Structurally modified opioids for the prevention and treatment of diseases and disorders

文档序号:245823 发布日期:2021-11-12 浏览:19次 中文

阅读说明:本技术 用于预防和治疗疾病和病症的结构修饰的阿片类药物 (Structurally modified opioids for the prevention and treatment of diseases and disorders ) 是由 保罗·L·曼佛雷蒂 查尔斯·E·因图里西 A·马塔雷 S·德马丁 J·斯格里尼亚尼 A·卡 于 2019-10-10 设计创作,主要内容包括:本发明的方面涉及结构修饰的阿片类药物(SMO),与具有NMDAR调节活性的现有药物相比,其导致改善的NMDAR调节活性和改善的PK和PD参数。导致SMO的阿片类或阿片类对映体的结构修饰可以通过从头开始合成过程来获得;通过在外消旋体或一种对映体合成过程中的任何中间步骤修改阿片类药物的合成工艺;或通过在合成后修饰阿片类或阿片类对映体的结构。硝酸酯取代特别相关,尤其是当与氘代取代和/或卤素取代相关时。(Aspects of the invention relate to Structurally Modified Opioids (SMO) that result in improved NMDAR modulating activity and improved PK and PD parameters compared to existing drugs with NMDAR modulating activity. Structural modifications of the opioid or opioid enantiomer that result in SMO can be obtained by starting the synthesis process from the beginning; modifying the synthesis process of the opioid by any intermediate step during the synthesis of the racemate or one of the enantiomers; or by modifying the structure of the opioid or opioid enantiomer after synthesis. Nitrate substitutions are particularly relevant, especially when associated with deuterated substitutions and/or halogen substitutions.)

1. A compound according to formula I structurally analogous to dextromethorphan:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium, or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy,alkoxy, aryl, aryloxy, heterocyclic, nitro, nitrate substituted;

R3is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate; and

n is 1 to 4.

2. A compound according to formula II structurally analogous to levopropoxyphene:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium, or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is optionally substituted in one or more positions by deuterium, halogen, hydroxy, alkoxy, nitrateA substituted alkyl group;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate; and

n is 1 to 4.

3. A compound according to formula III structurally analogous to dextro-isomethadone:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium, or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate; and

n is 1 to 4.

4. A compound according to formula IV having a structure similar to levomoramide:

wherein NR is1R2Optionally through C3-C12Cycloalkyl or heterocyclyl ring closure, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

if NR is present1R2Not cyclized, then R1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; r2Is hydrogen, deuterium, or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is hydrogen; or selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally deuterated at one or more positionsHalogen, alkyl, hydroxyl, alkoxy, aryl, aryloxy, heterocyclic, nitro, nitrate;

NR4R5optionally through C3-C12Cycloalkyl or heterocyclyl ring closure, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

if NR is present1R2Not cyclized, then R1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; r2Is hydrogen, deuterium, or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; and

n is 1 to 4.

5. A compound according to formula V structurally analogous to N-methyl-dextromethorphan:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium, or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R5is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

X-is a nitrogen-counterion; and

n is 1 to 4.

6. A compound according to formula VI structurally analogous to levorphanol:

wherein R is1Is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate, or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate, or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally at one or morePosition substituted with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate, or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; and

R4is hydrogen or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate.

7. A compound according to formula VII structurally analogous to dextromethorphan or dextrorphan:

wherein R is1Is hydrogen, nitrate, or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate, or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate, or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen,alkyl, hydroxyl, alkoxy, aryl, aryloxy, heterocyclic, nitro, nitrate substituted;

R4is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate, or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; and

R5is hydrogen or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate.

Technical Field

The present invention relates generally to the development of structurally modified opioids for the prevention and treatment of various diseases and disorders.

Background

This section is intended to introduce the reader to various aspects of art, which may be related to various aspects of the present invention, which are described and/or claimed below. This discussion is believed to be helpful in providing the reader with background information to facilitate a better understanding of the various aspects of the present invention. Accordingly, it should be understood that these statements are to be read in this light, and not as admissions of prior art.

The present inventors have previously disclosed that certain pharmaceutical compounds currently classified as opioids and opioid enantiomers, including those having minimal or no clinically relevant opioid agonist activity, alone or in combination with other drugs (including opioid antagonists), may be useful in the treatment of certain diseases and conditions (see U.S. patent No. 6,008,258, U.S. patent No. 9,468,61, and international patent application No. PCT/US 2018/016159).

N-methyl-D-aspartate receptor (NMDAR) is increasingly recognized as a potential therapeutic target for a variety of human diseases caused by genetic or environmental factors, or a combination of genetic plus environmental (G + E) factors. However, a single NMDAR antagonist/modulator may be ineffective for multiple diseases associated with NMDAR dysfunction.

Disclosure of Invention

Certain exemplary aspects of the invention are set forth below. It should be understood that these aspects are presented merely to provide the reader with a brief summary of certain forms the invention might take and that these aspects are not intended to limit the scope of the invention. Indeed, the invention may encompass a variety of aspects that may not be set forth below.

In one aspect of the present invention, the present inventors now disclose that by modifying the structure of opioids and their enantiomers, new drugs (referred to herein as structurally modified opioids or SMO) can be designed and synthesized, those with unique Pharmacokinetic (PK) and Pharmacodynamic (PD) properties that have potentially safe and effective therapeutic effects, including effects on glutamate receptors, including NMDAR, as well as different effects on NMDAR subtypes, for the treatment and prevention of various diseases and disorders, including new drugs for improving cognitive and social skills.

In another aspect of the invention, structural modifications of the opioid or opioid enantiomer may be obtained as follows: (a) restarting the synthesis process; (b) altering the process of opioid synthesis at any intermediate step during the synthesis of the racemate or one of the enantiomers; or (c) altering the structure of the opioid or an enantiomer of the opioid after synthesis.

NMDAR may be a potential therapeutic target for SMO as described herein. In this regard, NMDAR is increasingly recognized as a potential therapeutic target for a variety of human diseases caused by genetic or environmental factors, or a combination of genetic plus environmental (G + E) factors. Furthermore, as described above, a single NMDAR antagonist/modulator may not be effective against multiple diseases associated with NMDAR dysfunction. However, the present inventors now disclose that newly designed molecules (SMOs) can be used to select diseases and disorders by preferentially targeting select cell populations, cell sites, brain regions, specific diseases, disease stages, disorders, and specific periods of an individual's life cycle. The SMO may be optimized for Pharmacokinetic (PK) parameters (e.g., optimal lipid solubility to reach receptor subtypes and receptor sites in selected brain regions and/or selected CNS) or changes in metabolic parameters that may alter drug half-life, including half-life in a selected patient population. In addition, the SMO can be optimized for Pharmacodynamic (PD) parameters, such as action at selected NMDAR domains and sites (e.g., the transmembrane domain and POP site of NMDAR and/or the extracellular domain and NO site of NMDAR). And SMO can be optimized for certain NMDAR subtypes (e.g. NR1, NR2A-D, NR3A-B, as detailed below), and possibly for other selective actions of different receptors (other than NMDAR) and transporters, as detailed below.

Other aspects of the invention may include or relate to, for example, compounds of formulae I-VII below:

a compound of formula I structurally analogous to dextromethorphan:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions by deuterium, halogenAlkyl, hydroxyl, alkoxy, aryl, aryloxy, heterocyclic, nitro, nitrate substituted;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate; and n is 1 to 4.

A compound of formula II structurally similar to levopropoxyphene:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally deuterated, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl at one or more positionsNitro, nitrate substitution;

R2is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate; and n is 1 to 4.

A compound of formula III structurally analogous to dextro-isomethadone:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy,heterocyclyl, nitro, nitrate;

R3is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate; and n is 1 to 4.

A compound of formula IV structurally similar to levomoramide:

wherein NR is1R2Optionally through C3-C12Cycloalkyl or heterocyclyl ring closure, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

if NR is present1R2Not cyclized, then R1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; r2Is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally deuterated, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy in one or more positionsHeterocyclyl, nitro, nitrate;

R3is hydrogen; or selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

NR4R5optionally through C3-C12Cycloalkyl or heterocyclyl ring closure, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

if NR is present1R2Not cyclized, then R1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; r2Is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; and

n is 1 to 4.

A compound of formula V having a structure analogous to N-methyl-dextromethorphan:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclic radicals, ofAny one optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R5is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

X-is a nitrogen-counterion; and

n is 1 to 4.

A compound of formula VI structurally similar to levorphanol:

wherein R is1Is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium, halogen, hydroxyl, nitro, nitrate or is selected from alkyl, alkoxyAryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; and

R4is hydrogen or selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate.

A compound of formula VII structurally analogous to dextromethorphan or dextrorphan:

wherein R is1Is hydrogen, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclic groups, any of which is optionally deuterated at one or more positionsHalogen, alkyl, hydroxyl, alkoxy, aryl, aryloxy, heterocyclic, nitro, nitrate;

R4is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; and

R5is hydrogen or selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate.

General examples of such compounds also include: dextromethorphan fluoro derivatives (-F), including fluoro dextromethorphan; dextromethorphan nitro derivative (-NO)2) Including nitro-dextromethorphan; dextromethorphan fluoronitro derivatives, including fluoronitro dextromethorphan; and deuterated dextromethorphan derivatives for dextromethorphan modified as described above (deuterated dextromethorphan fluoro derivatives (-F) including fluoro dextromethorphan; deuterated dextromethorphan nitro derivatives (-NO)2) Including nitro dextromethorphan; and deuterated dextromethorphan fluoronitro derivatives, including fluoronitrodextromethorphan).

General examples of such compounds may also include dextro-isomethadone (dexteroisothiadadone) derivatives, including: dextro-isomethadone fluoro derivatives including fluoro dextro-isomethadone; dextro-isomethadone nitro derivatives including nitro-dextro-isomethadone; dextromethorphan fluoronitro derivatives, including fluoronitro dextromethorphan; and deuterated dextro-isomethadone derivatives modified with dextro-isomethadone as described above.

General examples of such compounds may also include N-methyl-dextromethorphan derivatives, including: n-methyl-dextromethorphan fluoro derivatives including fluoro-N-methyl-dextromethorphan; n-methyl-dextromethorphan nitro derivatives, including nitro-N-methyl-dextromethorphan; n-methyl-dextromethorphan fluoronitro derivatives, including fluoronitro-N-methyl-dextromethorphan; and deuterated N-methyl-dextromethorphan derivatives modified as described above.

General examples of such compounds may also include levomoramide derivatives, including: levo-morpholino fluoro derivatives including fluoro-levo-morpholino; levo-morpholinone nitro derivatives, including nitro-levo-morpholinone; levo-morpholino fluoronitro derivatives including fluoro-nitro-levo-morpholino; and deuterated levorphalamda derivatives modified as described above.

General examples of such compounds may also include levopropoxyphene derivatives, including: levopropoxyphene fluoro derivatives, including fluoro-levopropoxyphene; levopropoxyphene nitro derivatives, including nitro-levopropoxyphenyl; levopropoxyphene fluoronitro derivatives, including fluoro-nitro-levopropoxyphene; and deuterated levopropoxyphene derivatives modified as described above.

General examples of such compounds may also include levorphanol derivatives, including: levorphanol fluoro derivatives including fluoro-levorphanol; levorphanol nitro derivatives, including nitro levorphanol; levorphanol fluoronitro derivatives including fluoro-nitro-levorphanol; and deuterated levorphanol derivatives modified as described above for levorphanol.

General examples of such compounds may also include dextromethorphan and dextrorphan (dextrorphan) derivatives, including: dextromethorphan and dextrorphan-fluoro derivatives, including fluoro-dextromethorphan and nitro-dextrorphan; dextromethorphan and dextrorphan-nitro derivatives, including nitro-dextromethorphan and nitro-dextrorphan; dextromethorphan and dextrorphan fluoronitro derivatives, including fluorodextromethorphan and fluoro-nitro-dextrorphan; and deuterated dextromethorphan and deuterated dextrorphan derivatives modified as described above.

Drawings

The accompanying drawings, which are incorporated in and constitute a part of this specification, illustrate embodiments of the invention and, together with a general description of the invention given above, and the detailed description of the embodiments given below, serve to explain the principles of the invention.

Figure 1 shows the effect of ketamine and d-methadone on immobility, climbing and swimming counts in the forced swim test ("FST"). Data represent mean ± SEM. For immobility: p-0.0034 for ketamine, 10mg/kg for d-methadone, 0.0007, and p <0.0001 for d-methadone 20 and 40mg/kg compared to vehicle group ANOVA. For climbing: p <0.05 for d-methadone 40mg/kg compared to the carrier. For swimming: p <0.05 for ketamine and d-methadone 10mg/kg, p <0.0001 for d-methadone 20mg/kg, and p <0.0003 for d-methadone 40mg/kg, compared to the carrier ANOVA.

Figures 2A-2E show the effect of d-methadone and ketamine on the female urine sniff test ("futt") and the novel inhibition feeding test ("NSFT"). In fig. 2A, a time table of dosing and testing rats is shown, wherein d-methadone or ketamine is administered, followed by various tests. Fig. 2B shows rats tested in futt 24 hours later. Figure 2C shows rats tested for autonomous activity (LMA) after 2 days. Figure 2D shows rats subjected to NSFT testing 72 hours later. FIG. 2E shows home cage rearing. Results are mean ± s.e.m. FUST: one-way ANOVA, F3,42 ═ 3.26, p ═ 0.031; LSD of Fisher (Fisher): veh × Met, p ═ 0.025; veh × Ket, p ═ 0.046; n is 9-12/group. NSFT: one-way ANOVA, F3,27 ═ 4.87, p ═ 0.008; snowy LSD: veh × Met, p ═ 0.035; veh × Ket, p ═ 0.005; n is 7-8/group.

Figures 3A-3E show that a single dose of D-methadone prevents depressive behavior caused by chronic unpredictable stress ("CUS") exposure. Fig. 3A shows the time course of the CUS protocol, drug dosage and behavioral analysis. D-methadone and ketamine prevent the behavioral effects of CUS in fig. 3B [ Sucrose Preference Test (SPT) (F3,45 ═ 2.99) ], fig. 3C [ FUST (F3,46 ═ 5.43) ], and fig. 3D [ NSFT (F3,46 ═ 6.79) ]. No difference was found in water sniffing or (fig. 3E) home cage food consumption. Results are mean ± s.e.m., n-9-15/group. P <0.05, one-way ANOVA and Duncan (Duncan) post hoc test.

Fig. 4A and 4B show the effect of d-methadone on mTORCI signaling and synaptophin. Rats were given d-methadone and the levels of mTORCI signaling protein and synaptophin were measured in PFC and hippocampus.

5A and 5B show that brain-derived neurotrophic factor (BDNF) plasma levels from a 25mg group MAD study were tested prior to any treatment and 4 hours after 25mg (6 patients) or placebo (2 patients) administration of d-methadone on days 2, 6 and 10. In the d-methadone-treated group, 6 of 6 subjects showed an increase in BDNF levels after d-methadone treatment compared to the pre-treatment levels, with the BDNF plasma levels at day 10 after treatment being 2-17 times the pre-treatment BDNF levels. In contrast, BDNF plasma levels remained unchanged in both placebo subjects. When placebo subjects were included in the analysis, plasma BDNF levels measured on days 2 and 10 significantly correlated with plasma levels of d-methadone. Day 2 p is 0.028, day 6 p is 0.043, day 10 p is 0.028; all compared to BDNF plasma levels before treatment.

Figure 6 shows cell viability of ARPE-19 cells after treatment with NMDAR agonist L-glutamic acid alone or in combination with NMDAR antagonists MK-801 and dextromethorphan. Dextromethorphan may also be referred to herein as "REL-1017". More specifically, FIG. 6 shows cell viability of ARPE-19 cells after treatment with NMDAR agonist L-glutamic acid (1mM L-Glu) alone or in combination with NMDAR antagonists MK-801(1mM L-Glu + MK-801) and REL-1017(1mM L-Glu + REL-1017). P <0.01 compared to vehicle (one-way ANOVA followed by Dunnett post-test). The concentration of dextromethorphan (Rel-1017) was 30 micromolar in all experiments.

FIG. 7 shows ROS production in ARPE-19 cells. Treatment with L-glutamic acid (1mM L-Glu), pretreatment with MK-801(1mM L-Glu + MK-801) and REL-1017(1mM L-Glu + REL-1017).

FIGS. 8A-8D show immunofluorescence of p65 of ARPE-19 after treatment with L-glutamic acid (1mML-Glu) and pretreatment with MK-801(1mML-Glu + MK-801) and REL-1017(1mML-Glu + REL-1017). Nuclei were labeled with DAPI. In FIGS. 8A-8D, the labeled nuclei appear white, while the immunofluorescence of p65 appears gray.

FIGS. 9A and 9B show graphical representations of fluorescence intensity of p65(A) and co-localized p65-DAPI (B) in immunocytochemistry experiments. Treatment with L-glutamic acid (1m ML-GLU), pretreatment with MK-801(1m ML-GLU + MK-801) and REL-1017(1m ML-GLU + REL-1017). Fluorescence intensity and Pearson r indicated that the degree of co-localization between p65 and the nuclear-labeled DAPI was calculated using ImageJ software. P <0.05 compared to vehicle (one-way ANOVA followed by Dunnett post test).

Figure 10 shows the relative quantification of NMDAR1 gene expression in ARPE-19 cells after the following treatment conditions: l-glutamic acid (1mML-Glu), pre-treated with MK-801(1mML-Glu + MK-801) or REL-1017(1mML-Glu + REL-1017). Compared to vehicle, p <0.0001 (one-way ANOVA followed by Dunnett post-test).

FIGS. 11A and 11B show the relative quantification of p65 gene expression in ARPE-19 cells subjected to the following treatment conditions. Treatment with L-glutamic acid (1mML-Glu), pretreatment with MK-801(1mML-Glu + MK-801) and REL-1017(1mML-Glu + REL-1017). P <0.05 compared to vehicle (one-way ANOVA followed by Dunnett post test). In the scatter plot, the vector-treated cells and the REL-1017-treated cells were compared. Compared to vehicle,. p <0.01 (student t-test of unpaired data).

FIG. 12 shows the relative quantification of TNF- α gene expression in ARPE-19 cells subjected to the following treatment conditions: l-glutamic acid (1mML-Glu), pretreated with MK-801(1mML-Glu + MK-801) and REL-1017(1mML-Glu + REL-1017). P <0.01 compared to vehicle (one-way ANOVA followed by Dunnett post-test).

FIG. 13 shows the relative quantification of IL-6 gene expression in ARPE-19 cells subjected to the following treatment conditions: l-glutamic acid (1m ML-Glu), pretreated with MK-801(1m ML-Glu + MK-801) and REL-1017(1m ML-Glu + REL-1017).

Detailed Description

One or more specific embodiments of the present invention will be described below. In an effort to provide a concise description of these embodiments, all features of an actual implementation may not be described in the specification. It should be appreciated that in the development of any such actual implementation, as in any engineering or design project, numerous implementation-specific decisions must be made to achieve the developers' specific goals, such as compliance with system-related and business-related constraints, which may vary from one implementation to another. Moreover, it should be appreciated that such a development effort might be complex and time consuming, but would nevertheless be a routine undertaking of design, fabrication, and manufacture for those of ordinary skill having the benefit of this disclosure.

For the purposes of this disclosure, the inventors define "disease" as human and veterinary diseases and conditions at various stages, from preclinical to late stage, (including symptoms and signs of disease, including prodromal symptoms and other manifestations of disease), and aging (including accelerated aging and disease due to physical and chemical factors, including environmental factors, toxins, and drugs, food, and lack of nutrients and vitamins).

For the purposes of this disclosure, the inventors define "disorders" as poor cognitive performance relative to personal potential and goals, as well as social skills deficits relative to personal potential and goals, and a deficient special feeling relative to personal potential and goals.

For the purposes of this disclosure, "treatment" is defined by the inventors as the treatment, prevention, and amelioration of diseases and conditions.

For the purposes of this disclosure, the inventors define "structurally modified opioids" (SMO), the composition of the subject matter of this disclosure, as the new active pharmaceutical ingredient designed or derived from the structural modification of opioids, including the modification of the molecule in any intermediate step of opioid synthesis.

For the purposes of this disclosure, the inventors define "opioids" as synthetic and natural drugs that bind to opioid receptors, including agonists, partial agonists and antagonists, including opioid enantiomers, particularly opioid and opioid enantiomers, which have minimal or no clinically relevant opioid agonist activity at doses that exert other pharmacological effects (e.g., NMDAR modulation), and particularly enantiomers with ion channel modulating activity, particularly N-methyl-D-aspartate receptor (NMDAR) and modulators of other ion channels, and particularly opioid and opioid enantiomers, which have minimal or no clinically relevant opioid activity with other Nervous System (NS) receptors and transporters, including resulting in effects such as 5-hydroxytryptamine, Neurotransmitter changes in NE, DA, GABA, or changes in neurotrophic factors such as BDNF, or changes in synaptophysins such as PD95, GluR1, synaptophysin, NMDAR1, and include particularly novel compounds that have a role in both NMDAR and nitric oxide pathways.

As noted above, in one aspect of the present invention, the present inventors now disclose that by modifying the structure of opioids and their enantiomers, new drugs (referred to herein as structurally modified opioids or SMOs) can be designed and synthesized that have unique Pharmacokinetics (PKs) and Pharmacodynamic (PD) properties with potentially safe and effective therapeutic effects, including effects on glutamate receptors, including NMDAR, as well as different effects on NMDAR subtypes, for the treatment and prevention of various diseases and conditions, including drugs for improving cognitive and social skills.

In another aspect of the invention, structural modifications of the opioid or opioid enantiomer may be obtained as follows: (a) restarting the synthesis process; (b) altering the process of opioid synthesis at any intermediate step during the synthesis of the racemate or one of the enantiomers; or (c) altering the structure of the opioid or an enantiomer of the opioid after synthesis.

NMDAR may be a potential therapeutic target for SMO as described herein. In this regard, NMDAR is increasingly recognized as a potential therapeutic target for a variety of human diseases caused by genetic or environmental factors, or a combination of genetic plus environmental (G + E) factors. Furthermore, as described above, a single NMDAR antagonist/modulator may not be effective against multiple diseases associated with NMDAR dysfunction. However, the present inventors now disclose that newly designed molecules (SMOs) can be used to select diseases and disorders by preferentially targeting select cell populations, cell sites, brain regions, specific diseases, disease stages, disorders, and specific periods of an individual's life cycle. The SMO may be optimized for Pharmacokinetic (PK) parameters (e.g., optimal lipid solubility to reach receptor sites in selected brain regions and/or selected CNS) or changes in metabolic parameters that may alter drug half-life, including half-life in a selected patient population. In addition, the SMO can be optimized for Pharmacodynamic (PD) parameters, such as action at selected NMDAR domains and sites (e.g., the transmembrane domain and POP site of NMDAR and/or the extracellular domain and NO site of NMDAR). And SMO can be optimized for certain NMDAR subtypes (e.g. NR1, NR2A-D, NR3A-B, as detailed below), and potentially for other selective actions of different receptors (other than NMDAR) and transporters, as detailed below.

The tetrameric form of NMDAR is formed from more than 3000 amino acids. The protein complex is surrounded by the extracellular medium, cytoplasm and cell membrane [ NMDAR-dependent regions/domains: amino terminal domain (AMT), Agonist Binding Domain (ABD), transmembrane domain (TMD), Carboxy Terminal Domain (CTD)]. The complex structure of this four-fusion system, and the many variables it brings, including the variation of the NMDAR subunit, [ NR1, NR2A-D, NR3A-B, encoded by seven different genes with genetic and epigenetic differences, including alternative splice variants (NR1 contains 8 different splice variant isoforms) and SNPs of subunits]Thus, the NMDAR becomes a potential culprit for various diseases and targets of various different medicines. Differences in the spatial (brain regions and circuits and neuronal subpopulations and non-neuronal cells, including astrocytes, even additional CNS cell populations expressing NMDAR) and temporal (age-related), the location of the NMDAR on the cell membrane (pre-and/or post-synaptic and extra-synaptic), the absolute number of NMDARs on the cell membrane (varying by the patient and the disease and/or disorder and during development and throughout the life cycle and disease process of the patient), the number of NMDA channels opened and closed at a given time, the time of opening and closing states during physiological activity, pathological activity and during occlusion caused by toxins or drugs, desensitization during physiological conditions, pathological conditions (including modulation of protons and pH, including long-term exposure to agonists/co-agonists, and comprises a pair of Mg2+、Zn2+、Ca2+NO, etc. mediationOr the length of time that a channel remains open or closed following drug or toxin modulation, including activation or inactivation of endogenous toxins (e.g., quinolinic acid) or foods (e.g., polyamine-rich foods, alcohol), including excess neurotransmitters (e.g., glutamate)), with different effects on the open and closed state of specific membrane regions, specific neuronal and astrocyte populations, and specific regions of the brain (even additional CNS), as well as temporal (age-related) changes in the spatial (within specific brain regions, even outside the brain), functional (within different cell populations), and subunit, and on receptor tetramer complexes for agonists (glutamate/NMDA), co-agonists (glycine and d-serine), and modulators [ (Mg and NO protons, Zn-serine)2+(blockers) and polyamines (activators)]The diversity of binding sites for drugs (aminoglycosides, cisplatin) and toxins (domoic acid) and antibodies (NMDAR encephalitis), the diversity and variability of electrical events affecting NMDAR activity (changes in membrane potential), including minimal events, and their biochemical pathological and physiological consequences, as well as events mediated by ionic currents (including biochemical events), in particular Ca2+Can be differently influenced by new chemical entities (SMO) specifically designed and optimized for modulation of NMDAR, with the potential to be new drugs.

These potential drugs (SMOs) may have different effects in different cell populations, including modulating inhibitory function of certain interneurons, which may be hyperactive or hypoactive, and play a role in diseases such as depression or restless leg syndrome and other diseases such as neurodevelopmental diseases or neurodegeneration, and different effects based on genetically determined receptor variation between individuals, or changes in physiological and pathological states in the presence of certain environmental factors (G + E) (neurodevelopmental diseases) or simply predisposition to certain diseases, alterations in receptor molecular structure and finally deregulation and impairment of NMDAR in pathological conditions (human diseases, including CVS degenerative diseases, including those caused by environmental factors (toxins), medical and ageing or even food products, such as polyamine-rich food products or alcohol or lack of certain nutrients and vitamins).Injury, altered membrane potential, biochemical reactions (including enzymatic and signaling cascades (e.g., Ras-ERK)) and their consequences, by altered ionic currents (including Ca) through NMDA receptor pores2+) And (5) triggering. These are all factors highlighting the multiple role and complexity of NMDAR in physiological and pathological states, but also their importance and potential as viable therapeutic targets for new safe and effective drugs, in particular new drugs with unique PK and PD properties, which may be selective or simply safer and/or more effective against specific diseases (HansenKB, YiF, Perszyk RE, Furukawa H, Wollmuth LP, Gibb AJ, Traynelis sf. structural, functional and allosteric modulation of NMDA receptors (Structure, function, and allosteric modulation of receptors), J Gen Physiol, 8.6.2018.150 (8): 1081-1105. doi: 10.1085/jjj.201812032. epub.8.7.23.23).

Channel pore blocking agent

Very subtle structural differences between different NMDAR blockers/modulators, including structural differences between drugs acting on the same domain (transmembrane domain) and site (PCP site) of an NMDAR, such as amantadine and memantine or PCP and ketamine, may alter their Pharmacokinetic (PK) and Pharmacodynamic (PD) properties, including different activities at receptor subtypes, and thus novel drugs, including drugs SMO, drugs with similar but not identical chemical structures compared to opioid and opioid enantiomers, may offer potential advantages for selected diseases or conditions and selected patient populations. These advantages (or disadvantages) may even result from very similar intra-molecular subtle structural differences. These structural molecular modifications may result in changes in PK and PD (and thus changes in the effect on, for example, any of the NMDAR variables described above), resulting in an effect that may be beneficial to a patient subpopulation.

Amantadine is FDA approved for parkinson's disease, while memantine is structurally very similar to amantadine, but has a higher affinity for the NR1-NR2BNMDAR subtype than the NR1-NR2A subtype, and is FDA approved for alzheimer's disease (only late and late); FDA approval of dextromethorphan (used in combination with quinidine to offset PK deficiency due to its very short half-life) for mood-unstable patients secondary to pseudobulbar palsy; ketamine is approved for use as an anesthetic, and is currently used in many specialty clinics in the united states (including clinics affiliated with the harvard medical school) for the treatment of depression. Esketamine (esketamine), the levorotatory enantiomer of ketamine, has recently received FDA approval for the treatment of depression. All of these drugs are believed to exert therapeutic effects on the very specific diseases and conditions listed above through modulation of dysfunctional, hyperactive receptors by NMDAR. Although their putative roles in common domains and sites of NMDAR (transmembrane domain and PCP site) have similar micromolar affinities, these drugs show different PK and PD characteristics, which is also true for structurally very similar drugs such as memantine and amantadine or PCP and ketamine. These PK/PD differences may explain their selective effectiveness for one disease but not another or for indications related to NMDAR dysfunction, including effectiveness at specific stages of the same disease but not other stages, such as memantine for the treatment of alzheimer's disease.

Furthermore, in the current clinical NMDAR antagonists used (amantadine for parkinson's disease, memantine for alzheimer's disease, dextromethorphan for pseudobulbar palsy, ketamine for anesthesia and depression) -all of these appear to act at the same site of the channel within the NMDAR (or close to each other), close to Mg2+Sites, in the so-called PCP site, located within the transmembrane portion of the NMDAR, have similar affinity-indications varying widely in the micromolar range. While dysfunctional NMDARs may be present in a variety of diseases, including the diseases cited above and those listed in international patent application No. PCT/US2018/016159, the extent of dysfunction, NMDAR location on cell membranes, cell type, presence in specific cell populations and circuits and brain regions (or any of the variables listed above) may vary greatly between different diseases and may be affected differently by different drugs; the stage of the disease may determine the effectiveness or ineffectiveness of the same drug, even within the same indication (memantine is approved for moderate and severe alzheimer's disease, as described above, but not shownEfficacy on mild alzheimer's disease). In addition, amantadine and memantine are very similar in structure, but differ in their indications and effects. Similarly, modification of the PCP molecule derived from Ketamine and the differences in the "capture, onset, offset" of these two drugs determine the clinical utility of Ketamine and the lack of clinical indications and toxicity of PCP (Zanos P, Moaddel R, Morris PJ, Riggs LM, Highland JN, Georgiou P, Pereira EFR, Albuquerque EX, Thomas CJ, Zarate CA Jr, Gould TD. Ketamine and Ketamine Metabolite Pharmacology: Insights into the mechanism of treatment (Ketamine and Ketamine metabolism Pharmacology: insight into pharmacological Mechanisms of therapeutics, 7.2018, 70(3): 621).

Thus, the NMDAR complex can be affected differently not only by drugs acting on different binding sites of the receptor, but also by drugs acting on putatively the same site (PCP site) or sites adjacent thereto and having similar but not exactly the same mechanism and affinity (differences in initiation/migration/capture and different affinities for specific NMOAR subtypes).

This premise underscores the great potential in the area of discovery of new NMDAR antagonists, since the PK and PD properties characteristic of new molecules will lead to different effects and influences on cells. Thus, these effects and influences (e.g., blocking/modulating effects at NMDAR), including their mechanisms (e.g., non-competitive), may prove beneficial for targeting specific diseases and disease stages. Furthermore, certain novel drugs (SMOs) disclosed by the present inventors may exert other effects at sites other than NMDAR, as is the case with dextromethorphan, and these effects may also be beneficial for particular diseases and patients (the "other effects" of dextromethorphan are detailed below as examples of potential "other effects" of SMO).

Potential of selected drugs in the opioid family for NMDAR modulation

As the inventors disclose in patent application No. PCT/US2018/016159, the selected opioids and their enantiomers have NMDAR modulating effects and may be directed to specific diseases and conditions associated with NMDAR dysfunction. Dextromethorphan, the d isomer of the racemate-racemic buprenorphine, is currently the only example of the opioid enantiomer with NMDAR blocking and weak or clinically negligible opioid (opioideergic) activity that is FDA approved for use in combination with quinidine for specific neurological indications, treating the emotional instability of pseudobulbar palsy. The development and approval of dextromethorphan in combination with quinidine, a drug with several disadvantages detailed below, for very specific indications (emotional lability of pseudobulbar palsy) is one example of the clinical usefulness of specific NMDAR modulators for specific indications, emphasizing the unmet need for a drug library with clinically tolerable NMDAR modulation effects and favorable PK and PD characteristics to target specific diseases. Synthetic opioids have been designed and synthesized to optimize targeting of opioid receptors. By modifying the structure of the selected opioid, the inventors now optimize the structure of the opioid for the first time to target NMDAR and NMDAR subtypes. In order to avoid or minimize the effects of opiate drugs mediated by opiate receptors, the development of opiate enantiomers with little or no opiate activity as NMDAR antagonists has previously been the subject of choice (see US patent No. 6,008,258; US patent No. 9,468,611; international patent application No. PCT/US 2018/016159). While opioid effects appear to be stereospecific, NMDAR blocking effects are generally not stereospecific, and therefore less opiate enantiomers can be selected as novel potential NMDAR modulators for specific diseases. Combinations of opioid drugs with opioid antagonists have also been proposed for use with opiate drugs having NMDAR antagonism, such as levorphanol (international patent application No. PCT/US2018/016159), to block the effects of opioid drugs while maintaining the modulatory effects of NMDAR. While the opioid and opioid enantiomer drugs of choice may be directed against diseases caused by NMDAR dysfunction, new modifications of their structure (as proposed herein for SMO) will likely result in new molecular entities that retain certain NMDAR modulating properties while exhibiting favorable PK parameters (e.g., enhanced lipid solubility or favorable metabolic parameters) and PD parameters (e.g., optimization of SAR at the NMDAR), which is favorable for a particular disease, disorder and/or patient population.

As previously disclosed in the present inventors' international patent application No. PCT/US2018/016159, the half-life of dextromethorphan is very short and when used as a single drug may not be effective for many diseases and patient subpopulations. However, the combination of dextromethorphan with quinidine avoids the very short half-life of dextromethorphan alone and has been found to be effective in Pseudobulbar paralysis (Ahmed A. et al, Pseudobulbar effects: epidemic and Management. treatment and Clinical Risk Management (Pseudomonas bulbar infection: prediction and Management. therapeutics and Clinical Risk Management) 2013; 9: 483-489). Thus, the U.S. Food and Drug Administration (FDA) has approved dextromethorphan HBr and quinidine sulfate 20mg/10mg capsules (Avanir Pharmaceuticals, Inc) as the first treatment for pseudobulbar effect (PBA). Unfortunately, quinidine has a potentially fatal risk of arrhythmia and thrombocytopenia, which makes it desirableBecome a poor candidate for further development of other disease treatments. In addition, dextromethorphan has an active metabolite and is affected by the Polymorphism of the CYP2D6 gene, leading to alterations in pharmacokinetics and responses in the human population (Polymorphism of human cytochrome P4502D6 of Zhou SF. and its clinical significance: part 11 (Polymorphism of human cytochrome P4502D6 and its clinical significance: part II), Clin pharmacopoene et al, 48: 761-one 804, 2009). These are drawbacks compared to dextromethorphan, and to its derivatives with NMDAR antagonism, and to other opioids and their derivatives disclosed in international patent application No. PCT/US2018/016159, including SMO disclosed in this application, including structurally modified dextromethorphan molecules: modification of the dextromethorphan molecule, similar to that of SMO outlined below, may alter its PK and PD characteristics and render the new modified drug safe and effective for the selected condition.

As mentioned above, the present inventors have conducted preclinical (in vitro and in vivo) and clinical development work for more than twenty years using racemic methadone and its isomers, in particular dextromethadone. Dextromethorphan [ d-methadone; (+) -methadone); s-methadone ] is one of the two opioid enantiomers of the racemate dl-methadone (methadone). Methadone racemate dl-methadone has been used clinically for over 60 years for the treatment of pain and opioid addiction. Dextromethorphan, a Novel NMDA Antagonist, is currently being developed for a variety of clinical indications with clinically negligible Opioid effects and favorable PK and PD profiles, as discovered by the inventors in Phase 1 clinical Studies (Bernstein G, Davis K, Mills C, Wang L, McDonnell M, Oldenhof J, Inturrisi C, Manfredi PL, vitamin OV. D-Methadone, a Novel N-Methyl-D-Aspartate Receptor Antagonist, its Safety and Pharmacokinetic Profile in Healthy, Opioid-free individuals, Characterization of the Results of Two Phase Studies (chromatography of the Safety and pharmaceutical Profile of D-Methadone, a Novel N-Methyl-D-Aspartate Receptor Antagonist, N-Methyl-D-vitamin Receptor Antagonist, vitamin N-vitamin P-D-P-P.E.W.W.C.A.N-Methyl-D-P.K.K.K.W.No. 12, and PCT-P.W.P.7.W.W.No. 23.W.W.W.W.No. 7.No. 7.7.No. 7.7.W.7.7.No. 7. Information disclosed in/US 2018/016159. While dextromethorphan may prove useful in its own right for treating one or more diseases, one or more structural modifications of the dextromethorphan molecule disclosed in the present application and Structural Modifications (SMO) of other selected opioids may make the PK/PD parameters of the resulting new chemical entity more suitable for a particular disease and condition, and provide PK and PD advantages over dextromethorphan and other available NMDAR antagonists, and may be useful in different patient populations and for selected diseases.

NMDAR dysfunction including glutamate excitotoxicity

Excitotoxicity is cellular damage caused by excess extracellular glutamate (the major excitatory amino acids), resulting in over-activation of NMDAR (and other ionotropic glutamate-activated membrane receptors, such as AMPA receptors and kainic acid receptors). Importantly, if NMDAR is not over-stimulated by glutamate, but is over-stimulated by, for example, endogenous toxins (such as quinolinic acid) or exogenous toxins (aminoglycoside toxicity is an example of NMDAR over-stimulation, which helps to damage selected cell populations, inner ear hair cells, and thus hair cell loss and deafness at physiological glutamate levels; dietary polyamines may also cause NMDAR over-stimulation), NMDAR over-activation may also occur in the presence of normal glutamate levels. Other toxins, including autoantibodies, as in NMDAR encephalitis, can cause NMDAR overstimulation in the presence of physiological glutamate levels, and can cause toxicity in selected neuronal populations and circuits, and cause disease in normal and/or genetically predisposed patients. This is considered when considering that dysfunction of NMDAR is associated with a variety of diseases. Even if the disease is apparently caused by genetic or sporadic genetic mutations, the mechanism of cellular injury may be over-activation of NMDAR, even at physiological glutamate levels. Thus, SMO may represent a potential treatment for a variety of diseases, and selected SMO may be selective for certain genetic and/or environmental causes (e.g., diseases caused by exposure to toxins, including drug therapies, even diseases caused by a deficiency in specific nutrients) or perhaps more commonly diseases caused by genetic (NMDAR propensity to toxicity) + environmental (toxins or lack of specific nutrients) (thus falling within the G + E paradigm described above).

In the mildest form, over-activation of NMDAR by excess glutamate or other causes may be limited to temporary synaptic dysfunction or loss of dendrites or other slight abnormalities (e.g., dendritic pruning defects), which may represent a reversible condition. This concept of reversible NMDAR-related toxicity has important therapeutic and prophylactic implications for conditions such as normal aging of neuronal populations, as well as diseases and conditions such as depression, ADHD, PTSD, anxiety disorders (including SAD, RLS, transient cognitive disorders) and many other diseases and conditions in which neurons are irreversibly damaged or killed, although possibly in severe form, but may not be the driver. Among these diseases, alterations in neuronal function or neuronal circuits account for the major pathogenesis, rather than the diseases in which actual neuronal loss appears to be more prominent (e.g., alzheimer's disease, parkinson's disease, ALS, etc.). As mentioned above, even potential "reversible" NMDAR-related toxicities may trigger apoptotic cascades with irreversible neuronal damage and death, for example in a variety of neurodevelopmental and neurodegenerative diseases, underscoring the potential of NMDAR modulators in the secondary prevention of diseases and disorders.

In the classical excitotoxicity paradigm, sudden release of excess glutamate in acute situations such as trauma and ischemia leads to excitotoxicity of surrounding tissues and amplifies acute necrosis and cell death from major events. Glutamate is the most important excitatory neurotransmitter and is responsible for more than 90% of excitatory communications between neurons. Glutamate is predominantly intracellular, where it is internalized into synaptic vesicles. The brain contains approximately 10mMol of intracellular glutamate and 0.6 micromolar of extracellular glutamate. Physiological extracellular concentrations during excitatory neuronal communication may be as high as 1mM, but such high extracellular concentrations only last for a few milliseconds under physiological conditions. Activation of NMDAR-opening of the channel-is triggered by glutamate under physiological conditions for a duration longer than the duration of the pulsed stimulation of glutamate, from tens to hundreds of milliseconds. In other words, the NMDAR channel remains open for a longer period of time than the extracellular glutamate takes to decay to resting levels. If exposed for a long period of time, the toxic extracellular concentration of extracellular glutamate can be as low as 2-4 μ M, and therefore the resting synaptic physiological concentration (0.6 μ M) approaches that toxic to the cell, while the "reserve" of intracellular glutamate is very high (10 mM). In the case of excessive glutamate release (excitotoxicity), this very high intracellular glutamate content has a high probability of causing damage to peripheral neurons. As described above, NMDAR may also be overactive when brain glutamate is at physiological levels, and therefore NMDAR blockers and modulators may also play a therapeutic role when glutamate is at physiological concentrations, whereas NMDAR is deregulated for reasons other than glutamate overactivity (e.g., selected toxins).

Glutamate-mediated physiological neuronal activity interferes with many sensory, motor and associated physiological neuronal pathways and is therefore critical for normal NS function, including sensory (including special senses such as vision, hearing, smell and taste) and motor activity. Furthermore, by interfering with neuronal plasticity mediated by NMDAR and other ionotropic glutamate receptors (AMPA and kainic acid receptors), glutamate is essential for LTP, LTD, empirically determined synaptic refinement, which is essential for proper development, and thus for memory formation, learning, mood regulation, and ultimately thinking and behavior. Finally, by affecting neuronal activity and survival, for example by modulating BDNF or modulating synaptoprotein, as previously shown by the present inventors, or by other mechanisms, NMDAR may also affect not only the function, nutrition and aging of the nervous system, but also of all organs and systems, since proper neuronal function affects the function, nutrition and aging of all other tissues, organs and systems through innervation. In addition, NMDAR also has the following important functions: additional CNS cells (Glutamate in peripheral organs of Jie Du, Xiao-Hui Li, Yuan-Jian Li.: biological and pharmacological (Glutamate in peripheral organs: Biology and pharmacological), European Journal of pharmacological, 784(2016)42-48) have an effect on the treatment of diseases secondary to NMDAR dysfunction in non-nervous system cells. Deliberate design of new molecules with little or no CNS penetration (in the context of the present disclosure, new SMOs), such as polarized or larger molecules that cannot cross the blood-brain barrier and thus have no potential CNS effects, may be advantageous for the development of new drugs for the treatment of diseases caused by NMDAR dysfunction outside the CNS.

The inventors performed experiments to confirm the presence of NMDAR on retinal epithelial pigment cells. The inventors then demonstrated the sensitivity of these cells to glutamate-induced toxicity and toxicity by inflammatory mediators, and finally the inventors showed that the NMDAR modulator dextromethorphan prevents toxicity in these cells (see example section below). These new data underscore the possibility of SMO for the treatment of diseases originating from dysfunctional NMDARs located outside the CNS.

NMDAR is a positive allosteric system that requires an agonist (glutamate) and a synergistic agonist (glycine or d-serine),Modulating factors (e.g., Zn +, NO, protons (pH), polyamines, and modulating physiological channel blockers (Mg)2+) Contribution of) balanced binding to function properly.

The NMDAR tetrameric complex is assembled from seven different protein subtypes. NR1 is mandatory and necessary for membrane expression; NR2A-D and NR3A-B are other subtypes. The seven proteins are encoded by seven different genes with different expression depending on regional (different brain regions, neuronal populations, different NMDA subtypes of neuronal circuits) and temporal factors (e.g., NR3A expression is relatively high in the young, decreasing in adulthood, while NR3B expression increases progressively with development; NR1 protein fluctuates less between different ages; NR2A and NR2B also exhibit differential expression at different stages of development). In addition, NR2B may be more prominent at additional synaptic sites, and its presence may therefore make cells more susceptible to excitotoxicity (memantine preferentially blocks this receptor subtype, the NR1-NR2B complex, may make this drug particularly suitable for preventing excitotoxicity in certain diseases). Complexes with NR1-NR2A and NR1-NR2B and triisotetrameric NR1-NR2A-NR2B (requiring voltage-gated expulsion of Mg2+NMDAR expressing NR2C and NR2D and NR3A and NR3B may be more resistant to magnesium blockade than activation (opening of the channel pore), mediated by AMPAR), and thus may be active (open channel) even when the neuron is in a hyperpolarized state. The NR3 NMDAR subtype is resistant to magnesium blockade and Ca2+Is less permeable and can intervene during minimal synaptic events and affect the regulation of dendritic proteins and thus synaptic plasticity. Furthermore, only glycine is required to activate NR3 NMDAR, whereas glutamate and NMDA do not act as agonists for these receptors, so, although classified as NMDAR due to structural similarity to other NMDAR subtypes, the MR 3-bearing receptors are functionally different because they lack NMDA activation, on Mg2+Blocked resistance and to Ca2+Is relatively impermeable. Furthermore, MK-801 and memantine are less active at NR3A-B (Chian-Ming Low and Karen Siaw-Link Wee, New instruments) than blocking channels containing NR2 subunitsThe Not-So-New NR3 Subunits of N-Methyl-D-aspartate Receptor Localization, Structure, and function. mol Pharmacol 78: 1-11, 2010). While most SMOs are more likely to act on an NMDAR with NR2 subunits, it is also likely that selected SMOs may instead target an NMDAR with NR3A-B subunits or mixed NR1-NR2/NR3 triisotetrameric subtypes and therefore they may help to define the role of the N3A-B receptor subtype in disease and as a target for SMO therapy.

Because of the widespread existence of NMDAR and its important role in almost all physiological NS activities (as well as many additional nervous system activities), it is not surprising that competitive and synergistic agonists interfere too strongly and/or are too extensive and unpredictable with normal physiological activities, and are clinically intolerable due to side effects. It is not surprising that competitive agonists and synergistic agonists should have unpredictable and modulatory (modulatory) NMDAR effects to treat a particular disease. High affinity NMDA channel blockers are also less suitable as therapeutic agents because they are likely to become trapped in the channel and cause a sustained block, leading to serious side effects. The design of the drug MK-801 (dezocine) may cause coma; the illicit drug phencyclidine (PCP; "angel powder") causes hallucinations. Ketamine is an FDA-approved anesthetic derived from PCP, a low affinity NMDAR non-competitive antagonist, with a faster "off rate" compared to dezocyclopine and PCP, resulting in less "capture"; however, ketamine may remain in clinically tolerable pathways outside of anesthetic or specific psychiatric indications for too long a period of time. In fact, ketamine determines clinical effects like sedation and dissociation, which may be beneficial for anesthetic indications (higher ketamine doses for anesthesia compared to doses for depression). However, these ketamine effects (sedation and dissociation) remain undesirable when the indication is major depression rather than anesthesia; in selected cases, for example, severe drug resistant depression, sedation and separation (disassociation) may be an acceptable side effect, such as intranasal injection of esketamine, with a short lifespan and recent FDA approval for certain depression patients. Once an NMDAR antagonist/modulator binds to a target binding site in a recipient, it is released shortly thereafter-without becoming trapped in the pores of the NMDAR complex, and therefore this release should occur within a reasonable time-the "excursion rate" (excursion) -otherwise it will stay too long within the channel and interfere with physiological activity (learning, memory formation, etc.) and cause sedation and psychomimetic side effects. MK-801, PCP and ketamine were "captured" at decreasing rates: 100% MK-801, 98% PCP, and 86% ketamine (Zanos et al, 2018). The timing of NMDAR antagonist entry into the open channel is also a concern: when the channel is only open for a few milliseconds during normal phasic physiological activity, such as LTP during memory formation and learning, it is preferred that the drug should not enter the open channel, so the "opening rate" (onset), i.e., the time required for the drug to enter the open channel, should be close to 1 second (hundreds of milliseconds) rather than tens of milliseconds of physiological activity. Therefore, NMDAR modulators should have a narrow window for "turn-on rate" -start- [ should not be too fast (side effects) or too slow (ineffective) ] and "drift" -drift- [ should also not be too fast (ineffective, such as magnesium blockade) or too slow (side effects, MK-801 and PCP type blockade) ]. Other factors may also play a role, such as dose and serum drug levels (e.g., different doses of ketamine for anesthesia and depression) as well as clinical indications and disease stages within the same clinical indication (e.g., memantine is effective for moderate and advanced alzheimer's disease, but not mild alzheimer's disease). Higher doses of ketamine for anesthesia and lower doses of ketamine for depression are one example of different clinical uses of the same drug for different indications, even within a narrow therapeutic window. Another less common form of narrow therapeutic window may not refer to dose, but rather to stage of disease: while memantine's NMDA receptor blocking property-its opening rate of 1 second, its excursion rate close to 5 seconds-may lead to the drug being used for moderate to severe alzheimer's disease, its effect was found to be ineffective for mild alzheimer's disease where drugs with slightly different onset and offset effects may instead be effective.

The ideal NMDAR modulator candidates are likely to be different drugs for different diseases, even different drugs for different stages of the same disease, and may vary depending on the particular patient subpopulation and age of the patient. By providing different PK and PD profiles, each new SMO drug may offer advantages for specific diseases and conditions as well as patient populations. More lipophilic drugs may preferentially target more difficult to reach NMDARs, such as those portions of the super complex (see below), and thus in the SMO disclosed herein, trihalo compounds or fluoro derivatives may be particularly beneficial for certain diseases or subpopulations of patients in which the super complex is affected. Starting from the initial phase of new drug development, starting from the design of novel and unique chemical formulas, among other features, drug polarity and molecular size across the Blood Brain Barrier (BBB) and preferential targeting of selected brain regions and specific SAR of NMDAR (which can be seen in the newly designed compound library discussed and described below as "design, molecular modeling, synthesis and testing of structurally modified opioids with NMDAR modulating activity"), the selection of specific indications for specific SMO may become more and more definite.

Design, molecular modeling, synthesis and testing of structurally modified opioids with NMDAR modulating activity

In order to optimize the chemical structure of SMO for their activity on NMDAR, including differential activity on NMDAR subtypes that may be preferentially regulated or blocked against specific diseases and conditions triggered, maintained or worsened by dysfunctional NMDAR, the inventors designed a first group of SMO, new chemical entities whose Structural Activity Relationship (SAR) to NMDAR is optimized based on the known NMDAR affinity of the selected opioid and computer-simulated testing (in-silico stimulating) of the newly designed SMO. In addition to PD parameters related to SAR for NMDAR, SMO is optimized for PK parameters, such as enhanced lipid solubility. New mechanisms of potential attachment sites for the NMDAR block are also considered in the design of SMO, for example, for nitro derivatives of opioids and opioid enantiomers, especially nitro derivatives of dextromethorphan: as detailed in the application, SMO nitro derivatives may have additional NMDA modulatory effects by linking to the amino-terminal domain of the NMDA receptor. The inventors then continued to test the affinity of these compounds for the putative PCP binding site of NMDAR in a new computational model, a specially developed NMDAR transmembrane domain (in silico static + dynamic modeling). In addition to direct selection of SMO by determining assumed ligand/receptor affinity, in computer simulated ranking (static and dynamic) helps to determine the priority of synthesis of SMO designated for further testing, also generates information about SAR, thus also helping to design other SMO optimized for NMDAR channel blocking activity.

Different lipophilicity gradients will determine different binding to CNS NMDARs which may be more or less accessible, e.g., NMDARs that are part of a super complex may be less accessible to lipophilic molecules. Notably, dextromethorphan increased PD95 levels in a rat model of depression (as detailed below, and in fig. 4B); PD95 protein is critical for the formation of NMDAR-containing supercomplexes. The inventors have also designed compounds that do not cross the BBB for targeting peripheral NMDAR while purposefully retaining NMDAR located in the CNS; these compounds will be tested for clinical indications where dysfunctional NMDAR is located primarily outside the CNS. In addition to the lipophilicity gradient, the molecular weight, synthetic feasibility and putative stability of SMO were all considered in the design of these new molecules (SMO) and in the development of new clinically safe SMO optimized for selective NMDAR activity.

As described above, throughout the application, a subset of SMOs are designed to incorporate a nitrate group, thereby generating nitro SMOs, to potentially modulate NMDA receptor activity through S-nitrosylation of a thiol group on the N-terminus of a cysteine residue (or the extracellular amino domain of an NMDAR). As mentioned above, this may lead to different PD effects at the level of NMDAR open channels in the extracellular domain, not just in the transmembrane domain of NMDAR, which may be the case for dextromethorphan and other non-nitro SMOs. Nitrosylation can also provide protection against the Reactive Nitrogen Species (RNS) described in the above-mentioned application, thereby providing an additional means of cytoprotection, in addition to the effect on the open channel of the NMDAR, thereby providing additional therapeutic properties.

Once the synthetic work for SMO selected according to in silico results was completed, the results of electrophysiological studies from different groups of SMO, in addition to informing each compound of its NMDAR blocking activity, including different affinities for NMDAR subtypes, would also provide additional insight into the SAR of additional new agents in order to design subsequent generations of NMDAR blockers by further modifying the fragments of SMO molecules most relevant for NMDAR blocking.

Fluoro-, nitro-, and fluoro-nitro-derivatives as well as deuterated fluoro-, deuterated nitro-, and deuterated fluoro-nitro-derivatives are of particular interest for optimization of SAR with NMDAR because of their potential to improve PK parameters, especially for fluoro derivatives, and due to additional NMDAR regulatory mechanisms and to prevent RNS cell damage, especially for nitro derivatives. The inventors have shown that selected deuterated dextromethorphan molecules can preferentially act on selected NMDAR receptor subtypes (GLUN1-GLUN2B tetramer) and that dextromethorphan increases PD95, PD95 being the basic component of a super-complex at post-synaptic density containing NMDAR. The inventors also showed that dextromethorphan increases GluR1 in vivo (fig. 4B) and increases mRNA of NMDAR1 in vitro (fig. 10). Thus, the expression of both receptors AMPAR and NMDAR may be modulated by dextromethorphan. In addition to fluoro-derivatives, nitro-derivatives and fluoro-nitro-derivatives, molecules obtained by deuteration in combination with fluoro and nitro-derivatives are of particular interest for the present disclosure.

In addition to extensive research on dextromethorphan, the present disclosure also allows for characterization of the NMDAR affinity of these compounds in a newly developed ad hoc molecular model of the NMDAR transmembrane domain, and by design of structural modifications of these opioids, followed by further in silico testing, and then continuing with the development program for dextromethorphan described above (further in silico testing, improved SAR definition, molecular synthesis, electrophysiological testing, in vitro and in vivo testing, and again improved SAR definition and design of additional new molecules).

In general, the following formulas (formulas I-VII below) are examples of newly designed compounds relevant to the present invention. These compounds are as follows:

a compound of formula I structurally analogous to dextromethorphan:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate; and n is 1 to 4.

A compound of formula II structurally similar to levopropoxyphene:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate; and n is 1 to 4.

A compound of formula III structurally analogous to dextro-isomethadone:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally deuterated at one or more positionsHalogen, alkyl, hydroxyl, alkoxy, aryl, aryloxy, heterocyclic, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate; and n is 1 to 4.

A compound of formula IV structurally similar to levomoramide:

wherein NR is1R2Optionally through C3-C12Cycloalkyl or heterocyclyl ring closure, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

if NR is present1R2Not cyclized, then R1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of themOptionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; r2Is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is hydrogen; or selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

NR4R5optionally through C3-C12Cycloalkyl or heterocyclyl ring closure, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

if NR is present1R2Not cyclized, then R1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; r2Is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl radicals, of whichAny one optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; and

n is 1 to 4.

A compound of formula V having a structure analogous to N-methyl-dextromethorphan:

wherein R is1Selected from alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR1selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

AR2selected from the group consisting of aryl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium or is selected from the group consisting of alkyl, aryl, C3-C12Cycloalkyl or heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R4is alkyl optionally substituted at one or more positions with deuterium, halogen, hydroxy, alkoxy, nitrate;

R5is optionally substituted in one or more positions by deuterium, halogen, hydroxy, alkaneOxy, nitrate substituted alkyl;

X-is a nitrogen-counterion; and

n is 1 to 4.

A compound of formula VI structurally similar to levorphanol:

wherein R is1Is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; and

R4is hydrogen or selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate.

A compound of formula VII structurally analogous to dextromethorphan or dextrorphan:

wherein R is1Is hydrogen, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R2is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R3is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate;

R4is hydrogen, deuterium, halogen, hydroxy, nitro, nitrate or is selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate; and

R5is hydrogen or selected from the group consisting of alkyl, alkoxy, aryl, aryloxy, heterocyclyl, any of which is optionally substituted at one or more positions with deuterium, halogen, alkyl, hydroxy, alkoxy, aryl, aryloxy, heterocyclyl, nitro, nitrate.

In addition, for all SMOs, including those listed above with formulas I-VII and for all listed substitutions, nitrate substitutions are relevant, particularly when associated with deuterated substitutions and/or halogen substitutions.

TABLE 1A List of the first set of designed SMOs tested in bioinformatic molecular models blocking the transmembrane domain of the NR1-NR2B subtype of NMDAR

TABLE 1b List of a second set of designed SMOs tested in a computer-simulated molecular model blocked the NR1-NR2B subtype transmembrane domain of NMDAR

After designing these molecules, the next step is to test these designed molecules in silico to select the best candidates (detailed information and test results of a particularly developed novel molecular model are discussed below in the section labeled "molecular modeling study of selective SMO binding to the transmembrane site of NMDA receptor GluN1-GluN2B tetrameric subtype in its blocked state"), and synthesis of the selected molecules, followed by more advanced and specific in vitro and in vivo testing of NMDAR activity, including electrophysiological testing of NMDAR to characterize relative affinities (methods of which are described in international patent application No. PCT/US2018/016159) and testing of blocking mechanisms (e.g., drugs with non-competitive blocking may be safer, more effective because they selectively act at sites of NMDAR dysfunction rather than at sites with physiological activity). The inventors have begun to validate excitotoxicity protection in vitro and are evaluating the safety and activity of selected SMO in vitro experimental models. Finally, after entering the clinical development phase, the inventors will confirm tolerance and efficacy in human trials, first in healthy volunteers, then in patients with specific diseases and conditions, as the inventors did in the right methadone program, currently in phase 2 clinical development for treatment resistant depression and RLS.

Selective binding to the transmembrane site of the NMDA receptor GluN1-GluN2B tetrameric subtype in its occluded state Molecular modeling study of SMO

Until recently, the structure of the transmembrane domain of NMDAR has not been characterized at the atomic level due to technical limitations in the expression and purification of NMDAR transmembrane proteins. In 2014, Gouaux and colleagues analyzed the structure of the Xenopus GluN1-GluN2B NMDA receptor by X-ray crystallography (Lee, Lu et al, 2014). This structure was obtained in the presence of the partial agonist Ro25-6981 and the ion channel blocker MK-801, representing the off state of the NMDAR. In view of the high similarity of this structure to human sequences, the inventors used the structure identified by Protein Database (PDB) code 4TLM as the starting point for their computational studies. The inventors investigated the following drugs shown in table 2 c: (a) putative NMDAR antagonist: levo-molamine, d-isomethadone, levopropoxyphene, N-methyl d-methadone; (b) dextromethorphan, a mature NMDAR antagonist, is currently in clinical development for a variety of indications; (c) all known NMDA open channel blockers with positive controls (ketamine, memantine, dextromethorphan, amantadine, MK-801, PCP) with known affinity and known clinical effect act on the PCP site of the transmembrane domain; the first four drugs have been used clinically, while PCP is a class I drug, MK-801 is a high affinity antagonist with severe side effects that prevent its clinical use; (d) morphine, a drug with negligible NMDAR activity, was also tested as a negative control. As shown in table 2, docking and dynamic scores were similar to the range of scores for established NMDAR channel blockers. The inventors observed that morphine, naloxone and naltrexone (all of which have negligible affinity for NMDAR) have a hydrogen donor group, whereas dextromethorphan and methadone (which have known clinically relevant affinity for NMDAR in the low micromolar range) do not have this hydrogen donor group. This raw observation, together with the new results of the bioinformatic tests introduced in appendix 2 and the planned patch clamp studies, greatly aided the design of new SMO with improved SAR for NMDAR.

In addition to the information shown in table 2c, table 2a also shows the docking results for the SMO of the first batch of new designs. Table 2b shows the docking results for a second series of newly designed SMOs.

Although the currently clinically used NMDAR antagonists acting on the transmembrane domain of the receptor are thought to exert their effect by binding to open NMDAR, for the purposes of this computational model, the inventors investigated binding to the closed conformation of the channel: clinically effective NMDAR antagonist drugs also bind to the PCP site in the closed state (Zanos et al, 2018) and their "capture" index in the closed state, reflecting the relationship between "open" and "offset" action times, can be used as an indicator of clinical tolerance and effectiveness (Zanos et al, 2018; (hue-Sheng vision chemi and study a. lipon. the chemical biology of clinical tolerized NMDAreceptor antagonists. journal of Neurochemistry,2006,97, 1611-1626)). Thus, an effective NMDAR modulator should bind to open channels, but also briefly (a few milliseconds) to closed channels ("threshold effect" concept), while avoiding long-term "capture". In docking calculations, the ligand is built into the host binding site, so the closed conformation makes it easier to assess the ligand/site interaction: the docking calculation does not take into account the trajectory of the ligand to the binding site.

The calculated NMDAR subtype established for this purpose in silico tests was a GLUN1-GLUN2B tetramer consisting of 2 subunits of GLUN1 and 2 subunits of GLUN 2B. The N2B subunit is necessary for the formation of a super complex comprising an NMDAR. As described in detail in this application, the inventors found that dextromethorphan increased the levels of PD95, GluR1 in vivo (fig. 4B), while in vitro, dextromethorphan increased the mRNA of NMDAR1 (fig. 10), providing more insight into the PD and neuroplasticity potential of dextromethorphan. The carboxy-terminal intracytoplasmic tail of GluN2B is essential for NMDA supercomplex assembly and the PD95-PD93 subunit, as described herein above.

To increase the computational efficiency calculated by the inventors, only the transmembrane region of the receptor, i.e. the region where the PCP binding site is located, and the region where FDA approved and clinically tolerated NMDA antagonists act (dextromethorphan, ketamine/esketamine, amantadine, memantine), and the inventors postulate that putative NMDA antagonist opioid enantiomers and their SMO are included in the simulation model as the region where the subject of the present disclosure acts. The goal of this computational part of the inventors' work was to optimize the structure of selected opioids by modifying their structurally critical selection part to bind to the transmembrane domain of the NMDAR to achieve pore channel blockage when needed to modulate pore channels to prevent or treat a particular disease. Each SMO, in addition to having a unique on/off/capture and unique effect on the NMDAR subtype and variants described in the application, would likely be beneficial for the selected disease and would also have a unique PK profile, which would also likely provide benefits for the selected disease.

Receptor preparation

First, the receptor is byAvailable in the kit, "protein preparation guide" program prepared, the kit is from New York StateNY (https:// www.schrodinger.com /) was used for molecular modeling.

This process automatically assigns the correct protonation state, completes the missing side chain or small region, and assigns the correct name to the atom. The receptor model was then impregnated in a membrane model formed of 1-palmitoyl-2-oleoyl-sn-triorganyl-3-phosphorylcholine (POPC) molecules, taking into account the data available in the membrane database (OPM) in protein orientation (Lomize et al, 2012).

Docking of known drugs

Use of Glide software (available from NY, N.Y.)Obtaining (a)https:// www.schrodinger.com/glide), the first attempt was made to dock molecules to study the conformational interior of the receptor directly from X-ray studies. During docking, the protein regions where the drug is located can be defined manually.

In this case, the region is defined in view of the residues identified by Dougherty and co-workers that are critical for memantine binding due to the lack of co-crystallizing ligands (Limapichat et al, 2013).

The inventors reduced the van der waals (VdW) radius of the molecule to allow the molecule to more "flexibly" adapt to the receptor.

Docking of levopropoxyphene was computationally successful and resulted in a model of the drug receptor complex, while demonstrating the potential NMDAR blocking activity of levopropoxyphene.

Then, to allow the creation of sites more suitable for drug binding, the levopropoxyphene receptor complex immersed in the membrane was simulated for 250ns by Molecular Dynamics (MD) simulation.

A new docking calculation is then performed using the final conformation of the receptor in the same setting as originally applied.

The calculations were successful and the structure of the drug receptor complex was obtained for the tested drug.

Molecular Dynamics (MD) simulation of receptor drug complexes

The system consisting of the drug, receptor and membrane was then simulated by MD for 1 microsecond. The inventors generated a trace of the complex: l-morpholine amide, d-isomethadone, levopropoxyphene, dextromethadone, memantine, ketamine, amantadine, dextromethaphen, PCP, MK-801, morphine. The simulation results using N-methyl d-methadone are unstable, indicating that the molecule may be difficult to bind without structural modification.

Virtual prescreening

The resulting receptor models for docking L-morpholine amide, d-isomethadone, levopropoxyphene, dextromethadone, memantine, ketamine, amantadine, dextromethaphen, PCP, MK-801, morphine were used to pre-screen a first set of newly designed SMO, followed by a second set of SMO. For this purpose, the 2D chemical structure of the molecule is transformed in a 3D model, in which all possible protonation states are calculated. All ligands dock inside the receptor and their affinity is scored by glidisescore, which is based onOne specificity scoring function for drug-protein interactions (https:// www.schrodinqer.com/Glide) for Glide software from New York, NY is shown in Table 2a (first set of SMOs), Table 2b (second set of SMOs) and Table 2c (initial set of selection molecules).

TABLE 2a

Name:

TABLE 2b

TABLE 2c

Drug/receptor complexes that scored the highest 10% of the first set of SMOs were then simulated for 10ns by MD simulation. Finally, their binding energy to the receptor was estimated by MM-GBSA and reported in table 3.

A valid binding event is always characterized by a negative difference in free energy (Delta G) between bound and unbound states (i.e. the free energy of the complex is lower than that calculated for the separated ligand and target).

In the inventors' calculations, several molecules, including dextromethorphan, were predicted to have negative Delta G values. In particular, the ligand/receptor binding affinities of many new compounds tested-represented by Delta G values-are similar or more negative than those obtained for ketamine, a drug known for its activity in NMDAR, an FDA approved clinical indication for anesthesia. The more negative Delta G values obtained for different compounds tested on the developed protein model relative to ketamine and other reference molecules indicate potential differences in drug receptor interactions, different on/off and capture values, and more potent binding affinities that result in different clinical effects that may ultimately be more appropriate for one or more diseases. As SMO experiments progressed, the inventors were likely able to characterize each new molecule with unique PD and PK parameters, which may prove beneficial for selected diseases and conditions.

TABLE 3 results of computational evaluation of the first set of ligands expressed as Delta G values for more active molecules in the pre-screen.

As described above, in order to select new compounds that are synthesized and tested in an in vitro excitotoxicity protection model, the present inventors developed a new in silico NMDAR model and conducted preliminary validation studies with MK-801 (control), dextromethorphan, and newly synthesized compounds, including those described and shown in tables 1a, 1b, 2a, 2b, and 2c above. In silico SMO, newly designed and tested, prior to in vivo experimental trials, is now undergoing in vitro synthesis and further testing.

In addition to NMDAR modulation, other contributions to the mechanism of action may also be useful, as the present inventors discuss dextromethorphan below. As mentioned above, the uptake, distribution, metabolism, excretion (PK) and specific PD characteristics of different effects of the NMDAR complex (including affinity to binding sites, "turn on" - "shift" rates and capture indices, -receptor pharmacokinetics-and binding mechanisms-noncompetitive and/or noncompetitive), as well as other mechanisms of action of drugs, in addition to NMDAR blockade, may vary even with minimal modification of the molecular structure of very similar drugs, and thus these minor modifications may provide particular advantages in the same disease (including different stages of the disease) or in different diseases and conditions (including cellular aging). While this concept may be applicable to most drugs, it is particularly relevant to NMDAR antagonists and modulators, because of the complexities outlined in the present disclosure, including those of drugs with multiple effects, such as dextromethorphan (described below) and other drugs in the same family (opioids and opioid enantiomers, including SMO), and including a variety of diseases and disorders involving these ubiquitous complex ion channel dysfunctions, as well as the effects on cellular processes by dextromethorphan and other mechanisms that may be modulated by SMO, such as upregulation of BDNF, as previously disclosed by the present inventors. To emphasize the importance of mechanisms other than NMDAR modulation, it was recently discovered that ketamine (a known NMDAR antagonist demonstrated antidepressant activity in experimental models and human studies) was not effective in depression when administered with opioid antagonists, which means that ketamine may also modulate opioid receptors other than NMDAR and dopamine systems, and that the opioid system needs to function in order for ketamine to exert its antidepressant effect.

It appears to date that an ideal NMDAR modulator for the treatment of one or more NMDAR-dysregulated diseases should provide effective but transient blockade of open channels, more prominent (or unique) when receptors are over-stimulated, and thus when the channels remain open for more than a few tens of milliseconds-in order to maintain receptor function under physiological conditions, and therefore should have low affinity, relatively slow "on-rates" (and thus not interfere with normal activities) and relatively fast "off-rates" to avoid trapping (so as not to cause side effects, as is the case with MK-801). An ideal NMDAR modulator should act by non-competitive antagonism (increased channel blockade in the case of increased receptor overstimulation-in other words, a constant level of drug will be more active with increased levels of NMDAR overstimulation while minimally interfering with NMDAR's physiological neurotransmission, e.g. during LTP, learning and memory formation). For certain diseases, preferential activity of a particular receptor subtype, as well as the cellular location of the receptor (synaptic, perisynaptic or extrasynaptic) or the location of the receptor along the neurotransmission pathway (presynaptic or postsynaptic) may also be useful. For other diseases, a particular drug may be particularly active for a particular subset of neurons or neuronal circuits, where the MNDAR may be overactive due to the disease, thereby making the drug more disease selective. While dextromethorphan appears to have the basic characteristics of an ideal NMDAR antagonist, including favorable PK and safety and tolerability, different diseases and different stages of the same disease may benefit from specific structural changes of the dextromethorphan molecule or of other opioids and their enantiomers, which create new chemical entities that act similarly to but not exactly the same as that of dextromethorphan: these structural changes may provide better safety and efficacy for specific diseases and patient subpopulations.

Therefore, it may be useful to develop a dextromethorphan like drug (SMO) platform with NMDAR modulating effect that may prove itself to be a better therapeutic agent than the currently approved NMDAR antagonists in certain specific cases related to specific diseases and patient variables as described above. It must be understood that while these new molecules may share some structural features with dextromethorphan or other opioids and their enantiomers, they are actually new molecular entities, with different PK and PD properties, that need to be characterized by a comprehensive drug development program, separate from the dextromethorphan development program, starting with the design of new chemical formulas, tested in new molecular model analyses, the development of new synthetic methods and molecular syntheses and a series of new experimental work in vitro, followed by experimental work in vivo and then a complete clinical phase, starting with a first phase of safety studies.

As mentioned above, NMDAR antagonists are becoming increasingly recognized for their great potential as therapeutic agents for a variety of diseases. Amantadine is well known to be effective in parkinson's disease; memantine is similar in structure to amantadine, and is effective against moderate to severe alzheimer's disease; dextromethorphan can effectively control emotional lability of pseudobulbar paralysis; ketamine is effective as an anesthetic and has been FDA approved for the treatment of patients with resistant depression in experimental models of depression.

Ketamine and memantine have been shown to affect synaptoprotein synthesis and increase the number of synapses in certain neuronal populations in the experimental setting, and therefore these drugs may play a role in neuronal plasticity. These effects may be beneficial for certain diseases: for example, Ketamine has recently been shown to be effective in experimental models of Rett Syndrome, which is a serious developmental disorder (Patrizi A, Picard N, Simon AJ, Gunner G, Centofante E, Andrews NA, Fagiolini M.Chronic addition of the N-Methyl-D-aspartic Receptor or oligonucleotide ketone precursors improvements synthesized Kidney synergistic photon catalysis, biol Psychiatry.2016May 1; 79(9):755-64.doi:10.1016/j. biopsych.2015.08.018.Epub 2015Aug 24).

Dextromethorphan was also found to increase the levels of GluR1 and PSD95 in a rat model of depression (data in this regard are shown in the section below entitled "development of NMDAR antagonist d-methadone for treatment of depression and other CNS disorders"), and these effects may be shared by selected SMO subjects of the present disclosure at different activity levels.

Development of NMDAR antagonist d-methadone for treatment of depression and other CNS disorders

In this regard, NMDA receptor (NMDAR) modulators are potential agents for the treatment of several Central Nervous System (CNS) disorders, including major depression, as previously disclosed by the present inventors in U.S. patent No. 9,468,611, international patent application No. PCT/US 2018/016159. Furthermore, racemic methadone and its stereoisomers, L-methadone and d-methadone, bind to NMDAR with similar affinity as established NMDAR antagonists, whereas only L-methadone and racemic methadone bind to opioid receptors with high affinity. The inventors found that D-methadone had no clinically significant opioid effect at therapeutic doses mediated by its NMDAR antagonism (SAD and MAD studies are detailed in the section below entitled clinical studies).

In view of this background, the present inventors have conducted several preclinical studies comparing the effects of d-methadone and ketamine in different behavioral animal models commonly used to assess antidepressant activity. These include forced swim tests, female urine sniffing tests, and novelty inhibition feeding tests. The inventors also performed behavioral analyses of the effect of d-methadone and ketamine on rats exposed to Chronic Unpredictable Stress (CUS) regimens.

In all of the above tests, d-methadone, such as ketamine, produced a significant improvement in drug treated animals compared to vehicle treated animals. Furthermore, the inventors observed a positive effect on the expression of synaptoproteins and receptors that are critically involved in synaptic plasticity. These biochemical effects are also accompanied by favorable changes in electrophysiology.

Figure 1 shows the effect of ketamine and d-methadone on immobility, climbing and swimming counts in FST. Data represent mean ± SEM. For immobility: compared to the carrier group ANOVA, p is 0.0034 for ketamine, 0.0007 for d-methadone, and <0.0001 for d-methadone 20 and 40 mg/kg. For climbing: p <0.05 for d-methadone 40mg/kg compared to the carrier. For swimming: compared to the carrier ANOVA, p <0.05 for ketamine and d-methadone, p <0.0001 for d-methadone 20mg/kg, and p <0.0003 for d-methadone 40 mg/kg.

Figures 2A-2E show the effect of d-methadone and ketamine on the female urine sniff test ("futt") and the novel inhibition feeding test ("NSFT"). In fig. 2A, a time table of dosing and testing rats is shown, wherein d-methadone or ketamine is administered, followed by various tests. Fig. 2B shows rats tested in futt 24 hours later. Figure 2C shows rats tested for autonomous activity (LMA) after 2 days. Figure 2D shows rats subjected to NSFT testing 72 hours later. FIG. 2E shows home cage rearing. Results are mean ± s.e.m. FUST: one-way ANOVA, F3,42 ═ 3.26, p ═ 0.031; snowy LSD: veh × Met, p ═ 0.025; veh × Ket, p ═ 0.046; n is 9-12/group. NSFT: one-way ANOVA, F3,27 ═ 4.87, p ═ 0.008; snowy LSD: veh × Met, p ═ 0.035; veh × Ket, p ═ 0.005; n is 7-8/group.

Figures 3A-3E show that a single dose of D-methadone prevents depressive behavior caused by chronic unpredictable stress ("CUS") exposure. Fig. 3A shows the time course of the CUS protocol, drug dosage and behavioral analysis. D-methadone and ketamine prevent the behavioral effects of CUS in fig. 3B [ Sucrose Preference Test (SPT) (F3,45 ═ 2.99) ], fig. 3C [ FUST (F3,46 ═ 5.43) ], and fig. 3D [ NSFT (F3,46 ═ 6.79) ]. No difference was found in water sniffing or (fig. 3E) home cage food consumption. Results are mean ± s.e.m., n-9-15/group. P <0.05, one-way ANOVA and duncan post hoc test.

Fig. 4A and 4B show the effect of d-methadone on mTORCI signaling and synaptophin. Rats were given d-methadone and the levels of mTORCI signaling protein and synaptophin were measured in PFC and hippocampus.

Levels of phosphorylated protein were normalized to total protein and levels of synapsin were normalized to GAPDH. Results are mean ± s.e.m., n-10-12/group. P <0.05 compared to vector (student's t-test).

Clinical research

The present inventors also investigated the safety, tolerability and Pharmacokinetic (PK) profile of d-methadone in healthy, naive volunteers taking opioids in two phase 1, double-blind, randomized, placebo-controlled, single and multiple ascending dose (SAD and MAD) studies, as detailed in international patent application No. PCT/US 2018/016159.

Single increment dose (SAD) studies included parallel group, double-blind, placebo-controlled designs. The purpose of this study was to determine PK, PD and safety for single dose administration. Administration involves groups of 5, 20, 60, 100, 150, 200mg and N-42. The SAD study concluded the following: (1) maximum Tolerated Dose (MTD) 150mg (single dose); (2) PK shows CmaxAnd AUC0_,1ifLinear proportion to dose; (3) dextromethorphan of no more than 150mg has no clinically significant opioid effects.

The objective of Multiple Ascending Dose (MAD) studies was to determine PK, PD and safety for once daily, 10 day dosing. Dosing involved groups of 25, 50, 75mg and N-24. The MAD study concluded the following: (1) dose tolerance of up to 75mg per day is good, and (2) single dose parameter C on day 1 is demonstratedmaxAnd AUCtauAnd steady state parameter C at day 10max、AUCtauAnd the dose ratio of Css.

Thus, for most single dose and multiple dose parameters, d-methadone exhibits a dose-proportional linear PK. Single doses up to 150mg and daily doses up to 75mg, lasting 10 days, are well tolerated, mostly mild cases of adverse events, with no serious or critical adverse events. At the tested doses, d-methadone did not cause dissociative or psychomimetic adverse events, had no clinically relevant opioid effects, and had no signs or symptoms of withdrawal upon sudden withdrawal.

Brain-derived neurotrophic factor (BDNF) plasma levels in the 25mg cohort in the MAD study were measured before any treatment and 4 hours after 25mg d-methadone (6 patients) or placebo (2 patients) on days 2, 6 and 10. Referring to fig. 5A and 5B, in the d-methadone treatment group, 6 of 6 individuals had increased BDNF levels after d-methadone treatment compared to pre-treatment levels, with BDNF plasma levels ranging from two-fold to 17-fold from pre-treatment BDNF levels at day 10 post-treatment. In contrast, BDNF plasma levels remained unchanged in both placebo subjects. When placebo subjects were included in the analysis, plasma BDNF levels measured on days 2 and 10 significantly correlated with plasma levels of d-methadone. These data are also described in more detail in International patent application No. PCT/US 2018/016159.

In summary, evidence collected to date continues to support the development of d-methadone in depression and other CNS disorders for which NMDAR modulation may be an effective mechanism of action for potential treatment.

In addition to the above information on "development of NMDAR antagonist d-methadone for the treatment of depression and other CNS disorders", it was found that dextromethadone instead of MK-801 can increase the expression of mRNA encoding NMDAR1 (see example below and fig. 10). While a neuroplasticity event may be a downstream effect of NMDAR antagonist activity shared by both drugs (ketamine and d-methadone), the degree, outcome and location of the neuroplasticity induced by the different NMDAR antagonists derived from human brain opioid drugs (SMO) is likely to vary between the different chemical entities in the group, and thus there may be only one specific NMDAR antagonist/modulator (for the purposes of this disclosure, only one specific new chemical entity in many SMO's disclosed) that may prove to be effective and fully developed as an effective drug for one or more selected diseases and conditions.

A recent paper (Frank RAW1, Zhu F2, Komiyama NH2, Grant SGN2.Hierarchical organization and genetic separated sub-compounds of PSD95 postsynthetic super compounds. JNeeurochem.2017 Aug; 142(4): 504-511. doi:10.1111/jnc.14056.Epub 2017Jul 25) describes supramolecular organization of NMDAR in the synaptic proteome, with postsynaptic density consisting of about 1000 proteins. While the NMDAR complex is a 0.8MDa heterotetramer formed only from its ion channel subunits, the NMDAR super complex is a component of 1.5MDa, including the NMDAR receptor bound to approximately 50 different proteins, including other ion channels, receptors, adhesion proteins, signaling proteins, scaffold proteins. Interestingly, while NMDAR complexes can be either di-hetero-tetramers (GluN1-GluN2A and GluN1-GluN2B) or tri-hetero-tetramers (GluN1-GluN2A-GluN2B), NMDAR super-complexes comprise only GluN2B di-hetero-tetramers and tri-hetero-tetramers (the carboxy-terminal intracytoplasmic tail of GluN2B together with the PD95-PD93 subunit is critical for NMDA super-complex assembly). A drug that is more selective for NMDAR diisotetrameric GluN1-GluN2A will be less likely to affect NMDA super-complexes and will have different PD effects than a drug that is more active for NMDAR assembled with the GluN2B subunit, such as memantine compared to amantadine and deuterated dextromethorphan compared to dextromethorphan (international patent application No. PCT/US 2018/016159).

Furthermore, the data in the above "development of NMDAR antagonist d-methadone for the treatment of depression and other CNS disorders" indicate that dextromethadone increases PSD95 in the rat model of depression in addition to GluR 1. These findings indicate that the effects of dextromethorphan and possibly other SMOs are not limited to NMDAR complexes but may also affect AMPA receptors and involve NMDAR super complexes. PSD95 is a participant with PSD93 and GluN2B in a three-way rule that explains the organization of a subset of NMDAR in a super-complex (Frank RAW and Grant SGN, 2017), and thus the increase in PD95 observed in dextromethorphan-treated rats and in vitro data on the increase in mRNA expression encoding NMDAR1 induced by dextromethorphan (see examples below and fig. 10), provides additional insight into the biochemical outcome and the neuroplasticity potential of drugs such as dextromethorphan and the neuroplasticity potential of new chemical entities in the group of SMO subjects of the present disclosure, and ultimately better defines the potential of this novel pool of NMDAR antagonists (SMO) for developing drugs that may be effective in treating the diseases and disorders defined in the present application.

Dextromethorphan is currently undergoing preclinical and clinical trials for several indications, including depression, Rett syndrome, restless leg syndrome, amyotrophic lateral sclerosis, ocular disease, and other potential indications. While dextromethorphan may prove effective for one or more of these indications, it may be effective for only one indication. Structural modification of the dextromethorphan molecule or of another opioid with NMDAR activity (SMO) or opioid enantiomer is one option that may provide additional and different effects at NMDAR or even other sites (see below, "other therapeutic targets for SMO"), with potential therapeutic efficacy for different diseases and patient populations, as compared to dextromethorphan.

Other therapeutic targets for SMO

In addition to their activity at NMDA receptors and their downstream consequences, which the inventors have disclosed in the listed patents and patent applications, including the upregulation of synaptophysins, including AMPA receptors, NMDAR1, PD95, as based on the data provided herein, dextromethorphan also exerts other effects, possibly therapeutic on selected diseases: dextromethorphan inhibits the noradrenaline transporter ("NET") system and the serotonin transporter ("SERT") system (Codd EE, Shank RP, Schuppky JJ, Raffa RB. Serotonin and norreppinephrine uptake activity of center acting and analytical biology. J. Pharmacol Exp ther.1995 Sep; 274(3): 1263-70.); dextromethorphan affects and upregulates psychotrophic factors, such as brain-derived neurotrophic factor ("BDNF"), and regulates reproductive hormones, such as testosterone (international patent application No. PCT/US 2018/016159); it is to K+、Ca2+And Na+Cellular currents play a role (Horrigan FT 1, Gilly WF. Methhadone block of K+current in liquid giant fiber loop neurones.J. Gen physiol.1996 Feb; 107(2) 243-60); it down-regulates blood pressure and potentially blood glucose in humans (International patent application No. PCT/US)2018/016159). Thus, dextromethorphan has a potential role in the treatment of one or more of NS, endocrine, metabolic and nutritional, and aging processes. Finally, dextromethorphan exerts mild Opioid activity in different Opioid Receptor subtypes (Codd et al, 1995), while these effects, while mild and insignificant in terms of Opioid side effects (Bernstein G, Davis K, Mills C, Wang L, McDonnell M, Oldenhout J, Inturrisi C, Manfredi PL, vitamin OV. Characterisation of the Safety and pharmaceutical Profile of D-Methyl, aNovel N-Methyl-D-Asparatate Receptor Antagonist in Healthy, Opoid-Naive Subjects: Results of Two Phase 1Studies. J Clin psychopharmacology May/Jun; 39: 3) although they may, if given with certain Opioid antagonists, provide significant advantages for certain Opioid diseases such as depression disorders, or in the case of painful conditions (even weak opioid effects may represent a therapeutic advantage), its antidepressant action may not be exerted.

SMO may retain some of the dextromethorphan effects listed above but not others, or may have entirely different effects, which will be fully revealed as part of a drug development program, and these effects may be selectively beneficial in treating one or more diseases.

In fact, there may be other drugs that have been used clinically for a variety of clinical indications that can exert therapeutic effects by modulating the NMDAR complex, but this mechanism of action of these drugs has not been recognized and their clinical effects may be attributed to other mechanisms. Amantadine is an NMDAR antagonist and may exert its therapeutic effect by different mechanisms. For example, the mechanism of action of amantadine in parkinson's disease has been thought to be dopaminergic or anticholinergic for many years; at present, NMDAR antagonism is considered to be an important mechanism of the antiparkinson action of amantadine. Although the NMDAR effect of amantadine may be critical to its effectiveness in parkinson's disease, its "other" effects, dopaminergic or anticholinergic, cannot be completely overlooked, and more than one mechanism of action may contribute to the therapeutic benefit of amantadine for parkinson's syndrome.

Variables as therapeutic targets affecting NMDAR

The following is a list of potentially advantageous variables that may be preferentially targeted by one SMO over another, resulting in a beneficial therapeutic profile for a particular new drug over another for the treatment of one or more diseases and disorders. The following list of variables underscores the unmet clinical need addressed by the present application, namely the pool of potentially safe and effective NMDAR modulators derived from opioids and opioid enantiomers (SMO disclosed in the present application):

(a) pre-and post-synaptic blockade of NMDAR: very different effects will occur with one or the other of the more selective drugs (Banerjee A, Larsen RS, Philpot BD, Paulsen O.Roles of Presynaptic NMDA Receptors in neurosensrisation and Plastic. trends in Neurosciences 2015, Volume 39, Issue 1).

(b) Synaptic and extrasynaptic blockade: preferential extrasynaptic blockade may be preferred over synaptic blockade because it may better prevent excitotoxicity with less interference with physiological neuronal activity occurring at or near the synapse. For example, memantine may preferably target extra-synaptic NMDAR (Huei-Shenng Vincent Chen and Stuart A. Lipton. the chemical biology of clinical tolerized NMDA regenerative studies. journal of Neurochemistry,2006,97, 1611-.

(c) Mechanism of ion channel blocking: for example, non-competitive versus non-competitive blocking: non-competitive blockade allows for increased blockade with increased overstimulation while the drug maintains a stable concentration. For example, memantine (Chen and Lipton,2006) and dextromethorphan may exert this type of blocking effect.

(d) The number and location (expression) of NMDAR and other glutamate receptors, including AMPA receptors on different areas of the cell membrane and cell membranes of selected neuronal populations (dextromethorphan increases GluR1 in rat models of depression and mRNA encoding NMDAR1 in retinal cells): different drugs may be more selective for certain neuronal populations and certain neuronal circuits (hansen et al, 2018).

(e) The relative number of ion channels that were open and closed at a particular time and the number and location of NMDA channels that were overactive (at risk of inducing excitotoxicity) (hansen et al, 2018).

(f) The time at which the drug enters and leaves the receptor (start, offset, capture and "step-in-gate" concepts) is also related to the relative amounts of NMDAR and other glutamate ion channel type receptors in the open or closed state.

(g) Affinity of the drug for the receptor. Improved affinity, when not associated with capture, may allow for lower and better tolerated doses.

(h) Drug selectivity for receptors, including NMDAR subtypes, and even within-subtype genetically and epigenetically determined variants, such as NR1, NR2A-D, and NR3A-B (Low and Wee, 2010). Altering the level of activity of a drug with NMDAR modulating activity based on receptor subunit composition may be important for designing a drug that is effective in patients within a specific age range, altering its potential therapeutic efficacy in children and adults ADHD. Furthermore, preferential blocking of NMDAR assembled in supercomplexes (NMDAR assembled in supercomplexes requires NR 2B-tripartite rules) may also be beneficial for selected diseases. Approximately 3% of PD95 superflex comprises NMDAR (Frank et al, 2017). The discovered increase in dextromethadone-induced PD95 (from the data above "development of NMDAR antagonist d-methadone for treatment of depression and other CNS diseases", and figures 1-5, particularly figure 4B) indicates a specific effect of dextromethadone on NMDAR, which is part of a super complex. The activity of a drug on the NMDAR in the super-complex may also be a factor in its PK parameters, e.g. a drug with a favourable partition coefficient (e.g. a fluoro derivative of dextromethorphan) may be more able to reach the NMDAR fraction of the super-complex.

(i) Activity of drugs on selected neuronal populations and circuits of NMDAR hyperactivity caused by genetic, environmental or genetic + environmental triggers.

(j) Activity of the drug in selected brain regions, neuronal subpopulations, brain circuits.

(k) The activity of drugs in selected pathological states, including different stages of the same disease, e.g. memantine, is used for the treatment of moderate and severe alzheimer's disease, but not for early stage alzheimer's disease.

(l) Alterations in absorption, distribution, metabolism, excretion may prove beneficial for the selected disease.

(m) the activity of specific new drugs on targets other than NMDAR may make it a better therapeutic option for specific diseases, such as amantadine for the treatment of parkinson's disease and dextromethorphan for the treatment of depression.

(n) although only the progress of a properly designed and implemented drug development program could best characterize whether a particular structural modification applied to an opioid drug with potential Therapeutic NMDAR modulating effects would lead to PK and PD advantages, more specifically, whether changes that would lead to interaction between a new drug and NMDAR might prove beneficial for a particular disease [ among other modifications, nitro derivatives of drugs targeting NMDA channel pores, such as dextromethorphan and potentially other SMOs, might impose additional NMDA modulation by mechanisms other than the PCP site, such as S-Nitrosylation of NMDAR subunits (NMDA Nakamura and Stuart A. Lipton. protein S-nitroso as a Therapeutic Target for neurogenetic separation. trends in pharmaceutical Sciences, January 2016, NMDA Vol.37 No. 1; mStastal. US, Int. patent No. 5593876A; Pearlait. 78. metadata, Pentium. catalog, peptide, catalog, publication No. 54, polypeptide, environmental research, 1994, issue 1). However, it should be considered that if nitro derivatives of NMDAR antagonist drugs, including SMO, should prove effective for a particular disease, this therapeutic effect may result from the above theoretical mechanism [ protein channel (NMDAR subunit) S-nitrosylation overactive NMDAR, NO induced channel closure ] or it may be entirely for another reason, one of the many reasons outlined in this disclosure, including improving the opening/migration of NMDA receptor pore channels not associated with S-nitrosylation. The inventors have recognized that simply increasing the level of channel blockade does not necessarily have a therapeutic advantage (MK-801, PCP, and ketamine are examples of drugs that provide "stronger", "capture" NMDAR blockade leading to adverse side effects); on the other hand, changes in the molecular structure of selected opioids that determine their PK and PD functional changes may prove beneficial for the selected disease, and thus, the synthesis of fluoro and nitro derivatives, as well as other possible structural modifications of opioids, may lead to new potentially effective NMDARs. Furthermore, although reactive free radicals, Reactive Oxygen Species (ROS) and RNS are normal components of cellular metabolism, the overproduction of these types of free radicals can result in the failure of the cell to regulate them, leading to an imbalance in redox and the development of oxidative stress. Nitro derivatives of opioids with NMDAR activity and the tropism of NMDAR can modulate the production of these reactive free radicals and prevent or reduce cellular damage.

(o) as described above, on the activity of NMDAR outside the CNS, and on the design of SMO that does not cross the BBB and may be active on neurons (e.g., sensory or autonomic neurons) or non-neuronal cells (e.g., pancreatic or cardiac cells) in the PNS, where modulation of NMDAR may provide therapeutic or prophylactic benefit.

The present inventors have discovered that selecting selected opioid drugs with baseline NMDAR modulation potential, after structural modification, including structural modifications that result in fluoro and nitro derivatives as well as fluoro-nitro derivatives and deuterated forms thereof (SMO), for one or more of the reasons and mechanisms listed above, are potential candidates for drug development programs for particular diseases and conditions exacerbated by NMDAR dysfunction. Thus, the design and PK and PD characterization of SMOs with potential regulatory effects at NMDAR including transmembrane and/or extracellular domains is an object of the present disclosure.

Astrocytes and NMDAR: astrocyte morphology and gene expression vary greatly depending on location, local contact and microenvironment. Astrocytes provide key regulation of synaptic glutamate concentration through bidirectional and unidirectional transporters. Astrocytes also bind to neurons, including glutamatergic and gabaergic interneurons, and regulate neuronal activity by modulating neurotransmitter levels. In addition, astrocytes are interconnected in a network by gap junctions. Complex brain activityActions, such as default mode networks and ultimately even conscious thought, may be related to astrocytic modulation of neuronal activity, but not the opposite. In addition, astrocytes express all seven subtypes of NMDAR (Ming-Chak Lee; Ka KaTing; Adams series. Characterisation of the Expression of NMDA Receptors in Human astrocytes. PLoS One, 11/2010, Volume 5, Issue 11). Calcium influx studies indicate that both glutamate and quinolinic acid can over-activate the NMDAR of astrocytes, resulting in Ca2+Influx cells and astrocytes are dysfunctional and even dead. NMDAR antagonists (MK-801 and memantine) prevent glutamate and quinolinate induced excitotoxicity. Astrocytic NMDARs may also play an important role in promoting glial signaling in the CNS, and therefore they are not critical for dysfunction. Modulation of NMDAR (including subtypes thereof) of dysfunctional astrocytes includes a therapeutic target that may represent a potential for treatment of a variety of diseases. SMO and its different activities on NMDAR and NMDAR subtypes, unique PK and PD, including initiation/offset/capture differences of NMDAR blockade, as well as potential differences and spatiotemporal differences in activity (cells, cell populations, circuits and brain regions) of NMDAR that preferentially block neurons or astrocytes, provide a platform for the development of new drugs that may be helpful in treating a variety of diseases and disorders caused by dysfunctional NMDAR, not only on neurons, but also on astrocytes.

Neural plasticity: neural plasticity is crucial to normal development, memory formation and learning, and ultimately determines individual cognitive function, personality, behavior and mood. Sporadic or genetic diseases may be driven by abnormal neuroplasticity resulting from normal sensory stimuli and experience. An experimental model of Rett syndrome gives an example of abnormal neural plasticity. In this mouse model, normal visual stimuli have negative effects on vision early in development (Patrizi et al, 2016), while ketamine, an NMDAR antagonist, prevents these negative effects. In addition to inherited or sporadic neurological genetic diseases, the genetic predisposition of certain diseases is associated with one or more specific environmental factors (packages)Including the different toxins (G + E concept) above) may lead to abnormal CNS plasticity and to neuropsychiatric disorders. In the presence of severe genetic disease (e.g., the Rett syndrome animal model discussed above), the "toxic" environmental factor may be a normal sensory experience (visual stimulus). Conversely, when an individual is genetically predisposed to exhibiting enhanced susceptibility to one or more environmental insults, these insults may be very diverse, such as endogenous toxins (e.g., quinolinic acid); or food (e.g., polyamine-rich food, alcohol); or drugs (e.g., aminoglycosides and cisplatin), or excess neurotransmitters, such as excitotoxicity caused by glutamate, or even autoantibodies against NMDAR. Furthermore, these environmental factors may be known, such as those above, even unknown, yet undefined, factors that affect neuroplasticity only in the presence of genetic predisposition by NMDAR. If the toxic substance is sufficiently harmful, it will determine that the neural circuit is abnormal in most "normal" individuals. In the case of certain neuropsychiatric disorders, the trigger for susceptible individuals may not be a chemical or physical factor, but may also be a particularly stressful (toxic) "life history", as is the case for PTSD patients and some patients with depression and anxiety.

Regardless of the trigger for cellular dysfunction, nitro dextromethorphan drugs may prove clinically useful when such dysfunction is mediated by NMDAR and/or NO pathways. In a 1994 paper (Inturrisi, ce. nmda receptors, nitrile oxide and opioid toxin Peptides,1994, Volume 54, Issue1), Charles Inturrisi, one of the inventors of the present application, predicted how morphine administration leads to tolerability and hyperalgesia and how NMDAR and NO pathways participate in the development of these side effects of morphine. The present inventors have now proposed a new pool of molecules (SMO) with potential NMDAR antagonism based on the new in silico test results shown in the present disclosure, and these SMO may have a modulating effect on NMDAR and NO pathways due to specific structural molecular modifications, such as NO or nitrate substitutions, applied to opioids. Thus, these novel molecules may play a role in the treatment of diseases, where modulation of NMDAR activity and/or modulation of NO pathways are involved in the pathophysiological mechanisms of diseases and disorders. These include all diseases and conditions disclosed in international patent application No. PCT/US2018/016159, and all diseases and conditions defined in the first paragraph of this application.

In view of the new explanations set forth throughout the application, morphine tolerance and hyperalgesia can be seen as a manifestation of abnormal neuroplasticity induced by toxins (in this case morphine). Many other chemical substances (e.g., aminoglycosides, cisplatin, domoic acid, polyamines, quinolinic acid, etc.) or physical factors, including trauma or brain radiation therapy, or electroconvulsive therapy (ECT) or even vocal and other sensory stimuli, as well as many diseases and conditions, including neuropsychiatric diseases caused or exacerbated by NMDAR dysfunction or NO pathway dysfunction, can therefore be ameliorated by dextromethorphan nitro derivatives. The "toxic life history" that leads to neuroplasticity abnormalities and NS-loop abnormalities in susceptible individuals may also be a fundamental or contributing factor to neuropsychiatric diseases such as depression, anxiety, PTSD, ADHD, schizophrenia, etc. (Chen and Baran, 2016).

Novel chemical entities having modulatory activity in the NMDAR and NO pathway, such as opioids and nitro derivatives of their enantiomers (SMO), can prevent or ameliorate these abnormal circuits triggered or maintained by "toxic experiences" or any of the chemical or physical factors listed above, which may underlie a variety of neuropsychiatric disorders. The present inventors have previously found that dextromethorphan increases human BDNF levels (international patent application No. PCT/US2018/016159), and in this submission, the present inventors disclosed that dextromethorphan increases expression of mRNA encoding NMDAR1 induced by dextromethorphan (see examples below, and fig. 10). Structural modifications of the dextromethorphan molecule, such as dextromethorphan nitro derivatives, have the potential to modulate the NO pathway, which may further enhance the neuroplasticity potential of the parent molecule, thereby expanding the therapeutic potential of dextromethorphan to treat one or more diseases and disorders.

In addition, the novel compounds disclosed herein, particularly the dextromethorphan nitro derivatives, and NMDAR antagonists in general, including dextromethorphan, may be particularly useful when administered to patients receiving electroconvulsive therapy (ECT). ECT may disrupt abnormal neural circuits resulting from abnormal neural plasticity that are manifested in patients with depression or other symptoms of neuropsychiatric disease. These new compounds, SMO, may help to restore and maintain a normal circuit when used alone or in combination with other drugs, ECT and even psychotherapy. In addition to SMO, including opioid nitro derivatives and ECT, psychological treatment may be a third useful therapeutic modality, as it may help SMO restore and preserve healthy neural circuits during and after ECT or other forms of treatment.

One of the applicants of the present disclosure, Charles Inturrisi, in 1994, states that "collectively, these results indicate that μ tolerance can be modulated at either the NMDA receptor or NOS (or both) and that both systems can be targeted for the development of new drugs. ", (Inturrisi, CE. NMDA receptors, nitrile oxide and opioid Peptides,1994, Volume 54, Issue1), as demonstrated by the new data provided throughout the application, can now be applied to SMO, including opioid nitro derivatives, and a variety of diseases and conditions, not just mu opioid tolerance due to morphine-induced aberrant neuroplasticity. SMO, including dextromethorphan nitro derivatives, may therefore be directed against NMDAR dysfunction and/or abnormal neuroplasticity due to a variety of causes.

Targeting, off-target and mixed targeting/off-target effects of opioids and SMOs

In addition to the diversity of targeting NMDAR and its subtypes and variants, and the nitric oxide pathway, as described in this application, and the effects on other receptors and systems detailed throughout the application, the inventors will also determine and characterize the manipulation of each SMO at the relevant non-targeted site, as appropriate. In particular, the inventors are investigating SMO in view of its opioid receptor activity, selecting molecules with lower affinity for these receptors or with potentially advantageous partial agonist or mixed agonist antagonist activity, including activity more specific for one or the other opioid receptor subtype, seeking better clinical tolerance than strong mu opioid receptor agonists, possibly even increased effectiveness: for example, if the novel SMO, in addition to being an NMDAR open channel blocker, is also a kappa opioid antagonist, it may provide additional therapeutic effects for the treatment of depression (Lowe, Stephen L; Wong, Conrad J; Witcher, Jennifer. safety, tolerability, and pharmacological evaluation of single-and multiple-influencing groups of a novel kappa optical receptor antagonist, the Journal of Clinical Pharmacology,09/2014, Volume 54, Issue 9).

In addition to the opioid receptors, potassium channels represent another potential off-target site of action to be studied during development programs: SMO, which has less of a possibility of blocking potassium channels associated with QT interval prolongation, may be advantageous for developing programs. However, as with opioid receptors, potassium channels may also be present at the targeted site of action, for example if the potassium channel blocking effect does not cause heart disease, but provides other therapeutic advantages, as described for dextromethorphan in international patent application No. PCT/US2018/016159, as described for dextromethorphan, Wulff et al, 2009 (Wulff H, Castle NA, Pardo la. voltage-gated potassiums channels as a therapeutic target. nat Rev Drug Discov 2009 Dec; 8(12): 982) 1001).

SMO scheme

With the aim of developing a new safe and effective NMDAR modulator library with specific PK and PD characteristics, which is most suitable for the chosen indication, the applicant, in collaboration with the university of Padova (italy) and the institute of biological research at the university of italy, switzerland (switzerland), implemented a drug development protocol comprising the design of new chemical formulas derived from SMO, including the opioid enantiomers with low affinity for the opioid receptors and with NMDAR antagonistic potential. A first group of novel chemical entities designed for their potential activity on NMDAR and/or for targeting NO pathways is provided in the present application (table 1). The newly designed molecules (SMO) were then tested in a new static and dynamic in silico model of the transmembrane domain of NMDAR (tables 2a-c) and table 3).

After selecting more promising molecules, and after completing the synthetic work for the selected molecules, the present inventors are performing in vitro and in vivo experimental work with potential clinically useful NMDAR and other targeting effects and potential off-target effects for treating diseases in order to fully characterize and define the potential safety and PK and PD properties of the new molecules. These new chemical entities and potential new drugs include dextromethorphan derivatives and other opioid derivatives (SMO).

Structural modifications that result in potentially favorable NMDAR subtype binding affinities in silico and in vitro also provide information on Structural Activity Relationships (SAR), allowing further improvements in silico models and improved selection of new molecules (SMO). As this protocol progressed, while defining the differential effects of NMDAR subtypes by electrophysiological testing of cells transfected with specific NMDAR subtypes, the inventors were confirming the biological activity of SMO on cellular models of excitotoxicity and ion channel hyperactivity, including CNS cells and other cells, including retinal cells and other specialized cells. The present inventors are identifying specific diseases in order to perform an in vitro preclinical test for cell-specific effects associated with these diseases (ex vivo dextromethorphan studies on retinal cells treated with inflammatory mediators-see examples below), followed by a preclinical disease model (see from "development of NMDAR antagonist d-methadone for treatment of depression and other CNS diseases", see above and figures 1-5), and then finally into a clinical development phase for more promising molecules (e.g. ongoing studies of phase 2 clinical dextromethorphan treatment resistant depression). These SMOs may offer PK and PD advantages over dextromethorphan and other currently available NMDAR antagonists, and they may offer improved selectivity for NMDAR, NMDAR subtypes, brain regions, neuronal and astrocytic subpopulations, as well as disease and condition-affected CNS circuits, and they may offer spatial or temporal or global receptor affinity advantages, ultimately leading to disease-specific advantages, as detailed above. In addition to the neuron population, the effects of SMO may be useful for the neuron outer cell populations and circuits described in the application.

The present inventors have shown and previously disclosed (international patent application No. PCT/US2018/016159) that certain deuterated dextromethorphan molecules may have an affinity for NMDAR subtypes that is different from the affinity exhibited by dextromethorphan and other NMDAR antagonists (international patent application No. PCT/US2018/016159), particularly with D9 having twice the affinity for the NR2B receptor subtype than for NR2A in patch clamp studies, and thus, as mentioned above, deuteration and other structural modifications may result in potentially advantageous drug profiles compared to dextromethorphan and other NMDAR antagonists for specific indications. By this relatively small modification (deuteration) of the dextromethorphan molecule, the inventors were able to influence the relative affinity of dextromethorphan for the receptor subtype (NR1-NR2A vs N1-NR2B tetramer complex) and were also able to alter the PK parameters in vitro: the inventors tested dextromethorphan and deuterated dextromethorphan in an in vitro metabolic assay and demonstrated how deuteration altered the results of an in vitro metabolic assay in an individual. The inventors also compared these results to similar tests using dextromethorphan, which showed significantly shorter half-lives compared to dextromethorphan and deuterated dextromethorphan (D9, D10, D16) (international patent application No. PCT/US 2018/016159). By further modifying the structure of dextromethorphan and/or its deuterated and tritiated derivatives, the present inventors are designing and testing molecules with the potential to improve PK and PD characteristics that may prove more beneficial for selected diseases than dextromethorphan and deuterated dextromethorphan, as well as other opioids and other NMDAR antagonists. The same principle (deuterated SMO can produce new molecules with potential PK and PD advantages) may be more relevant to the more substantial structural modifications disclosed below, including fluoro and nitro derivatives as well as fluoro and deuterated nitro and deuterated fluoronitro derivatives.

Furthermore, the present inventors have shown that different NMDAR antagonists (ketamine, memantine, PCP) act at or near the same NMDA receptor site as dextromethorphan (PCP site), with similar but different affinities compared to dextromethorphan and its deuterated derivatives, including different affinities for different receptor subunit complexes (e.g., NR1-NR2A or NR1-NR2B and other possible combinations with other subunits) (international patent application No. PCT/US 2018/016159).

There are also differences in the "capture", opening and shift of NMDAR blockade in SMO, as shown by structural modification of PCP, resulting in ketamine being a lower capturing activity drug than PCP (Zanos et al, 2018), and amantadine, as shown by structural modification of amantadine, resulting in memantine being a lower affinity drug for NR1-NR2A than NR1-NR 2B. A difference in blockade to the NMDAR is expected for each SMO object of the present disclosure. These different SMOs may have therapeutic significance for the different affinities of the NMDAR.

The burden of CNS disease is enormous and treatment is rare and often only partially effective. Novel safe and effective NMDAR modulators with potential therapeutic advantages for selected diseases represent a highly unmet medical need.

While there are indeed hundreds of diseases that may benefit from an NMDAR modulator, as described above and detailed by the inventors in international patent application No. PCT/US2018/016159 and throughout the present application, there are only four FDA approved drugs targeting the NMDAR transmembrane domain for diseases that have NMDAR dysfunction as a common drug target, one of which is a combination drug: amantadine, memantine, esketamine, and the combination dextromethorphan + quinidine. The fifth FDA-approved NMDAR modulator, ketamine, was approved for anesthesia, but could not be used to treat diseases and disorders.

The present inventors disclose the potential for newly designed SMOs to have safety and efficacy against one or more specific diseases and conditions, including diseases and conditions for which NMDAR blockade or modulation may be beneficial, including the diseases and conditions disclosed in international patent application No. PCT/US2018/016159 and the diseases and conditions defined in the present application.

Based on the results from FST, futt, NFST, CUS and the immunohistochemical and morphological and electrophysiological data described above in "development of NMDAR antagonist d-methadone for the treatment of depression and other CNS disorders" indicating that the antidepressant and neuroplasticity effects of dextromethadone are similar to ketamine, dextromethadone and SMO are not only useful for the treatment of psychiatric disorders and symptoms, including various forms of depression, various forms of anxiety, PTSD, addictive behaviors and drug addiction, but may be prevented when administered during periods of anticipated stress or stress prior to development of psychiatric disorders or symptoms. By promoting neuroplasticity and other mechanisms, such as modulation of NMDAR, SERT, NET and BDNF, dextromethorphan and SMO taken during periods of high stress of life (experience of CNS toxicity) or when stressful events are expected, may increase resistance to development of psychiatric diseases and symptoms, and thus dextromethorphan and SMO may help prevent psychiatric diseases and symptoms, including those triggered by mental stress due to a variety of causes, including social stress, sadness, illness, loss including economic loss and loss of relative pain, stress related to marital and family, war, natural disasters, and the like. This is also supported by the results of the Brachman et al ketamine experiments (Brachman RA, McGowan JC, Perusini JN, et al, Ketamine as a functional age Stress-Induced depression-like behavor. biol Psychiatry. 2015; 79(9): 776-.

Some of the disclosed compounds are optimized with the aim of obtaining clinically tolerated NMDAR antagonists which are active in the NO pathway and/or which have the potential for specific therapeutic effects on selected diseases and disorders based on different effects in selected areas of the CNS and additional CNS, because of specific PK parameters (e.g. lipid solubility of fluoro derivatives, such as DMD35, LMA9, dim 6, LPP6, NMeDMD9 and other examples of the above compounds, as well as halogen compounds, such as DAN-DMD38, DMD63, DMD41) or PD parameters (e.g. different effects and affinities for NMDAR, including potentially different effects and affinities for receptor subtypes-e.g. SMO disclosed herein, including those with additional blocking effects and additional neuroprotective effects on NMDAR, such as nitro-derivatives, e.g. nitro-DMD 1, LMA8, dii 8, NMeDMD8, including deuterated fluoro and nitro derivatives, or may produce additional effects on different receptors and transporters, including those that result in changes in neurotransmitters, e.g. serotonin and NE, opioids and DA and GABA pathways, or changes in neurotrophic factors, such as BDNF, or changes in synaptophysin, such as PD95, and thus different effects on the supercomplexes, or effects on GluR1 and NMDAR1 and consequent neuroplasticity effects.

The present inventors disclose compounds, including salts thereof, as outlined below, for the treatment and prevention of human and veterinary diseases and disorders, including diseases and disorders for improving cognitive and social functions, and for anti-aging use, including the prevention and treatment of accelerated aging factors or drug therapy caused by the environment, particularly if caused by NMDAR dysfunction or NO pathway dysfunction of clinically tolerated and effective NMDAR and NO pathway modulators, including effects on RNS, having specific PK and PD characteristics, including different affinities for NMDAR subtypes, may be advantageous.

The molecules shown in table 1 were designed for potential NMDAR modulation.

The molecules shown in tables 2a and 2b show in silico NMDAR affinity in a static model to assess potential activity against the NR2B subtype of NMDAR receptor. Each molecule designed has unique PK and PD characteristics, including effects on NMDAR and other receptors that may be used to treat disease. These compounds are shown in tables 2a, 2 b.

The molecules shown in table 3 show in silico NMDAR affinity in static and dynamic models to assess potential activity against the NMDAR receptor NR2B subtype. Each molecule designed has unique PK and PD characteristics, including effects on NMDAR and other receptors that may be used to treat disease. These compounds are shown in table 3.

Finally, the inventors disclose as examples of compounds according to the principles of the various aspects of the present invention, dextromethorphan fluoro derivatives (-F), including fluoro-dextromethorphan; dextromethorphan nitro derivative (-NO)2) Including nitro dextromethorphan; dextromethorphan fluoronitro derivatives including fluoro-nitro-dextromethorphan; and deuterated dextromethorphan derivatives (deuterated dextromethorphan fluoro derivatives (-F) including fluoro-dextromethorphan, deuterated dextromethorphan nitro derivatives (-NO) modified as above for dextromethorphan2) Including nitro dextromethorphan; and deuterationDextromethorphan fluoronitro derivatives, including fluoro-nitro-dextromethorphan).

General examples of such compounds may also include dextro-isomethadone derivatives, including: dextro-isomethadone fluoro derivatives including fluoro-dextro-isomethadone; dextro-isomethadone nitro derivatives including nitro dextro-isomethadone; dextromethorphan fluoronitro derivatives including fluoro-nitro-dextromethorphan; and deuterated dextro-isomethadone derivatives modified with dextro-isomethadone as described above.

General examples of such compounds may also include N-methyl-dextromethorphan derivatives, including: n-methyl-dextromethorphan fluoro derivatives including fluoro-N-methyl-dextromethorphan; n-methyl-dextromethorphan nitro derivatives, including nitro-N-methyl-dextromethorphan; n-methyl-dextromethorphan fluoronitro derivatives including fluoro-nitro-N-methyl-dextromethorphan; and deuterated N-methyl-dextromethorphan derivatives modified as described above.

General examples of such compounds may also include levomoramide derivatives, including: levo-morpholino fluoro derivatives including fluoro-levo-morpholino; levo-morpholinone nitro derivatives, including nitro-levo-morpholinone; levorphanol fluoro-nitro derivatives, including fluoro-nitro-levorphanol, and deuterated levorphanol derivatives modified as described above for levorphanol.

General examples of such compounds may also include levopropoxybenzene derivatives, including: levopropoxyphene derivatives, including levopropoxyphene fluoro derivatives, including fluoro-levopropoxybenzene; levopropoxyphene nitro derivatives, including nitro-levopropoxyphene; levopropoxyphene fluoronitro derivatives, including fluoro-nitro-levopropoxyphene; and deuterated levopropoxyphene derivatives modified as described above.

General examples of such compounds may also include levorphanol derivatives, including: levorphanol fluoro derivatives including fluoro-levorphanol; levorphanol nitro derivatives, including nitro-levorphanol; levorphanol fluoronitro derivatives including fluoro-nitro-levorphanol; and deuterated levorphanol derivatives modified as described above.

General examples of such compounds may also include dextromethorphan and dextrorphan derivatives, including: dextromethorphan and dextrorphan fluoro derivatives, including fluoro-dextromethorphan and nitro-dextrorphan; dextromethorphan and dextrorphan nitro derivatives, including nitrodextromethorphan and nitrodextrorphan; dextromethorphan and dextrorphan fluoronitro derivatives, including fluoro-dextromethorphan and fluoro-nitro-dextrorphan; and deuterated dextromethorphan and deuterated dextrorphan derivatives modified as described above.

Examples

Effect of NMDAR antagonism on ARPE-19 Activity

From this experiment, the present inventors determined whether inhibition of the NMDAR receptor by MK-801 and dextromethorphan (REL-1017) rescued the L-glutamic acid-induced reduction in cell viability of ARPE-19. As shown in fig. 6, the cell viability of cells incubated with 1mM L-glutamate was reduced (p <0.01), which was almost negligible when the cells were pretreated with the two NMDA receptor antagonists MK-801 and dextromethorphan (REL-1017) prior to L-glutamate treatment, indicating that these compounds have a protective effect on cell viability.

Effect of NMDAR antagonism on Reactive Oxygen Species (ROS) production

Increased ROS production following NMDAR activation has been widely demonstrated in neuronal cells in vitro. This study was also performed on the retinal cell line ARPE-19 to verify how NMDAR activation and blockade affect ROS production. As shown in FIG. 7, there was no significant increase in ROS production in cells treated with 1mM L-glutamic acid. A trend of decrease was observed in cells exposed to NMDA receptor antagonists (1mM L-Glu + 30. mu.M MK-801, 1mM L-Glu + 30. mu.M REL-1017) without reaching statistical significance.

Effect of NMDAR antagonism on expression and nuclear translocation of the inflammatory transcription factor p65

To determine whether the excitotoxicity produced by the glutamine stimulation of the NMDAR receptor would stimulate or interfere with the activation of the retinal cell inflammatory mechanism, the inventors stained protein p65 belonging to the NF-kB family, thereby performing immunofluorescence coupled with a confocal microscope. It is well known that nuclear translocation of the transcription factor p65 leads to increased synthesis of molecules involved in pro-inflammatory responses. In addition, the effects of using the receptor antagonists MK-801 and dextromethorphan (REL-1017) on the expression and translocation of this protein were also evaluated.

As shown in fig. 8A-8D, increased expression of p65 in L-glutamate exposed cells (p <0.05) compared to the vector can be observed, whereas p 65-related fluorescence decreases when the cells are pre-treated with two NMDAR receptor antagonists. P65 was also evaluated in response to nuclear translocation of L-glutamate, as p65, when translocated to the nucleus, acts as a transcription factor and promotes expression of pro-inflammatory genes. The degree of co-localization between p65 and the nuclear marker DAPI is determined by the Pearson r coefficient, which represents the index of correlation between the two variables. This coefficient varies between-1 and 1, the closer to positive values, the more p65 and DAPI have the same (nuclear) localization. Figures 9A and 9B show that nuclear expression of p65 protein increased after treatment with L-glutamate (p <0.05) and decreased after pretreatment with NMDAR antagonist, comparable to vector treated cells. This experiment demonstrates that NMDA receptor activation is involved in the establishment of an inflammatory response following glutamine stimulation, as evidenced by an increase in nuclear translocation of p 65.

Effect of NMDAR antagonism on expression of target genes NMDAR1, p65, IL-6, TNF-a

After confirming the effect of NMDAR antagonists on nuclear translocation of p65, the present inventors investigated the changes in gene expression levels of the pro-inflammatory cytokines IL-6 (interleukin-6) and TNF-a, the transcription factor p65 and the NMDA receptor in order to confirm their anti-inflammatory effects.

Quantification of relative NMDAR mRNA expression levels

The first gene to assess gene expression levels is the 1A subunit of the NMDAR receptor dimer (referred to as NMDAR 1).

FIG. 10 shows that stimulation of the NMDA receptor with L-glutamate results in a slight decrease in the basal gene expression of the receptor, possibly through a negative feedback mechanism, although this decrease is not statistically significant. In contrast, when receptor activation is blocked by the antagonist dextromethorphan (REL-1017), the cells significantly increase their expression (p <0.0001) up to 9-fold compared to the expression levels of vector-treated cells. This increase in NMDAR1 gene expression was only slightly apparent when MK-801 was used as an antagonist.

Quantification of relative p65 mRNA expression levels

Gene expression analysis of p65 (fig. 11A and 11B) showed that activation of the NMDA receptor by the agonist L-glutamate resulted in a significant increase in its gene expression (p <0.05), whereas in cells pretreated with the receptor antagonist dextromethorphan (REL-1017), the gene expression of p65 was reduced (p < 0.01). These data are consistent with those observed in immunocytochemistry analyses. Furthermore, the inventors could conclude that L-glutamic acid triggers an inflammatory response, which is uniquely protected by dextromethorphan (REL-1017).

Finally, gene expression levels of proinflammatory cytokines have been investigated under different experimental conditions.

Quantification of TNF-a and IL-6mRNA expression levels

FIG. 12 shows that TNF-a gene expressing cells treated with L-glutamic acid were significantly increased (p <0.01), whereas the expression of this proinflammatory cytokine was restored to normal levels when the receptor antagonists MK-801 and dextromethorphan (REL-1017) were added. The reduction in TNF-a gene expression levels was slightly more pronounced in samples pretreated with dextromethorphan compared to MK-801.

FIG. 13 shows that IL-6 gene expression tends to increase when the NMDA receptor is stimulated by glutamate; there was a tendency to decrease when cells were pretreated with the receptor antagonists MK-801 and dextromethorphan. There was no significant difference in the reduction of expression in the samples antagonized by MK-801 and dextromethorphan (REL-1017) receptors. In contrast, there was no significant change in IL-6 gene expression levels following NMDA receptor activation by L-glutamate or inhibition by MK-801 and dextromethorphan (REL-1017). These data indicate that TNF-a appears to be the major cytokine involved in cytotoxicity and inflammation following glutamine stimulation, since inhibition of NMDAR activation by MK-801 and dextromethorphan (REL-1017) has a significant effect on the reduction of its gene expression. Following the excitotoxicity associated with NMDAR activation, the increase in TNF-a gene expression levels is consistent with activation of the NF- κ B transcription complex and the resulting translocation of p65 into the nucleus. In fact, TNF- α plays a role in promoting NF-. kappa.B activation in retinal cells. These findings are also consistent with the observation that TNF- α induces optic nerve degeneration and may delay retinal neuronal cell death, and that an increase in p65 expression in the optic nerve may be associated with TNF- α induced axonal degeneration.

The embodiments of the invention recited herein are intended to be merely exemplary and those skilled in the art will be able to make numerous variations and modifications thereto without departing from the spirit of the invention. Nevertheless, certain variations and modifications, while producing less than desirable results, may still produce satisfactory results. All such variations and modifications are intended to fall within the scope of the present invention as defined by the appended claims.

103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:用于运动障碍的治疗药物

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!