HK1166280B - Composition for treatment of tuberculosis - Google Patents
Composition for treatment of tuberculosis Download PDFInfo
- Publication number
- HK1166280B HK1166280B HK12107097.3A HK12107097A HK1166280B HK 1166280 B HK1166280 B HK 1166280B HK 12107097 A HK12107097 A HK 12107097A HK 1166280 B HK1166280 B HK 1166280B
- Authority
- HK
- Hong Kong
- Prior art keywords
- phenyl
- formula
- dichloromethane
- vacuo
- substituted
- Prior art date
Links
Description
Technical Field
The present invention relates to pharmaceutical compositions for the treatment of tuberculosis and related diseases.
Background
Up to nine million people infect tuberculosis every year, and currently there are fifty million people infected with tubercle bacillus (WHO, Fact sheet No.104, March 2007) that is resistant to both the first-line drug isoniazid and rifampicin. Ethionamide (2-ethylthionicotinamide, 2-ethylpyrimidine-4-thiocarboxamide), a structural analogue of isoniazid, is the last line-of-defense drug for the treatment of multiple drug resistant tuberculosis (MDR-TB) at present. During 35 years of its clinical use ethionamide fortunately developed little cross-resistance with isoniazid, since both prodrugs had to be activated by different mycobacterial enzymes to develop their antibacterial activity. Ethionamide remains administered at hepatotoxic doses, as EthR inhibits ethA, i.e. catalytically activates the pro-drug ethionamide to the monooxygenase of the antimycobacterial nicotinamide adenine dinucleotide derivative. Acceptable concentrations in blood require up to 1 g/day (holdesses, m.r., Clin Pharmacokinet1984, 9, 511-44), which are associated with serious side effects, including neurotoxicity and lethal hepatotoxicity.
WO 2008/003861 describes compounds having a potentiating effect on ethionamide activity in the treatment of tuberculosis and related diseases. The present inventors have found that 2-phenylethylbutyrate, an approved food additive, and related compounds potentiate the activity of thioamides or thioureas such as ethionamide in the treatment of tuberculosis (WO 2009/080432).
Summary of The Invention
The present invention relates to a compound comprising a compound of the following formula 1
Wherein R is1Is unsubstituted phenyl or phenyl substituted with 1, 2 or 3 substituents selected from: c1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy, C1-C6-alkylcarbonyl group, C1-C6-alkylcarbonyloxy, C1-C6Alkylthio, nitro, amino, C1-C6-alkylamino, di-C1-C6Alkylamino, pyrrolidino, piperidino, morpholino, C1-C6-alkylcarbonylamino and halogen;
unsubstituted 2-, 3-or 4-pyridyl or 2-, 3-or 4-pyridyl substituted with 1 or 2 substituents selected from the group consisting of: c1-C6-alkyl, trifluoromethyl, C1-C6Alkoxy, nitro, amino, C1-C6-alkylamino, di-C1-C6-alkylamino radical, C1-C6-alkylcarbonylamino and halogen; or
An unsubstituted indolyl group or an indolyl group substituted with 1 or 2 substituents selected from the group consisting of: c1-C6-alkyl, trifluoromethyl, C1-C6Alkoxy, nitro, amino, C1-C6-alkylamino, di-C1-C6-alkylamino radical, C1-C6-alkylcarbonylamino and halogen;
R2is (CH)2)nWherein n is 0, 1, 2, 3 or 4;
R3is (CH)2)mR3AWherein m is 0, 1, 2, 3 or 4;
R3Ais CH3、CH(CH3)2、C(CH3)3、OCH3、OH、OR3BC ≡ CH, C ≡ N, unsubstituted phenyl or substituted by 1, 2 or 3 substituents selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy, C1-C6-alkylcarbonyl group, C1-C6-alkylcarbonyloxy, C1-C6Alkylthio, nitro, amino, C1-C6-alkylamino, di-C1-C6Alkylamino, pyrrolidino, piperidino, morpholino, C1-C6-phenyl substituted with substituents of alkylcarbonylamino and halogen;
unsubstituted 2-or 3-furyl or substituted by 1 or 2 radicals selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy and halogen substituted 2-or 3-furyl; or
Unsubstituted 2-or 3-thienyl or substituted by 1 or 2 groups selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy and halogen substituted 2-or 3-thienyl; and wherein
R3BIs unsubstituted phenyl or phenyl substituted with 1, 2 or 3 substituents selected from: c1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy, C1-C6-alkylcarbonyl group, C1-C6-alkylcarbonyloxy, C1-C6Alkylthio, nitro, amino, C1-C6-alkylamino, di-C1-C6Alkylamino, pyrrolidino, piperidino, morpholino, C1-C6-alkylcarbonylamino and halogen;
a is selected from
Wherein-represents the linkage C (═ X)2)-R3And → represents the connection R2-R1A bond of (a);
or a and-C (═ X)2)-R3Together form a ring
Wherein → represents the connection R2-R1A bond of (a);
X1is O, S, NH, N (CH)3) Or CH2(ii) a And
X2is O, S or NH;
and a compound of the following formula 2
Wherein R is4Is optionally substituted phenyl, optionally substituted pyridyl, optionally substituted indolyl, -NR7R8or-NH-N ═ CH-R9;
R5Is hydrogen, C1-C6-an alkyl group, an optionally substituted phenyl group, an optionally substituted pyridyl group, or a sugar residue;
R6is hydrogen or C1-C6-alkyl, or R5And R6Together with the N-atom to which they are bonded are pyrrolidine, piperidine or morpholine;
R7is hydrogen, C1-C6-an alkyl group, an optionally substituted phenyl group, an optionally substituted pyridyl group, or a sugar residue;
R8is hydrogen or C1-C6-alkyl, or R7And R8Together with the N-atom to which they are bonded are pyrrolidine, piperidine or morpholine; and
R9is optionally substituted phenyl.
Particularly preferred is a composition comprising a compound of formula 1 and a compound of formula 2, the compound of formula 2 being selected from: ethionamide, pentoxyphenylthiourea (isoxyl) or N-arabinofuranosyl-N' - [ p- (isopentyloxy) phenyl ] -thiourea or thiosemicarbazide, in particular ethionamide.
The present invention also relates to novel compounds of formula 1 as defined herein above.
Likewise, the invention relates to the use of pharmaceutical compositions comprising compounds of formula 1 and compounds of formula 2 (e.g., ethionamide) in the treatment of tuberculosis and related diseases, and methods in which pharmaceutical compositions comprising compounds of formula 1 and compounds of formula 2 (e.g., ethionamide) are used to treat tuberculosis and related diseases.
Detailed Description
The present invention relates to a pharmaceutical composition comprising a compound of formula 1 and a compound of formula 2 as defined hereinafter. The present invention is based on the discovery that: compounds that inhibit the binding of EthR to the ethA promoter greatly increase the activity of ethionamide in treating tuberculosis, but also increase the activity of other thioamides or thioureas. Comprehensive networks in mammalian cells can be used to screen for sites (O) that inhibit EthR-VP16 binding to its cognate operatorethR-Phsp70min) And thereby transactivating the expression of the reporter gene (SEAP). The active compound removes the EthR protein from the hybrid promoter, thereby further inhibiting the expression of the reporter gene SEAP. This test proves to be very accurate in identifying the active compound and the minimum inhibitory concentration determined is very good in comparison with the respective results of the ELISA screening and the screening of the microbial pathogensGood agreement. Thus, the results of the mammalian comprehensive screening network demonstrate a high degree of compatibility with endogenous resistance-modulating networks observed in pathogenic settings (WO 2009/080432; Weber et al, PNAS 2008, 105, 9994-8).
It has now been found that the compounds of formula 1 act as compounds inhibiting the binding of EthR to the ethA promoter,
wherein R is1Is unsubstituted phenyl or phenyl substituted with 1, 2 or 3 substituents selected from: c1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy, C1-C6-alkylcarbonyl group, C1-C6-alkylcarbonyloxy, C1-C6Alkylthio, nitro, amino, C1-C6-alkylamino, di-C1-C6Alkylamino, pyrrolidino, piperidino, morpholino, C1-C6-alkylcarbonylamino and halogen;
unsubstituted 2-, 3-or 4-pyridyl or 2-, 3-or 4-pyridyl substituted with 1 or 2 substituents selected from the group consisting of: c1-C6-alkyl, trifluoromethyl, C1-C6Alkoxy, nitro, amino, C1-C6-alkylamino, di-C1-C6-alkylamino radical, C1-C6-alkylcarbonylamino and halogen; or
An unsubstituted indolyl group or an indolyl group substituted with 1 or 2 substituents selected from the group consisting of: c1-C6-alkyl, trifluoromethyl, C1-C6Alkoxy, nitro, amino, C1-C6-alkylamino, di-C1-C6-alkylamino radical, C1-C6-alkylcarbonylamino and halogen;
R2is (CH)2)nWherein n is 0, 1, 2, 3 or 4;
R3is (CH)2)mR3AWherein m is 0, 1, 2, 3 or 4;
R3Ais CH3、CH(CH3)2、C(CH3)3、OCH3、OH、OR3BC ≡ CH, C ≡ N, unsubstituted phenyl or substituted by 1, 2 or 3 substituents selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy, C1-C6-alkylcarbonyl group, C1-C6-alkylcarbonyloxy, C1-C6Alkylthio, nitro, amino, C1-C6-alkylamino, di-C1-C6Alkylamino, pyrrolidino, piperidino, morpholino, C1-C6-phenyl substituted with substituents of alkylcarbonylamino and halogen;
unsubstituted 2-or 3-furyl or substituted by 1 or 2 radicals selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy and halogen substituted 2-or 3-furyl; or
Unsubstituted 2-or 3-thienyl or substituted by 1 or 2 groups selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy and halogen substituted 2-or 3-thienyl; and wherein
R3BIs unsubstituted phenyl or phenyl substituted with 1, 2 or 3 substituents selected from: c1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy, C1-C6-alkylcarbonyl group, C1-C6-alkylcarbonyloxy, C1-C6Alkylthio, nitro, amino, C1-C6-alkylamino, di-C1-C6Alkylamino, pyrrolidino, piperidino, morpholino, C1-C6-alkylcarbonylamino and halogen;
a is selected from
Wherein-represents the linkage C (═ X)2)-R3And → represents the connection R2-R1A bond of (a);
or a and-C (═ X)2)-R3Together form a ring
Wherein → represents the connection R2-R1A bond of (a);
X1is O, S, NH, N (CH)3) Or CH2(ii) a And
X2is O, S or NH;
accordingly, the present invention relates to a pharmaceutical composition comprising such a compound of formula 1 together with a compound of formula 2.
In the partial structures a-i of A presented, the bond represented as-or → which is not located at a particular ring position indicates that the bond may be attached at any position in the ring which represents a carbon atom which has not been completely substituted. For example, in part structure f, with R3May be located other than with ═ X2At any ring carbon position of the carbon (b).
The compounds of formula 2 in the pharmaceutical compositions of the present invention are those compounds
Wherein R is4Is optionally substituted phenyl, optionally substituted pyridyl, optionally substituted indolyl, -NR7R8or-NH-N ═ CH-R9;
R5Is hydrogen, C1-C6-an alkyl group, an optionally substituted phenyl group, an optionally substituted pyridyl group, or a sugar residue;
R6is hydrogen or C1-C6-alkyl, or R5And R6Together with the N-atom to which they are bonded are pyrrolidine, piperidine or morpholine;
R7is hydrogen, C1-C6-an alkyl group, an optionally substituted phenyl group, an optionally substituted pyridyl group, or a sugar residue;
R8is hydrogen or C1-C6-alkyl, or R7And R8Together with the N-atom to which they are bonded are pyrrolidine, piperidine or morpholine; and
R9is optionally substituted phenyl.
Unless otherwise indicated, general terms used in the context of the present disclosure preferably have the following meanings.
Alkyl radicals, especially C1-C6Alkyl radicals, e.g. C1-C4-an alkyl group. C1-C4Alkyl is methyl, ethyl, propyl, such as n-propyl or isopropyl, or butyl, such as n-butyl, isobutyl or tert-butyl. C1-C6Alkyl is methyl, ethyl, propyl or butyl as described above, or also pentyl, for example n-pentyl or isopentyl, or hexyl, for example n-hexyl or isohexyl.
Optionally substituted phenyl is unsubstituted phenyl or phenyl substituted with 1, 2 or 3 substituents selected from the group consisting of: c1-C6Alkyl radicals, e.g. methyl, trifluoromethyl, C1-C6Alkoxy, e.g. methoxy, ethoxy or isopentoxy, C1-C6-alkylcarbonylRadicals, e.g. acetyl, C1-C6Alkylcarbonyloxy, e.g. acetoxy, C1-C6Alkylthio, e.g. methylthio, nitro, amino, C1-C6Alkylamino, e.g. methylamino or ethylamino, di-C1-C6Alkylamino, e.g. dimethylamino or diethylamino, pyrrolidino, piperidino, morpholino, C1-C6Alkylcarbonylamino, such as acetylamino, and halogen. Halogen is fluoro, chloro, bromo or iodo, especially fluoro or chloro. Preferred optionally substituted phenyl is phenyl or phenyl substituted by 1 or 2 of the substituents mentioned, especially one of the substituents mentioned which is located in ortho, meta or para position, preferably meta or para position. For example, optionally substituted phenyl is phenyl, methyl-or dimethylphenyl, trifluoromethylphenyl, methoxyphenyl, ethoxyphenyl, acetoxyphenyl, nitrophenyl, dinitrophenyl, aminophenyl, methylaminophenyl, dimethylaminophenyl, fluorophenyl, chlorophenyl or dichlorophenyl.
Optionally substituted pyridyl is 2-, 3-or 4-pyridyl, unsubstituted or substituted with 1 or 2 substituents selected from the group consisting of: c1-C6Alkyl radicals, e.g. methyl or ethyl, trifluoromethyl, C1-C6Alkoxy, e.g. methoxy or ethoxy, nitro, amino, C1-C6Alkylamino, e.g. methylamino or ethylamino, di-C1-C6Alkylamino, e.g. dimethylamino or diethylamino, C1-C6Alkylcarbonylamino, such as acetylamino, and halogen. Halogen is fluoro, chloro, bromo or iodo, especially fluoro or chloro.
Optionally substituted indolyl is 1H-2-, 3-, 4-, 5-, 6-or 7-indolyl, unsubstituted or substituted with 1 or 2 substituents selected from the group consisting of: c1-C6Alkyl radicals, e.g. methyl or ethyl, trifluoromethyl, C1-C6Alkoxy, e.g. methoxy or ethoxy, nitro, amino, C1-C6Alkylamino, e.g. methylamino or ethylamino, di-C1-C6Alkylamino, e.g. dimethylamino or diethylamino, C1-C6Alkylcarbonylamino, such as acetylamino, and halogen.
Optionally substituted furyl is 2-or 3-furyl, unsubstituted or substituted by 1 or 2 substituents selected from the group consisting of: c1-C6Alkyl radicals, e.g. methyl or ethyl, trifluoromethyl, C1-C6Alkoxy, for example methoxy or ethoxy, and halogen, such as fluoro, chloro, bromo or iodo, especially fluoro or chloro.
Optionally substituted thienyl is 2-or 3-furyl, unsubstituted or substituted with 1 or 2 substituents selected from the group consisting of: c1-C6Alkyl radicals, e.g. methyl or ethyl, trifluoromethyl, C1-C6Alkoxy, for example methoxy or ethoxy, and halogen, such as fluoro, chloro, bromo or iodo, especially fluoro or chloro.
The sugar residue is
An L-or D-furanosyl group of formula 3 selected from the group consisting of aldopentose, arabinose, lyxose, ribose and xylose;
an L-or D-hexofuranosyl (hexofuranosyl) of formula 4 selected from the group consisting of aldohexose, allose, altrose, glucose, mannose, gulose, idose, galactose and talose;
an L-or D-hexofuranose group of formula 5 selected from the group consisting of hexulose, fructose, psicose, sorbose, and tagatose;
an L-or D-pyranosyl group of formula 6, selected from aldohexose, allose, altrose, glucose, mannose, gulose, idose, galactose and talose; or
An L-or D-pyranosyl group of formula 7 selected from the group consisting of hexulose, fructose, psicose, sorbose and tagatose;
wherein 1, 2, 3 or 4 of the hydroxyl groups may be methylated, benzylated or acetylated, or one of the hydroxyl groups may be replaced by hydrogen, halogen, methylamino, ethylamino or acetamido.
As mentioned above, the compounds of formula 1 have valuable properties. These properties were determined using the following tests:
clonal populations stably expressing pWW489 and pWW491 cell lines (Clonal posts) (Weber et al, PNAS 2008, 105, 9994-8) were treated with different amounts of each compound shown in table 1. After 48 hours of compound or solvent addition (w/o), the supernatants containing the secreted reporter gene SEAP were analyzed. The SEAP level in the untreated condition (w/o) was set to 100%. Table 1 summarizes the relative EthR-VP16 activity of SEAP expression reflexes in the presence of various compounds ranging between 500nMol (0.5) and 300. mu. Mol (300) at different concentrations.
Growth of mycobacterium tuberculosis (m. tuberculosis) was significantly disrupted in the presence of ethionamide due to EthA-mediated conversion of the drug to an antimycobacterial nicotinamide adenine dinucleotide derivative. EthR-mediated inhibition of ethaa transcription requires a rather high clinical dose of ethionamide (up to 1 g/day, Holdiness, m.r., Clin Pharmacokinet1984, 9, 511-44), which is associated with severe side effects, including neurotoxicity and fatal hepatotoxicity, however such high doses are often not sufficient to achieve minimal inhibition levels in the bloodstream. Thus, the 2-phenylethylbutyrate-triggered cleavage of EthR in the ethA promoter leading to ethA a derepression can increase the sensitivity of mycobacteria to ethionamide-based therapies. In the presence of sub-inhibitory concentrations of ethionamide (0.16 and 5. mu.g/ml), this concentration is readily achieved by therapeutic doses (c)max[250mg oral administration]=2μg/ml,t1/22H), the growth of mycobacterium tuberculosis H37Rv was inhibited dose-dependently by 0.5-40.5 μ Mol of the following three compounds: 2- (4-fluorophenoxy) -1- (3-phenylpyrrolidin-1-yl) ethan-1-one (example)15 compounds), 3-phenyl-1- (3-phenylpyrrolidin-1-yl) propan-1-one (example 16 compound) and 2-phenyl-1- (3-phenylpyrrolidin-1-yl) ethan-1-one (example 17 compound), see table 2. Since these three compounds do not exhibit any growth inhibitory effect by themselves, they must act synergistically with ethionamide to kill pathogenic bacteria.
TABLE 1The selected compound pairEthREthR-VP 16-mediated dose response curves in HEK-SEAP cells
TABLE 2Synergistic effect of ethionamide with the compounds of examples 15, 16 and 17 on growth inhibition by Mycobacterium tuberculosis laboratory strain H37Rv
(R: resistance, no effect of the combined drug application; I: intermediate; S: sensitive)
Preferably the present invention relates to pharmaceutical compositions comprising a compound of formula 1, preferably specified below, and a compound of formula 2, preferably specified below, and also to novel compounds of formula 1, preferably specified below, per se.
In particular, the present invention relates to a pharmaceutical composition comprising
A compound of formula 1, wherein
R1Is unsubstituted phenyl or phenyl substituted with 1, 2 or 3 substituents selected from: c1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy, C1-C6-alkylcarbonyl group, C1-C6-alkylcarbonyloxy, C1-C6Alkylthio, nitro, amino, C1-C6-alkylamino, di-C1-C6Alkylamino, pyrrolidino, piperidino, morpholino, C1-C6-alkylcarbonylamino and halogen;
unsubstituted 2-, 3-or 4-pyridyl or 2-, 3-or 4-pyridyl substituted with 1 or 2 substituents selected from the group consisting of: c1-C6-alkyl, trifluoromethyl, C1-C6Alkoxy, nitro, amino, C1-C6-alkylamino, di-C1-C6-alkylamino radical, C1-C6-alkylcarbonylamino and halogen; or
Unsubstituted indolyl or indolyl substituted with 1 or 2 substituents selected from: c1-C6-alkyl, trifluoromethyl, C1-C6Alkoxy, nitro, amino, C1-C6-alkylamino, di-C1-C6-alkylamino radical, C1-C6-alkylcarbonylamino and halogen;
R2is (CH)2)nWherein n is 0, 1, 2, 3 or 4;
R3is (CH)2)mCH3、(CH2)mOCH3、(CH2)mOH、(CH2)mC ≡ CH or (CH)2)mC≡N,
Wherein m is 0, 1, 2 or 3;
a is selected from the partial structures a-d and f-i as defined above;
X1is O, S, NH, N (CH)3) Or CH2(ii) a And
X2is O, S or NH;
and compounds of formula 2 as defined above, for example compounds selected from ethionamide, pentyloxyphenylurea or N-arabinofuranosyl-N' - [ p- (isopentyloxy) phenyl ] -thiourea or thiosemicarbazide, especially ethionamide.
Preferred compounds of formula 1 in the pharmaceutical compositions of the present invention are those compounds wherein R is1Is unsubstituted phenyl or phenyl monosubstituted by 1 substituent selected from: trifluoromethyl, C1-C3-alkoxy, C1-C3-alkylcarbonyl group, C1-C3Alkylthio, nitro, amino, C1-C3-alkylamino and halogen; unsubstituted 2-, 3-or 4-pyridyl or 2-, 3-or 4-pyridyl monosubstituted by 1 substituent selected from the group consisting of: trifluoromethyl, C1-C3Alkoxy, nitro, amino, C1-C3-alkylamino and halogen; or an unsubstituted indolyl group or an indolyl group monosubstituted with 1 substituent selected from: trifluoromethyl, C1-C3Alkoxy, nitro, amino, C1-C3-alkylamino and halogen;
R2is (CH)2)nWherein n is 0, 1 or 2;
R3is (CH)2)mR3AWherein m is 0, 1, 2, 3 or 4;
R3Ais CH3、CH(CH3)2、C(CH3)3、OCH3、OH、OR3BC ≡ CH, C ≡ N, unsubstituted phenyl or substituted by 1, 2 or 3 substituents selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy radicalBase, C1-C6-alkylcarbonyl group, C1-C6-alkylcarbonyloxy, C1-C6Alkylthio, nitro, amino, C1-C6-alkylamino, di-C1-C6Alkylamino, pyrrolidino, piperidino, morpholino, C1-C6-phenyl substituted with substituents of alkylcarbonylamino and halogen;
unsubstituted 2-or 3-furyl or substituted by 1 or 2 radicals selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy and halogen substituted 2-or 3-furyl; or
Unsubstituted 2-or 3-thienyl or substituted by 1 or 2 groups selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy and halogen substituted 2-or 3-thienyl; and wherein
R3BIs unsubstituted phenyl or phenyl substituted with 1, 2 or 3 substituents selected from: c1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy, C1-C6-alkylcarbonyl group, C1-C6-alkylcarbonyloxy, C1-C6Alkylthio, nitro, amino, C1-C6-alkylamino, di-C1-C6Alkylamino, pyrrolidino, piperidino, morpholino, C1-C6-alkylcarbonylamino and halogen;
a is selected from the partial structures a-i as defined above;
X1is O, S, NH, N (CH)3) Or CH2(ii) a And
X2is O, S or NH.
Equally preferred are compounds of formula 1 in the pharmaceutical compositions of the invention, wherein R1Is unsubstituted phenyl or phenyl monosubstituted by 1 substituent selected from: c1-C3-alkyl, trifluoromethyl、C1-C3-alkoxy, C1-C3-alkylcarbonyl group, C1-C3-alkylcarbonyloxy, C1-C3Alkylthio, nitro, amino, C1-C3-alkylamino, di-C1-C3Alkylamino, pyrrolidino, piperidino, morpholino, C1-C3-alkylcarbonylamino and halogen;
unsubstituted 2-, 3-or 4-pyridyl or 2-, 3-or 4-pyridyl monosubstituted by 1 substituent selected from the group consisting of: c1-C3-alkyl, trifluoromethyl, C1-C3Alkoxy, nitro, amino, C1-C3-alkylamino, di-C1-C3-alkylamino radical, C1-C3-alkylcarbonylamino and halogen; or an unsubstituted indolyl group or an indolyl group monosubstituted with 1 substituent selected from: c1-C3-alkyl, trifluoromethyl, C1-C3Alkoxy, nitro, amino, C1-C3-alkylamino, di-C1-C3-alkylamino radical, C1-C3-alkylcarbonylamino and halogen;
R2is (CH)2)nWherein n is 0, 1 or 2;
R3is (CH)2)mR3AWherein m is 0, 1, 2, 3 or 4;
R3Ais CH3、CH(CH3)2、C(CH3)3、OCH3、OH、OR3BC ≡ CH, C ≡ N, unsubstituted phenyl or substituted by 1, 2 or 3 substituents selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy, C1-C6-alkylcarbonyl group, C1-C6-alkylcarbonyloxy, C1-C6Alkylthio, nitro, amino, C1-C6-alkylamino, di-C1-C6Alkylamino, pyrrolidino, piperidino, morpholinoQuinoline and C1-C6-phenyl substituted with substituents of alkylcarbonylamino and halogen;
unsubstituted 2-or 3-furyl or substituted by 1 or 2 radicals selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy and halogen substituted 2-or 3-furyl; or
Unsubstituted 2-or 3-thienyl or substituted by 1 or 2 groups selected from C1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy and halogen substituted 2-or 3-thienyl; and wherein
R3BIs unsubstituted phenyl or phenyl substituted with 1, 2 or 3 substituents selected from: c1-C6-alkyl, trifluoromethyl, C1-C6-alkoxy, C1-C6-alkylcarbonyl group, C1-C6-alkylcarbonyloxy, C1-C6Alkylthio, nitro, amino, C1-C6-alkylamino, di-C1-C6Alkylamino, pyrrolidino, piperidino, morpholino, C1-C6-alkylcarbonylamino and halogen;
a is selected from the partial structures a-i as defined above;
X1is O, NH or N (CH)3) (ii) a And
X2is O, S or NH.
More preferred compounds of formula 1 in the pharmaceutical compositions of the present invention are those compounds wherein R is1Is unsubstituted phenyl; unsubstituted 2-, 3-, or 4-pyridyl; or an unsubstituted indolyl group;
R2is (CH)2)nWherein n is 0, 1 or 2;
R3is (CH)2)mR3AWherein m is 0, 1, 2, 3 or 4;
R3Ais CH3、CH(CH3)2、C(CH3)3、OCH3、OH、OR3BC ≡ CH, C ≡ N, unsubstituted phenyl or substituted by 1, 2 or 3 substituents selected from C1-C6-alkyl, trifluoromethyl, C1-C6-phenyl substituted with substituents of alkoxy and halogen; unsubstituted 2-or 3-furyl or unsubstituted 2-or 3-thienyl; and wherein
R3BIs unsubstituted phenyl or substituted by 1, 2 or 3 substituents selected from C1-C6-alkyl, trifluoromethyl, C1-C6-phenyl substituted with substituents of alkoxy and halogen;
a is selected from the partial structures a-i as defined above;
X1is O, NH or N (CH)3) (ii) a And
X2is O.
Equally preferred are compounds of formula 1 in the pharmaceutical compositions of the present invention, wherein
R1Is unsubstituted phenyl; unsubstituted 2-, 3-, or 4-pyridyl; or an unsubstituted indolyl group;
R2is (CH)2)nWherein n is 0, 1 or 2;
R3is (CH)2)mCH3、(CH2)mC ≡ CH or (CH)2)mC ═ N, where m is 2 or 3;
a is selected from the partial structures a-d and f-i as defined above;
X1is O, NH or N (CH)3) (ii) a And
X2is O.
Even more preferred are compounds of formula 1 in the pharmaceutical compositions of the present invention, wherein
R1Is phenyl or 2-, 3-or 4-pyridyl;
R2is (CH)2)nWherein n is 0, 1 or 2;
R3is (CH)2)mR3AWherein m is 1, 2, 3 or 4;
R3Ais CH3、CH(CH3)2、C(CH3)3、OCH3;OR3B(ii) a Unsubstituted phenyl or substituted by 1 or 2 substituents selected from C1-C6-alkyl, trifluoromethyl, C1-C6-phenyl substituted with substituents of alkoxy and halogen; 2-or 3-furyl; or 2-or 3-thienyl; and wherein
R3BIs unsubstituted phenyl or substituted by 1 or 2 substituents selected from C1-C6-alkyl, trifluoromethyl, C1-C6-phenyl substituted with substituents of alkoxy and halogen;
a is selected from the partial structures a-h as defined above;
X1is O or NH; and
X2is O.
Particularly preferred are compounds of formula 1 in the pharmaceutical compositions of the present invention, wherein
R1Is phenyl or 2-, 3-or 4-pyridyl;
R2is (CH)2)nWherein n is 0, 1 or 2;
R3is (CH)2)mR3AWherein m is 1, 2, 3 or 4;
R3Ais CH3、CH(CH3)2、C(CH3)3、OCH3;OR3B(ii) a A phenyl group; 2-furyl; or 2-thienyl; and wherein
R3BIs unsubstituted phenyl or phenyl substituted by halogen;
a is selected from the partial structures a, b, c, e, f, g and h as defined above;
X1is O or NH; and
X2is O.
The most preferred compounds of formula 1 in the pharmaceutical compositions of the present invention are those of the examples.
Preferred compounds of formula 2 in the pharmaceutical compositions of the present invention are those compounds wherein
R4Is optionally substituted pyridyl, -NR7R8or-NH-N ═ CH-R9;
R5Is hydrogen, optionally substituted phenyl or a sugar residue;
R6is hydrogen;
R7is an optionally substituted phenyl or sugar residue;
R8is hydrogen; and
R9is optionally substituted phenyl.
Even more preferred are compounds of formula 2 in the pharmaceutical compositions of the present invention, wherein
R4Is substituted pyridyl, NR7R8or-NH-N ═ CH-R9;
R5Is hydrogen, substituted phenyl or a sugar residue;
R6is hydrogen;
R7is a substituted phenyl or sugar residue;
R8is hydrogen; and
R9is a substituted phenyl group。
Most preferred are compounds of formula 2 in the pharmaceutical compositions of the present invention, wherein
R4Is a quilt C1-C6Alkyl, NR7R8or-NH-N ═ CH-R9A substituted pyridyl group;
R5is hydrogen, C1-C6-alkoxy substituted phenyl, or a sugar residue;
R6is hydrogen;
R7is a quilt C1-C6-alkoxy substituted phenyl, or a sugar residue;
R8is hydrogen; and
R9is a quilt C1-C6-alkylcarbonylamino substituted phenyl.
Particularly preferred are compounds of formula 2 in the pharmaceutical compositions of the present invention, wherein R4Is a quilt C1-C6-alkyl substituted 4-pyridyl; r5Is hydrogen or a sugar residue; and R6Is hydrogen; in particular ethionamide of formula 8:
a compound of formula 2 wherein R4is-NH-N ═ CH-R9;R5Is hydrogen or a sugar residue; r6Is hydrogen; and R9Is a quilt C1-C6-alkylcarbonylamino substituted phenyl, in particular thiosemicarbazides of formula 9:
a compound of formula 2 wherein R4is-NR7R8;R5Is a quilt C1-C6-alkoxy-substituted phenyl; r6Is hydrogen; r7Is a quilt C1-C6-an alkoxy-substituted phenyl or sugar residue; and R8Is hydrogen; in particular pentylenethiophenyl (isoxyl) of formula 10:
or pentoxyphenylthiourea analogs N-arabinofuranosyl-N' - [ p- (isopentyloxy) phenyl ] -thiourea of formula 11:
most preferred is a composition comprising a compound of formula 1 and a compound of formula 2, the compound of formula 2 being selected from the group consisting of ethionamide, pentyloxyphenylurea, N-arabinofuranosyl-N' - [ p- (isopentyloxy) phenyl ] -thiourea or thiosemicarbazide, especially ethionamide.
It will be appreciated that the pharmaceutical composition of the present invention comprising a compound of formula 1 as described herein and a compound of formula 2 as described herein may be a pharmaceutical composition comprising a mixture of a compound of formula 1 and a compound of formula 2, or two separate formulations of a compound of formula 1 and a compound of formula 2, packaged together or provided separately.
The compounds of formula 1 are known compounds or may be prepared according to methods well known in the art.
The compounds of formula 2 are known compounds or can be prepared according to the following method.
Thioamides, i.e. compounds of formula 2, in which R4Is optionally substituted phenyl, optionally substituted pyridyl or optionally substituted indolyl, obtainable by reacting a compound of formula HNR5R6With an amine of the formula R4Reaction of carboxylic acid of-COOH to form the formula R4-CO-NR5R6The amide of (a). The amide is reacted with Lawesson's reagent or phosphorus pentasulfide to give the thioamide of formula 2. Another method for synthesizing thioamides is the Kindler modification by the Willgeodt reaction using the formula R4Aldehydes of the formula-CH ═ O and HNR5R6And reacting them in the presence of sulfur.
Thiourea, i.e. a compound of formula 2, wherein R4is-NR7R8The formula R can be obtained by reacting bromide with potassium thiocyanate5-N ═ C ═ S or R7An isothiocyanate of the formula-N ═ C ═ S, each of which is reacted with a compound of the formula HNR7R8Or HNR5R6Is obtained by reacting the amine of (a). Corresponding hydrazones, i.e. in which R is4is-NH-N ═ CH-R9By reacting a compound of formula R9Aldehydes of the formula-CH ═ O and R5R6N-(C=S)-NH-NH2Hydrazinothiocarboxamide.
The invention also relates to novel compounds of formula 1 of the kind defined above.
In particular, the invention relates to compounds of formula 1
Wherein R is1Is phenyl or 2-pyridyl; r2Is (CH)2)nWherein n is 0, 1 or 2; r3Is (CH)2)mR3AWherein m is 1, 2 or 3; r3AIs CH3Or phenyl; a is
Wherein-represents a linkIs linked to C (═ X)2)-R3And → represents the connection R2-R1A bond of (a);
or a and-C (═ X)2)-R3Together forming a ring
Wherein → represents the connection R2-R1A bond of (a);
X1is O; and X2Is O;
or therein
R1Is phenyl; r2Is a bond; r3Is (CH)2)4CH3;
A is
Wherein-represents the linkage C (═ X)2)-R3And → represents the connection R2-R1A bond of (a);
X1is O; and X2Is O.
More particularly, the present invention relates to compounds of formula 1, wherein
R1Is phenyl; r2Is (CH)2)nWherein n is 0 or 1;
R3is (CH)2)mR3AWherein m is 1, 2 or 3, and R3AIs CH3Or phenyl;
a is
X2Is O.
Likewise, the invention relates to compounds of formula 1, wherein
R1Is phenyl; r2Is a bond; r3Is (CH)2)4CH3;
A is
Wherein-represents the linkage C (═ X)2)-R3And → represents the connection R2-R1A bond of (a);
X1is O; and X2Is O.
Most preferred are example compounds 2, 4-12, 22-27, 31 and 33.
The invention is also directed to the use of compositions comprising a compound of formula 1 and a compound of formula 2 (e.g., ethionamide) in the treatment of tuberculosis and related diseases, and to methods in which compositions comprising a compound of formula 1 and a compound of formula 2 (e.g., ethionamide) are used to treat tuberculosis and related diseases.
The present invention relates to a pharmaceutical composition comprising a compound of formula 1 as defined above and a compound of formula 2 as defined above, preferably ethionamide.
Since the compound of formula 1, thioamide or thiourea, i.e. the compound of formula 2, which inhibits the binding of EthR to the ethA promoter, is added to the pharmaceutical composition for the treatment of tuberculosis, the dosage thereof can be greatly reduced, thereby reducing the familiar side effects without reducing the utility thereof.
The pharmaceutical composition of the present invention is useful not only for treating tuberculosis, i.e., diseases caused by Mycobacterium tuberculosis, but also for treating diseases caused by related bacteria having an EthR-associated protein binding to a corresponding ethA-associated promoter, particularly Mycobacterium leprae, Mycobacterium ulcerosa (Mycobacterium ulcerocerans), Mycobacterium marinum (Mycobacterium marinum), Mycobacterium strain MCS (Mycobacterium sp.MCS), Mycobacterium strain KMS (Mycobacterium sp.KMS), Mycobacterium strain JLS (Mycobacterium sp.JLS), Mycobacterium vacaum, Mycobacterium paratuberculosis subspecies (Mycobacterium subspecies, Mycobacterium parakuwanensis), Mycobacterium avium (Mycobacterium avium), Mycobacterium smegmatis (Mycobacterium smegmatis), Mycobacterium phlei (Mycobacterium phlei), Mycobacterium smegmatis (Mycobacterium phlei), Mycobacterium phlei (Mycobacterium phlei), Mycobacterium smegmatis), Mycobacterium phlei (Mycobacterium phlei), Mycobacterium phlei (Mycobacterium phle, Bacillus licheniformis (Bacillus licheniformis), Clostridium spirochetum (Clostridium spiroforme) and Bacillus anthracis (Bacillus anthracaris). These diseases are leprosy, bruuli-ulcer disease, atypical mycobacteriosis, Johne's and crohn's disease, bathhouse lung (hot tub lung), female winddermere syndrome, chronic lung disease, post-traumatic wound infection, post-tympanostomy duct eardrops, disseminated skin disease, acinetobacter baumannii caused infection, pharyngitis, pustule, erysipelas, cellulitis, necrotizing fasciitis, scarlet fever, toxic shock septicemia, peritonitis, ophthalmia, diarrhea and malignant anthrax.
The pharmaceutical compositions of the invention are compositions for enteral, such as intranasal, buccal, rectal or, especially, oral administration, and for parenteral, such as intravenous, intramuscular or subcutaneous administration. The compositions comprise a compound of formula 1 and a compound of formula 2, and preferably a pharmaceutically acceptable carrier. The dosage of the active ingredient depends on the patient, his age, weight and personal condition, the pharmacokinetic data of the individual and the mode of administration.
The invention also relates to the use of said pharmaceutical composition in a method for the prophylaxis or, in particular, the treatment of tuberculosis, leprosy, purulent ulcers, atypical mycobacteriosis, john's and Crohn's disease, bathylar lung, female winddermere's syndrome, chronic lung disease, post-traumatic wound infection, posttympanostomy duct otorrhea, disseminated skin disease, acinetobacter baumannii-induced infection, pharyngitis, pustule, erysipelas, cellulitis, necrotizing fasciitis, scarlet fever, toxic shock septicemia, peritonitis, ophthalmia, diarrhea and malignant anthrax in a human or animal body.
The pharmaceutical composition comprises from about 5% to 95% of a mixture of the compound of formula 1 and the compound of formula 2, the relative molar amount of the compound of formula 1 and the compound of formula 2 being between 1: 1 and up to 1: 10,000, preferably 1: 10 and up to 1: 5000. The single-dose administration forms comprise from about 20% to about 90% of the mentioned mixture and from about 5% to about 20% of the mentioned mixture, which are not of the single-dose type. For example, the unit dosage form may be coated and uncoated tablets, ampoules, vials, suppositories or capsules. Other dosage forms are, for example, ointments, creams, pastes, foams, tinctures, lipsticks, drops, syrups, sprays and the like. An example is a capsule containing about 0.05g to about 1.0g of the active ingredient mixture.
It is also possible to use a mixture of the compound of formula 1 and the compound of formula 2 in two separate pharmaceutical unit dosage forms, and such a combination also forms part of the present invention. For example, a compound of formula 2, e.g., ethionamide, may be used in combination in a unit dosage form in an amount of 0.01g to about 0.5g, e.g., 0.05g to about 0.5g commercially available, of ethionamide, with a different or the same unit dosage form containing a compound of formula 1 in an amount of 0.5g to about 5.0g, in kit of parts.
The pharmaceutical compositions of the present invention may be prepared in a manner known per se, for example by means of conventional mixing, granulating, coating, dissolving, emulsifying or lyophilizing processes. The compound of formula 1 may optionally be formulated into a liposome formulation.
For parenteral administration, solutions of the active ingredient are preferred, as are suspensions, emulsions, or dispersions, especially isotonic aqueous solutions, dispersions, emulsions, or suspensions, which can be formulated prior to use, for example, as a lyophilized composition containing the active ingredient alone or together with a carrier. The pharmaceutical compositions may be sterilized and/or may contain excipients, for example preservatives, stabilizers, wetting and/or emulsifying agents, cosolvents, salts for regulating the osmotic pressure and/or buffers, and may be prepared in a manner known per se, for example by means of conventional dissolving and freeze-drying processes. The solution or suspension may contain a viscosity enhancing agent, typically sodium carboxymethylcellulose, dextran, polyvinylpyrrolidone or gelatin agent, or may also contain a co-solvent such as tween 80 (polyoxyethylene (20) sorbitan monooleate).
Suspensions in oils comprise, as oily component, vegetable oils, synthetic or semisynthetic oils of the type commonly used for injection purposes. Oils which are mentioned in particular for this purpose consist of liquid fatty acid esters which contain, as acid component, long-chain fatty acids having 8 to 22, in particular 12 to 22, carbon atoms. The alcohol component of these fatty acid esters has up to 6 carbon atoms and is a monovalent or polyvalent alcohol, for example a mono-, di-or trivalent alcohol, especially glycols and glycerol. As mixtures of fatty acid esters, vegetable oils, such as cottonseed oil, almond oil, olive oil, castor oil, sesame oil, soybean oil and peanut oil, are particularly suitable.
The preparation of injections is usually carried out under sterile conditions, for example when filling into ampoules or vials and sealing the containers.
Suitable carriers for oral compositions are, in particular, fillers, for example sugars, such as lactose, sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates, for example tricalcium phosphate or calcium hydrogen phosphate, and also binders, for example starches, such as corn, wheat, rice or potato starch, methyl cellulose, hydroxypropylmethyl cellulose, sodium carboxymethyl cellulose and/or polyvinylpyrrolidone, and/or, if desired, disintegrants, such as the starches mentioned above, also carboxymethyl starch, crosslinked polyvinylpyrrolidone, alginic acid or a salt thereof, such as sodium alginate. Further excipients are, in particular, flow regulators and lubricants, for example silicic acid, talc, stearic acid or salts thereof, such as magnesium or calcium stearate, and/or polyethylene glycol or derivatives thereof.
The tablet cores may be provided with a suitable coating, optionally an enteric coating, by using, inter alia, concentrated sugar solutions which may contain gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide, or coating solutions in suitable organic solvents or solvent mixtures, or solutions of suitable cellulose preparations for the preparation of enteric coatings, such as acetyl cellulose phthalate or hydroxypropylmethyl cellulose phthalate. Dyes or pigments may be added to the tablets or tablet coatings, for example for identification purposes or to indicate different doses of the active ingredient.
Pharmaceutical compositions for oral administration also include hard capsules composed of gelatin and soft, sealed capsules composed of gelatin and a plasticizer (e.g., glycerol or sorbitol). Hard capsules may contain the active ingredient in the form of granules, for example in admixture with fillers, such as corn starch, binders and/or glidants, such as talc or magnesium stearate, and optionally stabilizers. In soft capsules, the active ingredient is preferably dissolved, emulsified or suspended in suitable liquid excipients, such as fatty oils, paraffin oils or liquid polyethylene glycols or fatty acid esters of ethylene glycol or propylene glycol, to which stabilizers and detergents, for example of the polyoxyethylene sorbitan fatty acid ester type, may also be added.
Pharmaceutical compositions suitable for rectal administration are, for example, suppositories which are composed of the active ingredient in combination with a suppository base. Suitable suppository bases are, for example, natural and synthetic triglycerides, alkanes, polyethylene glycols or higher alkanols.
Furthermore, the present invention relates to a method for the treatment of tuberculosis and related diseases, which comprises administering to a warm-blooded animal in need of such treatment a mixture of a compound of formula 1 and a compound of formula 2 in an amount effective against the disease. The mixture may be administered prophylactically or therapeutically to a warm-blooded animal, such as man, in need of such treatment, preferably in an amount effective against tuberculosis and related diseases, in the form of a pharmaceutical composition comprising the mixture, or the components may also be administered separately, simultaneously or at separate times during the day. In an individual case weighing about 70kg, the daily dose of the mixture administered is from about 0.01g to about 50g, preferably from about 0.05g to about 10g of the mixture, including the components in relative amounts of between 1: 1 and 1: 10' 000.
The invention also relates in particular to the use of a compound of formula 1 in combination with a compound of formula 2, such as ethionamide, as such or in a pharmaceutical formulation containing at least one pharmaceutically acceptable carrier, for the therapeutic and also prophylactic control of tuberculosis, the two compounds being administered, for example, separately or in a fixed composition. Preferred dosages, compositions and methods of preparation of the pharmaceutical formulations to be used in each case are described above.
The following examples are intended to illustrate the invention and do not limit the scope of the invention.
Examples
Media design
pWW489(PSV40-ethR-vp16-pA) was constructed by PCR-mediated amplification of ethR in genomic m.bovis DNA using oligonucleotides OWW400 and OWW401 (Weber w.et al, PNAS 2008, 105, 9994-8), followed by definition and ligation (EcoRI/bshii) pWW35 (Weber, w.et al, Nat Biotechnol 2002, 20, 901-7). pWW491 (O)ethR-Phsp70minSEAP-pA) by reacting the synthesized OethRThe sequence (Weber W.et al, PNAS 2008, 105, 9994-8) (AatII/SbfI) was directly cloned into pMF172 (Weber, W et al, supra). pWW871 (5' LTR-psi)+-ethR-vp16-PPGK-neoR-3' LTR) was designed by cloning the ethR-vp16 of pWW489(EcoRI/BamHI) as pMSCVneo (Clontech).
Cell culture
Human embryonic kidney cells (HEK-293, ATCC CRL-1573) were cultured in Dulbecco's modified Eagle's medium (DMEM; Invitrogen, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (Pan Biotech GmbH, Aidenbach, Germany, cat. No.3302, lot P231902) and 1% penicillin/streptomycin solution (Sigma, St. Louis, MO, USA, cat. No. 4458). Cell transfection was performed using standard calcium phosphate methods (Weber W. et al, supra) followed by generation of retroviral particles (Clontech) according to the manufacturer's protocol.EthRHEK, transgenic constitutive EthR-VP16 expression was constructed by conversion of HEK-293 with pWW 871-derived retroviral particles followed by selection and single cell cloning in DMEM containing 200 μ g/ml neomycin.EthRCo-transfection of HEK with pWW491 and pPUR (Clontech), followed by selection in 200. mu.g/ml neomycin, 1. mu.g/ml puromycin, and subsequent single cell cloning resulted in the generation ofEthRHEK-SEAP. SEAP generation was quantified as described in schlator, s.et al, Gene 2002, 282, 19-31.
Mycobacterium culture and susceptibility testing Using the MGIT 960 System with EpiCenter TB eXiST software
Mycobacterium tuberculosis H37Rv (ATCC27294) was grown in Middlebrook 7H9 supplemented with oleic acid, albumin, glucose, catalase (Difco) and tween 80 (0.05%) until mid-log.
For drug susceptibility testing, the MGIT 960 system (Becton Dickinson) was used according to the manufacturer's manual. 0.8ml of MGIT 960SIRE supplement (Becton Dickson) and 0.2ml of drug solution were added to the MGIT tube. Each tube was inoculated with 0.5ml of the test strain suspension. As a control, MGIT tubes without drug were inoculated with 0.5ml of a 1: 100 diluted (sterile water) suspension of the test strain. The growth of the bacteria was monitored by EpiCenter software (version 5.6.6) equipped with a TB eXiST module (Becton Dickinson) and expressed in Growth Units (GU). When the test tube reached ≥ 100GU before the drug-free control tube reached 400 GU-value, the strain was considered drug-resistant (R) to the drug. The susceptibility of the strain is determined (S) when the drug control reaches 400GU and the test tube remains ≤ 100GU more than 7 days after the drug control reaches 400 GU. The strain was considered to be in the middle (I) when the test tube reached ≥ 100GU within 7 days after the control tube reached 400 GU.
Liquid Chromatography (LC)
HPLC column: reverse phase, Zorbax SB-C181.8 m 4.6x15mm fast split column; wavelength: 210nm-400 nm; HPLC instrument model: agilent 1100, diode array detector (PDA); MS instrument model: agilent SL, positive/negative mode switching, ionization mode ESI.
LCMS system 01(LCMS 01): gradient 01
Solvent A: acetonitrile/water (95: 5), 0.1% formic acid; solvent B: water, 0.1% formic acid;
| time (min) | Flow rate (mL/min) | %A | %B |
| 0.00 | 3 | 0 | 100 |
| 0.01 | 3 | 0 | 100 |
| 0.50 | 3 | 100 | 0 |
| 0.95 | 3 | 100 | 0 |
| 0.96 | 3 | 0 | 100 |
| 1.00 | 3 | 0 | 100 |
LCMS system 02(LCMS 02): gradient 02
Solvent A: water, 0.1% formic acid; solvent B: methanol, 0.1% formic acid
| Time (min) | Flow rate (mL/min) | %A | %B |
| 0.00 | 3 | 100 | 0 |
| 0.02 | 3 | 100 | 0 |
| 1.50 | 3 | 0 | 100 |
| 2.30 | 3 | 0 | 100 |
| 2.4 | 3 | 100 | 0 |
| 2.5 | 3 | 100 | 0 |
Example 1: 1- (4-Benzylpiperazin-1-yl) butan-1-one
A solution of 1-benzylpiperazine (352mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath, followed by the addition of butyryl chloride (266mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5 eq). After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane was removed in vacuo and the crude material was purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 1). The fractions containing 1- (4-benzylpiperazin-1-yl) butan-1-one were collected and the solvent was removed in vacuo to give 432mg of a colourless oil.
LCMS ESI+:247.0(M+H)+,Rt=0.511min(LCMS 01)
Example 2: 1- (4- (pyridin-2-ylmethyl) piperazin-1-yl) butan-1-one
To a solution of 1eq 1-Boc-piperazine and 3eq triethylamine in Tetrahydrofuran (THF) was added dropwise a solution of 1eq (2-bromomethyl) pyridine hydrobromide in tetrahydrofuran. After stirring overnight at room temperature, the reaction mixture was concentrated in vacuo to remove tetrahydrofuran and then resuspended in diethyl ether. The solution was filtered to remove triethylamine hydrobromide. The residue was washed with diethyl ether and the filtrate was concentrated in vacuo. The crude product was purified by column chromatography.
1-Boc-4- (pyridin-2-ylmethyl) piperazine was dissolved in diethyl ether and treated with saturated HCl/diethyl ether to remove the Boc-group. After stirring for 1 hour, the solvent was removed in vacuo to give 1- (pyridin-2-ylmethyl) piperazine hydrochloride.
To a solution of 1eq 1- (pyridin-2-ylmethyl) piperazine hydrochloride in anhydrous dichloromethane was added 1.5eq butyryl chloride and 2.5eq triethylamine. After stirring at room temperature for 6 hours, the solvent was removed in vacuo and the residue was redissolved in ethyl acetate. Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4Drying, removing ethyl acetate in vacuum and purifying the crude product by column chromatography.
Example 3 1- (2-Phenylmorpholino) butan-1-one
A solution of 2-phenylmorpholine (326mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath, followed by the addition of butyryl chloride (266mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5 eq). After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane was removed in vacuo and the crude material was purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 3). The fractions containing 1- (2-phenylmorpholin-4-yl) butan-1-one were collected and the solvent was removed in vacuo to give 378mg of a colourless oil.
1H-NMR(D6-DMSO/CCl4,400MHz)δ:7.38(m,5H),4.37(m,2H),4.00(m,1H),3.85(m,1H),3.55(m,1H),3.14(m,1H),2.66(m,1H),1.86(m,2H),1.54(m,2H),0.92(t,3H)。
Example 4: 1- (2- (pyridin-4-yl) morpholino) butan-1-one
To a solution of 1eq 2-amino-1- (pyridin-3-yl) ethanol and 3eq triethylamine in Tetrahydrofuran (THF), 1eq 2-bromoethanol was added dropwise. After stirring overnight at room temperature, the reaction mixture was concentrated in vacuo to remove tetrahydrofuran and then resuspended in diethyl ether. The solution was filtered to remove triethylamine hydrobromide. The residue was washed with diethyl ether and the filtrate was concentrated in vacuo. The crude product, 2- (2-hydroxyethylamino) -1- (pyridin-4-yl) ethanol, was purified by recrystallization.
Preparation of 2- (pyridin-4-yl) morpholine according to the method of F.Zymalkowski and F.Koppe (Arch.Pharmaz.1961, 294, 453-468) 2- (2-hydroxyethylamino) -1- (pyridin-4-yl) ethanol was treated with oleum at 170 ℃.
To a solution of 2- (pyridin-4-yl) morpholine in anhydrous dichloromethane was added 1.5eq butyryl chloride and 2.5eq triethylamine. After stirring at room temperature for 6 hours, the solvent was removed in vacuo and the residue was redissolved in ethyl acetate. Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4Drying and removing the ethyl acetate in vacuo and purifying the crude product 1- (2- (pyridin-4-yl) morpholino) butan-1-one by column chromatography.
Example 5: 1-phenethyl-3-propylpyrrolidin-2-one
The preparation of 1-phenethyl-2-pyrrolidone was carried out using 2-pyrrolidone and 2-bromoethylbenzene in N, N-dimethylformamide in the presence of sodium hydride (M.Matsukawa et al. neuro toxicology 2004, 25, 293-.
To a solution of 2M lithium diisopropylamide in tetrahydrofuran at-78 deg.C was added 1-phenethyl-2-pyrrolidone and the mixture was stirred at-78 deg.C for 1.5 h. Then 1-propyl bromide is addedThe reaction was stirred for a further 1.5h at-78 ℃. The reaction mixture was warmed to not more than 0 ℃, and then acetic acid and ice-cooled water were added, followed by extraction with dichloromethane 3 times. The dichloromethane was concentrated in vacuo, the residue was dissolved in dichloromethane and saturated NaHCO3And a brine wash. The solvent was removed in vacuo to give crude 1-phenethyl-3-propylpyrrolidin-2-one. The product was purified by column chromatography.
Example 6: 3-propyl-1- (2- (pyridin-2-yl) ethyl) pyrrolidin-2-one
The preparation of 1- (2- (pyridin-2-yl) ethyl) pyrrolidin-2-one was carried out using 2-pyrrolidone and 2-bromoethylbenzene in N, N-dimethylformamide in the presence of sodium hydride (M.Matsukawa et al, neuro Toxicology 2004, 25, 293-.
To a 2M solution of lithium diisopropylamide in tetrahydrofuran at-78 deg.C was added 1- (2- (pyridin-2-yl) ethyl) pyrrolidin-2-one and the mixture was stirred at-78 deg.C for 1.5 h. 1-propyl bromide was then added and the reaction was stirred for an additional 1.5h at-78 ℃. The reaction mixture was warmed to 0 ℃, then acetic acid and ice-cooled water were added, followed by extraction with dichloromethane 3 times. The dichloromethane was concentrated in vacuo, the residue was dissolved in dichloromethane and saturated NaHCO3And a brine wash. The solvent was removed in vacuo to give crude 3-propyl-1- (2- (pyridin-2-yl) ethyl) pyrrolidin-2-one. The product was purified by column chromatography.
Example 7: 5-benzyl-3-propylpyrrolidin-2-one
Synthesis of 5-benzylpyrrolidin-2-one was carried out using pyrrolidine-2, 5-dione and benzyl bromide using the method of S.Lebrun et al (Tetrahedron Asymmetry 2003, 14, 2625-2632).
To a solution of 2M lithium diisopropylamide in tetrahydrofuran at-78 deg.C was added 5-benzylpyrrolidin-2-one and the mixture was stirred at-78 deg.C for 1.5 h. 1-propyl bromide was then added and the reaction was stirred for an additional 1.5h at-78 ℃. The reaction mixture was warmed to 0 ℃, then acetic acid and ice-cooled water were added, followed by extraction with dichloromethane 3 times. The dichloromethane was concentrated in vacuo, the residue was dissolved in dichloromethane and saturated NaHCO3And a brine wash. The solvent was removed in vacuo to give crude 5-benzyl-3-propylpyrrolidin-2-one. The product was purified by column chromatography.
Example 8: 3-propyl-5- (pyridin-2-ylmethyl) pyrrolidin-2-one
Synthesis of 5- (pyridin-2-ylmethyl) pyrrolidin-2-one was carried out using pyrrolidine-2, 5-dione and 2- (bromomethyl) pyridine using the method of S.Lebrun et al (Tetrahedron Asymmetry 2003, 14, 2625-2632).
To a 2M solution of lithium diisopropylamide in tetrahydrofuran at-78 deg.C was added 5- (pyridin-2-ylmethyl) pyrrolidin-2-one and the mixture was stirred at-78 deg.C for 1.5 h. 1-propyl bromide was then added and the reaction was stirred for an additional 1.5h at-78 ℃. The reaction mixture was warmed to 0 ℃, then acetic acid and ice-cooled water were added, followed by extraction with dichloromethane 3 times. The dichloromethane was concentrated in vacuo, the residue was dissolved in dichloromethane and saturated NaHCO3And a brine wash. The solvent was removed in vacuo to give crude 3-propyl-5- (pyridin-2-ylmethyl) pyrrolidin-2-one. The product was purified by column chromatography.
Example 9: 4-Ethyl-1-phenethyl-1H-pyrrol-2 (5H) -one
Synthesis of 4-ethyl-1-phenethyl-1H-pyrrol-2 (5H) -one according to R.Fisher's method (DE 4127111, 1992), by cyclocondensation between methyl 3-formylvalerate and 2-phenylethylamine in glacial acetic acid at 70 ℃.
Example 10: 4-Ethyl-1- (2- (pyridin-2-yl) ethyl) -1H-pyrrol-2 (5H) -one
Synthesis of 4-ethyl-1- (2- (pyridin-2-yl) ethyl) -1H-pyrrol-2 (5H) -one was carried out according to the method of R.Fisher (DE 4127111, 1992) by cyclocondensation between methyl 3-formylvalerate and 2- (pyridin-2-yl) ethylamine in glacial acetic acid at 70 ℃.
Example 11: 5-benzyl-3-propyldihydrofuran-2 (3H) -one
4-oxo-5-phenylpentanoic acid was prepared from the amino acids valine and 2-phenylacetic acid as starting materials according to the procedure described by W.Steglich and P.Gruber (Angew.chem., 1971, 83, 727-one 728).
Reacting 4-oxo-5-phenylpentanoic acid with NaBH4Reduction followed by treatment with hydrogen chloride gave 5-benzylpyrrolidin-2-one (C.Ketterer et al, Tetrahedron Asymmetry 2006, 17, 3046-.
To a solution of 2M lithium diisopropylamide in tetrahydrofuran at-78 deg.C was added 5-benzylpyrrolidin-2-one and the mixture was stirred at-78 deg.C for 1.5 h. 1-propyl bromide was then added and the reaction was stirred for an additional 1.5h at-78 ℃. Warming the reaction mixtureHeated to 0 ℃, then acetic acid and ice-cooled water were added, followed by extraction with dichloromethane 3 times. The dichloromethane was concentrated in vacuo, the residue was dissolved in dichloromethane and saturated NaHCO3And a brine wash. The solvent was removed in vacuo to give crude 5-benzyl-3-propyldihydrofuran-2 (3H) -one. The product was purified by column chromatography.
Example 12: 3-propyl-5- (pyridin-2-ylmethyl) dihydrofuran-2 (3H) -one
4-oxo-5- (pyridin-2-yl) pentanoic acid is synthesized from valine and 2- (pyridin-2-yl) acetic acid according to the method of W.Steglich and P.Gruber (Angew.chem., 1971, 83, 727-728).
With NaBH4Reduction of 4-oxo-5- (pyridin-2-yl) pentanoic acid followed by treatment with hydrogen chloride gave 5- (pyridin-2-ylmethyl) dihydrofuran-2 (3H) -one (adapted from C.Ketterer et al, Tetrahedron Asymmetry 2006, 17, 3046-.
To a 2M solution of lithium diisopropylamide in tetrahydrofuran at-78 deg.C was added 5- (pyridin-2-ylmethyl) dihydrofuran-2 (3H) -one and the mixture was stirred at-78 deg.C for 1.5H. 1-propyl bromide was then added and the reaction was stirred for an additional 1.5h at-78 ℃. The reaction mixture was warmed to 0 ℃, then acetic acid and ice-cooled water were added, followed by extraction with dichloromethane 3 times. The dichloromethane was concentrated in vacuo, the residue was dissolved in dichloromethane and saturated NaHCO3And a brine wash. The solvent was removed in vacuo to give crude 3-propyl-5- (pyridin-2-ylmethyl) dihydrofuran-2 (3H) -one. The product was purified by column chromatography.
Example 13: 1- (3-phenylpyrrolidin-1-yl) butan-1-one
A solution of 3-phenylpyrrolidine (294mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and butyryl chloride (266mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 4). The fractions containing 1- (3-phenylpyrrolidin-1-yl) butan-1-one were collected and the solvent was removed in vacuo to yield 378mg of a colorless oil.
1H-NMR(D6-DMSO/CCl4,400MHz)δ:7.25(m,5H),3.87(m,1H),3.65(m,1H),3.35(m,3H),2.20(m,3H),2.00(m,1H),1.60(m,2H),0.97(t,3H)。
LCMS ESI+:218.2(M+H)+,Rt=1.086min(LCMS 02)。
Example 14: 1- (2-phenylmorpholin-4-yl) -2- (thien-2-yl) ethan-1-one
A solution of 2-phenylmorpholine (326mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and thien-2-ylacetyl chloride (401.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 3). The fraction containing 1- (2-phenylmorpholin-4-yl) -2- (thien-2-yl) ethan-1-one was collected and the solvent was removed in vacuo to give 511mgA colorless oil.
LCMS ESI+:288.2(M+H)+,Rt=1.546min(LCMS 02)。
Example 15: 2- (4-fluorophenoxy) -1- (3-phenylpyrrolidin-1-yl) ethan-1-one
A solution of 3-phenylpyrrolidine (294mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and (4-fluorophenoxy) acetyl chloride (471.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 4). The fractions containing 2- (4-fluorophenoxy) -1- (3-phenylpyrrolidin-1-yl) ethan-1-one were collected and the solvent was removed in vacuo to yield 495mg of a colorless oil.
1H-NMR(D6-DMSO,300MHz)δ:7.30(m,5H),6.94(m,4H),4.63(m,2H),3.95(m,1H),3.68(m,2H),3.40(m,2H),2.31(m,1H),2.03(m,1H)。
Example 16: 3-phenyl-1- (3-phenylpyrrolidin-1-yl) propan-1-one
A solution of 3-phenylpyrrolidine (294mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and 3-phenyl-propionyl chloride (421.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was stirredDilute with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 5). The fractions containing 3-phenyl-1- (3-phenylpyrrolidin-1-yl) propan-1-one were collected and the solvent was removed in vacuo to yield 521mg of a colorless oil.
1H-NMR(D6-DMSO/CCl4,300MHz)δ:7.15(m,10H),3.85(m,1H),3.62(m,1H),3.45(m,3H),2.82(m,2H),2.55(m,2H),2.28(m,1H),1.94(m,1H)。
Example 17: 2-phenyl-1- (3-phenylpyrrolidin-1-yl) ethan-1-one
A solution of 3-phenylpyrrolidine (294mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and phenyl-acetyl chloride (386.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 5). The fractions containing 2-phenyl-1- (3-phenylpyrrolidin-1-yl) ethan-1-one were collected and the solvent was removed in vacuo to give 465mg of a colorless oil.
1H-NMR(D6-DMSO/CCl4,300MHz)δ:7.12(m,10H),3.96(m,1H),3.61(m,3H),3.38(m,3H),2.39(m,1H),2.01(m,1H)。
Example 18: 1- (4-Benzylpiperazin-1-yl) -3- (furan-2-yl) propan-1-one
A solution of 1-benzylpiperazine (352mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and 3- (furan-2-yl) propionyl chloride (396.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 2). The fractions containing 1- (4-benzylpiperazin-1-yl) -3- (furan-2-yl) propan-1-one were collected and the solvent was removed in vacuo to give 497mg of a colorless oil.
LCMS ESI+:299.2(M+H)+,Rt=0.361min(LCMS 01)。
1H-NMR(D6-DMSO/CCl4,400MHz)δ:7.35(s,1H),7.27(s,4H),7.21(s,1H),6.26(s,1H),6.02(s,1H),3.45(m,6H),2.83(m,2H),2.59(m,2H),2.34(s,4H)。
Example 19: 3-methyl-1- (2-phenylmorpholin-4-yl) butan-1-one
A solution of 2-phenylmorpholine (326mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and 3-methylbutanoyl chloride (301.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane was removed in vacuo and the crude product was flash driedChromatography (silica gel, ethyl acetate/toluene 1: 3). The fractions containing 3-methyl-1- (2-phenylmorpholin-4-yl) butan-1-one were collected and the solvent was removed in vacuo to give 465mg of a colorless oil.
1H-NMR(CDCl3,400MHz)δ:7.43(m,5H),4.56(m,1H),4.36(m,1H),4.03(m,1H),3.74(m,1H),3.63(m,1H),3.20(m,1H),2.74(m,1H),2.18(m,3H),0.95(m,6H)。
Example 20: 3, 3-dimethyl-1- (2-phenylmorpholin-4-yl) butan-1-one
A solution of 2-phenylmorpholine (326mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and 3, 3-dimethylbutyrylchloride (336.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 4). The fractions containing 3, 3-dimethyl-1- (2-phenylmorpholin-4-yl) butan-1-one were collected and the solvent was removed in vacuo to give 438mg of a colourless oil.
1H-NMR(CDCl3,400MHz)δ:7.34(m,5H),4.60(m,1H),4.35(m,1H),4.03(m,1H),3.80(m,1H),3.61(m,1H),3.18(m,1H),2.71(m,1H),2.24(m,2H),1.03(s,9H)。
Example 21: 3-phenyl-1- (2-phenylmorpholin-4-yl) propan-1-one
A solution of 2-phenylmorpholine (326mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and 3-phenylpropionyl chloride (421.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 4). Fractions containing 3-phenyl-1- (2-phenylmorpholin-4-yl) propan-1-one were collected and the solvent was removed in vacuo to give 528mg of a colorless oil.
1H-NMR(D6-DMSO,400MHz)δ:7.36(m,5H),7.25(m,4H),7.17(m,1H),4.33(m,1H),4.30(m,1H),3.94(m,1H),3.81(m,1H),3.47(m,1H),3.11(m,1H),2.67(m,5H)。
Example 22: 3-phenyl-1- (2-phenylazetidin-1-yl) propan-1-one
A solution of 2-phenylazetidine (266mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and 3-phenylpropionyl chloride (421.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 4). The fractions containing 3-phenyl-1- (2-phenylazetidin-1-yl) propan-1-one were collected and the solvent was removed in vacuo to give 397mg of a colourless oil. LCMS ESI+:266.0(M+H)+,Rt=1.193min(LCMS 02)。
Example 23: 4-phenyl-1- (2-phenylazetidin-1-yl) butan-1-one
A solution of 2-phenylazetidine (266mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and 4-phenylbutyryl chloride (456.6mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 5). The fractions containing 4-phenyl-1- (2-phenylazetidin-1-yl) butan-1-one were collected and the solvent was removed in vacuo to yield 488mg of a colorless oil. LCMS ESI+:280.2(M+H)+,Rt=1.239min(LCMS 02)。
Example 24: 3-phenyl-1- (3-phenylazetidin-1-yl) propan-1-one
A solution of 3-phenylazetidine (266mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and 3-phenylpropionyl chloride (421.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 4). Collecting the fraction containing 3-phenyl-1- (3-phenylazetidin-1-yl) propan-1-one, and removing the solvent in vacuo to give 421mg of a colorless oil.
Example 25: 1- (3-benzyl azetidin-1-yl) -3-phenylpropan-1-one
A solution of 3-benzylazetidine (294mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and 3-phenylpropionyl chloride (421.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 5). The fractions containing 1- (3-benzylazetidin-1-yl) -3-phenylpropan-1-one were collected and the solvent was removed in vacuo to give 432mg of a colorless oil. LCMS ESI+:280.0(M+H)+,Rt=1.216min(LCMS 02)。
Example 26: 4-phenyl-1- (3-phenylazetidin-1-yl) butan-1-one
A solution of 3-phenylazetidine (266mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and 4-phenylbutyryl chloride (456.6mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 5). Collecting the nitrogen containing 4-phenyl-1- (3-phenyl nitrogen heterocycle)Butan-1-yl) butan-1-one fraction and the solvent was removed in vacuo to give 467mg of a colourless oil.
Example 27: 1- (3-benzyl azetidin-1-yl) -4-phenylbutan-1-one
A solution of 3-benzylazetidine (294mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and 4-phenylbutyryl chloride (456.6mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 5). The fractions containing 1- (3-benzylazetidin-1-yl) -4-phenylbutan-1-one were collected and the solvent was removed in vacuo to yield 479mg of a colorless oil. LCMS ESI+:294.2(M+H)+,Rt=1.305min(LCMS 02)。
Example 28: 1- (4-Benzylpiperazin-1-yl) pentan-1-one
A solution of 1-benzylpiperazine (352mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and valeryl chloride (301mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 1). Collecting a mixture containing 1-, (Fractions of 4-benzylpiperazin-1-yl) pentan-1-one and solvent removed in vacuo to give 416mg of a colourless oil.
LCMS ESI+:261.2(M+H)+,Rt=0.705min(LCMS 01)。
Example 29: 1- (4-benzylpiperazin-1-yl) hex-1-one
A solution of 1-benzylpiperazine (352mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and hexanoyl chloride (336.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 1). Fractions containing 1- (4-benzylpiperazin-1-yl) hex-1-one were collected and the solvent was removed in vacuo to give 453mg of a colorless oil.
1H-NMR(D6-DMSO, 400MHz) δ: 7.31(m, 4H), 7.36(m, 1H), 3.44(m, 2H), 2.30(m, 8H), 1.49(m, 2H), 1.26 (width s, 6H), 0.85(t, 3H).
LCMS ESI+:275.2(M+H)+,Rt=0.794min(LCMS 01)。
Example 30: 1- (2-phenylmorpholin-4-yl) pentan-1-one
A solution of 2-phenylmorpholine (326mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and pentane was addedAcid chloride (301mg, 2.5mmol, 1.25eq) and triethylamine (417 μ L, 3mmol, 1.5 eq). After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 4). Fractions containing 1- (2-phenylmorpholin-4-yl) pentan-1-one were collected and the solvent was removed in vacuo to give 412mg of a colourless oil.
1H-NMR(D6-DMSO,400MHz)δ:7.37(m,5H),4.36(m,2H),3.98(bs,1H),3.85(m,1H),3.55(m,1H),3.14(m,1H),2.66(m,1H),2.35(m,2H),1.50(bs,2H),1.32(bs,2H),0.89(t,3H)。
LCMS ESI+:248.2(M+H)+,Rt=1.138min(LCMS 02)。
Example 31: 1- (2-phenylmorpholin-4-yl) hex-1-one
A solution of 2-phenylmorpholine (326mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and hexanoyl chloride (336.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 4). Fractions containing 1- (2-phenylmorpholin-4-yl) hex-1-one were collected and the solvent was removed in vacuo to give 437mg of a colorless oil.
1H-NMR(D6-DMSO,400MHz)δ:7.38(m,5H),4.36(m,2H),3.98(bs,1H),3.84(m,1H),3.55(m,1H),3.13(m,1H),2.66(m,1H),2.35(m,2H),1.51(bs,2H),1.28(bs,4H),0.87(bs,3H)。
Example 32: 1- (3-phenylpyrrolidin-1-yl) pentan-1-one
A solution of 3-phenylpyrrolidine (294mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and valeryl chloride (301mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3mmol, 1.5eq) were added. After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 4). The fractions containing 1- (3-phenylpyrrolidin-1-yl) pentan-1-one were collected and the solvent was removed in vacuo to yield 378mg of a colorless oil.
1H-NMR(D6-DMSO/CCl4400MHz) δ: 7.26(m, 5H), 3.84(m, 1H), 3.65(m, 1H), 3.45(m, 3H), 2.23 (width s, 3H), 2.00(m, 1H), 1.55(m, 2H), 1.36(m, 2H), 0.95(m, 3H).
LCMS ESI+:232.2(M+H)+,Rt=1.192min(LCMS 02)。
Example 33: 1- (3-phenylpyrrolidin-1-yl) hex-1-one
A solution of 3-phenylpyrrolidine (294mg, 2mmol) in anhydrous dichloromethane (6mL) was cooled in an ice bath and hexanoyl chloride (336.5mg, 2.5mmol, 1.25eq) and triethylamine (417. mu.L, 3 mmol) were added1.5 eq). After stirring at room temperature for 6 hours, the reaction mixture was diluted with dichloromethane (10 mL). Subjecting the organic solution to 1M KHSO4、5%NaHCO3And brine sequentially. Through Na2SO4After drying, the dichloromethane is removed in vacuo and the crude product is purified by flash chromatography (silica gel, ethyl acetate/toluene 1: 4). Fractions containing 1- (3-phenylpyrrolidin-1-yl) hex-1-one were collected and the solvent was removed in vacuo to yield 405mg of a colorless oil.
1H-NMR(D6-DMSO/CCl4400MHz) δ: 7.16(m, 5H), 3.94(m, 1H), 3.60(m, 1H), 3.32(m, 3H), 2.24 (width s, 3H), 1.95(m, 1H), 1.52(m, 2H), 1.28(m, 4H), 0.98(m, 3H).
LCMS ESI+:246.2(M+H)+,Rt=1.274min(LCMS 02)。
Claims (1)
1. A pharmaceutical composition comprising a compound selected from the group consisting of:
。
2. The pharmaceutical composition of claim 1, comprising a compound selected from the group consisting of:
。
3. The pharmaceutical composition of claim 1 wherein the compound selected from examples 1, 3, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 25, 27, 28, 29, 30, 31, 32 and 33 and ethionamide are present in two separate pharmaceutical unit dosage forms.
4. The pharmaceutical composition of claim 2 wherein the compound selected from examples 15, 16 and 17 and ethionamide are present in two separate pharmaceutical unit dosage forms.
5. Use of a compound selected from examples 1, 3, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 25, 27, 28, 29, 30, 31, 32 and 33 according to claim 1 and ethionamide in the manufacture of a medicament for the treatment of tuberculosis and related diseases.
6. Use of a compound selected from examples 15, 16 and 17 according to claim 2 and ethionamide in the manufacture of a medicament for the treatment of tuberculosis and related diseases.
Applications Claiming Priority (3)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP09163765.2 | 2009-06-25 | ||
| EP09163765 | 2009-06-25 | ||
| PCT/EP2010/059044 WO2010149761A1 (en) | 2009-06-25 | 2010-06-25 | Composition for treatment of tuberculosis |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| HK1166280A1 HK1166280A1 (en) | 2012-10-26 |
| HK1166280B true HK1166280B (en) | 2015-07-31 |
Family
ID=
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| CN102458393B (en) | Composition for treatment of tuberculosis | |
| US11339130B1 (en) | Calpain modulators and therapeutic uses thereof | |
| TW409125B (en) | Antivirally active heterocyclic azahexane derivatives | |
| KR20050071472A (en) | Process for preparing quinolin antibiotic intermediates | |
| CA2860187C (en) | Cyclic amide derivatives as inhibitors of 11-beta-hydroxysteroid dehydrogenase and uses thereof | |
| CA2734415A1 (en) | Picolinamide derivatives as kinase inhibitors | |
| US20130289027A1 (en) | Isoquinoline Compounds And Methods For Treating HIV | |
| JP2013056926A (en) | Aza-peptide protease inhibitor | |
| AU2019355218B2 (en) | Monomer and multimeric anti-HBV agents | |
| KR20100041798A (en) | Novel hiv reverse transcriptase inhibitors | |
| JP2020527602A (en) | RAD51 inhibitor | |
| JP2019501919A (en) | Sulfamide derivative and its production method and application | |
| US20220363686A1 (en) | Novel 6,7-dihydro-4h-pyrazolo[1,5-a]pyrazine indole-2-carboxamides active against the hepatitis b virus (hbv) | |
| US20230112832A1 (en) | Chemokine CXCR4 Receptor Modulators and Uses Related Thereto | |
| CA3226724A1 (en) | Cyanopyridine and cyanopyrimidine bcl6 degraders | |
| EP3668856B1 (en) | Novel tetrazole compounds and their use in the treatment of tuberculosis | |
| AU2010275375A1 (en) | Spectinamides as anti-tuberculosis agents | |
| TW202229309A (en) | Compositions and methods for treatment of coronavirus infection | |
| TW200529809A (en) | HIV protease inhibiting compounds | |
| WO2014053666A1 (en) | Inhibitors of viral replication, their process of preparation and their therapeutical uses | |
| HK1166280B (en) | Composition for treatment of tuberculosis | |
| CN105820163A (en) | Substituted 1,3-miscellaneous azole compound, preparation method thereof, pharmaceutical composition containing substituted 1,3-miscellaneous azole compound and purpose thereof | |
| CN114957235A (en) | Benzothiazinone derivative and preparation method and application thereof | |
| AU2011265439B2 (en) | Picolinamide derivatives as kinase inhibitors | |
| US20250188092A1 (en) | Rifamycin analogs |