CN106243052B - Substituted heterocyclic compounds, process for their preparation and their use - Google Patents

Substituted heterocyclic compounds, process for their preparation and their use Download PDF

Info

Publication number
CN106243052B
CN106243052B CN201610404146.1A CN201610404146A CN106243052B CN 106243052 B CN106243052 B CN 106243052B CN 201610404146 A CN201610404146 A CN 201610404146A CN 106243052 B CN106243052 B CN 106243052B
Authority
CN
China
Prior art keywords
compound
disorders
compounds
group
independently
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201610404146.1A
Other languages
Chinese (zh)
Other versions
CN106243052A (en
Inventor
金传飞
高金恒
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.)
Guangdong HEC Pharmaceutical
Original Assignee
Guangdong HEC Pharmaceutical
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 Guangdong HEC Pharmaceutical filed Critical Guangdong HEC Pharmaceutical
Publication of CN106243052A publication Critical patent/CN106243052A/en
Application granted granted Critical
Publication of CN106243052B publication Critical patent/CN106243052B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/041,2,3-Triazoles; Hydrogenated 1,2,3-triazoles
    • C07D249/061,2,3-Triazoles; Hydrogenated 1,2,3-triazoles with aryl radicals directly attached to ring atoms

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to a substituted heterocyclic compound and a preparation method and application thereof, and further relates to a pharmaceutical composition containing the heterocyclic compound and application thereof; the compounds or pharmaceutical compositions are useful for antagonizing orexin receptors. The invention also relates to processes for preparing such compounds and pharmaceutical compositions, and to their use in the manufacture of medicaments for the treatment or prophylaxis of neurological and psychiatric disorders and diseases of the central nervous system in mammals, particularly humans.

Description

Substituted heterocyclic compounds, process for their preparation and their use
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a compound and a composition for treating or preventing nervous and mental disorders and diseases of a central nervous system, a preparation method and application thereof. More specifically, the invention relates to heterocyclic compounds which are useful as orexin receptor antagonists.
Background
Orexin (orexin), also known as hypothalamin, orexin, includes orexin a and orexin B (or hypothalamin-1 and hypothalamin-2), which are neuroendocrine hormones secreted by the hypothalamus and which play a role in the central nervous system, with its main physiological role: 1. appetite regulation, orexin can significantly promote food intake and exhibit a dose-dependent response, and activates the neurons that regulate food intake; 2. participating in the regulation of energy metabolism, the orexin can obviously increase the metabolic rate; 3. participating in sleep-wake regulation, orexin can inhibit rapid eye movement sleep, prolong wake time, and block orexin effect to promote sleep; 4. participating in endocrine regulation, and the influence of the orexin on the endocrine of pituitary hormone is obvious; 5. associated with sense of reward, learning, and memory; 6. promoting gastric acid secretion; 7. promoting the increase of drinking water; 8. raising blood pressure; 9. play a role in The treatment of alcohol addiction, drug addiction, alcoholism, etc. (Piperet et al, The novel blue neuroepidetide, orexin-A, models The sleep-like cycle of rates. Eur. J. neuroscience,2000,12(2), 726-730).
Orexin exerts its biological effects by binding to orexin receptors, a G-protein coupled receptor, of two types, called OX1Receptors and OX2Receptors, which are distributed only in brain tissue, with a significant difference in their distribution, wherein OX1Receptors are mainly distributed in the hypothalamic ventral-medial nucleus, paraventricular nucleus, hippocampal structure, dorsal raphe nucleus and locus coeruleus, OX2Receptors are distributed predominantly in the cerebral cortex, hypothalamus, paraventricular nuclei and pre-optic nuclei (Sakurai T. et al, Orexins and dororexin receptors: a family of hypothalamic neuropeptides and G protein-copolymers that regulate the growth of the receptor cell 1998,92(4), 573-585).
Orexin is of great importance to mammals, especially humans, and is widely involved in various regulatory activities of the central nervous system, and has great significance in pathology. There are a large number of neurological and psychiatric disorders and diseases associated with orexin and orexin receptors, among which depression, anxiety, seasonal affective disorder, mania, bipolar disorder, obsessive compulsive disorder, jetlag-induced insomnia and fatigue, schizophrenia, convulsions, panic attacks, melancholia, alcohol addiction, drug addiction, alcoholism, substance abuse, drug addiction withdrawal symptoms, insomnia, psychotic disorders, epilepsy, sleep disorders, sleep disturbances, sleep apnea syndrome, compulsive eating disorders, fibromyalgia, stress, obesity, Parkinson's disease, senile dementia, cognitive disorders, memory disorders, premenstrual tension syndrome, migraine, memory loss, Alzheimer's disease or other various disorders associated with normal or aging, these diseases have severely affected the well-being of society and the quality of life of the patients and their families, and some may even lead to death.
In view of the above, orexin receptor antagonists have become a hot spot for the development of drugs against nervous and psychiatric disorders and diseases of the central nervous system.
The invention provides compounds with orexin receptor antagonism, which have better clinical application prospect. Compared with the existing similar compounds, the compound provided by the invention has better pharmacodynamic activity, and in addition, the compound provided by the invention also has excellent physicochemical property and pharmacokinetic property.
Summary of The Invention
The following is a summary of some aspects of the invention only and is not intended to be limiting. These aspects and others are described more fully below. All references in this specification are incorporated herein by reference in their entirety. When the disclosure of the present specification differs from the cited documents, the disclosure of the present specification controls.
The present invention provides a class of compounds having orexin receptor antagonist activity for the manufacture of medicaments for the prevention or treatment of neurological and psychiatric disorders and diseases of the central nervous system, such as depression, anxiety, mania, alcohol addiction, drug addiction, alcoholism, bipolar disorder, obsessive-compulsive disorder, stress, sleep disorders, jetlag-induced insomnia and fatigue and insomnia in humans. The invention also provides processes for the preparation of these compounds, pharmaceutical compositions containing these compounds and methods of using these compounds or compositions in the treatment of the above-mentioned diseases in mammals, especially humans.
Specifically, the method comprises the following steps:
in one aspect, the invention relates to a compound of formula (I) or a stereoisomer, a tautomer, a nitrogen oxide, a solvate, a metabolite, a pharmaceutically acceptable salt of a compound of formula (I), or a prodrug thereof,
Figure BDA0001012285000000021
wherein X is-NR5-、-C(=O)NR5-or-S (═ O)2NR5-;
Each R1Independently H, D, F, Cl, Br, I, OH, NH2、NO2、CN、N3、C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C1-4Haloalkyl, C1-4Alkoxy radical, C1-4Hydroxyalkyl radical, C1-4Alkylamino radical, C3-8Carbocyclyl, heterocyclyl consisting of 3 to 8 atoms, C6-10Aryl or heteroaryl of 5 to 6 atoms, wherein each R1Independently optionally substituted by 1,2,3,4 or 5R6Substituted by a group;
R2and R3Each independently is H, D, F, Cl, Br, I, OH, NH2、NO2、CN、N3、C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C1-4Alkoxy radical, C1-4Alkylamino radical, C1-4Hydroxyalkyl or C1-4A haloalkyl group;
each R4Independently H, D, F, Cl, Br, I, OH, NH2、NO2、CN、N3、C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C3-8Carbocyclyl, 3-8 atomsSub-group of heterocyclic radicals, C6-10Aryl, 5-6-membered heteroaryl, - (CR)7R7a)q-OR10、-(CR7R7a)q-NR8R9、-(CR7R7a)qS(=O)rR10、-(CR7R7a)qS(=O)2NR8R9、-(CR7R7a)qC(=O)R10、-(CR7R7a)qOC(=O)R10、-(CR7R7a)qC(=O)OR10、-(CR7R7a)q-N(R8)C(=O)R10、-C(=NR10)NR8R9、-N(R10)C(=O)NR8R9、-(CR7R7a)q-N(R8)S(=O)rR10Or- (CR)7R7a)qC(=O)NR8R9Wherein each R is4Independently optionally substituted by 1,2,3,4 or 5R6Substituted by a group;
R5is H, D, C1-4Alkyl radical, C1-4Haloalkyl, C3-8Carbocyclyl, heterocyclyl consisting of 3 to 6 atoms, C6-10Aryl or heteroaryl of 5 to 6 atoms;
each R6Independently H, D, F, Cl, Br, I, OH, NH2、NO2、CN、N3、C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C1-4Alkoxy radical, C1-4Alkylamino radical, C1-4Haloalkyl, C1-4Haloalkoxy, C1-4Hydroxyalkyl or C1-4A haloalkylamino group;
each R7And R7aIndependently H, D, F, Cl, Br, I, CN, C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C1-4Alkoxy radical, C1-4Alkylamino radical, C1-4Haloalkoxy, C1-4Haloalkylamino, C3-8Carbocyclic group, 3-8 atomsHeterocyclic group of (A), C6-10Aryl or heteroaryl of 5 to 6 atoms;
each R8、R9And R10Independently H, C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C3-8Carbocyclyl group, (C)3-6Cycloalkyl) - (C)1-4Alkylene) -, heterocyclic group consisting of 3 to 8 atoms, (heterocyclic group consisting of 3 to 6 atoms) - (C)1-4Alkylene) -, phenyl, (C)6-10Aryl group) - (C1-4Alkylene) -, heteroaryl of 5-6 atoms or heteroaryl of 5-6 atoms) - (C1-4Alkylene) -, or R8And R9Together with the nitrogen atom to which they are attached, form a heterocyclyl group of 3 to 6 atoms, wherein C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl or C3-8Carbocyclyl is independently optionally substituted with 1,2,3,4 or 5 substituents independently selected from D, F, Cl, Br, OH, NH2、CN、N3、C1-4Alkyl radical, C1-4Haloalkyl, C1-4Alkoxy or C1-4Substituted with a substituent of alkylamino;
m is 0,1, 2,3,4, 5, 6 or 7; p is 1,2,3 or 4;
r and k are each independently 0,1 or 2; and
each n and q is independently 0,1, 2,3 or 4.
In some embodiments, each R1Independently H, F, Cl, Br, OH, NH2、NO2CN, methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, vinyl, ethynyl, methoxy, ethoxy, methylamino, ethylamino, hydroxymethyl, hydroxyethyl, trifluoromethyl, cyclopropylalkyl, piperidinyl, piperazinyl, morpholinyl, phenyl, pyrrolyl, imidazolyl, thiazolyl, or thienyl.
In some embodiments, R2And R3Each independently of the other is H, F, Cl, OH, NH2、NO2CN, methyl, ethyl, propyl, isopropyl, vinyl, allyl, propargyl, methoxy, ethoxy, methylamino, ethylamino, hydroxymethyl, hydroxyethyl, or trifluoromethyl.
In some embodiments, each R4Independently F, Cl, OH, NH2、NO2CN, methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, vinyl, ethynyl, methoxy, ethoxy, methylamino, ethylamino, hydroxymethyl, hydroxyethyl, trifluoromethyl, cyclopropylalkyl, piperidinyl, piperazinyl, morpholinyl, phenyl, pyrrolyl, imidazolyl, thiazolyl, thienyl, -C (═ O) NH2or-COOH.
In another aspect, the invention relates to a pharmaceutical composition comprising a compound disclosed herein. In some embodiments, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle, or combination thereof.
In other embodiments, the pharmaceutical composition of the present invention further comprises other drugs for preventing or treating cns neurological and psychiatric disorders and diseases, said other drugs for preventing or treating cns neurological and psychiatric disorders and diseases being antidepressants, anxiolytics, lithium salts as affective stabilizers, antipsychotics, atypical antipsychotics, antiepileptics, anti-parkinson's disease drugs, sedative hypnotics, antihistamines, GABA receptor agonists and/or GABA reuptake inhibitors, drugs as monoamine oxidase inhibitors, drugs as melatonin receptor agonists and drugs as orexin receptor antagonists or any combination thereof.
In another aspect, the invention relates to the use of a compound or composition disclosed herein for the preparation of a medicament for the prevention, treatment or alleviation of central nervous system neurological and psychiatric disorders and diseases in a mammal, including a human.
In some embodiments, the cns neurological and psychiatric disorders and diseases are depression, anxiety, seasonal affective disorder, mania, bipolar disorder, obsessive-compulsive disorder, jetlag, insomnia and fatigue due to jet lag, schizophrenia, convulsions, panic attacks, melancholia, alcohol addiction, drug addiction, alcohol intoxication, substance abuse, drug addiction withdrawal symptoms, insomnia, psychotic disorders, epilepsy, sleep disorders, sleep disturbances, sleep apnea syndrome, compulsive eating disorders, fibromyalgia, stress responses, obesity, parkinson's disease, senile dementia, cognitive disorders, memory disorders, premenstrual tension syndrome, migraine, memory loss, alzheimer's disease or other various disorders associated with normal or pathological aging.
In another aspect, the invention relates to the use of a compound or composition disclosed herein for the preparation of a medicament for selectively antagonizing an orexin receptor.
In another aspect, the invention relates to methods for the preparation, isolation and purification of compounds encompassed by formula (I).
Biological test results show that the compound provided by the invention can be used as a better orexin receptor antagonist.
Any embodiment of any aspect of the invention may be combined with other embodiments, as long as they do not contradict. Furthermore, in any embodiment of any aspect of the invention, any feature may be applicable to that feature in other embodiments, so long as they do not contradict.
The foregoing merely summarizes certain aspects of the invention and is not intended to be limiting. These and other aspects will be more fully described below.
Detailed description of the invention
Definitions and general terms
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated by the accompanying structural and chemical formulas. The invention is intended to cover alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. Those skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated documents, patents, and similar materials differ or contradict this application (including but not limited to defined terminology, application of terminology, described techniques, and the like), this application controls.
It will be further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
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.
The following definitions as used herein should be applied unless otherwise indicated. For the purposes of the present invention, the chemical elements are in accordance with the CAS version of the periodic Table of the elements, and the handbook of chemistry and Physics, 75 th edition, 1994. In addition, general principles of Organic Chemistry can be found in the descriptions of "Organic Chemistry", Thomas Sorrell, University Science Books, Sausaltito: 1999, and "March's Advanced Organic Chemistry" by Michael B.Smith and Jerry March, John Wiley & Sons, New York:2007, the entire contents of which are incorporated herein by reference.
The articles "a," "an," and "the" as used herein are intended to include "at least one" or "one or more" unless otherwise indicated or clearly contradicted by context. Thus, as used herein, the articles refer to articles of one or more than one (i.e., at least one) object. For example, "a component" refers to one or more components, i.e., there may be more than one component contemplated for use or use in embodiments of the described embodiments.
The term "subject" as used herein refers to an animal. Typically the animal is a mammal. Subjects, e.g., also primates (e.g., humans, males or females), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds, etc. In certain embodiments, the subject is a primate. In other embodiments, the subject is a human.
The term "patient" as used herein refers to humans (including adults and children) or other animals. In some embodiments, "patient" refers to a human.
The term "comprising" is open-ended, i.e. includes the elements indicated in the present invention, but does not exclude other elements.
"stereoisomers" refers to compounds having the same chemical structure but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric isomers (cis/trans), atropisomers, and the like.
"chiral" is a molecule having the property of not overlapping its mirror image; and "achiral" refers to a molecule that can overlap with its mirror image.
"enantiomer" refers to two isomers of a compound that are not overlapping but are in mirror image relationship to each other.
"diastereomer" refers to a stereoisomer having two or more chiral centers and whose molecules are not mirror images of each other. Diastereomers have different physical properties, such as melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may be separated by high resolution analytical procedures such as electrophoresis and chromatography, e.g., HPLC.
The stereochemical definitions and rules used in the present invention generally follow the general definitions of S.P. Parker, Ed., McGraw-Hilldictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; andEliel, E.and Wilen, S., "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994.
Many organic compounds exist in an optically active form, i.e., they have the ability to rotate the plane of plane polarized light. In describing optically active compounds, the prefixes D and L or R and S are used to denote the absolute configuration of a molecule with respect to one or more of its chiral centers. The prefixes d and l or (+) and (-) are the symbols used to specify the rotation of plane polarized light by the compound, where (-) or l indicates that the compound is left-handed. Compounds prefixed with (+) or d are dextrorotatory. A particular stereoisomer is an enantiomer and a mixture of such isomers is referred to as an enantiomeric mixture. A50: 50 mixture of enantiomers is referred to as a racemic mixture or racemate, which may occur when there is no stereoselectivity or stereospecificity in the chemical reaction or process.
Any asymmetric atom (e.g., carbon, etc.) of a compound disclosed herein can exist in racemic or enantiomerically enriched forms, such as the (R) -, (S) -or (R, S) -configuration. In certain embodiments, each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R) -or (S) -configuration.
Depending on the choice of starting materials and methods, the compounds of the invention may exist as one of the possible isomers or as mixtures thereof, for example as racemates and mixtures of non-corresponding isomers (depending on the number of asymmetric carbon atoms). Optically active (R) -or (S) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituents may be in the E or Z configuration; if the compound contains a disubstituted cycloalkyl group, the substituents of the cycloalkyl group may have cis or trans configuration.
Any resulting mixture of stereoisomers may be separated into pure or substantially pure geometric isomers, enantiomers, diastereomers, depending on differences in the physicochemical properties of the components, for example, by chromatography and/or fractional crystallization.
The racemates of any of the resulting end products or intermediates can be resolved into the optical enantiomers by known methods using methods familiar to those skilled in the art, e.g., by separation of the diastereomeric salts obtained. The racemic product can also be separated by chiral chromatography, e.g., using a chiral adsorbentHigh Performance Liquid Chromatography (HPLC). In particular, Enantiomers can be prepared by asymmetric synthesis, for example, see Jacques, et al, Enantiomers, racemases and solutions (Wiley Interscience, New York, 1981); principles of Asymmetric Synthesis (2)ndEd.Robert E.Gawley,JeffreyAubé,Elsevier,Oxford,UK,2012);Eliel,E.L.Stereochemistry of Carbon Compounds(McGraw-Hill,NY,1962);Wilen,S.H.Tablesof Resolving Agents and Optical Resolutions p.268(E.L.Eliel,Ed.,Univ.of NotreDame Press,Notre Dame,IN 1972);Chiral Separation Techniques:A PracticalApproach(Subramanian,G.Ed.,Wiley-VCH Verlag GmbH&Co.KGaA,Weinheim,Germany,2007)。
The term "tautomer" or "tautomeric form" refers to structural isomers having different energies that can interconvert by a low energy barrier (lowenergy barrier). If tautomerism is possible (e.g., in solution), then the chemical equilibrium of the tautomer can be reached. For example, proton tautomers (also known as proton transfer tautomers) include interconversions by proton migration, such as keto-enol isomerization and imine-enamine isomerization. Valence tautomers (valenctautomers) include interconversion by recombination of some of the bonding electrons. A specific example of keto-enol tautomerism is the tautomerism of the pentan-2, 4-dione and 4-hydroxypent-3-en-2-one tautomers. Another example of tautomerism is phenol-ketone tautomerism. One specific example of phenol-ketone tautomerism is the tautomerism of pyridin-4-ol and pyridin-4 (1H) -one tautomers. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
The compounds of the invention may be optionally substituted with one or more substituents, as described herein, in compounds of the general formula above, or as specifically exemplified, sub-classes, and classes of compounds encompassed by the invention.
In general, the term "substituted" means that one or more hydrogen atoms in a given structure are replaced with a particular substituent. Unless otherwise indicated, a substituted group may have one substituent substituted at each substitutable position of the group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, the substituents may be substituted at each position, identically or differently.
The term "unsubstituted" means that the specified group bears no substituents.
The term "optionally substituted with … …" is used interchangeably with the term "unsubstituted or substituted with … …", i.e., the structure is unsubstituted or substituted with one or more substituents described herein. Substituents described herein include, but are not limited to, D, F, Cl, Br, I, N3、CN、NO2、OH、SH、NH2Alkyl, haloalkyl, alkenyl, alkynyl, alkoxy, alkylamino, cycloalkyl, heterocyclyl, aryl, heteroaryl, - (CR)7R7a)q-OR10、-(CR7R7a)q-NR8R9、-(CR7R7a)qS(=O)rR10、-(CR7R7a)qS(=O)2NR8R9、-(CR7R7a)qC(=O)R10、-(CR7R7a)qOC(=O)R10、-(CR7R7a)qC(=O)OR10、-(CR7R7a)q-N(R8)C(=O)R10、-C(=NR10)NR8R9、-N(R10)C(=O)NR8R9、-(CR7R7a)q-N(R8)S(=O)rR10、-(CR7R7a)qC(=O)NR8R9And so on. Wherein R is7、R7a、R8、R9、R10Q and r have the meanings given in the description of the invention.
In addition, unless otherwise explicitly indicated, the descriptions of the terms "… independently" and "… independently" and "… independently" used in the present invention are interchangeable and should be understood in a broad sense to mean that the specific items expressed between the same symbols do not affect each other in different groups or that the specific items expressed between the same symbols in the same groups do not affect each other.
In the various parts of this specification, substituents of the disclosed compounds are disclosed in terms of group type or range. It is specifically intended that the invention includes each and every independent subcombination of the various members of these groups and ranges. For example, the term "C1-6Alkyl "means in particular independently disclosed methyl, ethyl, C3Alkyl radical, C4Alkyl radical, C5Alkyl and C6An alkyl group.
In each of the parts of the invention, linking substituents are described. Where the structure clearly requires a linking group, the markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for the variable recites "alkyl" or "aryl," it is understood that the "alkyl" or "aryl" represents an attached alkylene group or arylene group, respectively.
The term "alkyl" or "alkyl group" as used herein, denotes a saturated straight or branched chain monovalent hydrocarbon radical, wherein the alkyl group may be optionally substituted with one or more substituents as described herein. Unless otherwise specified, alkyl groups contain 1-20 carbon atoms. In one embodiment, the alkyl group contains 1 to 12 carbon atoms; in another embodiment, the alkyl group contains 3 to 12 carbon atoms; in another embodiment, the alkyl group contains 1 to 6 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 4 carbon atoms. Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, n-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2, 3-dimethyl-2-butyl, 3, 3-dimethyl-2-butyl, n-heptyl, n-octyl, and the like.
The term "alkylene" refers to a saturated divalent hydrocarbon radical resulting from the removal of two hydrogen atoms from a saturated straight or branched chain hydrocarbon radical. Unless otherwise specified, the alkylene group contains 1 to 12 carbon atoms. In one embodiment, the alkylene group contains 1 to 6 carbon atoms; in another embodiment, the alkylene group contains 1 to 4 carbon atoms; in yet another embodiment, the alkylene group contains 1 to 3 carbon atoms; in yet another embodiment, the alkylene group contains 1 to 2 carbon atoms. Examples of such include methylene, ethylene, isopropylene and the like.
The term "alkenyl" denotes a straight or branched chain monovalent hydrocarbon radical having at least one carbon-carbon sp2A double bond, wherein the alkenyl group may be optionally substituted with one or more substituents described herein, including the positioning of "cis" and "trans", or the positioning of "E" and "Z". In one embodiment, the alkenyl group contains 2 to 12 carbon atoms; in another embodiment, the alkenyl group contains 2 to 6 carbon atoms; in yet another embodiment, the alkenyl group contains 2 to 4 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl, allyl, and the like.
The term "alkynyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one carbon-carbon sp triple bond, wherein the alkynyl radical may be optionally substituted with one or more substituents as described herein. In one embodiment, alkynyl groups contain 2-12 carbon atoms; in another embodiment, alkynyl groups contain 2-6 carbon atoms; in yet another embodiment, alkynyl groups contain 2-4 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl, propargyl, 1-propynyl, and the like.
The term "alkoxy" means an alkyl group attached to the rest of the molecule through an oxygen atom, wherein the alkyl group has the meaning as described herein. Unless otherwise specified, the alkoxy group contains 1 to 12 carbon atoms. In one embodiment, the alkoxy group contains 1 to 6 carbon atoms; in another embodiment, the alkoxy group contains 1 to 4 carbon atoms; in yet another embodiment, the alkoxy group contains 1 to 3 carbon atoms. The alkoxy group may be optionally substituted with one or more substituents described herein. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, 1-propoxy, 2-propoxy, 1-butoxy, 2-methyl-l-propoxy, 2-butoxy, 2-methyl-2-propoxy, and the like.
The term "alkylamino" includes "N-alkylamino" and "N, N-dialkylamino" in which the amino groups are each independently substituted with one or two alkyl groups; the alkyl group has the meaning described in the present invention. In some of these embodiments, the alkylamino group is one or two C1-6The alkyl group is attached to a lower alkylamino group formed on the nitrogen atom. In other embodiments, the alkylamino group is one or two C1-4To the nitrogen atom to form an alkylamino group. Suitable alkylamino groups can be monoalkylamino or dialkylamino, and such examples include, but are not limited to, N-methylamino, N-ethylamino, N-dimethylamino, N-diethylamino, and the like.
The term "hydroxyalkyl" denotes an alkyl group substituted with one or more hydroxyl groups, wherein the alkyl group has the meaning as described herein; examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2-hydroxy-1-propyl, 3-hydroxy-1-propyl, 2, 3-dihydroxypropyl, and the like.
The term "aminoalkyl" denotes an alkyl group substituted by one or more amino groups, wherein the alkyl group has the meaning as described herein. Examples include, but are not limited to, aminomethyl, 2-aminoethyl, 3-amino-1-propyl, 4-amino-1-butyl, and the like.
The terms "haloalkyl", "haloalkenyl", "haloalkoxy" or "haloalkylamino" denote alkyl, alkenyl, alkoxy or alkylamino groups substituted with one or more halogen atoms, wherein the alkyl, alkenyl, alkoxy or alkylamino groups have the meaning as described herein, examples of which include, but are not limited to, trifluoromethyl, 2,3, 3-tetrafluoropropyl, trifluoromethoxy, trifluoromethylamino and the like.
The term "cycloalkyl" denotes a monovalent or polyvalent saturated monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 ring carbon atoms. In one embodiment, cycloalkyl groups contain from 7 to 12 ring carbon atoms; in yet another embodiment, cycloalkyl contains 3 to 8 ring carbon atoms; in yet another embodiment, cycloalkyl contains 3 to 6 ring carbon atoms. The cycloalkyl groups may be independently unsubstituted or substituted with one or more substituents described herein.
The term "carbocyclyl" or "carbocycle" denotes a monovalent or multivalent, saturated or partially unsaturated monocyclic, bicyclic or tricyclic ring system containing 3 to 12 ring carbon atoms and not being aromatic. Carbobicyclic groups include spirocarbocyclic and fused carbocyclic groups, and suitable carbocyclic groups include, but are not limited to, cycloalkyl, cycloalkenyl and cycloalkynyl groups. In one embodiment, carbocyclyl contains 3 to 8 ring carbon atoms; in yet another embodiment, carbocyclyl contains 3 to 6 ring carbon atoms. Examples of carbocyclyl groups further include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopentyl-1-alkenyl, 1-cyclopentyl-2-alkenyl, 1-cyclopentyl-3-alkenyl, cyclohexyl, 1-cyclohexyl-1-alkenyl, 1-cyclohexyl-2-alkenyl, 1-cyclohexyl-3-alkenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like. The carbocyclyl groups may independently be unsubstituted or substituted with one or more substituents described herein.
The terms "heterocyclyl" and "heterocycle" are used interchangeably herein and refer to a saturated or partially unsaturated, non-aromatic, monovalent or polyvalent, monocyclic, bicyclic, or tricyclic ring containing from 3 to 12 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur, and oxygen atoms. Unless otherwise specified, heterocyclyl may be carbon-or nitrogen-based, and-CH2-the group may optionally be replaced by-C (═ O) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxygen compound. Examples of heterocyclic groups include, but are not limited toLimited to: oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuryl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiocyclopentyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiaxanyl, homopiperazinyl, homopiperidinyl, diazepanyl, oxepanyl, thiepanyl, oxazepanyl, thiazepanyl, oxazepanyl, and pyrrolidinyl
Figure BDA0001012285000000071
Radical diaza
Figure BDA0001012285000000072
Radical, sulfur nitrogen hetero
Figure BDA0001012285000000073
Radicals, indolinyl, 1,2,3, 4-tetrahydroisoquinolinyl, 1, 3-benzodioxolyl, 2-oxa-5-azabicyclo [2.2.1]Hept-5-yl. In heterocyclic radicals of-CH2Examples of-groups substituted by-C (═ O) -include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidinonyl, 3, 5-dioxopiperidinyl and pyrimidinedione. Examples of the sulfur atom in the heterocyclic group being oxidized include, but are not limited to, sulfolane group, 1-dioxothiomorpholinyl group. The heterocyclyl group may be optionally substituted with one or more substituents as described herein.
The term "heterocycloalkyl" refers to a monovalent or polyvalent saturated monocyclic, bicyclic, or tricyclic ring system containing 3 to 12 ring atoms, wherein at least one ring atom is selected from nitrogen, sulfur, or oxygen atoms.
The term "Z-atom constituent" in "Z-atom constituent heterocyclyl" and "Z-atom constituent heteroaryl" wherein Z is an integer typically describes the number of ring-forming atoms in a molecule in which the number of ring-forming atoms is Z. For example, piperidinyl is a heterocycloalkyl group of 6 atoms, and decahydronaphthyl is a carbocyclyl group of 10 atoms.
The term "unsaturated" as used herein means that the group contains one or more unsaturations.
The term "heteroatom" refers to O, S, N, P and Si, including N, S and any oxidation state form of P; primary, secondary, tertiary amines and quaternary ammonium salt forms; or a form in which a hydrogen on a nitrogen atom in the heterocycle is substituted, for example, N (like N in 3, 4-dihydro-2H-pyrrolyl), NH (like NH in pyrrolidinyl) or NR (like NR in N-substituted pyrrolidinyl).
The term "halogen" refers to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I).
The term "aryl" denotes a monocyclic, bicyclic or tricyclic carbocyclic ring system containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein each ring is a ring of 3 to 7 atoms and at least one ring is aromatic; the aryl group has one or more attachment points to the rest of the molecule. The term "aryl" may be used interchangeably with the term "aromatic ring". In one embodiment, aryl is a carbocyclic ring system consisting of 6 to 10 ring atoms. Examples of the aryl group may include phenyl, naphthyl and anthracenyl. The aryl group may independently be optionally substituted with one or more substituents described herein.
The term "heteroaryl" denotes a monocyclic, bicyclic or tricyclic ring containing 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 6 ring atoms, wherein at least one ring is aromatic and at least one ring contains one or more heteroatoms; each ring system in the heteroaryl group comprises a ring of 5 to 7 atoms with one or more attachment points to the rest of the molecule. The term "heteroaryl" may be used interchangeably with the terms "heteroaromatic ring" or "heteroaromatic compound". The heteroaryl group is optionally substituted with one or more substituents described herein. In one embodiment, heteroaryl is a 5-12 atom heteroaryl comprising 1,2,3, or 4 heteroatoms independently selected from O, S and N; in another embodiment, heteroaryl is 5-6 atom consisting of 1,2,3, or 4 heteroatoms independently selected from O, S and N.
Examples of heteroaryl groups include, but are not limited to, 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, isothiazolyl, tetrazolyl (e.g., 2H-1,2,3, 4-tetrazolyl, 1H-1,2,3, 4-tetrazolyl), triazolyl (e.g., 2H-1,2, 3-triazolyl, 1H-1,2, 4-triazolyl, 4H-1,2, 4-triazolyl), 2-thienyl, 3-thienyl, 1H-pyrazolyl (e.g., 1H-pyrazol-3-yl), 1H-pyrazol-4-yl, 1H-pyrazol-5-yl), N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl; the following bicyclic rings are also included, but are in no way limited to these: benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl), purinyl, quinolyl (e.g., 2-quinolyl, 3-quinolyl, 4-quinolyl), imidazo [1,2-a ] pyridyl, pyrazolo [1,5-a ] pyrimidyl, and the like.
The term "carboxy", whether used alone or in combination with other terms, such as "carboxy", denotes-CO2H; the term "carbonyl", whether used alone or in combination with other terms, such as "aminocarbonyl" or "acyloxy", denotes- (C ═ O) -.
As described herein, the ring system formed by a substituent drawing a bond to the central ring (as shown in formulas b and e) represents that the substituent may be substituted at any substitutable position on the ring. For example, formula e represents ring a or ring B, and any possible substituted position may be substituted by R, as shown in formulas f 1-f 7; the substituent R represented by the formula b can be mono-substituted or poly-substituted at any possible substituted position on the C ring, as shown in formulas C1-C17.
Figure BDA0001012285000000081
As described herein, the ring system formed by a linker attached to the center of the ring (as shown in formula d) represents that the linker may be attached to the rest of the molecule at any available position on the ring system. Formula D represents that any possible attachment position on the D ring can be attached to the rest of the molecule.
Figure BDA0001012285000000091
The radical-C (═ O) NR, as described in the present invention5"two attachment sites are available for attachment to the rest of the molecule, and the attachment of the two attachment sites may be interchanged. For example, the group of formula a may be attached to the remainder of the molecule (e.g., the phenylene group of the structure of formula (I) of the present invention) via the E-terminus or the E' -terminus.
Figure BDA0001012285000000092
The term "protecting group" or "PG" refers to a substituent that, when reacted with other functional groups, is generally used to block or protect a particular functionality. For example, "amino protecting group" means a substituent attached to an amino group to block or protect the functionality of the amino group in a compound, and suitable amino protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC ), benzyloxycarbonyl (CBZ ) and 9-fluorenylmethyleneoxycarbonyl (Fmoc). Similarly, "hydroxyl protecting group" refers to the functionality of a substituent of a hydroxyl group to block or protect the hydroxyl group, and suitable protecting groups include acetyl and silyl groups. "carboxy protecting group" refers to the functionality of a substituent of a carboxy group to block or protect the carboxy group, and typical carboxy protecting groups include-CH2CH2SO2Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrobenzenesulfonyl) ethyl, 2- (diphenylphosphino) ethyl, nitroethyl, and the like. General descriptions of protecting groups can be found in the literature: greene, Protective Groups in Organic Synthesis, John Wiley&Sons,New York,1991;and P.J.Kocienski,Protecting Groups,Thieme,Stuttgart,2005.
The term "prodrug", as used herein, represents oneThe compounds are converted in vivo to compounds of formula (I). Such conversion is effected by hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue. The prodrug compound of the invention can be ester, and in the prior invention, the ester can be used as the prodrug and comprises phenyl ester and aliphatic (C)1-C24) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, a compound of the present invention contains a hydroxy group, i.e., it can be acylated to provide the compound in prodrug form. Other prodrug forms include phosphate esters, such as those obtained by phosphorylation of a hydroxyl group on the parent. For a complete discussion of prodrugs, reference may be made to the following: T.Higuchi and V.Stella, Pro-drugs as Novel delivery systems, Vol.14 of the A.C.S.Sympossium Series, Edward B.Roche, ed., Bioredeployers in Drug designs, American Pharmaceutical Association and PergammonPress, 1987, J.Rautio et al, Prodrugs in Design and Clinical Applications, Nature Review Drug Discovery,2008,7,255 and 270, and S.J.Herer et al, Prodrugs of pharmaceuticals and pharmaceuticals, Journal of chemical Chemistry,2008,51,2328 and 2345.
"metabolite" refers to the product of a particular compound or salt thereof obtained by metabolism in vivo. Metabolites of a compound can be identified by techniques well known in the art, and its activity can be characterized by assay methods as described herein. Such products may be obtained by administering the compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage, and the like. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting a compound of the present invention with a mammal for a sufficient period of time.
As used herein, "pharmaceutically acceptable salts" refer to organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable salts are well known in the art, as are: berge et al, descriptive acceptable salts in detail in J. pharmaceutical Sciences,1977,66:1-19. Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, inorganic acid salts such as hydrochloride, hydrobromide, phosphate, sulfate, perchlorate, and organic acid salts such as acetate, oxalate, maleate, tartrate, citrate, succinate, malonate, or salts obtained by other methods described in the literature above, such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, cyclopentylpropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, malates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, palmitates, pamoates, pectinates, persulfates, 3-phenylpropionates, picrates, pivalates, propionates, stearates, thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like. Salts obtained with appropriate bases include alkali metals, alkaline earth metals, ammonium and N+(C1-4Alkyl radical)4A salt. The present invention also contemplates quaternary ammonium salts formed from compounds containing groups of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C1-8Sulfonates and aromatic sulfonates.
"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that is water.
When the solvent is water, the term "hydrate" may be used. In some embodiments, a molecule of a compound of the present invention may be associated with a molecule of water, such as a monohydrate; in other embodiments, one molecule of the compound of the present invention may be associated with more than one molecule of water, such as a dihydrate, and in still other embodiments, one molecule of the compound of the present invention may be associated with less than one molecule of water, such as a hemihydrate. It should be noted that the hydrates of the present invention retain the biological effectiveness of the compound in its non-hydrated form.
The term "treating" any disease or condition, as used herein, means all that can slow, halt, arrest, control or halt the progression of the disease or condition, but does not necessarily mean that all the symptoms of the disease or condition have disappeared, and also includes prophylactic treatment of the symptoms, particularly in patients susceptible to such disease or disorder. In some of these embodiments, refers to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one clinical symptom thereof). In other embodiments, "treating" or "treatment" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" or "treatment" refers to modulating the disease or disorder, either physically (e.g., stabilizing a perceptible symptom) or physiologically (e.g., stabilizing a parameter of the body), or both. In other embodiments, "treating" or "treatment" refers to preventing or delaying the onset, occurrence, or worsening of a disease or disorder.
The term "therapeutically effective amount" or "therapeutically effective dose" as used herein refers to an amount of a compound of the invention that is capable of eliciting a biological or medical response (e.g., reducing or inhibiting enzyme or protein activity, or ameliorating symptoms, alleviating a disorder, slowing or delaying the progression of a disease, or preventing a disease, etc.) in a subject. In one non-limiting embodiment, the term "therapeutically effective amount" refers to an amount that, when administered to a subject, is effective for: (1) at least partially alleviating, inhibiting, preventing and/or ameliorating a condition or disease (i) mediated by orexin, or (ii) associated with orexin receptor activity, or (iii) characterized by abnormal activity of orexin receptors; or (2) reduces or inhibits orexin receptor activity; or (3) reduces or inhibits orexin receptor expression. In another embodiment, the term "therapeutically effective amount" means an amount that, when administered to a cell, or organ, or non-cellular biological substance, or vehicle, at least partially reduces or inhibits orexin receptor activity; or an amount of a compound of the invention effective to at least partially reduce or inhibit orexin receptor expression.
The terms "administration" and "administering" of a compound as used herein shall be understood as providing a compound of the invention or a prodrug of a compound of the invention to a subject in need thereof. It will be appreciated that one skilled in the art may have an effect on neurological and psychiatric disorders by treating a patient currently suffering from such a disorder, or prophylactically treating a patient suffering from such a disorder, with an effective amount of a compound of the invention.
The term "composition" as used herein refers to a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. The meaning of such terms in relation to pharmaceutical compositions includes products comprising the active ingredient(s) and the inert ingredient(s) that make up the carrier, as well as any product which results, directly or indirectly, from mixing, complexation or aggregation of any two or more of the ingredients, or from decomposition of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention include any composition prepared by admixing a compound of the present invention and a pharmaceutically acceptable carrier.
Description of the Compounds of the invention
The present invention discloses a class of heterocyclic compounds, pharmaceutically acceptable salts thereof, pharmaceutical formulations and compositions thereof, which are potentially useful as orexin receptor antagonists for the treatment of neurological and psychiatric disorders and diseases of the central nervous system in humans, such as depression, anxiety, alcohol addiction, drug addiction, alcohol intoxication, mania, bipolar disorder, obsessive-compulsive disorder, stress, sleep disorders, jetlag-induced insomnia and fatigue and insomnia.
In one aspect, the invention relates to a compound of formula (I) or a stereoisomer, a tautomer, a nitrogen oxide, a solvate, a metabolite, a pharmaceutically acceptable salt of a compound of formula (I), or a prodrug thereof,
Figure BDA0001012285000000111
wherein R is1,m,n,p,k,R4,R6,R2And R3Have the meaning as described in the present invention.
In some embodiments, X is-NR5-、-C(=O)NR5-or-S (═ O)2NR5-; wherein R is5Have the meaning as described in the present invention.
In some embodiments, each R1Independently H, D, F, Cl, Br, I, OH, NH2、NO2、CN、N3、C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C1-6Haloalkyl, C1-6Alkoxy radical, C1-6Hydroxyalkyl radical, C1-6Alkylamino radical, C3-12Carbocyclyl, heterocyclyl consisting of 3 to 12 atoms, C6-10Aryl or heteroaryl of 5 to 12 atoms, wherein each R1Independently optionally substituted by 1,2,3,4 or 5R6Substituted by a group; wherein each R is6Have the meaning as described in the present invention.
In some embodiments, R2And R3Each independently is H, D, F, Cl, Br, I, OH, NH2、NO2、CN、N3、C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C1-4Alkoxy radical, C1-4Alkylamino radical, C1-4Hydroxyalkyl or C1-4A haloalkyl group.
In some embodiments, each R4Independently H, D, F, Cl, Br, I, OH, NH2、NO2、CN、N3、C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-12Carbocyclyl, heterocyclyl consisting of 3 to 12 atoms, C6-10Aryl, 5-12-membered heteroaryl, - (CR)7R7a)q-OR10、-(CR7R7a)q-NR8R9、-(CR7R7a)qS(=O)rR10、-(CR7R7a)qS(=O)2NR8R9、-(CR7R7a)qC(=O)R10、-(CR7R7a)qOC(=O)R10、-(CR7R7a)qC(=O)OR10、-(CR7R7a)q-N(R8)C(=O)R10、-C(=NR10)NR8R9、-N(R10)C(=O)NR8R9、-(CR7R7a)q-N(R8)S(=O)rR10Or- (CR)7R7a)qC(=O)NR8R9Wherein each R is4Independently optionally substituted by 1,2,3,4 or 5R6Substituted by a group; wherein each R is6,R7,q,r,R7a,R8,R9And R10Have the meaning as described in the present invention.
In some embodiments, R5Is H, D, C1-4Alkyl radical, C1-4Haloalkyl, C3-8Carbocyclyl, heterocyclyl consisting of 3 to 6 atoms, C6-10Aryl or heteroaryl of 5 to 6 atoms.
In some embodiments, each R6Independently H, D, F, Cl, Br, I, OH, NH2、NO2、CN、N3、C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C1-4Alkoxy radical, C1-4Alkylamino radical, C1-4Haloalkyl, C1-4Haloalkoxy, C1-4Hydroxyalkyl or C1-4A haloalkylamino group.
In some embodiments, each R7And R7aIndependently H, D, F, Cl, Br, I, CN, C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C1-4Alkoxy radical, C1-4Alkylamino radical, C1-4Haloalkoxy, C1-4Haloalkylamino, C3-8Carbocyclyl, heterocyclyl consisting of 3 to 8 atoms, C6-10Aryl or heteroaryl of 5 to 6 atoms.
In some embodiments, each R8、R9And R10Independently H, C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C3-8Carbocyclyl group, (C)3-6Cycloalkyl) - (C)1-4Alkylene) -, heterocyclic group consisting of 3 to 8 atoms, (heterocyclic group consisting of 3 to 6 atoms) - (C)1-4Alkylene) -, phenyl, (C)6-10Aryl group) - (C1-4Alkylene) -, heteroaryl of 5-6 atoms or heteroaryl of 5-6 atoms) - (C1-4Alkylene) -, or R8And R9Together with the nitrogen atom to which they are attached, form a heterocyclyl group of 3 to 6 atoms, wherein C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl or C3-8Carbocyclyl is independently optionally substituted with 1,2,3,4 or 5 substituents independently selected from D, F, Cl, Br, OH, NH2、CN、N3、C1-4Alkyl radical, C1-4Haloalkyl, C1-4Alkoxy or C1-4Substituted by a substituent of alkylamino.
In some embodiments, m is 0,1, 2,3,4, 5, 6, or 7.
In some embodiments, p is 1,2,3, or 4.
In some embodiments, r is 0,1, or 2.
In some embodiments, k is 0,1, or 2.
In some embodiments, each n and q is independently 0,1, 2,3, or 4.
In some embodiments, each R1Independently H, D, F, Cl, Br, I, OH, NH2、NO2、CN、N3、C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C1-4Haloalkyl, C1-4Alkoxy radical, C1-4Hydroxyalkyl radical, C1-4Alkylamino radical, C3-8Carbocyclyl, heterocyclyl consisting of 3 to 8 atoms, C6-10Aryl or heteroaryl of 5 to 6 atoms, wherein each R1Independently optionally substituted by 1,2,3,4 or 5R6Substituted by a group; wherein each R is6Have the meaning as described in the present invention.
In some embodiments, each R1Independently H, F, Cl, Br, OH, NH2、NO2CN, methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, vinyl, ethynyl, methoxy, ethoxy, methylamino, ethylamino, hydroxymethyl, hydroxyethyl, trifluoromethyl, cyclopropylalkyl, piperidinyl, piperazinyl, morpholinyl, phenyl, pyrrolyl, imidazolyl, thiazolyl, or thienyl.
In some embodiments, R2And R3Each independently of the other is H, F, Cl, OH, NH2、NO2CN, methyl, ethyl, propyl, isopropyl, vinyl, allyl, propargyl, methoxy, ethoxy, methylamino, ethylamino, hydroxymethyl, hydroxyethyl, or trifluoromethyl.
In some embodiments, each R4Independently D, F, Cl, Br, I, OH, NH2、NO2、CN、N3、C1-4Alkyl radical, C2-4Alkenyl radical, C2-4Alkynyl, C3-8Carbocyclyl, heterocyclyl consisting of 3 to 8 atoms, C6-10Aryl, 5-6-membered heteroaryl, - (CR)7R7a)q-OR10、-(CR7R7a)q-NR8R9、-(CR7R7a)qS(=O)rR10、-(CR7R7a)qS(=O)2NR8R9、-(CR7R7a)qC(=O)R10、-(CR7R7a)qOC(=O)R10、-(CR7R7a)qC(=O)OR10、-(CR7R7a)q-N(R8)C(=O)R10、-C(=NR10)NR8R9、-N(R10)C(=O)NR8R9、-(CR7R7a)q-N(R8)S(=O)rR10Or- (CR)7R7a)qC(=O)NR8R9Wherein each R is4Independently optionally substituted by 1,2,3,4 or 5R6Substituted by a group; each R6,R7,q,r,R7a,R8,R9And R10Have the meaning as described in the present invention.
In some embodiments, each R4Independently F, Cl, OH, NH2、NO2CN, methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, vinyl, ethynyl, methoxy, ethoxy, methylamino, ethylamino, hydroxymethyl, hydroxyethyl, trifluoromethyl, cyclopropylalkyl, piperidinyl, piperazinyl, morpholinyl, phenyl, pyrrolyl, imidazolyl, thiazolyl, thienyl, -C (═ O) NH2or-COOH.
In some embodiments, the present invention relates to a compound, or a stereoisomer, tautomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof, of one of the following, but in no way limited to these compounds:
Figure BDA0001012285000000121
Figure BDA0001012285000000131
stereoisomers, tautomers, solvates, metabolites, salts and pharmaceutically acceptable prodrugs of the compounds of formula (I) are included within the scope of the present invention unless otherwise indicated.
The compounds of the present disclosure may contain asymmetric or chiral centers and thus may exist in different stereoisomeric forms. The present invention contemplates that all stereoisomeric forms of the compounds of formula (I), including but not limited to diastereomers, enantiomers, atropisomers and geometric (or conformational) isomers, and mixtures thereof, such as racemic mixtures, are integral to the invention.
In the structures disclosed herein, when the stereochemistry of any particular chiral atom is not specified, then all stereoisomers of that structure are contemplated as within this invention and are included as disclosed compounds in this invention. When stereochemistry is indicated by a solid wedge (solid wedge) or dashed line representing a particular configuration, then the stereoisomers of the structure are so well-defined and defined.
The compounds of formula (I) may exist in different tautomeric forms and all such tautomers are included within the scope of the invention.
The compounds of formula (I) may be present in the form of salts. In one embodiment, the salt refers to a pharmaceutically acceptable salt. The term "pharmaceutically acceptable" means that the substance or composition must be compatible chemically and/or toxicologically with the other ingredients comprising the formulation and/or the mammal being treated therewith. In another embodiment, the salts need not be pharmaceutically acceptable salts and may be intermediates useful in the preparation and/or purification of compounds of formula (I) and/or in the isolation of enantiomers of compounds of formula (I).
Pharmaceutically acceptable acid addition salts may be formed from the compounds disclosed herein by the action of an inorganic or organic acid, for example, acetate, glycolate, ethanesulfonate, aspartate, benzoate, benzenesulfonate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlorotheyl, citrate, ethanedisulfonate, fumarate, glucoheptonate, gluconate, glucuronate, hippurate, hydroiodide, isethionate, lactate, lactobionate, lauryl sulfate, malate, maleate, malonate, mandelate, methanesulfonate, methylsulfate, naphthoate, naphthalenesulfonate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, salts of the acids mentioned above, salts of the acids mentioned materials mentioned above, Pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, polypyrolactobionate, propionate, stearate, succinate, sulphosalicylate, tartrate, p-toluenesulphonate and trifluoroacetate.
Pharmaceutically acceptable base addition salts may be formed from the disclosed compounds by reaction with an inorganic or organic base. The inorganic base includes, for example, ammonium salts and metals of groups I to XII of the periodic Table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts; organic bases include primary, secondary and tertiary amines, and substituted amines include naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Some organic amines include, for example, isopropylamine, benzathine (benzathine), choline salts (cholinate), diethanolamine, diethylamine, lysine, meglumine (meglumine), piperazine, and tromethamine.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, basic or acidic moiety, by conventional chemical methods. In general, such salts can be prepared by reacting the free acid forms of these compounds with a stoichiometric amount of the appropriate base (e.g., Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, etc.), or by reacting the free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are usually carried out in water or an organic solvent or a mixture of both. Generally, where appropriate, it is desirable to use a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile. In, for example, "Remington's Pharmaceutical Sciences", 20 th edition, Mack Publishing Company, Easton, Pa., (1985); and "handbook of pharmaceutically acceptable salts: properties, Selection and application (Handbook of pharmaceutical salts: Properties, Selection, and Use) ", Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002) may find some additional lists of suitable salts.
In addition, the compounds disclosed herein, including their salts, may also be obtained in the form of their hydrates or in the form of solvents containing them (e.g., ethanol, DMSO, etc.), for their crystallization. The compounds disclosed herein may form solvates with pharmaceutically acceptable solvents (including water), either inherently or by design; thus, the present invention is intended to include both solvated and unsolvated forms of the disclosed compounds.
Any formulae given herein are also intended to represent the non-isotopically enriched forms as well as the isotopically enriched forms of these compounds. Isotopically enriched compounds have the structure depicted by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as2H、3H、11C、13C、14C、15N、17O、18O、18F、31P、32P、35S、36Cl and125I。
in another aspect, the compounds of the invention include isotopically enriched compounds as defined herein, e.g. wherein a radioisotope, e.g. is present3H、14C and18those compounds of F, or in which a non-radioactive isotope is present, e.g.2H and13those of C. The isotopically enriched compounds can be used for metabolic studies (use)14C) Reaction kinetics study (using, for example2H or3H) Detection or imaging techniques such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution determination, or may be used in radiotherapy of a patient.18F-enriched compounds are particularly desirable for PET or SPECT studies. Isotopically enriched compounds of formula (I) can be prepared by conventional techniques known to those skilled in the art or by the procedures and examples described in the present specification using a suitable isotopically labelled reagent in place of the original used unlabelled reagent.
In addition, heavier isotopes are, in particular, deuterium (i.e.,2substitution of H or D) may provide certain therapeutic advantages resulting from greater metabolic stability. For example, increased in vivo half-life or decreased dosage requirements or improved therapeutic index. It is to be understood that deuterium in the present invention is to be considered as a substituent of the compound of formula (I). The concentration of such heavier isotopes, particularly deuterium, can be defined by isotopic enrichment factors. The term "isotopic enrichment factor" as used herein refers to the ratio between the isotopic and natural abundance of a given isotope. If a substituent of a compound of the invention is designated as deuterium, the compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). Pharmaceutically acceptable solvates of the invention include those in which the crystallization solvent may be isotopically substituted, e.g. D2O, acetone-d6、DMSO-d6Those solvates of (a).
In another aspect, the invention relates to intermediates for the preparation of compounds of formula (I).
In another aspect, the invention relates to methods for the preparation, isolation and purification of compounds of formula (I).
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention. In one embodiment, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable carrier, excipient, adjuvant, vehicle or combination thereof. In another embodiment, the pharmaceutical composition may be in a liquid, solid, semi-solid, gel, or spray dosage form.
Pharmaceutical compositions, formulations and administration of the compounds of the invention
The present invention provides a pharmaceutical composition comprising a compound disclosed herein, for example, as set forth in the examples; and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or combination thereof.
The present invention provides methods of treating, preventing or ameliorating a disease or condition comprising administering a safe and effective amount of a combination comprising a compound of the present disclosure and one or more therapeutically active agents. Wherein the combination comprises one or more additional agents for the treatment of neurological and psychiatric disorders and diseases of the central nervous system.
Other agents for treating neurological and psychiatric disorders and diseases of the central nervous system include, but are not limited to: antidepressant drugs, anxiolytic drugs, lithium salt drugs as mood stabilizers, antipsychotic drugs, atypical antipsychotic drugs, antiepileptic drugs, anti-parkinson drugs, sedative hypnotic drugs, antihistamine drugs, GABA receptor agonists and/or GABA reuptake inhibitors drugs, drugs as monoamine oxidase inhibitors, drugs as melatonin receptor agonists and drugs as orexin receptor antagonists or any combination thereof.
Other drugs mentioned for the treatment of neurological and psychiatric disorders and diseases of the central nervous system are amitriptyline (amitriptyline), desipramine (desipramine), mirtazapine (mirtazapine), bupropion (bupropion), reboxetine (reboxetine), fluoxetine (fluoxetine), trazodone (trazodone), sertraline (sertraline), duloxetine (duloxetine), fluvoxamine (fluvoxamine), milnacipran (milnacipran), levomilnacipran (levomilnacipran), desvenlafaxine (desvenlafaxine), vilazodone (vilazodone), venlafaxine (venlafaxine), dapoxetine (dapoxetine), nefazodone (nefazodone), phenacetine (moxastine), chlorpromazine (ziprasidone), ziprasidone (sulpridone (sulprine), ziprasidone (sulprine (sulpridone), ziprasidone (sulprine), ziprasidone (sulprine (e), ziprasidone (sulprine), ziprasidone (e), ziprasidone (sulprine), ziprasidone (e), ziprasidone (sulprizone), ziprasidone (sulprione), ziprasidone (sulprine), ziprasidone (sulprine, Piperacillin (perzoprione), clozapine (clozapine), modafinil (modafinil), mecamylamine (mecamylamine), cabergoline (cabergoline), adamantane (amantane), imipramine (imipramine), pramipexole (pramipexole), thyroxine (thyroxine), dextromethorphan (dextromethorphan), quinidine (quinidine), naltrexone (naltrexone), samindorphin, buprenorphine (buprenorphine), melatonin (melatonin), alprazolam (alprazolam), pipamperone (pipamperone), vepipitant (vestitanate), perphenazine (perphenazine), midazolam (midelam), triazolam (triazezazolelam), tezomepin (diazepin), diazepizone (diazafloxacin), diazafloxacin (diazaflor (diazafloxacin), diazafloxacin (diazaflm), diazaflor (diazafloxacin), diazafloxacin (mepiridm), diazafloxacin (diazaflm (diazafloxacin (mepiride), diazafloxacin (mepiridm), diazafloxacin (mepiridine), diazaflamide (mepiridine), diazaflamide (mepiridine (triazaflamide), diazaflamide (triazaflacrm), diazaflacrm), diazaflamide (triazafloxacin (triazaflamide (triazaflacrm), triazaflamide (triazaflacrm, Gaboxadol (gaboxadol), clomipramine (clomipramine), doxepin (doxepin), chloral hydrate (chloroplate), haloperidol (haloperidol), chlorpromazine (chlorpromazine), carbamazepine (carbamazepine), promethazine (promethazine), lorazepam (lorazepam), hydroxyzine (hydroxyzine), aspirin (aspirin), diphenhydramine (diphenhydramine), chlorpheniramine (chlorphenamine), brotizolam (lentirmin), rameton (ramelton), teslamelmatone (tasimelteon), agomelatine (omelatine), mianserine (miseranine), famoxadine (famoxadine), doxylamine (gabapentine), doxepin (flavodoxepin), gazepine (gabapentin), gazepine (oxyphendazole), doxepin (oxyphenbutadine), or any combination thereof.
The amount of compound in the pharmaceutical compositions disclosed herein is that amount which is effective to detect antagonism of the orexin receptor in the biological sample or subject. The dosage of the active ingredient in the composition of the present invention may vary, however, the amount of the active ingredient must be such that a suitable dosage form is obtained. The active ingredient may be administered to patients (animals and humans) in need of such treatment at dosages that provide optimal pharmaceutical efficacy. The selected dosage depends on the desired therapeutic effect, on the route of administration and on the duration of the treatment. The dosage will vary from patient to patient depending on the nature and severity of the disease, the weight of the patient, the particular diet of the patient, the concurrent use of drugs, and other factors that will be recognized by those skilled in the art. The dosage range is generally about 0.5mg to 1.0g per patient per day and may be administered in a single dose or in multiple doses. In one embodiment, the dosage range is from about 0.5mg to 500mg per patient per day; from about 0.5mg to 200mg per patient per day in another embodiment; and in yet another embodiment from about 5mg to 50mg per patient per day.
It will also be appreciated that certain compounds of the invention may be present in free form and used in therapy, or if appropriate in the form of a pharmaceutically acceptable derivative thereof. Pharmaceutically acceptable derivatives include pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any additional adduct or derivative that upon administration to a patient in need thereof provides, directly or indirectly, a compound of the present invention or a metabolite or residue thereof.
The medicaments or pharmaceutical compositions disclosed herein may be prepared and packaged in bulk (bulk) form, wherein a safe and effective amount of the compound of formula (I) may be extracted and then administered to a patient in the form of a powder or syrup. Typically, the administration is to the patient at a dosage level of between 0.0001 and 10mg/kg body weight per day to achieve effective antagonism of the orexin receptor. Alternatively, the pharmaceutical compositions disclosed herein can be prepared and packaged in unit dosage forms, wherein each physically discrete unit contains a safe and effective amount of a compound of formula (I). When prepared in unit dosage form, the disclosed pharmaceutical compositions can generally contain, for example, from 0.5mg to 1g, or from 1mg to 700mg, or from 5mg to 100mg of the disclosed compounds.
When the pharmaceutical composition of the invention contains one or more other active ingredients in addition to the compound of the invention, the compound weight ratio of the compound of the invention to the second active ingredient may vary and depends on the effective dose of each ingredient. Generally, an effective dose of each is used. Thus, for example, when a compound of the present invention is mixed with another pharmaceutical agent, the weight ratio of the compound of the present invention to the other pharmaceutical agent typically ranges from about 1000: 1 to about 1: 1000, e.g., from about 200: 1 to about 1: 200. Mixtures of the compounds of the invention with other active ingredients are generally also within the above-mentioned ranges, but in each case an effective dose of each active ingredient should be used.
As used herein, "pharmaceutically acceptable excipient" means a pharmaceutically acceptable material, mixture or vehicle, which is compatible with the dosage form or pharmaceutical composition to be administered. Each excipient, when mixed, must be compatible with the other ingredients of the pharmaceutical composition to avoid interactions that would substantially reduce the efficacy of the disclosed compounds and which would result in a pharmaceutical composition that is not pharmaceutically acceptable when administered to a patient. Furthermore, each excipient must be pharmaceutically acceptable, e.g., of sufficiently high purity.
Suitable pharmaceutically acceptable excipients will vary depending on the particular dosage form selected. In addition, pharmaceutically acceptable excipients may be selected for their specific function in the composition. For example, certain pharmaceutically acceptable excipients may be selected to aid in the production of a uniform dosage form. Certain pharmaceutically acceptable excipients may be selected to aid in the production of stable dosage forms. Certain pharmaceutically acceptable excipients may be selected to facilitate carrying or transporting the disclosed compounds from one organ or portion of the body to another organ or portion of the body when administered to a patient. Certain pharmaceutically acceptable excipients may be selected that enhance patient compliance.
Suitable pharmaceutically acceptable excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, taste masking agents, colorants, anti-caking agents, humectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants and buffers. The skilled artisan will recognize that certain pharmaceutically acceptable excipients may provide more than one function, and provide alternative functions, depending on how many such excipients are present in the formulation and those other excipients are present in the formulation.
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 disclosed compounds, 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 disclosed herein 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).
Thus, in another aspect, the invention relates to a process for preparing a pharmaceutical composition comprising a compound of the present disclosure and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or combination thereof, which process comprises admixing the ingredients. Pharmaceutical compositions comprising the disclosed compounds may be prepared by mixing, for example, at ambient temperature and atmospheric pressure.
The compounds disclosed herein 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.
In one embodiment, the compounds disclosed herein may be formulated in oral dosage forms. In another embodiment, the compounds disclosed herein may be formulated in an inhalation dosage form. In another embodiment, the compounds disclosed herein can be formulated for nasal administration. In yet another embodiment, the compounds disclosed herein can be formulated for transdermal administration. In yet another embodiment, the compounds disclosed herein may be formulated for topical administration.
The pharmaceutical compositions provided by the present invention may be provided as compressed tablets, milled tablets, chewable lozenges, fast-dissolving tablets, double-compressed tablets, or enteric-coated, sugar-coated or film-coated tablets. Enteric coated tablets are compressed tablets coated with a substance that is resistant to the action of gastric acid but dissolves or disintegrates in the intestine, thereby preventing the active ingredient from contacting the acidic environment of the stomach. Enteric coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalate. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which can help to mask unpleasant tastes or odors and prevent oxidation of the tablet. Film-coated tablets are compressed tablets covered with a thin layer or film of a water-soluble substance. Film coatings include, but are not limited to, hydroxyethyl cellulose, sodium carboxymethyl cellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coatings are endowed with the same general characteristics as sugar coatings. A tabletted tablet is a compressed tablet prepared over more than one compression cycle, including a multi-layer tablet, and a press-coated or dry-coated tablet.
Tablet dosage forms may be prepared from the active ingredient in powder, crystalline or granular form, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled release polymers, lubricants, diluents and/or colorants. Flavoring and sweetening agents are particularly useful in forming chewable tablets and lozenges.
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. The 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.
The pharmaceutical compositions provided herein may be provided in liquid and semi-solid dosage forms, including emulsions, solutions, suspensions, elixirs and syrups. Emulsions are two-phase systems in which one liquid is dispersed throughout another in the form of globules, which can be either oil-in-water or water-in-oil. Emulsions may include pharmaceutically acceptable non-aqueous liquids and solvents, emulsifiers and preservatives. Suspensions may include a pharmaceutically acceptable suspending agent and a preservative. The aqueous alcoholic solution may comprise pharmaceutically acceptable acetals, such as di (lower alkyl) acetals of lower alkyl aldehydes, e.g. acetaldehyde diethyl acetal; and water-soluble solvents having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, hydroalcoholic solutions. Syrups are concentrated aqueous solutions of sugars, such as sucrose, and may also contain preservatives. For liquid dosage forms, for example, a solution in polyethylene glycol may be diluted with a sufficient amount of a pharmaceutically acceptable liquid carrier, such as water, for precise and convenient administration.
Other useful liquid and semi-solid dosage forms include, but are not limited to, those comprising the active ingredients provided herein and a secondary mono-or poly-alkylene glycol, including: 1, 2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, where 350, 550, 750 refer to the approximate average molecular weight of the polyethylene glycol. These formulations may further include one or more antioxidants, such as Butylated Hydroxytoluene (BHT), Butylated Hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
Dosage unit formulations for oral administration may be microencapsulated, where appropriate. They may also be prepared as extended or sustained release compositions, for example by coating or embedding the particulate material in a polymer, wax or the like.
The oral pharmaceutical composition provided by the invention can also be provided in the form of liposome, micelle, microsphere or nano system. Micellar dosage forms can be prepared using the methods described in U.S. Pat. No.6,350,458.
The pharmaceutical compositions provided herein can be provided as non-effervescent or effervescent granules and powders for reconstitution into liquid dosage forms. Pharmaceutically acceptable carriers and excipients used in non-effervescent granules or powders may include diluents, sweeteners and wetting agents. Pharmaceutically acceptable carriers and excipients used in effervescent granules or powders may include organic acids and sources of carbon dioxide.
Coloring and flavoring agents may be used in all of the above dosage forms.
The disclosed compounds may also be conjugated to soluble polymers as targeted drug carriers. Such polymers include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspartamidephenol or polyoxyethylene polylysine substituted with palmitoyl residues. In addition, the disclosed compounds may be combined with a class of biodegradable polymers used in achieving controlled release of a drug, such as polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and crosslinked or amphiphilic block copolymers of hydrogels.
The pharmaceutical compositions provided by the present invention may be formulated into immediate or modified release dosage forms, including delayed-, sustained-, pulsed-, controlled-, targeted-, and programmed release forms.
The pharmaceutical compositions provided by the present invention may be co-formulated with other active ingredients that do not impair the intended therapeutic effect, or with substances that supplement the intended effect.
The pharmaceutical compositions provided by the present invention may be administered parenterally by injection, infusion or implantation for local or systemic administration. Parenteral administration as used herein includes intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial and subcutaneous administration.
The pharmaceutical compositions provided herein can be formulated in any dosage form suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems and solid forms suitable for solution or suspension in a liquid prior to injection. Such dosage forms may be prepared according to conventional methods known to those skilled in The art of pharmaceutical Science (see Remington: The Science and Practice of Pharmacy, supra).
Pharmaceutical compositions intended for parenteral administration may include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives to inhibit microbial growth, stabilizers, solubility enhancers, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, thickening agents, pH adjusting agents, and inert gases.
Suitable aqueous carriers include, but are not limited to: water, saline, normal saline or Phosphate Buffered Saline (PBS), sodium chloride injection, Ringers injection, isotonic glucose injection, sterile water injection, dextrose and lactated Ringers injection. Non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and the medium chain triglycerides of coconut oil, and palm seed oil. Water-miscible vehicles include, but are not limited to, ethanol, 1, 3-butanediol, liquid polyethylene glycols (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerol, N-methyl-2-pyrrolidone, N-dimethylacetamide, and dimethylsulfoxide.
Suitable antimicrobial agents or preservatives include, but are not limited to, phenol, cresol, mercurial, benzyl alcohol, chlorobutanol, methyl and propyl parabens, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl and propyl parabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerol and glucose. Suitable buffers include, but are not limited to, phosphate and citrate. Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfite. Suitable local anesthetics include, but are not limited to, procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcellulose, hydroxypropylmethylcellulose and polyvinylpyrrolidone. Suitable emulsifiers include those described herein, including polyoxyethylene sorbitan monolaurate. Polyoxyethylene sorbitan monooleate 80 and triethanolamine oleate. Suitable sequestering or chelating agents include, but are not limited to, EDTA. Suitable pH adjusters include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid. Suitable complexing agents include, but are not limited to, cyclodextrins, including alpha-cyclodextrin, beta-cyclodextrin, hydroxylsPropyl-beta-cyclodextrin, sulfobutyl ether-beta-cyclodextrin and sulfobutyl ether 7-beta-cyclodextrin (C:)CyDex,Lenexa,KS)。
The pharmaceutical compositions provided herein may be formulated for single or multiple dose administration. The single dose formulations are packaged in ampoules, vials or syringes. The multi-dose parenteral formulation must contain a bacteriostatic or fungistatic concentration of the antimicrobial agent. All parenteral formulations must be sterile, as is known and practiced in the art.
In one embodiment, the pharmaceutical composition is provided as a ready-to-use sterile solution. In another embodiment, the pharmaceutical compositions are provided as sterile dried soluble products, including lyophilized powders and subcutaneous injection tablets, which are reconstituted with a carrier prior to use. In yet another embodiment, the pharmaceutical composition is formulated as a ready-to-use sterile suspension. In yet another embodiment, the pharmaceutical composition is formulated as a sterile, dry, insoluble product that is reconstituted with a carrier prior to use. In yet another embodiment, the pharmaceutical composition is formulated as a sterile emulsion ready for use.
The pharmaceutical composition may be formulated as a suspension, solid, semi-solid, or thixotropic liquid for depot administration for implantation. In one embodiment, the disclosed pharmaceutical compositions are dispersed in a solid internal matrix surrounded by an outer polymeric membrane that is insoluble in body fluids but allows diffusion therethrough of the active ingredient in the pharmaceutical composition.
Suitable internal matrices include polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinyl chloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene vinyl acetate copolymers, silicone rubber, polydimethylsiloxane, silicone carbonate copolymers, hydrogels of hydrophilic polymers such as esters of acrylic and methacrylic acid, collagen, crosslinked polyvinyl alcohol, and partially hydrolyzed polyvinyl acetate of the class of copolymers.
Suitable outer polymeric films include polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate copolymers, silicone rubber, polydimethylsiloxane, neoprene, chlorinated polyethylene, polyvinyl chloride, copolymers of chlorinated ethylene and vinyl acetate, vinylidene chloride, ethylene and propylene, ionomers polyethylene terephthalate, butyl rubber chlorohydrin rubber, ethylene/vinyl alcohol copolymers, ethylene/vinyl acetate/vinyl alcohol terpolymers, and ethylene/ethyleneoxyethanol copolymers.
In another aspect, the disclosed pharmaceutical compositions may be formulated in any dosage form suitable for administration to a patient by inhalation, such as a dry powder, aerosol, suspension, or solution composition. In one embodiment, the disclosed pharmaceutical compositions may be formulated in a dosage form suitable for inhalation administration to a patient as a dry powder. In yet another embodiment, the disclosed pharmaceutical compositions may be formulated in a dosage form suitable for inhalation administration to a patient via a nebulizer. Dry powder compositions for delivery to the lung by inhalation typically comprise a finely powdered compound disclosed herein and one or more finely powdered pharmaceutically acceptable excipients. Pharmaceutically acceptable excipients that are particularly suitable for use as dry powders are known to those skilled in the art and include lactose, starch, mannitol, and mono-, di-and polysaccharides. Fine powders may be prepared, for example, by micronization and milling. Generally, the size-reduced (e.g., micronized) compound may pass through a D of about 1 to 10 microns50Values (e.g., measured by laser diffraction).
Aerosols can be formulated by suspending or dissolving the disclosed compounds in a liquefied propellant. Suitable propellants include chlorinated hydrocarbons, hydrocarbons and other liquefied gases. Representative propellants include: trichlorofluoromethane (propellant 11), dichlorofluoromethane (propellant 12), dichlorotetrafluoroethane (propellant 114), tetrafluoroethane (HFA-134a), 1-difluoroethane (HFA-152a), difluoromethane (HFA-32), pentafluoroethane (HFA-12), heptafluoropropane (HFA-227a), perfluoropropane, perfluorobutane, perfluoropentane, butane, isobutane and pentane. Aerosols comprising the compounds disclosed herein are typically administered to a patient via a Metered Dose Inhaler (MDI). Such devices are known to those skilled in the art
The aerosol may contain additional pharmaceutically acceptable excipients that may be used by MDIs, such as surfactants, lubricants, co-solvents, and other excipients, to improve the physical stability of the formulation, to improve valve characteristics, to improve solubility, or to improve taste.
Pharmaceutical compositions suitable for transdermal administration may be prepared as discrete patches intended to remain in intimate contact with the epidermis of the patient for an extended period of time. For example, the active ingredient may be delivered from a patch agent by iontophoresis, as generally described in Pharmaceutical Research,3(6),318 (1986).
Pharmaceutical compositions suitable for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils. For example, ointments, creams and gels may be formulated with a water or oil base, and suitable thickeners and/or gelling agents and/or solvents. Such bases may include, water, and/or oils such as liquid paraffin and vegetable oils (e.g., peanut oil or castor oil), or solvents such as polyethylene glycol. Thickeners and gelling agents used according to the nature of the base include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycol, lanolin, beeswax, carbopol and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifiers.
Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents or thickening agents.
Powders for external use may be formed in the presence of any suitable powder base, for example talc, lactose or starch. Drops may be formulated with an aqueous or non-aqueous base containing one or more dispersing agents, solubilising agents, suspending agents or preservatives.
Topical formulations may be administered by application to the affected area one or more times per day; an occlusive dressing covering the skin is preferably used. Adhesive depot systems allow for continuous or extended administration.
Use of the Compounds and compositions of the invention
The compounds or pharmaceutical compositions disclosed herein may be used in the manufacture of a medicament for the treatment, prevention, amelioration, control or alleviation of neurological and psychiatric disorders and diseases of the central nervous system of mammals, including humans, and in the manufacture of other medicaments for antagonism of orexin receptors.
In particular, the compounds of the present invention are present in the compositions in amounts effective to detectably antagonize orexin receptors and are useful as agents for the prevention or treatment of neurological and psychiatric disorders and diseases of the Central Nervous System (CNS) in humans such as depression, anxiety, seasonal affective disorder, mania, bipolar disorder, obsessive-compulsive disorder, jetlag-induced insomnia and fatigue, schizophrenia, convulsions, panic attacks, melancholia, alcohol addiction, drug addiction, alcoholism, substance abuse, drug addiction withdrawal symptoms, insomnia, psychotic disorders, epilepsy, sleep disorders, sleep disturbances, sleep apnea syndrome, compulsive eating disorders, fibromyalgia, stress, obesity, Parkinson's disease, senile dementia, cognitive disorders, memory disorders, premenstrual syndrome, migraine, Memory loss, Alzheimer's disease, or other various disorders associated with normal or pathological aging.
The compounds or compositions of the present invention may be used in, but are in no way limited to, the prevention, treatment, or alleviation of central nervous system neurological and psychiatric disorders and diseases in mammals, including humans, by administering to a patient an effective amount of a compound or composition of the present invention. The human CNS neurological and psychiatric disorders and diseases further include, but are not limited to, depression, anxiety, seasonal affective disorder, mania, bipolar disorder, obsessive-compulsive disorder, insomnia and fatigue due to jet lag, schizophrenia, convulsion, panic attack, melancholia, alcohol addiction, drug addiction, alcohol intoxication, substance abuse, drug addiction withdrawal symptoms, insomnia, psychotic disorders, epilepsy, sleep disorders, sleep disturbances, sleep apnea syndrome, compulsive eating disorders, fibromyalgia, stress response, obesity, Parkinson's disease, senile dementia, cognitive disorders, memory disorders, premenstrual tension syndrome, migraine, memory loss, Alzheimer's disease or other disorders associated with normal or pathological aging.
In addition to being beneficial for human therapy, the compounds and pharmaceutical compositions of the present invention may also find application in veterinary therapy for pets, animals of the introduced species and mammals in farm animals. Examples of other animals include horses, dogs, and cats. Herein, the compound of the present invention includes pharmaceutically acceptable derivatives thereof.
Method of treatment
In one embodiment, the presently disclosed methods of treatment comprise administering to a patient in need thereof a safe and effective amount of a compound of the present invention or a pharmaceutical composition comprising a compound of the present invention. Various embodiments of the present disclosure include methods of treating the above-mentioned diseases by administering to a patient in need thereof a safe and effective amount of a disclosed compound or a pharmaceutical composition comprising a disclosed compound.
In one embodiment, the disclosed compounds or pharmaceutical compositions comprising the disclosed compounds may be administered by any suitable route of administration, including systemic 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 disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered orally. In another embodiment, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered by inhalation. In yet another embodiment, the presently disclosed compounds or compositions comprising the presently disclosed compounds may be administered intranasally.
In one embodiment, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered once or several times at different time intervals over a specified period of time 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 the disclosed compounds or pharmaceutical compositions comprising the disclosed compounds depend on the pharmacokinetic properties of the compound, such as dilution, distribution and half-life, which can be determined by the skilled person. In addition, suitable dosing regimens for the compounds or pharmaceutical compositions comprising the disclosed compounds, including the duration of the regimen, will depend on 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 other factors within the knowledge and experience of the skilled artisan. Such a skilled artisan will also appreciate that adjustments to the subject's response to the dosage regimen, or the need for changes in the subject's patient over time, may be required.
The compounds disclosed herein may be administered simultaneously, or before or after, one or more other therapeutic agents. The compounds of the invention may be administered separately from the other therapeutic agents, by the same or different routes of administration, or together with them in pharmaceutical compositions.
For an individual of about 50-70kg, the disclosed pharmaceutical compositions and combinations may be in unit dosage form containing from about 1-1000mg, or from about 1-500mg, or from about 1-250mg, or from about 1-150mg, or from about 0.5-100mg, or from about 1-50mg of the active ingredient. The therapeutically effective amount of the compound, pharmaceutical composition or combination thereof will depend on the species, weight, age and condition of the individual, 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. The compounds disclosed herein are used in vitro in the form of solutions, e.g. aqueous solutions, and also enterally, parenterally, especially intravenously, in vivo, e.g. in the form of suspensions or aqueous solutions.
In one embodiment, a therapeutically effective dose of a compound of the present disclosure 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.
In addition, the compounds disclosed herein may be administered in the form of a prodrug. In the present invention, a "prodrug" of a disclosed compound is a functional derivative that, when administered to a patient, is ultimately released in vivo. When administering the compounds disclosed herein in the form of a prodrug, one skilled in the art can practice one or more of the following: (a) altering the in vivo onset time of the compound; (b) altering the duration of action of the compound in vivo; (c) altering the in vivo delivery or distribution of the compound; (d) altering the in vivo solubility of the compound; and (e) overcoming side effects or other difficulties faced by the compounds. Typical functional derivatives useful for preparing prodrugs comprise variants of the compounds which are cleaved in vivo either chemically or enzymatically. These variants, which involve the preparation of phosphates, amides, esters, thioesters, carbonates and carbamates, are well known to those skilled in the art.
General synthetic procedure
To illustrate the invention, the following examples are set forth. It is to be understood that the invention is not limited to these embodiments, but is provided as a means of practicing the invention.
In general, the compounds of the present invention may be prepared by the methods described herein, wherein the substituents are as defined in formula (I), unless otherwise indicated. The following reaction schemes and examples serve to further illustrate the context of the invention.
Those skilled in the art will recognize that: the chemical reactions described herein may be used to suitably prepare a number of other compounds of the invention, and other methods for preparing the compounds of the invention are considered to be within the scope of the invention. For example, the synthesis of those non-exemplified compounds according to the present invention can be successfully accomplished by those skilled in the art by modification, such as appropriate protection of interfering groups, by the use of other known reagents in addition to those described herein, or by some routine modification of reaction conditions. In addition, the reactions disclosed herein or known reaction conditions are also recognized as being applicable to the preparation of other compounds of the present invention.
The examples described below, unless otherwise indicated, are all temperatures set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Arco Chemical Company and Alfa Chemical Company and were used without further purification unless otherwise indicated. General reagents were purchased from Shantou Wen Long chemical reagent factory, Guangdong Guanghua chemical reagent factory, Guangzhou chemical reagent factory, Tianjin Haojian Yunyu chemical Co., Ltd, Tianjin Shucheng chemical reagent factory, Wuhan Xin Huayuan scientific and technological development Co., Ltd, Qingdao Tenglong chemical reagent Co., Ltd, and Qingdao Kaolingyi factory.
The anhydrous tetrahydrofuran, dioxane, toluene and ether are obtained through reflux drying of metal sodium. The anhydrous dichloromethane and chloroform are obtained by calcium hydride reflux drying. Ethyl acetate, petroleum ether, N-hexane, N, N-dimethylacetamide and N, N-dimethylformamide were used as they were previously dried over anhydrous sodium sulfate.
The following reactions are generally carried out under positive pressure of nitrogen or argon or by sleeving a dry tube over an anhydrous solvent (unless otherwise indicated), the reaction vial being stoppered with a suitable rubber stopper and the substrate being injected by syringe. The glassware was dried.
The column chromatography is performed using a silica gel column. Silica gel (300 and 400 meshes) was purchased from Qingdao oceanic chemical plants.
1H NMR spectra were recorded using a Bruker 400MHz or 600MHz NMR spectrometer.1H NMR Spectrum in CDC13、DMSO-d6、CD3OD or acetone-d6TMS (0ppm) or chloroform (7.26ppm) was used as a reference standard for the solvent (in ppm). When multiple peaks occur, the following abbreviations will be used: s (singleton), d (doublet), t (triplet), m (multiplet), br (broad), dd (doublet of doublets), dt (doublet of triplets). Coupling constants are expressed in hertz (Hz).
The conditions for determining low resolution Mass Spectrometry (MS) data were: agilent 6120 four-stage rod HPLC-M (column model: Zorbax SB-C18,2.1X 30mm,3.5 micron, 6min, flow rate 0.6 mL/min. mobile phase: 5% -95% (CH with 0.1% formic acid)3CN) in (H containing 0.1% formic acid)2O) by electrospray ionization (ESI) at 210nm/254nm, with UV detection.
Pure compounds were detected by UV at 210nm/254nm using Agilent 1260 pre-HPLC or Calesep pump 250 pre-HPLC (column model: NOVASEP 50/80 mmDAC).
The following acronyms are used throughout the invention:
CH2Cl2DCM dichloromethane; cs2CO3Cesium carbonate; CDC13Deuterated chloroform; CuI cuprous iodide; DMF N, N-dimethylformamide; DMSO dimethyl sulfoxide; et (Et)3N, TEA triethylamine; EtOAc, EA ethyl acetate; EtOH ethanol; g, g; h hours; min; h2Hydrogen gas; HCl hydrochloric acid; KOH potassium hydroxide; k2CO3Potassium carbonate; MeCN, CH3CN acetonitrile; na (Na)2CO3Sodium carbonate; NaOH sodium hydroxide; na (Na)2SO4Sodium sulfate; MeOH AAn alcohol; MgSO (MgSO)4Magnesium sulfate; mL, mL; pd (PPh)3)4Tetrakis (triphenylphosphine) palladium; PE petroleum ether (60-90 deg.C); RT, RT, r.t. room temperature; rt retention time; SOCl2Thionyl chloride, thionyl chloride; THF tetrahydrofuran
A typical synthetic procedure for the preparation of the disclosed compounds is shown in scheme 1 below. Unless otherwise stated, each of n, m, R1And R4Having the definitions as described in the present invention.
Intermediate synthesis scheme
Figure BDA0001012285000000231
O-iodobenzoic acid(s) substituted with different substituents1) With 2H-1,2, 3-triazole in [ Cu ]]In the presence of a catalyst and a suitable base, under heating to obtain a compound intermediate (2). Compound (A) to (B)2) With a chlorinating agent under heating to give compound (3).
Synthesis scheme 1
Figure BDA0001012285000000232
Compound (A) to (B)8) And a compound of (A), (B), (C), (9) Can be prepared by the following processes respectively:
substituted 1-naphthaldehydes (4) Reacting with nitromethane under the action of ammonium acetate to generate a compound (A)5) Then sequentially carrying out two-step reactions of sodium borohydride reduction and catalytic hydrogenation to obtain a compound (A)7) Compound (A) to (B)7) With an intermediate compound (3) Directly reacting to obtain a compound (A)8). Compound (A) to (B)8) Finally reacting with bromoethane to obtain a compound (A)9)。
The compounds, pharmaceutical compositions and uses thereof provided by the present invention are further illustrated below in connection with the examples.
Examples
Example 1 5-methyl-N- (2- (naphthalen-1-yl) ethyl) -2- (2H-1,2, 3-triazol-2-yl) benzamide
Figure BDA0001012285000000233
Step 1 Synthesis of (E) -1- (2-nitrovinyl) naphthalene
1-Naphthalenecarboxaldehyde (0.156g,1.0mmol) and ammonium acetate (0.116g,1.5mmol) were sequentially added to a 100mL single-necked flask, nitromethane (10mL) was added thereto, and the mixture was gradually heated to reflux. After 7 hours, the reaction was stopped, cooled, the solvent was evaporated under reduced pressure, the residue was dissolved in dichloromethane (30mL), the mixture was washed with equal volume of tap water and saturated brine, the organic layer was dried over anhydrous sodium sulfate, filtered, and the filtrate was evaporated under reduced pressure and purified by column chromatography (petroleum ether/ethyl acetate (v/v) ═ 100/1) to give the title compound (yellow solid, 0.15g, 75%).
MS(ESI,pos.ion)m/z:200.0[M+H]+
1HNMR(600MHz,CDCl3)δ(ppm):8.82(d,J=13.4Hz,1H),8.12(d,J=8.4Hz,1H),8.00(d,J=8.2Hz,1H),7.91(d,J=8.1Hz,1H),7.74(d,J=7.2Hz,1H),7.65-7.62(m,2H),7.60-7.57(m,1H),7.52-7.50(m,1H).
Step 2) Synthesis of 1- (2-Nitroethyl) naphthalene
(E) -1- (2-nitrovinyl) naphthalene (0.598g,3.0mmol) and absolute methanol (20mL) were sequentially added to a 100mL single-neck flask, sodium borohydride (0.290g,7.5mmol) was added in portions slowly at 0 ℃, and after 2 hours, the reaction was completed and stopped. An ice-water mixture (30mL) was slowly added to the reaction mixture, and the mixture was extracted with methylene chloride (30 mL. times.3), and the organic phases were combined and washed with saturated brine (30mL), and the organic layer was dried over anhydrous sodium sulfate. The filtrate was filtered, evaporated to dryness under reduced pressure and purified by column chromatography (petroleum ether/ethyl acetate (v/v) ═ 100/1) to give the title compound (yellow liquid, 0.344g, 57%).
1H NMR(600MHz,CDCl3)δ(ppm):7.99(d,J=8.3Hz,1H),7.90(d,J=8.0Hz,1H),7.81(d,J=8.1Hz,1H),7.59(t,J=7.6Hz,1H),7.54(t,J=7.3Hz,1H),7.43(t,J=7.5Hz,1H),7.37(d,J=6.8Hz,1H),4.73(t,J=7.6Hz,2H),3.81(t,J=7.6Hz,2H).
Step 3) Synthesis of 2- (Naphthalen-1-yl) ethylamine
1- (2-Nitroethyl) naphthalene (0.373g,1.85mmol), absolute methanol (15mL) and 10% palladium on carbon (0.04g) were sequentially charged into a 100mL single-neck flask, and after purging with hydrogen three times, the reaction was completed after reacting at room temperature for 10 hours under a hydrogen atmosphere. The reaction was stopped, filtered, and the filtrate was evaporated to dryness and purified by column chromatography (dichloromethane/methanol (v/v) ═ 20/1) to give the title compound (yellow liquid, 0.266g, 84%).
MS(ESI,pos.ion)m/z:172.1[M+H]+
1H NMR(600MHz,CDCl3)δ(ppm):8.05(d,J=8.4Hz,1H),7.87-7.85(m,1H),7.74(d,J=8.2Hz,1H),7.53-7.47(m,2H),7.42-7.40(m,1H),7.35(d,J=6.8Hz,1H),3.25(t,J=7.0Hz,2H),3.12(t,J=7.0Hz,2H).
Step 4) synthesis of 5-methyl-2- (2H-1,2, 3-triazole-2-yl) benzoic acid
2H-1,2, 3-triazole (3.45g,50mmol), 2-iodo-5-methylbenzoic acid (5.24g,20mmol), cesium carbonate (11.72g,36mmol), trans-N, N' -dimethyl-1, 2-cyclohexanediamine (0.51g,3.6mmol), cuprous iodide (0.38g,2mmol), and N, N-dimethylformamide (30mL) were sequentially added to a 100mL single-neck round-bottom flask, and the temperature was gradually raised to 100 ℃ under nitrogen protection for 4 hours. The reaction was stopped, cooled, diluted with tap water and extracted with ethyl acetate (200 mL. times.2). The aqueous layer was acidified to pH 1-2 with concentrated hydrochloric acid, extracted with ethyl acetate (200mL × 2), the subsequent organic layers were combined and dried over anhydrous sodium sulfate, filtered, and the filtrate was evaporated to dryness under reduced pressure and purified by column chromatography (dichloromethane/methanol (v/v) ═ 50/1) to give the title compound (yellow solid, 2.76g, 68%).
MS(ESI,neg.ion)m/z:202.1[M-H]-
1HNMR(CD3OD,600MHz)δ(ppm):7.88(s,2H),7.66(d,1H),7.59(d,J=8.2Hz,1H),7.50-7.48(dd,J=8.1Hz,J=1.1Hz,1H),2.45(s,3H).
13C NMR(CD3OD,151MHz)δ(ppm):169.8,140.7,137.5,136.7,133.5,131.5,129.3,126.0,21.0.
Step 5) synthesis of 5-methyl-2- (2H-1,2, 3-triazole-2-yl) benzoyl chloride
5-methyl-2- (2H-1,2, 3-triazole-2-yl) benzoic acid (2.03g,10mmol) is added into a 100mL single-neck round-bottom flask, dissolved by anhydrous dichloromethane (20mL), added with thionyl chloride (15mL,200mmol) and pyridine (0.15mL,2mmol) slowly, and heated to reflux reaction for 3 hours gradually. The reaction was stopped, cooled, and the solvent was slowly evaporated under reduced pressure, and the resulting product was used directly in the next reaction.
Step 6) Synthesis of 5-methyl-N- (2- (naphthalen-1-yl) ethyl) -2- (2H-1,2, 3-triazol-2-yl) benzamide Become into
Under the condition of ice-water bath, 5-methyl-2- (2H-1,2, 3-triazole-2-yl) benzoyl chloride (0.266g,1.2mmol) is added into a 50mL single-neck flask, dissolved by anhydrous dichloromethane (15mL), added with triethylamine (0.3mL,2.0mmol) and 2- (naphthalene-1-yl) ethylamine (0.20g,1.0mmol) slowly, stirred for 10min, removed of the ice-water bath, and reacted at room temperature overnight. The reaction mixture was washed with equal volumes of tap water and saturated brine in this order, and the organic layer was dried over anhydrous sodium sulfate, filtered, and the filtrate was evaporated to dryness under reduced pressure and then subjected to column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound (white solid, 214mg, 60%).
MS(ESI,pos.ion)m/z:357.3[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm):8.11(d,J=8.2Hz,1H),7.86(d,J=7.6Hz,1H),7.75(t,J=7.0Hz,1H),7.68(s,2H),7.58(d,J=8.2Hz,1H),7.56-7.46(m,2H),7.41-7.33(m,2H),7.29(dd,J=13.0,6.8Hz,2H),5.92(s,1H),3.71(dd,J=13.2,6.8Hz,2H),3.29(t,J=7.0Hz,2H),2.40(d,J=10.8Hz,3H).
13C NMR(101MHz,CDCl3)δ(ppm):167.6,139.0,135.5,134.9,134.8,134.0,132.0,131.3,131.2,129.7,128.8,127.3,126.6,126.2,125.7,125.5,124.4,123.7,40.6,32.4,21.0.
Example 2N-Ethyl-5-methyl-N- (2- (naphthalen-1-yl) ethyl) -2- (2H-1,2, 3-triazol-2-yl) benzamide
Figure BDA0001012285000000251
Under the condition of ice-water bath, 5-methyl-N- (2- (naphthalene-1-yl) ethyl) -2- (2H-1,2, 3-triazole-2-yl) benzamide (0.14g,0.39mmol) is added into a 50mL single-neck bottle, dissolved by dry N, N-dimethylformamide (3mL), sodium hydride (0.05g,1.25mmol) with the content of 60 percent is slowly added, stirred for 10min, bromoethane (0.055g,0.5mmol) is added, the ice-water bath is removed, and the reaction is carried out for 2 hours at room temperature. The reaction mixture was slowly poured into water (20mL), extracted with ethyl acetate (30mL × 3), the organic layers were combined, washed with water (30mL × 3), washed with saturated brine (30mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was evaporated to dryness under reduced pressure and then subjected to column chromatography for purification (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound (pale yellow viscous substance, 129mg, 86%).
MS(ESI,pos.ion)m/z:385.3[M+H]+.
Example 3N- (2- (4-fluoronaphthalen-1-yl) ethyl) -5-methyl-2- (2H-1,2, 3-triazol-2-yl) benzamide
Figure BDA0001012285000000252
Step 1 Synthesis of (E) -1-fluoro-4- (2-nitrovinyl) naphthalene
This step title compound was prepared as described in example 1, step 1, by reacting 4-fluoro-1-naphthaldehyde (0.871g,5.0mmol), ammonium acetate (0.578g,7.5mmol) in nitromethane (25mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 150/1) to give the title compound as a yellow solid (0.968 g, 89%).
MS(ESI,pos.ion)m/z:217.9[M+H]+
1HNMR(600MHz,CDCl3)δ(ppm):8.77(d,J=13.4Hz,1H),8.20(d,J=8.3Hz,1H),8.15(d,J=8.6Hz,1H),7.75-7.71(m,2H),7.68-7.62(m,2H),7.21(dd,J=9.7,8.1Hz,1H).
Step 2) Synthesis of 1-fluoro-4- (2-nitroethyl) naphthalene
The title compound was prepared by the method described in example 1, step 2, i.e. (E) -1-fluoro-4- (2-nitrovinyl) naphthalene (0.489g,2.25mmol), sodium borohydride (0.217g,5.62mmol) in a mixed solvent of tetrahydrofuran (9mL) and anhydrous methanol (36mL), and the crude product was isolated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 150/1) to give the title compound (yellow liquid, 0.20g, 40%).
1H NMR(600MHz,CDCl3)δ(ppm):8.18(d,J=8.2Hz,1H),7.96(d,J=8.4Hz,1H),7.66-7.63(m,1H),7.60(t,J=7.3Hz,1H),7.30(dd,J=7.8,5.3Hz,1H),7.09(dd,J=10.1,7.9Hz,1H),4.72-4.69(m,2H),3.77(t,J=7.6Hz,2H).
Step 3) Synthesis of 2- (4-fluoronaphthalen-1-yl) ethylamine
The title compound was prepared as described in example 1, step 3 by reacting 1-fluoro-4- (2-nitroethyl) naphthalene (0.197g,0.9mmol), 10% palladium on charcoal (0.02g) in dry methanol (10mL) under hydrogen, and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20/1) to give the title compound (yellow liquid, 0.160g, 94%).
MS(ESI,pos.ion)m/z:190.2[M+H]+
1H NMR(600MHz,CDCl3)δ(ppm):8.14(d,J=8.1Hz,1H),8.03(d,J=8.0Hz,1H),7.58-7.53(m,2H),7.26(d,J=12.9Hz,1H),7.07(dd,J=10.1,8.0Hz,1H),3.18(d,J=6.0Hz,2H),3.12-3.08(m,2H).
Step 4) N- (2- (4-fluoronaphthalene-1-yl) ethyl) -5-methyl-2- (2H-1,2, 3-triazole-2-yl) benzamide Synthesis of (2)
The title compound was prepared as described in example 1, step 6, by reacting 2- (4-fluoronaphthalen-1-yl) ethylamine (0.41g,2.2mmol), 5-methyl-2- (2H-1,2, 3-triazol-2-yl) benzoyl chloride (0.585g,2.64mmol) with triethylamine (0.76mL,5.4mmol) and dichloromethane (20mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 4/1) to give the title compound as a white solid (0.231 g, 28%).
MS(ESI,pos.ion)m/z:374.5(M+);
1H NMR(400MHz,CDCl3)δ(ppm):8.15(dd,J=15.2,7.6Hz,2H),7.73(s,2H),7.68-7.55(m,3H),7.39(s,1H),7.36(d,J=8.2Hz,1H),7.23(dd,J=7.6,5.4Hz,1H),7.06(dd,J=10.2,8.0Hz,1H),5.91(s,1H),3.70(dd,J=13.4,6.8Hz,2H),3.27(t,J=7.2Hz,2H),2.43(s,3H).
13C NMR(151MHz,DMSO-d6)δ(ppm):167.3,158.2,156.6,138.5,136.3,135.1,133.3(d,J=4.2Hz),132.4(d,J=4.2Hz),132.2,131.0,129.6,127.7,124.7(d,J=2.4Hz),124.0,123.7,123.6,120.9(d,J=5.5Hz),109.7,109.6,40.6,32.1,20.9.
EXAMPLE 4N-Ethyl-N- (2- (4-fluoronaphthalen-1-yl) ethyl) -5-methyl-2- (2H-1,2, 3-triazol-2-yl) benzamide
Figure BDA0001012285000000261
The title compound was prepared as described in example 2, i.e. N- (2- (4-fluoronaphthalen-1-yl) ethyl) -5-methyl-2- (2H-1,2, 3-triazol-2-yl) benzamide (0.146g,0.39mmol), ethyl bromide (0.055g,0.5mmol) was reacted at a 60% sodium hydride content (0.05g,1.25mmol) and N, N-dimethylformamide (3mL) and the crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 4/1) to give the title compound (pale yellow dope, 0.094g, 60%).
MS(ESI,pos.ion)m/z:403.3[M+H]+.
Example 5N- (2- (naphthalen-1-yl) ethyl) -2- (2H-1,2, 3-triazol-2-yl) benzamide
Figure BDA0001012285000000262
Step 1) synthesis of 2- (2H-1,2, 3-triazole-2-yl) benzoic acid
The title compound was prepared as described in example 1, step 4 by reacting 2H-1,2, 3-triazole (0.7g,10.08mmol), 2-iodobenzoic acid (1g,4.03mmol), cesium carbonate (2.36g,7.2mmol), trans-N, N' -dimethyl-1, 2-cyclohexanediamine (0.103g,0.752mmol) and cuprous iodide (0.077g,0.403mmol) in N, N-dimethylformamide (18mL) and purifying the crude by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 30/1) to give the title compound as a yellow solid (0.511 g, 67%).
MS(ESI,neg.ion)m/z:188.1[M-H]-
1H NMR(600MHz,DMSO-d6)δ(ppm):13.06(br.s,1H),8.08(s,2H),7.78-7.75(m,2H),7.72-7.68(m,1H),7.60-7.57(m,1H);
13C NMR(151MHz,DMSO-d6)δ(ppm):167.7,137.5,136.3,131.7,129.6,128.9,128.5,124.4.
Step 2) synthesis of 2- (2H-1,2, 3-triazole-2-yl) benzoyl chloride
The title compound of this step was prepared as described in example 1, step 5, by dissolving 2- (2H-1,2, 3-triazol-2-yl) benzoic acid (0.37g,1.96mmol) in dry dichloromethane (20mL), slowly adding thionyl chloride (6mL,82.7mmol) and pyridine (0.04mL,0.5mmol), and gradually heating to reflux for 3H. The reaction was stopped, cooled, and the solvent was slowly evaporated under reduced pressure, and the resulting product was used directly in the next reaction.
Step 3) synthesis of N- (2- (naphthalene-1-yl) ethyl) -2- (2H-1,2, 3-triazole-2-yl) benzamide
The title compound was prepared as described in example 1, step 6, by reacting 2- (naphthalen-1-yl) ethylamine (0.260g,1.52mmol), 2- (2H-1,2, 3-triazol-2-yl) benzoyl chloride (0.405g,1.95mmol) with triethylamine (0.85mL,6.0mmol) and dichloromethane (10mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a yellow solid (0.279 g, 54%).
MS(ESI,pos.ion)m/z:342.9[M+H]+
1H NMR(600MHz,CDCl3)δ(ppm):8.11(d,J=8.4Hz,1H),7.87(d,J=7.5Hz,1H),7.77-7.73(m,2H),7.72(s,2H),7.57(dd,J=7.6,1.4Hz,1H),7.56-7.52(m,2H),7.51-7.48(m,1H),7.44(td,J=7.6,1.1Hz,1H),7.40-7.36(m,1H),7.30(d,J=6.8Hz,1H),5.86(s,1H),3.75(q,J=7.0Hz,2H),3.32(t,J=7.1Hz,2H);
13C NMR(151MHz,CDCl3)δ(ppm):167.6,137.0,135.8,134.8,134.0,132.0,131.4,130.6,129.1,128.8,128.7,127.4,126.6,126.3,125.8,125.5,124.3,123.7,40.6,32.5.
Example 6N-Ethyl-N- (2- (naphthalen-1-yl) ethyl) -2- (2H-1,2, 3-triazol-2-yl) benzamide
Figure BDA0001012285000000271
This step was performed as described in example 2 for the title compound prepared by reacting N- (2- (naphthalen-1-yl) ethyl) -2- (2H-1,2, 3-triazol-2-yl) benzamide (0.171g,0.5mmol), bromoethane (0.06mL,0.75mmol) in 60% sodium hydride (0.05g,1.25mmol) and N, N-dimethylformamide (5mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a pale yellow viscous material (0.171g, 92%).
MS(ESI,pos.ion)m/z:371.0[M+H]+.
Example 7N- (2- (4-fluoronaphthalen-1-yl) ethyl) -2- (2H-1,2, 3-triazol-2-yl) benzamide
Figure BDA0001012285000000272
The title compound was prepared as described in example 1, step 6, by reacting 2- (4-fluoronaphthalen-1-yl) ethylamine (0.159g,0.84mmol), 2- (2H-1,2, 3-triazol-2-yl) benzoyl chloride (0.227g,1.09mmol) with triethylamine (0.473mL,3.36mmol) and dichloromethane (10mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a pale yellow solid (0.261 g, 86%).
MS(ESI,pos.ion)m/z:360.9[M+H]+
1H NMR(600MHz,CDCl3)δ(ppm):8.14(d,J=7.8Hz,1H),8.11(d,J=8.3Hz,1H),7.76(dd,J=8.1,0.9Hz,1H),7.73(s,2H),7.62-7.52(m,4H),7.45(td,J=7.6,1.1Hz,1H),7.22(dd,J=7.8,5.4Hz,1H),7.04(dd,J=10.3,7.8Hz,1H),5.88(s,1H),3.70(q,J=7.1Hz,2H),3.27(t,J=7.2Hz,2H);
13C NMR(151MHz,CDCl3)δ(ppm):167.6,158.9,157.2,137.0,135.8,133.1(d,J=4.4Hz),131.3,130.7,129.1,128.7,127.2,126.2(d,J=8.2Hz),126.1(d,J=1.8Hz),124.4,124.2,124.1,123.8(d,J=2.7Hz),121.3(d,J=5.8Hz),108.9,108.8,40.7,32.1.
Example 8N-Ethyl-N- (2- (4-fluoronaphthalen-1-yl) ethyl) -2- (2H-1,2, 3-triazol-2-yl) benzamide
Figure BDA0001012285000000281
The title compound was prepared as described in example 2 by reacting N- (2- (4-fluoronaphthalen-1-yl) ethyl) -2- (2H-1,2, 3-triazol-2-yl) benzamide (0.18g,0.5mmol), bromoethane (0.06mL,0.75mmol) in 60% sodium hydride (0.05g,1.25mmol) and N, N-dimethylformamide (5mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a pale yellow viscous mass, 0.187g, 96%).
MS(ESI,pos.ion)m/z:389.3[M+H]+.
Example 9N- (2- (4-Methoxynaphthalen-1-yl) ethyl) -5-methyl-2- (2H-1,2, 3-triazol-2-yl) benzamide
Figure BDA0001012285000000282
Step 1 Synthesis of (E) -1-methoxy-4- (2-nitrovinyl) naphthalene
The title compound was prepared as described in example 1, step 1, by reacting 4-methoxy-1-naphthaldehyde (0.930g,5.0mmol), ammonium acetate (0.578g,7.5mmol) in nitromethane (25mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 150/1) to give the title compound as a yellow solid (0.973 g, 85%).
MS(ESI,pos.ion)m/z:230.2[M+H]+.
Step 2) Synthesis of 1-methoxy-4- (2-nitroethyl) naphthalene
The title compound was prepared by the method described in example 1, step 2, i.e. (E) -1-methoxy-4- (2-nitrovinyl) naphthalene (0.515g,2.25mmol), sodium borohydride (0.217g,5.62mmol) in a mixed solvent of tetrahydrofuran (9mL) and anhydrous methanol (36mL), and the crude product was isolated and purified by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 150/1) to give the title compound (yellow liquid, 0.26g, 50%).
1H NMR(600MHz,CDCl3)δ(ppm):8.16(d,J=8.2Hz,1H),7.99(d,J=8.4Hz,1H),7.67-7.63(m,1H),7.60(t,J=7.3Hz,1H),7.31(dd,J=7.8,5.3Hz,1H),7.09(dd,J=10.1,7.9Hz,1H),4.71-4.69(m,2H),3.83(s,3H)3.76(t,J=7.6Hz,2H).
Step 3) Synthesis of 2- (4-methoxynaphthalen-1-yl) ethylamine
The title compound was prepared as described in example 1, step 3, by reacting 1-methoxy-4- (2-nitroethyl) naphthalene (0.208g,0.9mmol), 10% palladium on charcoal (0.02g) in dry methanol (10mL) under hydrogen, and purifying the crude product by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 20/1) to give the title compound (yellow liquid, 0.163g, 90%).
MS(ESI,pos.ion)m/z:202.5[M+H]+.
Step 4) N- (2- (4-methoxy naphthalene-1-yl) ethyl) -5-methyl-2- (2H-1,2, 3-triazole-2-yl) benzyl Synthesis of amides
The title compound was prepared as described in example 1, step 6, by reacting 2- (4-methoxynaphthalen-1-yl) ethylamine (0.44g,2.2mmol), 5-methyl-2- (2H-1,2, 3-triazol-2-yl) benzoyl chloride (0.585g,2.64mmol) with triethylamine (0.76mL,5.4mmol) and dichloromethane (20mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 4/1) to give the title compound as a white solid (0.255 g, 30%).
MS(ESI,pos.ion)m/z:387.3(M+);
1H NMR(400MHz,CDCl3)δ(ppm):8.16(dd,J=15.2,7.6Hz,2H),7.73(s,2H),7.65-7.55(m,3H),7.40(s,1H),7.37(d,J=8.2Hz,1H),7.21(dd,J=7.6,5.4Hz,1H),7.06(dd,J=10.2,8.0Hz,1H),5.91(s,1H),3.84(s,3H),3.68(dd,J=13.4,6.8Hz,2H),3.27(t,J=7.2Hz,2H),2.43(s,3H).
Example 10N-Ethyl-N- (2- (4-methoxynaphthalen-1-yl) ethyl) -5-methyl-2- (2H-1,2, 3-triazol-2-yl) benzamide
Figure BDA0001012285000000291
The title compound was prepared as described in example 2 by reacting N- (2- (4-methoxynaphthalen-1-yl) ethyl) -5-methyl-2- (2H-1,2, 3-triazol-2-yl) benzamide (0.146g,0.39mmol), ethyl bromide (0.055g,0.5mmol) in 60% sodium hydride (0.05g,1.25mmol) and N, N-dimethylformamide (3mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 4/1) to give the title compound as a pale yellow viscous mass, 0.11g, 65%).
MS(ESI,pos.ion)m/z:415.5[M+H]+.
Biological assay
1Example A antagonism of humanized OX receptors
Test method
Evaluation of compounds on humanized OX transfected in Chinese Hamster Ovary (CHO) cells using fluorescence detection of free calcium ion concentration in cytoplasm1Antagonistic ability of the receptor. Cells were suspended in cell culture medium (invitrogen) and then plated at 2X 104The density of cells/well is averaged into the microwell reaction plate. After mixing the fluorescent probe (Fluo4NW, Invitrogen) with probenecid Hank's balanced salt solution (Invitrogen), 20mM hydroxyethylpiperazine ethanethiosulfonic acid (Invitrogen) (pH7.4) was added and finally added to the microwells containing the cells. Cells were incubated at 37 ℃ for 60min and then equilibrated at 22 ℃ for 15 min. The plates were placed in a microplate reader (CellLux, PerkinElmer), test compound solutions of different concentrations or Hank's balanced salt solution were added, incubated for 5min and 3nM of orexin a or balanced salt solution (control) was added, and the change in fluorescence intensity, which was proportional to the intracytoplasmic calcium ion concentration, was measured.
Data analysis
Experimental results Each compound was recorded with a basal control of 0 and orexin A of 3nM as 100%Inhibition rate of the substance. In each experiment, several different concentrations are measured respectively, dose-response curves are made, and IC of each compound is calculated50The value is obtained.
The results of the tests show that the compounds of the invention are on OX in the above tests1The receptor has better antagonism.
2Example B antagonism of humanized OX receptors
Test method
Evaluation of Compounds on humanized OX transfected in HEK-293 cells Using fluorescence detection of free calcium ion concentration in cytoplasm2Antagonistic ability of the receptor. Cells were suspended in cell culture medium (invitrogen) and then plated at 3X 104The density of cells/well is averaged into the microwell reaction plate. After mixing the fluorescent probe (Fluo4NW, Invitrogen) with probenecid Hank's balanced salt solution (Invitrogen), 20mM hydroxyethylpiperazine ethanethiosulfonic acid (Invitrogen) (pH7.4) was added and finally added to the microwells containing the cells. Cells were incubated at 37 ℃ for 60min and then equilibrated at 22 ℃ for 15 min. The plates were placed in a microplate reader (CellLux, PerkinElmer), test compound solutions of different concentrations or Hank's balanced salt solution were added, incubated for 5min and 10nM orexin B or balanced salt buffer (control) was added, and the change in fluorescence intensity, which was proportional to the cytosolic calcium ion concentration, was measured.
Data analysis
Results the inhibition of each compound was recorded at 0 for the control and 100% for 10nM orexin B. In each experiment, several different concentrations are measured respectively, dose-response curves are made, and IC of each compound is calculated50The value is obtained. See table 1 for results.
TABLE 1 Compound pairs OX as provided in the examples of the invention2Results of receptor antagonism experiments
Figure BDA0001012285000000301
The results of the tests show that the compounds of the invention are on OX in the above tests2The receptor has better antagonism.
Example C pharmacokinetic evaluation of rat, dog and monkey following intravenous or oral quantification of the Compounds of the invention
The present invention evaluates pharmacokinetic studies of the compounds of the invention in rats, dogs or monkeys. The compounds of the invention were administered as 5% DMSO + 5% Kolliphor HS 15+ 2% (2% HCl) + 88% Saline solution or 10% DMSO + 10% Kolliphor HS 15+ 80% Saline solution. For intravenous administration, animals are given a dose of 1mg/kg, bled (0.3mL) at time points of 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24 hours, and centrifuged at 3,000 or 4,000rpm for 10 minutes. For oral (p.o.) administration, animals were given a dose of 5mg/kg, bled (0.3mL) at time points of 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, and 24 hours, and centrifuged at 3,000 or 4,000rpm for 10 minutes. Plasma solutions were collected and stored at-20 ℃ or-70 ℃ until LC/MS/MS analysis.
Test results show that the compound has better pharmacokinetic properties in rats, dogs or monkeys.
Finally, it should be noted that there are other ways of implementing the invention. Accordingly, the embodiments of the present invention will be described by way of illustration, but not limitation to the description of the present invention, and modifications made within the scope of the present invention or equivalents added to the claims are possible. All publications or patents cited herein are incorporated by reference.

Claims (9)

1. A compound which is a compound represented by formula (I) or a stereoisomer, tautomer, or pharmaceutically acceptable salt of a compound represented by formula (I),
wherein X is
Figure FDA0002227762620000012
The group being linked via the E-terminus toPhenylene in the structure shown in the formula (I) is connected;
each R1Independently is H, D, F, Cl, Br, I or C1-4An alkoxy group;
each R2And R3Independently is H;
each R4Independently is H, D, F, Cl, Br, I or C1-4Alkyl radical, wherein, C1-4Alkyl is optionally substituted by 1,2,3,4 or 5R6Substituted by a group;
R5is H, D, C1-4Alkyl or C1-4A haloalkyl group;
each R6Independently is D, F, Cl, Br or I;
m is 0,1, 2,3,4, 5, 6 or 7;
n is 0,1, 2,3 or 4;
p is 1,2,3 or 4;
k is 0,1 or 2.
2. The compound of claim 1, wherein each R1Independently H, F, Cl, Br, methoxy or ethoxy.
3. The compound of claim 1, wherein each R4Independently H, F, Cl, methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl or trifluoromethyl.
4. The compound of claim 1, having the structure of one of:
Figure FDA0002227762620000013
Figure FDA0002227762620000021
or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof.
5. A pharmaceutical composition comprising a compound of any one of claims 1-4, and a pharmaceutically acceptable excipient, carrier, adjuvant, or combination thereof.
6. The pharmaceutical composition according to claim 5, further comprising other agents for the prevention or treatment of CNS neurological and psychiatric disorders and diseases, which are antidepressants, anxiolytics, lithium salts as affective stabilizers, antipsychotics, antiepileptics, antiparkinson drugs, sedative hypnotics, antihistamines, GABA receptor agonists and/or GABA reuptake inhibitors, drugs as monoamine oxidase inhibitors, drugs as melatonin receptor agonists and drugs as orexin receptor antagonists or any combination thereof.
7. Use of a compound according to any one of claims 1 to 4 or a pharmaceutical composition according to any one of claims 5 to 6 in the manufacture of a medicament for the prevention, treatment or alleviation of central nervous system neurological and psychiatric disorders and diseases in mammals, including humans.
8. The use according to claim 7, wherein the neurological and psychiatric disorders and diseases of the central nervous system are depression, anxiety disorders, seasonal affective disorders, mania, bipolar disorders, obsessive-compulsive disorders, insomnia and fatigue due to jet lag, schizophrenia, convulsions, panic attacks, melancholia, alcohol addiction, drug addiction, alcoholism, substance abuse, drug addiction withdrawal symptoms, insomnia, psychotic disorders, epilepsy, sleep disorders, sleep disturbances, sleep apnea syndrome, compulsive eating disorders, fibromyalgia, stress responses, obesity, Parkinson's disease, senile dementia, cognitive disorders, memory disorders, premenstrual tension syndrome, migraine, memory loss, Alzheimer's disease or other disorders associated with normal or pathological aging.
9. Use of a compound according to any one of claims 1 to 4 or a pharmaceutical composition according to any one of claims 5 to 6 in the manufacture of a medicament for selectively antagonizing an orexin receptor.
CN201610404146.1A 2015-06-09 2016-06-08 Substituted heterocyclic compounds, process for their preparation and their use Active CN106243052B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2015103118781 2015-06-09
CN201510311878 2015-06-09

Publications (2)

Publication Number Publication Date
CN106243052A CN106243052A (en) 2016-12-21
CN106243052B true CN106243052B (en) 2020-01-21

Family

ID=57612928

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201610404146.1A Active CN106243052B (en) 2015-06-09 2016-06-08 Substituted heterocyclic compounds, process for their preparation and their use

Country Status (1)

Country Link
CN (1) CN106243052B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106674207B (en) * 2016-12-09 2019-04-19 广东东阳光药业有限公司 Substituted aryl heteroaryl compound and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101155792A (en) * 2005-04-12 2008-04-02 默克公司 Amidopropoxyphenyl orexin receptor antagonists
JP2014015452A (en) * 2012-06-15 2014-01-30 Taisho Pharmaceutical Co Ltd Medicine containing pyrazole derivative
CN104364238A (en) * 2012-06-15 2015-02-18 大正制药株式会社 Branched chain alkyl heteroaromatic ring derivative

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2012519682A (en) * 2009-03-03 2012-08-30 ファイザー・インク Novel phenylimidazoles and phenyltriazoles as gamma-secretase modulators

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101155792A (en) * 2005-04-12 2008-04-02 默克公司 Amidopropoxyphenyl orexin receptor antagonists
JP2014015452A (en) * 2012-06-15 2014-01-30 Taisho Pharmaceutical Co Ltd Medicine containing pyrazole derivative
CN104364238A (en) * 2012-06-15 2015-02-18 大正制药株式会社 Branched chain alkyl heteroaromatic ring derivative

Also Published As

Publication number Publication date
CN106243052A (en) 2016-12-21

Similar Documents

Publication Publication Date Title
JP7299837B2 (en) Compounds, compositions and methods of use
US11420955B2 (en) Compounds as neuronal histamine receptor-3 antagonists and uses thereof
CN109574993B (en) Substituted pyrimidinylpiperazine compounds and uses thereof
US11639337B2 (en) Bicyclic heteroaryl derivatives and preparation and uses thereof
CN111770914B (en) Compounds as neurokinin-1 receptor antagonists and uses thereof
EP3144308B1 (en) Nitrogen-containing heterocyclic compound
KR20110096032A (en) Organic compounds
BR112015009168B1 (en) COMPOUND OF STRUCTURAL FORMULA XI OR A SALT THEREOF, USE OF A COMPOUND AND PHARMACEUTICAL COMPOSITION
TW201206937A (en) Organic compounds
CN107106563B (en) Compounds and methods
CN106831614B (en) Substituted benzodiazacyclo compound and its preparation method and use
CN107759620B (en) Octahydropyrrolo [3,4-c ] pyrrole derivatives, methods of use, and uses thereof
JP6849668B2 (en) Pyridine or pyrimidine derivatives
CN109988170B (en) Octahydropyrrolo [3,4-c ] pyrrole derivatives and uses thereof
CN106986859B (en) Indole derivatives and uses thereof
BR112018015191B1 (en) 6,7-DI-HYDRO-5H-PYRAZOLO[5,1-B][1,3]OXAZINE-2-CARBOXAMIDE COMPOUNDS, THEIR USE AND PHARMACEUTICAL COMPOSITION COMPRISING THEM
AU2016295693B2 (en) Substituted quinazoline compounds and preparation and uses thereof
CN108299437B (en) Octahydropyrrolo [3,4-c ] pyrrole derivatives and uses thereof
CN106243052B (en) Substituted heterocyclic compounds, process for their preparation and their use
JP7447020B2 (en) Triazole, imidazole and pyrrole-fused piperazine derivatives and their use as modulators of mGlu5 receptors
CN106065018B (en) Substituted indole compounds, methods of use and uses thereof
CN112679497B (en) Azaindole amide compound and preparation method and application thereof
CA3068209A1 (en) Heterocyclic compound
CN109956945B (en) Octahydropyrrolo [3,4-c ] pyrrole derivatives and uses thereof

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
CP03 Change of name, title or address
CP03 Change of name, title or address

Address after: 523808 No.1, Gongye North Road, Songshanhu Park, Dongguan City, Guangdong Province

Patentee after: Guangdong Dongyangguang Pharmaceutical Co.,Ltd.

Address before: 523808 No. 1 Industrial North Road, Songshan Industrial Park, Songshan, Guangdong, Dongguan, Hubei

Patentee before: SUNSHINE LAKE PHARMA Co.,Ltd.