专利摘要:
Benzopyrans prenylated PPAR agonists. The present invention discloses compounds of formula I comprising a core structure of prenylated benzopyran {IMAGE-01}. Said compounds of formula I, stereoisomers and pharmaceutically acceptable salts, exhibit activity as PPAR α, PPAR γ agonists, or dual PPAR α agonist activity; and PPAR γ There are thus described compounds with said prenylated benzopyran structure of formula I, their pharmaceutical compositions, their first medical use, as well as the use of compounds of prenylated benzopyran structure in the treatment of diseases responsive to the administration of PPAR α agonists; and/or PPAR γ (Machine-translation by Google Translate, not legally binding)
公开号:ES2718000A1
申请号:ES201731470
申请日:2017-12-26
公开日:2019-06-26
发明作者:Martínez Diego Miguel Cortés;Ferrando María Jesús Sanz;Escrig Nuria Cabedo;Del Castillo Almudena Bermejo;Ruiz Laura Piqueras;Sánchez Aída Collado;Marques Patrice Gomes
申请人:Universitat de Valencia;Fundacion Incliva;
IPC主号:
专利说明:

[0001]
[0002]
[0003] OBJECT OF THE INVENTION
[0004]
[0005] The present invention relates to compounds that comprise a central structure of pre-piled benzopyran. These compounds have activity as agonists of PPARa, PPARy, or dual agonist activity of PPARa and PPARy. The invention thus relates to compounds with said benzylated benzopyran structure of formula I, its pharmaceutical compositions, first medical use, as well as the use of compounds of benzopyranylated structure in the treatment of diseases that respond to the administration of PPARa agonists and / or PPARy. Such diseases are disorders such as type 2 diabetes (DT2), obesity, metabolic syndrome, cardiovascular disorders, dyslipidemias, such as hypercholesterolemia, familial hypertriglyceridemia (severe and moderate), primary chylomicronemia, familial dysbetalipoproteinemia or hyperlipoproteinemia, inflammation and neurodegenerative diseases, such as Parkinson's, Alzheimer's or amyotrophic lateral sclerosis (ALS).
[0006]
[0007] BACKGROUND OF THE INVENTION
[0008]
[0009] Peroxisome proliferation activating receptors (PPARs) belong to the family of nuclear hormone receptors. There are three types of PPAR: PPARa (NR1C1), PPARp / 5 (NUCI; NR1C2), and PPARy (NR1C3). All act as heterodimers with 9-cis-retinoic acid receptors (X retinoid receptor; RXR) and play a fundamental role in the regulation of various metabolic pathways, including lipid biosynthesis and glucose metabolism, as well as in differentiation and cell proliferation and apoptosis (Cheang et al., 2015, Ahmadian et al., 2013 and Tan et al., 2017).
[0010]
[0011] PPARs are considered powerful therapeutic tools in the control of different pathologies, such as type 2 diabetes (DT2), obesity, hyperlipidemia, cardiovascular disorders, inflammation and neurodegenerative diseases. In addition, PPARs have an additional importance since they are involved in the control of inflammatory processes, so they can regulate inflammation, vascular function, and vascular remodeling (Cheang et al., 2015, Ahmadian et al., 2013 and Tan et al., 2017).
[0012] PPARa are found mainly in the liver, kidneys, heart, muscle and adipose tissue, and play a fundamental role in the oxidation of fatty acids and the metabolism of lipoproteins.
[0013]
[0014] PPARy predominantly predominates in adipose tissue, macrophages, monocytes, intestinal cells, skeletal muscle and endothelium. They are related to lipid metabolism, adipogenesis, glucose homeostasis, and insulin sensitization.
[0015]
[0016] PPAR ligands and in particular PPARa and PPAR agonists and inhibit the activation of gene expression and of inflammation may adversely interfere with signaling pathways of proinflammatory transcription factors in vascular and inflammatory cells.
[0017]
[0018] PPARa are activated by fibrate structure compounds: fenofibrate, chlorfibrate, benzafibrate, and similar compounds such as WY-14643:
[0019]
[0020]
[0021]
[0022]
[0023] PPARa agonists are able to lower triglyceride levels and increase HDL-c, being very effective in the treatment of hypertriglyceridemia.1
[0024]
[0025] Among the known PPAR ligands of these receptors, thiazolidinediones (TZD) agonists, among which rosiglitazone and pioglitazone, have been used, which have been used clinically for the treatment of DT2 due to their ability to lower glucose levels in blood and improve insulin sensitivity (Ahmadian etal., 2013).
[0026]
[0027]
[0028] The development of PPARa or PPAR agonists and , or dual PPARa / PPARY agonists, has led to the synthesis of a new class of candidates with therapeutic potential (Cheang et al., 2015 and Tan et al., 2017). In fact, dual PPARa / Y agonists are considered more potent compounds for correcting lipid and glucose homeostasis in DT2 and metabolic syndrome, than selective agonists (Tan et al., 2017). Despite the clinical efficacy of PPARa and PPARy agonists in the treatment of dyslipidemia and DT2, respectively, the occurrence of numerous adverse effects has limited the use of these selective agonists, forcing the withdrawal of numerous drugs. In particular, PPARa agonists can produce an increase in creatinine and transaminase levels, and more rarely myopathy, while PPARy agonists are related to weight gain, edema and myocardial infarction (Cheang et al., 2015). The possibility of finding a single compound capable of activating the two PPARa and PPARy receptors, a dual agonist, is presented as a potential alternative in the treatment of DT2 and metabolic syndrome, with lower side effects (Cheang et al., 2015 and Tan et al., 2017). Recently, the use of two PPARa / Y dual agonists belonging to the group of glands, saroglitazar in India (Lipaglyn ™, by the Zydus Cadila research center) and lobeglitazone in Korea (Duvie ™, by Chong Kun Dang) has been approved, for lipid and glycemic control (Tan et al., 2017).
[0029]
[0030]
[0031]
[0032]
[0033] However, other dual agonists such as farglitazar and muraglitazar have been withdrawn from the market due to their side effects of edema, myocardial infarction, stroke and heart failure, as well as aleglitazar (in clinical phase II), ragaglitazar and tesaglitazar since they have shown carcinogenicity in rodent models and an increase in plasma creatinine (Tan et al., 2017).
[0034]
[0035] The development of hPPARa and hPPARy dual agonists allows combining the properties of PPARa agonists (fibrates) on the regulation of lipid metabolism including decreased triglycerides (TG), with the additional effect of improving insulin sensitivity produced by the agonists of PPAR and (TZD). Generally, these dual hPPARa / y ligands have shown in animal models greater efficacy and lower toxicity in the treatment of metabolic dysfunctions, than the use of selective compounds for each of the receptors. In fact, clinical studies show that dual PPARa agonists / and decrease insulin resistance in type 2 diabetes, blood glucose and TG values, and participate in the control of vascular inflammation. Therefore, the search for dual agonists PPARa / y is important for the treatment of various metabolic dysfunctions such as metabolic syndrome, DT2 and cardiovascular diseases.
[0036]
[0037] Therefore, there is a need to develop PPARa agonists and / or and that show greater efficacy than the current ones, and / or that also show less toxicity.
[0038]
[0039] DESCRIPTION
[0040]
[0041] The present invention describes agonist compounds of PPARa and / or PPAR and pharmaceutical compositions comprising them. Such compounds, and compositions comprising them are useful in the treatment of diseases that respond to administration of PPAR agonists and / or PPAR. These compounds are, therefore, useful in the treatment of cardiovascular disorders, type 2 diabetes (DT2), obesity, metabolic syndrome, hypercholesterolemia, hypertriglyceridemia, primary chylomicronemia, hyperlipoproteinemia, familial dysbetalipoproteinemia, inflammation and neurodegenerative diseases, such as Parkinson's, Alzheimer's or amyotrophic lateral sclerosis (ALS). The present invention also describes the uses of such compounds agonists PPARa and / or PPAR and pharmaceutical compositions comprising, in the treatment of a disease which responds to administration of agonists PPARa and / or PPAR.
[0042]
[0043] An embodiment of the PPARa and / or PPAR agonist compounds described refers to a compound of general formula I, stereoisomers and pharmaceutically acceptable salts thereof:
[0044]
[0045]
[0046] where
[0047]
[0048] - Ri is independently selected from H or a protective group of a phenol;
[0049] - R 2 is independently selected from an alkyl ester group of structure:
[0050]
[0051]
[0052]
[0053] j
[0054]
[0055] a derivative of a prenyl group of structure:
[0056]
[0057]
[0058]
[0059]
[0060] or an acyl group -C (O) ORb, where Rb is a C1-6 alkyl group,
[0061]
[0062] where
[0063]
[0064] - A is independently selected from an O atom, an S atom or an NR7 group, where R7 is independently selected from H, OR8 or a C1-6 alkyl group, and where R8 is an H group or a C1-6 group I rent;
[0065]
[0066] - R5 is independently selected from H or a carboxylic acid protecting group;
[0067]
[0068] - R6 is C1-6 alkyl or OR9 where R9 is C1-6 alkyl;
[0069]
[0070] and where
[0071]
[0072] - R3 is H or a C1-6 alkyl group;
[0073] - R4 is a C1-6 alkyl group or an alkoxide .;
[0074] - R10 is independently selected from a C1-6 alkyl, allyl, alkylamine, alkylamide, alkylaryl, alkyl ether, haloalkyl, silyl, carbamate, alkylsulfone or haloalkylsulfone group.
[0075]
[0076] In one embodiment of the present invention when R1 is H and R3 is methyl, R5 is different from H or methyl, and when R1 is methyl and R3 is methyl, R5 is different from H or methyl.
[0077] One embodiment relates to a compound of general formula I, described above, to its enantiomers, isomers E, Z and pharmaceutically acceptable salts thereof.
[0078]
[0079] For the purposes of the present invention, the term "phenol protecting group" is defined as groups that replace the H of the phenol OH group to prevent it from being reactive or groups that favor the pharmacodynamics and pharmacokinetics of the molecule. They are non-limiting examples of phenol protecting groups, among others: C1-6 alkyl (eg methyl, ethyl, propyl, etc.); allyl; alkylamine (eg methyl isoquinoline); alkylamide (eg acetamide); alkylaryl ( eg benzyl, p-fluorobenzyl, p-methoxybenzyl); alkyl ether (eg 2-methoxyethoxymethyl); halo-alkyl (eg trifluoromethyl, fluoroethyl, chloroethyl); silyl (eg alkyltrimethylsilane) , allyltrimethylsilane); C (O) NRmRp carbamate, where Rm and / or Rp are each and independently H, C1-6 alkyl or aryl; an acyl group (eg benzoyl), an alkylsulfone or a halo-alkylsulfone ( eg trifluoromethylsulfone).
[0080]
[0081] In one embodiment of the present invention the group R1 is a protective group of a phenol that is independently selected from the group consisting of H; C1-6 alkyl; allyl alkylamide; alkylamine; alkylaryl; alkyl ether; halo-alkyl; silyl; C (O) NRmRp, where Rm and / or Rp are each and independently H, C 1-6 alkyl or aryl; acyl C (O) Ra, where Ra is an aryl group; alkylsulfone or halo-alkylsulfone.
[0082]
[0083] In one embodiment R1 is independently selected from the group consisting of H, C1-6 alkyl, allyl, alkylamide, alkylaryl, alkyl ether, alkylbenzoyl, haloalkyl, silyl, haloalkylsulfone or an acyl group -C (O) Ra, where Ra is an aryl group.
[0084]
[0085] In another embodiment R1 is independently selected from the group consisting of H, C1-6 alkyl, alkylamide, alkylaryl, or an acyl group -C (O) Ra, where Ra is an aryl group.
[0086] The term "protecting group of a carboxylic acid" is defined, for the purposes of the present invention, as groups that replace the H of the carboxylic acid -COOH group to prevent it from being reactive or groups that favor the pharmacodynamics and pharmacokinetics of the molecule Non-limiting examples of carboxylic acid protecting groups are, among others: C1-6 alkyl, allyl, alkylamide, alkylamine, alkylaryl, alkyl ether, alkyl ester, haloalkyl, silyl, C (O) NRmRp carbamate, where Rm and / or Rp are each and independently H, C1-6 alkyl or aryl, C (O) acyl Ra, where Ra is an aryl, alkylsulfone or halo-alkylsulfone group.
[0087] In one embodiment R5 is independently selected from H; C1-6 alkyl; allyl alkylamide; alkylamine; alkylaryl; alkyl ether; halo-alkyl; silyl; C (O) NRmRp, where Rm and / or Rp are each and independently H, C1-6 alkyl or aryl; acyl; alkylsulfone or haloalkylsulfone.
[0088] In one embodiment R5 is independently selected from the group consisting of H, C1-6 alkyl, alkylaryl or alkyl ester.
[0089]
[0090] In another embodiment R5 is independently selected from the group consisting of H, C1-6 alkyl, aryl, alkylaryl, alkyl ether, alkyl ester, alkylsulfone, haloalkyl, silyl, haloalkylsulfone.
[0091]
[0092] An embodiment of the invention relates to a compound of general formula I, stereoisomers and pharmaceutically acceptable salts thereof:
[0093]
[0094]
[0095]
[0096]
[0097] where
[0098]
[0099] - R 1 is independently selected from H; C 1-6 alkyl; allyl alkylamide; alkylamine;
[0100] alkylaryl; alkyl ether; halo-alkyl; silyl; C (O) NRmRp, where Rm and / or Rp are each and independently H, C 1-6 alkyl or aryl; acyl C (O) Ra, where Ra is an aryl group; alkylsulfone or halo-alkylsulfone;
[0101] - R 2 is independently selected from an alkyl ester group of structure:
[0102]
[0103]
[0104]
[0105]
[0106] a derivative of a prenyl group of structure:
[0107]
[0108]
[0109] or an acyl group -C (O) ORb, where Rb is a C i-6 alkyl group,
[0110]
[0111] where
[0112]
[0113] - A is independently selected from an O atom, an S atom or an NR7 group, where R7 is independently selected from H, OR8 or a C1-6 alkyl group; and where R8 is an H group or a C1-6 alkyl group;
[0114]
[0115] - R5 is independently selected from H; C1-6 alkyl; allyl alkylamide;
[0116] alkylamine; alkylaryl; alkyl ether; halo-alkyl; silyl; C (O) NRmRp, where Rm and / or Rp are each and independently H, C1-6 alkyl or aryl; acyl; alkylsulfone or halo-alkylsulfone;
[0117]
[0118] - R6 is C1-6 alkyl or OR9 where R9 is C1-6 alkyl;
[0119]
[0120] and where
[0121]
[0122] - R 3 is H or a C 1-6 alkyl group;
[0123] - R 4 is a C 1-6 alkyl group or an alkoxide .;
[0124] - R 10 is independently selected from a C 1-6 alkyl, allyl, alkylamine, alkylamide, alkylaryl, alkyl ether, haloalkyl, silyl, carbamate, alkylsulfone or haloalkylsulfone group.
[0125]
[0126] In one embodiment of the present invention when R 1 is H and R 3 is methyl, R 5 is different from H or methyl, and when R 1 is methyl and R 3 is methyl, R 5 is different from H or methyl.
[0127]
[0128] For the purposes of the present invention, a compound of formula I includes all stereoisomers of said formula I.
[0129]
[0130] The term "stereoisomer" refers, for the purposes of the present invention, to molecules that have the same molecular formula and the same sequence of bonded atoms (same constitution), with the same bonds between their atoms, but which differ in the three-dimensional orientation of Your atoms in space. Enantiomers and diastereomers are two different types of stereoisomers.
[0131]
[0132] The term "enantiomer" refers, for the purposes of the present invention, to two stereoisomers that are mirror images and not superimposable to each other.
[0133]
[0134] In particular, one embodiment of the present invention comprises the R and S enantiomers of the compounds of formula I, wherein said enantiomers are the 2 possible enantiomers with respect to the C-2 position of the benzopyran nucleus, thus including the configuration R and S of the radical R 10 .
[0135] The term "diasteromer" refers, for the purposes of the present invention, to two stereoisomers that are not mirror images. Diasteromers include meso compounds, E / Z isomers and non-enantiomeric optical isomers.
[0136]
[0137] The E / Z isomers refer to compounds comprising double C = C bonds, ie alkenes or cycloalkenes. If in a double C = C link the two highest priority substituents, in accordance with the rules of the IUPAC (International Union of Pure and Applied Chemistry), are on the same side, the provision is Z. Instead, if they are in opposite sides the arrangement is E.
[0138]
[0139] One embodiment relates to a compound of general formula I, described above, to its enantiomers, isomers E, Z and pharmaceutically acceptable salts thereof.
[0140]
[0141] An embodiment comprises the R enantiomer with respect to the C-2 position of the benzopyran nucleus.
[0142]
[0143] Another embodiment comprises the S enantiomer with respect to the C-2 position of the benzopyran nucleus.
[0144]
[0145] An embodiment comprises the E-isomer with respect to the double bond of the C-‘3 position of the pre-stacked side chain.
[0146]
[0147] An embodiment comprises the Z isomer with respect to the double bond of the C-3 position of the pre-stacked side chain.
[0148]
[0149] The term "pharmaceutically acceptable salts", for the purposes of the present invention, refers to any salt that administered to a patient is capable of providing (directly or indirectly) the compound in question. Said salts are preferably addition salts of organic or inorganic acids, or of organic or inorganic bases. Examples of acid addition salts include, but are not limited to, inorganic acid addition salts such as, for example, chloride, bromide, iodide, sulfate, nitrate, phosphate, or organic acid addition salts such as, by example, acetate, trifluoroacetate, maleate, fumarate, citrate, oxalate, succinate or tartrate. Examples of base addition salts include, but are not limited to, inorganic base addition salts such as, for example, sodium, potassium, calcium or ammonium, or organic base addition salts such as, for example, ethylenediamine, ethanolamine, N, N-dialkylene ethanol, triethanolamine and basic amino acid salts. Salt formation procedures are conventional procedures in the state of the art.
[0150] The term "Ci-6 alkyl" refers, for the purposes of the present invention, to a fully saturated hydrocarbon chain of 1 to 6 carbon atoms, which can be linear or branched.
[0151]
[0152] The term "halo-alkyl" refers, for the purposes of the present invention, to a hydrocarbon chain, which can be linear or branched, in which at least one H is substituted by an atom of a halogen.
[0153]
[0154] The term "allyl" refers, for the purposes of the present invention, to a hydrocarbon chain, which can be linear or branched, comprising at least one group -CH = CH-CH2-.
[0155]
[0156] The term "alkylamide" refers, for the purposes of the present invention, to a hydrocarbon chain, which can be linear or branched, substituted with an amide group.
[0157]
[0158] The term "alkylamine" refers, for the purposes of the present invention, to a hydrocarbon chain, which can be linear or branched, substituted with an amino group.
[0159]
[0160] The term "aryl" refers, for the purposes of the present invention, to a group derived from an aromatic hydrocarbon that is formed by extracting a hydrogen atom from an aromatic ring in said aromatic hydrocarbon. For the purposes of the present invention said aryl groups can be optionally substituted Examples of non-limiting substitutions are OH, alkoxide, alkyl, halogen, halogenated alkyl, cyano, thiol, amine or alkylamine.
[0161]
[0162] The term "alkylaryl" refers, for the purposes of the present invention, to a hydrocarbon chain substituted with an aryl group, which in turn is defined as described above.
[0163]
[0164] The term "alkoxide" refers, for the purposes of the present invention, to an oxygen atom attached to a hydrocarbon chain, wherein said hydrocarbon chain may comprise an alkyl group and / or an aryl group, as defined above.
[0165]
[0166] The term "alkyl ether" refers, for the purposes of the present invention, to a saturated hydrocarbon chain substituted with an alkoxide group, as defined above.
[0167] The term "acyl" refers, for the purposes of the present invention, to a carbonyl group attached to a hydrocarbon chain, wherein said hydrocarbon chain may comprise an alkyl group and / or an aryl group, as defined above.
[0168]
[0169] The term "ester", for the purposes of the present invention, refers to a group derived from a carboxylic acid -COOH, in which the hydroxyl -OH group is substituted by an alkoxide group, as defined in accordance with the present invention
[0170] The term "alkyl ester" refers, for the purposes of the present invention, to a saturated hydrocarbon chain substituted with an ester group, as defined above.
[0171] In a preferred embodiment A is an O atom.
[0172]
[0173] In another preferred embodiment A is an atom of S.
[0174]
[0175] In another preferred embodiment A is an NR7 group, where R7 is independently selected from H, -OR8 or a C1-6 alkyl group, and where R8 is an H group or a C1-6 alkyl group. More preferably R7 is H, -OH or -OCH3.
[0176]
[0177] In a preferred embodiment R1 is H.
[0178]
[0179] In another preferred embodiment R1 is a C1-6 alkyl group. More preferably R1 is a methyl or propyl group.
[0180]
[0181] In another preferred embodiment R1 is a alkylaryl group. More preferably R1 is a benzyl or 4-fluorobenzyl group.
[0182]
[0183] In another preferred embodiment R1 is an alkylamide group. More preferably R1 is an acetamide group.
[0184]
[0185] In another preferred embodiment R1 is an acyl group -C (O) Ra. More preferably R1 is an acyl group -C (O) Ra, where Ra is an aryl group. Even more preferably Ra is a phenyl group.
[0186]
[0187] In a preferred embodiment R3 is H.
[0188]
[0189] In another preferred embodiment R3 is methyl.
[0190]
[0191] In a preferred embodiment R4 is C1-6 alkyl or an alkoxide group. In a preferred embodiment the alkoxide group is a halogenated alkoxide group.
[0192]
[0193] In a more preferred embodiment R4 is methyl.
[0194]
[0195] In a more preferred embodiment R4 is -OCH2CF3.
[0196]
[0197] In a preferred embodiment R5 is H.
[0198]
[0199] In another preferred embodiment R5 is C1-6 alkyl. More preferably R5 is methyl, ethyl, propyl, butyl or tert-butyl. Even more preferably R5 is methyl, ethyl or propyl.
[0200]
[0201] In another preferred embodiment R5 is alkylaryl. More preferably R5 is benzyl.
[0202]
[0203] In another preferred embodiment R5 is alkyl ester. More preferably R5 is -CH2COOCH3 or -CH (COOCH3) 2.
[0204] In a preferred embodiment R 6 is methyl.
[0205]
[0206] In another preferred embodiment R6 is ORg where R9 is C1-6 alkyl. More preferably R9 is ethyl.
[0207]
[0208] In a preferred embodiment R10 is C1-6 alkyl. More preferably R10 is methyl.
[0209]
[0210] A preferred embodiment of the invention relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof, independently selected from the group consisting of 3, 4, 4a, 4b, 4c, 6, 8, 9, 10, 11 , 12 and 13:
[0211] Ċ
[0212] Ċ
[0213] 5
[0214]
[0215]
[0216] One embodiment comprises the R enantiomer of compounds 3, 4, 4a, 4b, 4c, 6, 7, 8, 9, 10, 11, 12 and 13, relative to the C-2 position of the benzopyran nucleus.
[0217] Another embodiment comprises the S enantiomer of compounds 3, 4, 4a, 4b, 4c, 6, 7, 8, 9, 10, 11, 12 and 13, relative to the C-2 position of the benzopyran nucleus.
[0218]
[0219] One embodiment comprises the isomer (E, Z) of compounds 3, 4, 4a, 4b, 4c, 6, 7, 8, 10, 11 and 12, relative to positions C-'3 and C-'7 of the side chain stacked.
[0220]
[0221] Another embodiment comprises the isomer (E, E) of compounds 3, 4, 4a, 4b, 4c, 6, 7, 8,10, 11 and 12, relative to positions C-'3 and C-'7 of the side chain stacked.
[0222]
[0223] One embodiment comprises the isomer (Z, Z) of compounds 3, 4, 4a, 4b, 4c, 6, 7, 8, 10, 11 and 12, relative to positions C-'3 and C-'7 of the side chain stacked.
[0224]
[0225] Another embodiment comprises the isomer (Z, E) of compounds 3, 4, 4a, 4b, 4c, 6, 7, 8, 10, 11 and 12, with respect to positions C-'3 and C-'7 of the side chain stacked.
[0226]
[0227] An embodiment comprises the E- isomer of compounds 9 and 13, relative to the C-'3 position of the pre-stacked side chain.
[0228]
[0229] Another embodiment comprises the Z isomer of compounds 9 and 13, relative to the C-'3 position of the pre-stacked side chain.
[0230] The compounds of formula I, their stereoisomers and pharmaceutically acceptable salts, with PPAR agonist activity comprise a common structure derived from the natural precursor benzopyrans polycrasoidol and polycrasoidin (Gonzalez et al., 1995 and 1996), natural isomers of compounds 1 and 2 herein. described: The isomer (3E, 7Z) of compound 1 corresponds to the natural compound polycrasoidol, while the isomer (3E, 7E) is the natural compound isopolycerasoidol, and the polycrasoidin corresponds to the isomer (3E, 7Z) of compound 2:
[0231]
[0232]
[0233]
[0234] These documents by Gonzalez et al (1995 and 1996) describe the isolation of the natural compounds polycrasoidol and policerasoidin from the bark of trees of the genus Polyalthia, indicating that some of the compounds of the family of pre-piled hydroquinones, to which they belong, are known for having cytotoxic or antioxidant properties, without referring or giving clues about a possible activity as PPAR agonists. In the same vein, other documents disclose said compounds, or compounds of similar structure, although said compounds also do not exhibit activity as PPAR agonists. In particular, the document Taha et al, 2015 and Karimian et al. (2015) describes the use of polycrasoidol, its methyl ester, and polycrasoidin as inducers of apoptosis in breast cancer tumor cells. Zhao et al. (2010) analyzes different metabolites derived from tocopherol and tocotrienols detected in urine, serum or liver in human and mouse samples, among which polycrasoidol is described. Zafra-Polo et al. (1996) describes polyalthidin, related to the basic structure of the compounds of the invention, but which is nevertheless only described in relation to its activity as a mitochondrial respiratory chain inhibitor. Finally, document EP0421419A describes benzopyran nucleus compounds with three pre-piled chains referring to their use to lower serum cholesterol levels. None of these documents describe, or give clues to, the possible utility of similar compounds, with a benzopyran nucleus, as PPAR agonists.
[0235]
[0236] An embodiment comprises the R enantiomer of compounds 1 and 2, relative to the C-2 position of the benzopyran nucleus.
[0237] Another embodiment comprises the S enantiomer of compounds 1 and 2, relative to the C-2 position of the benzopyran nucleus.
[0238]
[0239] One embodiment comprises the isomer (E, E) of compounds 1 and 2, with respect to positions C-'3 and C-'7 of the pre-stacked side chain.
[0240]
[0241] Another embodiment comprises the isomer (E, Z) of compounds 1 and 2, with respect to positions C-'3 and C-'7 of the pre-stacked side chain.
[0242]
[0243] One embodiment comprises the isomer (Z, Z) of compounds 1 and 2, relative to positions C-‘3 and C-‘7 of the pre-piled side chain.
[0244]
[0245] Another embodiment comprises the isomer (Z, E) of compounds 1 and 2, with respect to positions C-'3 and C-'7 of the pre-stacked side chain.
[0246]
[0247] A preferred embodiment relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof:
[0248]
[0249]
[0250]
[0251] wherein R3, R10 and R4 are methyl and R2 is a derivative of a prenyl group of structure:
[0252]
[0253]
[0254] , where A is an atom of O, and R6 is methyl,
[0255] and wherein said compound of formula I is a compound of formula II, stereoisomers and pharmaceutically acceptable salts thereof:
[0256]
[0257]
[0258]
[0259]
[0260] wherein R 1 and R 5 are defined according to the embodiments of the present invention described above.
[0261]
[0262] Said compounds of formula II therefore include compounds 1, 2 and derivative compounds with different R 1 and R 5 groups .
[0263]
[0264] In a preferred embodiment, when R 1 is H, R 5 is different from H or methyl, and when R 1 is Me, R 5 is different from H or methyl.
[0265]
[0266] One embodiment comprises the R enantiomer of the compounds of formula II, relative to the C-2 position of the benzopyran nucleus.
[0267]
[0268] Another embodiment comprises the R enantiomer of the compounds of formula II, relative to the C-2 position of the benzopyran nucleus.
[0269]
[0270] An embodiment comprises the isomer (E, Z) of the compounds of formula II, with respect to positions C-'3 and C-'7 of the pre-stacked side chain.
[0271]
[0272] Another embodiment comprises the isomer (E, E) of the compounds of formula II, with respect to positions C-‘3 and C-‘7 of the pre-piled side chain.
[0273]
[0274] One embodiment comprises the isomer (Z, Z) of the compounds of formula II, with respect to positions C-'3 and C-'7 of the pre-stacked side chain.
[0275]
[0276] Another embodiment comprises the isomer (Z, E) of the compounds of formula II, with respect to positions C-‘3 and C-‘7 of the pre-piled side chain.
[0277]
[0278] In a preferred embodiment, R 1 is H and R 5 is H.
[0279]
[0280] In another preferred embodiment, R 1 is C 1-6 alkyl and R 5 is H. More preferably R 1 is methyl and R 5 is H.
[0281] In another preferred embodiment, Ri is H and R5 is C1-6 alkyl. More preferably Ri is H and R5 is methyl or propyl.
[0282]
[0283] In a preferred embodiment, R1 is H and R5 is alkylaryl. More preferably R1 is H and R5 is benzyl.
[0284]
[0285] In another preferred embodiment, R1 is alkylamide and R5 is alkylaryl. More preferably R1 is acetamide, and R5 is benzyl.
[0286]
[0287] In another preferred embodiment, R1 and R5 are alkylaryl. More preferably R1 and R5 are benzyl.
[0288]
[0289] In another preferred embodiment, R1 is C1-6 alkyl and R5 is H. More preferably R1 is propyl is R5 is H.
[0290]
[0291] In another preferred embodiment, R1 is an acyl group -C (O) Ra, where Ra is an aryl group, and R5 is an alkyl ester group. More preferably R1 is a benzoyl group and R5 is a -CH2COOCH3 group.
[0292]
[0293] In another preferred embodiment, R1 is a C1-6 alkyl group and R5 is an alkyl ester group. More preferably R1 is a methyl group and R5 is a -CH2COOCH3 group or a -CH (COOCH3) 2 group.
[0294]
[0295] Both compounds 1 and 2 show a similar structure and only differ in the functional group of the C-6 position, a hydroxyl in compound 1 and a methoxy in compound 2. From compounds 1 and 2 the compounds can be synthesized 3, 4, 4a, 4b, 4c, 6, 7 or 8 of formula I, by substituting positions C-9 'and C-6, as described in Example 1, below.
[0296]
[0297] An embodiment of the compound of formula II, stereoisomers and pharmaceutically acceptable salts thereof, as described above, is a compound that is independently selected from compound 1, 2, 3, 4, 4a, 4, b, 4c, 6, 7 or 8.
[0298]
[0299] A preferred embodiment of the present invention relates to a compound of formula II, stereoisomers and pharmaceutically acceptable salts thereof, independently selected from the group consisting of 3, 4, 4a, 4b, 4c, 6, 7 and 8. In one Preferred embodiment compounds 3, 4, 4a, 4b, 4c, 6, 7 and 8 are isomers (E, Z), in relation to the double bonds of the side chain stacked in positions C-3 'and C-7'.
[0300]
[0301] Another preferred embodiment of the present invention relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof:
[0302]
[0303] wherein R3 is H, R4 and R10 are methyl and R2 is a derivative of a prenyl group of structure:
[0304]
[0305]
[0306]
[0307] , where A is an atom of O,
[0308]
[0309] and wherein said compound of formula I is a compound of formula III, stereoisomers and pharmaceutically acceptable salts thereof:
[0310]
[0311]
[0312]
[0313]
[0314] wherein R1, R5, and R6 are defined according to the embodiments of the present invention described above.
[0315]
[0316] Said compounds of formula III also have a structure derived from compounds 1 and 2, but unlike the compounds of formula II, they present H instead of methyl as group R3, and in addition to the modifications at positions C-6 (- OR1) and C-9 '(-COOR5), also have modifications in position C-8' (R6).
[0317]
[0318] One embodiment comprises the R enantiomer of the compounds of formula III, relative to the C-2 position of the benzopyran nucleus.
[0319] Another embodiment comprises the R enantiomer of the compounds of formula III, with respect to the C-2 position of the benzopyran nucleus.
[0320]
[0321] One embodiment comprises the isomer (E, Z) of the compounds of formula III, with respect to positions C-'3 and C-'7 of the pre-stacked side chain.
[0322]
[0323] Another embodiment comprises the isomer (E, E) of the compounds of formula III, with respect to positions C-‘3 and C-‘7 of the pre-piled side chain.
[0324]
[0325] One embodiment comprises the isomer (Z, Z) of the compounds of formula III, with respect to positions C-‘3 and C-‘7 of the pre-piled side chain.
[0326]
[0327] Another embodiment comprises the isomer (Z, E) of the compounds of formula III, with respect to positions C-‘3 and C-‘7 of the pre-piled side chain.
[0328]
[0329] In a preferred embodiment, R1 is an alkylaryl group, R6 is a -OR9 group, where R9 is C1-6 alkyl, and R5 is H or a C 1-6 alkyl group. More preferably R1 is a benzyl group, R6 is a -OCH2CH3 group, and R5 is H or ethyl.
[0330]
[0331] More preferably R1 is a 4-fluorobenzyl group, R6 is a -OCH2CH3 group, and R5 is ethyl.
[0332] An embodiment of the compound of formula III, stereoisomers and pharmaceutically acceptable salts thereof, as described above, is a compound that is independently selected from the group consisting of compound 10, 11 and 12:
[0333]
[0334]
[0335]
[0336]
[0337] Another preferred embodiment of the present invention relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof:
[0338]
[0339]
[0340]
[0341]
[0342] wherein R3 is H, R4 and R10 are methyl and R2 is an alkyl ester group of structure:
[0343]
[0344]
[0345] , where A is an atom of O,
[0346]
[0347] and wherein said compound of formula I is a compound of formula IV, stereoisomers and pharmaceutically acceptable salts thereof:
[0348]
[0349]
[0350] wherein Ri and R 5 are defined according to the embodiments of the present invention described above.
[0351]
[0352] Said compounds of formula IV also have a structure derived from the compounds of formula I 1 and 2. However, the compounds of formula IV are compounds of formula I where the radical R 3 is H, and not methyl as compounds 1 present. and 2, and the radical R 2 is an alkyl ester group which, unlike compounds 1 and 2 , does not have a second prenyl group.
[0353]
[0354] One embodiment comprises the R enantiomer of the compounds of formula IV, relative to the C-2 position of the benzopyran nucleus.
[0355]
[0356] Another embodiment comprises the S enantiomer of the compounds of formula IV, with respect to the C-2 position of the benzopyran nucleus.
[0357]
[0358] One embodiment comprises the E- isomer of the compounds of formula IV, relative to the double bond of the pre-piled side chain.
[0359]
[0360] Another embodiment comprises the Z isomer of the compounds of formula IV, with respect to the double bond of the pre-piled side chain.
[0361]
[0362] In a preferred embodiment R 1 is alkylaryl and R 5 is C 1-6 alkyl. More preferably R 1 is 4-fluorobenzyl and R 5 is ethyl.
[0363]
[0364] An embodiment of the compound of formula IV, stereoisomers and pharmaceutically acceptable salts thereof, as described above, is compound 9:
[0365]
[0366]
[0367] Another preferred embodiment of the present invention relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof:
[0368]
[0369]
[0370]
[0371]
[0372] wherein R 3 is H, R 10 is methyl and R 2 is -C (O) ORb, and wherein said compound of formula I is a compound of formula V, stereoisomers and pharmaceutically acceptable salts thereof:
[0373]
[0374]
[0375]
[0376]
[0377] wherein Rb is a C1-6 alkyl group and, R1 and R4 are defined according to the embodiments of the present invention described above.
[0378]
[0379] Said compounds of formula V also have a structure derived from the compounds of formula I1 and 2, but wherein in said formula I the radical R2 is an ester group instead of a pre-alkyl ester-ester group having compounds 1 and 2 .
[0380]
[0381] An embodiment comprises the R enantiomer of the compounds of formula V, relative to the C-2 position of the benzopyran nucleus.
[0382]
[0383] Another embodiment comprises the S enantiomer of the compounds of formula V, with respect to the C-2 position of the benzopyran nucleus.
[0384]
[0385] An embodiment comprises the E- isomer of the compounds of formula V, relative to the double bond of the pre-piled side chain.
[0386]
[0387] Another embodiment comprises the Z isomer of the compounds of formula V, with respect to the double bond of the pre-piled side chain.
[0388]
[0389] In a preferred embodiment R1 is alkylaryl, Rb is C1-6 alkyl and R4 is an alkoxide group. More preferably R1 is benzyl, Rb is ethyl and R4 is -OCH2CF3.
[0390] An embodiment of the compound of formula V, stereoisomers and pharmaceutically acceptable salts thereof, as described above, is compound 13:
[0391]
[0392]
[0393]
[0394]
[0395] An embodiment of the present invention relates to a pharmaceutical composition comprising an effective amount of at least one compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof, as described above, and at least one pharmaceutically acceptable excipient.
[0396]
[0397] Effective amount is defined, for the purposes of the present invention, that amount of the compound that provides an objectively identifiable improvement in the patient's condition, recognized by a qualified observer, and wherein said patient is treated with a pharmaceutical composition comprising said amount of the compound.
[0398]
[0399] Pharmaceutically acceptable excipients, for the purposes of the present invention, are inert ingredients such as, but not limited to, co-solvents, surfactants, oils, humectants, emollients, preservatives, stabilizers and antioxidants.
[0400]
[0401] An embodiment of the present invention relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof:
[0402]
[0403]
[0404]
[0405]
[0406] where
[0407]
[0408] - R 1 is H or a protective group of a phenol;
[0409] - R 2 is independently selected from an alkyl ester group of structure:
[0410]
[0411]
[0412] j
[0413]
[0414] a derivative of a prenyl group of structure:
[0415]
[0416]
[0417]
[0418]
[0419] or an acyl group -C (O) ORb, where Rb is a C i-6 alkyl group,
[0420]
[0421] where
[0422]
[0423] - A is independently selected from an O atom, an S atom or an NR7 group, where R7 is independently selected from H, OR8 or a C1-6 alkyl group, and where R8 is an H group or a C 1- group 6 alkyl;
[0424]
[0425] - R5 is independently selected from H or a carboxylic acid protecting group;
[0426]
[0427] - R6 is C1-6 alkyl or OR9 where R9 is C1-6 alkyl;
[0428]
[0429] where
[0430]
[0431] - R3 is H or a C1-6 alkyl group; Y
[0432] - R4 is a C1-6 alkyl group or an alkoxide;
[0433] - R10 is independently selected from a C1-6 alkyl, allyl, alkylamine, alkylamide, alkylaryl, alkyl ether, haloalkyl, silyl, carbamate, alkylsulfone or haloalkylsulfone group; Y
[0434]
[0435] where when R1 is H and R3 is methyl, R5 is different from H or methyl, and when R1 is methyl and R3 is methyl, R5 is different from H or methyl; or a pharmaceutical composition comprising it, for use as a medicine.
[0436]
[0437] Another embodiment of the present invention relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof:
[0438]
[0439]
[0440] where
[0441]
[0442] - Ri is independently selected from the group consisting of H; C i -6 alkyl; allyl alkylamide; alkylamine; alkylaryl; alkyl ether; halo-alkyl; silyl; C (O) NRmRp, where Rm and / or Rp are each and independently H, C 1-6 alkyl or aryl; acyl C (O) Ra, where Ra is an aryl group; alkylsulfone or halo-alkylsulfone;
[0443] - R2 is independently selected from an alkyl ester group of structure:
[0444]
[0445]
[0446]
[0447]
[0448] a derivative of a prenyl group of structure:
[0449]
[0450]
[0451]
[0452]
[0453] or an acyl group -C (O) ORb, where Rb is a C1-6 alkyl group,
[0454]
[0455] where
[0456]
[0457] - A is independently selected from an O atom, an S atom or an NR7 group, where R7 is independently selected from H, OR8 or a C1-6 alkyl group, and where R8 is an H group or a C1-6 group I rent;
[0458] - R5 is independently selected from H; C1-6 alkyl; allyl alkylamide; alkylamine; alkylaryl; alkyl ether; halo-alkyl; silyl; C (O) NRmRp, where Rm and / or Rp are each and independently H, C1-6 alkyl or aryl; acyl C (O) Ra, where Ra is an aryl group; alkylsulfone or halo-alkylsulfone;
[0459] - R6 is C 1-6 alkyl or OR9 where R9 is C1-6 alkyl;
[0460]
[0461] where
[0462]
[0463] - R 3 is H or a C 1-6 alkyl group; Y
[0464] - R 4 is a C 1-6 alkyl group or an alkoxide;
[0465] - R 10 is independently selected from a C 1-6 alkyl, allyl, alkylamine, alkylamide, alkylaryl, alkyl ether, haloalkyl, silyl, carbamate, alkylsulfone or haloalkylsulfone group; Y
[0466]
[0467] where when R 1 is H and R 3 is methyl, R 5 is different from H or methyl, and when R 1 is methyl and R 3 is methyl, R 5 is different from H or methyl; or a pharmaceutical composition comprising it, for use as a medicine.
[0468]
[0469] An embodiment of the compound of formula I, or of a composition comprising it, for use as a medicament, as described above, comprises any of its enantiomers or mixtures thereof, and any of its E / Z isomers and mixtures thereof, and its pharmaceutically acceptable salts. One embodiment comprises the R enantiomer. Another embodiment comprises the S enantiomer. One embodiment comprises the E isomer. Another embodiment comprises the Z isomer.
[0470]
[0471] Another preferred embodiment relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof, which is independently selected from the group consisting of 3, 4, 4a, 4b, 4c, 6, 7, 8, 9, 10, 11, 12 and 13, or a pharmaceutical composition comprising it, for use as a medicine.
[0472]
[0473] The compounds of formula I described are agonists of PPARa and / or PPARy. Therefore, said compounds are useful in the treatment of diseases that respond to the administration of PPARa and / or PPARy agonists, that is, in diseases mediated by PPARa and / or PPARy agonists, which is equivalent to diseases whose treatment benefits from the administration of agonists of PPARa and PPARy.
[0474]
[0475] As indicated above, PPARs are considered powerful therapeutic tools in the control of cardiovascular disorders, type 2 diabetes (DT2), obesity, metabolic syndrome, hypercholesterolemia, hypertriglyceridemia, primary chylomicronemia, hyperlipoproteinemia, familial dysbetalipoproteinemia, inflammation and diseases neurodegeneratives, such as Parkinson's, Alzheimer's or amyotrophic lateral sclerosis (ALS) (Ahmadian et al., 2013). In addition, PPARs have an additional importance as they are involved in the control of inflammatory processes, so they can regulate inflammation, vascular function, and vascular remodeling.
[0476]
[0477] An embodiment of the present invention relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof:
[0478]
[0479]
[0480]
[0481]
[0482] where
[0483]
[0484] - R 1 is H or a protective group of a phenol;
[0485] - R 2 is independently selected from an alkyl ester group of structure:
[0486]
[0487]
[0488]
[0489] j
[0490]
[0491] a derivative of a prenyl group of structure:
[0492]
[0493]
[0494]
[0495]
[0496] or an acyl group -C (O) ORb, where Rb is a C 1-6 alkyl group,
[0497]
[0498] where
[0499] - A is independently selected from an O atom, an S atom or an NR7 group, where R7 is independently selected from H, OR8 or a C1-6 alkyl group, and where R8 is an H group or a C1-6 group I rent;
[0500] - R5 is H or a protecting group of a carboxylic acid;
[0501] - R6 is C 1-6 alkyl or OR9 where R9 is C1-6 alkyl;
[0502]
[0503] and where
[0504]
[0505] - R3 is H or a C1-6 alkyl group; Y
[0506] - R4 is a C1-6 alkyl group or an alkoxide; Y
[0507] - R10 is independently selected from a C1-6 alkyl, allyl, alkylamine, alkylamide, alkylaryl, alkyl ether, haloalkyl, silyl, carbamate, alkylsulfone or haloalkylsulfone group;
[0508]
[0509] or a pharmaceutical composition comprising it, for use in the treatment of a disease that responds to the administration of PPARa and / or PPARy agonists.
[0510]
[0511] In one embodiment, the disease that responds to the administration of PPARa and / or PPAR agonists is independently selected from cardiovascular disorders, type 2 diabetes (DT2), obesity, metabolic syndrome, hypercholesterolemia, hypertriglyceridemia, primary chylomicronemia, hyperlipoproteinemia, familial dysbetalipoproteinemia, inflammation. and neurodegenerative diseases.
[0512]
[0513] In one embodiment, cardiovascular disorders refer to atherosclerosis.
[0514]
[0515] In one embodiment, neurodegenerative diseases are independently selected from Parkinson's, Alzheimer's or amyotrophic lateral sclerosis (ALS).
[0516]
[0517] An embodiment of the present invention relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof:
[0518]
[0519]
[0520]
[0521]
[0522] where
[0523] - Ri is independently selected from the group consisting of H; C i -6 alkyl; allyl alkylamide; alkylamine; alkylaryl; alkyl ether; halo-alkyl; silyl; C (O) NRmRp, where Rm and / or Rp are each and independently H, C 1-6 alkyl or aryl; acyl C (O) Ra, where Ra is an aryl group; alkylsulfone or halo-alkylsulfone;
[0524] - R2 is independently selected from an alkyl ester group of structure:
[0525]
[0526]
[0527]
[0528]
[0529] a derivative of a prenyl group of structure:
[0530]
[0531]
[0532]
[0533]
[0534] or an acyl group -C (O) ORb, where Rb is a C1-6 alkyl group,
[0535]
[0536] where
[0537]
[0538] - A is independently selected from an O atom, an S atom or an NR7 group, where R7 is independently selected from H, OR8 or a C1-6 alkyl group, and where R8 is an H group or a C1-6 group I rent;
[0539] - R5 is independently selected from H; C1-6 alkyl; allyl alkylamide; alkylamine; alkylaryl; alkyl ether; halo-alkyl; silyl; C (O) NRmRp, where Rm and / or Rp are each and independently H, C1-6 alkyl or aryl; acyl C (O) Ra, where Ra is an aryl group; alkylsulfone or halo-alkylsulfone;
[0540] - R6 is C 1-6 alkyl or OR9 where R9 is C1-6 alkyl;
[0541]
[0542] and where
[0543]
[0544] - R3 is H or a C1-6 alkyl group; Y
[0545] - R4 is a C1-6 alkyl group or an alkoxide; Y
[0546] - Rio is independently selected from a group Ci-6 alkyl, allyl, alkylamine, alkylamide, alkylaryl, alkyl ether, haloalkyl, silyl, carbamate, alkylsulfone or haloalkylsulfone;
[0547]
[0548] or a pharmaceutical composition comprising it, for use in the treatment of a disease that responds to the administration of PPARa and / or PPARy agonists.
[0549]
[0550] In one embodiment, the disease that responds to the administration of PPARa and / or PPAR agonists is independently selected from cardiovascular disorders, type 2 diabetes (DT2), obesity, metabolic syndrome, hypercholesterolemia, hypertriglyceridemia, primary chylomicronemia, hyperlipoproteinemia, familial dysbetalipoproteinemia, inflammation. and neurodegenerative diseases.
[0551]
[0552] In one embodiment, neurodegenerative diseases are independently selected from Parkinson's, Alzheimer's or amyotrophic lateral sclerosis (ALS).
[0553]
[0554] One embodiment refers to a compound of formula I or a pharmaceutical composition comprising it, as described above, for use in the treatment of autoimmune diseases, such as type 2 diabetes (DT2).
[0555]
[0556] Another embodiment relates to a compound of formula I or a pharmaceutical composition comprising it, as described above, for use in the treatment of atherosclerosis. Another embodiment relates to a compound of formula I or a pharmaceutical composition comprising it, as described above, for use in the treatment of atherogenesis.
[0557]
[0558] Another embodiment relates to a compound of formula I or a pharmaceutical composition comprising it, as described above, for use in the treatment of inflammation and diseases related to inflammatory processes.
[0559]
[0560] Another embodiment relates to a compound of formula I or a pharmaceutical composition comprising it, as described above, for use in the treatment of hypercholesterolemia.
[0561]
[0562] Another embodiment relates to a compound of formula I or a pharmaceutical composition comprising it, as described above, for use in the treatment of hypertriglyceridemia.
[0563]
[0564] Another embodiment relates to a compound of formula I or a pharmaceutical composition comprising it, as described above, for use in the treatment of metabolic syndrome.
[0565] Another embodiment relates to a compound of formula I or a pharmaceutical composition comprising it, as described above, for use in the treatment of obesity.
[0566] Another embodiment relates to a compound of formula I or a pharmaceutical composition comprising it, as described above, for use in the treatment of primary chylomicronemia.
[0567]
[0568] Another embodiment relates to a compound of formula I or a pharmaceutical composition comprising it, as described above, for use in the treatment of hyperlipoproteinemia.
[0569]
[0570] Another embodiment relates to a compound of formula I or a pharmaceutical composition that
[0571] 0 comprises, as described above, for use in the treatment of dysbetalipoproteinemia.
[0572]
[0573] In one embodiment of the present invention when R 1 is H and R 3 is methyl, R 5 is different from H or methyl, and when R 1 is methyl and R 3 is methyl, R 5 is different from H or methyl.
[0574]
[0575] An embodiment of the compound of formula I, or of a composition comprising it, for use in the treatment of a disease that responds to the administration of PPARa and / or PPAR agonists, as described above, comprises any of its enantiomers or mixtures of these, and any of their E / Z isomers and mixtures thereof, and their pharmaceutically acceptable salts. One embodiment comprises the R enantiomer. Another embodiment comprises the S enantiomer. One embodiment comprises the E isomer. Another 0 embodiment comprises the Z isomer.
[0576]
[0577] Another embodiment of the present invention relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof, which is independently selected from the group consisting of II, III, IV or V:
[0578]
[0579] where Ri, R 4 , R 5 , R6 and Rb are defined as described above, or a pharmaceutical composition comprising it, for use in the treatment of a disease that responds to the administration of PPARa and / or PPAR agonists and .
[0580]
[0581] An embodiment of the compounds of formula II, III, IV or V, or of a composition comprising one of said compounds, for use in the treatment of a disease that responds to the administration of PPARa and / or PPAR agonists and , such As described above, it comprises any of its enantiomers or mixtures thereof, and any of its E / Z isomers and mixtures thereof, and their pharmaceutically acceptable salts.
[0582]
[0583] Another embodiment of the present invention relates to a compound of formula I, stereoisomers and pharmaceutically acceptable salts thereof, which is independently selected from a compound 1, 2, 3, 4, 4a, 4b, 4c, 5, 6, 7, 8, 9, 10, 11, 12 or 13, or a pharmaceutical composition comprising it, for use in the treatment of a disease that responds to the administration of PPARa and / or PPAR agonists and .
[0584]
[0585] Compound 5 refers to the compound of formula I:
[0586]
[0587] In one embodiment the compounds 1, 2, 3, 4, 4a, 4b, 4c, 5, 6 , 7, 8 , 9, 10, 11, 12 and 13, for use in the treatment of a disease that responds to administration of PPARa and / or PPARy agonists, described above, comprise the R enantiomer.
[0588]
[0589] In another embodiment the compounds 1, 2, 3, 4, 4a, 4b, 4c, 5, 6 , 7, 8 , 9, 10, 11, 12 and 13, for use in the treatment of a disease that responds to administration of PPARa and / or PPARy agonists, described above, comprise the S enantiomer.
[0590]
[0591] In one embodiment the compounds 1,2, 3, 4, 4a, 4b, 4c, 5, 6 , 7, 8 , 10, 11 and 12, for use in the treatment of a disease that responds to the administration of PPARa agonists and / or PPAR and , described above, comprise the isomer (E, Z).
[0592]
[0593] In another embodiment the compounds 1, 2, 3, 4, 4a, 4b, 4c, 5, 6 , 7, 8 , 10, 11 and 12, for use in the treatment of a disease that responds to the administration of PPARa agonists and / or PPARy, described above, comprise the isomer (E, E).
[0594]
[0595] In one embodiment the compounds 1,2, 3, 4, 4a, 4b, 4c, 5, 6 , 7, 8 , 10, 11 and 12, for use in the treatment of a disease that responds to the administration of PPARa agonists and / or PPARy, described above, comprise the isomer (Z, Z).
[0596]
[0597] In another embodiment the compounds 1, 2, 3, 4, 4a, 4b, 4c, 5, 6 , 7, 8 , 10, 11 and 12, for use in the treatment of a disease that responds to the administration of PPARa agonists and / or PPARy, described above, comprise the isomer (Z, E).
[0598]
[0599] In one embodiment, compounds 9 and 13, for use in the treatment of a disease that responds to the administration of PPARa and / or PPARy agonists, described above, comprise isomer E.
[0600]
[0601] In another embodiment, compounds 9 and 13, for use in the treatment of a disease that responds to the administration of PPARa and / or PPAR agonists, described above, comprise the Z isomer.
[0602] Another embodiment relates to the use of compounds of formula I in the treatment of diseases that respond to administration of PPAR agonists and / or PPAR.
[0603]
[0604] Another embodiment relates to a method of treatment comprising the administration of an effective amount of at least one compound of formula I, or of a pharmaceutical composition comprising it, to a patient of a disease that responds to the administration of PPARa agonists. and / or PPAR and .
[0605]
[0606] Another embodiment of the present invention relates to a compound of formula I independently selected from the group consisting of 1, 2, 3, 4c, 5, 7, 8, 9, 10, 11, 12 and 13, for use in the treatment of a disease that responds to the administration of dual agonists of PPARa and PPAR and .
[0607] In one embodiment, the disease that responds to the administration of dual PPARa / Y agonists is independently selected from cardiovascular disorders, type 2 diabetes (DT2), obesity, metabolic syndrome, hypercholesterolemia, hypertriglyceridemia, primary chylomicronemia, hyperlipoproteinemia, familial dysbetalipoproteinemia, inflammation and neurodegenerative diseases.
[0608]
[0609] In a more preferred embodiment, neurodegenerative diseases are independently selected from Parkinson's, Alzheimer's or amyotrophic lateral sclerosis (ALS).
[0610] Another embodiment relates to a treatment method comprising the administration of an effective amount of at least one compound of formula I independently selected from the group consisting of 1, 2, 3, 4c, 5, 7, 8, 9, 10 , 11, 12 and 13, or of a pharmaceutical composition comprising it, to a patient of a disease that responds to the administration of dual agonists of PPARa and PPARy.
[0611]
[0612] An embodiment of the invention relates to the process of synthesis of the compounds of formula I, as described above, wherein said process comprises:
[0613]
[0614] (a) condensing a 2,5-dihydroxyacetophenone and an alkyl ketoester to obtain a benzopyran-4-one of formula A;
[0615] (c) protect the phenol group from benzopyran-4-one to obtain a benzopyran ester of formula (B);
[0616]
[0617]
[0618]
[0619]
[0620] (d) reduce the ester group with suitable reagents to obtain an aldehyde;
[0621]
[0622]
[0623]
[0624]
[0625] (e) reacting the aldehyde group (C), obtained in (d), with a Grignard reagent to obtain an allyl alcohol;
[0626]
[0627]
[0628]
[0629]
[0630] and subjecting said allyl alcohol to the Johnson-Claisen transposition conditions obtaining a benzopyran ester (D);
[0631]
[0632]
[0633]
[0634]
[0635] where optionally,
[0636]
[0637] - the aldehyde (C) obtained in step (d) is subjected to:
[0638] (f) Horner-Wadsworth-Emmons olefination conditions using a phosphonate comprising a radical -CH (COORb) R 4 , or
[0639]
[0640] (g) Wittig olefining conditions using a phosphorus ilide comprising a radical -C (COORb) R 4 ;
[0641]
[0642]
[0643]
[0644]
[0645] - or where once step (e) has been performed, the benzopyran ester (D) obtained is subjected to step (d):
[0646]
[0647]
[0648]
[0649]
[0650] to obtain a new aldehyde (E); and where said new aldehyde (E) is subjected to
[0651] (f) - Horner-Wadsworth-Emmons olefination conditions using a phosphonate comprising a radical -CH (COOR 5 ) R 6 , or
[0652] (g) Wittig olefining conditions using a phosphorus ilide comprising a radical -C (COOR 5 ) R 6 ;
[0653]
[0654] and where, optionally, the phenol group, and / or the carboxylic group is subsequently deprotected in the compound resulting from step (f) or (g);
[0655]
[0656] and where R 1 , R 3 , R 4 , R 5 , R6, R 10 and Rb are defined as described above in the embodiments described in this invention.
[0657] By said synthesis process the compounds 1, 2, 3, 4, 4a, 4b, 4c, 5, 6, 7, 8, 9, 10, 11, 12 and 13 described above are obtained.
[0658]
[0659] Said synthesis procedure of the compounds of formula I can be schematized as follows:
[0660]
[0661] where:
[0662]
[0663] Reagents and conditions: (a) pyrrolidine, EtOH, 45 ° C, 24 h; (b) Zn, HCl-AcOH (2: 1, v / v), temp amb, 1 h; (c) R 1 X where X is a halogen, K 2 CO 3 , EtOH, reflux, 4 h; (d) 1M DIBAL-H (diisobutylaluminum hydride) in THF, -78 ° C, 20 min; (e) 1) R 4 -MgBr, - 78 ° C, 3 h; 2) MeC (OR 6 ) 3 , isobutyric acid (2 drops), 140 ° C, 2 h; (f) (g) tBuOK or NaH, THF, 0 ° C at room temperature, 16h.
[0664]
[0665] The formation of the chroman nucleus has been carried out, therefore, by (a) condensation of 2,5-dihydroxy acetophenone and alkyl ketoester, giving rise to benzopyran-4-one (Kabbe et al., 1982). Then, (b) the carbonyl group was reduced by reduction of Clemmensen, and (c) the phenol group was protected. Elongation of the side chain was performed (d) by reduction of the ethyl ester forming the corresponding aldehyde. Compound 13 is obtained by subjecting said aldehyde obtained in step (d), to an olefination by reaction of Wittig (f) or Horner-Wadsworth-Emmons (g) (Bisceglia et al., 2012 and Toshima et al., 2001 ).
[0666] To obtain compound 9 , (e) the aldehyde obtained in step (d) was treated, first, with a Grignard reagent generating an allylic alcohol that was subjected to the Johnson-Claisen transposition conditions (Sen et al., 1990) to finally obtain the ester of compound 9 . Compounds 1-8 and 10-12 are obtained by a second elongation of the side chain using again the conditions of step (d) described above to perform the reduction of the ester group of compound 9 forming the corresponding aldehyde, where said aldehyde is subjected to an olefination by reaction of Wittig (f) or Horner-Wadsworth-Emmons (g).
[0667]
[0668] DESCRIPTION OF THE FIGURES
[0669]
[0670] Figure 1 Effect on cell viability by the MTT assay of polycarrasoidol (1), policerasoidin (2), compound 8 , WY-14643 and rosiglitazone. In human neutrophils ( Fig. 1A ) and HUVEC ( Fig. 1B ). Data represented as mean ± SEM of n = 5 independent experiments. * p <0.05 or ** p <0.01 relative to the vehicle group.
[0671]
[0672] Figure 2 Effect on cell viability by MTT assay of the synthesized analog 9. In human neutrophils ( A ) and HUVEC ( B ). Data represented as mean ± SEM of n = 5 independent experiments. * p <0.05 or ** p <0.01 relative to the vehicle group.
[0673]
[0674] Figure 3. Effect of policerasoidol (1), policerasoidin (2), compound 8 , WY-14643 and rosiglitazone, on cell viability by flow cytometry. Percentage of apoptotic neutrophils ( Fig. 3A ), survival ( Fig. 3B ), apoptosis ( Fig. 3C ) and survival in HUVEC ( Fig. 3D ) after 24 h of incubation with compounds 1 , 2 , 8 , WY-14643 and rosiglitazone. Apoptotic cells were quantified as a percentage of the total population of late V + / PI-apoptotic annexes and / or necrotics such as annexin V + / PI +, and viable non-apoptotic cells such as annexin V '/ PI'. The columns are the mean ± SEM of n = 3 independent experiments. Representative flow cytometry panels ( Fig. 3E ) showing the effects of the vehicle and compounds 1 , 2 , 8 , WY-14643 and rosiglitazone on apoptosis / neutrophil survival have been included. * p <0.05 or ** p <0.01 relative to the vehicle group.
[0675]
[0676] Figure 4. Effect on cell viability of compound 9 by flow cytometry. Percentage of neutrophil apoptosis ( Fig. 4A ), survival ( Fig. 4B ), HUVEC apoptosis ( Fig. 4C ) and survival ( Fig. 4D ) after 24 h of incubation. Apoptotic cells were quantified as a percentage of the total annexin V + / PI 'population, late and / or necrotic apoptotics such as annexin V + / PI +, and viable non-apoptotic cells such as annexin V' / PI '. The columns are the mean ± SEM of n = 3 independent experiments. Representative flow cytometry panels ( Fig. 4E ) showing the effects of the vehicle and compound 9 on neutrophil apoptosis / survival have been included. * p <0.05 or ** p <0.01 relative to the vehicle group.
[0677]
[0678] Figure 5. Policerasoidol (1) inhibits the adhesion of mononuclear cells to HUVEC induced by TNFa under conditions of physiological flow. The cells were pretreated for 20 h with 1 pM of polycrasoidol (1), polycrasoidin (2), compound 8, WY-14643 or rosiglitazone, prior to stimulation with TNFa (20 ng / mL, 24 h). Freshly isolated human neutrophils or mononuclear cells were perfused on endothelial monolayers for 5 min at 0.5 dynes / cm2 and adhesion of neutrophils ( Fig. 5A ) and mononuclear cells ( Fig. 5B ) was quantified. The dose-response curves were performed for polycrasoidol (1) and rosiglitazone with HUVEC stimulated with TNFa (20 ng / mL, 24 h) and perfusion of freshly isolated human mononuclear cells ( Fig. 5C ). The results are the mean ± SEM of 4-6 independent experiments. ** p <0.01 relative to the vehicle group and np <0.01 relative to cells stimulated with TNFa.
[0679] Figure 6. Compound 9 inhibits the adhesion of mononuclear cells to HUVEC induced by TNFa under physiological flow conditions. Some cells were pretreated for 20 h with 3, 30 or 100 pM of compound 9 , prior to stimulation with TNFa (20 ng / mL, 24 h). Freshly isolated human neutrophils or mononuclear cells were perfused on endothelial monolayers for 5 min at 0.5 dynes / cm2 and adhesion of neutrophils ( Fig. 6A ) and mononuclear cells (Fig. 6B ) was quantified. Concentration-response curves (0.1-100 pM) were performed for compound 9 with HUVEC stimulated with TNFa (20 ng / mL, 24 h) and perfusion of freshly isolated human mononuclear cells ( Fig . 6C ). The results are the mean ± SEM of 4-6 independent experiments. ** p <0.01 relative to the vehicle group and + p <0.01 relative to cells stimulated with TNFa.
[0680] Figure 7. Endothelial cells were transfected with control siRNA or specific siRNA of RXRa, PPARa or PPAR. 48 h after transfection, the protein expression of RXRa ( Fig. 7A ), PPARa ( Fig. 7B ) or PPAR and ( Fig. 7C ) was determined by Western blot. The results (mean ± SEM of n = 4 independent experiments) are expressed in relation to p-actin. The images of representative gels are also shown. ** p <0.01 with respect to the values in the respective siRNA control group.
[0681]
[0682] Figure 8. The silencing of PPAR and or RXRa by siRNA stops the inhibitory effect of polycarrasoidol (1) in the interactions between endothelial cells-mononuclear leukocytes induced by TNFa. HUVEC were transfected with the control, specific siRNA of RXRa, PPARa or PPAR and .48h after transfection, the cells were pretreated with polychorasoidol (10 pM) or rosiglitazone (1 pM) for 20h and then stimulated with TNFa (20 ng / mL, 4h). Mononuclear cell adhesion was quantified. The results are the mean ± SEM of 3 4 independent experiments. * p <0.05 or ** p <0.01 relative to the respective vehicle group, fp <0.05 onp <0.01 relative to the respective TNFa stimulated cells ( Fig. 8A, 8B, 8C ). The RXRa / PPARY interaction was measured by immunoprecipitation of PPAR and followed by Western blotting for RXRa. Chemiluminescence films were quantified by image analysis. The blot represents a n = 3 of independent experiments ( Fig. 8D ).
[0683]
[0684] Figure 9. The RXRa / PPARY interaction of compound 9 was measured by immunoprecipitation of PPAR and followed by Western blotting for RXRa. Chemiluminescence films were quantified by image analysis. The blot represents a n = 3 of independent experiments.
[0685]
[0686] Figure 10. Policerasoidol (1) inhibits the expression in HUVEC of ICAM-1, VCAM-1 and CX 3 CL 1 induced by TNFa. Some cells were pretreated with polycarrasoidol (10 pM) 20 h, before stimulation with TNFa (20 ng / mL, 24 h for ICAM-1, VCAM-1 and CX 3 CL 1 ). The expression of ICAM-1 ( Fig. 10A ), VCAM-1 ( Fig. 10B ) and CX 3 CL 1 ( Fig. 10C ) was determined by flow cytometry. The results (mean ± SEM) were expressed as a mean fluorescence intensity of n = 3-6 independent experiments. The expression of ICAM-1, VCAM-1, and CX 3 CL 1 was also visualized in HUVEC not permeabilized by immunofluorescence, using alexa-fluor 488 (green) ( Fig. 10E ). The nuclei were contrasted with Hoechst dye (blue). * p <0.05 or ** p <0.01 relative to the vehicle group, fp <0.05 onp <0.01 relative to TNFa stimulated cells.
[0687]
[0688] Figure 11. Compound 9 inhibits the expression in HUVEC of VCAM-1, CX 3 CL 1 and CXCL16 induced by TNFa, but not ICAM-1. Some cells were pretreated with compound 9 (3 pM) 20 h, prior to stimulation with TNFa (20 ng / mL, 24 h). By cytometry of The expression of ICAM-1 ( Fig. 11A ), VCAM-1 ( Fig. 11B ), CX 3 CLI ( Fig. 11C ) and CXCL16 ( Fig. 11D ) was determined. The results (mean ± SEM) were expressed as a mean fluorescence intensity of n = 3-6 independent experiments. The expression of ICAM-1, VCAM-1, CX 3 CL 1 and CXCL16 was also visualized in HUVEC not permeabilized by immunofluorescence, using alexa-fluor 488 (green) ( Fig. 11E ). The nuclei were contrasted with Hoechst dye (blue). ** p <0.01 relative to the vehicle group, fp <0.05 onp <0.01 relative to cells stimulated with TNFa.
[0689]
[0690] Figure 12. Policerasoidol (1) inhibits the activation of p38 MAPK and NF- k B (p65) induced by TNFa in HUVEC. The cells were stimulated for 1 h with TNFa (20 ng / mL). Some cells were pretreated with polycarrasoidol (10 pM) 20 h before TNFa stimulation. P38 MAPK activation (Fig. 12A) and NF- k B (Fig. 12B) was determined by flow cytometry. The results (mean ± SEM) were expressed as the mean fluorescence intensity of n = 3-6 independent experiments ( Fig. 12C and 12D ). Representative histograms are shown. ** p <0.01 relative to the vehicle group fp <0.05 and np <0.01 relative to cells stimulated with TNFa.
[0691]
[0692] Figure 13. Compound 9 inhibits the activation of p38 MAPK and NF- k B (p65) induced by TNFa in HUVEC. The cells were stimulated for 1 h with TNFa (20 ng / mL). Some cells were pretreated with compound 9 (3 pM) 20 h before stimulation with TNFa. The activation of p38 MAPK ( Fig. 13A ) and NF- k B ( Fig. 13B ) was determined by flow cytometry. The results (mean ± SEM) were expressed as the mean fluorescence intensity of n = 3-6 independent experiments (Fig. 13C and 13D). Representative histograms are shown. ** p <0.01 relative to the vehicle group and fp <0.05 relative to TNFa stimulated cells.
[0693]
[0694] Figure 14. Compound 9 does not produce significant differences in weight gain (g) and feeding efficiency in ob / ob mice after 4 days of treatment. The histograms show the weight gain ( Fig. 14A ) that was expressed as the difference in weight between day 5 and day 0 (d 5 -d 0 ), and the food efficiency ( Fig. 14B ) that was calculated as the quotient between weight gain and food consumed. No significant differences in weight gain or feeding efficiency were observed between mice treated with compound 9 and control groups. The columns represent the mean ± SEM of n = 5-6 independent experiments.
[0695]
[0696] Figure 15. Compound 9 and rosiglitazone reduce plasma glucose values in ob / ob mice after 4 days of treatment. The variation of glucose with respect to d 0 has been represented, which has been calculated using the following formula: ((d 5 -d 0 ) / d 0 ) x 100. Columns represent the mean ± SEM of n = 5-6 independent experiments. * p <0.05 relative to the group of ob / ob mice treated with vehicle.
[0697]
[0698] Figure 16. Compound 9 does not produce significant differences in the weight gain of white adipose tissue (WAT) in ob / ob mice after 4 days of treatment. The columns show the mean ± SEM of n = 5-6 independent experiments
[0699]
[0700] Figure 17. Compound 9 significantly reduces the average area of WAT adipocytes in a dose-dependent manner in ob / ob mice after 4 days of treatment. Average area of adipocytes, in pm2 ( Fig. 17A ), and their respective representative images ( Fig. 17B ). The columns represent the mean ± SEM of n = 5-6 independent experiments. * p <0.05 or ** p <0.01 relative to the group of ob / ob mice treated with vehicle.
[0701]
[0702] Figure 18. Study of the anti-inflammatory effect on white adipose tissue (WAT) in ob / ob mice after 4 days of treatment. MCP-1 mRNA expression level in WAT ( Fig. 18A ) and Quantification of F4 / 80 + macrophages in WAT by immunohistochemistry ( Fig. 18B ). Compound 9 decreases MCP-1 mRNA expression and the number of macrophages infiltrated in white adipose tissue (WAT). The columns are the mean ± SEM of n = 5-6 independent experiments. * p <0.05 relative to the group of ob / ob mice treated with vehicle.
[0703]
[0704] Figure 19. Compound 9 and WY-14643 increase levels of total cholesterol and HDL-c, and decrease levels of triglycerides and free fatty acids in plasma. Total cholesterol values ( Fig. 19A ), HDL-c ( Fig. 19B ), triglycerides ( Fig. 19C ) and free fatty acids ( Fig . 19D ). The columns represent the mean ± SEM of n = 5-6 independent experiments . * p <0.05 or ** p <0.01 relative to the group of ob / ob mice treated with vehicle.
[0705]
[0706] Figure 20. Compound 9 did not significantly affect liver weight or accumulated triglycerides, compared to the control group (vehicle). Liver weight gain ( Fig. 20A ) and triglyceride levels in the liver ( Fig. 20B ). The columns represent the mean ± SEM of n = 5-6 independent experiments. * p <0.05 relative to the group of ob / ob mice treated with vehicle.
[0707]
[0708] Figure 21. Compound 9 reduced circulating levels of ALT and AST Levels of alanine amino transferase (ALT) and aspartic transaminase (AST) in plasma of ob / ob mice after 4 days of treatment. The columns represent the mean ± SEM of n = 5-6 independent experiments. * p <0.05 relative to the group of ob / ob mice treated with vehicle.
[0709] EXAMPLES
[0710]
[0711] The examples described below are illustrative and are not intended to limit the scope of the present invention. All research with human samples of this study has been carried out in compliance with the principles of the Declaration of Helsinki and with the approval of the institutional ethical Committee of the Clinical Hospital of Valencia (Valencia, Spain).
[0712]
[0713] Example 1: Obtaining compounds of formula I
[0714]
[0715] All reagents and solvents were purchased from commercial sources and used directly. All moisture sensitive reactions were carried out in oven dried flasks, under nitrogen atmosphere and with dry solvents. All reactions were monitored by analytical TLC with silica gel 60 F254 (Merck 5554) with UV detection at 254 nm. The residues were purified through a 60H silica gel column (5-40 pm, Merck 7736) and by flash chromatography (230-400 pm, Merck 9385). Preparative TLC was performed using 0.5 mm silica gel plates (Merck). Hewlett-Packard (HP-1100) or TripleTOF5600 (ABSciex) was used for LC-MS analysis, with the API source (atmospheric pressure ionization) configured as APCI (atmospheric pressure chemical ionization) or APIES (electrospray ionization) in positive or negative mode. NMR spectra ( 1 H,
[0716] 13 C, COZY 45, DEPT, HSQC and HMBC) were referenced for the CDCh solvent signal in a Bruker AC 300 or AC-500. The multiplicity of resonances of 13C NMR were assigned by DEPT experiments. NMR assignments were supported by COZY 45, DEPT, HSQC and HMBC experiments.
[0717]
[0718] 1.1. Obtaining compounds 1 (polycarrasoidol) and 2 (polycarrasoidin)
[0719]
[0720] Policerasoidol ( 1 ) and policerasoidin ( 2 ) were isolated from a methanolic extract of stem barks of Polyalthia cerasoides and P. sclerophylla and were identified based on their chromatographic and spectral characteristics (Gonzalez et al., 1995 and 1996) .
[0721]
[0722] 1.2. Obtaining compounds 3 to 8
[0723]
[0724] Compound 3 : To a suspension of polycrasoidol ( 1 ) (80 mg, 0.2 mmol) and K 2 CO 3 (80 mg) in acetone (2 mL), 1-bromopropane CH 3 CH 2 CH 2 Br (20 pL 0.2) was added mmol) and the mixture was stirred at reflux for 4 h. The reaction mixture was concentrated to dryness and re-dissolved in 10 mL of CH 2 Cl 2 . The organic phase was washed with brine (3 x 10 mL) and H 2 O (3 x 10 mL), dehydrated with anhydrous Na 2 SO 4 and evaporated to dryness on the rotary evaporator. The reaction crude was purified by silica gel column chromatography (hexane-AcOEt, 90:10) to obtain compound 3 (50 mg, 63%). 1H NMR (400 MHz, CDCh) or 6.49 (d, J = 2.6 Hz, H-7), 6.39 (d, J = 2.6 Hz, H-5), 5.91 (td, J = 1.3, 7.2 Hz, H- 7 ', 5.15 (td, J = 0.9, 7.1 Hz, H-3 '), 4.11 (t, J = 6.6 Hz, 2H, COOCH 2 CH 2 CH 3 - 9 '), 2.68 (t, J = 6.6 Hz, 2H, CH2-4), 2.57 (ddd, J = 1.2, 7.5, 15.1 Hz, 2H, CH 2 - 6 '), 2.12 (s, 3H, CH3-13'), 2.10-2.03 (m, 4H, CH 2 - 2 'and CH2-5'), 1.89 ( d, 3H, J = 1.6 Hz, CH 3 - 10 '), 1.75 (sex, J = 7.3 Hz, 2H, COOCH 2 CH 2 CH 3 - 9 '), 1.86-1.60 (m, 4H, CH 2 - 3 , CH 2 - 1 '), 1.59 (s, 3H, CH3-11'), 1.25 (s, 3H, CH3-12 '), 0.97 (t, J = 7.3 Hz, 3H, COOCH 2 CH 2 CH 3 - 9 '); 13C NMR (125 MHz, CDCl 3 ) 5 168.4 (COOCH 2 CH 2 CH 3 - 9 '), 147.9 (C-6), 145.8 (C-8a), 142.8 (CH-7'), 134.3 (C-4 '), 127.2 (C-8'), 127.0 (C-8), 124.9 (CH-3 '), 121.1 (C-4a), 115.6 (CH-7), 112.6 (CH-5), 75.2 (C -2), 65.8 (COOCH 2 CH 2 CH 3 - 9 '), 39.6 (CH 2 - 1 '), 39.0 (CH2-5 '), 31.3 (CH2-3), 27.9 (CH 2 - 6 '), 23.9 (CH3-12 '), 22.4 (CH2-4), 22.1 (COOCH 2 CH 2 CH 3 - 9 '), 22.0 (CH 2 - 2 '), 20.6 (CH3-10'), 16.0 (CH3-13 '), 15.7 (CH 3 -H'), 10.5 (COOCH 2 CH 2 CH 3 - 9 '); LC-MS (APCI positive mode) m / z 401.1 (100) [MH] +; HREIMS m / z 400.263746 [M] + (400.261360 calcd for C 25 H 36 O 4 ).
[0725]
[0726] Compound 4: To a suspension of polycarrasoidol ( 1 ) (80 mg, 0.2 mmol) and K 2 CO 3 (80 mg) in acetone (2 mL), ClCH 2 Ph benzyl chloride (25 pL 0.2 mmol) was added and the The mixture was stirred at reflux for 4 h. After usual extraction, the reaction crude was purified by silica gel column chromatography (hexane-AcOEt, 90:10) to obtain compound 4 (60 mg, 67%). 1H NMR (400 MHz, CDCh) 57.39-7.29 (m, 5H, COOCH2Pft-9 '), 6.50 (dd, J = 0.4, 2.7 Hz, H-7), 6.40 (dd, J = 0.4, 2.7 Hz, H -5), 5.95 (td, J = 1.3, 7.2 Hz, H-7 '), 5.20 (s, 2H, COOCHPh-9'), 5.12 (td, J = 1.1, 7.1 Hz, H-3 '), 2.70 (td, J = 2.5, 6.7 Hz, 2H, CH2-4), 2.57 (ddd, J = 1.3, 7.4, 15.2 Hz, 2H, CH 2 - 6 '), 2.14 (s, 3H, CH3-13' ), 2.12-1.90 (m, 4H, CH 2 - 2 'and CH2-5'), 1.92 (d, 3H, J = 1.6 Hz, CH3-10 '), 1.82-1.60 (m, 4H, CH2-3 and CH 2 - 1 '), 1.56 (s, 3H, CH 3 -H'), 1.26 (s, 3H, CH3-12 '); 13C NMR (125 MHz, CDCh) 5 167.9 (COOCH2Ph-9 '), 147.8 (C-6), 145.8 (C-8a), 143.6 (CH-7'), 134.1 (C-1 '' of COOCH2Pft-9 '), 134.3 (C-4'), 128.5 (2CH of COOCH 2 Pñ-9 '), 128.0 (3CH of COOCH2Pñ-9'), 127.3 (C-8 '), 126.7 (C-8), 124.8 ( CH-3 '), 121.2 (C-4a), 115.6 (CH-7), 112.6 (CH-5), 76.0 (C-2), 66.0 (COOCHPh-9'), 39.6 (CH 2 - 1 ') , 39.0 (CH2-5 '), 31.3 (CH2-3), 28.0 (CH 2 - 6 '), 24.0 (CH3-12 '), 22.4 (CH2-4), 22.1 (CH 2 - 2 '), 20.6 (CH3-10), 16.0 (CH3-13), 15.7 (CH 3 to 11 '); LC-MS (APCI positive mode) m / z 472.2 (100) [MH Na] +; EIMS m / z 357 [M-CH2Ph] +.
[0727]
[0728] Compound 4a: To a suspension of compound 4 (45 mg, 0.1 mmol) and K 2 CO 3 (45 mg) in acetonitrile (5 mL), 2-chloroacetamide ClCH 2 CONH 2 (0.1 mmol) was added and the mixture was stirred at reflux for 9 h. After usual extraction, the reaction crude was purified by silica gel column chromatography (hexane- AcOEt , 90:10) to obtain compound 4a (25 mg, 50%). 1H NMR (400 MHz, CDCh) 57.38-7.29 (m, 5H, COOCH2Pñ-9 '), 6.60 (d, J = 3.0 Hz, H-7), 6.46 (dd, J = 3.0 Hz, H-5), 5.93 (td, J = 1.3, 7.3 Hz, H-7 '), 5.18 (s, 2H, COOCHPh-9'), 5.11 (td, J = 1.2, 7.3 Hz, H-3 '), 4.41 (s, 2H, OCH 2 CONH 2 - 6 ), 2.72 (td, J = 2.5, 6.5 Hz, 2H, CH 2 -4), 2.56 (ddd, J = 1.2, 7.3, 14.8 Hz, 2H, CH 2 - 6 ') , 2.16 (s, 3H, CH3-13 '), 2.10-2.04 (m, 4H, CH 2 2 'and CH2-5'), 1.92 (d, 3H, J = 1.2 Hz, CH 3 -IO '), 1.81-1.58 (m, 4H, CH 2 - 3 , CH 2 -I'), 1.55 (s, 3H, CH 3 -II '), 1.26 (s, 3H, CH3-12'); 13C NMR (100 MHz, CDCl 3 ) 5 171.8 (OCH 2 CONH 2 - 6 ), 167.7 (COOCH2Ph-9 '), 149.8 (C-6), 147.0 (C-8a), 143.5 (CH-7'), 136.2 (C-1 '' of COOCH2Pft-9 '), 134.4 (C-4'), 128.4 (2CH of COOCH2Pft-9 '), 128.0 (3CH of COOCH2Pft-9'), 126.7 (C-8 '), 126.7 (C-8), 124.7 (CH-3 '), 121.2 (C-4a), 115.6 (CH-7), 112.0 (CH-5), 75.5 (C-2), 67.9 (OCH 2 CONH 2 - 6 ), 65.9 (COOCHPh-9 '), 39.6 (CH 2 - 1 '), 39.0 (CH2-5 '), 31.2 (CH2-3), 28.0 (CH 2 -6'), 24.0 (CH3-12 ' ), 22.6 (CH2-4), 22.1 (CH 2 - 2 '), 20.6 (CH3-10'), 16.2 (CH3-13 '), 15.7 (CH 3 - H'); HREIMS m / z 505.290785 [M] + (505.282824 calcd for C 31 H 39 NO 5 ).
[0729]
[0730] Compound 4b. To a suspension of compound 4 (35 mg, 0.1 mmol) and K 2 CO 3 (35 mg) in acetone (5 mL), ClCH 2 Ph benzyl chloride (10 pL, 0.1 mmol) was added and the mixture was stirred at reflux for 15 h. After usual extraction, the reaction crude was purified by silica gel column chromatography (hexane-AcOEt, 90:10) to obtain compound 4b (20 mg, 40%). 1H NMR (400 MHz, CDCh) 57.44-7.29 (m, 10H, OCH 2 Pñ- 6 , COOCH2Pñ-9 '), 6.66 (d, J = 3.0 Hz, H-7), 6.54 (dd, J = 3.0 Hz , H-5), 5.94 (td, J = 1.2, 7.2 Hz, H-7 '), 5.19 (s, 2H, COOCHPh-9'), 5.12 (td, J = 1.2, 7.2 Hz, H-3 ' ), 4.98 (s, 2H, OCHPh-6), 2.73 (td, J = 2.8, 6.7 Hz, 2H, CH2-4), 2.56 (ddd, J = 1.2, 7.2, 15.2 Hz, 2H, CH 2 - 6 '), 2.16 (s, 3H, CH3-13'), 2.11-2.04 (m, 4H, CH 2 - 2 'and CH2-5'), 1.92 (d, 3H, J = 1.2 Hz, CH 3 - 10 '), 1.81-1.60 (m, 4H, CH 2-3, CH 2-1'), 1.56 (s, 3H, CH3-11), 1.27 (s, 3H, CH3-12 '); 13C NMR (100 MHz, CDCh) 5167.0 (COOCH 2 Ph-9 '), 151.4 (C-6), 146.2 (C-8a), 143.5 (CH-7'), 137.6 and 136.2 (2C, C-1 ''of OCH 2 Pñ- 6 , C-1''' of COOCH2Pñ-9 '), 134.3 (C-4'), 128.5-127.5 (10CH, OCH2Pñ-6, COOCH2Pñ-9 '), 127.2 (C-8 '), 126.7 (C-8), 124.9 (CH-3'), 120.9 (C-4a), 115.7 (CH-7), 112.2 (CH-5), 75.3 (C-2), 70.5 (OCHP h -6), 65.9 (COOCHP h-9 '), 39.7 (CH 2 - 1 '), 39.0 (CH2-5 '), 31.4 (CH2-3), 28.0 (CH 2 - 6 '), 24.0 (CH 3 -12 '), 22.6 (CH 2 -4), 22.1 (CH 2 - 2 '), 20.6 (CH 3 -10 '), 16.2 (CH 3 -13'), 15.7 (CH 3 -H '); EIMS m / z 538 [M] + (70), 447 [M-CH 2 Ph] +, 356 (10) [M-2 x CH 2 Ph] +, 91 (100).
[0731]
[0732] Compound 4c. To a suspension of compound 4 (70 mg, 0.2 mmol) and K 2 CO 3 (70 mg) in acetone (5 mL), 1-bromopropane CH 3 CH 2 CH 2 Br (20 pL, 0.2 mmol) was added and the The mixture was stirred at reflux for 10 h. After usual extraction, the reaction crude with the protected phenol group was dissolved in 10 mL of a 20% (v / v) EtOH-KOH mixture and refluxed for 10 h. Subsequently, the reaction mixture was acidified with 1N HCl, concentrated to dryness and redissolved in 10 mL of CH 2 Ch. After usual extraction, the reaction crude was purified by silica gel column chromatography (hexane-AcOEt, 90 : 10) to obtain 4c (34 mg, 43%). 1H NMR (300 MHz, CDCh) 56.58 (d, J = 2.9 Hz, H-7), 6.45 (d, J = 2.9 Hz, H-5), 6.06 (td, J = 1.3, 7.3 Hz, H-7 '), 5.16 (td, J = 0.9, 6.6 Hz, H-3'), 3.83 (t, J = 6.6 Hz, 2H, OCH 2 CH 2 CH 3 - 6 ), 2.72 (t, J = 6.8 Hz, 2H, CH2-4), 2.62 (dd, J = 7.5, 14.4 Hz, 2H, CH 2 - 6 '), 2.15 (s, 3H, CH3-13'), 2.11-2.05 (m, 4H, CH 2 - 2 'and CH2-5'), 1.90 (d, 3H, J = 1.5 Hz, CH 3 - 10 '), 1.79-1.72 (m, 6H, OCH 2 CH 2 CH 3 - 6 , CH 2- 3 and CH 2 -I '), 1.60 (s, 3H, CH 3 - II'), 1.26 (s, 3H, CH 3 -12 '), 1.01 (t, J = 6.6 Hz, 3H, OCH 2 CH 2 CH 3 - 6 ); 13C NMR (75 MHz, CDCI 3 ) 5 173.0 (COOH-9 '), 151.6 (C-6), 146.3 (C-8a), 145.9 (CH-7'), 134.2 (C-4 '), 127.1 ( C-8 '), 126.1 (C-8), 125.1 (CH-3'), 120.9 (C-4a), 115.4 (CH-7), 111.8 (CH-5), 75.2 (C-2), 70.0 (OCH 2 CH 2 CH 3 - 6 ), 39.7 (CH 2 -1 '), 39.0 (CH2-5'), 31.4 (CH2-3), 28.1 (CH 2 - 6 '), 24.0 (CH3-12' ), 22.7 (CH2-4), 22.6 (OCH 2 CH 2 CH 3 - 6 ), 22.1 (CH 2 - 2 '), 20.5 (CH3-10'), 16.2 (CH3-13 '), 15.7 (CH 3 -H), 6.22 (OCH 2 CH 2 CH 3 to 6); LC-MS (APCI positive mode) m / z 424.2 (100) [MH Na] +.
[0733]
[0734] Compound 5. To a solution of polycarrasoidol ( 1 ) (10 mg, 0.03 mmol) in methanol (5 mL) was added dropwise conc HCl (1.5 mL) at 0 ° C. The mixture was stirred at reflux 7 h. After usual extraction, the reaction crude was purified by preparative silica gel TLC (hexane-AcOEt, 90:10) to obtain 5 (6 mg, 54%). 1H NMR (300 MHz, CDCh) 56.48 (d, J = 2.9 Hz, H-7), 6.38 (d, J = 2.9 Hz, H-5), 5.94 (td, J = 1.2, 7.4 Hz, H-7 '), 5.16 (td, J = 1.2, 7.4 Hz, H-3'), 3.73 (s, 3H, COOCH 3 - 9 '), 2.80-2.57 (m, 4H, CH2-4, CH 2 - 6 ' ), 2.35-2.00 (m, 4H, CH 2 - 2 'and CH2-5'), 2.12 (s, 3H, CH3-13 '), 1.90-1.82 (m, 4H, CH2-3, CH 2 - 1 '), 1.90 (d, 3H, J = 1.3 Hz, CH 3 -10'), 1.58 (s, 3H, CH3-11 '), 1.25 (s, 3H, CH3-12'); 13C NMR (75 MHz, CDCl 3 ) 5169.1 (COOCH 3 -9 '), 147.3 (C-6), 146.0 (C-8a), 144.8 (CH-7'), 134.2 (C-4 '), 127.2 ( C-8 '), 127.1 (C-8), 124.9 (CH-3'), 121.1 (C-4a), 115.5 (CH-7), 112.5 (CH-5), 75.1 (C-2), 51.1 (COOCH 3 - 9 '), 39.5 (CH 2 - 1 '), 39.0 (CH2-5 '), 31.3 (CH2-3), 27.9 (CH 2 - 6 '), 23.9 (CH3-12 '), 22.3 (CH2-4), 22.0 (CH 2 - 2 '), 20.5 (CH3-10'), 16.0 (CH3-13 '), 15.7 (CH3-11'); HREIMS m / z 372.236641 [M] + (372.230060 calcd for C23H32O4).
[0735]
[0736] Compound 6. To a suspension of polycarrasoidol ( 1 ) (100 mg, 0.3 mmol) and K 2 CO 3 (100 mg) in acetone (5 mL), Benzoyl PhCOCl chloride (25 pL, 0.2 mmol) was added and the mixture stirred at reflux for 2 h. After this time, methyl bromoacetate BrCH 2 COOCH 3 (20 pL, 0.2 mmol) was added and the mixture was stirred at reflux for 15 h. After usual extraction, the reaction crude was purified by silica gel column chromatography (hexane-AcOEt, 90:10) to obtain compound 6 (55 mg, 35%). 1H NMR (400 MHz, CDCh) 5 8.18 (dd, 2H, J = 1.5, 7.7 Hz, H-2 '' and H-6 '' of OCOPh-6), 7.62 (td, J = 1.5, 7.7 Hz, H-4 '' of OCOPh-6), 7.50 (td, 2H, J = 1.5, 7.7 Hz, H-3 '' and H-5 '' of OCOPh-6), 6.80 (dd, J = 0.8, 2.8 Hz, H-7), 6.75 (dd, J = 0.8, 2.8 Hz, H-5), 6.02 (td, J = 1.2, 7.2 Hz, H-7 '), 5.17 (td, J = 1.2, 7.2 Hz , H-3 '), 4.69 (s, 2H, COOCH 2 COOCH 3 - 9 '), 3.77 (s, 3H, COOCH 2 COOCH 3 - 9 '), 2.77 (m, 2H, CH 2 -4), 2.61 (m, 2H, CH 2 - 6 '), 2.18 (s, 3H, CH3-13'), 2.15-2.06 (m, 4H, CH 2 - 2 ', CH2-5'), 1.94 (d, 3H, J = 1.6 Hz, CH3-10 '), 1.80-1.62 (m, 4H, CH 2 - 3 , CH 2 - 1 '), 1.57 (s, 3H, CH 3 -H '), 1.30 (s, 3H, CH 3 -12 '); 13C NMR (100 MHz, CDCl 3 ) 5170.0-165.0 (3C, OCOPh-6 and COOCH 2 COOCH 3 - 9 '), 149.5 (C-6), 145.0 (CH-7'), 142.7 (C-8a), 134.4 (C-1 '' of OCOPh-6), 134.2 (C-4 '), 130.1-128.5 (5CH, of OCOPh-6), 127.4 (C-8'), 125.9 (C-8), 124.9 ( CH-3 '), 121. 2 (CH-7), 120.9 (C-4a), 119.2 (CH-5), 75.9 (C-2), 60.4 (COOCH 2 COOCH 3 - 9 '), 52.2 (COOCH 2 COOCH 3 - 9 '), 39.8 (CH 2 -I '), 39.0 (CH2-5' ), 31.0 (CH2-3), 28.0 (CH 2 - 6 '), 24.1 (CH 3 - I 2 '), 22.4 (CH2-4), 22.1 (CH 2 - 2 '), 20.5 (CH3-10' ), 16.1 (CH3-13 '), 15.7 (CH3-11'); HREIMS m / z 534.267390 [M] + (534.261754 calcd for C 32 H 38 O 7 ).
[0737]
[0738] Compound 7. To a suspension of polycarrasoidin ( 2 ) (20 mg, 0.05 mmol) and K 2 CO 3 (20 mg) in acetone (5 mL), BrCH dimethyl bromomalonate (COOCH 3 ) 2 (5 pL, 0.05 mmol) was added ) and the mixture was stirred at reflux for 6 h. After this time, methyl bromoacetate BrCH 2 COOCH 3 (20 pL, 0.2 mmol) was added and the mixture was stirred at reflux for 15 h. After usual extraction, the reaction crude was purified by preparative TLC (hexane-AcOEt, 80:20) to obtain compound 7 (17 mg, 67%). 1H NMR (300 MHz, CDCh) 56.56 (d, J = 2.8 Hz, H-7), 6.44 (d, J = 2.8 Hz, H-5), 6.08 (td, J = 1.3, 7.2 Hz, H-7 '), 5.62 (s, COOCH (COOCH3) 2-9'), 5.16 (t, J = 6.6 Hz, H-3 '), 3.83 (s, 6H, COOCH (COOCH3) 2-9'), 3.73 ( s, 3H, COOCH 3 - 6 ), 2.75 2.71 (m, 2H, CH2-4), 2.66-2.59 (m, 2H, CH 2 - 6 '), 2.18-2.05 (m, 4H, CH 2 - 2 ' and CH2-5 '), 2.15 (s, 3H, CH3-13'), 1.96 (d, 3H, J = 1.3 Hz, CH3-10 '), 1.90-1.73 (m, 4H, CH2-3, CH 2 - 1 '), 1.59 (s, 3H, CH3-11'), 1.26 (s, 3H, CH3-12 '); 13C NMR (75 MHz, CDCh) 5 169.0-167.2 (3C, COOCH (COOCH3) 2-9 '), 151.5 (C-6), 145.4 (C-8a), 144.4 (CH-7'), 133.7 (C -4 '), 126.6 (C-8'), 126.1 (COOCH (COOCH3) 2-9 '), 125.3 (C-8), 124.4 (CH-3'), 121.0 (C-4a), 115.0 (CH -7), 111.0 (CH-5), 75.4 (C-2), 56.1 (OCH 3 - 6 ), 52.1 (2C, COOCH (COOCH3) 2-9 '), 40.0 (CH 2 - 1 '), 39.0 (CH 2 -5 '), 31.3 (CH2-3), 28.3 (CH 2 - 6 '), 24.1 (CH3-12 '), 23.3 (CH2-4), 22.2 (CH 2 - 2 '), 20.3 ( CH3-10 '), 16.3 (CH3-13), 16.0 (CH 3 to 11'); HREIMS m / z 502.263390 [M] + (502.256669 calcd for C 28 H 38 O 8 ).
[0739]
[0740] Compound 8. To a suspension of polycarrasoidin ( 2 ) (20 mg, 0.05 mmol) and K 2 CO 3 (20 mg) in acetone (5 mL), methyl bromoacetate BrCH 2 COOCH 3 (5 pL, 0.05 mmol) was added and The mixture was stirred at reflux for 6 h. After usual extraction, the reaction crude was purified by preparative silica gel TLC (hexane-AcOEt, 80:20) to obtain compound 8 (15 mg, 67%). 1H NMR (300 MHz, CDCh) 56.56 (d, J = 2.7 Hz, H-7), 6.44 (d, J = 2.7 Hz, H-5), 6.00 (td, J = 1.5, 7.2 Hz, H-7 '), 5.15 (td, J = 1.5, 7.2 Hz, H-3'), 4.68 (s, 2H, COOCH 2 COOCH 3 -9 '), 3.76 (s, 3H, COOCH 2 COOCH 3 - 9 '), 3.73 (s, 3H, COOCH 3 - 6 ), 2.73 (t, 2H, J = 7.0 Hz, CH 2 -4), 2.59 (dd, 2H, J = 7.2, 14.8 Hz, CH 2 - 6 '), 2.15 (s, 3H, CH3-13 '), 2.10-2.04 (m, 4H, CH 2 - 2 ' and CH2-5 '), 1.93 (d, 3H, J = 1.4 Hz, CH3-10'), 1.79- 1.56 (m, 4H, CH2-3, CH 2-1 '), 1.59 (s, 3H, CH 3 -11), 1.26 (s, 3H, CH3-12'); 13C NMR (75 MHz, CDCh) 5168.8 and 167.4 (2C, COOCH 2 COOCH 3 -9 '), 152.5 (C-6), 146.4 (C-8a), 145.4 (CH-7'), 134.7 (C-4 '), 127.6 (C-8'), 126.3 (C-8), 125.4 (CH-3 '), 121.3 (C-4a), 115.2 (CH-7), 111.4 (CH-5), 75.7 (C -2), 60.8 (COOCH 2 COOCH 3 - 9 '), 56.0 (OCH 3 - 6 ), 52.6 (COOCH 2 COOCH 3 - 9 '), 40.1 (CH 2 - 1 '), 39.4 (CH2-5') , 31.8 (CH2-3), 28.4 (CH 2 6 '), 24.4 (CH3-12'), 23.1 (CH2-4), 22.6 (CH 2 - 2 '), 20.9 (CH 3 -IO'), 16.6 (CH3-13 '), 16.1 ( CH 3 - II '); HREIMS m / z 444.252007 [M] + (444.251189 calcd for C 26 H 36 O 6 ).
[0741] 1.3. Synthesis of compound 9:
[0742]
[0743]
[0744]
[0745]
[0746] (a) Synthesis of benzopiran-4-one: 2,5-dihydroxyacetophenone (0.5 g, 3.28 mmol), ethyl levulinate (0.46 mL, 3.28 mmol) and pyrrolidine (0.87 mL, 9.86 mmol), dissolved in absolute EtOH (10 mL) with 3-Á molecular sieve (100 mg) .56 * 10 The mixture was stirred at 45 ° C for 24 h under N 2 atmosphere. After this time, the reaction mixture was filtered, and the filtrate was diluted with CH 2 Cl 2 (100 mL) and washed with 1N HCl (3 x 50 mL), H 2 O (3 x 50 mL) and die salt (3 x 50 mL), dried over anhydrous Na 2 SO 4 and evaporated on the rotary evaporator. The residue was purified by silica gel column chromatography (hexane-AcOEt, 70:30) to obtain benzopyran-4-one (729 mg, 2.62 mmol, 80%) as a yellow oil. 1H NMR (400 MHz, CDCh) or 7.34 (d, J = 3.0 Hz, H-5), 7.07 (dd, J = 8.9, 3.0 Hz, H-7), 6.80 (d, J = 8.9 Hz, H- 8), 4.14 (q, J = 7.1 Hz, 2H, COOCH 2 CH 3 ), 2.78 (d, J = 16.7 Hz, 1H, CH2a-3), 2.63 (d, J = 16.7 Hz, 1H, CH2b-3 ), 2.53-2.46 (m, 2H, CH 2 - 2 '), 2.1 (m, 2H, CH 2 - 1 '), 1.38 (s, 3H, CH3-4 '), 1.25 (t, J = 7.1 Hz , 3H, COOCH 2 CH 3 ); 13 * 15C NMR (100 MHz, CDCh) or 193.0 (C-4), 173.3 (COOCH 2 CH 3 - 3 '), 153.7 (C-8a), 150.2 (C-6), 125.2 (CH-7), 120.1 (C-4a), 119.5 (CH-8), 110.6 (CH-5), 79.8 (C-2), 60.8 (COOCH 2 CH 3 ), 47.2 (CH2-3), 34.1 (CH 2 - 1 ' ), 28.7 (CH 2 - 2 '), 23.3 (CH3-4'), 14.1 (COOCH 2 CHs); EIMS m / z 278 [M: C i 5 HisO 5 ] + (35), 233 (25), 177 (100), 137 (75); HREIMS (%) m / z 278.11408 [M] + (278.11542 calcd for C 15 H 18 O 5 ).
[0747]
[0748] (b) Reduction of the ketone group: The benzopiran-4-one (0.4 g, 1.44 mmol) was dissolved in a mixture of AcOH-H 2 O (2: 1, v / v) (14 mL). Then, Zn powder (1.69 g, 25.92 mmol) and HClconc (9 mL) were added gradually over 30 min and the mixture was stirred for 1 h at room temperature. H 2 O (15 mL) was added to the reaction mixture and filtered and the filtrate was extracted with AcOEt (3 x 15 mL). The organic phases were combined, dried over anhydrous Na 2 SO 4 and evaporated on the rotary evaporator. The residue was purified by silica gel column chromatography (hexane-AcOEt, 85:15) to obtain the benzopyran ester (220.3 mg, 0.834 mmol, 58%) as a colorless oil. 1H NMR (300 MHz, CDCh) 56.55 (m, 3H, H-5, H-7, H-8), 4.12 (q, J = 7.1 Hz, 2H, COOCH 2 CH 3 ), 2.70 (m, 2H, CH 2 -4), 2.44 (t, J = 7.7 Hz, 2H, H-2 '), 2.05-1.70 (m, 4H, CH 2 - 3 , CH 2 - 1 '), 1.23 (s, 3H, CH 3 - 2), 1.24 (t, J = 7.1 Hz, 3H, COOCH 2 CH 3); 1 3 C NMR (75 MHz, CDCh) 5 174.1 (COOCH 2 CH 3 - 3 '), 148.8 (C-6), 147.4 (C-8a), 121.6 (CH-7), 117.8 (CH-8), 115.4 (C-4a), 114.6 (CH-5), 74.6 (C-2), 60.5 (COOCH 2 CH 3 ), 34.3 (CH2-3), 31.1 (CH 2 - 1 '), 28.8 (CH 2 - 2 '), 34.3 (CH2-3), 23.7 (CH 3 - 2 ), 22.1 (CH2-4), 14.1 (COOCH 2 CH 3 ); EIMS m / z 264 [M: C15H20O4] + (100), 218 (50), 163 (35), 123 (55). HREIMS (%) m / z 264.13562 [M] + (264.13616 calcd for C 15 H 20 O 4 ) (100).
[0749]
[0750] (c) O-protection of the benzopyran ester: A suspension of benzopyran ester (0.5 g, 1.89 mmol), p-flurobenzyl chloride pF (Ph) CH 2 Cl (0.3 mL, 2.46 mmol), K 2 CO 3 anhydrous (0.4 g, 2.83 mmol) in absolute EtOH (20 mL) was stirred at 65 ° C under N 2 atmosphere for 4 h. The reaction mixture was evaporated on the rotary evaporator, H 2 O (3 x 20 mL) was added and extracted with CH 2 Cl 2 (3 x 15 mL). The organic phases were combined, washed with 1M HCl (2 x 15 mL) and brine (2 x 15 mL), dehydrated with anhydrous Na 2 SO 4 and evaporated under reduced pressure. The residue was purified by silica gel column chromatography (hexane-AcOEt, 90:10) to obtain the O-protected benzopyran ester (556 mg, 1.49 mmol, 79%) as a colorless oil. 1H NMR (300 MHz, CDCh) 57.39 (m, 2H, H-2``, H-6 ''), 7.05 (m, 2H, H-3``, H-5 ''), 6.70 (m, 3H, H-5, H-7, H-8), 4.94 (s, 2H, pF-PhCH 2 O), 4.13 (q, J = 7.1 Hz, 2H, CO 2 CH 2 CH 3 ), 2.76 (t , 2H, J = 6.7 Hz, CH2-4), 2.48 (t, J = 7.7 Hz, 2H, CH 2 - 2 '), 1.91 (m, 4H, CH 2 - 3, CH 2 - 1 '), 1.24 (t, J = 7.2 Hz, 3H, CO 2 CH 2 CH 3 - 3 '); 13C NMR (75 MHz, CDCh) 5 173.6 (COOCH 2 CH 3 - 3 '), 162.3 (d, Jcf = 243.7 Hz, C-4''), 152.0 (C-6), 147.9 (C-8a), 133.1 (d, Jcf = 3 Hz, C-1 ''), 129.1 (d, 2C, Jcf = 8.3 Hz, C-2 '', C-6 ''), 121.4 (C-4a), 117.7 (CH -5), 115.2 (d, 2C, Jcf = 24.8 Hz, CH-3 '', CH-5 ''), 115.1 and 114.4 (CH-7 and CH-8), 74.6 (C-2), 69.9 ( pF-PhCH 2 O), 60.3 (CO 2 CH 2 CH 3 ), 34.3 (CH 2 - 1 '), 31.0 (CH2-3), 28.7 (CH 2 - 2 '), 23.6 (CH 3 - 2 ), 22.2 (CH2-4), 14.1 (CO 2 CH 2 CH 3 ); HREIMS m / z (%) 372.1730 [M] + (372.173688 calcd for C 22 H 25 O 4 F) (83).
[0751] (d) Reduction of the O- protected benzopyran ester An O-protected benzopyran ester solution (0.2 g, 0.54 mmol) in anhydrous CH 2 CI 2 (6 mL) at -78 ° C under N 2 atmosphere was stirred for 15 min. To this solution was added dropwise a solution of 1M diisobutylaluminum hydride (DIBAL-H) in THF (2.47 mL, 2.47 mmol). After 20 min, the reaction was stopped by adding MeOH (1 mL) and stirred for 15 min, and then a saturated aqueous solution of NH 4 Cl (1 mL) was added. The mixture was stirred for 1 h at room temperature, filtered and the filtrate was extracted with AcOEt (3 x 15 mL). The organic phases were combined, dried over anhydrous Na 2 SO 4 and evaporated under reduced pressure. The residue was purified by column chromatography (hexane-AcOEt, 90:10) to obtain the aldehyde (128 mg, 0.34 mmol, 63%) as a colorless oil. 1H NMR (300 MHz, CDCh) 5 9.78 (t, J = 1.6 Hz, 1H, CHO), 7.39 (m, 2H, H-2 '' and H-6 ''), 7.06 (m, 2H, H- 3 '' and H-5 ''), 6.73 (m, 3H, H-5, H-7, H-8), 4.94 (s, 2H, OCHPh-pF), 2.75 (m, 2H, CH2 -4), 2.60 (m, 2H, CH 2 -2 '), 1.90 (m, 4H, CH 2 - 3 , CH 2 - 1 '), 1.30 (s, 3H, CH 3 - 2 ); 13C NMR (75 MHz, CDCh) 5202 (CHO), 162.4 (d, J cf = 244.4 Hz, C-4``), 152.1 (C-6), 147.7 (C-8a), 133.1 (d, J cf = 3.3 Hz, C-1 ''), 129.3 and 129.2 (d, 2C, J cf = 8.3 Hz, C-2 '', C-6 ''), 121.4 (C-4a), 117.7 (CH-5 ), 115.2 (d, 2C, J cf = 24.8 Hz, CH-3 '', CH-5 ''), 115.2 and 114.5 (CH-7 and CH-8), 74.6 (C-2), 69.9 (OCH 2 Ph-pF), 38.4 (CH 2 -2 '), 31.8 (CH 2 - 1 '), 31.2 (CH2-3), 23.7 (CH 3 - 2 ), 22.3 (CH2-4); HREIMS m / z (%) 329.1552 [M] + (329.1547 calcd for C 20 H 21 O 3 F), 343.1710 (343.1704 calcd for C 21 H 23 O 3 F).
[0752]
[0753] (e) Grignard reaction and Johnson-Claisen transposition: A solution of the aldehyde intermediate (0.2 g, 0.61 mmol) in anhydrous THF (5 mL) was stirred at -78 ° C under N 2 atmosphere for 15 min, and then It was treated with a 0.5 M solution of isopropenylmagnesium bromide (7.32 mL, 3.66 mmol) (Sen et al., 1990) .7 The mixture was stirred at -78 ° C for 3 h. The reaction mixture was treated with a saturated NH 4 Cl solution and stirred for 15 min at room temperature. H 2 O was added and extracted with with AcOEt (3 x 15 mL). The organic phases were combined and the resulting organic phase was washed with H 2 O (3 x 15 mL) and dead salt (3 x 15 mL), dehydrated with anhydrous Na 2 SO 4 and concentrated on the rotary evaporator.
[0754]
[0755] The residue obtained (250 mg) was used in the next unpurified reaction. The residue was treated with 10 mL of triethyl orthoacetate and catalytic amounts of isobutyric acid (3 drops) (Sen et al., 1990) .7 The mixture was stirred at 140 ° C for 2 h. After cooling, the mixture was concentrated on the rotary evaporator to remove excess triethylortoacetate. The residue was diluted with CH 2 Cl 2 (20 mL) and washed with H 2 O (3 x 15 mL), dehydrated with anhydrous Na 2 SO 4 and concentrated on the rotary evaporator. The residue was purified by silica gel column chromatography (hexane / AcOEt, 98: 2) to obtain 126 mg of compound 9 (0.29 mmol, 47%). 1H NMR (500 MHz, CDCh) 5 7.39 (m, 2H, H-2 '' and H-6 ''), 7.06 (m, 2H, H-3 '' and H-5 ''), 6.73 (m , 3H, H-5, H-7, H-8), 5.14 (t, J = 7.0 Hz, 1H, CH-3 '), 4.94 (s, 2H, OCHgPh-pF), 4.11 (q, J = 7.4 Hz, 2H, CO OR CH 2 CH 3 ), 2.72 (t , J = 6.8, 2H, CH2-4), 2.39 (m, 2H, CH 2 - 6 '), 2.28 (m, 2H, CH2-5'), 2.08 (m, 2H, CH 2 - 2 '), 1.82 (m, 2H, CH2-3), 1.67 (m, 6H, CH 2 - 1 '), 1.60 (s, 3H, CH 3 - 4 '), 1.27 (s, 3H, CH 3 - 2 ), 1.23 (t, J = 7.3 Hz, 3H, CO OR CH 2 CH 3 ); 13C NMR (125 MHz, CDCl3) 5 173.4 (CO), 162.4 (d, J cf = 245 Hz, C-4``), 151.9 (C-6), 148.2 (C-8a), 133.5 (C-4 '), 133.3 (d, J cf = 3 Hz, C-1''), 129.2 (d, 2C, J cf = 8.3 Hz, C-2'',C-6''), 124.9 (C-3 '), 121.7 (C-4a), 117.8 (CH-5), 115.3 (d, 2C, J cf = 24.8 Hz, CH-3'',CH-5''), 115.2 and 114.4 (CH-7 and CH-8), 75.6 (C-2), 70.1 (OCH 2 Ph-pF), 60.2 (COOCH 2 CH 3 ), 39.2 (CH 2 - 1 '), 34.6 (CH 2 - 6 '), 33.2 (CH2 -5 '), 30.9 (CH2-3), 24.1 (CH 3 - 2 ), 22.4 (CH2-4), 22.2 (CH 2 -2'), 15.8 (CH 3 - 4 '), 14.2 (COO CH 2 CH 3 ); HREIMS m / z (%) 441.2441 [M] + (441.2436 calcd for
[0756] 27 H 33 FO 4 ).
[0757]
[0758] 1.4. Synthesis of compounds 10 to 12:
[0759]
[0760] Steps (a), (b), (c), (d) and (e) are performed as described in section 1.3 for compound 9, although the O-protection of the benzopyran ester is performed in the case of compounds 10 and 11 with benzyl chloride (R 1 = benzyl) and in the case of compound 12 with p-flurobenzyl chloride (R 1 = p-FluoroBn).
[0761]
[0762] Once step (e) has been carried out, a reduction of the ester group, obtained by transposition of Johnson-Claisen, is performed again to obtain an aldehyde, according to the conditions described in step (d) of section 1.3 .
[0763]
[0764] Compounds 10 and 12 are obtained by olefining the aldehyde obtained by carrying out the Wittig reaction with a phosphorus ilide (Ph) 3 P = C (CH 2 CH 3 ) (COOCH 2 CH 3 ) or the Horner-Wadsworth reaction -Emmons with a phosphonate (EtO) 2 P (O) -CH (CH 2 CH 3 ) (COOCH 2 CH 3 ). Compound 11 is obtained by hydrolysis of the ester group, by treatment with KOH at reflux of compound 10.
[0765]
[0766]
[0767] 1.5. Synthesis of compound 13:
[0768]
[0769] Steps (a), (b), (c) and (d) are carried out as described in section 1.3, wherein the O-protection of the benzopyran ester is carried out with benzyl chloride. The aldehyde obtained after step (d) is subjected to olefination by carrying out the Wittig reaction with a phosphorus ilide (Ph) 3 P = C (CH 2 CF 3 ) (COOCH 2 CH 3 ) or the Horner reaction. Wadsworth-Emmons with a phosphonate (EtO) 2P (O) -CH (CH2CF3) (COOCH2CH3).
[0770]
[0771]
[0772] In vitro studies
[0773]
[0774] Example 2: Evaluation of hPPARa and hPPARy transactivation
[0775]
[0776] Natural compounds 1 and 2 , compounds 3-8 and compound ( 9 ), were studied in vitro by PPARa / Y transactivation assays. A transcription assay was used on Cos-7 cells transiently transfected with a luciferase reporter plasmid in the presence of pGAL4hPPARa and pGAL4hPPARY expression vectors (Carmona etal., 2007). Cos-7 cells were obtained from the ATCC (CRL-1651). The cells were maintained under standard culture conditions (minimum essential Eagle medium modified by Dulbecco, DMEM) supplemented with 10% fetal bovine serum (FCS) at 37 ° C in an atmosphere of 5% humidity CO 2 . The medium was changed every 2 days. Cos-7 cells were seeded in 60 mm discs at a density of 5.5 x 105 cells / disc in DMEM supplemented with 10% FCS and incubated at 37 ° C for 16 h before transfection. The cells were transfected in DMEM, using jetPEI, with reporter (pG5-TK-pGL3) and expression plasmids (pGal4-h, hPPARa or hPPARY). The pCMV-pgalactosidase expression plasmid was co-transfected as a control for transfection efficiency. After 16 h the transfection was stopped by adding DMEM supplemented with 10% FCS, and the cells were trypsinized and seeded in 96-well plates and incubated for 6 h in DMEM containing 10% FCS. Subsequently, the cells were incubated for 24 h in DMEM containing 0.2% FCS and increasing concentrations of the compounds or the vehicle (DMSO, maximum concentration 0.1%) were tested. Finally, the cells were washed once with ice-cold PBS, lysed and luciferase and pgalactosidase assays were performed.
[0777]
[0778] To evaluate the compounds on PPAR transcriptional activity, the assays were carried out in a chimera of a human PPAR / Gal4 luciferase reporter gene system to determine the maximum transactivation response of each compound. The different compounds were tested in a concentration range of 0.001 to 10 pM. The results obtained from all the compounds were compared with the data of the reference compounds W Y-14643 and rosiglitazone in hPPARa and hPPARY, respectively.
[0779]
[0780] The results are summarized in Table 1 .
[0781] Table 1 hPPARa hPPARY
[0782] EC compound fifty (nM) Efficiency (%) CE fifty (nM) Efficiency (%)
[0783] Rosiglitazone 10000 15 4 100
[0784] W Y-14643 10000 100 NA NA
[0785] Policerasoidol, 1 184 107 84 95
[0786] Policerasoidin, 2 1000 82 1000 55
[0787] 3 10000 100 991 86
[0788] 4th 10,000 18 10,000 0
[0789] 4b 10000 28 10000 0
[0790] 4c salt 10000 191 1692 88
[0791] 5 10000 27 574 35
[0792] 6 10000 14 10000 0
[0793] 7 1616 76 10000 37
[0794] 8 10000 39 12 24
[0795] 9 300 251 400 57
[0796] 10 10,000 60 2000 183
[0797] 11 1500 96 300 62
[0798] 12 10,000 113 2000 183
[0799] 13 100 166 166 400 39
[0800] (a) Maximum activity obtained with each compound expressed as a percentage of maximum activity of WY-14643 for PPARa (EC 50 12 pM) and rosiglitazone for PPARy (EC 50 4 nM) respectively. (NA: not active on the hPPAR tested).
[0801]
[0802] Among all the compounds tested, the natural product polycrasoidol ( 1 ) showed the highest dual agonist activity PPARa / Y and its maximum response was comparable to that obtained by the reference compounds (WY-14643 and rosiglitazone). Data from EC 50 showed that natural compound 1 , and its semi-synthetic derivative 3 , with the free phenolic group, show total agonist activity on hPPARa and hPPARY, and high transactivation percentages (107-100% and 95-86% for hPPARa and hPPARY, respectively). Compound 4c , the O-propylated derivative with a free carboxylic function at C-9 ', showed the highest percentage of transactivation (191% and 88% for hPPARa and hPPARY, respectively). The other polycarrasoidol derivatives ( 4a , 4b and 5 ) showed lower transactivation percentages than compounds 1 , 3 and 4c at the concentrations tested. Modifications on the C-9 'carboxylic group of the side chain in position 2, introducing the COOCH group (COOCH 3 ) 2 gave rise to compound 7 with selectivity on PPARa activation (76% for hPPARa and 37% for hPPARY) . However, derivative 8 , with a COOCH 2 COOCH 3 group in C-9 ', showed lower PPARa activation (39% for hPPARa and 24% for hPPARY). Compound 9 also showed a dual agonist activity and a high transactivation percentage (251% for hPPARa and 57% for hPPARY, respectively).
[0803]
[0804] Compounds 10 , 11 , 12 and 13 also showed dual agonist activity PPARa and PPAR and , highlighting the potency of compound 11, with transactivation percentages of 96% and 62% for hPPARa and hPPARY respectively.
[0805]
[0806] It should be noted that compound 1 (natural) and 9 (synthetic) showed a potent dual agonist action of the PPARa and PPAR and receptors. Compound 2 had a moderate dual activity PPARa / Y and compound 8 shifted its selectivity towards PPARy activation ( Table 1 ).
[0807]
[0808] Example 3: Cytotoxicity studies in human neutrophils and endothelial cells of human umbilical cord veins (HUVEC)
[0809]
[0810] Previously it has been mentioned that there are important limitations in the use of PPAR agonists for use in humans, due to its adverse effects such as hepatotoxicity, cancer, cardiotoxicity or other cardiovascular effects. Glitazares are the most studied compounds with PPAR agonist activity, however, as we mentioned in the introduction, only the use of dual agonists is approved saroglitazar (Lipaglyn ™) in India since 2013 and lobeglitazone in Korea (DuvieTM) with antidiabetic and lipid lowering effects. Currently, there is a greater interest in developing dual PPARa and PPARy agonists that show fewer adverse effects than selective ao and ligands (Tan et al., 2017).
[0811]
[0812] In order to evaluate the possible adverse effects (cytotoxicity) of the compounds of the invention, compounds 1 , 2 and 8 as well as the reference compounds, were evaluated at concentrations of 30 and 100 pM in human neutrophils ( Fig. 1A ) and primary cell cultures of human umbilical cord vein endothelial cells (HUVEC) ( Fig. 1B ) by two different techniques: the MTT colorimetry assay and the flow cytometry analysis of cell apoptosis and survival. In addition, the synthesized compound 9 was evaluated at 10, 30 and 100 pM ( Fig. 2 ).
[0813]
[0814] MTT test. The viability of neutrophils and HUVEC was determined by the MTT colorimetry test (3 (4,5-dimethylthiazol-2-yl) -2,5-diphenyltetrazolium bromide).
[0815]
[0816] Neutrophils were obtained from the buffy coats fraction of healthy donors by centrifugation with Ficoll-Hypaque density gradient.
[0817]
[0818] Human umbilical cord vein endothelial cells (HUVEC) were isolated by collagenase treatment and maintained in human specific endothelial basal medium (EBM-2) supplemented with endothelial growth medium (EGM-2) and 10% FCS. Pass 1 cells were grown to confluence in 24-well plates. Before starting each experiment, the cells were incubated 24 h in medium containing 1% FBS.
[0819]
[0820] 100 pL of neutrophils and the HUVEC suspension in supplemented RPMI medium (2 x 105 cells / mL) were added to each well of a 96-well plate. The cells were incubated in the absence and / or presence of the compounds at 37 ° C for 24h. A solution of MTT (2 mg / mL in PBS) was prepared. 100 pL of the MTT solution was added to each well and incubated at 37 ° C for another 3 h. The supernatants were discarded and 200 pL of DMSO was added to each well to dissolve the formazan precipitate. The optical densities at two wavelengths (560 and 630 nm) were measured in a spectrophotometer (Infinite M200, Tecan, Mannedorf, Switzerland).
[0821]
[0822] All compounds showed significant cytotoxicity at 100 pM on both types of cells ( Figures 1 and 2 ). Policerasoidin ( 2 ) and derivative 8 also presented toxicity at 30 pM in neutrophils and HUVEC, and WY-14643 produced cytotoxicity at 30 pM (9%) but only in human neutrophils ( Fig. 1 ). The synthesized compound 9 showed only cytotoxicity in HUVEC at 30 pM ( Fig. 2 ) but 12% with respect to the vehicle.
[0823] In addition, the rate of apoptosis and survival in human neutrophils and HUVEC was determined by double staining with annexin V and propidium iodide (PI) by flow cytometry.
[0824] Cytofluorometry analysis of apoptosis and survival. The FITC Annexin V Apoptosis Detection kit (BD Bioscience, San Jose, CA) was used. The staining protocol and all reagents were supplied in the commercial kit. The HUVEC were separated from the culture flasks by acutase (StemPro® Accutase® Cell dissociation reagent, Thermofischer Scientific Waltham, MA). Both cell types were incubated at 37 ° C for 24 hours in the presence or absence of the compounds to be tested. Freshly isolated confluent HUVEC and neutrophils were washed, resuspended in 1x binding buffer (1 x 106 cells / mL) and stained with annexin V conjugated with FITC and propidium iodide (PI), as described in the manufacturer's instructions. The cells were analyzed on a BD FACS Verse Flow Cytometer flow cytometer (BD Biosciences, San Jose, CA) and differentiated as early apoptosis cells (annexin V + and PI-), late apoptosis and / or necrotic (annexin V + and PI +) , and living cells (annexin V- and PI-).
[0825]
[0826] In neutrophils, only compound 8 and rosiglitazone at the dose of 100 pM caused a minimal effect on cell survival (5.3% and 8.5% respectively) ( Figure 3B) . Synthesis compound 9 also caused a minimal effect on neutrophil survival at doses of 30 and 100 pM (1.07% and 1.55%, respectively), compared to the vehicle ( Figure 4B ). On the contrary, all compounds increased apoptosis significantly and decreased survival from 30 pM in HUVEC cells, with the exception of policerasoidol ( 1 ) ( Figure 3C and D ) and compound 9 ( Figure 4C and D ). In HUVEC, the most toxic compounds were policerasoidin ( 2 ), its analogue 8 and rosiglitazone, which decreased cell survival by 38.9, 61.3 and 79.2%, respectively, to 100 pM and exerted some significant effects at 30 pM ( Figure 3C and D ). Note the absence of toxicity of compound 9 in HUVEC at the doses tested, including at 100 pM, compared to the vehicle ( Figure 4 C and D ).
[0827]
[0828] In the introduction we mentioned the limited use in humans of numerous PPARa and PPAR agonists and due to toxicity problems that lead to significant adverse effects. In fact, despite the fact that rosiglitazone has numerous beneficial effects in patients with type 2 diabetes, the evidence of its cardiovascular adverse effects that include increased risk of myocardial infarction and heart failure, has forced the withdrawal of the drug in many countries of the European Community (Palee et al., 2011). Thus, toxicity studies are important in this type of compounds and through MTT and flow cytometry tests we have shown that compounds 1 and 9 are less cytotoxic than the specific reference agonists for PPARa and PPARy.
[0829] Example 4: Effect of compounds 1, 2, 8 and 9 on the leukocyte-endothelium interaction induced by TNFa under flow conditions
[0830]
[0831] The endothelium that covers the blood vessels internally has several functions, whose alterations ("endothelial dysfunction") have been related to cardiovascular diseases, thromboembolism and atherosclerosis. In fact, endothelial dysfunction is one of the first manifestations of the onset of atherosclerosis that leads to a proinflammatory and prothrombotic phenotype of the endothelium, causing the release and migration of leukocytes into the subendothelial space. PPARs, in particular PPARa and PPARy, are expressed in most vascular cells, including endothelial and smooth muscle cells, where they participate in specific anti-inflammatory effects and lipid control (Rosenson et al., 2012) .12 PPARy agonists (rosiglitazone) are able to inhibit the inflammatory process, decreasing the accumulation of neutrophils and mononuclear cells (Palee et al., 2011).
[0832] The parallel flow chamber technique was used to visualize cell adhesion under dynamic physiological flow conditions.
[0833]
[0834] In carrying out this study, neutrophils and mononuclear cells were perfused through HUVEC monolayers previously stimulated or not with TNFa (20 ng / mL) for 24 h. For this, human neutrophils and mononuclear cells were obtained in suspension from healthy volunteers using the Ficoll-Hypaque method of density gradient centrifugation. Freshly isolated cells (1 x 106 / mL) were perfused through the endothelial cell monolayer (HUVEC) stimulated or not, with TNFa for 24 h. In all experiments, leukocyte interactions were determined after perfusing for 5 min at 0.5 dynes / cm2. The HUVEC plates were placed in the flow chamber (GlycoTech, Gaithersburg, MD) and a camera placed on an inverted phase contrast microscope (Axio Observer A1 Carl Zeiss microscope, Thornwood, NY) was coupled to visualize cell adhesion ( 20x objective and a 10x eyepiece). In addition, the images were recorded on video for later analysis.
[0835]
[0836] The HUVECs were pretreated with polycarrasoidol ( 1 ), polycarrasoidin ( 2 ), compound ( 8 ), or compound 9 at doses of 1, 3, 10, 30 and 100 pM, as well as WY-14643 or rosiglitazone at 1 pM, 20 hours before stimulation with TNFa (Sanz et al., 2012).
[0837]
[0838] TNFa caused a significant increase in adhesion to endothelial cells of mononuclear leukocytes and neutrophils ( Figures 5 and 6 ). None of the compounds significantly inhibited HUVEC-neutrophil interactions at the concentration of 1 pM, however, it should be noted that pretreatment with polycyrasoidol ( 1 ), synthesized analog 9 and rosiglitazone significantly decreased HUVEC-mononuclear cell adhesion ( Figures 5 and 6 ) in a concentration-dependent manner ( Figures 5C and 6B ). In this test, policerasoidol ( 1 ) was 5 times less potent than rosiglitazone, while compound 9 showed a potency of approximately 3.6 and 16 times greater than rosiglitazone and polychorasoidol, respectively (IC 50 values of 0.30 pM vs. 1.1 pM and 4.9 pM, respectively). Taking into account these results and the cytotoxicity studies on apoptosis and survival in neutrophils, we can say that policerasoidol ( 1 ) and the synthesized compound 9 could act by inhibiting the recruitment of mononuclear cells without compromising the immune response of neutrophils, necessary for defense of the host Therefore, it seems that both compounds are interesting candidates for further immuno-pharmacological evaluation, due to the ability of these compounds to reduce the recruitment of mononuclear cells, the power as dual agonists of PPARa / Y and the low cytotoxicity.
[0839]
[0840] Example 5: Policerasoidol (1) and compound 9 inhibited mononuclear cell adhesion to the endothelium, induced by TNFa via RXRg / PPARv interaction
[0841] Policerasoidol ( 1 ) and the synthesized compound 9 are dual agonist ligands PPARa / Y and rosiglitazone is a selective agonist PPARy. To suppress PPARa or PPARy expression in endothelial cells, we have applied a gene silencing approach using RNA interference (siRNA).
[0842]
[0843] Confluent HUVEC were transfected with siRNAs from RXRa, PPARa, specific PPARs (Dharmacon, Lafayette, CO) using lipofectamine RNAiMAX (Invitrogen, Carslbad, CA) for 48 h (Sanz et al., 2012) .13 Next, the cells were they treated for 20 h with polycrasoidol ( 1 ) at 10 pM or rosiglitazone at 1 pM and then stimulated for 4 h with TNFa (10 ng / mL). In order to confirm the PPARa, PPARy and RXRa silencing via siRNA, the analysis was performed by Western blot (Sanz et al., 2012) .13 After silencing, the cells were washed, scraped, collected and centrifuged at 15,000 g at 4 ° C for 30 min to obtain the total protein extract. The protein concentration was determined by the Bradford method. The samples were denatured, subjected to polyacrylamide gel electrophoresis with 10% SDS (SDS-PAGE) and transferred to a nitrocellulose membrane. The membranes were blocked with 5% BSA-PBS with 0.05% Tween 20 and incubated with the corresponding antibody. Polyclonal rabbit antibodies against RXRa and PPARy (H-100) and mouse monoclonal antibody against PPARa were purchased from Santa Cruz Biotechnology (dilution: 1: 400, Santa Cruz, CA). Secondary anti-rabbit or anti-mouse IgG antibodies bound to horseradish peroxidase were supplied by DakoCytomation (dilution 1: 2000, Denmark).
[0844] After 48 hours of transfection with specific siRNA of PPARa or PPARy, the HUVEC showed> 68% and 66% protein reduction, respectively, compared to the levels detected in control cells treated with siRNA ( Figure 7 ). In the control of transfected cells-siRNA, stimulation with TNFa induced significant increases in the adhesion of mononuclear cells that were inhibited when endothelial cells were pretreated with polycarrasoidol ( 1 ) or rosiglitazone ( Figure 8A and B ). It should be noted that, while these responses were not affected by PPARa silencing ( Figure 8A ), the responses induced by policerasoidol ( 1 ) were canceled in PPARy-deficient HUVEC ( Figure 8B ). PPARs form heterodimers with RXR that in some cases respond synergistically to both agonists, RXR and the receptor. Since PPARy can interact with RXRa to produce its anti-inflammatory activity (Sanz et al., 2012), the directed deletion of RXRa expression in HUVEC with specific RXRa siRNA (> 72% protein reduction, Figure 7 ) also suppressed completely the inhibitory effect of polycarrasoidol ( 1 ) on the adhesion of mononuclear cells induced by TNFa ( Figure 8C ).
[0845]
[0846] In addition, to confirm the interaction between PPAR and RXRa, the HUVECs were immunoprecipitated with an anti-PPARY antibody and subjected to electrophoresis and westernblot with an anti-RXRa antibody, and the increase in the RXRa / PPARY interaction in those cells was detected. pretreated with polychorasoidol ( 1 ) or rosiglitazone ( Figure 8D ).
[0847]
[0848] For immunoprecipitation, protein extracts were obtained in 50 mM Tris-HCl (pH 8), 150 mM NaCl, 1% Nonidet P-40 and protease (1 mM PMSF, 40 pg / mL aprotinin and 40 pg / mL leupeptin) and phosphatase inhibitors Then, protein extracts (~ 200 pg) were incubated with 5 pg of antibody against PPARy. Immunocomplexes were precipitated using an anti-rabbit IgG beads (cat # 8800, eBioscience, San Diego, CA) and suspended in sample buffer with 50 mM dithiothreitol (DTT). Western blotting was carried out with an antibody against RXRa and the membranes were incubated with the secondary HRP-conjugate Rabbit TrueBlot (eBioscience, San Diego, CA) as a secondary antibody. Chemiluminescence signals were developed with ECL (GE Healthcare, Madrid, Spain).
[0849]
[0850] This same immunoprecipitation experiment was carried out with the synthesis compound 9 ( Figure 9 ). Taking into account the results, we can conclude that polychorasoidol ( 1 ) and compound 9 do not require the activation of PPARa to inhibit the recruitment of mononuclear cells, but they need the activation of RXRa and PPARy to exert their anti-inflammatory effect, as occurs with others PPAR and RXRa selective agonists (Sanz et al., 2012).
[0851] Example 6: Policerasoidol (1) and synthesized compound 9 decrease the expression of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and fractalkine (CX 3 CLI). TNFa induced
[0852]
[0853] In cardiovascular and cardiometabolic diseases (hypertension, obesity, diabetes, metabolic syndrome) high levels of circulating mediators have been detected, among others, cytokines and chemokines that may reflect alterations in endothelial function. Pro-inflammatory cytokines stimulate the endothelium to express cell adhesion molecules (CAM), such as vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1), increasing leukocyte adhesion to vascular endothelial cells, which constitutes an important process of the initial stage of inflammatory and immunological responses (Zernecke et al., 2010). PPAR and is expressed in vascular tissues and leukocytes, and its activation may decrease the expression of CAMs such as ICAM-1 or VCAM-1, as well as the expression of CX 3 CL 1 (fractalkine). CX 3 CL 1 is a chemokine that is expressed on the surface of endothelial cells, and can bind to its CX 3 CR 1 receptor, expressed in monocytes, T lymphocytes and NK cells, promoting cell adhesion. CX 3 CL 1 can also be released from the cell surface by the action of ADAM10 and ADAM17, releasing a soluble form capable of acting as a chemoattractant for leukocytes expressing its receptor. In addition to CX 3 CL 1 , there is another transmembrane chemokine, CXCL16, which, like CX 3 CL 1 , is also expressed in endothelial cells and can be released by the action of ADAM10. It exerts its action interacting with its CXCR6 receptor, present in certain leukocyte subtypes, promoting their adhesion to the endothelial surface. Thus, these chemokines and adhesion molecules regulate the adhesion and migration of leukocytes to the vascular wall, and there are indications that an increase in their endothelial expression is associated with an increase in atherosclerosis (Zernecke et al., 2008).
[0854]
[0855] To investigate the expression of VCAM-1, ICAM-1, fractalkine (CX 3 CL 1 ) and CXCL16, HUVEC were pretreated with polychorasoidol ( 1 ) (10 pM) or compound 9 (3 pM) for 24 h, and stimulated with TNFa (20 ng / mL) for an additional 24 h. Cells were collected from the bottles using accutase (StemPro® Accutase® Cell Dissociation Reagent, ThermoFischer Scientific, Waltham, MA). Then, they were washed and incubated for 1 h with an APC-conjugated mAb antibody against human VCAM-1 (100 pg / mL; STA clone, BioLegend, San Diego, CA), FITC-conjugated mAb against human ICAM-1 ( 400 pg / mL; clone HA58, BioLegend, San Diego, CA), PE-conjugated mAb versus human CX 3 CLI (1.25 pg / mL; clone 51637, R&D Systems, Minneapolis, MN) or with an APC monoclonal antibody -conjugated against human CXCL16 (clone 256213, R&D Systems, Abingdon, UK), all of them at 1:25 dilution in 3% BSA / PBS. Samples were analyzed in a parallel flow cytometer (FACSVerse, BD Biosciences, San Jose, CA).
[0856]
[0857] In this study we have shown that the pretreatment of HUVEC with policerasoidol ( 1 ) for 20 h at 10 pM significantly reduced the expression of ICAM-1 and VCAM -1, induced by TNFa ( Fig. 10) , the most pronounced effect on VCAM - 1 (87.8% inhibition) than ICAM-1 (23.8% inhibition). To investigate the effect of policerasoidol ( 1 ) on the expression of fractalkine (CX 3 C LI), the cells were stimulated with TNFa and the responses were evaluated by flow cytometry and immunofluorescence after 24 h.
[0858]
[0859] The results showed that polycrasoidol ( 1 ) at 10 pM reduced the expression of this chemokine by 77.6% ( Figure 10C ). The expression of adhesion molecules such as VCAM-1, ICAM-1 or chemokines such as CX 3 CL 1 or CXCL16, was also determined for compound 9 to 3 pM, which showed a significant decrease in the expression of VCAM -1, CX 3 CL 1 and CXCL 16 ( Figure 11 ).
[0860]
[0861] To carry out the confluent HUVEC immunofluorescence assays, they were cultured on crystals and some cells were incubated 24 h with polychorasoidol ( 1 ) (10 pM) or compound 9 (3 pM), before being treated with TNFa (20 ng / mL) during Additional 24 h. Then, the cells were washed twice with PBS (1x), fixed with 4% paraformaldehyde and blocked in PBS solution (1x) containing 1% BSA. HUVEC were incubated at 4 ° C overnight with a mouse primary monoclonal antibody against human VCAM -1 (1: 200 dilution; clone 1.G 11B 1, Serotec, Kidlington, UK), ICAM-1 (dilution 1 : 200; clone 6.5B5, Serotec, Kidlington, UK), CX 3 CL 1 (1: 200 dilution; clone 81506, R&D Systems, Minneapolis, MN) or CXCL 16 (1: 200 dilution; Immunostep, Salamanca, Spain) in a 0.1% BSA / PBS solution, followed by incubation with a secondary antibody alexa-fluor 488-goat anti-rabbit conjugate (1: 1000 dilution; A11034, Life Technologies, Carlsbad, CA) at room temperature for 1 h . Cell nuclei were contrasted with Hoechst (diluted 1: 4000 in PBS, Sigma-Aldrich, Madrid, Spain). Fluorescence microscope images (Axio Observer A1, Carl Zeiss, Thornwood, NY) equipped with a 40x lens objective and a 10x eyepiece were captured.
[0862]
[0863] Example 7: Policerasoidol (1) and compound 9 inhibit the activation of p38 MAPK and NF-
[0864] kB induced by TNFa in HUVEC
[0865]
[0866] Numerous evidence has revealed that PPAR ligands are capable of modulating MAPK signaling pathways (Park et al., 2011). Therefore, we have investigated the intracellular signaling pathways that underlie the inhibitory responses shown by polycerasoidol ( 1 ) and the synthesized compound 9 .
[0867] The effect of polychorasoidol ( 1 ) or compound 9 on the phosphorylation of p38 MAPK and p65 (NF-
[0868] kB) induced by TNFa was determined in HUVEC by flow cytometry. HUVEC were previously incubated for 24 h with compound 1 (10 pM) or 9 (3 pM) and stimulated for 1 h with TNFa (20 ng / mL). Endothelial cells were fixed with BD Cytofix Fixation Buffer (BD Biosciences, San Jose, CA) and permeabilized with BD Sol Perm III solution (BD Biosciences, San Jose, CA) and stained sequentially with a 1:10 dilution of an antibody PE-conjugated mAb against human p65 (pS529, clone K10-895.12.50; BD Biosciences, San Jose, CA) and a 1:10 dilution of Alexa Fluor-conjugated Alexa antibody against human p38 MAPK (pT80 / pY182, clone 36 / p38; BD Biosciences, San Jose, CA). Cells were analyzed using a BD FACSVerse flow cytometer (BD Biosciences, San Jose, CA).
[0869]
[0870] The results showed that polycrasoidol ( 1 ) was able to significantly decrease (63.4%) the activation of p38 MAPK, as well as compound 9 (71.9%) ( Figures 12A and 13A ). In addition, the activation of different compounds of the MAPK family is associated with the transactivation of N F- k B (p65), and the mobilization of N F- k B to the nucleus activates the transcription of genes that encode the expression of numerous inflammatory mediators such as endothelial adhesion molecules or the synthesis of cytokines and chemokines (Monaco et al., 2004). We were able to determine that polycrasoidol ( 1 ) and compound 9 were able to significantly block the activation of N F- k B induced by TNFa ( Figure 12B and 13B ).
[0871]
[0872] In vivo studies
[0873]
[0874] Example 8: Treatment of ob / ob mice with compound 9.
[0875]
[0876] The protocols in accordance with the guidelines of the European Union for the care and protection of animals were approved by the Ethical Committee of the University of Valencia. Every effort was made to minimize the number of animals used, as well as their suffering.
[0877]
[0878] Ob / ob mice have a leptin deficiency, and are associated with hyperphagia, and obesity, in addition to hyperglycemia and hyperinsulinemia. Male ob / ob mice (C57BL / 6J) (Charles River laboratories, Chatillon-sur-Chalaronne, France) were used in this study.
[0879] The mice were housed in individual cages in a room where temperature, humidity and light were controlled. They were maintained and raised (UCIM Statute, University of Valencia). in pathogen-free conditions with free access to food and autoclaved water, at a humidity of 60-65% and at a constant temperature of 22 ± 2 ° C with one cycle dark / bright from 12 h (light from 06: 00-18: 00 h, darkness from 18: 00-06: 00 h). Mice were allowed to acclimatize for ten days before the study.
[0880]
[0881] Male ob / ob mice 7 to 8 weeks of age were treated daily between 9:00 am and 11:00 am with compound 9 orally by an orogastric tube (10 mg / kg / d and 30 mg / kg) / d), rosiglitazone (10 mg / Kg / d) and / or W Y-14643 (30 mg / Kg / d) for 4 days, and were compared with the group of control ob / ob mice treated only with the vehicle ( physiological serum with 1% HEC). The doses were chosen based on previous results, obtained from an experiment with ob / ob mice (n = 3) in which compound 9 and the reference compounds, rosiglitazone and W Y-14643, were administered intraperitoneally (data not shown ). 6 animals per group were used in independent experiments (n = 6). The compounds were prepared daily as suspensions in 1% HEC for oral administration. Randomization was performed on blood glucose values at day 0. During the duration of the experiment, weighed daily for each mouse: amount of food ingested and body weight. Daily, between 9:00 am and 11:00 hours (prior to treatment) blood glucose was measured using (Contour next USB Blood glucose meter, BAYER, Basel, Switzerland). On the fifth day of the experiment, the mice were anesthetized with isofluorane and blood was drawn by cardiac puncture. Blood was collected in tubes containing EDTA or heparin. Once the samples were taken, the animals were sacrificed by cervical dislocation and subsequently, various organs such as white adipose tissue (WAT) and liver were removed for later study. The organs were weighed and frozen for further RNA analysis and to be able to carry out histological studies. Plasma samples were obtained by centrifugation and stored at -80 ° C.
[0882]
[0883] Histological analysis
[0884]
[0885] Perigonal white adipose tissue (WAT) and liver samples were fixed in 4% paraformaldehyde, embedded in paraffin, sectioned using a microtome (Leica RM2245, Leica Biosystems, Wetzlar, Germany) and mounted on Superfrost® plus microscope slides ( Thermo Fisher Scientific, Waltham, MA). Cross sections of WAT and liver tissue were obtained. At least 10 slides were examined, containing “4 tissue cross sections (5 m thick) of each animal. Hematoxylin and eosin (H&E) staining of WAT and liver was performed. Inflammatory infiltration in WAT was measured as previously described (Toyoda et al., 2008). After inactivation of peroxidase (H 2 O 2 3%) and blocking with goat serum (Abcam, Cambridge, United Kingdom), cross sections of WAT were incubated overnight (4 ° C) with the following monoclonal antibody ( mAb) Primary: F4 / 80 rabbit anti-mouse mAb (1: 100 dilution, Abcam, Cambridge, UK). A specific label was detected with an antibody secondary of goat anti-rabbit conjugated with HRP (dilution 1: 500, Dako, Copenhagen, Denmark). In order to confirm the specificity of the antibodies, control antibodies of the same isotype (Abcam, Cambridge, United Kingdom) or secondary antibody were used only as negative controls. Samples were revealed using a solution containing 3, 3'-diaminobenzidine (DAB, Vector Laboratories Inc, Burlingame, CA), subsequently stained with Harris hematoxylin (Sigma-Aldrich, Madrid, Spain) and dehydrated. Five fields of each section of WAT tissue were photographed (Axio Observer A1, Carl Zeiss microscope, Thornwood, NY) digitized and analyzed (Im ageJ software 1.48V, Bethesda, MA). The quantification was carried out double blind and through codes.
[0886]
[0887] MRNA extraction , real-time PCR
[0888]
[0889] Total RNA was extracted from the WAT fat of mice by homogenization. Reverse transcription was performed with 1000 ng of total RNA using the TaqMan reverse transcription kit (Applied Biosystems, Thermo Fisher Scientific, Waltham, MA) and converted to cDNA by standard methods. The cDNA was amplified with primers specific for mouse monocytic chemoattractant protein 1 (MCP-1 or CCL2, Mm00441242_m1) in a 7900HT Real-Time PCR apparatus (Applied Biosystems, Thermo Fisher Scientific, Waltham, MA) using the Mix Universal Master (Applied Biosystems, Thermo Fisher Scientific, Waltham, MA) All primers were pre-designed by Applied Biosystems. The relative quantification of the different transcripts was determined with the 2-AACt method using GAPDH (Applied Biosystems, Thermo Fisher Scientific, Waltham, MA) as an endogenous and standardized control for the control group.
[0890]
[0891] Metabolite Measurements
[0892]
[0893] Plasma free fatty acids (FFA), the enzyme aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were determined by means of a colorimetric assay kit (Sigma-Aldrich, Madrid, Spain). In addition, plasma lipid levels of total cholesterol and triglycerides (TG) were determined by enzymatic procedures (WAKO, Cape Charles, VA). The concentration of HDL-cholesterol (HDL-c) was also determined with the same method used to measure total cholesterol, after the precipitation of apolipoprotein-B (apoB) with dextran sulfate / MgCh (Sigma-Aldrich, Madrid , Spain) as previously described (Zieske et al., 2005).
[0894] 8.1 Compound 9 does not cause variations in body weight and nutritional efficiency.
[0895]
[0896] An adverse effect during treatment with TZD in individuals with DT2 is weight gain (Ahmadian e ta l., 2013). To determine the effects of treatment with compound 9 , mice and ingested food were weighed daily. Weight gain was calculated at the end of treatment (d 5 -d 0 ) ( Figure 14A ). Treatment with compound 9 at doses of 10 and 30 mg / Kg / d did not cause significant differences in weight gain in relation to the control group. The gain increase was 1.70 ± 0.25, 2.5 ± 0.32 and 1.88 ± 0.52 g, for control mice, treated with rosiglitazone and compound 9 (30 mg / Kg / d) respectively. Food efficiency followed this same trend, and no significant differences were observed in the mice treated with compound 9 ( Figure 14B ).
[0897]
[0898] 8.2 Compound 9 lowers blood glucose values
[0899]
[0900] The 4-day treatment allowed us to observe a 47% increase in the percentage of blood glucose (248 vs. 300 mg / dL) in the ob / ob mice of the control group treated with vehicle, while the administration of compound 9 caused a 20% decrease in glucose levels (261 vs. 194 mg / dL) at the dose of 30 mg / kg / d compared to day 0 (d 0 ), finally, treatment with rosiglitazone lowered glucose by 32 % (217 vs. 135 mg / dL) ( Figure 15) .
[0901]
[0902] 8.3 Effect of compound 9 on white adipose tissue (WAT)
[0903]
[0904] Treatment of ob / ob mice with compound 9 did not significantly increase the weight of perigonadal white adipose tissue (WAT) compared to the group of control mice ( Figure 16 ).
[0905]
[0906] On the other hand, the histological examination of WAT revealed changes in cell morphology, with a significant decrease in the average area of adipocytes in mice treated with compound 9 in a dose-dependent manner, similar to the effect produced by W Y-14643 ( Figure 17 ), suggesting the ability of compound 9 to prevent adipocyte hypertrophy. In contrast, treatment with rosiglitazone increased the number of small adipocytes in WAT deposits, decreasing the average value of the adipocyte area and confirming its adipogenic effect.
[0907]
[0908] 8.4 Anti-inflammatory effect of compound 9 on white adipose tissue (WAT)
[0909] There is evidence that in obese individuals with DT2 a chronic inflammatory process begins in adipose tissue. In WAT of obese mice are found Increased levels of gene expression in inflammatory pathways, this is reflected in an increase in inflammatory chemokines in the adipose tissue of obese individuals, such as monocyte chemoattractant protein (MCP-1), which participates in monocyte recruitment in the area of inflammation (Yoshimura et al., 1989), as well as an increase in macrophage infiltration into adipose tissue (MacKnight et al., 1996).
[0910]
[0911] Studies were carried out on the inflammatory process of W AT and we could observe that in ob / ob mice treated with compound 9 at 30 mg / Kg / d, the expression of M CP-1 decreased significantly ( Figure 18A ). In addition, quantification of macrophages in WAT by immunohistochemistry using an antibody against F4 / 80, showed us that compound 9 was able to significantly decrease the number of macrophages at the dose of 30 mg / Kg / d, indicating the capacity of the compound 9 to reduce macrophage infiltration in WAT, similar to the effect produced by rosiglitazone ( Figure 18B ).
[0912]
[0913] 8.5 Compound 9 improves blood lipid parameters
[0914]
[0915] The effects on dyslipidemia of ob / ob mice have been studied by determining various plasma parameters such as total cholesterol, HDL-c, triglycerides and free fatty acids. Treatment with compound 9 at the dose of 30 mg / Kg / d and W Y-14643 significantly increased total cholesterol levels compared to the group of ob / ob control mice (50% and 64%, respectively) ( Figure 19A ). Compound 9 at doses of 10 and 30 mg / Kg / d and W Y-14643 also significantly increased HDL-c, relative to the group of ob / ob control mice ( Figure 19B ). In addition, compound 9 and W Y-14643 showed a decrease in plasma triglyceride levels, although not significantly and that could be due to the short period of treatment (122 mg / dL or 101 mg / dL at the dose of 10 or 30 mg / Kg / d, respectively, vs. 137 mg / dL of the control ob / ob) ( Figure 19C ). Compound 9 to 30 mg / Kg / d and W Y-14643 were able to significantly lower levels of free fatty acids in blood (0.25 and 0.33 mol / pL, respectively, vs. 0.67 mol / pL of the ob / ob control ) ( Figure 19D ).
[0916]
[0917] 8.6 Effect of compound 9 on the liver
[0918]
[0919] The effect of compound 9 on a primary organ such as the liver has been studied, observing a non-significant weight gain with respect to the ob / ob control group ( Figure 20A ). The quantification of triglyceride content showed that in the short period of treatment with compound 9 at 30 mg / kg / d, triglycerides did not accumulate significantly in the liver compared to the ob / ob control group ( Figure 20B ).
[0920] 8.7. Compound 9 lowered the levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST)
[0921] Compound 9 lowered the levels of ALT and AST, the latter suffering a statistically significant reduction at the concentration of 30 mg / Kg / d (28%) compared to the ob / ob control group after 4 days of treatment ( Figure 21 ) . This suggests that compound 9 could reduce liver damage, showing a hepatoprotective effect.
[0922]
[0923] REFERENCES
[0924] 1. Cheang, WS et al. The peroxisome proliferator-activated receptors in cardiovascular diseases: experimental benefits and clinical challenges. Br. J. Pharm acol. 2015, 172, 5512-5522.
[0925] 2. Ahmadian, M. et al. PPARy signaling and metabolism: the good, the bad and the future.
[0926] Nat. Med. 2013, 19, 557-566.
[0927] 3. Tan, CK etal . Synthetic and natural Peroxisome Proliferator-Activated Receptor (PPAR) agonists as candidates for the therapy of the metabolic syndrome. E xpert Opin. Ther. Targets 2017, 21, 333-348.
[0928] 4. González, MC etal . Polycerasoidin and polycerasoidol, two new prenylated benzopyran derivatives from Polya lth ia cerasoides. J. Nat. Prod. 1995, 58, 1278-1284.
[0929] 5. González, MC et al. Prenylated benzopyran derivatives from two Polya lth ia species.
[0930] Phytochem 1996, 43, 1361-1364.
[0931] 6. Kabbe, H.-J., Widdig, A. Synthesis and reactions of 4-chromanones. Angew Chem
[0932] Int. Ed. Engl. 1982, 21,247-256.
[0933] 7. Sen, SE et al. Inhibition of verterate squalene epoxidase by extended and truncated analogues of trisnorsqualene alcohol. J. Med. Chem. 1990, 33, 1698-1701.
[0934] 8. Bisceglia, J. A .; Orelli, L. R. Recent Applications of the Horner-Wadsworth-Emmons Reaction to the Synthesis of Natural Products. Curr. Org. Chem. 2012, 16, 2206-2230.
[0935] 9. Toshima, K. etal . The First Total Synthesis of Concanamycin F (Concanolide A). J.
[0936] Org. Chem. 2001, 66, 1708-1715.
[0937] Carmona, MC et al. S 26948: a new specific peroxisome proliferator-activated receptor-modulator with potent antidiabetes and antiatherogenic effects. Diabetes 2007, 56, 2797-2808.
[0938]
[0939] Palee, S. et al. PPARgamma activator, rosiglitazone: Is it beneficial or harmful to the cardiovascular system W orld J. Cardiol. 2011, 3, 144-152.
[0940]
[0941] Rosenson, RS et al. Modulating peroxisome proliferator-activated receptors for therapeutic benefit Biology, clinical experience, and future prospects. Am. H eart J.
[0942] 2012, 164, 672-680.
[0943]
[0944] Sanz, MJ et al. Retinoid X receptor agonists impair arterial mononuclear cell recruitment through peroxisome proliferator-activated receptor-gamma activation. J. Im m unol. 2012, 189, 411-424.
[0945]
[0946] Zernecke, A .; Weber, C., Chemokines in the vascular inflammatory response of atherosclerosis. Cardiovasc. Res. 2010, 86, 192-201.
[0947]
[0948] Zernecke, A. et al. Chemokines in atherosclerosis an update. Artherioscler Thromb. Vasc Biol. 2008, 28, 1897-1908.
[0949]
[0950] Park, JB et al. Peroxisome proliferator-activated receptor-gamma agonists suppress tissue factor overexpression in rat balloon injury model with paclitaxel infusion. PLoS One 2011, 6, e2832.
[0951]
[0952] Monaco, C .; Paleolog, E., Nuclear factor kappaB: a potential therapeutic target in atherosclerosis and thrombosis. Cardiovasc. Res. 2004, 61, 671-678.
[0953]
[0954] Toyoda, T. et al. Effect of peroxisoma proliferator-activated receptor-a ligands in the interaction between adipocytes and macrophages in obese adipose tissue. Or besity 2008, 16, 1199-1207.
[0955]
[0956] Zieske AW et al. Elevated serum c-reactive protein levels and advanced atherosclerosis in youth. To rterioscler. Thromb. Vasc Biol. 2005, 25, 1237-1243.
[0957]
[0958] Yoshimura, T. et al. Human monocyte chemoattractant protein-1 (MCP-1). Full-length cDNA cloning, expression in mitogen-stimulated blood mononuclear leukocytes, and sequence similarity to mouse competence gene JE. FE BS Lett. 1989, 244, 487-49321.
[0959] McKnight, AJ et al. Molecular cloning of F4 / 80, a murine macrophage-restricted cell surface glycoprotein with homology to the G-protein-linked transmembrane 7 hormone receptor family. J. Biol. Chem. 1996, 271, 486-489.
[0960] 22. Taha H et al., (2015) PLoS ONE 10 (5) e0126126.do¡: 10.1371 / journal.pone.0126126.
[0961] 23. Zhao Yang et al., Journal of Agricultural and Food Chemistry (2010), 58 (8), 4844-4852.
[0962] 24. Karimian et al., (2015) PLoS ONE 10 (5) e0127434.doi: 10.1371 / journal.pone.0127434.
[0963] 25. Zafra-Polo et. Al., J. Nat. Prod., 1996, 59 (10), 913-916.
权利要求:
Claims (13)
[1]
1. Compound of general formula I, stereoisomers and pharmaceutically acceptable salts thereof:

[2]
2. Compound according to claim 1, selected from the group consisting of 3, 4, 4a, 4b, 4c, 6, 7, 8, 9, 10, 11, 12 or 13:

[3]
3. Compound according to any of claims 1 or 2, for use as a medicament.
[4]
4. Compound according to any of claims 1 or 2, for use in the treatment of a disease that responds to the administration of PPARa and / or PPAR agonists.
[5]
5. Compound for use according to claim 4, wherein said disease that responds to the administration of PPARa and / or PPAR agonists is independently selected from cardiometabolic disorders, type 2 diabetes (DT2), obesity, metabolic syndrome, hypercholesterolemia, hypertriglyceridemia, primary chylomicronemia, hyperlipoproteinemia, familial dysbetalipoproteinemia and neurodegenerative diseases.
[6]
6. Pharmaceutical composition comprising an effective amount of at least one compound of formula I according to any one of claims 1 or 2 and at least one pharmaceutically acceptable excipient.
[7]
7. Pharmaceutical composition according to claim 6, for use as a medicament.
[8]
8. Pharmaceutical composition according to claim 6, for use in the treatment of a disease that responds to the administration of PPARa and / or PPARy agonists.
[9]
9. Pharmaceutical composition for use according to claim 8, wherein said disease that responds to the administration of PPARa and / or PPAR agonists is independently selected from cardiometabolic disorders, type 2 diabetes (DT2), obesity, metabolic syndrome, hypercholesterolemia , hypertriglyceridemia, primary chylomicronemia, hyperlipoproteinemia, familial dysbetalipoproteinemia and neurodegenerative diseases.
[10]
10. Compound of general formula I, stereoisomers and pharmaceutically acceptable salts thereof:

[11]
11. Compound for use according to claim 10, wherein the disease that responds to the administration of PPARa and / or PPAR agonists is independently selected from cardiovascular disorders, type 2 diabetes (DT2), obesity, metabolic syndrome, hypercholesterolemia, hypertriglyceridemia, primary chylomicronemia, hyperlipoproteinemia, familial dysbetalipoproteinemia, inflammation and neurodegenerative diseases.
[12]
12. Pharmaceutical composition comprising an effective amount of at least one compound according to claim 10 and at least one pharmaceutically acceptable excipient, for use in the treatment of a disease that responds to the administration of PPARa and / or PPARy agonists.
[13]
13. Pharmaceutical composition for use according to claim 12, wherein the disease that responds to the administration of PPARa and / or PPAR agonists and is independently selected for cardiovascular disorders, type 2 diabetes (DT2), obesity, metabolic syndrome, hypercholesterolemia , hypertriglyceridemia, primary chylomicronemia, hyperlipoproteinemia, familial dysbetalipoproteinemia, inflammation and neurodegenerative diseases.
类似技术:
公开号 | 公开日 | 专利标题
RU2572596C2|2016-01-20|Therapeutic agents based on diglycylyl ether derivatives and methods for using them
Choi et al.2013|Saussurea lappa Clarke-derived costunolide prevents TNFα-induced breast cancer cell migration and invasion by inhibiting NF-κB activity
US20050137251A1|2005-06-23|Dexanabinol and dexanabinol analogs regulate inflammation related genes
CN107082770A|2017-08-22|The alpha inhibitors of IRE 1
AU2003214608A1|2003-09-29|Dexanabinol and dexanabinol analogs regulate inflammation related genes
TW201039815A|2010-11-16|Compositions and methods for the treatment of inflammation
EP3458448A1|2019-03-27|Methods of using fasn inhibitors
CA3103958A1|2020-01-09|Activators of the unfolded protein response
JP2019507111A|2019-03-14|Metabolically resistant fenfluramine analogues and methods of use thereof
ES2718000A1|2019-06-26|BENZOPIRANS PRENILATED AGGREGATES OF PPAR |
US20210002222A1|2021-01-07|Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents
Cheng et al.2020|Fucoidan inhibits NLRP3 Inflammasome activation by enhancing p62/SQSTM1-dependent selective autophagy to alleviate atherosclerosis
US20090047221A1|2009-02-19|Compounds and compositions useful in the treatment of neoplasia
Li et al.2012|Short‐term lenalidomide | administration ameliorates cardiomyocyte contractile dysfunction in ob/ob obese mice
JP2021527069A|2021-10-11|Compounds for the treatment or prevention of liver disease
CA2926928A1|2017-10-11|Aminobenzoic acid derivatives for use as anti-inflammatory agents, anti-metastatic agents and/or anticancer agents
JP2019031495A|2019-02-28|Selective MAO-B inhibitor compound, pharmaceutical composition thereof and use thereof
AU2020253578A1|2021-11-04|Very-long-chain polyunsaturated fatty acids, elovanoid hydroxylated derivatives, and methods of use
WO2019089577A1|2019-05-09|Small molecule inhibitors of cancer stem cells and mesenchymal cancer types
Cui et al.2020|Andrographolide attenuates choroidal neovascularization by inhibiting the HIF-1α/VEGF signaling pathway
RU2527526C2|2014-09-10|Indolyl-substituted pyrazino-quinolines and using them for treating cancer
US10519179B2|2019-12-31|Highly efficient NRF2 activators-co-releasing molecule hybrids, their use in the treatment of inflammatory or cardiovascular diseases and their process of preparation
WO2014041393A1|2014-03-20|Combination of compounds derived from gallic acid for the treatment of cancer
KR102221723B1|2021-03-03|Compound for activating ampk and uses thereof
KR20200094215A|2020-08-06|Benzenesulfonamide derivatives and lipid raft adjustment method
同族专利:
公开号 | 公开日
WO2019129909A1|2019-07-04|
EP3733658A4|2020-12-16|
EP3733658A1|2020-11-04|
ES2718000B2|2019-12-20|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题
EP0421419A2|1989-10-04|1991-04-10|Bristol-Myers Squibb Company|Tocotrienols in the treatment of hypercholesterolemia, hyperlipidemia and thromboembolic disorders|
WO2013176745A1|2012-05-22|2013-11-28|The Board Of Trustees Of The University Of Arkansas|Methods for making tocoflexols and analogues thereof|
EP1324995A2|2000-09-27|2003-07-09|Merck & Co., Inc.|Benzopyrancarboxylic acid derivatives for the treatment of diabetes and lipid disorders|
法律状态:
2019-06-26| BA2A| Patent application published|Ref document number: 2718000 Country of ref document: ES Kind code of ref document: A1 Effective date: 20190626 |
2019-12-20| FG2A| Definitive protection|Ref document number: 2718000 Country of ref document: ES Kind code of ref document: B2 Effective date: 20191220 |
优先权:
申请号 | 申请日 | 专利标题
ES201731470A|ES2718000B2|2017-12-26|2017-12-26|BENZOPIRANOS PRENILADOS AGONISTAS DE PPAR|ES201731470A| ES2718000B2|2017-12-26|2017-12-26|BENZOPIRANOS PRENILADOS AGONISTAS DE PPAR|
EP18897200.4A| EP3733658A4|2017-12-26|2018-12-21|Prenylated benzopyranes as ppar agonists|
PCT/ES2018/070826| WO2019129909A1|2017-12-26|2018-12-21|Prenylated benzopyranes as ppar agonists|
[返回顶部]