Aromatic molecules for the treatment of pathological conditions

文档序号:538258 发布日期:2021-06-01 浏览:11次 中文

阅读说明:本技术 用于治疗病态状况的芳香型分子 (Aromatic molecules for the treatment of pathological conditions ) 是由 V·莱因穆勒 R·马蒂 O·瓦涅里斯 J·B·瓜尔蒂耶罗蒂 V·库珀斯 于 2019-08-23 设计创作,主要内容包括:本发明包含新型芳香型分子,其可在人和兽医学中用于治疗病态状况,诸如癌症、皮肤病、肌肉病症,和免疫系统相关病症,诸如包括血液系统在内的造血系统的病症。(The present invention comprises novel aromatic molecules that are useful in human and veterinary medicine for the treatment of pathological conditions such as cancer, skin disorders, muscle disorders, and immune system-related disorders such as those involving the blood systemDisorders of the hematopoietic system.)

1. A compound according to the general formula (I) as defined herein, or a salt or solvate thereof:

R1=C1–C12alkyl, preferably C4–C12Alkyl radical, C2–C12Alkenyl, preferably C4–C12Alkenyl radical, C2–C12Alkynyl, preferably C4–C12Alkynyl, C3–C8Cycloalkyl radical, C5–C8Cycloalkenyl radical, C5–C12Bicycloalkyl radical, C7–C12Bicycloalkenyl, C8–C14Tricycloalkyl, -OC1–C12Alkyl, preferably-OC3–C12Alkyl, -OC2–C12Alkenyl, preferably-OC 3–C12Alkenyl, -OC2–C12Alkynyl, preferably-OC3–C12Alkynyl, -OC3–C8Cycloalkyl, -OC5–C8Cycloalkenyl radical, -OC5–C12Bicycloalkyl, -OC7–C12Bicycloalkenyl, -OC8–C14Tricycloalkyl, -SC1–C12Alkyl, preferably-SC3–C12Alkyl, -SC2–C12Alkenyl, preferably-SC3–C12Alkenyl, -SC2–C12Alkynyl, preferably-SC3–C12Alkynyl, -SC3–C8Cycloalkyl, -SC5–C8Cycloalkenyl radical, -SC5–C12Bicycloalkyl, -SC7–C12Bicycloalkenyl, -SC8–C14Tricycloalkyl, -NHR7or-NR7R8Wherein R is7And R8Independently of one another, selected from: c1–C12Alkyl, preferably C3–C12Alkyl radical, C2–C12Alkenyl, preferably C3–C12Alkenyl radical, C2–C12Alkynyl, preferably C3–C12Alkynyl, C3–C8Cycloalkyl radical, C5–C8Cycloalkenyl radical, C5–C12Bicycloalkyl radical, C7–C12Bicycloalkenyl, C8–C14Tricycloalkyl, or wherein R7Can be reacted with R8Together form a ring structure, wherein the ring structure comprising an N atom is selected from a 3-to 8-membered ring structure or a 5-to 12-membered bicyclic structure, and wherein all of the ring structures may additionally compriseReplacing a carbon atom contained in said ring structure with one or more heteroatoms independently selected from O, S and N, particularly wherein such replacement results in the residue containing at least twice as many C atoms as heteroatoms independently selected from O, S and N;

wherein is contained in R1、R7And R8All alkyl, alkenyl and alkynyl residues in the definition of (A) are straight or branched chain and are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH 2、-NO2、=O、C3–C8Cycloalkyl radical, C5–C8Cycloalkenyl radical, C5–C12Bicycloalkyl radical, C7–C12Bicycloalkenyl, C8–C14Tricycloalkyl, straight or branched-OC1–C5Alkyl radicals such as-OCH3、-OC3–C5Cycloalkyl radicals such as-O (cyclopropyl), straight-chain or branched-NH (C)1–C5Alkyl), straight or branched-N (C)1–C5Alkyl) (C1–C5Alkyl), -NH (C)3–C5Cycloalkyl) such as-NH (cyclopropyl), -N (C)3–C5Cycloalkyl) (C)3–C5Cycloalkyl), straight or branched-chain-N (C)1–C5Alkyl) (C3–C5Cycloalkyl) substituted with a substituent;

wherein when contained in R1、R7And R8When alkyl, alkenyl and alkynyl residues in the definition of (a) are substituted with one or more substituents which are ═ O, such substitution with ═ O cannot result in the direct attachment of one of the groups selected from C ═ O, S ═ O and N ═ O to the aromatic ring;

wherein is contained in R1、R7And R8All cyclic, bicyclic and tricyclic structures in the definition of (A) include cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl and tricycloalkyl residues unsubstituted or substituted with one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2O, straight or branched C1–C5Alkyl radicals such as-CH3Linear chain, linear chainOr branched-OC1–C5Alkyl radicals such as-OCH3Straight or branched-NH (C)1–C5Alkyl), straight or branched-N (C)1–C5Alkyl) (C1–C5Alkyl), -NH (C)3–C5Cycloalkyl) such as-NH (cyclopropyl), -N (C) 3–C5Cycloalkyl) (C)3–C5Cycloalkyl), straight or branched-chain-N (C)1–C5Alkyl) (C3–C5Cycloalkyl) substituted with a substituent;

wherein is contained in R1、R7And R8All alkyl, alkenyl and alkynyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of a carbon atom, and wherein such substitutions result in the residue containing at least 2 times the number of C atoms as many as the number of heteroatoms independently selected from O, S and N; and wherein such substitution additionally does not result in the direct attachment of one of the groups selected from C ═ O, S ═ O and N ═ O to the aromatic ring;

wherein is contained in R1、R7And R8All cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl and tricycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of a carbon atom, and wherein such substitution results in the residue containing at least the same number of C atoms as the number of heteroatoms independently selected from O, S and N;

wherein is contained in R1、R7And R8All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl and tricycloalkyl residues in the definition of (a) may be partially or fully halogenated, in particular fluorinated, more in particular perfluorinated;

wherein bicyclic and tricyclic residues include fused, bridged and spiro ring systems;

R2–R5Independently of one another, from the group consisting of-H, -F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C1–C4Alkyl, straight or branched C2–C4Alkenyl, straight-chain or branched C2–C4Alkynyl, C3–C6Cycloalkyl, -CH2(C3–C6Cycloalkyl), linear or branched-OC1–C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1–C3Alkyl), straight or branched-N (C)1–C3Alkyl) (C1–C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1–C3Alkyl) (cyclopropyl);

wherein is contained in R2–R5All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (A) are unsubstituted or independently selected from one or more of-F, -Cl, -Br, -I, -CH3、-CF3-OH and-OCH3、-OCF3、-NH2、-NHCH3、-N(CH3)2Substituted with the substituent(s);

wherein is contained in R2–R5All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in the substitution of carbon atoms, and wherein such substitution does not result in one of the groups selected from C ═ O and S ═ O being bonded directly to the aromatic ring;

X1–X4are independently selected from N, CR9、CR10、CR11、CR12

R9-R12Independently of one another, from the group consisting of-H, -F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C1–C4Alkyl, straight or branched C2–C4Alkenyl, straight-chain or branched C2–C4Alkynyl, C 3–C6Cycloalkyl, -CH2(C3–C6Cycloalkyl), linear or branched-OC1–C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1–C3Alkyl), straight or branched-N (C)1–C3Alkyl) (C1–C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1–C3Alkyl) (cyclopropyl);

wherein is contained in R9-R12All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (A) are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CH3、-CF3-OH and-OCH3、-OCF3、-NH2、-NHCH3、-N(CH3)2Substituted with the substituent(s);

wherein is contained in R9-R12All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in the substitution of carbon atoms, and wherein such substitution does not result in one of the groups selected from C ═ O and S ═ O being bonded directly to the aromatic ring;

wherein R is9-R12Preferably selected from the group consisting of-H, -F, -Cl, -Br, -CH3、-CF3、-OH、-OCH3、-OCF3Cyclopropyl, oxirane, -C (CH)3)3、-N(CH3)2、-NH2、-CN、-CH2OCH3、-OCH(CH3)2、-CH2NH2、-CH2N(CH3)2、-CH2OH、-NO2、-CH2-N-morpholinyl;

R6=-H、C1–C8alkyl, preferably C1–C4Alkyl radical, C2–C8Alkenyl, preferably C2–C4Alkenyl radical, C2–C8Alkynyl, preferably C2–C4Alkynyl, C3–C6Cycloalkyl radical, C5–C6Cycloalkenyl radical, C5–C12Bicycloalkyl radical, C7–C12Bicycloalkenyl, C8–C14Tricycloalkyl and aromatic and heteroaromatic residues, preferably a 6-membered aromatic ring and a 5-to 6-membered heteroaromatic ring;

and wherein bicyclic and tricyclic residues include fused, bridged and spiro ring systems;

Wherein is contained in R6Said cycloalkyl, cycloalkenyl, or heterocyclyl group in the definition of,Bicycloalkyl, bicycloalkenyl, tricycloalkyl, aromatic and heteroaromatic residues optionally through C1Alkylene or C2Alkylene or C3An alkylene linker to R6Bound N;

wherein is contained in R6All aromatic and heteroaromatic residues in the definition of (A) are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C1–C3Alkyl radical, C2–C3Alkenyl radical, C2–C3Alkynyl, cyclopropyl, straight or branched-OC1–C3Alkyl radicals such as-OCH3-O (cyclopropyl), straight or branched-NH (C)1–C3Alkyl), straight or branched-N (C)1–C3Alkyl) (C1–C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1–C3Alkyl) (cyclopropyl) substituents;

wherein is contained in R6All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl and tricycloalkyl residues and alkylene linkers in the definitions of (A) are straight or branched chain and are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2O, straight or branched C1–C3Alkyl radical, C2–C3Alkenyl radical, C2–C3Alkynyl, cyclopropyl, straight or branched-OC 1–C3Alkyl radicals such as-OCH3-O (cyclopropyl), straight or branched-NH (C)1–C3Alkyl), straight or branched-N (C)1–C3Alkyl) (C1–C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1–C3Alkyl) (cyclopropyl) substituents;

wherein is contained in R6All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl, tricycloalkyl and heteroaryl residues in the definitions of (A) and the alkylene linker may compriseContaining one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

wherein R is6preferably-H, -CH3、-CH2CH3N-propyl, isopropyl, cyclopropyl, -CF3and-CF2CF3Benzyl, tert-butyl, phenyl, cyclohexyl, 1-phenylethyl, 2-dimethyl-1-phenylpropyl, (1-naphthyl) -methyl, 4-methoxybenzyl, 4-trifluoromethylbenzyl, tetrahydropyranyl;

wherein is contained in R2–R6And R9–R12All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl, tricycloalkyl, aromatic and heteroaromatic residues in the definition of (a) may be partially or fully halogenated, in particular fluorinated, more in particular perfluorinated;

y ═ H, straight or branched C1–C6Alkyl, straight or branched C2–C6Alkenyl, straight-chain or branched C2–C6Alkynyl, C 3–C6Cycloalkyl radical, C5–C6Cycloalkenyl, -OH, straight or branched-OC1–C6Alkyl, straight or branched-OC2–C6Alkenyl, straight or branched-OC2–C6Alkynyl, -OC3–C6Cycloalkyl, -OC5–C6Cycloalkenyl, -CN, aromatic and heteroaromatic residues, preferably six-membered aromatic rings and five-to six-membered heteroaromatic rings, -S (O) R13and-S (O)2R13Wherein R is13Selected from straight or branched C1–C6Alkyl, straight or branched C2–C6Alkenyl, straight-chain or branched C2–C6Alkynyl, C3–C6Cycloalkyl radical, C5–C6Cycloalkenyl radical, -CF3and-C6H4CH3

Wherein all cycloalkyl, cycloalkenyl, aromatic and heteroaromatic residues comprised in the definition of Y may optionally pass through C1Alkylene, or C2Alkylene, or C3Alkylene, or-O-CH2-, or-O-CH2-CH2-the linker is linked to N to which Y is bound;

wherein all aromatic and heteroaromatic residues comprised in the definition of Y are unsubstituted or are selected from the group consisting of-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C1–C3Alkyl radical, C2–C3Alkenyl radical, C2–C3Alkynyl, cyclopropyl, straight or branched-OC1–C3Alkyl radicals such as-OCH3-O (cyclopropyl), straight or branched-NH (C)1–C3Alkyl), straight or branched-N (C)1–C3Alkyl) (C1–C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1–C3Alkyl) (cyclopropyl) substituents;

wherein all alkyl, alkenyl, alkynyl, cycloalkyl and cycloalkenyl residues and alkylene linkers contained in the definition of Y are straight or branched chain and are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH 2O, straight or branched C1–C3Alkyl radical, C2–C3Alkenyl radical, C2–C3Alkynyl, cyclopropyl, straight or branched-OC1–C3Alkyl radicals such as-OCH3-O (cyclopropyl), straight or branched-NH (C)1–C3Alkyl), straight or branched-N (C)1–C3Alkyl) (C1–C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1–C3Alkyl) (cyclopropyl) substituents;

wherein all alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl and heteroaryl residues contained in the definition of Y and the alkylene linker may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

wherein all alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aromatic and heteroaromatic residues and alkylene linkers comprised in the definition of Y may be partially or fully halogenated, in particular fluorinated, more in particular perfluorinated;

wherein Y may be substituted with R6Together form a ring structure, wherein the ring structure comprising an N atom of formula I is selected from the group consisting of 3-membered rings, 4-membered rings, 5-membered rings, 6-membered rings, 5-to 12-membered bicyclic residues, 8-to 14-membered tricyclic residues and heteroaromatic residues, wherein all rings, bicyclic, tricyclic and heteroaromatic residues may additionally comprise one or more heteroatoms independently selected from O, S and N in place of carbon atoms comprised in the ring structure, and wherein all rings, bicyclic, tricyclic and heteroaromatic residues are unsubstituted or substituted with one or more heteroatoms independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -OCH 3、-NH2、-NHCH3、-N(CH3)2、=O、-CH3、-CF3Morpholinyl;

and wherein bicyclic and tricyclic residues include fused, bridged and spiro ring systems;

Z1and Z2Selected from the following groups:

wherein Z1Selected from straight or branched C1–C3Alkyl, preferably-CH3Cyclopropyl, oxiranyl, N-methyl-aziridinyl, thietanyl, -CN, -N3、-CF3、-CF2CF3And wherein Z is2Independently selected from-H and straight or branched C1–C3Alkyl, preferably-CH3、-CF3、-CF2CF3(Ia);

Or wherein Z1And Z2Together are ═ O, ═ S, ═ NR14(Ib); wherein R is14Selected from-H, -OH, -OCH3、-CN、-S(O)C(CH3)3、-S(O)2CH3、-S(O)2CF3Straight or branched chain C1–C3Alkyl, preferably-CH3Cyclopropyl, -CF3、-CF2CF3、-CH2CF3、-C6H5、-CH2C6H5

Or wherein Z1And Z2Comprises the carbon atoms to which they are bound together form a cyclic residue (Ic); wherein the cyclic residue is selected from the group consisting of a three-membered ring, a four-membered ring, a five-membered ring and a six-membered ring, wherein all rings optionally may comprise one or more heteroatoms independently selected from O, S and N in place of carbon atoms comprised in the ring structure; wherein all rings are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -OCH3、-NH2、-NHCH3、-N(CH3)2、=O、-CH3and-CF3Substituted with the substituent(s);

wherein is contained in Z1And Z2All alkyl and cyclic residues in the definition of (a) may be partially or fully halogenated, in particular fluorinated, more in particular perfluorinated.

2. A compound according to formula (Ia) as claimed in claim 1 or a salt or solvate thereof.

3. A compound according to formula (Ib) as claimed in claim 1 or a salt or solvate thereof.

4. A compound according to formula (Ic) according to claim 1 or a salt or solvate thereof.

5. The compound of any one of claims 1-4, with the proviso that

(i) Excluding the compounds shown in table 1,

(ii) excluding the compounds shown in Table 2, and/or

(iii) The compounds shown in table 3 were excluded.

6. The compound of any one of claims 1-5

And wherein R1Selected from methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, isopropylAlkyl, sec-butyl, tert-pentyl, tert-octyl, 3-pentyl, -CF3、-CF2CF3、-(CF2)2CF3、-CH(CF3)2、-CH2SCH3、-CH2CH2SCH3、-CH2SCH2CH3、-CH2CH2SCH2CH3Methoxymethyl, methoxyethyl, methoxypropyl, ethoxymethyl, ethoxyethyl, propoxymethyl, dimethyl-aminomethyl, dimethyl-aminoethyl, diethyl-aminomethyl, ethyl-methyl-aminomethyl, cyclopropyl, methyl-cyclopropyl, ethyl-cyclopropyl, trifluoromethyl-cyclopropyl, perfluoroethyl-cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, dicyclopentyl, dicyclohexyl, bicycloheptyl, preferably norbornyl, bicyclooctyl, bicyclooctenyl, bicyclononyl, methylbicyclononyl, adamantyl, tricyclodecyl, oxiranyl, oxetanyl, tetrahydrofuranyl, methyltetrahydrofuranyl, trimethyltetrahydrofuranyl, tetrahydropyranyl, aziridinyl, N-methylaziridine, oxaziridinyl, and the like, Azetidinyl, N-methylazetidinyl, difluoroazetidinyl, pyrrolidinyl, N-methylpyrrolidinyl, piperidinyl, N-methylpiperidinyl, difluoropiperidinyl, thienylpropyl, thienylbutyl, tetrahydrothienyl, tetrahydrothiopyranyl, dioxanyl, piperazinyl, dimethylpiperazinyl, dithianyl, morpholinyl, N-methylmorpholinyl, thiomorpholinyl, N-methylthiomorpholinyl, oxa-azaspiroheptyl, N-methyloxa-azaspiroheptyl, N-methylazepiroheptyl, thia-azaspiroheptyl, N-methylthio-azaspiroheptyl, difluorothia-azaspiroheptyl, azaspirooctyl, N-methylazepirooctyl, oxa-azaspirooctyl, N-methyloxa-azaspirooctyl, Oxa-azaspiro nonyl, N-methyloxa-azaspiro nonyl, N-methylazaspiro nonyl, oxa-azaspiro decyl, N-methyloxa-azaspiro decyl, N-methylazaspiro decyl, dihydro-oxazinyl, N-methyldihydro-oxazinyl, oxazolidinyl, N-methyloxazolidinyl, dioxolanyl, imidazolidinyl N-methylimidazolidyl, N-dimethylimidazolidinyl, azepanyl, N-methylazepanyl, azaspirohexyl, N-methylazepanyl, oxa-azadispirodecyl, N-methyloxa-azadispirodecyl, N-methylazepanyl, oxa-azabicyclooctyl, N-methyloxa-azabicyclooctyl, N-methylazebicyclooctyl, azabicycloheptyl, N-methylazebicyclononyl, azaadamantyl, -O (adamantyl), oxa-azabicyclononyl, N-methyloxa-azabicyclononyl, oxaazabicycloheptyl, azabicyclononyl, N-methyloxa-azabicyclononyl, oxa-azabicycloheptyl, azacycloheptyl, azepanyl, aze, N-methyldiazabicycloheptyl, diazabicyclooctyl, N-methyldiazabicyclooctyl, N-dimethyldiazabicyclooctyl, diazabicycloheptyl, N-methyldiazabicycloheptyl, N-dimethyldiazabicycloheptyl; 4-oxocyclohexyl, 3-oxocyclopentyl; 2-oxocyclobutyl, 4-oxobicyclo [4.1.0]Hept-1-yl.

7. The compound according to any one of claims 1 to 6,

wherein R is1Is selected from C4–C12Alkyl radical, C4–C12Alkenyl radical, C4–C12Alkynyl, cyclic, bicyclic and tricyclic residues, wherein said alkyl, alkenyl and alkynyl residues are preferably branched, including:

8. The compound according to any one of claims 1 to 7,

wherein R is2–R3Each is-H, R4preferably-H or-F, and/or R5is-H, -F, -Cl, -Br, -CH3、-CF3、-CH=CH2、-C≡CH、-CH2OH、-CH2NHCH3、-OH、-OCH3、-OCF3Cyclopropyl, oxirane, -CH2-N-morpholinyl, -C (CH)3)3、-CH2OCH3、-NO2、-CN、-NH2、-N(CH3)2、-OCH(CH3)2、-CH2NH2、-CH2N(CH3)2

9. The compound of any one of claims 1 to 8,

wherein the substituents R are as defined in the general formula (I)1To R5The six-membered aromatic ring in combination being selected from

10. The compound of any one of claims 1-9

Wherein X is as defined in formula (I)1–X4The six-membered aromatic ring of (a) is selected from:

11. the compound of any one of claims 1-10

Wherein Y is-H, -CH3、-CH2CH3N-propyl, isopropyl, cyclopropyl, cyclohexyl, tetrahydropyranyl, -CF3、-CF2CF3、-OH、-OCH3、-OCH2CH3、-OCH2(cyclopropyl), -CN, -S (O) C (CH)3)3、-S(O)2CH3、-S(O)2CF3、-S(O)2C6H4CH3、-OCH2C6H5and-OC6H5(ii) a And for R6is-H or-CH3Or benzyl, then Y is preferably-OH, -OCH3、-OCH2CH3、-OCH2(cyclopropyl).

12. The compound of any one of claims 1-11

Wherein Y and R of formula I6Together, the ring structure containing an N atom is selected from aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, difluoropiperidinyl, morpholinyl azetidinyl, hydroxyazetidiyl, azetidinyl, difluoroazetidinyl, azaspirohexyl, azaspiroheptyl, difluoroazaspiroheptyl, hydroxyazapiroheptyl, methylhydroxyazaspiroheptyl, trifluoromethylhydroxyazapiroheptyl, azaspirooctyl, azaspirononyl, oxa-azaspiroheptyl, oxa-azaspirooctyl, oxa-azaspirononyl, thia-azaspiroheptyl, oxazolidinyl, tetrahydro-oxazinyl, isoxazolidinyl, oxazolidinyl, isoxazolidinyl, piperazine.

13. The compound of any one of claims 1-12

Wherein Y and R of formula I6The ring structures comprising N atoms taken together are selected from:

14. the compound of any one of claims 1-13

Wherein Z1is-CH3、-CF3-CN, cyclopropyl; and/or wherein Z2preferably-H, -CH3and-CF3(ii) a For example:

15. the compound of any one of claims 1-13

Wherein Z1And Z2Together are preferably ═ O, ═ NR14(ii) a Wherein R is14Preferably selected from-H, -CH3Cyclopropyl, -OH, -OCH3、-CN:

16. The compound of any one of claims 1-13

Wherein Z1And Z2Together comprising the carbon atoms to which they are bound to form a 3-or 4-membered cyclic residue; wherein the cyclic residue is preferably selected from cyclopropyl, cyclobutyl, oxirane, oxetanyl, aziridinyl, azetidinyl and thietanyl; and wherein the cyclic residue is optionally preferably substituted by-F, -OH, -OCH3、-NH2、-NHCH3、-N(CH3)2、=O、-CH3and-CF3Substitution;

and wherein the cyclic residue is even more preferably selected from:

17. the compound of any one of claims 1-16

Wherein Y is selected from the residues contained in the general definition of Y, which together with the oxygen atom are bound to the N to which Y is bound.

18. The compound of any one of claims 1-17

Wherein R is1Is selected from R1Contains 4 or more, preferably 6 or more, even more preferably 7 or more carbon atoms, and wherein R is1No heteroatoms.

19. As claimed in claim 18The compound of (1), wherein R1Comprising a ring structure selected from the group consisting of cyclic, bicyclic, and tricyclic structures.

20. The compound of claim 18 or 19, wherein R1Selected from the group consisting of cyclohexyl, norbornyl, bicyclooctyl, bicyclononyl, methylbicyclononyl, tricyclodecyl and adamantyl.

21. The compound of any one of claims 1-17, wherein R1Selected from the group consisting of R1Contains 4 or more, preferably 6 or more, even more preferably 7 or more carbon atoms, and wherein R is1Comprising one or more, preferably 1 to 2, heteroatoms independently selected from O, S and N for substitution of R1Carbon atoms contained in (a).

22. The compound of claim 21, wherein R1Selected from tetrahydropyranyl, N-methylpiperidyl, morpholinyl, 4-oxocyclohexyl, azabicyclohexyl, N-methylazabicyclohexyl, oxa-azabicyclohexyl, N-methyldiazabicyclohexyl, azabicyclooctyl, diazabicyclooctyl, N-methyldiabicyclooctyl, oxa-azabicyclooctyl, azabicyclononyl, azaadamantyl and-O (adamantyl).

23. The compound of any one of claims 1-22, wherein the compound has the following structure (I-1)

And wherein Z1And Z2As defined in general formula (I), including general formula (Ia), general formula (Ib) and general formula (Ic), including substitutions and preferred definitions, optionally with the proviso that Z is in the case of general formula (Ib)1And Z2Together with O, is different from O,

and wherein R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein Y, R2–R6、R9–R13Or X1-X4As defined in formula (I), including substituted and preferred definitions.

24. The compound of any one of claims 1-23, wherein the compound has the following structure (I-4):

wherein R is6As defined in formula (I), including substituted and preferred definitions, with the proviso that R6In contrast to the case of the group-H,

and wherein Z1And Z2As defined in general formula (I), including general formula (Ia), general formula (Ib) and general formula (Ic), including substituted and preferred definitions,

and wherein R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R1–R5、R7–R12And X1–X4As defined in formula (I), including substituted and preferred definitions.

25. The compound of any one of claims 1-24, wherein the compound has the following structure (Ib-1):

Wherein R is1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Comprises 6 or more carbon atoms optionally independently replaced by a heteroatom selected from O, S and N as defined in formula (I), and wherein R is1Selected from the group consisting of cyclic, bicyclic and tricyclic structures,

and wherein R5As defined in formula (I), including substituted and preferred definitions, with the proviso that R5In contrast to the case of the group-H,

and wherein Z1、Z2And R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R2–R4、R6–R13And X1–X4And Y is as defined in formula (I), including substituted and preferred definitions.

26. The compound of any one of claims 1-25, wherein the compound has the following structure (Ib-2):

wherein R is1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Containing six or more carbon atoms, optionally independently substituted by a heteroatom selected from O, S and N as defined in formula (I), and wherein R is1Selected from the group consisting of cyclic, bicyclic and tricyclic structures,

and wherein Z1、Z2And R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R2–R13、X1–X4And Y is as defined for formula (I), including substituted and preferred definitions.

27. A compound as described in any one of tables 6 to 54, or a salt or solvate thereof.

28. A compound as claimed in any one of claims 1 to 27 for use in medicine, for example in human or veterinary medicine.

29. A compound according to any one of claims 1 to 27 for use in the treatment of a disorder associated with, accompanied by and/or caused by dysfunctional Notch signaling.

30. A compound of any one of claims 1-27 for use as an enhancer of Notch signaling.

31. The compound of any one of claims 1-27 for use in treating a hyperproliferative disorder, including malignant and non-malignant hyperproliferative disorders.

32. A compound according to any one of claims 1 to 27 for use in the treatment of diseases of the skin, mucosa, skin and mucosal appendages, cornea and epithelial tissues and malignant, non-malignant and hyperproliferative disorders, including cancers such as non-melanoma skin cancers including squamous cell carcinoma and basal cell carcinoma and precancerous lesions including actinic keratosis, skin and/or mucosal disorders with keratotic defects and/or abnormal keratinocyte proliferation, viral infections, atopic dermatitis and acne related, associated and/or caused skin and/or mucosal diseases, and for promoting wound healing of the skin and mucosa.

33. A compound as claimed in any one of claims 1 to 27 for use in the treatment of hyperproliferative disorders, cancers or pre-cancerous conditions of the skin, oral mucosa, tongue, lung, stomach, breast, cancer of the neuroendocrine system such as medullary thyroid cancer, brain cancer, pancreatic cancer, liver cancer, thyroid cancer and cancer of the genitourinary tract, including cervical cancer and ovarian cancer.

34. A compound as claimed in any one of claims 1 to 27 for use in the treatment of malignant and non-malignant muscle diseases, including muscular dystrophy, or for use in muscle regeneration, or for use in muscle hyperproliferative disorders, such as muscle hyperplasia and muscle hypertrophy.

35. A compound as claimed in any one of claims 1 to 27 for use in the treatment of immune system related disorders, including disorders of the hematopoietic system including the blood system, such as cancers of the hematopoietic and blood systems, such as leukemias and lymphomas, such as myeloid malignancies, e.g. acute and chronic myeloid leukemia and acute and chronic promyelocytic leukemia, and lymphoid malignancies, e.g. acute and chronic T-cell leukemia and acute and chronic B-cell leukemia, and cutaneous T-cell lymphoma.

36. A compound as claimed in any one of claims 1 to 27 for use in therapeutic immune system related applications, including immunotherapy and other immunotherapy approaches, such as use as an immunoadjuvant or vaccine adjuvant.

37. A method of treating a hyperproliferative disorder, comprising administering to a subject, in particular a human subject, in need thereof a therapeutically effective amount of a compound according to any one of claims 1-27.

38. A method of treating a dysfunctional Notch signaling-related, concomitant and/or caused disorder, comprising administering to a subject, particularly a human subject, in need thereof a therapeutically effective amount of a compound according to any one of claims 1-27.

Disclosure of Invention

The present invention encompasses novel molecules that exhibit significant biological activity on cells of human and animal origin. The corresponding compounds were found to affect the growth and survival of cancer cells and primary non-cancer cells. In particular, molecules that are capable of completely or partially inhibiting cell growth or causing cell death have been identified. Furthermore, some compounds were found to affect cell signaling pathways, in particular the Notch signaling pathway. The corresponding molecules were found to enhance the Notch signaling pathway.

The present invention therefore relates to compounds having antiproliferative activity as defined herein, which are useful in the treatment of benign and malignant hyperproliferative disorders in human and veterinary medicine. In particular, the present invention relates to compounds as defined herein for use in the treatment of the following conditions in human and veterinary medicine: hematopoietic disorders (including hematologic and immune system-related disorders), malignancies involving the myeloid and lymphoid lineages, malignant and non-malignant diseases of the skin and mucosa (e.g., keratoses), malignant and non-malignant diseases of the muscle (including muscle hyperproliferative diseases such as muscle proliferation and muscle hypertrophy), neuroendocrine system disorders, hyperproliferative disorders of the skin and mucosa, cancer and precancerous cross-lesions, such as non-melanoma skin cancers (including squamous cell carcinoma and basal cell carcinoma), actinic keratosis, hyperproliferative diseases and cancers of the oral cavity and tongue, hyperproliferative disorders and cancers of the neuroendocrine system (such as medullary thyroid carcinoma), hyperproliferative diseases and cancers of the hematopoietic system (including the hematologic system) such as leukemias and lymphomas, hyperproliferative disorders of the lung, breast, stomach, urogenital tract and cancers, for example, cervical cancer, including ovarian cancer.

The biological activity, e.g., antiproliferative activity, of the claimed compounds may be attributed to, but is not limited to, Notch signaling enhancing activity. Thus, the present invention also relates to compounds as defined herein which characterize Notch enhancing activity and which are useful in the treatment of pathological conditions in human and veterinary medicine which respond to Notch modulation, such as cancer, skin diseases, muscle disorders, disorders of the hematopoietic system, including disorders related to the blood system and the immune system.

The compounds of the present invention are directed to bisarylether structures consisting of two six-membered aromatic rings, wherein one aromatic ring is an unsubstituted or substituted benzyl ring and the other aromatic ring is an unsubstituted or substituted aryl ring, which optionally contains an N atom and is thus optionally a six-membered heteroaromatic ring. All such bisaryl ether structures share a common feature of containing substituents at both para positions relative to the ether linkage, wherein such substituents on the benzyl ring that cannot be heteroaromatic are preferably selected from non-polar residues and/or sterically demanding residues; and wherein such substituents on the aryl ring, optionally the heteroaromatic ring, are selected from structural units preferably containing a significant number of heteroatoms.

The first aspect of the present invention relates to compounds of the general formula (I):

R1=C1-C12Alkyl, preferably C4-C12Alkyl radical, C2-C12Alkenyl, preferably C4-C12Alkenyl radical, C2-C12Alkynyl, preferably C4-C12Alkynyl, C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl, C8-C14Tricycloalkyl, -OC1-C12Alkyl, preferably-OC3-C12Alkyl, -OC2-C12Alkenyl, preferably-OC3-C12Alkenyl, -OC2-C12Alkynyl, preferably-OC3-C12Alkynyl, -OC3-C8Cycloalkyl, -OC5-C8Cycloalkenyl radical, -OC5-C12Bicycloalkyl, -OC7-C12Bicycloalkenyl, -OC8-C14Tricycloalkyl, -SC1-C12Alkyl, preferably-SC3-C12Alkyl, -SC2-C12Alkenyl, preferably-SC3-C12Alkenyl, -SC2-C12Alkynyl, preferably-SC3-C12Alkynyl, -SC3-C8Cycloalkyl, -SC5-C8Cycloalkenyl radical, -SC5-C12Bicycloalkyl, -SC7-C12Bicycloalkenyl, -SC8-C14Tricycloalkyl, -NHR7or-NR7R8Wherein R is7And R8Independently of one another, selected from: c1-C12Alkyl, preferably C3-C12Alkyl radical, C2-C12Alkenyl, preferably C3-C12Alkenyl radical, C2-C12Alkynyl, preferably C3-C12Alkynyl, C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl, C8-C14Tricycloalkyl, or wherein R7Can be reacted with R8Together forming a ring structure, wherein said ring structure comprising an N atom is selected from a 3-to 8-membered ring structure or a 5-to 12-membered ring structure, and wherein all of said ring structures may additionally comprise one or more heteroatoms independently selected from O, S and N in place of carbon atoms contained in said ring structure, particularly wherein such replacement results in a residue containing at least twice the number of C atoms as heteroatoms independently selected from O, S and N;

Which comprisesAt R1、R7And R8All alkyl, alkenyl and alkynyl residues in the definition of (A) are straight or branched chain and are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2、=O、C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl, C8-C14Tricycloalkyl, straight or branched-OC1-C5Alkyl radicals such as-OCH3、-OC3-C5Cycloalkyl radicals such as-O (cyclopropyl), straight-chain or branched-NH (C)1-C5Alkyl), straight or branched-N (C)1-C5Alkyl) (C1-C5Alkyl), -NH (C)3-C5Cycloalkyl) such as-NH (cyclopropyl), -N (C)3-C5Cycloalkyl) (C)3-C5Cycloalkyl), straight or branched-chain-N (C)1-C5Alkyl) (C3-C5Cycloalkyl) substituted with a substituent;

wherein when contained in R1、R7And R8When alkyl, alkenyl and alkynyl residues in the definition of (a) are substituted with one or more substituents which are ═ O, such substitution with ═ O cannot result in the direct attachment of one of the groups selected from C ═ O, S ═ O and N ═ O to the aromatic ring;

wherein is contained in R1、R7And R8All cyclic, bicyclic and tricyclic structures in the definition of (A) include cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl and tricycloalkyl residues unsubstituted or substituted with one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH 2、-NO2O, straight or branched C1-C5Alkyl radicals such as-CH3Straight or branched-OC1-C5Alkyl radicals such as OCH3Straight or branched-NH (C)1-C5Alkyl), straight or branched-N (C)1-C5Alkyl) (C1-C5Alkyl), -NH (C)3-C5Cycloalkyl) such as-NH (cyclopropyl), -N (C)3-C5Cycloalkyl) (C)3-C5Cycloalkyl), straight or branched-chain-N (C)1-C5Alkyl) (C3-C5Cycloalkyl) substituted with a substituent;

wherein is contained in R1、R7And R8All alkyl, alkenyl and alkynyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of a carbon atom, and wherein such substitutions result in the residue containing at least 2 times the number of C atoms as many as the number of heteroatoms independently selected from O, S and N; and wherein such substitution additionally does not result in the direct attachment of one of the groups selected from C ═ O, S ═ O and N ═ O to the aromatic ring;

wherein is contained in R1、R7And R8All cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl and tricycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of a carbon atom, and wherein such substitution results in the residue containing at least the same number of C atoms as the number of heteroatoms independently selected from O, S and N;

wherein is contained in R1、R7And R8All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl and tricycloalkyl residues in the definition of (a) may be partially or fully halogenated, in particular fluorinated, more in particular perfluorinated;

Wherein bicyclic and tricyclic residues include fused, bridged and spiro ring systems;

and wherein R1Selected from the group consisting of methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, isopropyl, sec-butyl, tert-pentyl, tert-octyl, 3-pentyl, -CF3、-CF2CF3、-(CF2)2CF3、-CH(CF3)2、-CH2SCH3、-CH2CH2SCH3、-CH2SCH2CH3、-CH2CH2SCH2CH3Methoxymethyl, methoxyethyl, methoxypropyl, ethoxymethyl, ethoxyethyl, propoxymethylAlkyl, dimethyl-aminomethyl, dimethyl-aminoethyl, diethyl-aminomethyl, ethyl-methyl-aminomethyl, cyclopropyl, methyl-cyclopropyl, ethyl-cyclopropyl, trifluoromethyl-cyclopropyl, perfluoroethyl-cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, dicyclopentyl, dicyclohexyl, bicycloheptyl (preferably norbornyl), bicyclooctyl, bicyclononyl, methylcyclononyl, adamantyl, tricyclodecyl, oxiranyl, oxetanyl, tetrahydrofuranyl, methyltetrahydrofuranyl, trimethyltetrahydrofuranyl, tetrahydropyranyl, aziridinyl, N-methylaziridine, azetidinyl, N-methylazetidiyl, difluoroazetidinyl, pyrrolidinyl, N-methylpyrrolidinyl, N-methylazetidinyl, N-methylazetidiyl, difluoroazetidinyl, pyrrolidinyl, N-methylpyrrolidinyl, Piperidinyl, N-methylpiperidinyl, difluoropiperidinyl, thienylpropyl (thiairanyl), thienylbutyl (thietanyl), tetrahydrothienyl, tetrahydrothiopyranyl (tetrahydrothiopyranyl), dioxanyl (dioxanyl), piperazinyl, dimethylpiperazinyl, dithianyl (dithianyl), morpholinyl, N-methylmorpholinyl, thiomorpholinyl, N-methylthiomorpholinyl, oxa-azaspiroheptyl, N-methyloxa-azaspiroheptyl, N-methylazaspiroheptyl, thia-azaspiroheptyl, N-methylthio-azaspiroheptyl, difluorothia-azaspiroheptyl, azaspirooctyl, N-methylazaspirooctyl, oxa-azaspirooctyl, N-methyloxa-azaspirooctyl, oxa-azaspirononyl, N-methyloxa-azaspiro nonyl, N-methylazaspiro nonyl, oxa-azaspiro decyl, N-methyloxa-azaspiro decyl, N-methylazaspiro decyl, dihydro-oxazinyl, N-methyldihydro-oxazinyl, oxazolidinyl, N-methyloxazolidinyl, dioxolanyl, imidazolidinyl, N-methylimidazolidyl, N-dimethylimidazolidinyl, azepanyl, N-methylazepanyl, azaspirohexyl, N-methylazepanyl, oxa-azadispirodecyl, N-methyloxa-azadispirodecyl, N-methylazepanyl, oxa-azabicyclooctyl, oxa-azadispirodecyl, N-methylazepanyl, N-azadispirodecyl, N-azadicyloctyl, N-, N-methyloxa-azabicyclooctyl, N-methylazabicyclooctyl, azabicycloheptyl, N-methylazabicycloheptyl A group, azabicyclononyl, N-methylazabicyclononyl, azaadamantyl, -O (adamantyl), oxa-azabicyclononyl, N-methyloxa-azabicyclononyl, oxaazabicycloheptyl, N-methyldiazabicycloheptyl, diazabicyclooctyl, N-methyldiabicyclooctyl, N-dimethyldiazabicyclooctyl, diazabicycloheptyl, N-methyldiabicycloheptyl, N-dimethyldiazabicycloheptyl; 4-oxocyclohexyl, 3-oxocyclopentyl; 2-oxocyclobutyl, 4-oxobicyclo [4.1.0]Hept-1-yl;

and wherein R1Even more preferably from C4-C12Alkyl radical, C4-C12Alkenyl radical, C4-C12Alkynyl, cyclic, bicyclic and tricyclic residues, wherein the alkyl, alkenyl and alkynyl residues are preferably branched, including:

R2-R5independently of one another, from the group consisting of-H, -F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C2-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl radical, CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C) 1-C3Alkyl) (cyclopropyl);

wherein is contained in R2-R5All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (A) are unsubstituted or independently selected from one or more of-F, -Cl, -Br, -I, -CH3、-CF3-OH and-OCH3、-OCF3、-NH2、-NHCH3、-N(CH3)2Substituted with the substituent(s);

wherein is contained in R2-R5All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in the substitution of carbon atoms, and wherein such substitution does not result in one of the groups selected from C ═ O and S ═ O being bonded directly to the aromatic ring;

wherein R is2-R3Each is preferably-H, R4preferably-H or-F, R5preferably-H, -F, -Cl, -Br, -CH3、-CF3、-CH=CH2、-C≡CH、-CH2OH、-CH2NHCH3、-OH、-OCH3、-OCF3Cyclopropyl, oxirane, -CH2-N-morpholinyl, -C (CH)3)3、-CH2OCH3、-NO2、-CN、-NH2、-N(CH3)2、-OCH(CH3)2、-CH2NH2、-CH2N(CH3)2

Wherein the substituents R are as defined in the general formula (I)1To R5The six-membered aromatic ring incorporated is preferably selected from:

X1-X4are independently selected from N, CR9、CR11、CR12

R9-R12Independently of one another, from the group consisting of-H, -F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C 1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl):

wherein is contained in R9-R12All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (A) are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CH3、-CF3-OH and-OCH3、-OCF3、-NH2、-NHCH3、-N(CH3)2Substituted with the substituent(s);

wherein is contained in R9-R12All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in the substitution of carbon atoms, and wherein such substitution does not result in one of the groups selected from C ═ O and S ═ O being bonded directly to the aromatic ring:

wherein R is9-R12Preferably selected from the group consisting of-H, -F, -Cl, -Br、-CH3、-CF3、-OH、-OCH3、-OCF3Cyclopropyl, oxirane, -C (CH)3)3、-N(CH3)2、-NH2、-CN、-CH2OCH3、-OCH(CH3)2、-CH2NH2、-CH2N(CH3)2、-CH2OH、-NO2、-CH2-N-morpholinyl;

and wherein X is contained as defined in the general formula (I)1-X4The six-membered aromatic ring(s) is preferably selected from:

R6=-H、C1-C8alkyl, preferably C1-C4Alkyl radical, C2-C8Alkenyl, preferably C2-C4Alkenyl radical, C2-C8Alkynyl, preferably C2-C4Alkynyl, C3-C6Cycloalkyl radical, C5-C6Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl, C8-C14Tricycloalkyl and aromatic and heteroaromatic residues, preferably a 6-membered aromatic ring and a 5-to 6-membered heteroaromatic ring;

and wherein bicyclic and tricyclic residues include fused, bridged and spiro ring systems;

wherein is contained in R6Said cycloalkyl, cycloalkenyl bicycloalkyl, bicycloalkenyl, tricycloalkyl, aromatic and heteroaromatic residues in the definition of (1) are optionally interrupted by C 1Alkylene or C2Alkylene or C3An alkylene linker to R6Bound N;

wherein is contained in R6All aromatic and heteroaromatic residues in the definition of (A) are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2,-NO2Straight or branched chain C1-C3Alkyl radical, C2-C3Alkenyl radical, C2-C3Alkynyl, cyclopropyl, straight or branched-OC1-C3Alkyl radicals such as-OCH3-O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl) substituents;

wherein is contained in R6All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl and tricycloalkyl residues and alkylene linkers in the definitions of (A) are straight or branched chain and are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2O, straight or branched C1-C3Alkyl radical, C2-C3Alkenyl radical, C2-C3Alkynyl, cyclopropyl, straight or branched-OC1-C3Alkyl radicals such as-OCH3-O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl) 2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl) substituents;

wherein is contained in R6All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, bicycloalkyl, bicycloalkenyl, tricycloalkyl and heteroaryl residues and alkylene linkers in the definitions of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

wherein R is6preferably-H, -CH3、-CH2CH3N-propyl, isopropyl, cyclopropyl, -CF3and-CF2CF3Benzyl, tert-butyl, phenyl, cyclohexyl, 1-phenylethyl, 2-dimethyl-1-phenylpropyl, (1-naphthyl) -methyl, 4-methoxybenzyl, 4-trifluoromethylbenzyl, tetrahydropyranyl;

wherein is contained in R2-R6And R9-R12All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl in the definition of (a), bicycloalkyl, bicycloalkenyl, tricycloalkyl, aromatic and heteroaromatic residues may be partially or fully halogenated, in particular fluorinated, more in particular perfluorinated;

y ═ H, straight or branched C1-C6Alkyl, straight or branched C2-C6Alkenyl, straight-chain or branched C2-C6Alkynyl, C3-C6Cycloalkyl radical, C5-C6Cycloalkenyl, -OH, straight or branched-OC1-C6Alkyl, straight or branched-OC2-C6Alkenyl, straight or branched-OC2-C6Alkynyl, -OC3-C6Cycloalkyl, -OC 5-C6Cycloalkenyl, -CN, aromatic and heteroaromatic residues, preferably six-membered aromatic rings and five-to six-membered heteroaromatic rings, -S (O) R13and-S (O)2R13Wherein R is13Selected from straight or branched C1-C6Alkyl, straight or branched C2-C6Alkenyl, straight-chain or branchedChain C2-C6Alkynyl, C3-C6Cycloalkyl radical, C5-C6Cycloalkenyl radical, -CF3and-C6H4CH3

Wherein all cycloalkyl, cycloalkenyl, aromatic and heteroaromatic residues comprised in the definition of Y may optionally pass through C1Alkylene, or C2Alkylene, or C3Alkylene, or-O-CH2-, or-O-CH2-CH2-the linker is linked to N to which Y is bound;

wherein all aromatic and heteroaromatic residues comprised in the definition of Y are unsubstituted or are selected from the group consisting of-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C1-C3Alkyl radical, C2-C3Alkenyl radical, C2-C3Alkynyl, cyclopropyl, straight or branched-OC1-C3Alkyl radicals such as-OCH3-O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl) substituents;

wherein all alkyl, alkenyl, alkynyl, cycloalkyl and cycloalkenyl residues and alkylene linkers contained in the definition of Y are straight or branched chain and are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH 2O, straight or branched C1-C3Alkyl radical, C2-C3Alkenyl radical, C2-C3Alkynyl, cyclopropyl, straight or branched-OC1-C3Alkyl radicals such as-OCH2-O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl) substituents;

wherein all alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl and heteroaryl residues contained in the definition of Y and the alkylene linker may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

wherein all alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aromatic and heteroaromatic residues and alkylene linkers comprised in the definition of Y may be partially or fully halogenated, in particular fluorinated, more in particular perfluorinated;

wherein Y is preferably-H, -CH3、-CH2CH3N-propyl, isopropyl, cyclopropyl, cyclohexyl, tetrahydropyranyl, -CF3、-CF2CF3、-OH、-OCH3、-OCH2CH3、-OCH2(cyclopropyl), -CN, -S (O) C (CH)3)3、-S(O)2CH3、-S(O)2CF3、-S(O)2C6H4CH3、-OCH2C6H5and-OC6H5(ii) a And for R6is-H or-CH3Or benzyl, then Y is preferably-OH, -OCH3、-OCH2CH3、-OCH2(cyclopropyl);

wherein Y may be substituted with R6Together form a ring structure, wherein the ring structure comprising an N atom of formula I is selected from the group consisting of 3-membered rings, 4-membered rings, 5-membered rings, 6-membered rings, 5-to 12-membered bicyclic residues, 8-to 14-membered tricyclic residues and heteroaromatic residues, wherein all rings, bicyclic, tricyclic and heteroaromatic residues may additionally comprise one or more heteroatoms independently selected from O, S and N in place of carbon atoms comprised in the ring structure, and wherein all rings, bicyclic, tricyclic and heteroaromatic residues are unsubstituted or substituted with one or more heteroatoms independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -OCH 3、-NH2、-NHCH3、-N(CH3)2、=O、-CH3、-CF3Morpholinyl;

and wherein bicyclic and tricyclic residues include fused, bridged and spiro ring systems;

wherein formula I contains NY and R of an atom6The ring structures taken together are preferably selected from aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, difluoropiperidinyl, morpholinyl azetidinyl, hydroxyazetidinyl, azetidinyl (azetinidinonyl), azetidinyl, difluoroazetidinyl, azaspirohexyl, azaspiroheptyl, difluoroazaspiroheptyl, hydroxyazapiroheptyl, methylhydroxyazaspiroheptyl, trifluoromethylhydroxyazapiroheptyl, azaspirooctyl, azaspirononyl, oxa-azaspiroheptyl, oxa-azaspirooctyl, oxa-azaspirononyl, thia-azaspiroheptyl, oxazolidinyl, tetrahydro-oxazinyl, isoxazolidinyl, oxazinane, isoxazolidine, piperazine;

and wherein Y and R of formula I containing an N atom6Even more preferably the ring structures together are selected from:

Z1and Z2Selected from the following groups:

wherein Z1Selected from straight or branched C1-C3Alkyl, preferably-CH3Cyclopropyl, oxiranyl, N-methyl-aziridinyl, thietanyl, -CN, -N3、-CF3、-CF2CF3And wherein Z is 2Independently selected from-H and straight or branched C1-C3Alkyl, preferably-CH3、-CF3、-CF2CF3(general formula Ia);

wherein Z1Is preferably-CH3、-CF3-CN, cyclopropyl; and/or wherein Z2preferably-H, -CH3and-CF3(ii) a For example:

or wherein Z1And Z2Together are ═ O, ═ S, ═ NR14(formula Ib); wherein R is14Selected from-H, -OH, -OCH3、-CN、-S(O)C(CH3)3、-S(O)2CH3、-S(O)2CF3Straight or branched chain C1-C3Alkyl, preferably-CH3Cyclopropyl, -CF3、-CF2CF3、-CH2CF3、-C6H5、-CH2C6H5

Wherein Z1And Z2Together are preferably ═ O, ═ NR14(ii) a Wherein R is14Preferably selected from-H, -CH3Cyclopropyl, -OH, -OCH3、-CN:

Or wherein Z1And Z2Together form a cyclic residue containing the carbon atom to which they are bound (formula Ic); wherein the cyclic residue is selected from the group consisting of a three-membered ring, a four-membered ring, a five-membered ring and a six-membered ring, wherein all rings optionally may comprise one or more heteroatoms independently selected from O, S and N in place of carbon atoms comprised in the ring structure; wherein all rings are unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -OCH3、-NH2、-NHCH3、-N(CH3)2、=O、-CH3and-CF3Substituted with the substituent(s);

wherein Z1And Z2Together preferably form a 3-or 4-membered cyclic residue comprising the carbon atom to which they are bound; wherein the cyclic residue is preferably selected from the group consisting of cyclopropyl, cyclobutyl, oxirane, oxetanyl, aziridinyl (aziridyl), azetidinyl (azetidinyl), and thietanyl; and wherein the cyclic residue is optionally preferably substituted by-F, -OH, -O CH3、-NH2、-NHCH3、-N(CH3)2、=O、-CH3and-CF3Substitution;

and wherein the cyclic residue is even more preferably selected from:

wherein is contained in Z1And Z2All alkyl and cyclic residues in the definition of (a) may be partially or fully halogenated, in particular fluorinated, more in particular perfluorinated.

R1-R14、X1-X4、Z1、Z2And Y the following preferred definitions of Y may optionally apply independently and/or in combination to all aspects including preferred and specific aspects, all embodiments including preferred and specific embodiments, and all subgenera defined in the present invention:

1)R1preferably four or more, preferably six or more, even more preferably seven or more carbon atoms;

2)R1preferably selected from branched alkyl, alkenyl and alkynyl residues;

3)R1preferably selected from cyclic, bicyclic and tricyclic ring structures, wherein the bicyclic and tricyclic residues include fused, bridged and spiro ring systems;

4)R1preferably no heteroatoms;

5)R1preferably selected from cyclohexyl, norbornyl, bicyclooctyl, bicyclononyl, methylbicyclononyl, tricyclodecyl, most preferably adamantyl, e.g. 1-adamantyl and 2-adamantyl;

6)R1preferably comprising one or more heteroatoms, preferably one, two or three heteroatoms independently selected from O, S and N, for substitution of R1Carbon atoms contained in (1);

7)R1Preferably selected from tetrahydropyranyl, N-methylpiperidinyl, morpholinyl, 4-oxocyclohexyl, azabicycloheptyl, N-methylazabicycloheptylHeptyl, oxa-azabicycloheptyl, N-methyldiazabicycloheptyl, azabicyclooctyl, diazabicyclooctyl, N-methyldiazabicyclooctyl, oxa-azabicyclooctyl, azabicyclononyl, azabicycloalkyl, and-O (adamantyl);

8) preferably two, or more preferably three are independently selected from R2-R5The substituents of (A) are-H, i.e. preferably two, more preferably one independently selected from R2-R5Is different from-H;

9) at two independently selected from R2-R5In the case where the substituents of (A) are different from-H and are in the ortho position relative to the ether linkage, the two substituents are preferably different from-F, -Cl, -Br, -I and-NO2More preferably different from each other;

10) from X1-X4The composition of the defined ring atoms is preferably selected from the following: all X1-X4Independently selected from CR9、CR10、CR11、CR12Or X1-X4One of which is N and the other three are independently selected from CR9、CR10、CR11、CR12Or X1-X4Two of which are N and the other two are independently selected from CR9、CR10、CR11、CR12(ii) a I.e. the aromatic or heterocyclic ring is selected from benzene, pyridine, pyrimidine, pyridazine and pyrazine;

11) preferably two, or more preferably three are independently selected from R 9-R12The substituents of (A) are-H, i.e. preferably two, more preferably one independently selected from R9-R12Is different from-H;

12) in two independent choices of R9-R12In the case where the substituents of (A) are different from-H and are in the ortho position relative to the ether linkage, the two substituents are preferably different from-F, -Cl, -Br, -I and-NO2More preferably different from each other;

13) y is preferably selected from the residues comprised in the general definition of Y, which together with the oxygen atom are bound to the N to which Y is bound;

a preferred aspect of the present invention relates to compounds of the general formula (Ib)A salt and solvate thereof, wherein Z1And Z2Together is ═ O, and wherein R6And at least one of Y is different from H,

and R is1-R5、R7-R13And X1-X4As defined in formula (I), including substituted and preferred definitions.

Another preferred aspect of the invention relates to compounds of general formula (I) and salts and solvates thereof, wherein Y is selected from the group consisting of the residues comprised in the general definition of Y, which together with the oxygen atom are bound to the N to which Y is bound,

and wherein Y is even more preferably-OH, -OCH3、-OCH2CH3、-OCH2(cyclopropyl), -OC6H5and-OCH2C6H5

And R is1-R12、R14、X1-X4、Z1And Z2As defined in formula (I), including substituted and preferred definitions.

Another preferred aspect of the invention relates to compounds of the general formula (I) wherein R is 1Selected from the group consisting of1A residue in the general definition of (1), which comprises 4 or more, preferably 6 or more, even more preferably 7 or more carbon atoms,

and wherein R1The catalyst does not contain a heteroatom,

and wherein R1More preferably selected from cyclic, bicyclic and tricyclic structures,

and wherein R1Even more preferably selected from the group consisting of cyclohexyl, norbornyl, bicyclooctyl, bicyclononyl, methylbicyclononyl, tricyclodecyl and adamantyl,

and wherein R1Most preferably an adamantyl group, or a substituted or unsubstituted group,

and R is2-R6、R9-R14、X1-X4、Z1、Z2And Y is as defined in formula (I), including substituted and preferred definitions.

Another preferred aspect of the present invention relates toCompounds of formula (I) and salts and solvates thereof, wherein R1Is selected from R1A residue comprising 4 or more, preferably 6 or more, even more preferably 7 or more carbon atoms,

and wherein R1Comprising one or more (preferably 1 to 2) heteroatoms independently selected from O, S and N for substitution of R1The carbon atom(s) contained in (a),

and wherein R1Even more preferably selected from cyclic, bicyclic and tricyclic structures, or wherein R is1Selected from the group consisting of residues comprising cyclic, bicyclic and tricyclic structures,

and wherein R1Even more preferably tetrahydropyranyl, N-methylpiperidinyl, morpholinyl, 4-oxocyclohexyl, azabicycloheptyl, N-methylazabicycloheptyl, oxa-azabicycloheptyl, N-methyldiazabicycloheptyl, azabicyclooctyl, diazabicyclooctyl, N-methyldiazabicyclooctyl, oxa-azabicyclooctyl, azabicyclononyl, azaadamantyl and-O (adamantyl),

And wherein R1Most preferably tetrahydropyranyl, N-methylpiperidinyl, morpholinyl, 4-oxocyclohexyl, azabicycloheptyl, azaadamantyl and-O (adamantyl),

and R is2-R14、X1-X4、Z1、Z2And Y is as defined in formula (I), including substituted and preferred definitions.

Yet another preferred aspect of the invention relates to compounds of general formula (I) and salts and solvates thereof, which fall within the subgenus defined herein:

s.1 if Z1And Z2As defined in formula (I), including its substitutions and preferred definitions, with the proviso that Z1And Z2Different from together being either O or S,

then R is1-R13、X1-X4And Y is as defined in formula (I), including substituted and preferred definitions thereof.

S.2 if R6As defined in formula (I), including the derivatives thereofGeneration and preferred definition, with the proviso that R6Other than-H, or linear unsubstituted or branched unsubstituted-C1-C6An alkyl group, a carboxyl group,

then R is1-R5、R7-R14、X1-X4、Z1And Z2As defined in formula (I), including substituted and preferred definitions thereof.

S.3 if Y is as defined in the formula (I), including the substitutions and preferred definitions, with the proviso that Y is different from-H, straight-chain unsubstituted or branched unsubstituted C1-C6An alkyl group or an-OH group,

then R is1-R14、X1-X4、Z1And Z2As defined in formula (I), including substituted and preferred definitions.

S.3 if Y is as defined in the formula (I), including the substitutions and preferred definitions, with the proviso that Y is different from-H, straight-chain unsubstituted or branched unsubstituted C 1-C6An alkyl group or an-OH group,

then R is1-R14、X1-X4、Z1And Z2As defined in formula (I), including substituted and preferred definitions.

S.4 if Z1And Z2Together is ═ O or ═ S, and Y is — OH,

then R is6As defined in formula (I), including substituted and preferred definitions, with the proviso that R6In contrast to the case of the group-H,

and then R1-R5、R7-R12And X1-X4As defined in formula (I), including substituted and preferred definitions.

S.5 if Z1And Z2Together is ═ O or ═ S, and R6is-H, and Y is-H, linear unsubstituted or branched unsubstituted C1-C6An alkyl group or an-OH group,

or if Z is1And Z2Together is ═ O or ═ S, and R6is-H or straight-chain unsubstituted or branched unsubstituted C1-C6Alkyl, and Y is-H. Straight-chain unsubstituted or branched unsubstituted C1-C6An alkyl group, a carboxyl group,

then R is1=C1-C12Alkyl, preferably C1-C6Alkyl radical, C2-C12Alkenyl, preferably C2-C6Alkenyl radical, C2-C12Alkynyl, preferably C2-C6Alkynyl, C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl, C8-C14tricycloalkyl-OC1-C12Alkyl, preferably-OC1-C6Alkyl, -OC2-C12Alkenyl, preferably-OC2-C6Alkenyl, -OC2-C12Alkynyl, preferably-OC2-C6Alkynyl, -OC3-C8Cycloalkyl, -OC5-C8Cycloalkenyl radical, -OC5-C12Bicycloalkyl, -OC7-C12Bicycloalkenyl, -OC8-C14Tricyclic alkyl-SC1-C12Alkyl, preferably-SC1-C6Alkyl, -SC2-C12Alkenyl, preferably-SC2-C6Alkenyl, -SC 2-C12Alkynyl, preferably-SC2-C6Alkynyl, -SC3-C8Cycloalkyl, -SC5-C8Cycloalkenyl radical, -SC5-C12Bicycloalkyl, -SC7-C12Bicycloalkenyl, -SC8-C14Tricycloalkyl, -NHR7or-NR7R8Wherein R is7And R8Independently of one another, selected from: c1-C12Alkyl, preferably C1-C6Alkyl radical, C2-C12Alkenyl, preferably C2-C6Alkenyl radical, C2-C12Alkynyl, preferably C2-C6Alkynyl, C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl, C8-C14Three ringsAlkyl, or wherein R7Can be reacted with R8Together form a ring structure, wherein the ring structure comprising an N atom is selected from a 3-to 8-membered ring structure or a 5-to 12-membered ring structure, and wherein all of the ring structures may additionally comprise one or more heteroatoms independently selected from O, S and N in place of carbon atoms contained in the ring structures, wherein all C atoms are C atoms1-C12Alkyl radical, C2-C8Alkenyl radical, C2-C8Alkynyl, C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl, norbornyl, and adamantyl residues are straight or branched chain and are substituted with one or more substituents (referred to herein as pendant substituents) independently selected from: -OH, -NH2、-NO2、=O、C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl (including norbornyl), C7-C12Bicycloalkenyl, C8-C14Tricycloalkyl (including adamantyl), straight or branched-OC1-C5Alkyl radicals such as-OCH3、-OC3-C5Cycloalkyl radicals such as-O (cyclopropyl), straight-chain or branched-NH (C) 1-C5Alkyl), straight or branched-N (C)1-C5Alkyl) (C1-C5Alkyl), -NH (C)3-C5Cycloalkyl) such as-NH (cyclopropyl), -N (C)3-C5Cycloalkyl) (C)3-C5Cycloalkyl), straight or branched-chain-N (C)1-C5Alkyl) (C3-C5Cycloalkyl groups); and wherein all of said C1-C12Alkyl radical, C2-C8Alkenyl radical, C2-C8Alkynyl, C3-C8Cycloalkyl radical, C5-C8The cycloalkenyl, adamantyl or norbornyl residue may optionally be substituted additionally comprising one or more substituents independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS;

and all C9-C12Alkenyl radical, C9-C12Alkynyl, -OC1-C12Alkyl, -OC2-C12An alkenyl group,-OC2-C12Alkynyl, -SC1-C12Alkyl, -SC2-C12Alkenyl, -SC2-C12Alkynyl and radicals contained in R7And R8All alkyl, alkenyl and alkynyl residues in the definition of (a) are straight or branched chain and are unsubstituted or substituted with one or more substituents (referred to herein as pendant substituents) independently selected from: -F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2、=O、C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl, C8-C14Tricycloalkyl, straight or branched-OC1-C5Alkyl radicals such as-OCH3、-OC3-C5 RingAlkyl radicals such as-O (cyclopropyl), straight-chain or branched-NH (C)1-C5Alkyl), straight or branched-N (C)1-C5Alkyl) (C1-C5Alkyl), -NH (C)3-C5Cycloalkyl) such as-NH (cyclopropyl), -N (C) 3-C5Cycloalkyl) (C)3-C5Cycloalkyl), straight or branched-chain-N (C)1-C5Alkyl) (C3-C5Cycloalkyl groups); wherein all of the-OC3-C8Cycloalkyl, -OC5-C8Cycloalkenyl radical, -SC3-C8Cycloalkyl, -SC5-C8Cycloalkenyl residue, and the radicals contained in R7And R8All cycloalkyl and cycloalkenyl residues in the definition of (1) and included in the selection referred to as side substituents, and also included in R1、R7And R8All bicyclic and tricyclic structures (including bicycloalkyl, bicycloalkenyl and tricycloalkyl residues) within the definition of (A) provided that they are different from adamantyl and norbornyl are unsubstituted or substituted with one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2O, straight or branched C1-C5Alkyl radicals such as-CH3Straight or branched-OC1-C5Alkyl radicals such as-OCH3Straight or branched-NH (C)1-C5Alkyl), straight or branched-N (C)1-C5Alkyl) (C1-C5Alkyl), -NH (C)3-C5Cycloalkyl) such as-NH (cyclopropyl), -N (C)3-C5Cycloalkyl) (C)3-C5Cycloalkyl), straight or branched-chain-N (C)1-C5Alkyl) (C3-C5Cycloalkyl) substituted with a substituent;

wherein is contained in R7And R8All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl bicycloalkyl, bicycloalkenyl and tricycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms; and wherein is contained in R 1All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenylbicycloalkyl, bicycloalkenyl and tricycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms, optionally with the proviso that if not explicitly contained in R1In the definition of (1), then the combination of said heteroatoms in terminal positions is different from the residues-CN, -NCO, -NCS and-N3

Wherein is contained in R1、R7And R8All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenylbicycloalkyl, bicycloalkenyl and tricycloalkyl residues in the definition of (a) may be partially or fully halogenated, in particular fluorinated, more in particular perfluorinated;

wherein bicyclic and tricyclic residues include fused, bridged and spiro ring systems;

and R is2-R5、R9-R12And X1-X4As defined in formula (I), including substituted and preferred definitions thereof.

S.6 if Z1And Z2Together is ═ O or ═ S, and R6is-H, and Y is-H, linear unsubstituted or branched unsubstituted C1-C6An alkyl group or an-OH group,

or if Z is1And Z2Together is ═ O or ═ S, and R6is-H or straight-chain unsubstituted or branched unsubstituted C1-C6Alkyl, and Y is-H, straight chain unsubstituted or branched unsubstituted C1-C6An alkyl group, a carboxyl group,

then R is2Selected from the group consisting of-CN, -NCO, -NCS, -OH, -NH 2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl);

wherein all C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl and C3-C4The cycloalkyl residue is substituted by one or more groups independently selected from-OH, -OCH3、-OCF3、-NH2、-NHCH3and-N (CH)3)2Substituted with the substituent(s);

wherein said C5-C6The cycloalkyl residue is unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CH3、-CF3、-OH、-OCH3、-OCF3、-NH2、-NHCH3and-N (CH)3)2Substituted with the substituent(s);

wherein is contained in R2All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

and R is3-R5Independently of one another, from the group consisting of-H, -F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C) 1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl);

wherein is contained in R3-R5All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (A) are unsubstituted or independently selected from one or more of-F, -Cl, -Br, -I, -CH3、-CF3-OH and-OCH3、-OCF3、-NH2、-NHCH3、-N(CH3)2Substituted with the substituent(s);

wherein is contained in R3-R5All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

and R is1,R7-R12And X1-X4As defined in formula (I), including substituted and preferred definitions thereof.

S.7 if Z1And Z2Together is ═ O or ═ S, and R6is-H, and Y is-H, linear unsubstituted or branched unsubstituted C1-C6An alkyl group or an-OH group,

or if Z is1And Z2Together is ═ O or ═ S, and R6is-H or straight-chain unsubstituted or branched unsubstituted C1-C6Alkyl, and Y is-H, straight chain unsubstituted or branched unsubstituted C1-C6An alkyl group, a carboxyl group,

then X1Is CR9

And R is9Selected from the group consisting of-CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl radical、C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C) 1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl);

wherein all C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl and C3-C4The cycloalkyl residue is substituted by one or more groups independently selected from-OH, -OCH3、-OCF3、-NH2、-NHCH3and-N (CH)3)2Substituted with a substituent group (b);

wherein C is5-C6The cycloalkyl residue is unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CH3、-CF3、-OH、-OCH3、-OCF3、-NH2、-NHCH3and-N (CH)3)2Substituted with the substituent(s);

wherein is contained in R9All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

and then R1-R5、R7、R8、R10-R12And X2-X4As defined in formula (I), including substituted and preferred definitions.

S.8 if Z1And Z2Together is ═ O or ═ S, and R6is-H, and Y is-H, linear unsubstituted or branched unsubstituted C1-C6An alkyl group or an-OH group,

or if Z is1And Z2Together is ═ O or ═ S, and R6is-H or straight-chain unsubstituted or branched unsubstituted C1-C6Alkyl, and Y is-H, straight chain unsubstituted or branched unsubstituted C1-C6An alkyl group, a carboxyl group,

then X2Is CR9

And R is9Selected from the group consisting of-CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C 2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl);

wherein all C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl and C3-C4The cycloalkyl residue is substituted by one or more groups independently selected from-OH, -OCH3、-OCF3、-NH2、-NHCH3and-N (CH)3)2Substituted with the substituent(s);

wherein C is5-C6The cycloalkyl residue is unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CH3、-CF3、-OH、-OCH3、-OCF3、-NH2、-NHCH3and-N (CH)3)2Substituted with the substituent(s);

wherein is contained in R9All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

and then R1-R7、R7、R8、R10-R12、X1、X3And X4As defined in formula (I), including substituted and preferred definitions thereof.

S.9 if Z1And Z2Together is ═ O or ═ S, and R6is-H, Y is-H, linear unsubstituted or branched unsubstituted C1-C6An alkyl group or an-OH group,

or if Z is1And Z2Together is ═ O or ═ S, and R6is-H or straight-chain unsubstituted or branched unsubstituted C1-C6Alkyl, and Y is-H, straight chain unsubstituted or branched unsubstituted C 1-C6An alkyl group, a carboxyl group,

then X3Is CR9

And R is9Selected from the group consisting of-CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl);

wherein all C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl and C3-C4The cycloalkyl residue is substituted by one or more groups independently selected from-OH, -OCH3、-OCF3、-NH2、-NHCH3and-N (CH)3)2Substituted with the substituent(s);

wherein C is5-C6The cycloalkyl residue is unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CH3、-CF3、-OH、-OCH3、-OCF3、-NH2、-NHCH3and-N (CH)3)2Substituted with the substituent(s);

wherein is contained in R9All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

and R is1-R5、R7、R10-R12、X1、X2And X4As defined in formula (I), including substituted and preferred definitions thereof.

S.10 if Z1And Z2Together is ═ O or ═ S, and R6is-H, and Y is-H, linear unsubstituted or branched unsubstituted C1-C6An alkyl group or an-OH group,

or if Z is1And Z2Together is ═ O or ═ S, and R 6is-H or straight-chain unsubstituted or branched unsubstituted C1-C6Alkyl, and Y is-H, straight chain unsubstituted or branched unsubstituted C1-C6An alkyl group, a carboxyl group,

then X4Is CR9

And R is9Selected from the group consisting of-CN, -NCO, -NCS, -OH, -NH2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl);

wherein all C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl and C3-C4The cycloalkyl residue is substituted by one or more groups independently selected from-OH, -OCH3、-OCF3、-NH2、-NHCH3and-N (CH)3)2Substituted with the substituent(s);

wherein C is5-C6The cycloalkyl residue is unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CH3、-CF3、-OH、-OCH3、-OCF3、-NH2、-NHCH3and-N (CH)3)2Substituted with the substituent(s);

wherein is contained in R9All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

and R is1-R5、R7、R8、R10-R12And X1-X3As defined in formula (I), including substituted and preferred definitions thereof.

S.11 if Z1And Z2Together is ═ O or ═ S, and R 6is-H, and Y is-H, linear unsubstituted or branched unsubstituted C1-C6An alkyl group or an-OH group,

or if Z is1And Z2Together is ═ O or ═ S, and R6is-H or straight-chain unsubstituted or branched unsubstituted C1-C6Alkyl, and Y is-H, straight-chain unsubstituted or branched unsubstituted C1-C6An alkyl group, a carboxyl group,

then X1、X2And X3Each is N

And then R1-R5、R7-R12And X4As defined in formula (I), including substituted and preferred definitions.

S.12 if Z1And Z2Together is ═ O or ═ S, and R6is-H, and Y is-H, linear unsubstituted or branched unsubstituted C1-C6An alkyl group or an-OH group,

or if Z is1And Z2Together is ═ O or ═ S, and R6is-H or straight-chain unsubstituted or branched unsubstituted C1-C6Alkyl, and Y is-H, straight chain unsubstituted or branched unsubstituted C1-C6An alkyl group, a carboxyl group,

then X1、X2And X4Each is N

And then R1-R5、R7-R12And X3As defined in formula (I), including substituted and preferred definitions.

S.13 if Z1And Z2Together is ═ O or ═ S, and R6is-H, and Y is-H, linear unsubstituted or branched unsubstituted C1-C6An alkyl group or an-OH group,

or if Z is1And Z2Together is ═ O or ═ S, and R6is-H or straight-chain unsubstituted or branched unsubstituted C1-C6Alkyl, and Y is-H, straight chain unsubstituted or branched unsubstituted C 1-C6An alkyl group, a carboxyl group,

then X1、X3And X4Each is N

And then R1-R5、R7-R12And X2As defined in formula (I), including substituted and preferred definitions.

S.14 if Z1And Z2Together is ═ O or ═ S, and R6is-H, and Y is-H, linear unsubstituted or branched unsubstituted C1-C6An alkyl group or an-OH group,

or if Z is1And Z2Together is ═ O or ═ S, and R6is-H or straight-chain unsubstituted or branched unsubstituted C1-C6Alkyl, and Y is-H, straight-chain unsubstituted or branched unsubstituted C1-C6An alkyl group, a carboxyl group,

then X2、X3And X4Each is N

And then R1-R5、R7-R12And X1As defined in the general formula (I), including the substituted and preferred definitions thereof

S.15 if R1As defined in formula (I), including substituted and preferred definitions, with the proviso that R1Comprising one or more heteroatoms independently selected from O, S and N, with the proviso that the combination of heteroatoms in terminal positions is different from the residues-CN, -NCO, -NCS,

then R is2-R14、X1-X4、Y、Z1And Z2As defined in formula (I), including substituted and preferred definitions thereof.

S.16 if Z1And Z2As defined in formula (I), including its substitutions and preferred definitions, with the proviso that Z1And Z2Are not taken together as ═ O,

then R is1-R14、X1-X4And Y is as defined in formula (I), including substituted and preferred definitions thereof.

S.17 if R6As defined in formula (I), including substituted and preferred definitions, with the proviso that R 6and-H, or-OC1-C6Alkyl, or-C3-C6The cycloalkyl groups are different from each other in that,

wherein all of said-C1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

then R is1-R5、R7-R14、X1-X4、Y、Z1And Z2As defined in formula (I), including substituted and preferred definitions.

S.18 if Y is as defined in formula (I), including the substitutions and preferred definitions, with the proviso that Y is different from-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, or-OH, or-OC1-C6An alkyl group, a carboxyl group,

wherein all of said C1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more groups independently selected from-F, -Cl, and,-Br、-I、C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said-C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

Then R is1-R14、X1-X4、Z1And Z2As defined in formula (I), including substituted and preferred definitions thereof.

S.19 if Z1And Z2Together is ═ O, and R6is-H, or-C1-C6Alkyl or-C3-C6Cycloalkyl, and Y is-H, or C1-C6Alkyl or C3-C6Cycloalkyl, or-OH, or-OC1-C6An alkyl group, a carboxyl group,

wherein all of said-C1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said-C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

then R is1=C1-C12Alkyl, preferably C1-C6Alkyl radical, C2-C12Alkenyl, preferably C2-C6Alkenyl radical, C2-C12Alkynyl, preferably C2-C6Alkynyl, C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl, C8-C14Tricycloalkyl, -OC1-C12Alkyl, preferably-OC1-C6Alkyl, -OC2-C12Alkenyl, preferably-OC2-C6Alkenyl, -OC2-C12Alkynyl, preferably-OC2-C6Alkynyl, -OC3-C8Cycloalkyl, -OC5-C8Cycloalkenyl radical, -OC5-C12Bicycloalkyl, -OC7-C12Bicycloalkenyl, -OC8-C14Tricycloalkyl, -SC1-C12Alkyl, preferably-SC1-C6Alkyl, -SC 2-C12Alkenyl, preferably-SC2-C6Alkenyl, -SC2-C12Alkynyl, preferably-SC2-C6Alkynyl, -SC3-C8Cycloalkyl, -SC5-C8Cycloalkenyl radical, -SC5-C12Bicycloalkyl, -SC7-C12Bicycloalkenyl, -SC8-C14Tricycloalkyl, -NHR7or-NR7R8

Wherein R is7And R8Independently of one another, selected from: c1-C12Preferably C1-C6Alkyl radical, C2-C12Alkenyl, preferably C2-C6Alkenyl radical, C2-C12Alkynyl, preferably C2-C6Alkynyl, C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl, C8-C14Tricycloalkyl, or wherein R7Can be reacted with R8Together form a ring structure, wherein the ring structure comprising an N atom is selected from a 3-to 8-membered ring structure or a 5-to 12-membered bicyclic structure, and wherein all of the ring structures may additionally comprise one or more heteroatoms independently selected from O, S and N in place of carbon atoms comprised in the ring structure;

wherein all C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl and C8-C14The tricycloalkyl residue is straight or branched chain and is substituted with one or more substituents (referred to herein as pendant substituents) independently selected from the group consisting of: -OH, -NH2、-NO2、=O、C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl, C8-C14Tricycloalkyl straight or branched-OC4-C5Alkyl, -OC3-C5Cycloalkyl radicals such as-O (cyclopropyl), straight-chain or branched-NH (C) 1-C5Base), straight or branched-N (C)1-C5Alkyl) (C1-C5Alkyl), -NH (C)3-C5Cycloalkyl) such as-NH (cyclopropyl), -N (C)3-C5Cycloalkyl) (C)3-C5Cycloalkyl), straight or branched-chain-N (C)1-C5Alkyl) (C3-C5Cycloalkyl groups); and wherein all of said C1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl and C8-C14The tricycloalkyl residue may optionally additionally comprise one or more substituents independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS;

and all-OC1-C12Alkyl, -OC2-C12Alkenyl, -OC2-C12Alkynyl, -SC1-C12Alkyl, -SC2-C12Alkenyl, -SC2-C12Alkynyl and radicals contained in R6And R7All alkyl, alkenyl and alkynyl residues in the definition of (1) are straight-chain or branched and unsubstituted or substituted by one or moreSubstituted with a group (referred to herein as a pendant substituent) independently selected from: -F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2、=O、C3-C8Cycloalkyl radical, C5-C8Cycloalkenyl radical, C5-C12Bicycloalkyl radical, C7-C12Bicycloalkenyl, C8-C14Tricycloalkyl straight or branched-OC1-C5Alkyl radicals such as-OCH3、-OC3-C5Cycloalkyl radicals such as-O (cyclopropyl), straight-chain or branched-NH (C)1-C5Alkyl), straight or branched-N (C)1-C5Alkyl) (C1-C5Alkyl), -NH (C)3-C5Cycloalkyl) such as-NH (cyclopropyl), -N (C)3-C5Cycloalkyl) (C) 3-C5Cycloalkyl), straight or branched-chain-N (C)1-C5Alkyl) (C3-C5Cycloalkyl groups);

wherein all of the-OC3-C8Cycloalkyl, -OC5-C8Cycloalkenyl radical, -OC5-C12Bicycloalkyl, -OC7-C12Bicycloalkenyl, -OC8-C14Tricyclic alkyl-SC3-C8Cycloalkyl, -SC5-C8Cycloalkenyl radical, -SC5-C12Bicycloalkyl, -SC7-C12Bicycloalkenyl, -SC8-C14A tricycloalkyl residue, and as contained in R7And R8All cycloalkyl, cycloalkenylbicycloalkyl, bicycloalkenyl and tricycloalkyl residues in the definitions and included in the selection as pendant substituents are substituted or by one or more substituents independently selected from: -F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH, -NH2、-NO2O, straight or branched C1-C5Alkyl radicals such as-CH3Straight or branched-OC1-C5Alkyl radicals such as-OCH3Straight or branched-NH (C)1-C5Alkyl), straight or branched-N (C)1-C5Alkyl) (C1-C5Alkyl), -NH (C)3-C5Cycloalkyl) such as-NH (cyclopropyl), -N (C)3-C5Cycloalkyl) (C)3-C5Cycloalkyl), straight or branched-chain-N (C)1-C5Alkyl) (C3-C5Cycloalkyl groups);

wherein is contained in R7And R8All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl bicycloalkyl, bicycloalkenyl and tricycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms; and wherein is contained in R 1All alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenylbicycloalkyl, bicycloalkenyl and tricycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms, provided that if not explicitly contained in R1In the definition of (1), then the combination of said heteroatoms in terminal positions is different from the residues-CN, -NCO, -NCS and-OC1-C3An alkyl group;

wherein bicyclic or tricyclic residues include fused, bridged and spiro ring systems;

and R is2-R5、R9-R12And X1-X4As defined in formula (I), including substituted and preferred definitions thereof.

S.20 if Z1And Z2Together is ═ O, and R6is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, and Y is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, or-OH or-OC1-C6An alkyl group, a carboxyl group,

wherein all of said C1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said-C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

Then R is2Selected from the group consisting of-F, -Cl, -Br, -I, -CN, -NCO, -NCS, -OH and-NH2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl);

wherein is contained in R2All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (A) are unsubstituted or independently selected from one or more of-F, -Cl, -Br, -I, -CH3、-CF3-OH and-OCH3、-OCF3、-NH2、-NHCH3、-N(CH3)2Substituted with the substituent(s);

wherein is contained in R2All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of a carbon atom;

and then R1、R3-R5、R7-R12And X1-X4As defined in formula (I), including substituted and preferred definitions thereof.

S.21 if Z1And Z2Together is ═ O, and R6is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, and Y is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, or-OH or-OC1-C6An alkyl group, a carboxyl group,

whereinHaving the-OC1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more independently selected from-F, -Cl, -Br, -I, -C 1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

then X1Is CR9

And R is9Selected from the group consisting of-Cl, -Br, -I, CN, -NCO, -NCS, -OH and-NH2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl);

r wherein all alkyl, alkenyl, alkynyl and cycloalkyl residues are contained in the definition9Unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CH3、-CF3-OH and-OCH3、-OCF3、-NH2、NHCH3、N(CH3)2Substituted with the substituent(s);

r wherein all alkyl, alkenyl, alkynyl and cycloalkyl residues are contained in the definition9May contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

and then R1-R5、R7、R8、R10-R12And X2-X4As defined in formula (I), including substituted and preferred definitions.

S.22 if Z1And Z2Together is ═ O, and R6is-H, or C1-C6Alkyl or C3-C6Cycloalkyl, and Y is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, or-OH, or-OC1-C6An alkyl group, a carboxyl group,

wherein all of said C1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

then X2Is CR9

And R is9Selected from the group consisting of-Cl, -Br, -I, CN, -NCO, -NCS, -OH and-NH2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl);

wherein is contained in R9All alkanes in the definition ofThe radicals, alkenyl, alkynyl and cycloalkyl residues being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CH 3、-CF3-OH and-OCH3、-OCF3、-NH2、NHCH3、N(CH3)2Substituted with the substituent(s);

wherein is contained in R9All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

and then R1-R5、R7、R8、R10-R12、X1、X3And X4As defined in formula (I). Including the substitution and preferred definitions thereof.

S.23 if Z1And Z2Together is ═ O, and R6is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, and Y is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, or-OH or-OC1-C6An alkyl group, a carboxyl group,

wherein all of said C1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

then X3Is CR9

And R is9Selected from the group consisting of-F, -Cl, -Br, -I, CN, -NCO, -NCS, -OH and-NH2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC 1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl);

wherein is contained in R9All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (A) are unsubstituted or independently selected from one or more of-F, -Cl, -Br, -I, -CH3、-CF3-OH and-OCH3、-OCF3、-NH2、NHCH3、N(CH3)2Substituted with the substituent(s);

wherein is contained in R9All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

and R is1-R5、R7、R8、R10-R12、X1、X2And X4As defined in formula (I), including substituted and preferred definitions thereof.

S.24 if Z1And Z2Together is ═ O, and R6is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, and Y is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, or-OH or-OC1-C6An alkyl group, a carboxyl group,

wherein all of said C1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said C3-C6Cycloalkyl residues not being takenSubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C 1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

then X4Is CR9

And R is9Selected from the group consisting of-F, -Cl, -Br, -I, CN, -NCO, -NCS, -OH and-NH2、-NO2Straight or branched chain C1-C4Alkyl, straight or branched C2-C4Alkenyl, straight-chain or branched C2-C4Alkynyl, C3-C6Cycloalkyl, -CH2(C3-C6Cycloalkyl), linear or branched-OC1-C3Alkyl, -O (cyclopropyl), straight or branched-NH (C)1-C3Alkyl), straight or branched-N (C)1-C3Alkyl) (C1-C3Alkyl), -NH (cyclopropyl), -N (cyclopropyl)2Straight or branched chain-N (C)1-C3Alkyl) (cyclopropyl);

wherein is contained in R8All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (A) are unsubstituted or independently selected from one or more of-F, -Cl, -Br, -I, -CH3、-CF3-OH and-OCH3、-OCF3、-NH2、NHCH3、N(CH3)2Substituted with the substituent(s);

wherein is contained in R9All alkyl, alkenyl, alkynyl and cycloalkyl residues in the definition of (a) may contain one or more heteroatoms independently selected from O, S and N in place of carbon atoms;

and then R1-R5、R7、R8、R10-R12And X1-X3As defined in formula (I), including substituted and preferred definitions thereof.

S.25 if Z1And Z2Together is ═ O, and R6is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, and Y is-H, or C 1-C6Alkyl, or C3-C6Cycloalkyl, or-OH or-OC1-C6An alkyl group, a carboxyl group,

wherein all of said C1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

then X3Is N

And then R1-R5、R7-R12、X1、X2And X4As defined in formula (I), including substituted and preferred definitions thereof.

S.26 if Z1And Z2Together is ═ O, and R6is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, and Y is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, or-OH or-OC1-C6An alkyl group, a carboxyl group,

wherein all of said C1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

And wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

then X4Is N

And then R1-R5、R7-R12、X1To X3As defined in formula (I), including substituted and preferred definitions thereof.

S.27 if Z1And Z2Together is ═ O, and R6is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, and Y is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, or-OH or-OC1-C6An alkyl group, a carboxyl group,

wherein all of said C1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

then X1And X4Each is N

And then R1-R5、R7-R12、X3And X4As defined in formula (I), including substituted and preferred definitions thereof.

S.28 if Z1And Z2Together is ═ O, and R6is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, and Y is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, or-OH or-OC1-C6An alkyl group, a carboxyl group,

wherein all of said C1-C6Alkyl radicalThe residue is straight-chain or branched, and is unsubstituted or substituted by one or more independently selected from-F, -Cl, -Br, -I, C 1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

then X1And X3Each is N

And R is1-R5、R7-R12、X2And X4As defined in formula (I), including substituted and preferred definitions thereof.

S.29 if Z1And Z2Together is ═ O, and R6is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, and Y is-H, or C1-C6Alkyl, or C3-C6Cycloalkyl, or-OH or-OC1-C6An alkyl group, a carboxyl group,

wherein all of said C1-C6The alkyl residue is straight-chain or branched and is unsubstituted or substituted by one or more independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said C3-C6The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl and cycloalkyl residues may optionally be halogenated or perhalogenated,

then X1And X4Each is N

And R is1-R5、R7-R12、X2And X3As defined by formula (I), including substituted and preferred definitions thereof.

S.30 if R1As defined in formula (I), including substituted and preferred definitions, with the proviso that R1Is different from C3-C8A cycloalkyl group,

wherein said C3-C8The cycloalkyl residue being unsubstituted or substituted by one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, C1-C3Alkyl radicals and-OC1-C3the substituent of the alkyl group is substituted,

wherein said C3-C8The cycloalkyl residue may optionally be perhalogenated

And wherein said C3-C8The cycloalkyl residues being chosen from C at the same carbon atom (said carbon atom being bound to the phenyl ring defined in formula (I))1-C12Alkyl radical, C2-C12Alkenyl radical, C2-C12Alkynyl, C3-C8Cycloalkyl or C5-C8The substituent group of the cycloalkenyl is substituted,

wherein all of said alkyl, alkenyl and alkynyl residues are straight or branched chain and unsubstituted or substituted one or more is independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS and-OC1-C3The substituent of the alkyl group is substituted,

wherein all of said cycloalkyl and cycloalkenyl residues are unsubstituted or substituted with one or more groups independently selected from-F, -Cl, -Br, -I, -CN, -NCO, -NCS, C1-C3Alkyl and-OC1-C3The substituent of the alkyl group is substituted,

and wherein all of said alkyl, alkenyl, alkynyl, cycloalkyl and cycloalkenyl residues may optionally be perhalogenated,

then R is2-R14、X1-X4、Y、Z1And Z2As defined by formula (I), including substituted and preferred definitions thereof.

In one embodiment, the invention relates to compounds of formula (I) and salts thereofAnd solvates of which R1Is an adamantyl group, or a substituted or unsubstituted alkyl group,

and wherein Z1And Z2As defined in general formula (I) (including general formula (Ia), general formula (Ib) and general formula (Ic)), including substitutions and preferred definitions, optionally with the proviso that in the case of general formula (Ib) Z1And Z2Together with O, is different from O,

and wherein R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein Y, R2-R6、R9-R13、X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (I-1):

and wherein the compounds of structure (I-1) are preferably for use in human and veterinary medicine, in particular for medical uses as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0014, XPW-0028, XPW-0042, XPW-0182, XPW-0924, XPW-3038, XPW-3052, XPW-4633, XPW-4642 and XPW-4643.

In another particular embodiment, the invention relates to compounds of the general formula (I) and salts and solvates thereof, wherein Y and R6As defined in formula (I), including substituted and preferred definitions, wherein Y and R6Together form a ring structure, and wherein such ring structure contains an O atom instead of directly with Y and R6One of the ring C atoms to which the bonded N atoms are attached,

and wherein Z1And Z2As defined in general formula (I), including general formula (Ia), general formula (Ib) and general formula (Ic), including substituted and preferred definitions,

and wherein R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R1-R5、R7-R12、X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (I-2):

and wherein the compounds of structure (I-2) are preferably used in human and veterinary medicine, particularly for medical uses as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, stomach cancers, breast cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-4637 and XPW-4638.

In another particular embodiment, the present invention relates to compounds of formula (I) and salts and solvates thereof, wherein Y is selected from the group consisting of-S (O) R13and-S (O)2R13

And wherein R1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Comprising 4 or more, preferably 6 or more carbon atoms optionally independently replaced by a heteroatom selected from O, S and N as defined in formula (I),

and wherein Z1And Z2As defined in general formula (I), including general formula (Ia), general formula (Ib) and general formula (Ic), including substituted and preferred definitions,

and wherein R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R2-R13And X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (I-3):

and wherein the compounds of structure (I-3) are preferably for use in human and veterinary medicine, in particular for medical uses as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0547, XPW-0548, XPW-0552, XPW-0560, XPW-0566, XPW-0574, XPW-0575, XPW-0576, XPW-0580, XPW-0588, XPW-0603, XPW-0604, XPW-0608, XPW-0616, XPW-2675, XPW-2676, XPW-2688, XPW-2703, XPW-2704, XPW-2708, XPW-2716, XPW-2732, XPW-2744, XPW-4633 and XPW-4642.

In another particular embodiment, the invention relates to compounds of formula (I) and salts and solvates thereof, wherein Y is-OH,

and wherein R6As defined in formula (I), including substituted and preferred definitions, with the proviso that R6In contrast to the case of the group-H,

and wherein Z1And Z2Such as general formula (I) (including general formula (Ia), general formula (Ib) and general formula (Ic)), including substitutions and preferred definitions,

and wherein R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R1-R5、R7-R12And X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (I-4):

and wherein the compounds of structure (I4) are preferably for use in human and veterinary medicine, in particular for medical uses as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0182, XPW-0674, XPW-0675, XPW-0678, XPW-0679, XPW-0686, XPW-0700, XPW-0734, XPW-0742, XPW-1750, XPW-2805, XPW-2806, XPW-4612, XPW-4614, XPW-4616, XPW-4617, XPW-4618, XPW-4619, XPW-4620, XPW-4621, XPW-4622, XPW-4626, XPW-4631, XPW-4632, XPW-4640, XPW-4644, XPW-4646 and XPW-4647.

In another particular embodiment, the invention relates to compounds of general formula (I) and salts and solvates thereof, wherein Y is-OCH3

And wherein R1R as defined in formula (I), including substituted and preferred definitions, including any substituents1Does not contain a heteroatom selected from O, S, N, optionally with the proviso that R1Containing 2 or more carbon atoms in the molecule(s),

and wherein Z1And Z2As defined in general formula (I), including general formula (Ia), general formula (Ib) and general formula (Ic), including substituted and preferred definitions,

and wherein R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R2-R6、R9-R12And X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (I-5):

and wherein the compounds of structure (I-5) -especially without additional conditions-are preferably used in human and veterinary medicine, in particular for medical uses as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, gastric cancers, breast cancers.

Examples are the compounds XPW-0702, XPW-0706, XPW-0714, XPW-0716, XPW-0720, XPW-0728, XPW-2833, XPW-2834, XPW-2847, XPW-2848 and XPW-4605.

In another particular embodiment, the invention relates to compounds of the general formula (I) and salts and solvates thereof, wherein R6is-H and Y is-OCH3

And wherein R1As defined in formula (I), including substituted and preferred definitions, with the proviso that R1Selected from the group consisting of cyclic, bicyclic and tricyclic structures,

and wherein Z1And Z2As defined in general formula (I), including general formula (Ia), general formula (Ib) and general formula (Ic), including substituted and preferred definitions,

and wherein R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R2-R5、R7-R12And X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (I-6):

and wherein the compounds of structure (I-6) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers.

Examples are the compounds XPW-0706, XPW-0714, XPW-2833 and XPW-2834.

In another particular embodiment, the invention relates to compounds of the general formula (I) and salts and solvates thereof, wherein Y and R6As defined in formula (I), including substituted and preferred definitions, wherein Y and R6Together form a ring structure that is,

and wherein R1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Selected from the group consisting of cyclic, bicyclic and tricyclic structures, optionally with the proviso that R includes any substituent1Containing no heteroatom selected from O, S, N or containing one heteroatom selected from O, S, N,

and wherein Z1、Z2And R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R2-R5、R7-R12And X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (I-7):

and wherein the compounds of structure (I-7) -especially in the absence of additional conditions-are preferably used in human and veterinary medicine, especially in medical uses as described herein, preferably in immune system-related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system-related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, gastric cancers, breast cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0762, XPW-0770, XPW-0776, XPW-0784, XPW-0790, XPW-0798, XPW-0818, XPW-2890, XPW-2898, XPW-2904, XPW-2912, XPW-2918, XPW-2926, XPW-4576, XPW-4577, XPW-4578, XPW-4579, XPW-4580, XPW-4581, XPW-4583, XPW-4586, XPW-4589, XPW-4592 and XPW-4594.

In another particular embodiment, the invention relates to compounds of formula (Ia) and salts and solvates thereof, wherein Z1is-CF3

And wherein Y is as defined in formula (I), including substitutions and preferred definitions, optionally with the proviso that Y is different from-H,

and wherein Z2As defined in formula (Ia), including substituted and preferred definitions,

and wherein R1-R13And X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ia-1):

and wherein the compounds of structure (Ia-1) -especially in the absence of additional conditions-are preferably for use in human and veterinary medicine, in particular for medical uses as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, gastric cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0014, XPW-0020, XPW-0028, XPW-0042, XPW-0182, XPW-4633, XPW-4642 and XPW-4643.

In another particular embodiment, the invention relates to compounds of formula (Ia) and salts and solvates thereof, wherein Z1Is CF3

And wherein R6As defined in formula (I), including substituted and preferred definitions, optionally with the proviso that R is6In contrast to the case of the group-H,

and wherein Z2As defined in formula (Ia), including substituted and preferred definitions,

and wherein R1-R5、R7-R13、X1-X4And Y is as defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ia-2):

and wherein the compounds of structure (Ia-2) -especially in the absence of additional conditions-are preferably for use in human and veterinary medicine, in particular for medical uses as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, gastric cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0014, XPW-0020, XPW-0028, XPW-0042, XPW-0182, XPW-4633, XPW-4642 and XPW-4643.

In another particular embodiment, the invention relates to compounds of formula (Ia) and salts and solvates thereof, wherein Z1is-CF3And wherein Y and R6Each of which is-H, and,

and wherein R1As defined in formula (I), including substituted and preferred definitions, optionally with the proviso that R is1Containing 5 or more, preferably 6 or more carbon atoms, optionally independently replaced by a heteroatom selected from O, S and N as defined in formula (I),

and wherein Z2As defined in formula (I), including substituted and preferred definitions,

and wherein R2-R5、R7-R12And X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ia-3):

and wherein the compounds of structure (Ia-3) -especially in the absence of additional conditions-are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the haematopoietic and haematological system (such as leukaemia and lymphoma), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, ovarian cancers and cancers of the neuroendocrine system.

An example is the compound XPW-0014.

In another particular embodiment, the invention relates to compounds of formula (Ia) and salts and solvates thereof, wherein Z1Is a group of-CN which is,

and wherein R1As defined in formula (I), including substituted and preferred definitions, and wherein R1Comprises 3 or more, preferably 6 or more carbon atoms, optionally independently substituted by a heteroatom selected from O, S and N as defined in formula (I) optionally with the proviso that R includes any substituents1Free of a heteroatom selected from the group consisting of O, S, N,

and wherein Z2As defined in formula (Ia), including substituted and preferred definitions,

and wherein R2-R13、X1-X4And Y is as defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ia-4):

and wherein the compounds of structure (Ia-4) -especially in the absence of additional conditions-are preferably for use in human and veterinary medicine, in particular for medical uses as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemia) and skin cancers.

An example is the compound XPW-0314.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein R1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Comprises 6 or more carbon atoms optionally independently replaced by a heteroatom selected from O, S and N as defined in formula (I), and wherein R is1Selected from the group consisting of cyclic, bicyclic and tricyclic structures,

and wherein R5As defined in formula (I), including substituted and preferred definitions, with the proviso that R5In contrast to the case of the group-H,

and wherein Z1、Z2And R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R2-R4、R6-R13And X1-X4And Y is as defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-1):

and wherein the compounds of structure (Ib-1) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-4575, XPW-4577, XPW-4578, XPW-4579, XPW-4580, XPW-4581, XPW-4583, XPW-4584, XPW-4585, XPW-4586, XPW-4587, XPW-4588, XPW-4589, XPW-4590, XPW-4591, XPW-4592, XPW-4593, XPW-4594 and XPW-4595.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein X4The content of the N is N,

and wherein R1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Containing six or more carbon atoms, optionally independently substituted by a heteroatom selected from O, S and N as defined in formula (I), and whereinR1Selected from the group consisting of cyclic, bicyclic and tricyclic structures,

and wherein Z1、Z2And R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R2-R13、X1-X4And Y is as defined for formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-2):

and wherein the compounds of structure (Ib-2) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-4623, XPW-4624, XPW-4628, XPW-4629, XPW-4630, XPW-4631, XPW-4632, XPW-4634, XPW-4635, XPW-4636 and XPW-4644.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein Z1And Z2Together is ═ O, and wherein Y is-OCH3

And wherein R1R as defined in formula (I), including substituted and preferred definitions, including any substituents1Does not contain a heteroatom selected from O, S, N, optionally with the proviso that R1Containing two or more carbon atoms

And wherein R2-R6、R9-R12And X1-X4As defined in formula (I), including substitutionsAnd the definition of the preferred one is,

and wherein the compounds share the following structure (Ib-3):

and wherein the compounds of structure (Ib-3) -especially without additional conditions-are preferably for use in human and veterinary medicine, in particular for medical use as described in the present invention, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined in the present invention, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological system (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, gastric cancers, breast cancers.

Examples are the compounds XPW-0702, XPW-0706, XPW-0714, XPW-0716, XPW-0720, XPW-0728, XPW-2833, XPW-2834, XPW-2947, XPW-2848 and XPW-4605.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein Y and R6As defined in formula (I), including substituted and preferred definitions, wherein Y and R6Together form a ring structure that is,

and wherein R1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Selected from the group consisting of cyclic, bicyclic and tricyclic structures, optionally with the proviso that R includes any substituent1Containing no heteroatom selected from O, S, N or containing one heteroatom selected from O, S, N,

and wherein Z1、Z2And R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R2-R5、R7-R12And X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-4):

and wherein the compounds of structure (Ib-4) -especially in the absence of additional conditions-are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the haematopoietic and haematological system (such as leukaemia and lymphoma), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0762, XPW-0770, XPW-0776, XPW-0784, XPW-0790, XPW-0798, XPW-0818, XPW-2890, XPW-2898, XPW-2904, XPW-2912, XPW-2918, XPW-2926, XPW-4576, XPW-4577, XPW-4578, XPW-4579, XPW-4580, XPW-4581, XPW-4583, XPW-4586, XPW-4589, XPW-4592 and XPW-4594.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein Z1And Z2Together is ═ O, and wherein R1Is an adamantyl group, or a substituted or unsubstituted alkyl group,

and wherein R5As defined in formula (I), including substituted and preferred definitions, with the proviso that R5In contrast to the case of the group-H,

and wherein Y, R2-R4、R6、R9-R13And X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-5):

and wherein the compounds of structure (Ib-5) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-4585, XPW-4586, XPW-4587, XPW-4591, XPW-4592 and XPW-4593.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein Z1And Z2Together is ═ O, and wherein R1Is adamantyl, and wherein X4The content of the N is N,

and wherein R2-R6、R9-R13、X1-X3And Y is as defined under (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-6):

and wherein the compounds of structure (Ib-6) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-4623, XPW-4624, XPW-4628, XPW-4629, XPW-4630, XPW-4631, XPW-4632, XPW-4634, XPW-4635, XPW-4636 and XPW-4644.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein Z1And Z2Together is ═ O, and wherein Y is-OH, and wherein R is6Is a compound of the formula-H,

and wherein R1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Comprising 6 or more carbon atoms optionally independently replaced by a heteroatom selected from O, S and N as defined in formula (I),

and wherein R5As defined in formula (I), including substituted and preferred definitions, with the proviso that R5In contrast to the case of the group-H,

and wherein R2-R4、R7-R12And X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-7):

and wherein the compounds of structure (Ib-7) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-4584, XPW-4587, XPW-4590 and XPW-4593.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein Z1And Z2Together is ═ O, and wherein Y is-OH, and wherein R is6is-H, andin (C) X4The content of the N is N,

and wherein R1-R5、R7-R12And X1-X3As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-8):

and wherein the compounds of structure (Ib-8) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-4623, XPW-4628, XPW-4630 and XPW-4636.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein Z 1And Z2Together is ═ O, and wherein Y is-OH, and wherein R is6is-H, and wherein X1And X2Each of which is N, and each of which is N,

and wherein R1-R5、R7-R12、X3And X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-9):

and wherein the compounds of structure (Ib-9) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

An example is the compound XPW-4625.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein Z1And Z2Together is ═ O, and wherein Y is-OH, and wherein R is6is-H, and wherein X1Is N, and wherein X4Is CR10And wherein R is10As defined in formula (I), including substituted and preferred definitions, with the proviso that R 10In contrast to the case of the group-H,

and wherein R1-R5、R7-R9、R11、R12、X2And X3As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-10):

and wherein the compounds of structure (Ib-10) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

An example is the compound XPW-4639.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein Z1And Z2Together is ═ O, and wherein Y is-CH3And wherein R is6is-CH3

And wherein R1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Comprising 6 or more carbon atoms optionally independently replaced by a heteroatom selected from O, S and N as defined in formula (I),

And wherein R5As defined in the general formula (I), including substituted and preferred definitions, wherein R5With the optional proviso that R is different from-H5Is different from-OCH3

And wherein R2-R4、R7-R12And X1-X4As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-11):

and wherein the compounds of structure (Ib-11) -especially in the absence of additional conditions-are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, gastric cancers, breast cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-4575, XPW-4585, XPW-4588, XPW-4591 and XPW-4595.

In another particular embodiment, the invention relates to compounds of general formula (Ib) and salts and solvates thereofIn which R is1As defined in the general formula (I), including substituted and preferred definitions, wherein R 1Selected from the group consisting of optionally substituted and substituted cycloalkyl and cycloalkenyl, wherein such cyclic rings contain 4 or more, preferably 6 or more, carbon atoms which cannot be replaced by a heteroatom selected from the group consisting of O, S and N,

and wherein R5As defined in formula (I), including substituted and preferred definitions, with the proviso that R5In contrast to the case of the group-H,

and wherein Z1、Z2And R14As defined in formula (Ib), including substituted and preferred definitions,

and wherein R2-R4、R6-R13、X1-X4And Y is as defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-12):

and wherein the compounds of structure (Ib-12) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-4575, XPW-4585, XPW-4578, XPW-4579, XPW-4580, XPW-4581, XPW-4583, XPW-4584, XPW-4588, XPW-4589, XPW-4590, XPW-4594 and XPW-4595.

In another particular embodiment, the invention relates to compounds of general formula (1b) and salts and solvates thereof, wherein Z1And Z2Together are ═ NR14

And wherein R1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Selected from cyclic, bicyclic and tricyclic structures, optionally with the proviso that R includes any substituent1Containing no heteroatom selected from O, S, N, or containing one heteroatom selected from O, S, N,

and wherein R14As defined by the general formula (Ib), including substituted and preferred definitions,

and wherein R2-R13、X1-X4And Y is as defined for formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-13):

and wherein the compounds of structure (Ib-13) -especially in the absence of additional conditions-are preferably for use in human and veterinary medicine, especially for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the haematopoietic and haematological system (such as leukaemia and lymphoma), skin cancers, oral mucosal cancers, tongue cancers and breast cancers,

Examples are the compounds XPW-0832 and XPW-4574.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein Z1And Z2Together is ═ O, and wherein Y is-OH, and wherein R is6is-H, and wherein X1Is CR11,X2Is CR8,X3Is CR9And X4Is CR10

And wherein R1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Containing 4 or more, preferably 6 or more carbon atoms,optionally independently substituted by a heteroatom such as O, S and N as defined in formula (I), provided that R includes any substituent1Containing 1 or 2 heteroatoms selected from O, S, N,

and wherein R2-R5And R7-R12As defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-14):

and wherein the compounds of structure (Ib-14) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the hematopoietic and hematological systems (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0661, XPW-0665, XPW-0667 and XPW-4613.

In another particular embodiment, the invention relates to compounds of the general formula (Ib) and salts and solvates thereof, wherein Z1And Z2Together is ═ O, and wherein R6is-CH3And wherein X1Is CR11、X2Is CR8、X3Is CR9And X4Is CR10

And wherein R1As defined in the general formula (I), including substituted and preferred definitions, wherein R1Selected from the group consisting of cyclic, bicyclic and tricyclic structures, provided that R includes any substituent1Containing 1 or 2 heteroatoms selected from O, S, N,

and wherein R2-R5、R7-R13And YAs defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ib-15):

and wherein the compounds of structure (Ib-15) are preferably for use in human and veterinary medicine, in particular for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the haematopoietic and haematological system (such as leukemias and lymphomas), skin cancers, oral mucosal cancers, tongue cancers, breast cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0539, XPW-0541 and XPW-0679.

In another particular embodiment, the invention relates to compounds of general formula (Ic) and salts and solvates thereof, wherein Z1And Z2Together form a cyclic structure containing the carbon atom to which they are bound, and wherein Z1And Z2As defined in formula (Ic), including substituted and preferred definitions,

and wherein R6As defined in formula (I), including substituted and preferred definitions, optionally with the proviso that R is6In contrast to H, the reaction mixture is,

and R is1-R5、R7-R13、X1-X4And Y is as defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ic-1):

and wherein the compounds of structure (Ic-1) -especially in the absence of additional conditions-are preferably for use in human and veterinary medicine, especially for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the haematopoietic and haematological system (such as leukaemia and lymphoma), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0902, XPW-0916, XPW-0924, XPW-0930, XPW-3038 and XPW-3052.

In another particular embodiment, the invention relates to compounds of general formula (Ic) and salts and solvates thereof, wherein Z1And Z2Together form a cyclic structure containing the carbon atom to which they are bound, and wherein Z1And Z2 is as defined in formula (Ic), including substitutions and preferred definitions, optionally with the proviso that the cyclic residue contains one or more heteroatoms independently selected from O, S and N in place of carbon atoms contained in the ring structure and/or the cyclic residue is substituted with one or more substituents defined in formula (Ic),

and R is1-R13、X1-X4And Y is as defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ic-2):

and wherein the compounds of structure (Ic-2) -especially in the absence of additional conditions-are preferably for use in human and veterinary medicine, especially for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the haematopoietic and haematological system (such as leukaemia and lymphoma), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0902, XPW-0916, XPW-0924, XPW-0930, XPW-3038 and XPW-3052.

In another particular embodiment, the invention relates to compounds of general formula (Ic) and salts and solvates thereof, wherein Z1And Z2Together form a cyclic residue comprising the carbon atom to which they are bound, and wherein Z1And Z2As defined in formula (Ic), including substitutions and preferred definitions, optionally with the proviso that the cyclic residue is a 4-membered ring,

and wherein R2-R13、X1-X4And Y is as defined in formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ic-3):

and wherein the compounds of structure (Ic-3) -especially in the absence of additional conditions-are preferably for use in human and veterinary medicine, especially for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the haematopoietic and haematological system (such as leukaemia and lymphoma), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system.

Examples are the compounds XPW-0902, XPW-0916, XPW-0924, XPW-0930, XPW-3038 and XPW-3052.

In another particular embodiment, the invention relates to compounds of formula (Ic) and salts and solvates thereof, wherein Z1And Z2Together form a cyclic residue containing the carbon atom to which they are boundAnd wherein Z is1And Z2As defined for formula (Ic), including substituted and preferred definitions,

and wherein Y and R6Each of which is-H, and,

and wherein R1As defined in formula (I), including substituted and preferred definitions, optionally with the proviso that R is1Comprising 5 or more, preferably 6 or more carbon atoms, optionally independently substituted by a heteroatom such as O, S and N as defined in formula (I),

and wherein R2-R5、R7-R12And X1-X4As defined by the general formula (I), including substituted and preferred definitions,

and wherein the compounds share the following structure (Ic-4):

and wherein the compounds of structure (Ic-4) -especially in the absence of additional conditions-are preferably for use in human and veterinary medicine, especially for medical use as described herein, preferably for immune system related applications including immunotherapy and other immunotherapy methods as defined herein, and for the treatment of immune system related disorders, skin diseases, muscle diseases, hyperproliferative disorders and cancers, including cancers of the haematopoietic and haematological system (such as leukaemia and lymphoma), skin cancers, oral mucosal cancers, tongue cancers, lung cancers, stomach cancers, breast cancers, cervical cancers, ovarian cancers and cancers of the neuroendocrine system,

Examples are the compounds XPW-0916, XPW-0924 and XPW-3052.

In some embodiments, the following compounds shown in tables 1 to 3 are specifically excluded from the scope of the present invention:

the compounds of table 1 specifically identified by CAS accession numbers have been identified by the present inventors as state of the art. In embodiments where these compounds are encompassed by general formula (I) or any subgeneric formula defined herein, they are expressly excluded from the scope of the invention in terms of compound protection. To the knowledge of the present inventors, any medical use of these compounds is not known. Accordingly, the present invention encompasses any medical use of the compounds of table 1.

The compounds of table 2 specifically identified by CAS accession numbers have been identified by the present inventors as state of the art. In embodiments, when these compounds are encompassed by general formula (I) or any subgeneric formula defined herein, they are specifically excluded from the scope of the invention with respect to compound protection. To the best of the inventors' knowledge, these compounds are not recognized for any of the medical uses defined herein. Thus, for the medical uses defined herein, particularly in the treatment of non-malignant or malignant hyperproliferative diseases, the compounds of table 2 are expressly included within the scope of the present invention.

The compounds of table 3 specifically identified by CAS accession numbers have been identified by the present inventors as state of the art. In embodiments, when these compounds are encompassed by general formula (I) or any subgeneric formula defined herein, they are specifically excluded from the scope of the invention with respect to compound protection. In addition, to the best of the inventors' knowledge, these compounds are known for medical use, which in some embodiments may be encompassed by the medical uses defined herein. Thus, in some embodiments defined herein, the compounds of table 3 may be specifically excluded from the scope of the present invention with respect to compound protection and certain medical uses.

Specific examples of compounds falling within the scope of the compounds contained in pending application PCT/EP2018/054686 have been identified in the present application for new medical uses, in particular muscle cells, cells and malignant cells having growth inhibitory properties on keratinocytes and cells selected from cervical cancer, cutaneous T-cell lymphoma, acute promyelocytic leukemia, acute myeloid leukemia, oral and tongue squamous cell carcinoma, epidermoid squamous cell carcinoma and lung squamous cell carcinoma cells.

Thus, these compounds and their salts and solvates are particularly useful in the treatment of hyperproliferative muscle disorders, hyperproliferative skin diseases, as defined herein, and in the treatment of cervical cancer, cutaneous T-cell lymphoma, acute promyelocytic leukemia, acute myeloid leukemia, epidermoid skin cancers such as non-melanoma skin cancers, oral cancers, tongue cancers and lung cancers, as defined herein.

Specific examples of compounds falling within the scope of the compounds contained in pending application PCT/EP2018/054686 have been identified in the present application for further novel medical uses, in particular growth inhibitory properties on cancers selected from T-cell leukemia, B-cell leukemia, gastric cancer, breast cancer and medullary thyroid cancer.

Thus, these compounds and their salts and solvates are particularly useful in the treatment of hematopoietic disorders, including hematopoietic disorders such as T-cell leukemia, B-cell leukemia, and gastric, breast, ovarian and neuroendocrine system cancers as defined herein.

Table 4 and table 5 show the herein identified novel medical uses of specific compounds falling within the scope of the compounds contained in the pending application PCT/EP2018/054686, wherein the medical uses are selected from the group of treatments of hyperproliferative muscle diseases (a), hyperproliferative skin diseases (B), cervical cancer (C), cutaneous T-cell lymphoma (D), acute promyelocytic leukemia (E), acute myeloid leukemia (F), epidermoid skin cancer (G), oral cancer (H), tongue cancer (I), lung cancer (J), T-cell leukemia (K), B-cell leukemia (L), gastric cancer (M), breast cancer (N), ovarian cancer (O) and neuroendocrine system cancer (P) as defined herein.

The following compounds described in PCT/EP2018/054686 are specifically claimed for specified medical uses.

Table 4:

the following compounds described in PCT/EP2018/054686 are specifically claimed for specified medical uses.

Table 5:

specific examples of the compounds falling within the range of formula (I) are shown in tables 6 to 54. The intermediate is denoted "XPW-I".

Other definitions:

the term "C1-C12Alkyl "includes all isomers of the corresponding saturated aliphatic hydrocarbon groups containing 1 to 12 carbon atoms; this includes methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, sec-pentyl, 3-pentyl, 2-methylbutyl, isopentyl, 2-methylbut-2-yl, 3-methylbut-2-yl, all hexyl isomers, all heptyl isomers, all octyl isomers, all nonyl isomers, all decyl isomers, all undecyl isomers and all dodecyl isomers.

The term "C2-C12Alkenyl "includes all isomers of the corresponding unsaturated olefinic hydrocarbon groups containing 2 to 12 carbon atoms connected by (i.e., containing) one or more double bonds; this includes ethenyl, all propenyl isomers, all butenyl isomers, all pentenyl isomers, all hexenyl isomers, all heptenyl isomers, all octenyl isomers, all nonenyl isomers, all decenyl isomers, all undecenyl isomers, and all dodecenyl isomers.

The term "C2-C12Alkynyl "includes all isomers of the corresponding unsaturated acetylenic hydrocarbyl radicals containing 2 to 12 carbon atoms joined by (i.e., containing) one or more triple bonds; this includes ethynyl, all propynyl isomers, all butynyl isomers, all pentynyl isomers, all hexynyl isomers, all heptynylThe isomers include, but are not limited to, the base isomer, all octynyl isomers, all nonynyl isomers, all decynyl isomers, all undecylynyl isomers, and all dodecylynyl isomers. The term "alkynyl" also includes compounds having one or more triple bonds and one or more double bonds.

The term "C3-C8Cycloalkyl "includes the corresponding saturated hydrocarbon groups containing 3 to 8 carbon atoms arranged in a monocyclic ring structure; this includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl.

The term "C5-C8Cycloalkenyl includes the corresponding unsaturated nonaromatic and nonaromatic hydrocarbon radicals having 5 to 8 carbon atoms in which at least one is sp3Hybridized, and they are arranged in a single ring structure and are linked by (i.e., contain) one or more double bonds; this includes all cyclopentenyl isomers, all cyclohexenyl isomers, all cycloheptenyl isomers, all cyclooctenyl isomers.

The term "C5-C12Bicycloalkyl "includes the corresponding saturated hydrocarbon groups of 5 to 12 carbon atoms arranged in a bicyclic structure; wherein these bicyclic structures include fused, bridged and spiro ring systems;

the term "C7-C12Bicycloalkenyl "includes the corresponding unsaturated non-aromatic and non-heteroaromatic hydrocarbon radicals containing from 7 to 12 carbon atoms arranged in a bicyclic structure and connected by, i.e., containing) one or more double bonds; wherein these bicyclic structures include fused, bridged and spiro ring systems;

the term "C8-C14Tricycloalkyl "includes the corresponding saturated hydrocarbon groups containing 8 to 14 carbon atoms arranged in a tricyclic structure; wherein these tricyclic structures include fused, bridged and spiro ring systems;

for R1By way of illustration, the terms "cyclic", "bicyclic", "tricyclic", "cycloalkyl", "cycloalkenyl", "bicycloalkyl", "bicycloalkenyl" and "tricycloalkyl" mean that such cyclic, bicyclic or tricyclic residues are directly connected to R by a chemical bond1On the aromatic ring to which it is bonded; and R is1Of the substituentsThe terms "cyclic", "bicyclic", "tricyclic", "cycloalkyl", "cycloalkenyl", "bicycloalkyl", "bicycloalkenyl" and "tricycloalkyl" mean that such cyclic, bicyclic or tricyclic residues are directly connected to R by a chemical bond 1On one of the C atoms or N atoms or O atoms or S atoms contained; for example, "R1Is cyclohexyl "means a cyclohexyl residue and R1The bonded aromatic rings are linked; "R1Is methyl, and R1By cyclohexyl "is meant the resulting-CH2(cyclohexyl) residue with R1The attached aromatic rings are linked.

If a carbon atom is substituted by a heteroatom selected from O, N or S, the number of substituents on the corresponding heteroatom is adjusted according to its valency, e.g. -CR2The-group may be substituted by-NR-, -NR-2 +-, -O-or-S-groups.

The term "perhalogenated" relates to the complete halogenation of a carbon scaffold; the corresponding residues include the corresponding perfluorinated, perchlorinated, perbrominated and perchlorinated groups. Preferably, the term "perhalogenated" relates to perfluorinated or perchlorinated groups, more preferably perfluorinated groups.

The following includes definitions of terms used in this specification. Unless otherwise indicated, the initial definitions provided herein for a group or term apply to the group or term throughout the specification, individually or as part of another group.

It is also within the scope of the present invention that the compounds of the present invention may form salts. Unless otherwise indicated, reference herein to a compound of the invention is to be understood as including reference to a salt thereof. As used herein, the term "salt(s)" means an acidic and/or basic salt formed from an inorganic and/or organic acid and a base. Zwitterions (internal salts) or internal salts) are included in the term "salt(s)" as used herein (and may be formed, for example, where the substituent comprises an acid moiety, such as a carboxyl group, and an amino group). Also included herein are quaternary ammonium salts, such as alkyl ammonium salts. A salt of a compound can be formed, for example, by reacting the compound with an amount of an acid or base, such as an equivalent amount of an acid or base, in a medium such as one in which the salt precipitates or in an aqueous medium, followed by lyophilization.

Exemplary salts derived from the addition of acid include acetates (such as those formed from acetic acid or trihaloacetic acid, e.g., trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, hydroiodides, chlorates, bromates, iodates, 2-hydroxyethanesulfonates, lactates, maleates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, oxalates, pectinates, persulfates, 3-phenylpropionates, phosphates, picrates, nitrates, 2-hydroxyethanesulfonates, lactates, maleates, methanesulfonates, nicotinates, nitrates, oxalates, pectinates, persulfates, 3-phenylpropionates, phosphates, picrates, Pivalate, propionate, salicylate, succinate, sulfate (such as those formed with sulfuric acid), sulfonate (such as those mentioned herein), tartrate, thiocyanate, tosylate such as tosylate, undecanoate, and the like.

Exemplary salts resulting from addition of a base (e.g., formed where the substituent includes an acidic moiety such as a carboxyl group) include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (e.g., organic amines) such as benzathine, dicyclohexylamine, hydrazylamine (hydrabamine), N-methyl-D-glucamine, N-methyl-D-glucamide, t-butylamine, and salts with amino acids such as arginine, lysine, and the like. Basic nitrogen-containing groups can be quaternized with agents such as lower alkyl halides (e.g., methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides), dialkyl sulfates (e.g., dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (e.g., decyl, dodecyl, tetradecyl, and octadecyl chlorides, bromides, and iodides), aralkyl halides (e.g., benzyl and phenethyl bromides), and the like.

The invention also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein the parent compound is modified by conversion of an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; basic or organic salts of acidic residues such as carboxylic acids; and so on. Pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound, for example, salts formed from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, which contains a basic or acidic moiety, by conventional chemical methods. In general, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile are preferred. A list of suitable salts can be found in Remington's Pharmaceutical Sciences, 17 th edition, Mack Publishing Company, Easton, Pa., 1985, page 1418, and Journal of Pharmaceutical Science 1977, 66(2), each of which is incorporated herein by reference in its entirety.

The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

Furthermore, in the case where the compounds of the present invention contain asymmetric carbon atoms or atropisomeric bonds, the present invention relates to mixtures of forms D, L and D, L, and to diastereomeric forms when more than one asymmetric carbon atom or atropisomeric bond is present. The compounds of the invention which contain asymmetric carbon atoms or atropisomeric bonds and are usually obtained in the form of racemates can be separated into the optically active isomers in a known manner, for example using optically active acids. However, it is also possible to use the optically active starting materials from the outset and then to obtain the corresponding optically active or diastereomeric compounds as end products.

The compounds of the present invention also include tautomeric forms. The tautomeric forms result from the exchange of a single bond with an adjacent double bond and the concomitant migration of protons. Tautomeric forms include proton tautomers (prototropic tautomers), which are isomeric protonation states having the same empirical formula (empirical formula) and total charge. Examples of proton tautomers include keto-enol pairs, amide-imidic acid pairs, lactam-lactim pairs, amide-imidic acid pairs, enamine-imine pairs and cyclic rings, wherein protons may occupy two or more positions of the heterocyclic ring system, such as 1H-and 3H-imidazole, 1H-, 2H-and 4H-1, 2, 4-triazole, 1H-and 2H-isoindoles and 1H-and 2H-pyrazoles. Tautomeric forms can be in equilibrium or can be sterically locked into one form by appropriate substitution.

The compounds described herein can be asymmetric (e.g., have one or more stereogenic centers). Unless otherwise indicated, all stereoisomers, such as enantiomers and diastereomers, are intended to be included. The compounds of the present invention containing an asymmetrically substituted carbon atom may be isolated in optically active or racemic forms. Methods of how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C ═ N double bonds, and the like, may also be present in the compounds described herein, and all such stable isomers are contemplated within the scope of the present invention. Cis and trans geometric isomers of the compounds of the invention are described and may be separated as mixtures of isomers or as isolated isomeric forms.

The compounds of the invention may also include all of the atomic isotopes that occur in the intermediates or final compounds. Isotopes include atoms of the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium.

Solvates and hydrates of the compounds of the invention and of their pharmaceutically acceptable salts are also included.

The term "compound" as used herein, unless otherwise indicated, is intended to include all stereoisomers, geometric isomers, tautomers, rotational isomers and isotopes of the structure recited.

In some embodiments, the compound may be provided in a prodrug form. As used herein, the term "prodrug" refers to a compound that undergoes chemical conversion by metabolic or chemical processes, resulting in a compound of the present invention, or a salt and/or solvate thereof, upon administration to a subject.

In some embodiments, the compounds of the present invention and salts thereof are substantially isolated. By "substantially isolated" is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected. Partial separation may include, for example, enrichment of a composition with a compound of the invention. Substantial separation may include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of a compound of the invention or a salt thereof.

Pharmaceutical process

It has been found that the compounds according to the invention have pharmacologically important properties and can be used in therapy. The compounds of the present invention may be used alone, in combination with each other or in combination with other active compounds.

In certain embodiments, the compounds of the present invention may exhibit growth inhibitory properties during hyperproliferative processes.

The antiproliferative activity of the compounds belonging to formulae (Ia), (Ib) and (Ic), respectively, was studied on cells or cell lines derived from the hematopoietic system, including myeloid and lymphoid cells (T and B cells), neuroendocrine system, cervix, breast, ovary, lung, gastrointestinal tract and mucosal epithelium, as well as from the cutaneous epithelium and from the muscle. For this purpose, HL-60 cells, NB-4 cells, HH cells, RPMI-8402 cells, TANOIUE cells, TT cells, HeLa cells, MDA-MB-231 cells, FU-OV-1 cells, LOU-NH91 cells, 23132/87 cells, CAL-27 cells, BHY cells, SCC-25 cellsA-431 cells, human-representative keratinocytes (HPEK) and C2C12 cells were seeded in 96-well plates (CORNING #3598) suitable for fluorescence assay at the following initial cell numbers: HL-60 1000 cells per well; NB-4 1000 cells per well; HH 5000 cells per well; RPMI-8402 had 5000 cells per well; TANOUUE 1500 cells per well; TT 9000 cells per well; HeLa 2000 cells per well; 3000 cells per well MDA-MB-231; FU-OV-1 per hole 3000 cells; LOU-NH91 4000 cells per well; 23132/87 cells per well; CAL-27 cells/well 2000; BHY 1500 cells per well; SCC-25 1500 cells per well; a-431 cells per well; HPEK 1000 cells per well; C2C12 500 cells per well. Cells were diluted to 0.1% v/v in H with the indicated final concentration (from 1000-fold stock solution in DMSO to 2Compound of O (final DMSO concentration in water for injection, WFI, fisher scientific #10378939) for 5 days, or 0.1% v/v of the empty vehicle DMSO as control for 5 days. On day 5 after initiation of treatment, cells were subjected to the manufacturer's protocolProliferation assay (Bio-Rad Serotec GmbH, BUF 012B). The readout was performed in fluorescence mode using a multiwell plate reader using filters (excitation at 560nm (bandwidth 10nm), excitation at 590nm (bandwidth 10 nm)). Control treatments for growth inhibition with commercial compounds such as Methotrexate (MTREX) and Resveratrol (RES) were included on each plate. Some test compounds of the invention were obtained and used in the form of their salts. The column "Specification" in tables 55 to 92 and the general chemical formula thereof in table 93 show the corresponding cases.

The assays were performed in independent single experiments repeated in duplicate or more, each experiment being repeated 6 times for each condition. For each individual plate, the measured fluorescence intensity values under compound-treated conditions were normalized to the corresponding equally weighted arithmetic mean (equivalent weighted arithmetric mean) of the fluorescence intensity values of the six DMSO-treated control wells to obtain a relative value for the baseline level of 1.0.

Two independent outlier analyses were performed according to the method of Peirce and Chauvenet (Ross, Journal of Engineering Technology 2003, 1-12). Outliers identified by at least one method were excluded from the calculations, but in a single experiment, no more than 1 out of 6 values for each compound. The weighted arithmetic mean (abbreviated here as AVE) for each compound was calculated from the normalized values of all independent replicates of a single experiment repeated 6 times eachw). The respective standard deviations of the weighted arithmetic mean are calculated according to the method described by Bronstein et al (Bronstein, Semendjajew, Musiol, Muhlig, Taschenbuch der Mathematik, 5 th edition 2001(Gerrnan), pubisher: Verlag Harri Deutsch, Frankfurt am Main and Thun) and combined with a Gaussian error propagation (Gau β' error propagation) which is associated with the normalization calculation performed. The resulting standard deviation is referred to herein as the "combined standard deviation".

In the case of a considerable variation of the normalized equal weighted arithmetic mean derived from two independent repetitions, the number of independent repetitions is increased to three or more. In the case of four or more independent replicates, a two-line outlier analysis was performed on all normalized equal-weighted recalculation averages according to the method of Peirce and Chauvenet described above.

In certain embodiments, the compounds of the present invention may be growth inhibitors in hyperproliferative processes, including malignant and non-malignant hyperproliferative processes.

In one embodiment, several compounds of the present invention were found to inhibit the growth of HL-60 cells (human acute myeloid leukemia cells), which are available from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ), under the accession number ACC 3. At 37 ℃ and 5% CO2HL-60 cells were cultured in RPMI 1640 medium (Fisherschelentipic, #11554526) containing 10% fetal bovine serum (Fisherschelentipic, # 15517589).

If the normalized fluorescence intensity values after addition of the respective combined standard deviations are weighted arithmetic mean relative to an overall base level of 1.0 at a reference concentration of 20. mu.MValues that add up to or below 0.9, in particular that add up to or below 0.8, that add up to or below 0.7, that add up to or below 0.6, that add up to or below 0.4, that add up to or below 0.2 are considered inhibitors of the growth of HL-60 cells. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10 -2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of HL-60 cells according to the above method. The HL-60 growth inhibitors identified to date relate to the compounds listed in tables 55 and 56. The entries of tables 55 and 56 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 55: proliferation assay using HL-60 cells at 20. mu.M

Table 56: at 20. mu.M with HL-60 cellsProliferation assay of

The data in table 55 relate to novel compounds, wherein the data in table 56 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the present invention were found to inhibit the growth of NB-4 cells (human acute promyelocytic leukemia cells) available from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 207. At 37 ℃ and 5% CO2NB-4 cells were cultured in RPMI 1640 medium (Fisherschelentific, #11554526) containing 10% fetal bovine serum (Fisherschelentific, # 15517589).

A compound is considered to be an inhibitor of the growth of NB-4 cells if the weighted arithmetic means of the normalized fluorescence intensity values after addition of the respective combined standard deviations add up to or below 0.9, in particular add up to or below 0.8, add up to or below 0.7, add up to or below 0.6, add up to or below 0.4, add up to or below 0.2 relative to an overall basal level of 1.0 at a reference concentration of 20. mu.M. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10-2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of NB-4 cells according to the above method. The NB-4 growth inhibitors identified to date relate to the compounds listed in tables 57 and 58. The entries of tables 57 and 58 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 57: proliferation assay with NB-4 cells at 20. mu.M

Table 58: proliferation assay with NB-4 cells at 20. mu.M

The data in table 57 relate to novel compounds, wherein the data in table 58 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the invention were found to inhibit the growth of HH cells (human skin T-cell lymphoma cells) available from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 707. At 37 ℃ and 5% CO2HH cells were cultured in RPMI 1640 medium (Fisherschelentipic, #11554526) containing 10% fetal bovine serum (Fisherschelentipic, # 15517589).

A compound is considered to be a growth inhibitor for HH cells if the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the respective combined standard deviations, relative to an overall base level of 1.0, at a reference concentration of 20 μ M, add up to or below 0.9, in particular add up to or below 0.8, add up to or below 0.7, add up to or below 0.6, add up to or below 0.4, add up to or below 0.2. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10 -2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors for HH cells according to the above method. The HH growth inhibitors identified to date relate to the compounds listed in table 59 and table 60. The entries of tables 59 and 60 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 59: proliferation assay with HH cells at 20 μ M

Table 60: proliferation assay with HH cells at 20 μ M

The data in table 59 relate to novel compounds, wherein the data in table 60 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the present invention were found to inhibit the growth of RPMI-8402 cells (human T-cell acute lymphoblastic leukemia cells) available from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under accession number ACC 290. At 37 ℃ and 5% CO2RPMI-8402 cells were cultured in RPMI 1640 medium (Fisherschelentipic, #11554526) containing 10% fetal bovine serum (Fisherschelentipic, # 15517589).

A compound is considered to be an inhibitor of the growth of RPMI-8402 cells if the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the respective combined standard deviations adds up to or below 0.9, in particular to or below 0.8, to or below 0.7, to or below 0.6, to or below 0.4, to or below 0.2 relative to an overall base level of 1.0 at a reference concentration of 20 μ M. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10-2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of RPMI-8402 cells according to the above method. The RPMI-8402 growth inhibitors identified to date relate to the compounds listed in tables 61 and 62. The entries of tables 61 and 62 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 61: proliferation assay using RPMI-8402 cells at 20 μ M

Table 62: proliferation assay using RPMI-8402 cells at 20 μ M

The data in table 61 relate to novel compounds, wherein the data in table 62 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the invention were found to inhibit the growth of TANOUE cells (human B-cell leukemia cells) obtainable from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 399. At 37 ℃ and 5% CO2TANOIUE cells were cultured in RPMI 1640 medium (Fisherschelientific, #11554526) containing 10% fetal bovine serum (Fisherschelientific, # 15517589).

A compound is considered to be an inhibitor of the growth of tanoee cells if the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the respective combined standard deviations adds up to or below 0.9, in particular to or below 0.8, to or below 0.7, to or below 0.6, to or below 0.4, to or below 0.2 relative to an overall basal level of 1.0 at a reference concentration of 20 μ M. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10 -2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of TANOUE cells according to the above method. The RPMI-8402 growth inhibitors identified to date relate to the compounds listed in tables 63 and 64. The entries of tables 63 and 64 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 63: proliferation assay using TANOIUE cells at 20. mu.M

Table 64: proliferation assay using TANOIUE cells at 20. mu.M

The data in table 63 relate to novel compounds, wherein the data in table 64 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the present invention were found to inhibit the growth of TT cells (human medullary thyroid carcinoma cells) available from the American Type Culture Collection (ATCC) under the accession number ATCC-CRL-1803. At 37 ℃ and 5% CO2TT cells were cultured in F-12K medium (Fisherschelientific, #11580556, or ATCC, # ATCC-30-2004) containing 10% fetal bovine serum (Fisherschelientific, # 15517589).

A compound is considered to be a growth inhibitor of TT cells if the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the respective combined standard deviations, at a reference concentration of 20 μ M, is added up to or below 0.9, in particular to or below 0.8, to or below 0.7, to or below 0.6, to or below 0.4, to or below 0.2, relative to an overall base level of 1.0. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10-2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of TT cells according to the above method. The TT growth inhibitors identified to date involve the compounds listed in tables 65 and 66. The entries of tables 65 and 66 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 65: proliferation assay with TT cells at 20 μ M

Table 66: proliferation assay with TT cells at 20 μ M

The data in table 65 relate to novel compounds, wherein the data in table 66 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the present invention were found to inhibit the growth of HeLa cells (human cervical adenocarcinoma cells) available from the American Type Culture Collection (ATCC) under the accession number ATCC-CCL-2. At 37 ℃ and 5% CO2HeLa cells were cultured in a DMEM medium (Fisherscifieic, #11584456) containing 10% fetal bovine serum (Fisherscifieic, # 15517589).

If the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the respective combined standard deviations is at or below 0.9, in particular at or below 0.8, at or below 0.7, at or below 0.6, at or below 0.4, at or below 0.0, relative to the overall base level of 1.0, at a reference concentration of 20. mu.M, the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the respective combined standard deviations is added up to or below 0.9, in particular up to or below 0.8, up to or below 0.7, up to orUp to or below 0.2, the compound is considered to be a growth inhibitor for HeLa cells. Similar to the calculations performed for the test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10 -2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of HeLa cells according to the above method. The HeLa growth inhibitors identified to date relate to the compounds listed in table 67 and table 68. The entries of tables 67 and 68 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 67: proliferation assay with HeLa cells at 20. mu.M

Table 68: proliferation assay with HeLa cells at 20. mu.M

The data in table 67 relate to novel compounds, wherein the data in table 68 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

Growth of MDA-MB-231 cells (human breast cancer cells) from Deutsche Sammlung von Mikroorganismen und Zellkulturen Gmbh (dsmz), accession number ACC 732. At 37 ℃ and 5% CO2MDA-MB-231 cells were cultured in Leibovitz's L-15 (phenol red free) medium (Fisherschelentipic, #11540556) containing 10% fetal bovine serum (Fisherschelentipic, # 15517589).

A compound is considered to be an inhibitor of the growth of MDA-MB-231 cells if the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the respective combined standard deviations adds up to or below 0.9, in particular to or below 0.8, to or below 0.7, to or below 0.6, to or below 0.4, to or below 0.2 relative to the overall basal level of 1.0 at a reference concentration of 20. mu.M. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10 -2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of MDA-MB-231 cells according to the above method. The MDA-MB-231 growth inhibitors identified to date involve the compounds listed in tables 69 and 70. The entries of tables 69 and 70 are sorted by the corresponding weighted arithmetic mean of the compounds without regard to the respective standard deviation and thus fall within the activity ranges shown.

Table 69: proliferation assay with MDA-MB-231 cells at 20. mu.M

Table 70: proliferation assay with MDA-MB-231 cells at 20. mu.M

The data in table 69 relate to novel compounds, wherein the data in table 70 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the present invention were found to inhibit the growth of FU-OV-1 cells (human ovarian cancer cells) obtainable from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 444. At 37 ℃ and 5% CO2The cells were cultured in Ham's F-12/DMEM (1: 1) medium (Fisher) containing 10% fetal bovine serum (Fisher scientific, #15517589) and 1mM sodium pyruvate (Fisher scientific, #11501871) scientific, #11514436) were cultured in FU-OV-1 cells.

A compound is considered to be an inhibitor of the growth of FU-OV-1 cells if the weighted arithmetic means of the normalized fluorescence intensity values after addition of the respective combined standard deviations add up to or below 0.9, in particular add up to or below 0.8, add up to or below 0.7, add up to or below 0.6, add up to or below 0.4, add up to or below 0.2, relative to an overall base level of 1.0 at a reference concentration of 20 μ M. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10-2

Several molecules falling within the scope of the compounds defined in formulae (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of FU-OV-1 cells according to the above method. The FU-OV-1 growth inhibitors identified thus far relate to the compounds listed in tables 71 and 72. The entries of tables 71 and 72 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 71: proliferation assay with FU-OV-1 cells at 20 μ M

Table 72: proliferation assay with FU-OV-1 cells at 20 μ M

The data in table 71 relate to novel compounds, wherein the data in table 72 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the present invention were found to inhibit the growth of LOU-NH91 cells (human lung squamous cell carcinoma cells) available from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 393. At 37 ℃ and 5% CO2LOU-NH91 cells were cultured in RPMI 1640 medium (Fisherschelentipic, #11554526) containing 10% fetal bovine serum (Fisherschelentipic, # 15517589).

A compound is considered to be an inhibitor of growth of LOU-NH91 cells if the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the respective combined standard deviations adds up to or below 0.9, in particular to or below 0.8, to or below 0.7, to or below 0.6, to or below 0.4, to or below 0.2 relative to an overall base level of 1.0 at a reference concentration of 20 μ M. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10 -2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of LOU-NH91 cells according to the above method. The LOU-NH91 growth inhibitors identified to date relate to the compounds listed in tables 73 and 74. The entries of tables 73 and 74 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 731: at 20 μ M profitProliferation assay with LOU-NH91 cells

Table 742: proliferation assay at 20 μ M using LOU-NH91 cells

The data in table 73 relate to novel compounds, wherein the data in table 74 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the present invention were found to inhibit the growth of 23132/87 cells (human gastric adenocarcinoma cells) available from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 201. At 37 ℃ and 5% CO223132/87 cells were cultured in RPMI 1640 medium (Fisherschelentipic, #11554526) containing 10% fetal bovine serum (Fisherschelentipic, # 15517589).

If at the reference concentration of 20. mu.M, the corresponding combinations are combined with respect to the overall basal level of 1.0The weighted arithmetic means of the normalized fluorescence intensity values after addition of the standard deviations add up to or below 0.9, in particular add up to or below 0.8, add up to or below 0.7, add up to or below 0.6, add up to or below 0.4, add up to or below 0.2, then the compound is considered to be an 23132/87 cell growth inhibitor. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10-2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of 23132/87 cells according to the above method. The 23132/87 growth inhibitors identified to date relate to the compounds listed in tables 75 and 76. The entries of tables 75 and 76 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 75: proliferation assay using 23132/87 cells at 20 μ M

Table 76: proliferation assay using 23132/87 cells at 20 μ M

The data in table 75 relate to novel compounds, wherein the data in table 76 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the present invention were found to inhibit the growth of CAL-27 cells (human squamous cell carcinoma of tongue cells) available from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 446. At 37 ℃ and 5% CO2CAL-27 cells were cultured in a DMEM medium (Fisherschelientific, #11584456) containing 10% fetal bovine serum (Fisherschelientific, # 15517589).

A compound is considered to be a growth inhibitor of CAL-27 cells if the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the respective combined standard deviations, at a reference concentration of 20 μ M, is added up to or below 0.9, in particular to or below 0.8, to or below 0.7, to or below 0.6, to or below 0.4, to or below 0.2, relative to a global base level of 1.0. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10 -2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of CAL-27 cells according to the above method. The CAL-27 growth inhibitors identified to date relate to the compounds listed in tables 77 and 78. The entries of tables 77 and 78 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 773: in thatProliferation assay at 20 μ M using CAL-27 cells

Table 784: proliferation assay using CAL-27 cells at 20. mu.M

The data in table 77 relate to novel compounds, wherein the data in table 78 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the invention were found to inhibit the growth of BHY cells (human oral squamous cell carcinoma cells) available from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession number ACC 404. At 37 ℃ and 5% CO2BHY cells were cultured in a DMEM medium (Fisherschelientific, #11584456) containing 10% fetal bovine serum (Fisherschelientific, # 15517589).

If the reference concentration is 20. mu.M, the corresponding is compared with the overall basal level of 1.0The weighted arithmetic means of the normalized fluorescence intensity values after addition of the standard deviations of the combinations add up to or below 0.9, in particular add up to or below 0.8, add up to or below 0.7, add up to or below 0.6, add up to or below 0.4, add up to or below 0.2, the compound is considered as a growth inhibitor for BHY cells. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10-2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of BHY cells according to the above method. The BHY growth inhibitors identified so far relate to the compounds listed in table 79, table 80 and table 81. The entries of table 79, table 80 and table 81 are sorted by the corresponding weighted arithmetic mean of the compounds without regard to the respective standard deviation and thus fall within the activity ranges shown.

TABLE 795: proliferation assay using BHY cells at 20 μ M

Table 806: proliferation assay using BHY cells at 20 μ M

Table 817: proliferation assay using BHY cells at 20 μ M

The data in table 79 relate to novel compounds, wherein the data in table 80 and table 81 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the present invention were found to inhibit the growth of SCC-25 cells (human squamous cell carcinoma of the tongue) available from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under accession number ACC 617. At 37 ℃ and 5% CO2SCC-25 cells were cultured in Ham's F-12/DMEM (1: 1) medium (Fisherschelentipic, #11514436) containing 10% fetal bovine serum (Fisherschelentipic, #15517589) and 1mM sodium pyruvate (Fisherschelentipic, # 11501871).

If the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the respective combined standard deviations is at or below 0.9, in particular at or below 0.8, at or below 0.7, at or below 0.6, at or below 0.4, at or below 0.0, at a reference concentration of 20. mu.M, relative to an overall base level of 1.0 Or below 0.2, the compound is considered to be a growth inhibitor of SCC-25 cells. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10-2

According to the above method, several molecules falling within the scope of the compounds defined in formulae (Ia) (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of SCC-25 cells. The SCC-25 growth inhibitors identified to date relate to the compounds listed in tables 82 and 83. The entries of tables 82 and 83 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 828: proliferation assay with SCC-25 cells at 20 μ M

Table 839: proliferation assay with SCC-25 cells at 20 μ M

The data in table 82 relate to novel compounds, wherein the data in table 83 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the present invention were found to inhibit the growth of a-431 cells (human epidermoid squamous Cell carcinoma cells), available from Cell Lines Service GmbH (CLS), under accession number 300112. At 37 ℃ and 5% CO 2A-431 cells were cultured in a DMEM medium (Fisherschelientific, #11584456) containing 10% fetal bovine serum (Fisherschelientific, # 15517589).

A compound is considered to be an inhibitor of the growth of a-431 cells if the weighted arithmetic mean of the normalized fluorescence intensity values after addition of the respective combined standard deviations, at a reference concentration of 20 μ M, is added up to or below 0.9, in particular to or below 0.8, to or below 0.7, to or below 0.6, to or below 0.4, to or below 0.2, relative to an overall base level of 1.0. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10-2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of a-431 cells according to the above method. The A-431 growth inhibitors identified to date relate to the compounds listed in tables 84 and 85. The entries of tables 84 and 85 are sorted by the corresponding weighted arithmetic mean of the compounds, regardless of the respective standard deviation, and thus fall within the activity ranges shown.

Table 84: proliferation assay using A-431 cells at 20. mu.M

Table 85: proliferation assay using A-431 cells at 20. mu.M

The data in table 84 relate to novel compounds, wherein the data in table 85 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the invention were found to inhibit the growth of human epidermal keratinocyte progenitor cells (HPEKp, pooled), available from cellnet Advanced Cell Systems AG under the accession number HPEKp. At 37 ℃ and 5% CO2Human epithelial cells were cultured in CnT-Prime epithelial medium (cellnet tec, # CnT-PR, fully defined low calcium preparation completely free of animal or human derived components) without addition of additional components.

Normalized fluorescence intensity relative to an overall basal level of 1.0 if the corresponding combined standard deviations are added at a reference concentration of 10. mu.MA weighted arithmetic mean of the values equal to or lower than 0.9, in particular equal to or lower than 0.8, equal to or lower than 0.7, equal to or lower than 0.6, equal to or lower than 0.4, equal to or lower than 0.2, is then considered as an inhibitor of the growth of HPEKp cells. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10 -2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of HPEKp cells according to the above method. The HPEKp growth inhibitors identified to date relate to the compounds listed in table 86, table 87 and table 88. The entries of tables 86, 87 and 88 are sorted by the corresponding weighted arithmetic mean of the compounds without regard to the respective standard deviation and thus fall within the activity ranges shown.

Table 86: proliferation assay using HPEKp cells at 10. mu.M

Table 87: proliferation assay using HPEKD cells at 10. mu.M

Table 8810: proliferation assay using HPEKp cells at 10. mu.M

The data in table 86 relate to novel compounds, wherein the data in table 87 and table 88 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In one embodiment, several compounds of the invention were found to inhibit the growth of C2C12 cells (murine myoblasts), which C2C12 cells are available from Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ) under accession No. ACC 565. At 37 ℃ and 5% CO2The cells were cultured in RPMI 1640 medium (Fisherschelientific, #15517589) containing 10% fetal bovine serum (Fisherschelientific, #15517589) 11554526) in culture with C2C12 cells.

A compound is considered to be a growth inhibitor of C2C12 cells if the weighted arithmetic means of the normalized fluorescence intensity values after addition of the respective combined standard deviations add up to or below 0.9, in particular add up to or below 0.8, add up to or below 0.7, add up to or below 0.6, add up to or below 0.4, add up to or below 0.2 relative to the overall base level of 1.0 at a reference concentration of 20 μ M. Similar to the calculations performed for test compounds, the total basal level was calculated as the weighted arithmetic mean of all normalized values from DMSO control measurements. The corresponding combined standard deviation of DMSO values is less than 1.10-2

Several molecules falling within the scope of the compounds defined in formulae (Ia), (Ib) and (Ic) herein, respectively, have been identified as growth inhibitors of C2C12 cells according to the above method. The C2C12 growth inhibitors identified to date relate to the compounds listed in table 89, table 90 and table 91. The entries of table 89, table 90 and table 91 are sorted by the corresponding weighted arithmetic mean of the compounds without regard to the respective standard deviation and thus fall within the activity ranges shown.

Table 89: proliferation assay using C2C12 cells at 20. mu.M

Table 90: proliferation assay using C2C12 cells at 20. mu.M

Table 9111: proliferation assay using C2C12 cells at 20. mu.M

The data in table 89 relate to novel compounds, wherein the data in table 90 and table 91 relate to novel medical uses of the compounds disclosed in PCT/EP 2018/054686.

In certain embodiments, the compounds of the present invention may be modulators, in particular enhancers of Notch signaling.

Intercellular communication by Notch signaling (reviewed in Kopan et al, Cell 2009, 137, 216-: notch receptors distributed on the cell membrane of the signaling cells and Notch ligands covering the cell membrane of the signaling cells. Mechanistically, Notch signaling is activated by receptor-ligand interactions, which result in the intracellular domain (NICD) protein-water interpretation of membrane-bound Notch receptors being placed inside signal-receiving cells. NICD is subsequently transferred into the nucleus, which in turn leads to transcriptional activation of certain cell-type specific genes. Notch-mediated alteration of the cellular previous gene expression program is manifested by corresponding cellular changes that represent the response of the cell to Notch signaling.

By measuring the expression level of Notch-specific target genes, the level of activation of Notch signaling can be reliably quantified in vitro. This can be achieved by quantitative detection of the corresponding mRNA or protein of a particular Notch target gene. Alternatively, the cells may be genetically modified to carry a luciferase gene as an artificial Notch target gene, the expression of which is dependent on Notch activity. In this case, the level of Notch signaling can be quantified by measuring luciferase-derived bioluminescence values.

The ability of the claimed compounds to enhance Notch signaling in cellular systems is quantitatively determined here using a corresponding Notch reporter assay, i.e. based on the luminescence readout of luciferase. For this purpose, use is made ofHD (Promega, # E2311) as a transfection reagent, HeLa cells available from the American Type Culture Collection (ATCC) with accession number ATCC-CCL-2 were transiently transfected for 24 hours with a membrane-line form of the constitutively active intracellular domain of human Notch1 receptor (hNotch1 Δ E) that activates the Notch signaling cascade (BPS Bioscience, a customized human analog of Notch pathway reporter kit #60509 component C), luciferase expressed under the control of a Notch responsive promoter to monitor Notch signaling (BPS Bioscience, Notch pathway reporter kit #60509, CSL luciferase reporter vector from component a not pre-mixed with renilla luciferase vector), and an expression vector for renilla luciferase expressed constitutively in a manner independent of Notch signaling to include a measure of the number of cells per sample, which were deposited as ATCC-CCL-2. HeLa cells were cultured in DMEM medium (Fisherschelientific, #11584456) containing 10% fetal bovine serum (Fisherschelientific, # 15517589). Transfection was performed in 100 mm-dishes (StarLab, # CC7682-3394) with cells correctly attached to the plates at 80-90% cell confluence in a total volume of 7mL of medium. For each dish to be transfected, 40. mu.L hNotch 1. delta.E expression vector (100 ng/. mu.L), 80. mu.L CSL luciferin were added to 238. mu.L Opti-MEM (Fisherschelentipic, #10149832) Enzyme reporter vector (40 ng/. mu.L), 4. mu.L PR1-SV 40-Renilla luciferase vector (10 ng/. mu.L), and 18.1. mu.L was added in the last stepHD to prepare transfection mixtures. In the process of addingAfter HD, the transfection mixture was allowed to stand at room temperature for 15 minutes and then evenly distributed into the petri dish. 24 hours after transfection, the transfected cells were carefully detached from the petri dish with 0.5mM EDTA in PBS and seeded into 96-well plates (CORNING, #3610) suitable for luminescence readings at a density of 10,000 cells per well. The cells were then diluted to 0.1% v/v in H with a final concentration of 10. mu.M (from a stock solution of 10mM in DMSO to2O (final DMSO concentration in water for injection, WFI, fisher scientific #10378939) or incubated with 0.1% v/v of the empty vehicle DMSO as a control for 20 hours. After this time, the cells were washed once with PBS, and then washed with 30 μ L/well of passive lysis buffer (Promega, # E194A,component of reporter assay, # E1910) was subjected to lysis. Immediately after lysis, the firefly luciferase value was first measured and then the Renilla luciferase value was measured sequentially from the same well using a luminescence reader, directly after application of 15. mu.L/well of each of the respective enzyme substrates required for generating a luminescent signal (Promega, Reporter assay system, # E1910).

The applicability of assays for monitoring Notch signaling was controlled by additionally including a generally accepted commercial Notch inhibitor (i.e., DAPT) as a negative control, and a reported Notch enhancer Resveratrol (RES) as a positive control (Pinchot et al, Cancer 2011, 117, 1386-. Two control compounds were also tested at 10 μ M.

Six replicates of each compound were performed per single experiment. This experiment was independently repeated three or more times for each compound. Values of Notch reporter luciferase were normalized by dividing by the corresponding individual Notch independent Renilla values to eliminate the effect of absolute cell number variation between samples. For each individual plate, a second normalization was performed in a single experiment on the equiweighted arithmetic mean (abbreviated here as AVE) of the six relevant renilla normalized DMSO-control values to obtain a relative value relative to the baseline level of 1.0. Statistical calculations were performed similarly to the proliferation assay described above. For this purpose, two independent outlier analyses were performed according to the method of Peirce and Chauvenet (Ross, Journal of Engineering Technology 2003, 1-12). Outliers identified by at least one method were excluded from the calculation, but in a single experiment, did not exceed one of the 6 values for each compound. Weighted arithmetic mean AVE for each compound wCalculated from the double normalized values of all independent replicates of a single experiment each comprising six replicates. The corresponding standard deviation of the weighted arithmetic mean is calculated according to the method described by Bronstein et al (Bronstein, Semendjjajew, Musiol, Muhlig, Taschenbuch der Mathemeitk, 5 th edition 2001(German), publishers: Verlag Harri Deutsch, Frankfurt am Main and Thun) and combined with the Gaussian error propagation associated with performing the normalization calculation. The resulting standard deviation is referred to herein as the "combined standard deviation".

If the double normalized equal-weight arithmetic mean values from three independent repeats differ considerably, the number of independent repeats is increased to four or more. In the case of four or more independent replicates, two-line outlier analysis was performed on all double normalized equiweighted arithmetic means according to the method of Peirce and Chauvenet described above.

A compound is considered to be a Notch signaling enhancing molecule, i.e. an enhancer of Notch signaling, if the weighted arithmetic mean of the luminescence values minus the respective combined standard deviations, relative to the overall base level of 1.0, reaches 1.1 or higher, in particular 1.2 or higher, 1.3 or higher, 1.4 or higher, 1.5 or higher, 1.7 or higher and 2.0 or higher. The overall basal level was calculated as the weighted arithmetic mean of all double normalized values of the DMSO control measurements, similar to the calculation performed for the test compounds. The corresponding combined standard deviation of DMSO values is less than 1.10 -2

Several molecules falling within the scope of the compounds defined in formulae (Ib) and (Ic) herein, respectively, have been identified as enhancers of Notch signaling according to the above methods. The enhancers of Notch signaling identified to date relate to the compounds listed in table 92. The entries in table 92 are sorted according to the corresponding weighted arithmetic mean of the compounds without regard to the respective standard deviation, and thus fall within the activity ranges shown.

Table 92: notch receptor assay

Several other molecules have not been identified as enhancers of Notch signaling according to the above methods.

In some cases, the growth inhibitory properties are associated with Notch enhancing properties, in other cases, the growth inhibitory properties are not associated with Notch enhancing properties.

The biological activity of the claimed compounds can be attributed to, but is not limited to, Notch signaling enhancing activity. The Notch modulating properties of the claimed compounds may alternatively or in combination with mechanisms leading to an antiproliferative effect be used in pharmaceutical therapy, preferably in the treatment of hyperproliferative disorders, including cancer and non-malignant hyperproliferative disorders.

In one aspect, the invention relates to the treatment of skin, skin appendages, mucous membranes, mucous membrane appendages, cornea, and all kinds of epithelial tissue. The term "skin" relates to tissue including the epidermis and dermis. The term "mucosa" relates to the mucosa and submucosal tissues including the oral, nasal, ocular, otic, respiratory, genital, urothelial, anal and rectal mucosa. The term "appendages" relates to tissues including hair follicles, hairs, fingernails, toenails, and glands, including sebaceous glands, sweat glands, such as the apocrine or eccrine sweat glands, and mammary glands.

In one embodiment, the invention relates to the treatment of: non-melanoma skin cancers and precancerous lesions such as Basal Cell Carcinoma (BCC), Squamous Cell Carcinoma (SCC), sebaceous gland carcinoma, merkel cell carcinoma, angiosarcoma, cutaneous B-cell lymphoma, cutaneous T-cell lymphoma, cutaneous fibrosarcoma, Actinic Keratosis (AK) or Bowen's Disease (BD), as well as other squamous epithelial cancers and precancerous lesions such as skin SCC, lung SCC, head and neck SCC, oral SCC, tongue SCC, esophageal SCC, cervical SCC, periocular SCC, thyroid SCC, penile SCC, vaginal SCC, prostate SCC and bladder SCC.

In another embodiment, the invention relates to the treatment of: skin and mucosal disorders with keratosis defects (keratoses) and/or abnormal keratinocyte proliferation, such as psoriasis, Darie disease, lichen planus, lupus erythematosus, ichthyosis, or verruca vulgaris (senile).

In another embodiment, the invention relates to the treatment of: skin and mucosal diseases and skin and mucosal cancers each associated with and/or caused by viral infection, such as warts, and warts, papillomas, HPV-associated papillomas, papillomas and HPV-associated papillomas associated with HPV (human papilloma virus), e.g., warts (plantar warts), flat warts (flat warts)/plane warts (plane warts)), filiform warts (filiform warts), mosaic warts, periungual warts, subungual warts, oral warts, genital warts, fibroepithelial papillomas (fibroEpoepidermal papilloma), intraductal papillomas, inverted papillomas, basal cell papillomas, squamous papillomas, cutaneous papillomas, fibrovascular papillomas, plexas, nasal papillomas, cutaneous papillomas, and papillomas, Lymphostatic dermal Papillomatosis (Papillomas Papillomatosis), confluent reticular Papillomatosis (Papillomas confluens et regressions), or laryngeal Papillomatosis (respiratory Papillomatosis), herpes-related diseases such as cold sores, genital herpes, herpes zoster, corneal herpes or Kaposi's sarcoma and HPV-related cancers of the cervix, vulva, penis, vagina, anus, oropharynx, tongue and oral cavity.

In another embodiment, the invention relates to the treatment of atopic dermatitis.

In another embodiment, the present invention relates to the treatment of acne.

In another embodiment, the present invention relates to the treatment of skin wounds, wherein the process of wound healing is accelerated.

In another embodiment, the invention relates to the treatment of cancer associated with and/or caused by viral infection, i.e. cancer viral infection, for example cancer associated with HBV and HCV (hepatitis b and c virus), such as liver cancer, cancer associated with EBV (epstein-barr virus), such as burkitt lymphoma, hodgkin lymphoma and non-hodgkin lymphoma and gastric cancer, cancer associated with HPV (human papilloma virus), such as cervical cancer, cancer associated with HHV (human herpes virus), such as kaposi's sarcoma, and cancer associated with HTLV (human T-cell lymphotrophic virus), such as T-cell leukemia and T-cell lymphoma.

Another aspect of the invention relates to the treatment of immune system related disorders. As used herein, the term "immune system-related disorder" applies to pathological conditions of the hematopoietic system, including the blood system, in particular to immune cells belonging to the innate or adaptive immune system.

Another aspect of the invention relates to therapeutic use in immune system related applications. As used herein, the term "immune system-related application" applies to the intervention of the proliferation, differentiation and/or activation of cell lineages of the hematopoietic system, including the hematopoietic system, to modulate the immune response (immunomodulation). As used herein, the term "immune system-related application" also applies to the intervention of the cellular and non-cellular microenvironment at the site of immune cell action to support and/or enable immune cells to exert their properties. In particular, the intervention defined herein by the term "immune system-related application" relates to immune cells belonging to the innate or adaptive immune system.

Thus, the compounds of the invention may be used in immunotherapy alone or in combination with other immunotherapeutic methods or compounds (as immunoadjuvants, e.g. as vaccine adjuvants, or as adjuvants for immunotherapy). As used herein, the term "immunotherapy" applies to activated immunotherapy of patients without immunodeficiency or with acquired or congenital immunodeficiency, as well as immune recovery to enhance the functionality of the immune system in response to pathogens or pathologically transformed endogenous cells (such as cancer cells).

As used herein, the term "other immunotherapeutic methods" applies to vaccination, antibody therapy, cytokine therapy, the use of immune checkpoint inhibitors and immune response stimulating drugs, as well as to the autologous transplantation of genetically modified or unmodified immune cells that can be stimulated with intercellular signals, signaling molecules, antigens or antibodies, i.e., adoptive immune cell transfer.

The methods of use of the invention in immune system related applications and other immunotherapeutic methods involve in vivo, in vitro and ex vivo use, respectively.

Specific examples are the activation and/or enhancement of the activation of peripheral T lymphocytes, including T helper cells and cytotoxic T cells, to enhance the immune response, in particular to stimulate proliferation upon antigen recognition and/or the production and/or secretion of cytokines and/or cytotoxic agents, to amplify the immune response, such as activating B lymphocytes and/or enhancing the activation of B lymphocytes, to amplify the immune response, in particular to stimulate proliferation and/or antibody production and/or secretion, such as by increasing the number of specific immune cell subtypes, by modulating differentiation and/or cell fate decisions during immune cell development (e.g. modulating, in particular increasing, the number of immune cells belonging to T cell and B cell lineages, including marginal zone B cells, cytotoxic T cells or T helper (Th) subsets (in particular Th1, T helper (Th) subsets, Th2, Th17) and regulatory T cells) to enhance the immune response; or as an immunological adjuvant such as a vaccine adjuvant.

Another aspect of the invention relates to the treatment of muscle diseases, including diseases of skeletal muscle, cardiac muscle and smooth muscle.

In one embodiment, the invention relates to the treatment of Muscular Dystrophy (MD).

Specific examples are Duchenne type MD, Becker type MD, congenital MD, Limb-Girdle type MD, facioscapulohumeral type MD, Emery-Dreifuss type MD, distal MD, myotonic MD or eupharyneal MD.

In another embodiment, the invention relates to the treatment of muscle hyperproliferative disorders, including myoblastoma, rhabdomyoma and rhabdomyosarcoma, as well as muscle hyperplasia and muscle hypertrophy.

In another embodiment, the compounds of the invention may be used in muscle atrophy associated with aging or in muscle atrophy associated with disease (such as myositis and fibromyositis or polio), for example for muscle regeneration following pathological muscle degeneration or atrophy caused by trauma, caused by muscle ischemia or caused by inflammation.

Another aspect relates to the treatment of: disorders of the neuroendocrine system, such as cancers of the neuroendocrine system, including neuroendocrine small cell carcinomas, neuroendocrine large cell carcinomas, and carcinoid tumors, for example, tumors of the brain, thyroid, pancreas, gastrointestinal tract, liver, esophagus, and lung, such as neuroendocrine tumors of the pituitary gland, neuroendocrine tumors of the adrenal gland, Medullary Thyroid Carcinoma (MTC), C-cell hyperplasia, Anaplastic Thyroid Carcinoma (ATC), parathyroid adenoma, intrathyroid nodules, island carcinomas, hyaline trabecular tumors, paragangliomas, lung carcinoid tumors, neuroblastoma, gastrointestinal carcinoid, Goblet-cell carcinoid, pancreatic carcinoid, gastrinoma, glucagonomas, somatostatin tumors, viromas (VIPona), insulinoma, nonfunctional islet cell tumor, multiple endocrine tumor type 1, or lung carcinoid.

Another aspect relates to the treatment of lung disorders such as lung cancer, including Small Cell Lung Cancer (SCLC) and non-small cell lung cancer (NSCLC), including squamous cell carcinoma of the lung, adenocarcinoma of the lung, and large cell carcinoma of the lung.

Another aspect relates to the treatment of: cancers or precancerous lesions of brain, pancreas, breast, ovary, liver, thyroid, genitourinary tract, gastrointestinal tract and endothelial tissue, including glioma, mixed glioma, glioblastoma multiforme, astrocytoma, anaplastic astrocytoma, glioblastoma multiforme, oligodendroglioma, anaplastic oligoastrocytoma, ependymoma, anaplastic ependymoma, mucinous ependymoma, subintimal tumor, brainstem glioma, optic glioma and forebrain tumor, pancreatic adenocarcinoma, pancreatic ductal adenocarcinoma, pancreatic acinar cell carcinoma, pancreatic pseudopapillary tumor, pancreatic ductal papillary-mucinous tumor, pancreatic mucinous cystadenocarcinoma, pancreatic glioblastoma and pancreatic intraepithelial tumors, hepatocellular carcinoma, fibrolamellar hepatocellular carcinoma, papillary thyroid carcinoma and follicular thyroid carcinoma, Cervical cancer, hormone receptor positive breast cancer and hormone receptor negative breast cancer, ovarian cancer, gastric cancer and angiosarcoma.

As used herein, the term "treating" or "treatment" refers to one or more of the following: (1) inhibiting the disease; for example, inhibiting a disease, disorder, or condition in an individual who is experiencing or exhibiting a pathology or symptomatology of the disease, disorder, or condition (i.e., arresting further development of the pathology and/or symptomatology); and (2) ameliorating the disease; for example, ameliorating a disease, disorder or condition in an individual who is experiencing or exhibiting a pathology or symptom of the disease, disorder or condition (i.e., reversing the pathology and/or symptom), such as reducing the severity of the disease. The term "treatment" also encompasses post-treatment care.

In some embodiments, administration of a compound of the invention or a pharmaceutically acceptable salt thereof is effective to prevent disease; for example, a disease, disorder or condition is prevented in an individual who may be predisposed to the disease, disorder or condition but has not yet experienced or exhibited the pathology and/or symptomology of the disease.

The compounds of the invention are useful in human and veterinary medicine, including the treatment of companion animals such as horses, dogs, cats, rabbits, guinea pigs, fish (e.g., koi), birds (e.g., falcon); and livestock (e.g., cattle), poultry, pigs, sheep, goats, donkeys, yaks, and camels.

Pharmaceutical composition

The invention also provides a pharmaceutical composition for use in medicine (e.g., human or veterinary medicine) comprising a compound described herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the composition further comprises a pharmaceutically acceptable carrier.

An effective dose of a compound according to the invention or a salt, solvate or prodrug thereof is used in addition to a physiologically acceptable carrier, diluent and/or adjuvant to produce a pharmaceutical composition. The dosage of the active compound may vary depending on the route of administration, the age and weight of the patient, the nature and severity of the disease to be treated and similar factors. The daily dose may be administered in a single dose, which may be administered at one time, or may be subdivided into two or more daily doses, typically 0.001 to 2000 mg. It is particularly preferred to provide a daily administration dose of 0.1 to 500mg (e.g. 0.1 to 100 mg).

Suitable administration forms are topical or systemic, including enteral, oral, rectal, and parenteral, such as infusion and injection, intravenous, intraarterial, intraperitoneal, intramuscular, intracardiac, epidural, intracerebral, intracerebroventricular, intraosteal, intraarticular, intraocular, intravitreal, intrathecal, intravaginal, intracavernosal, intravesical, subcutaneous, intradermal, transdermal, transmucosal, inhalation, intranasal, buccal, sublingual, and intralesional formulations. Oral, parenteral (e.g. intravenous or intramuscular), intranasal (e.g. dry powder or sublingual) formulations of the compounds according to the invention are particularly preferred. Conventional galenic forms can be used, such as tablets, sugar-coated tablets, capsules, dispersible powders, granules, aqueous solutions, alcoholic aqueous solutions, aqueous or oily suspensions, gels, hydrogels, ointments, creams, lotions, shampoos (shammpoos), lipsticks, mouthwashes, foams, pastes, tinctures, skin patches and tapes, occlusive forms or forms in combination with time-release drug delivery systems, with electrophoretic skin delivery systems including implants and devices, and with jet injectors, liposomes and carrier vesicles (transfersome vesicles), vapors, sprays, syrups, juices or drops and eye drops.

Solid pharmaceutical forms may contain inert ingredients and carrier substances such as calcium carbonate, calcium phosphate, sodium phosphate, lactose, starch, mannitol, alginates, gelatin, guar gum, magnesium stearate, aluminium stearate, methyl cellulose, talc, highly dispersed silicic acids, silicone oils, high molecular weight fatty acids (such as stearic acid), gelatin, agar or vegetable or animal fats and oils, or solid high molecular weight polymers (such as polyethylene glycol); formulations suitable for oral administration may contain additional flavouring and/or sweetening agents, if desired.

The liquid pharmaceutical forms can be sterilized and/or contain auxiliary substances, such as preservatives, stabilizers, wetting agents, osmotic agents, emulsifiers, spreading agents, solubilizers, salts, sugars or sugar alcohols (for regulating the osmotic pressure or for buffering), and/or viscosity regulators, as appropriate. Examples of such additives are tartrate and citrate buffers, ethanol and chelating agents (such as ethylenediaminetetraacetic acid and its non-toxic salts). High molecular weight polymers, such as liquid polyethylene oxide, microcrystalline cellulose, carboxymethylcellulose, polyvinylpyrrolidone, dextran or gelatin, are suitable for adjusting the viscosity. Examples of solid carrier materials are starch, lactose, mannitol, methylcellulose, talc, highly disperse silicic acids, high molecular weight fatty acids (such as stearic acid), gelatin, agar, calcium phosphate, magnesium stearate, animal and vegetable fats and solid high molecular weight polymers, such as polyethylene glycol.

Oily suspensions for parenteral or topical use may be vegetable, synthetic or semisynthetic oils, such as liquid fatty acid esters having in each case from 8 to 22 carbon atoms in the fatty acid chain, for example palmitic, lauric, tridecanoic, margaric, stearic, arachidic, myristic, behenic, pentadecanoic, linoleic, elaidic, brasidic, erucic or oleic acid, which is esterified with mono-to trihydric alcohols having from 1 to 6 carbon atoms, such as methanol, ethanol, propanol, butanol, pentanol or isomers thereof, ethylene glycol or glycerol. Examples of such fatty acid esters are, inter alia, commercial miglitol, isopropyl myristate, isopropyl palmitate, isopropyl stearate, PEG 6-decanoic acid, caprylic/capric acid esters of saturated fatty alcohols, polyoxyethylene glycerol trioleate, ethyl oleate, waxy fatty acid esters, such as, for example, artificial duck tail fat, isopropyl cocoate fatty acid, oleyl oleate, decyl oleate, ethyl lactate, dibutyl phthalate, diisopropyl adipate, polyol fatty acid esters. Silicone oils of different viscosities or fatty alcohols, such as isotridecanol, 2-octyldodecanol, cetostearyl alcohol or oleyl alcohol, or fatty acids, such as oleic acid, are also suitable. Vegetable oils such as castor oil, almond oil, olive oil, sesame oil, cottonseed oil, peanut oil or soybean oil may also be used.

Suitable solvents, gelling agents and solubilizers are water or water-miscible solvents. Examples of suitable substances are alcohols such as ethanol or isopropanol, benzyl alcohol, 2-octyldodecanol, polyethylene glycol, phthalate, adipate, propylene glycol, glycerol, dipropylene glycol or tripropylene glycol, waxes, methyl cellosolve, esters, morpholine, dioxane, dimethyl sulfoxide, dimethylformamide, tetrahydrofuran, cyclohexanone, and the like.

Cellulose ethers which are soluble or swellable in water or organic solvents, such as hydroxypropyl methylcellulose, methylcellulose or ethylcellulose, or soluble starches, can be used as film formers.

Mixtures of gelling agents and film formers are also fully useful. In this case, ionic macromolecules are used in particular, such as sodium carboxymethylcellulose, polyacrylic acids, polymethacrylic acids and salts thereof, sodium amylopectin hemiglycolate, alginic acid or propylene glycol alginate (as sodium salt), gum arabic, xanthan gum, guar gum or carrageenan. The following materials may be used as additional formulation aids: glycerol, paraffin of different viscosities, triethanolamine, collagen, allantoin and phenylbenzimidazole sulfonic acid (novantisolidic acid). The formulations may also require the use of surfactants, emulsifiers or wetting agents, for example sodium lauryl sulphate, fatty alcohol ether sulphates, disodium N-lauryl- β -iminodipropionate, polyethoxylated castor oil or sorbitan monooleate, sorbitan monostearate, polysorbates (e.g. Tween), cetyl alcohol, lecithin, glycerol monostearate, polyoxyethylene stearate, alkylphenol polyglycol ethers, cetyltrimethylammonium chloride or mono/dialkyl polyglycol ether orthophosphoric acid monoethanolamine salts. Stabilizers for stabilizing emulsions or for preventing the decomposition of active substances such as antioxidants (for example tocopherol or butylhydroxyanisole) or preservatives (for example parabens), i.e. montmorillonite or colloidal silicic acid, are likewise useful for preparing the desired formulations.

Formulations for parenteral administration may be presented in individual dosage unit form, such as ampoules or vials. Preference is given to using solutions, preferably aqueous solutions, in particular isotonic solutions, and also suspensions of the active compounds. These injectable forms can be prepared as ready-to-use formulations or simply immediately before use, by mixing the active compound (for example lyophilizate, where appropriate together with other solid carrier materials) with the desired solvents or suspending agents.

Intranasal formulations may be presented as aqueous or oily solutions or aqueous or oily suspensions. They may also be presented in the form of a lyophilizate, which is prepared prior to use using a suitable solvent or suspending agent.

Inhalable formulations may be presented as powders, solutions or suspensions. Preferably, the inhalable formulation is in powder form, e.g. a mixture of the active ingredient with suitable formulation auxiliaries such as lactose.

The formulations are produced, aliquoted and sealed under conventional antibacterial and aseptic conditions.

As indicated above, the compounds of the present invention may be administered as a combination therapy, sequential therapy or concurrent combination therapy with other active agents (e.g., therapeutically active compounds for the treatment of the above-mentioned conditions). These therapeutically active compounds may include, but are not limited to, chemotherapeutic agents such as nucleoside and nucleobase analogs, e.g., cytarabine, gemcitabine, azathioprine, mercaptopurine, fluorouracil, thioguanine, azacytidine, capecitabine, doxifluridine; such as platinum-based drugs, e.g., cisplatin, oxaliplatin, carboplatin, and nedaplatin; such as anthracyclines, for example doxorubicin, epirubicin, valrubicin, idarubicin, daunorubicin, sarubicin, Pixantrone and mitoxantrone; such as peptide antibiotics, e.g., actinomycin and bleomycin; such as alkylating agents, for example Mechlorethamine (Mechlorethamine), chlorambucil, melphalan, nitrosoureas, dacarbazine, temozolomide and cyclophosphamide; such as antimitotic agents including taxanes and vinca alkaloids, e.g., docetaxel, paclitaxel, Abraxane, cabazitaxel, vinblastine, vindesine, vinorelbine, and vincristine; such as topoisomerase inhibitors, e.g., irinotecan, topotecan, teniposide, and etoposide; such as other cytostatic agents, for example hydroxyurea and methotrexate; such as proteasome inhibitors, e.g., bortezomib, ixazoib; and other targeted therapeutics such as kinase inhibitors, cell cycle inhibitors, modulators, i.e., inhibitors and activators of signaling pathways including growth factor signaling, cytokine signaling, NF-kb signaling, AP1 signaling, JAK/STAT signaling, EGFR signaling, TGF- β signaling, Notch signaling, Wnt signaling, Hedgehog signaling, hormone and nuclear receptor signaling, e.g., erlotinib, lapatinib, dasatinib, imatinib, afatinib, vemurafenib, darafenib (Dabrafenib), nilotinib, cetuximab, trametinib, palexib, cobicistinib (Cobimetinib), carboazatinib, pegaptanib sodium (pegaptanib), crizotinib, lapatinib, panitumumab, cabozatinib (Cabozantinib), agolinatinib, reginib (entratinib), ritinib (entecatinib), ritinib (entecanib), and the like, Ranibizumab, ibrutinib (ibrutinib), trastuzumab, rituximab, alemtuzumab, gefitinib, bevacizumab, lenvatinib (lenvatinib), bosutinib, axitinib, pazopanib, everolimus, temsirolimus, ruxolitib, tofacitinib, sorafenib, sunitinib, aflibercept, vandetanib, vismodegib (vismodegib), and sonidegu (sonidegib); retinoids such as retinol, tretinoin, isotretinoin, alitretinoin, bexarotene, tazarotene, acitretin, adapalene and etretinate; hormone signaling modulators, including estrogen receptor modulators, androgen receptor modulators, and aromatase inhibitors, such as raloxifene, tamoxifen, fulvestrant, lasofoxifene, toremifene, bicalutamide, flutamide, anastrozole, letrozole, and exemestane; histone deacetylase inhibitors such as Volinostat, Romidepsin (Romidepsin), Panostat, Berlin stat and Cedar benamine; and Ingenol Mebutate (Ingenol Mebutate); and other Notch enhancers not encompassed by the compounds of the invention, such as valproic acid, resveratrol, hesperetin, chrysin, phenethyl isothiocyanate, thiocoraline; n-methylhydroxyethyl chloride and a Notch signaling activating peptide or antibody; and immune response modifiers, including immune checkpoint inhibitors, such as imiquimod, ipilimumab, atilizumab, ofatumumab, rituximab, nivolumab, and parboluzumab (Pembrolizumab); and anti-inflammatory agents, including glucocorticoids and non-steroidal anti-inflammatory agents, such as cortisol-based formulations, dexamethasone, betamethasone, prednisone, prednisolone, methylprednisolone, triamcinolone acetonide-hexaacetonide (triamcinolone-hexetonide), mometasone furoate (mometasone furoat), clobetasol propionate (clobetasol propinoat), acetylsalicylic acid, salicylic acid and other salicylates, diflunisal, ibuprofen, dexibuprofen, naproxen, fenoprofen, ketoprofen, dexketoprofen, loxoprofen, flurbiprofen, oxaprozin, indomethacin, ketorolac, tolmetin, diclofenac, etodolac, aceclofenac, nabumetone, sulindac, Mefenamic acid (Mefenamic acid), meclofenamic acid, flufenamic acid, tolfenamic acid, etocelipran, sertib, and felicic; and an ACE inhibitor; and a beta-blocker; and a myostatin inhibitor; and a PDE-5 inhibitor; and an antihistamine. For combination therapy, the active ingredients may be formulated as a composition comprising several active ingredients in a single dosage form and/or as a kit comprising the individual active ingredients in separate dosage forms. The active ingredients used in the combination therapy may be administered together or separately.

The compounds of the invention may be administered in the form of antibody-drug conjugates.

The compounds of the invention may be administered in combination with surgery, cryotherapy, electro-desiccation, radiation therapy, photodynamic therapy, laser therapy, chemotherapy, targeted therapy, immunotherapy, gene therapy, antisense therapy, cell-based transplantation therapy, stem cell therapy, physical therapy and occupational therapy.

Chemical synthesis

Abbreviations

Ac acetyl group

aq aqueous

Alk alkyl group

Bn benzyl group

BRSM based on recycled raw material (yield)

Bu butyl

mCPBA chloroperoxybenzoic acid

NMR nuclear magnetic resonance spectroscopy

DCE 1, 2-dichloroethane

DCM dichloromethane

DIBAL-H diisobutylaluminum hydride

DMF N, N-dimethylformamide

DMSO dimethyl sulfoxide

DMAP dimethylaminopyridine

equiv equivalent of

ESI electrospray ionization

EDC N- (3-dimethylaminopropyl) -N' -ethylcarbodiimide

HOBt 1-hydroxybenzotriazole

HATU hexafluorophosphate azabenzotriazole tetramethylurea

Et Ethyl group

LiHMDS lithium bis (trimethylsilyl) amide

Me methyl group

Ms methanesulfonyl

PE Petroleum Ether

PG protecting group

PTSA para-toluenesulfonic acid

Sat

TBAF tetrabutylammonium fluoride

Tf trifluoromethanesulfonyl

THF tetrahydrofuran

TMS trimethylsilyl group

Ts p-toluenesulfonyl group

UV ultraviolet light

General rule

The compounds listed in tables 93 and 94 have been identified by TLC using pre-coated silica TLC plates and common organic solvents such as petroleum ether, ethyl acetate, dichloromethane, methanol, toluene, triethylamine or acetic acid (as eluents, preferably as binary or ternary solvent mixtures thereof). The compounds are visualized using UV light at wavelengths of 254nm or 366nm and/or common staining solutions such as phosphomolybdic acid, potassium permanganate or ninhydrin.

In addition, the compounds listed in table 93 were also identified by mass spectrometry (formic acid was used to detect positive ions in the mobile phase, and no additives were used to detect negative ions). Ammonium carbonate is used if the molecule is difficult to ionize in negative mode. Representative compounds and those that showed poor ionization in the mass spectrum were also identified by nmr spectroscopy (table 94). Chemical shifts (δ) are reported in parts per million (ppm) relative to the residual solvent peak, rounded to the nearest 0.01ppm for protons and to the nearest 0.1ppm for carbon (reference: CHCl) 3[1H:7.26ppm,13C:77.2ppm],DMSO[1H:2.50ppm,13C:39.5ppm]). The coupling constant (J) is reported in Hz to the nearest 0.1 Hz. Peak multiplicities are represented as follows: s (singlet), d (doublet), t (triplet), q (quartet), hept (heptad), m (multiplet) and br (broad).

Synthesis of the said compounds

The above-described compounds of the invention falling within the scope of formula I can be synthesized and purified by one skilled in the art, and preferably synthesized according to the general procedures (a to R) described herein, as shown in scheme 1.

Scheme 1: general synthetic schemes.

A) To the corresponding mono-or disubstituted phenol (1.0-1.5 equivalents) and 4-alkyl ester halo (hetero) aryl (1 equivalent) dissolved in DMSO (0.5M) was added K under argon and stirring2CO3(1.5 equivalents) and the mixture is stirred at room temperature or heated between 40 ℃ and 160 ℃ until complete conversion. The mixture is brought to room temperature and partitioned between organic solvents, preferably petroleum ether and water. The aqueous layer was extracted two more times, then the combined organic phases were washed with NaOH (aq, 2M), then brine, over Na2SO4Dried, filtered and concentrated in vacuo. Then the residue is passed throughFlash chromatography (SiO)2Gradient petroleum ether/AcOEt, DCM/MeOH or petroleum ether/AcOEt/NEt3) Purification to give the desired bis (hetero) aryl ether ethyl ester.

B) The corresponding bis (hetero) aryl ether alkyl ester (1 eq) was dissolved in anhydrous THF (0.2M) under argon and stirring, and the resulting solution was cooled to 0 ℃ with an ice bath. DIBAL-H (2.5 eq., 1.2M in toluene) was then added dropwise and the mixture was stirred at this temperature until complete conversion. The reaction is stopped by the Fieser method, filtered, concentrated in vacuo and the residue is then chromatographed by flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield the desired alcohol.

C) Depending on scale and substrate, either of these procedures was used.

To the corresponding alcohol (1 eq) dissolved in DCM (0.2M) was added MnO with vigorous stirring2(2-4 equivalents). The resulting suspension was stirred at room temperature or 40 ℃ until complete conversion. The reaction was then diluted with AcOEt, filtered over celite, and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to give the desired aldehyde.

To the corresponding alcohol (1 eq) dissolved in DCM or DMSO (0.2M) was added dess-Martin iodophor (Des-Martin periodinane) (1.2 eq) with vigorous stirring. The resulting suspension was stirred at room temperature until complete conversion. The solution was diluted in AcOEt and saturated NaHCO 3The aqueous solution was quenched and then phase separated. The aqueous layer was extracted two more times and the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to give the desired aldehyde.

To a solution of oxalyl chloride (2 equiv.) in DCM (0.2M) was added anhydrous DMSO (4 equiv.) at-78 ℃ and the mixture was stirred for 30 min. Then a solution of the corresponding alcohol (1 eq.) in DCM (0.2M) was added, followed by the addition of freshly distilled NEt3(8 equivalents). The resulting solution was stirred for 1 hour and then slowly returned to room temperature. The solution was diluted in AcOEt and quenched with aqueous HCl 1M solutionThe phases were separated. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to give the desired aldehyde.

D) To the corresponding aldehyde (1 equivalent) dissolved in anhydrous THF (0.2M) was added TMSCF with stirring at 0 deg.C under argon atmosphere3(2 equiv.) followed by TBAF (1 mol%) to obtain the corresponding CF-containing3Or adding Grignard reagent (2 equivalents) to obtain the corresponding secondary alkyl alcohol. In both cases, the resulting solution is stirred at this temperature until complete conversion. Aqueous HCl (2.5M) was then added and the reaction stirred for an additional 1 hour. The reaction was then partitioned between AcOEt and water. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na 2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to give the desired secondary alcohol.

E) To a stirred solution of the corresponding secondary alcohol (1 eq) in chloroform (0.2M) was added dess-Martin periodinane (1.5 eq.) at 0 ℃. After the reaction was complete, it was reacted with AcOEt and saturated NaHCO3The aqueous solution was partitioned. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield the desired ketone.

F) To a stirred solution of the corresponding ketone (1 equivalent) in ethanol (0.2M) is added an amine (2.5-40 equivalents) followed by a catalytic amount of PTSA in the case of an aliphatic amine or a base (2.5-40 equivalents) in the case of hydroxylamine. The reaction was then refluxed for 24-72 hours. Thereafter, celite was added and the volatiles were evaporated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to give the desired imine.

G) DCM (0.25M) obtained before purification in procedure (F)) To a solution of the corresponding aldehyde or crude imine, sodium borohydride (4 equivalents) was added and the solution was stirred for an additional 2 hours. The reaction was then incubated with AcOEt and saturated NaHCO 3The water is distributed among the water. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield the desired product.

H) To a stirred solution of the corresponding aldehyde (1 eq) in toluene (0.2M) under argon was added an amine (2 eq) followed by TMSCN (2 eq) and the reaction was stirred for 16 hours. The reaction was then quenched with AcOEt and saturated NaHCO3The aqueous solution was partitioned. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield the desired cyanamide.

I) To the corresponding bis (hetero) aryl ether alkyl ester (1 eq) dissolved in EtOH or THF (0.5M) was added aqueous NaOH 2M (2 eq) and the reaction was stirred until completion. The reaction was then partitioned between AcOEt and aqueous HCl (1M). The aqueous layer was extracted two more times and the combined organic phases were washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The residue is then purified by flash chromatography (SiO2, gradient petroleum ether/AcOEt) or recrystallization (AcOEt) to give the desired carboxylic acid.

J) Depending on the amine used, either of these procedures is employed.

SOCl was first added to the corresponding bis (hetero) aryl ether carboxylic acid (1 equivalent) suspended in stirred toluene (0.2M) under argon2(2.5 eq.) DMF (1 mol%) was then added and the mixture was heated to 80 ℃ for 3 hours. The reaction mixture was then evaporated to dryness and the residue obtained was again placed under argon and redissolved in THF (0.2M). To this solution was added trimethylamine (2.5 equiv.), DMAP (1 mol%) and the corresponding amine or amide (1.2-1.5 equiv.) in that order, and the suspension was stirred for 16 h. Then the reaction is carried outPartition was performed between AcOEt and aqueous HCl (1M). The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then purified by flash chromatography (SiO)2Gradient petroleum ether/AcOEt or petroleum ether/AcOEt/AcOH or DCM/MeOH) to afford the desired amide.

To the corresponding bis (hetero) aryl ether carboxylic acid (1 eq) in DCM (0.2M) was added NEt in sequence3(3 equiv.) and HOBt/EDCI (1.5 equiv/1.5 equiv.) or HATU (1.5 equiv.). The reaction mixture was then stirred for 5-60 minutes, then the corresponding amine (1.25 equivalents) was added and the mixture was stirred to completion. The reaction was then partitioned between AcOEt and aqueous HCl (1M). The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na 2SO4Dried, filtered and concentrated in vacuo. The residue was then purified by flash chromatography (SiO)2Gradient petroleum ether/AcOEt or petroleum ether/AcOEt/AcOH or DCM/MeOH) to afford the desired amide.

SOCl was first added to the corresponding bis (hetero) aryl ether carboxylic acid (1 equivalent) suspended in stirred toluene (0.2M) under argon2(2.5 eq.) DMF (1 mol%) was then added and the mixture was heated to 80 ℃ for 3 hours. The reaction mixture was then evaporated to dryness and the residue obtained was again placed under argon and redissolved in toluene (0.2M). This solution was added to a saturated aqueous solution of the corresponding hydroxylamine. Stirred NaHCO3And the reaction mixture until complete. The reaction was then partitioned between AcOEt and aqueous HCl 1M or water. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then purified by flash chromatography (SiO)2Gradient petroleum ether/AcOEt or petroleum ether/AcOEt/AcOH or DCM/MeOH) to afford the desired amide.

K) To the corresponding 4-substituted phenol (1 eq) and 1, 4-dibromoaryl (2.5 eq) dissolved in DMF (0.2M) was added Cs2CO3(2 equiv.), CuI (10 mol%) and tBuXPos (20 mol%). Mixing using freeze-pump-thaw method The mixture was degassed, placed under argon, stirred vigorously and refluxed (165 ℃) for 72 hours. The mixture was returned to room temperature and partitioned between petroleum ether and 2M aqueous NaOH. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield the desired biaryl ether bromide.

L) to the corresponding 4-substituted phenol (1.2-1.5 equiv.) and 1, 4-dibromo (hetero) aryl (1 equiv.) dissolved in DMSO (0.5M) under argon and stirring, K was added2CO3(1.5 equivalents) and the mixture is heated between 80 ℃ and 160 ℃ until complete conversion. The mixture was returned to room temperature and partitioned between petroleum ether and 2M aqueous NaOH. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield the desired bis (hetero) aryl ether bromide.

M) the corresponding bis (hetero) aryl ether bromide (1 eq) was dissolved in anhydrous THF (0.2M) under argon and with stirring, and the resulting solution was cooled to-78 ℃ with a dry ice/acetone bath. N-or t-BuLi (1.1-2.2 equivalents, 1.9-2.5M in hexane or pentane) was then added dropwise and the mixture stirred at this temperature for 30 minutes and then at-50 ℃ until complete consumption of the starting material (monitored by TLC in pentane). The mixture was then cooled to-78 ℃ and a solution of the corresponding electrophile (2 eq, 0.5M) in anhydrous THF was added and the reaction was allowed to slowly return to room temperature over 16 hours. Then the reaction mixture is mixed with saturated NH in AcOEt 4Partition between aqueous Cl solutions, re-extract the aqueous layer twice more, then wash the combined organic phases with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient Petroleum Ether/AcOEt/NEt3) Purification to give the desired compound.

N) to the corresponding protected amine compound dissolved in THF (0.1-0.2M)To this (1 eq) was added HCl (0.5M in methanol, 2-6 eq) and the reaction was stirred until complete. The reaction was then evaporated to dryness to give the desired amine as the hydrochloride salt, or the reaction was incubated with AcOEt and saturated NaHCO3The aqueous solution was partitioned. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt or petroleum ether/AcOEt/AcOH or DCM/MeOH) to give the desired free amine.

O) to the corresponding 4-substituted phenol (1 equivalent) and 4-cyano (hetero) haloaryl (2.5 equivalents) dissolved in DMSO (0.5M) under argon and stirring was added K2CO3(1.5 equivalents) and the mixture is heated between 80 ℃ and 160 ℃ until complete conversion. The mixture was allowed to return to room temperature and partitioned between petroleum ether and 2M aqueous NaOH. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na 2SO4Dried, filtered and concentrated in vacuo. The residue was then purified by flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield the desired bis (hetero) aryl ether cyanide.

P) to the corresponding bis (hetero) aryl ether cyanide (1 eq) dissolved in THF/MeOH (1: 1, 0.1M) was added NaH (1.1 eq) at 0 ℃ under argon and stirring. After 4 hours, the ice bath was removed, cyanamide (1.5 eq) was added and the mixture was stirred for a further 16 hours. The reaction was then partitioned between AcOEt and water. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt/MeOH) to afford the desired amine.

Q) to the corresponding bis (hetero) arylether alcohol, hydroxamic acid or amide (1 equivalent) dissolved in THF/DMF (1: 0 to 2: 8 mixture, 0.2M) under argon with stirring, NaH, NaOAc or Cs is added2CO3(1.2-2 equivalents). After 30 minutes, the alkyl (di) halide or acid chloride (1.2-2 equivalents) is addedIn the case of alkyl (di) bromides KI (1.2 equivalents) was added. The mixture is then stirred at room temperature for a further 16 hours or, in the case of alkyl (di) bromides, at 50 ℃ for a further 16 hours. The reaction was then partitioned between AcOEt and aqueous HCl (1M). The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na 2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt/AcOH) to give the desired amide.

R) to the corresponding bis (hetero) aryl ether amide (1 eq) dissolved in THF (0.2M) at 0 ℃ under argon and stirring was added NaBH4(1.1 equiv.). After 1 hour, the reaction was then quenched with AcOEt and saturated NaHCO3The aqueous solution was partitioned. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then purified by flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to give the desired amide.

S) to the corresponding aldehyde (1 equivalent) in anhydrous THF (0.2M) at 0 ℃ was added the corresponding Wittig reagent (1.5 equivalents). To this stirred mixture was added LiHMDS (1.3 et al, 1M in THF) dropwise. The reaction was stirred to completion and then partitioned between AcOEt and aqueous HCl (1M). The aqueous layer was extracted twice more, and the combined organic phases were then washed with saturated NaHCO3Washed with aqueous solution, then brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then purified by flash chromatography (SiO) 2Gradient petroleum ether/AcOEt) to yield the desired olefin.

T) for adding a methyl group to the amine:

to the corresponding free amine (1 equivalent) in acetonitrile (0.2M) was added formaldehyde (6 equivalents, 37% w/w in water) followed by NaBH3CN (2 equivalents). The reaction mixture was stirred until completion, then AcOEt with saturated NaHCO3Partition between aqueous solutions, re-extract the aqueous layer twice more, then wash the combined organic phases with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then purified by flash chromatography (SiO)2Gradient DCM/MeOH/NEt3) Purification to give the desired compound.

For the addition of isopropyl group to the amine:

five portions of NaBH were added to the corresponding free amine (1 equivalent) in acetone (0.2M) every 15 minutes3CN (10 equivalents) while maintaining the pH at about 5 with acetic acid. The reaction mixture was then stirred in AcOEt with saturated NaHCO3Partitioning between aqueous carbons, the aqueous layer was extracted two more times, and the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient DCM/MeOH/NEt3) Purification to give the desired compound.

U) to the corresponding amine (1 eq) in DCM (0.1M) was added mCPBA (1.2 eq) and the mixture was stirred at room temperature until complete conversion. The reaction mixture was then washed with AcOEt and NaHCO 3Partitioning between aqueous carbons, the aqueous layer was extracted two more times, and the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to afford the desired compound.

V) to the corresponding nitrile (1 eq) in ethanol (0.2M) were added hydroxylamine hydrochloride (2.5 eq) and sodium hydroxide (2.5 eq). The reaction was heated to 80 ℃ overnight, then filtered through celite, concentrated in vacuo, and the residue was purified by flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to afford the desired compound.

W) to the corresponding aldehyde (1 eq) in methanol (0.1M) was added K2CO3(2 eq.) followed by Ohira-Bestmann reagent (1.1 eq.) and the reaction was stirred until completion. The reaction was then partitioned between ethyl acetate and sodium bicarbonate. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) purification to obtainTo the desired alkyne.

Analyzing data

The following compounds were synthesized according to the above protocol and characterized by mass spectrometry (table 93) or NMR (table 94).

Table 93:

table 94:

for illustrative purposes, the synthesis and characterization of the following examples are described in detail.

XPW-05474- (4-butylphenoxy) -N- (methylsulfonyl) benzamide

To 4- (4-butylphenoxy) benzoic acid (104mg, 0.38mmol, 1 equiv.) suspended in stirred toluene (1.85mL, 0.2M) was first added SOCl under argon2(67. mu.L, 0.93mmol, 2.5 equiv.) then DMF (0.3. mu.L, 3.7. mu. mol, 1 mol%) is added and the mixture is heated to 80 ℃ for 3 hours. The reaction mixture was then evaporated to dryness and the resulting residue was again placed under argon and redissolved in THF (1.85mL, 0.2M). To this solution was added trimethylamine (0.13mL, 0.93mmol, 2.5 equiv.), DMAP (0.45mg, 3.7. mu. mol, 1 mol%) and methanesulfonamide (42.3mg, 0.45mmol, 1.2 equiv.) successively, and the suspension was stirred for 16 h. The reaction was then partitioned between AcOEt and aqueous hydrochloric acid (1M). The aqueous layer was extracted twice more, and the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to give 118mg of 4- (4-butylphenoxy) ) -N- (methylsulfonyl) benzamide (88%).

MS:m/z[M-H]-,[C18H20NO4S]-346.11; found 346.24

1H-NMR(300MHz DMSO-d6)δ12.03(s,1H),7.97(d,J=8.9Hz,2H),7.32-7.24(m,2H),7.08-6.97(m,4H),3.36(s,3H),2.66-2.56(m,2H),1.58(tt,J=8.8,6.8Hz,2H),1.41-1.24(m,2H),0.92(t,J=7.3Hz,3H).

13C-NMR(75MHz,DMSO-d6)δ166.1,162.1,153.2,139.4,131.4,130.5,126.3,120.4,117.2,41.8,34.6,33.6,22.2,14.3.

XPW-2890(6- (4-cyclohexylphenoxy) pyridin-3-yl) (morpholino) methanone

To 6- (4-Cyclohexylphenoxy) nicotinic acid (50mg, 0.17mmol, 1 equiv.) suspended in stirred toluene (0.85mL, 0.2M) was first added SOCl under argon2(31 μ L, 0.43mmol, 2.5 equiv.) DMF (0.14 μ L, 1.7 μmol, 1 mol%) was then added and the mixture was heated to 80 ℃ for 3 hours. The reaction mixture was then evaporated to dryness and the resulting residue was again placed under argon and redissolved in THF (0.85mL, 0.2M). To this solution was added trimethylamine (9.4. mu.L, 0.68mmol, 2.5 equiv.), DMAP (0.2mg, 1.7. mu. mol, 1 mol%) and morpholine (20. mu.L, 0.23mmol, 1.5 equiv.) in that order, and the suspension was stirred for 16 h. The reaction was then partitioned between AcOEt and aqueous HCl (1M). The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then purified by flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield 45mg of (6- (4-cyclohexylphenoxy) pyridin-3-yl) (morpholino) methanone (71%).

MS:m/z[M+H]+,[C22H27N2O3]+367.20; found 367.32

1H-NMR(300MHz,CDCl3)δ8.20(d,J=2.3Hz,1H),7.73(dd,J=8.6,2.2Hz,1H),7.18(d,J=7.7Hz,2H),7.06-6.92(m,2H),6.87(d,J=8.5Hz,1H),3.63(s,8H),2.50-2.37(m,1H),1.89-1.62(m,5H),1.38-1.08(m,5H).

13C-NMR(75MHz,CDCl3)δ167.7,164.7,151.2,146.6,145.1,139.5,128.1,125.5,121.0,111.3,66.8,53.4,44.0,34.5,26.9,26.1.

XPW-0636N-cyano-4- (4-cyclohexylphenoxy) benzamide

To 4- (4-Cyclohexylphenoxy) benzoic acid (60mg, 0.2mmol, 1 equiv.) suspended in stirred toluene (0.8mL, 0.2M) was first added SOCl under argon2(37. mu.L, 0.0.5mmol, 2.5 equiv.) DMF (0.15. mu.L, 2.0. mu. mol, 1 mol%) was then added and the mixture was heated to 80 ℃ for 3 hours. The reaction mixture was then evaporated to dryness and the resulting residue was again placed under argon and redissolved in THF (0.8mL, 0.2M). To this solution was added trimethylamine (57 μ L, 0.5mmol, 2.5 equiv.), DMAP (0.24mg, 2.0 μmol, 1 mol%) and cyanamide (12.6mg, 0.3mmol, 1.5 equiv.) in that order, and the suspension was stirred for 16 h. The reaction was then partitioned between AcOEt and aqueous hydrochloric acid (1M). The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield 15.4mg of N-cyano-4- (4-cyclohexylphenoxy) benzamide (24%).

MS:m/z[M-H]-,[C20H19N2O2]-319.15; found 319.28

1H-NMR,(300MHz,CDCl3)δ7.93(d,J=8.8Hz,2H),7.31(d,J=8.5Hz,2H),7.23-6.85(m,4H),3.37(brs,1H),2.59-2.51(m,1H)1.91-1.63(m,5H),1.57-1.12(m,5H).

13C-NMR,(75MHz,CDCl3)δ166.5,161.8,152.7,144.2,130.8,128.4,124.8,119.9,117.1,110.0,43.1,34.0,26.3,25.5.

XPW-0675N-hydroxy-4- (4- (2-methoxyethyl) phenoxy) -N-methylbenzamide

To 4- (4- (2-methoxyethyl) phenoxy) benzoic acid (125mg, 0.46mmol, 1 eq) suspended in stirred toluene (2.3mL, 0.2M) was first added SOCl under argon2(84. mu.L, 1.15mmol, 2.5 equiv.) DMF (0.35. mu.L, 4.6. mu. mol, 1 mol%) was then added and the mixture was heated to 80 ℃ for 3 hours. The reaction mixture was then evaporated to dryness and the resulting residue was again placed under argon and redissolved in THF (2.3mL, 0.2M). To this solution was added sequentially trimethylamine (144 μ L, 1.13mmol, 2.5 equiv.), DMAP (0.56mg, 4.6 μmol, 1 mol%) and N-methylhydroxylamine hydrochloride (58m8, 0.69mmol, 1.5 equiv.) and the suspension was stirred for 16 h. The reaction was then partitioned between AcOEt and aqueous hydrochloric acid (1M). The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield 103.1mg of N-hydroxy-4- (4- (2-methoxyethyl) phenoxy) -N-methylbenzamide (74%).

MS:m/z[M+H]+,[C17H20NO4]+302.14; found 302.17

1H-NMR,(300MHz,CDCl3)δ10.00(s,1H),7.73-7.61(m,2H),7.37-7.20(m,2H),7.07-6.82(m,4H),3.55(t,J=6.8Hz,2H),3.26(s,3H),3.25(s,3H),2.82(t,J=6.8Hz,2H).

13C-NMR,(75MHz,CDCl3)δ168.6,159.2,154.3,135.6,131.2,130.9,129.5,119.9,117.1,73.2,58.3,37.9,35.1.

XPW-0832N' -cyano-6- (4-cyclohexylphenoxy) nicotinamide

To 6- (4-cyclohexylphenoxy) nicotinonitrile (41mg, 0.15mmol, 1 equiv.) dissolved in THF/MeOH (1: 1, 1.5mL, 0.1M) was added NaH (6.6mg, 0.17mmol, 1.1 equiv., 60% in oil) at 0 deg.C under argon and stirring. After 4 h, the ice bath was removed, cyanamide (9.5mg, 0.23mmol, 1.5 eq) was added and the mixture was stirred for a further 16 h. The reaction was then partitioned between AcOEt and water. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt/MeOH) to yield 32mg of N' -cyano-6- (4-cyclohexylphenoxy) nicotinimide (67%).

MS:m/z[M+H]+,[C20H22N3O]+320.18; found 320.27

1H-NMR(300MHz,DMSO-d6)δ7.79(d,J=8.9Hz,2H),7.25-7.17(m,2H),7.07-6.94(m,4H),3.83(s,2H),2.63-2.41(m,1H),1.94-1.55(m,5H),1.48-1.16(m,5H).

13C-NMR(75MHz,DMSO-d6)δ152.9,144.9,129.3,129.3,128.4,124.4,120.1,117.4,116.4,113.3,77.5,44.0,34.6,26.9,26.1.

XPW-0902: n- (1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoroethyl) -2-methylpropane-2-sulfinamide

(3r, 5r, 7r) -1- (4- (4-bromophenoxy) phenyl) adamantane (100mg, 0.26mmol, 1 equiv.) was dissolved in anhydrous THF (1.3mL, 0.2M) under argon with stirring, and the resulting solution was cooled to-78 ℃ with a dry ice/acetone bath. Then dropwise adding nBuLi (0.11mL, 0.26mmol, 1.0 equiv, 2.3M in pentane), and the mixture was stirred at this temperature for 30 minutes, then at-50 ℃ for another 30 minutes. The mixture was then cooled back to-78 ℃. A solution of 2-methyl-N- (2, 2, 2-trifluoroethylene) propane-2-sulfinamide (79mg, 0.39mmol, 1.5 equivalents) in THF (0.39mL, 1M) was added dropwise, the reaction stirred for 1 hour and then allowed to slowly return to room temperature overnight. Then the reaction mixture is mixed with saturated NH in AcOEt4The partition was made between the Cl aqueous solutions. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield 81mg of N- (1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoroethyl) -2-methylpropane-2-sulfinamide (62%).

MS:m/z[M+H]+,[C28H35F3NO2S]+506.23; found 506.70

1H NMR(400MHz,CDCl3)δ7.38-7.32(m,4H),7.03-6.95(m,4H),4.83(qd,J=7.1,3.5Hz,1H),3.88(d,J=3.5Hz,1H),2.10(p,J=3.5Hz,3H),1.91(d,J=2.9Hz,6H),1.83-1.68(m,6H),1.23(s,9H).

13C NMR(101MHz,CDCl3)δ159.29,153.59,147.36,130.78,126.31,125.24,124.53(q,J=281.3Hz),119.39,117.93,59.87(q,J=30.4Hz),56.31,43.33,36.76,35.91,28.95,22.41.

19F NMR(376MHz,CDCl3)δ-74.56(d,J=7.2Hz).

XPW-30523- (6- (4- ((adamantan-1-yl) phenoxy) pyridin-3-yl) oxetan-3-amine

To a solution of N- (3- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) oxetan-3-yl) -2-methylpropane-2-sulfinamide (31mg, 0.065mmol, 1 eq) in THF (0.32mL, 0.2M) was added HCl (0.8mL, 0.5M in MeOH, 6 eq) and the reaction was stirred until completion. The reaction was then incubated with AcOEt and NaHCO 3The aqueous solution (1M) was partitioned. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient DCM/MeOH) to yield 17.4mg of 3- (6- (4- ((adamantan-1-yl) phenoxy) pyridin-3-yl) oxetan-3-amine (71%).

MS:m/z[M+H]+,[C24H29N2O2]+377.22; found 377.34

1H-NMR,(300MHz,DMSO-d6)δ8.33(dd,J=2.6,0.7Hz,1H),8.02(dd,J=8.6,2.6Hz,1H),7.50-7.36(m,2H),7.14-6.96(m,3H),4.70(d,J=6.3Hz,2H),4.64(d,J=6.3Hz,2H),2.65(brs,2H),2.18-1.99(m,3H),1.96-1.83(m,6H),1.75(s,6H).

13C-NMR,(75MHz,DMSO-d6)δ162.6,152.3,147.4,144.9,138.0,136.4,126.4,121.0,111.3,85.7,57.6,43.2,36.6,35.9,28.8.

XPW-4642N- (1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoroethyl) -N, 2-dimethylpropane-2-sulfinamide

To a solution of N- (1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoroethyl) -2-methylpropane-2-sulfinamide (360mg, 0.71mmol, 1 eq) in THF (72mL, 0.1M) was added Cs2CO3(464mg, 1.42mmol, 2 equiv.). After 30 min, iodomethane (0.089mL, 1.42mmol, 2 equiv.) was added. The mixture was then heated at 50 ℃ for a further 16 hours. The reaction was then partitioned between AcOEt and water. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then purified by flash chromatography (SiO2, gradient petroleum ether/AcOEt) to yield 300mg of N- (1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoroethyl) -N, N-dimethylpropane-2-sulfinamide (81%).

MS:m/z[M+H]+,[C29H37F3NO2S]+520.25; found 520.69

1H NMR(400MHz,CDCl3)δ7.42(d,J=8.6Hz,2H),7.37-7.32(m,2H),7.02-6.96(m,4H),4.92(q,J=8.6Hz,1H),2.71(s,3H),2.11(s,3H),1.91(d,J=2.9Hz,6H),1.85-1.71(m,6H),1.20(s,9H).

19F NMR(376MHz,CDCl3)δ-67.23(d,J=8.2Hz).

XPW-0028: 1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoro-N-methylethan-1-amine hydrochloride

To N- (1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoroethyl) -N, 2-dimethylpropane-2-sulfinamide (300mg, 0.58mmol, 1 eq) dissolved in THF (5.8mL, 0.1M) was added HCl (2.3mL, 1.15mmol, 0.5M in MeOH, 2 eq) and the reaction was stirred until completion. The reaction was then evaporated to dryness to give 255mg of 1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoro-N-methylethan-1-amine hydrochloride (quantitative).

MS:m/z[M+H]+,[C25H29F3NO]+416.22; found 416.69

1H NMR(400MHz,CDCl3)δ11.04(brs,2H),7.62(d,J=8.5Hz,2H),7.43-7.33(m,2H),7.08(d,J=8.4Hz,2H),7.05-6.96(m,2H),4.71-4.36(m,1H),2.69(s,3H),2.14(s,3H),1.94(d,J=2.9Hz,6H),1.88-1.73(m,6H).

XPW-0182:

N- (1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoroethyl) -N-methylhydroxylamine

To 1- (4- (4- (3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoro-N-methylethane-1-amine hydrochloride (50mg, 0.12mmol, 1 equivalent) in DCM (1.2mL, 0.1M) was added mCPBA (35mg, 0.144mmol, 70%, 1.2 equivalents), and the mixture was stirred at room temperature for 30 min. The reaction mixture was then stirred in AcOEt with saturated NaHCO 3Partitioning between aqueous carbons, the aqueous layer was extracted two more times, and the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield 44.5mg of N- (1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoroethyl) -N-methylhydroxylamine (86%).

1H NMR(400MHz,CDCl3)δ7.41-7.33(m,4H),7.05-6.96(m,4H),4.32(q,J=7.6Hz,1H),2.87(s,3H),2.16-2.07(m,3H),1.92(d,J=2.9Hz,6H),1.85-1.67(m,6H).

19F NMR(376MHz,CDCl3)δ-67.82(d,J=7.5Hz).

13C NMR(101MHz,CDCl3)δ159.24,153.61,147.38,131.35,126.33,124.40,124.18(q,J=282.5Hz),119.37,117.73,74.70(q,J=29.0Hz),50.31,43.34,36.76,35.92,28.96.

XPW-0042: 1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoro-N, N-dimethylethan-1-amine

To 1- (4- (4- (3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoro-N-methylethane-1-amine hydrochloride (20mg, 0.05mmol, 1 eq) in acetonitrile (0.25mL, 0.2M) was added formaldehyde (0.025mL, 0.29mmol, 6 eq, 37% w/w in water), followed by NaBH3CN (6.1mg, 0.10mmol, 2 equiv.). The reaction mixture was stirred until complete, then AcOEt with saturated NaHCO3Partitioning between aqueous carbons, the aqueous layer was extracted two more times, and the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient DCM/MeOH) to give 15m8 of 1- (4- (4- ((3r, 5r, 7r) -adamantan-1-yl) phenoxy) phenyl) -2, 2, 2-trifluoro-N, N-dimethylethan-1-amine (75%).

MS:m/z[M+H]+,[C26H31F3NO]+430.27; found 430.71

1H NMR(400MHz,CDCl3)δ7.39-7.28(m,4H),7.04-6.93(m,4H),3.95(q,J=8.7Hz,1H),2.36(s,6H),2.10(s,3H),1.91(d,J=2.9Hz,6H),1.85-1.69(m,6H).

19F NMR(376MHz,CDCl3)δ-67.30.

XPW-03142- (4- (4-cyclohexylphenoxy) phenyl) -2- (dimethylamino) acetonitrile

To a stirred solution of 4- (4-cyclohexylphenoxy) benzaldehyde (50mg, 0.18mmol, 1 eq) in toluene (0.90.9mL, 0.2M) under argon was added dimethylamine (0.18mL, 0.36mmol, 2 eq) followed by TMSCN (0.05mL, 0.36mmol, 2 eq) and the reaction stirred for 16 h. The reaction was then quenched with AcOEt and saturated NaHCO3The aqueous solution was partitioned. The aqueous layer was extracted twice more, then the combined organic phases were washed with brine, over Na2SO4Dried, filtered and concentrated in vacuo. The residue was then passed through flash chromatography (SiO)2Gradient petroleum ether/AcOEt) to yield 29mg of 2- (4- (4-cyclohexylphenoxy) phenyl) -2- (dimethylamino) acetonitrile (48%).

MS:m/z[M+H]+,[C22H27N2O]+335.21; found 335.31

1H-NMR,(300MHz,CDCl3)δ7.46-7.32(m,2H),7.16-7.07(m,2H),6.99-6.82(m,4H),4.75(s,1H),2.43(ddt,J=11.7,8.2,5.0Hz,1H),2.27(s,6H),1.87-1.61(m,5H),1.42-1.24(m,5H).

13C NMR(75MHz,CDCl3)δ158.5,154.2,143.8,129.3,128.1,119.3,118.3,115.0,62.5,43.9,41.7,34.6,26.9,26.1。

266页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:用于治疗和预防乙型肝炎病毒疾病的新型三环化合物

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!