OA16265A - Anti-inflammatory agents. - Google Patents

Anti-inflammatory agents. Download PDF

Info

Publication number
OA16265A
OA16265A OA1201200503 OA16265A OA 16265 A OA16265 A OA 16265A OA 1201200503 OA1201200503 OA 1201200503 OA 16265 A OA16265 A OA 16265A
Authority
OA
OAPI
Prior art keywords
group
compound
caprolactam
lactam
amino
Prior art date
Application number
OA1201200503
Inventor
David John Grainger
David John Fox
Original Assignee
Cambridge Enterprise Limited
Boehringer Ingelheim International Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cambridge Enterprise Limited, Boehringer Ingelheim International Gmbh filed Critical Cambridge Enterprise Limited
Publication of OA16265A publication Critical patent/OA16265A/en

Links

Abstract

Disclosed herein are methods of preventing or treating inflammatory diseases using 3aminolactam compounds, and sulfonamide analogs thereof, each with aromatic "tail groups". Compounds as defined by formulae (I) and (I'), and the medical uses of the compounds, are described herein.

Description

The invention relates to aryl substituted 3-aminolactam dérivatives and their use in preventing or treating inflammatory diseases.
Inflammation is an important component of physiologîcal host defence. Increasingly, however, it is clear that temporally or spatially inappropriate inflammatory responses play a part in a wide range of diseases, including those with an obvious leukocyte component (such as autoimmune diseases, asthma or atherosclerosis) but also in diseases that hâve not traditionally been considered to involve leukocytes (such as osteoporosis or Alzheimer’s disease).
The chemokines are a large family of signal ling molécules with homology to interleukin-8 which hâve been implicated in regulating leukocyte trafïickîng both in physiologîcal and pathological conditions. With more than fifty ligands and twenty receptors involved in chemokine signalling, the System has the requisite information density to address leukocytes through the complex immune regulatory processes from the bone marrow, to the periphery, then back through secondary lymphoid organs. However, this complexity of the chemokine System has at first hindered pharmacological approaches to modulatîng inflammatory responses through chemokine receptor blockade. It has proved difficult to détermine which chemokine receptor(s) should be inhibîted to produce therapeutic benefit in a gîven inflammatory 20 disease.
More recently, a family of agents which block signalling by a wide range of chemokines simultaneously has been described (see Reckless et al., Biochem J. (1999) 340: 803-811). The first such agent, a peptide termed “Peptide 3”, was found to inhibit leukocyte migration induced by 5 different chemokines, while leaving migration in response to other chemoattractants (such as fMLP or TGF-beta) unaltered. This peptide, and its analogs such as NR58-3.14.3 (i.e. c(DCys-DGln-DIleDTrp-DLys-DGln-DLys-DPro-DAsp-DLeu-DCys)-NH2 [SEQ ID NO: 1]), are collectively termed “Broad Spectrum Chemokine Inhibitors” (BSCIs). Grainger et al. (2003, Biochem. Pharm. 65: 1027-1034) hâve subsequently shown BSCIs to hâve potentîally useful anti-inflammatory activity in a range of animal models of diseases. Interestingly, simultaneous blockade of multiple chemokines is not apparently / associated with acute or chronic toxîcity, suggesting this approach may be a useful strategy for developing new anti-inflammatory médications with similar benefits to steroids but with reduced side-effects. This bénéficiai risk:benefit profile most likely results from the unexpected mechanism of action of these compounds (see International Patent Appl. No. PCT/GB2010/000354 in the name of Cambridge Enterprise Limited filed 28 February 2010, and International Patent Appl. No. PCT/GB2010/000342 in the name of Cambridge Enterprise Limited filed 26 February 2010).
However, peptides and peptoid dérivatives such as N R5 8-3.14.3, may not be optimal for use in vivo. They are qui te expensive to synthesise and hâve relatively unfavourable pharmacokinetic and pharmacodynamie properties. For example, NR583.14.3 is not orally bioavailable and is cleared from blood plasma with a half-life period of less than 30 minutes after intravenous injection.
Two parallel strategies hâve been adopted to identify novel préparations that retain the anti-inflammatory properties of peptide 3 and NR58-3.14.3, but hâve improved characteristics for use as pharmaceutîcals. Fîrstly, a sériés of peptide analogs hâve been developed, some of which hâve longer plasma half-lives than NR58-3.14.3 and which are considerably cheaper to synthesise (see for example W02009/017620). Secondly, a detailed structure: activity analysis of the peptides has been carried out to identify the key pharmacophores and design small non-peptidic structures which retain the bénéficiai properties of the original peptide.
This second approach yielded several structurally distinct sériés of compounds that retained the anti-inflammatory properties of the peptides, including 16-amino and 16aminoalkyl dérivatives of the alkaloid yohimbine, as well as a range of N-substituted 3-aminoglutarimides, identified from a small combînatorial library (see Fox étal., 2002, J Med Chem 45: 360-370; WO 99/12968 and WO 00/42071). Ail of these compounds are broad-spectrum chemokine inhibitors that retain selectivity over nonchemokine chemoattractants, and a number of them hâve been shown to block acute inflammation in vivo.
The most potent and sélective of the above-mentioned aminoglutarimides was (S)-3(undec-lO-enoyl)-aminoglutarimide (NR58,4), which inhibited chemokine-induced d migration in vitro with an ED50 of 5nM. This compound was orders of magnitude more potent than 3-aminoglutarimides with more complex acyl side chains (such as benzoyl or tert-butyloxo (Boc) groups). As a resuit, subséquent studies of aminoglutarimide and aminolactam BSCIs hâve focussed almost exclusively on compounds with simple linear and branched alkyl side chains.
However, further studies revealed that the aminoglutarimide ring was susceptible to enzymatîc ring opening in sérum. Consequently, for some applications (for example, where the inflammation under treatment is chronîc, such as in autoimmune diseases) these compounds may not hâve optimal properties, and a more stable compound with similar anti-inflammatory properties may be superior.
As an approach to identifying such stable analogs, various dérivatives of (S)- 3(undec-lO-enoyl)-aminoglutarimide hâve been tested for their stability in sérum. One dérivative, the 6-deoxo analog (S)-3-(undec-IO-enoyl)-tetrahydropyridin-2-one, is completely stable in human sérum for at least 7 days at 37°C, but has consîderably reduced potency compared with the parental molécule.
One such family of stable, broad spectrum chemokine inhibitors (BSCIs) are the 3amino caprolactams, with a seven-membered monolactam ring (see, for example, W02005/053702 and W02006/016l 52). However, further useful anti-inflammatory compounds hâve also been generated from other 3-aminolactams with different ring size (see for example W02006/134385). Other modifications to the lactam ring, including introduction of heteroatoms and bicyclolactam ring Systems, also yield compounds with BSCI activity (see, for example, W02006/018609 and W02006/085096).
In general, these earlier studies hâve demonstrated that the BSCI activity is conferred on the molécule by the cyclic “head group” (a 3-amino lactam or imide) and defined, to an extent, the structural limitations for activity (for example, bulky substituents on the ring nitrogen are detrimental for activity, but variations in ring size hâve little impact). To be active as a BSCI, this “head group” must hâve an acyl “tail group” attached. Compounds with a 3-amino group, either free or N-alkyl substituted, bearing a positive charge at physiological pH are completely inactive as BSCIs. Préviens -A disclosures have shown that this “tail group” can be linked to the “head group” through simple amide, sulfonamide, urea or carbamate linkers.
While the structure of the “head” group and linker are cri ti cal for B SCI activity, it has been shown that a wide variety of “tail groups” can be selected with out affecting the primary phannacology of the compound, at Ieast in vitro. As a resuit, modification of the “tail group” has been extensively used to optimise the physical and pharmaceutical properties of the compounds. Changes in the structure of the “tail group” can, for example, change the primary route of metabolism or excrétion, modify the phannacokinetics or oral bioavailability, and thus act as the primary déterminant of the ADME properties of a selected compound.
Although the universe of possible “tail groups” known to retain BSCI activity for suitable aminolactam “head groups” is very large, some “tail groups” have been described as preferred. In some cases, structural features of the “tail group” have been identified which increase the potency of BSCI activity of the aminolactam compound. The most obvious such example is the introduction of 2’,2’ disubstitution, with a tetrahedral sp3 arrangement at the 2’ carbon centre in the tail group (the so-called “key carbon”), which confers a I0-fold increase in potency as a BSCI, at Ieast in vitro, compared to a related compound lacking 2’2’-disubstitution. For example, 2'2'dimethyldodecenanoyl-3-aminocaprolactam is l O-fold more potent as a BSCI in the MCP-l induced THP-l cell migration than assay than dodecanoyl-3aminocaprolactam (as disclosed previously in W02005/053702), or indeed any other related compound with a linear alkyl “tail group”. The increased potency for branched alkyl “tail groups” is restricted to branching at the 2* position -3’3’dimethyldodecanoyl-3-aminocaprolactam is no more potent than the linear alkyl analogs.
In other cases, structural features of the “tail group” have been identified which are associated with improved ADME properties. For example, the pivoyl “tail group” of 2’2’-dimethylpropanoyl-3-aminovalerolactam contributes to the unexpected, and particularly favourable, pharmaceutical properties of this molécule (as disclosed previously in W02009/016390). In particular, the pivoyl group is résistant to metabolism, and therefore contributes to the unusually prolonged biological half-life of this compound.___
In marked contrast, other possible “tail groups” hâve generally been less preferred. For example, compounds with a planar (sp2) carbon centre at the 2’ position (such as dodec-2’,3’-enoyl-3-aminocaprolactam) hâve markedly lower potency as BSCIs than compounds with corresponding saturated alkyl “tail groups”. Similarly, the data from the original library of glutarimides suggested that aromatic rings at the 2-position were also substantîally less active (Fox et al., 2002, J Med Chem 45: 360-370). Taken together, these two findings hâve led to the reasonable assumption that aminolactams with aromatic “tail groups”, such as benzoyl or substituted benzoyl, would not be useful as BSCIs. As a resuit, previous disclosures of compounds with BSCI actîvity hâve ail excluded such aromatic “tail groups”.
The présent invention discloses a sériés of 3-aminolactam compounds with aromatic “tail groups”, as well as pharmaceutical compositions comprising the compounds, and medical uses of the compounds and compositions such as for the treatment of inflammatory diseases. Surprisingly, ail of the compounds as set out below hâve substantial BSCI actîvity (greater than either 2’,3’-unsaturated acyl 3-aminolactams or benzoylaminoglutarimides).
In one aspect of the invention, there is provided according to the invention is a compound of general formula (I), or a pharmaceutically acceptable sait thereof, for use in the treatment of an inflammatory disorder:
(») wherein n is an integer from l to 4;
k is an integer from 0 to 5, representing the number of groups substituting C2, Ci, C4,
C5 and/or Cf, of the benzyl ring; and
X are linear or branched groups substituting the benzyl ring independently selected from any one of the group consisting of: alkyl, haloalkyl, hydroxyalkyl, hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, carboxy, and halogen;
with the proviso that:
when on the benzyl ring C2, C5 and C(. are unsubstituted, and C4 is unsubstituted or is substituted with an hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, or halogen group, then C3 is substituted with a halogen group; and when on the benzyl ring C2, C5 and C& are unsubstituted, and C3 îs unsubstituted or is substituted with an alkyl, haloalkyl, hydroxyalkyl, hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl or carboxy group, then C4 is substituted with any one of the group consisting of: alkyl group, haloalkyl group, hydroxyalkyl group, and carboxy group.
The carbon atom at position 3 of the lactam ring is asymmetric and consequently, the compounds according to the présent invention hâve at least two possible enantiomeric forms, that is, the “R” and “S” configurations. The présent invention encompasses each of the two enantiomeric forms and ail combinations of these forms, including the racemîc “RS” mixtures. With a view to simplicity, when no spécifie configuration is shown in the structural formula, it should be understood that each of the two enantiomeric forms and their mixtures are represented.
Further provided is a compound of formula (Γ), or a pharmaceutically acceptable sait thereof, for use in the treatment of an inflammatory disorder:
wherein n, k and X are defined as for general formula (I) compounds above.
Compounds (!’), having the (Si-configuration at the stereocentre, are 5-100 fold more potent as a BSCIs than the (R)-enantiomer of the same compound.
The invention also provides the use of a compound of general formula (I), or a pharmaceutically acceptable sait thereof, in the manufacture of a médicament for the treatment of an inflammatory disorder:
wherein n is an integer from l to 4;
k is an integer from 0 to 5, representing the number of groups substituting (b, C3, C4, C5 and/or C& of the benzyl ring; and
X are linear or branched groups substituting the benzyl ring independently selected from any one of the group consisting of: alkyl, haloalkyl, hydroxyalkyl, hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, carboxy, and halogen;
with the proviso that:
when on the benzyl ring C2, C5 and Cfi are unsubstituted, and C4 is unsubstituted or is substituted with an hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, or halogen group, then C3 is substituted with a halogen group; and when on the benzyl ring C2, C5 and C6 are unsubstituted, and C3 is unsubstituted or is substituted with an alkyl, haloalkyl, hydroxyalkyl, hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl or carboxy group, then C4 is substituted with any one of the group consisting of: alkyl group, haloalkyl group, hydroxyalkyl group, and carboxy group.
Additionally provided is the use of a compound of formula (P), or a pharmaceutically acceptable sait thereof, in the manufacture of a médicament for the treatment of an inflammatory disorder:
(I’) wherein n, k and X are defïned as for general formula (I) above.
Certain compounds hâve been found to be novel per se. Thus, in another aspect of the invention, there is provided a compound of general formula (I):
wherein n is an înteger from l to 4;
k is an integer from l to 5, representing the number of groups substituting C2, C3, C4, C5 and/or Cé of the benzyl ring;
when n is l or 2, X are linear or branched groups independently selected from any one of the group consisting of: C7 or higher alkyl, haloalkyl with a C7 or higher alkyl group, hydroxyalkyl with a C7OThigher alkyl group, C7orgreater alkoxy, aminoalkyl with a C4 ür higher alkyl group, aminodialkyl with two C40rhlgher alkyl groups, and carboxy; and when n is 3 or 4, X are linear or branched groups independently selected from any one of the group consisting of: alkyl, haloalkyl, hydroxyalkyl, hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, carboxy, and halogen;
with the proviso that:
when n is 3 or 4 and on the benzyl ring C2, C5 and C6 are unsubstituted, and C4 is unsubstituted or is substituted with an hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, or halogen group, then C; is substituted with a halogen group;
when n is 3 or 4 and on the benzyl ring C2, C5 and C& are unsubstituted, and C3 is unsubstituted or is substituted with an alkyl, haloalkyl, hydroxyalkyl, hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl or carboxy group, then C4 is substituted with any one of the group consisting of: alkyl group, haloalkyl group, hydroxyalkyl group, and carboxy group; and when n=3, X is other than 4’-methoxy, 3’-trifluoromethyl, or 3*,4’,5’-trimethoxy, provided that the compound is none of the group consisting of: 3-(3'trifluoromethylbenzoylamino)-caprolactam, 3-(4'-methylbenzoylamino)-caprolactam, 3-(2'-amînobenzoylamino)-caprolactain, 3-(3',4'-dimethoxybenzoylamino)caprolactam, S-p’^'-di-teri-butyl -4'- hydroxybenzoylamino)-caprolactam, 3-(2’,4'dimethoxybenzoylamino)-caprolactam, 3-(3’-methoxybenzoylamino)-caprolactam, 3(4’-tnfluoromethylbenzoylamino)-caprolactam, 3-(2',3’,4,-trimethoxybenzoylamino)caprolactam, 3-(2,,6'-dîfluoromethylbenzoylamino)-caprolactam, 3-(2’fluoromethylbenzoylamino)-caprolactam, 3-(2’-amino-3’-hydroxy-4'methylbenzoylamino)-caprolactam, and 3-(3’,5’-dimethylbenzoylamino)-caprolactam.
Also provided is a compound of formula (P):
(Γ) wherein η, k and X are defined herein for general formula (I), provided that the compound is none of the group consisting of: (S)-3-(3’-trifluoromethylbenzoylamino)-caprolactam, (S)-3-(4’-methyIbenzoylamino)-caprolactam, (S)3-(4’-methoxybenzoylamino)-caprolactam, (S)-3-(2'-carboxybenzoylamino)caprolactam, and (S)-3-(3‘, 4’,5,-trimethoxybenzoylamino)-caprolactam.
W02007/0038669 teaches diarylamine-containing compounds and their use as moduclators of c-kit receptors. Various întermediate compounds are used in the synthesis of the diarylamine-containing compounds. Any overlap of the întermediate compounds îs hereby disclaîmed from the présent invention.
EP0462949 together with the related publication Angelucci et al., 1993, J Médicinal Chemistry 36: 1512-1519 teach 7-membered 3-acylamino lactams as enhancers of leaming and memory. Any overlap of spécifie compounds [such as (R)- and (S)-3-(3’trifluoromethylbenzoylamino)-caprolactam, (S)-3-(4’-methoxybenzoylamino)caprolactam, and (S)-3-(3’, 4',5'-trimethoxybenzoylamino)-caprolactam] or generic compounds (notably where n=3 in compounds of generic formulae (I) and/or (I’) according to the présent invention) mentioned in these documents is hereby disclaimed from the présent invention.
JP03206042 discloses the préparation of 5,6,7,8-tetrahydo-4H-thiazolo[5,4-b]azepine dérivatives with potassium channel activation activity, for use as antihypertensives. Various întermediate compounds (notably where n=3 in compounds of generic formulae (I) and/or (I’) according to the présent invention) are used in the synthesis of the dérivatives. Any overlap of the întermediate compounds is hereby disclaimed from the présent invention. ->
The prior art also discloses spécifie compounds, for example:
S-iT-methylbenzoylaminoi-caprolactam is disclosed in Uchikawa et al. ( 1996) Chemical & Pharmaceutical Bulletin 44: 2070-2077;
- 3-(2'-aminobenzoylamino)-caprolactam is disclosed in Uchikawa et al. ( 1994) J Heterocyclic Chemistry 31: 877-887;
(S)-3-(2,-carboxybenzoylamino)-caprolactam is disclosed in Belyaev (1995) Tetrahedron Letters 36: 439-440 ;
- 3-(3’-trifluoromethylbenzoylamino)-caprolactam (in both (S)- and (R)- forms) and (S)-3-(3', 4,,5'-trimethoxybenzoylamîno)-caprolactam are disclosed in EP462949A1; 3-(3'-trifluoromethylbenzoylamino)-caprolactam is also disclosed in EP351856A2;
- S-ÎS'^'-dimethoxybenzoylaminoJ-caprolactam, 3-(3',5’-di-ter/-butyl -4’hydroxybenzoylamino)-caprolactam, 3-(2,,4'-dimethoxybenzoylamino)caprolactam, 3-(3'-methoxybenzoylamino)-caprolactam, 3-(4'trifluoromethylbenzoylamino)-caprolactam, 3-(2’,3',4'trimethoxybenzoylamino)-caprolactam, 3-(2',6’-difluoromethylbenzoylamino)caprolactam, and 3-(2'-fluoromethylbenzoylamino)-caprolactam are disclosed in JP03206042A;
3-(2’-amino-3’-hydroxy-4'-methylbenzoylamino)-caprolactam is disclosed in Kameda et al. (1968) Chemical & Pharmaceutical Bulletin 16: 480-485; and
3-(3’5’-dimethylbenzoylamino)-caprolactam is disclosed in the Aurora Screening Library Catalogue published on 10 March 2010 (Order No. K.07.167.701 ; CHEMCATS Acc. NO. 0015557046).
However, none of the above prior art compounds hâve been shown to hâve BSCI activity, or to be useful for the treatment of inflammatory diseases. As a resuit, compounds disclosed in the prior art documents mentioned herein in no way teach or suggest our unexpected ftnding that the class of aryl-substituted aminolactams and analogs as defined herein hâve useful BSCI activity, and the prior art compounds are hereby disclaimed. _
In another aspect of the invention, there is provided a pharmaceutical composition comprising, as active ingrédient, a compound per se as defined above, or a pharmaceutically acceptable sait thereof, and at least one pharmaceutically acceptable excipient and/or carrier.
By pharmaceutically acceptable sait is meant in particular the addition salts of înorganic acids such as hydrochloride, hydrobromide, hydroîodide, sulphate, phosphate, dîphosphate and nitrate or of organic acids such as acetate, maleate, fumarate, tartrate, succinate, citrate, lactate, methanesulphonate, p-toluenesulphonate, palmoate and stéarate. Also within the scope of the présent invention, when they can be used, are the salts formed from bases such as sodium or potassium hydroxide. For other examples of pharmaceutically acceptable salts, reference can be made to “Sait sélection for basic drugs” (1986) Int. J. Pharm. 33: 201-217.
The pharmaceutical composition can be in the form of a solid, for example powders, granules, tablets, gelatîn capsules, liposomes or suppositories. Appropriate solid supports can be, for example, calcium phosphate, magnésium stéarate, talc, sugars, lactose, dextrin, starch, gelatîn, cellulose, methyl cellulose, sodium carboxymethyl cellulose, polyvinylpyrrolidine and wax. Other appropriate pharmaceutically acceptable excipients and/or carriers will be known to those skilled in the art.
The pharmaceutical compositions according to the invention can also be presented in liquid form, for example, solutions, émulsions, suspensions or syrups. Appropriate liquid supports can be, for example, water, organic solvents such as glycerol or glycols, as well as their mixtures, in varying proportions, in water.
Exemplar compounds according to general formula (I) and formula (P) for medical uses according to the invention may be selected from the group consisting of:
(S)-3-(4'-methylbenzoylamino)'Caprolactam, and (5)-3-(3 ’ ,5 '-dimethylbenzoylami no)-caprol actam, and pharmaceutically acceptable salts thereof.
Exemplarper se compounds of the invention according to general formula (I’), and/or exemplar compounds according to general formula (I) and formula (I’) for medical uses according to the invention, may be selected from the group consisting of:
(5)-3-Fl uoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-2-Fluoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-4-Fluoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-N-(2-Oxopiperidin-3-yl)-4-(trifluoromethyl)benzamide, (5)-N-(2-Oxopiperidin-3-yl)-3-(trifluoromethyl)benzamÎde, (5)-N-(2-Oxopiperidin-3-yl)-2-(trifluoromethyl)benzamide, (5)-2,3-difluoro-N-(2-oxopiperidin-3-yl)benzamide, (5f)-2,4-difluoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-2,5-di fl uoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-2,6-di fl uoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-3,4-di fl uoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-3,5-difluoro-N-(2-oxopiperidin-3-yl)benzamide, (S)-3-(3’-Butylbenzoylamino)-azepan-2-one, (S)-3-(4’-Ethylbenzoylamino)-tetrahydropyridin-2-one, (S)-3-(4'-Butylbenzoylamino)-tetrahydropyridin-2-one, (S)-3-(4'-tert-Butylbenzoylamino)-tetrahydropyridin-2-one, and (S)-3-(4'-Hexylbenzoylamino)-tetrahydropyridin-2-one, and pharmaceutically acceptable salts thereof.
The compound (5)-4-Fluoro-N-(2-oxopiperidîn-3-yl)benzamide is also known as (S)3-(4’-fluorobenzoylamino)-tetrahydropyridin-2-one ( see Example 3 below).
Exemplarper se compounds of the invention according to general formula (I) or (I’), and/or exemplar compounds according to general formula (I) and formula (I’) for medical uses according to the invention, may be selected from the group consisting of:
(S)-3 -(4’- Ethylbenzoyl amino)-azepan-2-one, (S)-3-(4'-Butylbenzoylamino)-azepan-2-one, (S)-3-(4'-tert-Bütylbenzoyl amino)-azepan-2-one, (S)-3 -(4’- H exylbenzoyl amino )-azepan-2-one.
(S)-3-(4'-Octylbenzoylamino)-azepan-2-one, and (S)-3-(4'-Octylbenzoylamino)-tetrahydropyridin-2-one, and pharmaceutically acceptable salts thereof.
Ex.emplar compounds according to general formula (I) for medical uses according to the invention, may be selected from the group consisting of:
( R)-3 -(4'-B uty lbenzoylami no)-tetrahydropyridi n-2-one, (R)-3-(4'-tert-Butylbenzoytemino)-tetrahydropyridin-2-one, and (R)-3-(4'-Hexylbenzoylamino)-tetrahydropyridin-2-one, and pharmaceutically acceptable salts thereof.
Exempter per se compounds of the invention according to general formula (I), and/or exempter compounds according to general formula (I) for medical uses according to the invention, may be (R)-3-(4'-Octylbenzoylamino)-tetrahydropyridin-2-one or a pharmaceutically acceptable sait thereof.
The compounds (R)-3-(4'-Butylbenzoytemino)-tetrahydropyridin-2-one, (R)-3-(4'-fôri- Buty lbenzoylami no )-tetrahydropyridin-2-one, (R)-3-(4'-Hexylbenzoylamino)-tetrahydropyridin-2-one, and (R)-3-(4'-Octylbenzoylamino)-tetrahydropyridin2-one, and pharmaceutical salts of each, are a further aspect of the invention.
According to the invention, inflammatory disorders (which term is used herein interchangeably with “inflammatory disease”) intended to be prevented or treated by the compounds of formula (I) or (I*), or pharmaceutically acceptable salts thereof or pharmaceutical compositions or médicaments containing them as active ingrédients, include notably:
autoimmune dîseases, for example such as multiple sclerosis, rheumatoid arthritis, lupus, irritable bowel syndrome, Crohn’s disease;
vascular disorders including stroke, coronary artery dîseases, myocardial infarction, unstable angina pectoris, atherosclerosis or vasculitis, e. g., Behçet’s syndrome, giant cell arteritis, polymyalgia rheumatica, Wegener's granulomatosis,
Churg-Strauss syndrome vasculitis, Henoch-Schônlein purpura and Kawasaki disease;
asthma, and related respiratory disorders such as allergie rhinitis and COPD;
- organ transplant rejection and/or delayed graft or organ function, e.g. in rénal transplant patients;
psoriasis;
skin wounds and other fibrotic disorders including hypertrophie scarring (keloid formation), adhesion formations following general or gynaecologîcal surgery, lung fibrosis, liver fibrosis (including alcoholic liver disease) or kidney fibrosis, whether idiopathic or as a conséquence of an underlying disease such as diabètes (dîabetic nephropathy); or
- allergies.
The infiammatory disorder may be selected from the group consisting of autoimmune diseases, asthma, rheumatoid arthritîs, a disorder characterised by an elevated TNF-ct level, psoriasis, allergies, multiple sclerosis, fibrosis (including dîabetic nephropathy), and formation of adhesions.
The above clinical indications fall under the general définition of infiammatory disorders or disorders characterized by elevated TNFa levels.
In one aspect of the invention, merely in order to circumvent any potentially confiicting prior art (for example as noted above), the term infiammatory disorder may exclude cognitive disorders such as Alzheimer’s disease and/or memory loss.
Compounds of formula (I) or (Γ) are particularly useful for local delîvery, and also for the préparation of médicaments for local delîvery, including creams and ointments for topical delîvery, powders, aérosols or émulsions for inhaled delîvery, and solutions or émulsions for injection. Pharmaceutical compositions containing one or more excipients suitable for such local delîvery are therefore envisaged, and subsequently claimed. —
Also provided according to the invention is a method of treatment, amelioration or prophylaxis of the symptoms of an inflammatory disease (including an adverse inflammatory reaction to any agent) by the administration to a patient of an antiinflammatory amount of a compound, pharmaceutical composition or médicament as defîned herein.
Administration of a compound, composition or médicament according to the invention can be carried out by topical, oral, parentéral route, by intramuscular injection, etc.
The administration dose envisaged for a compound, composition or médicament according to the invention is comprised between 0.1 mg and 10 g dependîng on the formulation and route of administration used.
The invention further encompasses a library consisting of éléments ail of which hâve structures according to the formula (I) or (P), and hence which ail hâve antiinflammatory activity, useful for screening compounds for novel or improved properties in a particular assay of anti-inflammatory activity.
The invention includes compounds, compositions and uses thereof as defîned, wherein the compound is in hydrated or solvated form. Unless specified otherwise, compounds of the invention include tautomers, resolved enantiomers, resolved diastereomers, racemic mixtures, solvatés, métabolites, salts and prodrugs thereof, including pharmaceutically acceptable salts and prodrugs.
In any of the compounds described above, n may be 2. Altematively, n may be 3.
X may be haloalkyl, for example trifluoromethyl.
An exemplar group of compounds per se and/or for medical use according to any aspect of the invention is selected from among compounds according to formula (I) or (F) where X is halogen or haloakyl and where k is between 1 and 3. For example, X may be fluoro or fluoroalkyl (such as trifluoromethyl) and k may be between 1 and 3.
Where permissible according to the formulae herein, the benzyl ring may be monosubstituted with a group X as defîned above (i.e. k = 1). For example, the benzyl ring may be monosubstituted with an alkyl group (such as other than para-methyl or other than Ci-6 alkyl), haloalkyl (such as trifluoromethyl, for example para- X tri fl uoro methyl [i.e. 4’-trifluoromethyl], ortho-trifluoromethyl [i.e. 2’-triflouromethyl] or meta-trifluoromethyl [i.e. 3’-trifluoromethyl]). The benzyl ring may be monosubstîtuted with an haloalkyl other than a C|„6 haloalkyl. The benzyl ring may be monosubstituted with halogen. The benzyl ring may be monosubstîtuted with orthocarboxy [i.e. 2’-carboxy].
The single substitution group X may in particular be located in the meta (i.e. 3’-) position on the benzyl ring.
In one aspect, the above features for k=l apply when n=2.
Where permissible according to the formulae herein, n may be 3 and the benzyl ring may be monosubstituted with a group X as defined above (i.e. k = l ). For example, the benzyl ring may be monosubstituted with an alkyl group other than a C].& alkyl.
According to the invention, the compounds of general formula (I) or (I’) can be prepared using the processes described hereafter.
DEFINITIONS
The term “about” refers to an interval around the considered value. As used in this patent application, “about X” means an interval from X minus 10% of X to X plus 10% of X, and preferably an interval fforn X minus 5% of X to X plus 5% of X.
The use of a numerical range in this description is întended unambiguously to include within the scope of the invention ail îndividual integers within the range and ail the combinations of upper and lower limit numbers within the broadest scope of the given range. Hence, for example, the range of 0.1 mg to 10g specified in respect of (inter alla) a dose of a compound or composition of the invention to be used is intended to include ail doses between O.img and 10g and ail sub-ranges of each combination of upper and lower numbers, whether exemplified explicitly or not.
As used herein, the term “comprising” is to be read as meanîng or encompassing both comprising and consisting of. Consequently, where the invention relates to a “pharmaceutical composition comprising as active ingrédient” a compound, this terminology is intended to cover both compositions in which other active ingrédients — may be présent and also compositions which consist only of one active ingrédient as defined.
The term “alkyl” or “alkyl group” as used herein refers to a saturated linear or branched- chain monovalent hydrocarbon radical, for example of one to twenty carbon atoms, one to twelve carbon atoms, one to six carbon atoms, one to four carbon atoms, or as otherwise specified herein. Examples of alkyl groups include, but are not limited to, methyl (Me, -CH3), ethyl (Et, -CH2CH3), l-propyl (n-Pr, n-propyl, -CH2CH2CH3), 2-propyl (i-Pr, i-propyl, -CH(CHj)2), l-butyl (n-Bu, n- butyl, -CH2CH2CH2CH3), 2methyl-1-propyl (i-Bu, i-butyl, -CH2CH(CH3)2), 2-butyl (s- Bu, s-butyl, CH(CH3)CH2CH3), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CH3)3), 1-pentyl (n- pentyl, CH2CH2CH2CH2CH3), 2-pentyl (-CH(CH3)CH2CH2CH3), 3-pentyl (- CH(CH2CH3)2)5
2- methyl-2-butyl (-C(CHs)2CH2CH3), 3-methyl-2-butyl (- CH(CH3)CH(CH3)2), 3methyl-1 -butyl (-CH2CH2CH(CH3)2), 2-methyl-l -butyl (- CH2CH(CH3)CH2CH3), 1hexyl (-CH2CH2CH2CH2CH2CH3), 2-hexyl (- CH(CH3)CH2CH2CH2CH3), 3-hexyl (CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (- C(CHs)2CH2CH2CH3), 3-methyl-2pentyl (-CH(CH3)CH(CH3)CH2CH3), 4-methyl-2- pentyl (-CH(CH3)CH2CH(CH3)2),
3- methyl-3-pentyl (-C(CH3)(CH2CH3)2), 2-methyl-3- pentyl (CH(CH2CH3)CH(CH3)2), 2,3-dimethyl-2-butyl (-C(CH3)2CH(CH3)2), 3,3- dimethyl-2butyl (-CH(CH3)C(CH3)3, 1-heptyl, and 1-octyl.
The term “haloalkyl” or “haloalkyl group” as used herein refers to an alkyl group (as defined above) except that one or more or ail of the hydrogens of the alkyl group is replaced by a halogen, which replacement can be at any site on the alkyl, including the end. Examples include, but are not limited to, CH2F, CHF2, CF3, CH2CH2Fs CH2CHF2) CH2CF3, CHFCF3, CF2CF3, CH2C1, CHC12, CC13) CH2CH2C1, CH2CHC12, CH2CCI3, CHCICCI3, and CCI2CCI3.
The term “halogen” (which may be abbreviated to “halo”) or “halogen group” as used herein includes fluorine (F), bromine (Br), chlorine (Cl), and iodine (I).
The term “hydroxy” or “hydroxy group” dénotés the group “-OH”.
The term “hydroxyalkyl” or “hydroxyalkyl group” as used herein refers to an alkyl group (as defined above) except wherein one or more or ail of the hydrogens of the TO alkyl group îs replaced by an hydroxy group, which replacement can be at any site on the alkyl, including the end.
The term “alkoxy” or “alkoxy group” dénotés an alkyl group as defined above attached via a divalent oxygen atom to the rest of the molécule. Examples include but are not limited to methoxy (-OCH3), ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tertbutoxy, pentoxy, isopentoxy, neopentoxy, hexoxy, and 3-methylpentoxy.
The term “amino” or “amino group” dénotés the group “-NH2”.
The term “aminoalkyl” or “aminoalkyl group” refers to an amino group in which one of the hydrogen atoms has been replaced by an alkyl group as defined above.
The term “aminodialkyl” or “aminodialkyl group” refers to an amino group in which both of the hydrogen atoms hâve been replaced by an alkyl group as defined above. The alkyl groups attached to the nitrogen atom may be different or the same.
The term “carboxy” or “carboxy group” dénotés the group “-C(O)OH”.
The term “benzyl ring” (also known as a “phenyl group”) refers to a 6 carbon aryl group in compounds of general formulae (I) or (I’) shown above. For the purposes of the general formulae of the présent invention, numbering to locate the carbon atoms C2-C6 within the benzyl ring is in a clockwise direction from Ci which is linked to the 3-aminolactam group. However, numbering of ring carbons with respect to one or more substituent groups on the benzyl ring for spécifie compounds follows the IUPAC rule that the second substituent in a clockwise or counter clockwise direction is afforded the lower possible location number. Where two or more substituents are présent in a spécifie compound, the IUPAC rule is that they are listed in alphabetical order. Location numbers on the ring are assigned according to the IUPAC rule to the substituents so that they hâve the lowest possible number (starting from Ci which is linked to the 3-aminolactam group), counting in either a clockwise or counterclockwise direction.
As would be understood by a person skilled in the art, where there are fewer than 5 groups substituting the benzyl ring in compounds of general formulae (I) or (I’), i.e.,
I9 where k=0, 1 2, 3 or 4, the or each unsubstituted position is occupied by a hydrogen atom.
Unless otherwise defined, all the technîcal and scientific terms used here hâve the same meaning as that usually understood by an ordinary specialist in the field to which this invention belongs. Similarly, all the publications, patent applications, all the patents and all other references mentioned here are incorporated by way of reference (where legally permissible).
Préparation of the compounds of general formula (I) or (1’1
All the compounds of general formula (I) or (I’) can be prepared easîly according to general methods known to the person skilled in the art.
Typicalîy, such compounds are made by coupling the “tail group” in the form of a suitably activated acid (such as an acid chloride) with the appropriate 3-aminolactam. Methods for the préparation of 3-aminolactams with 5,6,7 and 8 membered rings, encompassing all the compounds claimed herein, hâve been extensively described in the literature. For example, we hâve provided suitable methods for the préparation of 6-membered aminolactams from omithine (see W02009/016390) and 7-membered aminolactams from lysine (see W02005/053702), as well as methods for 5- and 8membered aminolactams (see W02006/134385). We hâve described in particular detail various synthesis routes to the 6-membered aminolactam, including processes suitable for scaling up the manufacture to Kg quantifies (W02009/016390). Various other methods for the synthesis of 3-aminolactams of various ring sizes hâve also been described in the literature (see for example Pellegata et al., 1978, Synthesis 614-616 and Boyle et al., 1979, J Org Chem 44:4841-4847), and any suitable method for the préparation of the aminolactam “head group” may be employed in accordance with the method of the présent invention.
In the second step, the 3-aminolactam product is reacted with an appropriate acid chloride, for example as previously described for 7-ring aminolactams (Fox et ai, 2005, J Med Chem 48: 867-74). This reaction may be carried out, for example, in chloroform or dichloromethane. The most preferred reaction solvent is dichloromethane, and is preferably carried out in the presence of a base, for example
NibCO;. The above reaction may be carried out at ambîent température (about 25 °C) or more generally at a température between 20 and 50 °C. The two reactions may be carried out independently, with séparation and purification of the 3-aminolactam between the reactions, or altematively, the reactions may be performed in a single vessel without purification ofthe 3-aminolactam prior to its derivatisation with acid chloride.
As noted previously (see W02009/016390) care must be exercised during the acylation reaction when preparing an enantiomerically pure compound, according to formula (Γ) by acylating an enantiomeriocally pure 3-aminolactam. In particular, the base, such as sodium carbonate, must be added slowly continually monitoring the pH of the reaction vessel to ensure that the pH of the reaction remains below pH 9.0 throughout. Excess basicity, for example due to rapid or excessive addition of sodium carbonate, increases the racémisation ofthe 3-aminolactam and yields enantiomerically impure product.
The following examples are presented in order to illustrate the above procedures and should in no way be considered to limit the scope of the invention.
FIGURES
Fig. 1 shows the chemical structure of various examples of compounds according to the inventions and reference examples; and
Fig. 2 . is a graph showing the results of a murine sub-lethal endotoxemia test. In the graph, column A shows data from a control group (1% CMC lOml/kg p.o.), and column B shows data from a group treated with lmg/kg p.o. (S)-4-Fluoro-N-(2oxopiperidin-3-yl)benzamide (a compound according to one embodiment ofthe présent invention - see also Example 3 below) The y-axis shows levels of TNF-α in pg/ml.
EXAMPLES
In the following further examples, 'H-NMR and 13C-NMR spectra were recorded on a Bruker Avance DRX 400 MHz fourier transform machine and l<sF-NMR spectra were recorded on a Bruker Avance DRX 300. Chemical shifts are given in ppm and coupling constants, J, are given in Hz to the nearest 0.5. IR spectra were recorded on an Avatar 320. HRMS data was gained via an Esquire 2000. [a]n values were recorded on an optical activity AA 1000 polarimeter set at 598 nm (Sodium D line). The samples were made using spectroscopic grade MeOH.
Reference Example 1: 3-(Benzoylamino)tetrahydropyridin-2-one:
3-aminotetrahydropyridin-2-one hydrochloride ( 10 mmol) and K2CO3 (30 mmol) were added to water (20 mL) and stirred. A solution of benzoyl chloride (10 mmol) in CH2C12 (10 mL) was added and the reaction was stirred overnight at room température in an inert atmosphère (using dinitrogen). The réaction was extracted with CH2C12 (3 x 50 mL), and the combined organic layers where then dried (Na2SO4) and reduced in vacuo to give a crude product which was recrystallised from CH2CI2 / petroleum ether (bp 40-60 °C) to give the product (1.62 g, 74%):
Vmax/cm'1 3250 (N-H, amide), 1664, 1633, 1538 (secondary CONH, lactam), 1605, 1578, 1486 (aromatic ring), 766, 715, 704, 690 (monosubstituted benzene ring).
'H NMR: Ôh (400MHz, CDCI3) 7.80 (2H, br d, J7.0, ort/io-H), 7.47-7.40 (1H, m, para-H), 7.42-7.39 (1H, m, C6H5-CONH), 7.40-7.31 (2H, m, meta-H), 6.78 (1H, brs, CONtf-CH2), 4.41 (1H, dt, J 11.5, 5.5, CH-CO), 3.36-3.23 (2H, m, C/NNH). 2.59 (1H, dq,J 13.0,4.5, NHCH-C/7?), 1.94-1.81 (2H, m, lactam CH2), 1.64 (1H, tt, .7 12.5, 8.0, NHCH-C/Λ)· I3C NMR: ôc (100MHz, CDCI3) 171.9 (lactam C=O), 167.4 (aryl C=O), 134.0 (zpsoC), 131.4 (ortho-Cf 128.3 (me/a-C), 127.0 (para-Cf 50.8 (CH-CO), 41.5 (CH2NH), 27.0 (lactam CH2), 20.9 (lactam CH2).
HRMS (+ESI) C12HUN2O2 + Na+: calcd 241.0947; found 241.0950.
Reference Example 2: 3-(BenzoyIamino)azepan-2-one:
3-aminoazepan-2-one hydrochloride ( 10 mmol) and K.2CO3 (30 mmol) were added to water (20 mL) and stirred. A solution of benzoyl chloride (10 mmol) in CH2Cl2 (10 mL) was added and the reaction was stirred ovemight at room température in an inert atmosphère (using dinitrogen). The reaction was extracted with CH2CI2 (3 x 50 mL), and the combined organic layers where then dried (NaiSCL) and reduced in vacuo to give a crude product which was recrystallised from CH2C12 / petroleum ether (bp 4060 °C) to give the product (1.59 g, 68%):
Vm^/cm'1 3244, 3202 (N-H, amide), 1660, 1642, 1536 (secondary CONH, lactam), 1601, 1578, 1536 (aromatic ring), 771, 707 (monosubstituted benzene ring).
’H NMR: δΗ (400MHz, CDCI3) 7.86-7.80 (2H, m, orrôo-H), 7.65 (IH, d, J4.0, C6H5CON/7), 7.52-7.45 (IH, m,para-H), 7.46-7.39 (2H, m, meta-H), 6.11 (IH, br s, CON//-CH2). 4.72 (IH, ddd, J 11.0, 5.5, 1.5, CH-CO), 3.49-3.39 (2H, m, C/LNH).
2.25 (IH, br d, J 13.5, lactam CH2), 2.08-1.98 (IH, m, lactam CH2), 1.94-1.82 (2H, m, lactam CH2), 1.62-1.49 (IH, m, lactam CH2), 1.49-1.36 (IH, m, lactam CH2).
13C NMR: Ôc (100MHz, CDCI3) 175.9 (lactam OO), 166.4 (aryl C=O), 134.3 (ipsoC), 131.7 (ortho-C), 128.7 (meta-Cf 127.2 (para-C), 52.7 (CH-CO), 42.3 (CH2-NH), 31.7 (lactam CH2), 29.0 (lactam CH2), 28.1 (lactam CH2).
HRMS (+ESI) CI2H14N2O2 + H+: calcd 233.1285; found 233.1283.
Reference Example 3: 3-(4’-Methylbenzoylamino)tetrahydropyridin-2-one:
3-aminotetrahydropyridin-2-one hydrochloride (10 mmol), K.2CO3 (30 mmol) and 4methylbenzoyl chloride (10 mmol) were reacted according to the above procedure to give the product (1.62 g, 78%):
ν,,^/cm'1 3306, 3203 (N-H, amide), 1669, 1649 (secondary CONH, lactam), 1612, 1489 (aromatic ring), 842, 810 (para-disubstîtuted benzene ring).
lH NMR: Ôh (400MHz, CDCI3) 7.69 (2H, br d, J 8.0, orrôo-H), 7.20 (2H, br d, J 8.0, meta-W), 7.13 (IH, br d, J4.0, CON/Z-CH2), 6.03 (IH, br s, C7H7-CON/T), 4.41 (IH, >
dt, J l l.O, 5.5, CH-CO), 3.41-3.30 (2H, m, Œ2NH), 2.72 (ÎH, dq, J 13.0, 4.5, lactam
CH2), 2.36 (3H, s, CH3), 2.05-1.90 (2H, m, lactam CH2), 1.68-1.54 (1H, m, lactam
CH2).
13C NMR: Ôc (100MHz, CDCI3) 172.3 (lactam C=O), 167.7 (aryl OO), 142.1 fipsoC), 131.2 (para-C), 129.3 (aromatic-CH), 127.3 (aromatic-CH), 51.1 (CH-CO), 41.8 (CH2-NH), 27.4 (lactam CH2), 21.7 (CH3), 21.2 (lactam CH2).
HRMS (+ESI) Ci3Hl6N2O2 + Na+: calcd 255.1104; found 255.1104.
Référencé Example 4: 3-(4’-Methylbenzoylamino)azepan-2-one :
3-aminoazepan-2-one hydrochloride (10 mmol), K2CO3 (30 mmol) and 4methylbenzoyl chloride (10 mmol) were reacted according to the above procedure to give the product (1.63 g, 74%):
vmax/cnf1 3265, 3219 (N-H, amide), 1663, 1647 (secondary CONH, lactam), 1607, 1570, 1505 (aromatic ring), 838, 823 (para-disubstituted benzene ring).
lH NMR: δΗ (400MHz, CDCI3) 7.72 (2H, br d, J 8.0, οτϊΛο-Η), 7.58 (1H, br d, 74.50, CON/7-CH2), 7.21 (2H, d, J 8.0, meta-H), 5.98 ( 1 H, br s, C7H7-CONZ7), 4.69 (1H, ddd, 7 11.0, 5.5, 2.0, CH-CO), 3.40-3.321 (2H,m, Cf/2NH), 2.37 (3H, s, CH3), 2.23 ( 1 H, br d, lactam CH2), 2.08-1.99 ( 1 H, m, lactam CH2), 1.96-1.83 (2H, m, lactam CH2), 1.60-1.51 (2H, m, lactam CH2).
13C NMR: ôc (100MHz, CDCI3) 176.1 (lactam C=O), 166.3 (aryl C=O), 142.0 (ipsoC), 131.5 (para-C), 129.3 (aromatic-CH), 127.2 (aromatic-CH), 52.6 (CH-CO), 42.2 (CH2-NH), 31.7 (lactam CH2), 29.0 (lactam CH2), 28.1 (lactam CH2), 21.6 (CH3).
HRMS (+ESI) C14Hi8N2O2 + H+: calcd 247.1441; found 247.1453.
Reference Example 5: 3-(4’-Chlorobenzoylamino)tetrahydropyridin-2-one:
3-aminotetrahydropyridin-2-one hydrochloride (l 0 mmol), K2CO3 (30 mmol) and 4chlorobenzoyl chloride (ÎO mmol) were reacted according to the above procedure to give the product (0.87 g, 39%):
Vina^/cm'1 3295, 3202 (N-H, amide), 1668, 1648, 1629 (secondary CONH, lactam), 1594, 1486 (aromatic ring), 859, 845, 808, (para-disubstituted benzene ring), 750, 656 (C-Cl).
lH NMR: δΗ (400MHz, CDC13) 7.74 (2H, br d, J 8.5, ori/20-H), 7.38 (2H, br d, J 8.5, meta-H), 7.14 (1H, br d, J3.0, C6H4C1-CONH), 5.83 (IH, br s, CONH-CH2), 4.40 (1H, dt, 7 11.0, 5.5, CH-CO), 3.44-3.33 (2H, m, CH2NH), 2.73 (1H, dq, J 13.0, 4.5, NHCH-CZ/?), 2.05-1.93 (2H, m, lactam CH2), 1.66-1.56 (1H, m, lactam CH2).
I3C NMR: Ôc (100MHz, CDCI3) 180.2 (lactam OO), 171.8 (aryl C=O), 138.1 (ipsoC), 132.7 (C-Cl), 129.1 (aromatic-CH), 128.5 (aromatic-CH), 51.4 (CH-CO), 42.0 (CH2-NH), 27.2 (lactam CH2), 21.2 (lactam CH2).
HRMS (+ESI) Ci2H]3ClN2O2 + Na+: calcd 275.0558; found 275.0559.
Reference Ex ample 6: 3-(4’-Chlorobenzoylaniino)azepan-2-one:
3-aminoazepan-2-one hydrochloride (10 mmol), K2CO3 (30 mmol) and 4chlorobenzoyl chloride ( 10 mmol) were reacted according to the above procedure to give the product ( 1.79 g, 75%);
Vmax/crn'1 3243, 3200 (N-H, amide), 1662, 1643 (secondary CONH, lactam), 1595, 1484 (aromatic ring), 856, 841,819 (para-disubstituted benzene ring), 776, 732 (CCl).
'H NMR: ôh (400MHz, CDC13) 7,76 (2H, br d, J 8.5, ortho-H), 7.59 ( 1 H, br d, J 4.0, C6H4C1-CONH), 7.39 (2H, br d, J 8.5, meta-W), 6.00 ( 1 H, br s, CON7/-CH,), 4.67 (1H, ddd, 7 11.0, 5.5, 1.5, CH-CO), 3.39-3.22 (2H, m, CH2NH), 2.22 (1H, br d, 7 14.0, lactam CH2), 2.09-2.00 (IH, m, lactam CH2), 1.96-1.82 (2H, m, lactam CH2), 1.601.36 (2H, m, lactam CH2).
l3C NMR: ôc (100MHz, CDC13) 175.9 (lactam C=O), 165.3 (aryl C=0), 137.9 (ipsoC), 132.7 (C-Cl), 128.9 (aromatic-CH), 128.7 (aromatic-CH), 52.8 (CH-CO), 42.3 (CH2-NH), 31.7 (lactam CH2), 29.0 (lactam CH2), 28.1 (lactam CH2).
HRMS (+ESI) C]3H1SC1N2O2 + H+: calcd 267.0895; found 267.0890.
Reference Example 7: 3-(4*-Methoxybenzoylamino)tetrahydropyridin-2one:
3-aminotetrahydropyridin-2-one hydrochloride (10 mmol), K2CÛ3 (30 mmol) and 4methoxybenzoyl chloride (10 mmol) were reacted according to the above procedure to give the product (2.18 g, 98%):
vmax/cnf1 3305, 3212 (N-H, amide), 2854 (O-CH3), 1693, 1627 (secondary CONH, lactam), 1605, 1576, 1505 (aromatic ring), 837 (para-disubstituted benzene ring).
‘H NMR: δΗ (400MHz, CDCI3) 7.76 (2H, br d, J9.0, ortho-H), 7.19 ( 1 H, br d, J 5.0, C7H7O-CON7?), 6.87 (2H, br d, J 9.0, meta-H), 6.31 ( I H, br s, CON/f-CH2), 4.39 ( 1 H, dt, J 11.5, 5.5, CH-CO), 3.81 (3H, s, OCH3), 3.39-3.28 (2H, m, C/f2NH), 2.64 (IH, dq, J 13.0,4.5, NHCH-CW2), 2.01-1.88 (2H,m, lactam CH2), 1.68-1.55 (IH, m, lactam CH2).
l3C NMR: ôc (100MHz, CDCI3) 172.4 (lactam C=O), 167.3 (aryl C=O), 162.4 (COCH3), 129.1 (aromatic-CH), 126.6 (ipso-C), 113.8 (aromatic-CH), 55.6 (OCH3), 51.1 (CH-CO), 41.9 (CH2-NH), 27.4 (lactam CH2), 21.3 (lactam CH2).
HRMS (+ESI) Ci3H16N2O3 + Na+: calcd 271.1053; found 271.1057.
Reference Example 8: 3-(4’-Methoxybenzoylamino)azepan-2-one:
3-aminoazepan-2-one hydrochloride (ΙΟ mmol), K2CO3 (30 mmol) and 4methoxybenzoyl chloride (10 mmol) were reacted according to the above procedure to give the product (1.54 g, 65%):
Vmax/cm1 3270, 3205 (N-H, amide), 2839 (O-CH3), 1642 (secondary CONH, lactam), 1602, 1577, 1504 (aromatic ring), 854, 822 (p«ra-dîsubstituted benzene ring).
‘H NMR: Ôh (400MHz, CDCI3) 7.79 (2H, br d,79.0, ortho-W), 7.52 (IH, brd,75.0, C7H7O-CON/7), 6.91 (2H, br d, 7 9.0, meta-H), 5.94 (IH, br s, CONZ7-CH2), 4.69 ( 1 H, ddd, 7 11.0, 5.5, 1.5, CH-CO), 3.83 (3H, s, OCH3), 3.40-3.21 (2H, m, C/ή,ΝΗ), 2.22 (IH, br d,7 12.5, lactam CH2), 2.08-1.97 (IH, m, lactam CH2), 1.95-1.82 (2H, m, lactam CH2), 1.60-1.36 (2H, m, lactam CH2).
l3C NMR: ôc (100MHz, CDCI3) 176.2 (lactam CO). 165.9 (aryl C=O), 162.1 (C15 OCH3), 129.1 (aromatic-CH), 126.7 (ipso-C), 113.8 (aromatic-CH), 55.5 (OCH3), 52.7 (CH-CO), 42.3 (CH2-NH), 31.9 (lactam CH2), 29.1 (lactam CH2), 28.1 (lactam CH2).
HRMS (+ESI) C14H]8N2O3 + Na+: calcd 285.1210; found 285.1215.
Reference Example 9: 3-(4’-Fluorobenzoylamino)tetrahydropyridin-2-one:
3-aminotetrahydropyridin-2-one hydrochloride (10 mmol), K.2CO3 (30 mmol) and 4fluorobenzoyl chloride (10 mmol) were reacted according to the above procedure (except that CHCI3 was used instead of CH2CI2) to give the product (1.41 g, 54%):
vmax/cnfl 3216, 3075 (N-H, amide), 1650, 1555 (secondary CONH, lactam), 1595, 1491 (aromatic ring), 809, 844 (para-disubstituted benzene ring), 1105, 1158, 1226, 1328,756 (C-F).
'H NMR: δΗ (400MHz, CDCI3) 7.76 (2H, br dd, 7 9.0, 5.5, ortAo-H), 7.21 (1 H, br s, C6H4F-COHf7), 7.02 (2H, br t, 7 8.5, me/a-H), 6.08 (1 H, br s, CON/7-CH2), 4.35 ( 1 H, dt, 7 11.5, 5.5, CH-CO), 3.40-3.26 (2H, m, C/f2NH), 2.62 (IH, dq, 7 13.0, 4.5, NHCHCH2), 2.00-1.86 (2H, m, lactam CH2), 1.66-1.52 (IH, m, lactam CH2).
l3C NMR: Ôc (100MHz, CDCl3) 172.2 (lactam C'-O). 166.6 (aryl C=0), 164.9 (d, J 252.0, C-F), 130.4 (d, J 3.0, ipso-C), 129.7 (d, J 9.0, ortho-C), 115.6 (d, J 22.0, metaC), 51.1 (CH-CO), 41.9 (CH2-NH), 27.3 (lactam CH2), 21.3 (lactam CH2).
HRMS (+ESI) C]2Hi3FN2O2 + H+: calcd 237.1034; found 237.1034.
Reference Example 10: 3-(4’-Huorobenzoylamino)azepan-2-one:
3-aminoazepan-2-one hydrochloride (10 mmol), K2CO3 (30 mmol) and 4fluorobenzoyl chloride (10 mmol) were reacted according to the above procedure (except that CHC13 was used instead of CH2C12) to give the product (0.86 g, 45%):
Vmax/cm'1 3205, 3056 (N-H, amide), 1544 (secondary CONH, lactam), 1599, 1501 (aromatic ring), 821, 858 (p«ra-disubstituted benzene ring), 1164, 1222, 1291, 765, 696 (C-F).
’H NMR: Ôh (400MHz, CDC13) 7.84 (2H, br dd, 79.0, 5.5, ori/m-H), 7.57 (1H, br s, C6H4F-CONH), 7.09 (2H,br t,78.5, meta-W), 5.94 (1H,br s, CON7/-CH2), 4.67 (1H, ddd, 7 11.5, 5.5, 1.5, CH-CO), 3.40-3.22 (2H, m, C/72NH), 2.26-2.19 (1H, m, lactam CH2), 2.09-2.00 (1H, m, lactam CH2), 1.96-1.83 (2H, m, lactam CH2), 1.60-1.36 (2H, m, lactam CH2).
3C NMR: ôc (100MHz, d6-DMSO) 174.3 (lactam C=O), 164.1 (aryl C=O), 163.9 (CF,d7247), 130.7 (ipso-C), 129.8 (ortho-C, d,77), 115.2 (meta-C, d,722), 52.0 (CHCO), 40.6 (CH2-NH), 30.6 (lactam CH2), 28.9 (lactam CH2), 27.7 (lactam CH2).
HRMS (+ESI) C]3Hl5FN2O2 + H+: calcd 251.1190; found 251.1192.
Reference Example 11: 3-(Pyridin-3’-carbonylamino)tetrahydropyridin-2one: —-28
Oxalyl chloride (20 mmol) was added to a solution of nicotinic acid (10 mmol) in DCM (40 mL), along with one drop of catalytîc DMF. The reaction mixture was stirred for 16 h and then the solvent was removed under high vacuum. The resulting crystals were dissolved in DCM (10 mL). In a separate flask, 3aminotetrahydropyridin-2-one hydrochloride (10 mmol) and K2CO3 (30 mmol) were added to water (30 mL) and stirred, giving a solution to which the acid chloride solution was added. The reaction was worked-up as above to give the product (0.10 g, 5%):
vmax/cnfl 3257 (N-H, amide), 1642, 1541 (secondary CONH, lactam, NH), 1591, 1479 (aromatic pyridine ring).
'H NMR: δΗ (400MHz, CDCI3) 9.03 (1H, d, J2.0, 2’-aryl CH), 8.71 (1H, dd, J5.0,
1.5, 6’-aryl CH), 8.12 (1 H, dt, J 8.0, 2.0,4’-aryl CH), 7.36 (1 H, dd, J 8.0, 5.0, 5’-aryl CH), 7.27 (1H, br d, J 2.0, C5H4N-CON/7), 5.91 (1H, br s, CON77-CH2), 4.45 (1H, dt, J 11.0, 5.5, CH-CO), 3.44-3.32 (2H, m, (7Λ\Η). 2.72 (1H, dt, J 14.5, 4.5, NHCH(7Λ), 2.06-1.93 (2H, m, lactam CH2), 1.70-1.54 (lH,m, lactam CH2).
l3C NMR: Ôc (100MHz, CDCI3) 171.8 (lactam C-O). 166.0 (aryl C=O), 152.5 (aryl NCH), 148.6 (aryl N-CH), 135.3 (orrôo-C(-CH)), 133.4 (ipso-C), 123.6 (roeta-C), 51.2 (CH-CO), 42.0 (CH2-NH), 27.3 (lactam CH2), 21.3 (lactam CH2).
HRMS (+ESI)C]lHl3N3O2 + H+: calcd 220.1081; found 220.1085.
Reference Example 12: 3-(Pyridin-3’-carbonylamino)azepan-2-one:
Oxalyl chloride ( 1.69 mL, 20 mmol) was added to a solution of nicotinic acid ( 1.23 g, 10 mmol) in DCM (40 mL), along with one drop of catalytîc DMF. The reaction mixture was stirred for 16 h and then the solvent was removed under high vacuum. The resulting crystals were dissolved in DCM (10 mL). In a separate flask, 3aminoazepan-2-one hydrochloride (10 mmol) and K2CO3 (30 mmol) were added to water (30 mL) and stirred, giving a solution to which the acid chloride solution was added. The reaction was worked-up as above to give the product (0.66 g, 42%):
Vroax/cm'1 3200 (N-H, amide), 1642, 1548 (secondary CONH, lactam), 1590, 1476 (aromatic pyridine ring).
lH NMR: δκ (400MHz, CDC13) 9.06 (1H, d, J2.0, 2’-aryl CH), 8.72 (1H, dd, J5.0,
1.5, 6’-aryl CH), 8.11 (1 H, dt, J 8.0, 2.0, 4’-aryl CH), 7.72-7.62 (1H, m, C5H4NCON/7), 7.37 (1H, dd, 8.0, 5.0, 5’-aryl CH), 5.99 (1H, br s, CONZ7-CH2), 4.70 (1H, ddd, J 11.0, 5.5, 1.5, CH-CO), 3.40-3.23 (2H, m, (7/2NH). 2.24 (1 H, br d, J 14.5, lactam CH2), 2.11-2.02 (1H, m, lactam CH2), 1.97-1.83 (2H, m, lactam CH2), 1.631.38 (2H, m, lactam CH2).
13C NMR: Ôc (100MHz, CDC13) 175.7 (lactam C=O), 164.6 (aryl C=O), 152.4 (aryl NCH), 148.5 (aryl N-CH), 135.1 (orrtro-C(-CH)), 130.0 (/>o-C), 123.5 (meta-C\ 52.8 (CH-CO), 42.2 (CH2-NH), 31.6 (lactam CH2), 28.9 (lactam CH2), 28.1 (lactam CH2).
HRMS (+ESI) C12H15N3O2 + H+: calcd 234.1237; found 234.1239.
Reference Example 13: 3-(3’,5’-Dimethylbenzoylamino)tetrahydropyridin-2011e:
3-aminotetrahydropyridin-2-one hydrochloride (10 mmol), K2CO3 (30 mmol) and 3,5dimethylbenzoyl chloride ( 10 mmol) were reacted according to the above procedure (except that CHC13 was used înstead of CH2C12) to give the product (2.06 g, 94%):
Vmax/cm’1 3212 (N-H, amide), 1675, 1627, 1534 (secondary CONH, lactam), 1598, 1491 (aromatic ring), 890, 865, 817 (meta-trisubstituted benzene ring).
'H NMR: δΗ (400MHz, CDC13) 7.39 (2H, s, ortAo-H), 7.15 (1H, br d, J4.5, CSH9CONW), 7.09(1 H, s, para-H), 6.25 (1 H, br s, CON//-CH2), 4.41 (1H, dt, J 11.5, 5.5, CH-CO), 3.43-3.29 (2H, m, C7/2NHÏ, 2.69 (1H, dq, J 13.0, 4.5, NHCH-C7N). 2.31 (6H, s, CH3), 2.03-1.89 (2H, m, lactam CH2), 1.66-1.53 (1 H, m, lactam CH2).
,3C NMR: Ôc (100MHz, CDCI3) 172.2 (lactam C=0), 168.0 (aryl C=O), 138.2 (ipsoC), 134.3 (meta-C), 133.5 (aromatic CH), 125.1 (aromatic CH), 51.2 (CH-CO), 41.9 (CH2-NH), 27.2 (lactam CH2), 21.4 (CH3), 21.2 (lactam CH2).
HRMS (+ESI) C[4H[gN2O2 + H+: calcd 247.1441; found 247.1455.
Reference Example Î4: 3-(3’,5*-Dimethylbenzoylamino)azepan-2-one:
3-aminoazepan-2-one hydrochloride (10 mmol), K2CO3 (30 mmol) and 3,5dimethylbenzoyl chloride (10 mmol) were reacted according to the above procedure (except that CHCI3 was used instead of CH2C12) to give the product (2.26 g, 96%):
Vmax/cm'1 3319 (N-H, amide), 1682, 1635 (secondary CONH, lactam), 1600, 1498 (aromatic ring), 888, 866, 828 (meia-trisubstituted benzene ring).
lH NMR: δΗ (400MHz, CDCI3) 7.57 (IH, br d, J5.0, C\II9-(O\//). 7.42 (2H, s, ortho-H), 7.10 (IH, s,/«ira-H), 6.09 (IH, br s, CON/7-CH2), 4.69 (IH, ddd, J 11.0, 6.0, 1.5, CH-CO), 3.40-3.20 (2H, m, Ctt2NH), 2.33 (6H, s, CH3), 2.21 (IH, br d, J
12.5, lactam CH2), 2.08-1.98 (IH, m, lactam CH2), 1.97-1.82 (2H, m, lactam CH2), 1.59-1.35 (2H, m, lactam CH2).
l3C NMR: Ôc (100MHz, CDCI3) 176.0 (lactam C=O), 166.8 (aryl C=O), 138.3 (ipsoC), 134.3 (meta-C), 133.3 (aromatic CH), 124.9 (aromatic CH), 52.6 (CH-CO), 42.3 (CH2-NH), 31.8 (lactam CH2), 29.0 (lactam CH2), 28.1 (lactam CH2), 21.4 (CH3).
HRMS (+ESI) Ci5H20N2O2 + H+: calcd 261.1598; found 261.1602.
In the examples below, the general procedure for the synthesis of 3-acylamino-2oxopiperidînes was: potassium carbonate (3 mmol) and (S)-3-amino-2-oxopiperidine hydrochloride (1.5 mmol) were dissolved in water (5 ml) and the solution was cooled to 0 °C, and a solution of substituted benzoyl chloride (1 mmol) in tetrahydrofuran (5 mL) was added. The mixture was stirred for 16 hours, and then the reaction was extracted with dichloromethane or chioroform. The combined organic layers were dried over sodium sulfate and reduced in vacuo to give a solid. This solid was redîssolved in a minimum amount of dichloromethane and crystallîsed by addition of petroleum ether 40-60 °C. The solid product was isolated by filtration and dried over potassium pentoxide.
Example 1 : (S)-3-Fluoro-N-(2-oxopiperidm-3-yl)benzamide
0.147 g off-white coarse powder (41 %). mp 164-166 °C. [«]24d -5.50 (c 0.1, MeOH); Vmax/crn·' 1669, 1644 (C=O, amide), 1552 (N-H, amide), 1303 (C-F). Anal. (C12H13FN2O2) C, H, N: calcd C 61.01, H 5.55, N 11.86; found C 60.65, H 5.50, N 11.78. 'H-NMR Ôh. 7.52 (2H, tt, J 8 and 2 , Atf/4 and Ar//6), 7.38 (1H, td, J 8 and
5.5, Ar//2). 7.21-7.14 (2H, m, N//CH and Ar/73), 5.9 (1H, s, N//CH2), 4.41 (1H, dt, J
11.5 and 6 , CHNH), 3.42-3.28 (2H, m, C/72NH), 2.72 (1H, dq, J 13 and 5 , C/f2CH), 2.04-1.95 (2H, m, C//2CH2NH), 1.62 (1H, dq, J 16 and 5 , C//2CH). 13C-NMR Ôc 171.6 (CHCONH), 166.3 (CONHCH), 162.8 (d, J 247, ArC3), 136.4 (CCO), 130.17 (d, J 12, ArC5), 122.6 (ArC6), 118.6 (d, J 21, ArC2), 114.6 (d, J 21, ArC4), 51.2 (CHNH), 41.8 (CH2NH), 27.0 (CH2CHNH), 21.0 (CH2CH2NH). 19F-NMR ÔF -111.9. HRMS (+ESI) C12H13FN2O2Na: calcd 259.0853; found 259.0859.
Example 2: (S)-2-Fluoro-N-(2-oxopiperidin-3-yl)benzamide
0.222 g white flufïy powder (63 %). mp 166-168 °C. [a]24D -10.00 (c 0.1, MeOH).
Vmax/cm·’ 1647, 1613 (C=O, amide), 1512 (N-H, amide), 1277 (C-F). Anal.
(CizHoFNîCh) C, H, N: calcd C 6l.0l, H 5.55, N 11.86; found C 60.78, H 5.54, N 11.69. ’H-NMR ôh. 8.05 (ÎH, td, 7 8 and 2 , Ar//6), 7.62 (1H, dd, J 6 and 4, NZ/CH), 7.45 (1H, dddd, 7 8,7, 5 and 2, ArF/4), 7.21 (ÎH, td, 77.5 and 1, ArH5), 7.13 (1H, ddd, 7 12, 9 and 1, Ar//3), 6.01 (1H, s, NHCH2), 4.53 (1H, dt, 7 11 and 6, C7/NH), 3.45 (2H, td, 7 6 and 3, CF/2NH), 2.72 (IH, dq, 7 13 and 6, C//2CH), 2.02-1.95 (2H, m, CF/2CH2NH), 1.72-1.59 (1H, m, C//2CH). 13C-NMR ôc 171.3 (CHCONH), 163.4 (CONHCH), 160.9 (d, 7 248.5, ArC2), 130.17 (d, 7 12, ArC5), 133.4 (d, 7 9, ArC4),
131.9 (ArC6), 120.9 (d, 7 9, CCO), 116.1 (d, 7 21, ArC3), 51.3 (CHNH), 41.9 (CH2NH), 27.2 (CH2CHNH), 21.1 (CH2CH2NH). I9F-NMR ÔF -112.4. HRMS (+ESI) CI2Hi3FN2O2Na: calcd 259.0853; found 259.0858.
Example 3: (S)-4-Fluoro-N-(2-oxopiperidin-3-yl)benzamide
NH
0.145 g cream coloured fine crystals (41 %)mp 133-135 °C; [a]24o -7.90 (c 0.1, MeOH); v^/crn'1 1650, 1636 (C=O, amide), 1557 (N-H, amide), 1327 (C-F) Anal. (Ci2H[3FN2O2) C, H, N: calcd C 61.01, H 5.55, N 11.86; found C 60.71, H 5.38, N 11.44 (1/3 H2O). lH-NMR ÔH 7.98 (2H, q, J 5, A17/3 and Ar//5). 7.41 (1H, d, 7 6, NF/CH), 7.20 (2H, tt, 7 9,2, Ar//2 and Ar/76), 6.31 ( 1 H, s, NF/CH2), 4.52 ( 1 H, dt, 7 Il and 6, CF/NH), 3.52 (2H, td, 7 6 and 2, CFZ2NH), 2.84 (1H, dq, J 13 and 4, CF/2CH), 2.19-2.10 (2H, m, CF/2CH2NH), 1.81 (1H, dq, 7 12 and 8, C7/2CH). 13CNMR Ôc 171.7 (CHCONH), 166.6 (CONHCH), 164.9 (d, 7 251.5, ArC4), 130.3 (d, 7 4, CCO), 129.5 (d, J 9, ArC2/6), 115.5 (ArC3/5), 51.2 (CHNH), 41.8 (CH2NH), 27.1 (CH2CHNH), 21.0 (CH2CH2NH). HRMS (+ESI) C12H]3FN2O2Na: calcd 259.0853; found 259.0852.
Example 4: (S)-N-(2-Oxopiperidin-3-yl)-4-(trifluoromethyl)benzamide -—x
0.376 g whîte fine powder (87 %). mp 212-214 °C; [a]24D -6.35 (c 0.1, MeOH); v^/cm'1 1650, 1636 (C=O, amide), 1557 (N-H, amide), 1327 (C-F). Anal. (C13HI3F3N2O2) C, H, N: calcd C 54.55, H 4.58, N 9.79; found C 53.99, H 4.68, N 9.59. ’H-NMR Ôh ‘η-NMR δΗ. 7.51 (2H, d, J 8, Ar//2 and Ar/76), 7.22 (2H, d, J 8.5, Ar/73 and ArH5), 7.15 (1H, s, NWCH), 5.70 (1H, s, N/7CH2), 4.00 (1H, dt, J 11.5 and 6, C77NH), 2.98 (2H, td, J 6 and 2, C/AXH). 2.25 (1H, dq, J 13 and 4, C/72CH), 1.621.53 (2H, m, C//2CH2NH), 1.28 (1H, dq, J 12 and 8, C/FCH). nC-NMR ôc 171.8 (CHCONH), 166.3 (CONHCH), 137.3 (ArCl), 133.3 (q, J 31, ArC4), 127.6 (ArC2/6),
125.5.9 (q, .7 4, ArC3/5), 123.7 (q, J 271, CF3), 51.2 (CHNH), 41.9 (CH2NH), 27.0 (CH2CHNH), 21.1 (CH2CH2NH). 19F-NMR -62.9. HRMS (+ESI) CBH|3F3N2O2Na: calcd 309.0821; found 309.0822.
Example 5: (5)-N-(2-Oxopiperidin-3-yl)-3-(trifluoromethyl)benzamide
0.251 g cream fine powder (59 %). mp 148-150 °C; [cc]24d -10.20 (c 0.1, MeOH); Vmax/cm'1 1671, 1648 (C=O, amide), 1554 (N-H, amide), 1327 (C-F). Anal. (Ci3Hi3F3N2O2) C, H, N: calcd C 54.55, H 4.58, N 9.79; found C 53.90, H 4.56, N 9.60. ’H-NMR δΗ 7.80 (1H, s, Ar/72), 7.73 (1 H, d, J 7, Ar/74), 7.60 (1H, d, J 6, N/fCH), 7.43 (1H, d, J 7, ArZ/6), 7.23 (1H, d, J 8, Ar/75), 6.52 (1H, s, N/7CH2), 4.23 (1H, dt, J 11 and 6, CZ/NH), 3.17-3.06 (2H, m, C/72NH), 2.30 (1H, dq, J 13 and 6, C//2CH), 1.74-1.65 (2H, m, C772CH2NH), 1.62-1.42 (1H, m, C//2CH). l3C-NMR ôc 171.6 (CHCONH), 166.1 (CONHCH), 134,9 (ArCl), 131.1 (q, J 34, ArC3), 130.3 (ArC5), 129.1 (ArC6), 128.2 (q, J 4, ArC2), 124.3 (q, J 4, ArC4), 123.7 (q, J 271,
CF3), 51.2 (CHNH), 41.8 (CH2NH), 27.1 (CH2CHNH), 21.1 (CH2CH2NH).I9F-NMR
ÔF -62.7. HRMS (+ESI) C13H13F3N2O2Na: calcd 309.0821; found 309.0820.
Example 6: (5)-N-(2-Oxopiperidin-3-yl)-2-(trifluoromethyl)benzamide
0.262 g white fluffy powder (61 %). mp 155-156 °C; [a]24o -18.20 (c 0.1, MeOH); v,nax/cm1 1674, 1654 (C=O, amide), 1543 (N-H, amide), 1312 (C-F). Anal. (Cî3Hi3F3N2O2) C, H, N: calcd C 54.55, H 4.58, N 9.79; found C 54.25, H 4.51, N 9.70. 'H-NMR ôh7,64 (1H, d, J7, Ar/76), 7.54 (1H, d, J 6, Arf/3), 7.22-7.15 (2H, m, ArZ/4 and ArH5), 6.78 (1H, d, J 5, N//CH), 6.08 (1H, NRCH2), 4.42 (1H, dt, J 11 and 6, C/VNH), 3.35 (2H, td, J 6 and 2, C//2\H). 2.73 ( 1 H, dq, J 13 and 6, CH2CH), 1.991.91 (2H, m, C//2CH2NH). 1.59 (1H, dq, J 12 and 8, CVÇCH). 13C-NMR δΓ 171.0 (CHCONH), 167.8 (CONHCH), 135.5 (ArCl), 131.9 (ArC4), 129.9 (ArC5), 128.6 (ArC6), 127.4 (q, J 31, ArC2), 126.4 (q, J 4, ArC3), 123.6 (q, J 270, CF3), 51.3 (CHNH), 41.8 (CH2NH), 26.5 (CH2CHNH), 20.9 (CH2CH2NH). 19F-NMR ÔF -58.7. HRMS (+ESI) Ci3Hi3F3N2O2Na: calcd 309.0821; found 309.0818.
Example 7: (5)-2,3-difluoro-N-(2-oxopipciïdin-3-yl)benzamide
0.234 g white needle crystals (61 %). mp 160-162 °C; [a]24p -5.65 (c 0.1, MeOH); vinax/cm*' 1686, 1648 (C=O, amide), 1536 (N-H, amide), 1320 (C-F). Anal. (Ci2H]2F2N2O2) C, H, N: calcd C 56.99, H 4.76, N 11.02; found C 56.26, H 4.69, N 10.84. 'H-NMR δΗ. 7.76 (1H, ddt, J8, 6 and 2, Ar/76), 7.51 (1H, q, J 3, N//CH), 7.28 (1H, dddd, J 9.5, 8, 7.5, 2, ArR4), 7.15 (1 H, tdd, J 8, 5, 1.5, ArH5), 6.18 (1 H, s, 35
N/7CH2), 4.47 (IH, dt, J 12 and 4, C//NH). 3.38 (2H, td, J6 and 2, (7/2\'H), 2.69 (IH, dq, J 12.5 and 3.5, C//2CH), 2.02-1.93 (2H, m, Cff2CH2NH), 1.73-1.59 (IH, m, C/ACII). ,3C-NMR Ôc 171.2 (CHCONH), 162.5 (CCONH), 132.8 (dd, J250 and 15, ArC3), 149.1 (dd, J 251 and 14, ArC2), 126.2 (t, J 3, ArC5), 124.4 (t, J 4, ArC6), 123.3 (d, J 9, CCONH), 120.3 (d, J 17, ArC4), 51.4 (CHNH), 41.8 (CH2NH), 27.1 (CH2CHNH), 21.1 (CH2CH2NH). HRMS (+ESI) Ci2H[2F2N2O2Na: calcd 277.0759; found 277.0769.
Example 8: (5)-2,4-difluoro-N-(2-oxopiperidin-3-yl)benzamÎde
0.236 g off-white crystals (62 %). mp 140-141 °C; [a]24D -2.00 (c 0.1, MeOH); v^/cm’1 1682, 1638 (C=O, amide), 1526 (N-H, amide), 1289 (C-F). Anal. (Ci2H12F2N2O2.l/6 H2O) C, H, N: calcd C 56.03, H 4.83, N 10.89; found C 56.40, H 4.69, N 10.93. 'H-NMR ôh. 8.07 (IH, td, J 9 and 7, ArH6), 7.54 (IH, q, J5, N/fCH), 6.95 (IH, tdd, J8, 2.5, 1, Ar//5), 6.84 (IH, dq, J 8.5, 2, Ar/73), 6.43 (IH, s, NHCH2), 4.45 (IH, dt, J 11 and 6, COI H), 3.36 (2H, td, J6.5 and 2.5, CW2NH), 2.66 (IH, dq, J
12.5 and 5.5, CZf2CH), 2.00-1.91 (2H, m, C/f2CH2NH), 1.73-1.59 (IH, m, C172CH). 13C-NMR ôc 171.4 (CHCONH), 162.5 (CCONH), 164.9 (dd, J 255 and 12, ArC2),
161.2 (dd, J 253 and 12, ArC4), 133.6 (dd, J 10 and 4, ArC6), 117.4 (dd, J 12 and 4, CCONH), 112.2 (dd, J 21 and 3, ArC5), 104.3 (t, J 27, ArC3), 51.3 (CHNH), 41.8 (CH2NH), 27.2 (CHÆHNH), 21.1 (CH2CH2NH). HRMS (+ESI) Cl2H12F2N2O2Na: calcd 277.0759; found 277.0761.
Example 9: (5)-2,5-difluoro-N-(2-oxopiperidin-3-yl)benzamide
0.252 g off-white crystals (66 %). mp 140-142 °C; [a]24D +0.85 (c 0.1, MeOH); vmax/cm·' 1681, 1635 (C=O, amide), 1519 (N-H, amide), 1328 (C-F). Anal. (Ci2H12F2N2O2.1/6 H2O) C, H, N: calcd C 56.03, H 4.83, N 10.89; found C 55.47, H 4.72, N 10.67. ’H-NMR δΗ. 8.07 (IH, dt, J 11 and 8.5, ArH6), 7.54 (IH, dd, J 11, 5, NHCH), 6.68-6.54 (2H, m, Ar/73 and ArH4). 6.43 (IH, s, NHCH2), 4.45 (IH, dt, J 11 and 6, CHNH), 3.36 (2H, td, J 6.5 and 2.5, Cff2NH), 2.66 (IH, dq, J 12.5 and 5.5, (7/2CH), 1.99-1.90 (2H, m, CH2CH2NH), 1.64 (IH, dq, J 11 and 8 CH2CH). 13C-NMR Ôc 171.3 (CHCONH), 162.2 (CCONH), 158.6 (d, J 258, ArC5), 156.7 (d, J 258, ArC2), 133.6 (dd, J 10 and 4, ArC6), 122.4 (dd, J 8 and 6, CCONH), 120.0 (dd, J 24 and 8, ArC3), 118.0 (dd, J 26 and 4, ArC4), 117.5 (dd, J 28 and 7, ArC6), 51.4 (CHNH), 41.8 (CH2NH), 27.1 (CH2CHNH), 21.1 (CH2CH2NH). HRMS (+ESI) C12Hi2F2N2O2Na: calcd 277.0759; found 277.0766.
Example 10: (5)-2,6-difluoro-N-(2-oxopiperidin-3-yl)benzamide
0.209 g white fine powder (55 %). mp 190-194 °C; [a]24p -7.20 (c 0.1, MeOH); Vmax/cm’1 1672, 1638 (C=O, amide), 1492 (N-H, amide), 1329 (C-F). Anal. (C12H12F2N2O2) C, H, N: calcd C 56.99, H 4.76, N 11.02; found C 55.93, H 4.68, N 10.83 (1/6 H2O). lH-NMR δΗ. 7.30 (IH, tt, J 9 and 6, ArH4), 7.06 (IH, d, J 3.5, NHCH), 6.89 (2H, t, J3.5, ArH3 and ArH5), 6.23 (IH, s, NHCH2), 4.43 (IH, dt, J 11 and 5, CHNH), 3.37-3.32 (2H, m, CH2NH), 2.75 (IH, dq, J 14.5 and 4.5, CH2CH),
I. 99-1.90 (2H, m, CH2CH2NH), 1.68-1.54 (IH, m, CH2CH). 13C-NMR Ôc 171.1 (CHCONH), 160.5 (CCONH), 160.1 (dd, J 250 and 7.5, ArC2 and ArC6), 131.7 (t, J
II, ArC4), 114.1 (t, J 20, CCONH), 111.9 (dd, J 20 and 5, ArC3 and ArC5), 51.3 (CHNH), 41.7 (CH2NH), 26.8 (CH2CHNH), 20.9 (CH2CH2NH). ,9F-NMR δΡ -112.5. HRMS (+ESI) Ci2Hi2F2N2O2Na: calcd 277.0759; found 277.0760.
Example 11: (5)-3,4-difluoro-N-(2-oxopiperidin-3-yl)benzamide
0.226 g off-white fine powder (59%). mp 202-204 °C; [a]24o -7.50 (c O.l, MeOH); Vjnax/cm’1 1691, 1652 (C=O, amide), 1487 (N-H, amide), 1285 (C-F). Anal. (C12H12F2N2O2) C, H, N: calcd C 56.99, H 4.76, N 11.02; found C 56.06, H 4.68, N 10.88 (1/6 H2O). lH-NMR δΗ. 7.66 (1 H, qd, J 7 and 2, Ar/72), 7.56-7.50 (IH, m, ArH5), 7.24-7.14 (2H, m, Ar/76 and N//CH). 5.90 (IH, s, N77CH2), 4.38 (IH, dt, J
11.5 and 5, C/7NH), 3.39 (2H, td, J 7 and 3, CZ/2NH), 2.69 (IH, dq, J 14 and 5, C7/2CH), 2.03-1.94 (2H, m, C7/2CH2NH). 1.68-1.54 (IH, m, C//2CH). 13C-NMR ôc
171.5 (CHCONH), 165.4 (CCONH), 152.5 (dd, J 253 and 12, ArC3), 150.2 (dd, J 249 and 12, ArC4), 131.2 (t, J 4.5, CCONH), 123.5 (q, J 3.5, ArC6), 117.4 (d, J 18, ArC5),
116.9 (d, J 18, ArC2), 51.2 (CHNH), 41.8 (CH2NH), 26.9 (CH2CHNH), 21.1 (CH2CH2NH). HRMS (+ESI) C12H12F2N2O2Na: calcd 277.0759; found 277.0751.
Example 12: (5)-3,5-difluoro-N-(2-oxopipendin-3-yl)benzamide
NH
0.234 g white fine powder (61 %). mp 198-199 °C; [a]24D -9.50 (c 0.1, MeOH); Vmax/crn’1 1674, 1641 (C=O, amide), 1565 (N-H, amide), 1333 (C-F). Anal. (Ci2Hi2F2N2O2) C, H, N: calcd C 56.99, H 4.76, N 11.02; found C 56.20, H 4.69, N 10.93. ‘H-NMR δΗ. 7.31 (2H, dd, J 8 and 2.5, Ar//2 and Ar//6). 7.26 (IH, d, J 5, N7/CH), 6.9 (IH, tt, J 9 and 2, Arf/4), 6.01 (IH, s, NZ/CIh), 4.39 (IH, dt, J 12 and 5.5, CffllH), 3.41-3.36 (2H, m, C/72NH). 2.68 (IH, dq, 7 12 and 5, C/ACH). 2.03-1.94 (2H, m, CZ/2CH2NH), 1.69-1.55 (IH, m, C//2CH). 13C-NMR Ôc 171.3 (CHCONH),
165.2 (CCONH), 162.9 (dd, J 251.5 and 12, ArC3 and ArC5), 137.5 (t, J 9, CCONH),
110.4 (dd, J 20 and 7, ArC2 and ArC6), 107.0 (t, J 20, ArC4), 51.3 (CHNH), 41.8 TO
(CH2NH), 26.9 (CH2CHNH), 20.9 (CH2CH2NH). l9F-NMR δΡ -1Ο8.2. HRMS (+ESI)
Cl2H]2F2N2O2Na: calcd 277.0759; found 277.0750.
Example 13: (S)-3-(3’-ButylbenzoyIamino)-azepan-2-one
(S)-3-Amino-azepan-2-one hydrochloride (0.72 g, 4.39 mmoles) was dissolved in H2O (20 mL) and cooled to 0°C. 3-Butylbenzoyl chloride in dichloromethane was added and triethylamine (1.3 mL, 9 mmoles) and the reaction was stirred over night. H2O (20 mL) was added and the reaction was extracted with dichloromethane (3 * 20 mL), the organic layer was washed with a pH 2 buffer (3x15 mL), dried over Na2SO4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ether:ethyl acetate 75:25 to 0:100) to give the product as a white solid δΗ (400 MHz, CDC13) 7.71-7.62 (m, 3H, Ar & NHCH), 7.35-7.29 (m, 2H, Ar), 7.06 (br.t, 1 H, J 6, NFfCH2), 4.72 (dd, 1H, J 11,6, NHC/f), 3.37-3.32 (m, 2H, NHC/^), 2.65 (t, 2H, J 8, CH3CH2CH2C/ù), 2.22 (br.d, 1H, J 13.5, NHCHC/Λ), 2.03 (br.d, 1H, J 14, NHCHCH2C/ù), 1.95-1.81 (m, 2H, NH(HCH2Œ2C7/2), 1.61 (quintet, 2H, J Ί, CHY'ILC/T). 1.57-1.48 (m, 1H. MICHC/L), 1.46-1.25 (m, 3H, NHCHCH2CH2C7/2 and CH3C^) and 0.92 (t, 3H, J 7.5, H8); Ôc (100 MHz, CDC13) 176.0 (Ç=O), 166.6 (C=O), 143.4 (Ar quat, Ç-nBu), 134.2 (Ar quat, Ç-C=O), 128.4 (CH, Ar), 127.2 (CH, Ar), 126.8 (CH, Ar), 124.0 (CH, Ar), 52.6 (ÇH-NH), 42.1 (ÇH2-NH), 35.6 (ÇH2), 33.5 (ÇH2), 31.6 (ÇH2), 28.9 (CH2), 28.0 (ÇH2), 22.3 (ÇH2) and 13.9 (ÇH3).
Example 14: (S)-3-(4'-Ethylbenzoylamino)-azepan-2-one
(5)-3-amino-azepan-2-one hydrochloride (1.65 g, 10 mmoles) was dissolved in H2O (20 mL) and cooled to 0°C. 4-Ethylbenzoyl chloride in dichloromethane was added and triethylamine (4.2 mL, 30 mmoles) and the reaction was stirred over night. H2O (20 mL) was added and the reaction was extracted with dichloromethane (3 x 20 mL), the organic layer was washed with a pH 2 buffer (3 x 15 mL), dried over Na2SÛ4 and reduced in vacuo. The product was purified by recrystallîsation from chloroform and cold petroleum ether to give the product as a white solid 0.94 g (36 %); mp 218-219 ° C; δΗ (400 MHz, CDC13) 7.66 (d, 2H, J 8, CH-C-Et), 7.62 (d, 1 H, J 5.5, N/f-CH), 7.24 (d, 2H, J 8, CH-C-CO), 6.55 (br.t, 1H, J6, NH-CV), 4.70 (dd, 1H,-J 11, 5.5, CH-C4), 3.37-3.32 (m, 2H, Hl), 2.67 (q, 2H, J 7.5, H5), 2.21 (br.d, 1H, J 13, H4 équatorial), 2.02 (dt, 1H, J 14, 4, H3 équatorial), 1,95-1.82 (m, 2H, H2 équatorial & H3 axial), 1.53 (br.q, 1H, J 12.5, H4 axial), 1.40 (br.q, 1H, J 13, H2 axial) and 1.22 (t, 3H, J 7.5, H6); Ôc (100 MHz, CDCI3) 175.9 (Ç=O), 166.2 (C=O), 148.2 (Ç-Et), 131.6 (Ç-OO), 128.0, 127.2 (ÇH phenyl), 52.6 (ÇH-NH), 42.2 (Cl), 28.9 (C5), 28.8, 28.0 (C2, C3) and 15.4 (C6); tUcm4: 3200 (NH indole), 2956 (C-H), 1642 (amide C=O) and 1543 (aromatic); ESI m/z 100 %, 542.9 (M2Na+), 70%, 283.1 (MNa+) and 10 %, 261.2 (MH+); HR ESI m/z (Ci5H20N2O2Na requires 283.1417) found 283.1414; [a]23 D (c = 0.49, CHCI3) +70.48.
Example 15: (S )-3-(4'-Ethylbenzoylamino)-tetrahydropyridin-2-one
(S)-3-amino-tetrahydropyridin-2-one (20 mmoles) was dissolved in H2O (100 mL) and cooled to 0°C. 4-Ethylbenzoyl chloride (16 mmoles) in dichloromethane was added and triethylamine (6.7 mL, 48 mmoles) and the reaction was stirred over night. The reaction was extracted with dichloromethane (3 x 30 mL), the organic layer was washed with a pH 2 buffer (3 χ 20 mL), dried over Na2SO4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ether:ethyl acetate 50:50:0 to 0:80:20) to give the product as a white solid 1.46 g (37 %); mp 112-113 0 C;
δκ (400 MHz, CDClî) 7.71 (d, 2H, J 8.5, C/f-C-Et), 7.55 (d, ÎH, J 5.5, NH-CH), 7.19 (d, 2H, J 8.5, C/f-C-CO), 6.69 (br.s, 1H, NH-C1), 4.39 (dt, 1H, J 11, 5.5, CH-C3), 3.35-3.28 (m, 2H, Hl), 2.67 (q, 2H,7 7.5, H4), 2.63-2.56 (m, 1H, H3 équatorial), 1.941.87 (m, 2H, H2), 1.68 (tt, 1H, 7 12.5, 8, H3 axial), and 1.20 (t, 3H, 7 7.5, H5); 5C (100 MHz, CDC13) 172.2 (C=O), 167.5 (Ç=O), 148.2 (Ç-Et), 131.5 (Ç-C=O), 127.9, 127.2 (ÇH phenyl), 50.9 (ÇH-NH), 41.7 (Cl), 28.8 (C4), 27.2 (C3), 21.1 (C2) and 15.3 (C5);
3334, 3245 (NH), 2932 (C-H), 1656, 1634 (C=O) and 1528 (aromatic); ESI m/z 100 %, 514.9 (M2Na+) and 35%, 269.1 (MNa+); HR ESI m/z (C14H18N2O2Na requires 269.1260) found 269.1261; [a]23D(c = 0.491, CHCI3) +103.95.
Example 16: (S)-3-(4'-Biitylbenzoylamino)-azepan-2-one
(S)-3-amino-azepan-2-one hydrochloride (1.65 g, 10 mmoles) was dîssolved in H2O (20 mL) and cooled to 0°C. 4-Butylbenzoyl chloride (8 mmoles) in dichloromethane was added and triethylamine (4.2 mL, 30 mmoles) and the reaction was stirred over night. H2O (20 mL) was added and the reaction was extracted with dichloromethane (3 x 20 mL), the organic layer was washed with a pH 2 buffer (3*15 mL), dried over Na2SÜ4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum cthcriethyl acetate 75:25 to 0:100) to give the product as a white solid 0.42 g (18 %); mp 183-184 0 C; δΗ (400 MHz, CDCI3) 7.73 (d, 2H, 7 8, Ar), 7.63 (d, 1H, 7 5.5, NHCH), 7.21 (d, 2H, 7 8, Ar), 6.80 (br.t, 1H, 7 6, NHCH2), 4.69 (dd, 1H,7 10.5, 5.5, N H CH), 3.35-3.20 (m, 2H, CW2NH),2.61 (t, 2H,7 7.5, H5), 2.19 (br.d, 1H, 7 13.5, H4 équatorial), 2.00 (br.d, 1H, 7 12.5, H3 équatorial), 1.93-1.80 (m, 2H, H2 équatorial & H3 axial), 1.70-1.46 (m, 3H, H6 and H4 axial), 1.38 (br.q, 1H, 7 13, H2 axial), 1.31 (sextet, H2, J 7, H7) and 0.89 (t, 3H, 7 7.5, H8); Ôc (100 MHz, CDCI3) 176.0 (CO), 166.3 (Ç=O), 146.7 (Ç-nBu), 131.6 (Ç-C=O), 128.5, 127.1 (ÇH phenyl), 52.5 (ÇH-NH), 42.2 (Cl), 35.5 (C5), 33.4 (C4), 31.6 (C6), 28.9, 28.0 (C2, C3), 22.3 (C7) and I3.9 (C6); υ^/αη'': 3359, 3207 (NH), 2951 (C-H), I67l, 1650 (C=O) and 1543 (aromatic); ESI m/z 100 %, 311.2 (MNa+) and 22 %, 289.2 (MH+); HR ESI m/z (Ci7H24N2O2Na requires 311.1730) found 31 1.1732; [a]25D (c = 0.515, CHClj) +64.88.
Example 17: (S)-3-(4’-Butylbenzoylamino)-tetrahydropyridin-2-one
(5)-3-amino-tetrahydropyridin-2-one (20 mmoles) was dissolved in H2O (20 mL) and cooled to 0°C. The 4-butylbenzoyl chloride in dichloromethane was added and triethylamine (4.2 mL, 30 mmoles) and the reaction was stirred over night. H2O (20 mL) was added and the reaction was extracted with dichloromethane (3 x 20 mL), the organic layer was washed with a pH 2 buffer (3x15 mL), dried over Na2SÛ4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum etherethyl acetateimethanol 50:50:0 to 0:80:20) to give the product as a white solid 0.45 g (16 %); ôH (400 MHz, CDCfi) 7.70 (d, 2H, J 8, C/f-C-nBu), 7.36 (d, IH, J 6, NÇ- mp 117-118 ° C; CH), 7.17 (d, 2H, J 8, C/f-C-CO), 6.67 (br.s, IH, N/fCl), 4.69 (dt, IH, J 12, 6, C/AC4), 3.36-3.29 (m, 2H, Hl), 2.60 (t, 3H, J 7.5, H4 &
H3), 1.93-1.86 (m, 2H, H2), 1.67-1.54 (m, IH, H3), 1.55 (quintet, 2H, J 7.5, H5), 1.30 (sextet, 2H, J 7, H7.5, H6) and 0.89 (t, 3H, J 7.5, H7); δΓ (100 MHz, CDC13) 172.2 (Ç=O lactam), 167.5 (ÇO amide), 146.9 (Ç-nBu), 131.5 (Ç-CO), 128.5 (ÇH phenyl),
127.2 (ÇH phenyl), 50.8 (ÇH-NH), 41.7 (Cl), 35.5 (C4), 33.3 (C5), 27.2 (C3), 22.3 (C6), 21.1 (C2) and 13.9 (C7); ESI m/z 100 %, 297.2 (MNa+) and 26 %, 275.2 (MH+);
HR ESI m/z (C,6H22N2O2Na requires 297.1573) found 297.1573; [ct]24 D (c = 0.523, CHC13) +98.73.
Example 18: (R)-3-(4’-Butylbenzoylamino)-tetrahydropyridin-2-one
(7?)-3-amino-tetrahydropyridin-2-one (ΙΟ mmoles) was dissolved in H2O (30 mL) and cooled to 0°C. 4-Butylbenzoyl chloride (8.5 mmoles) in dichloromethane was added and triethylamine (4.2 mL, 30 mmoles) and the reaction was stirred over night. The reaction was extracted with dichloromethane (3 x 15 mL), the organic layer was washed with a pH 2 buffer (3 * 15 mL), dried over Na2SO4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum etheriethyl acetate:methanol 50:50:0 to 0:80:20) to give the product as a white solid 1.10 g (40 %); mp 116-117 0 C; ÔH (400 MHz, CDC13) 7.72 (d, 2H, J 8, CW-C-nBu), 7.25 (d, 1 H, J
5.5, X7/-CH), 7.20 (d, 2H, J 8, CH-C-CO), 6.41 (br.s, 1H, M-Cl), 4.41 (dt, 1H, J
11.5, 5.5, C/7-C4), 3.37-3.32 (m, 2H, Hl), 2.66 (ddt, 1H, J 13, 6, 4.5, H3 équatorial), 2.62 (t, 3H, J 8, H4), 1.98-1.90 (m, 2H, H2), 1.67-1.53 (m, 3H, H3 axial & H5), 1.32 (quintet, 2H, J 7.5, H6) and 0.90 (t, 3H, J 7, H7); 5C ( 100 MHz, CDC13) 172.1 (Ç=O lactam), 167.6 (Ç=O amide), 147.0 (Ç-nBu), 131.5 (Ç-C=O), 128.5 (ÇH phenyl), 127.2 (ÇH phenyl), 51.0 (ÇH-NH), 41.7 (Cl), 35.5 (C4), 33.3 (C5), 27.2 (C3), 22.3 (C6), 21.1 (C2) and 13.9 (C7); wW1: 3319, 3245 (NH), 2949 (C-H), 1651, 1634 (C=O) and 1521 (aromatic); ESI m/z 100 %, 297.1 (MNa+); HR ESI m/z (Ci6H22N2O2Na requires 297.1573) found 297.1574; [a]25D(c = 0.493, CHC13) -89.45.
Example 19: (S)-3-(4'-teri-Butylbenzoylamino)-azepan-2-one
(S)-3-amino-azepan-2-one hydrochloride (2.04 g, 12.44 mmoles) was dissolved in H2O (20 mL) and cooled to 0°C. The 4-lButylbenzoyl chloride in dichloromethane was added and triethylamine (4.2 mL, 30 mmoles) and the reaction was stirred over night. H2O (20 mL) was added and the reaction was extracted with dichloromethane (3 x 20 mL), the organic layer was washed with a pH 2 buffer (3 x 20 mL), dried over Na2SO4 and reduced in vacuo. The product was purified by recrystallisation from chloroform 5 and cold petroleum ether and washed with boiling ethyl acetate to give the product as a white solid 1.44 g (50 %); mp 204-205 0 C; δΗ (400 MHz, CDC13) 7.76 (d, 2H, J 8.5, Cif-C-’Bu), 7.66 (d, 1H, J 6, NH-CH), 7.42 (d, 2H, J 8.5, CFf-C-CO), 6.00 (br.s, 1H, N//-C1). 4.67 (ddd, 1 H, 7 11, 6, 1.5, C//-C4), 3.34-3.19 (m, 2H, Hl), 2.18 (br.d, 1H,7 13, H4 équatorial), 2.00 (br.d, 1H, J 12.5, H3 équatorial), 1.92-1.78 (m, 2H, H2 10 équatorial & H3 axial), 1.51 (q, 1H, J 13, H4 axial), 1.33 (br.q, 1H, J 11, H2 axial), and 1.29 (s, 3H, C(C/Û)3); ôc (100 MHz, CDC13) 176.0 (Ç=O), 166.2 (Ç=O), 155.0 (Ç-C(CH3)3), 131.4 (Ç-C=O), 127.9, 125.4 (CH phenyl), 52.5 (ÇH-NH), 42.1 (Cl), 34.9 (Ç(CH3)3), 31.6 (C4), 31.2 (C(ÇH3)3) and 28.9, 28.0 (C2, C3); ESI m/z 100 %, 598.8 (M2Na+) and 32 %, 289.2 (MH+); HR ESI m/z (Cl?H24N2O2Na requires 15 311.1730) found 311.1736; t)inax/crn3210 (NH indole), 2906 (C-H), 1640 (C=O) and 1566 (aromatic); [a]nD(c = 0.523, CHC13) +63.77.
Example 20: (S)-3-(4'-teri-Butylbenzoylamino)-tetrahydropyridin-2-one
(S)-3-amino-tetrahydropyridin-2-one (33 mmoles) was dissolved in H2O (100 mL) and cooled to 0°C. 4-’Butylbenzoyl chloride (20 mmoles) in dichloromethane was added and triethylamine (6.3 mL, 45 mmoles) and the reaction was stirred over night. The 25 reaction was extracted with dichloromethane (3 x 20 mL), the organic layer was washed with a pH 2 buffer (3 x 20 mL), dried over Na2SO4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ether: ethyl acetate:methanol 50:50:0 to 0:80:20) to give the product as a white solid 3.13 g (53 %) mp 195-196 °C; ÔH (400 MHz, CDC13) 7.24 (d, 2H, J 8.5, Cff-C-lBu), 7.49 (d, 1H, 7 6, 30 N/7-CH). 7.36 (d, 2H, J 8.5, C/f-C-CO), 6.94 (br.s, 1H, Nff-Cl), 4.62 (dt, 1H, J 11,6,
1.5, C//-C4), 3.31-3.26 (m, 2H, Hl), 2.52 (ddt, IH, J 13, 6, 4.5, H3 équatorial), 1.90183 (m, 2H, H2), 1.63 (tt, IH, J 12.5, 8.5, H3 axial) and 1.27 (s, 9H, C(CAf3)3); Ôc (100 MHz, CDC13) 172.3 (Ç=O), 167.4 (CO), 154.9 (Ç-C(CH3)3), 131.2 (Ç-C=O), 127.0, 126.7, 125.3 (ÇH phenyl), 50.8 (ÇH-NH), 41.6 (Cl), 34.9 (Ç(CH3)3), 31.2 (C(ÇH3)3),
27.2 (C3) and 21.1 (C2); ESI m/z 100 %, 297.2 (MNa+) and 38 %, 275.2 (MH+); Pmax/cnf': 3251 (NH), 2959 (C-H), 1683, 1648 (C=O) and 1558 (aromatic); HR ESI m/z (C16H23N2O2 requires 275.1754) found 275.1752; [ct]23 D (c = 0.515, CHC13) +82.52.
Example 21: (R)-3“(4*-terr-Butylbenzoylamino)-tetrahydropyridin-2-one
NH (/?)-3-amino-tetrahydropyridin-2-one (15 mmoles) was dissolved in H2O (100 mL) and cooied to 0°C. 4-'Butylbenzoyl chloride (10 mmoles) in dichloromethane was added and triethylamine (4.2 mL, 30 mmoles) and the reaction was stirred over night. The reaction was extracted with dichloromethane (3 * 20 mL), the organic layer was washed with a pH 2 buffer (3 χ 20 mL), dried over Na2SO4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ether:ethyl acetateimethanol 50:50:0 to 0:80:20) to give the product as a white solid 1.03 g (38 %) mp 193-194 °C; ÔH (400 MHz, CDC13) 7.72 (d, 2H, J 8.5, C/AC-'Bu), 7.49 (d, IH, J 6, N/f-CH), 7.36 (d, 2H, J 8.5, CÇ-C-CO). 6.93 (br.s, IH, N/7-C1), 4.39 (dt, 1 H, J 11.5, 6, C/7-C4), 3.31-3.26 (m, 2H, Hl), 2.52 (ddt, IH, J 12.5, 5.5,4.5, H3 équatorial), 1.901.87 (m, 2H, H2), 1.63 (tt, IH, J 12.5, 8.5, H3) and 1.27 (s, 9H, C(C//3)3); ôc (100 MHz, CDCI3) 172.3 (Ç=O), 167.4 (C=O), 154.9 (C-C(CH3)3), 131.2 (Ç-C=O), 127.0, 125.3 (ÇH phenyl), 50.8 (ÇH-NH), 41.6 (Cl), 34.9 (C(CH3)3), 31.2 (C(ÇH3)3), 27.2 (C3) and 21.1 (C2); tUcm’1: 3247 (NH), 2958 (C-H), 1682, 1647 (C=O) and 1544 (aromatic); ESI m/z 19 %, 297.2 (MNa+) and 13 %, 275.2 (MH+); HR ESI m/z (Ci6H23N2O2requires 297.1573) found 275.1750; [a]23 D(c = 0.512, CHC13) -84.08.
Example 22: (S)-3-(4'-Hexylbenzoylamino)-azepan-2-one
(5)-3-amino-azepan-2-one hydrochloride (0.95 g, 5.79 mmoles) was dissolved in H2O (15 mL) and cooled to 0°C. 4-Hexylbenzoyl chloride (1.2 mL, 5 mmoles) in dichloromethane was added and triethylamîne (2.1 mL, 15 mmoles) and the reaction was stirred over night. H2O (20 mL) was added and the réaction was extracted with dichloromethane (3 x 20 mL), the organic layer was washed with a pH 2 buffer (3x15 mL), dried over Na2SO4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ether.ethyl acetate 50:50:0 to 0:80:20) to give the product as a white solid 0.76 g (48 %); mp 167-168 ° C; ôh (400 MHz, CDCI3) 7.74 (d, 2H, J 8, C/f-C-Hex), 7.61 (d, 1H, J 6, N//-CH), 7.21 (d, 2H, J 8, C/f-C-CO), 6.54 (br.t, 1H, J 6, Nff-Cl), 4.69 (ddd, 1 H, J 11, 6, 1.5, C7/-C4), 3.37-3.22 (m, 2H, Hl), 2.62 (t, 2H, J 7.5, H5), 2.21 (d, 1H, J 13, H4 équatorial), 2.03 (dt, 1H, J 14, 3.5, H3 équatorial), 1.95-1.82 (m, 2H, H2 équatorial & H3 axial), 1.64-1.49 (m, 3H, H4 axial &H6), 1.41 (q, 1H, J 13, H2 axial) 1.33-1.23 (m, 6H, H7, H8 & H9) and 0.86 (t, 3H, J 7, H10); ôc (100 MHz, CDCh) 175.9 (Ç=O lactam), 166.3 (Ç=O amide), 147.0 (CHex), 131.6 (Ç-C=O). 128.5 (CH phenyl), 127.1 (ÇH phenyl), 52.6 (ÇH-NH), 42.2 (Cl), 35.8 (C5), 31.7 (C4), 31.2 (C6), 29.0, 28.9, 28.0, (C2, C3, C7, and C8), 22.6 (C9) and 14.1 (CIO); o^/cm’1: 3244 (NH), 2956 (C-H), 1658, 1644 (C=O) and 1543 (aromatic); ESI m/z 43 %, 317.2 (MH+), 6% 339.2 (MNa+) and 6 %, 654.7 (M2Na+); HR ESI m/z (C|9H28N2O2Na requires 339.2043) found 339.2050; [a]25 D (c = 0.507, CHCI3) +60.06.
Example 23: (S)-3-(4'-Hexylbenzoylamino)-tetrahydropyridin-2-one
(S)-3-amino-tetrahydropyridin-2-one (ΙΟ mmoles) was dissolved in H2O (35 mL) and cooled to 0°C. 4-Hexylbenzoyl chloride (1.2 mL, 5 mmoles) in dichlorométhane was added and triethylamine (2.1 mL, 15 mmoles) and the reaction was stirred over night. The reaction was extracted with dichlorométhane (3x15 mL), the organic layer was washed with a pH 2 buffer (3x15 mL), dried over Na2SÜ4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ether:ethyl acetate 50:50:0 to 0:80:20) to give the product as a white solid 0.95 g (63 %); tnp 118-119 0 C; ÔH (400 MHz, CDClj) 7.70 (d, 2H, J 8, (V/-C-Hex). 7.38 (d, 1 H, J 6, N//-CH), 7.17 (d, 2H, J 8, CJf-C-CO), 6.81 (br.s, IH, N7/-CI), 4.39 (dt, 1 H, 7 11.5, 6, C//-C3), 3.333.26 (m, 2H, Hl), 2.58 (t, 2H, J 7.5, H4), 2.57-2.52 (m, IH, H3 équatorial obscured by H4), 1.92-1.84 (m, 2H, H2), 1.67-1.52 (m, 3H, H3 axial & H5), 1.29-1.23 (m, 6H, H6, H7 & H8) and 0.84 (t, 3H, J 7.5, H9); ); Ôc (100 MHz, CDCI3) 172.3 (Ç=O lactam), 167.5 (Ç=O amide), 146.9 (C-Hex), 131.5 (Ç-C=O), 128.4 (ÇH phenyl), 127.2 (ÇH phenyl), 50.8 (ÇH-NH), 41.6 (Cl), 35.8 (C4), 31.7 (C3), 31.2 (C5), 28.9 (C6), 27.2, (C7), 22.6 (C8), 21.1 (C2) and 14.1 (C9); 3338, 3247 (NH), 2921 (C-H),
1656, 1637 (C=O) and 1562 (aromatic); ESI m/z 100 %, 325.2 (MNa+) and 37% 303.2 (MH+); HR ESI m/z (€ιΚΗ26Ν2Ο2Ν3 requires 325.1886) found 325.1883; [ct]23D (c = 0.511, CHCh) +79.55.
Example 24: (R)-3-(4'-Hexylbenzoylamino)-tetrahydropyridin-2-one
(7?)-3-amino-tetrahydropyridin-2-one (10 mmoles) was dissolved in H2O (20 mL) and cooled to 0°C. 4-Hexylbenzoyl chloride (1.2 mL, 5 mmoles) in dichlorométhane was S added and triethylamine (2.1 mL, 15 mmoles) and the reaction was stirred over night. The reaction was extracted with dichloromethane (3><15 mL), the organic layer was washed with a pH 2 buffer (3x15 mL), dried over Na2SÜ4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ether:ethyl acetate 50:50:0 to 0:80:20) to give the product as a white solid 0.53 g (35 %); mp 117-118 ° C; Ôh (400 MHz, CDClj) 7.71 (d, 2H, J 8, CH-C-Hex), 7.28 (d, 1H, J5, 7.19 (d,
2H, J 8, Cff-C-CO), 6.52 (br.s, 1H, NH-C1), 4.41 (dt, 1 H, J 11.5, 5.5, 3.353.31 (m, 2H, Hl), 2.67-2.60 (m, 1H, H3 équatorial), 2.61 (t, 2H, J 7.5, H4), 2.00-1.89 (m, 2H, H2), 1.67-1.54 (m, 3H, H3 équatorial & H5), 1.32-1.23 (m, 6H, H6, H7 & H8) and 0.85 (t, 3H, J 7, H9); ); Ôc (100 MHz, CDCh) 172.1 (Ç=O lactam), 167.6 (Ç=O amide), 147.0 (Ç-Hex), 131.5 (Ç-C=O), 128.5 (ÇH phenyl), 127.2 (ÇH phenyl), 50.9 (ÇH-NH), 41.7 (Cl), 35.8 (C4), 31.7 (C3), 31.2 (C5), 28.9 (C6), 27.2, (C7), 22.6 (C8), 21.1 (C2) and 14.1 (C9); wmax/cm'‘: 3328, 3240 (NH), 2954 (C-H), 1651, 1635 (C=O) and 1533 (aromatic); ESI m/z 100 %, 325.2 (MNa+) and 33% 303.2 (MH+); HR ESI m/z (Ci8H26N2O2Na requires 325.1886) found 325.1888; [a]25o (c = 0.496, CHC13) 81.17.
Example 25: (S)-3-(4’-Octylbenzoylamïno)-azepan-2-one
(5)-3-amino-azepan-2-one hydrochloride (0.91 g, 5.55 mmoles) was dissolved in H2O (15 mL) and cooled to 0°C. The 4-octylbenzoyl chloride in dichloromethane was added and triethylamine (0.84 mL, 6 mmoles) and the reaction was stirred over night. H2O (20 mL) was added and the reaction was extracted with dichloromethane (3 x 20 mL), the organic layer was washed with a pH 2 buffer (3 x 15 mL), dried over Na2SO4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ether;ethyl acetate 50:50:0 to 0:80:20) to give the product as a white solid 0.087 g (4 %); mp 159-160 0 C; ÔH (400 MHz, CDC13) 7.73 (d, 2H, J 8, CH-C-Oct),
7.64 (d, IH, J 5.5, N//-CH). 7.20 (d, 2H, J 8, CH-C-CO), 6.94 (br.t, IH, J 6, N//-C1), 4.68 (dd, IH, J 11, 6, C/f-C4), 3.35-3.20 (m, 2H, HI), 2.60 (t, 2H, J7.5, H5), 2.19 (d, IH, J 13, H4 équatorial), 2.00 (br.d, IH, H3 équatorial), l.92-1,79 (m, 3H, H2 & H3 axial), 1.62-1.46 (m, 3H, H4 axial & H6), 1.38 (q, 1 H, J 11.5, H2 axial) 1.30-1.19 (m, 10H, H7, H8, H9, H10 & Hll) and 0.84 (t, 3H, J 7.5, H12); ôc (100 MHz, CDCh) 176.0 (Ç=O lactam), 166.3 (Ç=O amide), 146.9 (Ç-Oct), 131.6 (Ç-C=O), 128.5 (ÇH phenyl), 127.1 (ÇH phenyl), 52.5 (ÇH-NH), 42.1 (Cl), 35.8 (C5), 31.9 (C4), 31.6 (C6), 31.2 (C7), 29.4, 29.3,28.9,28.0, (C2, C3, C8, C9 and C10), 22.7 (Cil) and 14.1 (C12); tw/cm’1: 3204 (NH indole), 2923 (C-H), 1637 (amide C=O) and 1544 (aromatic); ES1 m/z 100 %, 345.2 (MH+) and 9 %, 710.8 (M2Na+); HR ES1 m/z (C2[H32N2O2Na requires 367.2356) found 367.2361 ; [a]25 D(c = 0.124, CDC13) +68.01.
Example 26: (8)-3-(4'-Octyibenzoylamino)-tetrahydropyridin-2-one
(5)-3-amino-tetrahydropyridin-2-one (10 mm oies) was dissolved in H2O (35 mL) and cooled to 0°C. 4-Octylbenzoyl chloride (2 mmoles) in dichloromethane was added and triethylamine (0.85 mL, 6 mmoles) and the reaction was stirred over night. The reaction was extracted with dichloromethane (3 x 10 mL), the organic layer was washed with a pH 2 buffer (3x10 mL), dried over Na2SC>4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ether:ethyl acetate 50:50:0 to 0:80:20) to give the product as a white solid 0.55 g (83 %); mp 122-123 0 C; δΗ (400 MHz, CDCI3) 7.71 (d, 2H, J 8, CH-C-Oct), 7.26 (d, 1 H, J 5.5, N/f-CH), 7.19 (d, 2H, J 8, C7/-C-CO). 6.47 (br.s, IH, NH-C1), 4.41 (dt, IH, J 11.5, 5.5, CH-C3), 3.33-3.27 (m, 2H, Hl), 2.84-2.58 (m, IH, J 13, H3 équatorial obscured by H4), 2.60 (t, 2H, J 7.5, H4), 1.97-1.90 (m, 2H, H2), 1.67-1.54 (m, 3H, H3 axial & H5), 1.29-1.23 (m, 10H, H6, H7, H8, H9 & H10) and 0.86 (t, 3H, J 7, Hl 1); Ôc (100 MHz, CDCI3) 172.1 (Ç=O lactam), 167.6 (Ç=O amide), 147.0 (Ç-Oct), 131.5 (Ç-C-O), 128.5 (ÇH phenyl), 127.2 (ÇH phenyl), 50.9 (ÇH-NH), 41.7 (Cl), 35.8 (C4), 31.9 (C5), 31.2 (C3), 29.4 (C6), 29.3 (C7, C8), 27.2 (C9), 22.3 (CIO), 2l.l (C2) and 14.1 (Cil); ümax/cm·’: 3240 (NH), 2921 (C-H), 1653, 1615 (C=O) and 1563 (aromatic); ESI m/z 100 %, 353.2 (MNa+) and 47 %, 331.2 (MH+); HR ESI m/z (C20H3oN202Na requires 353.2199) found 353.2198; [a]23D(c = 0.509, CHC13) +75.74.
Example 27: (R)-3-(4’-Octylbenzoylamino)-tetrahydropyridin-2-one
(7?)-3-amino-tetrahydropyridin-2-one (5 mmoles) was dissolved in H2O (25 mL) and cooled to 0°C. 4-Octylbenzoyl chloride (2 mmoles) in dichloromethane was added and triethylamine (0.85 mL, 6 mmoles) and the reaction was stirred over night. The reaction was extracted with dichloromethane (3><10 mL), the organic layer was washed with a pH 2 buffer (3 x 10 mL), dried over Na2SÛ4 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ether:ethyl acetate 50:50:0 to 0:80:20) to give the product as a white solid 0.16 g (25 %); mp 122-123 0 C; δΗ (400 MHz, CDClj) 7.71 (d, 2H, J 8.5, C/f-C-Oct), 7.23 (d, i H, 7 6.5, N/ACH), 7.20 (d, 2H, J 8.5, CH-C-CO), 6.34 (br.s, 1H, N/AC1), 4.41 (dt, 1 H, J 11.5, 5.5, C/AC3), 3.37-3.33 (m, 2H, Hl), 2.68 (ddt, 1H, J 13, 5.5, 4.5, H3 équatorial), 2.61 (t, 2H, J 7.5, H4), 1.98-1.91 (m, 2H, H2), 1.67-1.55 (m, 3H, H3 axial & H5), 1.31-1.22 (m, 10H, H6, H7, H8, H9 & H10) and 0.86 (t, 3H,7 7, Hl 1); Ôc (100 MHz, CDC13) 172.1 (Ç=O lactam), 167.6 (C=O amide), 147.0 (Ç-Oct), 131.5 (Ç-C=O), 128.5 (ÇH phenyl), 127.2 (ÇH phenyl), 51.0 (ÇH-NH), 41.7 (Cl), 35.8 (C4), 31.9 (C5), 31.2 (C3), 29.4 (C6), 29.3 (C7, C8), 27.2 (C9), 22.7 (C10), 21.1 (C2) and 14.1 (Cil); ESI m/z 39 %, 353.2 (MNa+), 19 %, 331.2 (MH+) and 14 %, 682.7 (M2Na+); HR ESI m/z (C20H30N2O2H+ requires 331.2380) found 331.2381; υ^/cm'1: 3250 (NH), 2955 (C-H), 1653 (C=O) and 1540 (aromatic); [a]23o(c = 0.485, CHC13)-77.80. —
Pharmacological study of the products of the invention
A. Inhibition ofMCP-l induced leukocyte migration
Assay principle
The biological activity of the compounds of the current invention may be demonstrated using any of a broad range of functional assays of leukocyte migration in vitro, including but not limited to Boyden chamber and related transwell migration assays, under-agarose migration assays and direct visualisation chambers such as the Dunn Chamber.
For example, to demonstrate the inhibition of leukocyte migration in response to chemokines (but not other chemoattractants) the 96-well format micro transwell assay system from Neuroprobe (Gaîthersburg, MD, USA) has been used. In principle, this assay consists of two chambers separated by a porous membrane. The chemoattractant is placed in the lower compartment and the cells are placed in the upper compartment. After incubation for a period at 37°C the cells move towards the chemoattractant, and the number of cells in the lower compartment is proportional to the chemoattractant activity (relative to a sériés of controls).
This assay can be used with a range of different leukocyte populations. For example, freshly prepared human peripheral blood leukocytes may used. Altematively, leukocyte subsets may be prepared, including polymorphonuclear cells or lymphocytes or monocytes using methods well known to those skilled in the art such as density gradient centrifugation or magnetic bead séparations. Altematively, immortal cell lines which hâve been extensively validated as models of human peripheral blood leukocytes may be used, including, but not limited to THP-l cells as a model of monocytes or Jurkat cells as model of naïve T cells.
Although a range of conditions for the assay are acceptible to demonstrate the inhibition of chemokine-induced leukocyte migration, a spécifie example is hereby provided. -A
Materials S
The transwell migration Systems are manufactured by Neuroprobe, Gaithersburg, MD,
USA.
The plates used are ChemoTx plates (Neuroprobe 101-8) and 30 μΐ clear plates (Neuroprobe MP30).
Geys’ Balanced Sait Solution is purchased from Sigma (Sigma G-9779).
Fatty acid-free BSA is purchased from Sigma (Sigma A-8806).
MTT, i.e. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tétrazolium bromide, is purchased from Sigma (Sigma M-5655).
RPMI-1640 without phénol red is purchased from Sigma (Sigma R-8755).
The THP-l cell line (European Cell culture Collection) were used as the leukocyte cell population.
Test protocol
The following procedure is used for testing the invention compounds for MCP-l induced leukocyte migration:
First, the cell suspension to be placed in the upper compartment is prepared. The THP1 cells are pelleted by centrifugation (770 x g; 4 mins) and washed with Geys Balanced Sait Solution with 1 mg/ml BSA (GBSS + BSA). This wash is then repeated, and the cells repelleted before beîng resuspended in a small volume of GBSS + BSA for counting, for example using a standard haemocytometer.
The volume of GBSS + BSA is then adjusted depending on the number of cells présent so that the cells are at final density of 4.45 x 106 cells per ml of GBSS + BSA. This ensures that there are 100,000 THP-l cells in each 25μΐ of the solution that will be placed in the upper chamber of the plate.
To test a single compound for its ability to inhibit MCP-l induced migration, it is necessary to préparé two lots of cells. The suspension of THP-l cells at 4.45 x 106 ceils/ml is divided into two pots. To one pot the inhîbitor under test is added at an appropriate final concentration, in an appropriaie vehicle (for example at 1 μΜ in not 52 more than 1% DMSO). To the second pot an equal volume of GBSS + BSA plus vehicle as appropriate (e.g. not more than 1% DMSO) is added to act as a control.
Next, the chemoattractant solution to be placed in the lower compartment is prepared. MCP-1 îs diluted in GBSS + BSA to give a final concentration of 25 ng/ml. This is dîvided into two pots, as for the cell suspension. To one pot, the test compound is added to the same final concentration as was added to the cell suspension, while to the other pot an equal volume of GBSS + BSA plus vehicle as appropriate (e,g. not more than 1 % DMSO) îs added.
Note that the volume of liquid that needs to be added to make the addition of the text compound needs to be taken into account, when estabiishing the final concentration of MCP-1 in the solution for the lower compartment and the final concentration of cells in the upper compartment.
Once the chemoattractant solutions for the lower wells and cell solutions for the upper chambers hâve been prepared, the migration chamber should be assembled. Place 29 μΐ of the appropriate chemoattractant solution into the lower well of the chamber. Assays should be performed with at least triplicate déterminations of each condition. Once all the lower chambers hâve been filled, apply the prous membrane to the chamber in accordance with the manufacturer’s instructions. Finally, apply 25 μΐ of the appropriate cell solution to each upper chamber. A plastic lid is placed over the entire apparatus to prevent évaporation.
The assembled chamber îs incubated at 37 °C, 5% CO2, for 2 hours. A suspension of cells in GBSS + BSA is also incubated under identical conditions in a tube: these cells will be used to construct a standard curve for determining the number of cells that hâve migrated to the lower chamber under each condition.
At the end of the incubation, the liquid cell suspension is gently removed from the upper chamber, and 20μ1 of ice-cold 20mM EDTA in PBS is added to the upper chamber, and the apparatus is incubated at 4°C for 15 mins. This procedure causes any cells adhering to the underside of the membrane to fall into the lower chamber.
After this incubation the fil ter is carefully flushed with GBSS + BSA to wash off the EDTA, and then the filter îs removed.
The number of cells migrated into the lower chamber under each condition can then be determined by a number of methods, including direct counting, labelling with fluorescent or radioactive markers or through the use of a vital dye. Typically, we utilise the vital dye MTT, 3 μΐ of stock MTT solution are added to each well, and then the plate is incubated at 37 °C for l -2 hours during which time dehydrogenase enzymes within the cells couvert the soluble MTT to an insoluble blue formazan product that can be quantified spectrophotometrically.
In parallel, an 8-point standard curve is set up. Starting with the number of cells added to each upper chamber (100,000) and going down in 2-fold serial dilutions in GBSS + BSA, the cells are added to a plate in 25 μΐ, with 3 μΐ of MTT stock solution added. The standard curve plate is incubated along side the migration plate.
At the end of this incubation, the liquid is carefully removed from the lower chambers, taking care not to disturb the precipîtated formazan product. After allowing to air dry briefly, 20μ1 of DMSO is added to each lower chamber to solubilise the blue dye, and absorbance at 595nm is determined using a 96-weil plate reader. The absorbance of each well is then interpolated to the standard curve to estimate the number of cells in each lower chamber.
The MCP-1 stimulated migration is determined by subtracting the average number of cells that reached the lower compartment in wells where no MCP-1 was added from the average number of cells that reached the lower compartment where MCP-1 was présent at 25ng/ml.
The impact of the test substance is calcul ated b y comparing the MCP-l-induced migration which occurred in the presence or absence of various concentrations of the test substance. Typically, the inhibition of migration is expressed as a percentage of the total MCP-1 induced migration which was blocked by the presence of the compound. For most compounds, a dose-response graph is constructed by detennining the inhibition of MCP-1 induced migration which occurs at a range of different compound concentrations (typically ranging from 1 nM to 1 μΜ or higher in the case of poorly active compounds). The inhîbitory activity of each compound is then expressed as the concentration of compound required to reduce the MCP-l-induced migration by 50% (the ED50 concentration).
Results
The compounds of reference ex amples I to 14 were tested and were shown to hâve an ED50 of 100 nM or less in this test.
B. In vivo assay
The anti-înflammatory efficacy of an exemplary compound according to the présent invention was tested using the murine sub-lethal endotoxemia model. This model has been widely used to demonstrate the antî-inflainmatory effect of compounds in vivo Fox étal., 2009, J Med Chem. 52(11): 3591-3595.
Briefly, the method is as follows: Female CD1 mice (28-30g, ~7 weeks of âge) were dosed with their respective treatment in stérile filtered 1% CMC by oral gavage in a dose volume of lOml/kg one hour prior to an endotoxin (LPS) challenge. The endotoxîn challenge was injected by the intraperitoneal route contaîning 675,000 Endotoxin Units of LPS (E. coli strain 0111 :B4 (Code L4130)) in endotoxin free PBS. Mice were left for two hours and then exsanguinated under terminal anaesthesia and blood was taken. Sérum was prepared from this terminal bleed and aliquoted and stored at -20’C. Sérum TNF-α levels were measured by EL1SA per manufacturers instructions (R and D Systems).
Eight animais were treated in each group, and the data for the animal with the highest and lowest TNF- a level in each group were eliminated, and the mean and standard error reported for the remaining six animais. Data for untreated animais were taken from an historical control experiment.
A single dose of (S)-4-Fluoro-N-(2-oxopiperidin-3-yl)benzamide (also known as (S)3-(4’-fluorobenzoylamino)-tetrahydropyridin-2-one; see Example 3 above) administered by oral gavage, inhibited endotoxin-stimulated TNF-alpha levels by 50% (see Fig. 2, column B).
This experiment demonstrates that the compounds according to the invention hâve anti-înflammatory activîty in vivo.

Claims (20)

  1. Claims
    1. A compound of general formula (I), or a pharmaceutically acceptable sait thereof, for use in the treatment of an inflammatory disorder:
    (I) wherein n îs an integer from 1 to 4;
    k is an integer from 0 to 5, representing the number of groups substituting C2. C3, C4, C5 and/or C6 of the benzyl ring; and
    X are linear or branched groups substituting the benzyl ring independently selected from any one of the group consisting of: alkyl, haloalkyl, hydroxyalkyl, hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, carboxy, and halogen;
    with the proviso that:
    when on the benzyl ring C2, C5 and C& are unsubstituted, and C4 is unsubstituted or is substituted with an hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, or halogen group, then C3 is substituted with a halogen group; and when on the benzyl ring C2, C5 and C(1 are unsubstituted, and C3 is unsubstituted or is substituted with an alkyl, haloalkyl, hydroxyalkyl, hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl or carboxy group, then C4 is substituted with any one of the group consisting of: alkyl group, haloalkyl group, hydroxyalkyl group, and carboxy group.
  2. 2. A compound of formula (I’), or a pharmaceutically acceptable sait thereof, for use in the treatment of an inflammatory disorder: —+ (Γ) wherein η, k and X are defined as in claim l.
  3. 3. Use of a cornpound of general formula (I), or a pharmaceutically acceptable sait
    5 thereof, in the manufacture of a médicament for the treatment of an inflammatory disorder:
    wherein
    10 n is an integer from 1 to 4;
    k is an integer from 0 to 5, representing the number of groups substituting C2, C3, C4, C5 and/or Cf, of the benzyl ring; and
    X are linear or branched groups substituting the benzyl ring independently selected from any one of the group consisting of: alkyl, haloalkyl, hydroxyalkyl, hydroxy,
    15 alkoxy, amino, aminoalkyl, aminodialkyl, carboxy, and halogen;
    with the proviso that:
    when on the benzyl ring C2, Q and C(l are unsubstituted, and C4 is unsubstituted or is substituted with an hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, or halogen group, then C3 is substituted with a halogen group; and when on the benzyl ring C2, C5 and C(, are unsubstituted, and C3 is unsubstituted or is
    5 substituted with an alkyl, haloalkyl, hydroxyalkyl, hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl or carboxy group, then C4 is substituted with any one of the group consisting of: alkyl group, haloalkyl group, hydroxyalkyl group and carboxy group.
  4. 4. Use of a compound of formula (F), or a pharmaceutically acceptable sait thereof, in
    10 the manufacture of a médicament for the treatment of an infiammatory disorder:
    (Γ) wherein n, k and X are defïned as in claim 3.
  5. 5. A compound of general formula (I):
    wherein n is an integer from l to 4;
    k is an integer from l to 5, representing the number of groups substituting C2, C3, C4,
    C5 and/or C« of the benzyl ring;
    when n is l or 2, X are linear or bran ch ed groups independently selected from any one ofthe group consisting of: C7orhigher alkyl, haloalkyl with a C7orhigher alkyl group, hydroxyalkyl with a C7 or higher alkyl group, C7 or greater alkoxy, aminoalkyl with a C4 or higher alkyl group, aminodialkyl with two C4 or higher alkyl groups, and carboxy;
    when n is 3 or 4, X are linear or branched groups independently selected from any one of the group consisting of: alkyl, haloalkyl, hydroxyalkyl, hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, carboxy, and halogen;
    with the proviso that:
    when n is 3 or 4 and on the benzyl ring C2, C5 and C6 are unsubstituted, and C4 is unsubstituted or is substituted with an hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, or halogen group, then C3 is substituted with a halogen group;
    when n is 3 or 4 and on the benzyl ring C2, C5 and C\ are unsubstituted, and C3 is unsubstituted or is substituted with an alkyl, haloalkyl, hydroxyalkyl, hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl or carboxy group, then C4 is substituted with any one ofthe group consisting of: alkyl group, haloalkyl group, hydroxyalkyl group, and carboxy group; and when n=3, X is other than 4’-methoxy) 3’-trifluoromethyl, or 3’,4’,5’-trimethoxy, provided that the compound is none of the group consisting of: 3-(3'trifluoromethylbenzoylamino)-caprolactam, 3-(4'-methylbenzoylamino)-caprolactam, 3-(2’-aminobenzoylamino)-caprolactam, 3-(3',4'-dimethoxybcnzoylamino)caprolactam, 3-(3,,5'-di-i'er/-butyl -4'- hydroxybenzoylamino)-caprolactam, 3-(2’,4dimethoxybenzoylamino)-caprolactam, 3-(3’-methoxybenzoylamino)-caprolactam, 3(4'-trifluoromethylbenzoylamino)-caprolactam, 3-(2',3,,4'-trimethoxybenzoylamino)caprolactam, 3-(2',6'-difluoromethylbenzoylamino)-caproiactam, 3-(2'fluoromethylbenzoylamino)-caprolactam, 3-(2’-amino-3’-hydiOxy-4'methylbenzoylamino)-caprolactam, and 3-(3’,5’-dimethyIbenzoylamino)-caprolactam.
  6. 6. A compound of formula (I’):
    (Γ) wherein η, k and X are defined as in either of claim l or claim 5, provided that the compound is compound is none of the group consisting of: (S)-3-(4’methoxybenzoylamino)-caprolactam, (S)-3-(4’-methylbenzoylamîno)-caprolactam, (S)-3-(3’-trifluoromethylbenzoylamino)-caprolactam, (S)-3-(2'-carboxybenzoylamino)-caprolactam, and (S)-3-(3’, 4,,5'-trimethoxybenzoylamino)-caprolactam.
  7. 7. A pharmaceutical composition comprising, as active ingrédient, a compound as defined în either of claims 5 or 6, or a pharmaceutically acceptable sait thereof, and at least one pharmaceutically acceptable excipient and/or carrier.
  8. 8. The compound, use or composition according to any preceding claim, wherein n=2.
  9. 9. The compound, use or composition according to any preceding claim, wherein n=3.
  10. 10. The compound, use or composition according to any preceding claim, wherein X is haloalkyl, for example trifluoromethyl.
  11. 11. The compound according to claims 1 or 2, or the use according to claims 3 or 4, wherein the compound is selected from the group consisting of:
    (S)-3-(4'-methylbenzoylamino)-caprolactam, and (S)-3-(3',5’-dimethylbenzoylamino)-caprolactam, and pharmaceutically acceptable salts thereof.
  12. 12. The compound according to any of claims 1, 2 or 6, or the use according to claims
    3 or 4, wherein the compound is selected from the group consisting of:
    (S)-3-Fluoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-2-Fluoro-N-(2-oxopîperidin-3-yl)benzamide, (5)-4-Fluoro-N-(2-oxopiperidîn-3-yl)benzamide, (5)-N-(2-Oxopiperidin-3-yl)-4-(trifluoromethyl)benzamide, (5)-N-(2-Oxopiperidin-3 -yl )-3 -(tri fluoromethyl)benzamide, (5)-N-(2-Oxopiperidin-3-yl)-2-(trifluoromethyl)benzainîde, (5)-2,3-difluoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-2,4-dîfluoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-2,5-difluoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-2,6-difluoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-3,4-difluoro-N-(2-oxopiperidin-3-yl)benzamide, (5)-3,5-difluoro-N-(2-oxopiperidin-3-yl)benzamide, (S)-3-(3'-Butylbenzoylamîno)-azepan-2-one, (S)-3-(4'-Ethylbenzoylamino)-tetrahydropyridin-2-one, (S)-3-(4’-Butylbenzoylamino)-tetrahydropyridin-2-one, (S)-3-(4’-tert-Butylbenzoylamino)-tetrahydropyridin-2-one, and (S)-3-(4'-Hexylbenzoylamino)-tetrahydropyridin-2-one, and pharmaceutically acceptable salts thereof.
  13. 13. The compound according to any of daims 1, 2, 5 or 6, or the use according to daims 3 or 4, wherein the compound is selected from the group consisting of:
    (S)-3-(4'-Ethylbenzoylamino)-azepan-2-one, (S)-3-(4'-Butylbenzoylamino)-azepan-2-one, (S)-3-(4'-fôr/-Butylbenzoylamino)-azepan-2-one, ( S ) - 3 -(4'- Hexylbenzoyl amino )-azepan-2 -one, (S)-3-(4'-Octylbenzoylamino)-azepan-2-one, and (S)-3-(4'-OctylbenzoyIamino)-tetrahydropyridin-2-one, and pharmaceutically acceptable salts thereof.
  14. 14. The compound according to claim 1, or the use according to daim 3, wherein the compound is selected from the group consisting of:
    (R)-3-(4'-Butylbenzoylamino)-tetrahydropyridin-2-one, (R.)-3-(4,-fô/7-Butylbenzoylamîno)-tetrahydropyridin-2-one, and (R)-3-(4,-Hexylbenzoylamino)-tetrahydropyridin-2-one, and pharmaceutically acceptable salts thereof.
  15. 15. The compound according to either of claim 1 or claim 5, or the use according to claim 3, wherein the compound is (R)-3-(4,-Octylbenzoylamino)-tetrahydropyridin-2one or a pharmaceutically acceptable sait thereof.
  16. 16. The compound according to either of claims 1 or 2 or the use according to either of claims 3 or 4, wherein the inflammatory disorder is selected from the group consisting of autoimmune diseases, asthma, rheumatoid arthritis, a disorder characterised by an elevated TNF-α level, psoriasis, allergies, multiple sclerosis, fibrosis (including diabetic nephropathy), and formation of adhesions.
  17. 17. The compound or use according to claim 16, wherein the inflammatory disorder is formation of adhesions.
  18. 18. The compound or use according to claim 16 or claim 17, wherein the compound is administered locally.
  19. 19. Use of a compound, pharmaceutical composition or médicament as defined in any preceding claim in the manufacture of a médicament for the treatment, amelioration or prophylaxis of the symptoms of an inflammatory disease (including an adverse inflammatory reaction to any agent).
  20. 20. A library consisting of éléments ail of which hâve structures according to the formula (I) or (P) as defined in any of claims Ito 18, and hence which ail hâve antiinflammatory activity, useful for screening compounds for novel or improved properties in a particular assay of anti-inflammatory activity.
OA1201200503 2010-06-08 2011-06-08 Anti-inflammatory agents. OA16265A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB1009603.0 2010-06-08

Publications (1)

Publication Number Publication Date
OA16265A true OA16265A (en) 2015-04-10

Family

ID=

Similar Documents

Publication Publication Date Title
EP2134680B1 (en) Inhibitors of histone deacetylase
EP1896036B1 (en) Anti-inflammatory agents
US20140336227A1 (en) 3-amino lactams as anti-inflammatory agents
JP5248779B2 (en) Anti-inflammatory agent
US6962926B2 (en) Antagonist of MCP-1 function, and compositions and methods of use thereof
EP1646622B1 (en) Tacrine derivatives as inhibitors of acetylcholinesterase
GB2418425A (en) 3-Acylaminocaprolactams containing a mono- or poly-cyclic cyclo-alkane or -alkene group in the acyl moiety for treatment of inflammatory disorders
OA16265A (en) Anti-inflammatory agents.
AU2011263531A1 (en) Anti-inflammatory agents
JPH0441149B2 (en)
OA16264A (en) Anti-inflammatory agents.
AU2011263530A1 (en) Anti-inflammatory agents
KR100866285B1 (en) Novel catechol hydrazide derivatives and process for preparation thereof
KR100866286B1 (en) Novel catechol hydrazide derivatives and process for preparation thereof
KR100736838B1 (en) Novel catechol N-methylhydrazide derivatives and process for preparation thereof
WO2024040155A1 (en) Isoxazolidines as ripk1 inhibitors and use thereof
MXPA06006218A (en) Anti-inflammatory agents
GB2430674A (en) Anti-inflammatory 3-([1-substituted-carbocyclyl](carbonyl/sulphonyl)amino) lactams
KR20070038309A (en) Deformylase inhibitor, process for the preparation thereof, and composition comprising the same