Demethylanisolanetrithione derivatives for the treatment of diseases associated with mitochondrial Reactive Oxygen Species (ROS) production

文档序号:1602155 发布日期:2020-01-07 浏览:19次 中文

阅读说明:本技术 用于治疗与线粒体活性氧(ros)产生相关的疾病的去甲基茴三硫衍生物 (Demethylanisolanetrithione derivatives for the treatment of diseases associated with mitochondrial Reactive Oxygen Species (ROS) production ) 是由 P·迪奥莱兹 F·马林 O·佩提特金 于 2018-03-07 设计创作,主要内容包括:本发明涉及去甲基茴三硫(AOX)及其衍生物,特别是式(I)的衍生物,用于预防和治疗其开始和/或进展与线粒体来源的活性氧(ROS)的产生和作用有关的疾病。<Image he="161" wi="700" file="DDA0002289663480000011.GIF" imgContent="drawing" imgFormat="GIF" orientation="portrait" inline="no"></Image>(The present invention relates to demethylanethole trithione (AOX) and derivatives thereof, in particular of formula (I), for the prevention and treatment of diseases the onset and/or progression of which is associated with the production and action of Reactive Oxygen Species (ROS) of mitochondrial origin.)

1. A compound of formula (I):

Figure FDA0002261051960000011

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein:

x represents S, O or NHOH; preferably, X is S or O; more preferably, X is S;

y represents CH, C or N; preferably, Y is CH or N; more preferably, Y is CH;

R1、R2、R4and R5Each independently represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl;

R3is a hydroxyl group; or R3And R2Together with the carbon atom to which they are attached form a 5-membered heteroaryl moiety, wherein-R3-R2-represents-A-CR6or-B ═ CR6-a-; wherein:

a represents O, S or NR7(ii) a Wherein R is7Represents hydrogen, C1-C8 alkyl or alkoxycarbonyl;

b represents CH or N; and

R6represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl;

which are useful as inhibitors of Reactive Oxygen Species (ROS) production in the treatment and/or prevention of diseases associated with free oxygen radicals.

2. The compound for use according to claim 1, wherein the compound is selected from:

5- (4-hydroxyphenyl) -3H-1, 2-dithiole-3-thione;

5- (4-hydroxyphenyl) -3H-1, 2-dithiol-3-one;

5- (4-hydroxyphenyl) -3H-1, 2-dithiol-3-one oxime;

5- (4-hydroxyphenyl) -3H-1,2, 4-dithiazole-3-thione;

4- (4-hydroxyphenyl) -3H-1, 2-dithiole-3-thione;

5- (2-hydroxybenzo [ d ]]

Figure FDA0002261051960000023

5- (2-hydroxybenzo [ d ] thiazol-6-yl) -3H-1, 2-dithiole-3-thione;

5- (benzofuran-5-yl) -3H-1, 2-dithiole-3-thione; and

5- (3-thio-3H-1, 2-dithiolan-5-yl) -1H-indole-1-carboxylic acid methyl ester.

3. A compound for use according to claim 1 or claim 2, wherein the compound is 5- (4-hydroxyphenyl) -3H-1, 2-dithiole-3-thione.

4.A compound for use according to claim 1, which is a compound of formula (II) or (III):

Figure FDA0002261051960000021

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, X, Y, R1、R4、R5、R6A and B are as defined in claim 1.

5. Compound for use according to any of claims 1-4, wherein the compound inhibits the mitochondrial production of ROS, preferably wherein the compound inhibits I of complex I of mitochondriaQMitochondrial generation of ROS at the site.

6. The compound for use according to any one of claims 1-5, wherein the disease associated with free oxygen radicals is selected from the group comprising: cardiovascular disease, aging disease, autoimmune disease, premature aging syndrome, Parkinson's syndrome, neurological disease, ischemia reperfusion injury, infectious disease, muscle disease, lung, kidney and liver disease.

7. Compound for use according to claim 6, wherein the cardiovascular disease is selected from the group comprising: myocardial infarction, cardiotoxicity, pulmonary hypertension, heart failure, cardiopulmonary disease, ischemia, heart attack, stroke, thrombosis, and embolism.

8. A compound of formula (I'):

Figure FDA0002261051960000022

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein:

x represents S, O or NHOH; preferably, X is S or O; more preferably, X is S;

y represents CH, C or N; preferably, Y is CH or N; more preferably, Y is CH;

R1、R4and R5Each independently represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl;

R2’and R3’To which they are connectedThe carbon atoms together form a 5-membered heteroaryl moiety, wherein-R3’-R2’-represents-A-CR6or-B ═ CR6-a-; wherein:

a represents O, S or NR7(ii) a Wherein R is7Represents hydrogen, C1-C8 alkyl or alkoxycarbonyl;

b represents CH or N; and

R6represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl.

9. The compound of claim 8, of formula (IIa), (IIb), (IIIa), or (IIIb):

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, X, Y, A, B, R1、R4、R5And R6As defined in claim 8.

10. The compound according to claim 8 or claim 9, of formula (IIa-1), (IIa-2), (IIIa-1) or (IIIa-2):

Figure FDA0002261051960000041

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, X, Y, A, B, R1、R4、R5And R6As defined in claim 8.

11. The compound according to any one of claims 8-10, which is a compound of formula (IIa-1a), (IIa-1b), (IIa-1c), (IIa-1d), (IIa-1e), (IIa-2a), (IIa-2b), (IIa-2c), (IIa-2d), (IIa-2e), (IIIa-1a), (IIIa-1b), (IIIa-1c), (IIIa-1d), (IIIa-1e), (IIIa-2a), (IIIa-2b), (IIIa-2c), (IIIa-2d) or (IIIa-2 e):

Figure FDA0002261051960000042

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, X, R1、R4、R5、R6And R7As defined in claim 8.

12. A compound according to claim 8 or claim 10, of formula (IIb-1), (IIb-2), (IIb-3), (IIb-4), (IIb-5), (IIIb-1), (IIIb-2), (IIIb-3), (IIIb-4) or (IIIb-5):

Figure FDA0002261051960000061

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, X, R1、R4、R5、R6And R7As defined in claim 8.

13. A pharmaceutical composition comprising a compound according to any one of claims 8-12, or a pharmaceutically acceptable tautomer, salt, or solvate thereof, and at least one pharmaceutically acceptable excipient.

14.A medicament comprising a compound according to any one of claims 8-12, or a pharmaceutically acceptable tautomer, salt, or solvate thereof.

15. A process for the preparation of a compound of formula (IIa-1) according to claim 10 or claim 11, or a pharmaceutically acceptable tautomer, salt or solvate thereof, characterized in that said process comprises:

a) cyclizing a compound of formula (C) with a sulfur-based reagent in the presence of a siloxane,

Figure FDA0002261051960000071

wherein, A, B, R1、R4、R5And R6As defined in claim 10;

thereby obtaining a compound of formula (IIa-1'):

Figure FDA0002261051960000072

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, A, B, R1、R4、R5And R6As defined in claim 10;

and optionally:

b1) the compound of formula (IIa-1') may be reacted with an oxidizing agent; preferably, the oxidant is mercury acetate Hg (OAc)2(ii) a To obtain a compound of formula (IIa-1 "):

Figure FDA0002261051960000073

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, A, B, R1、R4、R5And R6As defined in claim 10;

or

b2) The compound of formula (IIa-1') may be reacted with hydroxylamine NH in the presence of a base2OH-HCl reaction; preferably, the base is sodium acetate (AcONa); to obtain a compound of formula (IIa-1'):

Figure FDA0002261051960000081

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, A, B, R1、R4、R5And R6As defined in claim 10.

Technical Field

The present invention relates to demethylanethole trithione (AOX) and derivatives thereof, in particular derivatives of formula (I) as disclosed in the claims, for use in the prevention and treatment of diseases whose onset and/or progression are linked to the production and action of Reactive Oxygen Species (ROS) of mitochondrial origin.

Background

Mitochondria are at the heart of the currently accepted "radical theory of aging" and are therefore involved in the pathogenesis of almost all diseases associated with aging, including cardiovascular diseases, neurodegenerative diseases (parkinson's disease, alzheimer's disease, etc.), cancer and diabetes, and tissue dysfunction caused by ischemia. This theory suggests that the accumulation of damage caused by Reactive Oxygen Species (ROS) affects many cellular functions, particularly mitochondrial function, which is critical for energy supply and optimal cellular function (functioning). Mitochondria therefore become a major target of ROS, as optimal cellular function is crucial for providing energy for cellular self-repair.

Interestingly, mitochondria are the main source of Reactive Oxygen Species (ROS) and are therefore particularly attacked by oxidative damage. Therefore, ROS produced by mitochondria themselves cause oxidative damage, resulting in mitochondrial dysfunction and cell death.

Various antioxidants have been tested against the physiological and pathological effects of ROS. Antioxidant research has provided many natural and engineered molecules that modulate ROS to have different selectivities against different origins (physiological (cell signaling) or pathological) of ROS. However, although ROS are associated with a variety of diseases and antioxidants have shown promise in many preclinical experiments, almost all clinical trials of antioxidant-based therapies show limited efficacy (Orr et al, 2013, freedric.biol.med.65: 1047-59).

In addition, some recent studies have also shown that excessive reduction of ROS in cells is detrimental, and that an adequate balance of ROS production appears to be necessary for cell function (Goodman et al, 2004, J.Natl. cancer Inst.96 (23): 1743-50; Bjelakovic et al, 2007, JAMA.297 (8): 842-57). As a result, there is growing interest in the selective inhibition of ROS production by mitochondria that do not affect the cellular signaling of cytosolic ROS production.

Because of the wide range of human diseases caused by oxidative damage to mitochondria, antioxidants designed for accumulation in the mitochondria have been developed. Of these mitochondrially targeted antioxidants, the most widely studied is MitoQ, which contains an antioxidant quinone moiety covalently linked to a lipophilic triphenylphosphonium cation. MitoQ has now been used in ratsAnd in a series of in vivo studies in mice and in two phase II human trials. Conditions of high ROS production can now be better characterized. It appears that ROS can be produced at multiple sites in the mitochondrial respiratory chain (Quinlan et al, 2013, Redox Biol, 1: 304-12). Maximum superoxide/H2O2The generation occurs under conditions of high reduction of the electron transporter (mainly quinone) and high values of mitochondrial membrane potential. Paradoxically, these conditions are met when mitochondrial oxidative phosphorylation is low (low muscle contraction) or under hypoxic conditions (hypoxia).

Applicants demonstrated herein that AOX (demethylanethole trithione) does not act as a classical non-specific antioxidant molecule, but more interestingly as I of complex I, which is predominantly in the mitochondrial respiratory chainQDirect selective inhibitors of oxygen Radical (ROS) production at the site, at the major mitochondrial site of ROS production, and at the major responsible site of mitochondrial dysfunction.

Accordingly, the present invention relates to AOX and its bioisostere derivatives for use in the treatment and/or prevention of diseases associated with free oxygen radicals.

Disclosure of Invention

The invention relates to compounds of formula (I)

Figure BDA0002261051970000021

Or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, X, Y, R1、R2、R3、R4、R5And R6Is defined as follows;

which are useful as inhibitors of Reactive Oxygen Species (ROS) production in the treatment and/or prevention of diseases associated with free oxygen radicals.

In one embodiment, the compound used in the present invention is selected from the group consisting of 5- (4-hydroxyphenyl) -3H-1, 2-dithiole-3-thione; 5- (4-hydroxyphenyl) -3H-1, 2-dithiol-3-one; 5- (4-hydroxyphenyl) -3H-1, 2-dithiol-3-one oxime; 5- (4-hydroxyphenyl) -3H-1,2, 4-dithiazole-3-thione; 4- (4-hydroxyphenyl) -3H-1, 2-dithiolaneAlkene-3-thiones; 5- (2-hydroxybenzo [ d ]]Oxazol-5-yl) -3H-1, 2-dithiole-3-thione; 5- (2-hydroxybenzo [ d ]]Thiazol-6-yl) -3H-1, 2-dithiole-3-thione; 5- (benzofuran-5-yl) -3H-1, 2-dithiole-3-thione; and methyl 5- (3-thio-3H-1, 2-dithiolan-5-yl) -1H-indole-1-carboxylate.

In one embodiment, the compound used in the present invention is 5- (4-hydroxyphenyl) -3H-1, 2-dithiole-3-thione (AOX).

In one embodiment, the compound used in the present invention is a compound of formula (II) or (III)

Figure BDA0002261051970000031

Or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, X, Y, R1、R4、R5、R6A and B are as defined below;

in one embodiment, the compounds of the invention inhibit the production of ROS by mitochondria. In one embodiment, the compounds of the invention inhibit I of complex I at mitochondriaQMitochondria at the site produce ROS.

In one embodiment, the disease associated with free oxygen radicals is selected from the group comprising: cardiovascular disease, aging disease, autoimmune disease, premature aging syndrome, Parkinson's syndrome, neurological disease, ischemia reperfusion injury, infectious disease, muscle disease, lung, kidney and liver disease.

In one embodiment, the cardiovascular disease is selected from the group comprising: myocardial infarction, cardiotoxicity (including cardiotoxicity of anthracyclines, cardiotoxicity of anticancer drugs, cardiotoxicity of quinolones and antiviral drugs, preferably anthracyclines), arterial pulmonary hypertension, heart failure, cardiopulmonary disease, ischemia, heart attack, stroke, thrombosis, and embolism.

In one embodiment, the compounds of the invention are used to prevent metastasis.

The invention also relates to compounds of formula (I

Or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, X, Y, R1、R2’、R3’、R4、R5And R6Is defined as follows;

in one embodiment, the compound of the invention is of formula (IIa), (IIb), (IIIa) or (IIIb), as defined below, or a pharmaceutically acceptable tautomer, salt or solvate thereof.

In one embodiment, the compound of the invention is a compound of formula (IIa-1), (IIa-2), (IIIa-1) or (IIIa-2), as defined below, or a pharmaceutically acceptable tautomer, salt or solvate thereof.

In one embodiment, the compounds of the invention are compounds of formula (IIa-1a), (IIa-1b), (IIa-1c), (IIa-1d), (IIa-1e), (IIa-2a), (IIa-2b), (IIa-2c), (IIa-2d), (IIa-2e), (IIIa-1a), (IIIa-1b), (IIIa-1c), (IIIa-1d), (IIIa-1e), (IIIa-2a), (IIIa-2b), (IIIa-2c), (IIIa-2d), or (IIIa-2e), or a pharmaceutically acceptable tautomer, salt, or solvate thereof, as defined below.

In one embodiment, the compounds of the present invention are compounds of formula (IIb-1), (IIb-2), (IIb-3), (IIb-4), (IIb-5), (IIIb-1), (IIIb-2), (IIIb-3), (IIIb-4) or (IIIb-5), as defined below, or a pharmaceutically acceptable tautomer, salt or solvate thereof.

The present invention also relates to a pharmaceutical composition comprising a compound of the present invention, or a pharmaceutically acceptable tautomer, salt, or solvate thereof, and at least one pharmaceutically acceptable excipient.

The invention also relates to a medicament comprising a compound of the invention or a pharmaceutically acceptable tautomer, salt, or solvate thereof.

The present invention also relates to a process for the preparation of a compound of formula (IIa-1), as defined below, or a pharmaceutically acceptable tautomer, salt or solvate thereof, characterized in that said process comprises:

a) cyclizing a compound of formula (c) with a sulfur-based reagent in the presence of a siloxane

Figure BDA0002261051970000051

Wherein, A, B, R1、R4、R5And R6Is defined as follows;

thereby obtaining a compound of formula (IIa-1'):

Figure BDA0002261051970000052

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, A, B, R1、R4、R5And R6Is defined as follows;

and optionally also,

b1) the compound of formula (IIa-1') may be reacted with an oxidizing agent; preferably, the oxidant is mercury acetate Hg (OAc)2(ii) a To obtain a compound of formula (IIa-1 "):

Figure BDA0002261051970000053

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, A, B, R1、R4、R5And R6Is defined as follows;

or

b2) The compound of formula (IIa-1') may be reacted with hydroxylamine NH in the presence of a base2OH-HCl reaction; preferably, the base is sodium acetate (AcONa); to obtain a compound of formula (IIa-1'):

Figure BDA0002261051970000054

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, A, B, R1、R4、R5And R6The definition is as follows.

Definition of

In the present invention, the following terms have the following meanings:

the term "about" preceding a digit means plus or minus 10% of the value of the digit.

The term "alkoxy" as used herein alone or as part of another substituent refers to the group-O-alkyl, wherein alkyl is as defined herein.

The term "alkyl" as used herein alone or as part of another substituent refers to the formula CnH2n+1Wherein n is a number of 1 or more. Typically, the alkyl groups of the present invention contain 1 to 8 carbon atoms, preferably 1 to 6 carbon atoms, more preferably 1 to 4 carbon atoms, more preferably 1 to 3 carbon atoms. The alkyl group may be linear or branched. Suitable alkyl groups include methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl and octyl.

The term "alkylamino" as used herein alone or as part of another substituent refers to the group-NH-alkyl, wherein alkyl is as defined herein.

The term "alkoxycarbonyl", as used herein alone or as part of another substituent, refers to the group-C (═ O) -O-alkyl, where alkyl is as defined herein. Preferably, the alkoxycarbonyl group is methoxycarbonyl.

The term "alkylsulfonyl" as used herein, alone or as part of another substituent, refers to the group-SO2-an alkyl group, wherein alkyl is as defined herein.

The term "amino" as used herein refers to the group-NH2

The term "aminoalkyl" as used herein, alone or as part of another substituent, refers to the group-alkyl-NH2Which isWherein alkyl is as defined herein.

The term "aminosulfonyl" as used herein alone or as part of another substituent refers to the group-SO2-NH2

The term "aryl" as used herein alone or as part of another substituent refers to a polyunsaturated aromatic hydrocarbon group having a single ring (i.e., phenyl) or multiple aromatic rings fused together (e.g., naphthyl), which typically contains 5 to 12 atoms; preferably 6 to 10 atoms. Non-limiting examples of aryl groups include phenyl, naphthylvinyl.

The term "bioisosteres" as used herein refers to compounds or groups that have approximately equal molecular shape and volume, approximately the same electron distribution, and that exhibit similar physical properties and similar biological activity.

The term "carboxy" as used herein refers to the group-COOH.

The term "carboxyalkyl" as used herein alone or as part of another substituent refers to the group-alkyl-COOH, wherein alkyl is as defined herein.

The term "halogen" refers to fluorine, chlorine, bromine or iodine.

The term "haloalkyl" as used herein alone or as part of another substituent refers to an alkyl group, as defined herein, wherein one or more hydrogens are replaced with a halogen, as defined herein. Non-limiting examples of haloalkyl groups include fluoromethyl, difluoromethyl, and trifluoromethyl.

The term "heteroaryl" as used herein alone or as part of another substituent refers to an aryl group as defined herein wherein at least one carbon atom is replaced by a heteroatom. In other words, it refers to an aromatic monocyclic ring of 5 to 12 carbon atoms or a ring system containing 2 rings fused together, usually containing 5 to 6 atoms; wherein one or more carbon atoms are substituted with oxygen, nitrogen and/or sulfur atoms, wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. Non-limiting examples of such heteroaryl groups include:

Figure BDA0002261051970000071

oxazolyl, thiazolyl, imidazolyl, furyl and pyrrolyl.

The term "IC50"or" maximum half inhibitory concentration "refers to the concentration of inhibitor required to inhibit 50% in vitro. Its "EC" with an agonist drug50"or" maximum half effective concentration "is equivalent. "EC 50" also indicates the plasma concentration required to obtain 50% of the maximal effect in vivo.

The term "nitroxyalkyl" as used herein, alone or as part of another substituent, refers to the group-alkyl-ONO2Wherein alkyl is as defined herein.

The expression "pharmaceutically acceptable excipient" refers to an excipient that does not produce negative, allergic or other adverse reactions when administered to an animal, preferably a human. It includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. For human administration, the formulations should meet sterility, thermogenicity, general safety and purity standards as required by regulatory authorities (e.g., FDA office or EMA).

The term "ROS" as used herein refers to reactive oxygen species. ROS are oxygen-containing chemically reactive chemicals. Examples include, but are not limited to, peroxides ([ O-O ]]2-And R-O-R, such as H2O2) Superoxide (O2.)-) Hydroxyl radical (. OH) and singlet oxygen ((OH))1O2). In cells, ROS are byproducts of oxygen metabolism. However, environmental stress can lead to increased cellular production of ROS, known as "oxidative stress", resulting in severe structural damage to the cell. To date, several different ROS production sites have been identified, including mitochondria (particularly site I of the mitochondrial respiratory chain)Q、IF、IIIQOSDH and mGPDH), microsomes (e.g., cytochrome P450 and diamine oxidase), peroxisomes, and some enzymes in the plasma membrane (e.g., NADPH oxidase and lipoxygenase). "cytoplasmic ROS" can be further distinguished according to the location of intracellular release and storage of ROS "And "mitochondrial ROS". For example, complex I (site I) of the mitochondrial respiratory chainQAnd IF) And site SDH of mitochondrial complex II produces and releases ROS to the mitochondrial cavity (lumen), and is therefore considered to be "mitochondrial ROS"; whereas complex III of the mitochondrial respiratory chain (site IIIQO) and site mGPDH produce and release ROS to the cell cytoplasm, are considered "cytoplasmic ROS".

The term "salts" of the compounds of the present invention is used herein to describe acid addition and base salts thereof. Suitable acid addition salts are formed from acids which form non-toxic salts. Non-limiting examples include acetate, trifluoroacetate, adipate, aspartate, benzoate, benzenesulfonate, bicarbonate/carbonate, bisulfate/sulfate, borate, tetrafluoroborate, camphorsulfonate, citrate, cyclamate, edisylate, ethanesulfonate, formate, fumarate, glucoheptonate, gluconate, glucuronate, hexafluorophosphate, salicylate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, methanesulfonate, methylsulfate, naphthoate, 2-naphthalenesulfonate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, dihydrogenphosphate, and the like, Pyroglutamate, saccharate, stearate, succinate, tannate, tartrate, tosylate, trifluoroacetate and xylonate (xinofoate). Suitable base salts are formed from bases which form non-toxic salts. Non-limiting examples include aluminum, arginine, benzathine, calcium, choline, diethylamine, glycolamine, glycinate, lysine, magnesium, meglumine, ethanolamine, potassium, sodium, tromethamine, 2- (diethylamino) ethanol, ethanolamine, morpholine, 4- (2-hydroxyethyl) morpholine and zinc. Hemisalts of acids and bases, such as hemisulfate and hemicalcium salts, may also be formed. Preferred pharmaceutically acceptable salts include hydrochloride/chloride, hydrobromide/bromide, bisulfate/sulfate, nitrate, citrate and acetate salts.

As herein describedAs used, the term "site IQ"refers to NADH: ubiquinone oxidoreductase (also known as mitochondrial complex I) binding site. Site IQROS are generated and released in the mitochondrial lumen.

As used herein, the term "site IF"refers to the flavin binding site of mitochondrial complex I. Site IFROS are produced and released in the mitochondrial lumen.

As used herein, the term "site IIIQO"refers to the ubiquinone binding site of the cytochrome bc1 complex (also known as mitochondrial complex III). Site IIIQOROS are produced and released into the cytoplasm.

As used herein, the term "site SDH" refers to succinate dehydrogenase (also known as mitochondrial complex II). The site SDH generates ROS, which are released in the mitochondrial lumen.

As used herein, the term "site mGPDH" refers to glycerol-3-phosphate dehydrogenase. The site mGPDH produces ROS, which are released into the cytoplasm.

As used herein, the term "solvate" is used to describe a compound of the invention comprising a stoichiometric or sub-stoichiometric amount of one or more pharmaceutically acceptable solvent molecules such as ethanol or water. The term "hydrate" refers to when the solvent is water.

The term "subject" refers to an animal, including a human. In the sense of the present invention, a subject may be a patient, i.e. a person receiving medical care, undergoing or having undergone medical treatment or being monitored for the development of a disease. In one embodiment, the subject is male. In another embodiment, the subject is a female.

The term "tautomer" refers to organic compounds that are interconvertible by a chemical reaction called tautomerization. The chemical reaction involves the migration of a hydrogen atom or proton, with the conversion of a single bond and an adjacent double bond.

The expression "therapeutically effective amount" refers to a level or amount of an agent that serves the following purpose without causing significant negative or adverse side effects to the target: (1) delaying or preventing the onset of a disease, condition, or condition associated with free oxygen radicals; (2) slowing or stopping the progression, exacerbation or worsening of one or more symptoms of the disease, condition or condition associated with free oxygen radicals; (3) ameliorating a disease, condition, or symptom of a condition associated with free oxygen radicals; (4) reducing the severity or incidence of a disease, condition, or condition associated with free oxygen radicals; or (5) cure of diseases, conditions, or conditions associated with free oxygen radicals. A therapeutically effective amount may be administered prior to the onset of a disease, condition, or condition associated with free oxygen radicals for prophylactic or preventative effects. Alternatively or additionally, a therapeutically effective amount may be administered after initiation of the disease, condition, or condition associated with free oxygen radicals for therapeutic effect.

The terms "treat," "treatment," or "ameliorating" refer to both therapeutic treatment and prophylactic or preventative measures; wherein the aim is to prevent or slow down targeting of a pathological condition or disease. Persons in need of treatment include those already with the disease as well as those prone to or to be prevented from the disease. A subject or mammal is successfully "treated" for the disease, infection, or condition if, after receiving treatment of the present invention, the subject or mammal exhibits an observable and/or measurable reduction or absence in one or more of the following: reducing the production of ROS; and/or relieve to some extent one or more symptoms associated with a particular disease or condition; reduced morbidity and mortality, and improved quality of life. The above parameters for assessing successful treatment and improvement of a disease can be readily measured by routine procedures familiar to physicians.

Drawings

FIG. 1 shows that AOL (5- (4-methoxyphenyl) -3H-1, 2-dithiole-3-thione) has no effect on mitochondrial respiration. FIG. A: after incubation in the presence of AOL (20 μ M in this example), mitochondria isolated from rat hearts oxidize glutamate + malate (GM) as substrate. Phosphorylation is triggered by Adenosine Diphosphate (ADP) and terminated by Atractyloside (ATR), a specific inhibitor of adenine transporter. Fig. B to D: classical studies of mitochondrial oxidative phosphorylation in the presence of various respiratory substrates were performed with increasing concentrations of AOL (5 μ M to 80 μ M). After addition of AOL, there was no statistical difference in mitochondrial respiration at different energy states. The oxidation rate after addition of ADP reflects the Adenosine Triphosphate (ATP) synthesis activity of the isolated mitochondria.

Figure 2 shows the major sites for oxygen radical production by isolated mitochondria in the presence of ATR (state 4), in the presence of substrates for both complexes I and III, to obtain maximal mitochondrial ROS production. As previously mentioned, mitochondrial ROS production is highly dependent on mitochondrial activity and conditions. Although AOL has been tested under a variety of conditions, for clarity we chose to provide only the most proven results here. The presence of all substrates that donate electrons to the entire chain (i.e. glutamate, malate, succinate) is closest to the in situ conditions in the cell. Under these substrate conditions, we evaluated the effect of the presence of AOL on ROS production by the intact chain under ATR (inhibition of phosphorylation: maximum yield) and by complex I (inhibition by rotenone) and complex II (inhibition by antimycin A). Color reference is made to fig. 3.

FIG. 3 shows the ROS/H of AOL (5. mu.M-80. mu.M) on isolated mitochondria in the presence of ATR (state 4) in the presence of substrates for complexes I and II2O2The resulting effects, rotenone, antimycin a and myxothiazole. In the absence of specific inhibitors of the complexUnder the conditions, ROS are produced most and mainly from site IQIs used (see fig. 4). By addition of inhibition IQAfter specific reverse electron-transport of rotenone, the yield decreases and almost exclusively occurs at site IIIQO. Subsequent addition of antimycin A, which prevents electron transfer to oxygen, increases site IIIQOAnd finally, the mythiazole prevents site III from being generatedQOROS production (see figure 2 for details).

FIG. 4 is a schematic diagram showing the sites of action of AOL on mitochondrial ROS production and the sites where AOL has no or little effect.

FIG. 5 is a histogram showing the effect of 10 μ M and 20 μ M AOL on glucose-stimulated insulin secretion (GSIS) in islets isolated from male C57Bl/6J mice. Combinations of three experiments are shown. Islets from two mice were used for each experiment; five islets per well; four to six wells are required for each condition. Insulin secretion data were normalized to the 11mM Glc-Veh group (considered 100%). P <0.05, p <0.01 and p <0.001 compared to 3mM Glc-Veh; # p <0.05, # # p <0.01, and # # # p <0.001, compared to 11mM Glc-Veh; one-way analysis of variance (ANOVA) and Bonferroni post hoc tests.

Fig. 6 is a histogram showing the amount of fat determined 3 weeks after treatment. The amount of fat is expressed in grams (g). Data are presented as mean ± SEM.

Figure 7 is a histogram showing the lean body mass measured 3 weeks after treatment. Lean body mass is expressed in grams (g). Data are presented as mean ± SEM.

FIG. 8 is a graph showing the effect of chronic treatment with AOL (5mg/kg and 10mg/kg) on glucose response during the insulin resistance test (ITT). The graph shows the change in blood glucose levels during ITT. Data are presented as mean ± SEM.

FIG. 9 is a histogram showing the effect of AOL on blood glucose levels. Five weeks after treatment, blood glucose was measured in mice fasted for 2 hours. Data are presented as mean ± SEM.

FIG. 10 is a histogram showing the neuroprotective effect of AOL (5mg/kg, bid, 11 days) on TH positive cell counts in SN in MPTP treated mice. Data are presented as mean ± SEM (n ═ 10-11) and analyzed using ANOVA and Dunnett (Dunnett) multiple comparison tests. P < 0.01; p <0.001c.f. mptp + vehicle (vehicle).

FIG. 11 is a graph showing the effect of AOL on cardiac contractile force recovery during the post-ischemic reperfusion phase. Data are presented as mean ± SEM of 6 independent experiments, control (black) and AOL treatment (grey).

Figure 12 is a graph showing the effect of AOL on infarct size of ischemic heart sections. At the end of the reperfusion period, the heart was stained with chlorotritetrazole (TTC). The living tissue appears red and the damaged tissue appears white.

Figure 13 is a set of two graphs showing the effect of AOL treatment on pulmonary artery pressure and cardiac remodeling. FIG. A: effect of AOL (shaded bars) on measured mean pulmonary arterial pressure (mPAP) in normoxic rats (N, white bars), chronic hypoxic rats (CH, light grey bars) and monocrotaline treated rats (MCT, dark grey bars). And B: right ventricular hypertrophy (i.e., the ratio of right ventricular weight (RV) to left ventricular plus septum weight (LV + S)) as expressed by the franton index. n is the number of rats. Each of the values represents a significant difference from N as P<0.05, 0.01 and 0.0001. # # indicates a significant difference in P compared to CH<0.05。

Figure BDA0002261051970000661

And

Figure BDA0002261051970000662

respectively representing a significant difference of P compared with N + AOL<0.05 and 0.01.

Figure BDA0002261051970000663

Representing a significant difference of P compared to MCT<0.05。

FIG. 14 is a set of graphs showing the effect of AOL on Pulmonary Artery (PA) remodeling. The effect of AOL (shaded bars) on PA remodeling was assessed by measuring the percentage of measured thickness within the PA in normoxic rats (N, white bars), chronic hypoxic rats (CH, light grey bars) and monocrotaline-treated rats (MCT, dark grey bars). To evaluateThe intraalveolar arteries observed for estimation of PA remodeling were divided into three groups with different cross-sectional diameters (panel A: below 50 μm; panel B: 50 μm-100 μm; panel C: 100 μm-150 μm). n is the number of vessels. Each of the values represents a significant difference from N as P<0.05, 0.01 and 0.0001. # # # and # # # indicate significant differences in P compared to CH<0.01 and 0.0001. The significant difference compared to N + AOL is P<0.05 and 0.01.

Figure BDA0002261051970000664

Representing a significant difference of P compared to MCT<0.05。

Fig. 15 is a set of graphs showing the effect of AOL on outer retinal nuclear layer (ONL) thickness in progressive light-induced retinal degeneration. FIG. A: effects of vehicle and AOL on "untransferred" animals. Animals were kept under low intensity cyclic illumination and received vehicle or AOL injections three times a day for 7 days. At 15 days after the end of the treatment, the retina was subjected to histological analysis. Data are expressed as mean ± SEM in μm of ONL thickness per 0.39mm in the superior and inferior poles from optic nerve and optic disc for untreated animals (light grey, square dots), vehicle treated animals (dark grey, triangular dots) and AOL treated animals (black, circular dots). And B: effects of vehicle and AOL on "transfer" animals. Animals were kept under low intensity cyclic illumination and transferred to high intensity cyclic illumination for 7 days during which the animals received 3 vehicle or AOL injections per day. At the end of the treatment, the animals were transferred back to low intensity cyclic lighting conditions and the retinas were histologically analyzed fifteen days later. Data are expressed as mean ± SEM in μm of ONL thickness per 0.39mm in the superior and inferior poles from optic nerve and optic disc for untreated animals (light grey, square dots), vehicle treated animals (dark grey, triangular dots) and AOL treated animals (black, circular dots).

FIG. 16 is a set of graphs showing lifetime SOD2-KO experiments for four groups of mice (WT-KOL: wild-type mice treated with vehicle; WT-AOL: wild-type mice treated with AOL; KO-KOL: SOD2-KO mice treated with vehicle; KO-AOL: SOD2-KO mice treated with AOL). Data are presented as mean values. FIG. A: mouse body weight (in grams) was varied over time (in days). And B: baseline correction for mouse body weight was as a percentage of body weight gain over time (in days). And (C) figure: survival (in percent) of the KO-KOL and KO-AOL groups over time (in days).

FIG. 17 is a graph showing Succinate Dehydrogenase (SDH) activity in the heart of five groups of mice (WT-KOL: wild-type mouse treated with vehicle; WT-AOL: wild-type mouse treated with AOL; KO-KOL: SOD2-KO mouse treated with vehicle; KO-AOL: SOD2-KO mouse treated with AOL; WT: wild-type untreated mouse). The optical density of SDH responses sampled from the cardiac section was measured with the Image analysis mkii (Akos) Image processing software. The density is expressed in the form of an average gray level, where average gray level is the sum of the grays/measured number of pixels. Data are presented as the average of the measured optical densities.

FIG. 18 is a set of graphs showing oil Red O (oil Red O) staining of liver sections of WT and SOD2-KO mice treated with AOL or without AOL. The histogram represents the mean size of the lipid droplets (panel a), the droplet density (number of droplets/liver area) (panel B) and the total lipid area (mean size × number of droplets) (panel C).

FIG. 19 shows the effect of AOX on mitochondrial oxidation. FIG. 19A shows a typical experiment showing mitochondrial respiration in the presence of AOX under various oxidative phosphorylation conditions. AOX was added to mitochondria and incubated for 5 minutes prior to oxidative phosphorylation assay. Oxygen consumption (dark grey traces) starts after addition of substrate (GM: glutamate-malate) and is activated by ADP (phosphorylation state). Phosphorylation is terminated by ATR (atractyloside) and low residual respiration reflects the integrity of the mitochondrial inner membrane. FIG. 19B shows the change in mitochondrial oxidation rate during the different respiratory states described above (Succ/ROT: GM substrate, succinate and rotenone) with increasing AOX concentration in DMSO.

FIG. 20 shows the effect of AOX on mitochondrial ATP phosphorylation. As described previously in Gouspillou et al (2014.Aging cell.13 (1): 39-48), both phosphorylation and oxidation rates have been determined and the effect of increased AOX concentrations has been reported. Results are expressed as relative percentage of control change relative to pH units/second (unit/sec).

Figure 21 shows the effect of increasing AOX concentration on oxygen radical production by isolated mitochondria in the presence of both complex I and III substrates (glutamate, malate and succinate), in the presence of ATR, thus under conditions predominantly produced by complex I. Measurement of mitochondrial generated ROS by use of the classical peroxidase-Amplex Red System, which measures H by oxidation of Amplex Red to produce fluorescent resorufin2O2Is present (see fig. 22B).

Figure 22 shows that AOX has no effect on non-mitochondrial ROS compared to AOL and Oltipraz in vitro assays. Obtaining non-mitochondrial ROS/H using commercially available NAD (P) H oxidases in the presence of reduced NAD (P) H2O2And then measured again using the classical peroxidase-Amplex Red system (fig. 22B). The effect of increasing concentration of different test molecules is shown in fig. 22A. The horizontal lines indicate the average percent of ROS production at all concentrations tested, AOL (99.2%) and AOX (109.9%).

FIG. 23 shows the effect of AOX on the viability of cancer cell lines A549 and H460, expressed as% relative to control (0 μ M AOX). Cells were incubated at increasing doses of 0 to 500 μ M AOX and cytotoxicity was assessed by sulforhodamine b (srb) assay (fig. 23A). The results are also expressed as the logarithm of the AOX concentration (FIG. 23B).

FIG. 24 shows that AOX and AOL have no effect on the respiration of H460 cells as demonstrated by electrography.

FIG. 25 shows the effect of AOX on metastatic activity of cancer cells using the Transwell assay. Briefly, H460 cancer cells are placed on top of a cell permeable membrane and after a period of incubation, cells that have migrated through the membrane are stained and counted. FIG. 25A shows photographs of cells stained in the lower compartment under different conditions (control, 5. mu.M AOX, 10. mu.M AOX and 10. mu.M NAC [ N-acetylcysteine ]). Fig. 25B is a histogram showing the result of fig. 25A.

FIG. 26 shows the contraction of the internal pulmonary arteries induced by 5-hydroxytryptamine (5HT) or endothelin (ET-1) in the presence of different concentrations of AOL (FIG. 26A) or AOX (FIG. 26B). Two-way ANOVA: p < 0.05; p < 0.01; p < 0.001.

FIG. 27 shows the proliferation of Pulmonary Artery Smooth Muscle Cells (PASMCs) following incubation in 10% Fetal Calf Serum (FCS), 0.2% FCS, and 0.2% FCS +100 μ M5-hydroxytryptamine (5HT), with or without AOL.

FIG. 28 shows the combination of succinate (energy substrate for respiratory complex 2) and known inhibitors of the respiratory chain when used (i.e. for site I)Q10mM succinate alone, for site IIIQ outer layerAOL, AOX and Otopraz (0 to 80. mu.M) targeting with 10mM succinate, 4. mu.M rotenone and 2.5. mu.M antimycin A), respectively, to isolated mitochondrial site IQAnd IIIQOROS/H of2O2The resulting effect. FIG. 28A shows these three molecular docking sites IQAnd FIG. 28B shows the effect on site IIIQOThe influence of (c).

FIG. 29 shows the combination of succinate (energy substrate for respiratory complex 2) and known inhibitors of the respiratory chain when used (i.e. for site I)Q10mM succinate alone, and for site IIIQOAOX analogue (Cp 1; cp 2; cp 3; cp 4; cp 5; cp6 a; cp 8; cp9a) (0 to 25. mu.M) site I on isolated mitochondriaQAnd IIIQOROS/H of2O2The resulting effect. FIG. 29A shows (upper panel) Cp1, Cp2, Cp3, Cp4, (lower panel) Cp5, Cp6a, Cp8 and Cp9A vs. site IQFIG. 29B shows Cp5, Cp6a and Cp9a vs. site IIIQOThe influence of (c).

Detailed Description

It is an object of the present invention to provide a method of treatment and/or prophylaxis; or for treating and/or preventing a disease associated with free oxygen radicals in a subject in need thereof, comprising administering an effective amount of an inhibitor of mitochondrial generation of Reactive Oxygen Species (ROS).

Another object of the invention is an inhibitor of mitochondrial generation of Reactive Oxygen Species (ROS) for use in therapy and/or prophylaxis; or for the treatment and/or prevention of diseases associated with free oxygen radicals, wherein the inhibitor inhibits mitochondrial production of ROS.

In one embodiment, the inhibitors of the invention do not affect physiological (cytosolic) ROS production. In one embodiment, physiological (cytosolic) ROS production is regulated by no more than 5% (either increased or decreased) in the presence of an inhibitor of the invention. In one embodiment, the modulation of physiological (cytosolic) ROS production in the presence of an inhibitor of the invention is no more than 2%, 4%, 6%, 8%, 10%, 12%, 14%, 16%, 18%, 20%, 25%, 30%, 35%, 40%, 45%, 50% (increased or decreased).

The term "does not affect" as used herein means that the inhibitors of the present invention do not function as measured by techniques known to those skilled in the art for determining the level of reactive oxygen species produced.

In another embodiment, the inhibitor of the invention is not an inhibitor of cytoplasmic ROS production.

In one embodiment, the inhibitors of the invention do not affect the activity of a compound selected from group IIIQOAnd physiological (cytosolic) ROS production at least one site of mGPDH.

In one embodiment, the inhibitors of the invention are not made in a mixture selected from group IIIQOAnd physiological (cytosolic) ROS production at least one site of mGPDH is reduced by greater than 2%, 4%, 6%, 8%, 10%, 12%, 14%, 16%, 18%, 20%, 25%, 30%, 35%, 40%, 45%, 50% or more.

In a preferred embodiment, the inhibitor of the invention does not reduce site III of complex III from the mitochondrial respiratory chainQOPhysiological (cytosolic) ROS production.

Cytoplasmic ROS production depends on the difference between total cellular ROS production and internal mitochondrial ROS production. Alternatively, cytosolic ROS production can be determined in an in vitro assay of nad (p) H oxidase ROS production.

In another embodiment, the inhibitors of the invention act upstream of ROS production.

Assays for detecting the production of cytoplasmic ROS are well known in the art and in the art.

Examples of such tests include, but are not limited to:

(1) measurement of total cellular reactive oxygen species ROS:

acetyl 5- (and-6) -chloromethyl-2 ', 7' -dichlorodihydrofluorescein diacetate (CM-H)2DCFDA) and/or H2DCFDA is an indicator of cytoplasmic Reactive Oxygen Species (ROS) in cells. CM-H2DCFDA passively diffuses into cells, its acetate group is cleaved by intracellular esterases, and its thiol-reactive chloromethyl group reacts with intracellular glutathione and other thiols. Subsequent oxidation produces fluorescent adducts that are trapped inside the cell, thereby facilitating long-term studies (Zhang et al, 2008.J. Cardiovasc. Pharmacol.51 (5): 443-9; Sarvazyan,1996.am. J. physiol.271(5 Pt 2): H2079-2085).

(2) Measurement of mitochondrial ROS production in cells:

measuring intracellular ROS in intact cells and determining the source of mitochondria is much more difficult. In recent years, the use of lipophilic triphenylphosphonium cation TPP+As a "delivery" conjugate, proton kinetics critical to mitochondrial function have been developed to target a variety of compounds to the highly negative mitochondrial matrix. Of these, MitoSOX red, also known as mitochondrial-ethidium hydride or mitochondrial-ethidium hydride, is commonly used for estimation of mitochondrial ROS. TPP of MitoSOX+ROS-sensitive ethidium hydride is partly allowed to accumulate in the mitochondrial matrix in large quantities and oxidized by superoxide to give the specific fluorescent oxidation product 2-hydroxyethidium (Zhao et al, 2005.Proc. Natl. Acad. Sci. USA.102 (16): 5727. sup. 5732; Polster et al, 2014.Methods Enzymol.547: 225. sup. 250).

In one embodiment, the inhibitors of the invention are selective inhibitors of mitochondrial production of Reactive Oxygen Species (ROS).

In one embodiment, the inhibitor of the present invention is in a compound selected from IQ、IFAnd a selective inhibitor of mitochondrial generation of ROS at least one site of the SDH.

In one embodiment, a wire according to the inventionSelective inhibitors of mitochondrial production selected from IQ、IFAnd SDH, while reducing mitochondrial ROS production at least one site by greater than 2%, 4%, 6%, 8%, 10%, 12%, 14%, 16%, 18%, 20%, 25%, 30%, 35%, 40%, 45%, 50% or more, while having less than 50%, 45%, 40%, 35%, 30%, 25%, 20%, 18%, 16%, 14%, 12%, 10%, 8%, 6%, 4%, 2% or less effect on mitochondrial ROS production at the remaining sites of ROS production.

In one embodiment, the inhibitor that is a selective inhibitor of mitochondrial ROS production from a single site of mitochondrial ROS production confers upon ROS production from site IQ、IFAnd SDH, while having less than 10% impact on ROS production from the remaining sites of ROS production.

In a preferred embodiment, the inhibitor of the invention is site I of complex I of the mitochondrial respiratory chainQAnd/or IFA selective inhibitor of mitochondrial generation of ROS.

Complex I of the mitochondrial respiratory chain can generate ROS from two different sites: a ubiquinone binding site and a flavin mononucleotide site.

Ubiquinone binding site of Complex I (I)Q)

In order to specifically analyze the gene from IQCan use 5mM succinate as substrate, providing electrons to the respiratory chain. I isQROS production at site is extremely sensitive to changes in proton dynamics (PMF) across the inner mitochondrial membrane (PMF ═ Δ Ψ m + Δ pH). Therefore, when in IQIn assessing the selectivity of hits (hits) in the ROS assay, a conservative threshold for the Δ Ψ m analysis may be used.

Reverse transport from the reduced Q cell to the substrate NAD through CI in the presence of strong PMF+From site IQThe leaked electrons are well characterized. From the experimental point of view, is in favor of IQThe conditions under which ROS are produced are considered to be physiologically far from each other, resulting in many people neglecting its relevance despite its high rate of capability. However, even when provided lowerAt both glutamate (for forward electron feed through CI) and succinate (for reverse electron feed), respiratory mitochondria still produce large amounts of rotenone-sensitive ROS (i.e., I)QROS). In addition, comparative analysis showed that the protein is derived from site IQ(instead of site I)F) Has an inverse relationship with the maximum ROS production of different vertebrate species (Lambert et al, 2007.Aging cell.6(5): 607-18; lambert et al, 2010.Aging cell.9(1): 78-91). Thus, IQSelective modulators of ROS provide unique opportunities to explore the putative role of mitochondrial ROS production in normal and pathological processes.

Flavin binding site of Complex I (I)F):

In order to specifically analyze the gene from IFThe substrate solution supplying electrons to the respiratory chain may comprise 5mM glutamate, 5mM malate and 4 μ M rotenone. Site IFThe rate of ROS production is directly proportional to the reduction status of the NADH pool in the mitochondrial matrix (Treberg et al, 2011.J.biol.chem.286(36): 31361-72). Blockade of site I with the insecticide rotenoneQEnhancement of the activity of the enzyme from site I by preventing oxidation of flavinFIs generated. Compared with site IQAnd IIIQOFrom Complex I (site I)F) The maximum ROS production of flavin binding sites of (a) is relatively low, which may lead to higher variability in the assay and subsequently to a higher false positive rate of hit finding (hit trapping) in the initial screen.

Rotenone and neurotoxin MPP+Inhibition of complex I activity was associated with parkinson's disease in both rodents and humans, suggesting a link between dysfunctional complex I, ROS production and neurodegeneration. Thus, compounds capable of inhibiting the production of ROS from complex I may be useful in therapy.

In another embodiment, the inhibitor or selective inhibitor of the invention is at site I of complex I of the mitochondrial respiratory chainQAnd (3) a selective inhibitor of mitochondrial generation of Reactive Oxygen Species (ROS) production.

The term "selectivity" as used hereinInhibitor "may refer to a compound capable of inhibiting site I in Complex IQAnd (c) ROS production at the same time as the compounds having minimal effects on ROS production at the remaining sites, mitochondrial membrane potential (Δ Ψ m), and oxidative phosphorylation. For example, on isolated mitochondria, when rotenone is present (i.e., when site I)QWhen ROS production is inhibited) and antimycin a (i.e., when ROS is produced primarily from complex III), the IC of the compound50The inhibition of ROS production is about 5, 6, 7, 8, 9, 10, 15, 20 times that in the absence of rotenone.

In one embodiment, the term "selective inhibitor" as used herein may also refer, exclusively or inclusively, to a compound capable of being at position I of complex I, in any one of the definitions given hereinQInhibit mitochondrial ROS production, IC thereof50From about 0.1. mu.M to about 20. mu.M, preferably about 10. mu.M. In one embodiment, the term "selective inhibitor" as used herein may also refer, exclusively or inclusively, to a compound capable of being at position I of complex I, in any one of the definitions given hereinQInhibit mitochondrial ROS production, IC thereof50About 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10.0, 10.5, 11.0, 11.5, 12.0, 12.5, 13.0, 13.5, 14.0, 14.5, 15.0, 15.5, 16.0, 16.5, 17.0, 17.5, 18.0, 18.5, 19.0, 19.5, 20 μ M. In another embodiment, the compound does not significantly inhibit cytosolic ROS production in an in vitro assay of nad (p) H oxidase ROS production.

The term "selective inhibitor" as used herein may also refer, exclusively or inclusively, to a compound capable of inhibiting I of complex I in any one of the definitions given hereinQROS production at the site with minimal effect on ROS production at the remaining sites and mitochondrial membrane potential (Δ Ψ m) and oxidative phosphorylation. For example, on isolated mitochondria, in the presence of rotenone (i.e., when site I)QWhen ROS production is inhibited), theIC for inhibiting ROS production by compound50About 5, 6, 7, 8, 9, 10, 15, 20 or more times that of antimycin a when present (i.e. added after rotenone, thus when ROS are mainly produced by complex III).

Site I of Complex I specifically detecting mitochondrial respiratory chain on isolated mitochondria from various tissuesQThe testing of generated ROS is well known to those skilled in the art.

High throughput assays are also described for identifying inhibitors of ROS production at defined sites in isolated mitochondria without altering energy production. This assay identifies site-specific modulators of ROS production, while also revealing less specific effectors such as broadly acting antioxidants and various inhibitors of mitochondrial bioenergetics. Thus, inhibitors can be identified that can distinguish between unwanted electrons leaking onto oxygen at specific locations in the electron transport chain (ROS production) without altering the normal energy-coupled electron and proton fluxes across the inner mitochondrial membrane. This assay adjusts standard assay methods for fluorescence-based mitochondrial ROS production using the dye Amplex UltraRed (Invitrogen) and Δ Ψ m using the potentiometric dye tmrm (Invitrogen) to a high-throughput microplate format. A core set of five ROS and one Δ Ψ m test is provided for reliably detecting functional modulation in just isolated skeletal muscle mitochondria. The five major sites of ROS production can be targeted separately by varying the substrate and the inhibitor added to the common assay mix (I)Q、IF、IIIQOSDH, and mGPDH). The counter screens used to monitor Δ Ψ m can be run in parallel to eliminate compounds that may be general inhibitors or uncouplers of normal mitochondrial energy production.

In another embodiment, the inhibitor is tested in duplicate at 2.5 μ M for all assays. Endpoint fluorescence was normalized to DMSO and known mitochondrial inhibitor control wells were included on each plate. The number of positive samples in each ROS assay may be initially filtered by applying a threshold in the assay that is preferably 15% lower, or more preferably 18% lower, or even more preferably 20% lower, or more lower. Each ROS assay can be used as a counter-screen relative to other ROS, while also eliminating compounds that alter Δ Ψ m in TMRM-based rescreening. Thus, the number of filtered samples can then be evaluated to eliminate those that change other ROS analyses by more than, for example, 20%, or 18%, or 15%, or change Δ Ψ m by more than, preferably, 10%, or more preferably, 5%, or even more preferably, 4%.

In another embodiment, the inhibitor of the invention does not significantly affect oxidative phosphorylation directly on mitochondria, preferably oxidative phosphorylation is modulated by less than 10%, 9%, 8%, 7%, 6%, 5%.

Diseases associated with free oxygen radicals involve an imbalance in oxidative stress and mitochondrial dysfunction. In particular, diseases involving mitochondrial dysfunction are caused by mitochondrial ROS production.

Diseases associated with free oxygen radicals include, but are not limited to: cardiovascular disease, aging disease, autoimmune disease, premature aging syndrome, Parkinson's syndrome, neurological disease, ischemia reperfusion injury, infectious disease, muscle disease, lung, kidney and liver disease.

Cardiovascular diseases associated with free oxygen radicals include, but are not limited to, hypertension, cardiotoxicity (including anthracycline cardiotoxicity, anticancer cardiotoxicity, quinolone cardiotoxicity, and antiviral cardiotoxicity), heart failure of whatever origin (heart failure regardless of origin), ischemia, myocardial infarction, heart attack, stroke, atherosclerosis, fibrillation, hypertension, thrombosis and embolism, allergic/inflammatory diseases such as bronchial asthma, rheumatoid arthritis, inflammatory bowel disease, type II diabetes, diabetes and deafness (DAD, also known as Barlinger-Wallace syndrome), inflammatory diseases, rheumatic fever, arterial pulmonary hypertension, complex cardiomyopathy (such as Bartt syndrome, Costello syndrome), Fredrichs (Friedreich) ataxia, LEOPARD (LEOPARD) syndrome, Noonan (Noonan) syndrome, cardiovascular system skin (cardiovascularis) syndrome, cardio-encephalomyopathy (cardioencephalyopathy) and alstonim (Alstrom) syndrome, innate immune response, and cardiopulmonary diseases (such as chronic obstructive pulmonary disease, pulmonary embolism, pericarditis, aortic stenosis, french tetrad, aortic stenosis, mitral valve stenosis, aortic valve regurgitation, mitral valve regurgitation, pneumoconiosis, bronchiectasis, cardiomyopathy, and/or endothelial nitroglycerin tolerance).

Aging disorders associated with free oxygen radicals include, but are not limited to: age-related macular degeneration (AMD), skin aging, ultraviolet damage to the skin, thinning, sagging, wrinkling, appearance of age spots, vascular rupture and dry areas, seborrheic keratosis, solar keratosis, Kindler's syndrome, Bowen's disease, skin cancer, arthritis, ankylosing spondylitis, inflammatory polyarthritis, gonarthritis, epidemic polyarthritis, psoriatic arthritis, cataracts, deafness, cancer, metastasis, prevention of metastatic processes, liver disease, transplantation, tumor and toxicity of anti-tumor or immunosuppressive agents and chemicals, osteoporosis, heteroderma, acrodermic prematurity, hereditary sclerosing heteroderma, congenital dyskeratosis, xeroderma pigmentosum, brunam (Bloom) syndrome, Fanconi anemia, Cockayne (Cockayne) syndrome, and pollution-induced disease.

Autoimmune diseases associated with free oxygen radicals include, but are not limited to: multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, type I diabetes, Crohn's (Crohn's) disease; myasthenia gravis, Grave's disease, scleroderma, Sjogren's syndrome, ulcerative colitis, primary biliary cirrhosis, autoimmune hepatitis, Hashimoto's thyroiditis, ankylosing spondylitis, psoriasis.

The autoimmune disease may be an autoimmune disease associated with a hematologic disease, such as autoimmune hemolytic anemia, pernicious anemia, and autoimmune thrombocytopenia.

Autoimmune diseases can also be temporal arteritis, antiphospholipid syndrome, vasculitis such as Wegener's granulomatosis and Behcet's disease.

Other autoimmune diseases include polymyositis, dermatomyositis, spondyloarthropathies (e.g., ankylosing spondylitis), antiphospholipid syndrome, and polymyositis.

Premature aging syndromes associated with free oxygen radicals include, but are not limited to: progeria, Bloom syndrome, Cockayne (Cockayne) syndrome, De brazil (De Barsy) syndrome, congenital dyskeratosis, restrictive skin diseases, rotmund-Thomson syndrome, low sulfur hair dystrophy, vorner (Werner) syndrome, vedmann-rautenstrach (Wiedemann-Rautenstrauch) syndrome and xeroderma pigmentosum.

Parkinsonism associated with free oxygen radicals includes, but is not limited to: parkinson's Disease (PD), progressive supranuclear palsy, multiple system atrophy, corticobasal degeneration or dementia with Lewy (Lewy) bodies, toxin-induced parkinson's disease, and early-onset variants of PD (such as parkinson's disease associated with autosomal recessive PARK6 or parkinson's disease associated with autosomal recessive PINK 1).

Neurological disorders associated with free oxygen radicals include, but are not limited to: dementia, Alzheimer's disease, Parkinson's disease and aging, Huntington's disease, Friedreich's ataxia, Wilson's disease, Leigh's syndrome, Kearns-Sayre syndrome, Leber hereditary optic neuropathy, cognitive disorders, mood disorders, movement disorders, tardive dyskinesia, brain injury, apoptosis, dementia, epilepsy, epileptic dementia, Alzheimer's disease, post-traumatic dementia, senile dementia, vascular dementia, HIV-1 related dementia, post-stroke dementia, schizophrenia, Down's syndrome, motor neuron disease, amyloidosis, type II diabetes-related amyloid protein, Kruez-Jakode-Jakob disease, Jazzer's necrosis, Trauberger's-Alkuler syndrome, Kuru and animal scrapie, amyloid associated with chronic hemodialysis, senile cardiac amyloid and familial amyloidosis polyneuropathy, encephalopathy, neurovisceral conditions (neuro-antichcnic disorder), memory loss, aluminum poisoning, decreased levels of iron in cells of living subjects, decreased levels of free transition metal ions in mammals, toxic amounts of metals in or in certain compartments of the body of a patient, multiple sclerosis, amyotrophic lateral sclerosis, motor agnosia, alcohol-related dementia, primary age-related proteinopathies, named alogens, agnosia, apraxia, speech agnosia, frontotemporal dementia, frontotemporal lobar degeneration, low-word (logapenic) progressive aphasia, neurofibrillary tangles, vocal agnosia, Pick's (Pick) disease, primary progressive aphasia, chronic non-fluent aphasia, chronic disorders, Semantic dementia, steroid dementia syndrome, toxic aphasia, secondary effects of aminoglycoside ototoxicity and ***e toxicity.

Ischemia reperfusion injury associated with free oxygen radicals includes, but is not limited to: stroke, cerebral ischemia, cerebral stem stroke syndrome, carotid endarterectomy, cerebellar stroke syndrome, central achromatopsia (cerebral achromatopsia), cerebral hemorrhage, cerebral infarction, cerebral venous sinus thrombosis, intraparenchymal hemorrhage, intracranial hemorrhage, lacunar stroke, lateral bulbar syndrome, lateral pontine syndrome, partial anterior infarction, posterior infarction, asymptomatic stroke (silent stroke), stroke-related (stroke Association), stroke in high-lift (stroke belt), stroke rehabilitation, transient ischemic attack, Watershed (Watershed) stroke, webber (Weber) syndrome, obesity, transplant preservation, ischemia reperfusion injury.

Infectious diseases associated with free oxygen radicals include, but are not limited to, hepatitis c, sepsis, infectious myopathy, and septic shock.

Muscle diseases associated with free oxygen radicals include, but are not limited to: myopathy, mitochondrial myopathy, facioscapulohumeral muscular dystrophy type 1, facioscapulohumeral muscular dystrophy type 2, Ryanodine (Ryanodine) receptor 1(RYR1) associated myopathy, selenoprotein 1(SEPN1) associated myopathy kaenssel-Sayre syndrome, cardiomyopathy, dyskinesia, immobilization-induced muscular dystrophy, skeletal muscle burn, and palmar aponeurotic contracture.

Lung, kidney and liver diseases associated with free oxygen radicals include, but are not limited to: cystic fibrosis, asthma, pollution-induced disease, cardiopulmonary disease, arterial pulmonary hypertension, chronic obstructive pulmonary disease, pulmonary embolism, pneumoconiosis, bronchiectasis, bronchial asthma, ventilator-induced diaphragm dysfunction, lung cancer, alcoholic fatty liver disease, diabetes, ex vivo kidney preservation, inflammation of the liver in hepatitis c, impaired renal function in type I diabetes, and cirrhosis of the liver.

In one embodiment, the diseases to be treated in particular in the present invention are age-related macular degeneration, parkinson's disease, alzheimer's disease, ischemia reperfusion injury, arterial pulmonary hypertension, scleroderma, atherosclerosis, heart failure, myocardial infarction, arthritis, pulmonary toxicity, cardiopulmonary disease, inflammatory diseases, cancer, metastasis, cardiotoxicity (including cardiotoxicity of anthracyclines, cardiotoxicity of anticancer drugs, cardiotoxicity of quinolones, and cardiotoxicity of antiviral drugs), heart failure regardless of origin, ischemia, heart attack, stroke, thrombosis and embolism, asthma, allergic/inflammatory diseases, bronchial asthma, rheumatoid arthritis, inflammatory bowel disease, Huntington's (huntton) chorea, cognitive disorders, presenile syndromes, epileptic dementia, presenile dementia, alzheimer's disease, inflammatory diseases, cancer, metastasis, cardiotoxicity (including cardiotoxicity of anthracyclines, cardiotoxicity of anticancer drugs, cardiotoxicity of quinolones, and cardiotoxicity of antiviral drugs), heart failure regardless of origin, ischemia, heart attack, Post-traumatic dementia, senile dementia, vascular dementia, HIV-1 associated dementia, post-stroke dementia, Down syndrome, motor neuron disease, amyloidosis, amyloid associated with type II diabetes, Creutzfeldt-Jakob disease, necrotic cell death, Gustmann-Straussler syndrome, Kuru and scrapie in animals, amyloid associated with chronic hemodialysis, senile cardiac amyloid and familial amyloidosis polyneuropathy, encephalopathy, neurovisceral conditions (neurospancnicdisc), memory loss, aluminum poisoning, decreased levels of iron in cells of living subjects, decreased levels of free transition metal ions in mammals, toxic amounts of metals in patients or certain body compartments, multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's disease, Alzheimer, Cataract, diabetes, cancer, liver disease, skin aging, transplantation, ototoxic secondary effects of aminoglycoside drugs, toxicity of tumors and anti-tumor or immunosuppressive agents and chemicals, innate immune response, and fradrichs (Friedreich) ataxia.

In one embodiment, the disease to be treated in particular in the present invention is a free oxygen radical related cardiovascular disease selected from the group consisting of: myocardial infarction, cardiotoxicity (including cardiotoxicity of anthracyclines, cardiotoxicity of anticancer drugs, cardiotoxicity of quinolones and antiviral drugs, preferably anthracyclines), arterial pulmonary hypertension, heart failure, cardiopulmonary disease, ischemia, heart attack, stroke, thrombosis, and embolism.

In one embodiment, the diseases to be treated in particular in the present invention are diseases caused by aging diseases, AMD, skin aging, cardiovascular diseases (e.g. cardiotoxicity of anthracyclines), presenile disorders and syndrome, parkinson's disease, alzheimer's disease, Friedreich's ataxia, ischemia-reperfusion injury, cardiopulmonary disease, asthma, cancer, metastasis and/or contamination.

In one embodiment, the disease to be specifically prevented and/or treated in the present invention is cardiotoxicity, preferably of anthracyclines. The mechanisms responsible for anthracycline toxicity refer to ROS production and site-specific DNA damage. Oxidative stress induces a profound role in the cardiotoxicity of anthracyclines by inducing DNA damage, sarcomere damage, mitochondrial dysfunction and loss of pro-survival signals, mediating both cardiomyocyte death and survival (Valcovici et al, 2016.arch.med.sci.12(2): 428-.

In one embodiment, the disease to be specifically prevented and/or treated in the present invention is pulmonary hypertension. Indeed, the deleterious effects of agents that promote ROS production on pulmonary blood vessels have been shown, whereas antioxidants have beneficial effects in animal models of pulmonary hypertension. Therefore, ROS production is directly related to all important features of pulmonary vascular remodeling, endothelial dysfunction, altered vasoconstrictive responses, inflammatory and extracellular matrix modification, pulmonary hypertension pathophysiology (Freund-Michelet al, 2013.Ther.adv.Respir.Dis.7(3): 175-200).

In one embodiment, the disease to be specifically prevented and/or treated in the present invention is ischemia reperfusion injury. Indeed, overproduction of ROS is a critical factor in the development of reperfusion injury (Granger et al, 2015.Redox biol.6: 524-51).

In one embodiment, the disease to be specifically prevented and/or treated in the present invention is diabetes. In fact, chronic hyperglycemia and subsequent rise in Reactive Oxygen Species (ROS) can disrupt cell function and increase insulin resistance, leading to exacerbation of type 2 diabetes (Kaneto et al 2010.mediators inflam., 2010:453892), but also other types of diabetes, such as MODY (juvenile adult-onset diabetes).

In one embodiment, the disease to be specifically prevented and/or treated in the present invention is parkinson's disease. Indeed, mitochondrial dysfunction and oxidative damage lead to increased ROS production, a condition that frequently occurs in the damaged brain regions of parkinson's disease: (et al.,2016.Parkinsons Dis.2016:7049108)。

In one embodiment, the disease to be specifically prevented and/or treated in the present invention is macular degeneration. In fact, excessive ROS production and accumulation, as well as oxidative stress, play a role in the pathogenesis of macular degeneration, particularly in retinal pigment epithelial cells. ROS levels increase in aging retinas, leading to oxidative stress and to damage of photoreceptors, retinal pigment epithelial cells and choroidal capillary layers during apoptosis (niti et al, 2016.oxid. med. cell. langev.2016: 3164734).

In one embodiment, the disease to be specifically prevented and/or treated in the present invention is scleroderma. In fact, NADPH oxidase has been shown to be an important source of ROS, up-regulated in scleroderma fibroblasts, leading to the accumulation of large amounts of ROS, which play a crucial role in cellular activity and DNA damage (Spadoni et al, 2015.Arthritis Rheumatotol.67 (6): 1611-22).

In one embodiment, one disease to be specifically prevented and/or treated in the present invention is metastasis. In fact, ROS production is associated with the mechanisms of tumor growth and metastasis (a mechanism termed "metastatic mitochondrial switch"): tumor Cell migration, invasion, clonogenic, metastatic uptake, spontaneous metastasis are promoted by natural selection of mitochondrial phenotypes associated with ROS production and aberrant TCA cycle activity (porporoto et al, 2014.Cell reports.8(3): 754-766). Overproduction of ROS also promotes angiogenesis, and conversely inhibitors of ROS production are anti-angiogenic products.

According to one embodiment, the inhibitor or selective inhibitor of the invention is AOX or a derivative thereof.

AOX corresponds to demethylanethotrithione, also known as 5- (4-hydroxyphenyl) -3H-1, 2-dithiole-3-thione (AOX):

Figure BDA0002261051970000211

according to one embodiment, the derivative of AOX is its bioisostere, preferably its phenolic bioisostere.

In one embodiment, the phenol bioisosteres are, for example, the following groups, tautomers and optionally substituted derivatives thereof:

in a particular embodiment, preferred phenol bioisosteres of the invention are those of the following, tautomers and optionally substituted derivatives thereof:

Figure BDA0002261051970000221

according to a preferred embodiment, the inhibitors of the invention are therefore compounds of formula (I)

Figure BDA0002261051970000222

Or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein:

x represents S, O or NHOH; preferably, X is S or O; more preferably, X is S;

y represents CH, C or N; preferably, Y is CH or N; more preferably, Y is CH;

R1、R2、R4and R5Each independently represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl;

R3is a hydroxyl group; or R3And R2Together with the carbon atom to which they are attached form a 5-membered heteroaryl moiety, wherein-R3-R2-represents-A-CR6or-B ═ CR6-a-; wherein:

a represents O, S or NR7(ii) a Wherein R is7Represents hydrogen, C1-C8 alkyl or alkoxycarbonyl;

b represents CH or N; and

R6represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl.

According to a preferred embodiment, in formula (I):

x represents S, O or NHOH; preferably, X is S;

y represents CH, C or N; preferably, Y is CH;

R1、R2、R4and R5Each independently represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfoneAcyl, cyano, nitro, carboxyl, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl; preferably, R1、R2、R4And R5Represents hydrogen.

R3Is a hydroxyl group; or R3And R2Together with the carbon atom to which they are attached form a 5-membered heteroaryl moiety, wherein-R3-R2-represents-A-CR6-B-; wherein

A represents O, S or NR7(ii) a Wherein R is7Represents hydrogen, C1-C8 represent alkyl;

b represents CH or N; and

R6represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl;

according to a preferred embodiment, X represents S. According to another preferred embodiment, X represents O. According to a preferred embodiment, Y represents CH. According to another preferred embodiment, Y represents N.

According to a preferred embodiment, R3And R2Together with the carbon atom to which they are attached form a 5-membered heteroaryl moiety, wherein-R3-R2-represents-A-CR6-B-; wherein:

a represents O, S or NR7(ii) a Wherein R is7Represents hydrogen, C1-C8 alkyl or alkoxycarbonyl; preferably, R7Represents hydrogen or alkoxycarbonyl;

b represents CH or N; and

R6represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl; preferably, R6Represents hydrogen or hydroxyl.

More preferably, -R3-R2-represents-O-c (oh) -N-or-N (coome) -CH-, more preferably-R3-R2-represents-O-c (oh) ═ N-.

According to another preferred embodiment, R3And R2Together with the carbon atom to which they are attached form a 5-membered heteroaryl moiety, wherein-R3-R2-represents-B ═ CR6-a-; wherein

A represents O, S or NR7(ii) a Wherein R is7Represents hydrogen, C1-C8 alkyl or alkoxycarbonyl; preferably, R7Represents hydrogen or alkoxycarbonyl;

b represents CH or N; and

R6represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl; preferably, R6Represents hydrogen or hydroxyl.

More preferably, -R3-R2-represents-N ═ c (oh) -S-.

According to a preferred embodiment, the inhibitors of the invention are therefore compounds of formula (I

Figure BDA0002261051970000241

Or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein:

x represents S, O or NHOH; preferably, X is S or O; more preferably, X is S;

y represents CH, C or N; preferably, Y is CH or N; more preferably, Y is CH;

R1、R4and R5Each independently represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl;

R2’and R3’Together with the carbon atom to which they are attached form a 5-membered heteroaryl moiety, wherein-R3’-R2’-represents-A-CR6or-B ═ CR6-a-; wherein:

a represents O, S or NR7(ii) a Wherein R is7Represents hydrogen, C1-C8 alkyl or alkoxycarbonyl;

b represents CH or N; and

R6represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl.

According to a preferred embodiment, the inhibitor of the invention is a compound of formula (II)

Figure BDA0002261051970000242

Or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein X, Y, R1、R4、R5、R6A and B are as defined for formula (I).

According to one embodiment, in formula (II):

x represents S, O or NHOH; preferably, X is S;

y represents CH, C or N; preferably, Y is CH;

a represents O, S or NR7(ii) a Wherein R is7Represents hydrogen or a C1-C8 alkyl group;

b represents CH or N;

R1、R4and R5Each independently represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl;

R6represents hydrogen, hydroxy, halogen, amino, alkylsulfonyl, aminosulfonyl, cyano, nitro, carboxy, aryl, alkoxy, haloalkyl, alkylamino, aminoalkyl, nitrooxyalkyl or carboxyalkyl.

According to a preferred embodiment, the compound of formula (II) is a compound of formula (IIa) or (IIb):

Figure BDA0002261051970000251

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein X, Y, A, B, R1、R4、R5And R6As defined above.

In a preferred embodiment, the inhibitor of the invention is a compound of formula (IIa).

According to a preferred embodiment, the compound of formula (IIa) is a compound of formula (IIa-1) or (IIa-2):

Figure BDA0002261051970000252

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, X, A, B, R1、R4、R5And R6As defined above.

In a preferred embodiment, the inhibitor of the invention is an inhibitor of formula (IIa-1). In another preferred embodiment, the inhibitor of the invention is an inhibitor of formula (IIa-2).

According to a preferred embodiment, the compound of formula (IIa-1) is a compound of formula (IIa-1 '), (IIa-1 ") or (IIa-1'"):

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, A, B, R1、R4、R5And R6As defined above.

In a preferred embodiment, the inhibitor of the invention is an inhibitor of formula (IIa-1 ') or (IIa-1 "), more preferably of formula (IIa-1').

According to a preferred embodiment, the compound of formula (IIa-2) is a compound of formula (IIa-2 '), (IIa-2 ") or (IIa-2'"):

Figure BDA0002261051970000262

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein A, B, R1、R4、R5And R6As defined above.

In a preferred embodiment, the inhibitor of the invention is an inhibitor of formula (IIa-2 ') or (IIa-2').

According to a preferred embodiment, the compounds of the formulae (IIa-1) and (IIa-2) have compounds of the formulae (IIa-1a), (IIa-1b), (IIa-1c), (IIa-1d), (IIa-1e), (IIa-2a), (IIa-2b), (IIa-2c), (IIa-2d) or (IIa-2 e):

Figure BDA0002261051970000271

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein X, R1、R4、R5、R6And R7As defined above.

In a preferred embodiment, the inhibitor of the invention is an inhibitor of formula (IIa-1a), (IIa-1b), (IIa-1e), (IIa-2a), (IIa-2b) or (IIa-2e), more preferably (IIa-1a) or (IIa-1 e); more preferred are inhibitors of formula (IIa-1 a).

According to a preferred embodiment, the compound of formula (IIb) is a compound of formulae (IIb '), (IIb ') and (IIb '):

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein A, B, R1、R4、R5And R6As defined above.

According to a preferred embodiment, the compound of formula (IIb) is a compound of formula (IIb-1), (IIb-2), (IIb-3), (IIb-4) or (IIb-5):

Figure BDA0002261051970000282

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein X, R1、R4、R5、R6And R7As defined above.

According to a preferred embodiment, the inhibitor of the invention is a compound of formula (III)

Figure BDA0002261051970000283

Or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein X, Y, R1、R4、R5、R6A and B are as defined for formula (I).

According to a preferred embodiment, the compound of formula (III) is a compound of formula (IIIa) or (IIIb)

Or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein X, Y, A, B, R1、R4、R5And R6As defined above.

In a preferred embodiment, the inhibitor of the invention is an inhibitor of formula (IIIa).

According to a preferred embodiment, the compound of formula (IIIa) is a compound of formula (IIIa-1) or (IIIa-2):

Figure BDA0002261051970000292

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein X, A, B, R1、R4、R5And R6As defined above.

In a preferred embodiment, the inhibitor of the invention is an inhibitor of formula (IIIa-1). In another preferred embodiment, the inhibitor of the present invention is an inhibitor of formula (IIIa-2).

According to a preferred embodiment, the compound of formula (IIIa-1) is a compound of (IIIa-1 '), (IIIa-1 ') or (IIIa-1 ')

Figure BDA0002261051970000293

Or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein A, B, R1、R4、R5And R6As defined above.

In a preferred embodiment, the inhibitor of the invention is an inhibitor of formula (IIIa-1 ') or (IIIa-1 "), more preferably of formula (IIIa-1').

According to a preferred embodiment, the compound of formula (IIIa-2) is a compound of (IIIa-2 '), (IIIa-2 ') or (IIIa-2 '):

Figure BDA0002261051970000301

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein A, B, R1、R4、R5And R6As defined above.

In a preferred embodiment, the inhibitor of the invention is an inhibitor of formula (IIIa-2 ') or (IIIa-2').

According to a preferred embodiment, the compounds of formulae (IIIa-1) and (IIIa-2) are compounds of formulae (IIIa-1a), (IIIa-1b), (IIIa-1c), (IIIa-1d), (IIIa-1e), (IIIa-2a), (IIIa-2b), (IIIa-2c), (IIIa-2d) or (IIIa-2 e):

Figure BDA0002261051970000302

Figure BDA0002261051970000311

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein X, R1、R4、R5、R6And R7As defined above.

In a preferred embodiment, the inhibitor of the invention is an inhibitor of formula (IIIa-1a), (IIIa-1b), (IIIa-1e), (IIIa-2a), (IIIa-2b) or (IIIa-2e), more preferably of formula (IIIa-1 b).

According to a preferred embodiment, the compound of formula (IIIb) is a compound of formulae (IIIb '), (IIIb ') and (IIIb ')

Figure BDA0002261051970000312

Or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein A, B, R1、R4、R5And R6As defined above.

According to a preferred embodiment, the compound of formula (IIIb) is a compound of formula (IIIb-1), (IIIb-2), (IIIb-3), (IIIb-4) or (IIIb-5):

Figure BDA0002261051970000321

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein X, R1、R4、R5、R6And R7As defined above.

According to a particular embodiment, the inhibitor of the invention is selected from:

5- (4-hydroxyphenyl) -3H-1, 2-dithiole-3-thione (AOX);

5- (4-hydroxyphenyl) -3H-1, 2-dithiolan-3-one (Cp 1);

5- (4-hydroxyphenyl) -3H-1, 2-dithiol-3-one oxime (Cp 2);

5- (4-hydroxyphenyl) -3H-1,2, 4-dithiazole-3-thione (Cp 3);

4- (4-hydroxyphenyl) -3H-1, 2-dithiole-3-thione (Cp 4);

5- (2-hydroxybenzo [ d ]]

Figure BDA0002261051970000322

Oxazol-5-yl) -3H-1, 2-dithiole-3-thione (Cp 5);

5- (2-hydroxybenzo [ d ] thiazol-6-yl) -3H-1, 2-dithiole-3-thione (Cp6 a);

5- (benzofuran-5-yl) -3H-1, 2-dithiole-3-thione (Cp 8); and

5- (3-thio-3H-1, 2-dithiolan-5-yl) -1H-indole-1-carboxylic acid methyl ester (Cp9 a).

According to a preferred embodiment, the inhibitor of the invention is selected from AOX, Cp1, Cp3, Cp4, Cp5, Cp6a and Cp9 a. According to a preferred embodiment, the inhibitor of the invention is selected from AOX, Cp1, Cp3, Cp5 and Cp6 a. According to a preferred embodiment, the inhibitor of the invention is AOX. According to a preferred embodiment, the inhibitor of the invention is Cp 1. According to a preferred embodiment, the inhibitor of the invention is Cp 3. According to a preferred embodiment, the inhibitor of the invention is Cp 5. According to a preferred embodiment, the inhibitor of the invention is Cp6 a.

The invention also relates to a process for the preparation of compounds of formula (IIa-1), more particularly compounds of formula (IIa-1 '), (IIa-1 ") and (IIa-1'") as defined above:

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, A, B, R1、R4、R5And R6As defined above;

the method comprises the following steps:

a) cyclizing a compound of formula (C) with a sulfur-based reagent in the presence of a siloxane,

Figure BDA0002261051970000332

wherein, A, B, R1、R4、R5And R6As defined above; preferably, the sulfur-based reagent is P4S10(ii) a Preferably, the siloxane is hexamethyldisiloxane (Me)3SiOSiMe3,HMDO);

Obtaining a compound of formula (IIa-1') as defined above;

and optionally also,

b1) the compound of formula (IIa-1') may be reacted with an oxidizing agent; preferably, the oxidant is mercury acetate Hg (OAc)2(ii) a To obtain a compound of formula (IIa-1') as defined above; or

b2) The compound of formula (IIa-1') may be reacted with hydroxylamine NH in the presence of a base2OH-HCl reaction; preferably, the base is sodium acetate (AcONa); to obtain a compound of formula (IIa-1') as defined above.

According to one embodiment, a process for the preparation of the compound of formula (IIa-1) may comprise a preliminary step of preparing intermediate (C) comprising a step of reaction between magnesium chloride (MgCl)2) Then reacting the acid derivative of formula (A) with 3-methoxy-3-oxopropanoic acid (B) in the presence of a diimidazolone and then HCl:

Figure BDA0002261051970000341

wherein, A, B, R1、R4、R5And R6As defined above;

Figure BDA0002261051970000342

thereby obtaining the compound of formula (C).

According to one embodiment, a process for the preparation of compounds of formula (IIa-1) may comprise a preliminary step for the preparation of intermediate (C), which comprises reacting an acid derivative of formula (a') with dimethyl carbonate in the presence of sodium hydride:

Figure BDA0002261051970000343

wherein, A, B, R1、R4、R5And R6As defined above;

thereby obtaining the compound of formula (C).

The invention also relates to the preparation of compounds of formula (IIa-2), more specifically of formulae (IIa-2 ') and (IIa-2'):

Figure BDA0002261051970000344

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein A, B, R1、R4、R5And R6As defined above;

the process comprises reacting a compound of formula (E) with carbon disulphide (CS) in the presence of a base2) The materials are subjected to ring-closing reaction,

Figure BDA0002261051970000351

a, B, R therein1、R4、R5And R6As defined above; preferably, the base is sodium hydride (NaH); thereby obtaining a compound of formula (IIa-2') as defined above;

and optionally, the compound of formula (IIa-2') may be reacted with hydroxylamine NH in the presence of a base2OH-HCl reaction; preferably, the base is sodium acetate (AcONa); obtaining the compound of formula (IIa-2') as defined above.

The invention also relates to a process for preparing a compound of formula (IIa-2'):

Figure BDA0002261051970000352

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein, A, B, R1、R4、R5And R6As defined above;

the process comprises reacting a compound of formula (E) with chlorocarbonyl sulfinyl chloride,

wherein, A, B, R1、R4、R5And R6As defined above;

thereby obtaining a compound of formula (IIa-2').

According to one embodiment, the intermediate (E) may be obtained by reacting the amide derivative (D) with Lawesson's reagent to obtain the compound of formula (E) as defined above

Figure BDA0002261051970000361

Wherein, A, B, R1、R4、R5And R6As defined above.

According to one embodiment, the amide intermediate (D) may be prepared by reacting the acid derivative of formula (a) with urea CO (NH) in the presence of a base2)2Carrying out reaction;

Figure BDA0002261051970000362

wherein, A, B, R1、R4、R5And R6As defined above;

preferably, the base is pyridine (C)5H5N); obtaining the compound of formula (D).

The present invention also relates to a process for the preparation of compounds of formula (IIb), more specifically compounds of formulae (IIb '), (IIb ') and (IIb '):

Figure BDA0002261051970000363

or a pharmaceutically acceptable tautomer, salt, or solvate thereof, wherein,A、B、R1、R4、R5and R6As defined above;

the method comprises the following steps:

a) cyclizing a compound of formula (G) with a sulfur-based reagent in the presence of Lawesson's reagent;

Figure BDA0002261051970000364

wherein, A, B, R1、R4、R5And R6As defined above;

preferably, the sulfur-based reagent is octasulfur S8(ii) a Obtaining a compound of formula (IIb') as defined above;

and optionally:

b1) the compound of formula (IIb') may be reacted with an oxidizing agent; preferably, the oxidant is mercury acetate Hg (OAc)2(ii) a To obtain a compound of formula (IIb ") as defined above; or

The compounds of the formula (IIb') may be reacted with hydroxylamine NH in the presence of a base2OH-HCl reaction; preferably, the base is sodium acetate (AcONa); to obtain a compound of formula (IIb' ") as defined above.

According to one embodiment, the process for preparing the compound of formula (IIb) may comprise a preliminary step of preparing intermediate (G) comprising reacting an ester derivative of formula (F) with a lewis acid in the presence of a base;

Figure BDA0002261051970000371

wherein, A, B, R1、R4、R5And R6As defined above;

preferably, the Lewis acid is TiCl4(ii) a Preferably, the base is triethylamine (Et)3N); obtaining the compound of formula (G) as defined above.

The invention also relates to a composition comprising, consisting of, or consisting essentially of an inhibitor or selective inhibitor of the invention.

The present invention also relates to a composition comprising, consisting of, or consisting essentially of the inhibitor described above for use in, or in the treatment of, a free oxygen radical related disease in a subject in need thereof.

The invention also relates to a composition for use in, or in the treatment of, a disease associated with free oxygen radicals, wherein the composition comprises, consists essentially of, or consists of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

The invention also relates to a pharmaceutical composition comprising, consisting of, or consisting essentially of an inhibitor or selective inhibitor of the invention and at least one pharmaceutically acceptable excipient.

The invention also relates to a pharmaceutical composition for use in, or comprising, or consisting essentially of, an inhibitor as described above and at least one pharmaceutically acceptable excipient for use in treating a free oxygen radical related disease in a subject in need thereof.

The invention also relates to a pharmaceutical composition for use in, or in the treatment of, a disease associated with free oxygen radicals, wherein the pharmaceutical composition comprises, consists of, or consists essentially of an inhibitor or selective inhibitor of mitochondrial generation of ROS and at least one pharmaceutically acceptable excipient.

Suitable excipients include water, saline, ringer's solution, dextrose solution, and ethanol, glucose, sucrose, dextran, mannose, mannitol, sorbitol, polyethylene glycol (PEG), phosphate, acetate, gelatin, collagen, carbomerVegetable oil solutions, and the like. Suitable excipients may also beSuitable preservatives, stabilizers, antioxidants, antimicrobials and buffers are additionally included, for example BHA, BHT, citric acid, ascorbic acid, tetracycline and the like.

Other examples of pharmaceutically acceptable excipients that may be used in the compositions of the present invention include, but are not limited to: ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (such as human serum albumin), buffer substances (such as phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate), polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene block polymers, polyethylene glycol and wool fat.

Additionally, some excipients may include surfactants (e.g., hydroxypropyl cellulose); suitable carriers, for example, solvents and dispersion media containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils, for example, peanut oil and sesame oil; isotonic agents, for example sugars or sodium chloride; coating agents, such as lecithin; agents that delay absorption, such as aluminum monostearate and gelatin; preservatives, such as benzalkonium chloride, benzethonium chloride, chlorobutanol, thimerosal, and the like; buffers such as boric acid, sodium bicarbonate, potassium bicarbonate, sodium borate, potassium borate, sodium carbonate, potassium carbonate, sodium acetate, sodium dihydrogen phosphate, and the like; tonicity agents, such as dextran 40, dextran 70, dextrose, glycerol, potassium chloride, propylene glycol, sodium chloride; antioxidants and stabilizers such as sodium bisulfite, sodium metabisulfite, sodium thiosulfite, thiourea and the like; non-ionic wetting or clarifying agents such as polysorbate 80, polysorbate 20, poloxamer 282, and tyloxapol; viscosity modifiers such as dextran 40, dextran 70, gelatin, glycerin, hydroxyethyl cellulose, hydroxymethyl propyl cellulose, lanolin, methyl cellulose, petrolatum, polyethylene glycol, polyvinyl alcohol, polyvinyl pyrrolidone, carboxymethyl cellulose; and so on.

Other examples of pharmaceutically acceptable excipients are Cyclodextrins (CD) and derivatives thereof. Cyclodextrins may improve the dissolution and/or stability of the active ingredient. Preferably, the cyclodextrin is β -cyclodextrin. In one embodiment, the cyclodextrin is selected from SBE-cyclodextrin (SBE: sulfobutyl ether) and HP-cyclodextrin (HP: hydroxypropyl) or derivatives thereof. In one embodiment, the cyclodextrin is SBE-cyclodextrin, preferably SBE- β -cyclodextrin. In one embodiment, the cyclodextrin is HP-cyclodextrin, preferably HP- β -cyclodextrin.

The invention also relates to a medicament comprising, consisting of, or consisting essentially of an inhibitor or selective inhibitor of the invention.

The invention also relates to a medicament comprising, consisting of, or consisting essentially of an inhibitor as described above for use in, or in the treatment of, a free oxygen radical related disease in a subject in need thereof.

The invention also relates to a medicament for use in, or in the treatment of, a disease associated with free oxygen radicals, wherein the medicament comprises, consists essentially of or consists of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

The invention also relates to a cosmetic composition comprising the inhibitor of the invention.

The invention also relates to a cosmeceutical composition comprising the inhibitor of the invention.

Another object of the invention is a preservation medium or storage medium comprising, consisting of, or consisting essentially of an inhibitor of the invention.

In one embodiment, the preservation medium is used to store organs, biological tissues, and/or living cells. In one embodiment, the organ includes, but is not limited to, heart, liver, kidney, lung, pancreas, intestine, skin, and cornea. In one embodiment, the organ is used for transplantation, i.e., the transfer of any organ or body tissue from its point of orthotopic location to a recipient site. Specifically, during an allograft transplant, the original site of the transplanted organ is in the donor individual and the recipient site is in another recipient individual.

In one embodiment, the preservation medium comprises an inhibitor of the invention at a concentration in the range of 0.1. mu.M to 120. mu.M, i.e., at a concentration of about 0.1. mu.M, 0.5. mu.M, 1. mu.M, 2. mu.M, 3. mu.M, 4. mu.M, 5. mu.M, 10. mu.M, 15. mu.M, 20. mu.M, 30. mu.M, 40. mu.M, 50. mu.M, 75. mu.M, 100. mu.M or 120. mu.M.

In one embodiment, the composition, pharmaceutical, cosmetic composition or cosmeceutical composition of the present invention will be administered systemically or topically.

In one embodiment, the composition, pharmaceutical composition, medicament, cosmetic composition or cosmeceutical composition of the present invention will be administered orally, by injection, topically, nasally, buccally, rectally, vaginally, intratracheally, endoscopically, transmucosally and transdermally.

In one embodiment, the composition, pharmaceutical, cosmetic or cosmeceutical composition of the invention is injected, preferably systemically. Examples of formulations suitable for systemic injection include, but are not limited to, liquid solutions, or suspensions, solid forms suitable for dissolution or suspension in a liquid prior to injection. Examples of systemic injections include, but are not limited to, intravenous, subcutaneous, intramuscular, intradermal, and intraperitoneal injections, and infusion. In another embodiment, the composition, pharmaceutical, cosmetic composition or cosmeceutical composition of the present invention is sterile when injected. Methods for obtaining sterile pharmaceutical compositions include, but are not limited to, GMP synthesis (GMP stands for "good manufacturing practice").

In another embodiment, the composition, pharmaceutical, cosmetic composition, or cosmeceutical composition of the present invention is administered orally. Examples of formulations suitable for oral administration include, but are not limited to, solid forms, liquid forms, and gels. Examples of solid forms suitable for oral administration include, but are not limited to, pills, tablets, capsules, soft gelatin capsules, hard gelatin capsules, caplets, compressed tablets, cachets, wafers, dragees, sugar coated tablets, or dispersible and/or disintegrating tablets, powders, solid forms suitable for dissolution or suspension in a liquid prior to oral administration, and effervescent tablets. Examples of liquid forms suitable for oral administration include, but are not limited to, solutions, suspensions, drinkable solutions, elixirs (elixirs), sealed vials, potions (positions), drenches (drench), syrups, and liquids (liquor).

In another embodiment, the composition, pharmaceutical, cosmetic composition, or cosmeceutical composition of the present invention is administered topically. Examples of formulations suitable for topical application include, but are not limited to, sticks, lipsticks, waxes, creams, lotions, ointments, salves (balms), gels, lip gloss (gloss), sunscreen preparations, cosmetics, facial masks, leave-on lotions or cleansers, depilatories, and the like.

Topical application characterizes the direct application or application of the composition, pharmaceutical, cosmetic, or cosmeceutical composition of the present invention to a site of interest to produce an effect locally (typically on one or more exposed or outer surfaces thereof, e.g., the outermost layer of the epidermis exposed and visually visible), such as using the hand, fingers, or various cosmetic tools (roll-up, roll-on or other stick containers, tubular containers, cotton balls, powder puffs, cotton swabs, pumps, brushes, pads, cloths, etc.). Application may be by, for example, applying, placing, rubbing, wiping, pouring, spreading and/or massaging into or onto the skin, or by any other convenient or suitable method. Preferably, topical administration is effective without any significant absorption of components of the composition by the subject's blood stream (to avoid systemic effects).

The composition, pharmaceutical, cosmetic, or cosmeceutical composition of the present invention may be mixed with, for example, 1% or 2% (w/w) benzyl alcohol, emulsifying wax, glycerin, isopropyl palmitate, lactic acid, purified water, and sorbitol solution as a preservative to form a white, smooth, uniform, opaque cream or lotion. Additionally, the composition may comprise polyethylene glycol 400(PEG 400). They may be mixed with, for example, 2% (w/w) benzyl alcohol, white petrolatum, emulsifying wax and tenox II (butyl hydroxyanisole, propyl gallate, citric acid, propylene glycol) as preservatives to form an ointment. The woven pad or roll of bandage material, such as gauze, may be impregnated with a solution, lotion, cream, ointment, or other such form of composition that can be used for topical application.

In another embodiment, the composition, pharmaceutical, cosmetic, or cosmeceutical composition of the present invention may also be topically applied using a transdermal system, such as one of the acrylic polymer-based adhesives with a resin cross-linker impregnated with the composition and laminated to an impermeable backing.

In one embodiment, the composition, pharmaceutical composition, drug, cosmetic composition, or cosmeceutical composition of the present invention may be administered as a transdermal patch, more particularly as a sustained release transdermal patch. Transdermal patches may include any conventional form, such as an adhesive matrix, a polymer matrix, a reservoir patch, a matrix or a monolithic laminate structure, and are typically composed of one or more backing layers, an adhesive, a permeation enhancer, an optional rate controlling membrane, and a release liner that is removed prior to application to expose the adhesive. The polymer matrix patch further comprises a polymer matrix-forming material. Suitable transdermal patches are described in more detail in, for example, U.S. patent nos. 5,262,165, 5,948,433, 6,010,715 and 6,071,531, the disclosure of each of which is incorporated herein in its entirety.

Examples of formulations suitable for transdermal administration include, but are not limited to, ointments, pastes, creams, films, balms, patches (e.g., transdermal patches), gels, liposome forms, and the like.

In one embodiment, the transdermal composition is an ointment, paste, cream; film, balm, patch (e.g., transdermal patch), gel, liposome form, and the like.

In one embodiment of the invention, the ointment is an oily ointment; emulsifying ointments, e.g., oil-in-water or water-in-oil ointments; or water-soluble ointment, preferably oily ointment.

In one embodiment of the present invention, the oily ointment employs a base such as vegetable and animal oils; animal and vegetable fats; a wax; petrolatum, such as white petrolatum or petrolatum; and paraffin wax, such as liquid paraffin or paraffin oil.

In one embodiment of the invention, the transdermal composition further comprises one or more excipients. Suitable pharmaceutically acceptable excipients are well known to the skilled person. Examples of suitable excipients include, but are not limited to, carriers, emulsifiers, hardeners, rheology modifiers or thickeners, surfactants, emollients, preservatives, humectants, buffers, solvents, humectants, and stabilizers.

In another embodiment, a particular route of administration may be intraocular administration. In another embodiment, the route of administration may be topical ocular administration, such as administration of eye drops or by washing the eye in an ophthalmic solution comprising the inhibitor of the present invention.

By ophthalmic solution is meant a sterile liquid, semi-solid or solid formulation intended for administration on the eyeball and/or conjunctiva, or for insertion in the conjunctival sac or for administration at the posterior segment of the eye. As used herein, the term "posterior segment of the eye" refers to the posterior two-thirds of the eye, including the anterior vitreous membrane and structures behind it (vitreous humor, retina, choroid, optic nerve). In particular, the ophthalmic composition may be administered intravitreally, for example by intravitreal injection. Examples of ophthalmic compositions include, but are not limited to, eye drops, eye lotions, powders for eye drops and powders for eye lotions, and compositions injected into the conjunctival sac or vitreous body.

Examples of carriers include, but are not limited to, water; buffered saline; mineral oil (petrolatum, also known as white soft paraffin); petrolatum; oils, such as mineral oil, vegetable oil, animal oil, paraffin oil, castor oil or vaseline oil; organic and inorganic waxes such as microcrystalline wax, paraffin wax, beeswax and ozokerite; natural polymers, such as xanthan gum (xanthanes), gelatin, cellulose, collagen, starch or gum arabic; synthesizing a polymer; alcohols; polyols, and the like. In one embodiment of the invention, the carrier is a base cream comprising an emulsifier, an oil phase component and an aqueous phase component.

Examples of well known base excipients for ointments or lotions include, but are not limited to, petrolatum, PlastibaseTM(this is a matrix prepared from polyethylene (average molecular weight about 21000Da) and liquid paraffin) and ESMA-PTM(made from microcrystalline wax).

Examples of emulsifiers include, but are not limited to, cetyl alcohol, cetearyl alcohol, stearyl alcohol, carboxylated polyethylene, polycarbophil, polyethylene glycol and derivatives thereof, polyoxyethylene and derivatives thereof (e.g., polysorbate 20 or polysorbate 80), alone or in combination with fatty alcohols (e.g., cetyl alcohol, stearyl alcohol and cetearyl alcohol) and sorbitan esters (e.g., sorbitan fatty acid esters).

Examples of oil phase components include, but are not limited to, petrolatum, such as white petrolatum, yellow petrolatum, or petrolatum oil; paraffin wax, such as liquid paraffin or paraffin oil; dimethicone and mixtures thereof.

Examples of aqueous phase components include, but are not limited to, water, glycerol, and propylene glycol.

Examples of sclerosing agents include, but are not limited to, stearyl alcohol, cetearyl alcohol, and cetyl alcohol.

Examples of rheology modifiers or thickeners include, but are not limited to, carbomers (e.g., carbomers)

Figure BDA0002261051970000431

) And polyoxyethylene tallow amine (e.g., polyoxyethylene tallow amine)

Figure BDA0002261051970000432

)。

Examples of surfactants include, but are not limited to, anionic, cationic, amphoteric, and nonionic surfactants, such as sodium lauryl sulfate, cetearyl alcohol, cetyl alcohol, magnesium lauryl sulfate, waxes, or combinations thereof.

Examples of emollients include, but are not limited to, white or yellow petrolatum (white or yellow petrolatum), liquid petrolatum (liquid petrolatum), paraffin, or aquaphor.

Examples of preservatives include, but are not limited to, antimicrobial preservatives, such as paraben (methyl hydroxybenzoate), paraben (hydroxybenzoate), butyl paraben, ethyl paraben, methyl paraben, potassium propylparaben, sodium propylparaben; parabens; sorbic acid; potassium sorbate; benzoic acid; parabens; chlorobutanol; phenol; thimerosal; sodium benzoate and benzyl alcohol.

Examples of humectants include, but are not limited to, propylene glycol and propylene glycol alginate.

Examples of buffering agents include, but are not limited to, sodium hydroxide, citric acid, and potassium hydroxide.

Examples of solvents include, but are not limited to, water, isopropanol, benzyl alcohol, and propylene glycol.

Examples of humectants include, but are not limited to, glycerin, mineral oil, polyoxyethylene hardened castor oil and petrolatum, propylene glycol; paraffin wax; waxes, such as beeswax; polyethylene glycol or mixtures thereof, such as macrogol (macrogol is a mixture of polyethylene glycols of different molecular weights); stearyl alcohol; benzyl alcohol; parabens (parabens); a gelled hydrocarbon; citric acid; squalene; lanolin; glycerol; polyoxyethylene hardened castor oil; sorbitan fatty acid esters; glycerin fatty acid ester; animal and vegetable fats; an oil; starch; gum tragacanth; a cellulose derivative; a silicone; bentonite; silicic acid; bentonite; zinc oxide and mixtures thereof.

Examples of stabilizers include, but are not limited to, carbohydrates such as sucrose, lactose, and trehalose; sugar alcohols such as mannitol and sorbitol; amino acids such as histidine, glycine, phenylalanine and arginine.

In one embodiment of the present invention, the composition, pharmaceutical composition, drug, cosmetic composition, or cosmeceutical composition of the present invention may be used in combination with a delivery system that facilitates delivery of the agent to the central nervous system. For example, various permeability enhancers of the Blood Brain Barrier (BBB) can be used to transiently and reversibly increase the permeability of the blood brain barrier to therapeutic agents. Such permeability enhancers of the BBB include, but are not limited to, leukotrienes, bradykinin agonists, histamine, tight junction disrupters (e.g., zonulin, zot), hypertonic solutions (e.g., mannitol), cytoskeletal contractiles, and short chain alkyl glycerols (e.g., 1-O-pentylglycerol). Active agents can be delivered to the central nervous system by oral, sublingual, parenteral, transplantation, nasal and inhalation routes. In some embodiments, the compounds of the present invention may be administered to the central nervous system in a manner that has minimal impact on the peripheral nervous system.

The Blood Brain Barrier (BBB) is a system of physical barriers and cellular transport mechanisms between blood vessels in the Central Nervous System (CNS) and most areas of the CNS itself. The BBB maintains homeostasis by limiting the entry of potentially harmful chemicals in the blood and allowing the entry of essential nutrients. However, the BBB can be a powerful obstacle to delivering pharmacological agents to the CNS to treat disorders or to maintain or enhance normal and desirable brain functions, such as cognition, learning, and memory.

In one embodiment, the inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition is administered in a sustained release form. In another embodiment, the composition, pharmaceutical composition, or medicament comprises a delivery system that controls the release of the modulating agent.

In one embodiment, the inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition of the present invention is administered at a dosage determined by the skilled artisan and appropriate for each subject.

It is to be understood that the total daily amount of the inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition of the present invention will be determined by the attending physician within the scope of sound medical judgment. The specific therapeutically effective amount for any particular patient will depend upon a variety of factors including the disease being treated and the severity of the disease; the particular composition used, the age, weight, general health, sex, and diet of the subject; time of administration, route of administration, duration of treatment; a drug combined or used simultaneously with the inhibitor, the composition, the pharmaceutical composition, the drug, the cosmetic composition, or the cosmeceutical composition of the present invention; and factors well known in the medical arts. For example, it is within the skill of the art to start with a dosage level of the therapeutic compound that is lower than the dosage required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved; but conversely, starting with a loading dose (which is a way to achieve steady state plasma concentrations more quickly), and then administering a calculated maintenance dose to exactly compensate for the effect of the elimination, can be equally useful.

In one embodiment, a therapeutically effective amount of an inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition of the present invention is administered at least once daily, twice daily, at least three times daily.

In another embodiment, a therapeutically effective amount of an inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition of the present invention is administered every two, three, four, five, six days.

In another embodiment, a therapeutically effective amount of an inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition of the present invention is administered twice weekly, biweekly, monthly.

In one embodiment of the invention, the daily amount of inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition to be administered to a subject is from about 1 μ g/day to about 100 mg/day, from about 1 μ g/day to about 50 mg/day, from about 1 μ g/day to about 10 mg/day, from about 1 μ g/day to about 9 mg/day, from about 1 μ g/day to about 8 mg/day, from about 1 μ g/day to about 7 mg/day, from about 1 μ g/day to about 6 mg/day, from about 1 μ g/day to about 5 mg/day, from about 1 μ g/day to about 4 mg/day, from about 1 μ g/day to about 3 mg/day, from about 1 μ g/day to about 2 mg/day, or, About 1. mu.g/day to about 1 mg/day, about 1. mu.g/day to about 100. mu.g/day.

In one embodiment of the invention, the daily amount of the inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition to be administered to a subject is from about 1 μ g/day to about 10 mg/day, from about 5 μ g/day to about 10 mg/day, from about 10 μ g/day to about 7.5 mg/day, about 10. mu.g/day to about 5 mg/day, about 10. mu.g/day to about 2.5 mg/day, about 10. mu.g/day to about 2 mg/day, about 10. mu.g/day to about 1 mg/day, about 10. mu.g/day to about 0.75 mg/day, about 10. mu.g/day to about 1 mg/day, about 10. mu.g/day to about 0.5 mg/day, from about 10. mu.g/day to about 0.25 mg/day.

In one embodiment of the invention, the daily amount of inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition to be administered to a subject is from about 0.1 mg/day to about 2000 mg/day, from about 0.1 mg/day to about 1500 mg/day, from about 0.1 mg/day to about 1000 mg/day, from about 0.1 mg/day to about 500 mg/day, from about 0.1 mg/day to about 200 mg/day, from about 0.5 mg/day to about 2000 mg/day, from about 0.5 mg/day to about 1500 mg/day, from about 0.5 mg/day to about 1000 mg/day, from about 0.5 mg/day to about 500 mg/day, from about 0.5 mg/day to about 200 mg/day, from about 1 mg/day to about 2000 mg/day, from about 1 mg/day to about 1500 mg/day, or a combination thereof, From about 1 mg/day to about 1000 mg/day, from about 1 mg/day to about 500 mg/day, from about 1 mg/day to about 200 mg/day.

In one embodiment of the invention, the daily amount of inhibitor, composition, pharmaceutical, cosmetic, or cosmeceutical composition to be administered to a subject is about 1 μ g/day, about 2 μ g/day, about 4 μ g/day, about 6 μ g/day, about 8 μ g/day, about 10 μ g/day, about 15 μ g/day, about 20 μ g/day, about 25 μ g/day, about 30 μ g/day, about 35 μ g/day, about 40 μ g/day, about 45 μ g/day, about 50 μ g/day, about 55 μ g/day, about 60 μ g/day, about 65 μ g/day, about 70 μ g/day, about 75 μ g/day, about 80 μ g/day, about 85 μ g/day, about 90 μ g/day, about 95 μ g/day, about 65 μ g/day, or about, About 100. mu.g/day, about 150. mu.g/day, about 200. mu.g/day, about 250. mu.g/day, about 300. mu.g/day, about 350. mu.g/day, about 400. mu.g/day, about 450. mu.g/day, about 500. mu.g/day.

In one embodiment of the invention, the daily amount of the inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition to be administered to a subject is about 0.1 mg/day, 0.2 mg/day, 0.3 mg/day, 0.4 mg/day, 0.5 mg/day, 0.6 mg/day, 0.7 mg/day, 0.8 mg/day, 0.9 mg/day, 1 mg/day, 2 mg/day, 4 mg/day, 6 mg/day, 8 mg/day, 10 mg/day, 15 mg/day, 20 mg/day, 25 mg/day, 30 mg/day, 35 mg/day, 40 mg/day, 45 mg/day, 50 mg/day, 75 mg/day, 100 mg/day, 125 mg/day, 150 mg/day, 175 mg/day, 200 mg/day, or cosmeceutical composition, 300 mg/day, 400 mg/day, 500 mg/day, 600 mg/day, 700 mg/day, 800 mg/day, 900 mg/day, 1000 mg/day, 1200 mg/day, 1400 mg/day, 1600 mg/day, 1800 mg/day, 2000 mg/day.

In one embodiment of the invention, the daily amount of inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition to be administered to a subject is from about 0.1 μ g/kg/day to about 10 mg/kg/day, from about 0.1 μ g/kg/day to about 5 mg/kg/day, from about 0.1 μ g/kg/day to about 1 mg/kg/day, from about 0.1 μ g/kg/day to about 0.9 mg/kg/day, from about 0.1 μ g/kg/day to about 0.8 mg/kg/day, from about 0.1 μ g/kg/day to about 0.7 mg/kg/day, from about 0.1 μ g/kg/day to about 0.6 mg/kg/day, from about 0.1 μ g/kg/day to about 0.5 mg/kg/day, or, About 0.1 μ g/kg/day to about 0.4 mg/kg/day, about 0.1 μ g/kg/day to about 0.3 mg/kg/day, about 0.1 μ g/kg/day to about 0.2 mg/kg/day, about 0.1 μ g/kg/day to about 0.1 mg/kg/day, about 0.1 μ g/kg/day to about 10 μ g/kg/day.

In one embodiment of the invention, the daily amount of inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition to be administered to a subject is from about 0.1 μ g/kg/day to about 1 mg/kg/day, from about 0.5 μ g/kg/day to about 1 mg/kg/day, from about 1 μ g/kg/day to about 0.75 mg/kg/day, from about 1 μ g/kg/day to about 0.5 mg/kg/day, from about 1 μ g/kg/day to about 0.25 mg/kg/day, from about 1 μ g/kg/day to about 0.2 mg/kg/day, from about 1 μ g/kg/day to about 0.1 mg/kg/day, from about 1 μ g/kg/day to about 0.075 mg/kg/day, or, About 1. mu.g/kg/day to about 0.05 mg/kg/day, about 1. mu.g/kg/day to about 0.025 mg/kg/day.

In one embodiment of the invention, the daily amount of inhibitor, composition, pharmaceutical composition, drug, cosmetic composition, or cosmeceutical composition to be administered to a subject is from about 0.01 mg/kg/day to about 20 mg/kg/day, from about 0.01 mg/kg/day to about 15 mg/kg/day, from about 0.01 mg/kg/day to about 12 mg/kg/day, from about 0.01 mg/kg/day to about 10 mg/kg/day, from about 0.01 mg/kg/day to about 9 mg/kg/day, from about 0.01 mg/kg/day to about 8 mg/kg/day, from about 0.01 mg/kg/day to about 7 mg/kg/day, from about 0.01 mg/kg/day to about 6 mg/kg/day, From about 0.01 mg/kg/day to about 5 mg/kg/day, from about 0.01 mg/kg/day to about 4 mg/kg/day, from about 0.01 mg/kg/day to about 3 mg/kg/day, from about 0.01 mg/kg/day to about 2 mg/kg/day, from about 0.01 mg/kg/day to about 1 mg/kg/day.

In one embodiment of the invention, the daily amount of inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition to be administered to a subject is about 0.1 μ g/kg/day, about 0.2 μ g/kg/day, about 0.4 μ g/kg/day, about 0.6 μ g/kg/day, about 0.8 μ g/kg/day, about 1 μ g/kg/day, about 1.5 μ g/kg/day, about 2.0 μ g/kg/day, about 2.5 μ g/kg/day, about 3.0 μ g/kg/day, about 3.5 μ g/kg/day, about 4.0 μ g/kg/day, about 4.5 μ g/kg/day, about 5.0 μ g/kg/day, about 5.5 μ g/kg/day, about 6.0 μ g/kg/day, or a combination thereof, About 6.5. mu.g/kg/day, about 7.0. mu.g/kg/day, about 7.5. mu.g/kg/day, about 8.0. mu.g/kg/day, about 8.5. mu.g/kg/day, about 9.0. mu.g/kg/day, about 9.5. mu.g/kg/day, about 10.0. mu.g/kg/day, about 15.0. mu.g/kg/day, about 20.0. mu.g/kg/day, about 25.0. mu.g/kg/day, about 30.0. mu.g/kg/day, about 35.0. mu.g/kg/day, about 40.0. mu.g/kg/day, about 45.0. mu.g/kg/day, about 50.0. mu.g/kg/day.

In one embodiment of the invention, the daily amount of the inhibitor, composition, pharmaceutical composition, drug, cosmetic composition, or cosmeceutical composition to be administered to the subject is about 0.01 mg/kg/day, about 0.02 mg/kg/day, about 0.03 mg/kg/day, about 0.04 mg/kg/day, about 0.05 mg/kg/day, about 0.06 mg/kg/day, about 0.07 mg/kg/day, about 0.08 mg/kg/day, about 0.09 mg/kg/day, about 0.1 mg/kg/day, about 0.2 mg/kg/day, about 0.3 mg/kg/day, about 0.4 mg/kg/day, about 0.5 mg/kg/day, about 0.6 mg/kg/day, about 0.7 mg/kg/day, about 0.8 mg/kg/day, About 0.9 mg/kg/day, about 1 mg/kg/day, about 1.5 mg/kg/day, about 2 mg/kg/day, about 2.5 mg/kg/day, about 3 mg/kg/day, about 3.5 mg/kg/day, about 4 mg/kg/day, about 4.5 mg/kg/day, about 5 mg/kg/day, about 6 mg/kg/day, about 7 mg/kg/day, about 8 mg/kg/day, about 9 mg/kg/day, about 10 mg/kg/day, about 12 mg/day kg/day, about 14 mg/kg/day, about 16 mg/kg/day, about 18 mg/kg/day, about 20 mg/kg/day.

In one embodiment of the invention, the inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition of the invention is administered in an amount of from about 1 μ g to about 100mg, from about 1 μ g to about 50mg, from about 1 μ g to about 10mg, from about 1 μ g to about 9mg, from about 1 μ g to about 8mg, from about 1 μ g to about 7mg, from about 1 μ g to about 6mg, from about 1 μ g to about 5mg, from about 1 μ g to about 4mg, from about 1 μ g to about 3mg, from about 1 μ g to about 2mg, from about 1 μ g to about 1mg, from about 1 μ g to about 100 μ g.

In one embodiment of the invention, the inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition of the invention is administered in an amount of from about 1 μ g to about 10mg, from about 5 μ g to about 10mg, from about 10 μ g to about 7.5mg, from about 10 μ g to about 5mg, from about 10 μ g to about 2.5mg, from about 10 μ g to about 2mg, from about 10 μ g to about 1mg, from about 10 μ g to about 0.75mg, from about 10 μ g to about 0.5mg, from about 10 μ g to about 0.25 mg.

In another embodiment, the inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition of the present invention is administered in an amount of from about 0.02mg to about 2000mg, from about 0.02mg to about 1500mg, from about 0.02mg to about 1000mg, from about 0.02mg to about 500mg, from about 0.02mg to about 200mg, from about 0.02mg to about 100mg, from about 0.02mg to about 50mg, from about 0.02mg to about 25mg, from about 0.02mg to about 10mg, from about 0.02mg to about 5 mg.

In another embodiment, the inhibitor, composition, pharmaceutical, cosmetic composition, or cosmeceutical composition of the present invention is administered in an amount of about 0.02mg, 0.04mg, 0.06mg, 0.08mg, 0.1mg, 0.2mg, 0.3mg, 0.4mg, 0.5mg, 0.6mg, 0.7mg, 0.8mg, 0.9mg, 1mg, 1.5mg, 2mg, 2.5mg, 3mg, 3.5mg, 4mg, 4.5mg, 5mg, 5.5mg, 6mg, 6.5mg, 7mg, 7.5mg, 8mg, 8.5mg, 9mg, 9.5mg, 10mg, 15mg, 20mg, 25mg, 30mg, 35mg, 40mg, 45mg, 50mg, 75mg, 100mg, 125mg, 150mg, 175mg, 200mg, 300mg, 400mg, 600mg, 700mg, 800mg, 1200mg, 1400mg, 800mg, or more.

In one embodiment, the methods of the invention are used for chronic treatment. In another embodiment, the methods of the invention are used for acute treatment.

In one embodiment of the invention, the subject is diagnosed with a disease associated with free oxygen radicals. In another embodiment of the invention, the subject is at risk for having a disease associated with free oxygen radicals.

In one embodiment, the subject is an adult, adolescent, child, toddler, or neonate.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQTo inhibit the mitochondria in a subject in need thereofComprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method of inhibiting free oxygen radical production without inhibiting cytoplasmic ROS production in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method for treating and/or preventing at least one free oxygen radical related disease in a subject in need thereof, or a method for treating and/or preventing at least one free oxygen radical related disease in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQThe mitochondria of (a) to treat and/or prevent, or for use in the treatment and/or prevention of, at least one free oxygen radical-related disease in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method for treating and/or preventing at least one free oxygen radical related disease in a subject in need thereof, or for treating and/or preventing at least one free oxygen radical related disease in a subject in need thereof, without inhibiting production of cytosolic ROS, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method for the treatment and/or prevention of a cardiovascular disease associated with free oxygen radicals in a subject in need thereof, or for the treatment and/or prevention of a cardiovascular disease associated with free oxygen radicals in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQThe mitochondria of (a) to treat and/or prevent a cardiovascular disease associated with free oxygen radicals in a subject in need thereof, or a method for treating and/or preventing a cardiovascular disease associated with free oxygen radicals in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for treating and/or preventing a cardiovascular disease associated with free oxygen radicals in a subject in need thereof, or for treating and/or preventing a cardiovascular disease associated with free oxygen radicals in a subject in need thereof, without inhibiting production of cytosolic ROS, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method for treating and/or preventing myocardial infarction in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQThe mitochondria of (a) to treat and/or prevent myocardial infarction in a subject in need thereof, or a method for treating and/or preventing myocardial infarction in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method of treating and/or preventing myocardial infarction in a subject in need thereof, or for treating and/or preventing myocardial infarction in a subject in need thereof, without inhibiting the production of cytosolic ROS, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method for the treatment and/or prevention of heart failure in a subject in need thereof, or for the treatment and/or prevention of heart failure in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQThe mitochondria of (a) to treat and/or prevent heart failure in a subject in need thereof, or a method for treating and/or preventing heart failure in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for treating and/or preventing heart failure in a subject in need thereof, or for treating and/or preventing heart failure in a subject in need thereof, without inhibiting the production of cytosolic ROS, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the present invention is a method for the treatment and/or prevention of cardiotoxicity, preferably of anthracyclines, anticancer drugs, quinolones and/or antiviral drugs, more preferably of anthracyclines, in a subject in need thereof, or for the treatment and prevention of cardiotoxicity, preferably of anthracyclines; the method comprises administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQThe mitochondria of (a) to treat and/or prevent cardiotoxicity in, or for use in the treatment and prevention of cardiotoxicity in, a subject in need thereof, preferably cardiotoxicity of an anthracycline, an anticancer drug, a quinolone drug, and/or an antiviral drug, more preferably cardiotoxicity of an anthracycline; the method comprises administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the present invention is a method for treating and/or preventing cardiotoxicity, preferably of an anthracycline, an anticancer agent, a quinolone and/or an antiviral agent, more preferably of an anthracycline, in a subject in need thereof, or for treating and preventing cardiotoxicity, without inhibiting the production of cytosolic ROS; the method comprises administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for the treatment and/or prevention of pulmonary arterial hypertension in a subject in need thereof, or for the treatment and/or prevention of pulmonary arterial hypertension in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQThe mitochondria of (a) to treat and/or prevent pulmonary arterial hypertension in a subject in need thereof, or a method for treating and/or preventing pulmonary arterial hypertension in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for treating and/or preventing pulmonary arterial hypertension, or for treating and/or preventing pulmonary arterial hypertension, in a subject in need thereof, without inhibiting the production of cytosolic ROS, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method for treating and/or preventing ischemia reperfusion injury in a subject in need thereof, or for treating and/or preventing ischemia reperfusion injury in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQFor the treatment and/or prevention ofA method of treating and/or preventing ischemia reperfusion injury in a subject in need thereof, or for use in treating and/or preventing ischemia reperfusion injury in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method for treating and/or preventing ischemia reperfusion injury in a subject in need thereof, or for treating and/or preventing ischemia reperfusion injury in a subject in need thereof, without inhibiting production of cytosolic ROS, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method for treating and/or preventing an aging disease in a subject in need thereof, or for treating and/or preventing an aging disease in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQThe mitochondria of (a) to treat and/or prevent, or for the treatment and/or prevention of, a senescence disease in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for treating and/or preventing a senescence disease in, or for treating and/or preventing a senescence disease in, a subject in need thereof, without inhibiting production of cytosolic ROS, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method for increasing insulin secretion in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQTo increase the mitochondria in a subject in need thereofA method of insulin secretion comprising administering an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method of increasing insulin secretion in a subject in need thereof without inhibiting the production of cytosolic ROS, comprising administering an effective amount of an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method of protecting neurons in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQA method of protecting a neuron in a subject in need thereof comprising administering an effective amount of an inhibitor or selective inhibitor of mitochondrial generation of ROS.

Another object of the invention is a method for protecting neurons in a subject in need thereof without inhibiting cytoplasmic ROS production, comprising administering an effective amount of an inhibitor or selective inhibitor of mitochondrial ROS production.

Another object of the invention is a method of preserving an organ, biological tissue and/or living cells, preferably prior to a transplantation procedure, comprising contacting said organ, biological tissue and/or living cells with an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method for the preparation of a compound by acting on site I of complex IQThe mitochondria of (a) to preserve an organ, biological tissue and/or living cells, preferably prior to a transplantation procedure, comprising contacting the organ, biological tissue and/or living cells with an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is a method of preserving an organ, biological tissue and/or living cells, preferably prior to a transplantation procedure, without inhibiting the production of cytosolic ROS, comprising contacting said organ, biological tissue and/or living cells with an inhibitor or selective inhibitor of mitochondrial production of ROS.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a) inhibit the production of free oxygen radicals in a subject in need thereof.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in inhibiting the production of free oxygen radicals in a subject in need thereof, without inhibiting the production of cytoplasmic ROS.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in the treatment and/or prevention of at least one disease associated with free oxygen radicals, or for use in the treatment and/or prevention of at least one disease associated with free oxygen radicals.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS by acting on site I of Complex IQFor use in the treatment and/or prevention of at least one free oxygen radical related disease or for use in the treatment and/or prevention of at least one free oxygen radical related disease.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or in the treatment and/or prevention of, at least one disease associated with free oxygen radicals, without inhibiting cytoplasmic ROS generation.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in the treatment and/or prevention of at least one cardiovascular disease associated with free oxygen radicals, or for use in the treatment and/or prevention of at least one cardiovascular disease associated with free oxygen radicals.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS by acting on the site of Complex IIQFor use in the treatment and/or prevention of at least one cardiovascular disease associated with free oxygen radicals, or for use in the treatment and/or prevention of at least one cardiovascular disease associated with free oxygen radicals.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or in the treatment and/or prevention of, at least one cardiovascular disease associated with free oxygen radicals, without inhibiting cytoplasmic ROS generation.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial production of ROS for use in, or in, the treatment and/or prevention of myocardial infarction.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a) are useful in, or in the treatment and/or prevention of, myocardial infarction.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or in the treatment and/or prevention of, myocardial infarction, without inhibiting cytoplasmic ROS generation.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or in the treatment and/or prevention of, heart failure.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS by acting on site I of Complex IQFor use in the treatment and/or prevention of heart failure, or for use in the treatment and/or prevention of heart failure.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or in the treatment and/or prevention of, heart failure, without inhibiting cytoplasmic ROS generation.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or for use in, the treatment and/or prevention of cardiotoxicity, preferably of anthracyclines, anticancer drugs, quinolones and/or antiviral drugs, more preferably of anthracyclines.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a) are useful for treating and/or preventing cardiotoxicity, or for use in treating and/or preventing cardiotoxicity, preferably of an anthracycline, an anticancer agent, a quinolone and/or an antiviral agent, more preferably of an anthracycline.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or in the treatment and/or prevention of, cardiotoxicity, without inhibiting cytoplasmic ROS generation; the cardiotoxicity is preferably cardiotoxicity of anthracycline drugs, anti-cancer drugs, quinolone drugs and/or antiviral drugs, and more preferably the cardiotoxicity of anthracycline drugs; or in the course of treating and/or preventing same.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or in the treatment and/or prevention of, pulmonary arterial hypertension.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a) are used in, or in the treatment and/or prevention of, pulmonary hypertension.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or in the treatment and/or prevention of, pulmonary arterial hypertension, without inhibiting cytoplasmic ROS generation.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial production of ROS for use in, or in the treatment and/or prevention of, ischemia reperfusion injury.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a) are useful in treating and/or preventing ischemia reperfusion injury, or in treating and/or preventing ischemia reperfusion injury.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or in the treatment and/or prevention of, ischemia reperfusion injury, without inhibiting cytoplasmic ROS generation.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or in the treatment and/or prevention of, an ageing disease.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a) are used for treating and/or preventing aging diseases, or are used in treating and/or preventing aging diseases.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in, or in the treatment and/or prevention of, aging diseases, without inhibiting cytoplasmic ROS generation.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in increasing insulin secretion.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a) are used to increase insulin secretion.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in increasing insulin secretion without inhibiting cytoplasmic ROS generation.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for protecting neurons.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a) are used to protect neurons.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for protecting neurons without inhibiting cytoplasmic ROS generation.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in the preservation of organs, biological tissues and/or living cells, preferably prior to a transplantation procedure.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS, useful for inhibiting the production of ROS by acting on site I of Complex IQThe mitochondria of (a) to preserve an organ, biological tissue and/or living cells, preferably prior to a transplantation procedure.

Another object of the invention is an inhibitor or selective inhibitor of mitochondrial generation of ROS for use in preserving organs, biological tissues and/or living cells, preferably prior to a transplantation procedure, without inhibiting cytoplasmic ROS production.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a), for use in inhibiting the production of free oxygen radicals in a subject in need thereof.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for inhibiting the generation of free oxygen radicals in a subject in need thereof, without inhibiting the generation of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of at least one disease associated with free oxygen radicals.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a), for use in the treatment and/or prevention of at least one disease associated with free oxygen radicals.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of at least one disease associated with free oxygen radicals without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of at least one cardiovascular disease associated with free oxygen radicals.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQMitochondria of (a), for use in the treatment and/or prevention of at least one cardiovascular disease associated with free oxygen radicals.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of at least one cardiovascular disease associated with free oxygen radicals without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of myocardial infarction.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a), for use in the treatment and/or prevention of myocardial infarction.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of myocardial infarction without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of heart failure.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a), for use in the treatment and/or prevention of heart failure.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of heart failure without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of cardiotoxicity, preferably of anthracyclines, anticancer drugs, quinolones and/or antiviral drugs, more preferably of anthracyclines.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a) for use in the treatment and/or prevention of cardiotoxicity, preferably of an anthracycline, of an anticancer drug, of a quinolone drug and/or of an antiviral drug, more preferably of an anthracycline.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of cardiotoxicity, preferably of anthracyclines, anticancer drugs, quinolones and/or antiviral drugs, more preferably of anthracyclines, without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of pulmonary hypertension.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a), for use in the treatment and/or prevention of pulmonary hypertension.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of pulmonary hypertension without inhibiting the production of cytoplasmic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of ischemia reperfusion injury.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a), for use in the treatment and/or prevention of ischemia reperfusion injury.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of ischemia reperfusion injury without inhibiting the production of cytoplasmic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of aging diseases.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a), for use in the treatment and/or prevention of ageing diseases.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the treatment and/or prevention of aging diseases without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for increasing insulin secretion.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQMitochondria of (a) for increasingUse of insulin secretion.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for increasing insulin secretion, without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for protecting neurons.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a), for use in protecting neurons.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for protecting neurons, without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS for the preservation of organs, biological tissues and/or living cells, preferably prior to a transplantation procedure.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS by acting on site I of Complex IQThe mitochondria of (a), for use in preserving organs, biological tissues and/or living cells, preferably prior to a transplantation procedure.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS to preserve organs, biological tissues and/or living cells, preferably prior to a transplantation procedure, without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQThe mitochondria of (a) to inhibit the production of free oxygen radicals in a subject in need thereof.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for inhibiting the generation of free oxygen radicals, without inhibiting the generation of cytosolic ROS in a subject in need thereof.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of at least one disease associated with free oxygen radicals.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQTo a medicament for the treatment and/or prevention of at least one disease associated with free oxygen radicals.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of at least one disease associated with free oxygen radicals, without inhibiting the generation of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of at least one cardiovascular disease associated with free oxygen radicals.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQTo a medicament for the treatment and/or prevention of at least one cardiovascular disease associated with free oxygen radicals.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of at least one cardiovascular disease associated with free oxygen radicals, without inhibiting the generation of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of myocardial infarction.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQLine ofUse of mitochondria in a medicament for the treatment and/or prevention of myocardial infarction.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of myocardial infarction, without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of heart failure.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQThe use of mitochondria of (a) in the manufacture of a medicament for the treatment and/or prevention of heart failure.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of heart failure without inhibiting the production of cytoplasmic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of cardiotoxicity, preferably of an anthracycline, an anticancer agent, a quinolone and/or an antiviral agent, more preferably of an anthracycline.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQThe use of the mitochondria of (a) for the treatment and/or prevention of cardiotoxicity, preferably of an anthracycline, an anticancer agent, a quinolone and/or an antiviral agent, more preferably of an anthracycline.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of cardiotoxicity, preferably of anthracyclines, anticancer drugs, quinolones and/or antiviral drugs, more preferably of anthracyclines, without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of pulmonary hypertension.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQThe use of the mitochondria of (a) in the manufacture of a medicament for the treatment and/or prevention of pulmonary hypertension.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of pulmonary hypertension without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of ischemia reperfusion injury.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQThe use of the mitochondria of (a) in the manufacture of a medicament for treating and/or preventing ischemia reperfusion injury.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of ischemia reperfusion injury without inhibiting the production of cytosolic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of aging diseases.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQThe use of the mitochondria of (a) in the manufacture of a medicament for the treatment and/or prevention of an ageing disease.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the treatment and/or prevention of aging diseases, without inhibiting the production of cytoplasmic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for increasing insulin secretion.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQThe mitochondria of (a) to increase insulin secretion.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for increasing insulin secretion without inhibiting the production of cytoplasmic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for protecting neurons.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQThe mitochondria of (a) to protect neurons.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for protecting neurons without inhibiting the production of cytoplasmic ROS.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for the preservation of organs, biological tissues and/or living cells, preferably prior to a transplantation procedure.

Another object of the invention is the use of inhibitors or selective inhibitors of mitochondrial generation of ROS in the preparation of a medicament for inhibiting the production of ROS by acting on site I of Complex IQThe mitochondria of (a), for use in a medicament for preserving an organ, biological tissue and/or living cells, preferably for preserving an organ, biological tissue and/or living cells prior to a transplantation procedure.

Another object of the invention is the use of an inhibitor or selective inhibitor of mitochondrial generation of ROS in the manufacture of a medicament for preserving organs, biological tissues and/or living cells, preferably organs, biological tissues and/or living cells prior to a transplantation procedure, without inhibiting the production of cytosolic ROS.

104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:激酶网络抑制剂及其用途

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!