CN111302998A - Salts of heteroaromatic derivatives and use thereof - Google Patents

Salts of heteroaromatic derivatives and use thereof Download PDF

Info

Publication number
CN111302998A
CN111302998A CN202010181458.7A CN202010181458A CN111302998A CN 111302998 A CN111302998 A CN 111302998A CN 202010181458 A CN202010181458 A CN 202010181458A CN 111302998 A CN111302998 A CN 111302998A
Authority
CN
China
Prior art keywords
degrees
salt
salicylate
oxalate
ray powder
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202010181458.7A
Other languages
Chinese (zh)
Inventor
钟文和
金传飞
张英俊
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Dongguan Dongyangguang New Drug R & D Co ltd
Sunshine Lake Pharma Co Ltd
Guangdong HEC Pharmaceutical
Original Assignee
Dongguan Dongyangguang New Drug R & D Co ltd
Sunshine Lake Pharma Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Dongguan Dongyangguang New Drug R & D Co ltd, Sunshine Lake Pharma Co Ltd filed Critical Dongguan Dongyangguang New Drug R & D Co ltd
Priority to CN202010181458.7A priority Critical patent/CN111302998A/en
Publication of CN111302998A publication Critical patent/CN111302998A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C309/00Sulfonic acids; Halides, esters, or anhydrides thereof
    • C07C309/01Sulfonic acids
    • C07C309/28Sulfonic acids having sulfo groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • C07C309/29Sulfonic acids having sulfo groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton of non-condensed six-membered aromatic rings
    • C07C309/30Sulfonic acids having sulfo groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton of non-condensed six-membered aromatic rings of six-membered aromatic rings substituted by alkyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C55/00Saturated compounds having more than one carboxyl group bound to acyclic carbon atoms
    • C07C55/02Dicarboxylic acids
    • C07C55/06Oxalic acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C55/00Saturated compounds having more than one carboxyl group bound to acyclic carbon atoms
    • C07C55/02Dicarboxylic acids
    • C07C55/10Succinic acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C65/00Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C65/01Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing hydroxy or O-metal groups
    • C07C65/03Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing hydroxy or O-metal groups monocyclic and having all hydroxy or O-metal groups bound to the ring
    • C07C65/05Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing hydroxy or O-metal groups monocyclic and having all hydroxy or O-metal groups bound to the ring o-Hydroxy carboxylic acids
    • C07C65/10Salicylic acid
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Health & Medical Sciences (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biomedical Technology (AREA)
  • Public Health (AREA)
  • Psychology (AREA)
  • Psychiatry (AREA)
  • Child & Adolescent Psychology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to salts of aromatic heterocyclic derivatives and application thereof. The invention also relates to a pharmaceutical composition comprising said salt and the use of said salt or a pharmaceutical composition of said salt for the preparation of a medicament for the prevention, treatment or alleviation of 5-HT6Receptor-related diseases, in particular Alzheimer's disease.

Description

Salts of heteroaromatic derivatives and use thereof
Technical Field
The invention belongs to the technical field of medicines, relates to salts of aromatic heterocyclic derivatives and application thereof, and particularly relates to salts of 3- (difluoromethyl) -1- ((3-fluorophenyl) sulfonyl) -4- (piperazine-1-yl) -1H-indole and application thereof, and further relates to a pharmaceutical composition containing the salts.
Background
Various central nervous system disorders such as anxiety, depression, etc., are associated with disorders of the neurotransmitters 5-hydroxytryptamine (5-HT) or serotonin. The neurotransmitter 5-hydroxytryptamine (5-HT) functions as the primary regulatory neurotransmitter in the brain by being termed 5-HT1,5-HT2,5-HT3,5-HT4,5-HT5,5-HT6And 5-HT7Are mediated by a large family of receptors. Based on high levels of 5-HT in the brain6Receptor mRNA, 5-HT has been proposed6Receptors may play a role in the pathology and treatment of central nervous system disorders. In particular, 5-HT has been determined6Selective ligands have potential therapeutic utility in certain CNS (central nervous system) disorders, e.g. Parkinson's disease, Huntington's disease, anxiety, depression, manic depression, psychosis, epilepsy, obsessive-compulsive disorders, migraine, Alzheimer's disease (cognitive memory enhancement), sleep disorders, eating disorders (e.g. anorexia and bulimia), panic attacks, ADHD (attention deficit hyperactivity disorder), attention deficit disorder, drug abuse (e.g. ***e, ethanol, nicotine and benzodiazepines)
Figure BDA0002412651000000011
Class) and conditions associated with spinal trauma or head injury (e.g., hydrocephalus). The 5-HT is expected6Selective ligands may also be useful in the treatment of certain gastrointestinal disorders such as functional bowel disorders. (see, e.g., Roth, B.L., et al, J.Pharmacol.Exp.Ther.,1994,268, 1403-14120; Sibley, D.R., et al, Mol, Pharmacol, 1993,43, 320-327; sleight, a.j., et al, neurosransisition, 1995,11,1-5 and Sleight, a.j., et al, setonin ID Research Alert,1997,2(3), 115-.
It was found that known 5-HT6Receptor selective antagonists are capable of significantly increasing glutamate and aspartate levels in the frontal cortex without increasing norepinephrine, dopamine, or 5-HT levels. This selective increase in specific neurochemicals noted during memory and cognition strongly suggests 5-HT6Role of ligands in cognition (Dawson, L.A.; Nguyen, H.Q.; Li, P., British Journal of Pharmacology,2000,130(1), 23-26). With known selectivity for 5-HT6Receptor antagonists have some positive effects on studies of memory and learning in animals (Rogers, D.C.; Hatcher, P.D.; Hagan, J.J., Society of Neuroscience, Abstract, 2000, 26, 680). 5-HT6A related potential therapeutic use of ligands is in the treatment of attention deficit disorders in children and adults. Because of 5-HT6Receptor antagonists appear to increase the activity of the nigrostriatal dopamine pathway and, because ADHD is associated with abnormalities in the caudate nucleus (Ernst, M; Zametkin, A.J.; Matochik, J.H.; Jons, P.A.; Cohen, R.M., journal of Neuroscience, 1998, 18(5), 5901-6Receptor antagonists may be useful in the treatment of attention deficit disorders. 5-HT has also been determined6Receptor antagonists are potentially useful compounds for the treatment of obesity. See, e.g., Bentley et al, br.j.pharmac.1999, supplement 126; bentley et al, j.psychopharmacol.1997, supplement a 64: 255; wooley et al, Neuropharmacology 2001, 41: 210-129 and WO 02098878.
International application WO 2016004882A1 discloses the compound 3- (difluoromethyl) -1- ((3-fluorophenyl) sulfonyl) -4- (piperazin-1-yl) -1H-indole (compound of formula (I)) having 5-HT6Receptor antagonistic activity. However, no research on the salt of the compound or its crystal form has been made in the prior art.
Figure BDA0002412651000000021
Different salts and solid forms of a pharmaceutically active ingredient may have different properties. Different salts and solid forms may have significant differences in appearance, solubility, melting point, dissolution rate, bioavailability, etc., and may also have different effects on the stability, bioavailability, therapeutic effect, etc. of the drug. Therefore, in drug development, the problem of salt form and/or solid form of the drug should be fully considered.
The inventor discovers that the compound has poor water solubility and drug forming property when researching the compound, so that in order to find a solid form with better drug forming property, through a large amount of experimental researches, the preparation purity of a product is obviously improved after the compound shown in the formula (I) is salified, and the physical properties and various properties are more beneficial to the development of preparations.
Disclosure of Invention
Prior art WO 2016004882a1 discloses compounds of formula (I) as light yellow solids, but does not specifically disclose the specific solid form or salt thereof. Through a large number of experimental researches, the inventor finds that the salt of the compound shown in the formula (I) has good stability, good water solubility, good pharmacokinetic property in organisms and high preparation purity of products, namely, the physical properties and various properties of the salt are more beneficial to the development of preparations, so that the salt has more excellent drugability.
In particular, the invention relates to a salt of a compound shown as a formula (I), and the salt or a pharmaceutical composition containing the salt for preparing a medicament for preventing, treating or reducing the 5-HT6Receptor-related diseases, in particular Alzheimer's disease. The salts of the present invention may have a stable crystalline structure, and the salts may also be in the form of solvates, for example hydrates.
In one aspect, the invention provides a salt of a compound of formula (I),
Figure BDA0002412651000000022
in some embodiments, the salts described herein are salts of organic or inorganic acids.
In some embodiments, the inorganic acid salts of the present invention include, but are not limited to, hydrochloride, hydrobromide, phosphate, nitrate, or sulfate salts; the organic acid salt is acetate, oxalate, fumarate, maleate, tartrate, citrate, succinate, camphorsulfonate, malonate, benzoate, salicylate, benzenesulfonate, methanesulfonate or p-toluenesulfonate.
In some embodiments, the salt of the invention is a p-toluenesulfonate salt, wherein the p-toluenesulfonate salt is p-toluenesulfonate salt form a having an X-ray powder diffraction pattern with diffraction peaks at the following 2 θ angles: 6.00 degrees +/-0.2 degrees, 13.43 degrees +/-0.2 degrees, 16.22 degrees +/-0.2 degrees, 17.04 degrees +/-0.2 degrees, 17.25 degrees +/-0.2 degrees, 20.09 degrees +/-0.2 degrees, 20.77 degrees +/-0.2 degrees and 25.01 degrees +/-0.2 degrees.
In some embodiments, the salt of the invention is an oxalate salt, wherein the oxalate salt is oxalate form a having a diffraction peak at the following 2 Θ angles in an X-ray powder diffraction pattern: 7.69 ° ± 0.2 °,9.55 ° ± 0.2 °,12.66 ° ± 0.2 °,12.96 ° ± 0.2 °,19.17 ° ± 0.2 °,20.40 ° ± 0.2 °,22.49 ° ± 0.2 °,26.13 ° ± 0.2 °.
In some embodiments, the salt of the invention is succinate, wherein the succinate is succinate form a having an X-ray powder diffraction pattern with diffraction peaks at the following 2 Θ angles: 7.65 ° ± 0.2 °,8.88 ° ± 0.2 °,13.40 ° ± 0.2 °,17.91 ° ± 0.2 °,20.92 ° ± 0.2 °,27.05 ° ± 0.2 °.
In some embodiments, the salt of the invention is a salicylate, wherein the salicylate is salicylate form a, and wherein the salicylate form a has an X-ray powder diffraction pattern having diffraction peaks at the following 2 Θ angles: 8.30 ° ± 0.2 °,12.24 ° ± 0.2 °,13.20 ° ± 0.2 °,18.56 ° ± 0.2 °,19.23 ° ± 0.2 °,20.92 ° ± 0.2 °,22.86 ° ± 0.2 °,23.79 ° ± 0.2 °.
In some embodiments, the salt of the invention is a p-toluenesulfonate salt, wherein the p-toluenesulfonate salt is p-toluenesulfonate salt form a having an X-ray powder diffraction pattern with diffraction peaks at the following 2 θ angles: 6.00 degrees +/-0.2 degrees, 13.43 degrees +/-0.2 degrees, 15.90 degrees +/-0.2 degrees, 16.22 degrees +/-0.2 degrees, 17.04 degrees +/-0.2 degrees, 17.25 degrees +/-0.2 degrees, 18.14 degrees +/-0.2 degrees, 19.09 degrees +/-0.2 degrees, 20.09 degrees +/-0.2 degrees, 20.77 degrees +/-0.2 degrees, 21.76 degrees +/-0.2 degrees and 25.01 degrees +/-0.2 degrees.
In some embodiments, the salt of the invention is an oxalate salt, wherein the oxalate salt is oxalate form a having a diffraction peak at the following 2 Θ angles in an X-ray powder diffraction pattern: 6.37 ° ± 0.2 °,7.69 ° ± 0.2 °,12.66 ° ± 0.2 °,12.96 ° ± 0.2 °,15.83 ° ± 0.2 °,16.01 ° ± 0.2 °,19.17 ° ± 0.2 °,22.49 ° ± 0.2 °,23.06 ° ± 0.2 °,26.13 ° ± 0.2 °,28.47 ° ± 0.2 °.
In some embodiments, the salt of the invention is succinate, wherein the succinate is succinate form a having an X-ray powder diffraction pattern with diffraction peaks at the following 2 Θ angles: 7.65 ° ± 0.2 °,8.88 ° ± 0.2 °,11.88 ° ± 0.2 °,13.40 ° ± 0.2 °,17.91 ° ± 0.2 °,20.17 ° ± 0.2 °,20.92 ° ± 0.2 °,23.27 ° ± 0.2 °,24.48 ° ± 0.2 °,25.78 ° ± 0.2 °,27.05 ° ± 0.2 °.
In some embodiments, the salt of the invention is a salicylate, wherein the salicylate is salicylate form a, and wherein the salicylate form a has an X-ray powder diffraction pattern having diffraction peaks at the following 2 Θ angles: 8.30 ° ± 0.2 °,9.61 ° ± 0.2 °,9.92 ° ± 0.2 °,12.24 ° ± 0.2 °,13.20 ° ± 0.2 °,17.71 ° ± 0.2 °,18.56 ° ± 0.2 °,19.23 ° ± 0.2 °,20.92 ° ± 0.2 °,21.78 ° ± 0.2 °,22.86 ° ± 0.2 °,23.79 ° ± 0.2 °,24.69 ° ± 0.2 °.
In some embodiments, the salt of the invention is a p-toluenesulfonate salt, wherein the p-toluenesulfonate salt is p-toluenesulfonate salt form a having an X-ray powder diffraction pattern with diffraction peaks at the following 2 θ angles: 6.00 +/-0.2 degrees, 6.64 +/-0.2 degrees, 10.80 +/-0.2 degrees, 11.99 +/-0.2 degrees, 13.43 +/-0.2 degrees, 14.71 +/-0.2 degrees, 14.98 +/-0.2 degrees, 15.90 +/-0.2 degrees, 16.22 +/-0.2 degrees, 17.04 +/-0.2 degrees, 17.25 +/-0.2 degrees, 17.93 +/-0.2 degrees, 18.14 +/-0.2 degrees, 19.09 +/-0.2 degrees, 19.28 +/-0.2 degrees, 20.09 +/-0.2 degrees, 20.77 +/-0.2 degrees, 21.13 +/-0.2 degrees, 21.52 +/-0.2 degrees, 21.76 +/-0.2 degrees, 22.03 +/-0.2 degrees, 22.83 degrees, 0.23.2 +/-0.2 degrees, 0.95 +/-0.2 degrees, 0.25 +/-0.2 degrees, 0.29 degrees, 2 degrees, 2.2 degrees, 2 degrees, 2.2 degrees, 3 +/-0.3 degrees, 0.25 degrees, 0.2 degrees.
In some embodiments, the salt of the invention is an oxalate salt, wherein the oxalate salt is oxalate form a having a diffraction peak at the following 2 Θ angles in an X-ray powder diffraction pattern: 6.37 ° ± 0.2 °,7.69 ° ± 0.2 °,9.55 ° ± 0.2 °,12.66 ° ± 0.2 °,12.96 ° ± 0.2 °,13.47 ° ± 0.2 °,14.88 ° ± 0.2 °,15.83 ° ± 0.2 °,16.01 ° ± 0.2 °,17.07 ° ± 0.2 °,17.96 ° ± 0.2 °,19.17 ° ± 0.2 °,20.40 ° ± 0.2 °,20.84 ° ± 0.2 °,21.28 ° ± 0.2 °,21.74 ° ± 0.2 °,22.49 ° ± 0.2 °,23.06 ° ± 0.2 °,24.45 ° ± 0.2 °,25.00 ° ± 0.2 °,25.52 ° ± 0.2 °,26.13 ° ± 0.2 °,28.47 ° ± 0.2.2 ± 0.2 °, 360.2 ° ± 0.2 °,3 ° ± 0.2 ° 3.77 ° ± 0.2.2 °.
In some embodiments, the salt of the invention is succinate, wherein the succinate is succinate form a having an X-ray powder diffraction pattern with diffraction peaks at the following 2 Θ angles: 7.65 ° ± 0.2 °,8.88 ° ± 0.2 °,11.88 ° ± 0.2 °,13.40 ° ± 0.2 °,15.40 ° ± 0.2 °,16.39 ° ± 0.2 °,17.91 ° ± 0.2 °,18.53 ° ± 0.2 °,20.17 ° ± 0.2 °,20.92 ° ± 0.2 °,22.20 ° ± 0.2 °,23.27 ° ± 0.2 °,23.57 ° ± 0.2 °,24.48 ° ± 0.2 °,25.78 ° ± 0.2 °,27.05 ° ± 0.2 °,29.49 ° ± 0.2 °,30.15 ° ± 0.2 °,31.40 ° ± 0.2 °,33.09 ° ± 0.2 °,34.21 ° ± 0.2 °,36.37 ° ± 0.2 °,37.18 ° ± 0.2 °,37.81 ° ± 0.2 °,38.69 ° ± 0.2 °.
In some embodiments, the salt of the invention is a salicylate, wherein the salicylate is salicylate form a, and wherein the salicylate form a has an X-ray powder diffraction pattern having diffraction peaks at the following 2 Θ angles: 8.30 ° ± 0.2 °,9.61 ° ± 0.2 °,9.92 ° ± 0.2 °,12.24 ° ± 0.2 °,13.20 ° ± 0.2 °,15.60 ° ± 0.2 °,16.21 ° ± 0.2 °,16.45 ° ± 0.2 °,17.52 ° ± 0.2 °,17.71 ° ± 0.2 °,18.56 ° ± 0.2 °,19.23 ° ± 0.2 °,20.92 ° ± 0.2 °,21.78 ° ± 0.2 °,22.24 ° ± 0.2 °,22.86 ° ± 0.2 °,23.79 ° ± 0.2 °,24.45 ° ± 0.24 ° ± 0.69 ° ± 0.2 °,25.83 ° ± 0.2 °,26.60 ° ± 0.2 °,28.19 ° ± 0.2 °,29.10 ° ± 0.2.18 ° ± 0.2 °, 24.68 ° ± 0.68 ° ± 0.2 °, 3580 ° ± 0.31 ° ± 0.2 °.
In some embodiments, the salt of the invention is a p-toluenesulfonate salt, characterized in that the p-toluenesulfonate salt is form a p-toluenesulfonate salt having an X-ray powder diffraction pattern substantially as shown in fig. 1.
In some embodiments, the salt of the invention is an oxalate salt, wherein the oxalate salt is oxalate form a having an X-ray powder diffraction pattern substantially as shown in figure 2.
In some embodiments, the salt of the invention is a succinate salt, wherein the succinate salt is succinate salt form a having an X-ray powder diffraction pattern substantially as shown in figure 3.
In some embodiments, the salt of the invention is a salicylate, wherein the salicylate is salicylate form a having an X-ray powder diffraction pattern substantially as shown in figure 4.
In some embodiments, the salt of the invention is a p-toluenesulfonate salt, characterized in that the p-toluenesulfonate salt is form a p-toluenesulfonate salt, the differential scanning calorimetry trace of which comprises an endothermic peak at 203.19 ℃ ± 3 ℃.
In some embodiments, the salt of the invention is an oxalate salt, wherein the oxalate salt is form a oxalate salt, and wherein a differential scanning calorimetry trace of form a of the oxalate salt comprises an endothermic peak at 212.51 ℃ ± 3 ℃.
In some embodiments, the salt of the invention is a succinate salt, characterized in that the succinate salt is succinate form a having a differential scanning calorimetry trace comprising an endothermic peak at 179.31 ℃ ± 3 ℃.
In some embodiments, the salt of the invention is a salicylate, characterized in that the salicylate is salicylate form a, and the differential scanning calorimetry trace of salicylate form a comprises an endothermic peak at 221.92 ℃ ± 3 ℃.
In some embodiments, the salt of the invention is a p-toluenesulfonate salt, characterized in that the p-toluenesulfonate salt is form a p-toluenesulfonate salt having a differential scanning calorimetry trace substantially as shown in fig. 5.
In some embodiments, the salt of the invention is an oxalate salt, wherein the oxalate salt is oxalate form a having a differential scanning calorimetry pattern substantially as shown in figure 6.
In some embodiments, the salt of the invention is a succinate salt, wherein the succinate salt is succinate form a having a differential scanning calorimetry pattern substantially as shown in figure 7.
In some embodiments, the salt of the invention is a salicylate, characterized in that the salicylate is salicylate form a having a differential scanning calorimetry trace substantially as shown in figure 8.
In another aspect, the invention relates to a pharmaceutical composition comprising any one of the salts of the invention, and a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, or combination thereof.
In one aspect, the invention relates to the use of said salt or said pharmaceutical composition for the preparation of a medicament for the prevention, treatment or alleviation of 5-HT6A receptor associated disease.
In some such embodiments, the invention relates to 5-HT6The receptor-associated disease is a central nervous system disorder, a gastrointestinal disorder, or obesity.
In some such embodiments, the central nervous system disorder of the present invention is attention deficit hyperactivity disorder, anxiety, a stress-related disorder, schizophrenia, obsessive compulsive disorder, manic depression, a neurological disorder, a memory disorder, an attention deficit disorder, parkinson's disease, amyotrophic lateral sclerosis, alzheimer's disease, or huntington's chorea.
In another aspect, the invention relates to the use of said salt or said pharmaceutical composition for the preparation of a medicamentUse in an agent for antagonising 5-HT6A receptor.
In another aspect, the invention also relates to a preparation method of the salt of the compound shown in the formula (I) or the crystal form thereof.
The solvent used in the method for preparing the salt or the crystalline form thereof according to the present invention is not particularly limited, and any solvent that can dissolve the starting materials to an extent that does not affect the properties thereof is included in the present invention. Further, many equivalents, substitutions, or equivalents in the art to which this invention pertains, as well as different proportions of solvents, solvent combinations, and solvent combinations described herein, are deemed to be encompassed by the present invention. The invention provides a preferable solvent used in each reaction step.
The experiments for the preparation of the salts or crystalline forms of the invention are described in detail in the examples section. Meanwhile, the invention provides pharmacological property test experiments (such as pharmacokinetic experiments), solubility experiments, stability experiments, hygroscopicity experiments and the like of the salt or the crystal form thereof. Experiments prove that the salt has good stability and water solubility, so the salt has better biological activity and higher stability, and is more suitable for pharmaceutical use.
Definitions and general terms
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications referred to herein are incorporated by reference in their entirety. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods, devices, and materials are described herein.
"crystalline form" or "crystalline form" refers to a solid having a highly regular chemical structure, including, but not limited to, single or multicomponent crystals, and/or polymorphs, solvates, hydrates, clathrates, co-crystals, salts, solvates of salts, hydrates of salts of compounds. Crystalline forms of the substance can be obtained by a number of methods known in the art. Such methods include, but are not limited to, melt crystallization, melt cooling, solvent crystallization, crystallization in a defined space, e.g., in a nanopore or capillary, on a surface or template, e.g., on a polymer, in the presence of an additive such as a co-crystallizing counter molecule, desolventization, dehydration, rapid evaporation, rapid cooling, slow cooling, vapor diffusion, sublimation, reactive crystallization, anti-solvent addition, milling, and solvent drop milling, among others.
"amorphous" or "amorphous form" refers to a substance formed when particles (molecules, atoms, ions) of the substance are aperiodically arranged in three-dimensional space, and is characterized by a diffuse, non-peaked, X-ray powder diffraction pattern. Amorphous is a particular physical form of solid material, with locally ordered structural features suggesting a myriad of connections to crystalline materials. Amorphous forms of a substance can be obtained by a number of methods known in the art. Such methods include, but are not limited to, quenching, anti-solvent flocculation, ball milling, spray drying, freeze drying, wet granulation, and solid dispersion techniques, among others.
"solvent" refers to a substance (typically a liquid) that is capable of completely or partially dissolving another substance (typically a solid). Solvents useful in the practice of the present invention include, but are not limited to, water, acetic acid, acetone, acetonitrile, benzene, chloroform, carbon tetrachloride, methylene chloride, dimethyl sulfoxide, 1, 4-dioxane, ethanol, ethyl acetate, butanol, t-butanol, N-dimethylacetamide, N-dimethylformamide, formamide, formic acid, heptane, hexane, isopropanol, methanol, methyl ethyl ketone, mesitylene, nitromethane, polyethylene glycol, propanol, pyridine, tetrahydrofuran, toluene, xylene, mixtures thereof, and the like.
By "anti-solvent" is meant a fluid that facilitates precipitation of the product (or product precursor) from the solvent. The anti-solvent may comprise a cold gas, or a fluid that promotes precipitation by a chemical reaction, or a fluid that reduces the solubility of the product in the solvent; it may be the same liquid as the solvent but at a different temperature, or it may be a different liquid than the solvent.
"solvate" refers to a compound having a solvent on a surface, in a crystal lattice, or on and in a crystal lattice, which may be water, acetic acid, acetone, acetonitrile, benzene, chloroform, carbon tetrachloride, methylene chloride, dimethyl sulfoxide, 1, 4-dioxane, ethanol, ethyl acetate, butanol, t-butanol, N-dimethylacetamide, N-dimethylformamide, formamide, formic acid, heptane, hexane, isopropanol, methanol, methyl ethyl ketone, methyl pyrrolidone, mesitylene, nitromethane, polyethylene glycol, propanol, pyridine, tetrahydrofuran, toluene, xylene, mixtures thereof, and the like. A specific example of a solvate is a hydrate, wherein the solvent on the surface, in the crystal lattice or on the surface and in the crystal lattice is water. The hydrates may or may not have other solvents than water on the surface of the substance, in the crystal lattice or both.
Crystalline forms can be identified by a variety of techniques, such as X-ray powder diffraction (XRPD), infrared absorption spectroscopy (IR), melting point methods, Differential Scanning Calorimetry (DSC), thermogravimetric analysis (TGA), nuclear magnetic resonance methods, raman spectroscopy, X-ray single crystal diffraction, dissolution calorimetry, Scanning Electron Microscopy (SEM), quantitative analysis, solubility, and dissolution rate, and the like.
Information such as change, crystallinity, crystal structure state and the like of the crystal form can be detected by X-ray powder diffraction (XRPD), and the method is a common means for identifying the crystal form. In some embodiments, the crystalline form of the present invention is characterized by an XRPD pattern having certain peak positions, substantially as shown in the XRPD patterns provided in the figures of the present invention. Also, the 2 θ measurement of the XRPD pattern may have experimental error, and the 2 θ measurement of the XRPD pattern may be slightly different from instrument to instrument and from sample to sample, so the 2 θ value cannot be considered absolute. The diffraction peaks have a tolerance of ± 0.2 ° according to the conditions of the instrument used in the test.
Differential Scanning Calorimetry (DSC) is carried out by continuously heating or cooling under program control to measure sample and inert reference substance (usually α -Al)2O3) The energy difference therebetween varies with temperature. The endothermic peak height of the DSC curve depends onMany factors are involved in sample preparation and instrument geometry. Thus, in some embodiments, the crystalline form of the present invention is characterized by a DSC profile with characteristic peak positions substantially as shown in the DSC profiles provided in the figures of the present invention. Meanwhile, the DSC profile may have experimental errors, and the peak position and peak value of the DSC profile may slightly differ between different instruments and different samples, so the peak position or peak value of the DSC endothermic peak cannot be regarded as absolute. The endothermic peak has a tolerance of + -3 deg.C depending on the instrument used in the experiment.
Thermogravimetric analysis (TGA) is a technique for measuring the change in mass of a substance with temperature under program control, and is suitable for examining the loss of a solvent in a crystal or the sublimation and decomposition of a sample, and it can be presumed that the crystal contains crystal water or a crystal solvent. The change in mass shown by the TGA profile depends on many factors such as sample preparation and instrumentation; the mass change of the TGA detection varies slightly from instrument to instrument and from sample to sample. There is a tolerance of + -0.1% for mass change depending on the condition of the instrument used in the test.
In the context of the present invention, the 2 θ values in the X-ray powder diffraction pattern are all in degrees (°).
The term "substantially as shown" means that at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or at least 95%, or at least 99% of the peaks in the X-ray powder diffraction pattern or DSC pattern or raman spectrum or infrared spectrum are shown in the figure.
When referring to a spectrogram or/and data appearing in a graph, "peak" refers to a feature that one skilled in the art would recognize as not being attributable to background noise.
The present invention relates to said crystalline form of the salt of (3- (difluoromethyl) -1- ((3-fluorophenyl) sulfonyl) -4- (piperazin-1-yl) -1H-indole, present in substantially pure crystalline form.
By "substantially pure" is meant that a crystalline form is substantially free of one or more additional crystalline forms, i.e., the crystalline form is at least 80%, or at least 85%, or at least 90%, or at least 93%, or at least 95%, or at least 98%, or at least 99%, or at least 99.5%, or at least 99.6%, or at least 99.7%, or at least 99.8%, or at least 99.9% pure, or the crystalline form contains additional crystalline forms, the percentage of which in the total volume or weight of the crystalline form is less than 20%, or less than 10%, or less than 5%, or less than 3%, or less than 1%, or less than 0.5%, or less than 0.1%, or less than 0.01%.
By "substantially free" is meant that the percentage of one or more other crystalline forms in the total volume or weight of the crystalline form is less than 20%, or less than 10%, or less than 5%, or less than 4%, or less than 3%, or less than 2%, or less than 1%, or less than 0.5%, or less than 0.1%, or less than 0.01%.
"relative intensity" (or "relative peak height") in an XRPD pattern refers to the ratio of the intensity of the first strong peak to the intensity of the other peaks when the intensity of the first strong peak is 100% of all the diffraction peaks in the X-ray powder diffraction pattern.
In the context of the present invention, the word "about" or "approximately" when used or whether used, means within 10%, suitably within 5%, and especially within 1% of a given value or range. Alternatively, the term "about" or "approximately" means within an acceptable standard error of the mean, for one of ordinary skill in the art. Whenever a number is disclosed with a value of N, any number within the values of N +/-1%, N +/-2%, N +/-3%, N +/-5%, N +/-7%, N +/-8% or N +/-10% is explicitly disclosed, wherein "+/-" means plus or minus.
"room temperature" in the present invention means a temperature of from about 10 ℃ to about 40 ℃. In some embodiments, "room temperature" refers to a temperature of from about 20 ℃ to about 30 ℃; in other embodiments, "room temperature" refers to 20 ℃, 22.5 ℃,25 ℃, 27.5 ℃, and the like.
Pharmaceutical compositions, formulations, administration and uses of the salts of the invention
The pharmaceutical composition of the invention is characterized by comprising a salt of the compound shown in the formula (I) and a pharmaceutically acceptable carrier, adjuvant or excipient. The amount of the salt of the compound in the pharmaceutical composition of the present invention is effective to detectably treat or reduce central nervous system dysfunction in the patient. The pharmaceutical compositions of the present invention may also optionally comprise other therapeutic and/or prophylactic ingredients.
Suitable carriers, adjuvants and excipients are well known to those skilled in the art and are described in detail, for example, in Ansel h.c.et al, Ansel's Pharmaceutical Dosage Forms and Drug delivery systems (2004) Lippincott, Williams & Wilkins, philidelphia; gennaro a.r.et al, Remington: the Science and Practice of Pharmacy (2000) Lippincott, Williams & Wilkins, Philadelphia; and Rowe R.C., Handbook of Pharmaceutical Excipients (2005) Pharmaceutical Press, Chicago.
The skilled person is knowledgeable and skilled in the art to enable them to select suitable amounts of suitable pharmaceutically acceptable excipients for use in the present invention. Furthermore, there is a large amount of resources available to the skilled person, who describes pharmaceutically acceptable excipients and is used to select suitable pharmaceutically acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (The American Pharmaceutical Association and The Pharmaceutical Press).
Various carriers for formulating pharmaceutically acceptable compositions, and well known techniques for their preparation, are disclosed in Remington, The Science and Practice of Pharmacy,21st edition,2005, ed.D.B.Troy, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of pharmaceutical Technology, eds.J.Swarbrick and J.C.Boylan, 1988. sup. 1999, Marcel Dekker, New York, The contents of each of which are incorporated herein by reference. Except insofar as any conventional carrier is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or interacting in a deleterious manner with any other ingredient in a pharmaceutically acceptable composition, its use is contemplated as falling within the scope of the present invention.
The pharmaceutical compositions of the present invention are prepared using techniques and methods known to those skilled in the art. Some commonly used methods in the art are described in Remington's Pharmaceutical Sciences (Mack publishing company).
In another aspect, the invention relates to a process for preparing a pharmaceutical composition comprising a salt of a compound of the invention or a crystalline form thereof and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or combination thereof, which process comprises admixing the ingredients. Pharmaceutical compositions comprising salts of the compounds of the present invention may be prepared by mixing, for example, at ambient temperature and atmospheric pressure.
The salts of the compounds of the present invention or crystalline forms thereof are generally formulated in a dosage form suitable for administration to a patient by a desired route. For example, dosage forms include those suitable for the following routes of administration: (1) oral administration, such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets and cachets; (2) parenteral administration, such as sterile solutions, suspensions, and reconstituted powders; (3) transdermal administration, such as transdermal patches; (4) rectal administration, e.g., suppositories; (5) inhalation, such as aerosols, solutions, and dry powders; and (6) topical administration, such as creams, ointments, lotions, solutions, pastes, sprays, foams and gels.
The pharmaceutical composition provided by the present invention may be provided in soft or hard capsules, which may be prepared from gelatin, methylcellulose, starch or calcium alginate. Hard gelatin capsules, also known as Dry Fill Capsules (DFC), consist of two segments, one inserted into the other, thus completely encapsulating the active ingredient. Soft Elastic Capsules (SEC) are soft, spherical shells, such as gelatin shells, which are plasticized by the addition of glycerol, sorbitol or similar polyols. The soft gelatin shell may contain a preservative to prevent microbial growth. Suitable preservatives are those as described herein, including methyl and propyl parabens, and sorbic acid. The liquid, semi-solid and solid dosage forms provided by the present invention may be encapsulated in a capsule. Suitable liquid and semi-solid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils or triglycerides. Capsules containing such solutions may be as described in U.S. patent nos.4,328,245; 4,409,239 and 4,410,545. The capsules may also be coated as known to those skilled in the art to improve or maintain dissolution of the active ingredient.
In one embodiment, the treatment methods of the present invention comprise administering to a patient in need thereof a safe and effective amount of a salt of a compound of the present invention or a crystalline form thereof or a pharmaceutical composition comprising a salt of a compound of the present invention or a crystalline form thereof. Various embodiments of the present invention encompass the treatment of the diseases mentioned herein by administering to a patient in need thereof a safe and effective amount of a salt of a compound of the present invention or a crystalline form thereof or a pharmaceutical composition comprising a salt of a compound of the present invention or a crystalline form thereof.
In one embodiment, the salt of the compound of the present invention or a crystalline form thereof or a pharmaceutical composition comprising the salt of the compound of the present invention or a crystalline form thereof may be administered by any suitable route of administration, including systemic administration and topical administration. Systemic administration includes oral, parenteral, transdermal and rectal administration. Typical parenteral administration refers to administration by injection or infusion, including intravenous, intramuscular, and subcutaneous injection or infusion. Topical administration includes application to the skin and intraocular, otic, intravaginal, inhalation, and intranasal administration. In one embodiment, a salt of a compound of the invention or a crystalline form thereof or a pharmaceutical composition comprising a salt of a compound of the invention or a crystalline form thereof may be administered orally. In another embodiment, a salt of a compound of the invention or a crystalline form thereof or a pharmaceutical composition comprising a salt of a compound of the invention or a crystalline form thereof may be administered by inhalation. In yet another embodiment, the salt of the compound of the present invention or a crystalline form thereof or a pharmaceutical composition comprising the salt of the compound of the present invention or a crystalline form thereof may be administered intranasally.
In one embodiment, the salt of the compound of the present invention or a crystalline form thereof or a pharmaceutical composition comprising the salt of the compound of the present invention or a crystalline form thereof may be administered once or several times at different time intervals within a specified time period according to a dosing regimen. For example, once, twice, three times or four times daily. In one embodiment, the administration is once daily. In yet another embodiment, the administration is twice daily. The administration may be carried out until the desired therapeutic effect is achieved or the desired therapeutic effect is maintained indefinitely. Suitable dosing regimens for a salt of a compound of the invention or a crystalline form thereof, or a pharmaceutical composition comprising a salt of a compound of the invention or a crystalline form thereof, depend on the pharmacokinetic properties, such as absorption, distribution and half-life, of the salt of the compound or the crystalline form thereof, which can be determined by the skilled person. In addition, suitable dosing regimens of the salts of the compounds of the invention or crystalline forms thereof, or pharmaceutical compositions comprising the salts of the compounds of the invention or crystalline forms thereof, including the duration of the regimen, will depend upon the condition being treated, the severity of the condition being treated, the age and physical condition of the patient being treated, the medical history of the patient being treated, the nature of concurrent therapy, the desired therapeutic effect, and like factors within the knowledge and experience of the skilled artisan. Such a skilled artisan will also appreciate that appropriate dosage regimens may be required to be adjusted for the individual patient's response to the dosage regimen, or as the individual patient needs to change over time.
The salts of the compounds of the present invention or crystalline forms thereof may be administered concurrently with, before or after one or more other therapeutic agents. The salts of the compounds of the present invention or their crystalline forms may be administered separately from the other therapeutic agents by the same or different routes of administration, or in the form of the same pharmaceutical compositions.
The salts of the compounds of the present invention or their crystalline forms may be used in combination with, i.e. form a pharmaceutical combination according to the present invention, e.g.: donepezil, nalmefene, risperidone, vitamins E, SAM-760, AVN-211, AVN-101, RP-5063, tozadenant, PRX-3140, PRX-8066, RVT-101, naluzaton, idalopidine, tacrine, rivastigmine, galantamine, memantine, mirtazapine, venlafaxine, despramine, nortriptyline, zolpidem, zopiclone, nicergoline, piracetazone, selegiline, pentoxifylline, and salts and combinations thereof, etc., or a salt of a compound of the invention or a crystalline form thereof may be administered in combination with a physical method such as phototherapy or electrical stimulation.
For an individual of about 50-70kg, the pharmaceutical compositions and pharmaceutical combinations of the present invention may be in unit dosage forms containing about 1-1000mg, or about 1-500mg, or about 1-250mg, or about 1-150mg, or about 0.5-100mg, or about 1-50mg of the active ingredient. A therapeutically effective amount of a salt of a compound or a crystalline form thereof, a pharmaceutical composition or a combination thereof, will depend on the species, weight, age and condition of the subject, the disease (disorder) or illness (disease) being treated, or the severity thereof. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each active ingredient to prevent, treat or inhibit the progression of the disease (disorder) or condition (disease).
The above cited dose profiles have been demonstrated in vitro and in vivo tests using beneficial mammals (e.g., mice, rats, dogs, monkeys) or isolated organs, tissues and specimens thereof.
In one embodiment, the amount of the compound in a therapeutically effective dose of a salt of a compound of the invention or a crystalline form thereof is from about 0.1mg to about 2,000mg per day. The pharmaceutical composition thereof should provide a dose of the compound of about 0.1mg to about 2,000 mg. In a particular embodiment, the pharmaceutical dosage unit form is prepared to provide from about 1mg to about 2,000mg, from about 10mg to about 1,000mg, from about 20mg to about 500mg, or from about 25mg to about 250mg of the principal active ingredient or a combination of principal ingredients per dosage unit form. In a particular embodiment, the pharmaceutical dosage unit form is prepared to provide about 10mg,20mg,25mg,50mg,100mg,250mg,500mg,1000mg or 2000mg of the primary active ingredient.
The salt of the compound or the crystal form and the pharmaceutical composition thereof provided by the invention can be used for preparing medicaments for preventing, treating or relieving the Alzheimer's disease of mammals including human beings, and can also be used for preparing medicaments for preventing, treating or relieving the Alzheimer's disease of the mammals including human beings and 5-HT6Pharmaceutical compositions for the treatment of receptor-related diseases, and their use for antagonizing 5-HT6A pharmaceutical product of a recipient.
In particular, the amount of compound in the pharmaceutical compositions of the present invention is effective to detectably and selectively antagonize 5-HT6The receptor, the salt of the compound of the invention or the crystal forms thereof can be used for treating 5-HT6Receptor-related diseases such as Alzheimer's diseaseA medicine for treating the marine morbid disease.
The salts of the compounds of the present invention or their crystalline forms may be used in, but are in no way limited to, the prevention, treatment or alleviation of 5-HT by administering to a patient an effective amount of a salt of a compound of the present invention or a crystalline form or a pharmaceutical composition thereof6A receptor associated disease. The above-mentioned and 5-HT6The receptor-related disease is a central nervous system disorder, a gastrointestinal disorder, or obesity; wherein the central nervous system disorder is attention deficit hyperactivity disorder, anxiety, stress-related disorders, schizophrenia, obsessive compulsive disorder, manic depression, neurological disorders, memory disorders, attention deficit disorder, Parkinson's disease, amyotrophic lateral sclerosis, Alzheimer's disease or Huntington's chorea, or the like.
An "effective amount" or "effective dose" of a salt of a compound of the invention or a crystalline form or pharmaceutically acceptable composition thereof refers to an amount effective to treat or reduce the severity of one or more of the conditions mentioned herein. The salt of the compound of the present invention or a crystalline form or pharmaceutically acceptable composition thereof may be administered in any amount and by any route effective to treat or reduce the severity of the disease in accordance with the methods of the present invention. The exact amount necessary will vary depending on the patient, depending on the race, age, general condition of the patient, severity of infection, particular factors, mode of administration, and the like. The salts of the compounds of the present invention or crystalline forms or pharmaceutically acceptable compositions thereof may be administered in combination with one or more other therapeutic agents, as discussed herein.
The salts of the compounds of the present invention or their crystalline forms and pharmaceutical compositions are useful in veterinary therapy for pets, animals of the introduced breed and mammals in farm animals, in addition to human therapy. Examples of other animals include horses, dogs, and cats.
Drawings
FIG. 1 is an X-ray powder diffraction (XRPD) pattern of crystalline form A of the p-toluenesulfonate salt of the compound of formula (I).
Figure 2 is an X-ray powder diffraction (XRPD) pattern of oxalate form a of the compound of formula (I).
Figure 3 is an X-ray powder diffraction (XRPD) pattern of succinate form a of the compound of formula (I).
Figure 4 is an X-ray powder diffraction (XRPD) pattern of salicylate form a of the compound of formula (I).
FIG. 5 is a Differential Scanning Calorimetry (DSC) profile of crystalline form A of p-toluenesulfonate of the compound of formula (I).
FIG. 6 is a Differential Scanning Calorimetry (DSC) profile of oxalate form A of the compound of formula (I).
FIG. 7 is a Differential Scanning Calorimetry (DSC) profile of succinate salt form A of the compound of formula (I).
FIG. 8 is a Differential Scanning Calorimetry (DSC) profile of salicylate form A of the compound of formula (I).
Detailed Description
The invention is further illustrated by the following examples, which are not intended to limit the scope of the invention.
The X-ray powder diffraction analysis method used by the invention is that an Empyrean diffractometer obtains an X-ray powder diffraction pattern by using Cu-K α radiation (45KV,40mA), a powdery sample is prepared into thin layers on a monocrystalline silicon sample rack, the thin layers are placed on a rotary sample table, analysis is carried out in 0.0167 DEG step size within the range of 3-60 DEG, Data Collector software is used for collecting Data, HighScore Plus software is used for processing the Data, and Data Viewer software is used for reading the Data.
The Differential Scanning Calorimetry (DSC) analysis method used in the invention comprises the following steps: differential scanning calorimetry was performed using a TA Q2000 module with a thermoanalytical controller. Data were collected and analyzed using TA Instruments Thermal Solutions software. About 1-5mg of the sample was accurately weighed into a specially made aluminum crucible with a lid and the sample analysis was performed from room temperature to about 300 c using a 10 c/min linear heating device. During use, the DSC cell was purged with dry nitrogen.
The solubility of the invention is measured by an Agilent 1200 high performance liquid chromatograph DAD/VWD detector, and the type of a chromatographic column is Agilent XDB-C18(4.6 multiplied by 50mm, 5 mu m). The detection wavelength is 266nm, the flow rate is 1.0mL/min, the column temperature is 35 ℃, and the ratio of mobile phase A: acetonitrile/0.01M ammonium acetate 10/90(V/V) analytical method: acetonitrile/mobile phase a ═ 70/30(V/V), run time: for 10 minutes.
The moisture absorption of the invention is measured by adopting a DVS INT-Std type dynamic moisture and gas adsorption analyzer of Surface Measurement Systems company in England, and the humidity test range is as follows: 0% -95%, airflow: 200mL/min, temperature: 25 ℃, test point: one test point was taken per liter of 5% humidity.
Detailed description of the invention
A compound of formula (I): 3- (difluoromethyl) -1- ((3-fluorophenyl) sulfonyl) -4- (piperazin-1-yl) -1H-indole, the specific synthesis was as described in International application WO 2016004882A1, example 5.
Examples
Example 1 crystalline form a of p-toluenesulfonate according to the invention
1. Preparation of p-toluenesulfonate form A
The compound represented by the formula (I) (50mg) was added to EA (2mL), and p-toluenesulfonic acid (42mg) was further added to the mixture, and the mixture was reacted at room temperature overnight. Stopping the reaction, performing suction filtration, and drying to obtain white solid powder which is p-toluenesulfonate crystal form A.
2. Identification of p-toluenesulfonate form A
(1) Identified by Empyrean X-ray powder diffraction (XRPD) analysis as having the following characteristic peaks expressed in degrees 2 θ using Cu-K α radiation at 6.00 °,6.64 °,10.80 °,11.99 °,13.43 °,14.71 °,14.98 °,15.90 °,16.22 °,17.04 °,17.25 °,17.93 °,18.14 °,19.09 °,19.28 °,20.09 °,20.77 °,21.13 °,21.52 °,21.76 °,22.03 °,22.83 °,23.39 °,23.95 °,25.01 °,25.89 °,29.95 °, with a tolerance of error of ± 0.2 °.
(2) Identification by TA Q2000 Differential Scanning Calorimetry (DSC) analysis: the scan rate was 10 ℃/min, contained an endotherm peak at 203.19 ℃, with a margin of error of ± 3 ℃.
Example 2 crystalline form A of oxalate of the present invention
1. Preparation of oxalate form A
The compound represented by the formula (I) (50mg) was added to EA (5mL), and oxalic acid (25mg) was further added to the mixture to conduct a reaction at room temperature overnight. Stopping reaction, filtering, and drying to obtain white solid powder which is oxalate crystal form A.
2. Identification of oxalate form a
(1) Identified by Empyrean X-ray powder diffraction (XRPD) analysis as having the following characteristic peaks expressed in degrees 2 θ using Cu-K α with a tolerance of error of + -0.2 ° for 6.37 °,7.69 °,9.55 °,12.66 °,12.96 °,13.47 °,14.88 °,15.83 °,16.01 °,17.07 °,17.96 °,19.17 °,20.40 °,20.84 °,21.28 °,21.74 °,22.49 °,23.06 °,24.45 °,25.00 °,25.52 °,26.13 °,28.47 °,29.04 °,31.77 °,32.03 °,33.30 °.
(2) Identification by TA Q2000 Differential Scanning Calorimetry (DSC) analysis: the scan rate was 10 ℃/min, contained an endotherm peak at 212.51 ℃, with a margin of error of ± 3 ℃.
Example 3 succinate form a of the invention
1. Preparation of succinate form a
The compound represented by the formula (I) (50mg) was added to EA (5mL), and succinic acid (33mg) was further added to the mixture, and the mixture was reacted at room temperature overnight. Stopping reaction, filtering, and drying to obtain white solid powder as succinate crystal form A.
2. Identification of succinate form a
(1) Identified by Empyrean X-ray powder diffraction (XRPD) analysis as having the following characteristic peaks expressed in degrees 2 θ using Cu-K α with a tolerance of error of + -0.2 ° for 7.65 °,8.88,11.88 °,13.40 °,15.40 °,16.39 °,17.91 °,18.53 °,20.17 °,20.92 °,22.20 °,23.27 °,23.57 °,24.48 °,25.78 °,27.05 °,29.49 °,30.15 °,31.40 °,33.09 °,34.21 °,36.37 °,37.18 °,37.81 °,38.69 °.
(2) Identification by TA Q2000 Differential Scanning Calorimetry (DSC) analysis: the scan rate was 10 ℃/min, contained an endotherm peak at 179.31 ℃, with a margin of error of ± 3 ℃.
Example 4 salicylate form a of the invention
1. Preparation of salicylate form a
The compound represented by the formula (I) (50mg) was added to IPA (2mL), and salicylic acid (32mg) was further added to react at room temperature overnight. Stopping reaction, filtering, and drying to obtain white solid powder which is salicylate crystal form A.
2. Identification of salicylate form a
(1) Identified by Empyrean X-ray powder diffraction (XRPD) analysis as having the following characteristic peaks expressed in degrees 2 θ using Cu-K α radiation, 8.30 °,9.61 °,9.92 °,12.24 °,13.20 °,15.60 °,16.21 °,16.45 °,17.52 °,17.71 °,18.56 °,19.23 °,20.92 °,21.78 °,22.24 °,22.86 °,23.79 °,24.45 °,24.69 °,25.83 °,26.60 °,28.19 °,29.10 °,30.18 °,30.68 °,31.80 °,37.32 ° with a tolerance of ± 0.2 °.
(2) Identification by TA Q2000 Differential Scanning Calorimetry (DSC) analysis: the scan rate was 10 ℃/min, contained an endotherm peak at 221.92 ℃, with a margin of error of ± 3 ℃.
EXAMPLE 5 pharmacokinetic experiments on the salts of the invention
The test samples were encapsulated for oral administration.
3 male beagle dogs of 8-12kg were orally administered with capsules containing test samples at a dose of 5mg/kg, and blood was collected at time points of 0.25,0.5,1.0,2.0,4.0,6.0,8.0 and 24 hours. A standard curve of the appropriate range is established based on the sample concentration, and the concentration of the test sample in the plasma sample is determined in MRM mode using LC-MS/MS model AB SCIEX API4000 and subjected to quantitative analysis. According to the drug concentration-time curve, pharmacokinetic parameters are calculated by adopting a WinNonLin 6.3 software non-compartmental model method. Experiments prove that the salt has higher exposure in beagle dogs and better pharmacokinetic property.
Example 6 stability test of salts according to the invention
Taking a proper amount of sample to be tested, and respectively irradiating the sample under the condition of (4500 +/-500 lx and ultraviolet light is more than or equal to 0.7 w/m)2) Placing the sample under the conditions of high humidity (25 ℃, 75% +/-5% RH, 90% +/-5% RH) and high temperature (40 ℃ +/-2 ℃, 60 ℃ +/-2 ℃ and humidity control), carrying out influencing factor experiments, sampling respectively on days 5, 10, 15 and 30, carrying out XRPD detection, and inspecting the stability of the sample.
Experiments prove that the salt has good stability under various lofting conditions and is suitable for pharmaceutical application under high temperature (40 ℃ or 60 ℃), high humidity (25 ℃, RH 75% +/-5% or RH 90% +/-5%) and illumination conditions.
EXAMPLE 7 hygroscopicity test of the salts according to the invention
A proper amount of sample to be tested is taken, and the hygroscopicity of the sample is tested by adopting a dynamic moisture adsorption instrument. Experiments prove that the salt is not easy to deliquesce under the influence of high humidity.
Example 8 solubility testing of salts of the invention
Placing a sample to be tested in water with the temperature of 37 ℃ to prepare supersaturated turbid liquid, shaking for 24 hours, filtering, taking filtrate, and detecting the solubility of the target sample in the water by using an HPLC method. The experimental result shows that the salt has higher solubility in water, so the salt has better drug forming property and is suitable for preparation development.
The above description is only a basic description of the present invention, and any equivalent changes made according to the technical solution of the present invention should fall within the protection scope of the present invention.
In the description herein, references to the description of the term "one embodiment," "some embodiments," "an example," "a specific example," or "some examples," etc., mean that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the invention. In this specification, the schematic representations of the terms used above are not necessarily intended to refer to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, various embodiments or examples and features of different embodiments or examples described in this specification can be combined and combined by one skilled in the art without contradiction.
Although embodiments of the present invention have been shown and described above, it is understood that the above embodiments are exemplary and should not be construed as limiting the present invention, and that variations, modifications, substitutions and alterations can be made to the above embodiments by those of ordinary skill in the art within the scope of the present invention.

Claims (10)

1. A salt of a compound of formula (I),
Figure FDA0002412650990000011
wherein the salt is an organic acid salt or an inorganic acid salt.
2. The salt of claim 1, wherein the inorganic acid salt is a hydrochloride, hydrobromide, phosphate, nitrate, or sulfate; the organic acid salt is acetate, oxalate, fumarate, maleate, tartrate, citrate, succinate, camphorsulfonate, malonate, benzoate, salicylate, benzenesulfonate, methanesulfonate or p-toluenesulfonate.
3. The salt of claim 2, wherein the p-toluenesulfonate salt is form A p-toluenesulfonate salt having an X-ray powder diffraction pattern with diffraction peaks at the following 2 θ angles: 6.00 degrees +/-0.2 degrees, 13.43 degrees +/-0.2 degrees, 16.22 degrees +/-0.2 degrees, 17.04 degrees +/-0.2 degrees, 17.25 degrees +/-0.2 degrees, 20.09 degrees +/-0.2 degrees, 20.77 degrees +/-0.2 degrees and 25.01 degrees +/-0.2 degrees; or
The oxalate is oxalate crystal form A, and an X-ray powder diffraction pattern of the oxalate crystal form A has diffraction peaks at the following 2 theta angles: 7.69 ° ± 0.2 °,9.55 ° ± 0.2 °,12.66 ° ± 0.2 °,12.96 ° ± 0.2 °,19.17 ° ± 0.2 °,20.40 ° ± 0.2 °,22.49 ° ± 0.2 °,26.13 ° ± 0.2 °; or
The succinate salt is succinate salt crystal form A, and an X-ray powder diffraction pattern of the succinate salt crystal form A has diffraction peaks at the following 2 theta angles: 7.65 ° ± 0.2 °,8.88 ° ± 0.2 °,13.40 ° ± 0.2 °,17.91 ° ± 0.2 °,20.92 ° ± 0.2 °,27.05 ° ± 0.2 °; or
The salicylate is salicylate crystal form A, and an X-ray powder diffraction pattern of the salicylate crystal form A has diffraction peaks at the following 2 theta angles: 8.30 ° ± 0.2 °,12.24 ° ± 0.2 °,13.20 ° ± 0.2 °,18.56 ° ± 0.2 °,19.23 ° ± 0.2 °,20.92 ° ± 0.2 °,22.86 ° ± 0.2 °,23.79 ° ± 0.2 °.
4. The salt of claim 2, wherein the p-toluenesulfonate salt is form A p-toluenesulfonate salt having an X-ray powder diffraction pattern with diffraction peaks at the following 2 θ angles: 6.00 degrees +/-0.2 degrees, 13.43 degrees +/-0.2 degrees, 15.90 degrees +/-0.2 degrees, 16.22 degrees +/-0.2 degrees, 17.04 degrees +/-0.2 degrees, 17.25 degrees +/-0.2 degrees, 18.14 degrees +/-0.2 degrees, 19.09 degrees +/-0.2 degrees, 20.09 degrees +/-0.2 degrees, 20.77 degrees +/-0.2 degrees, 21.76 degrees +/-0.2 degrees and 25.01 degrees +/-0.2 degrees; or
The oxalate is oxalate crystal form A, and an X-ray powder diffraction pattern of the oxalate crystal form A has diffraction peaks at the following 2 theta angles: 6.37 ° ± 0.2 °,7.69 ° ± 0.2 °,12.66 ° ± 0.2 °,12.96 ° ± 0.2 °,15.83 ° ± 0.2 °,16.01 ° ± 0.2 °,19.17 ° ± 0.2 °,22.49 ° ± 0.2 °,23.06 ° ± 0.2 °,26.13 ° ± 0.2 °,28.47 ° ± 0.2 °; or
The succinate salt is succinate salt crystal form A, and an X-ray powder diffraction pattern of the succinate salt crystal form A has diffraction peaks at the following 2 theta angles: 7.65 ° ± 0.2 °,8.88 ° ± 0.2 °,11.88 ° ± 0.2 °,13.40 ° ± 0.2 °,17.91 ° ± 0.2 °,20.17 ° ± 0.2 °,20.92 ° ± 0.2 °,23.27 ° ± 0.2 °,24.48 ° ± 0.2 °,25.78 ° ± 0.2 °,27.05 ° ± 0.2 °; or
The salicylate is salicylate crystal form A, and an X-ray powder diffraction pattern of the salicylate crystal form A has diffraction peaks at the following 2 theta angles: 8.30 ° ± 0.2 °,9.61 ° ± 0.2 °,9.92 ° ± 0.2 °,12.24 ° ± 0.2 °,13.20 ° ± 0.2 °,17.71 ° ± 0.2 °,18.56 ° ± 0.2 °,19.23 ° ± 0.2 °,20.92 ° ± 0.2 °,21.78 ° ± 0.2 °,22.86 ° ± 0.2 °,23.79 ° ± 0.2 °,24.69 ° ± 0.2 °.
5. The salt of claim 2, wherein the p-toluenesulfonate salt is form A p-toluenesulfonate salt having an X-ray powder diffraction pattern with diffraction peaks at the following 2 θ angles: 6.00 ° ± 0.2 °,6.64 ° ± 0.2 °,10.80 ° ± 0.2 °,11.99 ° ± 0.2 °,13.43 ° ± 0.2 °,14.71 ° ± 0.2 °,14.98 ° ± 0.2 °,15.90 ° ± 0.2 °,16.22 ° ± 0.2 °,17.04 ° ± 0.2 °,17.25 ° ± 0.2 °,17.93 ° ± 0.2 °,18.14 ° ± 0.2 °,19.09 ° ± 0.2 °,19.28 ° ± 0.2 °,20.09 ° ± 0.2 °,20.77 ° ± 0.2 °,21.13 ° ± 0.2 ° ± 0.52 ° ± 0.2 °,21.76 ° ± 0.2 °,22.03 ° ± 0.2 °,22.83 ° ± 0.23.23 ° ± 0.2 ° ± 0.23 ° ± 0.2 °, 21.95 ° ± 0.25 ° ± 0.2 °,25 ° ± 0.2.29 ° ± 0.2 ° ± 0; or
The oxalate is oxalate crystal form A, and an X-ray powder diffraction pattern of the oxalate crystal form A has diffraction peaks at the following 2 theta angles: 6.37 ° ± 0.2 °,7.69 ° ± 0.2 °,9.55 ° ± 0.2 °,12.66 ° ± 0.2 °,12.96 ° ± 0.2 °,13.47 ° ± 0.2 °,14.88 ° ± 0.2 °,15.83 ° ± 0.2 °,16.01 ° ± 0.2 °,17.07 ° ± 0.2 °,17.96 ° ± 0.2 °,19.17 ° ± 0.2 °,20.40 ° ± 0.2 °,20.84 ° ± 0.2 °,21.28 ° ± 0.2 °,21.74 ° ± 0.2 °,22.49 ° ± 0.2 °,23.06 ° ± 0.2 °,24.45 ° ± 0.2 °,25.00 ° ± 0.2 °,25.52 ° ± 0.2 °,26.13 ° ± 0.2 °,28.47 ° ± 0.2.29.04 ° ± 0.2 ° 360.2 ° ± 0.31 ° ± 2.32 ° ± 52 ° ± 2.3.3 ° ± 52 ° ± 2 ° 3.2 °; or
The succinate salt is succinate salt crystal form A, and an X-ray powder diffraction pattern of the succinate salt crystal form A has diffraction peaks at the following 2 theta angles: 7.65 ° ± 0.2 °,8.88 ° ± 0.2 °,11.88 ° ± 0.2 °,13.40 ° ± 0.2 °,15.40 ° ± 0.2 °,16.39 ° ± 0.2 °,17.91 ° ± 0.2 °,18.53 ° ± 0.2 °,20.17 ° ± 0.2 °,20.92 ° ± 0.2 °,22.20 ° ± 0.2 °,23.27 ° ± 0.2 °,23.57 ° ± 0.2 °,24.48 ° ± 0.2 °,25.78 ° ± 0.2 °,27.05 ° ± 0.2 °,29.49 ° ± 0.2 °,30.15 ° ± 0.2 °,31.40 ° ± 0.2 °,33.09 ° ± 0.2 °,34.21 ° ± 0.2 °,36.37 ° ± 0.2 °,37.18 ° ± 0.2 °,37.81 ° ± 0.2 ° ± 38.69 ° ± 0.2 °; or
The salicylate is salicylate crystal form A, and an X-ray powder diffraction pattern of the salicylate crystal form A has diffraction peaks at the following 2 theta angles: 8.30 ° ± 0.2 °,9.61 ° ± 0.2 °,9.92 ° ± 0.2 °,12.24 ° ± 0.2 °,13.20 ° ± 0.2 °,15.60 ° ± 0.2 °,16.21 ° ± 0.2 °,16.45 ° ± 0.2 °,17.52 ° ± 0.2 °,17.71 ° ± 0.2 °,18.56 ° ± 0.2 °,19.23 ° ± 0.2 °,20.92 ° ± 0.2 °,21.78 ° ± 0.2 °,22.24 ° ± 0.2 °,22.86 ° ± 0.2 °,23.79 ° ± 0.2 °,24.45 ° ± 0.24 ° ± 0.69 ° ± 0.2 °,25.83 ° ± 0.2 °,26.60 ° ± 0.2 °,28.19 ° ± 0.2 °,29.10 ° ± 0.2.18 ° ± 0.2 °, 24.68 ° ± 0.68 ° ± 0.2 °, 3580 ° ± 0.31 ° ± 0.2 °.
6. The salt of any one of claims 2-5, wherein the p-toluenesulfonate salt is form A p-toluenesulfonate salt having an X-ray powder diffraction pattern substantially as shown in FIG. 1; or
The oxalate salt is oxalate form a having an X-ray powder diffraction pattern substantially as shown in figure 2; or
The succinate salt is succinate salt form a having an X-ray powder diffraction pattern substantially as shown in figure 3; or
The salicylate is salicylate form a having an X-ray powder diffraction pattern substantially as shown in figure 4.
7. The salt of any of claims 2-5, wherein the p-toluenesulfonate salt is form A p-toluenesulfonate salt, and wherein a differential scanning calorimetry trace of form A p-toluenesulfonate comprises an endothermic peak at 203.19 ℃ ± 3 ℃; or
The oxalate is oxalate form A, and a differential scanning calorimetry chart of the oxalate form A comprises an endothermic peak at 212.51 ℃ +/-3 ℃; or
The succinate salt is succinate salt form A, and a differential scanning calorimetry trace of succinate salt form A comprises an endothermic peak at 179.31 ℃ ± 3 ℃; or
The salicylate is salicylate form a, and a differential scanning calorimetry trace of salicylate form a comprises an endothermic peak at 221.92 ℃ ± 3 ℃.
8. The salt of claim 7, wherein the p-toluenesulfonate is form A p-toluenesulfonate having a differential scanning calorimetry pattern substantially as shown in FIG. 5; or
The oxalate salt is oxalate form a having a differential scanning calorimetry trace substantially as shown in figure 6; or
The succinate salt is succinate form a having a differential scanning calorimetry pattern substantially as shown in figure 7; or
The salicylate is salicylate form a having a differential scanning calorimetry pattern substantially as shown in figure 8.
9. A pharmaceutical composition comprising a salt of any one of claims 1-8, and a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, or combination thereof.
10. Use of a salt according to any one of claims 1 to 8 or a pharmaceutical composition according to claim 9 for the manufacture of a medicament for the prevention, treatment or alleviation of 5-HT6A receptor-associated disease;
wherein, the said and 5-HT6The receptor-related disease is a central nervous system disorder, a gastrointestinal disorder, or obesity;
wherein the central nervous system disorder is attention deficit hyperactivity disorder, anxiety, a stress-related disorder, schizophrenia, obsessive compulsive disorder, manic depression, a neurological disorder, a memory disorder, attention deficit disorder, Parkinson's disease, amyotrophic lateral sclerosis, Alzheimer's disease or Huntington's chorea.
CN202010181458.7A 2020-03-16 2020-03-16 Salts of heteroaromatic derivatives and use thereof Pending CN111302998A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010181458.7A CN111302998A (en) 2020-03-16 2020-03-16 Salts of heteroaromatic derivatives and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010181458.7A CN111302998A (en) 2020-03-16 2020-03-16 Salts of heteroaromatic derivatives and use thereof

Publications (1)

Publication Number Publication Date
CN111302998A true CN111302998A (en) 2020-06-19

Family

ID=71151177

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010181458.7A Pending CN111302998A (en) 2020-03-16 2020-03-16 Salts of heteroaromatic derivatives and use thereof

Country Status (1)

Country Link
CN (1) CN111302998A (en)

Similar Documents

Publication Publication Date Title
CN111233737B (en) Salts of heteroaromatic derivatives and use thereof
CN111233736B (en) Salts of heteroaromatic derivatives and use thereof
CN111362858B (en) Salts of heteroaromatic derivatives and use thereof
CN111362859B (en) Salts of heteroaromatic derivatives and use thereof
CN111302998A (en) Salts of heteroaromatic derivatives and use thereof
CN111233738A (en) Fumarate of aromatic heterocyclic derivative and application thereof
CN113861182B (en) Phosphate of pyrimidine thiazole carboxamide compound and crystal form thereof
CN112876478B (en) Crystalline forms of octahydropyrrolo [3,4-c ] pyrrole derivatives
CN112876480B (en) Crystalline forms of octahydropyrrolo [3,4-c ] pyrrole derivatives
CN112876479B (en) Crystalline forms of octahydropyrrolo [3,4-c ] pyrrole derivatives
CN114685444B (en) Salts of pyridyleiperidine derivatives and uses thereof
CN112876477B (en) Crystalline forms of octahydropyrrolo [3,4-c ] pyrrole derivatives
CN114685441B (en) Salts of pyridyleiperidine derivatives and uses thereof
CN112876481B (en) Crystalline forms of octahydropyrrolo [3,4-c ] pyrrole derivatives
CN114685443B (en) Salts of pyridyleiperidine derivatives and uses thereof
CN114685440B (en) Salts of pyridyleiperidine derivatives and uses thereof
US20230312551A1 (en) Salt of 2-(substituted pyrimidinyl) thiazole carboxamide compound, and composition and use thereof
CN112851642B (en) Salt of phenylpyrimidine piperazine compound and use thereof
CN114685445B (en) Salts of pyridyleiperidine derivatives and uses thereof
CN113861183B (en) Salts of substituted pyrimidine piperazine compounds and uses thereof
CN113861184B (en) Phosphate of 2- (substituted pyrimidinyl) thiazole carboxamide compound and use thereof
CN114989136A (en) Salts of indole derivatives and uses thereof
CN114835678A (en) Salts of indole derivatives and uses thereof
CN114835677A (en) Salts of indole derivatives and uses thereof
CN112876475A (en) Salts of octahydropyrrolo [3,4-c ] pyrrole derivatives

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20200619

WD01 Invention patent application deemed withdrawn after publication