Aminoadamantane nitrate compounds and their use for treating CNS disorders

文档序号:1342590 发布日期:2020-07-17 浏览:21次 中文

阅读说明:本技术 硝酸氨基金刚烷酯化合物和它们用于治疗cns病症的用途 (Aminoadamantane nitrate compounds and their use for treating CNS disorders ) 是由 C.K.贝克 M.S.文卡特拉曼 X.张 J.W.拉里克 于 2018-11-20 设计创作,主要内容包括:本公开提供了金刚烷基化合物,其具有一个或多个胺基和一个或多个硝酸酯基。所述硝酸氨基金刚烷酯化合物可以用于治疗中枢神经系统的病症,包括神经退行性疾病和非神经退行性疾病。(The present disclosure provides adamantyl compounds having one or more amine groups and one or more nitrate groups. The aminoadamantyl nitrate compounds are useful for treating disorders of the central nervous system including neurodegenerative and non-neurodegenerative diseases.)

1. A compound comprising formula I:

wherein:

R1and R2Independently hydrogen, a halide, a linear or branched alkyl, a linear or branched heteroalkyl, a linear or branched alkoxy, a linear or branched-O-heteroalkyl, a cycloalkyl, a heterocyclyl, an aryl, or a heteroaryl, each of which may be optionally substituted;

R3and R4Independently hydrogen or straight or branched C1-C6Alkyl, or R3、R4And the nitrogen atom to which they are attached form a 3-to 8-membered hetero atomA ring;

R5is hydrogen or straight or branched C1-C6An alkyl group;

x is a bond, straight or branched-alkyl-, straight or branched-heteroalkyl-, straight or branched-O-alkyl-, straight or branched-O-heteroalkyl-, - (CH)2)j-cycloalkyl- (CH)2)k-、-(CH2)j-heterocyclyl- (CH)2)k-、-(CH2)j-aryl- (O)h-(CH2)k-or- (CH)2)j-heteroaryl- (O)h-(CH2)k-, each of which may be optionally substituted;

y is-ONO2Or

m is 0, 1,2, 3, 4 or 5;

j is 0, 1,2 or 3;

k is 0, 1,2 or 3; and is

h is 0 or 1;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

2. The compound of claim 1, wherein R3Is hydrogen, R4Is hydrogen, and R5Is hydrogen to give formula Ia:

wherein:

n is 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

3. The compound of claim 2, wherein n is 1, R1Is methyl, R2Is methyl, X is- (CH)2) -, and Y is ONO2

4. The compound of claim 2, wherein n is 2, R1Is methyl, R2Is methyl, X is propyl and Y is ONO2

5. The compound of claim 2, wherein n is 1, R1Is methyl, R2Is methyl, X is a bond, and Y is ONO2

6. The compound of claim 1, wherein y is ONO2To give formula IA:

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

7. The compound of claim 6, wherein R3Is hydrogen, R4Is hydrogen, and R5Is hydrogen to give formula IAa:

wherein:

n is 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

8. The compound of claim 1,2 or 6, wherein X of the compound of formula I, Ia, IA or IAa is a bond, straight or branched C1-C6Or C1-C3Alkyl-or straight or branched C1-C6Or C1-C3-O-alkyl-.

9. The compound of claim 1, wherein the compound is selected from the group consisting of:

10. a compound comprising formula II:

wherein:

R1and R2Independently hydrogen, a halide, a linear or branched alkyl, a linear or branched heteroalkyl, a linear or branched alkoxy, a linear or branched-O-heteroalkyl, a cycloalkyl, a heterocyclyl, an aryl, or a heteroaryl, each of which may be optionally substituted;

R3and R4Independently hydrogen or straight or branched C1-C6Alkyl, or R3、R4And the nitrogen atom to which they are attached form a 3-to 8-membered heterocyclic ring;

R5is hydrogen or straight or branched C1-C6An alkyl group;

x is a bond, straight or branched-alkyl-, straight or branched-heteroalkyl-, straight or branched-O-alkyl-, straight or branched-O-heteroalkyl-, - (CH)2)j-, cycloalkyl- (CH)2)k-、-(CH2)j-heterocyclyl- (CH)2)k-、-(CH2)j-aryl- (O)h-(CH2)k-or- (CH)2)j-heteroaryl- (O)h-(CH2)k-, each of which may be optionally substituted;

y is-ONO2Or

m is 0, 1,2, 3, 4 or 5;

j is 0, 1,2 or 3;

k is 0, 1,2 or 3; and is

h is 0 or 1;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

11. The compound of claim 10, wherein R3Is hydrogen, R4Is hydrogen, and R5Is hydrogen to give formula IV:

wherein:

p is 0, 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

12. The compound of claim 11, wherein R1Is hydrogen, R2Is hydrogen to give formula IVa:

wherein:

p is 0, 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

13. The compound of claim 12, wherein p is 1, X is a bond, and Y is ortho-ONO2

14. The compound of claim 12, wherein p is 1 and X is ortho O- (CH)2)3-, and Y is ortho-ONO2

15. The compound of claim 11, wherein Y is-ONO2To obtain formula IVA:

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

16. The compound of claim 15, wherein R1Is hydrogen and R2Is hydrogen to give formula IVAa:

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

17. The compound of claim 10, wherein Y isTo give formula IIIB:

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

18. The compound of claim 17, wherein R3Is hydrogen, R4Is hydrogen, and R5Is hydrogen to give formula IVB:

wherein:

p is 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

19. The compound of claim 18, wherein R1Is hydrogen and R2Is hydrogen to give formula IVBa:

wherein:

p is 1,2, 3 or 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

20. The compound of any one of claims 10, 11, 12, 15, 16, 17, 18, or 19, wherein X is a bond, straight or branched C1-C6Or C1-C3Alkyl-or straight or branched C1-C6Or C1-C3-O-alkyl.

Background

The N-methyl-D-aspartate receptor (also known as NMDA receptor or NMDAR) is an excitatory glutamate receptor and ion channel protein found in neurons in the Central Nervous System (CNS). Activation of NMDA receptors requires binding of glutamate (or aspartate or NMDA, both of which are weaker stimulators) and glycine (or D-serine, a stronger co-agonist) released following presynaptic neuronal depolarization to effectively open the ion channel portion of the receptor. Activation of the NMDAR produces excitatory postsynaptic potentials that cause the opening of transmembrane ion channels and the flow of non-selective cations through it. Although the opening and closing of ion channels is primarily gated by ligand binding, the current flowing through an ion channel is voltage dependent. Extracellular magnesium (Mg)2+) Ions can bind to allosteric sites in the NMDAR channel at the resting membrane potential, blocking the passage of other cations through the open ion channel. Postsynaptic membrane depolarization in milliseconds mediated by another type of ion channel glutamate receptor, AMPA, will be driven out of the pore channel and repel Mg2+Ion, thereby allowing sodium (Na)+) Ions and calcium (Ca)2+) Ion influx into cells and potassium (K)+) Voltage-dependent flow of ions out of the cell. Ca2+The influx of (a) triggers intracellular signaling pathways, in which Ca is present2+Acting as a second messenger.

The ion channel of the NMDA receptor is only bound to the receptor when the co-agonists glutamate and glycine and the postsynaptic membrane is depolarized to remove the co-agonist Mg2+The resulting voltage-dependent channel is blocked and is opened and remains open. This property of the NMDA receptor is an important cellular mechanism for synaptic plasticity and supporting its long-term enhancement. NMDAR-mediated neurotransmission is a potentially major inter-neuronal communication of synaptic plasticity.

NMDA receptors are located at and outside synapses. The proportion of synaptic NMDARs increases with development, although there is still a significant amount of extra-synaptic NMDARs after adulthood. Ca2+Influx of NMDAR through activated synapses for controlling synaptic plasticity as a basis for memory, learning and neural network formation during CNS developmentSex and synapse formation are important. However, in pathological conditions, excessive extracellular glutamate levels can cause overstimulation of the extra-synaptic NMDAR and sustained depolarization of neurons, resulting in excessive Ca2+Flows into the neuron. Intracellular Ca2+Excessive concentrations disrupt calcium homeostasis and initiate a cascade of signaling pathways leading to upregulation of neuronal nitric oxide synthase, mitochondrial dysfunction, reactive oxygen species generation, dysregulation of oxidative phosphorylation, endoplasmic reticulum stress, release of lysosomal enzymes, and ultimately neuronal death. Resulting in excessive Ca2+Excessive activation of the inflowing extra-synaptic NMDAR may lead to excitotoxicity involving neurodegenerative disorders such as Alzheimer's disease, Huntington's disease and Parkinson's disease. Alzheimer's disease is the most common form of neurodegenerative disorders and dementia, afflicting at least 1800 million people worldwide, and will become more prevalent as the number of elderly people increases.

Disclosure of Invention

The present disclosure provides aminoadamantyl nitrate compounds that are selective, noncompetitive antagonists of activated extrasynaptic NMDA receptors. In some embodiments, the aminoadamantyl nitrate compound has the formula I:

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs, and stereoisomers thereof, wherein Y is a nitrate-containing group, and R is1、R2、R3、R4、R5X and m are as defined elsewhere herein.

In other embodiments, the aminoadamantyl nitrate has the formula II and formula III:

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs, and stereoisomers thereof, wherein Y is a nitrate-containing group, and R is1、R2、R3、R4、R5X and m are as defined elsewhere herein.

The aminoadamantyl nitrate compounds can be used to treat a wide range of neurodegenerative and other CNS disorders, including alzheimer's disease, vascular dementia, huntington's disease, parkinson's disease, cerebral ischemia, traumatic brain injury, epilepsy, and autism spectrum disorders.

Drawings

A better understanding of the features and advantages of the present disclosure will be obtained by reference to the following detailed description and accompanying drawings that set forth illustrative embodiments of the disclosure.

Figure 1 depicts the synthesis of various aminoadamantane nitrate ester compounds.

Detailed Description

While various embodiments of the present disclosure have been described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Many modifications and variations of the embodiments described herein, as well as variations and substitutions, will be apparent to those skilled in the art without departing from the disclosure. It should be understood that various alternatives to the embodiments described herein may be employed in practicing the disclosure. It should also be understood that each embodiment of the present disclosure may optionally be combined with any one or more of the other embodiments described herein consistent with that embodiment.

Where elements are presented in a list, for example in a Markush group, it will be appreciated that each possible sub-group of the elements is also disclosed, and that any one or more elements may be removed from the list or group.

It should also be understood that, unless clearly indicated to the contrary, in any methods described or claimed herein that include more than one action or step, the order of the actions or steps of the method is not necessarily limited to the order in which the actions or steps of the method are recited, but the disclosure encompasses embodiments in which the order is so limited.

It should also be understood that in general, where an embodiment in the specification or claims is referred to as including one or more features, the disclosure also encompasses embodiments consisting of, or consisting essentially of, the one or more features.

It should also be understood that any embodiment of the present disclosure, for example, any embodiment found in the prior art, may be explicitly excluded from the claims, whether or not a specific exclusion is recited in the specification.

Headings are included herein for reference and to aid in locating certain sections. The headings are not intended to limit the scope of the embodiments and concepts described in the sections below those headings, and those embodiments and concepts may be applicable in other sections throughout the disclosure.

All patent documents and all non-patent documents cited herein are incorporated by reference in their entirety to the same extent as if each patent document or non-patent document were specifically and individually indicated to be incorporated by reference in its entirety.

I.Definition of

The indefinite articles "a" and "an" as used in this specification and the appended claims may include plural referents and singular referents unless expressly stated otherwise or the context clearly dictates otherwise.

The term "about" or "approximately" means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term "about" or "approximately" means within one standard deviation. In some embodiments, when a specific error range is not recited (e.g., standard deviation from mean as given in a chart or data table), the term "about" or "approximately" is intended to cover the stated range of values and the range to be included by rounding up or down to the stated value, in view of the significant figures. In certain embodiments, the term "about" or "approximately" means within 20%, 15%, 10%, or 5% of the specified value. The term "about" or "approximately" applies to a series of two or more numerical values or a series of two or more numerical ranges when preceded by the first numerical value.

The term "at least" or "greater than" applies to each numerical value in a series of two or more numerical values when the term "at least" or "greater than" precedes the first numerical value in the series.

The term "not greater than" or "less than" applies to each numerical value in a series of two or more numerical values when preceded by the first numerical value.

The term "pharmaceutically acceptable" refers to a substance (e.g., an active ingredient or excipient) that is suitable for use in contact with the tissues and organs of a subject without excessive irritation, allergic response, immunogenicity, and toxicity, commensurate with a reasonable benefit/risk ratio, and effective for its intended use. The "pharmaceutically acceptable" excipient or carrier of the pharmaceutical composition is also compatible with the other ingredients of the composition.

The term "therapeutically effective amount" refers to an amount of a compound that, when administered to a subject, is sufficient to prevent the occurrence of, or alleviate to some extent, the medical condition being treated or one or more symptoms associated with the condition. The term "therapeutically effective amount" also refers to an amount of a compound that is sufficient to elicit the biological or medical response of a cell, tissue, organ, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or clinician.

The terms "treat", "treating" and "treatment" include reducing or eliminating a medical disorder or one or more symptoms associated with the disorder and reducing or eradicating one or more causes of the disorder. Reference to "treating" a disorder is intended to include preventing the disorder. The terms "preventing", "preventing" and "prevention" include preventing or delaying the onset of a medical condition or one or more symptoms associated with the condition, preventing the subject from acquiring the condition, and reducing the risk of the subject acquiring the condition. The term "medical condition" includes diseases and conditions.

The terms "disease" and "disorder" are used interchangeably herein.

The term "subject" refers to an animal, including, but not limited to, a mammal, such as a primate (e.g., a human, chimpanzee, or monkey), rodent (e.g., rat, mouse, guinea pig, gerbil, or hamster), lagomorph (e.g., rabbit), porcine (e.g., pig), equine (e.g., horse), canine (e.g., dog), or feline (e.g., cat). The terms "subject" and "patient" are used interchangeably herein when referring to, for example, a mammalian subject, such as a human subject.

The term "compound" encompasses salts, solvates, hydrates, clathrates and polymorphs of the compound. "solvates" of a compound include stoichiometric or non-stoichiometric solvents (e.g., water, acetone, or an alcohol [ e.g., ethanol ]) that are non-covalently bound to the compound. A "hydrate" of a compound includes stoichiometric or non-stoichiometric amounts of water that is non-covalently bound to the compound. A "clathrate" of a compound contains molecules of a substance (e.g., a solvent) enclosed in the crystal structure of the compound. A "polymorph" of a compound is a crystalline form of the compound. Unless the context clearly indicates otherwise, in certain instances of the present disclosure, a particular recitation of a "salt," "solvate," "hydrate," "clathrate," or "polymorph" of a compound should not be interpreted as an intent to omit any of these forms in other instances of the present disclosure where the term "compound" is used without recitation of any of these forms.

The terms "halogen", "halide" and "halo" refer to fluorine/fluoride, chlorine/chloride, bromine/bromide and iodine/iodide.

The term "alkyl" refers to a straight or branched chain saturated monovalent hydrocarbon group, wherein the alkyl group may be optionally substituted with one or more substituents as described herein. In certain embodiments, alkyl is of 1 to 10 (C)1-10) Or 1 to 6 (C)1-6) A straight-chain saturated monovalent hydrocarbon group of carbon atoms, or a saturated monovalent hydrocarbon group having 3 to 10 carbon atoms (C)3-10) Or 3 to 6 (C)3-6) Branched saturated monovalent hydrocarbon groups of carbon atoms. For example, the term "C1-6Alkyl "refers to a straight chain saturated monovalent hydrocarbon group having 1 to 6 carbon atoms or a branched chain saturated monovalent hydrocarbon group having 3 to 6 carbon atoms. Straight chain C1-6Alkyl and branched C3-6Alkyl groups may also be referred to as "lower alkyl". Non-limiting examples of alkyl groups include methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including all isomeric forms, such as n-butyl, isobutyl, sec-butyl and tert-butyl), pentyl (including all isomeric forms, such as n-pentyl), and hexyl (including all isomeric forms, such as n-hexyl).

The terms "alkylene" and "-alkyl-" refer to a divalent alkyl group, which may be optionally substituted with one or more substituents as described herein.

The term "heteroalkyl" refers to a straight or branched chain saturated monovalent hydrocarbon radical containing one or more heteroatoms independently selected from O, N and S. In some embodiments, one or more heteroatoms are in the backbone of the linear or branched hydrocarbon group. The terms "heteroalkylene" and "-heteroalkyl-" refer to a divalent heteroalkyl group. Heteroalkyl and-heteroalkyl-may be optionally substituted with one or more substituents as described herein. Examples of heteroalkyl and-heteroalkyl-include, but are not limited to- (CH)2)m- (O or S) - (CH)2)nCH3And- (CH)2)m- (O or S) - (CH)2)p-, where m is 1,2 or 3, n is 0, 1 or 2 and p is 1,2 or 3.

The term "alkoxy" refers to an-O-alkyl group, which may be optionally substituted with one or more substituents as described herein.

Examples of-O-heteroalkyl and-O-heteroalkyl-include, but are not limited to, ethylene glycol and polyethylene glycol (PEG) groups, including, but not limited to- (OCH)2CH2)n-OR and- (OCH)2CH2)n-O-, wherein R is hydrogen or alkyl and n is 1,2 or 3. It should be understood that for-O-heteroalkyl-ONO2A group, when-O-heteroalkyl-is an ethylene glycol or PEG group, the terminal oxygen atom of said ethylene glycol or PEG group is nitrate (-ONO)2) A portion of a group. -O-heteroalkyl and-O-heteroalkyl-may be optionally substituted with one or more substituents as described herein.

The term "haloalkyl" refers to an alkyl group substituted with one or more halogens/halogen atoms. The haloalkyl group can be optionally substituted with one or more additional substituents as described herein. Examples of haloalkyl groups include, but are not limited to, fluoroalkyl groups, such as-CH2F、-CHF2And- (CH)2)nCF3And perfluoroalkyl radicals, e.g. -CF3And- (CF)2)nCF3Wherein n is 1,2, 3, 4 or 5.

The term "-alkylaryl" refers to an alkyl group substituted with one or more aryl groups. The alkyl aryl group may be optionally substituted with one or more additional substituents as described herein.

The term "cycloalkyl" refers to a cyclic, saturated, bridged or non-bridged monovalent hydrocarbon radical, which may be optionally substituted with one or more substituents as described herein. In certain embodiments, the cycloalkyl group has 3 to 10 (C)3-10) Or 3 to 8 (C)3-8) Or 3 to 6 (C)3-6) A carbon atom. Non-limiting examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, norbornyl, decahydronaphthyl, and adamantyl. The term "-cycloalkyl-" refers to a divalent cycloalkyl group, which may be optionally substituted with one or more substituents as described herein.

The terms "heterocyclyl" and "heterocycle" refer to a monocyclic non-aromatic or polycyclic group containing at least one non-aromatic ring, at least one of which is non-aromaticThe aromatic ring contains one or more heteroatoms independently selected from O, N and S. A non-aromatic ring containing one or more heteroatoms may be linked or fused to one or more saturated, partially unsaturated, or aromatic rings. In certain embodiments, heterocyclyl or heterocyclic groups have from 3 to 10 or from 3 to 8 or from 3 to 6 ring atoms. In some embodiments, heterocyclyl or heterocyclic groups are monocyclic, bicyclic, or tricyclic systems, which may include fused or bridged ring systems, and in which nitrogen or sulfur atoms may be optionally oxidized, nitrogen atoms may be optionally quaternized, and one or more rings may be fully or partially saturated or aromatic. The heterocyclyl or heterocyclic group may be attached to the main structure at any heteroatom or carbon atom that results in the formation of a stable compound. Examples of heterocyclic or heterocyclic groups include, but are not limited to, azepinyl, azetidinyl, aziridinyl, benzodiazepineAlkyl, benzodioxolyl, benzofuranonyl, benzopyranoyl, benzotetrahydrofuranyl, benzothiophenyl, benzothiopyranyl, β -carbolinyl, chromanyl, decahydroisoquinolinyl, dihydrobenzisothiazinylOxazinyl, dihydrofuranyl, dihydropyranyl, dihydropyrazinyl, dihydropyridinyl, dihydropyrazolyl, dihydropyrimidinyl, dihydropyrrolyl, dioxolanyl, dithianyl, furanonyl, imidazolidinyl, imidazolinyl, indolinyl, indolizinyl, isobenzotetrahydrofuryl, isobenzotetrahydrothienyl, isobenzodihydropyranyl, isoindolinyl, isothiazolidinyl, and pyrazolidinylOxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, indolyl, or a pharmaceutically acceptable salt thereof,Oxazolidone group,Oxazolidinyl, oxiranyl, piperazinyl, piperidinyl, 4-piperidinonyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, tetrahydrofuranyl (tetrahydrofuryl), tetrahydrofuranyl (tetrahydrofuranyl) (oxolanyl), tetrahydroisoquinolinyl, tetrahydropyranyl, tetrahydrothienyl (tetrahydrothiophenyl/thiolanyl), thiomorpholinyl (thiomorpholinyl), thiazolidinyl, and 1,3, 5-trithianyl. The term "-heterocyclyl-" refers to divalent heterocyclic groups. Heterocyclyl or heterocyclic groups and-heterocyclyl-may be optionally substituted with one or more substituents as described herein.

The term "aryl" refers to a monocyclic aromatic hydrocarbon group or a polycyclic group containing at least one aromatic hydrocarbon ring. In certain embodiments, aryl groups have from 6 to 10 ring atoms. Non-limiting examples of aryl groups include phenyl, naphthyl, fluorenyl, azulenyl, anthracenyl, phenanthrenyl, biphenyl, and terphenyl. The arene ring of the aryl group may be linked or fused to one or more saturated, partially unsaturated, or aromatic rings, such as dihydronaphthyl, indenyl, indanyl, and tetrahydronaphthyl (tetralinyl). The term "-aryl-" refers to a divalent aromatic radical. Aryl and-aryl-may be optionally substituted with one or more substituents as described herein.

The term "heteroaryl" refers to a monocyclic aromatic group or a polycyclic group containing at least one aromatic ring, wherein at least one aromatic ring contains one or more heteroatoms independently selected from O, N and S. The heteroaromatic ring may be linked or fused to one or more saturated, partially unsaturated, or aromatic rings that may contain only carbon atoms or may contain one or more heteroatoms. The heteroaryl may be attached to the main structure at any heteroatom or carbon atom that results in the formation of a stable compound. In certain embodiments, heteroaryl groups have from 5 to 10 ring atoms. Examples of monocyclic heteroaryl groups include, but are not limited to, pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, and,Azolyl radical, isoOxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl (thienyl/thiophenyl),Oxadiazolyl, triazolyl, tetrazolyl, pyridyl, pyridonyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyridazinonyl and triazinyl groups. Non-limiting examples of bicyclic heteroaryls include indolyl, benzothiazolyl, benzothiadiazolylAzolyl, benzisoylAzolyl, benzothienyl (benzothienyl/benzothiophenyl), quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzotriazolyl, indolizinyl, benzofuranyl, isobenzofuranyl, chromonyl, coumarinyl, cinnolinyl, quinazolinyl, quinoxalinyl, indazolyl, naphthyridinyl, phthalazinyl, quinazolinyl, purinyl, pyrrolopyridyl, furopyridyl, thienopyridyl, dihydroisoindolyl, and tetrahydroquinolinyl. Examples of tricyclic heteroaryl groups include, but are not limited to, carbazolyl, benzindolyl, dibenzofuranyl, phenanthrolinyl, acridinyl, phenanthridinyl, xanthenyl, and phenothiazinyl. The term "-heteroaryl-" refers to a divalent heteroaryl group. Heteroaryl and-heteroaryl-may be optionally substituted with one or more substituents as described herein.

Each of the groups described herein (including but not limited to monovalent and divalent alkyl, heteroalkyl, -O-alkyl, -O-heteroalkyl, alkylaryl, cycloalkyl, heterocyclyl, aryl, and heteroaryl groups) may be optionally substituted, whether as a primary group or as a substituent, with one or more substituents. In certain embodiments, each group described herein may be optionally substituted with 1,2, 3, 4,5, or 6 substituents independentlyIs selected from the group consisting of halide, cyano, nitro, nitrate, hydroxy, mercapto (-SH), -NH2、-OR11、-SR11、-NR12R13、-C(=O)R11、-C(=O)OR11、-OC(=O)R11、-C(=O)NR12R13、-NR12C(=O)R11、-OC(=O)OR11、-OC(=O)NR12R13、-NR12C(=O)OR11、-NR11C(=O)NR12R13Alkyl, haloalkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl, wherein:

R11independently at each occurrence is hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl; and is

R12And R13Independently at each occurrence is hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, or R12And R13Form a heterocyclic or heteroaryl ring with the nitrogen atom to which they are attached.

II.Stereoisomers

It should be understood that the present disclosure encompasses all possible stereoisomers of the adamantyl compounds described herein having one or more stereocenters, including both enantiomers and all possible diastereomers in substantially pure form as well as mixtures of two enantiomers in any ratio (including racemic mixtures of enantiomers) and mixtures of two or more diastereomers in any ratio, and not just the particular stereoisomers as illustrated by the drawn structures or nomenclature. Some embodiments of the present disclosure relate to specific stereoisomers as indicated by the drawn structures or nomenclature. Unless the context clearly indicates otherwise, a specific recitation in certain instances of the present disclosure with respect to a phrase "or a stereoisomer thereof" or the like of a compound should not be interpreted as an intent to omit any of the other possible stereoisomers of that compound in other instances of the present disclosure where the term "compound" is used without recitation of the phrase "or a stereoisomer thereof" or the like.

III.Amino adamantane ester nitrate compound

The present disclosure provides novel aminoadamantyl nitrate compounds. In some embodiments, the compound has formula I:

wherein:

R1and R2Independently hydrogen, a halide, a linear or branched alkyl, a linear or branched heteroalkyl, a linear or branched alkoxy, a linear or branched-O-heteroalkyl, a cycloalkyl, a heterocyclyl, an aryl, or a heteroaryl, each of which may be optionally substituted;

R3and R4Independently hydrogen or straight or branched C1-C6Alkyl, or R3、R4And the nitrogen atom to which they are attached form a 3-to 8-membered heterocyclic ring;

R5is hydrogen or straight or branched C1-C6An alkyl group;

x is a bond, straight or branched-alkyl-, straight or branched-heteroalkyl-, straight or branched-O-alkyl-, straight or branched-O-heteroalkyl-, - (CH)2)j-cycloalkyl- (CH)2)k-、-(CH2)j-heterocyclyl- (CH)2)k-、-(CH2)j-aryl- (O)h-(CH2)k-or- (CH)2)j-heteroaryl- (O)h-(CH2)k-, each of which may be optionally substituted;

y is-ONO2Or

m is 0, 1,2, 3, 4 or 5;

j is 0, 1,2 or 3;

k is 0, 1,2 or 3; and is

h is 0 or 1;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

In certain embodiments, the compound has formula Ia:

wherein:

R1、R2x and Y are as defined for formula I; and is

n is 1,2, 3, 4,5 or 6;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

In some embodiments, the compound has formula IA:

wherein R is1、R2、R3、R4、R5X and m are as defined for formula I;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

In certain embodiments, the compound has the formula IAa:

wherein:

R1、R2and X is as defined for formula I; and is

n is 1,2, 3, 4,5 or 6;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

In other embodiments, the compound has formula IB:

wherein R is1、R2、R3、R4、R5X and m are as defined for formula I;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof. The stereocenter of the vicinal dinitrate moiety may have R-or S-type stereochemistry or may be racemic.

In certain embodiments, the compound has formula IBa:

wherein:

R1、R2and X is as defined for formula I; and is

n is 1,2, 3, 4,5 or 6;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof. The stereocenter of the vicinal dinitrate moiety may have R-or S-type stereochemistry or may be racemic.

In some embodiments, X of the compounds of formula I and subclasses thereof is a bond, straight or branched C1-C6Or C1-C3Alkyl-or straight or branched C1-C6Or C1-C3-O-alkyl-. In certain embodiments, X of the compounds of formula I and subclasses thereof is a bond or straight or branched C1-C3Alkyl- [ e.g. -CH2-、-(CH2)2-、-CHCH3、-(CH2)3-、-CHCH2CH3、-CH2CHCH3or-CH (CH)3)CH2-]。

In other embodiments, the aminoadamantyl nitrate compound has the formula II and formula III:

wherein:

R1and R2Independently hydrogen, a halide, a linear or branched alkyl, a linear or branched heteroalkyl, a linear or branched alkoxy, a linear or branched-O-heteroalkyl, a cycloalkyl, a heterocyclyl, an aryl, or a heteroaryl, each of which may be optionally substituted;

R3and R4Independently hydrogen or straight or branched C1-C6Alkyl, or R3、R4And the nitrogen atom to which they are attached form a 3-to 8-membered heterocyclic ring;

R5is hydrogen or straight or branched C1-C6An alkyl group;

x is a bond, straight or branched-alkyl-, straight or branched-heteroalkyl-, straight or branched-O-alkyl-, straight or branched-O-heteroalkyl-, - (CH)2)j-cycloalkyl- (CH)2)k-、-(CH2)j-heterocyclyl- (CH)2)k-、-(CH2)j-aryl- (O)h-(CH2)k-or- (CH)2)j-heteroaryl- (O)h-(CH2)k-, each of which may be optionally substituted;

y is-ONO2Or

m is 0, 1,2, 3, 4 or 5;

j is 0, 1,2 or 3;

k is 0, 1,2 or 3; and is

h is 0 or 1;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

In some embodiments, the compound has formula IV:

wherein:

R1、R2x and Y are as defined for formula II and formula III; and is

p is 0, 1,2, 3, 4,5 or 6;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

In certain embodiments, the compound has formula IVa:

wherein:

x and Y are as defined for formula II and formula III; and is

p is 0, 1,2, 3, 4,5 or 6;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

In further embodiments, the compound has formula IIA and formula IIIA:

wherein R is1、R2、R3、R4、R5X and m are as defined for formula II and formula III;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

In some embodiments, the compound has formula IVA:

wherein:

R1、R2and X is as defined for formula II and formula III; and is

p is 0, 1,2, 3, 4,5 or 6;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

In certain embodiments, the compound has formula iva:

wherein:

x is as defined for formula II and formula III; and is

p is 0, 1,2, 3, 4,5 or 6;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

In other embodiments, the compound has formula IIB and formula IIIB:

wherein R is1、R2、R3、R4、R5X and m are as defined for formula II and formula III;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof. The stereocenter of the vicinal dinitrate moiety may have R-or S-type stereochemistry or may be racemic.

In some embodiments, the compound has formula IVB:

wherein:

R1、R2and X is as defined for formula II and formula III; and is

p is 0, 1,2, 3, 4,5 or 6;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof. The stereocenter of the vicinal dinitrate moiety may have R-or S-type stereochemistry or may be racemic.

In certain embodiments, the compound has the formula IVBa:

wherein:

x is as defined for formula II and formula III; and is

p is 0, 1,2, 3, 4,5 or 6;

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof. The stereocenter of the vicinal dinitrate moiety may have R-or S-type stereochemistry or may be racemic.

For compounds of formula II and subclasses thereof, compounds of formula III and subclasses thereof, and compounds of formula IV and subclasses thereof, -X-Y, -X-ONO2or-X-CH (ONO)2)CH2-ONO2Moieties may be attached to the phenyl ring at the ortho, meta or para positions. In certain embodiments, -X-Y, -X-ONO2or-X-CH (ONO)2)CH2-ONO2The moiety is attached to the meta position of the phenyl ring.

For compounds of formula II and its subclasses, compounds of formula III and its subclasses, and compounds of formula IV and its subclasses, in some embodiments, X is a bond, straight or branched chain C1-C6Or C1-C3Alkyl-or straight or branched C1-C6Or C1-C3-O-alkyl-. In certain embodiments, X is a bond or a straight or branched chain C1-C3-O-alkyl- [ e.g. -O-CH2-、-O-(CH2)2-、-O-CHCH3、-O-(CH2)3-、-O-CHCH2CH3、-O-CH2CHCH3or-O-CH (CH)3)CH2-]。

For compounds of formula I and subclasses thereof, compounds of formula II and subclasses thereof, and compounds of formula III and subclasses thereof, the practice of 3-to 8-membered nitrogen-containing heterocyclesExamples include, but are not limited to, aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, azepanyl, and azacyclooctyl. In certain embodiments, R3、R4And the nitrogen atom to which they are attached form a 3-to 6-membered heterocyclic ring.

For compounds of formula I and its subclasses, compounds of formula III and its subclasses, and compounds of formula IV and its subclasses, in certain embodiments, m is 0, 1, or 2; n is 1,2 or 3; and p is 0, 1,2 or 3.

At or near the N +1 site of the GluN2 (or NR2) subunit in the channel-selective filtration region of an NMDAR channel, there may be steric effects of the (electrostatic) interaction of the amine group of the aminoadamantyl nitrate compound, which is protonated at physiological pH values. For compounds of formula I and its subclasses, compounds of formula II and its subclasses, and compounds of formula III and its subclasses, in some embodiments, R3And R4Both of which are hydrogen. In other embodiments, R3And R4One of which is hydrogen and the other is a straight or branched chain C1-C3An alkyl group. In certain embodiments, R3And R4One of which is hydrogen and the other is methyl or ethyl. In still other embodiments, R3And R4Independently is straight chain C1-C3Alkyl (e.g., methyl or ethyl), optionally the same alkyl.

For compounds of formula I and subclasses thereof and compounds of formula III and subclasses thereof, in some embodiments, R5Is hydrogen. In other embodiments, R5Is straight or branched C1-C3An alkyl group. In certain embodiments, R5Is methyl or ethyl. If the carbon atom to which the amine group is attached is a stereocenter, the stereocenter may have R-or S-type stereochemistry or may be racemic.

For compounds of formula I and its subclasses, compounds of formula II and its subclasses, compounds of formula III and its subclasses, and compounds of formula IV and its subclasses, in some embodiments, R1And R2Independently hydrogen or straight or branched C1-C6Or C1-C3An alkyl group. In certain embodiments, R1And R2Both of which are hydrogen. In other embodiments, R1Is hydrogen and R2Is straight or branched C1-C6Or C1-C3Alkyl, or R2Is hydrogen and R1Is straight or branched C1-C6Or C1-C3An alkyl group. In certain embodiments, R1Is hydrogen and R2Is methyl, ethyl or n-propyl, or R2Is hydrogen and R1Is methyl, ethyl or n-propyl. In still other embodiments, R1And R2Independently is a straight or branched chain C1-C6Or C1-C3Alkyl, optionally the same alkyl. In certain embodiments, R1And R2Independently methyl, ethyl or n-propyl, optionally the same alkyl group. In some embodiments, R1Is hydrogen and R2Is ethyl, or R2Is hydrogen and R1Is ethyl. In other embodiments, R1And R2Both of which are ethyl groups.

R1Or/and R2Can increase the binding affinity and residence time of the compound in the open ion channel of the activated NMDA receptor and slow its dissociation rate therefrom. Furthermore, R1、R2Or/and more hydrophobic groups of X (e.g. longer alkyl groups) may increase binding affinity, may compensate for lower affinity that may be associated with the presence of non-hydrogen groups at C-3, C-5 and C-7 of the adamantane scaffold, and may increase the affinity and selectivity of the compound for presynaptic NMDAR over synaptic NMDAR, although the degree of hydrophobicity of the compound or one or more of its groups may need to be balanced with its solubility in aqueous solution.

For compounds of formula I and its subclasses, compounds of formula II and its subclasses, compounds of formula III and its subclasses, and compounds of formula IV and its subclasses, in some embodiments, R1Radical, R2Radicals or X radicalsThe groups or any combination or all thereof are independently substituted with 1,2 or 3 substituents selected from straight or branched C1-C6Or C1-C3Alkyl, haloalkyl, -OR6、-NR7R8、-ONO2、-CN、-C(=O)R6、-C(=O)OR6、-OC(=O)R6、-C(=O)NR7R8、-NR7C(=O)R6、-OC(=O)OR6、-OC(=O)NR7R8、-NR7C(=O)OR6、-NR6C(=O)NR7R8Aryl and heteroaryl; or/and is substituted with 1 to 6 halogen (e.g. fluorine) or deuterium atoms, or all available hydrogen atoms are replaced by halogen (e.g. fluorine) or deuterium atoms, wherein:

R6independently at each occurrence is hydrogen or straight or branched chain C1-C6Or C1-C3An alkyl group; and is

R7And R8Independently at each occurrence is hydrogen or straight or branched chain C1-C6Or C1-C3Alkyl, or R7、R8And the nitrogen atom to which they are attached form a 3-to 6-membered ring.

In certain embodiments, R1Radical, R2The group or X group, or any combination or all thereof independently is a monovalent or divalent deuterated alkyl, fluoroalkyl, or alkyl-ONO2

For compounds of formula I and its subclasses, compounds of formula II and its subclasses, compounds of formula III and its subclasses, and compounds of formula IV and its subclasses, straight or branched chain C1-C6Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, and n-hexyl. Straight or branched C1-C3Examples of alkyl groups include methyl, ethyl, n-propyl and isopropyl.

For compounds of formula I and its subclasses, compounds of formula II and its subclasses, compounds of formula III and its subclasses, and compounds of formula IV and its subclasses, in some embodiments, X has 0, 1,2, 3, 4,5, or 6 carbon atoms. In certain embodiments, X has 0, 1,2, or 3 carbon atoms.

Table 1 shows representative compounds of formula IAa:

TABLE 1

And pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof. The present disclosure specifically discloses each of the 2160 compounds shown in table 1 and each of its possible stereoisomers. alkyl-ONO with stereocenter2A group may have R-or S-type stereochemistry at the stereocenter or may be racemic. Likewise, R with a stereocenter1Group or R2A group may have R-or S-type stereochemistry at the stereocenter or may be racemic.

In certain embodiments, the compound of formula IAa is selected from:

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof, wherein Et ═ ethyl and Pr ═ n-propyl. alkyl-ONO with stereocenter2A group may have R-or S-type stereochemistry at the stereocenter or may be racemic.

Table 2 shows representative compounds of formula IBa:

TABLE 2

And pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof. The present disclosure specifically discloses each of the 864 compounds shown in table 2 and each of its possible stereoisomers. Nitrated alkyl groups having a stereocenter at the branching point may have R-or S-type stereochemistry at the stereocenter or may be racemic. The stereocenter of the vicinal dinitrate moiety may have R-or S-type stereochemistry or may be racemic. Furthermore, R with a stereocenter1Group or R2A group may have R-or S-type stereochemistry at the stereocenter or may be racemic.

In certain embodiments, the compound of formula IBa is selected from:

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof, wherein Et ═ ethyl and Pr ═ n-propyl. Nitrated alkyl groups having a stereocenter at the branching point may have R-or S-type stereochemistry at the stereocenter or may be racemic. Likewise, the stereocenter of the vicinal dinitrate moiety may have R-or S-stereochemistry or may be racemic.

Table 3 shows representative compounds of formula IVA:

TABLE 3

And pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof. The present disclosure specifically discloses each of the 1036 compounds shown in table 3 and each of its possible stereoisomers. R with stereocenter1Group or R2A group may have R-or S-type stereochemistry at the stereocenter or may be racemic.

In certain embodiments, the compound of formula IVA is selected from:

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

Table 4 shows representative compounds of formula IVB:

TABLE 4

And pharmaceutical use thereofAcceptable salts, solvates, hydrates, clathrates, polymorphs, and stereoisomers. The present disclosure specifically discloses each of the 888 compounds shown in table 4 and each of their possible stereoisomers. The stereocenter of the vicinal dinitrate moiety may have R-or S-type stereochemistry or may be racemic. Likewise, R with a stereocenter1Group or R2A group may have R-or S-type stereochemistry at the stereocenter or may be racemic.

In certain embodiments, the compound of formula IVB is selected from:

and pharmaceutically acceptable salts, solvates, hydrates, clathrates, polymorphs and stereoisomers thereof. The stereocenter of the vicinal dinitrate moiety may have R-or S-type stereochemistry or may be racemic.

The amine group attached, indirectly or directly, to the C-1 position of the aminoadamantyl nitrate compound described herein may be an amide, carbamate, or urea, rather than an amine group. Mg in open NMDAR channels of amine groups of aminoadamantyl compounds protonated at physiological pH2+Hydrogen bonding at or near the binding site, with the protonated amine group serving as a hydrogen bond donor and the side chain of asparagine at position 616 of the GluN1 (or NR1) subunit serving as a hydrogen bond acceptor; and binds to the memantine/phencyclidine binding site by electrostatic interaction with the asparagine residue of the GluN2 (or NR2) subunit. -NH (C ═ O) R amide, -NH (C ═ O) OR carbamate, OR-NHC (═ O) NRaRbUrea groups may also be used as hydrogen bond donors. In some embodiments, -NR of aminoadamantyl nitrate compounds3R4The moiety being-NH (C ═ O) R6、-NH(C=O)OR6or-NHC (═ O) NR7R8Wherein R is6Is hydrogen (for formamide) or straight or branched C1-C6Alkyl, and R7And R8Independently of hydrogenOr straight or branched C1-C6Alkyl, or R7、R8And the nitrogen atom to which they are attached form a 3-to 6-membered ring. In certain embodiments, R6Is hydrogen (for formamide) or straight or branched C1-C3Alkyl (e.g. methyl or ethyl), and R7And R8Independently hydrogen or straight or branched C1-C3Alkyl (e.g., methyl or ethyl).

IV.Salt forms

Adamantyl compounds described herein have one or more amine groups (possibly except where the amine group attached indirectly or directly to the C-1 position of the adamantyl scaffold is an amide, carbamate, or urea) and may be present as a free base or salt, they may be used or administered as a free base or pharmaceutically acceptable salt, the amine groups may form addition salts with acids such as inorganic acids (e.g., HCl, HBr, HI, nitric acid, phosphoric acid, or sulfuric acid) or organic acids (e.g., carboxylic acids or sulfonic acids.) suitable acids for preparing pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, boric acid, (+) -camphoric acid, camphorsulfonic acid, (+) - (1S) -camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1, 2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, tartaric acid, malic acid, tartaric acid.

If the compound has an acidic group (e.g., carboxyl), the acidic group can form an addition salt with a base. Pharmaceutically acceptable base addition salts can be formed, for example, with metals (e.g., alkali metals or alkaline earth metals) or amines (e.g., organic amines). Examples of metals that can be used as cations include, but are not limited to, alkali metals (e.g., lithium, sodium, potassium, and cesium), alkaline earth metals (e.g., magnesium, calcium, and barium), aluminum, and zinc. The metal cations may be provided by, for example, inorganic bases such as hydroxides, carbonates and bicarbonates. Non-limiting examples of organic amines useful for forming base addition salts include chloroprocaine, choline, cyclohexylamine, benzhydrylamine, N' -benzhydrylethylenediamine, dicyclohexylamine, diethanolamine, ethylenediamine, N-ethylpiperidine, histidine, isopropylamine, N-methylglucamine, procaine, pyrazine, triethylamine, trimethylamine, and tromethamine. Pharmaceutically acceptable salts are discussed in detail in the handbook of pharmaceutical salts: properties, Selection and Use (Handbook of Pharmaceutical Salts, Properties, Selection and Use), P.Stahl and C.Wermuth, Wiley-VCH (2011).

In some embodiments, the aminoadamantyl nitrate compounds described herein are used or administered in the form of a pharmaceutically acceptable salt. In certain embodiments, the aminoadamantyl nitrate compound is used or administered in the form of the hydrochloride salt (HCl) or the hydrobromide salt (HBr).

V.Isotopic isomers comprising deuterated compounds

The present disclosure encompasses all isotopically enriched forms of the aminoadamantyl nitrate compounds described herein, including but not limited to those in2H (deuterium),13C、15N、17O or18O or any combination thereof, in enriched form at the content of one or more or all instances of the corresponding atom.

To eliminate foreign substances such as drugs, animals express various enzymes such as cytochrome P450Enzymes, esterases, proteases, reductases, dehydrogenases and monoamine oxidases, which react with foreign substances and convert them into foreign substancesMore polar intermediates or metabolites for renal excretion. Such metabolic reactions may involve oxidation of carbon-hydrogen (C-H) bonds to carbon-oxygen (C-O) or carbon-carbon (C ═ C) pi bonds, or oxidation of carbon-oxygen (C-O) single bonds to carbon-oxygen (C ═ O) double bonds. The resulting metabolites may be stable or unstable under physiological conditions and may have substantially different pharmacological, pharmacokinetic and pharmacodynamic properties and toxicity profiles than the parent compound. For many drugs, such metabolic oxidation may be rapid and may result in the need for higher doses or/and increased dosing frequency, which may cause greater side effects.

The present disclosure provides isotopic isomers corresponding to the aminoadamantyl nitrate compounds described herein, which are enriched with deuterium (deuterated) at one or more positions. Deuteration at one or more locations may have any one or more or all of the following benefits: (1) the half-life period is longer; (2) a reduction in the dose or/and a reduction in the number of doses required to achieve the desired effect; (3) a decrease in the blood or plasma level of the parent drug between subjects; (4) the efficacy is increased; (5) reduced side effects due to reduced amount of parent drug administered or/and reduced production of harmful metabolites; and (6) maximum tolerated dose increase.

Deuterium can replace hydrogen at any one or more or all available positions in an aminoadamantyl nitrate compound, including, but not limited to, an adamantyl scaffold, an alkyl group linking an amine group to the C-1 position of an adamantyl scaffold (unless the amine group is directly linked to the C-1 position), a X group (unless X is a bond), R1Radical (unless R1Is halide), R2Radical (unless R2Is a halide) or a compound of formula II, formula III or formula IV, or any combination thereof. In certain embodiments, the aminoadamantyl nitrate compound is deuterated at the carbon atom attached to the amine group indirectly attached to the C-1 position of the adamantyl scaffold (unless the amine group is directly attached to the C-1 position), or/and for one or more or all nitrate groups, at the carbon atom attached to the nitrate groupDepending on whether the compound has one or more nitrate groups (unless the compound has only one nitrate group and X is a bond). In another embodiment, aminoadamantyl nitrate is at R1Radical (unless R1Is halide) or/and R2Radical (unless R2Is a halide) is deuterated at one or more or all available sites.

In some embodiments, at least one of the available positions in the aminoadamantane nitrate has deuterium enrichment of at least about 10%, 25%, 50%, 75%, 90%, 95%, or 98%. In certain embodiments, at least one of the available positions has deuterium enrichment of at least about 90%, 95%, or 98%. In further embodiments, each position in the aminoadamantane nitrate enriched in deuterium (or deuterated) independently has deuterium enrichment of at least about 10%, 25%, 50%, 75%, 90%, 95%, or 98%. In certain embodiments, each position enriched with deuterium (or deuterated) independently has deuterium enrichment of at least about 90%, 95%, or 98%.

The term "deuterium enrichment" refers to the percentage of incorporation of deuterium in place of hydrogen at a given position in the molecule. For example, deuterium enrichment of 10% at a given position means that 10% of the molecules in a given sample contain deuterium at that position. Since the naturally occurring distribution of deuterium is about 0.0156%, the deuterium enrichment at any position in a molecule synthesized using non-deuterium enriched starting materials or reagents is about 0.0156%. Deuterium enrichment can be determined using conventional analytical methods known to those of ordinary skill in the art, including mass spectrometry and nuclear magnetic resonance spectroscopy.

The term "is deuterium" or "deuterated" when used to describe a given position in a molecule, or the symbol "D" when used to denote an element at a given position in a diagram of a molecule, means that the designated position is enriched with deuterium in a distribution that is higher than the naturally occurring distribution of deuterium. In some embodiments, deuterium enrichment at a given position is at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 99% (e.g., at least about 50%) deuterium. In certain embodiments, the deuterium enrichment at a given position is at least about 90%, 95%, or 98% deuterium.

VI.Therapeutic use of aminoadamantane nitrate ester compounds

Over-activation of NMDA receptors is relieved by Mg2+Resulting in voltage-dependent channel blocking and causing excess Ca2+Influx and subsequent oxidative stress due to overproduction of reactive oxygen species may lead to neuronal excitotoxicity and ultimately neuronal damage and death. Without intending to be bound by theory, it is believed that stimulation of the extra-synaptic NMDA receptor (eNMDAR) rather than the synaptic NMDA receptor (snmddar) results in excitotoxicity related to neurodegenerative disorders. Synaptic NMDAR activity is phased in nature and synaptic NMDAR is typically activated transiently and strongly by transsynaptic release of glutamate. In contrast, extrasynaptic NMDAR is typically activated chronically by elevated levels of environmental glutamate, whether due to synaptic release of glutamate or/and glutamate uptake dysfunction or reversal [ G.Hardingham et al, Nat.Rev.Neurosci, 11:682-696(2010)]. Since synaptic NMDARs are usually activated phasic/transiently, while extra-synaptic NMDARs are usually activated long-term/tense, it is mainly extra-synaptic NMDARs that are activated and thus allow Ca2+Excess and long-term influx through excess/sustained open ion channels. Furthermore, in the case of prolonged NMDAR activation, such as excessive levels of glutamate or NMDA, synaptic NMDAR activity is inhibited and the extrasynaptic NMDAR signaling pathway predominates [ F.Soriano et al, J.Neurosci.,26:4509-4518(2006)]。

Synaptic NMDAR activity promotes cell health and survival, while extrasynaptic NMDAR activity elicits a cell death pathway and antagonizes a synaptic NMDAR-induced cell survival pathway. Ca triggered by strong activation of synaptic NMDAR2+Influx does not cause interference with mitochondrial membrane potential and enhances mitochondrial health, primarily through nuclear Ca2+Signaling triggers genomic processes that make neurons more resistant to oxidative stress and apoptosis (including reduced expression of pro-apoptotic factors such as caspases). Synaptic NMDAR activity events promote "acquired" neuroprotectionAnd persists after most signaling pathways are no longer active. In contrast, comparable intracellular Ca induced by activation of the extra-synaptic NMDAR itself or in the presence of activation of the synaptic NMDAR2+Concentrations trigger mitochondrial dysfunction (including loss of mitochondrial membrane potential) and cell death (whether caused by apoptosis or necrosis). Mitochondrial dysfunction, activation of intrinsic mitochondrial apoptotic pathways, and oxidative stress due to overproduction of reactive oxygen species have been implicated in the pathogenesis of neurodegenerative diseases. The apparent difference in the results of synaptic and extrasynaptic NMDAR activation is due to the opposite effects on intracellular signaling pathways, many of which involve the same signaling protein, and the induction of very different gene expression programs. Nucleus Ca2+As an important gene expression regulator, plays a key role in the pro-survival role of synaptic NMDAR activation and is disrupted by extra-synaptic NMDAR activity. Many neurodegenerative disorders and other CNS disorders share a common signaling pathway downstream of extrasynaptic NMDAR activity leading to neurotoxicity. See, e.g., Hardingham (2010, supra) and M.Parsons et al, Neuron,82:279-293 (2014).

It is believed that activation of the synaptic NMDA receptor, but not the synaptic NMDA receptor, results in excitotoxicity related to neurodegenerative disorders, such as Alzheimer's disease, Huntington's disease, Parkinson's disease, and amyotrophic lateral sclerosis, as well as other CNS disorders, such as epilepsy, stroke, and traumatic brain injury [ Hardingham (2010, supra) and Parsons (2014, supra)]. Expression or/and activation of extra-synaptic NMDARs is elevated in disease states [ Parsons (2014, supra)]Amyloid β in studies with Alzheimer's Disease (AD)1-42Peptides stimulate α -7 nicotinic acetylcholine receptors and thus release excess glutamate from astrocytes, which in turn activates neuronal extra-synaptic NMDAR, mediating nitric oxide production, tau protein hyperphosphorylation, caspase-3 activation, and mitochondrial dysfunction, thus leading to neuronal spinal loss and loss of synapses associated with AD cognitive decline [ m.talantova et al, proc.natl.Acad.Sci.USA,110: E2518-E2527(2013)]. Loss of synapses may result in an imbalance in activity and signaling of synaptic and extra-synaptic NMDARs, which may lead to cell death associated with neurodegenerative diseases such as alzheimer's disease and huntington's disease.

NMDAR assembly into heterotetramers with two essential GluN1 subunits and two GluN2(GluN2A, 2B, 2C or 2D) or/and GluN3(GluN3A or 3B) subunits NMDAR assembly in CNS most NMDAR assemblies into GluN1/GluN2A or GluN1/GluN2B heterodimers or GluN1/GluN 632A/GluN 2B heterotrimers although both subtypes can be found outside synapses and synapses, GluN2A (or NR2A) are enriched in synaptic NMDAR whereas GluN2B (or NR2B) are more prevalent in extracellular NMDAR [ Ac29. et al, 2011 et al; Met 2 × 4675, Met 19, Met 5, N35, N5, D5, N35, N5, D, N5, III, N5, III, N5, III, N5, III.

Neurodegenerative disorders associated with excitotoxic events and other CNS disorders can be treated by blocking extra-synaptic NMDAR. The physiological activity of synaptic NMDARs is essential for normal neuronal function, and therefore an NMDAR antagonist needs to block activated extra-synaptic NMDARs without inhibiting the normal activity of synaptic NMDARs. This may be achieved by non-competitive antagonists or open channel blockers that selectively enter the open channels of the extra-synaptic NMDARs after they have been activated by the binding of a synergistic agonist and thereby block cations, particularly Ca2+The flow of (c).

The aminoadamantyl nitrate compounds described herein are useful as voltage-dependent, non-competitive antagonists that selectively block extra-synaptic NMDARs. They are directed against Mg in the channels of activated NMDAR2+The memantine/phencyclidine binding site at or near the binding site has a relatively low affinity and is relatively fastRapid off-rate so they do not accumulate in the channel when it is closed and so retain the physiological activity of the phasic activated synaptic NMDAR and avoid psychotic side effects associated with high affinity NMDAR antagonists [ S. L ipton, Curr. drug Targets,8:621-632(2007)]. However, aminoadamantyl nitrate compounds have a sufficiently high affinity for the memantine/phencyclidine binding site (e.g., K from about 200nM to about single digit. mu.M)i) And have a sufficiently long residence time in the ion channels of excessive/sustained opening of the tensilely activated extra-synaptic NMDAR that they block Ca2+Excessive influx through the channel and thereby prevent excitotoxic events. Whether administered as an acid addition salt or as a free base, the amine group of the adamantyl compound is protonated at physiological pH (pH of about 7.4). With Mg2+In the presence of Mg2+Binding at binding sites compared to Mg of protonated amine groups in channels of activated NMDAR2+Binding with higher affinity, slower dissociation/dissociation rates, and lower voltage dependence at or near the binding site, allowing adamantyl compounds to block Ca2+Flowing in through the channel for a long time. The protonated amine groups form hydrogen bonds with the side chain of asparagine at position 616 of the GluN1 (or NR1) subunit at the narrowest constriction site (channel selective filter) of the pore channel and also have significant electrostatic interactions with the carbonyl oxygen atom of the side chain of asparagine residue at N and N +1 sites of the GluN2 (or NR2) subunit therein [ H. -S.Chen et al, J.Pharmacol. Exper. Therap.,314:961-]。Mg2+The binding sites are within the electric field of the channel and thus in Mg by the (positively) charged agent2+The blocking of the channel by binding at or near the binding site is voltage dependent.

Hydrophobic interactions between the compound and subunits of the extra-synaptic NMDAR also result in an increase in the binding affinity of the aminoadamantyl nitrate compound for the extra-synaptic NMDAR. The two methyl groups of memantine are involved in hydrophobic interactions with the hydrophobic binding pocket formed by residues a645 and a644 in the third transmembrane helix of the GluN1 (or NR1) and GluN2B (or NR2B) subunits, respectively, which is greatly increased compared to amantadineBinding affinity of memantine to activated NMDAR [ W. L imapachat et al, ACS chem.Neurosci.,4:255-260(2013)]. The GluN2B (or NR2B) subtype is more prevalent in extra-synaptic NMDARs. Having R1And R2The non-hydrogen (e.g., alkyl) group of aminoadamantyl nitrate compounds can participate in hydrophobic interactions with the hydrophobic binding pocket of the GluN1 and GluN2B subunits of the extra-synaptic NMDAR. It is considered that R1The group is oriented towards the hydrophobic binding pocket of GluN 2B. A larger number of carbon atoms of the group increases its hydrophobicity/lipophilicity.

The aminoadamantyl nitrate compounds described herein selectively inhibit activated extra-synaptic NMDAR and have a dual inhibitory mechanism. They pass through Mg in the channels2+Binding at or near the binding site blocks open ion channels of the activated extra-synaptic NMDARs in a voltage-dependent manner, and they inhibit the activity of the activated extra-synaptic NMDARs in a voltage-independent manner by S-nitrosylation of cysteine residues of the redox regulatory site on the receptor. Modulation of NMDA receptor ion channel conductivity by nitrosylation [ Y.Choi et al, nat. Neurosci, 3:15-21(2000)]. Mg in the open ion channel of activated extra-synaptic NMDAR by protonated amine groups of aminoadamantyl nitrate2+Binding at or near the binding site brings the nitrate group of the compound into close proximity with the cysteine residue of the redox regulatory site in the extracellular domain of the receptor. Oxidation at the oxidation reduction site may cause the formation of one or more disulfide bonds involving two or more cysteine residues, thereby down-regulating/desensitizing NMDAR channel activity. Supplying/transferring NO (e.g. as NO) to cysteine residue of redox site from nitrate group of aminoadamantyl compound+In form (b) possibly with the aid of glutathione S-transferase. If the aminoadamantyl compound has two or more nitrate groups, the two or more nitrate groups can potentially transfer NO to two or more of the about five cysteine residues of the redox site. S-nitrosylation of one or more cysteine residues oxidizes the sulfhydryl group of the one or more cysteine residues, or may promote one or more cysteine residuesFormation of multiple disulfide bonds. The role of S-nitrosylation of one or more cysteine residues of the redox site is to inhibit the activity and Ca of extra-synaptic NMDAR channels2+Flows in through the extra-synaptic NMDAR channel. In neurons, Ca2+Influx through the NMDAR channel activates Nitric Oxide Synthase (NOS), causing Nitric Oxide (NO) production. NO downregulates NOS and inhibits Ca through negative feedback2+Inflow through NMDAR channels. The one or more cysteine residues of the aminoadamantyl compound that provide for targeting of the NO nitrate group to the redox site avoids the potential vascular and neurotoxic side effects of free nitric oxide, such as vasodilation, apoptosis and the formation of neurotoxic peroxynitrite. The resulting protonated aminoadamantyl compound with hydroxyl groups substituted for each donor nitrate group remains bound to Mg after S-nitrosylation by donating NO2+At or near the binding site unless its voltage-gated block to the ion channel is diminished by membrane depolarization. However, the redox regulatory site is outside the voltage field of the channel, and therefore, the inhibition of channel activity of activated extra-synaptic NMDARs by S-nitrosylation of cysteine residues of the redox site is voltage-independent and remains for a considerable time after the compound is excreted from the channel due to depolarization. Inhibition of activated extra-synaptic NMDAR by S-nitrosylation of cysteine residues is significantly increased under hypoxic conditions and is therefore particularly suitable for, but not limited to, preventing or reducing neuronal damage due to cerebral ischemia, such as stroke and vascular dementia [ h.takahashi et al, sci.rep.,5:14781(2015)]。

The aminoadamantyl nitrate compounds described herein selectively inhibit activated extra-synaptic NMDAR and thereby prevent or reduce excitotoxicity while retaining normal synaptic NMDAR activity. The NMDAR component that retains excitatory post-synaptic current (EPSC) is neurotrophic and avoids synaptic damage. Like nitromemantine, the aminoadamantyl nitrate compounds described herein have at least one nitrate group. Memantine preferentially blocks the excessive open channel of activated extra-synaptic NMDAR, while relatively retaining normal synaptic NMDAR activity, even ifExtracellular Mg2+This is also true in the presence of pathological depolarization [ P.Xia et al, J.Neurosci.,30:11246-11250(2010)]. Nitromentane more selectively inhibits synaptic NMDAR activity and retains synaptic NMDAR activity to a greater extent than memantine [ Takahashi (2015, supra)]The aminoadamantyl nitrate compounds described herein can prevent or reduce synaptic loss and neuronal death, increase synapse and dendrite density, and enhance cognitive function in Alzheimer's disease patients by, for example, long-term treatment to antagonize the extra-synaptic NMDAR and α -7 nicotinic acetylcholine receptors.

Furthermore, the aminoadamantyl nitrate compounds described herein may promote long-term enhancement (L TP) as a basis for synaptic plasticity and thus memory and learning, synaptic plasticity is the ability of chemical synapses to change their strength and allow the enhancement or attenuation of specific connections to be modulated in an organized manner. L TP is considered to be encoded by a change in synaptic strength is a persistent enhancement of synapses based on recent synaptic activity patterns, resulting in a persistent increase in signaling between two neurons, this process is considered to encode and store learning and long-term memory. L0 TP is reversed to long-term inhibition (L TD), which results in a persistent decrease in synaptic strength.activation of synaptic NMDA receptors induces L TP and inhibition of them impairs L TP Parsons (2014, supra), conversely, activation of synaptic NMDA receptors inhibits L and induces L TD [ L i et al, J.Neurosis, 31: 27 (2014, Bu TP et al) and activation of synaptic NMDA receptor 2011, and impairs 2011, 2011-99. Bull et al, and can help to restore synaptic plasticity and thus, by enhancing synaptic plasticity, by a compound, to restore synaptic plasticity, and by enhancing synaptic plasticity, and by Bu-95, Bull et 35, Bull et al, a physiological antagonism, a physiological, a decrease, a physiological, a synaptic plasticity, and a synaptic plasticity, which may help to restore synaptic plasticity, and thereby, a synaptic plasticity, a synaptic.

Non-limiting examples of neurodegenerative disorders that can be treated with the aminoadamantyl nitrate compounds described herein include dementia (e.g., Alzheimer 'S disease, vascular dementia, Lewy body dementia, frontotemporal dementia, and HIV-associated dementia), Huntington' S disease (often resulting in dementia), Parkinson 'S disease (often resulting in dementia), multiple system atrophy (sweet syndrome), cerebellar degeneration, ataxia (e.g., cerebellar ataxia, spinocerebellar ataxia, Friedreich' S ataxia), and ataxia-vasodilation [ Lewy-Pabar syndrome (L-syndrom), neuro-atrophy [ 19.35-neuro ataxia ], neuro-atrophy [ 11.35-neuro atrophy ], neuro-atrophy [ 11.35-neuro atrophy ], neuro-atrophy [ 11.35, neuro-atrophy ], neuro-atrophy [ 12.35, neuro-atrophy ], neuro-atrophy [ 11, neuro-atrophy ], neuro-atrophy [ 12, neuro-atrophy ], neuro-atrophy [ 11, neuro-atrophy [ 12, neuro-10, neuro-atrophy, neuro-3, neuro-atrophy [ 12, neuro-3, neuro-atrophy, neuro-atrophy, neuro-e, neuro-mediated neurodegeneration, neuro-mediated neurodegeneration, neuro-neurodegeneration, neuro-neurodegeneration, neuro-neurodegeneration.

In certain embodiments, aminoadamantyl nitrate compounds described herein are used to treat a neurodegenerative disorder selected from the group consisting of alzheimer ' S disease, vascular dementia, huntington ' S disease, parkinson ' S disease, and a L S.

Examples of other CNS disorders that may or may not involve neurodegeneration in the pathophysiology that can be treated with the aminoadamantyl nitrate compounds described herein include, but are not limited to, cerebrovascular disorders (including cerebral ischemia [ including acute ischemia, such as stroke, chronic ischemia, such as vascular dementia, cerebral ischemia/reperfusion injury, and nerve injury caused by hypoxia or/and glucose levels in the brain ], intracerebral hemorrhage and retinopathy), brain injury and trauma (including traumatic brain injury, diffuse axonal injury, primary and secondary brain injury, focal and diffuse brain injury, hypoxic and hypoxic brain injury, intracerebral hemorrhage and brain edema), spinal cord injury (due to, for example, trauma, ischemic or degenerative disorders), epilepsy (including nerve injury caused by seizure), movement disorders (such as levodopa-induced movement disorder and tardive movement disorder), spasticity, Pain (such as acute Pain, chronic Pain, trigeminal Pain, complex regional Pain syndrome [ CRPS ], fibromyalgia, hyperalgesia, inflammatory Pain, postoperative Pain, Headache-related Pain, and dysmotility disorder, and anxiety disorder [ including psycho stress, epilepsy [ 12, 20135, 121, 150, 2016, 92, 150, 2016, 92, 150, 12, 150, 12, 15, 150, 15, 150, 15, 150, 15, etc.;, D.

In certain embodiments, aminoadamantyl nitrate compounds described herein are used to treat a CNS disorder selected from the group consisting of cerebral ischemia, traumatic brain injury, epilepsy, pain, and autism spectrum disorders.

The therapeutically effective amount and frequency of administration of the aminoadamantyl nitrate compound to treat a neurodegenerative disorder or other CNS disorder can depend on various factors, including the type of disorder, the severity of the condition, the potency of the compound, the mode of administration, the age, weight, general health, sex, and diet of the subject, and the response of the subject to treatment, and can be determined by the treating physician. In some embodiments, an effective dose of aminoadamantyl nitrate per day is from about 1mg, 5mg, or 10mg to about 100mg, or as deemed appropriate by the treating physician, which can be administered in a single dose or in divided doses. In certain embodiments, the effective dose of aminoadamantyl nitrate per day is about 5mg or 10mg to about 50mg, or about 50mg to 100 mg. In additional embodiments, the effective dose of aminoadamantane nitrate per day is about 1mg, 5mg, 10mg, 15mg, 20mg, 25mg, 30mg, 40mg, 50mg, 60mg, 70mg, 80mg, 90mg, or 100 mg. In certain embodiments, an effective dose of aminoadamantane nitrate per day is about 10mg to 30mg, or about 10mg, 15mg, 20mg, 25mg, or 30 mg.

The dosage of aminoadamantane nitrate can be adjusted during the course of the treatment regimen, as can be determined by the treating physician. For example, the aminoadamantyl nitrate compound may be administered at an initial daily dose for the first week of treatment, and then the daily dose of the compound may be gradually or stepwise increased every week following treatment until the target or appropriate daily maintenance dose is administered for the duration of treatment, e.g., the fourth week of treatment and thereafter. For example, increasing the dosage of the drug gradually or stepwise during the initial phase of treatment will allow the treating physician to determine the optimal therapeutic dosage while avoiding or minimizing any potential side effects. The initial and maintenance doses can be any effective dose described herein. As another example, if it is desired to rapidly establish a therapeutic level of an aminoadamantyl nitrate compound to treat a CNS disorder, such as stroke, traumatic brain injury or pain, a first loading dose of the compound can be administered, for example, on day 1, an optional second loading dose can be administered, for example, on day 2, an optional third loading dose can be administered, for example, on day 3, and a maintenance dose of the compound can be administered daily for a subsequent duration of treatment. The loading dose can be, e.g., about 5-fold, 4-fold, 3-fold, 2.5-fold, 2-fold, or 1.5-fold of the maintenance dose, and the optional second and third loading doses can, e.g., be less than the previous loading dose. The maintenance dose can be any effective dose described herein.

The aminoadamantyl nitrate can be administered at any suitable frequency to treat neurodegenerative disorders or other CNS disorders, as can be determined by a treating physician. In some embodiments, the aminoadamantyl nitrate is administered daily (including once, twice or more daily), every second day, every third day, or weekly, or as frequently as the treating physician deems appropriate. In certain embodiments, the aminoadamantyl nitrate is administered once daily.

The aminoadamantyl nitrate can be administered for any suitable period of time to treat a neurodegenerative disorder or other CNS disorder, as can be determined by a treating physician. For the treatment of neurodegenerative disorders, in some embodiments, the aminoadamantane nitrate is administered for a period of at least about 3 months, 6 months, 1 year, 1.5 years, 2 years, 3 years, 4 years, 5 years, or longer. For the treatment of CNS disorders that may or may not involve neurodegeneration in pathophysiology, in certain embodiments, aminoadamantane nitrate is administered for a period of at least about 1 week, 2 weeks, 1 month, 2 months, 3 months, 6 months, 1 year, 1.5 years, 2 years, 3 years, or longer.

Aminoadamantyl nitrate may also be administered temporarily (as needed) to treat CNS disorders, as may be determined by the treating physician. For example, to treat pain or headache, aminoadamantane nitrate may be administered until the pain or headache dissipates. If pain or headache is reoccurring, the application of aminoadamantane nitrate can be resumed. The appropriate dosage, frequency of administration, and length of treatment for the aminoadamantyl nitrate for neurodegenerative or other CNS disorders can be determined by the treating physician.

The aminoadamantyl nitrate may be administered by any suitable route, and may be administered locally or systemically to treat neurodegenerative disorders or other CNS disorders, as may be determined by a treating physician. Potential routes of administration for aminoadamantane nitrate include, but are not limited to, oral, parenteral (including intradermal, subcutaneous, intramuscular, intravascular, intravenous, intraarterial, intraperitoneal, intramedullary, intrathecal, and topical), intracavitary and topical (including dermal/epidermal, transdermal, mucosal, transmucosal, intranasal [ e.g., via nasal spray or nasal drops ], intraocular [ e.g., via eye drops ], pulmonary [ e.g., via oral or nasal inhalation ], buccal, sublingual, rectal [ e.g., via suppository ], and vaginal [ e.g., via suppository ]). In certain embodiments, the aminoadamantyl nitrate is administered orally (e.g., as a tablet or capsule). In other embodiments, the aminoadamantyl nitrate is administered parenterally (e.g., intravenously, intramuscularly, or subcutaneously, whether by injection or infusion).

The aminoadamantyl nitrate compounds described herein can effectively cross the Blood Brain Barrier (BBB) to treat neurodegenerative disorders and other CNS disorders, or cross the Blood Retinal Barrier (BRB) to treat ocular disorders such as glaucoma, AMD and retinopathy. They are small molecules with hydrophobic/lipophilic scaffolds and can therefore penetrate into the brain or eye. Increasing the lipophilicity of one or more groups on the aminoadamantyl compounds increases their ability to cross the BBB or BRB. For example, memantine or amantadine (20 mg/kg/day and 100 mg/kg/day, respectively) is infused for 7 days such that the whole brain concentration of memantine or amantadine is 44 times and 16 times, respectively, the free concentration in serum [ M.Hesselink et al, pharm. Res.,16:637-642(1999) ].

Aminoadamantyl nitrate can optionally be used in combination with one or more additional therapeutic agents to treat neurodegenerative disorders or other CNS disorders.

VII.Pharmaceutical composition

Additional embodiments of the present disclosure relate to pharmaceutical compositions comprising an aminoadamantyl nitrate compound, or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, or polymorph thereof, as described herein, and one or more pharmaceutically acceptable excipients or carriers. The composition may optionally contain additional therapeutic agents. The pharmaceutical composition contains a therapeutically effective amount of aminoadamantyl nitrate, one or more pharmaceutically acceptable excipients or carriers, and optionally a therapeutically effective amount of an additional therapeutic agent, and is formulated for administration to a subject for therapeutic use.

The pharmaceutical composition contains aminoadamantyl nitrate in substantially pure form and optionally an additional therapeutic agent. In some embodiments, the purity of the aminoadamantane nitrate and optional additional therapeutic agent is independently at least about 95%, 96%, 97%, 98%, or 99%. In certain embodiments, the purity of the aminoadamantane nitrate and optional additional therapeutic agent is independently at least about 98% or 99%. In addition, the pharmaceutical composition is substantially free of contaminants or impurities. In some embodiments, the level of contaminants or impurities other than residual solvent in the pharmaceutical composition is no more than about 5%, 4%, 3%, 2%, or 1% relative to the combined weight of the intended active and inactive ingredients. In certain embodiments, the level of contaminants or impurities other than residual solvent in the pharmaceutical composition is no more than about 2% or 1% relative to the combined weight of the intended active and inactive ingredients. Pharmaceutical compositions are generally prepared in accordance with current Good Manufacturing Practice (GMP) as suggested or required by, for example, the Federal Food, Drug, and Cosmetic Act 501(a) (2) (B) and the International Conference on harmony (Q7) guidelines.

The pharmaceutical compositions/formulations may be prepared in sterile form. For example, pharmaceutical compositions/formulations for parenteral administration by injection or infusion are generally sterile. Sterile pharmaceutical compositions/formulations are compounded or manufactured according to pharmaceutical grade sterilization standards known to those skilled in the art, such as those disclosed or claimed in the United States Pharmacopeia (United States Pharmacopeia) chapters 797, 1072 and 1211, and Code of Federal Regulations section 211, part 21.

Non-limiting examples of excipients include liquid and solid fillers, diluents, binders, lubricants, glidants, surfactants, dispersants, disintegrants, emulsifiers, wetting agents, suspending agents, thickeners, solvents, isotonic agents, buffers, pH adjusting agents, absorption delaying agents, stabilizers, preservatives, antioxidants, antimicrobials, antibacterial agents, antifungal agents, adjuvants, sweeteners, flavoring agents, colorants, encapsulating materials and coating materials The use of such excipients in Pharmaceutical formulations is known in The art, for example, conventional vehicles and carriers include, but are not limited to, oils (e.g., vegetable oils such as sesame oil), aqueous solvents (e.g., saline, phosphate buffered saline [ PBS ] and isotonic solutions [ e.g., Ringer's solution ]) and solvents (e.g., dimethylsulfoxide [ DMSO ] and alcohols [ e.g., ethanol, glycerol and propylene glycol ]), unless any conventional excipient or carrier is incompatible with The active ingredient, The disclosure covers The use of amino alkyl nitrate containing compounds in formulations such as Pharmaceutical excipients and excipients, see The Pharmaceutical design of Pharmaceutical excipients, The Pharmaceutical Handbook of Pharmaceutical design of Pharmaceutical manufacturers (e.g., Pharmaceutical excipients, The Pharmaceutical Handbook, The publication [ e.g., The Pharmaceutical Handbook, The publication of Pharmaceutical Handbook, The publication [ e.g., Pharmaceutical Handbook, The publication [ 10, The publication [ e.g., Pharmaceutical Handbook, The publication [ e.g., Pharmaceutical publication of Pharmaceutical publication [ 10, The publication of Pharmaceutical publication [ e.g., Pharmaceutical publication of Pharmaceutical Handbook, The publication [ 10, The publication of Pharmaceutical publication [ e.g., The publication of Pharmaceutical publication of The publication of Pharmaceutical publication of.

The appropriate formulation may depend on various factors, such as the chosen route of administration. Potential routes of administration for pharmaceutical compositions comprising aminoadamantane nitrate compounds include, but are not limited to, oral, parenteral (including intradermal, subcutaneous, intramuscular, intravascular, intravenous, intraarterial, intraperitoneal, intramedullary, intrathecal, and topical), intracavitary, and topical (including dermal/epidermal, transdermal, mucosal, transmucosal, intranasal [ e.g., by nasal spray or nasal drops ], intraocular [ e.g., by eye drops ], pulmonary [ e.g., by oral or nasal inhalation ], buccal, sublingual, rectal [ e.g., by suppository ], and vaginal [ e.g., by suppository ]). Topical formulations can be designed to produce a local or systemic therapeutic effect.

For example, a formulation of an aminoadamantane nitrate ester compound suitable for oral administration can be, for example, a bolus; capsules (including push-fit capsules and soft capsules), tablets, pills, cachets, or lozenges; powder or granules; semi-solids, electuary, paste or gel; solutions or suspensions in aqueous or/and non-aqueous liquids; or in the form of an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.

Push-fit capsules or two-piece hard gelatin capsules may contain a mixture of aminoadamantyl nitrate with, for example, fillers or inert solid diluents (e.g., calcium carbonate, calcium phosphate, kaolin, or lactose), binders (e.g., starch), glidants or lubricants (e.g., talc or magnesium stearate) and disintegrants (e.g., crospovidone) and optionally stabilizers or/and preservatives. For soft or one-piece gelatin capsules, the aminoadamantyl nitrate may be dissolved or suspended in a suitable liquid (e.g., a liquid polyethylene glycol or an oil medium such as a fatty oil, peanut oil, olive oil, or liquid paraffin), and the liquid-filled capsules may contain one or more other liquid excipients or/and semisolid excipients such as stabilizers or/and amphiphiles (e.g., fatty acid esters of glycerol, propylene glycol, or sorbitol). In certain embodiments, the capsule (e.g., a hard gelatin capsule) comprises aminoadamantyl nitrate and sugar spheres, polyvinylpyrrolidone, hydroxypropylmethylcellulose, talc, polyethylene glycol, ethylcellulose, ammonium hydroxide, oleic acid, and medium chain triglycerides.

Tablets may contain a mixture of aminoadamantyl nitrate with, for example, fillers or inert diluents (e.g., calcium carbonate, calcium phosphate, lactose, mannitol, or microcrystalline cellulose), binders (e.g., starch, gelatin, gum arabic, alginic acid or a salt thereof, or microcrystalline cellulose), lubricants (e.g., stearic acid, magnesium stearate, talc, or silicon dioxide), and disintegrants (e.g., crospovidone, croscarmellose sodium, or colloidal silicon dioxide), and optionally surfactants (e.g., sodium lauryl sulfate). Tablets may be uncoated or may be coated with, for example, an enteric coating which protects the active ingredient from the acidic environment of the stomach or a material which delays disintegration and absorption of the active ingredient in the gastrointestinal tract and thereby provides a sustained action over a longer period. In certain embodiments, the tablets comprise aminoadamantyl nitrate and lactose monohydrate, microcrystalline cellulose, anhydrous colloidal silicon dioxide, talc, and magnesium stearate, and are coated with a film coating (e.g., a film coating comprising hydroxypropyl methylcellulose, titanium dioxide, and polyethylene glycol 400).

Compositions for oral administration may also be formulated as solutions or suspensions in aqueous liquids or/and non-aqueous liquids, or as oil-in-water or water-in-oil liquid emulsions. Dispersible powders or granules of aminoadamantyl nitrate can be mixed with any suitable combination of aqueous liquids, organic solvents or/and oils and any suitable excipients (e.g., any combination of dispersing, wetting, suspending, emulsifying, or/and preservative agents) to form a solution, suspension, or emulsion.

The aminoadamantyl nitrate compound can also be formulated for parenteral administration by injection or infusion to circumvent gastrointestinal absorption and first pass metabolism. An exemplary parenteral route is the intravenous route. Additional advantages of intravenous administration include the direct administration of the therapeutic agent into the systemic circulation to achieve rapid systemic action, and the ability to administer the therapeutic agent continuously or/and in large volumes, if desired. Formulations for injection or infusion may be in the form of solutions, suspensions or emulsions in oily or aqueous vehicles, for example, and may contain excipients such as suspending, dispersing or/and stabilizing agents. For example, an aqueous or non-aqueous (e.g., oily) sterile injection solution may contain aminoadamantyl nitrate in combination with excipients such as antioxidants, buffers, bacteriostats and solutes that render the formulation isotonic with the blood of the subject. Aqueous or non-aqueous sterile suspensions may contain an aminoadamantyl nitrate together with excipients such as suspending agents and thickening agents, and optionally stabilizers and agents that increase the solubility of the aminoadamantyl nitrate to allow for the preparation of more concentrated solutions or suspensions. As another example, a sterile aqueous solution for injection or infusion (e.g., subcutaneous or intravenous) may contain an aminoadamantyl nitrate, sodium chloride, a buffer (e.g., sodium citrate), a preservative (e.g., m-cresol), and optionally a base (e.g., NaOH) or/and an acid (e.g., HCl) to adjust the pH.

For topical administration, the aminoadamantyl nitrate may be formulated as, for example, buccal or sublingual tablets or pills. The advantages of buccal or sublingual tablets or pills include avoidance of gastrointestinal absorption and first pass metabolism, and rapid absorption into the systemic circulation. Buccal or sublingual tablets or pills can be designed to provide faster release of the aminoadamantyl nitrate so that it is taken up more quickly into the systemic circulation. In addition to the therapeutically effective amount of aminoadamantyl nitrate, the buccal or sublingual tablet or pill can contain suitable excipients including, but not limited to, any combination of fillers and diluents (e.g., mannitol and sorbitol), binders (e.g., sodium carbonate), wetting agents (e.g., sodium carbonate), disintegrants (e.g., crospovidone and croscarmellose sodium), lubricants (e.g., silicon dioxide [ including colloidal silicon dioxide ] and sodium stearyl fumarate), stabilizers (e.g., sodium bicarbonate), flavoring agents (e.g., spearmint flavor), sweeteners (e.g., sucralose), and coloring agents (e.g., yellow iron oxide).

For topical administration, the aminoadamantyl nitrate compound may also be formulated for intranasal administration. The nasal mucosa provides a large surface area, a porous endothelium, a high vascular subepithelial layer and a high absorption rate and thus allows a high bioavailability. Furthermore, intranasal administration avoids first-pass metabolism and significant concentrations of aminoadamantyl nitrate can be introduced into the CNS. Intranasal formulations may contain aminoadamantyl nitrate as well as excipients such as solubilizers (e.g. propylene glycol), humectants (e.g. mannitol or sorbitol), buffers and water, and optionally preservatives (e.g. benzalkonium chloride), mucoadhesive agents (e.g. hydroxyethyl cellulose) or/and penetration enhancers.

Another way of topically applying aminoadamantyl nitrate is pulmonary, including by oral inhalation and nasal inhalation. The lung serves as a portal for systemic circulation. Advantages of pulmonary drug delivery include, for example: 1) avoid first pass liver metabolism; 2) rapid drug action; 3) the alveolar region for absorption has a large surface area, the lung has high permeability (thin air-blood barrier), and the vascular system of the airways is abundant; 4) the extracellular enzyme levels are reduced due to the large alveolar surface area compared to the gastrointestinal tract; and 5) a lower dose can achieve equivalent therapeutic effects compared to other oral routes, and therefore, reduced systemic side effects. Advantages of oral inhalation over nasal inhalation include deeper penetration/deposition of the drug into the lungs. Oral or nasal inhalation may be achieved by means of, for example, a metered dose inhaler, a dry powder inhaler or a nebulizer, as is known in the art. In certain embodiments, a sterile aqueous solution for oral inhalation contains aminoadamantyl nitrate, sodium chloride, a buffering agent (e.g., sodium citrate), optionally a preservative (e.g., m-cresol), and optionally a base (e.g., NaOH) or/and an acid (e.g., HCl) to adjust the pH.

Topical formulations for application to the skin or mucosa may be used for transdermal or transmucosal administration of the therapeutic agent into the blood for systemic distribution. Advantages of topical administration may include circumventing gastrointestinal absorption and first pass metabolism; delivering a therapeutic agent with a short half-life and low oral bioavailability; more controlled and sustained release of therapeutic agents; a more uniform plasma dosing or delivery profile of the therapeutic agent; less frequent administration of the therapeutic agent; minimally invasive or non-invasive; easy self-administration; and increased patient compliance.

In general, compositions suitable for topical administration include, but are not limited to, liquid or semi-liquid preparations, such as sprays, gels, liniments, lotions, oil-in-water or water-in-oil emulsions, such as creams, foams, ointments and pastes, and solutions or suspensions, such as drops (e.g., eye drops, nose drops and ear drops). In some embodiments, the topical composition comprises an aminoadamantyl nitrate dissolved, dispersed, or suspended in a carrier. The carrier may be in the form of, for example, a solution, suspension, emulsion, ointment or gel base and may contain, for example, petrolatum, lanolin, waxes (e.g., beeswax), mineral oil, long chain alcohols, polyethylene or polypropylene glycols, diluents (e.g., water or/and alcohols [ e.g., ethanol or propylene glycol ]), gels, emulsifiers, thickeners, stabilizers or preservatives or any combination thereof. Topical compositions may include, for example, transdermal or transmucosal delivery devices, or topical formulations may be administered by means of, for example, transdermal or transmucosal delivery devices, such as transdermal patches, microneedle patches, or iontophoresis devices. The topical compositions can deliver aminoadamantane nitrate transdermally or transmucosally through a concentration gradient (with or without the use of chemical penetration enhancers) or an active mechanism (e.g., iontophoresis or microneedles).

For transdermal administration, in some embodiments, the topical composition (e.g., transdermal delivery system) comprises a chemical penetration enhancer (e.g., a surfactant [ e.g., sodium laureth sulfate ], optionally in combination with an aromatic compound [ e.g., phenylpiperazine ]) that aids in the transport of the aminoadamantane nitrate through the skin into the systemic circulation. In a further embodiment, the aminoadamantyl nitrate is administered via a transdermal patch. In certain embodiments, a transdermal patch comprises an impermeable backing membrane or layer, a drug reservoir, a semi-permeable membrane that can act as a rate-limiting or rate-controlling diffusion barrier, and an adhesive layer that contacts the skin. The semipermeable membrane may be constructed, for example, of a suitable polymeric material such as nitrocellulose or cellulose acetate, polyisobutylene, polypropylene, polyvinyl acetate, or polycarbonate. Transdermal drug delivery systems, including patches, can be designed to provide controlled and prolonged drug release over a period of about 1 week, 2 weeks, 1 month, or more.

In some embodiments, the aminoadamantyl nitrate is delivered from a sustained release composition. The term "sustained release composition" as used herein encompasses sustained release, extended release, slow release, and controlled release compositions, systems, and devices. The use of sustained release compositions can have benefits such as improved distribution of the amount of drug delivered to one or more target sites over a period of time, including delivery of a therapeutically effective amount of the drug over an extended period of time. In certain embodiments, the sustained release composition delivers the aminoadamantyl nitrate for at least about 1 day, 2 days, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, or longer. In some embodiments, the sustained release composition is a drug encapsulation system, such as a nanoparticle, microparticle, or capsule made from, for example, a biodegradable polymer or/and a hydrogel. In certain embodiments, the sustained release composition comprises a hydrogel. Non-limiting examples of polymers from which the hydrogel can be constructed include polyvinyl alcohol, acrylate polymers (e.g., sodium polyacrylate), and other homopolymers and copolymers having a large number of hydrophilic groups (e.g., hydroxyl or/and carboxylate groups). In other embodiments, the sustained release drug encapsulation system comprises a membrane enclosed reservoir, wherein the reservoir contains a drug and the membrane is permeable to the drug. Such a drug delivery system may be in the form of, for example, a transdermal patch.

In some embodiments, the sustained release composition is an oral dosage form, such as a tablet or capsule. For example, the drug may be embedded in an insoluble porous matrix so that the dissolved drug must first come out of the matrix before it can be absorbed through the gastrointestinal tract. Alternatively, the drug may be embedded in a matrix that swells to form a gel through which the drug is expelled. Sustained release may also be achieved by a single or multilayer osmotic controlled release oral delivery system (OROS). OROS is a tablet with a semipermeable outer membrane and one or more small laser drilled holes therein. As the tablet passes through the body, water is absorbed through the semipermeable membrane via osmotic action, and the resulting osmotic pressure pushes the drug out through one or more pores in the tablet and into the gastrointestinal tract, where it can be absorbed.

In other embodiments, the sustained release composition is formulated as a polymeric nanoparticle or microparticle, wherein the polymeric particle can be delivered, e.g., by injection or from an implant.

For delayed or sustained release of aminoadamantyl nitrate, the composition may also be formulated as a depot that may, for example, be implanted or injected intramuscularly or subcutaneously into a subject.

The aminoadamantyl nitrate compound may also be contained or dispersed in the matrix material. The matrix material may comprise a polymer (e.g., ethylene vinyl acetate) and control the release of the compound by controlling the dissolution or/and diffusion of the compound from, for example, a reservoir, and may enhance the stability of the compound when contained in the reservoir. Such "delivery systems" may be designed as sustained release systems, may be configured as, for example, transdermal or transmucosal patches, and may contain excipients that may facilitate release of the compound, such as water-swellable materials (e.g., hydrogels) that help to expel the compound from the reservoir. Examples of such delivery systems are described in U.S. patent nos. 4,144,317 and 5,797,898.

The delivery system may provide a time-modulated release profile (e.g., pulsatile release) when time-varying plasma levels are desired, or a more continuous or consistent release profile when constant plasma levels are desired. Pulsatile release can be achieved from a single reservoir or multiple reservoirs. For example, where a single pulse is provided per reservoir, multiple pulses are achieved by staggering in time the release of a single pulse from each of the multiple reservoirs ("pulsed" release). Alternatively, multiple pulses may be achieved from a single reservoir by incorporating multiple layers of delivery systems and other materials into the single reservoir. Continuous release may be achieved by incorporating a release system that degrades, dissolves or allows the compound to diffuse through it over a longer period of time. Furthermore, a continuous release can be approximated by several pulses of the compound being released in rapid succession ("digital" release). The active release system may be used alone or in combination with a passive release system, as described in U.S. Pat. No. 5,797,898.

Furthermore, the pharmaceutical composition comprising aminoadamantyl nitrate may be formulated, for example, as liposomes, micelles (e.g., those composed of biodegradable natural or/and synthetic polymers, such as liposomes), microparticles, microspheres, or nanoparticles, whether designed for sustained release or not.

The pharmaceutical compositions may be manufactured in any suitable manner known in the art, for example by means of conventional mixing, dissolving, suspending, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compressing processes.

The compositions may be presented in unit dosage form as a single dose, wherein all active and inactive ingredients are combined in a suitable system and no mixing of the components is required to form the composition to be administered. The unit dosage form contains an effective dose of aminoadamantyl nitrate. Representative examples of unit dosage forms are tablets, capsules or pills for oral ingestion.

Alternatively, the compositions may be presented as a kit, wherein the active ingredients, excipients and carrier (e.g., solvent) are provided in two or more separate containers (e.g., ampoules, vials, tubes, bottles or syringes) and need to be combined to form the composition to be administered. The kit may contain instructions for storing, preparing, and administering the composition (e.g., a solution to be injected intravenously).

The kit may contain all of the active and inactive ingredients in a unit dosage form, or may contain the active and inactive ingredients in two or more separate containers, and may contain instructions for using the pharmaceutical composition to treat a neurodegenerative disorder or other CNS disorder. In some embodiments, a kit contains an aminoadamantane nitrate or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, or polymorph thereof and instructions for administering the compound to treat a neurodegenerative disorder or other CNS disorder.

VIII.Synthesis of aminoadamantane nitrate ester compound

The synthesis of a representative aminoadamantyl nitrate compound is described in the examples and figure 1.

IX.Representative embodiments

The following embodiments of the present disclosure are provided by way of example only:

1. a compound of formula I:

wherein:

R1and R2Independently hydrogen, halide, linear or branched alkyl, linear or branched heteroalkyl, linear or branched alkoxyA group, straight or branched-O-heteroalkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, each of which may be optionally substituted;

R3and R4Independently hydrogen or straight or branched C1-C6Alkyl, or R3、R4And the nitrogen atom to which they are attached form a 3-to 8-membered heterocyclic ring;

R5is hydrogen or straight or branched C1-C6An alkyl group;

x is a bond, straight or branched-alkyl-, straight or branched-heteroalkyl-, straight or branched-O-alkyl-, straight or branched-O-heteroalkyl-, - (CH)2)j-cycloalkyl- (CH)2)k-、-(CH2)j-heterocyclyl- (CH)2)k-、-(CH2)j-aryl- (O)h-(CH2)k-or- (CH)2)j-heteroaryl- (O)h-(CH2)k-, each of which may be optionally substituted;

y is-ONO2Or

m is 0, 1,2, 3, 4 or 5;

j is 0, 1,2 or 3;

k is 0, 1,2 or 3; and is

h is 0 or 1;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

2. A compound of embodiment 1 having formula Ia:

wherein:

R1、R2x and Y are as defined in embodiment 1; and is

n is 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

3. A compound of embodiment 1 having formula IA:

wherein R is1、R2、R3、R4、R5X and m are as defined in embodiment 1;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

4. A compound of embodiment 3 having the formula IAa:

wherein:

R1、R2and X is as defined in embodiment 1; and is

n is 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

5. A compound of embodiment 1 having formula IB:

wherein R is1、R2、R3、R4、R5X and m are as defined in embodiment 1;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

6. A compound of embodiment 5 having formula IBa:

wherein:

R1、R2and X is as defined in embodiment 1; and is

n is 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

7. A compound of any of the preceding embodiments wherein X of the compound of formula I, Ia, IA, IAa, IB or IBa is a bond, straight or branched C1-C6Or C1-C3Alkyl-or straight or branched C1-C6Or C1-C3-O-alkyl-.

8. A compound of embodiment 7 wherein X of the compound of formula I, Ia, IA, IAa, IB or IBa is a bond or straight or branched chain C1-C3Alkyl- [ e.g. -CH2-、-(CH2)2-、-CHCH3、-(CH2)3-、-CHCH2CH3、-CH2CHCH3or-CH (CH)3)CH2-]。

9. A compound of formula II or formula III:

wherein:

R1and R2Independently hydrogen, a halide, a linear or branched alkyl, a linear or branched heteroalkyl, a linear or branched alkoxy, a linear or branched-O-heteroalkyl, a cycloalkyl, a heterocyclyl, an aryl, or a heteroaryl, each of which may be optionally substituted;

R3and R4Independently hydrogen or straight or branched C1-C6Alkyl, or R3、R4And the nitrogen atom to which they are attached form a 3-to 8-membered heterocyclic ring;

R5is hydrogen or straight or branched C1-C6An alkyl group;

x is a bond, straight or branched-alkyl-, straight or branched-heteroalkyl-, straight or branched-O-alkyl-, straight or branched-O-heteroalkyl-, - (CH)2)j-cycloalkyl- (CH)2)k-、-(CH2)j-heterocyclyl- (CH)2)k-、-(CH2)j-aryl- (O)h-(CH2)k-or- (CH)2)j-heteroaryl- (O)h-(CH2)k-, each of which may be optionally substituted;

y is-ONO2Or

m is 0, 1,2, 3, 4 or 5;

j is 0, 1,2 or 3;

k is 0, 1,2 or 3; and is

h is 0 or 1;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

10. A compound of embodiment 9 having formula IV:

wherein:

R1、R2x and Y are as defined in embodiment 9; and is

p is 0, 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

11. A compound of embodiment 10 having formula IVa:

wherein:

x and Y are as defined in embodiment 9; and is

p is 0, 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

12. A compound of embodiment 9 having formula IIA or formula IIIA:

wherein R is1、R2、R3、R4、R5X and m are as defined in embodiment 9;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

13. A compound of embodiment 12 having formula IVA:

wherein:

R1、R2and X is as defined in embodiment 9; and is

p is 0, 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

14. The compound of embodiment 13 having formula iva:

wherein:

x is as defined in embodiment 9; and is

p is 0, 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

15. A compound of embodiment 9 having formula IIB or formula IIIB:

wherein R is1、R2、R3、R4、R5X and m are as defined in embodiment 9;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

16. A compound of embodiment 15 having formula IVB:

wherein:

R1、R2and X is as defined in embodiment 9; and is

p is 0, 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

17. A compound of embodiment 16 having the formula IVBa:

wherein:

x is as defined in embodiment 9; and is

p is 0, 1,2, 3, 4,5 or 6;

or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

18. A compound according to any one of embodiments 9 to 17, wherein-X-Y, -X-ONO2or-X-CH (ONO)2)CH2-ONO2The moiety is attached to the meta position of the phenyl ring.

19. A compound of any one of embodiments 9 to 18 wherein formula II, IIA, IIB, III, IIIA, IIIB, IV, IVa,X of a compound of IVB or IVBa is a bond, straight or branched C1-C6Or C1-C3Alkyl-or straight or branched C1-C6Or C1-C3-O-alkyl-.

20. A compound of any of the preceding embodiments, wherein:

m is 0, 1 or 2;

n is 1,2 or 3; or

p is 0, 1,2 or 3.

21. A compound of any of the preceding embodiments wherein R3And R4Both of which are hydrogen.

22. A compound of any of embodiments 1 through 20 wherein R3And R4One of which is hydrogen and the other is a straight or branched chain C1-C3Alkyl (e.g., methyl or ethyl).

23. A compound of any of embodiments 1 through 20 wherein R3And R4Independently is straight chain C1-C3Alkyl (e.g., methyl or ethyl), optionally the same alkyl.

24. A compound of any of the preceding embodiments wherein R5Is hydrogen.

25. A compound of any of embodiments 1 through 23 wherein R5Is straight or branched C1-C3Alkyl (e.g., methyl or ethyl).

26. A compound of any of the preceding embodiments wherein R1And R2Independently hydrogen or straight or branched C1-C6An alkyl group.

27. A compound of embodiment 26 wherein R1And R2Independently hydrogen or straight or branched C1-C3Alkyl (e.g., methyl, ethyl, or n-propyl).

28. A compound of embodiment 26 or 27 wherein R1And R2Both of which are hydrogen.

29. A compound of embodiment 26 or 27 wherein R1Is hydrogen and R2Is straight or branched C1-C6Or C1-C3Alkyl (e.g. methyl, ethyl or n-propyl), or R2Is hydrogen and R1Is straight or branched C1-C6Or C1-C3Alkyl (e.g., methyl, ethyl, or n-propyl).

30. A compound of embodiment 26 or 27 wherein R1And R2Independently is a straight or branched chain C1-C6Or C1-C3Alkyl (e.g., methyl, ethyl, or n-propyl), optionally the same alkyl.

31. A compound of any of the preceding embodiments wherein R1Radical, R2The group or X group or any combination or all thereof is independently substituted with 1,2 or 3 substituents selected from straight or branched C1-C6Or C1-C3Alkyl, haloalkyl, -OR6、-NR7R8、-ONO2、-CN、-C(=O)R6、-C(=O)OR6、-OC(=O)R6、-C(=O)NR7R8、-NR7C(=O)R6、-OC(=O)OR6、-OC(=O)NR7R8、-NR7C(=O)OR6、-NR6C(=O)NR7R8Aryl and heteroaryl; or/and is substituted with 1 to 6 halogen (e.g. fluorine) or deuterium atoms, or all available hydrogen atoms are replaced by halogen (e.g. fluorine) or deuterium atoms, wherein:

R6independently at each occurrence is hydrogen or straight or branched chain C1-C6Or C1-C3An alkyl group; and is

R7And R8Independently at each occurrence is hydrogen or straight or branched chain C1-C6Or C1-C3Alkyl, or R7、R8And the nitrogen atom to which they are attached form a 3-to 6-membered ring.

32. A compound of embodiment 31 wherein R1Radical, R2The group or X group or any combination or all thereof independently is deuterated alkyl, fluoroalkyl or alkyl-ONO2

33. A compound of any of the preceding embodiments, wherein X has 0, 1,2, 3, 4,5, or 6 carbon atoms.

34. A compound of embodiment 33 wherein X has 0, 1,2 or 3 carbon atoms.

35. A compound of any of the preceding embodiments which is the hydrochloride (HCl) or hydrobromide (HBr) salt.

36. A compound of formula IAa selected from the compounds shown in table 1:

TABLE 1

And pharmaceutically acceptable salts (e.g., hydrochloride and hydrobromide), solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

37. A compound of embodiment 36 selected from:

and pharmaceutically acceptable salts (e.g., hydrochloride and hydrobromide), solvates, hydrates, clathrates, polymorphs and stereoisomers thereof, wherein Et ═ ethyl and Pr ═ n-propyl.

38. A compound of formula IBa selected from the compounds shown in table 2:

TABLE 2

And pharmaceutically acceptable salts (e.g., hydrochloride and hydrobromide), solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

39. A compound of embodiment 38 selected from:

and pharmaceutically acceptable salts (e.g., hydrochloride and hydrobromide), solvates, hydrates, clathrates, polymorphs and stereoisomers thereof, wherein Et ═ ethyl and Pr ═ n-propyl.

40. A compound of formula IVA selected from the compounds shown in table 3:

TABLE 3

And pharmaceutically acceptable salts (e.g., hydrochloride and hydrobromide), solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

41. A compound of embodiment 40 selected from:

and pharmaceutically acceptable salts (e.g., hydrochloride and hydrobromide), solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

42. A compound of formula IVB selected from the compounds shown in table 4:

TABLE 4

And pharmaceutically acceptable salts (e.g., hydrochloride and hydrobromide), solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

43. A compound of embodiment 42 selected from:

and pharmaceutically acceptable salts (e.g., hydrochloride and hydrobromide), solvates, hydrates, clathrates, polymorphs and stereoisomers thereof.

44. A pharmaceutical composition comprising a compound of any of the preceding embodiments, or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof, and one or more pharmaceutically acceptable excipients or carriers.

45. A method of treating a disorder of the Central Nervous System (CNS), comprising administering to a subject in need thereof a therapeutically effective amount of a compound of any one of embodiments 1 to 43, or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof.

46. The method of embodiment 45, wherein said CNS disorder is a neurodegenerative disorder.

47. The method of embodiment 46, wherein said neurodegenerative disorder is selected from the group consisting of dementia (e.g., Alzheimer 'S disease, vascular dementia, Lewy body dementia, frontotemporal dementia, and HIV-associated dementia), Huntington' S disease, Parkinson 'S disease, multiple system atrophy (Lewy syndrome), cerebellar degeneration, ataxia (e.g., cerebellar ataxia, spinocerebellar ataxia, Friedreich' S ataxia, and ataxia-telangiectasia [ Lewy-Barre syndrome ]), motor neuron disease (e.g., amyotrophic lateral sclerosis [ A L S ], primary lateral sclerosis [ P L S ], progressive muscular atrophy [ PMA ] and spinal muscular atrophy [ SMA ]), multiple sclerosis, vision disorders or impairment due to neurodegeneration of the visual pathway (e.g., optic neuropathy/atrophy, glaucoma, and age-related macular degeneration) and sensorineural hearing loss.

48. The method of embodiment 47, wherein said neurodegenerative disorder is Alzheimer ' S disease, vascular dementia, Huntington ' S disease, Parkinson ' S disease, or A L S.

49. The method of embodiment 45, wherein said CNS disorder is selected from the group consisting of cerebrovascular disorders (including cerebral ischemia [ including acute ischemia, such as stroke, chronic ischemia, such as vascular dementia, cerebral ischemia/reperfusion injury, and nerve injury caused by hypoxia or/and glucose levels in the brain ], intracerebral hemorrhage and retinopathy), brain injury and trauma (including traumatic brain injury, diffuse axonal injury, primary and secondary brain injury, focal and diffuse brain injury, hypoxic and hypoxic brain injury, intracerebral hemorrhage and brain edema), spinal cord injury (due to, e.g., trauma, ischemia or degenerative disorders), epilepsy (including nerve injury caused by epileptic seizures), movement disorders (e.g., levodopa-induced dyskinesia and tardive dyskinesia), spasticity, pain (e.g., acute pain, chronic pain, l pain, etc.), spasticity, and pain, Allodynia, complex regional pain syndrome [ CRPS ], fibromyalgia, hyperalgesia, inflammatory pain, neuropathic pain, post-operative pain, cancer-related pain, drug-induced pain, and injury-induced pain), headache (including primary headaches [ e.g., migraine, cluster headache, and tension headache ] and secondary headaches [ due to, e.g., cerebrovascular disorders, cerebral hemorrhage, brain injury, brain infection, or brain tumor), neurodevelopmental disorders (including MEF2C haploinsufficiency syndrome [ MCHS ], autism spectrum disorders [ including autism ], developmental retardation, mental disability, fragile X syndrome, attention deficit hyperactivity disorder [ ADHD ], and schizophrenia), mood disorders (including depression [ e.g., major and refractory depression ], bipolar disorders, and dementia-related mood disorders), and anxiety disorders (including generalized anxiety disorder, brain pain, brain injury, post-operative pain, cancer-related pain, brain injury-induced pain, brain injury-induced pain, brain tumor, Stress disorders [ e.g., acute stress disorder, post-traumatic stress disorder and chronic stress ] and obsessive-compulsive disorder).

50. The method of embodiment 49, wherein said CNS disorder is cerebral ischemia, traumatic brain injury, epilepsy, pain, or autism spectrum disorder.

51. The method of any one of embodiments 45 to 50, wherein the compound is administered orally.

52. The method of any one of embodiments 45 to 50, wherein the compound is administered parenterally (e.g., intravenously, intramuscularly, or subcutaneously).

53. The method of any one of embodiments 45 to 52, wherein the compound is administered in a daily dose of about 1mg, 5mg, or 10mg to about 100 mg.

54. The method of embodiment 53, wherein said compound is administered at a daily dose of about 1mg, 5mg, 10mg, 15mg, 20mg, 25mg, 30mg, 40mg, 50mg, 60mg, 70mg, 80mg, 90mg, or 100 mg.

55. The method of embodiment 53 or 54, wherein said compound is administered in a daily dose of about 5mg or 10mg to about 50 mg.

56. The method of any one of embodiments 45 to 55, further comprising administering an additional therapeutic agent.

57. A compound of any one of embodiments 1 to 43, or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof, for use as a medicament.

58. A composition comprising a compound of any one of embodiments 1 to 43, or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof, for use as a medicament.

59. Use of a compound of any one of embodiments 1 to 43, or a pharmaceutically acceptable salt, solvate, hydrate, clathrate, polymorph, or stereoisomer thereof, for the manufacture of a medicament.

60. A compound, composition or use of embodiment 57, 58 or 59, respectively, wherein the medicament is for treating a CNS disorder, such as a neurodegenerative disorder or a non-neurodegenerative disorder.

61. A kit, comprising:

83页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:马来酸曲美布汀的多晶型物及其使用方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!