WO2013153394A1 - Quinolonones with antibacterial properties - Google Patents

Quinolonones with antibacterial properties Download PDF

Info

Publication number
WO2013153394A1
WO2013153394A1 PCT/GB2013/050943 GB2013050943W WO2013153394A1 WO 2013153394 A1 WO2013153394 A1 WO 2013153394A1 GB 2013050943 W GB2013050943 W GB 2013050943W WO 2013153394 A1 WO2013153394 A1 WO 2013153394A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
alkyl
independently selected
heteroaryl
Prior art date
Application number
PCT/GB2013/050943
Other languages
French (fr)
Inventor
Neil Murray
Derek Lindsay
Ronald Palin
Mark Craighead
Original Assignee
Redx Pharma 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
Application filed by Redx Pharma Limited filed Critical Redx Pharma Limited
Publication of WO2013153394A1 publication Critical patent/WO2013153394A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/06Peri-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • 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
    • C07D401/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D401/10Heterocyclic 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 carbon chain containing aromatic rings
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/06Peri-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • This invention relates to derivatives of antibacterial drug compounds. It also relates to pharmaceutical formulations of derivatives of antibacterial drug compounds. It also relates to uses of the derivatives in treating bacterial infections and in methods of treating bacterial infections.
  • the fluoroquinolone antibacterial family are synthetic broad-spectrum antibiotics. They were originally introduced to treat Gram negative bacterial infections, but are also used for the treatment of Gram positive strains.
  • One problem with existing fluoroquinolones can be the negative side effects that may sometimes occur as a result of fluoroquinolone use. In general, the common side-effects are mild to moderate but, on occasion, more serious adverse effects occur.
  • Some of the serious side effects that occur, and which occur more commonly with fluoroquinolones than with other antibiotic drug classes, include central nervous system (CNS) toxicity and cardiotoxicity. In cases of acute overdose there may be renal failure and seizure.
  • CNS central nervous system
  • multidrug resistance has become the norm for some pathogens.
  • Staphylococcus aureus a Gram positive bacteria
  • MRSA methicillin resistant Staphylococcus aureus
  • MRSA methicillin resistant Staphylococcus aureus
  • Many strains of MRSA are also resistant to fluoroquinolones, in addition to ⁇ -lactam antibiotics such as methicillin.
  • antibiotic resistant Gram negative strains such as either E. Coli NDM- 1 (New Delhi metallo-p-lactamase) mutation or Klebsiella pneumoniae with the same mutation, are very difficult to treat, with only expensive antibiotics such as vancomycin and colistin being effective.
  • the present invention seeks to overcome the disadvantages of known fluoroquinolones.
  • antibiotics that can provide an effective treatment in a reliable manner.
  • antibiotic drugs which can avoid or reduce the side-effects associated with known antibiotics.
  • a further aim is to provide treatment which is effective in a selective manner at a chosen site of interest.
  • Another aim is to provide antibiotics having a convenient pharmacokinetic profile and a suitable duration of action following dosing.
  • a further aim of the present invention is to provide antibiotics in which the metabolised fragment or fragments of the drug after absorption are GRAS (Generally Regarded As Safe).
  • a further aim of the invention is to provide prodrugs which are not species dependent and/or which reduce inter-patient variability due to differences in metabolism.
  • Another aim of the invention is to provide prodrugs which are able to overcome the food effect in the sense that they can be administered to fed or fasted patients without the need to control carefully the dosing schedule relative to meal times.
  • the present invention satisfies some or all of the above aims.
  • X is C or N
  • Y is O or NR 3 ;
  • R 1 is independently selected from the group consisting: H, C 1-4 alkyl or Ac;
  • R 2 is independently selected from the group consisting: H, C Ce alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 - alkynyl, C Ce haloalkyi, C 3 -C 8 cydoalkyi, C 3 -C 8 heterocycloalkyl, -(CH 2 )n-C 3 -C8 cydoalkyi, - (CH 2 )n-C3-C 8 heterocycloalkyl, aryl, -(CH 2 ) n -aryl, -(CO)-aryl, -(CO)-(CH 2 ) n -aryl, heteroaryl, -(CO)- heteroaryl, -(CH 2 ) n -heteroaryl and -(CO)-(CH 2 ) n -heteroaryl; wherein each n is independently 1 , 2, 3 or 4;
  • R 3 is independently selected at each occurrence from: H, C 1-6 alkyl or Ac; or R 2 and R 3 , together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or NR 4 group;
  • R 4 is independently at each occurrence H, C 1-6 alkyl or C(0)-CrC 6 -alkyl;
  • R 5 is selected from the group consisting of: C r C 4 alkyl, C r C 4 haloalkyi, C 3 -C 5 cydoalkyi, C 3 -C 5 halocycloalkyl; unsubstituted phenyl; phenyl substituted with from 1 to 3 independently selected halogen atoms; unsubstituted pyridyl; and pyridyl substituted with from 1 to 3 independently substituents selected from the group consisting of: halo and NHR a ; wherein R a is H or Ac;
  • R 6 is selected from the group consisting of: H, C r C 4 alkyl and C r C 4 haloalkyi; or alternatively, R 5 and R 6 , together with the atoms to which they are attached to form a 4-6- membered ring which optionally contains an O or S atom; wherein the 4-6-membered ring is optionally substituted with 1 or 2 groups independently selected from halo and Ci-C 4 alkyl;
  • R 9 is selected from the group consisting: H, NHR a or C C 4 -alkyl; wherein R a is H or Ac;
  • R 10 is independently selected from the group: H or F;
  • R 11 is selected from the group consisting of: an N-heterocycloalkyl group and a C 3 -C 8 cydoalkyi group; wherein the N-heterocycloalkyl group comprises from 5 to 10 ring atoms and at least one nitrogen atom wherein the N-heterocycloalkyl group is optionally substituted with from 1-3 groups independently selected from halo, tri(C 1 -C 4 alkyl)silyloxy, hydroxyl, C C 4 alkyl, oxo or oxime and wherein any nitrogen which does not attach the N-heterocycloalkyl group to the rest of the compound of Formula (I) is an NR a group; and the C 3 -C 8 cydoalkyi group is optionally substituted with at least one NHR a group and optionally further substituted with from 1-3 groups independently selected from halo, hydroxyl, tri(C 1 -C 4 alkyl)silyloxy, C C 4 al
  • R 12 is absent or is selected from the group consisting of: H, OR 16 and halo; wherein R 16 is selected from the group consisting of: C C 4 alkyl and C C 4 haloalkyl; or R 12 and R 5 , together with the atoms to which they are attached form a saturated or unsaturated 5- to 7- membered ring which optionally contains an O, S or NR 4 group; wherein the 5-7-membered ring is optionally substituted with 1 or 2 groups independently selected from halo, C C 4 alkyl and C C 4 haloalkyl; wherein if X is N, R 12 is absent; wherein each of the aforementioned alkyl, haloalkyl, cydoalkyi, halocycloalkyl, aryl (e.g.
  • phenyl) and heteroaryl (e.g. pyridyl) groups are optionally substituted, where chemically possible, by 1 to 3 substituents which are each independently selected at each occurrence from the group consisting of: oxo, imino, oximo, halo, nitro, cyano, hydroxyl, amino, N- heterocycloalkyl, S0 3 R b , S0 2 R b , S0 2 NR b R b C0 2 R b C(0)R b , CONR b R b , C C 4 -alkyl, C C 4 haloalkyi, C C 4 alkoxy, and C C 4 haloalkoxy, wherein R is selected from H, C C 4 alkyl and C C 4 haloalkyi.
  • X is C or N
  • Y is O or NR 3 ;
  • R 1 is independently selected from the group consisting: H, C 1-4 alkyl or Ac;
  • R 2 is independently selected from the group consisting: H, C Ce alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 - alkynyl, C Ce haloalkyi, C 3 -C 8 cycloalkyi, C 3 -C 8 heterocycloalkyl, -(CH 2 )n-C 3 -C8 cycloalkyi, - (CH 2 )n-C 3 -C8 heterocycloalkyl, aryl, -(CH 2 ) n -aryl, -(CO)-aryl, heteroaryl, -(CO)-heteroaryl, and - (CH 2 )n-heteroaryl; wherein each n is independently 1 , 2, 3 or 4;
  • R 3 is independently selected at each occurrence from: H, C 1-6 alkyl or Ac; or R 2 and R 3 , together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or NR 4 group;
  • R 4 is independently at each occurrence H, C 1-6 alkyl or C(0)-CrC 6 -alkyl;
  • R 5 is selected from the group consisting of: C C 4 alkyl, C C 4 haloalkyi, C 3 -C 5 cycloalkyi, C 3 -C 5 halocycloalkyl; unsubstituted phenyl; phenyl substituted with from 1 to 3 independently selected halogen atoms; unsubstituted pyridyl; and pyridyl substituted with from 1 to 3 independently substituents selected from the group consisting of: halo and NHR a ; wherein R a is H or Ac;
  • R 6 is selected from the group consisting of: H, C C 4 alkyl and C C 4 haloalkyl;
  • R 5 and R 6 together with the atoms to which they are attached to form a 4-6- membered ring which optionally contains an O or S atom; wherein the 4-6-membered ring is optionally substituted with 1 or 2 groups independently selected from halo and C C 4 alkyl;
  • R 9 is selected from the group consisting: H, NHR a or CrC 4 -alkyl; wherein R a is H or Ac;
  • R 10 is independently selected from the group: H or F;
  • R 11 is selected from the group consisting of: an N-heterocycloalkyl group and a C 3 -C 8 cydoalkyi group; wherein the N-heterocycloalkyl group comprises from 5 to 10 ring atoms and at least one nitrogen atom wherein the N-heterocycloalkyl group is optionally substituted with from 1-3 groups independently selected from halo, tri(C 1 -C 4 alkyl)silyloxy, hydroxyl, C C 4 alkyl, oxo or oxime and wherein any nitrogen which does not attach the N-heterocycloalkyl group to the rest of the compound of Formula (I) is an NR a group; and the C 3 -C 8 cydoalkyi group is optionally substituted with at least one NHR a group and optionally further substituted with from 1-3 groups independently selected from halo, hydroxyl, tri ⁇ -C ⁇ alkyl)silyloxy, C C 4 alkyl,
  • R 12 is absent or is selected from the group consisting of: H, OR 16 and halo; wherein R 16 is selected from the group consisting of: C C 4 alkyl and C C 4 haloalkyl; wherein if X is N, R 12 is absent; wherein each of the aforementioned alkyl, haloalkyl, cydoalkyi, halocycloalkyl, aryl (e.g. phenyl) and heteroaryl (e.g.
  • pyridyl) groups are optionally substituted, where chemically possible, by 1 to 3 substituents which are each independently selected at each occurrence from the group consisting of: oxo, imino, oximo, halo, nitro, cyano, hydroxyl, amino, C0 2 H, C0 2 -(C 1 -C 4 alkyl), C(0)H, C C 4 -alkyl, C C 4 haloalkyl, C C 4 alkoxy, and C C 4 haloalkoxy.
  • the compound of formula (I) is a compound of formula (II):
  • R 12 and X are as described above.
  • the compound of formula (I) is a compound of formula
  • a derivative of an antibacterial compound selected from the group comprising: enoxacin, fleroxacin, lomefloxacin, nadifloxacin, norfloxacin, rufloxacin, balofloxacin, grepafloxacin, pazufloxacin, sparfloxacin, temafloxacin, tosufloxacin, besifloxacin, clinafloxacin, garenoxacin, gemifloxacin, gatifloxacin, sitafloxacin, trovafloxacin, prulifloxacin, ciprofloxacin, pefloxacin, moxifloxacin, ofloxacin, levofloxacin, delafloxacin and jnj-q2, in which the carboxylic acid of the antibacterial compound is replaced by a group having the following partial formula (IV):
  • Y is O or NR 3 ;
  • R 1 is independently selected from the group consisting: H, C 1-4 alkyl or Ac;
  • R 2 is independently selected from the group consisting: C Ce alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 - alkynyl, Ci-C 6 haloalkyl, C 3 -C 8 cycloalkyl, C 3 -C 8 heterocycloalkyl, -(CH 2 )n-C 3 -C8 cycloalkyl, - (CH 2 )n-C 3 -C8 heterocycloalkyl, aryl, -(CH 2 ) n -aryl, -(CO)-aryl, heteroaryl -(CO)-heteroaryl, and - (CH 2 )n-heteroaryl; wherein each n is independently 1 , 2, 3 or 4;
  • R 3 is independently selected at each occurrence from: H, C 1-6 alkyl or Ac; or R 2 and R 3 , together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or NR 4 group;
  • R 4 is independently at each occurrence H, C 1-6 alkyl or C ⁇ -C Ce-alkyl; wherein each of the aforementioned alkyl, haloalkyl, cycloalkyl, halocycloalkyl, aryl (e.g. phenyl) and heteroaryl (e.g.
  • pyridyl) groups are optionally substituted, where chemically possible, by 1 to 3 substituents which are each independently selected at each occurrence from the group consisting of: oxo, imino, oximo, halo, nitro, cyano, hydroxyl, amino, C0 2 H, C0 2 -(C 1 -C 4 alkyl), C(0)H, d-C-alkyl, C C 4 haloalkyl, C C 4 alkoxy, and C C 4 haloalkoxy; and wherein, if present, any one or more amine group in the fluoroquinolone may optionally be acetylated.
  • the antibacterial compound is selected from: fleroxacin, lomefloxacin, nadifloxacin, norfloxacin, rufloxacin, balofloxacin, grepafloxacin, pazufloxacin, sparfloxacin, temafloxacin, tosufloxacin, besifloxacin, clinafloxacin, garenoxacin, gemifloxacin, gatifloxacin, sitafloxacin, trovafloxacin, prulifloxacin, ciprofloxacin, pefloxacin, moxifloxacin, delafloxacin and jnj-q2
  • one amine group in the fluoroquinolone is acetylated.
  • the compounds of the invention are based on the parent approved pharmaceutically active compounds disclosed below.
  • the synthetic routes to each of the compounds are available in the literature and in the relevant EMA and FDA regulatory files and accordingly are not reproduced here.
  • These disclosures insofar as the synthetic procedures are concerned form part of the disclosure of the present invention. In the interests of brevity, the details of these synthetic procedures are not reproduced here but it is intended that this subject matter is specifically incorporated into the disclosure of these documents by reference. rs to a fluoroquinolone with the following structure:
  • Sensitive functional groups may need to be protected and deprotected during synthesis of a compound of the invention. This may be achieved by conventional methods, for example as described in "Protective Groups in Organic Synthesis” by TW Greene and PGM Wuts, John Wiley & Sons Inc (1999), and references therein.
  • each of the compounds of the present invention may be used as a medicament.
  • a derivative of an antibacterial compound as defined in this specification for the treatment of antibacterial infections.
  • the invention includes phamaceutical formulations comprising a compound of the invention and optionally, a pharmaceutically acceptable exceipient. Such formulations may also include other active agents.
  • the compounds and formulations of the present invention may be used in the treatment of a wide range of bacterial infections.
  • the compounds can be used to treat bacterial infections caused by one or more resistant strains of bacteria.
  • the compounds can be used to treat bacterial infections caused by one or more resistant strains of Gram positive bacteria.
  • the compounds can be used to treat bacterial infections caused by one or more resistant strains of Gram negative bacteria.
  • resistant strains is intended to mean strains of bacteria which have shown resistance to one or more known antibacterial drug. For example, it may refer to strains which are resistant to meticillin, strains that are resistant to other ⁇ -lactam antibiotics and/or strains that are resistant to fluoroquinolones which do not fall within this application.
  • a resistant strain is one in which the MIC 100 of a given compound or class of compounds for that strain has shifted to a significantly higher number than for the parent (susceptible) strain.
  • the compounds and formulations of the present invention can be used to treat or to prevent infections caused by bacterial strains associated with biowarfare. These may be strains which are category A pathogens as identified by the US government (e.g. those which cause anthrax, plague etc.) and/or they may be strains which are category B pathogens as identified by the US government (e.g. those which cause Glanders disease, mellioidosis etc).
  • the compounds and formulations of the present invention can be used to treat or to prevent infections caused by Gram positive bacterial strains associated with biowarfare (e.g. anthrax). More particularly, the compounds and formulations may be used to treat category A and/or category B pathogens as defined by the US government on 1 st April 2013.
  • the compounds and formulations of the present invention can be used to treat both Gram positive and Gram negative bacterial infections such as infections of the urinary tract, the respiratory tract, the ear, the skin, the throat, soft tissue, bone and joints (including infections caused by Staph Aureus).
  • the compounds can be used to treat pneumonia, sinusitis, acute bacterial sinusitis, bronchitis, acute bacterial exacerbation of chronic bronchitis, anthrax, chronic bacterial prostatitis, acute pyelonephritis, pharyngitis, tonsillitis, eColi, prophylaxis before dental surgery, cellulitis, acnes, cystitis, infectious diarrhoea, typhoid fever, infections caused by anaerobic bacteria, peritonitis, malaria, babesiosis bacterial vaginosis, pelvic inflammatory disease, pseudomembranous colitis, helicobacter pylori, amoebiasis, giardasis, acute gingivitis, Crohn's Disease, rosacea, fungating Tumours, MRSA, impetigo.
  • the compounds of the present invention may also be used in treating other conditions treatable by eliminating or reducing a bacterial infection.
  • the compounds of the present invention can be used in the treatment of the human body. They may be used in the treatment of the animal body. In particular, the compounds of the present invention can be used to treat commercial animals such as livestock. Alternatively, the compounds of the present invention can be used to treat companion animals such as cats, dogs, etc.
  • the parent compound upon which the derivatives of the invention are based may be selected from the group consisting of: Enoxacin, Fleroxacin, Lomefloxacin, Nadifloxacin, Norfloxacin, Rufloxacin, Balofloxacin, Grepafloxacin, Pazufloxacin, Sparfloxacin, Temafloxacin, Tosufloxacin, Besifloxacin, Clinafloxacin, Garenoxacin, Gemifloxacin, Gatifloxacin, Sitafloxacin, Trovafloxacin, Prulifloxacin, Ciprofloxacin, Pefloxacin, Moxifloxacin, Ofloxacin, jnj- q2, Levofloxacin and Delafloxacin.
  • Compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, or spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
  • Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallisation.
  • the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound of the invention contains an acidic or basic moiety, a base or acid such as 1-phenylethylamine or tartaric acid.
  • a suitable optically active compound for example, an alcohol, or, in the case where the compound of the invention contains an acidic or basic moiety, a base or acid such as 1-phenylethylamine or tartaric acid.
  • the resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person.
  • Chiral compounds of the invention may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1 % diethylamine. Concentration of the eluate affords the enriched mixture.
  • chromatography typically HPLC
  • a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1 % diethylamine.
  • racemic compounds such as the racemic compound (true racemate) referred to above wherein one homogeneous form of crystal is produced containing both enantiomers in equimolar amounts.
  • the second type is the racemic mixture or conglomerate wherein two forms of crystal are produced in equimolar amounts each comprising a single enantiomer. While both of the crystal forms present in a racemic mixture have identical physical properties, they may have different physical properties compared to the true racemate. Racemic mixtures may be separated by conventional techniques known to those skilled in the art - see, for example, "Stereochemistry of Organic Compounds" by E. L. Eliel and S. H. Wilen (Wiley, 1994).
  • the present invention provides a compound according to any one or more than one of the following formulae 1-27 taken alone or in any combination:
  • Y is O or NR ;
  • R 1 is independently selected from the group consisting: H , Ci -4 alkyl or Ac;
  • R 2 is independently selected from the group consisting: C Ce alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 - alkynyl, C Ce haloalkyl, C 3 -C 8 cycloalkyl, C 3 -C 8 heterocycloalkyl, -(CH 2 )n-C 3 -C8 cycloalkyl, - (CH 2 )n-C 3 -C8 heterocycloalkyl, aryl, -(CH 2 ) n -aryl, -(CO)-aryl, heteroaryl, -(CH 2 ) n -heteroaryl and - (CO)-heteroaryl; wherein each n is independently 1 , 2, 3 or 4;
  • R 3 is independently selected at each occurrence from: H , Ci -6 alkyl or Ac; or R 2 and R 3 , together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or N R 4 group;
  • R 4 is independently at each occurrence H, C 1-6 alkyl or C(0)-CrC 6 -alkyl; wherein each of the aforementioned alkyl, haloalkyl, cycloalkyl, halocycloalkyl, aryl (e.g. phenyl) and heteroaryl (e.g.
  • pyridyl) groups are optionally substituted, where chemically possible, by 1 to 3 substituents which are each independently selected at each occurrence from the group consisting of: oxo, imino, oximo, halo, nitro, cyano, hydroxyl, amino, C0 2 H, C0 2 -(C 1 -C 4 alkyl), C(0)H, d-C-alkyl, C C 4 haloalkyl, C C 4 alkoxy, and C C 4 haloalkoxy;
  • R a is independently at each occurrence H or Ac.
  • the compound may be selected from one, some or all of the group of compounds defined by formulae 1-27. Thus, it may be selected from a smaller group such as that defined by a single formula from within the formulae 1 to 27 (e.g. any one of formulae 4, 21 , 22, 23, 24 and 27), or from a group of compounds defined by more than one (e.g. from two to twenty) of any of the above formulae (e.g. the group defined by formulae 4, 21 , 22, 23, 24 and 27) taken together or in combination.
  • a smaller group such as that defined by a single formula from within the formulae 1 to 27 (e.g. any one of formulae 4, 21 , 22, 23, 24 and 27), or from a group of compounds defined by more than one (e.g. from two to twenty) of any of the above formulae (e.g. the group defined by formulae 4, 21 , 22, 23, 24 and 27) taken together or in combination.
  • the compound is not a compound of formula 1 , formula 26 or formula 27.
  • R 1 is H.
  • Y is NR 3 . In an alternative embodiment, Y is O.
  • R 2 is selected from the group consisting: Ci-C 6 alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 - alkynyl, CrC 6 haloalkyl, C 3 -C 8 cycloalkyl, C 3 -C 8 heterocycloalkyl, -(CH 2 )n-C 3 -C8 cycloalkyl, - (CH 2 )n-C 3 -C8 heterocycloalkyl, aryl, -(CH 2 ) n -aryl, -(CO)-aryl, heteroaryl, -(CO)-heteroaryl, and - (CH 2 )n-heteroaryl; wherein each n is independently 1 , 2, 3 or 4.
  • R 2 is H.
  • R 2 is independently selected from the group consisting: H, Ci-C 6 alkyl, C 2 - C 6 -alkenyl, C 2 -C 6 -alkynyl, CrC 6 haloalkyl, C 3 -C 8 cycloalkyl, C 3 -C 8 heterocycloalkyl, -(CH 2 ) n -C 3 - C 8 cycloalkyl, -(CH 2 ) n -C 3 -C 8 heterocycloalkyl, aryl, -(CH 2 ) n -aryl, heteroaryl, and -(CH 2 ) n - heteroaryl; wherein each n is independently 1 , 2, 3 or 4;
  • R 2 is selected from: CrC 6 alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 -alkynyl, CrC 6 haloalkyl, C 3 -C 5 cycloalkyl, aryl, -(CH 2 ) n -aryl, heteroaryl, -(CO)-heteroaryl and -(CH 2 ) n - heteroaryl.
  • R 2 is selected from: Ci-C 6 alkyl, C 2 -C 6 -alkenyl, Ci-C 6 haloalkyl, -(CO)-heteroaryl, aryl, -(CH 2 ) n -aryl and heteroaryl.
  • R 2 is selected from: C Ce alkyl, C 2 -C 6 -alkenyl, CrC 6 haloalkyl, aryl, -(CH 2 ) n -aryl and heteroaryl.
  • R 2 is C Ce alkyl. In an embodiment, R 2 is CrC 4 alkyl. Thus, R 2 may be methyl, ethyl, iso-propyl, n-propyl, iso-butyl, n-butyl, tert-butyl. In a particular embodiment, R 2 is ethyl (e.g. unsubstituted ethyl, 2-cyanoeth-1-yl or 2-hydroxyeth-1 -yl). In another, R 2 is tert- butyl. R 2 may be
  • R 2 is C 2 -C 6 alkenyl. In an embodiment, R 2 is C 3 -C 6 alkenyl. Thus, R 2 may be ally I .
  • R 2 is C Ce haloalkyl. In an embodiment, R 2 is C C 4 haloalkyl. Thus, R 2 may be trifluoromethyl or R 2 may be 1 , 1 , 1 -trifluoroeth-2-yl. In a particular embodiment, R 2 is 1 , 1 , 1-trifluoroeth-2-yl.
  • R 2 is selected from -(CO)-heteroaryl, -(CO)-aryl -(CO)-(CH 2 ) n - aryl, -(CO)-(CH 2 ) n -heteroaryl and -(CO ⁇ C Cealkyl.
  • R 2 is -(CO)-heteroaryl.
  • R 2 is -(CO)-pyridyl, e.g. -(CO)-3-pyridyl.
  • R 2 is aryl or -(CH 2 ) n -aryl.
  • R 2 may be aryl, e.g. a phenyl group optionally substituted with from 1 to 5 groups independently selected at each occurrence from: halo, nitro, cyano, hydroxyl, amino, N-heterocycloalkyl, S0 3 R B , S0 2 R B , S0 2 NR B R B C0 2 R B C(0)R B , CON R B R B , C C 4 -alkyl, C C 4 haloalkyl, C C 4 alkoxy, and C C 4 haloalkoxy.
  • substituent groups are independently selected from: halo; nitro; cyano; hydroxyl; NR 4 R 4 ; C0 2 H ; C(0)NR 4 R 4 ; C0 2 -(C C 6 alkyl); C(0)H; C C 6 -alkyl; C C 4 haloalkyl; C C 4 alkoxy; and Ci-C 6 haloalkoxy.
  • R 2 is unsubstituted phenyl.
  • R 2 is substituted phenyl.
  • R 2 is not an unsubstituted phenyl ring.
  • R 2 may be mono substituted or it may be disubstituted or it may be trisubstituted.
  • the phenyl in embodiments in which the phenyl is substituted, it may be substituted with at least one electron-withdrawing group. In embodiments in which the phenyl is substituted, it may be substituted with at least 1 group selected from the group consisting of: halo, nitro, S0 3 R B , S0 2 R B , S0 2 N R B R B C0 2 R B C(0)R B , CON R B R B , cyano, and C C 4 haloalkyl.
  • R 2 may be 2,6-dichloro-3-trifluoromethylphenyl.
  • R 2 may be a phenyl group substituted with one group selected from the group consisting of: halo, nitro, cyano, CO ⁇ CrCealkyl), and C C 4 haloalkyl.
  • R 2 may be a halophenyl group.
  • R 2 may be a chlorophenyl group, e.g. a 2-chlorophenyl or a 3-chlorophenyl or a 4-chlorophenyl group.
  • R 2 may be a fluorophenyl group, e.g. a 2- fluorophenyl or a 3-fluorophenyl or 4-fluorophenyl group.
  • R 2 may be a bromophenyl group, e.g.
  • R 2 may be a nitrophenyl group, e.g. a 2-nitrophenyl or a 3- nitrophenyl or a 4- nitrophenyl group.
  • R 2 may be a para- or meta- substituted phenyl group. This embodiment applies equally to monosubstituted rings, to bisubstituted rings and to trisubstituted phenyl rings. In other words, it may be that if R 2 is a substituted phenyl group it is not ortho substituted. In a particular embodiment, R 2 may be a mono- para- or meta substituted phenyl group.
  • the single substituent may be an electron withdrawing group e.g.
  • the single substituent may be any one of or may be selected from a group consisting of any more than one of: C0 2 H, S0 3 H, S0 2 Me, CN, S0 2 NH 2 , F, CI, Br, N0 2 , OCF 3 , CF 3 , CONMe 2 , CONHMe, CONH 2 .
  • the single substituent may also be any one of or may be selected from a group consisting of any more than one of: methyl, OMe, isopropyl, f-butyl.
  • R 2 may also be -(CH 2 ) n -aryl.
  • the integer n may be 1 or 2, e.g. 1 .
  • R 2 may be benzyl in which the aryl ring is optionally substituted with from 1 to 5 groups independently selected at each occurrence from: halo; nitro; cyano; hydroxyl; NR 4 R 4 ; C0 2 H; C(0)NR 4 R 4 ; C0 2 -(C C 6 alkyl); C(0)H; C C 6 -alkyl; C C 4 haloalkyl; C C 4 alkoxy; and C C 6 haloalkoxy.
  • R 2 is benzyl.
  • R 2 is pentafluorobenzyl.
  • R2 may be fluorobenzyl (e.g. 4-fluorophenyl)
  • R 2 is heteroaryl or C ⁇ Ce alkylheteroaryl.
  • R 2 may be a heteroaryl group, e.g. a monocyclic heteroaryl group.
  • R 2 may be a 6-membered heteroaryl group containing 1-3-nitrogen atoms.
  • R 2 may be pyridine, pyridazine, pyrimidine or pyrazine.
  • R 2 may be pyridine (e.g. 2-pyridine) or R 2 may be pyrimidine (e.g. 2-pyrimidine).
  • the heteroaryl ring is optionally substituted with from 1 to 5 groups independently selected at each occurrence from: halo, nitro, cyano, hydroxyl, amino, N-heterocycloalkyl, S0 3 R b , S0 2 R b , S0 2 NR b R b C0 2 R b C(0)R b , CONR b R b , C r C 4 -alkyl, C C 4 haloalkyl, C C 4 alkoxy, and C C 4 haloalkoxy.
  • substituent groups are independently selected from: halo; nitro; cyano; hydroxyl; NR 4 R 4 ; C0 2 H; C(0)NR 4 R 4 ; C0 2 -(C 1 -C 6 alkyl); C(0)H; C C 6 -alkyl; C C 4 haloalkyl; C C 4 alkoxy; and C C 6 haloalkoxy.
  • the heteroaryl may be unsubstituted.
  • R 2 and R 3 together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or NR 4 group.
  • R 2 and R 3 together with the nitrogen to which they are attached form a 6- membered ring which optionally contains an O, S or NR 4 group.
  • R 2 and R 3 together with the nitrogen to which they are attached form a 6- membered ring, e.g. a piperidine ring.
  • the heterocyclic ring formed by R 2 , R 3 and the nitrogen to which they are attached is optionally substituted with from 1 to 5 groups independently selected at each occurrence from: halo; nitro; cyano; hydroxyl; NR 4 R 4 ; C0 2 H; C(0)NR 4 R 4 ; CC CCrCealkyl); C(0)H; C Ce-alkyl; C C 4 haloalkyl; C C 4 alkoxy; and C Ce haloalkoxy.
  • the ring may be unsubstituted.
  • R 2 and R 3 together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or NR 4 group.
  • R 3 is H.
  • each R 4 is H.
  • X is N. In an alternative embodiment, X is C.
  • R 9 is H.
  • R 9 is NHR a .
  • R 9 is C C 4 alkyl, e.g. R 9 may be methyl, ethyl, propyl, butyl, iso-propyl or tert-butyl. In a particular embodiment, R 9 is methyl.
  • R 10 is H. In an alternative embodiment R 10 is F.
  • R 11 is a N-heterocycloalkyl group comprising from 5 to 10 ring atoms wherein at least one of the ring atoms is an NR a group wherein the N-heterocycloalkyl group is optionally substituted with from 1 -3 groups independently selected from halo, hydroxyl, C C 4 alkyl, oxo, oxime comprising from 5 to 10 ring atoms and at least one nitrogen atom which is optionally substituted with from 1 -3 groups independently selected from halo, hydroxyl, C C 4 alkyl, oxo, oxime.
  • the N-heterocycloalkyl group is attached to the remainder of the compound of Formula (I) via the N-atom.
  • R 11 is a piperazine ring which is optionally substituted with from 1-3 groups independently selected from halo, hydroxyl, C C 4 alkyl, oxo or oxime.
  • R 11 is a piperazine ring which is optionally substituted with from 1-3 independently selected C C 4 alkyl groups.
  • R 11 is a piperazine ring substituted with a methyl group, e.g. an N-methyl piperazine ring.
  • R 11 is a 3-methyl piperazine ring.
  • R 11 may be a 2-methyl piperazine ring.
  • R 11 is a piperazine ring which is optionally substituted with a C 1-4 alkanoyl group.
  • the R 11 group is an acetyl piperazine group.
  • R 11 is a piperidine ring which is optionally substituted with from 1-3 groups independently selected from halo, hydroxyl, Ci-C 4 alkyl, oxo, oxime.
  • the NH moiety in the structure depicted may alternatively be substituted with an acetyl group.
  • R 11 is C 3 -C 8 cycloalkyl group substituted with at least one NHR a group and optionally further substituted with from 1 -3 groups independently selected from halo, hydroxyl, C C 4 alkyl, oxo or oxime.
  • R 11 is a cyclopropyl group substituted with a NHR a group.
  • R 12 is H. In an alternative embodiment, R 12 is CI or F. In a further alternative, R 12 is OR 16 . In an embodiment, R 16 is C C 4 alkyl, e.g. R 16 may be methyl. In an alternative embodiment, R 16 is C C 4 haloalky, e.g. C C 4 fluoroalkyl such as trifluoromethyl or difluoromethyl.
  • R 12 and R 5 together with the atoms to which they are attached, form a 6-membered ring which optionally contains an O or S atom; wherein the 6-membered ring is optionally substituted with 1 or 2 groups independently selected from halo and C C 4 alkyl.
  • the ring contains an O atom.
  • the ring contains a S atom.
  • the ring may be substituted with a Ci-C 4 alkyl group, e.g. a methyl group.
  • R 5 is C C 4 alkyl, e.g. ethyl.
  • R 5 is C C 4 haloalkyi, e.g. a 2-fluoroethyl group.
  • R 5 is C 3 -C 5 cycloalkyl, e.g. cyclopropyl.
  • R 5 is C 3 -C 5 halocycloalkyl, e.g.fluoro cyclopropyl.
  • R 5 is phenyl which may be optionally substituted with from 1-3 halo groups, i.e. R 5 may be a difluorophenyl group, such as 2,4-difluorophenyl.
  • R 6 is H.
  • the compound of formula (I) is not a compound selected from:
  • the present invention provides a pharmaceutical formulation comprising a compound of the invention and a pharmaceutically acceptable excipient.
  • Aryl groups may be 6-membered aryl groups.
  • Aryl groups may be optionally substituted phenyl groups, optionally substituted biphenyl groups, optionally substituted naphthalenyl groups or optionally substituted anthracenyl groups.
  • Heteroaryl groups may be 5- or 6-membered heteroaryl groups. They may be monocyclic heteroaryl groups, (e.g. heteroaryl groups may be selected from: 5-membered heteroaryl groups in which the heteroaromatic ring is substituted with 1 -3 heteroatoms selected from O, S and N; and 6-membered heteroaryl groups in which the heteroaromatic ring is substituted with 1-2 nitrogen atoms) or they may be bicyclic heteroaryl groups (9-membered bicyclic heteroaryl groups in which the heteroaromatic system is substituted with 1 -4 heteroatoms selected from O, S and N; 10-membered bicyclic heteroaryl groups in which the heteroaromatic system is substituted with 1-4 nitrogen atoms).
  • heteroaryl groups may be selected from: 5-membered heteroaryl groups in which the heteroaromatic ring is substituted with 1 -3 heteroatoms selected from O, S and N; and 6-membered heteroaryl groups in which the heteroaromatic ring is substituted
  • heteroaryl groups may be selected from: pyrrole, furan, thiophene, pyrazole, imidazole, oxazole, isoxazole, triazole, oxadiazole, thiodiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine (e.g. 1 ,3,5-triazine or 1 ,3,4 triazine), indole, isoindole, benzofuran, isobenzofuran, benzothiophene, indazole,
  • benzimidazole benzoxazole, benzthiazole, benzisoxazole, purine, quinoline, isoquinoline, cinnoline, quinazoline, quinoxaline, pteridine, phthalazine, naphthyridine.
  • Heterocycloalkyi groups of the invention may be from 3-8 membered saturated rings in which the ring contains from 1 to 3 heteroatoms (e.g. O, N or S). Where the heterocycloalkyi group contains a nitrogen, the heterocycloalkyi group may be linked to the remainder of the molecule via the nitrogen or via one of the carbon atoms in the ring. Where the heterocycloalkyi group does not contain a nitrogen, the heterocycloalkyi group will be linked to the rest of the molecule via one of the carbon atoms in the ring.
  • heteroatoms e.g. O, N or S
  • the aryl and heteroaryl groups are optionally substituted with from 1 to 4 groups independently selected at each occurrence from: halo, nitro, cyano, hydroxyl, NHR a , C0 2 H, C0 2 -(CrC 4 alkyl), C(0)H, C C4-alkyl, C C 4 haloalkyl, C C 4 alkoxy, and C C 4 haloalkoxy.
  • the present invention also includes the synthesis of all pharmaceutically acceptable isotopically-labelled compounds of the invnetion wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • the dosage administered will, of course, vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated.
  • the daily dosage of the compound of the invention may be in the range from 0.01 micrograms per kilogram body weight ⁇ g/kg) to 100 milligrams per kilogram body weight (mg/kg).
  • a compound of the invention, or pharmaceutically acceptable salt thereof may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the compounds of the invention, or pharmaceutically acceptable salt thereof, is in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • a pharmaceutically acceptable adjuvant diluent or carrier.
  • Conventional procedures for the selection and preparation of suitable pharmaceutical formulations are described in, for example, "Pharmaceuticals - The Science of Dosage Form Designs", M. E. Aulton, Churchill Livingstone, 1988.
  • the pharmaceutical composition which is used to administer the compounds of the invention will preferably comprise from 0.05 to 99 %w (per cent by weight) compounds of the invention, more preferably from 0.05 to 80 %w compounds of the invention, still more preferably from 0.10 to 70 %w compounds of the invention, and even more preferably from 0.10 to 50 %w compounds of the invention, all percentages by weight being based on total composition.
  • compositions may be administered topically (e.g. to the skin) in the form, e.g., of creams, gels, lotions, solutions, suspensions, or systemically, e.g. by oral
  • a compound with an in vitro MIC100 of, for example, 16-64 may still provide an effective treatment against bacteria.
  • the compounds of the invention may be admixed with an adjuvant or a carrier, for example, lactose, saccharose, sorbitol, mannitol; a starch, for example, potato starch, corn starch or amylopectin; a cellulose derivative; a binder, for example, gelatine or polyvinylpyrrolidone; and/or a lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the like, and then compressed into tablets.
  • an adjuvant or a carrier for example, lactose, saccharose, sorbitol, mannitol
  • a starch for example, potato starch, corn starch or amylopectin
  • a cellulose derivative for example, gelatine or polyvinylpyrrolidone
  • a lubricant for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax
  • the cores may be coated with a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide.
  • a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide.
  • the tablet may be coated with a suitable polymer dissolved in a readily volatile organic solvent.
  • the compounds of the invention may be admixed with, for example, a vegetable oil or polyethylene glycol.
  • Hard gelatine capsules may contain granules of the compound using either the above-mentioned excipients for tablets.
  • liquid or semisolid formulations of the compound of the invention may be filled into hard gelatine capsules.
  • Liquid preparations for oral application may be in the form of syrups or suspensions, for example, solutions containing the compound of the invention, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol.
  • such liquid preparations may contain colouring agents, flavouring agents, sweetening agents (such as saccharine), preservative agents and/or carboxymethylcellulose as a thickening agent or other excipients known to those skilled in art.
  • the compounds of the invention may be administered as a sterile aqueous or oily solution.
  • the size of the dose for therapeutic purposes of compounds of the invention will naturally vary according to the nature and severity of the conditions, the age and sex of the animal or patient and the route of administration, according to well known principles of medicine
  • Dosage levels, dose frequency, and treatment durations of compounds of the invention are expected to differ depending on the formulation and clinical indication, age, and co-morbid medical conditions of the patient.
  • the standard duration of treatment with compounds of the invention is expected to vary between one and seven days for most clinical indications. It may be necessary to extend the duration of treatment beyond seven days in instances of recurrent infections or infections associated with tissues or implanted materials to which there is poor blood supply including bones/joints, respiratory tract, endocardium, and dental tissues.
  • isotopes suitable for inclusion in the compounds of the invention include isotopes
  • Radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e. 14 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
  • substitution with heavier isotopes such as deuterium, i.e. 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.
  • Isotopically-labelled compounds can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described using an appropriate isotopically-labelled reagent in place of the non-labelled reagent previously employed.
  • reaction mixture (0.05ml_, 0.5500mmol) at 0°C under Ar. This was allowed to stir at that temperature for 5 minutes. After which time the reaction mixture was allowed to warm to room temperature and hydrazine (0.5500mmol) was added in one portion. This was then allowed to stir at that temperature for 18 hours. After which time the reaction mixture was directly purified by automated flash chromatography (Grace Reveleris®) using an eluent system of DCM:MeOH.
  • the compound of the invention is any one or is selected from any group more than one of the following exemplary compounds:
  • Compound AG was prepared using method A.
  • Compound AR was prepared using method A.
  • (AS) ⁇ 1.5-1.6 (m,3H), 2.35 (s, 3H), 2.5-2.6 (m, 4H), 3.3-3.5 (m, 4H), 3.7 (s, 1H), 4.2-4.6 (m, 3H), 6.8 (m, 1H), 6.9 (m,1H), 7.0 (m, 1H), 7.3-7.5 (d, 1H), 8.2 (s, 1H), 8.3-8.6 (m, 2H), 12.0 (s, 1H).
  • Compound AS was prepared using method A.
  • Compound BD was prepared using method B.
  • Compound BJ was prepared using method B.
  • Compound BR was prepared using method B.
  • Compound BZ was prepared using method B.
  • Compound CF was prepared using method B. (CG) ⁇ 1 1.5 (s, 1 H), 8.6 (s, 1 H), 8.0 (s, 1 H), 7.10-7.65 (m, 3H), 4.4-4.2 (m, 4H), 3.35 (m, 4H), 2.50 (m, 4H), 2.4 (s, 3H), 1.5 (d, 3H). Compound CG was prepared using method B.
  • the inocula for each bacterial strain were prepared by picking 5-10 distinct colonies from the culture plates and suspending them in 3ml of sterile saline. The inoculum was resuspended by vigorous shaking on a vortex mixer for 15s. The turbidity was then adjusted to McFarland standard 0.5 (1 -5 x 106 CFU/ml). The inoculum was further diluted in Mueller Hinton Broth for MIC tests to give a final inoculum in each well of 2-8 x 105 CFU/ml. For Streptococcus pneumoniae the Mueller Hinton broth was supplemented with 5% lysed horse blood (MHLB).
  • MHLB 5% lysed horse blood
  • a stock solution was prepared at a concentration of 1.28 g/L in DMSO. The stock was further diluted in Mueller Hinton broth (or MHLB) to give a top starting concentration of 128 mg/L in the assay.
  • a comparator control was included for each strain. The final concentration range for the comparator control (ciprofloxacin) was 0.03 -16 ⁇ / ⁇ _.100 ⁇ _ of Mueller Hinton broth was dispensed into each well in columns 2-12. 200 ⁇ _ of the appropriate test compound solution (at 256mg/L) was dispensed into each well in column 1.
  • b A comparator control
  • Table 1 MIC 50 for selected compounds and levofloxacin against non-resistant (susceptible) strains of Gram negative and Gram positive bacteria (indicated by a + or a - in parentheses after the species name)
  • Table 2 MIC 10 o for selected compounds against a variety of resistant strains and non-resistant (susceptible) strains of Gram positive and Gram negative bacteria.
  • MICs were determined in MH broth in accordance with Clinical and Laboratory Standards Institute guidelines M07-A9 for aerobic bacteria, other than using flat-bottomed wells to allow spectrophotometric reading of MIC assays.
  • MH broth was prepared according to the manufacturer's instructions and either left unmodified, or buffered as detailed below. All broths were then filter-sterilised through a 0.22 ⁇ filter before use. Unbuffered MH broth was found to have a pH of 7.18. • For MH broth pH 6.2, 3-(N-morpholino)propanesulfonic acidacid (MOPS) was added to a final concentration of 20 mM, and the pH adjusted with 1 M HCI.
  • MOPS 3-(N-morpholino)propanesulfonic acidacid
  • HEPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid
  • Assay plates were incubated aerobically at 37°C for 18-20 h. Following incubation, plates were assessed both visually and spectrophotometrically at 490 nm.
  • Example 6 shows that there can be expected to be a considerable degree of activity variation in the activity at certain pHs for any given compound.
  • some compounds of the invention may be more active at low pH infection sites such as open wounds, whereas others may be more active at high pH infection sites such as urinary tract infections.
  • the following study demonstrates that compound O, which was shown in Example 4 to have low activity at low pH, does as expected have very little activity in acidic wound sites.
  • Sprague-Dawley male rats 225-250g (at delivery) were used in these studies which were supplied by Charles River UK Ltd and were specific pathogen free. Following receipt at our facility rats were allowed to acclimatise for 5-7 days (weigh at the start of the experiment ⁇ 250- 275g).
  • Rats were housed in sterilized individual ventilated cages that expose the animals at all times to HEPA filtered sterile air. Rats had free access to food and water (sterile) and were provided with sterile aspen chip bedding (changed every 3-4 days). Additionally, during infection, rats had additional access to wet food if required to ensure they remained fully hydrated. The room temperature was 22°C +/- 1°C, with a relative humidity of 60% and maximum background noise of 56dB. Rats were exposed to 12 hour light/dark cycles.
  • Staphylococcus aureus S. aureus ATCC 29213 (MSSA), a well characterised strain, was used throughout the study.
  • aureus ATCC 29213 by intramuscular injection of 0.1 mL of bacteria into both lateral thigh muscles under temporary inhaled anaesthesia using ⁇ 6.54x10 6 cfu/rat thigh. Rats were administered buprenorphine at ⁇ 0.03 mg/kg subcutaneously immediately post infection and 12 hours post infection for pain relief (buprenorphine causes delayed gastric emptying that can effect the PK following oral administration).
  • Table 7 shows the study treatment groups, treatment regimen and harvest (kill) time points. A total of 19 rats were used in the study (4 per time point plus 3 untreated controls).
  • Compound O was reconstituted freshly at each time point for immediate dosing in dimethyl sulfoxide (DMSO) giving a stock of 100mg/ml_, the DMSO stock was then diluted 1 :50 and 1 : 10 with water for injection (WFI) to provide dosing solutions at 2mg/ml_ and 10mg/ml_ respectively.
  • WFI water for injection
  • Dosing solutions were delivered at 10ml_/kg orally in order to achieve 20mg/kg and 100mg/kg per dose.
  • Vehicle solution (10% Dimethyl sulfoxide (DMSO), WFI, HCI), was prepared in the same manner minus the test article and delivered by oral administration at 10ml_/kg.
  • Comparator Levofloxacin (Tavanic-5mg/ml_, Sanofi Aventis, UK) was also diluted in water for injection to 2mg/ml_ and delivered orally at 10ml_/kg in order to achieve 20mg/kg per dose. Following reconstitution, all test article solutions remained clear and non-particulate for the duration of the dosing period.
  • Antibacterial treatment was initiated 1 hour post infection and delivered twice by oral gavage at 10 mL/kg (2.5ml_ per 250g rat) (see Table 1). All dosing solutions were well tolerated when administered via oral gavage at 10ml_/kg.
  • Table 7 Treatment groups, treatment regime, time of tissue harvest and number of rats per group.
  • the compounds of the invention are active in the treatment of one or more of the above strains.
  • the activity is broad spectrum. In other cases the activity is selective over Gram-negative bacteria or Gram positive bacteria. In yet further cases the activity is selective over one or more strains.
  • the compounds are active against bacterial strains associated with biowarfare, particularly Gram positive strains.

Abstract

This invention relates to derivatives of fluoroquinolone antibacterial drug compounds. It also relates to pharmaceutical formulations of derivatives of fluoroquinolone antibacterial drug compounds. It also relates to uses of the derivatives in treating bacterial infections and in methods of treating bacterial infections.

Description

QUINOLONONES WITH ANTIBACTERIAL PROPERTIES
This invention relates to derivatives of antibacterial drug compounds. It also relates to pharmaceutical formulations of derivatives of antibacterial drug compounds. It also relates to uses of the derivatives in treating bacterial infections and in methods of treating bacterial infections.
The fluoroquinolone antibacterial family are synthetic broad-spectrum antibiotics. They were originally introduced to treat Gram negative bacterial infections, but are also used for the treatment of Gram positive strains. One problem with existing fluoroquinolones can be the negative side effects that may sometimes occur as a result of fluoroquinolone use. In general, the common side-effects are mild to moderate but, on occasion, more serious adverse effects occur. Some of the serious side effects that occur, and which occur more commonly with fluoroquinolones than with other antibiotic drug classes, include central nervous system (CNS) toxicity and cardiotoxicity. In cases of acute overdose there may be renal failure and seizure.
A particular problem, both with antibiotics in general and with fluoroquinolones in particular, is the increasing frequency of resistant strains of bacterial pathogens such as Staphylococcus aureus, Streptococcus pneumonia, Clostridium difficile and Pseudomonas aeruginosa. In fact, multidrug resistance has become the norm for some pathogens. Of these, Staphylococcus aureus, a Gram positive bacteria, is the most concerning due to its potency and its capacity to adapt to environmental conditions. MRSA (methicillin resistant Staphylococcus aureus) is probably the most well known and has reached pandemic proportions. Of particular concern is the increasing incidence of 'community acquired' infections, those occurring in subjects with no prior hospital exposure. Many strains of MRSA are also resistant to fluoroquinolones, in addition to β-lactam antibiotics such as methicillin.
While less wide-spread, antibiotic resistant Gram negative strains, such as either E. Coli NDM- 1 (New Delhi metallo-p-lactamase) mutation or Klebsiella pneumoniae with the same mutation, are very difficult to treat, with only expensive antibiotics such as vancomycin and colistin being effective.
The present invention seeks to overcome the disadvantages of known fluoroquinolones. In spite of the numerous different antibiotics known in the art for a variety of different infections, there continues to be a need to provide antibiotics that can provide an effective treatment in a reliable manner. In addition, there remains a need for antibiotic drugs which can avoid or reduce the side-effects associated with known antibiotics. A further aim is to provide treatment which is effective in a selective manner at a chosen site of interest. Another aim is to provide antibiotics having a convenient pharmacokinetic profile and a suitable duration of action following dosing.
It is an aim of this invention to provide new antibiotics. In particular it is an aim to provide antibiotics which are active against resistant strains of Gram positive and/or Gram negative bacteria. It is also an aim to provide antibiotics which are active against strains of bacteria which may be associated with biowarfare. Such strains are typified by high mortality and/or rapid onset of disease.
A further aim of the present invention is to provide antibiotics in which the metabolised fragment or fragments of the drug after absorption are GRAS (Generally Regarded As Safe). A further aim of the invention is to provide prodrugs which are not species dependent and/or which reduce inter-patient variability due to differences in metabolism. Another aim of the invention is to provide prodrugs which are able to overcome the food effect in the sense that they can be administered to fed or fasted patients without the need to control carefully the dosing schedule relative to meal times.
The present invention satisfies some or all of the above aims.
According to a first aspect of the present invention, there is provided a compound of formula (I):
Figure imgf000004_0001
X is C or N;
Y is O or NR3;
R1 is independently selected from the group consisting: H, C1-4 alkyl or Ac;
R2 is independently selected from the group consisting: H, C Ce alkyl, C2-C6-alkenyl, C2-C6- alkynyl, C Ce haloalkyi, C3-C8 cydoalkyi, C3-C8 heterocycloalkyl, -(CH2)n-C3-C8 cydoalkyi, - (CH2)n-C3-C8 heterocycloalkyl, aryl, -(CH2)n-aryl, -(CO)-aryl, -(CO)-(CH2)n-aryl, heteroaryl, -(CO)- heteroaryl, -(CH2)n-heteroaryl and -(CO)-(CH2)n-heteroaryl; wherein each n is independently 1 , 2, 3 or 4;
R3 is independently selected at each occurrence from: H, C1-6 alkyl or Ac; or R2 and R3, together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or NR4 group;
R4 is independently at each occurrence H, C1-6 alkyl or C(0)-CrC6-alkyl;
R5 is selected from the group consisting of: CrC4 alkyl, CrC4 haloalkyi, C3-C5 cydoalkyi, C3-C5 halocycloalkyl; unsubstituted phenyl; phenyl substituted with from 1 to 3 independently selected halogen atoms; unsubstituted pyridyl; and pyridyl substituted with from 1 to 3 independently substituents selected from the group consisting of: halo and NHRa; wherein Ra is H or Ac;
R6 is selected from the group consisting of: H, CrC4 alkyl and CrC4 haloalkyi; or alternatively, R5 and R6, together with the atoms to which they are attached to form a 4-6- membered ring which optionally contains an O or S atom; wherein the 4-6-membered ring is optionally substituted with 1 or 2 groups independently selected from halo and Ci-C4 alkyl;
R9 is selected from the group consisting: H, NHRa or C C4-alkyl; wherein Ra is H or Ac;
R10 is independently selected from the group: H or F;
R11 is selected from the group consisting of: an N-heterocycloalkyl group and a C3-C8 cydoalkyi group; wherein the N-heterocycloalkyl group comprises from 5 to 10 ring atoms and at least one nitrogen atom wherein the N-heterocycloalkyl group is optionally substituted with from 1-3 groups independently selected from halo, tri(C1-C4 alkyl)silyloxy, hydroxyl, C C4 alkyl, oxo or oxime and wherein any nitrogen which does not attach the N-heterocycloalkyl group to the rest of the compound of Formula (I) is an NRa group; and the C3-C8 cydoalkyi group is optionally substituted with at least one NHRa group and optionally further substituted with from 1-3 groups independently selected from halo, hydroxyl, tri(C1-C4 alkyl)silyloxy, C C4 alkyl, oxo or oxime; wherein Ra is H or Ac;
R12 is absent or is selected from the group consisting of: H, OR16 and halo; wherein R16 is selected from the group consisting of: C C4 alkyl and C C4 haloalkyl; or R12 and R5, together with the atoms to which they are attached form a saturated or unsaturated 5- to 7- membered ring which optionally contains an O, S or NR4 group; wherein the 5-7-membered ring is optionally substituted with 1 or 2 groups independently selected from halo, C C4 alkyl and C C4 haloalkyl; wherein if X is N, R12 is absent; wherein each of the aforementioned alkyl, haloalkyl, cydoalkyi, halocycloalkyl, aryl (e.g. phenyl) and heteroaryl (e.g. pyridyl) groups are optionally substituted, where chemically possible, by 1 to 3 substituents which are each independently selected at each occurrence from the group consisting of: oxo, imino, oximo, halo, nitro, cyano, hydroxyl, amino, N- heterocycloalkyl, S03Rb, S02Rb, S02NRbRbC02Rb C(0)Rb, CONRbRb, C C4-alkyl, C C4 haloalkyi, C C4 alkoxy, and C C4 haloalkoxy, wherein R is selected from H, C C4 alkyl and C C4 haloalkyi.
In an embodiment, there is provided a compound of formula
Figure imgf000006_0001
; wherein
X is C or N;
Y is O or NR3;
R1 is independently selected from the group consisting: H, C1-4 alkyl or Ac;
R2 is independently selected from the group consisting: H, C Ce alkyl, C2-C6-alkenyl, C2-C6- alkynyl, C Ce haloalkyi, C3-C8 cycloalkyi, C3-C8 heterocycloalkyl, -(CH2)n-C3-C8 cycloalkyi, - (CH2)n-C3-C8 heterocycloalkyl, aryl, -(CH2)n-aryl, -(CO)-aryl, heteroaryl, -(CO)-heteroaryl, and - (CH2)n-heteroaryl; wherein each n is independently 1 , 2, 3 or 4;
R3 is independently selected at each occurrence from: H, C1-6 alkyl or Ac; or R2 and R3, together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or NR4 group;
R4 is independently at each occurrence H, C1-6 alkyl or C(0)-CrC6-alkyl;
R5 is selected from the group consisting of: C C4 alkyl, C C4 haloalkyi, C3-C5 cycloalkyi, C3-C5 halocycloalkyl; unsubstituted phenyl; phenyl substituted with from 1 to 3 independently selected halogen atoms; unsubstituted pyridyl; and pyridyl substituted with from 1 to 3 independently substituents selected from the group consisting of: halo and NHRa; wherein Ra is H or Ac;
R6 is selected from the group consisting of: H, C C4 alkyl and C C4 haloalkyl;
or alternatively, R5 and R6, together with the atoms to which they are attached to form a 4-6- membered ring which optionally contains an O or S atom; wherein the 4-6-membered ring is optionally substituted with 1 or 2 groups independently selected from halo and C C4 alkyl;
R9 is selected from the group consisting: H, NHRa or CrC4-alkyl; wherein Ra is H or Ac;
R10 is independently selected from the group: H or F;
R11 is selected from the group consisting of: an N-heterocycloalkyl group and a C3-C8 cydoalkyi group; wherein the N-heterocycloalkyl group comprises from 5 to 10 ring atoms and at least one nitrogen atom wherein the N-heterocycloalkyl group is optionally substituted with from 1-3 groups independently selected from halo, tri(C1-C4 alkyl)silyloxy, hydroxyl, C C4 alkyl, oxo or oxime and wherein any nitrogen which does not attach the N-heterocycloalkyl group to the rest of the compound of Formula (I) is an NRa group; and the C3-C8 cydoalkyi group is optionally substituted with at least one NHRa group and optionally further substituted with from 1-3 groups independently selected from halo, hydroxyl, tri^-C^ alkyl)silyloxy, C C4 alkyl, oxo or oxime; wherein Ra is H or Ac;
R12 is absent or is selected from the group consisting of: H, OR16 and halo; wherein R16 is selected from the group consisting of: C C4 alkyl and C C4 haloalkyl; wherein if X is N, R12 is absent; wherein each of the aforementioned alkyl, haloalkyl, cydoalkyi, halocycloalkyl, aryl (e.g. phenyl) and heteroaryl (e.g. pyridyl) groups are optionally substituted, where chemically possible, by 1 to 3 substituents which are each independently selected at each occurrence from the group consisting of: oxo, imino, oximo, halo, nitro, cyano, hydroxyl, amino, C02H, C02-(C1-C4alkyl), C(0)H, C C4-alkyl, C C4 haloalkyl, C C4 alkoxy, and C C4 haloalkoxy. In an embodiment, the compound of formula (I) is a compound of formula (II):
Figure imgf000008_0001
R12 and X are as described above.
In an embodiment, the compound of formula (I) is a compound of formula
Figure imgf000008_0002
X are as described above.
In an embodiment, there is provided a derivative of an antibacterial compound selected from the group comprising: enoxacin, fleroxacin, lomefloxacin, nadifloxacin, norfloxacin, rufloxacin, balofloxacin, grepafloxacin, pazufloxacin, sparfloxacin, temafloxacin, tosufloxacin, besifloxacin, clinafloxacin, garenoxacin, gemifloxacin, gatifloxacin, sitafloxacin, trovafloxacin, prulifloxacin, ciprofloxacin, pefloxacin, moxifloxacin, ofloxacin, levofloxacin, delafloxacin and jnj-q2, in which the carboxylic acid of the antibacterial compound is replaced by a group having the following partial formula (IV):
Figure imgf000008_0003
wherein:
Y is O or NR3;
R1 is independently selected from the group consisting: H, C1-4 alkyl or Ac;
R2 is independently selected from the group consisting: C Ce alkyl, C2-C6-alkenyl, C2-C6- alkynyl, Ci-C6 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, -(CH2)n-C3-C8 cycloalkyl, - (CH2)n-C3-C8 heterocycloalkyl, aryl, -(CH2)n-aryl, -(CO)-aryl, heteroaryl -(CO)-heteroaryl, and - (CH2)n-heteroaryl; wherein each n is independently 1 , 2, 3 or 4;
R3 is independently selected at each occurrence from: H, C1-6 alkyl or Ac; or R2 and R3, together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or NR4 group;
R4 is independently at each occurrence H, C1-6 alkyl or C^-C Ce-alkyl; wherein each of the aforementioned alkyl, haloalkyl, cycloalkyl, halocycloalkyl, aryl (e.g. phenyl) and heteroaryl (e.g. pyridyl) groups are optionally substituted, where chemically possible, by 1 to 3 substituents which are each independently selected at each occurrence from the group consisting of: oxo, imino, oximo, halo, nitro, cyano, hydroxyl, amino, C02H, C02-(C1-C4alkyl), C(0)H, d-C-alkyl, C C4 haloalkyl, C C4 alkoxy, and C C4 haloalkoxy; and wherein, if present, any one or more amine group in the fluoroquinolone may optionally be acetylated.
In an embodiment, the antibacterial compound is selected from: fleroxacin, lomefloxacin, nadifloxacin, norfloxacin, rufloxacin, balofloxacin, grepafloxacin, pazufloxacin, sparfloxacin, temafloxacin, tosufloxacin, besifloxacin, clinafloxacin, garenoxacin, gemifloxacin, gatifloxacin, sitafloxacin, trovafloxacin, prulifloxacin, ciprofloxacin, pefloxacin, moxifloxacin, delafloxacin and jnj-q2 In an embodiment, one amine group in the fluoroquinolone is acetylated.
In another aspect of the present invention, there is provided a method of preparing a formulation of a derivative of an antibacterial compound as defined above, the method comprising:
(i) obtaining the derivative of an antibacterial drug molecule; and
(ii) mixing the derivative with one or more pharmaceutically acceptable excipients to produce the pharmaceutical formulation.
Synthetic procedures for the formation of amides are well known in the art.
The compounds of the invention are based on the parent approved pharmaceutically active compounds disclosed below. The synthetic routes to each of the compounds are available in the literature and in the relevant EMA and FDA regulatory files and accordingly are not reproduced here. These disclosures insofar as the synthetic procedures are concerned form part of the disclosure of the present invention. In the interests of brevity, the details of these synthetic procedures are not reproduced here but it is intended that this subject matter is specifically incorporated into the disclosure of these documents by reference. rs to a fluoroquinolone with the following structure:
Figure imgf000010_0001
The skilled man will appreciate that adaptation of methods known in the art could be applied in the manufacture of the compounds of the present invention.
For example, the skilled person will be immediately familiar with standard textbooks such as "Comprehensive Organic Transformations - A Guide to Functional Group Transformations", RC Larock, Wiley-VCH (1999 or later editions), "March's Advanced Organic Chemistry - Reactions, Mechanisms and Structure", MB Smith, J. March, Wiley, (5th edition or later) "Advanced Organic Chemistry, Part B, Reactions and Synthesis", FA Carey, RJ Sundberg, Kluwer Academic/Plenum Publications, (2001 or later editions), "Organic Synthesis - The
Disconnection Approach", S Warren (Wiley), (1982 or later editions), "Designing Organic Syntheses" S Warren (Wiley) (1983 or later editions), "Guidebook To Organic Synthesis" RK Mackie and DM Smith (Longman) (1982 or later editions), etc., and the references therein as a guide.
The skilled chemist will exercise his judgement and skill as to the most efficient sequence of reactions for synthesis of a given target compound and will employ protecting groups as necessary. This will depend inter alia on factors such as the nature of other functional groups present in a particular substrate. Clearly, the type of chemistry involved will influence the choice of reagent that is used in the said synthetic steps, the need, and type, of protecting groups that are employed, and the sequence for accomplishing the protection / deprotection steps. These and other reaction parameters will be evident to the skilled person by reference to standard textbooks and to the examples provided herein.
Sensitive functional groups may need to be protected and deprotected during synthesis of a compound of the invention. This may be achieved by conventional methods, for example as described in "Protective Groups in Organic Synthesis" by TW Greene and PGM Wuts, John Wiley & Sons Inc (1999), and references therein.
Each of the compounds of the present invention may be used as a medicament. Thus, in another aspect of the invention, there is provided a derivative of an antibacterial compound as defined in this specification for the treatment of antibacterial infections.
The invention includes phamaceutical formulations comprising a compound of the invention and optionally, a pharmaceutically acceptable exceipient. Such formulations may also include other active agents.
The compounds and formulations of the present invention may be used in the treatment of a wide range of bacterial infections. In some embodiments, the compounds can be used to treat bacterial infections caused by one or more resistant strains of bacteria. In a further embodiment, the compounds can be used to treat bacterial infections caused by one or more resistant strains of Gram positive bacteria. In a further embodiment, the compounds can be used to treat bacterial infections caused by one or more resistant strains of Gram negative bacteria.
The term 'resistant strains' is intended to mean strains of bacteria which have shown resistance to one or more known antibacterial drug. For example, it may refer to strains which are resistant to meticillin, strains that are resistant to other β-lactam antibiotics and/or strains that are resistant to fluoroquinolones which do not fall within this application. A resistant strain is one in which the MIC 100 of a given compound or class of compounds for that strain has shifted to a significantly higher number than for the parent (susceptible) strain.
The compounds and formulations of the present invention can be used to treat or to prevent infections caused by bacterial strains associated with biowarfare. These may be strains which are category A pathogens as identified by the US government (e.g. those which cause anthrax, plague etc.) and/or they may be strains which are category B pathogens as identified by the US government (e.g. those which cause Glanders disease, mellioidosis etc). In a specific embodiment, the compounds and formulations of the present invention can be used to treat or to prevent infections caused by Gram positive bacterial strains associated with biowarfare (e.g. anthrax). More particularly, the compounds and formulations may be used to treat category A and/or category B pathogens as defined by the US government on 1 st April 2013.
The compounds and formulations of the present invention can be used to treat both Gram positive and Gram negative bacterial infections such as infections of the urinary tract, the respiratory tract, the ear, the skin, the throat, soft tissue, bone and joints (including infections caused by Staph Aureus). The compounds can be used to treat pneumonia, sinusitis, acute bacterial sinusitis, bronchitis, acute bacterial exacerbation of chronic bronchitis, anthrax, chronic bacterial prostatitis, acute pyelonephritis, pharyngitis, tonsillitis, eColi, prophylaxis before dental surgery, cellulitis, acnes, cystitis, infectious diarrhoea, typhoid fever, infections caused by anaerobic bacteria, peritonitis, malaria, babesiosis bacterial vaginosis, pelvic inflammatory disease, pseudomembranous colitis, helicobacter pylori, amoebiasis, giardasis, acute gingivitis, Crohn's Disease, rosacea, fungating Tumours, MRSA, impetigo. The compounds of the present invention may also be used in treating other conditions treatable by eliminating or reducing a bacterial infection. In this case they will act in a secondary manner alongside for example a chemotherapeutic agent used in the treatment of cancer.
The compounds of the present invention can be used in the treatment of the human body. They may be used in the treatment of the animal body. In particular, the compounds of the present invention can be used to treat commercial animals such as livestock. Alternatively, the compounds of the present invention can be used to treat companion animals such as cats, dogs, etc.
The parent compound upon which the derivatives of the invention are based may be selected from the group consisting of: Enoxacin, Fleroxacin, Lomefloxacin, Nadifloxacin, Norfloxacin, Rufloxacin, Balofloxacin, Grepafloxacin, Pazufloxacin, Sparfloxacin, Temafloxacin, Tosufloxacin, Besifloxacin, Clinafloxacin, Garenoxacin, Gemifloxacin, Gatifloxacin, Sitafloxacin, Trovafloxacin, Prulifloxacin, Ciprofloxacin, Pefloxacin, Moxifloxacin, Ofloxacin, jnj- q2, Levofloxacin and Delafloxacin.
Compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, or spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
Compounds of the invention containing one or more asymmetric carbon atoms can exist as two or more stereoisomers. Where a compound of the invention contains a double bond such as a C=C or C=N group, geometric cis/trans (or Z E) isomers are possible. Specifically, any oxime group present in the compounds of the invention may be present as the E-oxime, as the Z-oxime or as a mixture of both in any proprotion. Where structural isomers are interconvertible via a low energy barrier, tautomeric isomerism ('tautomerism') can occur. This can take the form of proton tautomerism in compounds of the invention containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism. Included within the scope of the present invention are all stereoisomers, geometric isomers and tautomeric forms of the compounds of the invention, including compounds exhibiting more than one type of isomerism, and mixtures of one or more thereof. Also included are acid addition or base salts wherein the counter ion is optically active, for example, d-lactate or l-lysine, or racemic, for example, dl-tartrate or dl-arginine.
Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallisation.
Conventional techniques for the preparation/isolation of individual enantiomers when necessary include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC).
Alternatively, the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound of the invention contains an acidic or basic moiety, a base or acid such as 1-phenylethylamine or tartaric acid. The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person.
Chiral compounds of the invention (and chiral precursors thereof) may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1 % diethylamine. Concentration of the eluate affords the enriched mixture.
When any racemate crystallises, crystals of two different types are possible. The first type is the racemic compound (true racemate) referred to above wherein one homogeneous form of crystal is produced containing both enantiomers in equimolar amounts. The second type is the racemic mixture or conglomerate wherein two forms of crystal are produced in equimolar amounts each comprising a single enantiomer. While both of the crystal forms present in a racemic mixture have identical physical properties, they may have different physical properties compared to the true racemate. Racemic mixtures may be separated by conventional techniques known to those skilled in the art - see, for example, "Stereochemistry of Organic Compounds" by E. L. Eliel and S. H. Wilen (Wiley, 1994).
In an embodiment, the present invention provides a compound according to any one or more than one of the following formulae 1-27 taken alone or in any combination:
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001

Figure imgf000018_0001
Figure imgf000019_0001

Figure imgf000020_0001
wherein:
Y is O or NR ;
R1 is independently selected from the group consisting: H , Ci -4 alkyl or Ac;
R2 is independently selected from the group consisting: C Ce alkyl, C2-C6-alkenyl, C2-C6- alkynyl, C Ce haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, -(CH2)n-C3-C8 cycloalkyl, - (CH2)n-C3-C8 heterocycloalkyl, aryl, -(CH2)n-aryl, -(CO)-aryl, heteroaryl, -(CH2)n-heteroaryl and - (CO)-heteroaryl; wherein each n is independently 1 , 2, 3 or 4;
R3 is independently selected at each occurrence from: H , Ci -6 alkyl or Ac; or R2 and R3, together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or N R4 group;
R4 is independently at each occurrence H, C1-6 alkyl or C(0)-CrC6-alkyl; wherein each of the aforementioned alkyl, haloalkyl, cycloalkyl, halocycloalkyl, aryl (e.g. phenyl) and heteroaryl (e.g. pyridyl) groups are optionally substituted, where chemically possible, by 1 to 3 substituents which are each independently selected at each occurrence from the group consisting of: oxo, imino, oximo, halo, nitro, cyano, hydroxyl, amino, C02H, C02-(C1-C4alkyl), C(0)H, d-C-alkyl, C C4 haloalkyl, C C4 alkoxy, and C C4 haloalkoxy;
Ra is independently at each occurrence H or Ac.
The compound may be selected from one, some or all of the group of compounds defined by formulae 1-27. Thus, it may be selected from a smaller group such as that defined by a single formula from within the formulae 1 to 27 (e.g. any one of formulae 4, 21 , 22, 23, 24 and 27), or from a group of compounds defined by more than one (e.g. from two to twenty) of any of the above formulae (e.g. the group defined by formulae 4, 21 , 22, 23, 24 and 27) taken together or in combination.
In an embodiment, the compound is not a compound of formula 1 , formula 26 or formula 27.
The following embodiments apply to any of the embodiments of the invention previously described, as appropriate:
In an embodiment R1 is H.
In an embodiment, Y is NR3. In an alternative embodiment, Y is O.
In an embodiment, R2 is selected from the group consisting: Ci-C6 alkyl, C2-C6-alkenyl, C2-C6- alkynyl, CrC6 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, -(CH2)n-C3-C8 cycloalkyl, - (CH2)n-C3-C8 heterocycloalkyl, aryl, -(CH2)n-aryl, -(CO)-aryl, heteroaryl, -(CO)-heteroaryl, and - (CH2)n-heteroaryl; wherein each n is independently 1 , 2, 3 or 4. In an alternative embodiment, R2 is H.
In an embodiment, R2 is independently selected from the group consisting: H, Ci-C6 alkyl, C2- C6-alkenyl, C2-C6-alkynyl, CrC6 haloalkyl, C3-C8 cycloalkyl, C3-C8 heterocycloalkyl, -(CH2)n-C3- C8 cycloalkyl, -(CH2)n-C3-C8 heterocycloalkyl, aryl, -(CH2)n-aryl, heteroaryl, and -(CH2)n- heteroaryl; wherein each n is independently 1 , 2, 3 or 4;
In an embodiment, R2 is selected from: CrC6 alkyl, C2-C6-alkenyl, C2-C6-alkynyl, CrC6 haloalkyl, C3-C5 cycloalkyl, aryl, -(CH2)n-aryl, heteroaryl, -(CO)-heteroaryl and -(CH2)n- heteroaryl. In a further embodiment, R2 is selected from: Ci-C6 alkyl, C2-C6-alkenyl, Ci-C6 haloalkyl, -(CO)-heteroaryl, aryl, -(CH2)n-aryl and heteroaryl. In yet another embodiment, R2 is selected from: C Ce alkyl, C2-C6-alkenyl, CrC6 haloalkyl, aryl, -(CH2)n-aryl and heteroaryl.
In an embodiment, R2 is C Ce alkyl. In an embodiment, R2 is CrC4 alkyl. Thus, R2 may be methyl, ethyl, iso-propyl, n-propyl, iso-butyl, n-butyl, tert-butyl. In a particular embodiment, R2 is ethyl (e.g. unsubstituted ethyl, 2-cyanoeth-1-yl or 2-hydroxyeth-1 -yl). In another, R2 is tert- butyl. R2 may be
In an embodiment, R2 is C2-C6 alkenyl. In an embodiment, R2 is C3-C6 alkenyl. Thus, R2 may be ally I .
In an alternative embodiment, R2 is C Ce haloalkyl. In an embodiment, R2 is C C4 haloalkyl. Thus, R2 may be trifluoromethyl or R2 may be 1 , 1 , 1 -trifluoroeth-2-yl. In a particular embodiment, R2 is 1 , 1 , 1-trifluoroeth-2-yl.
In an alternative embodiment, R2 is selected from -(CO)-heteroaryl, -(CO)-aryl -(CO)-(CH2)n- aryl, -(CO)-(CH2)n-heteroaryl and -(CO^C Cealkyl. In an embodiment, R2 is -(CO)-heteroaryl. In a further embodiment, R2 is -(CO)-pyridyl, e.g. -(CO)-3-pyridyl.
In a further alternative, R2 is aryl or -(CH2)n-aryl. Thus, R2 may be aryl, e.g. a phenyl group optionally substituted with from 1 to 5 groups independently selected at each occurrence from: halo, nitro, cyano, hydroxyl, amino, N-heterocycloalkyl, S03RB, S02RB, S02NRBRB C02RB C(0)RB, CON RBRB, C C4-alkyl, C C4 haloalkyl, C C4 alkoxy, and C C4 haloalkoxy. It may be that the substituent groups are independently selected from: halo; nitro; cyano; hydroxyl; NR4R4; C02H ; C(0)NR4R4; C02-(C C6alkyl); C(0)H; C C6-alkyl; C C4 haloalkyl; C C4 alkoxy; and Ci-C6 haloalkoxy. In an embodiment, R2 is unsubstituted phenyl. In an alternative embodiment, R2 is substituted phenyl. Thus, it may be that R2 is not an unsubstituted phenyl ring. Thus, R2 may be mono substituted or it may be disubstituted or it may be trisubstituted. In embodiments in which the phenyl is substituted, it may be substituted with at least one electron-withdrawing group. In embodiments in which the phenyl is substituted, it may be substituted with at least 1 group selected from the group consisting of: halo, nitro, S03RB, S02RB, S02N RBRB C02RB C(0)RB, CON RBRB, cyano, and C C4 haloalkyl. Thus, R2 may be 2,6-dichloro-3-trifluoromethylphenyl. R2 may be a phenyl group substituted with one group selected from the group consisting of: halo, nitro, cyano, CO^CrCealkyl), and C C4 haloalkyl. R2 may be a halophenyl group. Thus, R2 may be a chlorophenyl group, e.g. a 2-chlorophenyl or a 3-chlorophenyl or a 4-chlorophenyl group. R2 may be a fluorophenyl group, e.g. a 2- fluorophenyl or a 3-fluorophenyl or 4-fluorophenyl group. R2 may be a bromophenyl group, e.g. a 2-bromophenyl or a 3-bromophenyl or 4-bromophenyl group. In an embodiment, R2 may be a nitrophenyl group, e.g. a 2-nitrophenyl or a 3- nitrophenyl or a 4- nitrophenyl group.
In an embodiment, R2 may be a para- or meta- substituted phenyl group. This embodiment applies equally to monosubstituted rings, to bisubstituted rings and to trisubstituted phenyl rings. In other words, it may be that if R2 is a substituted phenyl group it is not ortho substituted. In a particular embodiment, R2 may be a mono- para- or meta substituted phenyl group. The single substituent may be an electron withdrawing group e.g. a group selected from: halo, nitro, S03Rb, S02Rb, S02NRbRb C02Rb C(0)Rb, CONRbRb, cyano, and C C4 haloalkyl. Thus, the single substituent may be any one of or may be selected from a group consisting of any more than one of: C02H, S03H, S02Me, CN, S02NH2, F, CI, Br, N02, OCF3, CF3, CONMe2, CONHMe, CONH2. The single substituent may also be any one of or may be selected from a group consisting of any more than one of: methyl, OMe, isopropyl, f-butyl.
We have found that in some cases the presence of a meta- or para- substituent has a beneficial effect on the activity.
R2 may also be -(CH2)n-aryl. The integer n may be 1 or 2, e.g. 1 . Thus, R2 may be benzyl in which the aryl ring is optionally substituted with from 1 to 5 groups independently selected at each occurrence from: halo; nitro; cyano; hydroxyl; NR4R4; C02H; C(0)NR4R4; C02-(C C6alkyl); C(0)H; C C6-alkyl; C C4 haloalkyl; C C4 alkoxy; and C C6 haloalkoxy. In a particular embodiment, R2 is benzyl. In another particular embodiment, R2 is pentafluorobenzyl. R2 may be fluorobenzyl (e.g. 4-fluorophenyl)
In another embodiment, R2 is heteroaryl or C^Ce alkylheteroaryl. Thus, R2 may be a heteroaryl group, e.g. a monocyclic heteroaryl group. In particular, R2 may be a 6-membered heteroaryl group containing 1-3-nitrogen atoms. In an embodiment, R2 may be pyridine, pyridazine, pyrimidine or pyrazine. Thus, R2 may be pyridine (e.g. 2-pyridine) or R2 may be pyrimidine (e.g. 2-pyrimidine). In all of these embodiments, the heteroaryl ring is optionally substituted with from 1 to 5 groups independently selected at each occurrence from: halo, nitro, cyano, hydroxyl, amino, N-heterocycloalkyl, S03Rb, S02Rb, S02NRbRb C02Rb C(0)Rb, CONRbRb, Cr C4-alkyl, C C4 haloalkyl, C C4 alkoxy, and C C4 haloalkoxy. It may be that the substituent groups are independently selected from: halo; nitro; cyano; hydroxyl; NR4R4; C02H; C(0)NR4R4; C02-(C1-C6alkyl); C(0)H; C C6-alkyl; C C4 haloalkyl; C C4 alkoxy; and C C6 haloalkoxy. Alternatively, the heteroaryl may be unsubstituted.
In an embodiment, R2 and R3 together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or NR4 group. In a further embodiment, R2 and R3 together with the nitrogen to which they are attached form a 6- membered ring which optionally contains an O, S or NR4 group. In a further embodiment, R2 and R3 together with the nitrogen to which they are attached form a 6- membered ring, e.g. a piperidine ring. In all of these embodiments, the heterocyclic ring formed by R2, R3 and the nitrogen to which they are attached is optionally substituted with from 1 to 5 groups independently selected at each occurrence from: halo; nitro; cyano; hydroxyl; NR4R4; C02H; C(0)NR4R4; CC CCrCealkyl); C(0)H; C Ce-alkyl; C C4 haloalkyl; C C4 alkoxy; and C Ce haloalkoxy. Alternatively, the ring may be unsubstituted.
In an embodiment, it is not the case that R2 and R3 together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or NR4 group.
In an embodiment, R3 is H.
In an embodiment, each R4 is H.
In an embodiment X is N. In an alternative embodiment, X is C.
In an embodiment R9 is H. Alternatively, R9 is NHRa. In a further alternative embodiment, R9 is C C4 alkyl, e.g. R9 may be methyl, ethyl, propyl, butyl, iso-propyl or tert-butyl. In a particular embodiment, R9 is methyl.
In an embodiment, R10 is H. In an alternative embodiment R10 is F.
In an embodiment, R11 is a N-heterocycloalkyl group comprising from 5 to 10 ring atoms wherein at least one of the ring atoms is an NRa group wherein the N-heterocycloalkyl group is optionally substituted with from 1 -3 groups independently selected from halo, hydroxyl, C C4 alkyl, oxo, oxime comprising from 5 to 10 ring atoms and at least one nitrogen atom which is optionally substituted with from 1 -3 groups independently selected from halo, hydroxyl, C C4 alkyl, oxo, oxime. In an embodiment, the N-heterocycloalkyl group is attached to the remainder of the compound of Formula (I) via the N-atom.
In an embodiment, R11 is a piperazine ring which is optionally substituted with from 1-3 groups independently selected from halo, hydroxyl, C C4 alkyl, oxo or oxime. In a preferred embodiment, R11 is a piperazine ring which is optionally substituted with from 1-3 independently selected C C4 alkyl groups. In a further preferred embodiment, R11 is a piperazine ring substituted with a methyl group, e.g. an N-methyl piperazine ring. In an alternative preferred embodiment, R11 is a 3-methyl piperazine ring. In a further alternative, R11 may be a 2-methyl piperazine ring. In another preferred embodiment, R11 is a piperazine ring which is optionally substituted with a C1-4 alkanoyl group. In an embodiment, the R11 group is an acetyl piperazine group.
In an alternative embodiment, R11 is a piperidine ring which is optionally substituted with from 1-3 groups independently selected from halo, hydroxyl, Ci-C4 alkyl, oxo, oxime.
In an alternative embodiment,
Figure imgf000025_0001
In an embodiment, the NH moiety in the structure depicted may alternatively be substituted with an acetyl group.
In an alternative embodiment, R11 is C3-C8 cycloalkyl group substituted with at least one NHRa group and optionally further substituted with from 1 -3 groups independently selected from halo, hydroxyl, C C4 alkyl, oxo or oxime. In a preferred embodiment, R11 is a cyclopropyl group substituted with a NHRa group.
In an embodiment, R12 is H. In an alternative embodiment, R12 is CI or F. In a further alternative, R12 is OR16. In an embodiment, R16 is C C4 alkyl, e.g. R16 may be methyl. In an alternative embodiment, R16 is C C4 haloalky, e.g. C C4 fluoroalkyl such as trifluoromethyl or difluoromethyl.
In an alternative embodiment, R12 and R5, together with the atoms to which they are attached, form a 6-membered ring which optionally contains an O or S atom; wherein the 6-membered ring is optionally substituted with 1 or 2 groups independently selected from halo and C C4 alkyl. In an embodiment, the ring contains an O atom. Alternatively, the ring contains a S atom. The ring may be substituted with a Ci-C4 alkyl group, e.g. a methyl group.
In an embodiment, R5 is C C4 alkyl, e.g. ethyl. In an embodiment, alternative embodiment R5 is C C4 haloalkyi, e.g. a 2-fluoroethyl group. In a further alternative embodiment, R5 is C3-C5 cycloalkyl, e.g. cyclopropyl. In yet a further alternative embodiment, R5 is C3-C5 halocycloalkyl, e.g.fluoro cyclopropyl. In other embodiments R5 is phenyl which may be optionally substituted with from 1-3 halo groups, i.e. R5 may be a difluorophenyl group, such as 2,4-difluorophenyl.
In an embodiment, R6 is H.
In an embodiment, the compound of formula (I) is not a compound selected from:
Figure imgf000026_0001
In another aspect the present invention provides a pharmaceutical formulation comprising a compound of the invention and a pharmaceutically acceptable excipient.
Aryl groups may be 6-membered aryl groups. Aryl groups may be optionally substituted phenyl groups, optionally substituted biphenyl groups, optionally substituted naphthalenyl groups or optionally substituted anthracenyl groups.
Heteroaryl groups may be 5- or 6-membered heteroaryl groups. They may be monocyclic heteroaryl groups, (e.g. heteroaryl groups may be selected from: 5-membered heteroaryl groups in which the heteroaromatic ring is substituted with 1 -3 heteroatoms selected from O, S and N; and 6-membered heteroaryl groups in which the heteroaromatic ring is substituted with 1-2 nitrogen atoms) or they may be bicyclic heteroaryl groups (9-membered bicyclic heteroaryl groups in which the heteroaromatic system is substituted with 1 -4 heteroatoms selected from O, S and N; 10-membered bicyclic heteroaryl groups in which the heteroaromatic system is substituted with 1-4 nitrogen atoms). Specifically, heteroaryl groups may be selected from: pyrrole, furan, thiophene, pyrazole, imidazole, oxazole, isoxazole, triazole, oxadiazole, thiodiazole, pyridine, pyridazine, pyrimidine, pyrazine, triazine (e.g. 1 ,3,5-triazine or 1 ,3,4 triazine), indole, isoindole, benzofuran, isobenzofuran, benzothiophene, indazole,
benzimidazole, benzoxazole, benzthiazole, benzisoxazole, purine, quinoline, isoquinoline, cinnoline, quinazoline, quinoxaline, pteridine, phthalazine, naphthyridine.
Heterocycloalkyi groups of the invention may be from 3-8 membered saturated rings in which the ring contains from 1 to 3 heteroatoms (e.g. O, N or S). Where the heterocycloalkyi group contains a nitrogen, the heterocycloalkyi group may be linked to the remainder of the molecule via the nitrogen or via one of the carbon atoms in the ring. Where the heterocycloalkyi group does not contain a nitrogen, the heterocycloalkyi group will be linked to the rest of the molecule via one of the carbon atoms in the ring.
The aryl and heteroaryl groups are optionally substituted with from 1 to 4 groups independently selected at each occurrence from: halo, nitro, cyano, hydroxyl, NHRa, C02H, C02-(CrC4alkyl), C(0)H, C C4-alkyl, C C4 haloalkyl, C C4 alkoxy, and C C4 haloalkoxy.
The present invention also includes the synthesis of all pharmaceutically acceptable isotopically-labelled compounds of the invnetion wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
For the above-mentioned compounds of the invention the dosage administered will, of course, vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated. For example, if the compound of the invention is administered orally, then the daily dosage of the compound of the invention may be in the range from 0.01 micrograms per kilogram body weight ^g/kg) to 100 milligrams per kilogram body weight (mg/kg).
A compound of the invention, or pharmaceutically acceptable salt thereof, may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the compounds of the invention, or pharmaceutically acceptable salt thereof, is in association with a pharmaceutically acceptable adjuvant, diluent or carrier. Conventional procedures for the selection and preparation of suitable pharmaceutical formulations are described in, for example, "Pharmaceuticals - The Science of Dosage Form Designs", M. E. Aulton, Churchill Livingstone, 1988. Depending on the mode of administration of the compounds of the invention, the pharmaceutical composition which is used to administer the compounds of the invention will preferably comprise from 0.05 to 99 %w (per cent by weight) compounds of the invention, more preferably from 0.05 to 80 %w compounds of the invention, still more preferably from 0.10 to 70 %w compounds of the invention, and even more preferably from 0.10 to 50 %w compounds of the invention, all percentages by weight being based on total composition.
The pharmaceutical compositions may be administered topically (e.g. to the skin) in the form, e.g., of creams, gels, lotions, solutions, suspensions, or systemically, e.g. by oral
administration in the form of tablets, capsules, syrups, powders or granules; or by parenteral administration in the form of a sterile solution, suspension or emulsion for injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion); or by rectal administration in the form of suppositories.
If administered topically high-dosages of the compounds of the invention can be administered. Thus, a compound with an in vitro MIC100 of, for example, 16-64 may still provide an effective treatment against bacteria.
For oral administration the compounds of the invention may be admixed with an adjuvant or a carrier, for example, lactose, saccharose, sorbitol, mannitol; a starch, for example, potato starch, corn starch or amylopectin; a cellulose derivative; a binder, for example, gelatine or polyvinylpyrrolidone; and/or a lubricant, for example, magnesium stearate, calcium stearate, polyethylene glycol, a wax, paraffin, and the like, and then compressed into tablets. If coated tablets are required, the cores, prepared as described above, may be coated with a concentrated sugar solution which may contain, for example, gum arabic, gelatine, talcum and titanium dioxide. Alternatively, the tablet may be coated with a suitable polymer dissolved in a readily volatile organic solvent.
For the preparation of soft gelatine capsules, the compounds of the invention may be admixed with, for example, a vegetable oil or polyethylene glycol. Hard gelatine capsules may contain granules of the compound using either the above-mentioned excipients for tablets. Also liquid or semisolid formulations of the compound of the invention may be filled into hard gelatine capsules. Liquid preparations for oral application may be in the form of syrups or suspensions, for example, solutions containing the compound of the invention, the balance being sugar and a mixture of ethanol, water, glycerol and propylene glycol. Optionally such liquid preparations may contain colouring agents, flavouring agents, sweetening agents (such as saccharine), preservative agents and/or carboxymethylcellulose as a thickening agent or other excipients known to those skilled in art.
For intravenous (parenteral) administration the compounds of the invention may be administered as a sterile aqueous or oily solution.
The size of the dose for therapeutic purposes of compounds of the invention will naturally vary according to the nature and severity of the conditions, the age and sex of the animal or patient and the route of administration, according to well known principles of medicine
Dosage levels, dose frequency, and treatment durations of compounds of the invention are expected to differ depending on the formulation and clinical indication, age, and co-morbid medical conditions of the patient. The standard duration of treatment with compounds of the invention is expected to vary between one and seven days for most clinical indications. It may be necessary to extend the duration of treatment beyond seven days in instances of recurrent infections or infections associated with tissues or implanted materials to which there is poor blood supply including bones/joints, respiratory tract, endocardium, and dental tissues.
Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes
2 3 1 1 13 14 36 of hydrogen, such as H and H, carbon, such as C, C and C, chlorine, such as CI, fluorine, such as 18F, iodine, such as 123l and 125l, nitrogen, such as 13N and 15N, oxygen, such as 150, 170 and 180, phosphorus, such as 32P, and sulphur, such as 35S.
Certain isotopically-labelled compounds, for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.
Substitution with positron emitting isotopes, such as 11C, 18F, 150 and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labelled compounds can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described using an appropriate isotopically-labelled reagent in place of the non-labelled reagent previously employed.
Throughout the description and claims of this specification, the words "comprise" and "contain" and variations of the words, for example "comprising" and "comprises", means "including but not limited to", and is not intended to (and does not) exclude other moieties, additives, components, integers or steps.
Throughout the description and claims of this specification, the singular encompasses the plural unless the context otherwise requires. In particular, where the indefinite article is used, the specification is to be understood as contemplating plurality as well as singularity, unless the context requires otherwise.
Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understood to be applicable to any other aspect, embodiment or example described herein unless incompatible therewith.
Example 1 - Preparation of quinolone imidazole derivatives (Method A).
Figure imgf000030_0001
Method
To a solution of the starting material carbonyl diimidazole (0.2g) in dimethylformamide (5ml) under argon atmosphere at room temperature was added the required substrate (1 .1 equivalent; levofloxacin is shown in the scheme above). The reaction was stirred overnight then quenched with water (20ml), extracted using dichloromethane (x 3) and dried (MgS04). The products were purified using automated grace flash chromatography. In the case of levofloxacin the gradient elution was MeOH 10%/ DCM 90%.
Example 2 - Preparation of compounds of the invention (Method A)
Figure imgf000031_0001
To a solution of the starting material imidazole (0.2g; levofloxacin imidazole is shown as an example in the scheme above), in dimethylformamide (5ml) under argon atmosphere at room temperature was added the relevant hydrazine, hydrazide or hydroxylamine (1 .1 equivalent). The reaction was stirred overnight then quenched with water (20ml), extracted using dichloromethane (x 3) and dried (MgS04). The products were purified using automated grace flash chromatography. In the case of levofloxacin the gradient elution MeOH 10%/ DCM 90%.
Example 3 - Preparation of compounds of the invention (Method B)
To a solution of fluoroquinolone (e.g. levofloxacin hemihydrate, 200. mg, 0.5500mmol) in DCM (2ml_) was added triethylamine (0.08ml_, 0.5500mmol) followed by ethyl chloroformate
(0.05ml_, 0.5500mmol) at 0°C under Ar. This was allowed to stir at that temperature for 5 minutes. After which time the reaction mixture was allowed to warm to room temperature and hydrazine (0.5500mmol) was added in one portion. This was then allowed to stir at that temperature for 18 hours. After which time the reaction mixture was directly purified by automated flash chromatography (Grace Reveleris®) using an eluent system of DCM:MeOH.
Example 4 - Preparation of compounds of the invention (Method C)
To a room temperature solution of fluoroquinolone (1.25mmol) in MeCN (10ml_) was added trifluoroacetic anhydride at room temperature (1.21 ml_, 8.72mmol). The solution was left to stir for two hours and hydrazine (0.22ml_, 2.49mmol) was added, followed by triethylamine
(0.52ml_, 3.74mmol) approx. 20 minutes later. The solution was left to stir overnight, after which time the solution was diluted with DCM (10ml) and washed with water (4 x 10ml). The organic layer was dried (MgS04) and the solvent removed under vacuum. The resulting residue was purified by automated GRACE chromatography to give the desired product.
Exemplary compounds of the invention:
In an embodiment, the compound of the invention is any one or is selected from any group more than one of the following exemplary compounds:
B2013/050943
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001

Figure imgf000039_0001
Unless indicated otherwise, the NMR data provided below was obtained in CDCI3 using a 400 MHz NMR machine.
(A) δ 1.59-1.57 (3H, m), 2.37 (3H, s), 2.56-2.59 (4H, m), 3.32-3.43 (4H, m), 4.38-4.39 (1H, m), 4.41-4.48 (2H, m), 4.50 (2H, d), 5.33-5.41 (2H, m), 6.03-6.11 (1H, m), 7.67- 7.70 (1H, d), 8.60 (1H, s), 12.17 (1H, s). Compound A was prepared using method A.
(B) δ 1.53-1.54 (3H, d), 2.36 (3H, s), 2.54-2.58 (4H, m), 3.31-3.41 (4H, m), 4.27-4.36 (3H, m), 5.02-5.09 (2H, q), 7.27-7.61 (6H, m), 8.57 (1H, s), 12.14 (1H, s). Compound B was prepared using method A.
(C) δ 1.58-1.60 (3H, d), 2.37 (3H, s) 2.58 (4H, 4), 3.32-3.43 (4H, m), 4.29-4.33 (1H, q), 4.40-4.47 (2H, m), 5.17-5.30 (2H, d), 7.63-7.66 (1H, d), 8.56 (1H, s), 12.21 (1H, S). Compound C was prepared using method A.
(D) δ 1.51-1.52 (3H, d), 2.37 (3H, s), 2.56 (4H, m), 3.32-3.48 (4H, m), 4.18-4.21 (1H, q), 4.28-4.31 (1H, q), 4.48-4.53 (1H, m), 7.00-7.03(1 H, m), 7.12-7.14(2H, m), 7.26-7.31 (2H, m) 7.70-7.73 (2H, d) 8.77 (1 H, s), 12.68 (1 H, s). Compound D was prepared using method A. (E) δ 1.37 (9H, s), 1.58-1.59 (3H, m), 2.37 (3H, s), 2.56 (4H, m), 3.35-3.40 (4H, m), 4.31-4.43 (3H, m), 7.69-7.72 (1H, d), 8.63 (1H, s), 11.96 (1H, s). Compound E was prepared using method A.
(F) δ 0.95-0.99 (2H, m), 1.12-1.16 (2H, m), 1.31-1.34 (2H, m), 1.36-1.39 (1H, t), 1.41-
I.43 (1H, s), 3.22-3.25 (4H, m), 3.50 (1H, m), 3.60-3.67 (4H, m), 4.15-4.19 (2H, m), 7.29-7.31 (1H, d), 7.95-7.98 (1H, d), 8.70 (1H, s). Compound F was prepared using method A.
(G) δ 1.52-1.57 (3H, m), 2.37 (3H, s), 2.57 (4H, m), 3.34-3.42 (4H, m), 3.38-3.40 (2H, m), 4.47-4.52 (1H, m), 7.61-7.62 (1H, m), 7.84 (1H, s), 7.98 (1H, s), 8.09 (1H, m), 8.20 (1 H, s), 8.64-8.67 (1 H, d), 11.91 (1 H, s). Compound G was prepared using method A.
(H) δ 1.59-1.62 (3H, m), 2.37 (3H, s), 2.56 (4H, m), 3.33-3.42 (4H, m), 4.33-4.40 (3H, m), 5.11-5.13 (1H, d), 7.55-7.58 (1H, d), 8.51 (1H, s), 11.19-11.21 (1H, d). Compound H was prepared using method A.
(I) δ 1.34 (3H, m), 2.38 (3H, s), 2.59 (4H, m), 3.35-3.42 (4H, m), 4.23-4.30 (2H, m), 4.52-4.57 (1H, q), 6.71-6.77(2H, m), 7.46-7.71 (3H, m), 8.20-8.21 (1H, m), 8.88 (1H, s),
I I .63 (1 H, d). Compound I was prepared using method A.
(J) MS (m/z, rel. intensity) 554.3 (M+H+, 100). Compound J was prepared using method A.
(K) δ 1.48-1.76 (12H, m), 1.88-1.93 (5H, m), 2.17 (3H, s), 2.73-2.78 (4H, m), 2.88-2.89 (3H, m), 3.13-3.16 (5H, m), 3.41-3.45 (4H, m), 4.30-4.45 (3H, m), 7.59-7.69 (1H, dd), 8.70 (1H, s), 10.90 (1H, s). Compound K was prepared using method A. (L) δ 1.53-1.59 (3H, m), 2.36-2.37 (3H, s), 2.55-2.57 (4H, m), 2.88-2.95 (1H, S), 3.34- 3.41 (4H, m), 4.39-4.59 (3H, 7.34-7.41 (2H, m), 8.19 (1H, m), 8.52 (1H, m), 8.60-8.69 (1H, m) 9.14 (1H, s), 12.52 (1H, s). Compound Lwas prepared using method A.
(M) δ 1.54-1.56 (3H, d), 2.36 (3H, s), 2.55 (4H, m), 3.31-3.42 (4H, m), 4.29-4.40 (3H, m), 7.07-7.08 (1 H, d), 7.51 (2H, s), 8.51 (1 H, s), 11.84-11.85 (1 H, d). Compound M was prepared using method A.
(O) δ 1.53-1.55 (3H, d), 2.39 (3H, s), 2.55-2.60 (4H, m), 3.33-3.50 (4H, m), 4.26-4.44 (3H, m), 6.49-6.50 (1H, d), 6.82-6.87 (2H, m), 7.12-7.17 (2H, m), 7.66-7.70 (1H, d), 8.61 (1 H, s), 11.45-11.46 (1 H, d). Compound O was prepared using method A.
(P) δ 1.54-1.56 (3H, d), 2.38 (3H, s), 2.56-2.59 (4H, m), 3.36-3.47 (4H, m), 4.31-4.52 (3H, m), 6.88-6.91 (1H, d), 7.70-7.75 (2H, m), 8.11-8.14 (1H, d), 8.58-8.61 (1H, s), 11.67-11.68 (1 H, d). Compound P was prepared using method A. ill δ 11.5 (1H, m), 8.6 (1H, s), 7.7 (1H, d), 7.3 (1H, m), 7.2 (1H, m), 6.9 (1H, m), 6.8 (1H, m), 6.7 (1H, m), 4.5 (1H, m), 4.3 (2H, m), 3.3-3.5 (4H, m), 2.6 (4H, m), 2.4 (3H, m), 1.5 (3H, m). Compound T was prepared using method A.
(U) δ 12.8 (1H, m), 10.4 (1H, m), 8.7 (1H, m), 8.4 (1H, m), 8.2 (1H, m), 7.8 (1H, m), 6.7 (1H, m), 4.4-4.6 (2H, m), 4.3 (1H, m), 3.9 (3H, s), 3.4 (4H, m), 2.6 (4H, m), 2.3 (3H, m), 1.6 (3H, m). Compound U was prepared using method A.
(V) δ 14.8 (1H, m), 8.7 (1H, m), 7.9-8.0 (5H, m), 7.4 (1H, m), 7.2 (1H, m), 7.1 (1H, m), 3.8-4.0 (4H, m), 3.6 (1H, m), 3.4 (4H, m), 2.0 (2H, m), 1.4 (2H, m), 1.2 (2H, m). Compound V was prepared using method A. (W) δ 8.6 (1 H, s), 8.2 (1 H, s), 7.8-8.0 (2H, m), 7.4 (2H, m), 7.2 (1 H, m), 7.1 (1 H, m), 3.8 (4H, m), 3.5 (1 H, m), 3.3 (4H, m), 1.4 (2H, m), 1.2 (2H, m). Compound W was prepared using method A.
(X) δ 8.6 (1 H, m), 7.8 (4H, m), 7.1 (2H, m), 4.2 (3H, m), 3.2 (4H, m), 2.8 (4H, m), 2.4 (3H, s), 1.7 (3H, m). Compound X was prepared using method A.
(Y) δ 1 1.4 (1 H, s), 8.6 (1 H, s), 7.8 (1 H, d), 7.2 (2H, m), 6.8 (2H, m), 6.3 (1 H, m), 4.3 (2H, m), 4.2 (1 H, m), 3.2 (4H, m), 2.6 (4H, m), 2.2 (3H, s), 1.5 (3H, s). Compound Y was prepared using method A.
(AA) δ 1 1.5 (1 H, m), 8.6 (1 H, m), 7.7 (1 H, d), 6.9-7.2 (4H, m), 4.5 (3H, m), 3.5 (4H, m), 2.7 (4H, m), 2.5 (3H, s), 1.6 (3H, m). Compound AA was prepared using method A.
(AE) δ 1.5 (d, 3H), 2.4 (s, 3H), 2.5 (m, 4H), 3.4 (m, 4H) 4.0 (s, 2H) 4.4 (m, 3H), 7-7.6 (m, 5H), 8.6 (s, 1 H). Compound AE was prepared using method A.
(AG) δ 1 1.5 (s, 1 H), 8.96 (m, 1 H), 8.0 (s, 2NH), 7.75 (m, 1 H), 7.1 (m, 1 H), 4.8 (m, 1 H), 4.4 (m, 1 H), 3.4 (m, 4H), 3.3 (m, 1 H), 2.45 (m, 4H), 2.35 (s, 3H), 2.3 (s, 3H), 1.57-1.59 (3H, d).
Compound AG was prepared using method A.
(AH) δ 8.96 (m, 1 H), 8.0 (s, 2NH), 7.8 (m, 1 H), 7.75 (m, 1 H), 7.4 (m, 2H), 7.1 (m, 1 H), 4.8 (m, 1 H), 4.4 (m, 1 H), 3.4 (m, 4H), 3.3 (m, 1 H), 2.45 (m, 4H), 2.3 (s, 3H), 1.57-1.59 (3H, d). MS (m/z, rel. intensity) 495.5 (M+ H+, 97). Compound AH was prepared using method A.
(AI) MS (m/z, rel. intensity) 524.5 (M+ H+, 100). Compound Al was prepared using method A.
(AJ) MS (m/z, rel. intensity) 432.5 (M+ H+, 92). Compound AJ was prepared using method A. (AK) MS (m/z, rel. intensity) 493.5 (M+ H+, 90). Compound AK was prepared using method A.
(ΑΙ_) δ 8.96 (m, 1 H), 8.0 (s, 2NH), 7.95 (m, 2H), 7.75 (m, 1 H), 7.7 (m, 2H), 6.75 (m, 1 H), 5.35 (s, 1 H) 4.8 (m, 2H), 4.4 (m, 2H), 3.4 (m, 4H), 3.3 (m, 1 H), 2.45 (m, 4H), 2.3 (s, 3H), 1.57-1.59 (3H, d) MS (m/z, rel. intensity) 495.5 (M+ H+, 95). MS (m/z, rel. intensity) 495.5 (M+ H+, 95). Compound AL was prepared using method A.
(AM) δ 8.5 (s, 1 H), 7.7 (m, 2H), 7.3 (m, 2H), 4.5-4.3 (m, 3H), 3.4 (m, 4H), 2.5 (m, 4H), 2.3 (s, 3H), 1.4 (3H, d). MS (m/z, rel. intensity) 514 (M+ H+, 100). Compound AM was prepared using method A.
(AN) δ 8.96 (m, 1 H), 8.05-7.5 (m, 6H), 4.4 (m, 3H), 3.4 (m, 4H), 2.45 (m, 4H), 2.3 (s, 3H), 1.6 (3H, d). MS (m/z, rel. intensity) 480 (M+ H+, 100). Compound AN was prepared using method A.
(AP) δ 1.208-1.226 (m, 3H), 1.472-1.489 (m, 3H), 2.297 (s, 3H), 2.494 (s, 4H), 3.286- 3.334 (m, 4H), 3.671-3.724 (m, 2H), 4.159-4.195 (q, 2H), 4.253-4.381 (m, 2H), 6.1 -6.2 (s, 1 H), 7.626 (d, 1 H), 8.477 (s, 1 H), 1 1.229 (s, 1 H). Compound AP was prepared using method A.
(AQ) δ 1.443-1.460 (d, 3H), 2.27 (s, 3H), 2.366 (s, 3H), 2.531-2.561 (m, 4H), 3.31 1 -3.450 (m, 5H) 4.189-4.274 (m, 2H), 6.787-6.814 (m, 2H), 6.982-7.048 (m, 2H), 7.600-7.613 (m, 2H), 8.678 (s, 1 H), 1 1.475 (s, 1 H). Compound AQ was prepared using method A.
(AR) δ 1.550-1.566 (m, 3H), 2.176 (s, 3H), 2.582 (s,4H), 3.366-3.417 (m, 4H), 4.370-4.404 (m, 3H), 7.043-7.1 1 1 (m, 4H), 7.676-7.707 (m, 1 H), 7.91 (s, 1 H), 8.60 (s, 1 H), 1 1.55 (s, 1 H).
Compound AR was prepared using method A. (AS) δ 1.5-1.6 (m,3H), 2.35 (s, 3H), 2.5-2.6 (m, 4H), 3.3-3.5 (m, 4H), 3.7 (s, 1H), 4.2-4.6 (m, 3H), 6.8 (m, 1H), 6.9 (m,1H), 7.0 (m, 1H), 7.3-7.5 (d, 1H), 8.2 (s, 1H), 8.3-8.6 (m, 2H), 12.0 (s, 1H). Compound AS was prepared using method A.
(AT) δ 1.5-1.6 (m, 3H), 2.3 (s, 3H), 2.4-2.6 (m, 4H), 3.2-3.4 (m, 4H), 4.2-4.4 (m, 3H), 6.8 (m, 3H), 7.0 (m, 2H), 7.6-7.8 (m, 1H), 8.5 (s, 1H), 11.4-11.5 (s, 1H). MS (m/z, rel. intensity) 536 (M+ H+, 100). Compound AT was prepared using method A.
(AU) δ 1.5-1.6 (m,3H), 2.35 (s, 3H), 2.5-2.6 (m, 4H), 3.3-3.5 (m, 4H), 4.3-4.6 (m, 3H), 6.8 (d,2H), 7.3 (s, 1H), 7.4(d, 2H), 7.6 (m, 1H), 8.6 (s, 1H), 11.5 (s, 1H). Compound AU was prepared using method A.
(AV) δ 1.5-1.6 (m,3H), 2.35 (s, 3H), 2.5-2.6 (m, 4H), 3.3-3.5 (m, 4H), ), 4.2-4.6 (m, 3H), 6.5 (bs,1H), 6.7 (m, 1H), 6.8 (m, 1H), 6.9 (m, 1H), 7.1 (s, 1H), 7.5-7.7 (m, 1H), 8.7 (s, 1H), 11.47 (s, 1H). MS (m/z, rel. intensity) 488 (M+H+, 100). Compound AVwas prepared using method A.
(AW) δ 1.4 (d, 3H), 2.4 (s, 3H), 2.6 (m, 4H), 3.4 (m, 4H) 4.3 (m, 3H), 6.4-6.9 (m, 3H), 7.7 (m, 3H), 8.6 (s, 1H), 9.5 (s, 1H), 11.6 (s, 1H). Compound AW was prepared using method A.
(AX) MS (m/z, rel. intensity) 466.4 (M+H+, 100). UV254nm. Compound AX was prepared using method A.
(AY) δ 1.44-1.46 (d, 3H), 2.27 (s, 3H), 2.366 (s, 3H), 2.531-2.561 (m, 4H), 3.311-3.450 (m, 5H) 4.189-4.274 (m, 2H), 4.6 (1H, m) 6.9-7.2 (m, 2H), 6.982-7.048 (m, 3H), 7.8 (d, 1H)-7.613 (m, 2H), 8.1 (s, 1H) 8.9 (s, 1H), 11.6 (s, 1H). MS (m/z, rel. intensity) 520.3 (M+, 100). UV 254nm. Compound AY was prepared using method A.
(AZ) δ 0.8 (s, 2H), 1.0 (s, 3 H), 1.1-1.3 (m, 2H), 1.5-1.6 (m, 3H), 1.8-1.9 (m,3H), 2.2-2.4 (m, 1H), 3.1-3.3 (m, 2H), 3.35-3.5 (m,3H), 3.6 (s, 3H), 3.8-3.9 (m, 3H), 4.0-4.1 (m, 2H), 4.5-4.6 (m, 1 H), 5.1 (m, 1 H), 7.7-7.8 (d, 1 H), 8.3 (s, 1 H), 8.8 (s, 1 H), 15.0 (s, 1 H). Compound AZ was prepared using method C.
(BB) MS (m/z, rel. intensity) 531 (M+ H+, 100). Compound BB was prepared using method B.
(BD) δ 1 1.1 1 (s, 1 H), 8.58 (s, 1 H), 7.70 (d, 1 H), 5.08 (m, 1 H), 4.46-4.31 (m, 4 H), 3.45-3.25 (m, 6 H), 2.62 (t, 2 H), 2.58 (br t, 4 H), 2.39 (s, 3 H), 2.28 (s, 3 H) and 1.60 (d, 3 H).
Compound BD was prepared using method B.
(BE) δ 1 1.51 (s, 1 H), 8.82 (s, 1 H), 7.59 (d, 1 H), 7.07 (m, 1 H), 6.92 (d, 1 H), 6.80 (m, 1 H), 6.39 (m, 1 H), 4.35 (m, 1 H), 4.1 1 (m, 2 H), 3.35 (m, 4 H), 2.55 (br t, 4 H), 2.36 (s, 3 H), 2.28 (s,
3 H) and 1.37 (d, 3 H). Compound BE was prepared using method B.
(BF) δ 12.02 (s, 1 H), 8.85 (s, 1 H), 7.44 (m, 3 H), 7.02 (m, 1 H), 4.59 (m, 1 H), 4.29 (m, 2 H), 3.39 (m, 4 H), 2.59 (br t, 4 H), 2.40 (s, 3 H) and 1.38 (d, 3 H). Compound BF was prepared using method B.
(BG) δ 10.79 (s, 1 H), 8.76 (s, 1 H), 7.70 (d, 1 H), 4.43 (m, 2 H), 4.30 (m, 1 H), 3.39 (m, 4 H), 2.72 (2 x (3 H, s)), 2.57 (br t, 4 H), 2.38 (s, 3 H), 1.58 (d, 3 H). MS (m/z, rel. intensity) 404 (M+ H+, 50). Compound BG was prepared using method B.
(BH) δ 1 1.53 (s, 1 H), 8.61 (s, 1 H), 7.76 (d, 1 H), 6.90 (m, 3 H), 4.36 (m, 3 H), 3.41 (m, 4 H), 2.60 (br t, 4 H), 2.40 (s, 3 H), 2.30 (s, 3 H), 2.25 (s, 3 H) and 1.57 (d, 3 H). Compound BH was prepared using method A.
(BI) δ 1 1.1 1 (s, 1 H), 8.59 (s, 1 H), 7.70 (d, 1 H), 4.43-3.34 (m, 7 H), 3.05 (br t, 4 H), 2.57 (br t,
4 H), 2.38 (s, 3 H) and 1.61 (d, 3 H). Compound Bl was prepared using method A. (BJ) δ 1 1.75 (s, 1 H), 8.70 (s, 1 H), 8.22 (m, 1 H), 7.78 (d, 1 H), 7.48 (m, 1 H), 7.21 (m, 1 H), 6.86 (m, 1 H), 4.42 (m, 3 H), 3.51 (m, 4 H), 2.77 (m, 4 H), 2.53 (s, 3 H) and 1.61 (d, 3 H).
Compound BJ was prepared using method B.
(BK) δ 1 1.93 (s, 1 H), 9.07 (s, 1 H), 7.63-7.10 (m, 5 H), 4.52 (m, 1 H), 4.24 (m, 1 H), 4.00 (m, 1 H), 3.39 (br t, 4 H), 2.60 (br t, 4 H), 2.41 (s, 3 H) and 1.07 (d, 3 H). MS (m/z, rel. intensity) 509 (M+ H+, 60). Compound BK was prepared using method B.
(BM) δ 12.13 (s, 1 H), 8.64 (s, 1 H), 8.15 (d, 1 H), 7.70 (d, 2 H), 6.18 (d, 1 H), 4.48 (m, 1 H), 4.40 (m, 2 H), 3.94 (s, 3 H), 3.40 (m, 4 H), 2.58 (br t, 4 H), 2.39 (s, 3 H) and 1.57 (d, 3 H). MS (m/z, rel. intensity) 485 (M+ H+, 100). Compound BM was prepared using method B.
(BN) δ 1 1.7 (1 H, s), 8.7 (1 H, s), 8.3 (1 H, s), 8.1 (1 H, d), 7.7 (1 H, d), 7.2 (1 H, br s), 6.9 (1 H, s), 4.6-4.3 (3H, m), 3.5-3.3 (4H, m), 2.7 (4H, s), 2.5 (3H, s), 1.6 (3H, d). MS (m/z, rel. intensity) 529 (M H", 95). Compound BN was prepared using method B.
(BO) δ 7.9 (1 H, s), 7.8 (1 H, s), 7.7 (1 H, d), 7.5 (2H, d), 6.7 (2H, d), 4.9-4.4 (3H, m), 3.6-3.4 (3H, s), 3.3 (4H, s), 2.9 (4H, s), 2.7 (3H, s), 1.5 (3H, d). MS (m/z, rel. intensity) 531 (M+ H+, 100). Compound BO was prepared using method B.
(BQ) δ 12.2 (1 H, s), 8.7 (1 H, s), 8.0 (1 H, d), 7.7 (1 H, d), 6.0 (1 H, s), 4.6-4.3 (3H, m), 3.8 (4H, d), 3.6 (4H, d), 3.4 (4H, d), 2.6 (4H, s), 2.4 (3H, s), 1.6 (3H, d). MS (m/z, rel. intensity) 538 (M+ H+, 10). Compound BQ was prepared using method B.
(BP) δ 1 1.5 (1 H, s), 8.8 (1 H, s), 7.6 (2H, d), 7.4 (2H, d), 6.9 (2H, d), 4.4-4.2 (3H, m), 3.5-3.2 (4H, m), 2.6 (4H, s), 2.4 (3H, s), 1.5 (3H, d). MS (m/z, rel. intensity) 520 (M+ H+, 100).
Compound BP was prepared using method B. (BR) δ 12.1 (1H, s), 9.8 (1H, s), 9.2 (1H, s), 8.8 (1H, s), 8.3 (1H, d), 7.7 (1H, d), 7.3 (1H, d), 4.6-4.2 (3H, m), 3.5-3.3 (4H, m), 2.7 (4H, s), 2.4 (3H, s) and 1.6 (3H, d). MS (m/z, rel.
intensity) 542 (M+H+, 100). Compound BR was prepared using method B.
(BS) δ 11.9 (1 H, s), 8.8 (1 H, s), 7.5 (1 H, d), 6.9 (1 H. s), 4.8 (1 H, s), 4.4 (2H, q), 3.4 (4H, s), 2.6 (4H, s), 2.4 (3H, s), 1.5 (3H, d). Compound BS was prepared using method B.
(BT) δ 11.8 (1H, s), 8.7 (1H, s), 7.8-7.6 (2H, m), 7.3 (1H, d), 7.1 (1H, s), 6.9 (1H, s), 4.5 (1H, d), 4.4-4.2 (2H, m), 3.5-3.3 (4H, m), 2.6 (4H, s), 2.4 (3H, s), 1.5 (3H, d). MS (m/z, rel. intensity) 521 (M+H+, 100). Compound BT was prepared using method B.
(BU) δ 12.1 (1H, s), 8.6 (1H, s), 8.1 (1H, s), 7.8 (1H, d), 7.5 (2H, d), 4.6 (1H, s), 4.5-4.3 (2H, m), 3.4 (4H, s), 2.5 (4H, s), 2.3 (3H, s), 1.5 (3H, d). MS (m/z, rel. intensity) 521 (M+ H+, 100). Compound BU was prepared using method B.
(BV) (NMR measured in D6-DMSO and not CDCI3) δ 10.8 (1H, s), 8.8 (1H, s), 8.4 (1H, s), 7.7 (1H, d), 6.6 (1H, brs), 4.9 (1H, d), 4.6 (1H, d), 4.4 (1H, d), 3.8 (4H, m) 3.3 (4H, m, H20), 2.8 (3H, s), 1.4 (3H, d). MS (m/z, rel. intensity) 488 (M+ H+, 97). Compound BVwas prepared using method B.
(BW) δ 11.2 (1H, s), 8.7 (1H, s), 7.8 (1H, d), 4.5 (2H, d), 4.4 (1H, d), 3.6-3.4 (4H, m), 2.6 (4H, s), 2.4 (3H, s), 1.6 (3H, s), 1.2 (9H, s). MS (m/z, rel. intensity) 432 (M+ H+, 20). Compound BWwas prepared using method B.
(BX) δ 11.3 (1H, s), 8.8 (1H, s), 8.7 (1H, s), 7.8 (2H, d), 7.6 (1H, d), 6.7 (2H, d), 4.9 (1H, d), 4.5 (1H, d), 4.4 (1H, d), 3.2 (2H, s), 2.5-2.3 (7H, m), 2.2 (3H, s), 1.4 (3H, d). MS (m/z, rel. intensity) 496 (M+H+, 100). Compound BX was prepared using method B.
(BY) δ 11.2 (1 H, s), 8.6 (1 H, s), 7.8 (1 H, s), 7.2 (2H, d), 6.9 (2H, d), 6.6 (2H, br s), 6.3 (1 H, s), 4.8 (1H, brs), 4.2-2.8 (6H, m), 1.5 (10H, m). Compound BY was prepared using method B. (BZ) δ 1.53-1.57 (3H, t), 2.26 (3H, s), 2.42 (3H, s), 2.67-2.69 (4H, t), 3.32-3.35 (4H, t), 4.25- 4.30 (2H, q), 6.40-6.41 (1H, d), 6.80-6.82 (1H, d), 6.86-6.89 (2H, d), 7.03-7.05 (2H, d), 8.09- 8.12 (1H, d), 8.67 (1H, s), 11.52-11.53 (1H, d). MS (m/z, rel. intensity) 438 (M+ H+, 100).
Compound BZ was prepared using method B.
(CA) δ 1.41-1.45 (1H, t), 1.57-1.61 (3H, t), 2.40 (3H, s), 2.56-2.62 (4H, m), 2.76 (2H, s), 3.34- 3.44 (4H, m), 4.31-4.38 (2H, m), 4.38-4.45 (2H, m), 7.68-7.72 (1H, m), 8.59 (1H, s), 11.07 (1H, s). MS (m/z, rel. intensity) 390 (M+H+, 35). Compound CAwas prepared using method B.
(CB) δ: 4.2 (m, 2H), 4.5 (m, 1H, 4.7 (m, 2H), 6.8 (m, 3H), 7.2 (d, 2H), 7.8 (m, 2H), 8.2 (s, 1H), 8.6 (s, 1 H), 11.0 (s, 1 H). Compound CB was prepared using method B.
(CC) (NMR measured in CD3OD and not CDCI3) δ 1.55 (m, 3H), 2.1 (s, 3H), 2.72 (m, 4H), 3.33 (m, 4H), 3.4 (m, 1H), 4.5-4.6 (m, 3H), 4.87 (s, H20), 6.85 (d, 2H), 7.19 (d, 2H), 8.03 (s, 1H), 8.8 (s, 1H). Compound CC was prepared using method B.
(CD) (NMR measured in CD3OD and not CDCI3) δ 1.25 (9H, s), 1.52-1.53 (3H, d), 2.96 (3H, s), 3.18-3.24 (4H, m), 3.30-3.32 (3H, m), 3.35 (2H, s), 3.55-3.70 (6H, m), 4.40-4.46 (1H, d), 4.52- 4.55 (1H, d), 4.72-4.73 (1H, d), 6.80-6.82 (2H, d), 7.23-7.25 (2H, d), 7.55-7.58 (1H, d), 8.79 (1H, s). MS (m/z, rel. intensity) 508 (M+H+, 100). Compound CD was prepared using method B.
(CE) δ 1.57-1.59 (3H, d), 2.37 (3H , s), 2.56-2.59 (4H, m), 3.32-3.42 (4H, m), 4.33-4.42 (2H, m), 4.46-4.47 (1H, d), 6.81-6.84 (1H, m), 7.74-7.77 (1H, d), 7.76-7.78 (1H, d), 7.94-7.96 (1H, d), 8.43-8.44 (1H, d), 8.60 (1H, s), 12.56-12.58 (1H, d). MS (m/z, rel. intensity) 519 (MH", 100). Compound CE was prepared using method B.
(CF) δ 11.5 (s, 1H), 8.75 (s, 1H), 7.70 (d, 1H), 7.1 (m, 1H), 6.72 (m, 3H), 6.5 (m, 1H), 4.4-4.2 (m, 3H), 3.35 (m, 4H), 2.55 (m, 4H), 2.35 (s, 3H), 2.3 (s, 3H), 1.45 (d, 3H). MS (m/z, rel.
intensity) 466 (M+H+, 100). Compound CF was prepared using method B. (CG) δ 1 1.5 (s, 1 H), 8.6 (s, 1 H), 8.0 (s, 1 H), 7.10-7.65 (m, 3H), 4.4-4.2 (m, 4H), 3.35 (m, 4H), 2.50 (m, 4H), 2.4 (s, 3H), 1.5 (d, 3H). Compound CG was prepared using method B.
(CH) δ 1 1.8 (s, 1 H), 8.55 (s, 1 H), 7.8 (d, 1 H) 6.95 (m, 3H), 4.4-4.1 (m, 3H), 3.4 (m, 4H), 2.50 (m, 4H), 2.4 (s, 3H), 1.5 (d, 3H). MS (m/z, rel. intensity) 488 (M+ H+, 95). Compound CH was prepared using method B.
(CI) δ 1 1.8 (s, 1 H), 9.25 (s, 1 H), 8.7 (s, 1 H), 8.15 (s, 1 H), 7.05-7.6 (m, 3H), 4.4-4.1 (m, 3H), 3.35 (m, 4H), 2.50 (m, 4H), 2.3 (s, 3H), 1.5 (d, 3H). MS (m/z, rel. intensity) 531 (M+ H+, 95). Compound CI was prepared using method B.
(CJ) MS (m/z, rel. intensity) 545.2 (M+ H+, 100). UV 254nm. Compound CJ was prepared using method B.
(CK) MS (m/z, rel. intensity) 496.35 (M+ H+, 100). UV 254nm. Compound CK was prepared using method B.
(CL) δ 1 1.6 (1 H, s), 8.8 (1 H, s), 8.0 (1 H, d), 7.2-6.5 (5H, m) 4.3 (1 H, m), 4.0 (2H, m), 3.3-2.5 (8H, m) 2.0-1.5 (6H, m), 1.3 (3H, d). MS (m/z, rel. intensity) 451.3 (M+ H+, 100). UV 254nm. Compound CL was prepared using method B.
Example 5
The in vitro efficacy of a series of compounds was assessed for activity against a range of bacterial strains. All test articles were stored in the dark at 4 °C following delivery. Immediately prior to use, approximately 1 mg of each compound was accurately weighed and dissolved in the appropriate volume of DMSO to give a stock concentration of 1.28g/L.
Strains Susceptibility tests were performed against a range of anaerobic bacterial strains: Details of the strains used are as follows.
Figure imgf000050_0001
Revival and Growth of the Strains
All strains were recovered from long-term storage at -80 °C by sub-culturing onto fresh blood agar plates and incubating at 35-37 °C in air, except for Streptococcus pneumoniae which was incubated in the presence of 5% C02, for 24 hours. Following visual checks to ensure purity and appropriate colony characteristics, isolates were deemed suitable for use.
Preparation of the Inoculum
The inocula for each bacterial strain were prepared by picking 5-10 distinct colonies from the culture plates and suspending them in 3ml of sterile saline. The inoculum was resuspended by vigorous shaking on a vortex mixer for 15s. The turbidity was then adjusted to McFarland standard 0.5 (1 -5 x 106 CFU/ml). The inoculum was further diluted in Mueller Hinton Broth for MIC tests to give a final inoculum in each well of 2-8 x 105 CFU/ml. For Streptococcus pneumoniae the Mueller Hinton broth was supplemented with 5% lysed horse blood (MHLB).
MIC Assay Conditions
MICs were tested in Mueller Hinton broth (MHLB for S. pneumoniae) in accordance with the appropriate CLSI guidelines. STEP 1: Addition of Test Article
a. A stock solution was prepared at a concentration of 1.28 g/L in DMSO. The stock was further diluted in Mueller Hinton broth (or MHLB) to give a top starting concentration of 128 mg/L in the assay. In addition, for each strain, a comparator control was included. The final concentration range for the comparator control (ciprofloxacin) was 0.03 -16 η^/Ι_.100μΙ_ of Mueller Hinton broth was dispensed into each well in columns 2-12. 200μΙ_ of the appropriate test compound solution (at 256mg/L) was dispensed into each well in column 1. b. 100μΙ_ aliquots were pipetted from column 1 wells and dispensed into column 2 with a multichannel pipette (± 2% coefficient of variation) thus diluting two-fold. 100 μΙ_ samples were then pipetted from column 2 wells and dispensed into column 3. The process was repeated through to column 10. The final 100 μΙ_ of diluted drug from column 10 was then discarded. Row 1 1 acted as a positive control (no drug or test article, organisms added), Row 12 acted as a negative control (no drug or test article, and no organisms added).
STEP 2: Addition of Bacterial Strains
100μΙ_ of the appropriate inoculum suspension in Mueller Hinton broth (or MHLB) was added to the appropriate wells. This resulted in a well containing 200μΙ_ final volume (made up of 100μΙ_ diluted compound or diluents and 100μΙ_ of inoculum or broth alone).
STEP 3: Incubation of Assay Plates
All plates were incubated in the dark at 35-37 °C in air for 18-24 hours. STEP 4: Reading of Plates
Plates were read visually and spectrophotometrically (450nm) where possible, 24 hours post inoculation. Endpoints of 50%, 80% and 100% inhibition were determined (or CLSI interpretation endpoints following visual examination). The results are presented in Table 1 (as MICsn along with levofloxacin) and further results are presented in Table 2 (as MICioo)- P201345WO specification as filed{804521 }[1 ].doc
Table 1 : MIC50 for selected compounds and levofloxacin against non-resistant (susceptible) strains of Gram negative and Gram positive bacteria (indicated by a + or a - in parentheses after the species name)
Klebsiella Streptococcus
Pseudomonas Escherichia Acinetobacter pneumoniae Staphylococcus pneumoniae Staphylococcus Enterococcu
Compound aeruginosa {-) coli (-) baumannii (-) (-) aureus (+) w epidermidis (+) s faecal is (+)
E 128 8 32 128 64 128 128 128
C 128 32 64 128 128 128 128 128
A 128 16 32 128 32 128 64 128
N 8 0.25 1 16 4 16 2 16
H 2 0.25 0.25 4 0.5 2 1 4
G 2 0.25 0.25 2 0.5 2 0.5 4
I 16 0.25 1 16 4 2 2 16
D 128 32 64 128 128 64 128 128
B 128 64 64 128 128 128 128 128
K 128 16 32 128 128 32 128 128
P201345WO specification as filed{804521 }[1 ].doc
F 64 4 8 128 1 128 0.25 4
<0.25 <0.25 <0.25 <0.25 <0.25 <0.25 <0.25 <0.25
0
<0.25 <0.25 <0.25 <0.25 <0.25
P 0.5 0.5 0.5
M 64 32 32 128 16 2 16 16
Q 0.5 0.25 0.25 2 0.25 0.25 0.25 1
8 0.25 0.5 8 2 8 1 8
S 2 0.25 0.25 2 0.25 1 0.25 4
T 2 0.25 0.25 8 0.25 0.25 0.25 4
<0.25 <0.25 <0.25 <0.25 <0.25
levofloxacin 0.5 0.5 0.5
Table 2: MIC10o for selected compounds against a variety of resistant strains and non-resistant (susceptible) strains of Gram positive and Gram negative bacteria.
Figure imgf000053_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000054_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000055_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000056_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000057_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000058_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000059_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000060_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000061_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000062_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000063_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000064_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000065_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000066_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000067_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000068_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000069_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000070_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000071_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000072_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000073_0001
P201345WO specification as filed{804521}[1 ].doc
Figure imgf000074_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000075_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000076_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000077_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000078_0001
P201345WO specification as filed{804521}[1].doc
Figure imgf000079_0001
Example 6
The in vitro efficacy of a series of compounds was assessed for activity against a range of bacterial strains in either unbuffered Mueller-Hinton (MH) broth, or MH broth buffered to pH 6.2 or pH 8.3, and their activity compared with that of levofloxacin.
Method
The bacterial strains used during this study are detailed below
Figure imgf000080_0001
All strains were recovered from long-term storage at -80°C by sub-culturing onto fresh agar plates and incubating aerobically overnight at 37°C. Following visual checks to ensure purity and appropriate colony characteristics, isolates were deemed suitable for use.
Compounds were stored at 4°C in the dark. All samples were received in 100μΙ aliquots at a concentration of 1.28 mg/mL.
MICs were determined in MH broth in accordance with Clinical and Laboratory Standards Institute guidelines M07-A9 for aerobic bacteria, other than using flat-bottomed wells to allow spectrophotometric reading of MIC assays. MH broth was prepared according to the manufacturer's instructions and either left unmodified, or buffered as detailed below. All broths were then filter-sterilised through a 0.22 μηη filter before use. Unbuffered MH broth was found to have a pH of 7.18. • For MH broth pH 6.2, 3-(N-morpholino)propanesulfonic acidacid (MOPS) was added to a final concentration of 20 mM, and the pH adjusted with 1 M HCI.
• For MH broth pH 8.2, 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) was added to a final concentration of 20 mM, and the pH adjusted with 1 M NaOH.
Five to 10 well-isolated colonies were picked and suspended in 3 mL sterile saline. The inoculum was suspended by vigorous shaking on a vortex mixer for 15 s. Turbidity was adjusted to McFarland standard 0.5 (1 -5 x 108 CFU/mL). The inoculum was further diluted in the corresponding MH broth for MIC tests to give a final inoculum in each well of 2-8 x 105 CFU/mL. a. Stock solutions of compounds were diluted in MH to give a maximum starting concentration of 64 μg/mL in the assay. 100 μΙ_ of MH was dispensed into each well in columns two to 12 of a 96 well plate. 200 μΙ_ of the appropriate test compound solution (128 μg/mL) was dispensed into each well in column one. b. Serial two-fold dilutions were performed from column one to column 10 to give a concentration range of 128 to 0.25 μg/mL of each compound in the assay. Columns 1 1 and 12 served as positive (no drug or test article, inoculum added), and negative (no drug, test article, or inoculum added) growth controls respectively.
For all strains, 100 μΙ of inoculum suspension was added to 100 μΙ_ of diluted compound to give a final volume of 200 μΙ_ and a final concentration range of 64 - 0.125 μg/mL.
Assay plates were incubated aerobically at 37°C for 18-20 h. Following incubation, plates were assessed both visually and spectrophotometrically at 490 nm.
Results
Table 3. MICs for 100, 80, and 50% inhibition of growth of E. coli ATCC 25922 at pH 6.2, 7.18 and 8.2. NG, No growth in positive control wells.
E. coli ATCC 25922 pH 6.2 (MOPS) Unbuffered (pH 7.18) pH 8.2 (HEPES)
Compound 100% 80% 50% 100% 80% 50% 100% 80% 50%
BX 4 4 2 0.25 0.25 <0.125 NG NG NG
AA <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 NG NG NG
0 0.5 0.5 0.25 <0.125 <0.125 <0.125 NG NG NG
Q 1 0.5 0.5 0.25 0.25 <0.125 NG NG NG
AV 0.25 0.25 0.25 <0.125 <0.125 <0.125 NG NG NG
AW 4 4 4 0.5 0.5 0.25 NG NG NG
CA 8 4 4 2 2 1 NG NG NG
CB <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 NG NG NG
Nadifloxacin <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 NG NG NG
Delafloxacin <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 NG NG NG
Levofloxacin <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 NG NG NG
Table 4. MICs for 100, 80, and 50% inhibition of growth of A. baumannii NCTC 13420 at pH 6.2, 7.18 and 8.2.
Figure imgf000082_0001
Table 5. MICs for 100, 80, and 50% inhibition of growth of S.aureus ATCC 29213 at pH 6.2, 7.18 and 8.2.
Figure imgf000082_0002
0 8 8 8 0.5 0.5 0.25 0.5 0.5 0.5
Q 4 4 2 2 2 2 4 4 4
AV 2 2 1 0.5 0.5 0.5 0.5 0.5 0.5
AW 8 8 8 4 4 4 2 1 1
CA 2 2 2 4 4 2 8 8 4
CB <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 <0.125
Nadifloxacin <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 <0.125
Delafloxacin <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 <0.125 <0.125
Levofloxacin 0.25 0.25 <0.125 <0.125 <0.125 <0.125 0.25 0.25 <0.125
Table 6. MICs for 100, 80, and 50% inhibition of growth of S.aureus NRS74 at pH 6.2, 7.2 and 8.2.
Figure imgf000083_0001
Example 7
The data provided in Example 6 shows that there can be expected to be a considerable degree of activity variation in the activity at certain pHs for any given compound. Thus some compounds of the invention may be more active at low pH infection sites such as open wounds, whereas others may be more active at high pH infection sites such as urinary tract infections. The following study demonstrates that compound O, which was shown in Example 4 to have low activity at low pH, does as expected have very little activity in acidic wound sites.
Sprague-Dawley male rats 225-250g (at delivery) were used in these studies which were supplied by Charles River UK Ltd and were specific pathogen free. Following receipt at our facility rats were allowed to acclimatise for 5-7 days (weigh at the start of the experiment ~250- 275g).
Rats were housed in sterilized individual ventilated cages that expose the animals at all times to HEPA filtered sterile air. Rats had free access to food and water (sterile) and were provided with sterile aspen chip bedding (changed every 3-4 days). Additionally, during infection, rats had additional access to wet food if required to ensure they remained fully hydrated. The room temperature was 22°C +/- 1°C, with a relative humidity of 60% and maximum background noise of 56dB. Rats were exposed to 12 hour light/dark cycles.
Staphylococcus aureus (S. aureus) ATCC 29213 (MSSA), a well characterised strain, was used throughout the study.
Method
In this efficacy model, group sizes of 4 rats per treatment group (test articles, comparator or vehicle control) per time-point were used. An additional group of 3 rats was included to determine burden 1 hour post-infection as pre-treatment group (total 19 rats). Rats were euthanized 24 hours post treatment. All rats were rendered neutropenic by immunosuppression with cyclophosphamide at 150mg/kg 3 days before infection and 75mg/kg 1 day before infection by intraperitoneal injection. The immunosuppression regime leads to neutropenia starting 24 hours post the first administration which continues throughout the study. 24 hours post the second round of immunosuppression, rats were infected with S. aureus ATCC 29213 by intramuscular injection of 0.1 mL of bacteria into both lateral thigh muscles under temporary inhaled anaesthesia using ~6.54x106 cfu/rat thigh. Rats were administered buprenorphine at ~0.03 mg/kg subcutaneously immediately post infection and 12 hours post infection for pain relief (buprenorphine causes delayed gastric emptying that can effect the PK following oral administration).
Table 7 shows the study treatment groups, treatment regimen and harvest (kill) time points. A total of 19 rats were used in the study (4 per time point plus 3 untreated controls).
Compound O was reconstituted freshly at each time point for immediate dosing in dimethyl sulfoxide (DMSO) giving a stock of 100mg/ml_, the DMSO stock was then diluted 1 :50 and 1 : 10 with water for injection (WFI) to provide dosing solutions at 2mg/ml_ and 10mg/ml_ respectively. Upon dilution solutions immediately turned cloudy, but after addition of 100- 200μΙ_ 1 M HCI the solutions became clear, non-particulate, straw coloured solutions (~pH3). Dosing solutions were delivered at 10ml_/kg orally in order to achieve 20mg/kg and 100mg/kg per dose. Vehicle solution (10% Dimethyl sulfoxide (DMSO), WFI, HCI), was prepared in the same manner minus the test article and delivered by oral administration at 10ml_/kg. Comparator Levofloxacin (Tavanic-5mg/ml_, Sanofi Aventis, UK) was also diluted in water for injection to 2mg/ml_ and delivered orally at 10ml_/kg in order to achieve 20mg/kg per dose. Following reconstitution, all test article solutions remained clear and non-particulate for the duration of the dosing period. Antibacterial treatment was initiated 1 hour post infection and delivered twice by oral gavage at 10 mL/kg (2.5ml_ per 250g rat) (see Table 1). All dosing solutions were well tolerated when administered via oral gavage at 10ml_/kg.
Table 7: Treatment groups, treatment regime, time of tissue harvest and number of rats per group.
Figure imgf000085_0001
Throughout the study rats had their clinical condition assessed. Animals were euthanized 24h after treatment. Following euthanasia rat weights were determined prior to both thighs being removed and weighed individually. Individual thigh tissue samples were homogenized in ice cold sterile phosphate buffered saline. Thigh homogenates were then quantitatively cultured on CLED agar, incubated at 37°C for up to 2 days and colonies counted. The data from the culture burdens were analyzed using the Kruskal-Wallis test, all pairwise comparisons (Conover-lnman), using Stats Direct v. 2.7.8. Culture burdens of the animals treated with test agents were compared to those of the vehicle control group. Note that each thigh has been treated as an independent sample for the statistical analysis.
Results
Table 8: Endpoint thigh tissue burdens (cfu/g; cfu = colony forming unit) for samples collected 24 hours post treatment
Figure imgf000086_0001
In conclusion, compound O failed to demonstrate any antibacterial efficacy in reducing Staphylococcus aureus ATCC 29213 (MSSA) burdens from the low pH localised sites of infection in this study. These results were entirely consistant with our understanding (see Example 6).
Example 8 - Biodefence
Several compounds of the invention were further tested for activity against strains which are known surrogates for bacterial strains associated with biowarfare. The testing methodology were the same as those described in Example 5. The activity is shown in Table 9 and the surrogate relationship is shown in Table 10.
Table 9: Activity of selected compounds against biodefense surrogate strains
Figure imgf000086_0002
Bacillus subtilis - QBR114769-04 positive 4
Burk olderia cepacia - QBR114769-09 negative 32
B Bacillus cereus - QBR114769-06 positive 1
Bacillus subtilis - QBR114769-05 positive 0.5000
Burkholderia cepacia - QBR114769-08 negative 8
Burkholderia cepacia - QBR114769-09 negative 4
Burkholderia cepacia - QBR114769-07 negative 16
Yersinia enterocolitica - QBR114769-03 negative 0.1200
Yersinia enterocolitica - QBR114769-02 negative 0.2500
Yersinia enterocolitica - QBR114769-01 negative 0.2500
Bacillus subtilis - QBR114769-04 positive 0.2500
P Bacillus subtilis - QBR114769-04 positive 1
Yersinia enterocolitica - QBR114769-01 negative 2
Burkholderia cepacia - QBR114769-09 negative 16
Yersinia enterocolitica - QBR114769-03 negative 1
Burkholderia cepacia - QBR114769-07 negative 32
Burkholderia cepacia - QBR114769-08 negative 32
Bacillus cereus - QBR114769-06 positive 4
Yersinia enterocolitica - QBR114769-02 negative 1
Bacillus subtilis - QBR114769-05 positive 2
Table 10: Surrogate relationships of biodefense strains
Figure imgf000087_0001
Preliminary studies show that compounds have comparable activity to the corresponding approved parent compounds.
This preliminary data indicates that the compounds have antibacterial activity and can be used as a medicament against one of the above bacterial strains.
Thus the compounds of the invention are active in the treatment of one or more of the above strains.
In some cases the activity is broad spectrum. In other cases the activity is selective over Gram-negative bacteria or Gram positive bacteria. In yet further cases the activity is selective over one or more strains.
In addition, the compounds are active against bacterial strains associated with biowarfare, particularly Gram positive strains.

Claims

Claims
1. A compound of formula (I):
Figure imgf000088_0001
X is C or N;
Y is O or NR3;
R1 is independently selected from the group consisting: H , C1 -4 alkyl or Ac;
R2 is independently selected from the group consisting: H , Ci-C6 alkyl, C2-C6-alkenyl, C2-C6- alkynyl, C C6 haloalkyi, C3-C8 cydoalkyi, C3-C8 heterocycloalkyl, -(CH2)n-C3-C8 cydoalkyi, - (CH2)n-C3-C8 heterocycloalkyl, aryl, -(CH2)n-aryl, -(CO)-aryl, -(CO)-(CH2)n-aryl, heteroaryl, -(CO)- heteroaryl, -(CH2)n-heteroaryl and -(CO)-(CH2)n-heteroaryl; wherein each n is independently 1 , 2, 3 or 4;
R3 is independently selected at each occurrence from: H , C1 -6 alkyl or Ac; or R2 and R3, together with the nitrogen to which they are attached form a 3- to 7- membered ring which optionally contains an O, S or NR4 group;
R4 is independently at each occurrence H , C1 -6 alkyl or C^-C Ce-alkyl;
R5 is selected from the group consisting of: C C4 alkyl, C C4 haloalkyi, C3-C5 cydoalkyi, C3-C5 halocycloalkyl; unsubstituted phenyl; phenyl substituted with from 1 to 3 independently selected halogen atoms; unsubstituted pyridyl; and pyridyl substituted with from 1 to 3 independently substituents selected from the group consisting of: halo and NHRa; wherein Ra is H or Ac;
R6 is selected from the group consisting of: H, C C4 alkyl and C C4 haloalkyl;
or alternatively, R5 and R6, together with the atoms to which they are attached to form a 4-6- membered ring which optionally contains an O or S atom; wherein the 4-6-membered ring is optionally substituted with 1 or 2 groups independently selected from halo and C C4 alkyl;
R9 is selected from the group consisting: H, NHRa or CrC4-alkyl; wherein Ra is H or Ac;
R10 is independently selected from the group: H or F;
R11 is selected from the group consisting of: an N-heterocycloalkyl group and a C3-C8 cycloalkyi group; wherein the N-heterocycloalkyl group comprises from 5 to 10 ring atoms and at least one nitrogen atom wherein the N-heterocycloalkyl group is optionally substituted with from 1 -3 groups independently selected from halo, tri(C1-C4 alkyl)silyloxy, hydroxyl, C C4 alkyl, oxo or oxime.and wherein any nitrogen which does not attach the N-heterocycloalkyl group to the rest of the compound of Formula (I) is an NRa group; and the C3-C8 cycloalkyi group is optionally substituted with at least one NHRa group and optionally further substituted with from 1-3 groups independently selected from halo, hydroxyl, tri(C1-C4 alkyl)silyloxy, C C4 alkyl, oxo or oxime; wherein Ra is H or Ac;
R12 is absent or is selected from the group consisting of: H, OR16 and halo; wherein R16 is selected from the group consisting of: C C4 alkyl and C C4 haloalkyl; or R12 and R5, together with the atoms to which they are attached form a saturated or unsaturated 5- to 7- membered ring which optionally contains an O, S or NR4 group; wherein the 5-7-membered ring is optionally substituted with 1 or 2 groups independently selected from halo, C C4 alkyl and C C4 haloalkyl; wherein if X is N, R12 is absent; wherein each of the aforementioned alkyl, haloalkyl, cycloalkyi, halocycloalkyl, aryl (e.g.
phenyl) and heteroaryl (e.g. pyridyl) groups are optionally substituted, where chemically possible, by 1 to 3 substituents which are each independently selected at each occurrence from the group consisting of: oxo, imino, oximo, halo, nitro, cyano, hydroxyl, amino, N- heterocycloalkyl, S03Rb, S02Rb, S02NRbRbC02Rb C(0)Rb, CONRbRb, C C4-alkyl, C C4 haloalkyi, C C4 alkoxy, and C C4 haloalkoxy, wherein Rb is selected from H, C C^alkyl and C C4-haloalkyl.
2. A compound of claim 1 , wherein the compound of formula (I) is a compound of formula (II):
Figure imgf000090_0001
A compound of claim 1 , wherein the compound of formula (I) is a compound of formuls
Figure imgf000090_0002
4. A compound of any one of claims 1 to 3, wherein R2 is independently selected from the group consisting: H, C C6 alkyl, C2-C6-alkenyl, C2-C6-alkynyl, C^Ce haloalkyi, C3-C8 cydoalkyi, C3-C8 heterocycloalkyl, -(CH2)n-C3-C8 cydoalkyi, -(CH2)n-C3-C8 heterocycloalkyl, aryl, -(CH2)n- aryl, heteroaryl, and -(CH2)n-heteroaryl; wherein each n is independently 1 , 2, 3 or 4.
5. A compound of any one of claims 1 to 3, wherein R2 is aryl.
6. A compound of claim 5, wherein R2 is a phenyl group substituted with from 1 to 5 groups independently selected at each occurrence from: halo, nitro, cyano, hydroxyl, amino, N- heterocycloalkyl, S03Rb, S02Rb, S02NRbRb C02Rb C(0)Rb, CONRbRb, C C4-alkyl, C C4 haloalkyl, C C4 alkoxy, and C C4 haloalkoxy.
7. A compound of claim 6, wherein R2 is a phenyl group substituted with at least one electron-withdrawing group.
8. A compound of claim 6 or claim 7, wherein R2 is a mono- para- substituted phenyl group.
9. A compound of any of claims 1 to 3, wherein R2 is a heteroaryl group.
10. A compound of claim 9, wherein R2 is a 6-membered heteroaryl group containing 1 -3- nitrogen atoms optionally substituted with from 1 to 4 groups independently selected at each occurrence from: halo, nitro, cyano, hydroxyl, amino, N-heterocycloalkyl, S03Rb, S02Rb, S02NRbRb C02Rb C(0)Rb, CONRbRb, C C4-alkyl, C C4 haloalkyl, C C4 alkoxy, and C C4 haloalkoxy.
1 1 . A compound of any of claims 1 to 3, wherein R2 is a Ci-C6 alkyl or Ci-C6 haloalkyl group.
12. A compound of any of claims 1 to 1 1 , for therapeutic use.
13. A compound of claim 12, wherein the compound is for use in treating bacterial infections.
14. A compound of claim 13, wherein the compound is for use in treating a bacterial infection caused by one or more resistant strains of bacteria.
15. A compound of claim 13, wherein the compound is for use in treating an infection caused by a bacterial strain associated with biowarfare.
PCT/GB2013/050943 2012-04-11 2013-04-11 Quinolonones with antibacterial properties WO2013153394A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1206384.8A GB201206384D0 (en) 2012-04-11 2012-04-11 Antibacterial drug derivatives
GB1206384.8 2012-04-11

Publications (1)

Publication Number Publication Date
WO2013153394A1 true WO2013153394A1 (en) 2013-10-17

Family

ID=46177152

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2013/050943 WO2013153394A1 (en) 2012-04-11 2013-04-11 Quinolonones with antibacterial properties

Country Status (2)

Country Link
GB (2) GB201206384D0 (en)
WO (1) WO2013153394A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017046605A1 (en) * 2015-09-18 2017-03-23 Redx Pharma Plc Antibacterial compounds
CN106674220A (en) * 2016-12-26 2017-05-17 郑州工业应用技术学院 N-methyl enoxacin aldehyde-4-aryl thiosemicarbazide derivative, and preparation method and application thereof
WO2018020004A1 (en) * 2016-07-29 2018-02-01 Abac Therapeutics, S.L 2-pyrrolidine phenylhydrazides antibacterial agents
CN113461612A (en) * 2021-07-20 2021-10-01 上海应用技术大学 Quinolone tankyrase 2 inhibitor and preparation method and application thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4880814A (en) * 1987-11-13 1989-11-14 Abbott Laboratories 7-cycloalkyl naphthyridines
US4959363A (en) * 1989-06-23 1990-09-25 Sterling Drug Inc. Quinolonecarboxamide compounds, their preparation and use as antivirals.
WO2004083207A1 (en) * 2003-03-12 2004-09-30 Abbott Laboratories Naphthyridine derivatives as antibacterial agents
WO2008131134A1 (en) * 2007-04-17 2008-10-30 Cylene Pharmaceuticals, Inc. Hydrazide compounds and uses thereof
US20090088420A1 (en) * 2007-04-12 2009-04-02 University Of Southern California Compounds with hiv-1 integrase inhibitory activity and use thereof as anti-hiv/aids therapeutics
WO2009046383A1 (en) * 2007-10-05 2009-04-09 Cylene Pharmaceuticals, Inc. Quinolone analogs and methods related thereto

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4880814A (en) * 1987-11-13 1989-11-14 Abbott Laboratories 7-cycloalkyl naphthyridines
US4959363A (en) * 1989-06-23 1990-09-25 Sterling Drug Inc. Quinolonecarboxamide compounds, their preparation and use as antivirals.
WO2004083207A1 (en) * 2003-03-12 2004-09-30 Abbott Laboratories Naphthyridine derivatives as antibacterial agents
US20090088420A1 (en) * 2007-04-12 2009-04-02 University Of Southern California Compounds with hiv-1 integrase inhibitory activity and use thereof as anti-hiv/aids therapeutics
WO2008131134A1 (en) * 2007-04-17 2008-10-30 Cylene Pharmaceuticals, Inc. Hydrazide compounds and uses thereof
WO2009046383A1 (en) * 2007-10-05 2009-04-09 Cylene Pharmaceuticals, Inc. Quinolone analogs and methods related thereto

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
CLARK R F ET AL: "Novel inhibitors of bacterial protein synthesis: structure-activity relationships for 1,8-naphthyridine derivatives incorporating position 3 and 4 variants", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, ELSEVIER SCIENCE, GB, vol. 14, no. 12, 21 June 2004 (2004-06-21), pages 3299 - 3302, XP004841393, ISSN: 0960-894X, DOI: 10.1016/J.BMCL.2004.03.091 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 10 July 2005 (2005-07-10), XP002697655, retrieved from CAS accession no. 854355-04-7 Database accession no. 854355-04-7 *
DATABASE REGISTRY [Online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 21 August 2001 (2001-08-21), XP002697669, retrieved from CAS accession no. 352213-91-3 Database accession no. 352213-91-3 *
E. L. ELIEL; S. H. WILEN: "Stereochemistry of Organic Compounds", 1994, WILEY
E. V. NOSOVA ET AL, CHEMISTRY OF HETEROCYCLIC COMPOUNDS, vol. 38, no. 8, 1 January 2002 (2002-01-01), pages 922 - 928, XP055063503, ISSN: 0009-3122, DOI: 10.1023/A:1020965312463 *
FA CAREY; RJ SUNDBERG: "Advanced Organic Chemistry, Part B, Reactions and Synthesis", 2001, KLUWER ACADEMIC/PLENUM PUBLICATIONS
GUO QIANG HU ET AL: "Synthesis and antitumor and antibacterial evaluation of fluoro-quinolone derivatives (III): Mono- and bis-Schiff-bases", CHINESE CHEMICAL LETTERS, vol. 23, no. 5, 29 March 2012 (2012-03-29), pages 515 - 517, XP028421600, ISSN: 1001-8417, [retrieved on 20120120], DOI: 10.1016/J.CCLET.2012.01.029 *
M. E. AULTON, PHARMACEUTICALS - THE SCIENCE OF DOSAGE FORM DESIGNS, 1988
M. SAEED ARAYNE ET AL: "Synthesis and biological evaluations of enoxacin carboxamide derivatives", ARCHIVES OF PHARMACAL RESEARCH, vol. 32, no. 7, 1 July 2009 (2009-07-01), pages 967 - 974, XP055063741, ISSN: 0253-6269, DOI: 10.1007/s12272-009-1700-5 *
MB SMITH; J. MARCH: "March's Advanced Organic Chemistry - Reactions, Mechanisms and Structure", WILEY
MICHAEL K SCHWAEBE ET AL: "Facile and efficient generation of quinolone amides from esters using aluminum chloride", TETRAHEDRON LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 52, no. 10, 21 December 2010 (2010-12-21), pages 1096 - 1100, XP028272837, ISSN: 0040-4039, [retrieved on 20110107], DOI: 10.1016/J.TETLET.2010.12.108 *
NAJMA SULTANA ET AL: "Synthesis, Characterization and Biological Evaluations of Ciprofloxacin Carboxamide Analogues", BULLETIN OF THE KOREAN CHEMICAL SOCIETY, vol. 32, no. 2, 20 February 2011 (2011-02-20), pages 483 - 488, XP055063500, ISSN: 0253-2964, DOI: 10.5012/bkcs.2011.32.2.483 *
RC LAROCK: "Comprehensive Organic Transformations - A Guide to Functional Group Transformations", 1999, WILEY-VCH
RK MACKIE; DM SMITH: "Guidebook To Organic Synthesis", 1982, LONGMAN
S WARREN: "Designing Organic Syntheses", 1983, WILEY
S WARREN: "Organic Synthesis - The Disconnection Approach", 1982, WILEY
TW GREENE: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS INC
VIKAS KUMAR ET AL: "Synthesis and antimicrobial activity of thiazolidinone hybrids", INDIAN JOURNAL OF CHEMISTRY, vol. 50B, 1 January 2011 (2011-01-01), pages 1496 - 1503, XP055064006 *
WENTLAND ET AL: "3-QUINOLINECARBOXAMIDES. A SERIES OF NOVEL ORALLY-ACTIVE ANTIHERPETIC AGENTS", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 36, no. 11, 1 January 1993 (1993-01-01), pages 1580 - 1596, XP002136767, ISSN: 0022-2623, DOI: 10.1021/JM00063A008 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017046605A1 (en) * 2015-09-18 2017-03-23 Redx Pharma Plc Antibacterial compounds
WO2018020004A1 (en) * 2016-07-29 2018-02-01 Abac Therapeutics, S.L 2-pyrrolidine phenylhydrazides antibacterial agents
US10919851B2 (en) 2016-07-29 2021-02-16 ABAC Therapeutics, S.L. 2-pyrrolidine phenylhydrazides antibacterial agents
CN106674220A (en) * 2016-12-26 2017-05-17 郑州工业应用技术学院 N-methyl enoxacin aldehyde-4-aryl thiosemicarbazide derivative, and preparation method and application thereof
CN106674220B (en) * 2016-12-26 2018-10-09 郑州工业应用技术学院 N- methyl Enoxacin aldehyde contracting 4- arylamino thiourea derivatives and its preparation method and application
CN113461612A (en) * 2021-07-20 2021-10-01 上海应用技术大学 Quinolone tankyrase 2 inhibitor and preparation method and application thereof

Also Published As

Publication number Publication date
GB201206384D0 (en) 2012-05-23
GB201306626D0 (en) 2013-05-29

Similar Documents

Publication Publication Date Title
TWI282335B (en) A61k 31/437 200601 a i vhtw a61p 31/04 200601 a i vhtw
RU2434851C1 (en) Cyclic n,n&#39;-diarylthioureas or n,n&#39;-diarylureas - antagonists of androgen receptors, anti-cancer medication, method of obtaining and application
US20130065895A1 (en) Pyrazolopyridine, Pyrazolopyrazine, Pyrazolopyrimidine, Pyrazolothiophene and Pyrazolothiazole Compounds as MGLUR4 Allosteric Potentiators, Compositions, and Methods of Treating Neurological Dysfunction
WO2008049538A1 (en) Substituted dihydropyrazolones and use thereof as hif-prolyl-4 -hydroxylase inhibitors
JP2020528399A (en) Antibiotic resistant breaker
US20150291565A1 (en) Indole compounds and their use as antimicrobials
US7884099B2 (en) Quinolone carboxylic acid-substituted rifamycin derivatives
CA2886402A1 (en) Tazobactam arginine antibiotic compositions
WO2013153394A1 (en) Quinolonones with antibacterial properties
JP2022548250A (en) Thyroid hormone receptor beta agonist compounds
TW201904970A (en) Antimicrobial agent and method of manufacture and use
US20030187008A1 (en) Quinolonecarboxylic acid derivative
WO2017046605A1 (en) Antibacterial compounds
CA2537066C (en) Benzoquinolizine-2-carboxylic acid arginine salt tetrahydrate
WO2014161412A1 (en) Tricyclic quinolone derivative and preparation method and use thereof
CN108976222B (en) Heterocyclic ring-containing biaryl oxazolidinone compound and preparation method thereof
CN102603738B (en) Stable moxifloxacin hydrochloride compound
US9085539B2 (en) Cyclic N,N′-diarylthiourea—androgen receptor antagonist, anti breast cancer composition and use thereof
US6818642B2 (en) Benzylaminopyrimidines
WO2015114317A1 (en) 5h-isothiazolo[4,5-c]pyridine-3,4-dione or 5h-pyrazolo[4,3-c]pyridin-3,4-dione as antibacterial compounds
WO2017137742A1 (en) Antibacterial compounds
US11780838B1 (en) Pyrrolo[3,2-b]quinoline compounds as antibacterial agents
WO2013072703A1 (en) Antibacterial drug derivatives
US20130131073A1 (en) Fused-imidazoyl compounds useful as antimicrobial agents
WO2017177828A1 (en) Novel oxazolidinone-fluoroquinolone derivative and uses

Legal Events

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

Ref document number: 13718617

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13718617

Country of ref document: EP

Kind code of ref document: A1