WO2022258007A1 - 含吡唑多环类衍生物的盐、晶型及其制备方法和应用 - Google Patents

含吡唑多环类衍生物的盐、晶型及其制备方法和应用 Download PDF

Info

Publication number
WO2022258007A1
WO2022258007A1 PCT/CN2022/097828 CN2022097828W WO2022258007A1 WO 2022258007 A1 WO2022258007 A1 WO 2022258007A1 CN 2022097828 W CN2022097828 W CN 2022097828W WO 2022258007 A1 WO2022258007 A1 WO 2022258007A1
Authority
WO
WIPO (PCT)
Prior art keywords
acid
crystal form
diffraction peak
alkyl
cyano
Prior art date
Application number
PCT/CN2022/097828
Other languages
English (en)
French (fr)
Inventor
董华
呙临松
Original Assignee
上海翰森生物医药科技有限公司
江苏豪森药业集团有限公司
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 上海翰森生物医药科技有限公司, 江苏豪森药业集团有限公司 filed Critical 上海翰森生物医药科技有限公司
Priority to IL309178A priority Critical patent/IL309178A/en
Priority to MX2023014525A priority patent/MX2023014525A/es
Priority to JP2023576189A priority patent/JP2024522207A/ja
Priority to AU2022290123A priority patent/AU2022290123A1/en
Priority to EP22819604.4A priority patent/EP4353725A1/en
Priority to CA3221791A priority patent/CA3221791A1/en
Priority to BR112023025543A priority patent/BR112023025543A2/pt
Priority to KR1020247000484A priority patent/KR20240019257A/ko
Priority to CN202280039138.5A priority patent/CN117412974A/zh
Publication of WO2022258007A1 publication Critical patent/WO2022258007A1/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
    • C07D471/12Heterocyclic 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 three hetero rings
    • C07D471/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • C07D471/12Heterocyclic 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 three hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the invention belongs to the field of biomedicine, and specifically relates to a salt, a crystal form, a preparation method and an application of a pyrazole-containing polycyclic derivative.
  • P2X receptors or P2X purinoreceptors are a family of cation-permeable ATP ligand-gated ion channels that bind to extracellular ATP.
  • P2X receptors have seven subunits and exist in the form of homotrimers or heterotrimers. They are mainly expressed in nerve terminals (pre-synaptic and post-synaptic) of the nervous system and regulate synaptic transmission.
  • the P2X3 receptor is one of the members of the P2X family. It is a key sensory receptor for sensing upper airway stimuli and triggering cough reflexes. It is believed to play a key role in the sensitization of specific sensory nerves, participate in pain and cough, and In bone cancer pain perception, blocking P2X3 suppresses cough signaling.
  • Cough is a defensive nerve reflex of the body, which is beneficial to clear respiratory secretions and harmful factors, but frequent and severe coughing will seriously affect the work, life and social activities of patients. Cough is divided into acute, subacute and chronic cough.
  • Chronic cough (Chronic cough) refers to the cough duration > 8 weeks, with cough as the main or only symptom, and no obvious lesions in the lungs by chest imaging examination. Chronic cough has long been recognized as a consequence of various conditions such as asthma/eosinophilic bronchitis, rhinitis, and gastroesophageal acid reflux disease. However, recent evidence suggests that chronic cough is a clinical symptom of neuroticism with unique intrinsic pathophysiological features.
  • Unexplained chronic cough or idiopathic cough which is mainly manifested by chronic irritating dry cough, is more sensitive to external stimuli, and cough hypersensitivity is common, and cough hypersensitivity is its physiological and pathological mechanism.
  • Cough-related afferent abnormalities may be the cause of refractory or unexplained chronic cough.
  • Chronic cough can cause complications in cardiovascular, digestive, nervous, urinary, musculoskeletal systems, such as urinary incontinence, syncope, insomnia, anxiety, etc.
  • treatment should aim at reducing cough sensitivity.
  • Current treatment options are limited, including pharmacological and non-pharmacological approaches.
  • Clinical research results show that the neuromodulator drug gabapentin is effective in treatment, and other drugs such as amitriptyline, baclofen, carbamazepine, and pregabalin can also be used.
  • Severe cough can be appropriately given antitussive treatment, and antitussive drugs are mainly divided into central antitussives and peripheral antitussives.
  • Central antitussive drugs are divided into dependent antitussives (morphine alkaloids and their derivatives) and non-dependent antitussives (synthetic dextromethorphan and pentoxyverine), the former is addictive and narcotic Sex and other side effects, the latter clinical application is very extensive.
  • Peripheral antitussives also known as peripheral antitussives, play an antitussive effect by inhibiting a certain link in the cough reflex arc, including local anesthetics (narcotine, benzonatate) and mucosal protective agents (benzonatate) properine and mogisteine).
  • P2X3 receptor antagonists there is no approved small-molecule drug for P2X3 receptor antagonists on the market.
  • the P2X3 receptor antagonist drugs currently in the clinical stage include Merck&Co's MK-7264, which is used to treat diseases such as chronic cough, pain and pulmonary fibrosis. It has low selectivity for P2X3/P2X2/3 and is safe. It has good sex but has side effects such as loss of taste.
  • the current indication for chronic cough has entered clinical phase III research. Bellus Health's BLU5937 has high selectivity and no side effects such as taste in Phase I clinical trials.
  • the object of the present invention is to provide an acid salt of a compound represented by general formula (I-a) or a stereoisomer thereof,
  • R is selected from hydrogen, deuterium, halogen, amino, hydroxyl, cyano, nitro, C 1-6 alkyl , C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 deuterated alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, halo C 1-6 alkoxy, C 1-6 hydroxyalkyl, C 3-12 cycloalkyl, -(CH 2 ) n C(O )R a , 3-12 membered heterocyclic group, C 6-14 aryl group or 5-14 membered heteroaryl group, the amino group, C 1-6 alkyl group, C 2-6 alkenyl group, C 2-6 Alkynyl, C 1-6 deuterated alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, halogenated C 1-6 alkoxy, C 1-6 hydroxyalkyl, C 3-12 ring Alkyl, -(CH 2 ) n C(O)
  • R is selected from hydrogen, deuterium , halogen, amino, nitro, hydroxyl, cyano, C 1-6 alkyl, C 1-6 deuterated alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl , C 1-6 alkoxy, C 1-6 haloalkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, C 6- 10 aryl or 5-10 membered heteroaryl, the amino, C 1-6 alkyl, C 1-6 deuterated alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1 -6 alkoxy, C 1-6 haloalkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, optionally deuterium, halogen
  • R is selected from hydrogen , deuterium, halogen, amino, hydroxyl, cyano, nitro, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 deuterated alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, halo C 1-6 alkoxy, C 1-6 hydroxyalkyl, C 3-12 cycloalkyl, 3-12 membered heterocyclyl, C 6-14 aryl or 5-14 membered heteroaryl, the amino, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 deuterated alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, halo C 1-6 alkoxy, C 1-6 hydroxyalkyl, C 3-12 cycloalkyl, 3-12 membered heterocyclyl, C 6 -14 aryl and 5-14 membered heteroaryl, optionally replaced by deuterium, hal
  • R is selected from hydrogen, deuterium, halogen, amino, hydroxyl, cyano, oxo, thio, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 Deuterated alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 alkoxy, C 1-6 haloalkoxy, C 3-8 cycloalkyl, 3-12 membered heterocycle base, C 6-14 aryl or 5-14 membered heteroaryl; and
  • x is an integer of 0 to 3, preferably 0, 1 or 2, more preferably 0 or 1;
  • n is an integer of 0 to 3, preferably 0, 1 or 2, more preferably 0 or 1;
  • the acid in the acid salt is an inorganic acid or an organic acid; preferably, the inorganic acid is selected from hydrochloric acid, sulfuric acid, nitric acid, hydrobromic acid, hydrofluoric acid, hydroiodic acid or phosphoric acid; the organic acid is selected from 2,5-dihydroxy Benzoic acid, 1-hydroxy-2-naphthoic acid, acetic acid, ethanesulfonic acid, dichloroacetic acid, trichloroacetic acid, acetohydroxamic acid, adipic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, benzoic acid, 4-Acetaminobenzoic acid, 4-aminobenzoic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamate, camphorsulfonic acid, aspartic acid, camphoric acid, gluconic acid, glucosinolate
  • said R is selected from hydrogen, deuterium, halogen, amino, hydroxyl, cyano, nitro, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl , C 1-6 deuterated alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, halogenated C 1-6 alkoxy, C 1-6 hydroxyalkyl, C 3-8 cycloalkyl , -(CH 2 ) n C(O) Ra , 3-8 membered heterocyclic group, C 6-10 aryl or 5-10 membered heteroaryl, the amino, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 deuterated alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, halogenated C 1-6 alkoxy, C 1- 6 hydroxyalkyl, C 3-8 cycloalkyl, -(CH 2 )
  • R a is selected from hydrogen, deuterium, halogen, amino, nitro, hydroxyl, cyano, C 1-3 alkyl, C 1-3 deuterated alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl , C 1-3 alkoxy, C 1-3 haloalkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, C 6- 10 aryl or 5-10 membered heteroaryl, the amino, C 1-3 alkyl, C 1-3 deuterated alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl, C 1 -3 alkoxy, C 1-3 haloalkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, C 6-10 aryl and 5-10 membered heteroaryl, optionally deuterium, hal
  • R is selected from hydrogen, halogen, amino, cyano, C 1-3 alkyl, C 2-6 alkenyl, C 1-6 haloalkyl, C 1-6 alkoxy, halogenated C 1 -6 alkoxy, C 1-6 hydroxyalkyl, C 3-8 cycloalkyl, -(CH 2 ) n C(O)R a , 3 containing 1-3 atoms selected from nitrogen, oxygen or sulfur -8-membered heterocyclic group, C 6-10 aryl group or 5-10 membered heteroaryl group containing 1-3 atoms selected from nitrogen, oxygen or sulfur, the amino group, C 1-6 alkyl group, C 2- 6 alkenyl, C 1-6 haloalkyl, C 1-6 alkoxy, halo C 1-6 alkoxy, C 1-6 hydroxyalkyl, C 3-8 cycloalkyl, containing 1-3 3-8 membered heterocyclic group selected from nitrogen, oxygen or sulfur atom, C 6-10 aryl group
  • R a is selected from hydrogen, deuterium, halogen, amino, cyano, C 1-3 alkyl, C 1-3 deuterated alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl, C 1-3 Alkoxy, C 1-3 haloalkoxy, C 3-6 cycloalkyl or a 4-6 membered heterocyclic group containing 1-2 selected from N or O, the amino C 1-3 alkyl, C 1-3 deuterated alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, C 3-6 cycloalkyl or 1 -2 4-6 membered heterocyclic groups selected from N or O, optionally replaced by deuterium, halogen, amino, hydroxyl, cyano, nitro, oxo, C 1-3 alkyl, C 1-3 Deuterated alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C
  • R is selected from the following groups :
  • R is selected from hydrogen , deuterium, halogen, amino, hydroxyl, cyano, nitro, C 1-3 alkyl, C 2-3 alkenyl, C 2-3 alkynyl, C 1-3 deuterated alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, halo C 1-3 alkoxy, C 1-3 hydroxyalkyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, preferably hydrogen, amino, cyano, fluorine, chlorine, bromine, methyl, isopropyl, trifluoromethyl, methoxy, cyclopropyl or morpholine base;
  • R is selected from hydrogen, deuterium, halogen, amino, hydroxyl, cyano, oxo, thio, C 1-3 alkyl, C 2-3 alkenyl, C 2-3 alkynyl, C 1-3 Deuterated alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, C 3-8 cycloalkyl, 3-8 membered heterocycle Base, C 6-10 aryl or 5-1. membered heteroaryl, preferably hydrogen or cyano.
  • the acid in the acid salt is selected from isethionic acid, hydrochloric acid, sulfuric acid, 1,5-naphthalene disulfonic acid, methanesulfonic acid, hydrobromic acid, ethanesulfonic acid, phosphoric acid, Benzenesulfonic acid, oxalic acid, maleic acid, adipic acid, hydrochloric acid, citric acid, malonic acid, L-malic acid, pamoic acid, p-toluenesulfonic acid or fumaric acid; preferably hydrochloric acid, sulfuric acid, methanesulfonic acid, hydrobromic acid or ethanesulfonic acid.
  • the acid salt of the compound is 0.2-3; preferably 0.2, 0.5, 1, 1.5, 2, 2.5 or 3; more preferably 0.5, 1, 2 or 3, 1 is further preferred.
  • the acid salt of the compound is a hydrate or anhydrous; when the acid salt is a hydrate, the number of water is 0.2-3; preferably 0.2, 0.5, 1, 1.5, 2, 2.5 or 3; more preferably 0.5, 1, 2 or 3.
  • the acid salt of the compound, the acid salt is a crystalline form
  • the crystal form is the compound 2-(2-(tert-butyl)-5-oxopyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl)-N Acid salt crystal form of -(5-fluoropyridin-2-yl)acetamide;
  • compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridine[3,2-e]pyrimidine is provided -4(5H)-yl)-N-(5-fluoropyridin-2-yl)acetamide mesylate form A, ethanesulfonate form A, sulfate form A-B, hydrochloride Crystal forms A-B and hydrobromide salt forms A-C.
  • the acid salt crystal form of the compound is mesylate salt crystal form A, and its X-ray powder diffraction pattern has a diffraction peak at 13.7 ⁇ 0.2°; or at 21.9 ⁇ 0.2° or have a diffraction peak at 20.4 ⁇ 0.2°; or have a diffraction peak at 15.4 ⁇ 0.2°; or have a diffraction peak at 19.6 ⁇ 0.2°; or have a diffraction peak at 16.4 ⁇ 0.2°; Or have a diffraction peak at 9.3 ⁇ 0.2°; or have a diffraction peak at 5.3 ⁇ 0.2°; or have a diffraction peak at 7.9 ⁇ 0.2°; or have a diffraction peak at 11.9 ⁇ 0.2°; preferably included in the above-mentioned diffraction peaks Any 2-5, or 3-5, or 3-6, or 3-8, or 5-8, or 6-8, more preferably including any 6, 7 or 8 of them ;
  • the X-ray powder diffraction pattern of mesylate salt crystal form A contains at least one or more diffraction peaks at 2 ⁇ of 13.7 ⁇ 0.2°, 16.4 ⁇ 0.2°, 21.9 ⁇ 0.2°, preferably two of them strips, more preferably comprising three strips; optionally, it may further comprise at least one of the One, preferably including 2, 3, 4 or 5 of them;
  • the X-ray powder diffraction pattern of the mesylate salt crystal form A optionally further comprises a position at 2 ⁇ of 7.9 ⁇ 0.2°, 19.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.8 ⁇ 0.2°, 21.0 ⁇ 0.2°,
  • One or more diffraction peaks in 23.3 ⁇ 0.2°, 24.1 ⁇ 0.2° preferably at least include any 2-3, or 4-5, or 6-7 of them; more preferably, include at least any 2 of them , 3 places, 4 places, 5 places, 6 places, 7 places;
  • the X-ray powder diffraction pattern of the mesylate salt crystal form A includes 2 ⁇ at 5.3 ⁇ 0.2°, 7.9 ⁇ 0.2°, 9.3 ⁇ 0.2°, 11.9 ⁇ 0.2°, 13.7 ⁇ 0.2°, 13.9 ⁇ 0.2 °, 15.4 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.8 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.4 ⁇ 0.2°, 21.0 ⁇ 0.2°, 21.9 ⁇ 0.2°, 23.3 ⁇ 0.2°, 24.1 ⁇ 0.2
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3, 2-e]Pyrimidin-4(5H)-yl)-N-(5-fluoropyridin-2-yl)acetamide mesylate salt form A, using Cu-K ⁇ radiation, at 2 ⁇ angle and interplanar spacing
  • the X-ray characteristic diffraction peaks represented by d values are shown in Table 1.
  • the acid salt crystal form of the compound is ethanesulfonate crystal form A, and its X-ray powder diffraction pattern has a diffraction peak at 15.0 ⁇ 0.2°; or at 21.1 ⁇ Diffraction peak at 0.2°; or diffraction peak at 23.1 ⁇ 0.2°; or diffraction peak at 19.8 ⁇ 0.2°; or diffraction peak at 12.5 ⁇ 0.2°; or diffraction peak at 9.0 ⁇ 0.2° or have a diffraction peak at 12.3 ⁇ 0.2°; or have a diffraction peak at 24.6 ⁇ 0.2°; or have a diffraction peak at 10.3 ⁇ 0.2°; or have a diffraction peak at 6.1 ⁇ 0.2°; preferably include the above-mentioned diffraction peaks Any 2-5, or 3-5, or 3-6, or 3-8, or 5-8, or 6-8, more preferably including any 6, 7 or 8 of them place;
  • the X-ray powder diffraction pattern of ethanesulfonate crystal form A contains at least one or more diffraction peaks at 2 ⁇ of 15.0 ⁇ 0.2°, 21.1 ⁇ 0.2°, and 23.1 ⁇ 0.2°, preferably including Two, more preferably three;
  • it may further include at least one of 2 ⁇ of 19.8 ⁇ 0.2°, 12.5 ⁇ 0.2°, 9.0 ⁇ 0.2°, 12.3 ⁇ 0.2°, 24.6 ⁇ 0.2°, preferably including 2, 3, 4 or 5 of them;
  • the X-ray powder diffraction pattern of ethanesulfonate salt crystal form A optionally further comprises a position at 2 ⁇ of 10.3 ⁇ 0.2°, 6.1 ⁇ 0.2°, 16.1 ⁇ 0.2°, 19.2 ⁇ 0.2°, 23.6 ⁇ 0.2° , 30.7 ⁇ 0.2°, 9.6 ⁇ 0.2°, one or more diffraction peaks; preferably at least include any 2-3, or 4-5, or 6-7; more preferably, at least include any 2 place, 3 places, 4 places, 5 places, 6 places, 7 places;
  • the X-ray powder diffraction pattern of ethanesulfonic acid salt crystal form A contains 2 ⁇ at 15.0 ⁇ 0.2°, 21.1 ⁇ 0.2°, 23.1 ⁇ 0.2°, 19.8 ⁇ 0.2°, 12.5 ⁇ 0.2°, 9.0 ⁇ One of 0.2°12.3 ⁇ 0.2°, 24.6 ⁇ 0.2°, 10.3 ⁇ 0.2°, 6.1 ⁇ 0.2°, 16.1 ⁇ 0.2°, 19.2 ⁇ 0.2°, 23.6 ⁇ 0.2°, 30.7 ⁇ 0.2°, 9.6 ⁇ 0.2° or multiple diffraction peaks, preferably, including optional 4, 5, 6, 8 or 10 diffraction peaks;
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridine[3,2-e] Form A of the ethanesulfonate salt of pyrimidin-4(5H)-N-(5-fluoropyridin-2-yl)acetamide, expressed in 2 ⁇ angles and interplanar spacing d values using Cu-K ⁇ radiation
  • the X-ray characteristic diffraction peaks are shown in Table 2.
  • the acid salt of the compound is sulfate crystal form A, and its X-ray powder diffraction pattern has a diffraction peak at 22.5 ⁇ 0.2°; or has a diffraction peak at 15.9 ⁇ 0.2° or have a diffraction peak at 22.3 ⁇ 0.2°; or have a diffraction peak at 16.8 ⁇ 0.2°; or have a diffraction peak at 22.9 ⁇ 0.2°; or have a diffraction peak at 32.1 ⁇ 0.2°; or have a diffraction peak at 14.0 ⁇ 0.2 or have a diffraction peak at 21.1 ⁇ 0.2°; or have a diffraction peak at 11.2 ⁇ 0.2°; or have a diffraction peak at 26.1 ⁇ 0.2°; preferably include any 2-5 of the above-mentioned diffraction peaks , or 3-5, or 3-6, or 3-8, or 5-8, or 6-8, more preferably including any 6, 7 or 8 of them;
  • the X-ray powder diffraction pattern of sulfate crystal form A contains at least one or more diffraction peaks located at 2 ⁇ of 22.5 ⁇ 0.2°, 15.9 ⁇ 0.2°, and 22.3 ⁇ 0.2°, preferably two of them, More preferably, it contains three; optionally, it can further include at least one of 2 ⁇ of 16.8 ⁇ 0.2°, 22.9 ⁇ 0.2°, 32.1 ⁇ 0.2°, 14.0 ⁇ 0.2°, 21.1 ⁇ 0.2°, preferably two of them , 3, 4 or 5;
  • the X-ray powder diffraction pattern of the sulfate salt crystal form A optionally further comprises 11.2 ⁇ 0.2°, 26.1 ⁇ 0.2°, 28.2 ⁇ 0.2°, 37.8 ⁇ 0.2°, 15.5 ⁇ 0.2°, One or more diffraction peaks in 26.5 ⁇ 0.2°, 36.4 ⁇ 0.2°; preferably at least include any 2-3, or 4-5, or 6-7 of them; more preferably, include at least any 2 of them , 3 places, 4 places, 5 places, 6 places, 7 places;
  • the X-ray powder diffraction pattern of the sulfate salt crystal form A includes 2 ⁇ at 22.5 ⁇ 0.2°, 15.9 ⁇ 0.2°, 22.3 ⁇ 0.2°, 16.8 ⁇ 0.2°, 22.9 ⁇ 0.2°, 32.1 ⁇ 0.2°, One or more of 14.0 ⁇ 0.2°, 21.1 ⁇ 0.2°, 11.2 ⁇ 0.2°, 26.1 ⁇ 0.2°, 28.2 ⁇ 0.2°, 37.8 ⁇ 0.2°, 15.5 ⁇ 0.2°, 26.5 ⁇ 0.2°, 36.4 ⁇ 0.2° Diffraction peaks, preferably, including optional 4, 5, 6, 8 or 10 diffraction peaks;
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridine[3,2-e] Form A of sulfate salt of pyrimidin-4(5H)-yl)-N-(5-fluoropyridin-2-yl)acetamide, X- The X-ray characteristic diffraction peaks are shown in Table 3.
  • the acid salt of the compound is sulfate crystal form B, and its X-ray powder diffraction pattern has a diffraction peak at 15.3 ⁇ 0.2°; or has a diffraction peak at 21.5 ⁇ 0.2° or have a diffraction peak at 10.6 ⁇ 0.2°; or have a diffraction peak at 19.8 ⁇ 0.2°; or have a diffraction peak at 20.1 ⁇ 0.2°; or have a diffraction peak at 12.6 ⁇ 0.2°; or have a diffraction peak at 25.2 ⁇ 0.2 or have a diffraction peak at 9.2 ⁇ 0.2°; or have a diffraction peak at 9.9 ⁇ 0.2°; or have a diffraction peak at 23.4 ⁇ 0.2°; preferably include any 2-5 of the above-mentioned diffraction peaks , or 3-5, or 3-6, or 3-8, or 5-8, or 6-8, more preferably including any 6, 7 or 8 of them;
  • the X-ray powder diffraction pattern of sulfate crystal form B contains at least one or more diffraction peaks located at 2 ⁇ of 15.3 ⁇ 0.2°, 21.5 ⁇ 0.2°, and 10.6 ⁇ 0.2°, preferably two of them, More preferably, it contains three; optionally, it can further include at least one of 2 ⁇ of 19.8 ⁇ 0.2°, 20.1 ⁇ 0.2°, 12.6 ⁇ 0.2°, 25.2 ⁇ 0.2°, 9.2 ⁇ 0.2°, preferably two of them , 3, 4 or 5;
  • the X-ray powder diffraction pattern of the sulfate crystal form B optionally further comprises One or more diffraction peaks in 0.2°, 16.3 ⁇ 0.2°; preferably at least include any 2-3, or 4-5, or 6-7; more preferably, at least include any 2, 3 place, 4 places, 5 places, 6 places, 7 places;
  • the X-ray powder diffraction pattern of sulfate salt crystal form B includes 15.3 ⁇ 0.2°, 21.5 ⁇ 0.2°, 10.6 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.1 ⁇ 0.2°, 12.6 ⁇ 0.2°, One or more of 25.2 ⁇ 0.2°, 9.2 ⁇ 0.2°, 9.9 ⁇ 0.2°, 23.4 ⁇ 0.2°, 6.3 ⁇ 0.2°, 16.7 ⁇ 0.2°, 23.9 ⁇ 0.2°, 33.8 ⁇ 0.2°, 16.3 ⁇ 0.2° Diffraction peaks, preferably, including optional 4, 5, 6, 8 or 10 diffraction peaks;
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridine[3,2-e] Form B of sulfate salt of pyrimidin-4(5H)-yl)-N-(5-fluoropyridin-2-yl)acetamide, X-
  • Table 4 The X-ray characteristic diffraction peaks are shown in Table 4.
  • the acid salt of the compound is hydrochloride crystal form A, and its X-ray powder diffraction pattern has a diffraction peak at 15.0 ⁇ 0.2°; or has a diffraction peak at 23.9 ⁇ 0.2° or have a diffraction peak at 9.7 ⁇ 0.2°; or have a diffraction peak at 5.3 ⁇ 0.2°; or have a diffraction peak at 24.8 ⁇ 0.2°; or have a diffraction peak at 29.5 ⁇ 0.2°; or have a diffraction peak at 7.5 ⁇ 0.2°
  • the X-ray powder diffraction pattern of hydrochloride crystal form A contains at least one or more diffraction peaks located at 2 ⁇ of 15.0 ⁇ 0.2°, 23.9 ⁇ 0.2°, 9.7 ⁇ 0.2°, preferably two of them , more preferably include three; optionally, further may include at least one of 2 ⁇ of 5.3 ⁇ 0.2°, 24.8 ⁇ 0.2°, 29.5 ⁇ 0.2°, 7.5 ⁇ 0.2°, 21.8 ⁇ 0.2°, preferably including 2 strips, 3 strips, 4 strips or 5 strips;
  • the X-ray powder diffraction pattern of the hydrochloride salt form A optionally further comprises One or more diffraction peaks in ⁇ 0.2°, 28.4 ⁇ 0.2°; preferably at least include any 2-3, or 4-5, or 6-7 of them; more preferably, at least include any 2, 3 places, 4 places, 5 places, 6 places, 7 places;
  • the X-ray powder diffraction pattern of the hydrochloride salt crystal form A contains 2 ⁇ at 15.0 ⁇ 0.2°, 23.9 ⁇ 0.2°, 9.7 ⁇ 0.2°, 5.3 ⁇ 0.2°, 24.8 ⁇ 0.2°, 29.5 ⁇ 0.2° , 7.5 ⁇ 0.2°, 21.8 ⁇ 0.2°, 21.3 ⁇ 0.2°, 10.6 ⁇ 0.2°, 16.9 ⁇ 0.2°, 16.0 ⁇ 0.2°, 18.4 ⁇ 0.2°, 25.8 ⁇ 0.2°, 28.4 ⁇ 0.2° or Multiple diffraction peaks, preferably, including optional 4, 5, 6, 8 or 10 diffraction peaks;
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridine[3,2-e] Pyrimidin-4(5H)-yl)-N-(5-fluoropyridin-2-yl)acetamide hydrochloride salt form A, using Cu-K ⁇ radiation, X in terms of 2 ⁇ angle and interplanar spacing d value - Ray characteristic diffraction peaks are shown in Table 5.
  • the acid salt of the compound is hydrochloride crystal form B
  • the X-ray powder diffraction pattern of hydrochloride crystal form B has a diffraction peak at 15.9 ⁇ 0.2°; or at 22.2° Diffraction peak at ⁇ 0.2°; or diffraction peak at 5.2 ⁇ 0.2°; or diffraction peak at 21.7 ⁇ 0.2°; or diffraction peak at 26.0 ⁇ 0.2°; or diffraction peak at 4.6 ⁇ 0.2° or have a diffraction peak at 28.4 ⁇ 0.2°; or have a diffraction peak at 9.2 ⁇ 0.2°; or have a diffraction peak at 17.3 ⁇ 0.2°; or have a diffraction peak at 15.2 ⁇ 0.2°; preferably include the above-mentioned diffraction Any 2-5, or 3-5, or 3-6, or 3-8, or 5-8, or 6-8 of the peaks, more preferably including any 6, 7 or 8 places;
  • the X-ray powder diffraction pattern of hydrochloride crystal form B contains at least one or more diffraction peaks located at 2 ⁇ of 15.9 ⁇ 0.2°, 22.2 ⁇ 0.2°, 5.2 ⁇ 0.2°, preferably two of them , more preferably include three; optionally, further may include at least one of 2 ⁇ at 21.7 ⁇ 0.2°, 26.0 ⁇ 0.2°, 4.6 ⁇ 0.2°, 28.4 ⁇ 0.2°, 9.2 ⁇ 0.2°, preferably including 2 strips, 3 strips, 4 strips or 5 strips;
  • the X-ray powder diffraction pattern of the hydrochloride salt crystal form B optionally further comprises a position at 2 ⁇ of 17.3 ⁇ 0.2°, 15.2 ⁇ 0.2°, 10.5 ⁇ 0.2°, 38.0 ⁇ 0.2°, 20.3 ⁇ 0.2° , 23.8 ⁇ 0.2°, 29.5 ⁇ 0.2°, one or more diffraction peaks; preferably at least include any 2-3, or 4-5, or 6-7; more preferably, at least include any 2 place, 3 places, 4 places, 5 places, 6 places, 7 places;
  • the X-ray powder diffraction pattern of the hydrochloride salt crystal form B includes 2 ⁇ at 15.9 ⁇ 0.2°, 22.2 ⁇ 0.2°, 5.2 ⁇ 0.2°, 21.7 ⁇ 0.2°, 26.0 ⁇ 0.2°, 4.6 ⁇ 0.2° , 28.4 ⁇ 0.2°, 9.2 ⁇ 0.2°, 17.3 ⁇ 0.2°, 15.2 ⁇ 0.2°, 10.5 ⁇ 0.2°, 38.0 ⁇ 0.2°, 20.3 ⁇ 0.2°, 23.8 ⁇ 0.2°, 29.5 ⁇ 0.2°, or Multiple diffraction peaks, preferably, including optional 4, 5, 6, 8 or 10 diffraction peaks;
  • the acid salt crystal form of the compound is hydrobromide salt crystal form A, and its X-ray powder diffraction pattern has a diffraction peak at 5.3 ⁇ 0.2°; or at 22.7 ⁇ 0.2° or have a diffraction peak at 14.8 ⁇ 0.2°; or have a diffraction peak at 10.5 ⁇ 0.2°; or have a diffraction peak at 22.5 ⁇ 0.2°; or have a diffraction peak at 28.0 ⁇ 0.2°; Or have a diffraction peak at 30.0 ⁇ 0.2°; or have a diffraction peak at 23.4 ⁇ 0.2°; or have a diffraction peak at 23.3 ⁇ 0.2°; or have a diffraction peak at 26.5 ⁇ 0.2°; preferably include the above-mentioned diffraction peaks Any 2-5, or 3-5, or 3-6, or 3-8, or 5-8, or 6-8, more preferably including any 6, 7 or 8 of them ;
  • the X-ray powder diffraction pattern of hydrobromide crystal form A contains at least one or more diffraction peaks located at 2 ⁇ of 5.3 ⁇ 0.2°, 22.7 ⁇ 0.2°, and 14.8 ⁇ 0.2°, preferably two of them Strips, more preferably three strips;
  • at least one of 2 ⁇ of 10.5 ⁇ 0.2°, 22.5 ⁇ 0.2°, 28.0 ⁇ 0.2°, 30.0 ⁇ 0.2°, 23.4 ⁇ 0.2° can be included, preferably included 2, 3, 4 or 5 strips;
  • the X-ray powder diffraction pattern of the hydrobromide salt crystal form A optionally further comprises a position at 2 ⁇ of 23.3 ⁇ 0.2°, 26.5 ⁇ 0.2°, 34.9 ⁇ 0.2°, 15.8 ⁇ 0.2°, 25.0 ⁇ 0.2°, One or more diffraction peaks in 31.9 ⁇ 0.2°, 37.0 ⁇ 0.2°; preferably at least include any 2-3, or 4-5, or 6-7; more preferably, include any 2, 3 places, 4 places, 5 places, 6 places, 7 places;
  • the X-ray powder diffraction pattern of the hydrobromide salt crystal form A comprises 2 ⁇ at 5.3 ⁇ 0.2°, 22.7 ⁇ 0.2°, 14.8 ⁇ 0.2°, 10.5 ⁇ 0.2°, 22.5 ⁇ 0.2°, 28.0 ⁇ 0.2°, 30.0 ⁇ 0.2°, 23.4 ⁇ 0.2°, 23.3 ⁇ 0.2°, 26.5 ⁇ 0.2°, 34.9 ⁇ 0.2°, 15.8 ⁇ 0.2°, 25.0 ⁇ 0.2°, 31.9 ⁇ 0.2°, 37.0 ⁇ 0.2°
  • One or more diffraction peaks preferably, including optional 4, 5, 6, 8 or 10 diffraction peaks;
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridine[3,2-e] Pyrimidin-4(5H)-yl)-N-(5-fluoropyridin-2-yl)acetamide hydrobromide salt form A using Cu-K ⁇ radiation, expressed in 2 ⁇ angle and interplanar spacing d value X-ray characteristic diffraction peaks are shown in Table 7.
  • the acid salt of the compound is hydrobromide crystal form B, and its X-ray powder diffraction pattern has a diffraction peak at 23.4 ⁇ 0.2°; or has a diffraction peak at 15.9 ⁇ 0.2° Diffraction peak; or have a diffraction peak at 16.2 ⁇ 0.2°; or have a diffraction peak at 14.2 ⁇ 0.2°; or have a diffraction peak at 5.3 ⁇ 0.2°; or have a diffraction peak at 10.6 ⁇ 0.2°; or have a diffraction peak at 23.1
  • the X-ray powder diffraction pattern of hydrobromide salt crystal form B includes at least one or more diffraction peaks at 2 ⁇ of 23.4 ⁇ 0.2°, 15.9 ⁇ 0.2°, and 16.2 ⁇ 0.2°, preferably including Two of them, more preferably including three;
  • it can further include at least one of 2 ⁇ being 14.2 ⁇ 0.2°, 5.3 ⁇ 0.2°, 10.6 ⁇ 0.2°, 23.1 ⁇ 0.2°, 24.1 ⁇ 0.2°, preferably Contains 2, 3, 4 or 5 of these;
  • the X-ray powder diffraction pattern of the hydrobromide salt crystal form B optionally further comprises a position at 2 ⁇ of 14.8 ⁇ 0.2°, 9.5 ⁇ 0.2°, 16.9 ⁇ 0.2°, 13.9 ⁇ 0.2°, 29.5 ⁇ 0.2°, One or more diffraction peaks in 32.2 ⁇ 0.2°, 22.2 ⁇ 0.2°; preferably at least include any 2-3, or 4-5, or 6-7; more preferably, include any 2, 3 places, 4 places, 5 places, 6 places, 7 places;
  • the X-ray powder diffraction pattern of the hydrobromide salt crystal form B includes 2 ⁇ at 23.4 ⁇ 0.2°, 15.9 ⁇ 0.2°, 16.2 ⁇ 0.2°, 14.2 ⁇ 0.2°, 5.3 ⁇ 0.2°, 10.6 ⁇ 0.2 °, 23.1 ⁇ 0.2°, 24.1 ⁇ 0.2°, 14.8 ⁇ 0.2°, 9.5 ⁇ 0.2°, 16.9 ⁇ 0.2°, 13.9 ⁇ 0.2°, 29.5 ⁇ 0.2°, 32.2 ⁇ 0.2°, 22.2 ⁇ 0.2° or multiple diffraction peaks, preferably, including optional 4, 5, 6, 8 or 10 diffraction peaks;
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridine[3,2-e] Pyrimidin-4(5H)-yl)-N-(5-fluoropyridin-2-yl)acetamide hydrobromide salt form B using Cu-K ⁇ radiation, expressed in 2 ⁇ angle and interplanar spacing d value X-ray characteristic diffraction peaks are shown in Table 8.
  • the acid salt of the compound is hydrobromide crystal form C, and its X-ray powder diffraction pattern has a diffraction peak at 5.2 ⁇ 0.2°; or has a diffraction peak at 15.7 ⁇ 0.2°; or Have a diffraction peak at 22.3 ⁇ 0.2°; or have a diffraction peak at 10.5 ⁇ 0.2°; or have a diffraction peak at 17.4 ⁇ 0.2°; or have a diffraction peak at 38.0 ⁇ 0.2°; or have a diffraction peak at 26.3 ⁇ 0.2°
  • the X-ray powder diffraction pattern of hydrobromide crystal form C contains at least one or more diffraction peaks located at 2 ⁇ of 5.2 ⁇ 0.2°, 15.7 ⁇ 0.2°, and 22.3 ⁇ 0.2°, preferably two of them Strips, more preferably three strips;
  • at least one of 2 ⁇ of 10.5 ⁇ 0.2°, 17.4 ⁇ 0.2°, 38.0 ⁇ 0.2°, 26.3 ⁇ 0.2°, 28.0 ⁇ 0.2° can be included, preferably included 2, 3, 4 or 5 strips;
  • the X-ray powder diffraction pattern of the hydrobromide salt crystal form C comprises 5.2 ⁇ 0.2°, 15.7 ⁇ 0.2°, 22.3 ⁇ 0.2°, 10.5 ⁇ 0.2°, 17.4 ⁇ 0.2°, 38.0 ⁇ 0.2 °, 26.3 ⁇ 0.2°, 28.0 ⁇ 0.2°, one or more diffraction peaks, preferably, including optional 4, 5, 6 or 8 diffraction peaks;
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridine[3,2-e] Pyrimidin-4(5H)-yl)-N-(5-fluoropyridin-2-yl)acetamide hydrobromide salt form C, using Cu-K ⁇ radiation, expressed in 2 ⁇ angle and interplanar spacing d value X-ray characteristic diffraction peaks are shown in Table 9.
  • the relative peak intensity is the top ten diffraction peak positions and the 2 ⁇ error of the diffraction peak at the corresponding position in Figure 1 is ⁇ 0.2° ⁇ 0.5°, preferably ⁇ 0.2° ⁇ 0.3°, most preferably ⁇ 0.2°.
  • a in the X-ray powder diffraction pattern relative peak intensity is the top ten diffraction peak positions and
  • the 2 ⁇ error of the diffraction peak corresponding to the position in Fig. 1 is ⁇ 0.2° ⁇ 0.5°, preferably ⁇ 0.2° ⁇ 0.3°, most preferably ⁇ 0.2°;
  • the relative peak intensity is the top ten diffraction peak positions and the corresponding positions in Figure 4
  • the 2 ⁇ error of the peak is ⁇ 0.2° ⁇ 0.5°, preferably ⁇ 0.2° ⁇ 0.3°, most preferably ⁇ 0.2°;
  • the error is ⁇ 0.2° ⁇ 0.5°, preferably ⁇ 0.2° ⁇ 0.3°, most preferably ⁇ 0.2°;
  • the error is ⁇ 0.2° ⁇ 0.5°, preferably ⁇ 0.2° ⁇ 0.3°, most preferably ⁇ 0.2°;
  • the relative peak intensity is the position of the top ten diffraction peaks and the corresponding position of the diffraction peaks in Figure 12
  • the 2 ⁇ error is ⁇ 0.2° ⁇ 0.5°, preferably ⁇ 0.2° ⁇ 0.3°, most preferably ⁇ 0.2°;
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl )-N-(5-fluoropyridin-2-yl)acetamide hydrochloride crystal form B X-ray powder diffraction pattern relative peak intensity for the top ten diffraction peak position and the corresponding position of the diffraction peak in Figure 15
  • the 2 ⁇ error is ⁇ 0.2° ⁇ 0.5°, preferably ⁇ 0.2° ⁇ 0.3°, most preferably ⁇ 0.2°;
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl )-N-(5-fluoropyridin-2-yl) acetamide hydrobromide crystal form A X-ray powder diffraction pattern relative peak intensity for the top ten diffraction peak positions and corresponding position diffraction peaks in Figure 16
  • the 2 ⁇ error is ⁇ 0.2° ⁇ 0.5°, preferably ⁇ 0.2° ⁇ 0.3°, most preferably ⁇ 0.2°;
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl )-N-(5-fluoropyridin-2-yl)acetamide hydrobromide crystal form B X-ray powder diffraction pattern relative peak intensity for the top ten diffraction peak positions and corresponding position diffraction peaks in Figure 18
  • the 2 ⁇ error is ⁇ 0.2° ⁇ 0.5°, preferably ⁇ 0.2° ⁇ 0.3°, most preferably ⁇ 0.2°;
  • the compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl In the X-ray powder diffraction pattern of the hydrobromide crystal form C of )-N-(5-fluoropyridin-2-yl)acetamide, the relative peak intensity is the top ten diffraction peak positions and the corresponding position diffraction peaks in Figure 21
  • the 2 ⁇ error is ⁇ 0.2° ⁇ 0.5°, preferably ⁇ 0.2° ⁇ 0.3°, most preferably ⁇ 0.2°.
  • the acid salt crystal form of the above compound is a hydrate or anhydrate, and when the acid salt crystal form is a hydrate, the number of water is 0.2-3, preferably 0.2, 0.5, 1 , 1.5, 2, 2.5 or 3, more preferably 0.5, 1, 2 or 3; further, the water in the hydrate is pipeline water or crystal water or a combination of both.
  • the preparation method of the above-mentioned acid salt comprises the following steps:
  • the solvent is an organic solvent, preferably methanol, ethanol, tetrahydrofuran, 2-methyltetrahydrofuran, toluene, isopropyl acetate, tert-butanol, n-butanol, acetone, 2-butanone, dichloromethane, ethyl acetate or 1, At least one of 4-dioxane;
  • Acid selected from hydrochloric acid, sulfuric acid, nitric acid, hydrobromic acid, hydrofluoric acid, hydroiodic acid, phosphoric acid, 2,5-dihydroxybenzoic acid, 1-hydroxy-2-naphthoic acid, acetic acid, dichloroacetic acid, trichloroacetic acid , Acetohydroxamic Acid, Adipic Acid, Benzenesulfonic Acid, 4-Chlorobenzenesulfonic Acid, Benzoic Acid, 4-Acetamidobenzoic Acid, 4-Aminobenzoic Acid, Capric Acid, Caproic Acid, Caprylic Acid, Cinnamic Acid, Citric Acid , Cyclohexanesulfamic acid, camphorsulfonic acid, aspartic acid, camphoric acid, gluconic acid, glucuronic acid, glutamic acid, isoascorbic acid, lactic acid, malic acid, mandelic acid, pyroglutamic acid, tartaric
  • the preparation method of the acid salt of the above-mentioned compound and its crystal form comprises the following steps:
  • the solvent is an organic solvent, preferably methanol, ethanol, tetrahydrofuran, 2-methyltetrahydrofuran, toluene, isopropyl acetate, tert-butanol, n-butanol, acetone, 2-butanone, dichloromethane, ethyl acetate or 1, At least one of 4-dioxane;
  • Acid selected from hydrochloric acid, sulfuric acid, nitric acid, hydrobromic acid, hydrofluoric acid, hydroiodic acid, phosphoric acid, 2,5-dihydroxybenzoic acid, 1-hydroxy-2-naphthoic acid, acetic acid, dichloroacetic acid, trichloroacetic acid , Acetohydroxamic Acid, Adipic Acid, Benzenesulfonic Acid, 4-Chlorobenzenesulfonic Acid, Benzoic Acid, 4-Acetamidobenzoic Acid, 4-Aminobenzoic Acid, Capric Acid, Caproic Acid, Caprylic Acid, Cinnamic Acid, Citric Acid , Cyclohexanesulfamic acid, camphorsulfonic acid, aspartic acid, camphoric acid, gluconic acid, glucuronic acid, glutamic acid, isoascorbic acid, lactic acid, malic acid, mandelic acid, pyroglutamic acid, tartaric
  • the present invention also provides a preferred solution, and also relates to a pharmaceutical composition, which contains a therapeutically effective amount of the acid salt of the above compound or its crystal form, and one or more pharmaceutically acceptable carriers and diluents or excipients.
  • the present invention further relates to the use of any acid salt of the compound of general formula (I-a) or its crystal form or the pharmaceutical composition in the preparation of P2X3 inhibitor drugs.
  • the pharmaceutically acceptable salts of the compounds of the present invention and their crystal forms or compositions are used in the preparation of drugs for the treatment of neurogenic diseases; preferably, the neurogenic diseases are selected from gynecological diseases, urinary tract A disease state, a respiratory disorder or a pain-related disease or condition, more preferably endometriosis, overactive bladder, pulmonary fibrosis or chronic cough.
  • said pain-related disease or condition is selected from neuropathic pain or pain or discomfort associated with uterine fibroids.
  • Another object of the present invention is to provide a method for preparing pyrazole-containing polycyclic derivatives
  • general formula (I) is:
  • R 1 , R 2 , and R 3 are independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxyl, cyano, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy radical, alkenyl, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl, aryloxy, heteroaryl, heteroaryloxy or -(CH 2 ) n1 C(O) R a ;
  • R 4 and R 5 are independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxyl, cyano, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl radical, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl, aryloxy, heteroaryl, heteroaryloxy, or -( CH2 ) n1C (O) Ra ;
  • R is selected from hydrogen or a protecting group
  • the protecting group can be an amino protecting group, such as a common amino protecting group, preferably tert-butoxycarbonyl, benzyloxycarbonyl, 2-biphenyl-2-propoxycarbonyl, p-toluenesulfonyl, trityl, methyl Acyl, trifluoroacetyl, etc.;
  • the compound of the general formula (I) whose R is a protecting group it can be obtained by reacting the compound of the general formula (I) whose R is H with a corresponding protecting reagent, and the protecting group can be removed as required;
  • R is selected from hydrogen, deuterium , halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, hetero Cycloalkyl, cycloalkyl, heterocyclyl, aryl, aryloxy, heteroaryl or heteroaryloxy;
  • n1 0, 1, 2, 3 or 4;
  • R 5 is not -C(CH 3 ) 3 or -COOCH 3 ;
  • R 1 is -CF 3
  • R 2 , R 3 , R 4 and R 6 are hydrogen at the same time
  • R 5 is not -CH 2 CH 3 , H or Br
  • R 4 is not -CN.
  • the present invention provides a compound of general formula (I-1), general formula (I-1) is:
  • the structure of the compound is selected from formula (I-1-1), formula (I-1-2) or formula (I-1-3):
  • R is selected from hydrogen, deuterium, halogen, cyano, C 1-8 alkyl , C 1-8 alkoxy, C 1-8 haloalkyl or C 1-8 haloalkoxy;
  • cyano Preferably cyano, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl or C 1-3 haloalkoxy;
  • R is selected from hydrogen, deuterium , halogen, cyano, C 1-8 alkyl, C 1-8 alkoxy, C 1-8 haloalkyl, C 1-8 haloalkoxy or -C (O) R a ;
  • R is selected from hydrogen, deuterium , halogen, amino, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl or C 1-3 haloalkoxy;
  • the present invention provides a kind of preparation method of the compound of general formula (I) or the compound of general formula (I '), comprising step (a):
  • the reaction is optionally carried out in the presence of an acid, the acid is preferably hydrochloric acid, more preferably an organic solution of hydrochloric acid, the concentration of hydrochloric acid in the organic solution of hydrochloric acid can be 1 to 10M/L, preferably 3 to 6M /L, more preferably 4M/L, the molar ratio of the acid to the compound of general formula (II) is more than 1, preferably 1 to 5:1, more preferably 3:1;
  • the reaction is optionally carried out in a solvent, the solvent is preferably an organic solvent, and the concentration of the compound of general formula (II) in the solvent is 0.05g/ml ⁇ 0.5g/ml;
  • the organic solvent is preferably dioxane, tetrahydrofuran, toluene, methyl tetrahydrofuran, ethyl acetate, trimethylbenzene, ethylene glycol, methanol, ethanol, isopropanol or dimethyl ether, more preferably dioxane;
  • the organic solvent can also preferably be N,N dimethylformamide, N,N dimethylacetamide, N-methylpyrrolidone, dimethyl sulfoxide, more preferably N,N dimethylformamide;
  • the molar ratio of the compound of general formula (III) to the compound of general formula (II) may be 0.5-5.0:1, preferably 1-1.5:1, more preferably 1.2:1;
  • the reaction temperature may be -20-200°C, preferably 50-150°C, more preferably 80-120°C, further preferably 105-110°C;
  • the reaction time may be 1 to 48 hours, preferably 10 to 30 hours, more preferably 16 to 24 hours;
  • the M 1 and M 2 are independently selected from H, Li, Na, K or Cs, M 1 is preferably H, and M 2 is preferably Na or K;
  • R 1 , R 2 , R 3 , R 4 , and R 5 are as described for the compound of general formula (I) or (I-1).
  • the present invention also provides a method for preparing a compound of general formula (VII), comprising step (b):
  • the compound of general formula (I) reacts with the compound of general formula (VI) to obtain the compound of general formula (VII);
  • the molar ratio of the compound of the general formula (VI) to the compound of the general formula (I) may be 0.8-3:1, preferably 0.9-1.2:1, more preferably 1:1;
  • reaction is optionally carried out in the presence of a base
  • reaction is optionally carried out in a solvent
  • the base is preferably one or more of organic bases and inorganic bases; more preferably one or more of sodium carbonate, potassium carbonate, sodium hydroxide, potassium hydroxide, triethylamine, DIPEA, DBU, DABCO , the molar ratio of the base to the compound of general formula (I) is more than 1, preferably 1 to 10:1, more preferably 2:1;
  • the solvent is preferably an organic solvent, more preferably one or more of DMF, DMA, THF; further preferably one or more of DMF, DMA, the concentration of the compound of general formula (I) in the solvent 0.05g/ml ⁇ 0.5g/ml;
  • the temperature of the reaction may be 0-50°C, preferably 20-25°C or 45°C;
  • X is halogen; preferably fluorine, chlorine or bromine; more preferably chlorine or bromine;
  • L 1 is selected from -(CH 2 ) n2 -, -(CH 2 ) n2 O-, -(CH 2 ) n2 S-, -(CH 2 ) n2 NR c -, -(CH 2 ) n2 C(O) NR c -or -(CH 2 ) n2 NR c C(O)-; preferably -CH 2 C(O)NH-;
  • R b and R c are independently selected from hydrogen, deuterium, halogen, amino, nitro, hydroxyl, cyano, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl radical, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl, aryloxy, heteroaryl or heteroaryloxy;
  • the amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, heterocyclylalkyl, cycloalkyl, heterocyclyl, aryl , aryloxy, heteroaryl, heteroaryloxy, -(CH 2 ) n2 -(-(CH 2 ) n2 - can also be -(CH 2 ) n2 O-, -(CH 2 ) n2 S -, -(CH 2 ) n 2 NR c -, -(CH 2 ) n 2 C(O)NR c - or -(CH 2 ) n 2 NR c C(O)- the alkylene chain in), optionally Hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl,
  • Ring A is selected from cycloalkyl, heterocyclyl, aryl or heteroaryl, preferably and / or,
  • the compound of general formula (VI) is preferably more preferably is further preferably
  • R 1 , R 2 , R 3 , R 4 , and R 5 are as described in the compound of general formula (I) or (I-1);
  • the preparation method of the compound of general formula (VII) further includes the step of preparing the compound of general formula (I).
  • the present invention also provides a method for preparing a compound of general formula (I-3), comprising step (c-1):
  • the compound of the general formula (I-2) is subjected to an amination reduction reaction under the condition of ammonia gas or an ammonia gas equivalent to obtain a compound of the general formula (I-3);
  • the ammonia gas equivalent is an organic solution of ammonia or ammonia water;
  • the organic solution of ammonia is preferably a methanol solution of ammonia, an ethanol solution of ammonia, an isopropanol solution of ammonia or a dioxane solution of ammonia;
  • the compound of general formula (I-2) carries out amination reduction reaction under the condition of ammoniacal liquor and/or the organic solution of ammonia, obtains the compound of general formula (I-3); More preferably, general formula (I- 2) the compound is subjected to an amination reduction reaction under the conditions of ammonia water and/or methanol solution of ammonia to obtain a compound of general formula (I-3);
  • the reaction temperature can be 0-50°C, preferably room temperature 25°C;
  • the reaction time is 1 to 72 hours, preferably 48 to 60 hours;
  • R a is selected from hydrogen, deuterium, halogen, hydroxyl, alkoxy, haloalkoxy, aryloxy or heteroaryloxy; the alkoxy, haloalkoxy, aryloxy, heteroaryl Oxygen, optionally replaced by hydrogen, deuterium, halogen, amino, nitro, hydroxy, cyano, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, alkenyl, alkyne
  • R 1 , R 2 , R 3 , R 4 , and R 6 are as described for the compound of general formula (I) or (I-1).
  • the present invention also provides a method for preparing a compound of general formula (I-4), comprising step (d-1):
  • the dehydrating agent is preferably one or more of acetic anhydride, trifluoroacetic anhydride, P 2 O 5 , cyanuric chloride, phosphorus oxychloride, phosphorus trichloride, and concentrated sulfuric acid.
  • the molar ratio of the compound of formula (I-3) is 1 or more, preferably 1-10:1, more preferably 2-2.5:1;
  • the reaction is optionally carried out in the presence of pyridine, and the molar ratio of the pyridine to the compound of general formula (I-3) is 1 or more, preferably 1-10:1, more preferably 2.5-3:1; More preferably 3:1;
  • the reaction is optionally carried out in a solvent;
  • the solvent is preferably an organic solvent, more preferably one or more of DMF, DMA, THF; further preferably one or more of DMF, THF, so
  • the compound concentration of general formula (I-3) in the solvent is 0.05g/ml ⁇ 0.5g/ml;
  • the temperature of the reaction may be -20 to 80°C, preferably room temperature;
  • the reaction time is 0.1 to 10 hours, preferably 0.5 to 4 hours, more preferably 1 to 2 hours;
  • R 1 , R 2 , R 3 , R 4 , and R 6 are as defined in general formula (I) or (I -1) Compounds described.
  • the present invention also relates to a preparation method of a compound of general formula (VII-4), characterized in that, the structure of the compound of general formula (VII-4) is:
  • L 1 , x, R b , ring A, R 1 , R 2 , R 3 , and R 4 are as described for the compound of general formula (I) or (I-1) and the compound of general formula (VII);
  • Described preparation method comprises the following steps:
  • the present invention also provides a compound of the general formula (I), a compound of the general formula (II) and a compound of the general formula (III), the compound of the general formula (I), the compound of the general formula (II) and the compound of the general formula
  • the compound of (III) is used as an intermediate for the preparation of P2X3 inhibitors, such as substances with P2X3 inhibitory activity disclosed in patent PCT/CN2020/134264, which is incorporated in this application in its entirety.
  • the P2X3 inhibitor is preferably a P2X3 inhibitor containing a core structure of 4,5-dihydropyrazolo[1,5-a]pyrido[3,2-e]pyrimidine, more preferably a general formula (VII- 4) compound;
  • L 1 , x, R b , ring A, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , M 1 , M 2 are defined as compounds of general formula (I) or (I-1), The compound of general formula (I') and the compound of general formula (VII) are described.
  • the present invention also relates to the preparation method of the compound of general formula (II') or the compound of general formula (V'), the method also includes step (e) and/or step (c); or optionally further includes step (g),
  • the base is preferably LDA, butyllithium or hydroxide Sodium etc.;
  • the solvent is preferably tetrahydrofuran, toluene, ethylene glycol dimethyl ether or dichloromethane;
  • the molar ratio of the compound of formula (II-1) to the base is 1:1-10, preferably 1:1-5, more preferably 1:1-1.5;
  • the solvent is preferably hydrazine hydrate, methanol, ethanol, isopropanol or tert-butanol;
  • the molar ratio of the compound of formula (II-2) to hydrazine hydrate is 1:1-10, preferably 1:1-5, more preferably 1:1-1.5;
  • the molar ratio of the compound of formula (V-1) to acetonitrile is 1:1-10, preferably 1:1-5, more preferably 1:1-1.5;
  • R 1 , R 2 , R 3 , R 4 , and R 5 are as described for the compound of general formula (I) or (I-1).
  • the preparation method of pyrazole-containing polycyclic derivatives of the present invention avoids the use of expensive raw materials such as pyrazolamide derivatives and noble metal catalysts, the reaction raw materials are cheap and easy to obtain, the cost is low, the reaction conditions are mild, and the yield is high. Mature, stable quality, suitable for industrial scale-up, more in line with the requirements of safety and environmental protection.
  • alkyl refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing 1 to 20 carbon atoms, preferably an alkyl group containing 1 to 8 carbon atoms, more preferably 1 to 6 carbon atoms An alkyl group, most preferably an alkyl group of 1 to 3 carbon atoms.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1 ,2-Dimethylpropyl, 2,2-Dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2- Methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3 -Dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2 -Methylhexyl, 3-methylhexyl, 4-methylhe
  • lower alkyl groups containing 1 to 6 carbon atoms include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl Base, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethyl Dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl group, 2,3-dimethylbutyl group, etc.
  • Alkyl groups may be substituted or unsubstituted, and when substituted, substituents may be substituted at any available point of attachment, said substituents being preferably one or more of the following groups independently selected from alkyl radical, alkenyl, alkynyl, alkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkane Oxygen, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo, carboxyl or carboxylate, preferably methyl, ethyl, isopropyl, tert-butyl, haloalkyl in the present invention , deuterated alkyl, alkoxy substituted alkyl and hydroxy substituted alkyl.
  • Deuterated alkyl refers to an alkyl group in which one or more hydrogens have been replaced by deuterium, wherein alkyl is as defined above.
  • alkylene means that one hydrogen atom of the alkyl group is further substituted, for example: "methylene” means -CH 2 -, "ethylene” means -(CH 2 ) 2 -, “propylene” refers to -(CH 2 ) 3 -, “butylene” refers to -(CH 2 ) 4 -, and the like.
  • alkenyl means an alkyl group as defined above consisting of at least two carbon atoms and at least one carbon-carbon double bond, for example vinyl, 1-propenyl, 2-propenyl, 1-, 2- or 3- -butenyl etc.
  • Alkenyl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring containing 3 to 20 carbon atoms, preferably containing 3 to 12 carbon atoms, more preferably containing 3 to 6 carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene group, cyclooctyl group, etc.; polycyclic cycloalkyl group includes spiro ring, fused ring and bridged ring cycloalkyl group, preferably cyclopropyl group, cyclobutyl group, cyclohexyl group, cyclopentyl group and cycloheptyl group.
  • the cycloalkyl ring may be fused to an aryl, heteroaryl or heterocycloalkyl ring where the ring bonded to the parent structure is a cycloalkyl, non-limiting examples include indanyl, tetrahydronaphthalene base, benzocycloheptyl, etc.
  • Cycloalkyl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, oxo, carboxyl or carboxylate.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent comprising 3 to 20 ring atoms, one or more of which is selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), but excluding ring portions of -OO-, -OS- or -SS-, the remaining ring atoms being carbon.
  • the membered heterocyclic group is optionally substituted by 1-2 oxygen atoms, sulfur atoms, or oxo groups, including nitrogen-containing monocyclic heterocyclic groups, nitrogen-containing spiroheterocyclic groups or nitrogen-containing condensed heterocyclic groups.
  • Non-limiting examples of monocyclic heterocyclyl groups include oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, tetrahydropyranyl, dihydroimidazolyl, Dihydrofuryl, dihydropyrazolyl, dihydropyrrolyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, azeptyl, 1,4-diazepine Cycloheptyl, pyranyl or tetrahydrothiopyranyl dioxide, etc., preferably oxetanyl, thietanyl, tetrahydrofuryl, tetrahydropyranyl, tetrahydrothiophenyl, tetrahydrothiopyranyl , tetrahydrothio
  • the heterocyclyl ring may be fused to an aryl, heteroaryl, or cycloalkyl ring where the ring bonded to the parent structure is a heterocyclyl, non-limiting examples of which include:
  • Heterocyclic groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alk Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, oxo, carboxyl or carboxylate.
  • aryl refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic (that is, rings sharing adjacent pairs of carbon atoms) group, preferably 6 to 12 membered, having a conjugated pi-electron system, such as benzene base and naphthyl. Phenyl is more preferred.
  • the aryl ring can be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, including benzo 5-10 membered heteroaryl, benzo 3-8 membered cycloalkyl and benzo 3-8 membered Heteroalkyl, preferably benzo 5-6 membered heteroaryl, benzo 3-6 membered cycloalkyl and benzo 3-6 membered heteroalkyl, wherein the heterocyclic group contains 1-3 nitrogen atoms, oxygen atoms, A heterocyclic group with a sulfur atom; or a three-membered nitrogen-containing condensed ring containing a benzene ring.
  • Aryl groups may be substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkylthio, Alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycle Alkylthio, carboxyl or carboxylate.
  • Aryloxy means -O-(aryl), wherein aryl is as defined above.
  • heteroaryl refers to a heteroaromatic system comprising 1 to 4 heteroatoms, 5 to 14 ring atoms, wherein the heteroatoms are selected from oxygen, sulfur and nitrogen.
  • Heteroaryl is preferably 5 to 12 membered, more preferably 5 or 6 membered, such as imidazolyl, furyl, thienyl, thiazolyl, pyrazolyl, oxazolyl, pyrrolyl, triazolyl, tetrazolyl , pyridyl, pyrimidyl, thiadiazole, pyridazinyl, pyrazinyl, etc., preferably pyridyl, oxadiazolyl, triazolyl, thienyl, imidazolyl, pyrazolyl, oxazolyl, pyrimidinyl, furyl, thienyl, pyridazinyl, pyrazinyl or thiazoly
  • Heteroaryl groups may be optionally substituted or unsubstituted, and when substituted, the substituents are preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkane Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, carboxyl or carboxylate.
  • Heteroaryloxy means -O-(heteroaryl), wherein heteroaryl is as defined above.
  • alkoxy refers to -O-(alkyl) and -O-(unsubstituted cycloalkyl), wherein alkyl is as defined above.
  • alkoxy include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy, alkoxy can is optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkane Amino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkane Thio group, carboxyl
  • alkylthio refers to -S-(alkyl) and -S-(unsubstituted cycloalkyl), wherein alkyl is as defined above.
  • alkylthio include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy
  • alkylthio can be is optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkane Amino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkane
  • Haloalkyl means an alkyl group substituted by one or more halogens, wherein alkyl is as defined above; eg trifluoromethyl.
  • Haloalkoxy means an alkoxy group substituted with one or more halogens, wherein alkoxy group is as defined above.
  • Hydroalkyl means an alkyl group substituted with a hydroxy group, wherein alkyl is as defined above.
  • alkenyl refers to alkenyl, also known as alkenyl, wherein said alkenyl can be further substituted by other related groups, such as: hydrogen, deuterium, amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkane radical, alkenyl, alkynyl, alkoxy, haloalkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl group, heterocyclyl, aryloxy, heteroaryloxy, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate.
  • Alkynyl refers to (CH ⁇ C-), wherein said alkynyl group can be further substituted by other related groups, such as: hydrogen, deuterium, amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkenyl, alkynyl, alkoxy, haloalkoxy, alkylthio, alkylamino, halogen, mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, Heterocyclyl, aryloxy, heteroaryloxy, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxyl or carboxylate.
  • other related groups such as: hydrogen, deuterium, amino, alkyl, deuterated alkyl, haloalkyl, hydroxyalkyl, alkenyl, alkynyl, alkoxy
  • alkenylcarbonyl refers to -C(O)-(alkenyl), wherein alkenyl is as defined above.
  • alkenylcarbonyl include: vinylcarbonyl, propenylcarbonyl, butenylcarbonyl.
  • Alkenylcarbonyl may be optionally substituted or unsubstituted, and when substituted, the substituent is preferably one or more of the following groups independently selected from alkyl, alkenyl, alkynyl, alkoxy, alkoxy Thio, alkylamino, halogen, mercapto, hydroxyl, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio , heterocycloalkylthio, carboxyl or carboxylate.
  • Haldroxy means an -OH group.
  • Halogen means fluorine, chlorine, bromine or iodine.
  • Amino refers to -NH2 .
  • Cyano refers to -CN.
  • Niro refers to -NO2 .
  • Carbonyl refers to -C(O)-.
  • Carboxy refers to -C(O)OH.
  • Alcohol solvents refer to alkane compounds containing hydroxyl groups in their molecules, such as methanol, ethanol, and isopropanol.
  • THF tetrahydrofuran
  • EtOAc means ethyl acetate
  • MeOH means methanol
  • DMF N,N-dimethylformamide
  • TFA trifluoroacetic acid
  • MeCN refers to acetonitrile
  • DMA refers to N,N-dimethylacetamide.
  • Et2O means diethyl ether.
  • DCE 1,2 dichloroethane
  • DIPEA N,N-diisopropylethylamine
  • DBU refers to 1,8-diazabicyclo[5.4.0]undec-7-ene.
  • DABCO refers to 1,4-diazabicyclo[2.2.2]octane.
  • NBS N-bromosuccinimide
  • NIS N-iodosuccinimide
  • Cbz-Cl refers to benzyl chloroformate
  • Pd 2 (dba) 3 refers to tris(dibenzylideneacetone)dipalladium.
  • Dppf refers to 1,1'-bisdiphenylphosphinoferrocene.
  • HATU refers to 2-(7-benzotriazole oxide)-N,N,N',N'-tetramethyluronium hexafluorophosphate.
  • KHMDS refers to potassium hexamethyldisilazide
  • LiHMDS refers to lithium bistrimethylsilylamide.
  • MeLi means methyllithium
  • n-BuLi refers to n-butyllithium
  • NaBH(OAc) 3 refers to sodium triacetoxyborohydride.
  • X is selected from A, B, or C
  • X is selected from A, B, and C
  • X is A, B, or C
  • X is A, B, and C
  • many of the multiple, multiple, etc. refer to 2, 3, 4, 5, 6, 7, etc.
  • the hydrogen atoms described in the present invention can be replaced by its isotope deuterium, and any hydrogen atom in the example compounds involved in the present invention can also be replaced by a deuterium atom.
  • Optional or “optionally” means that the subsequently described event or circumstance can but need not occur, and that the description includes instances where the event or circumstance occurs or does not occur.
  • a heterocyclic group optionally substituted with an alkyl group means that an alkyl group may but need not be present, and the description includes cases where the heterocycle group is substituted with an alkyl group and cases where the heterocycle group is not substituted with an alkyl group .
  • Substituted means that one or more hydrogen atoms in a group, preferably up to 5, more preferably 1 to 3 hydrogen atoms are independently substituted by the corresponding number of substituents. It goes without saying that substituents are only in their possible chemical positions and that a person skilled in the art can determine (by experiment or theory) possible or impossible substitutions without undue effort. For example, an amino or hydroxyl group with free hydrogen may be unstable when bonded to a carbon atom with an unsaturated (eg, ethylenic) bond.
  • “Pharmaceutical composition” means a mixture containing one or more compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, and other components such as a physiologically/pharmaceutically acceptable carrier and excipients.
  • the purpose of the pharmaceutical composition is to promote the administration to the organism, facilitate the absorption of the active ingredient and thus exert biological activity.
  • “Pharmaceutically acceptable salt” refers to the salt of the compound of the present invention, which is safe and effective when used in mammals, and has proper biological activity.
  • X-ray powder diffraction pattern refers to the experimentally observed diffraction pattern or the parameters derived from it, and the X-ray powder diffraction pattern is characterized by peak position (abscissa) and peak intensity (ordinate).
  • peak position abcissa
  • peak intensity ordinate
  • the relative intensity of the X-ray diffraction pattern may also vary with the experimental conditions, so the order of peak intensities cannot be used as the only or decisive factor.
  • the overall deviation of the peak angle will be caused, and a certain deviation is usually allowed. Therefore, those skilled in the art can understand that any crystal form having the same or similar characteristic peaks as the spectrum of the present invention falls within the scope of the present invention.
  • Figure 1 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl XRPD diagram of )-N-(5-fluoropyridin-2-yl)acetamide methanesulfonate Form A.
  • Figure 2 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl DSC diagram of Form A of )-N-(5-fluoropyridin-2-yl)acetamide mesylate salt.
  • Figure 3 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl TGA diagram of Form A of )-N-(5-fluoropyridin-2-yl)acetamide mesylate salt.
  • Figure 4 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl XRPD diagram of )-N-(5-fluoropyridin-2-yl)acetamide ethanesulfonate Form A.
  • Figure 5 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl DSC diagram of Form A of )-N-(5-fluoropyridin-2-yl)acetamide ethanesulfonate salt.
  • Figure 6 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl TGA diagram of Form A of )-N-(5-fluoropyridin-2-yl)acetamide ethanesulfonate salt.
  • Figure 7 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl XRPD diagram of )-N-(5-fluoropyridin-2-yl)acetamide sulfate salt Form A.
  • Figure 8 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl DSC diagram of )-N-(5-fluoropyridin-2-yl)acetamide sulfate salt Form A.
  • Figure 9 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl XRPD diagram of )-N-(5-fluoropyridin-2-yl)acetamide sulfate salt Form B.
  • Figure 10 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl DSC diagram of )-N-(5-fluoropyridin-2-yl)acetamide sulfate salt form B.
  • Figure 11 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl TGA diagram of )-N-(5-fluoropyridin-2-yl)acetamide sulfate salt Form B.
  • Figure 12 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl XRPD diagram of )-N-(5-fluoropyridin-2-yl)acetamide hydrochloride Form A.
  • Figure 13 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl DSC diagram of )-N-(5-fluoropyridin-2-yl)acetamide hydrochloride Form A.
  • Figure 14 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl TGA diagram of )-N-(5-fluoropyridin-2-yl)acetamide hydrochloride Form A.
  • Figure 15 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl XRPD diagram of )-N-(5-fluoropyridin-2-yl)acetamide hydrochloride Form B.
  • Figure 16 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl XRPD diagram of )-N-(5-fluoropyridin-2-yl)acetamide hydrobromide salt form A.
  • Figure 17 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl DSC diagram of Form A of )-N-(5-fluoropyridin-2-yl)acetamide hydrobromide salt.
  • Figure 18 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl XRPD diagram of )-N-(5-fluoropyridin-2-yl)acetamide hydrobromide salt form B.
  • Figure 19 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl DSC diagram of )-N-(5-fluoropyridin-2-yl)acetamide hydrobromide salt form B.
  • Figure 20 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl TGA diagram of Form B of )-N-(5-fluoropyridin-2-yl)acetamide hydrobromide salt.
  • Figure 21 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl XRPD diagram of )-N-(5-fluoropyridin-2-yl)acetamide hydrobromide salt form C.
  • Figure 22 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl XRPD pattern of )-N-(5-fluoropyridin-2-yl)acetamide free base Form I.
  • Figure 23 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl DSC diagram of )-N-(5-fluoropyridin-2-yl)acetamide free base Form I.
  • Figure 24 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl XRPD diagram of )-N-(5-fluoropyridin-2-yl)acetamide free base Form II.
  • Figure 25 is 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl DSC diagram of )-N-(5-fluoropyridin-2-yl)acetamide free base Form II.
  • the structures of the compounds of the present invention are determined by nuclear magnetic resonance (NMR) or/and liquid chromatography-mass chromatography (LC-MS). NMR chemical shifts ( ⁇ ) are given in parts per million (ppm).
  • the determination of NMR is to use Bruker AVANCE-400 nuclear magnetic apparatus, and the determination solvent is deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated methanol (CD 3 OD) and deuterated chloroform (CDCl 3 ), and the internal standard is four Methylsilane (TMS).
  • Agilent 1200 Infinity Series mass spectrometer was used for LC-MS determination.
  • the determination of HPLC uses Agilent 1200DAD high pressure liquid chromatograph (Sunfire C18 150 ⁇ 4.6mm chromatographic column) and Waters 2695-2996 high pressure liquid chromatograph (Gimini C 18 150 ⁇ 4.6mm chromatographic column).
  • Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plates are used for thin-layer chromatography silica gel plates.
  • the specifications used for TLC are 0.15mm-0.20mm, and the specifications used for thin-layer chromatography separation and purification products are 0.4mm-0.5mm.
  • Column chromatography generally uses Yantai Huanghai silica gel 200-300 mesh silica gel as the carrier.
  • HPLC detection method among the present invention is as follows:
  • the starting materials in the examples of the present invention are known and commercially available, or can be synthesized using or following methods known in the art.
  • Example 1-2 To a solution of Example 1-2 (1.5 g, 6.19 mmol) in DMF (30 mL) was added potassium carbonate (4.28 g, 30.96 mmol) and Example 1-3 (4.33 g, 18.57 mmol) at room temperature. The mixture was heated to 80°C and the reaction was stirred for 2h. After cooling, water was added, the precipitate was filtered and washed with ethyl acetate, purified to obtain Example 1 (656 mg, yield: 27%).
  • Example 2 (100mg, 0.24mmol), isopropenylboronic acid (41.2mg, 0.48mmol), [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium dichloromethane complex ( 19.2mg, 0.024mmol) and cesium carbonate (232.8mg, 0.72mmol) in dioxane (4mL) and water (1mL) were stirred at 100°C for 1 hour by microwave. The reaction solution was spin-dried and purified by preparative liquid phase to obtain Example 6 (54 mg, yield 60%).
  • Step 1 Preparation of 5-oxo-4,5-dihydropyrazol[1,5-a]pyridin[3,2-e]pyrimidine-2-carboxylic acid methyl
  • Example 8-1 refers to the synthetic method of Example 1-2, replace 3-(tert-butyl)-1H-pyrazole-5 with 5-amino-1H-pyrazole-3-carboxylic acid methyl ester -Amine, to obtain Example 8-1 (500 mg, 73%).
  • Example 8-2 For the synthesis method of Example 8-2, refer to the synthesis method of Example 1, and use Example 8-1 as the raw material to obtain the title compound Example 8-2 (500 mg, 51%).
  • the third step 4-(2-((5-fluoropyridin-2-yl)amino)-2-oxoethyl)-5-oxo-4,5-dihydropyrazol[1,5-a] Preparation of pyridin[3,2-e]pyrimidine-2-carboxylic acid
  • Example 8-2 To a solution of Example 8-2 (490 mg, 1.24 mmol) in tetrahydrofuran (10 mL) was added a solution of LiOH (519 mg, 12.36 mmol) in water (2 mL) at room temperature. The mixture was stirred at room temperature for 3 h, then the pH was adjusted to about 3 with 1M HCl, and concentrated to dryness to obtain Example 8-3 (470 mg, 99%).
  • Example 8-3 450 mg, 1.2 mmol
  • Et3N 33 ⁇ L, 0.24 mmol
  • BOP reagent 598 mg, 1.35 mmol
  • Sodium azide 160 mg, 2.46 mmol
  • tetrabutylammonium bromide 786 mg, 2.46 mmol
  • the reaction was then diluted with 1,4-dioxane (12 mL), 2M aqueous H 2 SO 4 (4 mL) was added, and heated at 100° C. for 2 h.
  • Example 8 (360 mg, 86%).
  • Example 9 The synthesis method of Example 9, referring to the synthesis method of Example 6, replaced ethylene propylene boronic acid with cyclopropylboronic acid to obtain the title compound Example 9 (8mg, 51%).
  • Example 10 For the synthesis method of Example 10, refer to the synthesis method of Example 1, and use 2-chloro-6-trifluoromethylnicotinic acid as a raw material to obtain the title compound Example 10 (25 mg, 46%).
  • Example 11 The synthesis method of Example 11 was referred to the synthesis method of Example 2 to obtain the title compound Example 11 (18 mg, 30%).
  • Step 1 Preparation of tert-butyl 5-amino-3-ethyl-1H-pyrazole-1-carboxylate
  • the second step the preparation of tert-butyl 5-amino-3-ethyl-1H-pyrazole-1-carboxylate
  • Example 12-1 (3.4g, 16.1mmol) was dissolved in anhydrous dichloromethane (60mL), triethylamine (5.4g, 53.1mmol) was added, and the now-prepared 2- Chloro-6-trifluoromethylnicotinic acid chloride (4.3g, 17.7mmol) in dichloromethane solution (50mL) was added and reacted at room temperature for 30 minutes.
  • the reaction solution was washed with water (200mL*2) and saturated sodium chloride solution (200mL) successively, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure.
  • the third step the preparation of N-(3-ethyl-1H-pyrazol-5-yl)-2-chloro-6-(trifluoromethyl)nicotinamide
  • Example 12-2 Dissolve Example 12-2 (2.6g, 6.2mmol) in anhydrous dichloromethane (10mL), add dioxane hydrochloride solution (4M, 20mL), and react at room temperature for 4 hours. The reaction solution was directly spin-dried to obtain Example 12-3 (1.9 g), yield: 96.0%.
  • Example 12-3 (1.9g, 6.0mmol) was dissolved in N,N-dimethylformamide (20mL), potassium carbonate (2.5g, 18.0mmol) was added, and heated to 120°C for 2 hours. The reaction solution was cooled to room temperature and used directly in the next reaction.
  • the fifth step 2-(2-ethyl-5-oxo-8-(trifluoromethyl)pyrazol[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl )-N-(5-fluoropyridin-2-yl)acetamide
  • Example 12-4 In the N,N-dimethylformamide (20mL) reaction solution of Example 12-4 (1.0g, 3.5mmol), add potassium carbonate (1.5g, 10.6mmol) and 2-bromo-N-(5- Fluoropyridin-2-yl)acetamide (0.99g, 4.2mmol) was reacted at 40°C for 2 hours.
  • the reaction solution was cooled to room temperature, poured into 300 mL of water, and extracted with ethyl acetate (200 mL*3).
  • Example 12 The organic phases were combined, washed with water (200mL*2) and saturated sodium chloride solution (200mL) successively, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the crude product was recrystallized from ethyl acetate to obtain Example 12.
  • Example 13 The synthesis method of Example 13 was referred to the synthesis method of Example 1 to obtain the title compound Example 13 (17 mg, 28%).
  • Example 14 The synthesis method of Example 14 was referred to the synthesis method of Example 4 to obtain the title compound Example 31 (10 mg, 22%).
  • Step 1 Preparation of tert-butyl 5-amino-3-bromo-1H-pyrazole-1-carboxylate
  • 3-Bromo-1H-pyrazol-5-amine (10.0 g, 61.7 mmol) was dissolved in anhydrous dichloromethane (100 mL), triethylamine (7.48 g, 74.1 mmol) and di-tert-butyl dicarbonate were added (16.0 g, 74.1 mmol), react at room temperature for 16 hours.
  • the second step the preparation of tert-butyl 5-amino-3-bromo-1H-pyrazole-1-carboxylate
  • Example 15-1 (14.5 g, 55.3 mmol) was dissolved in anhydrous dichloromethane (200 mL), and triethylamine ( 18.5g, 183mmol), under the protection of nitrogen, add dropwise the dichloromethane solution (50mL) of the now prepared 2-chloro-6-trifluoromethyl nicotinic acid chloride (13.0g, 61.0mmol) at 0°C, and react at room temperature 30 minutes.
  • the third step the preparation of N-(3-bromo-1H-pyrazol-5-yl)-2-chloro-6-(trifluoromethyl)nicotinamide
  • Example 15-3 (6.2 g, 16.8 mmol) was dissolved in N,N- Potassium carbonate (6.96 g, 50.4 mmol) was added to dimethylformamide (80 mL), and heated to 120° C. for 2 hours. The reaction solution was cooled to room temperature and used directly in the next reaction.
  • the fifth step 2-(2-bromo-5-oxo-8-(trifluoromethyl)pyrazol[1,5-a]pyridin[3,2-e]pyrimidin-4(5H)-yl)- Preparation of N-(5-fluoropyridin-2-yl)acetamide
  • Example 16 The synthesis method of Example 16 was referred to the synthesis method of Example 1 to obtain the title compound Example 16 (10 mg, 33%).
  • the first step 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e]pyrimidine-4(5H)- base)-N-(5-fluoropyridin-2-yl)acetamide
  • Example 15 (300mg, 0.619mmol) and Zn(CN) 2 (300mg, 2.56mmol), Pd 2 (dba) 3 (20mg, 0.022mmol), Pd(dppf)Cl 2 (30mg, 0.036mmol) were mixed at room temperature ) and Zn powder (10 mg, 0.154 mmol) were dissolved in DMA (10 mL), and nitrogen was blown inward for 2 minutes. Then microwave heating at 140°C for 8 hours. Cool to room temperature, and extract with ethyl acetate (50 mL), and wash the organic phase twice with saturated brine. The organic phase was dried ( Na2SO4 ) , concentrated under reduced pressure and sent to p-HPLC (FA) to give 100 mg (38% yield) of the title compound.
  • the title compound can also be prepared by:
  • the third step 2-cyano-5-oxo-8-(trifluoromethyl)-4,5-dihydropyrazolo[1,5-a]pyrido[3,2-e]pyrimidine preparation
  • Example 18-1 (2.0 g, 90%).
  • the second step the preparation of pyrazol[1,5-a]pyridin[3,2-e]pyrimidin-5(4H)-one
  • Example 18-2 To a solution of Example 18-2 (1.5 g, 8.06 mmol) in DMF (30 mL) were added potassium carbonate (2.23 g, 16.11 mmol) and Example 1-3 (2.25 g, 9.67 mmol) at room temperature. The mixture was heated to 80°C and the reaction was stirred for 2h. After cooling, water was added, the precipitate was filtered and washed with ethyl acetate, purified to obtain Example 18 (2.1 g, yield: 78%).
  • Example 19 Referring to the method of Example 1, the target compound (31 mg, 26 % yield).
  • Step 1 Preparation of tert-butyl 5-amino-4-cyano-1H-pyrazole-1-carboxylate
  • Example 20-1 Dissolve tert-butyl 5-amino-4-cyano-1H-pyrazole-1-carboxylate
  • Example 20-1 (3.5 g, 16.8 mmol) in anhydrous dichloromethane (50 mL) and add triethylamine (5.35g, 7.37mmol), under the protection of nitrogen, add dropwise the dichloromethane solution (50mL) of the now prepared 2-chloro-6-trifluoromethyl nicotinic acid chloride (4.3g, 17.6mmol) at 0 °C, and add React at room temperature for 1 hour.
  • the third step the preparation of 2-chloro-N-(4-cyano-1H-pyrazol-5-yl)-6-(trifluoromethyl)nicotinamide
  • Step 4 Preparation of 5-oxo-8-(trifluoromethyl)-4,5-dihydropyrazolo[1,5-a]pyridin[3,2-e]pyrimidine-3-carbonitrile
  • Example 20-4 (500mg, 1.79mmol) was dissolved in N,N-dimethylformamide (20mL), potassium carbonate (371mg, 2.69mmol) and 2-bromo-N-(5-fluoropyridin-2-yl)acetamide were added (501mg, 2.15mmol), react at 40°C for 2 hours. The reaction solution was cooled to room temperature, poured into 100 mL of water, and extracted with ethyl acetate (50 mL*2).
  • Example 22-1 100 mg, 0.22 mmol (the synthesis method of Example 21-1 refers to
  • Example 8-2 To a solution of Example 8-2) in THF (2 mL) was added diisobutylaluminum hydride (1M in toluene, 0.66 mL, 0.66 mmol), and the mixture was stirred at room temperature overnight. Rochelle's salt solution (1.0M, 5ml) was added; ethyl acetate (5mL) was then added, the resulting suspension was stirred at room temperature until separation of the clear phases was achieved, the organic phase was separated and the aqueous phase was extracted with EtOAc (3 x 40ml). The combined organic layers were washed with saturated aqueous sodium bicarbonate (50 mL) and saturated brine (50 mL), dried over anhydrous sodium sulfate, concentrated and purified to obtain the title compound (32 mg, 34% yield).
  • Test example 1 Determination of the influence of the compound of the present invention on the flow capacity of calcium ions in cells stably expressing 1321N1-hP2X3 receptors
  • the purpose of the experiment to determine the inhibitory activity of the compound on the 1321N1-hP2X3 receptor.
  • 384-well-cell plate (Corning; 3712); 384-well-compound plate (Corning; 3657);
  • DMEM Gibco; 11965
  • FBS Gibco; 10099-141
  • Hygromycin B (Invitrogen, 10687010); Matrix (Thermo; 5416);
  • DMSO (Sigma; D2650); HBSS (Invitrogen; 14025);
  • ⁇ , ⁇ -meATP (Sigma; M6517); ATP hydrolytic enzyme (Sigma; A7646);
  • Assay buffer 1*HBSS+20mM HEPES;
  • Cell culture medium DMEM+10%FBS+75 ⁇ g/mL hygromycin B+300 ⁇ g/mL G418;
  • Plating medium DMEM+10%DPBS
  • Cell lines are cultured in cell culture medium at 37°C, 5% CO 2 to 70% to 90% confluence, discard the medium, take out the cells, add 2mL Versene, place in a 37°C incubator for 2-5min, add 10mL for plating Cells were collected in the culture medium, counted, and 50 ⁇ L (1 ⁇ 10 4 cells/well density) was added to each well and seeded into a 384-well test plate and incubated for 16-24 hours (at least overnight).
  • On-machine detection Take 15 ⁇ L of 3X compound from each well and add it to the cell plate, add the sample to the FLIPR instrument, and detect the calcium signal. After 15 minutes, add 22.5 ⁇ L of the 3X agonist (EC 80 concentration) to each well to detect the calcium signal.
  • Calcium signal values were read by FLIPR.
  • the calculated output of each sampling time point in the experiment is the ratio of 340/510nm to 380/510nm wavelength signals.
  • the maximum minus minimum calculation is derived from the ratio signal curve.
  • IC50 values of compounds were calculated by fitting percent inhibition and ten-point concentration data to parametric nonlinear logic formulas using GraphPad prism.
  • Test Example 2 Determination of the Effects of Compounds of the Present Invention on Calcium Ion Flow Capacity in Cells Stably Expressing 1321N1-hP2X2/3 Receptors
  • the purpose of the experiment to determine the inhibitory activity of the compound on the 1321N1-hP2X2/3 receptor.
  • 384-well-cell plate (Corning; 3712); 384-well-compound plate (Corning; 3657);
  • DMEM Gibco; 11965
  • FBS Gibco; 10099-141
  • Hygromycin B (Invitrogen, 10687010); Matrix (Thermo; 5416);
  • DMSO (Sigma; D2650); HBSS (Invitrogen; 14025);
  • ⁇ , ⁇ -meATP (Sigma; M6517); ATP hydrolytic enzyme (Sigma; A7646);
  • Assay buffer 1*HBSS+20mM HEPES;
  • Cell culture medium DMEM+10%FBS+75 ⁇ g/mL hygromycin B+150 ⁇ g/mL G418;
  • Plating medium DMEM+10%DPBS
  • Cell lines are cultured in cell culture medium at 37°C, 5% CO 2 to 70% to 90% confluence, discard the medium, take out the cells, add 2mL Versene, place in a 37°C incubator for 2-5min, add 10mL for plating Cells were collected in the culture medium, counted, and 50 ⁇ L (1 ⁇ 10 4 cells/well density) was added to each well and seeded into a 384-well test plate and incubated for 16-24 hours (at least overnight).
  • On-machine detection Take 15 ⁇ L of 3X compound from each well and add it to the cell plate, add the sample to the FLIPR instrument, and detect the calcium signal. After 15 minutes, add 22.5 ⁇ L of the 3X agonist (EC 80 concentration) to each well to detect the calcium signal.
  • Calcium signal values were read by FLIPR.
  • the calculated output of each sampling time point in the experiment is the ratio of 340/510nm to 380/510nm wavelength signals.
  • the maximum minus minimum calculation is derived from the ratio signal curve.
  • IC50 values of compounds were calculated by fitting percent inhibition and ten-point concentration data to parametric nonlinear logic formulas using GraphPad prism.
  • Test example 3 Balb/C mouse pharmacokinetic determination
  • Test drug the embodiment of the present invention, self-made.
  • HEC hydroxyethyl cellulose
  • CMC-Na viscosity: 800-1200Cps
  • Tween80 10g
  • liquid A is 0.1% formic acid aqueous solution
  • liquid B is acetonitrile
  • Test example 4 rat pharmacokinetic determination
  • Test drug the embodiment of the present invention, self-made.
  • HEC hydroxyethyl cellulose
  • CMC-Na viscosity: 800-1200Cps
  • Tween80 10g
  • liquid A is 0.1% formic acid aqueous solution
  • liquid B is acetonitrile
  • the purpose of this experiment is to test the stability of the compounds of the examples in mice, rats, dogs and human liver microsomes.
  • liver microsome working solution dilute with 100mM phosphate buffer to a final concentration of 0.625mg/mL.
  • NADPH and UDPGA Weigh NADPH (reduced nicotinamide adenine dinucleotide phosphate) and UDPGA (uridine diphosphate glucuronic acid), add 100 mM phosphate buffer, and the final concentration is 20 mM.
  • reaction termination solution cold acetonitrile containing 100ng/mL labetalol hydrochloride and 400ng/mL tolbutamide as internal standard.
  • liver microsomes 0.5mg/mL compound 1 ⁇ M NADPH 2mM UDPGA 2mM Alamethicin 2.5 ⁇ g/mL
  • Ion source electrospray ionization source (ESI); dry gas: N 2 , temperature 500°C;
  • Electrospray voltage 5000V; detection method: positive ion detection;
  • Experimental purpose The purpose of this experimental method is to detect the plasma protein binding of the compounds of the examples in plasma.
  • the ion source is an electrospray ionization source (ESI); the drying gas (N 2 ) temperature is 500°C;
  • the electrospray voltage is 5500V; the detection method is positive ion detection;
  • the scanning method is selective reaction monitoring (MRM); the scanning time is 0.1s.
  • Test example 7 CYP enzyme single-point inhibition test
  • Solution preparation 2.5mM NADPH, weigh 4.165mg NADPH (reduced nicotinamide adenine dinucleotide phosphate) and add 100mM phosphate buffer to 2mL. 0.25mg/mL microsomes, 50 ⁇ L 20mg/mL microsomes, add 4mL 100mM phosphate buffer, mix well.
  • NADPH reduced nicotinamide adenine dinucleotide phosphate
  • test compound reaction solution Weigh the test compound, dilute to 10 mM with DMSO, and then dilute to 100 ⁇ M with 100 mM phosphate buffer.
  • CHO-hERG cells were cultured in a 175cm 2 culture flask. When the cell density grew to 60-80%, the culture medium was removed, washed once with 7mL PBS, and then digested by adding 3mL Detachin.
  • the single-cell high-impedance sealing and whole-cell pattern formation processes are all automatically completed by the Qpatch instrument.
  • the cell After obtaining the whole-cell recording pattern, the cell is clamped at -80 mV, before giving a 5-second +40 mV depolarization stimulus , give a pre-voltage of -50 mV for 50 milliseconds, then repolarize to -50 mV for 5 seconds, and then return to -80 mV. Apply this voltage stimulus every 15 seconds, record for 2 minutes, give extracellular fluid for recording for 5 minutes, and then start the administration process.
  • the compound concentration starts from the lowest test concentration, and each test concentration is given for 2.5 minutes. At least 3 cells are tested for each concentration. (n ⁇ 3).
  • the highest test concentration is 40 ⁇ M, and there are 6 concentrations in total of 40, 13.33, 4.44, 1.48, 0.49, and 0.16 ⁇ M respectively.
  • the experimental reagents used were purchased from Sigma, with a purity of >98%
  • Table 21 The results of inhibition of hERG current by the compounds of the examples at multiple concentrations hERG results
  • Inhibition of cardiac hERG potassium channels by drugs is the main cause of long QT syndrome caused by drugs. It can be seen from the experimental results that the compound of the embodiment of the present invention has no obvious inhibitory effect on the heart hERG potassium ion channel, and can avoid the cardiotoxic side effects at high doses.
  • mice BALB/c mice, 6-8 weeks old, ⁇ , purchased from Shanghai Xipuer-Bikay Experimental Animal Co., Ltd.
  • mice were randomly divided into groups according to the requirements according to body weight, and water and food were deprived 12-16 hours before administration.
  • Drug administration and animal quinine drinking water test drug administration and fasting: on the day of the experiment, the animals were weighed, fasted, litter changed, and administered according to the experimental design.
  • Rate of dysgeusia (Drinking water is quinine hydrochloric acid aqueous solution and given to the test drug group ⁇ pWW-drinking water is quinine hydrochloric acid aqueous solution and given solvent to the control group ⁇ pWW)/(Drinking water is ultrapure water and given solvent at the same time
  • the control group ⁇ pWW-drinking water is quinine hydrochloric acid aqueous solution and the solvent control group ⁇ pWW) ⁇ 100%.
  • Test example 10 drug effect study on guinea pig acute cough induced by citric acid
  • the purpose of this experiment is to evaluate the efficacy of the compound in the acute cough model of guinea pigs induced by citric acid.
  • Cough induction method put the guinea pig into the whole-body volume scanning box to adapt to it for 3-5 minutes, then perform 2 minutes of ATP atomization, with an interval of 3 minutes, and then give 5 minutes of citric acid atomization. From the start of citric acid nebulization, record the number of coughs and cough latency of animals within 10 minutes.
  • test compound was administered to guinea pigs by single gavage 2 hours before citric acid nebulization, and placed in the breath plethysmography cavity of DSI Buxco whole body plethysmography detection system (WBP) at the scheduled time for citric acid nebulization to induce cough. From the start of citric acid nebulization, the WBP system recorded the total number of coughs (CCnt) and cough latency (CIP) of guinea pigs within 10 minutes.
  • WBP whole body plethysmography detection system
  • Mobile phase A: water (0.05% trifluoroacetic acid); B: acetonitrile (0.05% trifluoroacetic acid)
  • Salt formation by dissolution or suspension reaction Weigh 10 mg of compound 2-(2-cyano-5-oxo-8-(trifluoromethyl)pyrazolo[1,5-a]pyridin[3,2-e ]pyrimidin-4(5H)-yl)-N-(5-fluoropyridin-2-yl)acetamide, add solvent 200 ⁇ L ⁇ 400 ⁇ L, heat and stir at 40 ⁇ 50°C, add different acids respectively, stir overnight, drop to room temperature, filtered and dried to obtain the salt of the compound.
  • liquid A is 0.1% formic acid aqueous solution
  • liquid B is acetonitrile

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Rheumatology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pain & Pain Management (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

一种式(I-a)所示的含吡唑多环类衍生物的酸式盐、晶型及其制备方法、含有治疗有效量的该晶型的药物组合物和制备该衍生物的中间体。特别地,涉及通式(I-a)化合物盐及晶型作为P2X3抑制剂,在制备治疗神经源性疾病药物中的用途。

Description

含吡唑多环类衍生物的盐、晶型及其制备方法和应用 技术领域
本发明属于生物医药领域,具体涉及一种含吡唑多环类衍生物的盐、晶型及其制备方法和应用。
背景技术
P2X受体(P2X receptors)或称为P2X嘌呤受体(P2X purinoreceptor)是一个阳离子渗透型ATP配体门控离子通道家族,能同细胞外的ATP结合。P2X受体具有七个亚基,以同源三聚体或异源三聚体的形式存在,主要表达于神经系统的神经末梢(突触前和突触后),调节突触传递。P2X3受体是P2X家族成员之一,是感受上呼吸道刺激和触发咳嗽反射的关键感觉受体,被认为在特定感觉神经的致敏(sensitisation)中发挥了关键作用,参与疼痛和咳嗽,并参与骨癌痛的感知,阻断P2X3可以抑制咳嗽的信号刺激。
咳嗽是机体的防御性神经反射,有利于清除呼吸道分泌物和有害因子,但频繁剧烈的咳嗽会对患者等工作、生活和社会活动造成严重影响。咳嗽分为急性、亚急性、慢性咳嗽。慢性咳嗽(Chronic cough)为咳嗽时间>8周,以咳嗽为主要或唯一症状者,胸部影像学检查肺内无明显病变。慢性咳嗽一直被认为是各种疾病的后果,如哮喘/嗜酸性支气管炎、鼻炎和胃食管反酸病。然而,最近的证据表明,慢性咳嗽是一种临床症状,具有独特的内在病理生理学特征的神经过敏。不明原因慢性咳嗽或特发性咳嗽,此类以慢性刺激性干咳为主要表现,对外界刺激较敏感,普遍存在咳嗽高敏感性,咳嗽高敏感性是其生理病理机制。咳嗽相关传入神经异常可能是难治性或原因不明的慢性咳嗽的原因。慢性咳嗽可引起心血管、消化、神经、泌尿、肌肉骨骼等多个系统的并发症,如尿失禁、晕厥、失眠、焦虑等。
基于咳嗽高敏综合症的病理生理学特征,治疗应以降低咳嗽敏感性为目的。目前治疗选择有限,包括药物治疗手段和非药物治疗手段。临床研究结果显示神经调节因子类药物加巴喷丁治疗有效,其他药物如阿米替林,巴氯芬、卡马西平、普瑞巴林等亦可选用。严重咳嗽可适当给予镇咳治疗,镇咳药物主要分为中枢性镇咳药和外周性镇咳药。中枢性镇咳药物分为依赖性镇咳药(吗啡类生物碱及其衍生物)和非依赖性镇咳药(人工合成的右美沙芬和喷托维林),前者具有成瘾性和麻醉性等副作用,后者临床应用十分广泛。外周性镇咳药也称为末梢镇咳药,通过抑制咳嗽反射弧中的某一环节而起到镇咳作用,包括局部麻醉药(那可丁、苯佐那酯)和黏膜防护剂(苯丙哌林和莫吉司坦)。
目前,市场上尚未有已批准上市的P2X3受体拮抗剂小分子药物。现在处在临床阶段的P2X3受体拮抗剂药物有默克(Merck&Co)的MK-7264,其用于治疗慢性咳嗽、疼痛和肺纤维化等疾病,其对P2X3/P2X2/3选择性低,安全性好但具有味觉丧失等副作用,目前适应症慢性咳嗽已经入临床III期研究。贝勒斯健康(Bellus Health)的BLU5937,具有较高的选择性,且在I期临床试验中未出现味觉等副作用,2020年7月6日,贝勒斯健康公布了BLU-5937的2期RELIEF试验在难治性慢性咳嗽患者中的主要研究结果:在II期临床研究中,RELIEF试验在任何剂量下均未达到安慰剂调整后咳嗽频率降低的主要终点的统计学意义。另外,拜耳(Bayer)的BAY-1817080和BAY-1902607,Shionogi公司的S-600918,适应症慢性咳嗽目前处在临床I/II期。因此,迫切需要开发安全性好、非成瘾麻醉性、具有高选择性的P2X3受体抑制剂药物用于治疗慢性咳嗽等疾病,以满足巨大的市场需求。
江苏豪森药业集团有限公司的专利申请(申请号:PCT/CN2020/134264)中公开了一系列含吡唑多环类衍生物抑制剂的结构,在后续的研发中,为了使产物易于处理、过滤、干燥,便于储存、产品长期稳定、生物利用度高等,本发明对上述物质的盐和晶型进行了全面的研究,致力于得到最适合的晶型,此外,目前制备含吡唑多环类衍生物的原料昂贵且反应路线条件苛刻,不适合工业化大生产,本发明开发出适合工业化生产的制备方法。
发明内容
专利申请PCT/CN2020/134264所涉及的所有内容均以引证的方式添加到本发明中。
本发明的目的在于提供一种通式(I-a)所示化合物或其立体异构体的酸式盐,
Figure PCTCN2022097828-appb-000001
其中:
R 1选自氢、氘、卤素、氨基、羟基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-12环烷基、-(CH 2) nC(O)R a、3-12元杂环基、C 6-14芳基或5-14元杂芳基,所述的氨基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-12环烷基、-(CH 2) nC(O)R a、3-12元杂环 基、C 6-14芳基和5-14元杂芳基,任选地被氘、卤素、氨基、羟基、氰基、硝基、氧代基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6羟烷基、氰基取代的C 1-6烷基、C 3-12环烷基、3-12元杂环基、C 6-12芳基和5-12元杂芳基中的一个或多个取代基所取代;
R a选自氢、氘、卤素、氨基、硝基、羟基、氰基、C 1-6烷基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基或5-10元杂芳基,所述的氨基、C 1-6烷基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基,任选地被氘、卤素、氨基、硝基、羟基、氰基、羧基、氧代基、硫代基、C 1-6烷基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基中的一个或多个取代基所取代;
R 2选自氢、氘、卤素、氨基、羟基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-12环烷基、3-12元杂环基、C 6-14芳基或5-14元杂芳基,所述的氨基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-12环烷基、3-12元杂环基、C 6-14芳基和5-14元杂芳基,任选地被氘、卤素、氨基、羟基、氰基、硝基、氧代基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6羟烷基、氰基取代的C 1-6烷基、C 3-12环烷基、3-12元杂环基、C 6-12芳基和5-12元杂芳基中的一个或多个取代基所取代;
R 3选自氢、氘、卤素、氨基、羟基、氰基、氧代基、硫代基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 3-8环烷基、3-12元杂环基、C 6-14芳基或5-14元杂芳基;且
x为0~3的整数,优选0、1或2,更优选0或1;
n为0~3的整数,优选0、1或2,更优选0或1;
酸式盐中的酸为无机酸或有机酸;优选地,无机酸选自盐酸、硫酸、硝酸、氢溴酸、氢氟酸、氢碘酸或磷酸;有机酸选自2,5-二羟基苯甲酸、1-羟基-2-萘甲酸、醋酸、乙烷磺酸、二氯醋酸、三氯醋酸、乙酰氧肟酸、己二酸、苯磺酸、4-氯苯磺酸、苯甲酸、4-乙酰氨基苯甲酸、4-氨基苯甲酸、癸酸、己酸、辛酸、肉桂酸、柠檬酸、环己烷氨基磺酸、樟脑磺酸、天门冬氨酸、樟脑酸、葡萄糖酸、葡糖醛酸、谷氨酸、异抗坏血酸、乳酸、苹果酸、扁桃酸、焦谷氨酸、酒石酸、十二烷基硫酸、二苯甲酰酒石酸、乙烷-1,2-二磺酸、乙烷磺酸、蚁酸、富马酸、半乳糖酸、龙胆酸、戊二酸、2-酮戊二酸、乙醇酸、马尿酸、羟乙基磺酸、乳糖酸、抗坏血酸、天冬氨酸、月桂酸、樟脑酸、马来酸、丙二酸、甲磺酸、1,5-萘 二磺酸、萘-2-磺酸、烟酸、油酸、乳清酸、草酸、棕榈酸、双羟萘酸、丙酸、水杨酸、4-氨基水杨酸、癸二酸、硬脂酸、丁二酸、硫氰酸、十一碳烯酸、三氟乙酸、苯磺酸、对甲基苯磺酸或L-苹果酸;优选盐酸、硫酸、磷酸、乙烷磺酸、苯磺酸、甲磺酸、富马酸、羟乙基磺酸、草酸或氢溴酸。
在本发明的优选方案中,通式(I-a)进一步如通式(II-a)所示:
Figure PCTCN2022097828-appb-000002
在本发明的优选方案中,所述R 1选自氢、氘、卤素、氨基、羟基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-8环烷基、-(CH 2) nC(O)R a、3-8元杂环基、C 6-10芳基或5-10元杂芳基,所述的氨基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-8环烷基、-(CH 2) nC(O)R a、3-8元杂环基、C 6-10芳基和5-10元杂芳基,任选地被氘、卤素、氨基、羟基、氰基、硝基、氧代基、C 1-3烷基、C 2-3烯基、C 2-3炔基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 1-3羟烷基、氰基取代的C 1-3烷基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基中的一个或多个取代基所取代,
R a选自氢、氘、卤素、氨基、硝基、羟基、氰基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基或5-10元杂芳基,所述的氨基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基,任选地被氘、卤素、氨基、硝基、羟基、氰基、羧基、氧代基、硫代基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基中的一个或多个取代基所取代;
优选地,R 1选自氢、卤素、氨基、氰基、C 1-3烷基、C 2-6烯基、、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-8环烷基、-(CH 2) nC(O)R a、含1-3个选自氮、氧或硫原子的3-8元杂环基、C 6-10芳基或含1-3个选自氮、氧或硫原子5-10元杂芳基,所述的氨基、C 1-6烷基、C 2-6烯基、C 1-6卤代烷基、C 1-6 烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-8环烷基、含1-3个选自氮、氧或硫原子的3-8元杂环基、C 6-10芳基或含1-3个选自氮、氧或硫原子5-10元杂芳基,任选地被氘、卤素、氨基、羟基、氰基、硝基、氧代基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 1-3羟烷基、氰基取代的C 1-3烷基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基中的一个或多个取代基所取代;
R a选自氢、氘、卤素、氨基、氰基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 3-6环烷基或含1-2个选自N或O的4-6元杂环基,所述的氨基C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 3-6环烷基或含1-2个选自N或O的4-6元杂环基,任选地被氘、卤素、氨基、羟基、氰基、硝基、氧代基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 1-3羟烷基、氰基取代的C 1-3烷基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基中的一个或多个取代基所取代;
更优选地,R 1选自以下基团:
-H、-NH 2、-F、-Cl、-Br、-CN、-CH 3、-CH 2CH 3、-CF 3
Figure PCTCN2022097828-appb-000003
Figure PCTCN2022097828-appb-000004
R 2选自氢、氘、卤素、氨基、羟基、氰基、硝基、C 1-3烷基、C 2-3烯基、C 2-3炔基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3烷氧基、卤代C 1-3烷氧基、C 1-3羟烷基、C 3-8环烷基、3-8元杂环基、C 6-10芳基或5-10元杂芳基,优选氢、氨基、氰基、氟、氯、溴、甲基、异丙基、三氟甲基、甲氧基、环丙基或吗啉基;
R 3选自氢、氘、卤素、氨基、羟基、氰基、氧代基、硫代基、C 1-3烷基、C 2-3 烯基、C 2-3炔基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 3-8环烷基、3-8元杂环基、C 6-10芳基或5-1.元杂芳基,优选氢或氰基。
在本发明的优选方案中,酸式盐中的酸选自羟乙基磺酸、盐酸、硫酸、1,5-萘二磺酸、甲磺酸、氢溴酸、乙烷磺酸、磷酸、苯磺酸、草酸、马来酸、己二酸、盐酸、柠檬酸、丙二酸、L-苹果酸、帕莫酸、对甲苯磺酸或富马酸;优选盐酸、硫酸、甲磺酸、氢溴酸或乙烷磺酸。
在本发明的优选方案中,所述化合物的酸式盐,酸的个数为0.2-3;优选0.2、0.5、1、1.5、2、2.5或3;更优选0.5、1、2或3,进一步优选1。
在本发明的优选方案中,所述化合物的酸式盐,酸式盐为水合物或无水物;当酸式盐为水合物时,水的个数为0.2-3;优选0.2、0.5、1、1.5、2、2.5或3;更优选0.5、1、2或3。
在本发明的优选方案中,所述化合物的酸式盐,所述酸式盐为晶型;
优选地,晶型为化合物2-(2-(叔丁基)-5-氧吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
2-(2-(叔丁基)-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
2-(2-乙基-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
2-(2-环丙基-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
2-(2,5-二甲基吡啶-4-基)-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
N-(5-氟吡啶-2-基)-2-(2-(1-甲基环丙基)-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)乙酰胺的酸式盐晶型;
2-(2-溴-5-氧-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
N-(5-氟吡啶-2-基)-2-(5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)乙酰胺的酸式盐晶型;
2-(3-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
更优选羟乙基磺酸盐晶型、硫酸盐晶型、盐酸盐晶型、1,5-萘二磺酸盐晶型、甲磺酸盐晶型、乙烷磺酸盐晶型、氢溴酸盐晶型、磷酸盐晶型、苯磺酸盐晶型、草酸盐晶型、马来酸盐晶型、己二酸盐晶型、盐酸盐晶型、柠檬酸盐晶型、 丙二酸盐晶型、L-苹果酸盐晶型、帕莫酸盐晶型、对甲苯磺酸盐晶型或富马酸盐晶型。
在本发明的优选方案中,提供化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的甲磺酸盐晶型A、乙烷磺酸盐晶型A、硫酸盐晶型A-B、盐酸盐晶型A-B和氢溴酸盐晶型A-C。
在本发明进一步优选的实施方式中,所述化合物的酸式盐晶型为甲磺酸盐晶型A,其X-射线粉末衍射图谱在13.7±0.2°处具有衍射峰;或者在21.9±0.2°处具有衍射峰;或者在20.4±0.2°处具有衍射峰;或者在15.4±0.2°处具有衍射峰;或者在19.6±0.2°处具有衍射峰;或者在16.4±0.2°处具有衍射峰;或者在9.3±0.2°处具有衍射峰;或者在5.3±0.2°处具有衍射峰;或者在7.9±0.2°处具有衍射峰;或者在11.9±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
优选地,甲磺酸盐晶型A的X-射线粉末衍射图谱至少包含位于2θ为13.7±0.2°、16.4±0.2°、21.9±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为13.9±0.2°、20.4±0.2°、15.4±0.2°、5.3±0.2°、11.9±0.2°、9.3±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
更优选地,甲磺酸盐晶型A的X-射线粉末衍射图谱任选还包含位于2θ为7.9±0.2°、19.6±0.2°、17.6±0.2°、18.8±0.2°、21.0±0.2°、23.3±0.2°、24.1±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
进一步优选地,甲磺酸盐晶型A的X-射线粉末衍射图谱包含位于2θ为5.3±0.2°、7.9±0.2°、9.3±0.2°、11.9±0.2°、13.7±0.2°、13.9±0.2°、15.4±0.2°、16.4±0.2°、17.6±0.2°、18.8±0.2°、19.6±0.2°、20.4±0.2°、21.0±0.2°、21.9±0.2°、23.3±0.2°、24.1±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
最优选地,在本发明进一步优选地实施方式中,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的甲磺酸盐晶型A,使用Cu-Kα辐射,以2θ角和晶面间距d值表示的X-射线特征衍射峰如表1所示。
表1
序号 2θ(±0.2°)
1 5.328
2 7.917
3 9.277
4 11.865
5 13.674
6 13.852
7 15.444
8 16.353
9 17.584
10 18.763
11 19.553
12 20.422
13 20.967
14 21.921
15 23.346
16 24.093
17 24.819
18 25.503
本发明化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的甲磺酸盐晶型A,其X-射线粉末衍射图谱基本如图1所示;其DSC图谱基本如图2所示;其TGA图谱基本如图3所示。
在本发明进一步优选的实施方式中,所述化合物的酸式盐晶型为乙烷磺酸盐晶型A,其X-射线粉末衍射图谱在15.0±0.2°处具有衍射峰;或者在21.1±0.2°处具有衍射峰;或者在23.1±0.2°处具有衍射峰;或者在19.8±0.2°处具有衍射峰;或者在12.5±0.2°处具有衍射峰;或者在9.0±0.2°处具有衍射峰;或者在12.3±0.2°处具有衍射峰;或者在24.6±0.2°处具有衍射峰;或者在10.3±0.2°处具有衍射峰;或者在6.1±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
优选地,乙烷磺酸盐晶型A的X-射线粉末衍射图谱至少包含位于2θ为15.0±0.2°、21.1±0.2°、23.1±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为19.8±0.2°、12.5±0.2°、9.0±0.2°、12.3±0.2°、24.6±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
更优选地,乙烷磺酸盐晶型A的X-射线粉末衍射图谱任选还包含位于2θ为10.3±0.2°、6.1±0.2°、16.1±0.2°、19.2±0.2°、23.6±0.2°、30.7±0.2°、9.6±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7 处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
进一步优选地,乙烷磺酸盐晶型A的X-射线粉末衍射图谱包含位于2θ为15.0±0.2°、21.1±0.2°、23.1±0.2°、19.8±0.2°、12.5±0.2°、9.0±0.2°12.3±0.2°、24.6±0.2°、10.3±0.2°、6.1±0.2°、16.1±0.2°、19.2±0.2°、23.6±0.2°、30.7±0.2°、9.6±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
在本发明进一步优选地实施方式中,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的乙烷磺酸盐晶型A,使用Cu-Kα辐射,以2θ角和晶面间距d值表示的X-射线特征衍射峰如表2所示。
表2
Figure PCTCN2022097828-appb-000005
本发明的化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的乙烷磺酸盐晶型A,其X-射线粉末衍射图谱基本如图4所示;其DSC图谱基本如图5所示;其TGA图谱基本如图6所示。
在本发明进一步优选地实施方式中,所述化合物酸式盐为硫酸盐晶型A,其 X-射线粉末衍射图谱在22.5±0.2°处具有衍射峰;或者在15.9±0.2°处具有衍射峰;或者在22.3±0.2°处具有衍射峰;或者在16.8±0.2°处具有衍射峰;或者在22.9±0.2°处具有衍射峰;或者在32.1±0.2°处具有衍射峰;或者在14.0±0.2°处具有衍射峰;或者在21.1±0.2°处具有衍射峰;或者在11.2±0.2°处具有衍射峰;或者在26.1±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
优选地,硫酸盐晶型A的X-射线粉末衍射图谱至少包含位于2θ为22.5±0.2°、15.9±0.2°、22.3±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为16.8±0.2°、22.9±0.2°、32.1±0.2°、14.0±0.2°、21.1±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
更优选地,或,硫酸盐晶型A的X-射线粉末衍射图谱任选还包含位于2θ为11.2±0.2°、26.1±0.2°、28.2±0.2°、37.8±0.2°、15.5±0.2°、26.5±0.2°、36.4±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
进一步优选地,硫酸盐晶型A的X-射线粉末衍射图谱包含位于2θ为22.5±0.2°、15.9±0.2°、22.3±0.2°、16.8±0.2°、22.9±0.2°、32.1±0.2°、14.0±0.2°、21.1±0.2°、11.2±0.2°、26.1±0.2°、28.2±0.2°、37.8±0.2°、15.5±0.2°、26.5±0.2°、36.4±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
在本发明进一步优选地实施方式中,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的硫酸盐晶型A,使用Cu-Kα辐射,以2θ角和晶面间距d值表示的X-射线特征衍射峰如表3所示。
表3
Figure PCTCN2022097828-appb-000006
Figure PCTCN2022097828-appb-000007
本发明的化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的硫酸盐晶型A,其X-射线粉末衍射图谱基本如图7所示;其DSC图谱基本如图8所示。
在本发明进一步优选地实施方式中,所述化合物酸式盐为硫酸盐晶型B,其X-射线粉末衍射图谱在15.3±0.2°处具有衍射峰;或者在21.5±0.2°处具有衍射峰;或者在10.6±0.2°处具有衍射峰;或者在19.8±0.2°处具有衍射峰;或者在20.1±0.2°处具有衍射峰;或者在12.6±0.2°处具有衍射峰;或者在25.2±0.2°处具有衍射峰;或者在9.2±0.2°处具有衍射峰;或者在9.9±0.2°处具有衍射峰;或者在23.4±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
优选地,硫酸盐晶型B的X-射线粉末衍射图谱至少包含位于2θ为15.3±0.2°、21.5±0.2°、10.6±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为19.8±0.2°、20.1±0.2°、12.6±0.2°、25.2±0.2°、9.2±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
更优选地,硫酸盐晶型B的X-射线粉末衍射图谱任选还包含位于2θ为9.9±0.2°、23.4±0.2°、6.3±0.2°、16.7±0.2°、23.9±0.2°、33.8±0.2°、16.3±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
进一步优选地,硫酸盐晶型B的X-射线粉末衍射图谱包含位于2θ为15.3±0.2°、21.5±0.2°、10.6±0.2°、19.8±0.2°、20.1±0.2°、12.6±0.2°、25.2±0.2°、9.2±0.2°、9.9±0.2°、23.4±0.2°、6.3±0.2°、16.7±0.2°、23.9±0.2°、33.8±0.2°、16.3±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
在本发明进一步优选地实施方式中,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的硫酸盐晶型B,使用Cu-Kα辐射,以2θ角和晶面间距d值表示的X-射线特征衍射峰如表4所示。
表4
Figure PCTCN2022097828-appb-000008
本发明的化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的硫酸盐晶型B,其X-射线粉末衍射图谱基本如图9所示;其DSC图谱基本如图10所示;其TGA图谱基本如图11所示。
在本发明进一步优选的实施方式中,所述化合物酸式盐为盐酸盐晶型A,其X-射线粉末衍射图谱在15.0±0.2°处具有衍射峰;或者在23.9±0.2°处具有衍射峰;或者在9.7±0.2°处具有衍射峰;或者在5.3±0.2°处具有衍射峰;或者在24.8±0.2°处具有衍射峰;或者在29.5±0.2°处具有衍射峰;或者在7.5±0.2°处具有衍射峰;或者在21.8±0.2°处具有衍射峰;或者在21.3±0.2°处具有衍射峰;或者在10.6±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
优选地,盐酸盐晶型A的X-射线粉末衍射图谱至少包含位于2θ为15.0±0.2°、 23.9±0.2°、9.7±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为5.3±0.2°、24.8±0.2°、29.5±0.2°、7.5±0.2°、21.8±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
更优选地,盐酸盐晶型A的X-射线粉末衍射图谱任选还包含位于2θ为21.3±0.2°、10.6±0.2°、16.9±0.2°、16.0±0.2°、18.4±0.2°、25.8±0.2°、28.4±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
进一步优选地,盐酸盐晶型A的X-射线粉末衍射图谱包含位于2θ为15.0±0.2°、23.9±0.2°、9.7±0.2°、5.3±0.2°、24.8±0.2°、29.5±0.2°、7.5±0.2°、21.8±0.2°、21.3±0.2°、10.6±0.2°、16.9±0.2°、16.0±0.2°、18.4±0.2°、25.8±0.2°、28.4±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
在本发明进一步优选地实施方式中,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的盐酸盐晶型A,使用Cu-Kα辐射,以2θ角和晶面间距d值表示的X-射线特征衍射峰如表5所示。
表5
Figure PCTCN2022097828-appb-000009
Figure PCTCN2022097828-appb-000010
本发明的化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的盐酸盐晶型A,其X-射线粉末衍射图谱基本如图12所示;其DSC图谱基本如图13所示;其TGA图谱基本如图14所示。
在本发明进一步优选的实施方式中,所述化合物酸式盐为盐酸盐晶型B,盐酸盐晶型B的X-射线粉末衍射图谱在15.9±0.2°处具有衍射峰;或者在22.2±0.2°处具有衍射峰;或者在5.2±0.2°处具有衍射峰;或者在21.7±0.2°处具有衍射峰;或者在26.0±0.2°处具有衍射峰;或者在4.6±0.2°处具有衍射峰;或者在28.4±0.2°处具有衍射峰;或者在9.2±0.2°处具有衍射峰;或者在17.3±0.2°处具有衍射峰;或者在15.2±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
优选地,盐酸盐晶型B的X-射线粉末衍射图谱至少包含位于2θ为15.9±0.2°、22.2±0.2°、5.2±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为21.7±0.2°、26.0±0.2°、4.6±0.2°、28.4±0.2°、9.2±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
更优选地,或,盐酸盐晶型B的X-射线粉末衍射图谱任选还包含位于2θ为17.3±0.2°、15.2±0.2°、10.5±0.2°、38.0±0.2°、20.3±0.2°、23.8±0.2°、29.5±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
进一步优选地,盐酸盐晶型B的X-射线粉末衍射图谱包含位于2θ为15.9±0.2°、22.2±0.2°、5.2±0.2°、21.7±0.2°、26.0±0.2°、4.6±0.2°、28.4±0.2°、9.2±0.2°、17.3±0.2°、15.2±0.2°、10.5±0.2°、38.0±0.2°、20.3±0.2°、23.8±0.2°、29.5±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
在本发明进一步优选地实施方式中,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的盐酸盐晶型B,使用Cu-Kα辐射,以2θ角和晶面间距d值表示的X-射线特征衍射峰如表6所示。
表6
Figure PCTCN2022097828-appb-000011
Figure PCTCN2022097828-appb-000012
本发明的化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的盐酸盐晶型B,其X-射线粉末衍射图谱基本如图15所示。
在本发明进一步优选的实施方式中,所述化合物的酸式盐晶型为氢溴酸盐晶型A,其X-射线粉末衍射图谱在5.3±0.2°处具有衍射峰;或者在22.7±0.2°处具有衍射峰;或者在14.8±0.2°处具有衍射峰;或者在10.5±0.2°处具有衍射峰;或者在22.5±0.2°处具有衍射峰;或者在28.0±0.2°处具有衍射峰;或者在30.0±0.2°处具有衍射峰;或者在23.4±0.2°处具有衍射峰;或者在23.3±0.2°处具有衍射峰;或者在26.5±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
优选地,氢溴酸盐晶型A的X-射线粉末衍射图谱至少包含位于2θ为5.3±0.2°、22.7±0.2°、14.8±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为10.5±0.2°、22.5±0.2°、28.0±0.2°、30.0±0.2°、23.4±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
更优选地,氢溴酸盐晶型A的X-射线粉末衍射图谱任选还包含位于2θ为23.3±0.2°、26.5±0.2°、34.9±0.2°、15.8±0.2°、25.0±0.2°、31.9±0.2°、37.0±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,包含其中任意2处、3处、4处、5处、6处、7处;
进一步优选地,或,氢溴酸盐晶型A的X-射线粉末衍射图谱包含位于2θ为5.3±0.2°、22.7±0.2°、14.8±0.2°、10.5±0.2°、22.5±0.2°、28.0±0.2°、30.0±0.2°、 23.4±0.2°、23.3±0.2°、26.5±0.2°、34.9±0.2°、15.8±0.2°、25.0±0.2°、31.9±0.2°、37.0±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
在本发明进一步优选地实施方式中,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的氢溴酸盐晶型A,使用Cu-Kα辐射,以2θ角和晶面间距d值表示的X-射线特征衍射峰如表7所示。
表7
Figure PCTCN2022097828-appb-000013
本发明的化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的氢溴酸盐晶型A,其X-射线粉末衍射图谱基本如图16所示;其DSC图谱基本如图17所示。
在本发明进一步优选的实施方式中,所述化合物酸式盐为氢溴酸盐晶型B,其X-射线粉末衍射图谱在23.4±0.2°处具有衍射峰;或者在15.9±0.2°处具有衍射峰;或者在16.2±0.2°处具有衍射峰;或者在14.2±0.2°处具有衍射峰;或者在5.3±0.2°处具有衍射峰;或者在10.6±0.2°处具有衍射峰;或者在23.1±0.2°处具有衍射峰;或者在24.1±0.2°处具有衍射峰;或者在14.8±0.2°处具有衍射峰;或者在9.5±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
优选地,或,氢溴酸盐晶型B的X-射线粉末衍射图谱至少包含位于2θ为23.4±0.2°、15.9±0.2°、16.2±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为14.2±0.2°、5.3±0.2°、10.6±0.2°、23.1±0.2°、24.1±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
更优选地,氢溴酸盐晶型B的X-射线粉末衍射图谱任选还包含位于2θ为14.8±0.2°、9.5±0.2°、16.9±0.2°、13.9±0.2°、29.5±0.2°、32.2±0.2°、22.2±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,包含其中任意2处、3处、4处、5处、6处、7处;
进一步优选地,氢溴酸盐晶型B的X-射线粉末衍射图谱包含位于2θ为23.4±0.2°、15.9±0.2°、16.2±0.2°、14.2±0.2°、5.3±0.2°、10.6±0.2°、23.1±0.2°、24.1±0.2°、14.8±0.2°、9.5±0.2°、16.9±0.2°、13.9±0.2°、29.5±0.2°、32.2±0.2°、22.2±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
在本发明进一步优选地实施方式中,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的氢溴酸盐晶型B,使用Cu-Kα辐射,以2θ角和晶面间距d值表示的X-射线特征衍射峰如表8所示。
表8
Figure PCTCN2022097828-appb-000014
Figure PCTCN2022097828-appb-000015
本发明的化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的氢溴酸盐晶型B,其X-射线粉末衍射图谱基本如图18所示;其DSC图谱基本如图19所示;其TGA图谱基本如图20所示。
在本发明优选的方案中,化合物酸式盐为氢溴酸盐晶型C,其X-射线粉末衍射图谱在5.2±0.2°处具有衍射峰;或者在15.7±0.2°处具有衍射峰;或者在22.3±0.2°处具有衍射峰;或者在10.5±0.2°处具有衍射峰;或者在17.4±0.2°处具有衍射峰;或者在38.0±0.2°处具有衍射峰;或者在26.3±0.2°处具有衍射峰;或者在28.0±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处。
优选地,氢溴酸盐晶型C的X-射线粉末衍射图谱至少包含位于2θ为5.2±0.2°、15.7±0.2°、22.3±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为10.5±0.2°、17.4±0.2°、38.0±0.2°、26.3±0.2°、28.0±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
更优选地,氢溴酸盐晶型C的X-射线粉末衍射图谱包含位于2θ为5.2±0.2°、15.7±0.2°、22.3±0.2°、10.5±0.2°、17.4±0.2°、38.0±0.2°、26.3±0.2°、28.0±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处或8处有衍射峰;
在本发明进一步优选地实施方式中,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的氢溴酸盐晶型C,使用Cu-Kα辐射,以2θ角和晶面间距d值表示的X-射线特征衍射峰如表9所示。
表9
Figure PCTCN2022097828-appb-000016
本发明的化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的氢溴酸盐晶型C,其X-射线粉末衍射图谱基本如图21所示。
在本发明进一步优选方案中,化合物的酸式盐晶型的X-射线粉末衍射图谱中相对峰强度为前十强的衍射峰位置与图1对应位置衍射峰的2θ误差为±0.2°~±0.5°,优选±0.2°~±0.3°,最优选±0.2°。
在本发明进一步优选方案中,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的甲磺酸盐晶型A的X-射线粉末衍射图谱中相对峰强度为前十强的衍射峰位置与图1对应位置衍射峰的2θ误差为±0.2°~±0.5°,优选±0.2°~±0.3°,最优选±0.2°;
或,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的乙烷磺酸盐晶型A的X-射线粉末衍射图谱中相对峰强度为前十强的衍射峰位置与图4对应位置衍射峰的2θ误差为±0.2°~±0.5°,优选±0.2°~±0.3°,最优选±0.2°;
或,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的硫酸盐晶型A的X-射线粉末衍射图谱中相对峰强度为前十强的衍射峰位置与图7对应位置衍射峰的2θ误差为±0.2°~±0.5°,优选±0.2°~±0.3°,最优选±0.2°;
或,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的硫酸盐晶型B的X-射线粉末衍射图谱中相对峰强度为前十强的衍射峰位置与图9对应位置衍射峰的2θ误差为±0.2°~±0.5°,优选±0.2°~±0.3°,最优选±0.2°;
或,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的盐酸盐晶型A的X-射线粉末衍射图谱中相对峰强度为前十强的衍射峰位置与图12对应位置衍射峰的2θ误差为±0.2°~±0.5°,优选±0.2°~±0.3°,最优选±0.2°;
或,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的盐酸盐晶型B的X-射线粉末衍射图谱中相对峰强度为前十强的衍射峰位置与图15对应位置衍射峰的2θ误差为±0.2°~±0.5°,优选±0.2°~±0.3°,最优选±0.2°;
或,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的氢溴酸盐晶型A的X-射线粉末衍射图谱中相对峰强度为前十强的衍射峰位置与图16对应位置衍射峰的2θ误差为±0.2°~±0.5°,优选±0.2°~±0.3°,最优选±0.2°;
或,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶 -4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的氢溴酸盐晶型B的X-射线粉末衍射图谱中相对峰强度为前十强的衍射峰位置与图18对应位置衍射峰的2θ误差为±0.2°~±0.5°,优选±0.2°~±0.3°,最优选±0.2°;
或,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的氢溴酸盐晶型C的X-射线粉末衍射图谱中相对峰强度为前十强的衍射峰位置与图21对应位置衍射峰的2θ误差为±0.2°~±0.5°,优选±0.2°~±0.3°,最优选±0.2°。
在本发明进一步优选方案中,上述化合物的酸式盐晶型为水合物或无水物,当酸式盐晶型为水合物时,水的个数为0.2-3,优选0.2、0.5、1、1.5、2、2.5或3,更优选0.5、1、2或3;进一步的,水合物中的水为管道水或结晶水或两者的结合。
在本发明进一步优选方案中上述酸式盐的制备方法,包括如下步骤:
1)称取适量的游离碱,加溶剂溶解;
2)加入适量的酸,搅拌;
3)快速离心或静置得到化合物的盐;
溶剂为有机溶剂,优选甲醇、乙醇、四氢呋喃、2-甲基四氢呋喃、甲苯、乙酸异丙酯、叔丁醇、正丁醇、丙酮、2-丁酮、二氯甲烷、乙酸乙酯或1,4-二氧六环中的至少一种;
酸选自盐酸、硫酸、硝酸、氢溴酸、氢氟酸、氢碘酸、磷酸、2,5-二羟基苯甲酸、1-羟基-2-萘甲酸、醋酸、二氯醋酸、三氯醋酸、乙酰氧肟酸、己二酸、苯磺酸、4-氯苯磺酸、苯甲酸、4-乙酰氨基苯甲酸、4-氨基苯甲酸、癸酸、己酸、辛酸、肉桂酸、柠檬酸、环己烷氨基磺酸、樟脑磺酸、天门冬氨酸、樟脑酸、葡萄糖酸、葡糖醛酸、谷氨酸、异抗坏血酸、乳酸、苹果酸、扁桃酸、焦谷氨酸、酒石酸、十二烷基硫酸、二苯甲酰酒石酸、乙烷-1,2-二磺酸、甲磺酸、乙烷磺酸、蚁酸、富马酸、半乳糖酸、龙胆酸、戊二酸、2-酮戊二酸、乙醇酸、马尿酸、羟乙基磺酸、乳糖酸、抗坏血酸、天冬氨酸、月桂酸、樟脑酸、马来酸、丙二酸、1,5-萘二磺酸、萘-2-磺酸、烟酸、油酸、乳清酸、草酸、棕榈酸、双羟萘酸、丙酸、水杨酸、4-氨基水杨酸、癸二酸、硬脂酸、丁二酸、硫氰酸、十一碳烯酸、三氟乙酸、苯磺酸、对甲基苯磺酸或L-苹果酸;优选盐酸、硫酸、磷酸、乙烷磺酸、苯磺酸、甲磺酸、富马酸、羟乙基磺酸、草酸或氢溴酸。
在本发明进一步优选方案中,上述化合物的酸式盐及其晶型的制备方法,包括如下步骤:
1)称取适量的游离碱,加反应溶剂溶解;
2)加入适量的酸,搅拌;
3)离心干燥后,得到化合物酸式盐的晶型;
溶剂为有机溶剂,优选甲醇、乙醇、四氢呋喃、2-甲基四氢呋喃、甲苯、乙酸异丙酯、叔丁醇、正丁醇、丙酮、2-丁酮、二氯甲烷、乙酸乙酯或1,4-二氧六环中的至少一种;
酸选自盐酸、硫酸、硝酸、氢溴酸、氢氟酸、氢碘酸、磷酸、2,5-二羟基苯甲酸、1-羟基-2-萘甲酸、醋酸、二氯醋酸、三氯醋酸、乙酰氧肟酸、己二酸、苯磺酸、4-氯苯磺酸、苯甲酸、4-乙酰氨基苯甲酸、4-氨基苯甲酸、癸酸、己酸、辛酸、肉桂酸、柠檬酸、环己烷氨基磺酸、樟脑磺酸、天门冬氨酸、樟脑酸、葡萄糖酸、葡糖醛酸、谷氨酸、异抗坏血酸、乳酸、苹果酸、扁桃酸、焦谷氨酸、酒石酸、十二烷基硫酸、二苯甲酰酒石酸、乙烷-1,2-二磺酸、甲磺酸、乙烷磺酸、蚁酸、富马酸、半乳糖酸、龙胆酸、戊二酸、2-酮戊二酸、乙醇酸、马尿酸、羟乙基磺酸、乳糖酸、抗坏血酸、天冬氨酸、月桂酸、樟脑酸、马来酸、丙二酸、1,5-萘二磺酸、萘-2-磺酸、烟酸、油酸、乳清酸、草酸、棕榈酸、双羟萘酸、丙酸、水杨酸、4-氨基水杨酸、癸二酸、硬脂酸、丁二酸、硫氰酸、十一碳烯酸、三氟乙酸、苯磺酸、对甲基苯磺酸或L-苹果酸;优选盐酸、硫酸、磷酸、乙烷磺酸、苯磺酸、甲磺酸、富马酸、羟乙基磺酸、草酸或氢溴酸。
本发明还提供了一种优选方案,还涉及一种药物组合物,其含有治疗有效量的上述化合物的酸式盐或其晶型,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
本发明进一步涉及任一所示的通式(I-a)化合物的酸式盐或其晶型或所述的药物组合物在制备P2X3抑制剂药物中的应用。
在一些实施例中,由本发明的化合物药学上可接受的盐及其晶型或组合物在制备治疗神经源性疾病药物中的用途;优选地所述神经源性疾病选自妇科疾病、泌尿道疾病状态、呼吸障碍疾病或疼痛相关疾病或病症,更优选子宫内膜异位症、膀胱过度活动症、肺纤维化或慢性咳嗽。
在本发明进一步优选的实施方式中,所述疼痛相关疾病或病症选自神经性疼痛或子宫肌瘤相关的疼痛或不适。
本发明另一目的在于提供一种制备含吡唑多环类衍生物的方法;
具体而言,本发明提供一种通式(I)的化合物,通式(I)为:
Figure PCTCN2022097828-appb-000017
其中:
R 1、R 2、R 3分别独立地选自氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、 氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧基、杂芳基、杂芳基氧基或-(CH 2) n1C(O)R a
R 4、R 5分别独立地选自氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧基、杂芳基、杂芳基氧基或-(CH 2) n1C(O)R a
R 6选自氢或保护基;
所述保护基可为氨基保护基,例如常见的氨基保护基,优选叔丁氧羰基、卞氧羰基、2-联苯基-2-丙氧羰基、对甲苯磺酰基、三苯甲基、甲酰基、三氟乙酰基等;
对于R 6为保护基的通式(I)的化合物,可由R 6为H的通式(I)的化合物与相应的保护试剂反应得到,并可以根据需要对保护基进行脱除;
R a选自氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧基、杂芳基或杂芳基氧基;
所述的氨基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧基、杂芳基、杂芳基氧基、-(CH 2) n1-,任选地被氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧、杂芳基或杂芳基氧基中的一个或多个取代基所取代;
n1为0、1、2、3或4;
优选地:
当R 1、R 2、R 3、R 4及R 6同时为氢时,R 5不为-C(CH 3) 3或-COOCH 3
当R 1为-CF 3,且R 2、R 3、R 4及R 6同时为氢时,R 5不为-CH 2CH 3,H或Br;
当R 1为-CF 3,且R 2、R 3、R 5及R 6同时为氢时,R 4不为-CN。
在一些具体实施方式中,本发明提供一种通式(I-1)的化合物,通式(I-1)为:
Figure PCTCN2022097828-appb-000018
优选地,所述化合物的结构选自式(I-1-1)、式(I-1-2)或式(I-1-3):
Figure PCTCN2022097828-appb-000019
其中,
R 1选自氢、氘、卤素、氰基、C 1-8烷基、C 1-8烷氧基、C 1-8卤代烷基或C 1-8卤代烷氧基;
优选氰基、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基或C 1-3卤代烷氧基;
更优选甲基、乙基、三氟甲基、甲氧基或氰基;
R 5选自氢、氘、卤素、氰基、C 1-8烷基、C 1-8烷氧基、C 1-8卤代烷基、C 1-8卤代烷氧基或-C(O)R a
优选氢、氘、卤素、氰基或-C(O)R a
更优选氢、氘、氟、氯、溴或-C(O)R a
R a选自氢、氘、卤素、氨基、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基或C 1-3卤代烷氧基;
优选氨基、甲氧基、乙氧基或异丙氧基。
本发明提供一种通式(I)的化合物或通式(I’)的化合物的制备方法,包括步骤(a):
Figure PCTCN2022097828-appb-000020
通式(II)的化合物与通式(III)的化合物在能得到通式(I’)的化合物或其盐的条件下反应得到通式(I’)的化合物或其盐;
所述反应任选地在酸存在下进行,所述酸优选为盐酸,更优选为盐酸的有机溶液,所述盐酸的有机溶液中盐酸的浓度可为1~10M/L,优选为3~6M/L,更优选为4M/L,所述酸与通式(II)的化合物的摩尔比为1以上,优选为1~5:1,更优选为3:1;
和/或,
所述反应任选地在溶剂中进行,所述溶剂优选为有机溶剂,所述溶剂中通式(II)的化合物浓度为0.05g/ml~0.5g/ml;
所述有机溶剂优选为二氧六环、四氢呋喃、甲苯、甲基四氢呋喃、乙酸乙酯、三甲苯、乙二醇、甲醇、乙醇、异丙醇或二甲醚,更优选为二氧六环;
所述有机溶剂还可以优选为N,N二甲基甲酰胺、N,N二甲基乙酰胺、N-甲基吡咯烷酮、二甲基亚砜,更优选为N,N二甲基甲酰胺;
通式(III)的化合物与通式(II)的化合物的摩尔比可为0.5~5.0:1,优选为1~1.5:1,更优选为1.2:1;
所述反应的温度可为-20~200℃,优选为50~150℃,更优选为80~120℃,进一步优选为105~110℃;
所述反应时间可为1~48小时,优选为10~30小时,更优选为16~24小时;
所述M 1、M 2分别独立地选自H、Li、Na、K或Cs,M 1优选为H,M 2优选为Na或K;
R 1、R 2、R 3、R 4、R 5的定义如通式(I)或(I-1)化合物所述。
本发明还提供一种通式(VII)的化合物的制备方法,包含步骤(b):
Figure PCTCN2022097828-appb-000021
通式(I)的化合物与通式(VI)的化合物反应得到通式(VII)的化合物;
所述通式(VI)的化合物与通式(I)的化合物的摩尔比可为0.8~3:1,优选为0.9~1.2:1,更优选为1:1;
所述反应任选地在碱存在条件下进行;
和/或,所述反应任选地在溶剂中进行;
其中,碱优选为有机碱、无机碱中的一种或多种;更优选碳酸钠、碳酸钾、氢氧化钠、氢氧化钾、三乙胺、DIPEA、DBU、DABCO中的一种或多种,所述碱与通式(I)的化合物的摩尔比为1以上,优选为1~10:1,更优选为2:1;
所述溶剂优选为有机溶剂,更优选为DMF、DMA、THF中的一种或多种;进一步优选为DMF、DMA中的一种或多种,所述溶剂中通式(I)的化合物浓度为0.05g/ml~0.5g/ml;
所述反应的温度可为0~50℃,优选为20~25℃或45℃;
X为卤素;优选为氟、氯或溴;更优选为氯或溴;
L 1选自-(CH 2) n2-、-(CH 2) n2O-、-(CH 2) n2S-、-(CH 2) n2NR c-、-(CH 2) n2C(O)NR c-或-(CH 2) n2NR cC(O)-;优选为-CH 2C(O)NH-;
n2为0、1、2或3;x为0、1、2或3;
R b、R c分别独立地选自氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、 环烷基、杂环基、芳基、芳基氧基、杂芳基或杂芳基氧基;
所述的氨基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧基、杂芳基、杂芳基氧基、-(CH 2) n2-(-(CH 2) n2-也可为-(CH 2) n2O-、-(CH 2) n2S-、-(CH 2) n2NR c-、-(CH 2) n2C(O)NR c-或-(CH 2) n2NR cC(O)-中的亚烷基链),任选地被氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧、杂芳基或杂芳基氧基中的一个或多个取代基所取代;R b优选为卤素;R c优选为H;
环A选自环烷基、杂环基、芳基或杂芳基,优选为
Figure PCTCN2022097828-appb-000022
和/或,
通式(VI)化合物优选为
Figure PCTCN2022097828-appb-000023
更优选为
Figure PCTCN2022097828-appb-000024
进一步优选为
Figure PCTCN2022097828-appb-000025
R 1、R 2、R 3、R 4、R 5的定义如通式(I)或(I-1)化合物所述;
任选地,所述通式(VII)的化合物的制备方法中还包括制备通式(I)的化合物的步骤。
本发明还提供一种通式(I-3)的化合物的制备方法,包括步骤(c-1):
Figure PCTCN2022097828-appb-000026
通式(I-2)的化合物在氨气或氨气等价物的条件下进行胺化还原反应,得到通式(I-3)的化合物;
所述氨气等价物为氨的有机溶液或氨水;所述氨的有机溶液优选为氨的甲醇溶液、氨的乙醇溶液、氨的异丙醇溶液或氨的二氧六环溶液;
优选地,通式(I-2)的化合物在氨水和/或氨的有机溶液的条件下进行胺化还原反应,得到通式(I-3)的化合物;更优选地,通式(I-2)的化合物在氨水和/或氨的甲醇溶液的条件下进行胺化还原反应,得到通式(I-3)的化合物;
所述反应的温度可为0~50℃,优选为室温25℃;
所述反应时间为1~72小时,优选为48~60小时;
R a选自氢、氘、卤素、羟基、烷氧基、卤代烷氧基、芳基氧基或杂芳基氧基;所述的烷氧基、卤代烷氧基、芳基氧基、杂芳基氧基,任选地被氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧、杂芳基或杂芳基氧基中的一个或多个取代基所取代;
R 1、R 2、R 3、R 4、R 6的定义如通式(I)或(I-1)化合物所述。
本发明还提供一种通式(I-4)的化合物的制备方法,包括步骤(d-1):
Figure PCTCN2022097828-appb-000027
通式(I-3)的化合物在脱水剂存在的条件下反应得到通式(I-4)的化合物;
所述脱水剂优选为乙酸酐、三氟乙酸酐、P 2O 5、三聚氯氰、三氯氧磷、三氯化磷、浓硫酸中的一种或多种,所述脱水剂与通式(I-3)的化合物的摩尔比为1以上,优选为1~10:1,更优选为2~2.5:1;
所述反应任选地在吡啶的存在下进行,所述吡啶与通式(I-3)的化合物的摩尔比为1以上,优选为1~10:1,更优选为2.5~3:1;进一步优选为3:1;
和/或,
所述反应任选地在溶剂中进行;所述溶剂优选为有机溶剂,更优选为DMF、DMA、THF中的一种或多种;进一步优选为DMF、THF中的一种或多种,所述溶剂中通式(I-3)的化合物浓度为0.05g/ml~0.5g/ml;
所述反应的温度可为-20~80℃,优选为室温;
所述反应时间为0.1~10小时,优选为0.5~4小时,更优选为1~2小时;R 1、R 2、R 3、R 4、R 6的定义如通式(I)或(I-1)化合物所述。
本发明还涉及一种通式(VII-4)的化合物的制备方法,其特征在于,通式(VII-4)的化合物的结构为:
Figure PCTCN2022097828-appb-000028
L 1、x、R b、环A、R 1、R 2、R 3、R 4的定义如通式(I)或(I-1)化合物和通式(VII)化合物所述;
所述制备方法包括以下步骤:
1-1:通过上述的制备方法由通式(I-4)的化合物制备通式(VII-4)的化合物;
1-2:任选地,还包括通过上述的制备方法由通式(I-3)的化合物制备通式(I-4)的化合物;
1-3:任选地,还包括通过上述的制备方法由通式(I-2)的化合物制备通式(I-3)的化合物;
1-4:任选地,还包括通过上述的制备方法制备通式(I-2)的化合物。
本发明还提供一种通式(I)的化合物、通式(II)的化合物和通式(III)的化合物,所述通式(I)的化合物、通式(II)的化合物和通式(III)的化合物作为中间体用于制备P2X3抑制剂,所述例如可为专利PCT/CN2020/134264中所公开的具有P2X3抑制活性的物质,专利PCT/CN2020/134264全文引入本申请中,所述P2X3抑制剂优选为含有母核结构为4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶的P2X3抑制剂,更优选为通式(VII-4)化合物;
Figure PCTCN2022097828-appb-000029
L 1、x、R b、环A、R 1、R 2、R 3、R 4、R 5、R 6、M 1、M 2的定义如通式(I)或(I-1)化合物、通式(I’)化合物和通式(VII)化合物所述。
在本发明中还涉及通式(II’)的化合物或通式(V’)的化合物的制备方法,所述方法还包括步骤(e)和/或步骤(c);或任选地进一步包括步骤(g),
Figure PCTCN2022097828-appb-000030
步骤(e):通式(II-1)的化合物在碱和CO 2存在的条件下,在溶剂中反应得到通式(II-2)的化合物;碱优选为LDA、丁基锂或氢氧化钠等;溶剂优选为四氢呋喃、甲苯、乙二醇二甲醚或二氯甲烷;
式(II-1)的化合物与碱的摩尔比为1:1~10,优选1:1~5,更优选1:1~1.5;
步骤(f):通式(II-2)的化合物在水合肼存在的条件下,在溶剂中反应得到通式(II’)的化合物;溶剂优选为水合肼、甲醇、乙醇、异丙醇或叔丁醇;
式(II-2)的化合物与水合肼的摩尔比为1:1~10,优选1:1~5,更优选1:1~1.5;
步骤(g):通式(V-1)的化合物在乙醇钠存在的条件下,与乙腈反应得到通式(V’)的化合物;
式(V-1)的化合物与乙腈的摩尔比为1:1~10,优选1:1~5,更优选1:1~1.5;
R 1、R 2、R 3、R 4、R 5的定义如通式(I)或(I-1)化合物所述。
本发明的含吡唑多环类衍生物的制备方法,避免使用吡唑胺衍生物、贵金属催化剂等价格昂贵的原料,反应原料价廉易得,成本低,反应条件温和,收率高,工艺成熟,质量稳定,适宜于进行工业化放大,更符合安全和环保的要求。
发明的详细说明
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。
术语“烷基”指饱和脂肪族烃基团,其为包含1至20个碳原子的直链或支链基团,优选含有1至8个碳原子的烷基,更优选1至6个碳原子的烷基,最优选1至3个碳原子的烷基。非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙基己基、2,2-二乙基己基,及其各种支链异构体等。更优选的是含有1至6个碳原子的低级烷基,非限制性实施例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基等。烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,所述取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基,本发明优选甲基、乙基、异丙基、叔丁基、卤代烷基、氘代烷基、烷氧基取代的烷基和羟基取代的烷基。
“氘代烷基”指烷基中的一个或多个氢被氘取代,其中烷基的定义如上所述。
术语“亚烷基”是指烷基的一个氢原子进一步被取代,例如:“亚甲基”指-CH 2-、 “亚乙基”指-(CH 2) 2-、“亚丙基”指-(CH 2) 3-、“亚丁基”指-(CH 2) 4-等。术语“烯基”指由至少由两个碳原子和至少一个碳-碳双键组成的如上定义的烷基,例如乙烯基、1-丙烯基、2-丙烯基、1-、2-或3-丁烯基等。烯基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基。
术语“环烷基”指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包含3至20个碳原子,优选包含3至12个碳原子,更优选包含3至6个碳原子。单环环烷基的非限制性实例包括环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等;多环环烷基包括螺环、稠环和桥环的环烷基,优选环丙基、环丁基、环己基、环戊基和环庚基。
所述环烷基环可以稠合于芳基、杂芳基或杂环烷基环上,其中与母体结构连接在一起的环为环烷基,非限制性实例包括茚满基、四氢萘基、苯并环庚烷基等。环烷基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其包含3至20个环原子,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包含3至12个环原子,其中1~4个是杂原子;更优选包含3至8个环原子;最优选包含3至8个环原子;进一步优选包含1-3氮原子的3-8元杂环基,任选地,被1-2个氧原子、硫原子、氧代基取代,包括含氮单环杂环基、含氮螺杂环基或含氮稠杂环基。
单环杂环基的非限制性实例包括氧杂环丁基、硫杂环丁基、吡咯烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、二氢咪唑基、二氢呋喃基、二氢吡唑基、二氢吡咯基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基、吖庚基、1,4-二氮杂环庚基、吡喃基或四氢噻喃二氧化物基等,优选氧杂环丁基、硫杂环丁基、四氢呋喃基、四氢吡喃基、四氢噻吩基、四氢噻喃基、四氢噻喃二氧化物基、吡咯烷基、吗啉基、哌啶基、吖庚基、1,4-二氮杂环庚基和哌嗪基;更优选氧杂环丁烷基、哌啶基、四氢吡喃基或四氢噻喃基;多环杂环基包括螺环、稠环和桥环的杂环基;其中涉及到的螺环、稠环和桥环的杂环基任选与其他基团通过单键相连接,或者通过环上的任意两个或者两个以上的原子与其他环烷基、杂环基、芳基和杂芳基进一步并环连接。
所述杂环基环可以稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基,其非限制性实例包括:
Figure PCTCN2022097828-appb-000031
等。
杂环基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基或羧酸酯基。
术语“芳基”指具有共轭的π电子体系的6至14元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为6至12元,例如苯基和萘基。更优选苯基。所述芳基环可以稠合于杂芳基、杂环基或环烷基环上,包括苯并5-10元杂芳基、苯并3-8元环烷基和苯并3-8元杂烷基,优选苯并5-6元杂芳基、苯并3-6元环烷基和苯并3-6元杂烷基,其中杂环基为含1-3氮原子、氧原子、硫原子的杂环基;或者还包含含苯环的三元含氮稠环。
芳基可以是取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
“芳基氧基”指-O-(芳基),其中芳基的定义如上所述。
术语“杂芳基”指包含1至4个杂原子、5至14个环原子的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至12元,更优选为5元或6元,例如咪唑基、呋喃基、噻吩基、噻唑基、吡唑基、噁唑基、吡咯基、三唑基、四唑基、吡啶基、嘧啶基、噻二唑、哒嗪基、吡嗪基等,优选吡啶基、噁二唑基、三唑基、噻吩基、咪唑基、吡唑基、噁唑基、嘧啶基、呋喃基、噻吩基、哒嗪基、吡嗪基或噻唑基;更优选吡啶基、呋喃基、噻吩基、嘧啶基、噁唑基、噁二唑基、吡唑基、吡咯基、噻唑基、哒嗪基、吡嗪基和噁唑基。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,其非限制性实例包括:
Figure PCTCN2022097828-appb-000032
Figure PCTCN2022097828-appb-000033
等。
杂芳基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、 杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
“杂芳基氧基”指-O-(杂芳基),其中杂芳基的定义如上所述。
术语“烷氧基”指-O-(烷基)和-O-(非取代的环烷基),其中烷基的定义如上所述。烷氧基的非限制性实例包括:甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基,烷氧基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
术语“烷硫基”指-S-(烷基)和-S-(非取代的环烷基),其中烷基的定义如上所述。烷硫基的非限制性实例包括:甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基,烷硫基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
“卤代烷基”指被一个或多个卤素取代的烷基,其中烷基如上所定义;例如三氟甲基。
“卤代烷氧基”指被一个或多个卤素取代的烷氧基,其中烷氧基如上所定义。
“羟烷基”指被羟基取代的烷基,其中烷基如上所定义。
“烯基”指链烯基,又称烯烃基,其中所述的烯基可以进一步被其他相关基团取代,例如:氢、氘、氨基、烷基、氘代烷基、卤代烷基、羟烷基、烯基、炔基、烷氧基、卤代烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、杂环基、芳基氧、杂芳基氧基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
“炔基”指(CH≡C-),其中所述的炔基可以进一步被其他相关基团取代,例如:氢、氘、氨基、烷基、氘代烷基、卤代烷基、羟烷基、烯基、炔基、烷氧基、卤代烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、杂环基、芳基氧、杂芳基氧基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。术语“烯基羰基”指-C(O)-(烯基),其中烯基的定义如上所述。烯基羰基的非限制性实例包括:乙烯基羰基、丙烯基羰基、丁烯基羰基。烯基羰基可以是任选取代的或非取代的,当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基或羧酸酯基。
“羟基”指-OH基团。
“卤素”指氟、氯、溴或碘。
“氨基”指-NH 2
“氰基”指-CN。
“硝基”指-NO 2
“羰基”指-C(O)-。
“羧基”指-C(O)OH。
“醇类溶剂”指分子中含有羟基的烷烃化合物,例如甲醇、乙醇、异丙醇。
“THF”指四氢呋喃。
“EtOAc”指乙酸乙酯。
“MeOH”指甲醇。
“DMF”指N,N-二甲基甲酰胺。
“TFA”指三氟乙酸。
“MeCN”指乙晴。
“DMA”指N,N-二甲基乙酰胺。“Et 2O”指乙醚。
“DCE”指1,2二氯乙烷。
“DIPEA”指N,N-二异丙基乙胺。
“DBU”指1,8-二氮杂二环[5.4.0]十一碳-7-烯。
“DABCO”指1,4-二氮杂二环[2.2.2]辛烷。
“NBS”指N-溴代琥珀酰亚胺。
“NIS”指N-碘代丁二酰亚胺。
“Cbz-Cl”指氯甲酸苄酯。
“Pd 2(dba) 3”指三(二亚苄基丙酮)二钯。
“Dppf”指1,1’-双二苯基膦二茂铁。
“HATU”指2-(7-氧化苯并三氮唑)-N,N,N’,N’-四甲基脲六氟磷酸酯。
“KHMDS”指六甲基二硅基胺基钾。
“LiHMDS”指双三甲基硅基胺基锂。
“MeLi”指甲基锂。
“n-BuLi”指正丁基锂。
“NaBH(OAc) 3”指三乙酰氧基硼氢化钠。
“X选自A、B、或C”、“X选自A、B和C”、“X为A、B或C”、“X为A、B和C”等不同用语均表达了相同的意义,即表示X可以是A、B、C中的任意一种或几种。
在本发明中,所述多个、多种等中的多是指2、3、4、5、6、7等。
本发明所述的氢原子均可被其同位素氘所取代,本发明涉及的实施例化合物中的任一氢原子也均可被氘原子取代。
“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明 包括该事件或环境发生或不发生的场合。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上/可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学/可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
“可药用盐”是指本发明化合物的盐,这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。
X-射线粉末衍射图谱(XRPD),是指实验观测到的衍射图或源自其的参数,通过峰位置(横坐标)及峰强度(纵坐标)表征X-射线粉末衍射图谱。本领域技术人员能够理解,其中的实验误差取决于仪器的条件、样品的准备和样品的纯度。特别是,本领域技术人员公知,X射线衍射图通常会随着仪器的条件而有所改变,本领域技术人员应理解XRPD合适的误差容限可以为:2θ±0.5°;2θ±0.4°;2θ±0.3°;2θ±0.2°。特别需要指出的是,X射线衍射图的相对强度也可能随着实验条件的变化而变化,所以峰强度的顺序不能作为唯一或决定性因素。另外,由于样品高度等实验因素的影响,会造成峰角度的整体偏移,通常允许一定的偏移。因而,本领域技术人员可以理解的是,任何具有与本发明图谱的特征峰相同或相似的晶型均属于本发明的范畴之内。
附图说明
图1为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺甲磺酸盐晶型A的XRPD图示。
图2为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺甲磺酸盐晶型A的DSC图示。
图3为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺甲磺酸盐晶型A的TGA图示。
图4为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺乙烷磺酸盐晶型A的XRPD图示。
图5为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺乙烷磺酸盐晶型A的DSC图示。
图6为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H) -基)-N-(5-氟吡啶-2-基)乙酰胺乙烷磺酸盐晶型A的TGA图示。
图7为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺硫酸盐晶型A的XRPD图示。
图8为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺硫酸盐晶型A的DSC图示。
图9为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺硫酸盐晶型B的XRPD图示。
图10为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺硫酸盐晶型B的的DSC图示。
图11为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺硫酸盐晶型B的TGA图示。
图12为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺盐酸盐晶型A的XRPD图示。
图13为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺盐酸盐晶型A的DSC图示。
图14为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺盐酸盐晶型A的TGA图示。
图15为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺盐酸盐晶型B的XRPD图示。
图16为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺氢溴酸盐晶型A的XRPD图示。
图17为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺氢溴酸盐晶型A的DSC图示。
图18为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺氢溴酸盐晶型B的XRPD图示。
图19为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺氢溴酸盐晶型B的DSC图示。
图20为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺氢溴酸盐晶型B的TGA图示。
图21为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺氢溴酸盐晶型C的XRPD图示。
图22为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺游离碱晶型I的XRPD图示。
图23为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺游离碱晶型I的DSC图示。
图24为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4 (5H)-基)-N-(5-氟吡啶-2-基)乙酰胺游离碱晶型II的XRPD图示。
图25为2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺游离碱晶型II的DSC图示。
具体实施方式
以下结合实施例进一步描述本发明,但这些实施例并非限制着本发明的范围。
实施例
本发明的化合物结构是通过核磁共振(NMR)或/和液质联用色谱(LC-MS)来确定的。NMR化学位移(δ)以百万分之一(ppm)的单位给出。NMR的测定是用Bruker AVANCE-400核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d 6),氘代甲醇(CD 3OD)和氘代氯仿(CDCl 3),内标为四甲基硅烷(TMS)。
液质联用色谱LC-MS的测定用Agilent 1200 Infinity Series质谱仪。HPLC的测定使用安捷伦1200DAD高压液相色谱仪(Sunfire C18 150×4.6mm色谱柱)和Waters 2695-2996高压液相色谱仪(Gimini C 18 150×4.6mm色谱柱)。
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,TLC采用的规格是0.15mm~0.20mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。柱层析一般使用烟台黄海硅胶200~300目硅胶为载体。
本发明中的HPLC检测方法如下:
Figure PCTCN2022097828-appb-000034
本发明实施例中的起始原料是已知的并且可以在市场上买到,或者可以采用或按照本领域已知的方法来合成。
在无特殊说明的情况下,本发明的所有反应均在连续的磁力搅拌下,在干燥氮气或氩气氛下进行,溶剂为干燥溶剂,反应温度单位为摄氏度。
中间体1
2-氯-6-三氟甲基烟酸的制备
Figure PCTCN2022097828-appb-000035
将2-氯-6-三氟甲基吡啶(50g,275.42mmol)和四氢呋喃(600mL)加入1L三口瓶中,氮气保护,反应液降温至-50℃。将二异丙基氨基锂(206.6mL,413.13mmol)慢慢滴加入反应液中,滴加完毕后,搅拌2h。称取干冰(750g),将反应液倒入盛放干冰的容器中,室温下搅拌3h,TLC检测至原料反应完全。将饱和氯化铵溶液(200mL)加入反应液,搅拌5min,然后加入水(200mL),用乙酸乙酯萃取(200mL×3),有机相用水(200ml×3)洗涤,收集水相。水相用4M盐酸调节pH至2,乙酸乙酯萃取(500mL×3),合并有机相,用饱和氯化钠溶液洗涤(300mL×2),无水硫酸钠干燥。过滤,滤液减压浓缩,用二氯甲烷(100mL)打浆纯化得到2-氯-6-三氟甲基烟酸(32g,白色固体),产率:51.5%,HPLC纯度为95.0%。
MS(ESI)m/z:226.55[M+1] +.
1H NMR(400MHz,DMSO-d 6):δ/ppm 12.67(s,1H),8.32(d,J=8.0Hz,1H),7.68(d,J=8.0Hz,1H).
中间体2
2-肼基-6-(三氟甲基)烟酸的制备
Figure PCTCN2022097828-appb-000036
开启搅拌,反应瓶中依次加入水合肼(210mL)和2-氯-6-(三氟甲基)烟酸(30g,0.133mol),搅拌溶清。油浴加热至内温75~80℃,保温反应12~16小时,原料反应完全。反应液降至室温,冰水浴冷却,用1N HCl调节pH=4~5,析出大量黄色固体,搅拌30分钟。抽滤,滤饼用水洗涤两次。滤饼真空干燥至恒重,得黄色固体,质量m=26.3g,收率89.4%,固体HPLC纯度为99.1%。
MS(ESI)m/z:222.14[M+1] +.
1H NMR(400MHz,DMSO-d 6):δ/ppm 12.67(s,1H),10.27(m,1H),7.82(d,J=8.0Hz,1H),7.70(d,J=8.0Hz,1H),4.61(m,2H).
中间体3
氰基丙酮酸乙酯钠盐的制备
Figure PCTCN2022097828-appb-000037
开启搅拌,加入乙醇钠(450ml,20%质量分数),氮气保护,冰水浴降温至0~5℃。将草酸二乙酯(96.6g,0.66mol)加入反应液中,有放热现象,30分 钟加完,搅拌20~30分钟。将乙腈(27.1g,0.66mol)加入反应液中,体系为澄清液。油浴升温至35℃反应20~24小时,逐渐析出大量固体。关闭加热,降至室温,20~25℃过滤,得到灰白色固体,滤饼用100ml冰乙醇洗涤。滤饼真空干燥至恒重,m=80g,收率74%。
MS(ESI)m/z:140.11[M-Na] -.
1H NMR(400MHz,DMSO--d 6):δ/ppm 7.52(s,1H),4.01(d,J=8.0Hz,2H),1.18(m,3H).
中间体4
2-溴-N-(5-氟吡啶-2-基)乙酰胺的制备
Figure PCTCN2022097828-appb-000038
开启搅拌,将2-氨基-5-氟吡啶(25g,0.223mol)和THF(250ml)加入500ml反应瓶中,氮气保护。控温20℃~25℃,加入DIPEA(37.4g,0.289mol),随后滴加溴乙酰溴(49.5g,0.245mol),有大量盐析出,颜色变深。滴加完毕,反应1小时,原料反应完全。将反应液倒入500ml水中,乙酸乙酯萃取(150ml×2),有机相合并用150ml水洗一次,150ml盐水洗一次,无水硫酸钠干燥。过滤,减压浓缩至剩余100ml乙酸乙酯时停止,滴加入100ml正庚烷搅拌析晶。抽滤,得到类白色固体,真空干燥至恒重,m=31.8g,收率61.2%,HPLC纯度为98.6%。
MS(ESI)m/z:234.04[M+1] +.
1H NMR(400MHz,DMSO-d 6):δ/ppm 10.92(m,1H),8.36(m,1H),8.12(m,1H),7.82-7.76(m,1H),4.36(m,2H).
实施例1
2-(2-(叔丁基)-5-氧吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺
Figure PCTCN2022097828-appb-000039
第一步:N-(3-(叔丁基)-1H-吡唑-5-基)-2-氯烟酰胺的制备
Figure PCTCN2022097828-appb-000040
冰浴条件下向2-氯烟酸(1.57g,9.96mmol)的DMF(30mL)溶液中依次加入3-(叔丁基)-1H-吡唑-5-胺(2.77g,19.93mmol),DIPEA(6.2g,49.8mmol)和HATU(5.4g,0.144mmol),撤去冰浴后搅拌1h。混合物经制备得到实施例1-1(2.5g,90%)。
MS m/z(ESI):279.7[M+H] +.
第二步:2-(叔丁基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-5(4H)-酮的制备
Figure PCTCN2022097828-appb-000041
向实施例1-1(2.5g,8.97mmol)的DMF(50mL)溶液中加入碳酸钾(1.61g,11.66mmol)and 1,4-二氮杂二环[2.2.2]辛烷(DABCO)(150.9mg,1.35mmol),反应液在室温下搅拌16小时。混合物经制备得到实施例1-2(2.1g,97%)。
MS m/z(ESI):279.7[M+H] +.
第三步:2-(2-(叔丁基)-5-氧吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的制备
Figure PCTCN2022097828-appb-000042
室温下向实施例1-2(1.5g,6.19mmol)的DMF(30mL)的溶液中加入碳酸钾(4.28g,30.96mmol)和实施例1-3(4.33g,18.57mmol)。将混合物加至热80℃,搅拌反应2h。冷却后,加入水,将沉淀过滤并用乙酸乙酯洗涤,纯化得到实施例1(656mg,收率:27%)。
MS m/z(ESI):395.4[M+H] +.
1H NMR(400MHz,DMSO-d6)δ11.01(s,1H),8.80–8.78(m,1H),8.47(d,J=7.6Hz,1H),8.30(d,J=2.8Hz,1H),8.01–7.94(m,1H),7.73–7.66(m,1H),7.49(dd,J=8.0,4.8Hz,1H),6.34(s,1H),4.87(s,2H),1.26(s,9H).
实施例2
2-(2-溴-5-氧吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺
Figure PCTCN2022097828-appb-000043
实施例2的合成参照实施例1的方法,以3-溴-1H-吡唑-5-胺替代3-(叔丁基)-1H-吡唑-5-胺,得到目标化合物(500mg,68%收率)。
MS m/z(ESI):418.2[M+H] +.
1H NMR(400MHz,DMSO-d6)δ11.32(s,1H),9.85(d,J=7.6Hz,1H),8.74(d,J=6.4Hz,1H),8.40(d,J=2.8Hz,1H),8.05-8.00(m,1H),7.78-7.73(m,1H),7.23-7.17(m,1H),6.31(s,1H),5.52(s,2H).
实施例3
N-(5-氟吡啶-2-基)-2-(2-甲基-5-氧代吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)基)乙酰胺
Figure PCTCN2022097828-appb-000044
实施例3的合成参照实施例1的方法,以3-甲基-1H-吡唑-5-胺替代3-(叔丁基)-1H-吡唑-5-胺,得到目标化合物(20mg,26%收率)。
MS m/z(ESI):353.3[M+H] +.
实施例4
N-(5-氟吡啶-2-基)-2-(2-乙基-5-氧代吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)基)乙酰胺
Figure PCTCN2022097828-appb-000045
实施例4的合成参照实施例1的方法,以3-乙基-1H-吡唑-5-胺替代3-(叔丁基)-1H-吡唑-5-胺,得到目标化合物(15mg,36%收率)。
MS m/z(ESI):367.4[M+H] +.
实施例5
N-(5-氟吡啶-2-基)-2-(2-异丙基-5-氧代吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)基)乙酰胺
Figure PCTCN2022097828-appb-000046
实施例5的合成参照实施例1的方法,以3-异丙基-1H-吡唑-5-胺替代3-(叔丁基)-1H-吡唑-5-胺,得到目标化合物(15mg,36%收率)。
MS m/z(ESI):381.4[M+H] +.
实施例6
N-(5-氟吡啶-2-基)-2-(2-异丙烯基-5-氧代吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)基)乙酰胺
Figure PCTCN2022097828-appb-000047
实施例2(100mg,0.24mmol),异丙烯基硼酸(41.2mg,0.48mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(19.2mg,0.024mmol)和碳酸铯(232.8mg,0.72mmol)在二氧六环(4mL)和水(1mL)中100℃微波搅拌1小时。旋干反应液,制备液相纯化得到实施例6(54mg,产率60%)。
MS m/z(ESI):379.4[M+H] +.
实施例7
N-(5-氟吡啶-2-基)-2-(5-氧代-2-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)乙酰胺
Figure PCTCN2022097828-appb-000048
实施例7的合成参照实施例1的方法,以3-三氟甲基-1H-吡唑-5-胺替代3-(叔丁基)-1H-吡唑-5-胺,得到目标化合物(15mg,36%收率)。
MS m/z(ESI):407.3[M+H] +.
1H NMR(400MHz,DMSO-d6)δ11.08(s,1H),8.96(dd,J=8.0,1.6Hz,1H),8.65(dd,J=8.0,1.6Hz,1H),8.37(d,J=3.2Hz,1H),8.07-8.02(m,1H),7.78-7.73(m,2H),7.05(s,1H),5.02(s,2H).
实施例8
2-(2-氨基-5-氧吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺
Figure PCTCN2022097828-appb-000049
第一步:5-氧代-4,5-二氢吡唑[1,5-a]吡啶[3,2-e]嘧啶-2-羧酸甲基的制备
Figure PCTCN2022097828-appb-000050
实施例8-1的合成方法,参考实施例1-2的合成方法,以5-氨基-1H-吡唑-3-羧酸甲酯代替3-(叔丁基)-1H-吡唑-5-胺,得到实施例8-1(500mg,73%)。
MS:m/z(ESI):245.2[M+H] +.
第二步:4-(2-((5-氟吡啶-2-基)氨基)-2-氧乙基)-5-氧代-4,5-二氢吡唑[1,5-a]吡啶[3,2-e]嘧啶-2-羧酸甲酯的制备
Figure PCTCN2022097828-appb-000051
实施例8-2的合成方法,参考实施例1的合成方法,以实施例8-1为原料,得到标题化合物实施例8-2(500mg,51%)。
MS m/z(ESI):397.3[M+H] +.
第三步:4-(2-((5-氟吡啶-2-基)氨基)-2-氧乙基)-5-氧代-4,5-二氢吡唑[1,5-a]吡啶[3,2-e]嘧啶-2-羧酸的制备
Figure PCTCN2022097828-appb-000052
室温下向实施例8-2(490mg,1.24mmol)的四氢呋喃(10mL)的溶液中加入LiOH(519mg,12.36mmol)的水(2mL)溶液。混合物室温搅拌反应3h,然后用1M HCl调节pH至3左右,浓缩至干得到实施例8-3(470mg,99%)。
MS m/z(ESI):383.3[M+H] +.
第四步:2-(2-氨基-5-氧吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的制备
Figure PCTCN2022097828-appb-000053
向实施例8-3(450mg,1.2mmol)在1,4-二氧六环(10mL)、Et3N(33μL,0.24mmol)和BOP试剂(598mg,1.35mmol)中的溶液中加入胺,并在室温下搅拌20min。添加叠氮化钠(160mg,2.46mmol)和四丁基溴化铵(786mg,2.46mmol),并继续搅拌1小时。然后用1,4-二氧六环(12mL)稀释反应,加入2M H 2SO 4(4mL)水溶液,并在100℃下加热2h。蒸发溶剂,将残渣稀释水,用乙酸乙酯萃取。用盐水清洗有机层,无水硫酸钠上干燥,并蒸发溶剂。柱层析纯化得到实施例8(360mg,86%)。
MS m/z(ESI):354.3[M+H] +.
实施例9
2-(2-环丙基-5-氧吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺
Figure PCTCN2022097828-appb-000054
实施例9的合成方法,参考实施例6的合成方法,以环丙基硼酸代替乙丙烯硼酸,得到标题化合物实施例9(8mg,51%)。
MS m/z(ESI):378.4[M+H] +.
实施例10
2-(2-(叔丁基)-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺
Figure PCTCN2022097828-appb-000055
实施例10的合成方法,参考实施例1的合成方法,以2-氯-6-三氟甲基烟酸为原料,得到标题化合物实施例10(25mg,46%)。
MS m/z(ESI):463.1[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.09(s,1H),7.78(d,J=8.0Hz,1H),8.38(d,J=2.8Hz,1H),8.09-8.04(m,1H),8.00(d,J=8.0Hz,1H),7.79-7.74(m,1H),6.52(s,1H),4.95(s,2H),1.34(s,9H).
实施例11
N-(5-氟吡啶-2-基)-2-(2-甲基-5-氧代-8-(三氟甲基)吡唑啉[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)乙酰胺
Figure PCTCN2022097828-appb-000056
实施例11的合成方法,参考实施例2的合成方法,得到标题化合物实施例 11(18mg,30%)。
MS m/z(ESI):421.1[M+H] +.
1H NMR(400MHz,DMSO-d6)δ11.06(s,1H),8.80(d,J=8.0Hz,1H),8.37(s,1H),8.07-8.03(m,1H),8.00(d,J=8.0Hz,1H),7.79-7.73(m,1H),6.28(s,1H),4.95(s,2H),2.33(s,3H).
实施例12
2-(2-乙基-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺
Figure PCTCN2022097828-appb-000057
第一步:5-氨基-3-乙基-1H-吡唑-1-羧酸叔丁酯的制备
Figure PCTCN2022097828-appb-000058
将3-乙基-1H-吡唑-5-胺(2.0g,18.0mmol)溶解在无水二氯甲烷(50mL)中,加入三乙胺(2.2g,21.6mmol)和二碳酸二叔丁酯(4.7g,21.6mmol),室温反应16小时。反应液依次用水(50mL*2)、饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗产品柱层析(乙酸乙酯/二氯甲烷=0~20%),得到标题产物实施例12-1(3.4g),产率:89.5%。
MS:m/z(ESI):212.1[M+H] +.
第二步:5-氨基-3-乙基-1H-吡唑-1-羧酸叔丁酯的制备
Figure PCTCN2022097828-appb-000059
将实施例12-1(3.4g,16.1mmol)溶解在无水二氯甲烷(60mL)中,加入三乙胺(5.4g,53.1mmol),氮气保护下在0℃滴加现制备的2-氯-6-三氟甲基烟酸酰氯(4.3g,17.7mmol)的二氯甲烷溶液(50mL),加完室温反应30分钟。反应液依次用水(200mL*2)、饱和氯化钠溶液(200mL)洗涤,无水硫酸钠 干燥,过滤,滤液减压浓缩,所得粗产品硅胶柱层析(乙酸乙酯/石油醚=0~20%),得到实施例12-2(2.6g),产率:38.2%。
MS:m/z(ESI):319.1[M-Boc+H] +.
第三步:N-(3-乙基-1H-吡唑-5-基)-2-氯-6-(三氟甲基)烟酰胺的制备
Figure PCTCN2022097828-appb-000060
将实施例12-2(2.6g,6.2mmol)溶解在无水二氯甲烷(10mL)中,加入盐酸二氧六环溶液(4M,20mL),室温反应4小时。反应液直接旋干,得到实施例12-3(1.9g),产率:96.0%。
MS:m/z(ESI):319.0[M+H] +.
第四步:2-乙基-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-5(4H)-酮
Figure PCTCN2022097828-appb-000061
将实施例12-3(1.9g,6.0mmol)溶解在N,N-二甲基甲酰胺(20mL)中,加入碳酸钾(2.5g,18.0mmol),加热至120℃反应2小时。反应液冷却至室温,直接用于下步反应。
MS:m/z(ESI):283.1[M+H] +.
第五步:2-(2-乙基-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺
Figure PCTCN2022097828-appb-000062
在实施例12-4(1.0g,3.5mmol)的N,N-二甲基甲酰胺(20mL)反应液中,加入碳酸钾(1.5g,10.6mmol)和2-溴-N-(5-氟吡啶-2-基)乙酰胺(0.99g,4.2mmol),40℃反应2小时。反应液冷却至室温,倒入300mL水中,乙酸乙酯萃取(200mL*3)。合并有机相,依次用水(200mL*2)、饱和氯化钠溶液(200mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,粗产品乙酸乙酯重 结晶,得到实施例12。
1H NMR(400MHz,DMSO-d 6)δ11.06(s,1H),8.79(d,J=7.6Hz,1H),8.37(s,1H),8.07–8.03(m,1H),8.00(d,J=8.0Hz,1H),7.79–7.72(m,1H),6.36(s,1H),4.96(s,2H),2.70(q,J=7.6Hz,2H),1.25(t,J=7.6Hz,3H).
MS m/z(ESI):435.1[M+H] +.
实施例13
2-(2-环丙基-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺
Figure PCTCN2022097828-appb-000063
实施例13的合成方法,参考实施例1的合成方法,得到标题化合物实施例13(17mg,28%)。
1H NMR(400MHz,DMSO-d6)δ11.05(s,1H),8.78(d,J=8.0Hz,1H),8.37(s,1H),8.08–8.02(m,1H),7.98(d,J=8.4Hz,1H),7.79-7.73(m,1H),6.23(s,1H),4.91(s,2H),2.11–2.04(m,1H),1.04-0.98(m,2H),0.82–0.78(m,2H).
MS m/z(ESI):447.1[M+H] +.
实施例14
N-(5-氟吡啶-2-基)-2-(2-异丙基-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)乙酰胺
Figure PCTCN2022097828-appb-000064
实施例14的合成方法,参考实施例4的合成方法,得到标题化合物实施例31(10mg,22%)。
MS m/z(ESI):449.1[M+H] +.
1H NMR(400MHz,DMSO-d6)δ11.06(s,1H),8.79(d,J=8.0Hz,1H),8.37(s, 1H),8.09-8.03(m,1H),8.00(d,J=8.0Hz,1H),7.79-7.74(m,1H),6.42(s,1H),4.96(s,2H),3.08-3.01(m,1H),1.29(s,3H),1.27(s,3H).
实施例15
2-(2-溴-5-氧-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的制备
第一步:5-氨基-3-溴-1H-吡唑-1-羧酸叔丁酯的制备
Figure PCTCN2022097828-appb-000065
将3-溴-1H-吡唑-5-胺(10.0g,61.7mmol)溶解在无水二氯甲烷(100mL)中,加入三乙胺(7.48g,74.1mmol)和二碳酸二叔丁酯(16.0g,74.1mmol),室温反应16小时。反应液依次用水(50mL*2)、饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗产品柱层析(乙酸乙酯/二氯甲烷=0~20%),得到标题产物5-氨基-3-溴-1H-吡唑-1-羧酸叔丁酯实施例15-1(14.5g),产率:89.5%。
MS:m/z(ESI):262.0[M+H] +
1H NMR(400MHz,DMSO-d6)δ6.62(s,2H),5.41(s,1H),1.56(s,9H).
第二步:5-氨基-3-溴-1H-吡唑-1-羧酸叔丁酯的制备
Figure PCTCN2022097828-appb-000066
将5-氨基-3-溴-1H-吡唑-1-羧酸叔丁酯实施例15-1(14.5g,55.3mmol)溶解在无水二氯甲烷(200mL)中,加入三乙胺(18.5g,183mmol),氮气保护下在0℃滴加现制备的2-氯-6-三氟甲基烟酸酰氯(13.0g,61.0mmol)的二氯甲烷溶液(50mL),加完室温反应30分钟。反应液依次用水(200mL*2)、饱和氯化钠溶液(200mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗产品硅胶柱层析(乙酸乙酯/石油醚=0~20%),得到5-氨基-3-溴-1H-吡唑-1-羧酸叔丁酯实施例15-2(9.5g),产率:38.2%。
MS:m/z(ESI):371.0[M-Boc+H] +
1H NMR(400MHz,DMSO-d6)δ11.08(s,1H),8.40(d,J=7.6Hz,1H),8.14(d,J=7.6Hz,1H),6.96(s,1H),1.58(s,9H).
第三步:N-(3-溴-1H-吡唑-5-基)-2-氯-6-(三氟甲基)烟酰胺的制备
Figure PCTCN2022097828-appb-000067
将5-氨基-3-溴-1H-吡唑-1-羧酸叔丁酯实施例51-2(8.0g,17.1mmol)溶解在无水二氯甲烷(20mL)中,加入盐酸二氧六环溶液(4M,40mL),室温反应4小时。反应液直接旋干,得到N-(3-溴-1H-吡唑-5-基)-2-氯-6-(三氟甲基)烟酰胺实施例15-3(6.2g),产率:98.4%。
MS:m/z(ESI):368.9[M+H] +
1H NMR(400MHz,DMSO-d6)δ11.50(s,1H),8.39(d,J=7.6Hz,1H),8.10(d,J=7.6Hz,1H),6.53(s,1H).
第四步:2-溴-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-5(4H)-酮的制备
Figure PCTCN2022097828-appb-000068
将N-(3-溴-1H-吡唑-5-基)-2-氯-6-(三氟甲基)烟酰胺实施例15-3(6.2g,16.8mmol)溶解在N,N-二甲基甲酰胺(80mL)中,加入碳酸钾(6.96g,50.4mmol),加热至120℃反应2小时。反应液冷却至室温,直接用于下步反应。
MS:m/z(ESI):333.0[M+H] +
第五步:2-(2-溴-5-氧-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的制备
Figure PCTCN2022097828-appb-000069
在2-溴-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-5(4H)-酮实施例51-4(NA,16.8mmol)的N,N-二甲基甲酰胺(80mL)反应液中,加入碳酸钾(6.96g,50.4mmol)和2-溴-N-(5-氟吡啶-2-基)乙酰胺(4.7g,20.2mmol),40℃反应2小时。反应液冷却至室温,倒入300mL水中,乙酸乙酯萃取(200mL*3)。合并有机相,依次用水(200mL*2)、饱和氯化钠溶液(200mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,粗产品乙酸乙酯重结晶,得到标题产物2- (2-溴-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺实施例15。
1H NMR(400MHz,DMSO-d6)δ11.05(s,1H),8.84(d,J=8.0Hz,1H),8.37(s,1H),8.09(d,J=8.0Hz,1H),8.07–8.02(m,1H),7.80–7.73(m,1H),6.78(s,1H),4.96(s,2H).
MS m/z(ESI):486.2[M+H] +.
实施例16
N-(5-氟吡啶-2-基)-2-(5-氧-2,8-双(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)乙酰胺
Figure PCTCN2022097828-appb-000070
实施例16的合成方法,参考实施例1的合成方法,得到标题化合物实施例16(10mg,33%)。
MS m/z(ESI):475.3[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.07(s,1H),8.91(d,J=8.0Hz,1H),8.37(d,J=3.1Hz,1H),8.19(d,J=8.1Hz,1H),8.14–7.98(m,1H),7.76(t,J=8.8Hz,1H),7.13(s,1H),5.04(s,2H).
实施例17
2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺
Figure PCTCN2022097828-appb-000071
第一步:2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的制备
Figure PCTCN2022097828-appb-000072
在室温下将实施例15(300mg,0.619mmol)和Zn(CN) 2(300mg,2.56mmol),Pd 2(dba) 3(20mg,0.022mmol),Pd(dppf)Cl 2(30mg,0.036mmol)和Zn粉(10mg,0.154mmol)溶在DMA(10mL)中,向里鼓氮气2分钟。然后微波加热140度反应8小时。冷却至室温,并用乙酸乙酯(50mL)萃取,有机相用饱和食盐水洗两遍。将有机相干燥(Na 2SO 4),减压浓缩,送p-HPLC(FA)得到100mg(38%收率)的标题化合物。
1H NMR(400MHz,DMSO-d6)δ11.07(s,1H),8.92(d,J=8.2Hz,1H),8.37(d,J=3.1Hz,1H),8.22(d,J=7.9Hz,1H),8.05(s,1H),7.77(t,J=8.6Hz,1H),7.24(s,1H),5.01(s,2H).
MS m/z(ESI):432.3[M+H] +.
标题化合物还可以采用以下方法制备:
第一步:2-羧酸乙酯-5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶的制备
Figure PCTCN2022097828-appb-000073
开启搅拌,反应瓶中加入氰基丙酮酸乙酯钠盐(17.7g,0.108mol),二氧六环(200ml),氮气保护,固体不溶。20~25℃加入4M/L的盐酸二氧六环溶液(68ml,0.27mol),搅拌15~30分钟。加入原料2-肼基-6-(三氟甲基)烟酸(20g,0.09mol),油浴加热至105~110℃,反应24小时。滴加400ml水(相对于有机溶剂为1.5V),析出褐色固体。过滤,滤饼用400ml水洗。所得粗品加入甲醇(100ml),室温搅拌2小时,过滤得白色固体,m=12.5g,收率40.3%,HPLC纯度为99.2%。
MS(ESI)m/z:327.24[M+1] +.
1H NMR(400MHz,DMSO-d 6):δ/ppm 12.68(s,1H),8.81(d,J=8.0Hz,1H),8.11(d,J=8.0Hz,1H),6.30(s,1H),4.35-4.40(m,2H),1.34-1.38(m,3H).
2-羧酸乙酯-5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶的制备还可以采用如下制备方法:
开启搅拌,反应瓶中加入氰基丙酮酸乙酯钠盐(35.4g,0.22mol),N,N-二甲基甲酰胺(400ml),氮气保护,固体不溶。控温0~10℃加入4M/L的盐酸二氧六环溶液(135ml,0.54mol),随后再加入原料2-肼基-6-(三氟甲基)烟酸(40g,0.18mol),油浴加热至105~110℃,反应24小时。滴加800ml水(相对于有机溶剂为1.5V),析出褐色固体。过滤,滤饼用800ml水洗。所得粗品加入甲醇(200ml),室温搅拌2小时,过滤得白色固体,m=33g,收率56.2%,HPLC纯度为99.4%。
MS(ESI)m/z:327.24[M+1] +.
1H NMR(400MHz,DMSO-d 6):δ/ppm 12.68(s,1H),8.81(d,J=8.0Hz,1H),8.11(d,J=8.0Hz,1H),6.30(s,1H),4.35-4.40(m,2H),1.34-1.38(m,3H).
第二步:2-甲酰胺-5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶的制备
Figure PCTCN2022097828-appb-000074
开启搅拌,25℃下,将2-羧酸乙酯-5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶(11.5g,35.3mmol),氨水(120ml)加入到500ml的单口瓶中,黄色固体不溶。室温搅拌48~60小时,原料反应完全,减压浓缩,然后加入120ml甲醇打浆1小时。冰水浴降至0~5℃,搅拌30分钟,抽滤,滤饼用少量冰甲醇淋洗,得到淡黄色固体,减压干燥至恒重,质量m=8.9g,收率85.0%,HPLC纯度为98.6%。
MS(ESI)m/z:298.20[M+1] +.
1H NMR(400MHz,DMSO-d 6):δ/ppm 11.68(s,1H),8.74(d,J=8.0Hz,1H),8.01(d,J=8.0Hz,1H),7.81(s,1H),7.46(s,1H),6.18(s,1H).
2-甲酰胺-5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶的制备还可以采用如下制备方法:
开启搅拌,25℃下,将2-羧酸乙酯-5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶(11.5g,35.3mmol),氨甲醇(7M/L,115ml)以及氨水(58ml)加入到500ml的单口瓶中,黄色固体不溶。室温搅拌48~60小时,原料反应完全,减压浓缩,然后加入120ml甲醇打浆1小时。冰水浴降至0~5℃,搅拌30分钟,抽滤,滤饼用少量冰甲醇淋洗,得到淡黄色固体,减压干燥至恒重,质量m=9.1g,收率87.0%,HPLC纯度为99.0%。
MS(ESI)m/z:298.20[M+1] +.
1H NMR(400MHz,DMSO-d 6):δ/ppm 11.68(s,1H),8.74(d,J=8.0Hz,1H),8.01(d,J=8.0Hz,1H),7.81(s,1H),7.46(s,1H),6.18(s,1H).
第三步:2-氰基-5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶的制备
Figure PCTCN2022097828-appb-000075
开启搅拌,将2-甲酰胺-5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶(6.2g,0.021mol)和N,N-二甲基甲酰胺(62ml)加入250ml反应瓶中,氮气保护。加入吡啶(4.9g,0.062mol),冰水浴降温,控温0℃~5℃,滴加三氯氧磷(8g,0.052mol),反应体系变为灰色浑浊液。滴加完毕后撤去冰水浴,升至室温反应1~2小时,原料反应完全。冰水浴降温,加入186ml水,搅拌20~30分钟,抽滤,滤饼用水洗涤,得类白色固体。将湿品转移至250ml单口瓶中,加入60ml的N,N-二甲基甲酰胺溶清,过滤除去机械杂质,滴加入180ml水,冰水浴降温搅拌30分钟。抽滤,得到类白色固体,真空干燥至恒重,质量m=4.7g,收率80.7%,HPLC纯度为99.0%。
MS(ESI)m/z:280.18[M+1] +.
1H NMR(400MHz,DMSO-d 6):δ/ppm 12.86(s,1H),8.82(d,J=8.0Hz,1H),8.15(d,J=8.0Hz,1H),6.67(s,1H).
2-氰基-5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶的制备还可以采用如下制备方法:
开启搅拌,将2-甲酰胺-5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶(15g,0.05mol)和四氢呋喃(150ml)加入500ml反应瓶中,氮气保护。加入吡啶(11.9g,0.15mol),冰水浴降温,控温0℃~5℃,滴加三氟乙酸酐(26.3g,0.125mol),反应体系变为灰色浑浊液。滴加完毕后撤去冰水浴,升至室温反应1~2小时,原料反应完全。冰水浴降温,加入450ml水,搅拌20~30分钟,抽滤,滤饼用水洗涤,得类白色固体。将湿品转移至500ml单口瓶中,加入145ml的N,N-二甲基甲酰胺溶清,过滤除去机械杂质,滴加入435ml水,冰水浴降温搅拌30分钟。抽滤,得到类白色固体,真空干燥至恒重,质量m=12.2g,收率87.1%,HPLC纯度为99.0%。
MS(ESI)m/z:280.18[M+1] +.
1H NMR(400MHz,DMSO-d 6):δ/ppm 12.86(s,1H),8.82(d,J=8.0Hz,1H),8.15(d,J=8.0Hz,1H),6.67(s,1H).
第四步:2-(2-氰基-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的制备
Figure PCTCN2022097828-appb-000076
将2-氰基-5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶(6.0g,0.0215mol)、N,N-二甲基甲酰胺(60mL)加入500ml三口瓶中,开启搅拌,固体溶清,加入碳酸钾(5.9g,0.043mol)。将2-溴-N-(5-氟吡啶-2-基)乙酰胺(5.0g,0.0215mol)用DMF(30ml)溶解滴加入反应体系中,升温至45℃搅拌1~2小时,原料反应完全。关闭加热,冷却到室温,加入水(135ml),析出大量固体,搅拌30分钟。抽滤,滤饼用水洗涤两次。将粗品用DCM/丙酮=3:1(250ml)溶解,加入活性炭(0.5g)以及无水硫酸钠(15g),40℃搅拌30分钟。过滤,滤液减压浓缩至干,用50ml丙酮带一次,蒸干。加入100ml丙酮,加热到65℃回流,仍未溶清,滴加200ml正庚烷,加热搅拌1小时,关闭加热,自然冷却到室温,继续搅拌1小时。过滤,得类白色固体,真空干燥至恒重,质量m=8.0g,收率86.4%,HPLC纯度为99.2%。
MS(ESI)m/z:432.31[M+1] +.
1H NMR(400MHz,DMSO-d 6):δ/ppm 11.06(s,1H),8.92(d,J=8.0Hz,1H),8.36(d,J=4.0Hz,1H),8.22(d,J=8.0Hz,1H),8.05(m,1H).7.76(m,1H),7.24(s,1H),5.01(m,2H).
实施例18
N-(5-氟吡啶-2-基)-2-(5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)乙酰胺
Figure PCTCN2022097828-appb-000077
第一步:N-(1H-吡唑-5-基)-2-氯烟酰胺的制备
Figure PCTCN2022097828-appb-000078
冰浴条件下向2-氯烟酸(1.57g,9.96mmol)的DMF(30mL)溶液中依次加入1H-吡唑-5-胺(1.66g,19.93mmol),DIPEA(6.2g,49.8mmol)和HATU(5.4g,0.144mmol),撤去冰浴后搅拌1h。混合物经制备得到实施例18-1(2.0g,90%)。
MS m/z(ESI):291.0[M+H] +.
第二步:吡唑[1,5-a]吡啶[3,2-e]嘧啶-5(4H)-酮的制备
Figure PCTCN2022097828-appb-000079
向实施例18-1(2.0g,8.97mmol)的DMF(50mL)溶液中加入碳酸钾(1.61g,11.66mmol)and 1,4-二氮杂二环[2.2.2]辛烷(DABCO)(150.9mg,1.35mmol),反应液在室温下搅拌16小时。混合物经制备得到实施例18-2(1.6g,97%)。
MS m/z(ESI):255.0[M+H] +.
第三步:2-(2-(叔丁基)-5-氧吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的制备
Figure PCTCN2022097828-appb-000080
室温下向实施例18-2(1.5g,8.06mmol)的DMF(30mL)的溶液中加入碳酸钾(2.23g,16.11mmol)和实施例1-3(2.25g,9.67mmol)。将混合物加至热80℃,搅拌反应2h。冷却后,加入水,将沉淀过滤并用乙酸乙酯洗涤,纯化得到实施例18(2.1g,收率:78%)。
1H NMR(400MHz,DMSO-d6)δ11.06(s,1H),8.83(d,J=8.0Hz,1H),8.37(d,J=3.2Hz,1H),8.06(d,J=8.0Hz,1H),8.05–8.02(m,1H),7.98(d,J=2.0Hz,1H),7.78–7.73(m,1H),6.46(s,1H),5.00(s,2H).
MS m/z(ESI):407.3[M+H] +.
实施例19
2-(2-氯-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺
Figure PCTCN2022097828-appb-000081
实施例19的合成参照实施例1的方法,以3-氯-1H-吡唑-5-胺替代3-(叔丁基)-1H-吡唑-5-胺,得到目标化合物(31mg,26%收率)。
1H NMR(400MHz,DMSO-d6)δ11.05(s,1H),8.84(d,J=8.1Hz,1H),8.36(s,1H),8.15–7.99(m,2H),7.76(t,J=9.0Hz,1H),6.73(s,1H),4.96(s,2H).
MS m/z(ESI):441.7[M+H] +.
实施例20
2-(3-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺
Figure PCTCN2022097828-appb-000082
实施例20的合成参照实施例1的方法,以4-氰基-1H-吡唑-5-胺替代3-(叔丁基)-1H-吡唑-5-胺,得到目标化合物。
第一步:5-氨基-4-氰基-1H-吡唑-1-羧酸叔丁酯的制备
Figure PCTCN2022097828-appb-000083
将5-氨基-1H-吡唑-4-甲腈(2.0g,18.5mmol)溶解在无水二氯甲烷(40mL)中,加入三乙胺(3.74g,37.0mmol)和二碳酸二叔丁酯(4.44g,20.4mmol),室温反应16小时。反应液减压浓缩,石油醚(50mL)打浆,得到标题产物5-氨基-4-氰基-1H-吡唑-1-羧酸叔丁酯实施例20-1(3.5g),产率:90.9%。
1H NMR(400MHz,DMSO-d6)δ7.77(s,1H),7.63(s,2H),1.56(s,9H).
第二步:5-(2-氯-6-(三氟甲基)烟酰胺)-4-氰基-1H-吡唑-1-羧酸叔丁酯的制备
Figure PCTCN2022097828-appb-000084
将5-氨基-4-氰基-1H-吡唑-1-羧酸叔丁酯实施例20-1(3.5g,16.8mmol)溶解在无水二氯甲烷(50mL)中,加入三乙胺(5.35g,7.37mmol),氮气保护下在0℃滴加现制备的2-氯-6-三氟甲基烟酸酰氯(4.3g,17.6mmol)的二氯甲烷溶液(50mL),加完室温反应1小时。反应液依次用水(50mL*2)、饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,所得粗产品硅胶柱层析(乙酸乙酯/石油醚=0~40%),得到5-(2-氯-6-(三氟甲基)烟酰胺)-4-氰基-1H-吡唑-1-羧酸叔丁酯实施例20-2(2.8g),产率:38.2%。
MS:m/z(ESI):432.8[M+NH 4] +
1H NMR(400MHz,DMSO-d6)δ11.87(s,1H),9.23(s,1H),8.43(d,J=7.6Hz,1H),8.13(d,J=7.6Hz,1H),1.59(s,9H).
第三步:2-氯-N-(4-氰基-1H-吡唑-5-基)-6-(三氟甲基)烟酰胺的制备
Figure PCTCN2022097828-appb-000085
将5-(2-氯-6-(三氟甲基)烟酰胺)-4-氰基-1H-吡唑-1-羧酸叔丁酯实施例20-2(2.8g,6.73mmol)溶解在无水二氯甲烷(10mL)中,加入盐酸二氧六环溶液(4M,30mL),室温反应5小时。反应液直接旋干,得到2-氯-N-(4-氰基-1H-吡唑-5-基)-6-(三氟甲基)烟酰胺实施例20-3(2.1g),产率:98.8%。
MS:m/z(ESI):315.8[M+H] +
第四步:5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶[3,2-e]嘧啶-3-甲腈的制备
Figure PCTCN2022097828-appb-000086
将2-氯-N-(4-氰基-1H-吡唑-5-基)-6-(三氟甲基)烟酰胺实施例20-3(2.1g,6.65mmol)溶解在N,N-二甲基甲酰胺(40mL)中,加入碳酸钾(1.84g,13.3mmol),加热至120℃反应2小时。反应液冷却至室温,1M的稀盐酸调pH 至6,乙酸乙酯萃取(100mL*2)。合并有机相,依次用水(100mL*2)、饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,乙酸乙酯(15mL)打浆,得到5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶[3,2-e]嘧啶-3-甲腈20-4(1.3g),产率:69.9%。
MS:m/z(ESI):279.8[M+H] +
第四步:2-(3-氰基-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的制备
Figure PCTCN2022097828-appb-000087
将5-氧代-8-(三氟甲基)-4,5-二氢吡唑并[1,5-a]吡啶[3,2-e]嘧啶-3-甲腈实施例20-4(500mg,1.79mmol)溶解在N,N-二甲基甲酰胺(20mL)中,加入碳酸钾(371mg,2.69mmol)和2-溴-N-(5-氟吡啶-2-基)乙酰胺(501mg,2.15mmol),40℃反应2小时。反应液冷却至室温,倒入100mL水中,乙酸乙酯萃取(50mL*2)。合并有机相,依次用水(50mL*2)、饱和氯化钠溶液(50mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,粗产品乙酸乙酯打浆,所得母液减压浓缩后反向HPLC制备,得到标题产物2-(3-氰基-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺实施例20。MS m/z(ESI):432.3[M+H] +.
1H NMR(400MHz,DMSO-d6)δ11.2(s,1H),8.93(d,J=8.0Hz,1H),8.58(s,1H),8.38(d,J=3.2Hz,1H),8.20(d,J=8.0Hz,1H),8.07-8.04(m,1H),7.81-7.75(m,1H),5.19(s,2H).
实施例21
N-(5-氟吡啶-2-基)-2-(2-(羟甲基)-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)乙酰胺
Figure PCTCN2022097828-appb-000088
第一步:N-(5-氟吡啶-2-基)-2-(2-(羟甲基)-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)乙酰胺的制备
Figure PCTCN2022097828-appb-000089
0℃下,向实施例22-1(100mg,0.22mmol)(实施例21-1的合成方法参照
实施例8-2)的THF(2mL)中的溶液中加入二异丁基氢化铝(1M的甲苯溶液,0.66mL,0.66mmol),将混合物在室温搅拌过夜。加入Rochelle's盐溶液(1.0M,5ml);然后加入乙酸乙酯(5mL),将得到的悬浮液在室温搅拌直至实现透明相分离,分离有机相,并用EtOAc(3×40ml)萃取水相。合并的有机层用饱和碳酸氢钠水溶液(50mL)和饱和食盐水(50mL)洗涤,经无水硫酸钠干燥,并浓缩纯化得到目标化合物(32mg,34%收率)。
MS m/z(ESI):437.1[M+H] +.
1H NMR(400MHz,DMSO)δ11.06(s,1H),8.82(d,J=7.9Hz,1H),8.37(s,1H),8.02(m,2H),7.76(s,1H),6.40(s,1H),5.44(s,1H),5.00(s,2H),4.56(s,2H).
生物学测试评价
以下结合测试例进一步描述解释本发明,但这些实施例并非意味着限制本发明的范围。
测试例1、本发明化合物在稳定表达1321N1-hP2X3受体细胞中对钙离子流动能力影响的测定
实验目的:测定化合物对1321N1-hP2X3受体的抑制活性。
实验仪器:
384孔-细胞板(Corning;3712);384孔-化合物板(Corning;3657);
384孔-试验板(LABCYTE;P-05525);FLIPR(Molecular Devices)。
实验试剂:
DMEM(Gibco;11965);FBS(Gibco;10099-141);
潮霉素B(Invitrogen,10687010);Matrix(Thermo;5416);
DMSO(Sigma;D2650);HBSS(Invitrogen;14025);
HEPES(Invitrogen;15630080);Probenecid(Sigma;P8761);
Versene(Gibco;15040066);G418(Sigma;G5013);
Figure PCTCN2022097828-appb-000090
Calcium 4 Assay Kit(Molecular Devices;R8141);
α,β-meATP(Sigma;M6517);ATP hydrolytic enzyme(Sigma;A7646);
稳转细胞株:1321N1-hP2X3(由上海睿智化学研究有限公司提供)。
实验方法:
1.试剂配制:
Assay buffer:1*HBSS+20mM HEPES;
细胞培养基:DMEM+10%FBS+75μg/mL潮霉素B+300μg/mL G418;
铺板培养基:DMEM+10%DPBS;
0.5*Dye:10*Dye stock+1.25 Probenecid+1*assay buffer+0.5U/mL ATP hydrolytic enzyme;
2.细胞株培养于细胞培养基中,37℃,5%CO 2至70%~90%融合度,弃培养基,取出细胞,加2mL Versene,置37℃培养箱2-5min,加10mL铺板培养基收集细胞,细胞计数,每孔加50μL(1×10 4个细胞/well的密度)播种到384孔-试验板孵育16-24小时(至少过夜)。
3.丢弃培养液,加入30μL 1X染料,37℃避光孵育60min。
4.用DMSO将化合物粉末溶解成20mM储备液,准备检测需要浓度的180X化合物并梯度稀释10个浓度点。
5.准备化合物板:用ECHO转移500nL 180X化合物到化合物板(FLIPR用的source板)中,每孔加入30μL assay buffer,轻微振摇20-40分钟。
6.上机检测:每孔取15μL 3X化合物加入细胞板,FLIPR仪器加样,检测钙信号,15分钟后,每孔加22.5μL 3X激动剂(EC 80浓度),检测钙信号。
实验数据处理方法:
通过FLIPR读取钙信号值。实验中每个采样时间点的计算后的输出结果是340/510nm与380/510nm波长信号的比值。最大值减去最小值的计算来源自比值信号曲线。使用GraphPad prism拟合百分比抑制率和十点浓度数据至参数非线性逻辑公式计算出化合物的IC 50值。
实验结果:本发明实施例化合物在1321N1-hP2X3受体细胞功能钙流试验中 的结果如表10所示:
表10
Figure PCTCN2022097828-appb-000091
实验结论:以上数据显示,本发明所示的化合物在1321N1-hP2X3受体细胞功能钙流试验中显示出良好的抑制作用。
测试例2、本发明化合物在稳定表达1321N1-hP2X2/3受体细胞中对钙离子流动能力影响的测定
实验目的:测定化合物对1321N1-hP2X2/3受体的抑制活性。
实验仪器:
384孔-细胞板(Corning;3712);384孔-化合物板(Corning;3657);
384孔-试验板(LABCYTE;P-05525);FLIPR(Molecular Devices)。
实验试剂:
DMEM(Gibco;11965);FBS(Gibco;10099-141);
潮霉素B(Invitrogen,10687010);Matrix(Thermo;5416);
DMSO(Sigma;D2650);HBSS(Invitrogen;14025);
HEPES(Invitrogen;15630080);Probenecid(Sigma;P8761);
Versene(Gibco;15040066);G418(Sigma;G5013);
Figure PCTCN2022097828-appb-000092
Calcium 4 Assay Kit(Molecular Devices;R8141);
α,β-meATP(Sigma;M6517);ATP hydrolytic enzyme(Sigma;A7646);
稳转细胞株:1321N1-hP2X2/3(由上海睿智化学研究有限公司提供)。
实验方法:
1.试剂配制:
Assay buffer:1*HBSS+20mM HEPES;
细胞培养基:DMEM+10%FBS+75μg/mL潮霉素B+150μg/mL G418;
铺板培养基:DMEM+10%DPBS;
0.5*Dye:10*Dye stock+1.25 Probenecid+1*assay buffer+0.5U/mL ATP hydrolytic enzyme;
2.细胞株培养于细胞培养基中,37℃,5%CO 2至70%~90%融合度,弃培养基,取出细胞,加2mL Versene,置37℃培养箱2-5min,加10mL铺板培养基收集细胞,细胞计数,每孔加50μL(1×10 4个细胞/well的密度)播种到384孔-试验板孵育16-24小时(至少过夜)。
3.丢弃培养液,加入30μL 1X染料,37℃避光孵育60min。
4.用DMSO将化合物粉末溶解成20mM储备液,准备检测需要浓度的180X化合物并梯度稀释10个浓度点。
5.准备化合物板:用ECHO转移500nL 180X化合物到化合物板(FLIPR用的source板)中,每孔加入30μL assay buffer,轻微振摇20-40分钟。
6.上机检测:每孔取15μL 3X化合物加入细胞板,FLIPR仪器加样,检测钙信号,15分钟后,每孔加22.5μL 3X激动剂(EC 80浓度),检测钙信号。
实验数据处理方法:
通过FLIPR读取钙信号值。实验中每个采样时间点的计算后的输出结果是340/510nm与380/510nm波长信号的比值。最大值减去最小值的计算来源自比值信号曲线。使用GraphPad prism拟合百分比抑制率和十点浓度数据至参数非线性逻辑公式计算出化合物的IC 50值。
实验结果:本发明实施例化合物对1321N1-hP2X2/3受体细胞功能钙流的试验,结果如表11所示:
表11
Figure PCTCN2022097828-appb-000093
实验结论:以上数据显示,本发明所示的化合物在1321N1-h2X2/3受体细胞功能钙流试验中显示出较小的抑制作用。
测试例3、Balb/C小鼠药代动力学测定
1.研究目的:以Balb/C小鼠为受试动物,研究以下化合物实施例,在5mg/kg剂量下口服给药在小鼠体内血浆的药代动力学行为。
2.试验方案
2.1试验药品:本发明实施例,自制。
2.2试验动物:Balb/C Mouse 6只/实施例,雄性,上海杰思捷实验动物有限公司,动物生产许可证号(SCXK(沪)2013-0006 N0.311620400001794)。
2.3药物配制:称取5g羟乙基纤维素(HEC,CMC-Na,粘度:800-1200Cps),溶于1000mL纯净水,加入10g Tween80。混合均匀成澄清溶液。
2.4给药:Balb/C小鼠,雄性;禁食一夜后分别p.o.,剂量为5mg/kg,给药体积10mL/kg。
2.5样品采集:小鼠给药前和给药后,在0、0.5、1、2、4、6、8和24小时,采用眼眶采血0.04mL,置于EDTA-K 2试管中,4℃ 6000rpm离心6min分离血浆,于-80℃保存。
2.6样品处理:
1)血浆样品20uL加入160uL乙腈沉淀,混合后3500×g离心5~20分钟。
2)取处理后上清溶液100uL进行LC/MS/MS分析待测化合物的浓度。
2.7液相分析
●液相条件:Shimadzu LC-20AD泵
●质谱条件:AB Sciex API 4000质谱仪
●色谱柱:phenomenex Gemiu 5um C18 50×4.6mm
●移动相:A液为0.1%甲酸水溶液,B液为乙腈
●流速:0.8mL/min
●洗脱时间:0-4.0分钟,洗脱液如下:
表12
Figure PCTCN2022097828-appb-000094
3.试验结果与分析
药代动力学主要参数用WinNonlin 8.2计算得到,小鼠药代实验结果见下表13:
表13 小鼠药代实验结果
Figure PCTCN2022097828-appb-000095
注:0.5%CMC-Na(1%吐温80)
4.实验结论:从表中小鼠药代实验结果可以看出,本发明实施例化合物表现出良好的代谢性质,暴露量AUC和最大血药浓度C max都表现良好。
测试例4、大鼠药代动力学测定
1.研究目的:以SD大鼠为受试动物,研究以下化合物实施例,在5mg/kg剂量下口服给药在大鼠体内血浆的药代动力学行为。
2.试验方案
2.1试验药品:本发明实施例,自制。
2.2试验动物:SD大鼠每组3只,雄性。上海杰思捷实验动物有限公司,动物生产许可证号(SCXK(沪)2013-0006 N0.311620400001794)。
2.3药物配制:称取5g羟乙基纤维素(HEC,CMC-Na,粘度:800-1200Cps),溶于1000mL纯净水,加入10g Tween80。混合均匀成澄清溶液。
2.4给药:SD大鼠每组3只,雄性,禁食一夜后分别PO,剂量为5mg/kg,给药体积10mL/kg。
2.5样品采集:大鼠给药前和给药后,在0、0.5、1、2、4、6、8和24小时,采用颈静脉采血0.2mL,置于EDTA-K 2试管中,4℃ 6000rpm离心6min分离血浆,于-80℃保存。
2.6样品处理:
1)血浆样品40uL加入160uL乙腈沉淀,混合后3500×g离心5~20分钟。
2)取处理后上清溶液100uL进行LC/MS/MS分析待测化合物的浓度。
2.7液相分析
●液相条件:Shimadzu LC-20AD泵
●质谱条件:AB Sciex API 4000质谱仪
●色谱柱:phenomenex Gemiu 5um C18 50×4.6mm
●移动相:A液为0.1%甲酸水溶液,B液为乙腈
●流速:0.8mL/min
●洗脱时间:0-4.0分钟,洗脱液如下:
表14
Figure PCTCN2022097828-appb-000096
3.试验结果与分析
药代动力学主要参数用WinNonlin 8.2计算得到,大鼠药代实验结果见下表15:
表15 大鼠药代实验结果
Figure PCTCN2022097828-appb-000097
注:0.5%CMC-Na(1%吐温80)
4.实验结论:从表中大鼠药代实验结果可以看出,5mg/kg剂量下,本发明实施例化合物表现出良好的代谢性质,暴露量AUC和最大血药浓度C max都表现良好。
测试例5、肝微粒体代谢稳定性试验
1.实验目的:本实验的目的是检测实施例化合物在小鼠、大鼠、犬和人肝微粒体中的稳定性情况。
2.实验步骤:
2.1配制化合物工作液:化合物的工作液配制:将化合物储备溶液加入磷酸缓冲液,终浓度为20μM。
2.2配制肝微粒体工作液:用100mM磷酸缓冲液稀释至终浓度为0.625mg/mL。
2.3准备NADPH和UDPGA:称取NADPH(还原型烟酰胺腺嘌呤二核苷酸磷酸)和UDPGA(尿苷二磷酸葡萄糖醛酸),加入100mM磷酸缓冲液,终浓度均为20mM。
2.4准备打孔剂:称取1mg Alamethicin(丙甲菌素)加入200μL DMSO,配制 成5mg/mL的溶液。再用磷酸缓冲液稀释至终浓度为50μg/mL。
2.5配制反应终止液,终止液:含有100ng/mL的盐酸拉贝洛尔和400ng/mL甲苯磺丁脲为内标的冷乙腈。
2.6孵育流程:在96孔板中依次加入400μL配制好的肝微粒体、25μL化合物工作液和25μL Alamethicin,于37℃预孵育10min。随后加入50μL配制好的NADPH/UDPGA启动反应,37℃孵育,反应体系的总体积为500μL,各成分最终含量如下:
表16
成分 含量
肝微粒体 0.5mg/mL
化合物 1μM
NADPH 2mM
UDPGA 2mM
Alamethicin 2.5μg/mL
2.7样品分析
2.7.1色谱条件:
仪器:岛津LC-30AD;色谱柱:
Figure PCTCN2022097828-appb-000098
C18(50*4.6mm,5μm粒径);
流动相:A:0.1%甲酸溶液,B:甲醇
冲洗梯度:0.2~1.6min 5%A到95%A,3.0~3.1min 95%A到5%A
运行时间:4.0min。
2.7.2质谱条件:
仪器:API5500型液相色谱质谱联用仪,AB Sciex公司;
离子源:电喷雾离子化源(ESI);干燥气体:N 2,温度500℃;
电喷雾电压:5000V;检测方式:正离子检测;
扫描方式:反应监测(MRM)方式。
3.实验结果:
表17 实施例化合物肝微粒体代谢稳定性结果
Figure PCTCN2022097828-appb-000099
4.实验结论:以上数据显示,本发明实施例化合物在小鼠、大鼠、犬和人 的肝微粒体中代谢稳定性良好。
测试例6、血浆蛋白结合率实验
1.实验目的:本实验方法的目的是检测实施例化合物在血浆中的血浆蛋白结合情况。
2.实验仪器及材料:液相质谱联用仪、离心机、涡旋仪、移液枪、连续加液器、96孔板、组织匀浆机(组织样品分析时使用)、50%的甲醇水溶液,加入内标的乙腈溶液、空白基质(血浆、尿液或组织匀浆液等)
3.实验步骤:
3.1待测物储备液的配制A:用DMSO将实施例化合物配制成1mM溶液A;
3.2血浆溶液的配制B:取溶液A加入到血浆溶液中,配制成5uM溶液B;
3.3处理流程
1)在膜内加入200uL溶液B;2)在膜外加入350uLPBS;
3)在37℃水浴锅内孵育6h;4)样品进行处理稀释并进质谱检测。
4.色谱条件:
仪器:岛津LC-20AD;色谱柱:Phenomenex
Figure PCTCN2022097828-appb-000100
C18(50*4.6mm,5μm粒径);
流动相:A:乙腈,B:0.1%甲酸溶液0~0.5min:5%A→90%A,2.0~2.1min:90%A→5%A;流速:0.8mL/min;运行时间:5.0min;进样体积:5μL。5.质谱条件:
仪器:API4000型液相色谱质谱联用仪,美国AB公司;
离子源为电喷雾离子化源(ESI);干燥气体(N 2)温度500℃;
电喷雾电压为5500V;检测方式为正离子检测;
扫描方式为选择反应监测(MRM)方式;扫描时间为0.1s。
6.实验结果:
表18:实施例化合物血浆蛋白结合率结果
Figure PCTCN2022097828-appb-000101
7.实验结论:以上数据显示,本发明实施例化合物显示出高血浆蛋白结合率,种属差异小。
测试例7、CYP酶单点抑制试验
1.实验目的:采用人肝微粒体孵育体系,利用单点法快速预测化合物对CYP450酶亚型的抑制情况。
2.实验步骤
2.1溶液配制:2.5mM NADPH,称重4.165mg NADPH(还原型烟酰胺腺嘌呤二核苷酸磷酸)加100mM磷酸缓冲液至2mL。0.25mg/mL微粒体,50μL 20mg/mL微粒体,加4mL 100mM磷酸缓冲液,混匀。
待测化合物反应液的配制:称取待测实施例化合物,用DMSO稀释至10mM,再用100mM磷酸缓冲液稀释至100μM。
2.2实验流程:
1.在96孔板中,加入40μL肝微粒体、10μL底物、10μL待测化合物,预孵育3min。
2.加入NADPH 40μL。
3.在20min时加入300μL含有内标的乙腈终止液。
4.离心进样。
3.实验结果:
表19 实施例化合物CYP酶单点抑制结果
Figure PCTCN2022097828-appb-000102
注:强抑制:IC 50<1μM;中等抑制:1μM<IC 50<10μM;弱抑制:IC 50>10μM4.实验结论:以上数据显示,本发明实施例化合物对各CYP酶亚型没有强抑制,DDI风险小。
测试例8、hERG钾离子通道抑制活性测试
1.细胞准备
7.1.1 CHO-hERG细胞培养于175cm 2培养瓶中,待细胞密度生长到60~80%,移走培养液,用7mL PBS洗一遍,然后加入3mL Detachin消化。
7.1.2待消化完全后加入7mL培养液中和,然后离心,吸走上清液,再加入5mL培养液重悬,以确保细胞密度为2~5×10 6/mL。
2.溶液配制
表20 细胞内液和外液的组成成分
Figure PCTCN2022097828-appb-000103
3.电生理记录过程
单细胞高阻抗封接和全细胞模式形成过程全部由Qpatch仪器自动完成,在获得全细胞记录模式后,细胞钳制在-80毫伏,在给予一个5秒的+40毫伏去极化刺激前,先给予一个50毫秒的-50毫伏前置电压,然后复极化到-50毫伏维持5秒,再回到-80毫伏。每15秒施加此电压刺激,记录2分钟后给予细胞外液记录5分钟,然后开始给药过程,化合物浓度从最低测试浓度开始,每个测试浓度给予2.5分钟,每个浓度至少测试3个细胞(n≥3)。
4.化合物准备
4.1将20mM的化合物母液用细胞外液进行稀释,取5μL 20mM的化合物母液加入2495μL细胞外液,500倍稀释至40μM,然后在含0.2%DMSO的细胞外液中依次进行3倍连续稀释得到需要测试的最终浓度。
4.2最高测试浓度为40μM,依次分别为40,13.33,4.44,1.48,0.49,0.16μM共6个浓度。
4.3最终测试浓度中的DMSO含量不超过0.2%,此浓度的DMSO对hERG钾通道没有影响。
5.数据分析:实验数据由XLFit软件进行分析。
6.质量控制
环境:湿度20~50%,温度22~25℃
试剂:所用实验试剂购买于Sigma公司,纯度>98%
报告中的实验数据必须满足以下标准:
全细胞封接阻抗>100MΩ
尾电流幅度>400pA
药理学参数:多浓度Cisapride对hERG通道的抑制效应设为阳性对照。
7.实验结果:
表21:实施例化合物在多浓度对hERG电流的抑制结果hERG结果
Figure PCTCN2022097828-appb-000104
Figure PCTCN2022097828-appb-000105
8.实验结论:
药物对于心脏hERG钾离子通道的抑制是药物导致QT延长综合症的主要原因。从实验结果可以看出,本发明实施例化合物对于心脏hERG钾离子通道没有明显抑制作用,可以避免高剂量时的心脏毒副作用。
测试例9、BALB/c小鼠味觉敏感性试验
1.实验目的:本实验通过奎宁苦水实验筛选出对动物味觉毒副作用较小的化合物。
2.实验主要仪器和材料
2.1仪器:
1、超净工作台(CJ-2F,苏州冯氏实验动物设备有限公司);
2、电子天平(CPA2202D,赛多利斯);
3、电子天平(BSA2202S-CW,赛多利斯);
4、纯水仪(Pacific TII,Thermo)。
2.2试剂:奎宁单盐酸盐二水合物(6119-47-7,Adamas)。
2.3动物:BALB/c小鼠,6-8周,♂,购自上海西普尔-必凯实验动物有限公司。
3.实验步骤:
3.1动物筛选:实验前1天,所有BALB/c小鼠称重,剔除体重过高或过低的动物。
3.2分组及禁水:根据体重将BALB/c小鼠按照要求进行随机分组,并于给药前12-16小时禁水、不禁食。
3.3奎宁水溶液的制备:称取适量的奎宁单盐酸盐二水合物,用超纯水配置成浓度为3mmol/L的奎宁盐酸水溶液,待用。
3.4待测化合物配制:称取适量的待测化合物,用对应的溶媒根据实验设计配制成目标浓度,待用。
3.5给药及动物奎宁饮水量测试:给药及禁食:实验当天,动物称重、禁食、更换垫料,按实验设计进行给药。
奎宁饮水量测试:
1、分别用超纯水和配制好的3mmol/L的奎宁盐酸水溶液润洗对应的干净的小鼠饮水瓶2-3次,之后装满称重并记录重量为Wi 0
2、按照实验设计,给药一定时长后,将装满的水瓶轻柔地放置于对应的小鼠笼架上,开始计时,30min后,轻柔地取出水瓶,称重并记录重量为Wi 30
3、每组动物饮水量计算:ΔWW(g)=Wi 30-Wi 0;单只小鼠饮水量计算:ΔpWW(g)=ΔWW/N,N为各组动物只数。
4、味觉障碍比率=(饮用水为奎宁盐酸水溶液同时给与受试药组ΔpWW-饮用水为奎宁盐酸水溶液同时给与溶剂对照组ΔpWW)/(饮用水为超纯水同时给与溶剂对照组ΔpWW-饮用水为奎宁盐酸水溶液同时给与溶剂对照组ΔpWW)×100%。用Excel等软件进行数据处理。
5、实验结束后安乐死动物。
4.试验结果:
表22 实施例化合物味觉结果
Figure PCTCN2022097828-appb-000106
5.实验结论:
从上述结果中可以看出,本专利的化合物对小鼠味觉毒副作用较小。
测试例10、对柠檬酸诱导的豚鼠急性咳嗽的药效研究
1.实验目的:本试验目的是评价化合物在柠檬酸诱导的豚鼠急性咳嗽模型中的药效。
2.实验仪器与试剂
2.1关键仪器
表23
Figure PCTCN2022097828-appb-000107
Figure PCTCN2022097828-appb-000108
2.2关键试剂
表24
试剂名称 厂家 货号
羧甲基纤维素钠 Sigma C5678
吐温80 Sigma P4780
ATP Sigma A2383
柠檬酸 Sigma C2404
3.实验操作及数据处理:
3.1动物:豚鼠(Hartley Guinea Pig),雄性,购自北京维通利华实验动物技术有限公司。
3.2实验流程:动物经适应性饲养,体重达标(300-400g)后,进行流水编号并根据体重进行随机分组。
引咳方法:先将豚鼠放入全身体积扫描箱适应3-5min后,进行2min ATP雾化,间隔3min,再给予5min的柠檬酸雾化。从柠檬酸雾化开始,记录10min内动物的咳嗽次数和咳嗽潜伏期。
3.3给药方案及咳嗽指标监测
受试化合物在柠檬酸雾化前2小时进行豚鼠单次灌胃给药处理,在预定时间放入DSI Buxco全身体积描记检测系统(WBP)的呼吸体积描记腔中进行柠檬酸雾化引咳。从柠檬酸雾化开始,WBP系统记录10分钟内豚鼠的咳嗽总次数(CCnt)和咳嗽潜伏期(CIP)。
3.4数据处理
所有的数据被录入到Excel文档中,并以平均值±标准误的方式表示。实验数据统计采用单因素方差分析方法(one-way ANOVA)将各组数据进行分析比较。统计分析结果p<0.05认为有显著差异。两两比较采用t-test方法比较差异性。
结果显示,本发明实施例化合物在柠檬酸诱导的豚鼠急性咳嗽模型中能有效改善咳嗽症状,咳嗽总次数下降率达59%以上。
三、化合物的盐及其晶型研究
本领域普通技术人员所熟知的是,上述实施例化合物被证明在1321N1-hP2X3受体细胞功能钙流试验中显示出良好的抑制作用时,其药学上可接受的盐往往会具有同样的药理药效活性。在此基础上,发明人进一步研究了相应化合物的盐型及晶型理化性质,但下述具体盐型或晶型的制备与表征并不代表 对本发明保护范围的限定,本领域普通技术人员可以以本发明为基础,获得本发明化合物更多的盐型和晶体,这些盐型和晶体均为本发明所保护的方案。具体如下:
1.实验仪器
1.1物理化学检测仪器的一些参数
表25
Figure PCTCN2022097828-appb-000109
1.2仪器与液相分析条件
1.2.1仪器与设备
表26
仪器名称 型号
分析天平 METTLER TOLEDO XA105
纯水机 Milli-Q Plus,Millipore
高效液相色谱仪 Agilent1260
Agilent G1311B
进样器 G1329B
柱温箱 G1316A
检测器 G1315D
1.2.2色谱条件
色谱柱:Zorbax BONUS RP(3.5μm,4.6*75mm)
流速:1.0mL/min
柱温:30℃
检测波长:262nm
进样体积:5.0μL
运行时间:15min
稀释剂:DMSO
流动相:A:水(0.05%三氟乙酸);B:乙腈(0.05%三氟乙酸)
表27
T(min) A(%) B(%)
0.00 60 40
12.00 25 75
12.01 60 40
15.00 60 40
2.化合物盐型的研究
2.1化合物的盐型筛选
2.1.1实验目的:筛选化合物的盐型。
2.1.2实验步骤:
1)仪器和设备
表28
名称 型号 来源
分析天平 XA105 METTLER TOLEDO
超声波清洗仪 SK5200LHC 上海科导超声仪器
移液枪 Eppendorf(50mL,100μL) Eppendorf
2)操作程序
溶析或悬浮反应成盐:称取10mg化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺,加溶剂200μL~400μL,40~50℃加热搅拌,分别加入不同的酸,搅拌过夜,降至室温,过滤,干燥,得到化合物的盐。
表29
Figure PCTCN2022097828-appb-000110
Figure PCTCN2022097828-appb-000111
2.1.3实验结果:通过盐型筛选实验的结果可知,化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的游离碱能够与盐酸、氢溴酸、硫酸、甲磺酸、乙烷磺酸成盐。
如上所述,本领域技术人员在本发明的基础上,可以获得更多的可药用盐。
2.2化合物盐型的定量分析
2.2.1化合物盐DAD定量
2.2.1.1实验目的:确定化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的盐中酸的个数。
2.2.1.2实验步骤:
1)色谱条件
表30
仪器 Thermo Ultimate 3000
稀释剂 DMSO
色谱柱 Waters x-bridge(150*4.6mm,3.5um)
流动相 A:25mM Phosphate buffer(NH 4H 2PO 4,pH2.0),B:MeOH
进样体积 5μL
流速 1.0ml/min
柱温箱稳定 35℃
运行时间 12min
梯度洗脱时间(min) B相(体积百分数)
0 50
10 80
10.01 50
12 50
2)操作:称取游离碱适量,加DMSO配制成浓度为0.006-0.68mg/mL范围的系 列线性溶液,作为外标溶液STD。
分别称取硫酸盐、甲烷磺酸盐以及乙烷磺酸盐化合物适量,加DMSO配制成含化合物盐浓度为0.3-0.5mg/mL的样品溶液。分别取上述线性溶液及样品溶液进样。
2.2.1.3实验结果:
表31
Figure PCTCN2022097828-appb-000112
DAD定量结果表明,2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的硫酸盐、甲磺酸酸盐和乙烷磺酸盐为1:1成盐。
2.2.2甲磺酸盐ELSD定量
2.2.2.1实验目的:确定化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺甲磺酸盐中甲磺酸的个数。
2.2.2.2实验步骤:
1)色谱条件
表32
仪器 Agilent 1200
稀释剂 DMSO
色谱柱 ZIC-HILIC(150*4.6mm,5μm)
流动相 A:75mM醋酸铵溶液(pH4.80)、B:乙腈(A:B=30:70)
进样体积 5μL
流速 1.0ml/min
ELSD温度 80℃
运行时间(min) 10
2)操作:称取甲烷磺酸适量,加DMSO配制成含甲烷磺酸浓度0.15-0.5mg/mL 范围的系列线性溶液。
称取甲烷磺酸盐适量,加DMSO配制成含化合物甲烷磺酸盐浓度为2.8mg/mL的样品溶液。分别取上述线性溶液及样品溶液进样。
2.2.2.3实验结果:
表33
Figure PCTCN2022097828-appb-000113
经计算化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的甲烷磺酸盐中甲烷磺酸的个数为1。
2.3甲磺酸盐的大鼠药代动力学测定
2.3.1研究目的:以SD大鼠为受试动物,研究2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的不同盐型,在10mg/kg剂量下口服给药在大鼠体内血浆的药代动力学行为。
2.3.2试验方案
2.3.2.1试验药品:本发明实施例,自制。
2.3.2.2试验动物:SD大鼠每组3只,雄性。动物生产许可证号(SCXK(沪)2013-0006 N0.311620400001794)。
2.3.2.3药物配制:分别将甲磺酸盐晶型A、乙烷磺酸盐晶型A、硫酸盐晶型B和氢溴酸盐晶型B用0.5%HPMC水溶液细胞粉碎和超声混悬均匀。
2.3.2.4给药:SD大鼠每组3只,雄性,禁食一夜后分别PO,剂量为10mg/kg,给药体积10mL/kg。
2.3.2.5样品采集:大鼠给药前和给药后,在0、0.5、1、2、4、6、8和24小时,采用颈静脉采血0.2mL,置于EDTA-K 2试管中,4℃ 6000rpm离心6min分离血浆,于-80℃保存。
2.3.2.6样品处理:
1)血浆样品40uL加入160uL乙腈沉淀,混合后3500×g离心5~20分钟。
2)取处理后上清溶液100uL进行LC/MS/MS分析待测化合物的浓度。
2.3.2.7液相分析
●液相条件:Shimadzu LC-20AD泵
●质谱条件:AB Sciex API 4000质谱仪
●色谱柱:phenomenex Gemiu 5um C18 50×4.6mm
●移动相:A液为0.1%甲酸水溶液,B液为乙腈
●流速:0.8mL/min
●洗脱时间:0-4.0分钟,洗脱液如下:
表34
Figure PCTCN2022097828-appb-000114
2.3.3.试验结果与分析
药代动力学主要参数用WinNonlin 8.2计算得到,大鼠药代实验结果见下表:
表35
Figure PCTCN2022097828-appb-000115
注:溶媒:0.5%HPMC
2.3.4.实验结论:
从表中大鼠药代实验结果可以看出,10mg/kg剂量下,本发明2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的甲磺酸盐(晶型A)、乙烷磺酸盐(晶型A)、硫酸盐(晶型B)和氢溴酸盐(晶型B)的暴露量AUC和最大血药浓度C max较游离碱均表现更好。
3.化合物盐的晶型研究
3.1 2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的盐的晶型研究
3.1.1实验目的:筛选化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡 啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺成晶型的盐。
3.1.2实验步骤:
1)仪器与设备
表36
Figure PCTCN2022097828-appb-000116
2)操作程序:溶析或悬浮反应成盐:称取10mg化合物,加溶剂200μL~400μL,40~50℃加热搅拌,分别加入不同的酸,搅拌过夜,降至室温,过滤,干燥,得到化合物的盐。
表37
Figure PCTCN2022097828-appb-000117
3.1.3实验结果:经过该化合物盐的晶型研究实验,得到有晶型的盐型为盐酸盐、氢溴酸盐、硫酸盐、甲磺酸盐、乙烷磺酸盐。
如上所述,本领域技术人员在本发明的基础上,运用常规的方法可以获得更多的可成晶型的可药用盐。
3.2化合物盐的晶型制备
3.2.1实验目的:制备化合物盐的晶型。
3.2.2实验步骤:
1)仪器和设备
表38
Figure PCTCN2022097828-appb-000118
2)操作程序
(1)甲磺酸盐晶型A的制备
称取150mg化合物游离碱,加入8mL二氯甲烷,40℃加热搅拌,固体溶不清,加入380uL(1.1e.q.)甲磺酸的乙醇溶液(1mol/L in EtOH),悬浮反应结晶,40℃保持过夜,降至室温后,快速离心,去除上清液,固体在40℃条件下真空干燥至恒重,得到甲磺酸盐晶型A,经检测分析,其具有如图1所示的XRPD图、如图2所示的DSC图以及如图3所示的TGA图。
或可按以下步骤制备:
将化合物游离碱(15.0g)加入反应瓶中,加入2-丁酮(600ml),升温到45~55℃搅拌溶清,趁热过滤,向滤液中滴加甲磺酸乙醇溶液(3.68g甲磺酸溶于35ml乙醇),滴加完毕保温搅拌过夜,降温至20~30℃,搅拌析晶30分钟,过滤,滤饼50℃真空干燥,得甲磺酸盐晶型A 16.2g。
(2)乙烷磺酸盐晶型A的制备
称取150mg化合物游离碱,加入8mL二氯甲烷,40℃加热搅拌,固体溶不清,加入380uL(1.1e.q.)乙烷磺酸的乙醇溶液(1mol/L in EtOH),悬浮反应结晶,体系逐渐变得粘稠,流动相差,补加3mL溶剂,40℃保持过夜,降至室温后,快速离心,去除上清液,固体在40℃条件下真空干燥至恒重,得到乙烷磺酸盐晶型A,经检测分析,其具有如图4所示的XRPD图、如图5所示的DSC图以及如图6所示的TGA图。
(3)硫酸盐晶型A的制备
称取30mg化合物游离碱,加入1mL四氢呋喃,50℃加热搅拌,固体溶解, 加入77uL(1.1e.q.)硫酸的乙醇溶液(1mol/L in EtOH),加酸后5min内析出沉淀,且体系逐渐粘稠,流动性变差,50℃保持过夜,降至室温后,然后快速离心,去除上清液,固体在40℃条件下真空干燥至恒重,经检测分析,其具有如图7所示的XRPD图和如图8所示的DSC图。
(4)硫酸盐晶型B的制备
称取150.54mg化合物游离碱,加入8mL二氯甲烷,40℃加热搅拌,固体溶不清,加入380uL(1.1e.q.)硫酸的甲醇溶液(1mol/L in MeOH),悬浮反应结晶,40℃保持过夜,降至室温后,快速离心,去除上清液,固体在40℃条件下真空干燥至恒重,得到硫酸盐晶型B。经检测分析,其有如图9所示的XRPD图、如图10所示的DSC图以及如图11所示的TGA图。
(5)盐酸盐晶型A的制备
称取150.76mg化合物游离碱,加入8mL二氯甲烷,40℃加热搅拌,固体溶不清,加入380uL(1.1e.q.)盐酸的乙醇溶液(1mol/L in EtOH),悬浮反应结晶,40℃保持过夜,降至室温后,快速离心,去除上清液,固体在40℃条件下真空干燥至恒重,得到盐酸盐晶型A。经检测分析,其有如下如图12所示的XRPD图、如图13所示的DSC图以及如图14所示的TGA图。
(6)盐酸盐晶型B的制备
称取30mg化合物游离碱,加入1mL四氢呋喃,50℃加热搅拌,固体溶解,加入77uL(1.1e.q.)盐酸的乙醇溶液(1mol/L in EtOH),立刻析出沉淀,50℃保持过夜,降至室温后,然后快速离心,去除上清液,固体在40℃条件下真空干燥至恒重,得到盐酸盐晶型B。经检测分析,其有如图15所示的XRPD图。
(7)氢溴酸盐晶型A的制备
称取10mg化合物游离碱,加入400uL溶剂丙酮,40℃加热搅拌,基本溶清,加入1.1e.q.氢溴酸的乙醇溶液(1mol/L in EtOH),悬浮反应结晶,40℃保持搅拌过夜,然后降至室温,过滤,固体进一步在真空干燥箱中干燥过夜(50℃减压真空干燥)至恒重,得到氢溴酸盐晶型A。经检测分析,其有如下如图16所示的XRPD图、如图17所示的DSC图。
(8)氢溴酸盐晶型B的制备
称取150mg化合物游离碱,加入8mL二氯甲烷,40℃加热搅拌,固体溶不清,加入380uL(1.1e.q.)氢溴酸的甲醇溶液(1mol/L in MeOH),悬浮反应结晶,40℃保持过夜,降至室温后,快速离心,去除上清液,固体在40℃条件下真空干燥至恒重,得到氢溴酸盐晶型B。经检测分析,其有如下如图18所示的XRPD图、如图19所示的DSC图以及如图20所示的TGA图。
(9)氢溴酸盐晶型C的制备
称取30mg化合物游离碱,加入1mL四氢呋喃,50℃加热搅拌,固体溶解, 加入77uL(1.1e.q.)氢溴酸的乙醇溶液(1mol/L in EtOH),立刻析出沉淀,50℃保持过夜,降至室温后,然后快速离心,去除上清液,固体在40℃条件下真空干燥至恒重,得到氢溴酸盐晶型C。经检测分析,其有如下如图21所示的XRPD图。
(10)化合物游离碱晶型I的制备
称取化合物游离碱6g,用DCM/丙酮=3:1(250ml)溶解,加入活性炭(0.5g)以及无水硫酸钠(15g),40℃搅拌30分钟。过滤,滤液减压浓缩至干,用50ml丙酮带一次,蒸干。加入100ml丙酮,加热到65℃回流,仍未溶清,滴加200ml正庚烷,加热搅拌1小时,关闭加热,自然冷却到室温,继续搅拌1小时。过滤,得类白色固体,真空干燥至恒重,得到游离碱晶型I,经检测分析,其有如图22所示的XRPD图、如图23所示的DSC图。
(11)游离碱晶型II的制备
称量10mg的化合物游离碱(晶型I)到2mL的玻璃瓶中,加入200μL的乙醇溶剂,得到混悬液;将混悬液置于磁力搅拌器上40℃打浆3天,离心,去除上清液,固体进一步在真空干燥箱中干燥过夜(50℃减压真空干燥),得到晶型II,经检测分析,其有如图24所示的XRPD图、如图25所示的DSC图。
4.固体稳定性实验
4.1化合物甲磺酸盐晶型A的固定稳定性实验
4.1.1实验目的:考察化合物晶型在高温、高湿、高温高湿条件下,化合物的物理化学稳定性,为晶型筛选与化合物晶型贮存提供依据。
4.1.2仪器和液相分析条件
表39
Figure PCTCN2022097828-appb-000119
4.1.3实验方案:称取甲磺酸盐晶型A、乙磺酸盐晶型A和硫酸盐晶型B适量, 分别在高温(60℃)、高湿(92.5%RH)、高温高湿(50℃&75%RH)条件下放置7天和14天后,用HPLC,外标法测定盐的含量,并采用色谱峰面积归一化法计算有关物质的变化。
4.1.4实验结果:
表40
Figure PCTCN2022097828-appb-000120
化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的甲磺酸盐晶型A在高温、高湿、高温高湿的条件下,比较稳定;乙磺酸盐晶型A在高温、高湿的条件下,比较稳定;硫酸盐晶型B在高湿的条件下,比较稳定。
5.不同介质中溶解度实验
5.1化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺在不同介质中溶解度实验
5.1.1实验目的:考察甲磺酸盐晶型A和化合物游离碱在不同pH值介质、水、人工模拟胃液(FaSSGF)、禁食人工模拟肠液(FaSSIF)及非禁食人工模拟肠液(FeSSIF)等媒介中溶解度大小,为盐可成药性评估提供依据。
5.1.2实验方案:分别称量1~2mg的化合物游离碱晶型II及不同的盐型到1.5mL的液相小瓶中,然后分别加入1mL的不同pH缓冲液、人工模拟胃液(FaSSGF)、禁食人工模拟肠液(FaSSIF)、非禁食人工模拟肠液(FeSSIF)和纯水,置于旋转混匀仪上过夜,温度为室温,24小时后将样品溶液用0.45μm混合水纤滤膜过滤后取续滤液,用HPLC测试其含量。
表41
Figure PCTCN2022097828-appb-000121
Figure PCTCN2022097828-appb-000122
5.1.3实验结果:如图所示:
表42
Figure PCTCN2022097828-appb-000123
化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的甲磺酸盐晶型相比游离碱晶型II,在相应介质中的溶解度提高四倍以上,表明成盐后可显著提高溶解度。
6.热力学稳定实验
6.1化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的甲磺酸盐的多晶型筛选
6.1.1实验目的:通过多晶筛选,获得热力学稳定的甲磺酸盐的晶型。
6.1.2实验方案:取3~5mg甲磺酸盐晶型A,加入100ul相应溶剂,置于35℃金属浴磁力搅拌打浆24h后,离心,固体干燥后测其XRPD。
6.1.3实验结果:如下表:
表43
Figure PCTCN2022097828-appb-000124
以上结果表明,2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的甲磺酸盐晶型A为甲磺酸盐的热力学稳定晶型。
7.引湿性实验
7.1化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的不同盐的晶型的引湿性实验
7.1.1实验目的:考察化合物2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的不同盐的晶型在不同相对湿度下的引湿性,为储存提供依据。
7.1.2实验方案:将化合物不同盐的晶型置于不同相对湿度的饱和水蒸气中,使化合物与水蒸气达到动态平衡,并计算平衡后化合物吸湿增重的百分数。
表44 动态水分吸附仪(DVS)仪器参数
仪器型号 SMS Intrinsic
实验温度 25℃
干燥时间 0%RH 120min
平衡dm/dt 0.02%/min(最小10min,最大180min)
RH(%)测量步长 10%
测量梯度 0-95-0%
循环次数 2
7.1.3实验结果
表45 甲磺酸盐晶型A的实验结果
Figure PCTCN2022097828-appb-000125
Figure PCTCN2022097828-appb-000126
表46 乙磺酸盐晶型A的实验结果
Figure PCTCN2022097828-appb-000127
Figure PCTCN2022097828-appb-000128
表47 硫酸盐晶型B的实验结果
Figure PCTCN2022097828-appb-000129
表48 盐酸盐晶型A的实验结果
Figure PCTCN2022097828-appb-000130
Figure PCTCN2022097828-appb-000131
表49 氢溴酸盐晶型B的实验结果
Figure PCTCN2022097828-appb-000132
Figure PCTCN2022097828-appb-000133
以上结果表明,2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的甲磺酸盐晶型A的引湿性最低,在80%相对湿度下引湿增重不超过0.5%,根据中国药典对引湿性(在温度为25℃±1℃,相对湿度为80%±2%下测定)特征描述与引湿性增重的界定,甲磺酸盐晶型A为略有引湿性。

Claims (20)

  1. 通式(I-a)所示化合物或其立体异构体的酸式盐,
    Figure PCTCN2022097828-appb-100001
    其中:
    R 1选自氢、氘、卤素、氨基、羟基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-12环烷基、-(CH 2) nC(O)R a、3-12元杂环基、C 6-14芳基或5-14元杂芳基,所述的氨基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-12环烷基、-(CH 2) nC(O)R a、3-12元杂环基、C 6-14芳基和5-14元杂芳基,任选地被氘、卤素、氨基、羟基、氰基、硝基、氧代基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6羟烷基、氰基取代的C 1-6烷基、C 3-12环烷基、3-12元杂环基、C 6-12芳基和5-12元杂芳基中的一个或多个取代基所取代;
    R a选自氢、氘、卤素、氨基、硝基、羟基、氰基、C 1-6烷基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基或5-10元杂芳基,所述的氨基、C 1-6烷基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基,任选地被氘、卤素、氨基、硝基、羟基、氰基、羧基、氧代基、硫代基、C 1-6烷基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基中的一个或多个取代基所取代;
    R 2选自氢、氘、卤素、氨基、羟基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-12环烷基、3-12元杂环基、C 6-14芳基或5-14元杂芳基,所述的氨基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-12环烷基、3-12元杂环基、C 6-14芳基和5-14元杂芳基,任选地被氘、卤素、氨基、羟基、氰基、硝基、氧代基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 1-6羟 烷基、氰基取代的C 1-6烷基、C 3-12环烷基、3-12元杂环基、C 6-12芳基和5-12元杂芳基中的一个或多个取代基所取代;
    R 3选自氢、氘、卤素、氨基、羟基、氰基、氧代基、硫代基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6羟烷基、C 1-6烷氧基、C 1-6卤代烷氧基、C 3-8环烷基、3-12元杂环基、C 6-14芳基或5-14元杂芳基;且
    x为0~3的整数,优选0、1或2,更优选0或1;
    n为0~3的整数,优选0、1或2,更优选0或1;
    酸式盐中的酸为无机酸或有机酸;优选地,无机酸选自盐酸、硫酸、硝酸、氢溴酸、氢氟酸、氢碘酸或磷酸;有机酸选自2,5-二羟基苯甲酸、1-羟基-2-萘甲酸、醋酸、乙烷磺酸、二氯醋酸、三氯醋酸、乙酰氧肟酸、己二酸、苯磺酸、4-氯苯磺酸、苯甲酸、4-乙酰氨基苯甲酸、4-氨基苯甲酸、癸酸、己酸、辛酸、肉桂酸、柠檬酸、环己烷氨基磺酸、樟脑磺酸、天门冬氨酸、樟脑酸、葡萄糖酸、葡糖醛酸、谷氨酸、异抗坏血酸、乳酸、苹果酸、扁桃酸、焦谷氨酸、酒石酸、十二烷基硫酸、二苯甲酰酒石酸、乙烷-1,2-二磺酸、乙烷磺酸、蚁酸、富马酸、半乳糖酸、龙胆酸、戊二酸、2-酮戊二酸、乙醇酸、马尿酸、羟乙基磺酸、乳糖酸、抗坏血酸、天冬氨酸、月桂酸、樟脑酸、马来酸、丙二酸、甲磺酸、1,5-萘二磺酸、萘-2-磺酸、烟酸、油酸、乳清酸、草酸、棕榈酸、双羟萘酸、丙酸、水杨酸、4-氨基水杨酸、癸二酸、硬脂酸、丁二酸、硫氰酸、十一碳烯酸、三氟乙酸、苯磺酸、对甲基苯磺酸或L-苹果酸;优选盐酸、硫酸、磷酸、乙烷磺酸、苯磺酸、甲磺酸、富马酸、羟乙基磺酸、草酸或氢溴酸。
  2. 根据权利要求1所述的酸式盐,其特征在于,通式(I-a)进一步如通式(II-a)所示:
    Figure PCTCN2022097828-appb-100002
  3. 根据权利要求1或2所述的酸式盐,其特征在于,
    R 1选自氢、氘、卤素、氨基、羟基、氰基、硝基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-8环烷基、-(CH 2) nC(O)R a、3-8元杂环基、C 6-10芳基或5-10元杂芳基,所述的 氨基、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6氘代烷基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-8环烷基、-(CH 2) nC(O)R a、3-8元杂环基、C 6-10芳基和5-10元杂芳基,任选地被氘、卤素、氨基、羟基、氰基、硝基、氧代基、C 1-3烷基、C 2-3烯基、C 2-3炔基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 1-3羟烷基、氰基取代的C 1-3烷基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基中的一个或多个取代基所取代;
    R a选自氢、氘、卤素、氨基、硝基、羟基、氰基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基或5-10元杂芳基,所述的氨基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基,任选地被氘、卤素、氨基、硝基、羟基、氰基、羧基、氧代基、硫代基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 2-6烯基、C 2-6炔基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基中的一个或多个取代基所取代;
    优选地,R 1选自氢、卤素、氨基、氰基、C 1-3烷基、C 2-6烯基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、C 3-8环烷基、-(CH 2) nC(O)R a、含1-3个选自氮、氧或硫原子的3-8元杂环基、C 6-10芳基或含1-3个选自氮、氧或硫原子5-10元杂芳基,所述的氨基、C 1-6烷基、C 2-6烯基、C 1-6卤代烷基、C 1-6烷氧基、卤代C 1-6烷氧基、C 1-6羟烷基、-(CH 2) nC(O)R a、C 3-8环烷基、含1-3个选自氮、氧或硫原子的3-8元杂环基、C 6-10芳基或含1-3个选自氮、氧或硫原子5-10元杂芳基,任选地被氘、卤素、氨基、羟基、氰基、硝基、氧代基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 1-3羟烷基、氰基取代的C 1-3烷基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基中的一个或多个取代基所取代;
    R a选自氢、氘、卤素、氨基、氰基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 3-6环烷基或含1-2个选自N或O的4-6元杂环基,所述的氨基C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 3-6环烷基或含1-2个选自N或O的4-6元杂环基,任选地被氘、卤素、氨基、羟基、氰基、硝基、氧代基、C 1-3烷基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 1-3羟烷基、氰基取代的C 1-3烷基、C 3-8环烷基、3-8元杂环基、C 6-10芳基和5-10元杂芳基中的一个或多个取代基所取代;
    更优选地,R 1选自以下基团:
    -H、-NH 2、-F、-Cl、-Br、-CN、-CH 3、-CH 2CH 3、-CF 3
    Figure PCTCN2022097828-appb-100003
    Figure PCTCN2022097828-appb-100004
    R 2选自氢、氘、卤素、氨基、羟基、氰基、硝基、C 1-3烷基、C 2-3烯基、C 2-3炔基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3烷氧基、卤代C 1-3烷氧基、C 1-3羟烷基、C 3-8环烷基、3-8元杂环基、C 6-10芳基或5-10元杂芳基,优选氢、氨基、氰基、氟、氯、溴、甲基、异丙基、三氟甲基、甲氧基、环丙基或吗啉基;
    R 3选自氢、氘、卤素、氨基、羟基、氰基、氧代基、硫代基、C 1-3烷基、C 2-3烯基、C 2-3炔基、C 1-3氘代烷基、C 1-3卤代烷基、C 1-3羟烷基、C 1-3烷氧基、C 1-3卤代烷氧基、C 3-8环烷基、3-8元杂环基、C 6-10芳基或5-1.元杂芳基,优选氢或氰基。
  4. 根据权利要求1-3任一项所述的酸式盐,其特征在于,化合物的具体结构如下:
    Figure PCTCN2022097828-appb-100005
    Figure PCTCN2022097828-appb-100006
    酸式盐中的酸选自羟乙基磺酸、盐酸、硫酸、1,5-萘二磺酸、甲磺酸、氢溴酸、乙烷磺酸、磷酸、苯磺酸、草酸、马来酸、己二酸、盐酸、柠檬酸、丙二酸、L-苹果酸、帕莫酸、对甲苯磺酸或富马酸;优选盐酸、硫酸、甲磺酸、氢溴酸或乙烷磺酸。
  5. 根据权利要求1-4任一项所述的酸式盐,其特征在于,酸式盐中酸的个数为0.2-3;优选0.2、0.5、1、1.5、2、2.5或3;更优选0.5、1、2或3,进一步优选1。
  6. 根据权利要求1-5任一项所述的酸式盐,其特征在于,酸式盐为水合物或无水物;当酸式盐为水合物时,水的个数为0.2-3;优选0.2、0.5、1、1.5、2、2.5或3;更优选0.5、1、2或3。
  7. 根据权利要求1-6任一项所述的酸式盐,其特征在于,所述酸式盐为晶型;
    优选地,晶型为化合物2-(2-(叔丁基)-5-氧吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
    2-(2-(叔丁基)-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
    2-(2-乙基-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
    2-(2-环丙基-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
    2-(2,5-二甲基吡啶-4-基)-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
    N-(5-氟吡啶-2-基)-2-(2-(1-甲基环丙基)-5-氧代-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)乙酰胺的酸式盐晶型;
    2-(2-溴-5-氧-8-(三氟甲基)吡唑[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
    2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
    N-(5-氟吡啶-2-基)-2-(5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)乙酰胺的酸式盐晶型;
    2-(3-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型;
    更优选地,酸式盐晶型为羟乙基磺酸盐晶型、硫酸盐晶型、盐酸盐晶型、1,5-萘二磺酸盐晶型、甲磺酸盐晶型、乙烷磺酸盐晶型、氢溴酸盐晶型、磷酸盐晶型、苯磺酸盐晶型、草酸盐晶型、马来酸盐晶型、己二酸盐晶型、盐酸盐晶型、柠檬酸盐晶型、丙二酸盐晶型、L-苹果酸盐晶型、帕莫酸盐晶型、对甲苯磺酸盐晶型或富马酸盐晶型。
  8. 根据权利要求7所述的酸式盐晶型,其特征在于,
    2-(2-氰基-5-氧代-8-(三氟甲基)吡唑并[1,5-a]吡啶[3,2-e]嘧啶-4(5H)-基)-N-(5-氟吡啶-2-基)乙酰胺的酸式盐晶型为:
    甲磺酸盐晶型A,其X-射线粉末衍射图谱在13.7±0.2°处具有衍射峰;或者在21.9±0.2°处具有衍射峰;或者在20.4±0.2°处具有衍射峰;或者在15.4±0.2°处 具有衍射峰;或者在19.6±0.2°处具有衍射峰;或者在16.4±0.2°处具有衍射峰;或者在9.3±0.2°处具有衍射峰;或者在5.3±0.2°处具有衍射峰;或者在7.9±0.2°处具有衍射峰;或者在11.9±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
    优选地,甲磺酸盐晶型A的X-射线粉末衍射图谱至少包含位于2θ为13.7±0.2°、16.4±0.2°、21.9±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为13.9±0.2°、20.4±0.2°、15.4±0.2°、5.3±0.2°、11.9±0.2°、9.3±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
    更优选地,甲磺酸盐晶型A的X-射线粉末衍射图谱任选还包含位于2θ为7.9±0.2°、19.6±0.2°、17.6±0.2°、18.8±0.2°、21.0±0.2°、23.3±0.2°、24.1±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
    进一步优选地,甲磺酸盐晶型A的X-射线粉末衍射图谱包含位于2θ为5.3±0.2°、7.9±0.2°、9.3±0.2°、11.9±0.2°、13.7±0.2°、13.9±0.2°、15.4±0.2°、16.4±0.2°、17.6±0.2°、18.8±0.2°、19.6±0.2°、20.4±0.2°、21.0±0.2°、21.9±0.2°、23.3±0.2°、24.1±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
    更进一步优选地,甲磺酸盐晶型A的X-射线粉末衍射图谱如图1所示;或其DSC图谱基本如图2所示,或其TGA图谱基本如图3所示;
    或,为乙烷磺酸盐晶型A,其X-射线粉末衍射图谱在15.0±0.2°处具有衍射峰;或者在21.1±0.2°处具有衍射峰;或者在23.1±0.2°处具有衍射峰;或者在19.8±0.2°处具有衍射峰;或者在12.5±0.2°处具有衍射峰;或者在9.0±0.2°处具有衍射峰;或者在12.3±0.2°处具有衍射峰;或者在24.6±0.2°处具有衍射峰;或者在10.3±0.2°处具有衍射峰;或者在6.1±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
    优选地,乙烷磺酸盐晶型A的X-射线粉末衍射图谱至少包含位于2θ为15.0±0.2°、21.1±0.2°、23.1±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为19.8±0.2°、12.5±0.2°、9.0±0.2°、12.3±0.2°、24.6±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
    更优选地,乙烷磺酸盐晶型A的X-射线粉末衍射图谱任选还包含位于2θ为10.3±0.2°、6.1±0.2°、16.1±0.2°、19.2±0.2°、23.6±0.2°、30.7±0.2°、9.6±0.2° 中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
    进一步优选地,乙烷磺酸盐晶型A的X-射线粉末衍射图谱包含位于2θ为15.0±0.2°、21.1±0.2°、23.1±0.2°、19.8±0.2°、12.5±0.2°、9.0±0.2°12.3±0.2°、24.6±0.2°、10.3±0.2°、6.1±0.2°、16.1±0.2°、19.2±0.2°、23.6±0.2°、30.7±0.2°、9.6±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
    更进一步优选地,乙烷磺酸盐晶型A的X-射线粉末衍射图谱如图4所示;或其DSC图谱基本如图5所示,或其TGA图谱基本如图6所示;
    或,为硫酸盐晶型A,其X-射线粉末衍射图谱在22.5±0.2°处具有衍射峰;或者在15.9±0.2°处具有衍射峰;或者在22.3±0.2°处具有衍射峰;或者在16.8±0.2°处具有衍射峰;或者在22.9±0.2°处具有衍射峰;或者在32.1±0.2°处具有衍射峰;或者在14.0±0.2°处具有衍射峰;或者在21.1±0.2°处具有衍射峰;或者在11.2±0.2°处具有衍射峰;或者在26.1±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
    优选地,硫酸盐晶型A的X-射线粉末衍射图谱至少包含位于2θ为22.5±0.2°、15.9±0.2°、22.3±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为16.8±0.2°、22.9±0.2°、32.1±0.2°、14.0±0.2°、21.1±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
    更优选地,硫酸盐晶型A的X-射线粉末衍射图谱任选还包含位于2θ为11.2±0.2°、26.1±0.2°、28.2±0.2°、37.8±0.2°、15.5±0.2°、26.5±0.2°、36.4±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
    进一步优选地,硫酸盐晶型A的X-射线粉末衍射图谱包含位于2θ为22.5±0.2°、15.9±0.2°、22.3±0.2°、16.8±0.2°、22.9±0.2°、32.1±0.2°、14.0±0.2°、21.1±0.2°、11.2±0.2°、26.1±0.2°、28.2±0.2°、37.8±0.2°、15.5±0.2°、26.5±0.2°、36.4±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
    更进一步优选地,硫酸盐晶型A的X-射线粉末衍射图谱如图7所示;或其DSC图谱基本如图8所示;
    或,为硫酸盐晶型B,其X-射线粉末衍射图谱在15.3±0.2°处具有衍射峰;或者在21.5±0.2°处具有衍射峰;或者在10.6±0.2°处具有衍射峰;或者在19.8±0.2°处具有衍射峰;或者在20.1±0.2°处具有衍射峰;或者在12.6±0.2°处具有衍射峰;或者在25.2±0.2°处具有衍射峰;或者在9.2±0.2°处具有衍射峰;或者在9.9±0.2° 处具有衍射峰;或者在23.4±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
    优选地,硫酸盐晶型B的X-射线粉末衍射图谱至少包含位于2θ为15.3±0.2°、21.5±0.2°、10.6±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为19.8±0.2°、20.1±0.2°、12.6±0.2°、25.2±0.2°、9.2±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
    更优选地,硫酸盐晶型B的X-射线粉末衍射图谱任选还包含位于2θ为9.9±0.2°、23.4±0.2°、6.3±0.2°、16.7±0.2°、23.9±0.2°、33.8±0.2°、16.3±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
    进一步优选地,硫酸盐晶型B的X-射线粉末衍射图谱包含位于2θ为15.3±0.2°、21.5±0.2°、10.6±0.2°、19.8±0.2°、20.1±0.2°、12.6±0.2°、25.2±0.2°、9.2±0.2°、9.9±0.2°、23.4±0.2°、6.3±0.2°、16.7±0.2°、23.9±0.2°、33.8±0.2°、16.3±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
    更进一步优选地,硫酸盐晶型B的X-射线粉末衍射图谱如图9所示;或其DSC图谱基本如图10所示,或其TGA图谱基本如图11所示;
    或,为盐酸盐晶型A,其X-射线粉末衍射图谱在15.0±0.2°处具有衍射峰;或者在23.9±0.2°处具有衍射峰;或者在9.7±0.2°处具有衍射峰;或者在5.3±0.2°处具有衍射峰;或者在24.8±0.2°处具有衍射峰;或者在29.5±0.2°处具有衍射峰;或者在7.5±0.2°处具有衍射峰;或者在21.8±0.2°处具有衍射峰;或者在21.3±0.2°处具有衍射峰;或者在10.6±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
    优选地,盐酸盐晶型A的X-射线粉末衍射图谱至少包含位于2θ为15.0±0.2°、23.9±0.2°、9.7±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为5.3±0.2°、24.8±0.2°、29.5±0.2°、7.5±0.2°、21.8±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
    更优选地,盐酸盐晶型A的X-射线粉末衍射图谱任选还包含位于2θ为21.3±0.2°、10.6±0.2°、16.9±0.2°、16.0±0.2°、18.4±0.2°、25.8±0.2°、28.4±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
    进一步优选地,盐酸盐晶型A的X-射线粉末衍射图谱包含位于2θ为15.0±0.2°、23.9±0.2°、9.7±0.2°、5.3±0.2°、24.8±0.2°、29.5±0.2°、7.5±0.2°、21.8±0.2°、 21.3±0.2°、10.6±0.2°、16.9±0.2°、16.0±0.2°、18.4±0.2°、25.8±0.2°、28.4±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
    更进一步优选地,盐酸盐晶型A的X-射线粉末衍射图谱如图12所示;或其DSC图谱基本如图13所示,或其TGA图谱基本如图14所示;
    或,为盐酸盐晶型B,其X-射线粉末衍射图谱在15.9±0.2°处具有衍射峰;或者在22.2±0.2°处具有衍射峰;或者在5.2±0.2°处具有衍射峰;或者在21.7±0.2°处具有衍射峰;或者在26.0±0.2°处具有衍射峰;或者在4.6±0.2°处具有衍射峰;或者在28.4±0.2°处具有衍射峰;或者在9.2±0.2°处具有衍射峰;或者在17.3±0.2°处具有衍射峰;或者在15.2±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
    优选地,盐酸盐晶型B的X-射线粉末衍射图谱至少包含位于2θ为15.9±0.2°、22.2±0.2°、5.2±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为21.7±0.2°、26.0±0.2°、4.6±0.2°、28.4±0.2°、9.2±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
    更优选地,盐酸盐晶型B的X-射线粉末衍射图谱任选还包含位于2θ为17.3±0.2°、15.2±0.2°、10.5±0.2°、38.0±0.2°、20.3±0.2°、23.8±0.2°、29.5±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,至少包含其中任意2处、3处、4处、5处、6处、7处;
    进一步优选地,盐酸盐晶型B的X-射线粉末衍射图谱包含位于2θ为15.9±0.2°、22.2±0.2°、5.2±0.2°、21.7±0.2°、26.0±0.2°、4.6±0.2°、28.4±0.2°、9.2±0.2°、17.3±0.2°、15.2±0.2°、10.5±0.2°、38.0±0.2°、20.3±0.2°、23.8±0.2°、29.5±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
    更进一步优选地,盐酸盐晶型B的X-射线粉末衍射图谱如图15所示;
    或,为氢溴酸盐晶型A,其X-射线粉末衍射图谱在5.3±0.2°处具有衍射峰;或者在22.7±0.2°处具有衍射峰;或者在14.8±0.2°处具有衍射峰;或者在10.5±0.2°处具有衍射峰;或者在22.5±0.2°处具有衍射峰;或者在28.0±0.2°处具有衍射峰;或者在30.0±0.2°处具有衍射峰;或者在23.4±0.2°处具有衍射峰;或者在23.3±0.2°处具有衍射峰;或者在26.5±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
    优选地,氢溴酸盐晶型A的X-射线粉末衍射图谱至少包含位于2θ为5.3±0.2°、22.7±0.2°、14.8±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三 条;任选的,进一步还可以包含位于2θ为10.5±0.2°、22.5±0.2°、28.0±0.2°、30.0±0.2°、23.4±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
    更优选地,氢溴酸盐晶型A的X-射线粉末衍射图谱任选还包含位于2θ为23.3±0.2°、26.5±0.2°、34.9±0.2°、15.8±0.2°、25.0±0.2°、31.9±0.2°、37.0±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,包含其中任意2处、3处、4处、5处、6处、7处;
    进一步优选地,氢溴酸盐晶型A的X-射线粉末衍射图谱包含位于2θ为5.3±0.2°、22.7±0.2°、14.8±0.2°、10.5±0.2°、22.5±0.2°、28.0±0.2°、30.0±0.2°、23.4±0.2°、23.3±0.2°、26.5±0.2°、34.9±0.2°、15.8±0.2°、25.0±0.2°、31.9±0.2°、37.0±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
    更进一步优选地,氢溴酸盐晶型A的X-射线粉末衍射图谱如图16所示;或其DSC图谱基本如图17所示;
    或,为氢溴酸盐晶型B,其X-射线粉末衍射图谱在23.4±0.2°处具有衍射峰;或者在15.9±0.2°处具有衍射峰;或者在16.2±0.2°处具有衍射峰;或者在14.2±0.2°处具有衍射峰;或者在5.3±0.2°处具有衍射峰;或者在10.6±0.2°处具有衍射峰;或者在23.1±0.2°处具有衍射峰;或者在24.1±0.2°处具有衍射峰;或者在14.8±0.2°处具有衍射峰;或者在9.5±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
    优选地,氢溴酸盐晶型B的X-射线粉末衍射图谱至少包含位于2θ为23.4±0.2°、15.9±0.2°、16.2±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为14.2±0.2°、5.3±0.2°、10.6±0.2°、23.1±0.2°、24.1±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
    更优选地,氢溴酸盐晶型B的X-射线粉末衍射图谱任选还包含位于2θ为14.8±0.2°、9.5±0.2°、16.9±0.2°、13.9±0.2°、29.5±0.2°、32.2±0.2°、22.2±0.2°中的一处或多处衍射峰;优选至少包含其中任意2-3处,或者4-5处,或者6-7处;进一步优选,包含其中任意2处、3处、4处、5处、6处、7处;
    进一步优选地,氢溴酸盐晶型B的X-射线粉末衍射图谱包含位于2θ为23.4±0.2°、15.9±0.2°、16.2±0.2°、14.2±0.2°、5.3±0.2°、10.6±0.2°、23.1±0.2°、24.1±0.2°、14.8±0.2°、9.5±0.2°、16.9±0.2°、13.9±0.2°、29.5±0.2°、32.2±0.2°、22.2±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处、8处或10处有衍射峰;
    更进一步优选地,氢溴酸盐晶型B的X-射线粉末衍射图谱如图18所示;或 其DSC图谱基本如图19所示;或其TGA图谱基本如图20所示;
    或,为氢溴酸盐晶型C,其X-射线粉末衍射图谱在5.2±0.2°处具有衍射峰;或者在15.7±0.2°处具有衍射峰;或者在22.3±0.2°处具有衍射峰;或者在10.5±0.2°处具有衍射峰;或者在17.4±0.2°处具有衍射峰;或者在38.0±0.2°处具有衍射峰;或者在26.3±0.2°处具有衍射峰;或者在28.0±0.2°处具有衍射峰;优选包含上述衍射峰中的任意2-5处,或者3-5处,或者3-6处,或者3-8处,或者5-8处,或者6-8处,更优选包含其中任意6处、7处或8处;
    优选地,氢溴酸盐晶型C的X-射线粉末衍射图谱至少包含位于2θ为5.2±0.2°、15.7±0.2°、22.3±0.2°中的一处或多处衍射峰,优选包含其中两条,更优选包含三条;任选的,进一步还可以包含位于2θ为10.5±0.2°、17.4±0.2°、38.0±0.2°、26.3±0.2°、28.0±0.2°中的至少一条,优选包含其中2条、3条、4条或5条;
    更优选地,氢溴酸盐晶型C的X-射线粉末衍射图谱包含位于2θ为5.2±0.2°、15.7±0.2°、22.3±0.2°、10.5±0.2°、17.4±0.2°、38.0±0.2°、26.3±0.2°、28.0±0.2°中的一处或多处衍射峰,优选的,包含其中任选的4处、5处、6处或8处有衍射峰;
    进一步优选地,氢溴酸盐晶型C的X-射线粉末衍射图谱如图21所示。
  9. 根据权利要求8所述的酸式盐晶型,其特征在于,
    各晶型中X-射线粉末衍射图谱中相对峰强度为前十强的衍射峰位置与其对应的X-射线粉末衍射附图位置衍射峰的2θ误差为±0.2°~±0.5°,优选±0.2°~±0.3°,最优选±0.2°。
  10. 根据权利要求8或9所述的酸式盐晶型,其特征在于,酸式盐晶型为水合物或无水物。
  11. 制备权利要求1-7任一项所述的酸式盐或权利要求8-10任一项所述的酸式盐晶型的方法,包括如下步骤:
    1)称取适量的游离碱,加溶剂溶解;
    2)加入适量的酸,搅拌;
    3)快速离心或静置得到酸式盐;
    或者,包括如下步骤:
    1)称取适量的游离碱,加溶剂溶解;
    2)加入适量的酸,搅拌;
    3)离心干燥后,得到酸式盐晶型;
    溶剂为有机溶剂,优选甲醇、乙醇、四氢呋喃、2-甲基四氢呋喃、甲苯、乙 酸异丙酯、叔丁醇、正丁醇、丙酮、2-丁酮、二氯甲烷、乙酸乙酯或1,4-二氧六环中的至少一种;
    酸选自盐酸、硫酸、硝酸、氢溴酸、氢氟酸、氢碘酸、磷酸、2,5-二羟基苯甲酸、1-羟基-2-萘甲酸、醋酸、二氯醋酸、三氯醋酸、乙酰氧肟酸、己二酸、苯磺酸、4-氯苯磺酸、苯甲酸、4-乙酰氨基苯甲酸、4-氨基苯甲酸、癸酸、己酸、辛酸、肉桂酸、柠檬酸、环己烷氨基磺酸、樟脑磺酸、天门冬氨酸、樟脑酸、葡萄糖酸、葡糖醛酸、谷氨酸、异抗坏血酸、乳酸、苹果酸、扁桃酸、焦谷氨酸、酒石酸、十二烷基硫酸、二苯甲酰酒石酸、乙烷-1,2-二磺酸、甲磺酸、乙烷磺酸、蚁酸、富马酸、半乳糖酸、龙胆酸、戊二酸、2-酮戊二酸、乙醇酸、马尿酸、羟乙基磺酸、乳糖酸、抗坏血酸、天冬氨酸、月桂酸、樟脑酸、马来酸、丙二酸、1,5-萘二磺酸、萘-2-磺酸、烟酸、油酸、乳清酸、草酸、棕榈酸、双羟萘酸、丙酸、水杨酸、4-氨基水杨酸、癸二酸、硬脂酸、丁二酸、硫氰酸、十一碳烯酸、三氟乙酸、苯磺酸、对甲基苯磺酸或L-苹果酸;优选盐酸、硫酸、磷酸、乙烷磺酸、苯磺酸、甲磺酸、富马酸、羟乙基磺酸、草酸或氢溴酸。
  12. 一种药物组合物,其含有治疗有效量的权利要求1-7任一项所述的酸式盐或权利要求8-10任一项所述的酸式盐晶型以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
  13. 根据权利要求1-7中任一项所述的酸式盐或权利要求8-10任一项所述的酸式盐晶型,或权利要求11所述的药物组合物在制备P2X3抑制剂药物中的应用。
  14. 根据权利要求1-7中任一项所述的酸式盐或权利要求8-10任一项所述的酸式盐晶型,或权利要求11所述的药物组合物在制备治疗神经源性疾病药物中的用途;
    优选地,所述神经源性疾病选自妇科疾病、泌尿道疾病状态、呼吸障碍疾病或疼痛相关疾病或病症;
    更优选地,所述神经源性疾病选自子宫内膜异位症、膀胱过度活动症、肺纤维化或慢性咳嗽;所述疼痛相关疾病或病症选自神经性疼痛或子宫肌瘤相关的疼痛或不适。
  15. 通式(I)的化合物或其药学上可接受的盐:
    Figure PCTCN2022097828-appb-100007
    其中:
    R 1、R 2、R 3分别独立地选自氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧基、杂芳基、杂芳基氧基或-(CH 2) n1C(O)R a
    R 4、R 5分别独立地选自氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧基、杂芳基、杂芳基氧基或-(CH 2) n1C(O)R a
    R 6选自氢或保护基;
    R a选自氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧基、杂芳基或杂芳基氧基;
    所述的氨基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧基、杂芳基、杂芳基氧基、-(CH 2) n1-,任选地被氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧、杂芳基或杂芳基氧基中的一个或多个取代基所取代;n1为0、1、2、3或4;
    优选地,
    当R 1、R 2、R 3、R 4及R 6同时为氢时,R 5不为-C(CH 3) 3或-COOCH 3
    当R 1为-CF 3,且R 2、R 3、R 4及R 6同时为氢时,R 5不为-CH 2CH 3、H或Br;
    当R 1为-CF 3,且R 2、R 3、R 5及R 6同时为氢时,R 4不为-CN;
    更优选地,所述化合物通式(I)的结构如通式(I-1)所示:
    Figure PCTCN2022097828-appb-100008
    进一步优选地,所述化合物的结构选自式(I-1-1)、式(I-1-2)或式(I-1-3):
    Figure PCTCN2022097828-appb-100009
    其中,
    R 1选自氢、氘、卤素、氰基、C 1-8烷基、C 1-8烷氧基、C 1-8卤代烷基或C 1-8卤代烷氧基,优选氰基、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基或C 1-3卤代烷氧基,更优选甲基、乙基、三氟甲基、甲氧基或氰基;
    R 5选自氢、氘、卤素、氰基、C 1-8烷基、C 1-8烷氧基、C 1-8卤代烷基、C 1-8卤代烷氧基或-C(O)R a,优选氢、氘、卤素、氰基或-C(O)R a,更优选氢、氘、氟、氯、溴或-C(O)R a
    R a选自氢、氘、卤素、氨基、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基或C 1-3卤代烷氧基,优选氨基、甲氧基、乙氧基或异丙氧基。
  16. 一种通式(I’)的化合物的制备方法,其特征在于,包括步骤(a):
    Figure PCTCN2022097828-appb-100010
    通式(II)的化合物与通式(III)的化合物在能得到通式(I’)的化合物或其盐的条件下反应得到通式(I’)的化合物或其盐;
    所述M 1、M 2分别独立地选自H、Li、Na、K或Cs,M 1优选为H,M 2优选为Na或K;
    R 1、R 2、R 3、R 4、R 5的定义如权利要求15所述。
  17. 一种通式(VII)的化合物的制备方法,其特征在于,包含步骤(b):
    Figure PCTCN2022097828-appb-100011
    通式(I)的化合物与通式(VI)的化合物反应得到通式(VII)的化合物;
    X为卤素;优选为氟、氯或溴;更优选为氯或溴;
    L 1选自-(CH 2) n2-、-(CH 2) n2O-、-(CH 2) n2S-、-(CH 2) n2NR c-、-(CH 2) n2C(O)NR c-或-(CH 2) n2NR cC(O)-;优选为-CH 2C(O)NH-;
    n2为0、1、2或3;
    x为0、1、2或3;
    R b、R c分别独立地选自氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧基、杂芳基或杂芳基氧基;
    所述的氨基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧基、杂芳基、杂芳基氧基、-(CH 2) n2-,任选地被氢、氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧、杂芳基或杂芳基氧基中的一个或多个取代基所取代;R b优选为卤素;R c优选为H;
    环A选自环烷基、杂环基、芳基或杂芳基,优选为
    Figure PCTCN2022097828-appb-100012
    和/或,
    通式(VI)化合物优选为
    Figure PCTCN2022097828-appb-100013
    更优选为
    Figure PCTCN2022097828-appb-100014
    进一步优选为
    Figure PCTCN2022097828-appb-100015
    R 1、R 2、R 3、R 4、R 5、R 6的定义如权利要求15所述;
    任选地,所述通式(VII)的化合物的制备方法中还包括制备通式(I)的化合物的步骤,所述制备通式(I)的化合物的步骤包括权利要求16的制备方法。
  18. 一种通式(I-3)的化合物的制备方法,其特征在于,包括步骤(c-1):
    Figure PCTCN2022097828-appb-100016
    通式(I-2)的化合物在氨气或氨气等价物的条件下进行胺化还原反应,得到通式(I-3)的化合物;
    所述氨气等价物为氨的有机溶液或氨水;所述氨的有机溶液优选为氨的甲醇溶液、氨的乙醇溶液、氨的异丙醇溶液或氨的二氧六环溶液;
    R a选自氢、氘、卤素、羟基、烷氧基、卤代烷氧基、芳基氧基或杂芳基氧基;所述的烷氧基、卤代烷氧基、芳基氧基或杂芳基氧基,任选地被氘、卤素、氨基、硝基、羟基、氰基、烷基、氘代烷基、卤代烷基、羟烷基、烷氧基、卤代烷氧基、烯基、炔基、杂环基烷基、环烷基、杂环基、芳基、芳基氧、杂芳基或杂芳基氧基中的一个或多个取代基所取代;
    R 1、R 2、R 3、R 4、R 6的定义如权利要求15所述。
  19. 一种通式(I-4)的化合物的制备方法,其特征在于,包括步骤(d-1):
    Figure PCTCN2022097828-appb-100017
    通式(I-3)的化合物在脱水剂存在的条件下反应得到通式(I-4)的化合物;所述脱水剂优选为乙酸酐、三氟乙酸酐、P 2O 5、三聚氯氰、三氯氧磷、三氯化磷或浓硫酸中的一种或多种;
    R 1、R 2、R 3、R 4、R 6的定义如权利要求15所述。
  20. 通式(I)的化合物、通式(II)的化合物、通式(III)的化合物的应用,所述通式(I)的化合物、通式(II)的化合物、通式(III)的化合物作为中间体用于制备P2X3抑制剂,所述P2X3抑制剂优选为含有母核结构为4,5-二氢吡唑并[1,5-a]吡啶并[3,2-e]嘧啶的P2X3抑制剂,更优选为通式(VII-4)化合物;
    Figure PCTCN2022097828-appb-100018
    L 1、x、R b、环A、R 1、R 2、R 3、R 4、R 5、R 6的定义如权利要求17所述;
    M 1、M 2的定义如权利要求16所述。
PCT/CN2022/097828 2021-06-09 2022-06-09 含吡唑多环类衍生物的盐、晶型及其制备方法和应用 WO2022258007A1 (zh)

Priority Applications (9)

Application Number Priority Date Filing Date Title
IL309178A IL309178A (en) 2021-06-09 2022-06-09 A salt and crystal form of a polycyclic derivative containing pyrazole, and the method of preparation thereof and its use
MX2023014525A MX2023014525A (es) 2021-06-09 2022-06-09 Forma salina y cristal del derivado policiclico que contiene pirazol, y metodo de preparacion del mismo y uso del mismo.
JP2023576189A JP2024522207A (ja) 2021-06-09 2022-06-09 ピラゾール含有多環式誘導体の塩及び結晶形、並びにその調製方法及びその使用
AU2022290123A AU2022290123A1 (en) 2021-06-09 2022-06-09 Salt and crystal form of pyrazole-containing polycyclic derivative, and preparation method therefor and use thereof
EP22819604.4A EP4353725A1 (en) 2021-06-09 2022-06-09 Salt and crystal form of pyrazole-containing polycyclic derivative, and preparation method therefor and use thereof
CA3221791A CA3221791A1 (en) 2021-06-09 2022-06-09 Salt and crystal form of pyrazole-containing polycyclic derivative, and preparation method therefor and use thereof
BR112023025543A BR112023025543A2 (pt) 2021-06-09 2022-06-09 Forma de sal e cristal de derivado policíclico contendo pirazol, e método de preparação do mesmo e uso do mesmo
KR1020247000484A KR20240019257A (ko) 2021-06-09 2022-06-09 피라졸-함유 다환식 유도체의 염 및 결정형, 및 이의 제조 방법 및 이의 용도
CN202280039138.5A CN117412974A (zh) 2021-06-09 2022-06-09 含吡唑多环类衍生物的盐、晶型及其制备方法和应用

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202110642255 2021-06-09
CN202110642255.8 2021-06-09
CN202110655580.8 2021-06-11
CN202110655580 2021-06-11

Publications (1)

Publication Number Publication Date
WO2022258007A1 true WO2022258007A1 (zh) 2022-12-15

Family

ID=84424768

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/097828 WO2022258007A1 (zh) 2021-06-09 2022-06-09 含吡唑多环类衍生物的盐、晶型及其制备方法和应用

Country Status (10)

Country Link
EP (1) EP4353725A1 (zh)
JP (1) JP2024522207A (zh)
KR (1) KR20240019257A (zh)
CN (1) CN117412974A (zh)
AU (1) AU2022290123A1 (zh)
BR (1) BR112023025543A2 (zh)
CA (1) CA3221791A1 (zh)
IL (1) IL309178A (zh)
MX (1) MX2023014525A (zh)
WO (1) WO2022258007A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4072680A (en) * 1977-04-28 1978-02-07 E. R. Squibb & Sons, Inc. Derivatives of pyrazolo[1,5-a]pyrido[3,2-e]pyrimidine
CN104540828A (zh) * 2012-05-21 2015-04-22 多曼治疗学公司 作为ii组代谢型谷氨酸受体的别构调节剂的取代的吡唑并喹唑啉酮和吡咯并喹唑啉酮
CN105683190A (zh) * 2013-10-29 2016-06-15 美迪维尔公司 基于喹唑啉的呼吸道合胞病毒抑制剂
CN108727377A (zh) * 2017-04-14 2018-11-02 四川大学 3-氰基吡唑并[1,5-a]嘧啶衍生物及其制备方法和用途
WO2021115225A1 (zh) * 2019-12-10 2021-06-17 上海翰森生物医药科技有限公司 含吡唑多环类衍生物抑制剂、其制备方法和应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4072680A (en) * 1977-04-28 1978-02-07 E. R. Squibb & Sons, Inc. Derivatives of pyrazolo[1,5-a]pyrido[3,2-e]pyrimidine
CN104540828A (zh) * 2012-05-21 2015-04-22 多曼治疗学公司 作为ii组代谢型谷氨酸受体的别构调节剂的取代的吡唑并喹唑啉酮和吡咯并喹唑啉酮
CN105683190A (zh) * 2013-10-29 2016-06-15 美迪维尔公司 基于喹唑啉的呼吸道合胞病毒抑制剂
CN108727377A (zh) * 2017-04-14 2018-11-02 四川大学 3-氰基吡唑并[1,5-a]嘧啶衍生物及其制备方法和用途
WO2021115225A1 (zh) * 2019-12-10 2021-06-17 上海翰森生物医药科技有限公司 含吡唑多环类衍生物抑制剂、其制备方法和应用

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
GHOZLAN SAID A. S., RAMADAN MUHAMMED A., ABDELMONIEM AMR M., ABDELHAMID ISMAIL A.: "N-(Pyrazol-5-yl)cyanoacetamide in heterocyclic synthesis: synthesis of novel N-(pyrazol-5-yl)pyridine-3,5-dicarbonitrile, pyrazolo[1,5-a]pyrido[3,2-e]pyrimidine-7-carbonitrile and pyrazolo[4,3-e]pyrido[1,2-a]pyrimidine-6,8-dicarbonitrile moieties", ARKIVOC, vol. 2019, no. 5, 30 December 2018 (2018-12-30), pages 30 - 41, XP093012949, DOI: 10.24820/ark.5550190.p010.801 *

Also Published As

Publication number Publication date
AU2022290123A1 (en) 2023-12-14
CN117412974A (zh) 2024-01-16
IL309178A (en) 2024-02-01
MX2023014525A (es) 2024-01-29
JP2024522207A (ja) 2024-06-11
KR20240019257A (ko) 2024-02-14
EP4353725A1 (en) 2024-04-17
BR112023025543A2 (pt) 2024-02-27
CA3221791A1 (en) 2022-12-15

Similar Documents

Publication Publication Date Title
IL297165A (en) Soysag tricyclic CRS inhibitors
WO2021129824A1 (zh) 新型K-Ras G12C抑制剂
CN112409331A (zh) 杂环类衍生物抑制剂、其制备方法和应用
KR20190014505A (ko) 이소퀴놀린-3-일 카르복스아마이드 및 이의 제제와 용도
CN104080784B (zh) 用作卵泡刺激素调节剂的苯甲酰胺衍生物
TWI824403B (zh) 一種雜環化合物、其中間體、其製備方法及其應用
WO2014114274A1 (zh) 氘代的苯基氨基嘧啶化合物以及包含该化合物的药物组合物
JP2012520246A (ja) Kcnq2/3モジュレータとしての置換された3−アミノイソオキサゾロピリジン
TW201605805A (zh) 喹唑啉酮及異喹啉酮衍生物
WO2016139292A1 (en) Pyridinone compound, pharmaceutical composition containing the same and use
CN101747282A (zh) 一类含有嘧啶酮苯基的化合物、其药物组合物及其制备方法和用途
WO2020151636A1 (zh) Pde9抑制剂及其用途
WO2019076336A1 (zh) 含吡唑基的三并环类衍生物、其制备方法和应用
TWI723480B (zh) 用作fgfr4抑制劑的稠環衍生物
WO2022111644A1 (zh) 含氮杂环类衍生物的盐、晶型及其制备方法和应用
CN114222746B (zh) 含吡唑多环类衍生物抑制剂、其制备方法和应用
WO2022258007A1 (zh) 含吡唑多环类衍生物的盐、晶型及其制备方法和应用
CN114901642B (zh) 一种稠环化合物及其应用
TWI820182B (zh) 四氫哌喃基胺基-吡咯并嘧啶酮化合物的固體形式
WO2022100738A1 (zh) 含二并环类衍生物抑制剂自由碱的晶型及其制备方法和应用
CN114478586A (zh) 一种含二并环类衍生物抑制剂盐或晶型及其制备方法和应用
TW202222795A (zh) 一種含吲哚類衍生物的鹽、晶型及其製備方法和應用
CN115448922A (zh) 含吡唑多环类衍生物的自由碱晶型及其制备方法和应用
CN114478519B (zh) 吡唑并吡啶类化合物或其盐及其制备方法和用途
WO2020200161A1 (zh) 含吲唑基的三并环类衍生物的盐及其晶型

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: 22819604

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2022290123

Country of ref document: AU

Ref document number: AU2022290123

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 202280039138.5

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: MX/A/2023/014525

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 12023553354

Country of ref document: PH

Ref document number: 3221791

Country of ref document: CA

Ref document number: 309178

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2301008032

Country of ref document: TH

ENP Entry into the national phase

Ref document number: 2023576189

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 18569184

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112023025543

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2022290123

Country of ref document: AU

Date of ref document: 20220609

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 202393526

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20247000484

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1020247000484

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2022819604

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022819604

Country of ref document: EP

Effective date: 20240109

ENP Entry into the national phase

Ref document number: 112023025543

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20231205