WO2011154696A1 - Anti-inflammatory agents - Google Patents

Anti-inflammatory agents Download PDF

Info

Publication number
WO2011154696A1
WO2011154696A1 PCT/GB2011/000863 GB2011000863W WO2011154696A1 WO 2011154696 A1 WO2011154696 A1 WO 2011154696A1 GB 2011000863 W GB2011000863 W GB 2011000863W WO 2011154696 A1 WO2011154696 A1 WO 2011154696A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
caprolactam
oxopiperidin
benzamide
Prior art date
Application number
PCT/GB2011/000863
Other languages
French (fr)
Inventor
David John Grainger
David John Fox
Original Assignee
Cambridge Enterprise Limited
Funxional Therapeutics Limited
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=42471347&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2011154696(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to SG2012081667A priority Critical patent/SG185129A1/en
Priority to US13/702,898 priority patent/US20130172318A1/en
Priority to MX2012014290A priority patent/MX2012014290A/en
Priority to AP2012006559A priority patent/AP2012006559A0/en
Priority to AU2011263531A priority patent/AU2011263531A1/en
Priority to CN201180028686XA priority patent/CN103119021A/en
Priority to JP2013513742A priority patent/JP2013531647A/en
Priority to BR112012030816A priority patent/BR112012030816A2/en
Priority to CA2798213A priority patent/CA2798213A1/en
Priority to EP11725490.4A priority patent/EP2580196A1/en
Application filed by Cambridge Enterprise Limited, Funxional Therapeutics Limited filed Critical Cambridge Enterprise Limited
Priority to KR1020127033497A priority patent/KR20130086952A/en
Priority to MA35548A priority patent/MA34370B1/en
Publication of WO2011154696A1 publication Critical patent/WO2011154696A1/en
Priority to TNP2012000521A priority patent/TN2012000521A1/en
Priority to IL222798A priority patent/IL222798A0/en
Priority to ZA2012/08367A priority patent/ZA201208367B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D223/00Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom
    • C07D223/02Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D223/06Heterocyclic compounds containing seven-membered rings having one nitrogen atom as the only ring hetero atom not condensed with other rings with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D223/12Nitrogen atoms not forming part of a nitro radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/45Non condensed piperidines, e.g. piperocaine having oxo groups directly attached to the heterocyclic ring, e.g. cycloheximide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/56Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/68Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D211/72Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D211/74Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/68Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D211/72Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D211/74Oxygen atoms
    • C07D211/76Oxygen atoms attached in position 2 or 6
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the invention relates to aryl substituted 3-aminolactam derivatives and their use in preventing or treating inflammatory diseases.
  • Inflammation is an important component of physiological 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 have not traditionally been considered to involve leukocytes (such as osteoporosis or Alzheimer's disease).
  • the chemokines are a large family of signalling molecules with homology to interleukin-8 which have been implicated in regulating leukocyte trafficking both in physiological and pathological conditions.
  • the system 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.
  • this complexity of the chemokine system has at first hindered
  • chemokine receptor blockade It has proved difficult to determine which chemokine receptor(s) should be inhibited to produce therapeutic benefit in a given inflammatory disease.
  • BSCIs Broad Spectrum Chemokine Inhibitors
  • peptides and peptoid derivatives such as NR58-3.14.3, may not be optimal for use in vivo. They are quite expensive to synthesise and have relatively
  • NR58- 3.14.3 is not orally bioavailable and is cleared from blood plasma with a half-life period of less than 30 minutes after intravenous injection.
  • aminoglutarimide and aminolactam BSCIs have focussed almost exclusively on compounds with simple linear and branched alkyl side chains.
  • aminoglutarimide ring was susceptible to enzymatic ring opening in serum. Consequently, for some applications (for example, where the inflammation under treatment is chronic, such as in autoimmune diseases) these compounds may not have optimal properties, and a more stable compound with similar anti-inflammatory properties may be superior.
  • BSCIs stable, broad spectrum chemokine inhibitors
  • 3- amino caprolactams with a seven-membered monolactam ring
  • further useful anti-inflammatory compounds have also been generated from other 3-aminolactams with different ring size (see for example WO2006/134385).
  • BSCI activity is conferred on the molecule 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 have little impact).
  • this "head group” must have 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.
  • tail groups 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 10-fold increase in potency as a BSCI, at least in vitro, compared to a related compound lacking 2'2'-disubstitution.
  • 2'2'- dimethyldodecenanoyl-3-aminocaprolactam is 10-fold more potent as a BSCI in the MCP-1 induced THP-1 cell migration than assay than dodecanoyl-3- aminocaprolactam (as disclosed previously in WO2005/053702), 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.
  • the pivoyl "tail group” of 2'2'-dimethylpropanoyl-3-aminovalerolactam contributes to the unexpected, and particularly favourable, pharmaceutical properties of this molecule (as disclosed previously in WO2009/016390).
  • the pivoyl group is resistant to
  • the present invention discloses a series 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.
  • all of the compounds as set out below have substantial BSCI activity (greater than either 2 ',3 '-unsaturated acyl 3-aminolactams or benzoylaminoglutarimides).
  • n is an integer from 1 to 4; k is an integer from 0 to 5, representing the number of groups substituting C 2 , C 3 , C 4 , C 5 and/or C 6 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 C 2 , C 5 and C 6 are unsubstituted, and C 4 is unsubstituted or is substituted with an hydroxy, alkoxy, amino, aminoalkyl, aminodialkyl, or halogen group, then C 3 is substituted with a halogen group; and when on the benzyl ring C 2 , C 5 and C 6 are unsub
  • the carbon atom at position 3 of the lactam ring is asymmetric and consequently, the compounds according to the present invention have at least two possible enantiomeric forms, that is, the "R” and “S” configurations.
  • the present invention encompasses each of the two enantiomeric forms and all combinations of these forms, including the racemic "RS" mixtures. With a view to simplicity, when no specific configuration is shown in the structural formula, it should be understood that each of the two enantiomeric forms and their mixtures are represented.
  • the invention also provides the use of a compound of general formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of an inflammatory disorder:
  • n is an integer from 1 to 4; k is an integer from 0 to 5, representing the number of groups substituting C 2 , C 3 , C 4 , C 5 and/or C 6 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:
  • n is an integer from 1 to 4; k is an integer from 1 to 5, representing the number of groups substituting C 2 , C 3 , C 4 , C 5 and/or C 6 of the benzyl ring; when n is 1 or 2, X are linear or branched groups independently selected from any one of the group consisting of: C 7 or higher alkyl, haloalkyl with a C 7 or higher alkyl group, hydroxyalkyl with a C 7 or higher alkyl group, C7 or g re ater alkoxy, aminoalkyl with a C 4 or higher alkyl group, aminodialkyl with two C 4 or higher 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,
  • n, k and X are defined herein for general formula (I), provided that the compound is none of the group consisting of: (S)-3-(3'-trifluoro- methylbenzoylamino)-caprolactam, (S)-3-(4' -methylbenzoylamino)-caprolactam, (S)- 3-(4'-methoxybenzoylamino)-caprolactam, (S)-3-(2'-carboxybenzoylamino)- caprolactam, and (S)-3-(3', 4 , ,5'-trimethoxybenzoylamino)-caprolactam.
  • WO2007/0038669 teaches diarylamine-containing compounds and their use as moduclators of c-kit receptors.
  • Various intermediate compounds are used in the synthesis of the diarylamine-containing compounds. Any overlap of the intermediate compounds is hereby disclaimed from the present invention.
  • EP0462949 together with the related publication Angelucci et al, 1993, J Medicinal Chemistry 36: 1512-1519 teach 7-membered 3-acylamino lactams as enhancers of learning and memory.
  • JP03206042 discloses the preparation of 5,6,7,8-tetrahydo-4H-thiazolo[5,4-b]azepine derivatives with potassium channel activation activity, for use as antihypertensives.
  • a pharmaceutical composition comprising, as active ingredient, a compound per se as defined above, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient and/or carrier.
  • salt in particular the addition salts of inorganic acids such as hydrochloride, hydrobromide, hydroiodide, sulphate, phosphate, diphosphate and nitrate or of organic acids such as acetate, maleate, fumarate, tartrate, succinate, citrate, lactate, methanesulphonate, p-toluenesulphonate, palmoate and stearate.
  • inorganic acids such as hydrochloride, hydrobromide, hydroiodide, sulphate, phosphate, diphosphate and nitrate
  • organic acids such as acetate, maleate, fumarate, tartrate, succinate, citrate, lactate, methanesulphonate, p-toluenesulphonate, palmoate and stearate.
  • bases such as sodium or potassium hydroxide.
  • Salt selection 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, gelatin capsules, liposomes or suppositories.
  • Appropriate solid supports can be, for example, calcium phosphate, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, 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, emulsions, 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 ( ⁇ ) for medical uses according to the invention may be selected from the group consisting of:
  • Exemplar per se compounds of the invention according to general formula ( ⁇ ), and/or exemplar compounds according to general formula ( ⁇ ) and formula (P) for medical uses according to the invention may be selected from the group consisting of:
  • the compound (5)-4-Fluoro-N-(2-oxopiperidin-3-yl)benzamide is also known as (S)- 3-(4'-fluorobenzoylamino)-tetrahydropyridin-2-one ( see Example 3 below).
  • Exemplar compounds according to general formula (I) for medical uses according to the invention may be selected from the group consisting of:
  • Exemplar per se compounds of the invention according to general formula (I), and/or exemplar 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 salt thereof.
  • inflammatory disorders (which term is used herein interchangeably with “inflammatory disease”) intended to be prevented or treated by the compounds of formula (I) or (P), or pharmaceutically acceptable salts thereof or
  • compositions or medicaments containing them as active ingredients include notably: autoimmune diseases, for example such as multiple sclerosis, rheumatoid arthritis, lupus, irritable bowel syndrome, Crohn's disease; - vascular disorders including stroke, coronary artery diseases, myocardial
  • infarction unstable angina pectoris, atherosclerosis or vasculitis, e. g., Behcet's syndrome, giant cell arteritis, polymyalgia rheumatica, Wegener's granulomatosis, Churg-Strauss syndrome vasculitis, Henoch-Schonlein purpura and Kawasaki disease; - asthma, and related respiratory disorders such as allergic rhinitis and COPD; organ transplant rejection and/or delayed graft or organ function, e.g. in renal transplant patients;
  • fibrosis skin wounds and other fibrotic disorders including hypertrophic scarring (keloid formation), adhesion formations following general or gynaecological surgery, lung fibrosis, liver fibrosis (including alcoholic liver disease) or kidney fibrosis, whether idiopathic or as a consequence of an underlying disease such as diabetes (diabetic nephropathy); or
  • the inflammatory disorder may be selected from the group consisting of autoimmune diseases, asthma, rheumatoid arthritis, a disorder characterised by an elevated TNF-a level, psoriasis, allergies, multiple sclerosis, fibrosis (including diabetic nephropathy), and formation of adhesions.
  • the term inflammatory disorder may exclude cognitive disorders such as Alzheimer's disease and/or memory loss.
  • Compounds of formula (I) or (F) are particularly useful for local delivery, and also for the preparation of medicaments for local delivery, including creams and ointments for topical delivery, powders, aerosols or emulsions for inhaled delivery, and solutions or emulsions for injection.
  • Pharmaceutical compositions containing one or more excipients suitable for such local delivery 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 medicament as defined herein.
  • Administration of a compound, composition or medicament according to the invention can be carried out by topical, oral, parenteral route, by intramuscular injection, etc.
  • the administration dose envisaged for a compound, composition or medicament according to the invention is comprised between 0.1 mg and 10 g depending on the formulation and route of administration used.
  • the invention further encompasses a library consisting of elements all of which have structures according to the formula (I) or ( ⁇ ), and hence which all have 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 defined, wherein the compound is in hydrated or solvated form.
  • compounds of the invention include tautomers, resolved enantiomers, resolved diastereomers, racemic mixtures, solvates, metabolites, salts and prodrugs thereof, including pharmaceutically acceptable salts and prodrugs.
  • n may be 2.
  • n may be 3.
  • 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 ( ⁇ ) where X is halogen or haloakyl and where k is between 1 and 3.
  • X may be fluoro or fluoroalkyl (such as trifluoromethyl) and k may be between 1 and 3.
  • 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- trifluoromethyl [i.e. 4'-trifluoromethyl], ortho-trifluoromethyl [i.e. 2'-triflouromethyl] or meta-trifluoromethyl [i.e. 3 '-trifluoromethyl]).
  • the benzyl ring may be
  • the benzyl ring may be monosubstituted with halogen.
  • the benzyl ring may be monosubstituted with ortho- carboxy [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.
  • the benzyl ring may be monosubstituted with an alkyl group other than a Ci -6 alkyl.
  • the compounds of general formula (I) or (F) can be prepared using the processes described hereafter.
  • compositions comprising as active ingredient a compound, this terminology is intended to cover both compositions in which other active ingredients may be present and also compositions which consist only of one active ingredient as defined.
  • 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.
  • alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), 1 -propyl (n-Pr, n-propyl, -CH 2 CH 2 CH 3 ), 2-propyl (i-Pr, i-propyl, -CH(CH 3 ) 2 ), 1 -butyl (n-Bu, n- butyl, -CH 2 CH 2 CH 2 CH 3 ), 2- methyl-1 -propyl (i-Bu, i-butyl, -CH 2 CH(CH 3 ) 2 ), 2-butyl (s- Bu, s-butyl, - CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CH 3 ) 3 ), 1-pentyl (n- pentyl, - CH 2 CH 2 CH 2 CH 3 ), 2-pentyl (n
  • haloalkyl or “haloalkyl group” as used herein refers to an alkyl group (as defined above) except that one or more or all of the hydrogens of the alkyl group is replaced by a halogen, which replacement can be at any site on the alkyl, including the
  • P668 5 4.WO01.Spec as filed 8.06.1 1 end.
  • Examples include, but are not limited to, CH 2 F, CHF 2 , CF 3 , CH 2 CH 2 F 5 CH 2 CHF 2 , CH 2 CF 3 , CHFCF 3 , CF 2 CF 3 , CH 2 C1, CHC1 2 , CC1 3 , CH 2 CH 2 C1, CH 2 CHC1 2 , CH 2 CCI 3 , CHC1CC1 3 , and CC1 2 CC1 3 .
  • halogen (which may be abbreviated to “halo") or "halogen group” as used herein includes fluorine (F), bromine (Br), chlorine (CI), and iodine (I).
  • hydroxyalkyl or "hydroxyalkyl group” as used herein refers to an alkyl group (as defined above) except wherein one or more or all of the hydrogens of the alkyl group is replaced by an hydroxy group, which replacement can be at any site on the alkyl, including the end.
  • alkoxy or "alkoxy group” denotes an alkyl group as defined above attached via a divalent oxygen atom to the rest of the molecule. Examples include but are not limited to methoxy (-OCH 3 ), ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert- butoxy, pentoxy, isopentoxy, neopentoxy, hexoxy, and 3-methylpentoxy.
  • amino or “amino group” denotes the group "-NH 2 ".
  • 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.
  • aminodialkyl or “aminodialkyl group” refers to an amino group in which both of the hydrogen atoms have been replaced by an alkyl group as defined above.
  • the alkyl groups attached to the nitrogen atom may be different or the same.
  • benzyl ring (also known as a "phenyl group”) refers to a 6 carbon aryl group in compounds of general formulae (I) or ( ⁇ ) shown above.
  • numbering to locate the carbon atoms C 2 -C 6 within the benzyl ring is in a clockwise direction from Ci which is linked to the
  • 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.
  • a suitably activated acid such as an acid chloride
  • 3-aminolactams with 5,6,7 and 8 membered rings encompassing all the compounds claimed herein, have been extensively described in the literature.
  • the 3-aminolactam product is reacted with an appropriate acid chloride, for example as previously described for 7-ring aminolactams (Fox et al, 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 Na 2 C0 3 .
  • a base for example Na 2 C0 3 .
  • the above reaction may be carried out at ambient temperature (about 25 °C) or more generally at a temperature between 20 and 50 °C.
  • the two reactions may be carried out independently, with separation and purification of the 3-aminolactam between the reactions, or alternatively, the reactions may be performed in a single vessel without purification of the 3-aminolactam prior to its derivatisation with acid chloride.
  • Fig. 1 shows the chemical structure of various examples of compounds according to the inventions and reference examples.
  • Fig. 2 is a graph showing the results of a murine sub-lethal endotoxemia test.
  • column A shows data from a control group (1% CMC lOml/kg p.o.), and
  • 3-aminotetrahydropyridin-2-one hydrochloride (10 mmol) and K 2 C0 3 (30 mmol) were added to water (20 mL) and stirred.
  • Vmax/cm " ' 3205, 3056 (N-H, amide), 1544 (secondary CONH, lactam), 1599, 1501 (aromatic ring), 821, 858 (para-disubstituted benzene ring), 1 164, 1222, 1291 , 765, 696 (C-F).
  • 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 catalytic 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, 3- aminoazepan-2-one hydrochloride (10 mmol) and 2 C0 3 (30 mmol) were added to water (30 mL) and stirred, giving a solution to which the acid chloride solution was added.
  • the product was purified by silica column chromatography (petroleum ethenethyl acetate:methanol 50:50:0 to 0:80:20) to give the product as a white solid 1.10 g (40 %); mp 1 16-1 17 0 C; ⁇ ⁇ (400 MHz, CDCI3) 7.72 (d, 2H, J 8, CH-C- n Bu), 7.25 (d, 1 ⁇ , J
  • 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 Na 2 S0 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 0.95 g (63 %); mp 1 18-119 0 C;
  • the product was purified by silica column chromatography (petroleum ethenethyl acetate 50:50:0 to 0:80:20) to give the product as a white solid 0.16 g (25 %); mp 122-123 ° C; ⁇ ⁇ (400 MHz, CDC1 3 ) 7.71 (d, 2H, J 8.5, C f-C-Oct), 7.23 (d, 1H, J 6.5, N//-CH), 7.20 (d, 2H, J 8.5, CH-C-CO), 6.34 (br.s, 1H, NH-C1), 4.41 (dt, 1H, J 1 1.5, 5.5, CH-C3), 3.37-3.33 (m, 2 ⁇ , HI ), 2.68 (ddt, 1 H, J 13, 5.5, 4.5, H3 equatorial), 2.61 (t, 2H, J 7.5, H4), 1.98-1.91 (m, 2H, H2), 1.67-1.55
  • This assay can be used with a range of different leukocyte populations.
  • 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 separations.
  • immortal cell lines which have been extensively validated as models of human peripheral blood leukocytes may be used, including, but not limited to THP-1 cells as a model of monocytes or Jurkat cells as model of naive T cells.
  • the transwell migration systems are manufactured by Neuroprobe, Gaithersburg, MD, USA.
  • the plates used are ChemoTx plates (Neuroprobe 101-8) and 30 ⁇ clear plates (Neuroprobe MP30).
  • Geys' Balanced Salt Solution is purchased from Sigma (Sigma G-9779).
  • Fatty acid-free BSA is purchased from Sigma (Sigma A-8806).
  • RPMI-1640 without phenol red is purchased from Sigma (Sigma R-8755).
  • the THP-1 cell line (European Cell culture Collection) were used as the leukocyte cell population.
  • the cell suspension to be placed in the upper compartment is prepared.
  • the THP- 1 cells are pelleted by centrifugation (770 x g; 4 mins) and washed with Geys
  • GBSS + BSA Balanced Salt Solution with Img/ml BSA
  • the volume of GBSS + BSA is then adjusted depending on the number of cells present so that the cells are at final density of 4.45 x 10 6 cells per ml of GBSS + BSA. This ensures that there are 100,000 THP-1 cells in each 25 ⁇ 1 of the solution that will be placed in the upper chamber of the plate.
  • THP-1 cells The suspension of THP-1 cells at 4.45 x 10 6 cells/ml is divided into two pots. To one pot the inhibitor under test is added at an appropriate final concentration, in an appropriate vehicle (for example at 1 ⁇ in not 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.
  • an appropriate vehicle for example at 1 ⁇ in not more than 1 % DMSO.
  • MCP-1 is diluted in GBSS + BSA to give a final concentration of 25 ng/ml. This is divided 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
  • the migration chamber should be assembled. Place 29 ⁇ of the appropriate chemoattractant solution into the lower well of the chamber. Assays should be performed with at least triplicate determinations of each condition. Once all the lower chambers have been filled, apply the prous membrane to the chamber in accordance with the manufacturer's instructions. Finally, apply 25 ⁇ of the appropriate cell solution to each upper chamber. A plastic lid is placed over the entire apparatus to prevent evaporation. The assembled chamber is incubated at 37 °C, 5% C0 2 , 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 have migrated to the lower chamber under each condition.
  • the liquid cell suspension is gently removed from the upper chamber, and 20 ⁇ 1 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.
  • the filter is carefully flushed with GBSS + BSA to wash off the EDTA, and then the filter is 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.
  • 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 ⁇ , with 3 ⁇ of MTT stock solution added. The standard curve plate is incubated along side the migration plate.
  • the liquid is carefully removed from the lower chambers, taking care not to disturb the precipitated formazan product.
  • 20 ⁇ 1 of DMSO is added to each lower chamber to solubilise the blue dye, and absorbance at 595nm is determined using a 96-well 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 present at 25ng/ml.
  • the impact of the test substance is calculated by comparing the MCP-1 -induced migration which occurred in the presence or absence of various concentrations of the test substance.
  • 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.
  • a dose-response graph is constructed by determining the inhibition of MCP-1 induced migration which occurs at a range of different compound concentrations (typically ranging from InM to 1 ⁇ or higher in the case of poorly active compounds). The inhibitory activity of each compound is then expressed as the concentration of compound required to reduce the MCP-1 -induced migration by 50% (the ED 50 concentration).
  • the compounds of reference examples 1 to 14 were tested and were shown to have an ED50 of 100 nM or less in this test.
  • mice Female CD1 mice (28-30g, ⁇ 7 weeks of age) were dosed with their respective treatment in sterile filtered 1% CMC by oral gavage in a dose volume of lOml kg one hour prior to an endotoxin (LPS) challenge.
  • LPS endotoxin
  • the endotoxin challenge was injected by the intraperitoneal route containing 675,000 Endotoxin Units of LPS (E. coli strain 01 1 1 :B4 (Code L4130)) in endotoxin free PBS. Mice were left for two hours and then exsanguinated under terminal anaesthesia and blood was taken. Serum was prepared from this terminal bleed and aliquoted and stored at -20°C.
  • Serum TNF-a levels were measured by ELISA per manufacturers instructions (R and D Systems). Eight animals 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 animals. Data for untreated animals were taken from an historical control experiment.

Abstract

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

Description

Anti-inflammatory agents
The invention relates to aryl substituted 3-aminolactam derivatives and their use in preventing or treating inflammatory diseases.
Inflammation is an important component of physiological 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 have not traditionally been considered to involve leukocytes (such as osteoporosis or Alzheimer's disease). The chemokines are a large family of signalling molecules with homology to interleukin-8 which have been implicated in regulating leukocyte trafficking both in physiological 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 modulating inflammatory responses through chemokine receptor blockade. It has proved difficult to determine which chemokine receptor(s) should be inhibited to produce therapeutic benefit in a given inflammatory 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: 803-81 1). 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-DIle- DT -DLys-DGln-DLys-DPro-DAsρ-DLeu-DCys)-NH2 [SEQ ID NO: 1]), are collectively termed "Broad Spectrum Chemokine Inhibitors" (BSCIs). Grainger et al. (2003, Biochem. Pharm. 65: 1027-1034) have subsequently shown BSCIs to have potentially useful anti-inflammatory activity in a range of animal models of diseases.
Interestingly, simultaneous blockade of multiple chemokines is not apparently
1
P66854. WOO L Spec as Hied 8.06.1 1 associated with acute or chronic toxicity, suggesting this approach may be a useful strategy for developing new anti-inflammatory medications with similar benefits to steroids but with reduced side-effects. This beneficial 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 derivatives such as NR58-3.14.3, may not be optimal for use in vivo. They are quite expensive to synthesise and have relatively
unfavourable pharmacokinetic and pharmacodynamic properties. For example, NR58- 3.14.3 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 have been adopted to identify novel preparations that retain the anti-inflammatory properties of peptide 3 and NR58-3.14.3, but have improved characteristics for use as pharmaceuticals. Firstly, a series of peptide analogs have been developed, some of which have longer plasma half-lives than NR58-3.14.3 and which are considerably cheaper to synthesise (see for example WO2009/017620). 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 beneficial properties of the original peptide.
This second approach yielded several structurally distinct series of compounds that retained the anti-inflammatory properties of the peptides, including 16-amino and 16- aminoalkyl derivatives of the alkaloid yohimbine, as well as a range of N-substituted 3-aminoglutarimides, identified from a small combinatorial library (see Fox et ai, 2002, J Med Chem 45: 360-370; WO 99/12968 and WO 00/42071 ). All of these compounds are broad-spectrum chemokine inhibitors that retain selectivity over non- chemokine chemoattractants, and a number of them have been shown to block acute inflammation in vivo. The most potent and selective of the above-mentioned aminoglutarimides was (S)-3-
(undec- 10-enoyl)-aminoglutarimide (NR58,4), which inhibited chemokine-induced
2
P66854.WO01 .Spec as filed 8.06.1 1 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 result, subsequent studies of
aminoglutarimide and aminolactam BSCIs have focussed almost exclusively on compounds with simple linear and branched alkyl side chains.
However, further studies revealed that the aminoglutarimide ring was susceptible to enzymatic ring opening in serum. Consequently, for some applications (for example, where the inflammation under treatment is chronic, such as in autoimmune diseases) these compounds may not have optimal properties, and a more stable compound with similar anti-inflammatory properties may be superior.
As an approach to identifying such stable analogs, various derivatives of (S)- 3- (undec-lO-enoyl)-aminoglutarimide have been tested for their stability in serum. One derivative, the 6-deoxo analog (S)-3-(undec-10-enoyl)-tetrahydropyridin-2-one, is completely stable in human serum for at least 7 days at 37°C, but has considerably reduced potency compared with the parental molecule.
One such family of stable, broad spectrum chemokine inhibitors (BSCIs) are the 3- amino caprolactams, with a seven-membered monolactam ring (see, for example, WO2005/053702 and WO2006/016152). However, further useful anti-inflammatory compounds have also been generated from other 3-aminolactams with different ring size (see for example WO2006/134385). Other modifications to the lactam ring, including introduction of heteroatoms and bicyclolactam ring systems, also yield compounds with BSCI activity (see, for example, WO2006/018609 and
WO2006/085096).
In general, these earlier studies have demonstrated that the BSCI activity is conferred on the molecule 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 have little impact). To be active as a BSCI, this "head group" must have 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. Previous
3
P66854. WOOL Spec as filed 8.06.1 1 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 critical for BSCI activity, it has been shown that a wide variety of "tail groups" can be selected with out affecting the primary pharmacology of the compound, at least in vitro. As a result, 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 excretion, modify the
pharmacokinetics or oral bioavailability, and thus act as the primary determinant 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 10-fold increase in potency as a BSCI, at least in vitro, compared to a related compound lacking 2'2'-disubstitution. For example, 2'2'- dimethyldodecenanoyl-3-aminocaprolactam is 10-fold more potent as a BSCI in the MCP-1 induced THP-1 cell migration than assay than dodecanoyl-3- aminocaprolactam (as disclosed previously in WO2005/053702), 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 molecule (as disclosed previously in WO2009/016390). In particular, the pivoyl group is resistant to
4
P66854. WOO 1. Spec as filed 8.06.1 1 metabolism, and therefore contributes to the unusually prolonged biological half-life of this compound.
In marked contrast, other possible "tail groups" have 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) have 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 substantially less active (Fox et al, 2002, J Med Chem 45: 360-370). Taken together, these two findings have 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 result, previous disclosures of compounds with BSCI activity have all excluded such aromatic "tail groups".
The present invention discloses a series 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, all of the compounds as set out below have substantial BSCI activity (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 salt thereof, for in the treatment of an inflammatory disorder:
Figure imgf000006_0001
(I)
P66854.WO01.Spec as filed 8.06.1 1 wherein 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 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 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; 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.
The carbon atom at position 3 of the lactam ring is asymmetric and consequently, the compounds according to the present invention have at least two possible enantiomeric forms, that is, the "R" and "S" configurations. The present invention encompasses each of the two enantiomeric forms and all combinations of these forms, including the racemic "RS" mixtures. With a view to simplicity, when no specific 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 salt thereof, for use in the treatment of an inflammatory disorder:
6
P66854.WO01.Spec as filed 8.06.1 1
Figure imgf000008_0001
d') wherein n, k and X are defined as for general formula (I) compounds above.
Compounds (Ι'), having the (S)-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 salt thereof, in the manufacture of a medicament for the treatment of an inflammatory disorder:
Figure imgf000008_0002
(I) wherein 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 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:
7
P66854.WO01 .Spec as filed 8.06.1 1 when 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; 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 (Γ), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of an inflammatory disorder:
Figure imgf000009_0001
(Γ) wherein n, k and X are defined as for general formula (I) above. Certain compounds have been found to be novel per se. Thus, in another aspect of the invention, there is provided a compound of general formula (I):
Figure imgf000009_0002
(I) wherein
8
P66854. WOO L Spec as filed 8.06.1 1 n is an integer from 1 to 4; k is an integer from 1 to 5, representing the number of groups substituting C2, C3, C4, C5 and/or C6 of the benzyl ring; when n is 1 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 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; 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 C 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 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; 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'-dimethoxybenzoylamino)- caprolactam, 3-(3',5'-di-iert-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, 3-(2'-
9
P66854. WOO 1. Spec as filed 8.06.1 1 fluoromethylbenzoylamino)-caprolactam, 3 -(2 '-amino-3 ' -hydroxy-4'- methylbenzoylamino)-caprolactam, and 3-(3 ' ,5 ' -dimethylbenzoylamino)-caprolactam.
Also provided is a compound of formula (P):
Figure imgf000011_0001
wherein n, k and X are defined herein for general formula (I), provided that the compound is none of the group consisting of: (S)-3-(3'-trifluoro- methylbenzoylamino)-caprolactam, (S)-3-(4' -methylbenzoylamino)-caprolactam, (S)- 3-(4'-methoxybenzoylamino)-caprolactam, (S)-3-(2'-carboxybenzoylamino)- caprolactam, and (S)-3-(3', 4,,5'-trimethoxybenzoylamino)-caprolactam.
WO2007/0038669 teaches diarylamine-containing compounds and their use as moduclators of c-kit receptors. Various intermediate compounds are used in the synthesis of the diarylamine-containing compounds. Any overlap of the intermediate compounds is hereby disclaimed from the present invention. EP0462949 together with the related publication Angelucci et al, 1993, J Medicinal Chemistry 36: 1512-1519 teach 7-membered 3-acylamino lactams as enhancers of learning and memory. Any overlap of specific 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 (Γ) according to the present invention) mentioned in these documents is hereby disclaimed from the present invention.
JP03206042 discloses the preparation of 5,6,7,8-tetrahydo-4H-thiazolo[5,4-b]azepine derivatives with potassium channel activation activity, for use as antihypertensives.
10
P66854.WO0l .Spec as filed 8.06.1 1 Various intermediate compounds (notably where n=3 in compounds of generic formulae (I) and/or (Γ) according to the present invention) are used in the synthesis of the derivatives. Any overlap of the intermediate compounds is hereby disclaimed from the present invention. The prior art also discloses specific compounds, for example:
3-(4'-methylbenzoylamino)-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'-trimethoxybenzoylamino)-caprolactam are disclosed in EP462949A1 ; 3-(3'-trifluoromethylbenzoylamino)-caprolactam is also disclosed in EP351856A2;
- 3-(3',4'-dimethoxybenzoylamino)-caprolactam, 3-(3',5'-di-tert-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.
07.167.701 ; CHEMCATS Acc. NO. 001 5557046).
1 1
P66854.WO01.Spec as filed 8.06.1 1 However, none of the above prior art compounds have been shown to have BSCI activity, or to be useful for the treatment of inflammatory diseases. As a result, compounds disclosed in the prior art documents mentioned herein in no way teach or suggest our unexpected finding that the class of aryl-substituted aminolactams and analogs as defined herein have 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 ingredient, a compound per se as defined above, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient and/or carrier.
By pharmaceutically acceptable salt is meant in particular the addition salts of inorganic acids such as hydrochloride, hydrobromide, hydroiodide, sulphate, phosphate, diphosphate and nitrate or of organic acids such as acetate, maleate, fumarate, tartrate, succinate, citrate, lactate, methanesulphonate, p-toluenesulphonate, palmoate and stearate. Also within the scope of the present 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 "Salt selection 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, gelatin capsules, liposomes or suppositories. Appropriate solid supports can be, for example, calcium phosphate, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, 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, emulsions, 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 (Γ) for medical uses according to the invention may be selected from the group consisting of:
12
P668S4.WO01 .Spec as filed 8.06.1 1 (S)-3 -(4'-methylbenzoylamino)-caprolactam, and
(S)-3-(3',5'-dimethylbenzoylamino)-caprolactam, and pharmaceutically acceptable salts thereof.
Exemplar per se compounds of the invention according to general formula (Γ), and/or exemplar compounds according to general formula (Γ) and formula (P) for medical uses according to the invention, may be selected from the group consisting of:
(5)-3-Fluoro-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)benzamide,
( )-N-(2-Oxopiperidin-3-yl)-2-(trifluoromethyl)benzamide,
(S)-2,3-difluoro-N-(2-oxopiperidin-3-yl)benzamide,
(5}-2,4-difluoro-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'-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-oxopiperidin-3-yl)benzamide is also known as (S)- 3-(4'-fluorobenzoylamino)-tetrahydropyridin-2-one ( see Example 3 below).
13
P66854. WOO L Spec as filed 8.06.1 1 Exemplar per se compounds of the invention according to general formula (I) or (P), and/or 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'-Ethylbenzoylamino)-azepan-2-one,
(S)-3-(4'-Butylbenzoylamino)-azepan-2-one,
(S)-3-(4'-tert-Butylbenzoylamino)-azepan-2-one,
(S)-3-(4'-Hexylbenzoylamino)-azepan-2-one,
(S)-3-(4'-Octylbenzoylamino)-azepan-2-one, and
(S)-3-(4'-Octylbenzoylamino)-tetrahydropyridin-2-one, and pharmaceutically acceptable salts thereof.
Exemplar compounds according to general formula (I) for medical uses according to the invention, may be selected from the group consisting of:
(R)-3-(4'-Butylbenzoylamino)-tetrahydropyridin-2-one,
(R)-3-(4'-tert-Butylbenzoylamino)-tetrahydropyridin-2-one, and
(R)-3-(4'-Hexylbenzoylamino)-tetrahydropyridin-2-one, and pharmaceutically acceptable salts thereof.
Exemplar per se compounds of the invention according to general formula (I), and/or exemplar 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 salt thereof.
The compounds (R)-3-(4'-Butylbenzoylamino)-tetrahydropyridin-2-one,
(R)-3-(4'-½rt-Butylbenzoylamino)-tetrahydropyridin-2-one, (R)-3-(4'-Hexylbenzoyl- amino)-tetrahydropyridin-2-one, and (R)-3-(4'-Octylbenzoylamino)-tetrahydropyridin- 2-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 (P), or pharmaceutically acceptable salts thereof or
14
P66854.WO01 .Spec as filed 8.06.1 1 pharmaceutical compositions or medicaments containing them as active ingredients, include notably: autoimmune diseases, for example such as multiple sclerosis, rheumatoid arthritis, lupus, irritable bowel syndrome, Crohn's disease; - vascular disorders including stroke, coronary artery diseases, myocardial
infarction, unstable angina pectoris, atherosclerosis or vasculitis, e. g., Behcet's syndrome, giant cell arteritis, polymyalgia rheumatica, Wegener's granulomatosis, Churg-Strauss syndrome vasculitis, Henoch-Schonlein purpura and Kawasaki disease; - asthma, and related respiratory disorders such as allergic rhinitis and COPD; organ transplant rejection and/or delayed graft or organ function, e.g. in renal transplant patients;
- psoriasis; skin wounds and other fibrotic disorders including hypertrophic scarring (keloid formation), adhesion formations following general or gynaecological surgery, lung fibrosis, liver fibrosis (including alcoholic liver disease) or kidney fibrosis, whether idiopathic or as a consequence of an underlying disease such as diabetes (diabetic nephropathy); or
- allergies. The inflammatory disorder may be selected from the group consisting of autoimmune diseases, asthma, rheumatoid arthritis, a disorder characterised by an elevated TNF-a level, psoriasis, allergies, multiple sclerosis, fibrosis (including diabetic nephropathy), and formation of adhesions.
The above clinical indications fall under the general definition of inflammatory disorders or disorders characterized by elevated TNFoc levels.
15
P66854. WOO 1 Spec as filed 8.06.1 1 In one aspect of the invention, merely in order to circumvent any potentially conflicting prior art (for example as noted above), the term inflammatory disorder may exclude cognitive disorders such as Alzheimer's disease and/or memory loss.
Compounds of formula (I) or (F) are particularly useful for local delivery, and also for the preparation of medicaments for local delivery, including creams and ointments for topical delivery, powders, aerosols or emulsions for inhaled delivery, and solutions or emulsions for injection. Pharmaceutical compositions containing one or more excipients suitable for such local delivery 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 medicament as defined herein. Administration of a compound, composition or medicament according to the invention can be carried out by topical, oral, parenteral route, by intramuscular injection, etc.
The administration dose envisaged for a compound, composition or medicament according to the invention is comprised between 0.1 mg and 10 g depending on the formulation and route of administration used. The invention further encompasses a library consisting of elements all of which have structures according to the formula (I) or (Γ), and hence which all have 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 defined, wherein the compound is in hydrated or solvated form. Unless specified otherwise, compounds of the invention include tautomers, resolved enantiomers, resolved diastereomers, racemic mixtures, solvates, metabolites, salts and prodrugs thereof, including pharmaceutically acceptable salts and prodrugs.
In any of the compounds described above, n may be 2. Alternatively, n may be 3.
16
P66854.WO01 Spec as filed 8.06.1 1 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 (Γ) 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 defined 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- trifluoromethyl [i.e. 4'-trifluoromethyl], ortho-trifluoromethyl [i.e. 2'-triflouromethyl] or meta-trifluoromethyl [i.e. 3 '-trifluoromethyl]). The benzyl ring may be
monosubstituted with an haloalkyl other than a Ci-6 haloalkyl. The benzyl ring may be monosubstituted with halogen. The benzyl ring may be monosubstituted with ortho- carboxy [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 = 1). For example, the benzyl ring may be monosubstituted with an alkyl group other than a Ci-6 alkyl.
According to the invention, the compounds of general formula (I) or (F) 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 from X minus 5% of X to X plus 5% of X.
The use of a numerical range in this description is intended unambiguously to include within the scope of the invention all individual integers within the range and all the
17
P66854.WO01.Spec as filed 8.06.1 1 combinations of upper and lower limit numbers within the broadest scope of the given range. Hence, for example, the range of 0. lmg to 1 Og specified in respect of (inter alia) a dose of a compound or composition of the invention to be used is intended to include all doses between O.lmg and lOg and all 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 meaning or encompassing both comprising and consisting of. Consequently, where the invention relates to a
"pharmaceutical composition comprising as active ingredient" a compound, this terminology is intended to cover both compositions in which other active ingredients may be present and also compositions which consist only of one active ingredient 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), 1 -propyl (n-Pr, n-propyl, -CH2CH2CH3), 2-propyl (i-Pr, i-propyl, -CH(CH3)2), 1 -butyl (n-Bu, n- butyl, -CH2CH2CH2CH3), 2- methyl-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), 3- methyl-1 -butyl (-CH2CH2CH(CH3)2), 2-methyl-l -butyl (- CH2CH(CH3)CH2CH3), 1 - hexyl (-CH2CH2CH2CH2CH2CH3), 2-hexyl (- CH(CH3)CH2CH2CH2CH3), 3-hexyl (- CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (- C(CHs)2CH2CH2CH3), 3-methyl-2- pentyl (-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- dimethyls- butyl (-CH(CH3)C(CH )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 all of the hydrogens of the alkyl group is replaced by a halogen, which replacement can be at any site on the alkyl, including the
18
P66854.WO01.Spec as filed 8.06.1 1 end. Examples include, but are not limited to, CH2F, CHF2, CF3, CH2CH2F5 CH2CHF2, CH2CF3, CHFCF3, CF2CF3, CH2C1, CHC12, CC13, CH2CH2C1, CH2CHC12, CH2CCI3, CHC1CC13, and CC12CC13.
The term "halogen" (which may be abbreviated to "halo") or "halogen group" as used herein includes fluorine (F), bromine (Br), chlorine (CI), and iodine (I).
The term "hydroxy" or "hydroxy group" denotes 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 all of the hydrogens of the alkyl group is replaced by an hydroxy group, which replacement can be at any site on the alkyl, including the end.
The term "alkoxy" or "alkoxy group" denotes an alkyl group as defined above attached via a divalent oxygen atom to the rest of the molecule. Examples include but are not limited to methoxy (-OCH3), ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert- butoxy, pentoxy, isopentoxy, neopentoxy, hexoxy, and 3-methylpentoxy. The term "amino" or "amino group" denotes 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 have 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" denotes the group "-C(0)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 (Γ) shown above. For the purposes of the general formulae of the present 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 specific compounds follows the IUPAC rule that the second substituent in a clockwise or counter clockwise direction is
19
P66854.WO0 l .Spec as filed 8.06.1 1 afforded the lower possible location number. Where two or more substituents are present in a specific 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 have 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 (P), i.e., where k=0, 1 2, 3 or 4, the or each unsubstiruted position is occupied by a hydrogen atom.
Unless otherwise defined, all the technical and scientific terms used here have 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).
Preparation of the compounds of general formula (I) or (F)
All the compounds of general formula (I) or (F) can be prepared easily according to general methods known to the person skilled in the art.
Typically, 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 preparation of 3-aminolactams with 5,6,7 and 8 membered rings, encompassing all the compounds claimed herein, have been extensively described in the literature. For example, we have provided suitable methods for the preparation of 6-membered aminolactams from ornithine (see WO2009/016390) and 7-membered aminolactams from lysine (see WO2005/053702), as well as methods for 5- and 8- membered aminolactams (see WO2006/134385). We have described in particular detail various synthesis routes to the 6-membered aminolactam, including processes suitable for scaling up the manufacture to Kg quantities (WO2009/016390). Various other methods for the synthesis of 3-aminolactams of various ring sizes have also been described in the literature (see for example Pellegata et al., 1978, Synthesis 614-616
20
P66854 WOO L Spec as filed 8.06.1 1 and Boyle et ai, 1 79, J Org Chem 44:4841-4847), and any suitable method for the preparation of the aminolactam "head group" may be employed in accordance with the method of the present 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 al, 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 Na2C03. The above reaction may be carried out at ambient temperature (about 25 °C) or more generally at a temperature between 20 and 50 °C. The two reactions may be carried out independently, with separation and purification of the 3-aminolactam between the reactions, or alternatively, the reactions may be performed in a single vessel without purification of the 3-aminolactam prior to its derivatisation with acid chloride. As noted previously (see WO2009/016390) 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 racemisation of the 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
21
P668 4.WO01.Spec as filed 8.06.1 1 column B shows data from a group treated with lmg/kg p.o. (5)-4-Fluoro-N-(2- oxopiperidin-3-yl)benzamide (a compound according to one embodiment of the present invention - see also Example 3 below) The y-axis shows levels of TNF-a in pg/ml.
EXAMPLES
In the following further examples, Ή-NMR and I3C-NMR spectra were recorded on a Bruker Avance DRX 400 MHz fourier transform machine and l9F-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. [CC]D 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 K2C03 (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 temperature in an inert atmosphere (using dinitrogen). The reaction was extracted with CH2C12 (3 χ 50 mL), and the combined organic layers where then dried (Na2S04) and reduced in vacuo to give a crude product which was recrystallised from CH2C12 / 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).
Ή NMR: δΗ (400MHz, CDC13) 7.80 (2H, br d, J 7.0, ortho-H), 7.47-7.40 (1 H, m, para-U), 7.42-7.39 (1 H, m, C6H5-CONH), 7.40-7.31 (2H, m, meta-W), 6.78 (1 H, br s, CONH-CH2), 4.41 (1 H, dt, J 1 1.5, 5.5, CH-CO), 3.36-3.23 (2H, m, CH2NH), 2.59 (1 H,
22
P66854. WOO L Spec as Hied 8.06.1 1 dq, J 13.0, 4.5, NHCH-CH;), 1.94-1.81 (2H, m, lactam CH2), 1.64 (IH, tt, J 12.5, 8.0, NHCH-CH^).
13C NMR: 5C (100MHz, CDC13) 171.9 (lactam C=0), 167.4 (aryl C=0), 134.0 (ipso- C), 131.4 (ortho-C), 128.3 (meta-C), 127.0 (para-C), 50.8 ( H-CO), 41.5 (CH2NH), 27.0 (lactam CH2), 20.9 (lactam CH2).
HRMS (+ESI) Ci2H,4N202 + Na+: calcd 241.0947; found 241.0950. Reference Example 2: 3-(Benzoylamino)azepan-2-one: 3-aminoazepan-2-one hydrochloride (10 mmol) and K2C03 (30 mmol) were added to water (20 mL) and stirred. A solution of benzoyl chloride (10 mmol) in CH2CI2 (10 mL) was added and the reaction was stirred overnight at room temperature in an inert atmosphere (using dinitrogen). The reaction was extracted with CH2C12 (3 x 50 mL), and the combined organic layers where then dried (Na2S04) and reduced in vacuo to give a crude product which was recrystallised from 0¾012 / petroleum ether (bp 40- 60 °C) to give the product (1.59 g, 68%): max cm"1 3244, 3202 (N-H, amide), 1660, 1642, 1536 (secondary CONH, lactam), 1601, 1578, 1536 (aromatic ring), 771 , 707 (monosubstituted benzene ring). Ή NMR: 8H (400MHz, CDC13) 7.86-7.80 (2H, m, ortho-H), 7.65 (IH, d, J 4.0, C6H5- CONH), 7.52-7.45 (1Η, m, para-H), 7.46-7.39 (2Η, m, meta-U), 6.1 1 (1Η, br s, CONH-CH2), 4.72 (IH, ddd, J 1 1.0, 5.5, 1.5, CH-CO), 3.49-3.39 (2H, m, CH2NH), 2.25 (I H, br d, J 13.5, lactam CH2), 2.08-1.98 (I H, 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: 6C (100MHz, CDC13) 175.9 (lactam C=0), 166.4 (aryl C=0), 134.3 (ipso- C), 131.7 (ortho-C), 128.7 (meta-C), 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) C|2H,4N202 + H+: calcd 233.1285; found 233.1283.
23
P66854. WOO L Spec as Hied 8.06.1 1 Reference Example 3: 3-(4'-Methylbenzoylamino)tetrahydropyridin-2-one:
3-aminotetrahydropyridin-2-one hydrochloride (10 mmol), K2C03 (30 mmol) and 4- methylbenzoyl chloride (10 mmol) were reacted according to the above procedure to give the product (1.62 g, 78%):
Vmax cm"1 3306, 3203 (N-H, amide), 1669, 1649 (secondary CONH, lactam), 1612, 1489 (aromatic ring), 842, 810 (para-disubstituted benzene ring). Ή NMR: δΗ (400MHz, CDC13) 7.69 (2H, br d, J 8.0, ortho-H), 7.20 (2H, br d, J 8.0, meta-H), 7.13 (I H, br d, J4.0, CONH-CH2), 6.03 (I H, br s, C7H7-CONH), 4.41 (1Η, dt, J 1 1.0, 5.5, CH-CO), 3.41-3.30 (2H, m, CH2NH), 2.72 (IH, 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 (I H, m, lactam CH2).
13C NMR: 6c (100MHz, CDC13) 172.3 (lactam C=0), 167.7 (aryl C=0), 142.1 (ipso- C), 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) Ci3H,6N202 + Na+: calcd 255.1 104; found 255.1 104.
Reference Example 4: 3-(4'-MethylbenzoyIaniino)azepan-2-one :
3-aminoazepan-2-one hydrochloride (10 mmol), 2C03 (30 mmol) and 4- methylbenzoyl chloride (10 mmol) were reacted according to the above procedure to give the product (1.63 g, 74%):
Vmax/cm"1 3265, 3219 (N-H, amide), 1663, 1647 (secondary CONH, lactam), 1607, 1570, 1505 (aromatic ring), 838, 823 (para-di substituted benzene ring).
Ή NMR: δΗ (400MHz, CDC13) 7.72 (2H, br d, J 8.0, ortho-H), 7.58 (I H, br d, J 4.50, CONH-CH2), 7.21 (2H, d, J 8.0, meta-H), 5.98 (I H, br s, C7H7-CONH), 4.69 (1 Η, ddd, J 1 1.0, 5.5, 2.0, CH-CO), 3.40-3.321 (2H, m, CH2NH), 2.37 (3H, s, CH3), 2.23
24
P66854.WO0l .Spec as filed 8.06.1 1 (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).
I3C NMR: 5c (100MHz, CDC13) 176.1 (lactam C=0), 166.3 (aryl C=0), 142.0 (ipso- C), 131.5 (para-C), 129.3 (aromatic-CH), 127.2 (aromatic-CH), 52.6 ( H-CO), 42.2 (CH2-NH), 31.7 (lactam CH2), 29.0 (lactam CH2), 28.1 (lactam CH2), 21.6 (CH3).
HRMS (+ESI) C14H18N202 + H+: calcd 247.1441 ; found 247.1453. Reference Example 5: 3-(4'-Chlorobenzoylamino)tetrahydropyridin-2-one:
3-aminotetrahydropyridin-2-one hydrochloride (10 mmol), K2C03 (30 mmol) and 4- chlorobenzoyl chloride (10 mmol) were reacted according to the above procedure to give the product (0.87 g, 39%): vmax/cm"' 3295, 3202 ( -H, amide), 1668, 1648, 1629 (secondary CONH, lactam), 1594, 1486 (aromatic ring), 859, 845, 808, (para-disubstituted benzene ring), 750, 656 (C-Cl). Ή NMR: δΗ (400MHz, CDC13) 7.74 (2H, br d, J 8.5, ortho- ), 7.38 (2H, br d, J 8.5, meta-H), 7.14 (1 H, br d, J 3.0, C6H4C1-C0NH), 5.83 (1 Η, br s, CONH-CH2), 4.40 (1H, dt, J 1 1.0, 5.5, CH-CO), 3.44-3.33 (2H, m, CH2NH), 2.73 (1 H, dq, J 13.0, 4.5, NHCH-CH2), 2.05-1.93 (2Η, m, lactam CH2), 1.66-1.56 (1 H, m, lactam CH2). 13C NMR: 5C (100MHz, CDC13) 180.2 (lactam C=0), 171.8 (aryl C=0), 138.1 (ipso- C), 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,3ClN202 + Na+: calcd 275.0558; found 275.0559.
Reference Example 6: 3-(4'-Chlorobenzoylamino)azepan-2-one:
25
P66854.WO01 .Spec as filed 8.06.1 1 3-aminoazepan-2-one hydrochloride (10 mmol), 2C03 (30 mmol) and 4- chlorobenzoyl chloride (10 mmol) were reacted according to the above procedure to give the product (1 .79 g, 75%): vmax/cm"' 3243, 3200 (N-H, amide), 1662, 1643 (secondary CONH, lactam), 1595, 1484 (aromatic ring), 856, 841 , 819 (par -disubstituted benzene ring), 776, 732 (C- Cl).
Ή NMR: δΗ (400MHz, CDC13) 7.76 (2H, br d, J 8.5, ortho-H), 7.59 (I H, br d, J 4.0, C6H4C1-C0NH), 7.39 (2Η, br d, J 8.5, meta-H), 6.00 (1Η, br s, CONH-CH2), 4.67 (I H, ddd, J 1 1.0, 5.5, 1.5, CH-CO), 3.39-3.22 (2H, m, CH2NH), 2.22 (I H, br d, J 14.0, lactam CH2), 2.09-2.00 (I H, m, lactam CH2), 1.96-1 .82 (2H, m, lactam CH2), 1 .60- 1.36 (2H, m, lactam CH2). 1 C NMR: 6C (100MHz, CDC13) 175.9 (lactam C=0), 165.3 (aryl C=0), 137.9 (ipso- C), 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) C13H,5C1N202 + H+: calcd 267.0895; found 267.0890.
Reference Example 7: 3-(4'-Methoxybenzoylamino)tetrahydropyridin-2- one:
3-aminotetrahydropyridin-2-one hydrochloride (1 0 mmol), 2C03 (30 mmol) and 4- methoxybenzoyl chloride (10 mmol) were reacted according to the above procedure to give the product (2.18 g, 98%): vmax/cm"' 3305, 3212 (N-H, amide), 2854 (0-CH3), 1693, 1627 (secondary CONH, lactam), 1605, 1576, 1505 (aromatic ring), 837 (pana-disubstituted benzene ring).
Ή NMR: δΗ (400MHz, CDC13) 7.76 (2H, br d, J 9.0, ortho-H), 7.19 (I H, br d, J 5.0, C7H7O-CONH), 6.87 (2H, br d, J 9.0, meta-H), 6.31 (I H, br s, CONH-CH2), 4.39 (I H, dt, J 1 1.5, 5.5, CH-CO), 3.81 (3H, s, OCH3), 3.39-3.28 (2H, m, CH2NH), 2.64 (I H, dq,
26
P66854.WO01.Spec as filed 8.06.1 1 J 13.0, 4.5, NHCH-CH2), 2.01 -1 .88 (2Η, m, lactam CH2), 1.68-1.55 (I H, m, lactam CH2).
I3C NMR: 8c (100MHz, CDC13) 172.4 (lactam C=0), 167.3 (aryl C=0), 162.4 (C- OCH3), 129.1 (aromatic-CH), 126.6 (ipso-C), 1 13.8 (aromatic-CH), 55.6 (OCH3), 51.1 (CH-CO), 41.9 (CH2-NH), 27.4 (lactam CH2), 21.3 (lactam CH2).
HRMS (+ESI) C13H16N203 + Na+: calcd 271.1053; found 271.1057. Reference Example 8: 3-(4'-Methoxybenzoylamino)azepan-2-one:
3-aminoazepan-2-one hydrochloride (10 mmol), 2C03 (30 mmol) and 4- methoxybenzoyl chloride (10 mmol) were reacted according to the above procedure to give the product (1.54 g, 65%):
Vmax cm"1 3270, 3205 (N-H, amide), 2839 (0-CH3), 1642 (secondary CONH, lactam), 1602, 1577, 1504 (aromatic ring), 854, 822 (para-disubstituted benzene ring).
Ή NMR: δΗ (400MHz, CDC13) 7.79 (2H, br d, J9.0, ortho-H), 7.52 (IH, br d, J 5.0, C7H70-CONH), 6.91 (2H, br d, J 9.0, meta-H), 5.94 (IH, br s, CONH-CH2), 4.69 (I H, ddd, J 1 1.0, 5.5, 1.5, CH-CO), 3.83 (3H, s, OCH3), 3.40-3.21 (2H, m, CH2NH), 2.22 (I H, br d, J 12.5, lactam CH2), 2.08-1.97 (I H, m, lactam CH2), 1.95-1.82 (2H, m, lactam CH2), 1.60-1.36 (2H, m, lactam CH2). 13C NMR: 5C (100MHz, CDC13) 176.2 (lactam C=0), 165.9 (aryl C=0), 162.1 (C- OCH3), 129.1 (aromatic-CH), 126.7 (ipso-C), 1 13.8 (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) C14Hi8N203 + Na+: calcd 285.1210; found 285.1215.
Reference Example 9: 3-(4'-Fluorobenzoylamino)tetrahydropyridin-2-one:
27
P668S4.WO01 Spec as filed 8.06.1 1 3-aminotetrahydropyridin-2-one hydrochloride (10 mmol), K2C03 (30 mmol) and 4- fluorobenzoyl chloride (10 mmol) were reacted according to the above procedure (except that CHC13 was used instead of CH2C12) to give the product (1.41 g, 54%): vmax/cm"' 3216, 3075 (N-H, amide), 1650, 1555 (secondary CONH, lactam), 1595, 1491 (aromatic ring), 809, 844 (para-disubstituted benzene ring), 1 105, 1 158, 1226, 1328, 756 (C-F).
Ή NMR: 6H (400MHz, CDC13) 7.76 (2H, br dd, J 9.0, 5.5, ortho-H), 7.21 (IH, br s, C6H4F-CONH), 7.02 (2Η, br t, J 8.5, meta- ), 6.08 (1Η, br s, CONH-CH2), 4.35 (IH, dt, J 1 1.5, 5.5, CH-CO), 3.40-3.26 (2H, m, CH2NH), 2.62 (IH, dq, J 13.0, 4.5, NHCH- CH2), 2.00-1.86 (2H, m, lactam CH2), 1.66-1.52 (I H, m, lactam CH2).
13C NMR: 5c (100MHz, CDC13) 172.2 (lactam C=0), 166.6 (aryl C=0), 164.9 (d, J 252.0, C-F), 130.4 (d, J3.0, ipso-C), 129.7 (d, J9.0, ortho-C), 1 15.6 (d, J 22.0, meta- C), 51.1 (CH-CO), 41.9 (CH2-NH), 27.3 (lactam CH2), 21.3 (lactam CH2).
HRMS (+ESI) Ci2Hi3FN202 + H+: calcd 237.1034; found 237.1034. Reference Example 10: 3-(4'-FIuorobenzoylamino)azepan-2-one:
3-aminoazepan-2-one hydrochloride (10 mmol), 2C03 (30 mmol) and 4- fluorobenzoyl chloride (10 mmol) were reacted according to the above procedure (except that CHC13 was used instead of CH2CI2) to give the product (0.86 g, 45%):
Vmax/cm"' 3205, 3056 (N-H, amide), 1544 (secondary CONH, lactam), 1599, 1501 (aromatic ring), 821, 858 (para-disubstituted benzene ring), 1 164, 1222, 1291 , 765, 696 (C-F). Ή NMR: δΗ (400MHz, CDC13) 7.84 (2H, br dd, J 9.0, 5.5, ortho-H), 7.57 (I H, br s, C6H4F-CONH), 7.09 (2H, br t, J 8.5, meta-H), 5.94 (IH, br s, CONH-CH2), 4.67 (1 H, ddd, J 1 1.5, 5.5, 1.5, CH-CO), 3.40-3.22 (2H, m, CH2NH), 2.26-2.19 (I H, m, lactam
28
P66854. WOO 1 . Spec as filed 8.06.1 1 CH2), 2.09-2.00 (I H, m, lactam CH2), 1.96-1.83 (2H, m, lactam CH2), 1.60-1.36 (2H, m, lactam CH2).
,3C NMR: 5C (100MHz, d6-DMSO) 174.3 (lactam C=0), 164.1 (aryl C=0), 163.9 (C- F,d J 247), 130.7 (ipso-C), 129.8 (ortho-C, d, J 7), 1 15.2 (meta-C, d, J 22), 52.0 (CH- CO), 40.6 (CH2-NH), 30.6 (lactam CH2), 28.9 (lactam CH2), 27.7 (lactam CH2).
HRMS (+ESI) C13H,5FN202 + H+: calcd 251.1 190; found 251.1 192. Reference Example 11 : 3-(Pyridin-3'-carbonylamino)tetrahydropyridin-2- one:
Oxalyl chloride (20 mmol) was added to a solution of nicotinic acid (10 mmol) in DCM (40 mL), along with one drop of catalytic 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, 3- aminotetrahydropyridin-2-one hydrochloride (10 mmol) and 2C03 (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 cm"' 3257 (N-H, amide), 1642, 1541 (secondary CONH, lactam, NH), 1591 , 1479 (aromatic pyridine ring). Ή NMR: δΗ (400MHz, CDC13) 9.03 (I H, d, J 2.0, 2'-aryl CH), 8.71 (I H, dd, J 5.0, 1.5, 6'-aryl CH), 8.12 (I H, dt, J 8.0, 2.0, 4'-aryl CH), 7.36 (IH, dd, J 8.0, 5.0, 5'-aryl CH), 7.27 (I H, br d, J2.0, C5H4N-CONH), 5.91 (IH, br s, CONH-CH2), 4.45 (IH, dt, J 1 1.0, 5.5, CH-CO), 3.44-3.32 (2H, m, CH2NH), 2.72 (IH, dt, J 14.5, 4.5, NHCH- C 2), 2.06-1.93 (2H, m, lactam CH2), 1.70-1.54 (I H, m, lactam CH2).
13C NMR: 6c (100MHz, CDC13) 171.8 (lactam C=0), 166.0 (aryl C=0), 152.5 (aryl N- CH), 148.6 (aryl N-CH), 135.3 (ortho-C(-C )), 133.4 (ipso-C), 123.6 (meta-C), 51.2 (CH-CO), 42.0 (CH2-NH), 27.3 (lactam CH2), 21.3 (lactam CH2).
29
P66854. WOO L Spec as filed 8.06.1 1 HRMS (+ESI) C, ,H 13N302 + H+: calcd 220.1081 ; found 220.1085. Reference Example 12: 3-(Pyridin-3'-carbonyIamino)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 catalytic 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, 3- aminoazepan-2-one hydrochloride (10 mmol) and 2C03 (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%): vmax/cm"1 3200 (N-H, amide), 1642, 1548 (secondary CONH, lactam), 1590, 1476 (aromatic pyridine ring).
Ή NMR: δΗ (400MHz, CDC13) 9.06 (1 H, d, J 2.0, 2'-aryl CH), 8.72 (1 H, dd, J 5.0, 1.5, 6'-aryl CH), 8.1 1 (1 H, dt, J 8.0, 2.0, 4'-aryl CH), 7.72-7.62 (1H, m, C5H4N- CONH), 7.37 (1H, dd, 8.0, 5.0, 5'-aryl CH), 5.99 (1 H, br s, CONH-CH2), 4.70 (1 H, ddd, J 1 1 .0, 5.5, 1.5, CH-CO), 3.40-3.23 (2H, m, CH2NH), 2.24 (1H, br d, J 14.5, lactam CH2), 2.1 1 -2.02 (1 H, m, lactam CH2), 1.97- 1.83 (2H, m, lactam CH2), 1.63- 1.38 (2H, m, lactam CH2).
13C NMR: 5C (l OOMHz, CDC13) 175.7 (lactam C=0), 164.6 (aryl C=0), 152.4 (aryl N- CH), 148.5 (aryl N-CH), 135.1 (ortho-C(-CH)), 130.0 (ipso-C), 123.5 (meta-C), 52.8 (CH-CO), 42.2 (CH2-NH), 3 1 .6 (lactam CH2), 28.9 (lactam CH2), 28.1 (lactam CH2).
HRMS (+ESI) C12H i5N302 + H+: calcd 234.1237; found 234.1239.
Reference Example 13: 3-(3',5'-Dimethylbenzoylamino)tetrahydropyridin-2- one:
30
P66854.WO01.Spec as filed 8.06.1 1 3-aminotetrahydropyridin-2-one hydrochloride (10 mmol), K2C03 (30 mmol) and 3,5- dimethylbenzoyl chloride (10 mmol) were reacted according to the above procedure (except that CHC13 was used instead 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 (weia-trisubstituted benzene ring).
Ή NMR: δΗ (400MHz, CDC13) 7.39 (2H, s, ortho-H), 7.15 (IH, br d, J 4.5, C8H9- CONH), 7.09 (1Η, s, para-U), 6.25 (1Η, br s, CONH-CH2), 4.41 (IH, dt, J 1 1 .5, 5.5, CH-CO), 3.43-3.29 (2H, m, CH2NH), 2.69 (I H, dq, J 13.0, 4.5, NHCH-CH?), 2.31 (6H, s, CH3), 2.03-1.89 (2H, m, lactam CH2), 1.66-1.53 (I H, m, lactam CH2).
I3C NMR: 5C (100MHz, CDCI3) 172.2 (lactam C=0), 168.0 (aryl C=0), 138.2 (ipso- C), 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) C14H|8N202 + H+: calcd 247.1441 ; found 247.1455.
Reference Example 14: 3-(3',5'-Dimethylbenzoylamino)azepan-2-one:
3-aminoazepan-2-one hydrochloride (10 mmol), 2C03 (30 mmol) and 3,5- dimethylbenzoyl 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"' 3319 (N-H, amide), 1682, 1635 (secondary CONH, lactam), 1600, 1498 (aromatic ring), 888, 866, 828 (meto-trisubstituted benzene ring).
Ή NMR: δΗ (400MHz, CDC13) 7.57 (I H, br d, J 5.0, C8H9-CONH), 7.42 (2Η, s, ortho-H), 7.10 (1Η, s, para-U), 6.09 (1 Η, br s, CONH-CH2), 4.69 (I H, ddd, J 1 1.0, 6.0, 1.5, CH-CO), 3.40-3.20 (2H, m, CH2NH), 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).
31
P66854.WO01 .Spec as filed 8.06.11 13C NMR: 5C (100MHz, CDC13) 176.0 (lactam C=0), 166.8 (aryl C=0), 138.3 (ipso- C), 134.3 (rneta-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) C15H20N2O2 + H+: calcd 261.1598; found 261.1602.
In the examples below, the general procedure for the synthesis of 3-acylamino-2- oxopiperidines was: potassium carbonate (3 mmol) and (5 -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 chloroform. The combined organic layers were dried over sodium sulfate and reduced in vacuo to give a solid. This solid was redissolved in a minimum amount of dichloromethane and crystallised 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-oxopiperidin-3-yl)benzamide
Figure imgf000033_0001
0.147 g off-white coarse powder (41 %). mp 164-166 °C. [a]24 D -5.50 (c 0.1 , MeOH); vmax/cm"' 1669, 1644 (C=0, amide), 1552 (N-H, amide), 1303 (C-F). Anal. (C,2H,3FN202) C, H, N: calcd C 61.01, H 5.55, N 1 1.86; found C 60.65, H 5.50, N 11.78. Ή-NMR δΗ. 7.52 (2H, tt, J 8 and 2 , ArH4 and ArH6), 7.38 (1Η, td, J 8 and 5.5, ArH2), 7.21 -7.14 (2Η, m, NHCH and ArH3), 5.9 (1Η, s, NHCH2), 4.41 (I H, dt, J 11.5 and 6 , CHNH), 3.42-3.28 (2H, m, CH2NH), 2.72 (I H, dq, J 13 and 5 , CH2CH), 2.04-1.95 (2H, m, G¾CH2NH), 1.62 (I H, dq, J 16 and 5 , CH2CH). 13C-NMR 5C
32
P66854. WOO 1 .Spec as filed 8.06.1 1 171.6 (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). I9F-NMR 6F -111.9. HRMS (+ESI) Ci2Hi3FN202Na: calcd 259.0853; found 259.0859.
Example 2: (S)-2-FIuoro-N-(2-oxopiperidin-3-yl)benzamide
Figure imgf000034_0001
0.222 g white fluffy powder (63 %). mp 166-168 °C. [a]24 D -10.00 (c 0.1, MeOH). Vmax/cm"1 1647, 1613 (C=0, amide), 1512 (N-H, amide), 1277 (C-F). Anal. (C12Hl3FN202) C, H, N: calcd C 61.01, H 5.55, N 11.86; found C 60.78, H 5.54, N 11.69. Ή-NMR δΗ.8.05 (IH, td, J 8 and 2 , ArH6), 7.62 (1Η, dd, J 6 and 4, NHCH), 7.45 (IH, dddd, J8,7, 5 and 2, ArH4), 7.21 (IH, td, J7.5 and 1, ArH5), 7.13 (1Η, ddd, J 12, 9 and 1, ArH3), 6.01 (1Η, s, NHCH2), 4.53 (IH, dt, J 11 and 6, CHNH), 3.45 (2H, td, J 6 and 3, CH2NH), 2.72 (IH, dq, J 13 and 6, CH2CH), 2.02-1.95 (2H, m, CH2CH2NH), 1.72-1.59 (IH, m, CH2CH). 13C-NMR 5C 171.3 (CHCONH), 163.4 (CONHCH), 160.9 (d, J 248.5, ArC2), 130.17 (d, J 12, ArC5), 133.4 (d, J 9, ArC4), 131.9 (ArC6), 120.9 (d, J 9, CCO), 116.1 (d, J 21, ArC3), 51.3 (CHNH), 41.9 (C¾NH), 27.2 (CH2CHNH), 21.1 (CH2CH2NH).19F-NMR 5F -112.4. HRMS (+ESI) C]2Hi3FN202Na: calcd 259.0853; found 259.0858.
Example 3: (S)-4-Fluoro-N-(2-oxopiperidin-3-yl)benzamide
Figure imgf000034_0002
0.145 g cream coloured fine crystals (41 %)mp 133-135 °C; [a]24 D -7.90 (c 0.1, MeOH); v„/cm'' 1650, 1636 (C=0, amide), 1557 (N-H, amide), 1327 (C-F) Anal.
33
P66854.WO01.Spec as filed 8.06.11 (Ci2H13FN202) C, H, N: calcd C 61.01 , H 5.55, N 1 1 .86; found C 60.71 , H 5.38, N 1 1 .44 (1/3 H20). Ή-NMR δΗ 7.98 (2H, q, J 5, ArH3 and ArH5), 7.41 (1 Η, d, J 6, NHCH), 7.20 (2H, tt, J 9,2, ArH2 and ArH6), 6.31 (1 Η, s, NHCH2), 4.52 (I H, dt, J 1 1 and 6, CHNH), 3.52 (2H, td, J 6 and 2, CH2NH), 2.84 (I H, dq, J 13 and 4, CH2CH), 2.19-2.10 (2H, m, CH2CH2NH), 1.81 (I H, dq, J 12 and 8, CH2CH). 13C- NMR 6C 171.7 (CHCONH), 166.6 (CONHCH), 164.9 (d, J 251.5, ArC4), 130.3 (d, J 4, CCO), 129.5 (d, J 9, ArC2/6), 1 15.5 (ArC3/5), 51.2 (CHNH), 41 .8 (CH2NH), 27.1 (CH2CHNH), 21.0 (CH2CH2NH). HRMS (+ESI) C,2H13FN202Na: calcd 259.0853; found 259.0852.
Example 4: (S)-N-(2-Oxopiperidin-3-yl)-4-(trifluoromethyl)benzamide
Figure imgf000035_0001
0.376 g white fine powder (87 %). mp 212-214 °C; [a] 24 D -6.35 (c 0.1 , MeOH); max cm"1 1650, 1636 (C=0, amide), 1557 (N-H, amide), 1327 (C-F). Anal. (C,3H,3F3N202) C, H, N: calcd C 54.55, H 4.58, N 9.79; found C 53.99, H 4.68, N 9.59. Ή-NMR δΗ Ή-NMR δΗ. 7.51 (2H, d, J 8, ArH2 and ArH6), 7.22 (2Η, d, J 8.5, ArH3 and ArH5), 7.15 (1 Η, s, NHCH), 5.70 (I H, s, NHCH2), 4.00 (I H, dt, J 1 1.5 and 6, CHNH), 2.98 (2H, td, J 6 and 2, CH2NH), 2.25 (I H, dq, J 13 and 4, CH2CH), 1.62- 1.53 (2H, m, CH2CH2NH), 1.28 (I H, dq, J 12 and 8, CH2CH). ,3C-NMR 6C 171.8 (CHCONH), 166.3 (CONHCH), 137.3 (ArCl), 133.3 (q, J 31, ArC4), 127.6 (ArC2/6), 125.5.9 (q, J 4, ArC3/5), 123.7 (q, J 271 , CF3), 51.2 (CHNH), 41 .9 (CH2NH), 27.0 (CH2CHNH), 21.1 (CH2CH2NH). 19F-NMR δΡ -62.9. HRMS (+ESI) C,3Hi3F3N202Na: calcd 309.0821 ; found 309.0822.
Example 5: (5)-N-(2-Oxopiperidin-3-yl)-3-(trifluoromethyl)benzamide
34
P66854.WO0 l .Spec as filed 8.06.1 1
Figure imgf000036_0001
0.251 g cream fine powder (59 %). mp 148-150 °C; [a] 24 D -10.20 (c 0.1 , MeOH); vmax/cm'' 1671 , 1648 (C=0, amide), 1554 (N-H, amide), 1327 (C-F). Anal. (Ci3H,3F3N202) C, H, N: calcd C 54.55, H 4.58, N 9.79; found C 53.90, H 4.56, N 9.60. Ή-NMR δΗ 7.80 (I H, s, ArH2), 7.73 (1 Η, d, J 7, ArH4), 7.60 (1 Η, d, J 6, NHCH), 7.43 (I H, d, J 7, ArH6), 7.23 (1Η, d, J 8, ArH5), 6.52 (1 Η, s, NHCH2), 4.23 (IH, dt, J 1 1 and 6, CHNH), 3.17-3.06 (2H, m, CH2NH), 2.30 (I H, dq, J 13 and 6, CH2CH), 1.74-1.65 (2H, m, CH2CH2NH), 1.62-1.42 (I H, m, GH2CH). 13C-NMR 5C 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).19F-NMR 6F -62.7. HRMS (+ESI) Ci3H13F3N202Na: calcd 309.0821 ; found 309.0820. Example 6: (5)-N-(2-Oxopiperidin-3-yI)-2-(trifluoromethyl)benzamide
Figure imgf000036_0002
0.262 g white fluffy powder (61 %). mp 155-156 °C; [a]24 D -18.20 (c 0.1 , MeOH); vmax/cm'' 1674, 1654 (C=0, amide), 1543 (N-H, amide), 1312 (C-F). Anal. (C,3H,3F3N202) C, H, N: calcd C 54.55, H 4.58, N 9.79; found C 54.25, H 4.51 , N 9.70. 1H-NMR δΗ 7.64 (I H, d, J 7, ArH6), 7.54 (1 Η, d, J 6, ArH3), 7.22-7.15 (2Η, m, ArH4 and ArH5), 6.78 (1 Η, d, J 5, NHCH), 6.08 (IH, NHCH2), 4.42 (I H, dt, J 1 1 and 6, CHNH), 3.35 (2H, td, J 6 and 2, CH2NH), 2.73 (I H, dq, J 13 and 6, CH2CH), 1 .99- 1 .91 (2H, m, CH2CH2NH), 1.59 (I H, dq, J 12 and 8, CH2CH). 13C-NMR 6C 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
35
P66854.WO0 I .Spec as filed 8.06.1 1 (CHNH), 41.8 (CH2NH), 26.5 (CH2CHNH), 20.9 (CH2CH2NH). 19F-NMR 6F -58.7. HRMS (+ESI) Ci3Hl 3F3N202Na: calcd 309.0821 ; found 309.0818.
Example 7: (5)-2,3-difluoro-N-(2-oxopiperidin-3-yl)benzamide
Figure imgf000037_0001
0.234 g white needle crystals (61 %). mp 160-162 °C; [a]2 D -5.65 (c 0.1 , MeOH); Vmax cm"1 1686, 1648 (C=0, amide), 1536 (N-H, amide), 1320 (C-F). Anal. (C,2Hi2F2N202) C, H, N: calcd C 56.99, H 4.76, N 1 1 .02; found C 56.26, H 4.69, N 10.84. Ή-NMR 6H. 7.76 (1 H, ddt, J 8, 6 and 2, ArH6), 7.51 (1 Η, q, J 3, NHCH), 7.28 (1 H, dddd, J 9.5, 8, 7.5, 2, ArH4), 7.15 (1 Η, tdd, J 8, 5, 1.5, ArH5), 6.18 (1 Η, s, NHC¾), 4.47 (1 Η, dt, J 12 and 4, CHNH), 3.38 (2H, td, J 6 and 2, CH2NH), 2.69 (1 H, dq, J 12.5 and 3.5, CH2CH), 2.02-1.93 (2H, m, CH2CH2NH), 1.73-1 .59 (1 H, m, CH2CH). 13C-NMR 5C 171.2 (CHCONH), 162.5 (CCONH), 132.8 (dd, J 250 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) Ci2Hl 2F2N202Na: calcd 277.0759; found 277.0769.
Example 8: (S)-2,4-difIuoro-N-(2-oxopiperidin-3-yI)benzamide
Figure imgf000037_0002
0.236 g off-white crystals (62 %). mp 140-141 °C; [a]24 D -2.00 (c 0.1 , MeOH); vmax/cm"' 1682, 1638 (C=0, amide), 1526 (N-H, amide), 1289 (C-F). Anal.
(C12H 12F2N202. l/6 H20) C, H, N: calcd C 56.03, H 4.83, N 10.89; found C 56.40, H
4.69, N 10.93. Ή-NMR 6H. 8.07 (1 H, td, J 9 and 7, ArH6), 7.54 (1 Η, q, J 5, NHCH),
36
P66854.WO01.Spec as filed 8.06.1 1 6.95 (IH, tdd, J 8, 2.5, 1 , ArH5), 6.84 (IH, dq, J 8.5, 2, ArH3), 6.43 (1 Η, s, NHCH2), 4.45 (IH, dt, J 1 1 and 6, CHNH), 3.36 (2H, td, J 6.5 and 2.5, CH2NH), 2.66 (I H, dq, J 12.5 and 5.5, CH2CH), 2.00-1.91 (2H, m, CH2CH2NH), 1.73-1.59 (I H, m, CH2CH). 13C-NMR 6c 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), 1 17.4 (dd, J 12 and 4, CCONH), 1 12.2 (dd, J 21 and 3, ArC5), 104.3 (t, J 27, ArC3), 51.3 (CHNH), 41.8 (CH2NH), 27.2 (CH2CHNH), 21.1 (CH2CH2NH). HRMS (+ESI) C,2H,2F2N202Na: calcd 277.0759; found 277.0761. Example 9: (S)-2,5-difluoro-N-(2-oxopiperidin-3-yl)benzamide
Figure imgf000038_0001
0.252 g off-white crystals (66 %). mp 140-142 °C; [a]24 D +0.85 (c 0.1, MeOH); v^/cm"1 1681 , 1635 (C=0, amide), 1519 (N-H, amide), 1328 (C-F). Anal. (C,2H12F2N202.1/6 H20) C, H, N: calcd C 56.03, H 4.83, N 10.89; found C 55.47, H 4.72, N 10.67. Ή-NMR δΗ. 8.07 (IH, dt, J 1 1 and 8.5, ArH6), 7.54 (1Η, dd, J 11 , 5, NHCH), 6.68-6.54 (2H, m, ArH3 and ArH4), 6.43 (1 Η, s, NHCH2), 4.45 (IH, dt, J 11 and 6, CHNH), 3.36 (2H, td, J 6.5 and 2.5, CH2NH), 2.66 (IH, dq, J 12.5 and 5.5, CH2CH), 1.99-1.90 (2H, m, CH2CH2NH), 1.64 (1 H, dq, J 1 1 and 8 CH2CH). 13C-NMR 6c 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), 1 18.0 (dd, J 26 and 4, ArC4), 1 17.5 (dd, J 28 and 7, ArC6), 51.4 (CHNH), 41.8 (CH2NH), 27.1 (CH2CHNH), 21.1 (CH2CH2NH). HRMS (+ESI) C12H12F2N202Na: calcd 277.0759; found 277.0766.
Example 10: (S)-2,6-difluoro-N-(2-oxopiperidin-3-yl)benzamide
37
P66854. WOO L Spec as filed 8.06.1 1
Figure imgf000039_0001
0.209 g white fine powder (55 %). mp 190-194 °C; [a] 24 D -7.20 (c 0.1, MeOH); vmax/cm"' 1672, 1638 (C=0, amide), 1492 (N-H, amide), 1329 (C-F). Anal. (Ci2H|2F2N202) 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 H20). Ή-NM δΗ. 7.30 (IH, tt, J 9 and 6, ArH4), 7.06 (IH, d, J 3.5, NHCH), 6.89 (2H, t, J 3.5, ArH3 and ArH5), 6.23 (1Η, 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 6C 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). 19F-NMR 5F -112.5. HRMS (+ESI) Ci2H,2F2N202Na: calcd 277.0759; found 277.0760. Example 11: (S)-3,4-difluoro-N-(2-oxopiperidin-3-yl)benzamide
Figure imgf000039_0002
0.226 g off-white fine powder (59%). mp 202-204 °C; [a]24 D -7.50 (c 0.1, MeOH); vmax/cm"' 1691, 1652 (C=0, amide), 1487 (N-H, amide), 1285 (C-F). Anal. (Ci2Hi2F2N202) 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 H20). Ή-NMR δΗ. 7.66 (IH, qd, J 7 and 2, ArH2), 7.56-7.50 (1Η, m, ArH5), 7.24-7.14 (2Η, m, ArH6 and NHCH), 5.90 (IH, s, NHCH2), 4.38 (IH, dt, J 11.5 and 5, CHNH), 3.39 (2H, td, J 7 and 3, CH2NH), 2.69 (IH, dq, J 14 and 5, CH2CH), 2.03-1.94 (2H, m, CH2CH2NH), 1.68-1.54 (IH, m, CH2CH). 13C-NMR 5C 171.5 (CHCONH), 165.4 (CCONH), 152.5 (dd,J253 and 12, ArC3), 150.2 (dd,J249 and 12, ArC4), 131.2 (t, J 4.5, CCONH), 123.5 (q, J 3.5, ArC6), 117.4 (d,J18, ArC5),
38
P66854. WOO 1. Spec as filed 8.06.11 1 16.9 (d, J 18, ArC2), 51.2 (CHNH), 41.8 (CH2NH), 26.9 (CH2CHNH), 21.1 (CH2CH2NH). HRMS (+ESI) C12H12F2N202Na: calcd 277.0759; found 277.0751.
Example 12: (S)-3,5-difluoro-N-(2-oxopiperidin-3-yl)benzamide
Figure imgf000040_0001
0.234 g white fine powder (61 %). mp 198-199 °C; [a]24 D -9.50 (c 0.1 , MeOH); max/crn 1 1674, 1641 (C=0, amide), 1565 (N-H, amide), 1333 (C-F). Anal. (Ci2H|2F2N202) C, H, N: calcd C 56.99, H 4.76, N 1 1.02; found C 56.20, H 4.69, N 10.93. Ή-NMR δΗ. 7.31 (2H, dd, J 8 and 2.5, ArH2 and ArH6), 7.26 (1 Η, d, J 5, NHCH), 6.9 (IH, tt, J 9 and 2, ArH4), 6.01 (I H, s, NHCH2), 4.39 (IH, dt, J 12 and 5.5, CHNH), 3.41-3.36 (2H, m, CH2NH), 2.68 (I H, dq, J 12 and 5, CH2CH), 2.03-1.94 (2H, m, CH2CH2NH), 1.69-1.55 (I H, m, CH2CH). I 3C-NMR 5C 171.3 (CHCONH), 165.2 (CCONH), 162.9 (dd, J 251.5 and 12, ArC3 and ArC5), 137.5 (t, J 9, CCONH), 1 10.4 (dd, J 20 and 7, ArC2 and ArC6), 107.0 (t, J 20, ArC4), 51.3 (CHNH), 41.8 (CH2NH), 26.9 (CH2CHNH), 20.9 ( H2CH2NH). 19F-NMR 5F -108.2. HRMS (+ESI) C,2H,2F2N202Na: calcd 277.0759; found 277.0750. Example 13: (S)-3-(3'-Butylbenzoylamino)-azepan-2-one
Figure imgf000040_0002
(5)-3-Amino-azepan-2-one hydrochloride (0.72 g, 4.39 mmoles) was dissolved in H20 (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. H20 (20 mL) was added and the reaction was extracted with dichloromethane (3 x 20 mL), the
39
P66854.WO0l .Spec as filed 8.06.1 1 organic layer was washed with a pH 2 buffer (3 x 15 mL), dried over Na2S04 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ethenethyl acetate 75:25 to 0:100) to give the product as a white solid 8H (400 MHz, CDC13) 7.71 -7.62 (m, 3H, Ar & NHCH), 7.35-7.29 (m, 2H, Ar), 7.06 (br.t, I H, J 6, NHCH2), 4.72 (dd, IH, J 1 1 , 6, NHCH), 3.37-3.32 (m, 2Η, NHCH?), 2.65 (t, 2Η, J 8, CH3CH2CH2CH2), 2.22 (br.d, 1 Η, J 13.5, NHCHCH?), 2.03 (br.d, 1Η, J 14, NHCHCH2CH ), 1.95-1.81 (m, 2Η, NHCHCH2CHjCHj), 1.61 (quintet, 2Η, J 7, CH3CH2CH2), 1.57-1.48 (m, 1 Η. NHCHCH^), 1.46-1.25 (m, 3Η, NHCHCHzC^CH^ and CH3CH2) and 0.92 (t, 3H, J 7.5, H8); 5C (100 MHz, CDC13) 176.0 (C=0), 166.6 (C=0), 143.4 (Ar quat, C-nBu), 134.2 (Ar quat, C-C=0), 128.4 (CH, Ar), 127.2 (CH, Ar), 126.8 (CH, Ar), 124.0 (CH, Ar), 52.6 (CH-NH), 42.1 (CH2-NH), 35.6 (CH2), 33.5 (CH2), 31.6 (CH2), 28.9 (CH2), 28.0 (CH2), 22.3 (CH2) and 13.9 (CH3).
Example 14: (S)-3-(4'-Ethylbenzoylamino)-azepan-2-one
Figure imgf000041_0001
(5)-3-amino-azepan-2-one hydrochloride (1.65 g, 10 mmoles) was dissolved in H20 (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. H20 (20 mL) was added and the reaction was extracted with dichloromethane (3 χ 20 mL), the organic layer was washed with a pH 2 buffer (3 x 15 mL), dried over Na2S04 and reduced in vacuo. The product was purified by recrystallisation from chloroform and cold petroleum ether to give the product as a white solid 0.94 g (36 %); mp 218-219 0 C; δΗ (400 MHz, CDC13) 7.66 (d, 2H, J 8, CH-C-Et), 7.62 (d, 1 Η, J 5.5, NH-CH), 7.24 (d, 2H, J 8, CH-C-CO), 6.55 (br.t, 1 Η, J 6, NH-Cl), 4.70 (dd, 1Η, J 1 1 , 5.5, CH-C4), 3.37-3.32 (m, 2Η, HI), 2.67 (q, 2H, J 7.5, H5), 2.21 (br.d, I H, J 13, H4 equatorial), 2.02 (dt, I H, J 14, 4, H3 equatorial), 1 ,95-1.82 (m, 2H, H2 equatorial & H3 axial), 1.53 (br.q, 1 H, J 12.5, H4 axial), 1.40 (br.q, 1 H, J 13, H2 axial) and 1.22 (t, 3H, J 7.5, H6); 6C (100 MHz, CDC13) 175.9 (C=0), 166.2 (C=0), 148.2 (C-Et), 131.6 (C-C=0), 128.0, 127.2 (CH phenyl), 52.6 (CH-NH), 42.2 (CI), 28.9 (C5), 28.8, 28.0 (C2, C3)
40
P66854.WO0l .Spec as filed 8.06.1 1 and 15.4 (C6); t /cm"1: 3200 (NH indole), 2956 (C-H), 1642 (amide C=0) and 1543 (aromatic); ESI m/z 100 %, 542.9 (M2Na+), 70%, 283.1 (M a+) and 10 %, 261.2 (MH+); HR ESI m/z (Ci5H2oN202Na requires 283.1417) found 283.1414; [a]23 D (c = 0.49, CHC13) +70.48.
Example 15: (S)-3-(4'-Ethylbenzoylamino)-tetrahydropyridin-2-one
Figure imgf000042_0001
(5)-3-amino-tetrahydropyridin-2-one (20 mmoles) was dissolved in H20 (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 Na2S04 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 1 12-113 0 C; δΗ (400 MHz, CDCI3) 7.71 (d, 2H, J 8.5, CH-C-Et), 7.55 (d, 1Η, J 5.5, NH-CH), 7.19 (d, 2H, J 8.5, CH-C-CO), 6.69 (br.s, 1H, NH-C1), 4.39 (dt, 1Η, J 11 , 5.5, CH-C3), 3.35-3.28 (m, 2Η, HI), 2.67 (q, 2H, J 7.5, H4), 2.63-2.56 (m, 1 H, H3 equatorial), 1.94- 1.87 (m, 2H, H2), 1.68 (tt, 1H, J 12.5, 8, H3 axial), and 1.20 (t, 3H, J 7.5, H5); 5C (100 MHz, CDCI3) 172.2 (C=0), 167.5 ( =0), 148.2 ( -Et), 131.5 (C-C=0), 127.9, 127.2 (CH phenyl), 50.9 (CH-NH), 41.7 (CI), 28.8 (C4), 27.2 (C3), 21.1 (C2) and 15.3 (C5); Umax/cm"1: 3334, 3245 (NH), 2932 (C-H), 1656, 1634 (C=0) and 1528 (aromatic); ESI m/z 100 %, 514.9 (M2Na+) and 35%, 269.1 (MNa+); HR ESI m/z (C14Hi8N202Na requires 269.1260) found 269.1261 ; [a]23 D (c = 0.491 , CHC13) +103.95.
Example 16: (S)-3-(4'-Butylbenzoylamino)-azepan-2-one
41
P66854. WOOL Spec as filed 8.06.1 1
Figure imgf000043_0001
(S)-3-amino-azepan-2-one hydrochloride (1.65 g, 10 mmoles) was dissolved in H20 (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. H20 (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 Na2S04 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ethenethyl 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, CDC13) 7.73 (d, 2H, J 8, Ar), 7.63 (d, 1H, J 5.5, NHCH), 7.21 (d, 2H, J 8, Ar), 6.80 (br.t, 1H, J 6, NHCH2), 4.69 (dd, 1 H, J 10.5, 5.5, NHCH), 3.35-3.20 (m, 2Η, CH^NH), 2.61 (t, 2H, J 7.5, H5), 2.19 (br.d, 1H, J 13.5, H4 equatorial), 2.00 (br.d, 1H, J 12.5, H3 equatorial), 1.93-1.80 (m, 2H, H2 equatorial & H3 axial), 1.70-1.46 (m, 3H, H6 and H4 axial), 1.38 (br.q, 1H, J 13, H2 axial), 1.31 (sextet, H2, J 7, H7) and 0.89 (t, 3H, J 7.5, H8); 6C (100 MHz, CDC13) 176.0 (C=0), 166.3 (C=0), 146.7 ( -nBu), 131.6 ( -C=0), 128.5, 127.1 (CH phenyl), 52.5 ( H-NH), 42.2 (CI), 35.5 (C5), 33.4 (C4), 31.6 (C6), 28.9, 28.0 (C2, C3), 22.3 (C7) and 13.9 (C6); i /cm"1 : 3359, 3207 (NH), 2951 (C-H), 1671 , 1650 (CO) and 1543 (aromatic); ESI m/z 100 %, 31 1.2 (M a+) and 22 %, 289.2 (MH+); HR ESI m/z (C,7H24N202Na requires 31 1.1730) found 31 1.1732; [a]25 D (c = 0.515, CHCI3) +64.88.
Example 17: (S)-3-(4'-Butylbenzoylamino)-tetrahydropyridin-2-one
Figure imgf000043_0002
42
P66854.WO01 Spec as filed 8.06.1 1 (5)-3-amino-tetrahydropyridin-2-one (20 mmoles) was dissolved in H20 (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. H20 (20 mL) was added and the reaction was extracted with dichloromethane (3 χ 20 mL), the organic layer was washed with a pH 2 buffer (3 x 15 mL), dried over Na2S04 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ethenethyl acetate: methanol 50:50:0 to 0:80:20) to give the product as a white solid 0.45 g (16 %); δΗ (400 MHz, CDC13) 7.70 (d, 2H, J 8, CH-C-nBu), 7.36 (d, 1H, J 6, NH- mp 1 17-1 18 0 C; CH), 7.17 (d, 2H, J 8, CH-C-CO), 6.67 (br.s, 1 H, NH- CI), 4.69 (dt, 1H, J 12, 6, CH-C4), 3.36-3.29 (m, 2H, HI ), 2.60 (t, 3H, J 7.5, H4 & H3), 1.93-1.86 (m, 2H, H2), 1.67- 1.54 (m, 1H, 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); 6C (100 MHz, CDCI3) 172.2 (C=0 lactam), 167.5 (C=0 amide), 146.9 (C-nBu), 131.5 (C-C=0), 128.5 (CH phenyl), 127.2 (CH phenyl), 50.8 (CH-NH), 41.7 (CI), 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 (C16H22N202Na requires 297.1573) found 297.1573; [a]24 D (c = 0.523,
Figure imgf000044_0001
Example 18: (R)-3-(4'-Butylbenzoylamino)-tetrahydropyridin-2-one
Figure imgf000044_0002
(^)-3-amino-tetrahydropyridin-2-one (10 mmoles) was dissolved in H20 (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 * 15 mL), the organic layer was washed with a pH 2 buffer (3 x 15 mL), dried over Na2S04 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ethenethyl acetate:methanol 50:50:0 to 0:80:20) to give the product as a white solid 1.10 g (40 %); mp 1 16-1 17 0 C; δΗ (400 MHz, CDCI3) 7.72 (d, 2H, J 8, CH-C-nBu), 7.25 (d, 1 Η, J
43
P668S4. WOOL Spec as filed 8.06.1 1 5.5, NH-CH), 7.20 (d, 2H, J 8, CH-C-CO), 6.41 (br.s, 1Η, NH-Cl ), 4.41 (dt, 1 Η, J 1 1.5, 5.5, CH-C4), 3.37-3.32 (m, 2Η, HI), 2.66 (ddt, IH, J 13, 6, 4.5, H3 equatorial), 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); δ0 (100 MHz, CDC13) 172.1 (C=0 lactam), 167.6 (C=0 amide), 147.0 (C-nBu), 131.5 (C-C=0), 128.5 (CH phenyl), 127.2 (CH phenyl), 51.0 (CH-NH), 41.7 (CI), 35.5 (C4), 33.3 (C5), 27.2 (C3), 22.3 (C6), 21.1 (C2) and 13.9 (C7); υ^/cm"1 : 3319, 3245 (NH), 2949 (C-H), 1651 , 1634 (C=0) and 1521 (aromatic); ESI m/z 100 %, 297.1 (MNa+); HR ESI m/z (Ci6H22N202Na requires 297.1573) found 297.1574; [ct]25 D (c = 0.493, CHC13) -89.45.
Example 19: (S)-3-(4'-½ri-Butylbenzoylamino)-azepan-2-one
Figure imgf000045_0001
3 2 (5)-3-amino-azepan-2-one hydrochloride (2.04 g, 12.44 mmoles) was dissolved in H20 (20 mL) and cooled to 0°C. The 4-tButylbenzoyl chloride in dichloromethane was added and triethylamine (4.2 mL, 30 mmoles) and the reaction was stirred over night. H20 (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 * 20 mL), dried over Na2S04 and reduced in vacuo. The product was purified by recrystallisation from chloroform 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 ° C; δΗ (400 MHz, CDC13) 7.76 (d, 2H, J 8.5, CH-C-'Bu), 7.66 (d, 1Η, J 6, NH-CH), 7.42 (d, 2H, J 8.5, CH-C-CO), 6.00 (br.s, 1Η, NH-Cl), 4.67 (ddd, 1Η, J 11, 6, 1.5, CH-C4), 3.34-3.19 (m, 2Η, HI), 2.18 (br.d, I H, J 13, H4 equatorial), 2.00 (br.d, I H, J 12.5, H3 equatorial), 1.92-1.78 (m, 2H, H2 equatorial & H3 axial), 1.51 (q, I H, J 13, H4 axial), 1.33 (br.q, I H, J 1 1 , H2 axial), and 1.29 (s, 3H, C(CH3)3); 6C (100 MHz, CDC13) 176.0 (C=0), 166.2 (C=0), 155.0 (C-C(CH3)3), 131.4 ( -C=0), 127.9, 125.4 (CH phenyl), 52.5 (CH-NH), 42.1 (CI), 34.9 (C(CH3)3), 31.6 (C4), 31.2 (C(CH3)3) and 28.9, 28.0 (C2, C3); ESI m/z 100 %,
44
P66854. WOO L Spec as filed 8.06.1 1 598.8 (M2Na+) and 32 %, 289.2 (MH+); HR ESI m/z (Ci7H24N202Na requires 31 1.1730) found 31 1.1736; t)raax/cm"': 3210 (NH indole), 2906 (C-H), 1640 (C=0) and 1566 (aromatic); [a]23 D (c = 0.523, CHC13) +63.77. Example 20: (S)-3-(4'-iert-Butylbenzoylamino)-tetrahydropyridin-2-one
Figure imgf000046_0001
(5r)-3-amino-tetrahydropyridin-2-one (33 mmoles) was dissolved in H20 (100 mL) and cooled to 0°C. 4-tButylbenzoyl chloride (20 mmoles) in dichloromethane was added and triethylamine (6.3 mL, 45 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 Na2S0 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; δΗ (400 MHz, CDC13) 7.24 (d, 2H, J 8.5, CH-C-¾u), 7.49 (d, I H, J 6, NH-CH), 7.36 (d, 2H, J 8.5, CH-C-CO), 6.94 (br.s, 1 Η, NH-Cl), 4.62 (dt, 1 Η, J 1 1 , 6, 1.5, CH-C4), 3.31-3.26 (m, 2Η, HI), 2.52 (ddt, IH, J 13, 6, 4.5, H3 equatorial), 1.90- 183 (m, 2H, H2), 1.63 (tt, IH, J 12.5, 8.5, H3 axial) and 1.27 (s, 9H, C(CHj)3); 5C (100 MHz, CDC13) 172.3 ( =0), 167.4 ( =0), 154.9 (C-C(CH3)3), 131.2 (C-CO), 127.0, 126.7, 125.3 ( H phenyl), 50.8 ( H-NH), 41.6 (CI), 34.9 (C(CH3)3), 31.2 (C(CH3)3), 27.2 (C3) and 21.1 (C2); ESI m/z 100 %, 297.2 (MNa+) and 38 %, 275.2 (MH+); Umax/cm"' : 3251 (NH), 2959 (C-H), 1683, 1648 (C=0) and 1558 (aromatic); HR ESI m/z (Ci6H23N202 requires 275.1754) found 275.1752; [a]23 D (c = 0.515, CHCI3) +82.52.
Example 21: (R)-3-(4'-feri-Butylbenzoylamino)-tetrahydropyridin-2-one
45
P668S4.WO0 l .Spec as filed 8.06.1 1
Figure imgf000047_0001
(fl)-3-amino-tetrahydropyridin-2-one ( 15 mmoles) was dissolved in H20 (100 mL) and cooled to 0°C. 4-lButylbenzoyl 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 x 20 mL), the organic layer was washed with a pH 2 buffer (3 * 20 mL), dried over Na2S04 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 1.03 g (38 %) mp 193-194 °C; δΗ (400 MHz, CDC13) 7.72 (d, 2H, J 8.5, CH-C-'Bu), 7.49 (d, 1H, J 6, NH-CH), 7.36 (d, 2H, J 8.5, CH-C-CO), 6.93 (br.s, 1H, NH-Cl), 4.39 (dt, 1 Η, J 1 1.5, 6, CH-C4), 3.31 -3.26 (m, 2Η, HI ), 2.52 (ddt, 1H, J 12.5, 5.5, 4.5, H3 equatorial), 1.90- 1.87 (m, 2H, H2), 1.63 (tt, 1H, J 12.5, 8.5, H3) and 1.27 (s, 9H, C(CH,)3); 6C (100 MHz, CDCI3) 172.3 (CO), 167.4 (0=0), 154.9 (C-C(CH3)3), 131.2 (C-C=0), 127.0, 125.3 ( H phenyl), 50.8 ( H-NH), 41.6 (CI), 34.9 (C(CH3)3), 31.2 (C( H3)3), 27.2 (C3) and 21.1 (C2); t /cm 1: 3247 (NH), 2958 (C-H), 1682, 1647 (C=0) and 1544 (aromatic); ESI m/z 19 %, 297.2 (MNa+) and 13 %, 275.2 (MH+); HR ESI m/z (CI 6H2 N202 requires 297.1573) found 275.1750; [a]23 D (c = 0.512, CHC13) -84.08. Example 22: (S)-3-(4'-HexyIbenzoylamino)-azepan-2-one
Figure imgf000047_0002
(S)-3-amino-azepan-2-one hydrochloride (0.95 g, 5.79 mmoles) was dissolved in H20 (15 mL) and cooled to 0°C. 4-Hexylbenzoyl chloride (1.2 mL, 5 mmoles) in
46
P668S4. WOO L Spec as filed 8.06.1 1 dichloromethane was added and triethylamine (2.1 mL, 15 mmoles) and the reaction was stirred over night. H20 (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 Na2S04 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ethenethyl acetate 50:50:0 to 0:80:20) to give the product as a white solid 0.76 g (48 %); mp 167-168 ° C; δΗ (400 MHz, CDC13) 7.74 (d, 2H, J 8, CH-C-Hex), 7.61 (d, 1 H, J 6, NH-CH), 7.21 (d, 2H, J 8, C/ -C-CO), 6.54 (br.t, 1H, J 6, NH-C1), 4.69 (ddd, 1H, J 1 1 , 6, 1.5, CH-C4), 3.37-3.22 (m, 2Η, HI), 2.62 (t, 2H, J 7.5, H5), 2.21 (d, IH, J 13, H4 equatorial), 2.03 (dt, 1 H, J 14, 3.5, H3 equatorial), 1.95-1.82 (m, 2H, H2 equatorial & H3 axial), 1.64-1.49 (m, 3H, H4 axial & H6), 1.41 (q, I H, J 13, H2 axial) 1.33-1.23 (m, 6H, H7, H8 & H9) and 0.86 (t, 3H, J 7, H 10); 8c (100 MHz, CDC13) 175.9 (C=0 lactam), 166.3 (C=0 amide), 147.0 (C- Hex), 131.6 ( -C=0), 128.5 (CH phenyl), 127.1 (CH phenyl), 52.6 (CH-NH), 42.2 (CI), 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); i /cm"1 : 3244 (NH), 2956 (C-H), 1658, 1644 (C=0) and 1543 (aromatic); ESI m/z 43 %, 317.2 (MH+), 6% 339.2 (MNa+) and 6 %, 654.7 (M2Na+); HR ESI m/z (C19H28N202Na requires 339.2043) found 339.2050; [a]25 D (c = 0.507, CHC13) +60.06. Example 23: (S)-3-(4'-Hexylbenzoylamino)-tetrahydropyridin-2-one
Figure imgf000048_0001
(5)-3-amino-tetrahydropyridin-2-one (10 mmoles) was dissolved in H20 (35 mL) and cooled to 0°C. 4-Hexylbenzoyl chloride (1.2 mL, 5 mmoles) in dichloromethane was added and triethylamine (2.1 mL, 15 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 Na2S04 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 %); mp 1 18-119 0 C;
47
P66854.WO01.Spec as filed 8.06.1 1 δΗ (400 MHz, CDCI3) 7.70 (d, 2H, J 8, CH-C-Hex), 7.38 (d, 1 H, J 6, NH-CH), 7.17 (d, 2H, J 8, CH-C-CO), 6.81 (br.s, 1H, NH-C1), 4.39 (dt, 1Η, J 1 1.5, 6, CH-C3), 3.33- 3.26 (m, 2Η, H I), 2.58 (t, 2H, J 7.5, H4), 2.57-2.52 (m, 1H, H3 equatorial 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); ); 8C (100 MHz, CDCI3) 172.3 (C=0 lactam), 167.5 (C=0 amide), 146.9 (C-Hex), 131.5 (C-C=0), 128.4 (CH phenyl), 127.2 (CH phenyl), 50.8 (CH-NH), 41.6 (CI), 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); ojcm"1: 3338, 3247 (NH), 2921 (C-H), 1656, 1637 (C=0) and 1562 (aromatic); ESI m/z 100 %, 325.2 (MNa+) and 37% 303.2 (MH+); HR ESI m/z (Ci8H26N202Na requires 325.1886) found 325.1883; [a]23 D (c = 0.51 1 , CHClj) +79.55.
Example 24: (R)-3-(4,-HexylbenzoyIamino)-tetrahydropyridin-2-one
Figure imgf000049_0001
(i?)-3-amino-tetrahydropyridin-2-one (10 mmoles) was dissolved in H20 (20 mL) and cooled to 0°C. 4-Hexylbenzoyl chloride (1.2 mL, 5 mmoles) in dichloromethane was added and triethylamine (2.1 mL, 15 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 Na2S04 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 117-1 18 0 C; δΗ (400 MHz, CDCI3) 7.71 (d, 2H, J8, CH-C-Hex), 7.28 (d, 1H, J 5, NH-CH), 7.19 (d, 2H, J 8, CH-C-CO), 6.52 (br.s, 1Η, NH-C1 ), 4.41 (dt, 1 Η, J 1 1.5, 5.5, CH-C3), 3.35- 3.31 (m, 2Η, H I ), 2.67-2.60 (m, 1 H, H3 equatorial), 2.61 (t, 2H, J 7.5, H4), 2.00-1.89 (m, 2H, H2), 1.67-1.54 (m, 3H, H3 equatorial & H5), 1.32-1.23 (m, 6H, H6, H7 & H8) and 0.85 (t, 3H, J 7, H9); ); 8C (100 MHz, CDC13) 172.1 (C=0 lactam), 167.6 (C=0 amide), 147.0 (C-Hex), 131.5 (C-C=0), 128.5 (CH phenyl), 127.2 (CH phenyl), 50.9 (CH-NH), 41.7 (CI ), 35.8 (C4), 31.7 (C3), 31.2 (C5), 28.9 (C6), 27.2, (C7), 22.6 (C8),
48
P66854.WO01.Spec as filed 8.06.11 21.1 (C2) and 14.1 (C9); i /cm 1: 3328, 3240 (NH), 2954 (C-H), 1651 , 1635 (C=0) and 1533 (aromatic); ESI m/z 100 %, 325.2 (MNa+) and 33% 303.2 (MH+); HR ESI m/z (C,8H26N202Na requires 325.1886) found 325.1888; [a]25 D (c = 0.496, CHC13) - 81.17.
Example 25: (S)-3-(4'-Octylbenzoylamino)-azepan-2-one
Figure imgf000050_0001
(5)-3-amino-azepan-2-one hydrochloride (0.91 g, 5.55 mmoles) was dissolved in H20 (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. H20 (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 Na2S04 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 ° C; δΗ (400 MHz, CDC13) 7.73 (d, 2H, J 8, CH-C-Oct), 7.64 (d, 1H, J 5.5, NH-CH), 7.20 (d, 2H, J 8, CH-C-CO), 6.94 (br.t, 1H, J 6, NH-C1), 4.68 (dd, 1H, J 1 1 , 6, CH-C4), 3.35-3.20 (m, 2Η, HI), 2.60 (t, 2H, J 7.5, H5), 2.19 (d, 1 H, J 13, H4 equatorial), 2.00 (br.d, 1H, H3 equatorial), 1.92-1.79 (m, 3H, H2 & H3 axial), 1.62-1.46 (m, 3H, H4 axial & H6), 1.38 (q, 1H, J 1 1.5, H2 axial) 1.30-1.19 (m, 10H, H7, H8, H9, H10 & Hl l) and 0.84 (t, 3H, J 7.5, H12); 5C (100 MHz, CDC13) 176.0 (C=0 lactam), 166.3 (C=0 amide), 146.9 ( -Oct), 131.6 (C-C=0), 128.5 (CH phenyl), 127.1 (CH phenyl), 52.5 (CH-NH), 42.1 (CI ), 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 CIO), 22.7 (CI 1 ) and 14.1 (C12); iw/cm"1 : 3204 (NH indole), 2923 (C-H), 1637 (amide C=0) and 1544 (aromatic); ESI m/z 100 %, 345.2 (MH+) and 9 %, 710.8 (M2Na+); HR ESI m/z (C2iH32N202Na requires 367.2356) found 367.2361 ; [ct]25 D (c = 0.124, CDC13) +68.01.
49
P66854. WOO L Spec as filed 8.06.1 1 Example 26: (S)-3-(4'-OctyIbenzoylamino)-tetrahydropyridin-2-one
Figure imgf000051_0001
2 (5)-3-amino-tetrahydropyridin-2-one (10 mmoles) was dissolved in H20 (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 (3 x 10 mL), dried over Na2S04 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ethenethyl acetate 50:50:0 to 0:80:20) to give the product as a white solid 0.55 g (83 %); mp 122-123 ° C; δΗ (400 MHz, CDC13) 7.71 (d, 2H, J 8, CH-C-Oct), 7.26 (d, 1Η, J 5.5, NH-CH), 7.19 (d, 2H, J 8, CH-C-CO), 6.47 (br.s, 1Η, NH-C1), 4.41 (dt, 1Η, J 1 1.5, 5.5, CH-C3), 3.33-3.27 (m, 2Η, HI), 2.84-2.58 (m, 1H, J 13, H3 equatorial 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, HI 1); 5C (100 MHz, CDC13) 172.1 (C=0 lactam), 167.6 (C=0 amide), 147.0 (C-Oct), 131.5 (C-C=0), 128.5 (CH phenyl), 127.2 (CH phenyl), 50.9 (CH-NH), 41.7 (CI), 35.8 (C4), 31.9 (C5), 31.2 (C3), 29.4 (C6), 29.3 (C7, C8), 27.2 (C9), 22.3 (CIO), 21.1 (C2) and 14.1 (CI 1); Omax/cm"' : 3240 (NH), 2921 (C-H), 1653, 1615 (C=0) and 1563 (aromatic); ESI m/z 100 %, 353.2 (MNa+) and 47 %, 331.2 (MH+); HR ESI m/z (C20H30N2O2Na requires 353.2199) found 353.2198; [a]23 D(c = 0.509, CHC13) +75.74.
Example 27: (R)-3-(4'-Octylbenzoylamino)-tetrahydropyridin-2-one
Figure imgf000051_0002
2
50
P66854. WOOl .Spec as filed 8.06.1 1 (/?)-3-amino-tetrahydropyridin-2-one (5 mmoles) was dissolved in H20 (25 mL) and cooled to 0°C. 4-Octyl benzoyl 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 >< 10 mL), dried over Na2S0 and reduced in vacuo. The product was purified by silica column chromatography (petroleum ethenethyl acetate 50:50:0 to 0:80:20) to give the product as a white solid 0.16 g (25 %); mp 122-123 ° C; δΗ (400 MHz, CDC13) 7.71 (d, 2H, J 8.5, C f-C-Oct), 7.23 (d, 1H, J 6.5, N//-CH), 7.20 (d, 2H, J 8.5, CH-C-CO), 6.34 (br.s, 1H, NH-C1), 4.41 (dt, 1H, J 1 1.5, 5.5, CH-C3), 3.37-3.33 (m, 2Η, HI ), 2.68 (ddt, 1 H, J 13, 5.5, 4.5, H3 equatorial), 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, J 7, HI 1); 5C (100 MHz, CDC13) 172.1 (C=0 lactam), 167.6 (C=0 amide), 147.0 ( -Oct), 131.5 ( -C=0), 128.5 (CH phenyl), 127.2 (CH phenyl), 51.0 (CH-NH), 41.7 (CI), 35.8 (C4), 31.9 (C5), 31.2 (C3), 29.4 (C6), 29.3 (C7, C8), 27.2 (C9), 22.7 (CIO), 21.1 (C2) and 14.1 (Cl l); ESI m/z 39 %, 353.2 (M a+), 19 %, 331.2 (MH+) and 14 %, 682.7 (M2Na+); HR ESI m/z (C20H30N2O2H+ requires 331.2380) found 331.2381 ; t cm"1: 3250 (NH), 2955 (C-H), 1653 (C=0) and 1540 (aromatic); [a]23 D (c = 0.485, CHC13) -77.80.
Pharmacological study of the products of the invention
A. Inhibition of MCP-1 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.
51
P66854.WO01.Spec as filed 8.06.1 1 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 (Gaithersburg, 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 series of controls).
This assay can be used with a range of different leukocyte populations. For example, freshly prepared human peripheral blood leukocytes may used. Alternatively, 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 separations. Alternatively, immortal cell lines which have been extensively validated as models of human peripheral blood leukocytes may be used, including, but not limited to THP-1 cells as a model of monocytes or Jurkat cells as model of naive T cells.
Although a range of conditions for the assay are acceptible to demonstrate the inhibition of chemokine-induced leukocyte migration, a specific example is hereby provided.
Materials
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 Salt Solution is purchased from Sigma (Sigma G-9779).
Fatty acid-free BSA is purchased from Sigma (Sigma A-8806).
52
P66854.WO0 l .Spec as filed 8.06.1 1 TT, i.e. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, is purchased from Sigma (Sigma M-5655).
RPMI-1640 without phenol red is purchased from Sigma (Sigma R-8755).
The THP-1 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- 1 induced leukocyte migration:
First, the cell suspension to be placed in the upper compartment is prepared. The THP- 1 cells are pelleted by centrifugation (770 x g; 4 mins) and washed with Geys
Balanced Salt Solution with Img/ml BSA (GBSS + BSA). This wash is then repeated, and the cells repelleted before being 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 present 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-1 cells in each 25μ1 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-1 induced migration, it is necessary to prepare two lots of cells. The suspension of THP-1 cells at 4.45 x 106 cells/ml is divided into two pots. To one pot the inhibitor under test is added at an appropriate final concentration, in an appropriate vehicle (for example at 1 μΜ in not 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 is diluted in GBSS + BSA to give a final concentration of 25 ng/ml. This is divided 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
53
P66854. WOOL Spec as filed 8.06.1 1 other pot an equal volume of GBSS + BSA plus vehicle as appropriate (e,g. not more than 1 % DMSO) is 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 establishing 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 have 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 determinations of each condition. Once all the lower chambers have 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 evaporation. The assembled chamber is incubated at 37 °C, 5% C02, 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 have 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 filter is carefully flushed with GBSS + BSA to wash off the EDTA, and then the filter is 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 1-2 hours during which time dehydrogenase
54
P668S4. WOO L Spec as filed 8.06.1 1 enzymes within the cells convert 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 precipitated 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-well 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 present at 25ng/ml.
The impact of the test substance is calculated by comparing the MCP-1 -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 determining the inhibition of MCP-1 induced migration which occurs at a range of different compound concentrations (typically ranging from InM to 1 μΜ or higher in the case of poorly active compounds). The inhibitory activity of each compound is then expressed as the concentration of compound required to reduce the MCP-1 -induced migration by 50% (the ED50 concentration).
Results
The compounds of reference examples 1 to 14 were tested and were shown to have an ED50 of 100 nM or less in this test.
55
P66854. WOOL Spec as filed 8.06.1 1 B. In vivo assay
The anti-inflammatory efficacy of an exemplary compound according to the present invention was tested using the murine sub-lethal endotoxemia model. This model has been widely used to demonstrate the anti-inflammatory effect of compounds in vivo - Fox et a 2009, J Med Chem. 52(1 1): 3591-3595.
Briefly, the method is as follows: Female CD1 mice (28-30g, ~7 weeks of age) were dosed with their respective treatment in sterile filtered 1% CMC by oral gavage in a dose volume of lOml kg one hour prior to an endotoxin (LPS) challenge. The endotoxin challenge was injected by the intraperitoneal route containing 675,000 Endotoxin Units of LPS (E. coli strain 01 1 1 :B4 (Code L4130)) in endotoxin free PBS. Mice were left for two hours and then exsanguinated under terminal anaesthesia and blood was taken. Serum was prepared from this terminal bleed and aliquoted and stored at -20°C. Serum TNF-a levels were measured by ELISA per manufacturers instructions (R and D Systems). Eight animals 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 animals. Data for untreated animals 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 have anti-inflammatory activity in vivo.
56
P66854.WO01.Spec as filed 8.06.1 1

Claims

Claims
1. A compound of general formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of an inflammatory disorder:
Figure imgf000058_0001
(I) wherein 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 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 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; 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.
57
P66854. WOOl Spec as filed 8.06.1 1
2. A compound of formula (Ι'), or a pharmaceutically acceptable salt thereof, for use in the treatment of an inflammatory disorder:
Figure imgf000059_0001
wherein n, k and X are defined as in claim 1.
3. Use of a compound of general formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of an inflammatory disorder:
Figure imgf000059_0002
(I) wherein 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 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:
58
P66854.WO01 .Spec as filed 8.06.1 1 when 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; 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.
4. Use of a compound of formula (F), or a pharmaceutically acceptable salt thereof, the manufacture of a medicament for the treatment of an inflammatory disorder:
Figure imgf000060_0001
( ) wherein n, k and X are defined as in claim 3.
5. A compound of general formula (I):
Figure imgf000060_0002
(I) wherein n is an integer from 1 to 4;
59
P66854.WO0 l .Spec as filed 8.06.1 1 k is an integer from 1 to 5, representing the number of groups substituting C2, C3, C4, C5 and/or C6 of the benzyl ring; when n is 1 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 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 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; and when
Figure imgf000061_0001
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'-dimethoxybenzoylamino)- caprolactam, 3-(3',5'-di-tert-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, 3-(2'- fluoromethylbenzoylamino)-caprolactam, 3-(2'-amino-3'-hydroxy-4'- methylbenzoylamino)-caprolactam, and 3 -(3 ',5 ' -dimethylbenzoylamino)-caprolactam.
60
P66854.WO01 .Spec as filed 8.06.1 1
6. A compound of formula (Γ):
Figure imgf000062_0001
d') wherein n, k and X are defined as in either of claim 1 or claim 5, provided that the compound is compound is none of the group consisting of: (S)-3-(4'- methoxybenzoylamino)-caprolactam, (S)-3-(4'-methylbenzoylamino)-caprolactam, (S)-3-(3'-trifluoromethylbenzoylamino)-caprolactam, (S)-3-(2'-carboxybenzoyl- amino)-caprolactam, and (S)-3-(3', 4',5'-trimethoxybenzoylamino)-caprolactam.
7. A pharmaceutical composition comprising, as active ingredient, a compound as defined in either of claims 5 or 6, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient and/or carrier.
8. The compound, use or composition according to any preceding claim, wherein n=2.
9. The compound, use or composition according to any preceding claim, wherein n=3.
10. The compound, use or composition according to any preceding claim, wherein X is haloalkyl, for example trifluoromethyl.
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. 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:
61
P66854. WOOL Spec as filed 8.06.1 1 (S)-3-Fluoro-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,
(S)-N-(2-Oxopiperidin-3 -yl)-3 -(trifluoromethyl)benzamide,
(S)-N-(2-Oxopiperidin-3-yl)-2-(trifluoromethyl)benzamide,
(. )-2,3-difluoro-N-(2-oxopiperidin-3-yl)benzamide,
(5 -2,4-difluoro-N-(2-oxopiperidin-3-yl)benzamide,
(-y)-2,5-difluoro-N-(2-oxopiperidin-3-yl)benzamide,
(S)-2,6-difluoro-N-(2-oxopiperidin-3-yl)benzamide,
(S -3,4-difluoro-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.
13. The compound according to any of claims 1 , 2, 5 or 6, or the use according to claims 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'-tert-Butylbenzoylamino)-azepan-2-one,
(S)-3-(4'-Hexylbenzoylamino)-azepan-2-one,
(S)-3-(4'-Octylbenzoylamino)-azepan-2-one, and
(S)-3-(4'-Octylbenzoylamino)-tetrahydropyridin-2-one, and pharmaceutically acceptable salts thereof.
14. The compound according to claim 1 , or the use according to claim 3, wherein the compound is selected from the group consisting of:
62
P66854. WOOL Spec as filed 8.06.1 1 (R)-3-(4'-Butylbenzoylamino)-tetrahydropyridin-2-one,
(R)-3-(4'-tert-Butylbenzoylamino)-tetrahydropyridin-2-one, and
(R)-3-(4'-Hexylbenzoylamino)-tetrahydropyridin-2-one, and pharmaceutically acceptable salts thereof.
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-2- one or a pharmaceutically acceptable salt thereof.
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-ct level, psoriasis, allergies, multiple sclerosis, fibrosis (including diabetic nephropathy), and formation of adhesions.
17. The compound or use according to claim 16, wherein the inflammatory disorder is formation of adhesions.
18. The compound or use according to claim 16 or claim 17, wherein the compound is administered locally.
19. 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 anti-inflammatory amount of a compound, pharmaceutical composition or medicament as defined in any preceding claim.
20. A library consisting of elements all of which have structures according to the formula (I) or (Γ) as defined in any of claims lto 18, and hence which all have antiinflammatory activity, useful for screening compounds for novel or improved properties in a particular assay of anti-inflammatory activity.
63
P66854. WOOL Spec as filed 8.06.1 1
PCT/GB2011/000863 2010-06-08 2011-06-08 Anti-inflammatory agents WO2011154696A1 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
MA35548A MA34370B1 (en) 2010-06-08 2011-06-08 ANTI-INFLAMMATORY AGENTS
EP11725490.4A EP2580196A1 (en) 2010-06-08 2011-06-08 Anti-inflammatory agents
CA2798213A CA2798213A1 (en) 2010-06-08 2011-06-08 Anti-inflammatory agents
AP2012006559A AP2012006559A0 (en) 2010-06-08 2011-06-08 Anti-inflammatory agents
US13/702,898 US20130172318A1 (en) 2010-06-08 2011-06-08 Anti-inflammatory agents
CN201180028686XA CN103119021A (en) 2010-06-08 2011-06-08 Anti-inflammatory agents
JP2013513742A JP2013531647A (en) 2010-06-08 2011-06-08 Anti-inflammatory agent
SG2012081667A SG185129A1 (en) 2010-06-08 2011-06-08 Anti-inflammatory agents
MX2012014290A MX2012014290A (en) 2010-06-08 2011-06-08 Anti-inflammatory agents.
AU2011263531A AU2011263531A1 (en) 2010-06-08 2011-06-08 Anti-inflammatory agents
BR112012030816A BR112012030816A2 (en) 2010-06-08 2011-06-08 anti-inflammatory agents
KR1020127033497A KR20130086952A (en) 2010-06-08 2011-06-08 Anti-inflammatory agents
TNP2012000521A TN2012000521A1 (en) 2010-06-08 2012-10-31 Anti-inflammatory agents
IL222798A IL222798A0 (en) 2010-06-08 2012-11-01 Anti-inflammatory agents
ZA2012/08367A ZA201208367B (en) 2010-06-08 2012-11-07 Anti-inflammatory agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1009603.0A GB201009603D0 (en) 2010-06-08 2010-06-08 Anti-inflammatory agent
GB1009603.0 2010-06-08

Publications (1)

Publication Number Publication Date
WO2011154696A1 true WO2011154696A1 (en) 2011-12-15

Family

ID=42471347

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/GB2011/000862 WO2011154695A1 (en) 2010-06-08 2011-06-08 Anti-inflammatory agents
PCT/GB2011/000863 WO2011154696A1 (en) 2010-06-08 2011-06-08 Anti-inflammatory agents

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/GB2011/000862 WO2011154695A1 (en) 2010-06-08 2011-06-08 Anti-inflammatory agents

Country Status (20)

Country Link
US (2) US20130203734A1 (en)
EP (2) EP2580197A1 (en)
JP (2) JP2013531647A (en)
KR (2) KR20130086952A (en)
CN (2) CN103080090A (en)
AP (2) AP2012006559A0 (en)
AU (1) AU2011263531A1 (en)
BR (2) BR112012031136A2 (en)
CA (2) CA2798213A1 (en)
CL (1) CL2012003460A1 (en)
CO (2) CO6670553A2 (en)
GB (1) GB201009603D0 (en)
IL (2) IL222798A0 (en)
MA (2) MA34369B1 (en)
MX (2) MX2012014290A (en)
PE (2) PE20131046A1 (en)
SG (2) SG185130A1 (en)
TN (2) TN2012000521A1 (en)
WO (2) WO2011154695A1 (en)
ZA (2) ZA201208367B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MD4788C1 (en) * 2017-09-07 2022-09-30 Ооо "Валента-Интеллект" Use of a glutarimide derivative to treat diseases related to the aberrant activity of cytokines

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0351856A2 (en) 1988-07-22 1990-01-24 Takeda Chemical Industries, Ltd. Thiazolo[5,4-b]azepine compounds
JPH03206042A (en) 1990-01-06 1991-09-09 Takeda Chem Ind Ltd Hypotensor
EP0462949A1 (en) 1990-06-21 1991-12-27 Sigma-Tau Industrie Farmaceutiche Riunite S.p.A. 1-alkyl-3-(acylamino)-epsilon-caprolactames as enhancers of learning and memory and pharmaceutical compositions containing same
WO1999012968A2 (en) 1997-09-11 1999-03-18 Neorx Corporation Chemokine peptides, variants, derivatives and analogs, their use in methods to inhibit or augment an inflammatory response
WO2000042071A2 (en) 1999-01-12 2000-07-20 Cambridge University Technical Services Ltd. Compounds and methods to inhibit or augment an inflammatory response
JP3206042B2 (en) 1991-10-31 2001-09-04 ミノルタ株式会社 Charge transport material using novel triamino compound, photoreceptor containing this triamino compound, and electroluminescent device
WO2005053702A2 (en) 2003-12-01 2005-06-16 Cambridge University Technical Services Limited Anti-inflammatory agents
WO2006016152A1 (en) 2004-08-11 2006-02-16 Cambridge Enterprise Limited Anti-inflammatory agents
WO2006018609A2 (en) 2004-08-18 2006-02-23 Cambridge Enterprise Limited Anti-inflammatory agents
WO2006085096A1 (en) 2005-02-11 2006-08-17 Cambridge Enterprise Limited Ligands for g-protein coupled receptors
WO2006134384A1 (en) * 2005-06-15 2006-12-21 Cambridge Enterprise Limited Anti-inflammatory agents
WO2006134385A2 (en) 2005-06-15 2006-12-21 Cambridge Enterprise Limited Anti-inflammatory agents
WO2007038669A2 (en) 2005-09-27 2007-04-05 Irm Llc Diarylamine-containing compounds and compositions, and their use as modulators of c-kit receptors
WO2009017620A1 (en) 2007-07-26 2009-02-05 Ipsen Pharma S.A.S. Chemokine analogs
WO2009016390A1 (en) 2007-08-02 2009-02-05 Cambridge Enterprise Limited Anti-inflammatory composition

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4117507A1 (en) 1991-05-24 1992-11-26 Schering Ag METHOD FOR PRODUCING N (ARROW HIGH) 6 (ARROW HIGH) SUBSTITUTED LYSINE DERIVATIVES
AU2003269850A1 (en) 2002-10-08 2004-05-04 Novo Nordisk A/S Hemisuccinate salts of heterocyclic dpp-iv inhibitors
WO2004084898A1 (en) * 2003-03-24 2004-10-07 Actimis Pharmaceuticals, Inc. 2-phenoxy- and 2-phenylsulfomamide derivatives with ccr3 antagonistic activity for the treatment of asthma and other inflammatory or immunological disorders
CA2544350A1 (en) * 2003-10-29 2005-05-12 Elan Pharmaceuticals, Inc. N-substituted benzene sulfonamides
CN101035765B (en) 2004-07-13 2010-10-13 弗·哈夫曼-拉罗切有限公司 Sulfonamide derivatives
JP4885958B2 (en) 2005-08-12 2012-02-29 エフ.ホフマン−ラ ロシュ アーゲー Fluoro-substituted 2-oxoazepane derivatives

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0351856A2 (en) 1988-07-22 1990-01-24 Takeda Chemical Industries, Ltd. Thiazolo[5,4-b]azepine compounds
JPH03206042A (en) 1990-01-06 1991-09-09 Takeda Chem Ind Ltd Hypotensor
EP0462949A1 (en) 1990-06-21 1991-12-27 Sigma-Tau Industrie Farmaceutiche Riunite S.p.A. 1-alkyl-3-(acylamino)-epsilon-caprolactames as enhancers of learning and memory and pharmaceutical compositions containing same
JP3206042B2 (en) 1991-10-31 2001-09-04 ミノルタ株式会社 Charge transport material using novel triamino compound, photoreceptor containing this triamino compound, and electroluminescent device
WO1999012968A2 (en) 1997-09-11 1999-03-18 Neorx Corporation Chemokine peptides, variants, derivatives and analogs, their use in methods to inhibit or augment an inflammatory response
WO2000042071A2 (en) 1999-01-12 2000-07-20 Cambridge University Technical Services Ltd. Compounds and methods to inhibit or augment an inflammatory response
WO2005053702A2 (en) 2003-12-01 2005-06-16 Cambridge University Technical Services Limited Anti-inflammatory agents
WO2006016152A1 (en) 2004-08-11 2006-02-16 Cambridge Enterprise Limited Anti-inflammatory agents
WO2006018609A2 (en) 2004-08-18 2006-02-23 Cambridge Enterprise Limited Anti-inflammatory agents
WO2006085096A1 (en) 2005-02-11 2006-08-17 Cambridge Enterprise Limited Ligands for g-protein coupled receptors
WO2006134384A1 (en) * 2005-06-15 2006-12-21 Cambridge Enterprise Limited Anti-inflammatory agents
WO2006134385A2 (en) 2005-06-15 2006-12-21 Cambridge Enterprise Limited Anti-inflammatory agents
WO2007038669A2 (en) 2005-09-27 2007-04-05 Irm Llc Diarylamine-containing compounds and compositions, and their use as modulators of c-kit receptors
WO2009017620A1 (en) 2007-07-26 2009-02-05 Ipsen Pharma S.A.S. Chemokine analogs
WO2009016390A1 (en) 2007-08-02 2009-02-05 Cambridge Enterprise Limited Anti-inflammatory composition

Non-Patent Citations (23)

* Cited by examiner, † Cited by third party
Title
"Salt selection for basic drugs", INT. J. PHARM., vol. 33, 1986, pages 201 - 217
ANGELUCCI ET AL., J MEDICINAL CHEMISTRY, vol. 36, 1993, pages 1512 - 1519
ANGELUCCI LUCIANO ET AL: "Synthesis and amnesia-reversal activity of a series of 7- and 5-membered 3-acylamino lactams", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 36, no. 11, 1 January 1993 (1993-01-01), pages 1511 - 1519, XP002180746, ISSN: 0022-2623, DOI: DOI:10.1021/JM00063A001 *
BELYAEV A A: "A Novel Synthetic Route to Enantiomers of epsilon-Hydroxynorleucine and epsilon-Chloronorleucine from L- and D,L-Lysine", TETRAHEDRON LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 36, no. 3, 16 January 1995 (1995-01-16), pages 439 - 440, XP004028844, ISSN: 0040-4039, DOI: DOI:10.1016/0040-4039(94)02278-J *
BELYAEV, TETRAHEDRON LETTERS, vol. 36, 1995, pages 439 - 440
BOYLE ET AL., J ORG CHEM, vol. 44, 1979, pages 4841 - 4847
CHERNEY ET AL.: "Gamma-Lactams as glycinamide replacements in cyclohexane-based CC chemokine receptor 2 (CCR2) antagonists", BIOORGANIC AND MEDICINAL CHEMISTRY LETTERS, vol. 20, 10 March 2010 (2010-03-10), pages 2425 - 2430, XP002655822 *
FOX D J ET AL: "Design, Synthesis, and Preliminary Pharmacological Evaluation of N-Acyl-3-aminoglutarimides as Broad-Spectrum Chemokine Inhibitors in Vitro and Anti-inflammatory Agents in Vivo", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 45, no. 2, 1 January 2002 (2002-01-01), pages 360 - 370, XP002267550, ISSN: 0022-2623, DOI: DOI:10.1021/JM010984I *
FOX D J ET AL: "Highly potent, orally available anti-inflammatory broad-spectrum chemokine inhibitors", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 52, no. 11, 11 June 2009 (2009-06-11), pages 3591 - 3595, XP002579389, ISSN: 0022-2623, [retrieved on 20090508], DOI: DOI:10.1021/JM900133W *
FOX ET AL., J MED CHEM, vol. 45, 2002, pages 360 - 370
FOX ET AL., J MED CHEM, vol. 48, 2005, pages 867 - 74
FOX ET AL., J MED CHEM., vol. 52, no. 11, 2009, pages 3591 - 3595
GRAINGER ET AL., BIOCHEM. PHARM., vol. 65, 2003, pages 1027 - 1034
IZUMIYA ET AL.: "ACTION OF TRYPSIN AND PAPAIN ON DERIVATIVES OF DIAMINOBUTYRIC ACID, ORNITHINE AND LYSINE", THE JOURNAL OF BIOCHEMISTRY, vol. 46, no. 10, 1959, pages 1347 - 1354, XP009150957 *
KAMEDA ET AL., CHEMICAL & PHARMACEUTICAL BULLETIN, vol. 16, 1968, pages 480 - 485
KAMEDA ET AL.: "Synthesis of Actinomycin Related Compounds I", CHEMICAL AND PHARMACEUTICAL BULLETIN, vol. 16, no. 3, 1968, pages 480 - 485, XP002655823 *
OSAMU UCHIKAWA: "Aminothiazole derivatives. I. A convenient synthesis of monocyclic and condensed 5-aminothiazole derivatives", JOURNAL OF HETEROCYCLIC CHEMISTRY, WILEY-BLACKWELL PUBLISHING, INC, US, vol. 31, no. 4, 1 July 1994 (1994-07-01), pages 877 - 887, XP002137194, ISSN: 0022-152X, DOI: DOI:10.1002/JHET.5570310432 *
PAQUET A ET AL: "Synthesis of conjugates of 3-hydroxy- and 3-phenoxybenzoic acid", JOURNAL OF ENVIRONMENTAL SCIENCES AND HEALTH B: PESTICIDES, TAYLOR AND FRANCIS GROUP, XX, vol. B28, no. 2, 1 January 1993 (1993-01-01), pages 171 - 191, XP009150354, ISSN: 0360-1234 *
PELLEGATA ET AL., SYNTHESIS, 1978, pages 614 - 616
RECKLESS, BIOCHEM J., vol. 340, 1999, pages 803 - 811
UCHIKAWA ET AL., CHEMICAL & PHARMACEUTICAL BULLETIN, vol. 44, 1996, pages 2070 - 2077
UCHIKAWA ET AL., J HETEROCYCLIC CHEMISTRY, vol. 31, 1994, pages 877 - 887
UCHIKAWA O ET AL: "IN VIVO BIOLOGICAL ACTIVITY OF ANTIOXIDATIVE AMINOTHIAZOLE DERIVATIVES", CHEMICAL AND PHARMACEUTICAL BULLETIN, PHARMACEUTICAL SOCIETY OF JAPAN, TOKYO, JP, vol. 44, no. 11, 1 January 1996 (1996-01-01), pages 2070 - 2077, XP001247776, ISSN: 0009-2363 *

Also Published As

Publication number Publication date
BR112012030816A2 (en) 2016-11-01
PE20131046A1 (en) 2013-09-23
MA34369B1 (en) 2013-07-03
GB201009603D0 (en) 2010-07-21
AU2011263531A1 (en) 2012-11-22
CN103119021A (en) 2013-05-22
SG185129A1 (en) 2012-12-28
AP2012006559A0 (en) 2012-12-31
PE20130772A1 (en) 2013-07-03
WO2011154695A1 (en) 2011-12-15
EP2580196A1 (en) 2013-04-17
JP2013528201A (en) 2013-07-08
KR20130115082A (en) 2013-10-21
ZA201208406B (en) 2013-07-31
IL222798A0 (en) 2012-12-31
MX2012014290A (en) 2013-05-01
CA2798129A1 (en) 2011-12-15
US20130172318A1 (en) 2013-07-04
KR20130086952A (en) 2013-08-05
CL2012003460A1 (en) 2013-08-16
EP2580197A1 (en) 2013-04-17
CA2798213A1 (en) 2011-12-15
TN2012000521A1 (en) 2014-04-01
JP2013531647A (en) 2013-08-08
MX2012014291A (en) 2013-05-01
IL222866A0 (en) 2012-12-31
CO6670553A2 (en) 2013-05-15
CO6670569A2 (en) 2013-05-15
MA34370B1 (en) 2013-07-03
BR112012031136A2 (en) 2017-08-08
CN103080090A (en) 2013-05-01
SG185130A1 (en) 2012-12-28
AP2012006560A0 (en) 2012-12-31
ZA201208367B (en) 2013-10-30
US20130203734A1 (en) 2013-08-08
TN2012000525A1 (en) 2014-04-01

Similar Documents

Publication Publication Date Title
EP2134680B1 (en) Inhibitors of histone deacetylase
EP1385840B1 (en) Sulfonamides
CA2576257C (en) Anti-inflammatory agents
US20140336227A1 (en) 3-amino lactams as anti-inflammatory agents
EP1691814B1 (en) Anti-inflammatory agents
WO2011154696A1 (en) Anti-inflammatory agents
EP1646622B1 (en) Tacrine derivatives as inhibitors of acetylcholinesterase
OA16265A (en) Anti-inflammatory agents.
AU771745B2 (en) New aminopyrroline compounds, a process for their preparation and pharmaceutical compositions containing them
AU2011263530A1 (en) Anti-inflammatory agents
OA16264A (en) Anti-inflammatory agents.
TWI307338B (en) Novel antidiuretic agents

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180028686.X

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11725490

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2798213

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 222798

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 12012502167

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 201291025

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 2011263531

Country of ref document: AU

Date of ref document: 20110608

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 10251/DELNP/2012

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2013513742

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2012/014290

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 002286-2012

Country of ref document: PE

WWE Wipo information: entry into national phase

Ref document number: 1201006335

Country of ref document: TH

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20127033497

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: A201214820

Country of ref document: UA

WWE Wipo information: entry into national phase

Ref document number: 2011725490

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12953

Country of ref document: GE

Ref document number: 13002648

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 13702898

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112012030816

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112012030816

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20121203