++H+)。
步骤2:4-(4-(三氟甲氧基)苯基)哌啶(A2)
Figure PCTCN2015086852-appb-000036
将4-(4-(三氟甲氧基)苯基)-5,6-二氢吡啶-1(2H)-碳酸叔丁酯(A1,25.6mmol,8.8g)和10%Pd/C(1.32g)溶于130ml甲醇中,反应体系用氢气球置换三次后,室温下搅拌24h。经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂,得到黄色油状物。再将该油状物溶于无水DCM(150mL)中,冰浴条件下缓慢加入TFA(70mL),室温下搅拌过夜。减压旋干溶剂,用乙酸乙酯溶解,再加入碳酸氢钠饱和溶液至水相pH=8.0,分离有机相后,用乙酸乙酯萃取水相三次。合并有机相,减压旋干溶剂得到A2,棕色粘稠状固体6.3g(100%)。
MS(ESI),m/z:246(M++H+)。
步骤3:4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯甲腈(A3)
Figure PCTCN2015086852-appb-000037
4-(4-(三氟甲氧基)苯基)哌啶(A2,6.56mmol,1.61g)、4-氯苯甲腈(32.8mmol,4.5g)和无水碳酸钾(13.12mmol,1.8g)溶于40mLDMSO中,加热至150℃反应过夜。冷却至室温加入大量水,将析出的固体用不是漏斗过滤,将滤渣用水洗三次后收集溶于乙酸乙酯,用无水硫酸钠干燥。经柱层析分离得到A3,白色固体1.70g(74.6%)。
MS(ESI),m/z:347(M++H+)。
步骤4:(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)甲胺盐酸盐A4
Figure PCTCN2015086852-appb-000038
4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯甲腈(A3,4.9mmol,1.7g)溶于无水THF(110mL),冰浴条件下加入LAH(15.2mmol,0.58g),搅拌半小时后升温回流过夜。冷却反应,冰浴条件下缓慢加入大量水淬灭反应。经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂。加入过量的氯化氢乙醇溶液使其成盐,旋干溶剂得到A4,固体2.02g(97.2%)。
MS(ESI),m/z:351(M+)。
步骤5:2-乙基-5-((叔丁氧羰基)胺)-吡唑[1,5-a]吡啶-3-碳酸乙酯B1
Figure PCTCN2015086852-appb-000039
将4-(叔丁氧羰基氨基)吡啶(1.2g,6.2mmol)和2,4-二硝基羟胺(DNPH,1.23g,6.2mmol)溶于28mL乙腈中升温至40℃,反应18h后,旋干溶剂。再向反应体系中加入无水DMF(15mL)、2-戊炔酸乙酯(0.78g,6.2mmol)和无水K2CO3(1.71g,12.4mmol)。室温下搅拌过夜,TLC检测反应完全。加入大量水,用乙酸乙酯萃取三次,收集有机相用盐水洗三次后经柱层析分离得到产物B1,0.71g(34.2%)。
MS(ESI),m/z:334(M++H+)。
步骤6:2-乙基5-胺基-吡唑[1,5-a]吡啶-3-碳酸乙酯B2
Figure PCTCN2015086852-appb-000040
将2-乙基-5-((叔丁氧羰基)胺)-吡唑[1,5-a]吡啶-3-碳酸乙酯(B1,0.69g,2.07mmol)溶于28mL无水DCM中,滴加1.8mLTFA。室温搅拌过夜。旋干,加入NaHCO3饱和溶液至pH=7-8,乙酸乙酯萃取三次,有机相用无水硫酸钠干燥,收集旋干得到产物B2,0.36g(75%)。
MS(ESI),m/z:234(M++H+)。
步骤7:2-乙基-5-氯-吡唑[1,5-a]吡啶-3-碳酸乙酯B3
Figure PCTCN2015086852-appb-000041
将2-乙基-5-胺基-吡唑[1,5-a]吡啶-3-碳酸乙酯(B2,0.1g,0.43mmol)和CuCl(0.11g,1.1mmol)溶于浓盐酸(1.5mL)中,冰浴条件下缓慢滴加0.4M的NaNO2溶液(0.038g,0.56mmol)。半小时后,将反应体系加热至80℃,反应15min后,TLC检测反应完全。冷却 反应,加入1M NaOH溶液调pH至10,经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂,经柱层析分离得到产物B3,0.08g(73.8%)。
MS(ESI),m/z:253(M++H+)。
步骤8:2-乙基-5-氯-吡唑[1,5-a]吡啶-3-羧酸B4
Figure PCTCN2015086852-appb-000042
将2-乙基-5-氯-吡唑[1,5-a]吡啶-3-碳酸乙酯(B3,0.27g,1.07mmol)溶解于9mL乙醇和3mL水的混合溶剂中,加入NaOH(0.13g,3.2mmol),加热至回流过夜。冷却反应,旋干溶剂,加入1N HCl使体系pH=4.0,将析出的固体抽滤再真空干燥得到B4,固体0.29g(100%)。
MS(ESI),m/z:225(M++H+)。
步骤9:5-氯-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ170298)
Figure PCTCN2015086852-appb-000043
将2-乙基-5-氯-吡唑[1,5-a]吡啶-3-羧酸(B4,0.25g,1.1mmol)、EDCI·HCl(0.32g,1.65mmol)、HOBT(0.074g,0.55mmol)溶于DMF(20mL)中,再加入三乙胺(0.66g,6.6mmol)和化合物A4(0.55g,1.3mmol)。将反应升温至80℃过夜。冷却反应后加入大量水用乙酸乙酯萃取三次,收集有机相用盐水洗三次后经柱层析分离得到TJ170298,固体0.46g(74.7%)。
1H NMR(400MHz,DMSO-d6)δ8.72(d,J=7.6Hz,1H),8.17(t,J=6.0Hz,1H),7.90(s,1H),7.41(d,J=8.8Hz,1H),7.28(d,J=8.0Hz,2H),7.22(d,J=8.8Hz,2H),7.02(dd,J=7.2,2.4Hz,1H),6.95(d,J=8.8Hz,2H),4.39(d,J=6.0Hz,2H),3.78(m,2H,3.00(q,J=7.6Hz,2H),2.73(m,3H),1.86(m,2H),1.74(m,2H),1.24(t,J=7.6Hz,2H)。
MS(ESI),m/z:557(M++H+)。
实施例2:5-氯-2-乙基-N-(4-(三氟甲氧基)苯基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ170322)
Figure PCTCN2015086852-appb-000044
合成方法如实施例1中的步骤5-9。
1H NMR(400MHz,DMSO-d6):δ8.74(d,J=7.2Hz,1H),8.32(t,J=6.0Hz,1H),7.96(s,1H),7.71(d,J=8.4Hz,2H),7.57(d,J=8.0Hz,2H),7.04(d,J=7.2Hz,1H),4.57(d,J=5.6Hz,2H),3.02(q,J=7.4Hz,2H),1.25(t,J=7.4Hz,2H)。
MS(ESI),m/z:382(M++H+)。
实施例3:5-氯-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ170371)
Figure PCTCN2015086852-appb-000045
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ8.82(d,J=7.2Hz,1H),8.72(s1H),8.63(s,1H),8.24(s,1H),7.40(d,J=8.4Hz,2H),7.28(d,J=8.0Hz,2H),7.20(d,J=8.0Hz,2H),7.13(d,J=7.2Hz,2H),6.94(d,J=8.0Hz,2H),4.38(d,J=5.2Hz,2H),3.77(m,2H),2.72(m,3H),1.86(m,2H),1.74(m,2H)。
MS(ESI),m/z:529(M++H+)。
实施例4:5-氯-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ170372)
Figure PCTCN2015086852-appb-000046
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ8.70(d,J=6.0Hz,1H),8.08(t,J=4.4Hz,1H),7.94(s,1H),7.40(d,J=6.8Hz,2H),7.29(d,J=6.8Hz,2H),7.22(d,J=6.8Hz,2H),7.03(d,J=6.0Hz,1H),6.95(d,J=6.8Hz,2H),4.39(d,J4.4Hz,2H),3.78(m,2H),2.73(m,3H),2.56(s,3H),1.86(m,2H),1.74(m,2H)。
MS(ESI),m/z:543(M++H+)。
实施例5:5-氯-2-环丙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ170375)
Figure PCTCN2015086852-appb-000047
合成方法如实施例1。
1H NMR(500MHz,DMSO-d6):δ8.66(d,J=7.0Hz,1H),8.17(s,1H),7.90(s,1H),7.40(d,J=8.0Hz,2H),7.28(d,J=8.0Hz,2H),7.23(d,J=8.0Hz,2H),7.00(d,J=6.5Hz,1H),6.96(d,J=8.0Hz,2H),4.14(d,J=5.5Hz,2H),3.78(m,2H),2.73(m,3H),2.54(m,3H),1.86(m,2H),1.74(m,2H),1.02(m,2H),=0.96(s,2H)。
MS(ESI),m/z:569(M++H+)。
实施例6:2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ170381)
Figure PCTCN2015086852-appb-000048
步骤1:2-甲基吡唑[1,5-a]吡啶-3-碳酸乙酯
Figure PCTCN2015086852-appb-000049
将吡啶(0.36g,4.5mmol)和2,4-二硝基羟胺(DNPH,1.0g,5mmol)溶于25mL乙腈中升温至40℃,反应18h后,旋干溶剂。再向反应体系中加入无水DMF(25mL)、2-丁炔酸乙酯(0.75g,6.75mmol)和无水K2CO3(1.24g,9mmol)。室温下搅拌过夜,TLC检测反应完全。加入大量水,用乙酸乙酯萃取三次,收集有机相用盐水洗三次后经柱层析分离得到固体0.53g(57.27%)。
1H NMR(400MHz,CDCl3):δ8.46(d,J=6.8Hz,1H),8.10(d,J=8.8Hz,1H),7.38(t,J=7.8Hz,1H),6.90(t,J=6.6Hz,2H),4.39(q,J=6.8Hz,2H),2.69(s,3H),1.43(t,J=7.0Hz,3H)。
MS(ESI),m/z:205(M++H+)。
步骤2:2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ170381)
合成方法如实施例1中的步骤8和9。
1H NMR(400MHz,DMSO-d6):δ8.64(d,J=6.8Hz,1H),8.01(s,1H),7.90(d,J=8.8Hz,1H),7.40(m,3H),7.28(d,J=8.0Hz,2H),7.23(d,J=8.0Hz,2H),6.95(m,3H),4.40(d,J=5.2Hz,2H),3.78(d,J=11.6Hz,2H),2.73(m,3H),2.56(s,3H),1.86(m,2H),1.74(m,2H)。
MS(ESI),m/z:509(M++H+)。
实施例7:2,5-二甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ170385)
Figure PCTCN2015086852-appb-000051
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.52(d,J=7.2Hz,1H),7.90(t,J=6.0Hz,1H),7.71(s,1H),7.40(d,J=8.4Hz,2H),7.28(d,J=8.0Hz,2H),7.22(d,J=8.4Hz,2H),6.95(d,J=8.4Hz,2H),6.80(d,J=6.8Hz, 1H),4.39(d,J=5.6Hz,2H),3.78(m,2H),2.73(m,3H),2.53(s,3H),2.37(s,3H),1.86(m,2H),1.75(m,2H)。
MS(ESI),m/z:523(M++H+)。
实施例8:5-氯-2-苯基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ170386)
Figure PCTCN2015086852-appb-000052
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.32(s,1H),7.42(m,3H),7.41(m,5H),7.29(d,J=8.0Hz,2H),7.18(d,J=8.0Hz,1H),7.10(d,J=6.4Hz,1H),6.96(d,J=8.4Hz,2H),4.35(d,J=5.2Hz,2H),3.80(m,2H),2.75(m,3H),1.87(m,2H),1.76(m,2H)。
MS(ESI),m/z:605(M++H+)。
实施例9:4-((5-氯-2-乙基吡唑[1,5-a]吡啶-3-甲酰胺)甲基)苯甲酸甲酯(TJ064814)
Figure PCTCN2015086852-appb-000053
合成方法如实施例1中步骤5-9。
1H NMR(400MHz,DMSO-d6):δ8.75(d,J=7.6Hz,1H),8.33(t,J=5.4Hz,1H),7.94(m,3H),7.48(d,J=7.6Hz,2H),7.03(d,J=7.2Hz,1H),4.56(d,J=5.6Hz,2H),3.84(s,3H),3.01(q,J=7.6Hz,2H),1.24(t,J=7.2Hz,2H)。
MS(ESI),m/z:372(M++H+)。
实施例10:5-氯-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064819)
Figure PCTCN2015086852-appb-000054
步骤1:4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯胺
Figure PCTCN2015086852-appb-000055
将中间体A2(0.5g,2mmol)溶于DMSO(12mL)中,加入无水K2CO3(0.55g,4mmol)和对氟硝基苯(0.56g,4mmol)。升温至130℃,反应过夜。冷却至室温,加入大量水,将析出的固体用布氏漏斗抽滤,水洗三次后将滤渣溶于乙酸乙酯。水洗三次,有机相用无水硫酸钠干燥后经柱层析纯化得到中间体1-(4-硝基苯)-4-(4-(三氟甲氧基)苯基)哌啶0.68g(92.0%)。
将得到的中间体1-(4-硝基苯)-4-(4-(三氟甲氧基)苯基)哌啶(0.68g,1.86mmol)溶于MeOH (30mL),加入催化量的Pd/C。氢气置换三次后,室温搅拌7h。反应完全后,经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂,经柱层析分离得到4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯胺0.44g(70.9%)。
MS(ESI),m/z:337(M++H+)。
步骤2:5-氯-2-乙基N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064819)
Figure PCTCN2015086852-appb-000056
将中间体B4(0.15g,0.67mmol)溶于DMF(10mL)中,加入HATU(0.38g,1.0mmol)和DIEA(0.26g,2.0mmol)。室温搅拌半小时后,加入中间体4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯胺(0.27g,0.8mmol)。室温搅拌过夜,TLC检测反应完全后,加入大量水。将析出的固体用布氏漏斗抽滤,水洗三次后用将滤渣溶于乙酸乙酯。盐水洗三次后,减压旋干溶剂,经柱层析分离得到产物TJ064819,94mg(26.0%)。
1H NMR(400MHz,DMSO-d6):δ9.64(s,1H),8.77(d,J=7.2Hz,1H),7.84(s,1H),7.54(d,J=8.8Hz,2H),7.43(d,J=8.4Hz,2H),7.30(d,J=8.0Hz,2H),7.05(d,J=7.2Hz,1H),6.98(d,J=8.8Hz,2H),3.76(m,2H),3.02(q,J=7.6Hz,2H),2.73(m,3H),1.88(m,2H),1.78(m,2H),1.27(t,J=7.6Hz,3H)。
MS(ESI),m/z:543(M++H+)。
实施例11:2,7-二甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064851)
Figure PCTCN2015086852-appb-000057
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ7.98(s,1H),7.82(d,J=8.8Hz,1H)7.40(d,J=8.4Hz,2H),7.31(m,3H),7.23(d,J=8.4Hz,2H),6.95(d,J=8.4Hz,2H),6.88(d,J=6.8Hz,1H),4.40(d,J=6.0Hz,2H),3.78(m,2H),2.73(m,3H),2.66(s,3H),2.60(s,3H),1.86(m,2H),1.73(m,2H)。
MS(ESI),m/z:523(M++H+)。
实施例12:2,6-二甲基N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064854)
Figure PCTCN2015086852-appb-000058
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.48(s,1H),7.93(s,1H),7.83(d,J=8.8Hz,1H),7.40(d,J=8.0Hz,2H),7.25(m,5H),6.95(d,J=8.0Hz,2H),4.39(d,J=4.8Hz,2H),3.78(d,J=11.2.0Hz,2H),2.73(m,3H),2.54(s,3H),2.30(s,3H),1.86(m,2H),1.76(m,2H)。
MS(ESI),m/z:523(M++H+)。
实施例13:2,4-二甲基N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3- 甲酰胺(TJ064872)
Figure PCTCN2015086852-appb-000059
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.57(s,1H),8.44(d,J=7.2Hz,1H),7.41(d,J=8.4Hz,2H),7.29(d,J=8.4Hz,2H),7.22(d,J=8.4Hz,2H),7.03(d,J=6.8Hz,1H),6.96(d,J=8.4Hz,2H),6.80(t,J=7.0Hz,2H),4.38(d,J=6.0Hz,2H),3.80(m,2H),2.74(m,3H),2.39(s,3H),2.37(s,3H),1.87(m,2H),1.76(m,2H)。
MS(ESI),m/z:523(M++H+)。
实施例14:5-甲氧基-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064889)
Figure PCTCN2015086852-appb-000060
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.51(d,J=7.6Hz,1H),7.90(t,J=6.0Hz,1H),7.41(d,J=8.8Hz,2H),7.28(d,J=8.4Hz,2H),7.22(d,J=8.8Hz,2H),7.19(d,J=2.8Hz,1H),6.95(d,J=8.8Hz,2H),6.62(dd,J=7.4,2.6Hz,1H),4.40(d,J=5.6Hz,2H),3.85(s,3H),3.78(m,2H),2.94(q,J=7.6Hz,2H),2.72(m,3H),1.86(m,2H),1.75(m,2H),1.23(t,J=7.6Hz,3H)。
MS(ESI),m/z:553(M++H+)。
实施例15:5-氯-2-乙基-N-((1-(4-(三氟甲氧基)苯基)-1H-1,2,3-三氮唑-4-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064983)
Figure PCTCN2015086852-appb-000061
步骤1:(1-(4-(三氟甲氧基)苯基)-1H-1,2,3-三氮唑-4-取代)甲胺
Figure PCTCN2015086852-appb-000062
将4-三氟甲氧基苯胺(1.3g,7.34mmol)溶于水(32mL)和浓盐酸(1.25mL)的混合溶液中。在冰浴条件下,向体系中缓慢滴加NaNO2水溶液(0.92M,7.36mmol),搅拌10min后,缓慢滴加NaN3水溶液(1.1M,8mL)。冰浴条件下继续搅拌45min后,升温至室温继续搅拌1h。TLC检测反应完全后用乙酸乙酯萃取三次,收集有机相用无水硫酸钠干燥,减压旋 干得到油状物,无需进一步纯化。
将上述油状物溶于四氢呋喃(27mL)中,加入炔丙胺(0.44g,8.07mmol)和水(13.5mL)。再向反应体系中加入现配置的抗坏血酸钠(30mg,0.15mmol)和Cu(OAc)2·H2O(14mg,0.075mmol)的水溶液(13.5mL)。室温条件下搅拌12h后加入盐水,用二氯甲烷萃取三次后,收集有机相经柱层析分离得到产物(1-(4-(三氟甲氧基)苯基)-1H-1,2,3-三氮唑-4-取代)甲胺,0.19g(两步总产率10.1%)。
MS(ESI),m/z:259(M++H+)。
步骤2:5-氯-2-乙基N-((1-(4-(三氟甲氧基)苯基)-1H-1,2,3-三氮唑-4-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064983)
Figure PCTCN2015086852-appb-000063
合成方法如实施例1中的步骤9,产率64.0%。
1H NMR(400MHz,DMSO-d6):δ8.73(m,2H),8.30(s,1H),8.06(d,J=9.2Hz,2H),7.99(s,1H),7.61(d,J=8.4Hz,2H),7.03(dd,J=7.6,2.0Hz,1H),4.62(d,J=5.2Hz,2H),3.02(q,J=7.6Hz,2H),1.26(t,J=7.4Hz,3H)。
MS(ESI),m/z:465(M++H+)。
实施例16:5-氯-2-乙基-N-甲基N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064985)
Figure PCTCN2015086852-appb-000064
将TJ170298(0.2g,0.34mmol)溶于无水DMF(7mL)中,冰浴条件下加入60%的NaH(0.02g,0.51mmol),搅拌0.5h后,加入CH3I(0.063g,0.44mmol)。反应移至室温,继续搅拌1h后,缓慢加入水淬灭反应。用乙酸乙酯萃取三次后,收集有机相,减压旋干溶剂,经柱层析分离得到产物TJ064985,0.18g(92.2%)。
1H NMR(400MHz,DMSO-d6):δ8.72(d,J=7.2Hz,1H),7.58(s,1H),7.40(d,J=8.8Hz,2H),7.29(d,J=8.4Hz,2H),7.09(m,2H),6.97(m,3H),4.52(s,2H),3.80(m,2H),2.83(s,3H),2.75(m,5H),1.86(m,2H),1.73(m,2H),1.25(t,J=7.6Hz,2H)。
MS(ESI),m/z:571(M++H+)。
实施例17:5-氯-2-乙基-N-(2-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)丙烷-2-取代)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064987)
Figure PCTCN2015086852-appb-000065
步骤1:2-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)丙烷-2-胺盐酸盐
Figure PCTCN2015086852-appb-000066
将中间体A3(1.5g,4.33mmol)溶于经CaH2预处理的干燥乙醚(75mL)中,Ar置换三次后,冰浴条件下缓慢滴加MeMgCl(8.7mL,26mmol,3M in THF),搅拌0.5h后,室温条件下滴加Ti(OPr-i)4(1.23g,4.33mmol),滴加完毕,升温至回流过夜。冷却反应,加入10%NaOH(4.5mmol,18mL),继续室温搅拌20min后,经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂,经柱层析分离得到产物。将产物溶于乙醇中,加入过量HCl的乙醇溶液,旋干溶剂得到2-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)丙烷-2-胺盐酸盐0.87g(44.6%)。
MS(ESI),m/z:379(M+)。
步骤2:5-氯-2-乙基-N-(2-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)丙烷-2-取代)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064987)
Figure PCTCN2015086852-appb-000067
合成方法如实施例1中的步骤9,产率59.8%。
1H NMR(400MHz,DMSO-d6):δ8.72(d,J=7.2Hz,1H),7.81(s,1H),7.78(d,J=2.0Hz,1H),7.42(d,J=8.8Hz,2H),7.28(m,4H),7.00(dd,J=7.4,2.2Hz,1H),6.92(d,J=8.8Hz,2H),3.77(m,2H),2.96(q,J=7.6Hz,2H),2.73(m,2H),1.88(m,2H),1.75(m,2H),1.66(s,6H),1.25(t,J=7.6Hz,3H)。
MS(ESI),m/z:585(M++H+)。
实施例18:5-三氟甲基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064995)
Figure PCTCN2015086852-appb-000068
步骤1:5-三氟甲基2-甲基-吡唑[1,5-a]吡啶-3-碳酸乙酯
Figure PCTCN2015086852-appb-000069
将2-[(氨基氧基)磺酰]-1,3,5-三甲基苯(1.63g,7.56mmol)溶于无水DCM(25mL)中,冰浴条件下将其缓慢滴加至4-三氟甲基吡啶(1.11g,7.56mmol)的无水DCM(25mL)溶液中,室温搅拌2h后,减压旋干溶剂。加入20mL无水DMF(20mL),并加入2-丁炔酸乙酯(0.84g,7.56mmol)和无水K2CO3(2.1g15.12mmol)。室温搅拌过夜,TLC检测反应完全。加入大量水,用乙酸乙酯萃取三次,收集有机相用盐水洗三次后经柱层析分离得到5-三氟甲基2-甲基-吡唑[1,5-a]吡啶-3-碳酸乙酯,固体0.94g(45.44%)。
MS(ESI),m/z:273(M++H+)。
步骤2:5-三氟甲基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ064995)
Figure PCTCN2015086852-appb-000070
合成方法如实施例1中的步骤8和9,两步总产率85.1%。
1H NMR(400MHz,DMSO-d6):δ8.89(d,J=6.8Hz,1H),8.29(t,J=5.8Hz,1H),8.24(s,1H),7.40(d,J=8.4Hz,2H),7.28(d,J=8.4Hz,2H),7.23(m,3H),6.96(d,J=8.4Hz,2H),4.42(d,J=5.6Hz,2H),3.78(m,2H),2.73(m,3H),2.61(s,3H),1.86(m,2H),1.74(m,2H)。
MS(ESI),m/z:577(M++H+)。
实施例19:2,5-二甲基-N-((1-(4-(三氟甲氧基)苯基)-1H-1,2,3-三氮唑-4-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830003)
Figure PCTCN2015086852-appb-000071
合成方法如实施例15。
1H NMR(400MHz,DMSO-d6):δ8.71(s,1H),8.53(d,J=6.8Hz,1H),8.06(d,J=9.2Hz,2H),8.01(t,J=5.6Hz,1H),7.76(s,1H),7.60(d,J=8.4Hz,2H),6.81(d,J=6.8Hz,1H),4.23(d,J=5.6Hz,2H),2.56(s,3H),2.38(s,3H)。
MS(ESI),m/z:431(M++H+)。
实施例20:5-溴-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830008)
Figure PCTCN2015086852-appb-000072
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ8.62(d,J=7.2Hz,1H),8.10(s,2H),8.08(t,J=6.2Hz,1H),7.41(d,J=8.8Hz,2H),7.28(d,J=8.4Hz,2H),7.22(d,J=8.4Hz,2H),7.11(dd,J=7.2,2.0Hz,1H),6.95(d,J=8.8Hz,2H),4.40(d,J=5.6Hz,2H),3.79(m,2H),2.73(m,3H),2.56(s,3H),1.86(m,2H),1.75(m,2H)。
MS(ESI),m/z:587(M++H+)。
实施例21:5-苯基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830012)
Figure PCTCN2015086852-appb-000073
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.72(d,J=7.2Hz,1H),8.13(m,2H),7.80(d,,J=7.6Hz,2H),7.53(t,J=7.6Hz,2H),7.45(m,1H),7.40(d,J=8.4Hz,2H),7.28(m,5H),6.96(d,J=8.4Hz,2H),4.43(d,J=6.0Hz,2H),3.78(m,2H),2.73(m,3H),2.58(s,3H),1.86(m,2H),1.74(m,2H)。
MS(ESI),mJz:585(M++H+)。
实施例22:5-氯-2-乙基-N-((6-(4-(三氟甲氧基)苯基)吡啶-3-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830028)
Figure PCTCN2015086852-appb-000074
步骤1:6-(4-(三氟甲氧基)苯基)-3-氰基吡啶
Figure PCTCN2015086852-appb-000075
将2-氯-5-氰基吡啶(5.0g,36.1mmol)和4-三氟甲氧基苯硼酸(11.12g,54.0mmol)溶于甲苯(100mL),加入四三苯基磷钯(0.5g,0.43mmol)和NaCO3(2M,36mL)。氩气置换三次后,升温至110℃,反应过夜后,冷却至室温。经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂,经柱层析分离得到产物6-(4-(三氟甲氧基)苯基)-3-氰基吡啶,7.62g(79.9%)。
MS(ESI),m/z:265(M++H+)。
步骤2:(6-(4-(三氟甲氧基)苯基)吡啶-3取代)甲胺盐酸盐
Figure PCTCN2015086852-appb-000076
将6-(4-(三氟甲氧基)苯基)-3-氰基吡啶(2.0g,7.57mmol)溶于甲醇(100mL)中,加入催化量雷尼镍。氢气球置换三次后,室温搅拌3.5h。反应结束后,经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂,经柱层析分离得到产物。将产物溶于乙醇,加入过量的HC1的乙醇溶液使其成盐,减压旋干溶剂得到(6-(4-(三氟甲氧基)苯基)吡啶-3取代)甲胺盐酸盐,固体1.5g(58.1%)。
MS(ESI),m/z:269(M+)。
步骤3:5-氯-2-乙基-N-((6-(4-(三氟甲氧基)苯基)吡啶-3-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830028)
Figure PCTCN2015086852-appb-000077
合成方法如实施例1中的步骤9,产率57.9%。
1H NMR(400MHz,DMSO-d6):δ8.74(dJ=7.6Hz,1H),8.68(s,1H),8.32(t,J=5.8Hz,1H),8.20(d,J=8.8Hz,2H),7.98(m,2H),7.88(dd,J=8.2,1.8Hz,1H),7.47(d,J=8.4Hz,2H),7.04(dd,J=7.2,2.0Hz,1H ),4.55(d,J=6.0Hz,2H),3.02(q,J=7.6Hz,2H),1.25(t,J=7.6Hz,3H)。
MS(ESI),m/z:475(M++H+)。
实施例23:2,5-二甲基-N-((6-(4-(三氟甲氧基)苯基)吡啶-3-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830025)
Figure PCTCN2015086852-appb-000078
合成方法如实施例22。
1H NMR(400MHz,DMSO-d6):δ8.68(s,1H),8.53(d,J=6.8Hz,1H),8.20(d,J=8.8Hz,2H),8.08(t,J=5.8Hz,1H),7.98(d,J=8.4Hz,2H),7.88(dd,J=8.2,2.2Hz,1H),7.74(s,1H),7.47(d,J=8.0Hz,2H),6.82(dd,J=7.2,1.6Hz,1H),4.55(d,J=5.6Hz,2H),2.56(s,3H),2.39(s,3H)。
MS(ESI),m/z:441(M++H+)。
实施例24:5-甲氧基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830047)
Figure PCTCN2015086852-appb-000079
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.49(d,J=7.2Hz,1H),7.82(t,J=6.0Hz,1H),7.41(d,J=8.4Hz,1H),7.29(d,J=8.0Hz,2H),7.24(m,3H),6.95(d,J=8.8Hz,2H),6.64(dd,J=7.2,2.4Hz,1H),4.40(d,J=6.0Hz,2H),3.85(s,3H),3.78(m,2H),2.73(m,3H),2.52(s,3H),1.86(m,2H),1.75(m,2H)。
MS(ESI),m/z:539(M++H+)。
实施例25:2-甲基-N-(4-(三氟甲氧基)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830069)
Figure PCTCN2015086852-appb-000080
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.66(d,J=6.8Hz,1H),8.18(t,J=5.8Hz,1H),7.94(d,J=8.8Hz,1H),7.70(d,J=8.4Hz,2H),7.58(d,J=8.0Hz,2H),7.40(m,1H),6.97(m,1H),4.57(d,J=5.6Hz,2H),2.58(s,3H)。
MS(ESI),m/z:334(M++H+)。
实施例26:5-氯-2-乙基-N-(1-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)乙醇)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830070)
Figure PCTCN2015086852-appb-000081
步骤1:1-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)乙酮
Figure PCTCN2015086852-appb-000082
室温下,将A2(1g,4.08mmol)和4-氟苯乙酮(1.69g,12.23mmol)溶于DMSO(25mL)中,加入DIPEA(1.56g,12.23mmol)。加热反应体系至150℃,过夜。冷却反应至室温,加入大量水,乙酸乙酯萃取三次后,收集有机相。经柱层析得到产物0.40g(27%)。
MS(ESI),m/z:364(M++H+)。
步骤2:1-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)乙醇
Figure PCTCN2015086852-appb-000083
将1-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)乙酮(0.4g,1.1mmol)溶于无水乙醇(10mL)中,缓慢分批次加入NaBH4(46g,1.21mmol)。室温搅拌过夜后,向反应体系缓慢加入冰水,搅拌10min后,乙酸乙酯萃取三次后,收集有机相。减压旋蒸溶剂得到产物0.40g(99.2%)。
MS(ESI),m/z:366(M++H+)。
步骤3:1-(4-(1-叠氮乙基)苯基-4-(4-(三氟甲氧基)苯基)哌啶
Figure PCTCN2015086852-appb-000084
将1-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)乙醇(0.4g,1.1mmol)溶于无水THF(10mL)中,氩气置换三次。在0℃条件下加入DPPA(0.6g,2.19mmol),搅拌5min后再缓慢滴加DBU(0.33g,2.19mmol)。升温至室温反应过夜后,加入大量水,乙酸乙酯萃取三次后,收集有机相。经柱层析得到产物0.32g(75.0%)。
MS(ESI),m/z:391(M++H+)。
步骤4:1-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)乙胺
Figure PCTCN2015086852-appb-000085
将1-(4-(1-叠氮乙基)苯基-4-(4-(三氟甲氧基)苯基)哌啶(0.32g,0.82mmol)溶于甲苯(8mL)和H2O(0.41mL,0.41mmol)的混合体系中,再加入三苯基膦(0.43g,1.64mmol)。室温条件下反应6h后,加入乙酸乙酯。向有机相加入1N的盐酸水溶液后收集水相,并用NaOH水溶液调pH至10,然后用DCM萃取三次,收集有机相,经柱层析得到产物0.22g(72.9%)。
MS(ESI),m/z:365(M++H+)。
步骤5:5-氯-2-乙基-N-(1-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)乙醇)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830070)
Figure PCTCN2015086852-appb-000086
合成方法如实施例1中的步骤9,产率51.1%。
1H NMR(400MHz,DMSO-d6):δ8.72(d,J=7.2Hz,1H),8.09(d,J=8.0Hz,1H),7.76(d,J=2.0Hz,1H),7.60(d,J=8.4Hz,0.5H),7.41(m,1.5H),7.34(d,J=8.0Hz,0.5H),7.28(m,3.5H),6.99(m,3H),5.10(m,1H),3.84(m,2H),3.45(m,0.5H),2.97(q,J=7.6Hz,2H),2.73(m,2H),2.70(m,0.5H),1.87(m,1.5H),1.76(m,1.5H),1.46(d,J=7.2Hz,3H),1.22(d,J=7.6Hz,3H)。
MS(ESI),m/z:571(M++H+)。
实施例27:2,5-二甲基-N-(4-(三氟甲氧基)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830072)
Figure PCTCN2015086852-appb-000087
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.54(d,J=6.8Hz,1H),8.07(t,J=5.8Hz,1H),7.74(s,1H),7.70(d,J=8.4Hz,2H),7.57(d,J=8.0Hz,2H),4.57(d,J=6.0Hz,2H),2.56(s,3H),2.38(s,3H)。
MS(ESI),m/z:348(M++H+)。
实施例28:5-异丙基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830073)
Figure PCTCN2015086852-appb-000088
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.54(d,J=7.2Hz,1H),7.93(t,J=6.0Hz,1H),7.71(s,1H),7.41(d,J=8.8Hz,2H),7.28(d,J=8.4Hz,2H),7.23(d,J=8.4Hz,2H),6.93(d,J=8.4Hz,2H),6.90(d,J=7.2Hz,1H),4.40(d,J=6.0Hz,2H),3.78(m,2H),2.96(m,1H),2.73(m,3H),2.54(s,3H),1.86(m,2H),1.75(m,2H),1.25(s,3H),1.23(s,3H)。
MS(ESI),m/z:551(M++H+)。
实施例29:5-叔丁基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830075)
Figure PCTCN2015086852-appb-000089
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.53(d,J=7.2Hz,1H),7.99(t,J=5.6Hz,1H),7.76(s,1H),7.41(d,J=8.4Hz,2H),7.28(d,J=8.0Hz,2H),7.23(d,J=8.4Hz,2H),7.04(d,J=6.0Hz,1H),6.95(d,J=8.4Hz,2H),4.40(d,J=5.6Hz,2H),3.78(m,2H),2.73(m,3H),2.54(s,3H),1.86(m,2H),1.75(m,2H),1.31(s,9H)。
MS(ESI),m/z:565(M++H+)。
实施例30:5-乙基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830082)
Figure PCTCN2015086852-appb-000090
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.54(d,J=7.2Hz,1H),7.92(t,J=5.8Hz,1H),7.71(s,1H),7.40(d,J=8.8Hz,2H),7.28(d,J=8.4Hz,2H),7.23(d,J=8.8Hz,2H),6.95(d,J=8.4Hz,2H),6.84(dd,J=7.2,1.6Hz,1H),4.40(d,J=5.6Hz,2H),3.78(m,2H),2.70(m,5H),2.54(s,3H),1.86(m,2H),1.74(m,2H),1.(t,J=7.6Hz,3H)。
MS(ESI),m/z:537(M++H+)。
实施例31:5-甲基-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830102)
Figure PCTCN2015086852-appb-000091
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.54(d,J=7.2Hz,1H),7.98(t,J=5.8Hz,1H),7.67(s,1H),7.40(d,J=8.4Hz,2H),7.28(d,J=8.4Hz,2H),7.22(d,J=8.8Hz,2H),6.95(d,J=8.8Hz,2H),6.79(dd,J=7.2,2.0Hz,1H),4.39(d,J=5.6Hz,2H),3.78(m,2H),2.98(q,J=7.6Hz,2H),2.72(m,3H),2.37(s,3H),1.86(m,2H),1.74(m,2H),1.24(t,J=7.6Hz,3H)。
MS(ESI),m/z:537(M++H+)。
实施例32:5-甲基-2-乙氧基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830108)
Figure PCTCN2015086852-appb-000092
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.48(d,J=7.6Hz,1H),7.80(t,J=6.0Hz,1H),7.40(d,J=8.8Hz,2H),7.28(d,J=8.4Hz,2H),7.23(m,3H),6.95(d,J=8.4Hz,2H),6.61(dd,J=7.6,2.8Hz,1H),4.39(d,J=5.6Hz,2H) ,4.10(q,J=6.8Hz,2H),3.78(m,2H),2.72(m,3H),2.51(s,3H),1.86(m,2H),1.74(m,2H),1.37(t,J=6.8Hz,3H)。
MS(ESI),m/z:553(M++H+)。
实施例33:5-氯-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯乙基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830128)
Figure PCTCN2015086852-appb-000093
步骤1:2-(4-溴苯乙基)异吲哚啉-1,3-二酮
Figure PCTCN2015086852-appb-000094
将异吲哚啉-1,3-二酮(0.33g,2.27mmol),2-(4-溴苯基)乙醇(0.46g,2.27mmol)和三苯基膦(0.59g,2,27mmol)溶于无水THF(7.5mL)中,再缓慢滴加DIAD(0.46g,2.27mmol)。室温条件下搅拌3h,后减压旋干,经柱层析得到产物0.56g(产率75%)。
MS(ESI),m/z:330(M++H+)。
步骤2:2-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯乙基)异吲哚啉-1,3-二酮
Figure PCTCN2015086852-appb-000095
将2-(4-溴苯乙基)异吲哚啉-1,3-二酮(0.56g,1.7mmol)和A2(0.63g,2.55mmol)溶于甲苯(15mL)中,再加入Pd(dba)2(59mg,0.1mmol)、XPhos(97mg,0.2mmol)和碳酸铯(2.21g,6.8mmol)。反应体系用氩气置换三次后,加热至110℃反应过夜。冷却反应至室温,经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂,经柱层析分离得到产物0.19g(26%)。
MS(ESI),m/z:495(M++H+)。
步骤3:2-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)乙胺
Figure PCTCN2015086852-appb-000096
将2-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯乙基)异吲哚啉-1,3-二酮(0.19g,0.38mmol)溶于乙醇(4mL)中,加入80%水合联氨(0.072mL)后加热回流(80℃)1.5h。 反应结束后,减压旋干溶剂,用乙酸乙酯溶解,盐水洗三次。向有机相加入过量的盐酸乙醇溶液,减压旋干后得到产物0.13g(78.7%)。
MS(ESI),m/z:365(M++H+)。
步骤4:5-氯-2-乙基N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯乙基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830128)
Figure PCTCN2015086852-appb-000097
合成方法如实施例1中的步骤9,产率63.8%。
1H NMR(400MHz,DMSO-d6):δ8.70(d,J=7.2Hz,1H),7.75(t,J=5.6Hz,1H),7.70(s,1H),7.41(d,J=8.4Hz,2H),7.29(d,J=8.0Hz,2H),7.12(d,J=8.4Hz,2H),6.61(dd,J=7.2,2.0Hz,1H),6.94(d,J=8.8Hz,2H),3.76(m,2H),3.46(q,J=6.8Hz,2H),2.93(q,J=7.6Hz,2H),2.74(m,5H),1.87(m,2H),1.75(m,2H),1.21(t,J=7.2Hz,3H)。
MS(ESI),m/z:571(M++H+)。
实施例34:2,5-二甲基-N-((5-(4-(三氟甲氧基)苯基)噻吩-2-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830132)
Figure PCTCN2015086852-appb-000098
步骤1:叔丁基(噻吩-2取代甲基)氨基甲酸酯
Figure PCTCN2015086852-appb-000099
将噻吩-2-取代甲胺(1.4g,12.36mmol)和碳酸氢钠(1.04g,12.36mmol)溶于无水THF(20mL)中,缓慢滴加Boc2O(2.76g,13.6mmol)。室温反应3h后,减压旋干溶剂,加入乙酸乙酯萃取。收集有机相经柱层析得到产物2.61g(99.1%)。
MS(ESI),m/z:214(M++H+)。
步骤2:叔丁基((5-溴噻吩-2取代)甲基)氨基甲酸酯
Figure PCTCN2015086852-appb-000100
将叔丁基(噻吩-2取代甲基)氨基甲酸酯(2.22g,10.41mmol)溶于DMF(11mL)中,再加入NBS(2.04g,11.45mmol)。室温反应5h后,加入大量水,乙酸乙酯萃取三次后,收集有机相。经柱层析得到产物2.56g(84.3%)。
MS(ESI),m/z:292(M++H+)。
步骤3:叔丁基((5-(4-(三氟甲氧基)苯基)噻吩-2-取代)氨基甲酸酯
Figure PCTCN2015086852-appb-000101
将叔丁基((5-溴噻吩-2取代)甲基)氨基甲酸酯(1.0g,3.42mmol)、4-三氟甲氧基苯硼酸(0.84g,4.11mmol)、Pd(PPh3)4(0.2g,0.17mmol)和碳酸钾(2.66g,12.5mmol)溶于H2O(6.3mL)和DME(8mL)中,氩气置换三次后,在80℃下反应过夜。冷却反应至室温,经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂,经柱层析分离得到产物1.2g(93.7%)。
MS(ESI),m/z:374(M++H+)。
步骤4:(5-(4-(三氟甲氧基)苯基)噻吩-2-取代)甲胺
将叔丁基((5-(4-(三氟甲氧基)苯基)噻吩-2-取代)氨基甲酸酯(1.2g,3.2mmol)溶于无水DCM(40mL)中,缓慢滴加TFA(2.76mL)。室温反应3h后,减压旋干溶剂,用乙酸乙酯溶解,再加入碳酸氢钠饱和溶液至水相pH=8.0,分离有机相后,用乙酸乙酯萃取水相三次。合并有机相,减压旋干溶剂得到产物0.58g(66.5%)。
MS(ESI),m/z:274(M++H+)。
步骤5:2,5-二甲基-N-((5-(4-(三氟甲氧基)苯基)噻吩-2-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830132)
Figure PCTCN2015086852-appb-000103
合成方法如实施例1中的步骤9,产率75.9%。
1H NMR(400MHz,DMSO-d6):δ8.53(d,J=6.8Hz,1H),8.15(t,J=6.0Hz,1H),7.73(m,3H),7.39(m,3H),7.05(d,J=3.6Hz,1H),6.82(d,J=7.2,1H),4.64(d,J=5.6Hz,2H),2.55(s,3H),2.39(s,3H)。
MS(ESI),m/z:446(M++H+)。
实施例35:2,5-二甲基-N-((5-(4-(三氟甲氧基)苯基)吡啶-2-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830133)
Figure PCTCN2015086852-appb-000104
合成方法如实施22。
1H NMR(400MHz,DMSO-d6):δ8.86(s,1H),8.55(d,J=7.2Hz,1H),8.09(m,2H),7.86(d,J=8.8Hz,2H),7.80(s,1H),7.48(m,3H),6.83(dd,J=7.2,1.6Hz,1H),4.64(d,J=6.0Hz,2H),2.59(s,3H),2.39(s,3H)。
MS(ESI),m/z:441(M++H+)。
实施例36:5-甲基-2-丙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830134)
Figure PCTCN2015086852-appb-000105
合成方法如实施例1。
1H NMR(400MHz,DMSO-d6):δ8.72(d,J=7.2Hz,1H),8.20(t,J=6.0Hz,1H),7.88(d,J=2.0Hz,1H),7.40(d,J=8.4Hz,2H),7.28(d,J=8.4Hz,2H),7.21(d,J=8.4Hz,2H),7.01(dd,J=7.6,2.4Hz,1H),6.95(d,J=8.4Hz,2H),4.39(d,J=5.6Hz,2H),3.78(m,2H),2.95(t,J=7.6Hz,2H),2.73(m,3H),1.86(m,2H),1.72(m,5H),0.90(t,J=7.6Hz,3H)。
MS(ESI),m/z:571(M++H+)。
实施例37:N-(4-(叔丁基)苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺(TJ830135)
Figure PCTCN2015086852-appb-000106
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.52(d,J=6.8Hz,1H),7.96(t,J=6.0Hz,1H),7.72(s,1H),7.35(d,J=8.0Hz,2H),7.28(d,J=8.4Hz,2H),6.80(dd,J=6.8,1.6Hz,1H),4.45(d,J=6.0Hz,2H),2.54(s,3H),2.38(s,3H),1.27(s,9H)。
MS(ESI),m/z:336(M++H+)。
实施例38:N-(4-(二甲氨基)苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺(TJ830136)
Figure PCTCN2015086852-appb-000107
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.51(d,J=7.2Hz,1H),7.86(t,J=5.6Hz,1H),7.69(s,1H),7.19(d,J=8.4Hz,2H),6.79(d,J=6.0Hz,1H),6.70(d,J=8.4Hz,2H),4.36(d,J=6.0Hz,2H),2.85(s,6H),2.52(s,3H),2.37(s,3H)。
MS(ESI),m/z:323(M++H+)。
实施例39:N-(4-(4-(4-甲氧基苯基)哌啶-1-取代)苄基)-2,5-二甲基-吡唑[1,5-a]吡啶-3-甲酰胺(TJ830140)
Figure PCTCN2015086852-appb-000108
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.52(d,J=6.8Hz,1H),7.90(t,J=6.0Hz,1H),7.71(s,1H),7.20(m,4H),6.94(d,J=8.4Hz,2H),6.86(d,J=8.8Hz,2H),6.80(dd,J=6.8,1.2Hz,1H),4.39(d,J=5.6Hz,2H),3.76(m,2H),3.72(s,3H),2.71(m,2H),2.60(m,1H),2.53(s,3H),2.37(s,3H),1.82(m,2H),1.71(m,2H)。
MS(ESI),m/z:469(M++H+)。
实施例40:N-(4-(4-(4-氟苯基)哌啶-1-取代)苄基)-2,5-二甲基-吡唑[1,5-a]吡啶-3-甲酰胺(TJ830141)
Figure PCTCN2015086852-appb-000109
合成方法如实施例6。
1H NMR(400MHz,DMSO-d6):δ8.52(d,J=7.2Hz,1H),7.90(t,J=6.0Hz,1H),7.71(s,1H),7.30(m,2H),7.22(d,J=8.4Hz,2H),7.11(m,2H),6.94(d,J=8.8Hz,2H),6.80(d,J=7.2Hz,1H),4.39(d,J=5.6Hz,2H),3.77(m,2H),2.69(m,3H),2.53(s,3H),2.37(s,3H),1.84(m,2H),1.73(m,2H)。
MS(ESI),m/z:457(M++H+)。
实施例41:2,5-二甲基-N-(4-(哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830146)
Figure PCTCN2015086852-appb-000110
合成方法如实施例1和6。
1H NMR(400MHz,DMSO-d6):δ8.51(d,J=7.6Hz,1H),7.88(t,J=5.6Hz,1H),7.70(s,1H),7.19(d,J=8.4Hz,2H),6.88(d,J=8.8Hz,2H),6.79(dd,J=6.8,1.6Hz,1H),4.37(d,J=6.0Hz,2H),3.08(m,4H),2.53(s,3H),2.37(s,3H),1.60(m,4H),1.51(m,2H)。
MS(ESI),m/z:363(M++H+)。
实施例42:N-(4-(4-氟苯氧基)苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺(TJ830147)
Figure PCTCN2015086852-appb-000111
步骤1:4-(4-氟苯氧基)苯甲腈
Figure PCTCN2015086852-appb-000112
将4-氟苯甲腈(0.6g,5mmol)、4-氟苯酚(0.85g,7.5mmol)和叔丁醇钾(1.1g,10mmol)溶于DMSO(10mL)中,在微波反应器中加热至120℃。反应15min后,冷却至室温,加入大量水,用乙酸乙酯萃取三次后,收集有机相。经柱层析得到产物0.42g(39.3%)
MS(ESI),m/z:214(M++H+)。
步骤2:N-(4-(4-氟苯氧基)苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺(TJ830147)
Figure PCTCN2015086852-appb-000113
合成方法如实施例1中的步骤4和9,两步产率80.6%。
1H NMR(400MHz,DMSO-d6):δ8.52(d,J=6.8Hz,1H),7.99(t,J=6.0Hz,1H),7.72(s,1H),7.37(d,J=8.4Hz,2H),7.20(m,2H),7.03(m,2H),6.97(d,J=8.4Hz,2H),6.80(dd,J=7.2,1.6Hz,1H),4.46(d,J=6.0Hz,2H),2.54(s,3H),2.38(s,3H)。
MS(ESI),m/z:390(M++H+)。
实施例43:2,5-二甲基-N-(4-(4-(三氟甲基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830149)
Figure PCTCN2015086852-appb-000114
合成方法如实施例1和6。
1H NMR(400MHz,DMSO-d6):δ8.52(d,J=6.8Hz,1H),7.90(t,J=6.0Hz,1H),7.70(s,1H),7.21(d,J=8.4Hz,2H),6.92(d,J=8.4Hz,2H),6.79(dd,J=6.8,1.6Hz,1H),4.38(d,J=5.6Hz,2H),3.74(m,2H),2.68(m,2H),2.53(s,3H),2.44(m,1H),2.37(s,3H),1.86(m,2H),1.54(m,2H)。
MS(ESI),m/z:431(M++H+)。
实施例44:2,5-二甲基-N-((2-(4-(三氟甲氧基)苯基)嘧啶-5-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830150)
Figure PCTCN2015086852-appb-000115
步骤1:5-溴-2-(4-(三氟甲氧基)苯基)嘧啶
Figure PCTCN2015086852-appb-000116
将5-溴2-碘嘧啶(6.0g,21.1mmol)、4-三氟甲氧基苯硼酸(4.77g,23.2mmol)、Pd(PPh3)4(0.24g,0.21mmol)和碳酸钠(4.46g,42.12mmol)溶于H2O(60mL)和甲苯(480mL)的混合溶剂中,加热至回流过夜。冷却反应至室温,经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂,经柱层析分离得到产物5.6g(83.6%)。
MS(ESI),m/z:319(M++H+)。
步骤2:2-(4-(三氟甲氧基)苯基)嘧啶-5-甲醛
Figure PCTCN2015086852-appb-000117
将5-溴-2-(4-(三氟甲氧基)苯基)嘧啶(3.2g,9.72mmol)溶于无水THF(100mL)中,氩气置换三次后降温至-80℃,向反应体系中缓慢滴加2.4M正丁基锂(4.86mL,11.66mmol),搅拌5min后,再缓慢滴加无水DMF(12.4mL)。保持-80℃,反应20min后,升温至室温。向反应体系中缓慢加入大量水,减压旋蒸溶剂,再用乙酸乙酯萃取。经柱层析 得到产物0.38g(14.8%)。
MS(ESI),m/z:269(M++H+)。
步骤3:(2-(4-(三氟甲氧基)苯基)嘧啶-5-取代)甲胺
Figure PCTCN2015086852-appb-000118
合成方法如实施例26中的步骤2、3和4。
MS(ESI),m/z:270(M++H+)。
步骤:4:2,5-二甲基-N-((2-(4-(三氟甲氧基)苯基)嘧啶-5-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830150)
Figure PCTCN2015086852-appb-000119
合成方法如实施例1中的步骤9。
1H NMR(400MHz,DMSO-d6):δ8.92(s,1H),8.54(d,J=7.2Hz,1H),8.49(d,J=8.8Hz,2H),8.10(t,J=5.6Hz,2H),7.75(s,1H),7.51(d,J=8.4Hz,2H),6.82(dd,J=6.8,1.2Hz,1H),4.54(d,J=6.0Hz,2H),2.55(s,3H),2.39(s,3H)。
MS(ESI),m/z:442(M++H+)。
实施例45:2,5-二甲基-N-((4’-(三氟甲氧基)-[1,1’-联苯]-4-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830151)
Figure PCTCN2015086852-appb-000120
步骤1:(4’-(三氟甲氧基)-[1,1’-联苯]-4-取代)甲胺
Figure PCTCN2015086852-appb-000121
将(4-溴苯基)甲胺(0.56g,3.0mmol)、4-三氟甲氧基苯硼酸(0.68g,3.3mmol)、Pd(PPh3)4(0.17g,0.15mmol)和碳酸钾(1.66g,12mmol)溶于H2O(6mL)和乙腈(34mL)的混合溶剂中,加热至90℃过夜。冷却反应至室温,经2cm硅胶短柱过滤,滤渣用乙酸乙酯洗三次,减压旋干溶剂,经柱层析分离得到产物0.72g(89.6%)。
MS(ESI),m/z:268(M++H+)。
步骤2:2,5-二甲基-N-((4’-(三氟甲氧基)-[1,1’-联苯]-4-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830151)
Figure PCTCN2015086852-appb-000122
合成方法如实施例1中的步骤9,产率67.3%。
1H NMR(400MHz,DMSO-d6):δ8.53(d,J=7.2Hz,1H),78.04(t,J=5.6Hz,1H),7.77(m,3H),7.66(d,J=8.0Hz,2H),7.45(m,4H),6.81(d,J=7.2Hz,1H),4.53(d,J=6.0Hz,2H),2.56(s,3H),2.38(s,3H)。
MS(ESI),m/z:440(M++H+)。
实施例46:N-(4-(4-(4-氟苯基)哌嗪-1-取代)苄基)-2,5-二甲基-吡唑[1,5-a]吡啶-3-甲酰胺(TJ830152)
Figure PCTCN2015086852-appb-000123
合成方法如实施例1和6。
1H NMR(400MHz,DMSO-d6):δ8.52(d,J=6.8Hz,1H),7.91(t,J=5.6Hz,1H),7.71(s,1H),7.24(d,J=8.4Hz,2H),7.02(m,6H),6.80(d,J=6.8Hz,1H)4.40(d,J=5.6Hz,2H),3.22(m,8H),2.53(s,3H),2.37(s,3H)。
MS(ESI),m/z:458(M++H+)。
实施例47:2,5-二甲基-N-(4-(4-(三氟甲氧基)苯基)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830153)
Figure PCTCN2015086852-appb-000124
合成方法如实施例42。
1H NMR(400MHz,DMSO-d6):δ8.53(d,J=7.2Hz,1H),8.00(s,1H),7.73(s,1H),7.39(m,4H),7.06(m,4H),6.81(d,J=6.8Hz,1H),4.48(d,J=5.6Hz,2H),2.55(s,3H),2.38(s,3H)。
MS(ESI),m/z:456(M++H+)。
实施例48:2,5-二甲基-N-(4-(4-(4-(三氟甲氧基)苯氧基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830161)
Figure PCTCN2015086852-appb-000125
步骤1:叔丁基4-(4-(三氟甲氧基)苯氧基)哌啶-1-羧酸酯
Figure PCTCN2015086852-appb-000126
将叔丁基4-羟基哌啶-1-羧酸酯(1.08g,5.37mmol)、4-三氟甲氧基苯酚(1.05g,5.9mmol) 和PPh3(1.55g,5.9mL)溶于THF(50mL)中,再向反应体系缓慢滴加DIAD(1.19g,5.9mmol)。反应2h后,减压旋蒸溶剂,加入大量水,用乙酸乙酯萃取三次,合并有机相,经柱层析得到产物1.37g(70.4%)。
tert-butyl 4-hydroxypiperidine-1-carboxylate
MS(ESI),m/z:362(M++H+)。
步骤2:(4-(4-(4-(三氟甲氧基)苯氧基)哌啶-1-取代)苯基)甲胺二盐酸盐
Figure PCTCN2015086852-appb-000127
合成方法如实施例1中的步骤2,3和4。
MS(ESI),m/z:367(M++H+)。
步骤3:2,5-二甲基-N-(4-(4-(4-(三氟甲氧基)苯氧基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ830161)
Figure PCTCN2015086852-appb-000128
合成方法如实施例1中的步骤9,产率68.0%。
1H NMR(400MHz,DMSO-d6):δ8.51(d,J=7.2Hz,1H),7.90(t,J=6.0Hz,1H),7.70(s,1H),7.27(d,J=8.4Hz,2H),7.21(d,J=8.4Hz,2H),7.08(d,J=9.2Hz,2H),6.93(d,J=8.8Hz,2H),6.79(dd,J=7.2,1.2Hz,1H),4.57(m,1H),4.38(d,J=6.0Hz,2H),3.48(m,2H),3.02(m,2H),2.53(s,3H),2.37(s,3H),2.03(m,2H),1.71(m,2H)。
MS(ESI),m/z:539(M++H+)。
实施例49:5-氯-2-乙基-N-(4-氟苄基)吡唑[1,5-a]吡啶-3-甲酰胺(TJ4570004)
Figure PCTCN2015086852-appb-000129
合成方法如实施例1。
1HNMR(400MHz,DMSO-d6):δ8.73(d,J=7.2Hz,1H),8.25(s,1H),7.92(s,1H),7.39(m,2H),7.16(m,2H),7.02(dd,J=7.2,1.6Hz,1H),4.46(d,J=6.0Hz,2H),3.00(q,J=7.2Hz,2H),1.24(t,J=7.6Hz,3H)。
MS(ESI),m/z:332(M++H+)。
实施例50:N-(4-氟苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺(TJ4570006)
Figure PCTCN2015086852-appb-000130
合成方法如实施例6。
1HNMR(400MHz,DMSO-d6):δ8.52(d,J=6.8Hz,1H),8.00(t,J=6.0Hz,1H),7.72(s,1H),7.39(m,2H),7.15(m,2H),6.80(d,J=6.8Hz,1H),4.46(d,J=6.0Hz,2H),2.54(s,3H),2.38(s,3H)。
MS(ESI),m/z:298(M++H+)。
实施例51:对自主发光结核菌的抑制活性。
1.自主发光结核菌的准备:
1.1培养发光菌
将-80℃冻存的无抗性筛选标记的稳定的原代发光菌以2mL接种至含有50mL7H9(含有0.1%土温80)培养基的锥形瓶中,培养至OD值达到0.3-1.0之间。
注:检测OD600值注意事项:将菌液加入检测杯后,用封口膜封住杯口,上下颠倒轻柔摇匀后立即检测,重复三次后取平均值。另外,在1.5mL EP管中检测荧光值,荧光值约在10-100万/mL以上为宜。
1.2获得检测用菌液并置于384孔板
将菌液以原液、稀释10、100及1000倍(稀释方法:取上一浓度100μL加入900μL 7H9培养基中为下一浓度,充分吹吸后继续依次稀释)。取荧光值达到2000-10000/25μL的稀释倍数稀释的菌液为检测用菌液。
将稀释好的菌液用排枪加入到384孔板中,每孔25μL,将384孔板放入37℃孵箱内孵育约1-2小时后,检测各孔的荧光值,并将该荧光值的平均值纪录为Day 0荧光值。
2.配制化合物溶液并置于384孔板保存
各检测样品统一用DMSO配置为10mg/mL的母液,并进行3倍梯度稀释,阳性药利福平(RIF)和异烟肼(INH)用DMSO配置两个浓度,2mg/mL和1mg/mL。将化合物加入到384孔板中。在384孔板中,每孔5%μL检测样品,-20℃保存。
3.加药
使用Echo520超声微量移液系统进行加药,检测样品每孔打样25nL(在总体积为0.025ml的7H9培养基中进行操作),形成以化合物终浓度从10μg/mL开始的3倍梯度稀释,终浓度分别为10mg/mL,3mg/mL,1mg/mL,0.3mg/mL,0.1mg/mL,0.03mg/mL,0.01mg/mL,0.003mg/mL,0.001mg/mL,0.0003mg/mL,0.0001mg/mL,0.00003mg/mL,0.00001mg/mL,0.000003mg/mL,0.000001mg/mL。阳性药分别从2mg/mL的母液孔加样125nL和12.5nL,及从1mg/mL的母液孔2.5nL到终体积0.025nL的培养基中,最后阳性药终浓度分别为10μg/mL,1μg/mL,0.1μg/mL。DMSO作为对照。在振荡器上震荡5min,继续培养。
4.检测及数据分析
37℃孵箱孵育,分别于24小时、48小时、72小时用envision酶标仪检测荧光值。以荧光值随时间逐渐降低的样品为有活性,其中,RLU为荧光值的相对光单位。结果如表1所示。
表1.化合物对自主发光结核菌的MIClux
化合物编号 MIClux   化合物编号 MIClux
TJ170298 A   TJ170322 A
TJ170371 B   TJ170372 A
TJ170375 A   TJ170381 A
TJ170385 A   TJ170386 C
TJ064814 B   TJ064819 C
TJ064851 B   TJ064854 A
TJ064872 C   TJ064889 A
TJ064983 C   TJ064985 B
TJ064987 C   TJ064995 B
TJ830003 C   TJ830008 A
TJ830012 C   TJ830028 A
TJ830025 A   TJ830047 A
TJ830069 B   TJ830070 C
TJ830072 A   TJ830073 B
TJ830075 C   TJ830082 A
TJ830102 A   TJ830108 A
TJ830128 B   TJ830132 A
TJ830133 A   TJ830134 A
TJ830135 A   TJ830136 A
TJ830140 A   TJ830141 A
TJ830146 A   TJ830147 A
TJ830149 A   TJ830150 A
TJ830151 A   TJ830152 A
TJ830153 A   TJ830161 A
TJ4570004 C   TJ4570006 B
活性范围:A表示<1μg/mL,B表示1-10μg/mL,C表示>10μg/mL。
实施例52:平板法测定本发明所述化合物的结核分枝杆菌MIC(最低抑菌浓度)
1.结核分枝杆菌标准株H37Rv的液体培养:在7H9培养基中培养,37℃摇床长培养至菌液600nm下吸光值(OD600)为0.3-0.7。
2.制作各个化合物不同浓度(化合物的浓度设计如表2所示)的7H11平板:所述化合物用DMSO溶解,配制的浓度按照1∶1000加入培养基中,而对照只用相同量的DMSO加入到没有药物的平板中。
3.MIC检测:用无菌水稀释结核分枝杆菌标准株H37Rv培养液至50倍得到母液,再将母液稀释成10-3和10-5的浓度,然后将这两个浓度的菌液各取0.5mL铺于上述7H11平板上,各两个重复。以加阳性药异烟肼0.1μg/mL为对照。
表2.化合物的浓度设计
Figure PCTCN2015086852-appb-000131
4.结果及结果分析:
在37℃环境中培养4周,统计每板菌落个数,分析数据,得出各化合物的MIC值,结果如表3所示。
表3.各化合物的MIC值
化合物 MIC(μg/mL)
TJ170322 0.1
TJ170372 0.01
TJ170381 0.1
TJ170385 0.01
TJ170375 0.1
阳性对照异烟肼 <=0.1
阴性对照DMSO -
实施例53:所述化合物对临床分选的耐多药结核菌(MDR-TB)菌株的敏感性测定
1.结核分枝杆菌标准株H37Rv及临床分选的MDR-TB菌P91,P105和P103的液体培养:在7H9培养基中培养,37℃摇床长培养至菌液600nm下吸光值(OD600)为0.7以上。
2.制作化合物TJ170298的不同浓度(化合物的浓度设计如表4所示)的7H11平板:所述化合物用DMSO溶解,配制的浓度按照1∶1000加入培养基中。
3.MIC检测:用无菌水稀释各菌株的培养液至50倍得到母液,再将母液稀释成10-3和10-5的浓度,然后将这两个浓度的菌液各取0.5mL铺于上述7H11平板,各两个重复。以加阳性药RIF 10μg/mL和1μg/mL为对照。
表4.研究药物的浓度设计
Figure PCTCN2015086852-appb-000132
4.结果及结果分析:检测化合物TJ170298对临床分选的MDR-TB菌及标准H37Rv的MIC如表5:
表5.各化合物对临床分选的MDR-TB菌及标准H37Rv的MIC值
Figure PCTCN2015086852-appb-000133
上述结果表明,临床分选的MDR-TB菌对化合物TJ170298有很好的敏感性。说明化合物TJ170298对临床分选的MDR-TB菌株具有较好的抗菌活性。
实施例54:本发明化合物体内抗结核病菌活性测定
1.结核分枝杆菌自主发光菌H37Ra的液体培养:在7H9培养基中培养,37℃摇床长培养至菌液发光值为22,000,000RLU/mL。
2.购买5-6周龄BALB/c雄性小鼠,进行感染实验:用自主发光菌H37Ra对每只进行BALB/c雄性小鼠尾静脉注射感染。1天后测定被感染的BALB/c雄性小鼠的发光值,对于发光值大于700RLU/只的BALB/c雄性小鼠进行随机分组。
3.化合物体内活性测定:将不同浓度,不同种类的化合物分别对感染的BALB/c雄性小鼠在感染2天后开始进行灌胃治疗,每天灌胃一次,连续灌胃6天,具体治疗用量如表6所示:
表6.化合物治疗用量
Figure PCTCN2015086852-appb-000134
4.实时监测化合物的体内抗结核病菌活性:分别测定day0,day2,day4活体小鼠内自主发光菌的发光数值,从而实时检测化合物体内抗结核病菌活性。
实验结果如图1-4所示,化合物L298,L385,L372对H37Ra有明显的体内抗结核病菌活性,活性与阳性对照利福平相当或更优,但对脾脏内抗结核病菌活性较阳性对照利福平差。
实施例55:化合物TJ170298和TJ170385的药代动力学研究
1.给药与样品采集
a.大鼠静脉给药:SD大鼠3只,体重180~220g。给药前禁食12h,试验期间少量进食,饮水自由。按2mg/kg的剂量静脉注射给予化合物TJ170298和TJ170385。给药后2min,10min,30min,1.0h,2.0h,3.0h,4.0h,6.0h,8.0h,12.0h,21h,24.0h,30h,36h,48h,60h,72h,分别经眼眶取血约0.3mL,置肝素化试管中,6000rpm离心10min,分离血浆,4℃保存待测。
b.大鼠口服给药:SD大鼠3只,体重180~220g。给药前禁食12h,试验期间少量进食,自由饮水。分别按10mg/kg的剂量口服给予化合物TJ170298和TJ170385。给药后5min,10min,30min,1.0h,2.0h,3.0h,4.0h,6.0h,8.0h,12.0h,21h,24.0h,30h,36h,48h,60h,72h,分别经眼眶取血约0.3mL置肝素化试管中,6000rpm离心10min,分离血浆,4℃保存待测。
2.血浆样品测定
a.血浆样品处理
向50μL上述血浆样品中加入150μL内标溶液(5μg/mL,乙腈溶液),混匀;涡流混合2min,离心30min(13000rpm,4℃),取上清于另一离心管中,取20μL进行LC/MS/MS分析。
b.标准曲线的配制
取大鼠空白血浆50μL,依次加入化合物标准系列溶液10μL,配制相当于血浆浓度为20,50,100,500,1000,2000,4000,6000,12000,40000ng/mL的化合物血浆样品,按“血浆样品处理”项下操作,建立标准曲线。以待测物浓度(x)为横坐标,待测物与内标物的峰面积比值(y)为纵坐标,用加权(W=1/x2)最小二乘法进行回归运算,求得直线回归方程,即为标准曲线。
c.数据处理和分析
采用DAS 2.0软件计算大鼠给药后的药代动力学参数。
表7.化合物在SD大鼠体内的药代动力学性质参数
Figure PCTCN2015086852-appb-000135
AUC(Area Under the Curve):血药浓度-时间曲线下的面积,代表药物的生物利用度(药物在人体中被吸收利用的程度),AUC大则生物利用度高,反之则低。AUC全称为area under concentration-time curve。
Cmax:药峰浓度(Peak Concentration)系指血药浓度——时间曲线上的最大血药浓度值,即用药后所能达到的最高血浆药物浓度。药峰浓度与药物的临床应用密切相关。药峰浓度达到有效浓度才能显效,而如高出了安全的范围则可显示毒性的反应。此外,药峰浓度还是衡量制剂吸收和安全性的重要指标。
T1/2(half life time):半衰期。是指体内药物浓度下降一半所需要的时间,反映药物通过生物转化或排泄从体内消除的快慢。
Tmax:(Peak Time)药峰时间,系指在给药后人体血浆药物浓度曲线上达到最高浓度(药峰浓度)所需的时间。药峰时间短,表示药物吸收快、起效迅速,但同时消除也快;而药峰时间长,则表明药物吸收和起效较慢,药物作用持续时间也往往延长。药峰时间是应用药物和研究制剂的一个重要指标。
BA(bioavailability):生物利用度。是指药物吸收进入大循环的速度和程度。生物利用度分为绝对生物利用度和相对生物利用度。绝对生物利用度是指该药物静脉注射时100%被利用,该药物的其它剂型与其剂量相等时被机体吸收利用的百分率;相对生物利用度则是以某种任意指定的剂型(如口服水制剂)为100%被利用,然后测定该药物其它剂型在相同条件下的百分利用率。
由表7知,化合物TJ170298和TJ170385均有很好的药代动力学特性。
以上所述实施例仅表达了本发明的几种实施方式,其描述较为具体和详细,但并不能因此而理解为对本发明专利范围的限制。应当指出的是,对于本领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干变形和改进,这些都属于本发明的保护范围。因此,本发明专利的保护范围应以所附权利要求为准。

Claims (12)

  1. 具有式(I)结构特征的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子:
    Figure PCTCN2015086852-appb-100001
    m为0,1,2,3或4;
    n为0或1;
    V为CH或N;
    W为CH或N;
    X任选自:CH,O,N或S;
    Y任选自:CH,O,N或S;
    Z任选自:CH,O,N或S;
    L任选自:C0~C5直链或支链的烷烃;
    R8为H或甲基;
    R1任选自:
    1)H;
    2)卤素;
    3)OaC1~C5烷基;
    4)C3~C6环烷基;
    5)芳基;
    6)氨基,羟基,氰基,硝基;
    7)杂芳基;
    所述烷基、芳基、环烷基和杂芳基分别任选被0,1,2或3个选自R6的取代基取代;
    R2任选自:
    1)H;
    2)OaC1~C5烷基;
    3)C3~C6环烷基;
    4)芳基;
    5)杂芳基;
    所述烷基、芳基、环烷基和杂芳基分别任选被0,1,2或3个选自R6的取代基取代;
    R3任选自:
    1)H;
    2)卤素;
    3)OaC1~C5烷基;
    4)C3~C6环烷基;
    5)
    Figure PCTCN2015086852-appb-100002
    6)
    Figure PCTCN2015086852-appb-100003
    b为0或1;U为CH或N;
    R4任选自:
    a)H;
    b)F,Cl,Br;
    c)C1~C3烷基;
    d)C1~C3烷氧基;
    e)苯氧基;
    7)
    Figure PCTCN2015086852-appb-100004
    R5任选自:
    a)H;
    b)F,Cl,Br;
    c)OaC1~C3烷基;
    8)
    Figure PCTCN2015086852-appb-100005
    9)(C=O)cOdR7,其中,c为0或1;d为0或1;
    R7任选自:
    a)CF3
    b)C1~C5烷基;
    10)-N(CH3)2
    11)
    Figure PCTCN2015086852-appb-100006
    12)
    Figure PCTCN2015086852-appb-100007
    所述烷基、芳基、环烷基、杂环烷基和杂芳基分别任选被0,1,2或3个选自R6取代基取代,
    其中,所述a为0或1;
    所述R6任选自:
    1)H;
    2)C3~C6环烷基;
    3)杂环基;
    4)C1~C3烷基;
    5)C1~C3含氟烷基;
    6)C0~C3亚烷基-杂环基;
    7)卤素。
  2. 根据权利要求1所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子,其特征在于,
    所述R1任选自:
    1)H;
    2)F,Cl,Br,I;
    3)OH,OCH3,OEt,OCF3
    4)甲基,乙基,异丙基,叔丁基;
    5)环丙基;
    6)CF3
    7)苯基;
    所述R2任选自:
    1)H;
    2)甲基,乙基,丙基,异丙基,叔丁基;
    3)环丙基;
    4)苯基;
    所述R3任选自:
    1)
    Figure PCTCN2015086852-appb-100008
    2)
    Figure PCTCN2015086852-appb-100009
    3)
    Figure PCTCN2015086852-appb-100010
    4)
    Figure PCTCN2015086852-appb-100011
    5)CF3
    6)(C=O)cOdR7,其中,c为0或1;d为0或1;
    R7任选自:
    a)CF3
    b)C1~C5烷基;
    7)H;
    8)卤素。
  3. 根据权利要求1所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子,其特征在于,所述
    Figure PCTCN2015086852-appb-100012
    选自:
    Figure PCTCN2015086852-appb-100013
    Figure PCTCN2015086852-appb-100014
  4. 根据权利要求1所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子,其特征在于,具有如式(II)所示结构特征:
    Figure PCTCN2015086852-appb-100015
    所述R1为甲基;R2为甲基;
    所述R3任选自:
    1)-N(CH3)2
    2)
    Figure PCTCN2015086852-appb-100016
    3)-C(CH3)3
    4)
    Figure PCTCN2015086852-appb-100017
    5)
    Figure PCTCN2015086852-appb-100018
    6)
    Figure PCTCN2015086852-appb-100019
    n=1。
  5. 根据权利要求1-3任一项所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子,其特征在于,具有如式(II)所示结构特征:
    Figure PCTCN2015086852-appb-100020
    所述R1任选自:
    1)H;
    2)F,Cl,Br,I;
    3)OH,OCH3,OEt,OCF3
    4)甲基,乙基,异丙基,叔丁基;
    5)环丙基;
    6)CF3
    7)苯基;
    所述R2任选自:
    1)H;
    2)甲基,乙基,丙基,异丙基;
    3)环丙基;
    4)苯基;
    所述R3任选自:
    1)
    Figure PCTCN2015086852-appb-100021
    2)
    Figure PCTCN2015086852-appb-100022
    3)
    Figure PCTCN2015086852-appb-100023
    4)
    Figure PCTCN2015086852-appb-100024
    5)CF3
    6)(C=O)OMe;
    7)H;
    8)卤素;
    所述
    Figure PCTCN2015086852-appb-100025
    选自
    Figure PCTCN2015086852-appb-100026
    Figure PCTCN2015086852-appb-100027
  6. 根据权利要求1所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子,其特征在于,具有如式(III)所示结构特征:
    Figure PCTCN2015086852-appb-100028
    所述R1任选自:
    1)H;
    2)F,Cl,Br;
    3)OH,OCH3,OEt;
    4)甲基,乙基,异丙基,叔丁基;
    5)CF3
    6)苯基;
    所述R2任选自:
    1)H;
    2)甲基,乙基,丙基;
    3)环丙基;
    4)苯基;
    所述R9任选自:
    1)F,Cl,Br;
    2)OCH3,OCF3
    所述Q任选自:CH,N。
  7. 根据权利要求6所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子,其特征在于,
    所述R1任选自:
    1)2-Cl,2-Br;
    2)2-OCH3,2-OEt;
    3)2-甲基,2-乙基,3-甲基,3-乙基;
    4)H;
    所述R2任选自:
    1)甲基,乙基,丙基;
    2)环丙基;
    所述R9任选自:
    1)F,Cl;
    2)OCH3,OCF3
    所述Q任选自:CH,N。
  8. 根据权利要求5所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子,其特征在于,具有如式(IV)所示结构特征:
    Figure PCTCN2015086852-appb-100029
    所述R1任选自:
    1)H;
    2)F,Cl,Br;
    3)OH,OCH3,OEt;
    4)甲基,乙基,异丙基,叔丁基;
    5)CF3
    6)苯基;
    所述R2任选自:
    1)H;
    2)甲基,乙基,丙基;
    3)环丙基;
    4)苯基;
    所述R10任选自:
    1)H;
    2)F,Cl,Br;
    3)CF3
    4)
    Figure PCTCN2015086852-appb-100030
    所述U任选自:CH,N。
  9. 根据权利要求5所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子,其特征在于,具有如式(II)所示结构特征:
    Figure PCTCN2015086852-appb-100031
    所述R1为甲基;R2为甲基;
    所述R3任选自:
    1)
    Figure PCTCN2015086852-appb-100032
    2)
    Figure PCTCN2015086852-appb-100033
  10. 根据权利要求1所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子,其特征在于,所述化合物选自:
    5-氯-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-2-乙基-N-(4-(三氟甲氧基)苯基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-2-环丙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-2-苯基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    4-((5-氯-2-乙基吡唑[1,5-a]吡啶-3-甲酰胺)甲基)苯甲酸甲酯
    5-氯-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,7-二甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,6-二甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,4-二甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-甲氧基-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-2-乙基-N-((1-(4-(三氟甲氧基)苯基)-1H-1,2,3-三氮唑-4-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-2-乙基-N-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-2-乙基-N-(2-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)丙烷-2-取代)吡唑[1,5-a]吡啶-3-甲酰胺
    5-三氟甲基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-((1-(4-(三氟甲氧基)苯基)-1H-1,2,3-三氮唑-4-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-溴-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-苯基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-2-乙基-N-((6-(4-(三氟甲氧基)苯基)吡啶-3-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-((6-(4-(三氟甲氧基)苯基)吡啶-3-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-甲氧基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    2-甲基-N-(4-(三氟甲氧基)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-2-乙基-N-(1-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯基)乙醇)吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-(4-(三氟甲氧基)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-异丙基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-叔丁基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-乙基-2-甲基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-甲基-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-甲基-2-乙氧基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-2-乙基-N-(4-(4-(4-(三氟甲氧基)苯基)哌啶-1-取代)苯乙基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-((5-(4-(三氟甲氧基)苯基)噻吩-2-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-((5-(4-(三氟甲氧基)苯基)吡啶-2-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-((5-(4-(三氟甲氧基)苯基)吡啶-2-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺
    N-(4-(叔丁基)苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺
    N-(4-(二甲氨基)苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺
    N-(4-(4-(4-甲氧基苯基)哌啶-1-取代)苄基)-2,5-二甲基-吡唑[1,5-a]吡啶-3-甲酰胺
    N-(4-(4-(4-氟苯基)哌啶-1-取代)苄基)-2,5-二甲基-吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-(4-(哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    N-(4-(4-氟苯氧基)苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-(4-(4-(三氟甲基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-((2-(4-(三氟甲氧基)苯基)嘧啶-5-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-((4’-(三氟甲氧基)-[1,1’-联苯]-4-取代)甲基)吡唑[1,5-a]吡啶-3-甲酰胺
    N-(4-(4-(4-氟苯基)哌嗪-1-取代)苄基)-2,5-二甲基-吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-(4-(4-(三氟甲氧基)苯基)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    2,5-二甲基-N-(4-(4-(4-(三氟甲氧基)苯氧基)哌啶-1-取代)苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    5-氯-2-乙基-N-(4-氟苄基)吡唑[1,5-a]吡啶-3-甲酰胺
    N-(4-氟苄基)-2,5-二甲基吡唑[1,5-a]吡啶-3-甲酰胺。
  11. 权利要求1-10任一项所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子在制备具有抗结核病作用的药物中的应用。
  12. 一种抗结核病药物组合物,其特征在于,其活性成分包括权利要求1-10任一项所述的吡唑并[1,5-a]吡啶类化合物或其药学上可接受的盐或其立体异构体或其前药分子。
PCT/CN2015/086852 2014-10-21 2015-08-13 吡唑并[1,5-a]吡啶类化合物及其应用 WO2016062151A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/520,765 US10155756B2 (en) 2014-10-21 2015-08-13 Pyrazolo[1,5-A]pyridine compounds and use thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201410562805 2014-10-21
CN201410562805.5 2014-10-21
CN201510460751.6 2015-07-29
CN201510460751.6A CN105524058B (zh) 2014-10-21 2015-07-29 吡唑并[1,5‑a]吡啶类化合物及其应用

Publications (1)

Publication Number Publication Date
WO2016062151A1 true WO2016062151A1 (zh) 2016-04-28

Family

ID=55760259

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/086852 WO2016062151A1 (zh) 2014-10-21 2015-08-13 吡唑并[1,5-a]吡啶类化合物及其应用

Country Status (3)

Country Link
US (1) US10155756B2 (zh)
CN (1) CN105524058B (zh)
WO (1) WO2016062151A1 (zh)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017001661A1 (en) * 2015-07-02 2017-01-05 Janssen Sciences Ireland Uc Antibacterial compounds
WO2017216283A1 (en) 2016-06-16 2017-12-21 Janssen Sciences Ireland Uc Heterocyclic compounds as antibacte rials
US11179396B2 (en) 2016-06-16 2021-11-23 Janssen Sciences Ireland Uc Heterocyclic compounds as antibacterials
US11224596B2 (en) 2017-03-01 2022-01-18 Janssen Sciences Ireland Unlimited Company PZA and cytochrome bc1 inhibitor combination treatment
US11820767B2 (en) 2015-09-17 2023-11-21 University Of Notre Dame Du Lac Benzyl amine-containing heterocyclic compounds and compositions useful against mycobacterial infection

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106866680B (zh) * 2017-03-31 2019-02-01 陕西理工大学 一种3-芳基-6-甲酰胺吡唑并[1,5-a]嘧啶类化合物及其应用
CN108159049B (zh) * 2018-02-01 2021-01-05 中国科学院广州生物医药与健康研究院 一种吡啶类化合物的新用途
TW202124379A (zh) * 2019-09-10 2021-07-01 日商鹽野義製藥股份有限公司 對分枝桿菌感染有用之含苄胺的5,6-雜芳族化合物
WO2021048342A1 (en) * 2019-09-13 2021-03-18 Janssen Sciences Ireland Unlimited Company Antibacterial compounds
CN111620881B (zh) * 2020-07-08 2023-03-31 浙江合聚生物医药有限公司 拉罗替尼衍生物及其制备方法和应用
CN112089713B (zh) * 2020-09-18 2022-02-22 中国科学院广州生物医药与健康研究院 一种基于吡唑并[1,5-a]吡啶类化合物的治疗结核分枝杆菌感染的药物组合物
CN112724132B (zh) * 2021-01-04 2022-05-20 药雅科技(上海)有限公司 一种3-卤代-5-三氟甲基-吡唑并[1,5-a]吡啶的合成方法
BR112023018662A2 (pt) * 2021-03-16 2023-12-19 Janssen Sciences Ireland Unlimited Co Compostos antibacterianos
TW202325274A (zh) * 2021-10-28 2023-07-01 愛爾蘭商健生科學愛爾蘭無限公司 抗菌化合物
WO2024089170A1 (en) * 2022-10-27 2024-05-02 Janssen Sciences Ireland Unlimited Company Antibacterial compounds

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103492384A (zh) * 2011-02-25 2014-01-01 Irm责任有限公司 作为trk抑制剂的化合物和组合物
CN104024252A (zh) * 2011-09-01 2014-09-03 诺华股份有限公司 用于治疗肺动脉高压的双环杂环衍生物

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060094699A1 (en) * 2003-04-11 2006-05-04 Kampen Gita Camilla T Combination therapy using an 11beta-hydroxysteroid dehydrogenase type 1 inhibitor and a glucocorticoid receptor agonist to minimize the side effects associated with glucocorticoid receptor agonist therapy
US7501405B2 (en) * 2003-04-11 2009-03-10 High Point Pharmaceuticals, Llc Combination therapy using an 11β-hydroxysteroid dehydrogenase type 1 inhibitor and an antihypertensive agent for the treatment of metabolic syndrome and related diseases and disorders
US7700583B2 (en) * 2003-04-11 2010-04-20 High Point Pharmaceuticals, Llc 11β-hydroxysteroid dehydrogenase type 1 active compounds
US7795273B2 (en) * 2005-12-08 2010-09-14 Novartis Ag Pyrazolo[1,5-a]pyridine-3-carboxylic acids as EphB and VEGFR2 kinase inhibitors
US8148380B2 (en) * 2007-07-23 2012-04-03 Crestone, Inc. Antibacterial amide and sulfonamide substituted heterocyclic urea compounds
EP2219646A4 (en) * 2007-12-21 2010-12-22 Univ Rochester METHOD FOR MODIFYING THE LIFETIME OF EUKARYOTIC ORGANISMS
US8927545B2 (en) * 2009-03-30 2015-01-06 Duke University Inhibiting Eph B-3 kinase
WO2011050245A1 (en) * 2009-10-23 2011-04-28 Yangbo Feng Bicyclic heteroaryls as kinase inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103492384A (zh) * 2011-02-25 2014-01-01 Irm责任有限公司 作为trk抑制剂的化合物和组合物
CN104024252A (zh) * 2011-09-01 2014-09-03 诺华股份有限公司 用于治疗肺动脉高压的双环杂环衍生物

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017001661A1 (en) * 2015-07-02 2017-01-05 Janssen Sciences Ireland Uc Antibacterial compounds
JP2018519302A (ja) * 2015-07-02 2018-07-19 ヤンセン・サイエンシズ・アイルランド・ユーシー 抗菌化合物
US10364232B2 (en) 2015-07-02 2019-07-30 Janssen Sciences Ireland Uc Antibacterial compounds
US11180472B2 (en) 2015-07-02 2021-11-23 Janssen Sciences Ireland Uc Antibacterial compounds
US11866419B2 (en) 2015-07-02 2024-01-09 Janssen Sciences Ireland Unlimited Company Antibacterial compounds
US11820767B2 (en) 2015-09-17 2023-11-21 University Of Notre Dame Du Lac Benzyl amine-containing heterocyclic compounds and compositions useful against mycobacterial infection
WO2017216283A1 (en) 2016-06-16 2017-12-21 Janssen Sciences Ireland Uc Heterocyclic compounds as antibacte rials
CN109415349A (zh) * 2016-06-16 2019-03-01 爱尔兰詹森科学公司 杂环化合物作为抗细菌剂
US11179396B2 (en) 2016-06-16 2021-11-23 Janssen Sciences Ireland Uc Heterocyclic compounds as antibacterials
US11224596B2 (en) 2017-03-01 2022-01-18 Janssen Sciences Ireland Unlimited Company PZA and cytochrome bc1 inhibitor combination treatment
US11918575B2 (en) 2017-03-01 2024-03-05 Janssen Sciences Ireland Unlimited Company PZA and cytochrome Bc1 inhibitor combination treatment

Also Published As

Publication number Publication date
US10155756B2 (en) 2018-12-18
CN105524058A (zh) 2016-04-27
US20170313697A1 (en) 2017-11-02
CN105524058B (zh) 2018-03-27

Similar Documents

Publication Publication Date Title
WO2016062151A1 (zh) 吡唑并[1,5-a]吡啶类化合物及其应用
KR102292293B1 (ko) 항감염성 화합물
JP4719151B2 (ja) イソキノリノンカリウムチャネル阻害剤
EP1831201B1 (en) Piperidine and azetidine derivatives as glyt1 inhibitors
AU2013290375B2 (en) 5-HT3 receptor antagonists
CN102105170A (zh) 抗真菌组合治疗
JP2007516218A (ja) イソキノリノンカリウムチャネル阻害剤
CA3021608A1 (en) Fluorinated cyclopropylamine compound, preparation method therefor, pharmaceutical composition thereof, and uses thereof
BR112014002958B1 (pt) 3,4-dihidro-1h-[1,8]naftiridinonas substituídas com piperidinila antibacterianas, composição farmacêutica compreendendo os referidos compostos, processos para preparação destes e uso
TW201625566A (zh) 作為gpr139的調節劑之4-側氧基-3,4-二氫-1,2,3-苯並三
CN107200734B (zh) 奎宁环衍生物及其制备方法和用途
RU2444515C2 (ru) Производное амида индазолакриловой кислоты
EP4121167A1 (en) Selective modulators of mutant lrrk2 proteolysis and associated methods of use
JP2020522517A (ja) バニン阻害薬としてのヘテロ芳香族化合物
JP2007506748A (ja) イソキノリノンカリウムチャネル阻害剤
WO2023030459A1 (zh) 一类含喹啉酮酰胺的化合物及其制备方法、药物组合物和用途
CN105367565B (zh) 哌嗪(啶)环己基衍生物及其治疗精神神经疾病的应用
TWI424998B (zh) 作為腺苷A3受體配體的三唑并[1,5-a]喹啉類
JP7110338B2 (ja) ムスカリンm1受容体陽性アロステリックモジュレーターである多環式アミド類
US20200377496A1 (en) INDOLE AND BENZIMIDAZOLE DERIVATIVES AS DUAL 5-HT2A and 5-HT6 RECEPTOR ANTAGONISTS
US11981668B2 (en) Indole and benzimidazole derivatives as dual 5-HT2A and 5-HT6 receptor antagonists
JP2024500179A (ja) ピロール誘導体、その調製方法及びその用途
EP4171569A1 (en) Quinazolinones derivatives for treatment of non-alcoholic fatty liver disease, preparation and use thereof
TW202328072A (zh) Ahr促效劑
WO2022057858A1 (zh) 酰胺膦氧类衍生物及其制备方法和用途

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15852500

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 15520765

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15852500

Country of ref document: EP

Kind code of ref document: A1