WO2003050107A1 - New generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments - Google Patents

New generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments Download PDF

Info

Publication number
WO2003050107A1
WO2003050107A1 PCT/IN2002/000232 IN0200232W WO03050107A1 WO 2003050107 A1 WO2003050107 A1 WO 2003050107A1 IN 0200232 W IN0200232 W IN 0200232W WO 03050107 A1 WO03050107 A1 WO 03050107A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
oxo
dihydro
quinoline
fluoro
Prior art date
Application number
PCT/IN2002/000232
Other languages
English (en)
French (fr)
Inventor
Noel J. De Souza
Mahesh V. Patel
Prasad K. Deshpande
Yati Chugh
Milind C Shukla
Original Assignee
Wockhardt 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
Priority claimed from US10/128,996 external-priority patent/US6964966B2/en
Application filed by Wockhardt Limited filed Critical Wockhardt Limited
Priority to AU2002361493A priority Critical patent/AU2002361493A1/en
Priority to JP2003551131A priority patent/JP2005511743A/ja
Priority to EP02796957A priority patent/EP1453824A1/en
Priority to CA002468190A priority patent/CA2468190A1/en
Publication of WO2003050107A1 publication Critical patent/WO2003050107A1/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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

Definitions

  • the present invention relates to novel antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives and pharmaceutically acceptable salts thereof. Methods of preparation of the compounds of the invention, compositions of compounds of the invention and their use are also described.
  • the quinolones are classified as first-, second-, and third- generation compounds (Gootz T D et al, Chemistry & Mechanism of Action of the Quinolone Antibacterials. In Andriole NT ed. The Quinolones, San Francisco, Academic Press, 1998, 28- 80).
  • First-generation compounds like piromidic acid and pipemidic acid provided coverage for gram-negative Enterohacteriaceae.
  • the second-generation compounds are divided into those with enhanced but predominant gram-negative activity, against pathogens like Escherischia coli and Pseudomonas aeruginosa, and those with balanced broad-spectrum activity (norfloxacin, pefloxacin, enoxacin, fleroxacin, lomefloxacin, ciprofloxacin, ofloxacin, ru loxacin, nadifloxacin).
  • Norfloxacin, ofloxacin and ciprofloxacin have, therefore, been used mainly for treatment of diseases including urinary tract infections, gastrointestinal infections, sexually transmitted diseases and the like.
  • Third-generation drugs are those with enhanced activity against gram-positive cocci (notably clinafloxacin, sitafloxacin, trovafloxacin for Streptococcus pneumoniae) and, for essentially all the third- generation quinolones, activity also against gram- negative Haemophilus influenzae and Legionella pneumophila, and against anaerobes and atypical pathogens (Ball P, The Quinolone.
  • Levofloxacin, moxifloxacin and gatifloxacin have, therefore, found use for community-acquired infections such as those of the upper and lower respiratory tract infections (RTI) like pneumonia, sinusitis and pharyngitis, and for skin and soft tissue infections (SSI) caused by gram-positive strains of staphylococci, pneumococci, streptococci and enterococci.
  • RTI upper and lower respiratory tract infections
  • SSI skin and soft tissue infections
  • quinolone inhibition Three categories of quinolone inhibition have been suggested.
  • Type I quinolones (norfloxacin, enoxacin, fleroxacin, ciprofloxacin, lomefloxacin, trovafloxacin, grepafloxacin, ofloxacin and levofloxacin) indicated a preference for topoisomerase IN inhibition.
  • Type II quinolones (nadifloxacin and sparfloxacin) indicated a preference for D ⁇ A gyrase inhibition.
  • Type III quinolones to which some of the third-generation quinolones belong display, however, a dual-targeting property, and equally influence D ⁇ A gyrase inhibition and topoisomerase IV inhibition.
  • D ⁇ A gyrase is the primary target in bacteria, and thus is explained the weaker activity in gram-positive bacteria of the preferred topoisomerase IN-targeting second-generation quinolones like norfloxacin, ciprofloxacin, ofloxacin, and levofloxacin.
  • third generation quinolones being primarily topoisomerase IN- targeting in gram-positive staphylococci, and D ⁇ A gyrase-targeting in gram-positive S. pneumoniae, explains the advantages provided by the dual-targeting third-generation quinolones like moxifloxacin and gatifloxacin.
  • the structural feature that remains constant throughout the drug class is the bicyclic aromatic core consisting of 2 fused 6-membered rings.
  • This core can contain a carbon at the 8- position, yielding a true quinolone, or a nitrogen which provides a ring system technically termed a naphthyridone, or an additional fused ring across the N-l and C-8 positions yielding tricyclic heterocycles, such as pyridobenzoxazines and benzoquinolizidines.
  • the nature of the amine group at the 7-position takes on special relevance. It is notable that in the cited second-generation quinolones the piperazine ring remains relatively constant and undisturbed as a 7-substituent, except for alkylation on the distal nitrogen, or less frequently on the ring carbons. In the third-generation quinolones, the continuing trend of use of a C-7 cyclic amino group is also almost universal. The presence of a second amine, in addition to the nitrogen bonded to C-7 of the quinolone nucleus has been found to be important. However, amongst these new quinolones, too, the frequent employment of mainly a C-7 piperazino or pyrrolidino variant is to be noted, but with only one example of a C-7 piperidino substituent.
  • Both these antibacterial agents have an 8-methoxy substituent in the fluoroquinolone core.
  • 7-substituents in the core there is for moxifloxacin a bicyclic pyrollidine as the amino moiety, and for gatifloxacin a substituted pyrollidine as the amino moiety.
  • a more recently described olamufloxacin which has been shown to have activity in murine models of system infections and urinary tract infections, has an 8-methyl substituent in its fluoroquinolone core in which the C-7 substituent is also a substituted pyrollidine.
  • N-1, C-5, C-8 variants of the quinolone core are generally those that contribute to increase in theophylline interactions, clastogenicity, phototoxicity, hepatotoxicity, cardiotoxic ⁇ ty, arthropathy and tendonitis. Notable is the pattern of (a) the N-1 cyclopropyl and C-8 fluorine, chlorine or methoxy substituted quinolone reported to show heightened cytotoxicity (Domagala J M, J. Antimicrob.
  • C-7 pyrrolidines tend to show increased cytotoxicity over piperazino substituents, with the combination of 3-substituted pyrrolidines at C-7 and halogens at C-8 providing the most cytotoxic compounds.
  • Resistance can be defined as existence of organisms, within a population of a given microbial species, that are less susceptible to the action of a given antimicrobial agent. This resistance is of particular concern in environments such as hospitals and nursing homes, where relatively high rates of infection and intense use of antibacterials are common.
  • Recent international conferences in 2002 on infectious diseases organised by the Centres for Disease Control and Prevention, USA, World Health Organisation and other groups have highlighted emerging infectious diseases, in which the word “emerging” refers to newly discovered infectious diseases or old ones that have rebounded, turned up in new places, or become drug resistant.
  • the mechanisms of bacterial resistance to fluoroquinolones is generally believed to function by two principal categories, both resulting from chromosomal mutations (DC Hooper, Drug Resis Updat 1999; 2:38-55).
  • One category is the alterations in drug target enzymes. Fluoroquinolone resistance mutations generally occurring stepwise have been localized to specific regions of the parC sa ⁇ parE genes (grlA and grlB in S. aureus) encoding topoisomerase IN, and the gyrA and gyrB genes encoding D ⁇ A gyrase.
  • S-(-)-levofloxacin as an example of a compound in which the chiral centre is close to the quinolone nucleus, is from 8-
  • Chiral centres at C-7 that are at some distance from the quinolone nucleus are said to contribute less significantly to biological activity.
  • the relative orientation of the methyl groups on the C-7 piperazine of sparfloxacin is important for bacterial enzymes versus mammalian enzyme selectivity.
  • Sparfloxacin bearing methyl groups with a cw-stereochemistry essential for its antibacterial activity, displays dramatic differential effects on mammalian topoisomerase-II with no or less interaction with the mammalian enzyme, in contrast to the traHs-isomer which does interact with the mammalian enzyme, while however retaining its antibacterial activity (Gootz T D et al., vide infra).
  • the present invention once again surprisingly shows that stereochemical differences of substituents on the C- 7 piperidino moiety, while dramatically affecting antibacterial activity, do not significantly influence cytotoxicity of mammalian cell lines, irrespective of whether the differences are enantiomeric or diastereomeric.
  • European Patent Application 241206A2 discloses a compound having a 7-cyclic amino substituent of the formula
  • European Patent Application 0394553B1 discloses a compound for the treatment of HIV infections having a 7-cyclic amino substituent of the formula
  • each symbol is as defined in the specification of the above-mentioned publication at the 7-position of the quinolone carboxylic acid with a 4-amino substituent and a single 3 -alkyl substituent or a 3-3-dialkyl substituent claimed.
  • specific examples of the compounds having a substituent at the 7-position as piperidine of the present invention with a 4-amino or 4- hydroxy substituent with 2-alkyl, 3- alkyl, 5-alkyl or 6-alkyl substituents, or with geminal 3,3- dialkyl, or 3,5-dialkyl, or 3,3,5-trialkyl substituents, located at a position adjacent to the substituent at the 4-position are not disclosed.
  • European Patent Application 0304087A2 discloses a compound having a 7-cyclic amino substituent of the formula
  • European Patent Application 0572259A1 discloses a compound having a 7-cyclic amino substituent of the formula
  • European Patent Application 0287951A2 discloses a compound having a 7-cyclic amino substituent as in the following formula
  • R 2 is substituent which is a 5- to 9- membered saturated or unsaturated heterocyclic ring which may be substituted, wherein each symbol is as defined in the specification of the above-mentioned publication at the 7-position of the quinolone carboxylic acid with a 4-hydroxy piperidinyl moiety.
  • the present inventors have extensively studied the subject by introducing various substituted piperidine groups in the 7-position of different fluoroquinolone cores and determining the microbiological/pharmacological properties of the compounds to develop the novel substituted piperidino compounds of the invention, which (a) show a potent hitherto- undescribed antibacterial potency against broad spectrum sensitive and existing/emerging resistant pathogenic strains, including ⁇ -lactam-resistant, macrolide-resistant and even fluoroquinolone-resistant strains, mycobacteria, anaerobes and atypical pathogens (b) prevent selection of resistant bacteria by apparently inhibiting both DNA gyrase and topoisomerase IN equally, or by potently inhibiting the enzyme it targets, (c) are not subjected to efflux or have facilitated uptake, (d) do not apparently act to merely form bacteriostatic quinolone- gyrase/topoisomerase-D ⁇ A complexes to inhibit cell growth, but also apparently extend the action to release the
  • an aspect of the present invention to provide new non-chiral and chiral 7- substituted piperidino-quinolone carboxylic acid derivatives, of the formula I, as defined below, which show potent antibacterial activity against a broad range of pathogenic microorganisms, including both gram-positive and gram-negative strains with advantages of activity against resistant microorganisms, reduced toxicity, and improved pharmacology and pharmacokinetics.
  • compositions containing 7-substituted piperidino-quinolone carboxylic acid derivatives of the formula I as an active component are provided.
  • This invention describes fluoroquinolones of the fonnula I
  • Ri is C ⁇ _5 alkyl, substituted C1-5 alkyl, C 3-6 cycloalkyl, substituted C 3-6 cycloalkyl, aryl, or substituted aryl; or when Q is CH and the nitrogen atom to which Ri is linked forms an optionally substituted 5-, 6- or 7-membered ring with the carbon atom of Q, the ring optionally containing one or more hetero atoms selected from nitrogen, oxygen or sulfur atoms, said heteroatom(s) represented by
  • T preferably Ri is CH 2 CH 2 -, CH 2 T-, CH 2 CH 2 CH 2 -, CH 2 CH 2 T-, CH 2 TCH 2 -, TCH 2 T- 3
  • R 3 is hydrogen
  • R 3 is C ⁇ -C 20 alkyl, such as straight chain or branched chain aliphatic residues
  • R 3 is aralkyl
  • R 3 is CH 2 CH(NH 2 )COOH; R 3 is (CH 2 ) n -CHR ⁇ o-OCOR ⁇ 1 or (CH2VCHR10-OCO2R11 wherein R 10 is H, or CH 3 ; n is 0-3 and
  • R 11 is C ⁇ -C 20 alkyl, substituted C ⁇ -C 6 alkyl or aralkyl or Rn is
  • R 3 is ⁇ -aminoalkanoyl or an alkanoylalkyl group; or R 3 is
  • A is CH or N, and when A is CH, Z is NH or NCH 3 , and when A is N, Z is CH, 0, NH,
  • R 2 is H, Ci-20 alkyl, C 3- 6 cycloalkyl, substituted C3.6 cycloalkyl, aryl, substituted aryl, or heteroaryl, all of which heteroaryl residues may be further substituted or unsubstituted; or R 2 is an amino acid residue derived from one of the 20 naturally occurring amino acids, or the optically active isomers thereof, or the racemic mixtures thereof; R 5 is H, C 1 . 5 alkyl, C1. 5 alkoxy, amino, C ⁇ - 5 alkylamino, or C s acylamino;
  • Q is -N-, -C(R 8 )- (R 8 being H, F, CI, bromo, C ⁇ - alkyl or unsubstituted or substituted C ⁇ -4 alkoxy, wherein when the alkoxy group is substituted it is substituted by one or more halogen atoms such as F, CI, or Br), or when Q is CH and the nitrogen atom to which Ri is linked forms an optionally substituted 5-, 6- or 7-membered ring with the carbon atom of Q, the ring optionally containing one or more hetero atoms selected from nitrogen, oxygen or sulfur atoms, said heteroatom(s) represented by
  • T preferably Ri is CH 2 CH 2 -, CH 2 T-, CH 2 CH 2 CH 2 -, CH 2 CH 2 T-, CH 2 TCH 2 -, TCH 2 T-,
  • the substituent is as defined above for Ri.
  • X is OR 4 , wherein R is hydrogen, C ⁇ -C 20 alkyl, glycosyl, aralkyl, C ⁇ -C 6 alkanoyl, aminoalkanoyl or an amino acid residue derived from one of the 20 naturally occurring amino acids, or the optically active isomers thereof, or the racemic mixtures thereof, or R_ ⁇ is 1-aminocyclohexylcarbonyl or
  • R ⁇ is as hereinbefore defined or R is -(CH2) n -CHR ⁇ o-OCOOR ⁇ where R 10 and R u are as hereinbefore defined, or R 4 is C 6 HnO 6 , PO 2 (CH 3 )H, PO 3 H 2 , PO 2 (OCH 3 )H or
  • R 6 is H, Ci- 20 alkyl, C 3-6 cycloalkyl, aralkyl, C 1-2 o alkanoyl, C ⁇ - 20 alkoxycarbonyl, aralkyloxycarbonyl, amino(C ⁇ -2o)alkanoyl, or an amino acid residue derived from one of the 20 naturally occurring amino acids or the optically active isomers thereof, or the racemic mixtures thereof.
  • the amino acid residue is derived from a single amino acid or from combinations of amino acids that form dipeptide, tripeptide or polypeptide amino acid unit residues, wherein a terminal carboxy group is optionally protected by C M alkyl or aralkyl groups and a terminal amino group is optionally protected by a '-Boc (tertiarybutyloxycarbonyl), F-Moc (fluorenylmethoxycarbonyl) or Cbz (benzyloxycarbonyl) group.
  • R 6 may also be COORn wherein R ⁇ as hereinbefore defined or R 6 is C 6 Hn ⁇ 6 thus giving the gluconic acid ester derivative of the compounds.
  • R 7 is H, C ⁇ -6 alkyl, C 3-6 cycloalkyl, aralkyl; C ⁇ -6 alkanoyl, aralkyloxycarbonyl or amino(C ⁇ -2 o) alkanoyl; or an amino acid residue derived from one of the 20 naturally occurring amino acids or the optically active isomers thereof, or the racemic mixtures thereof.
  • the amino acid residue is derived from a single amino acid or from combinations of amino acids that form dipeptide, tripeptide or polypeptide amino acid unit residues, wherein a terminal carboxy group is optionally protected by C ⁇ _ alkyl or aralkyl groups and a terminal amino group is optionally protected by a '-Boc (tertiarybutyloxycarbonyl), F-Moc (fluorenylmethoxycarbonyl) or Cbz (benzyloxycarbonyl) group or R 7 may be C 6 H ⁇ i O 6 .
  • R 8 /R 8 ' are substituents at the 3/3-position of the piperidino ring and are the same or different and represent H, C ⁇ -6 alkyl, substituted C ⁇ -6 alkyl, alkylamino, or aralkyl.
  • R is a substituent at the 4-position or 5-position of the piperidino ring and represents H, C ⁇ _6 alkyl, C ⁇ -5 alkylamino, C ⁇ -3 dialkylamino, aryl, aralkyl or a trihaloalkyl.
  • This invention also includes optical isomers, diastereomers, enantiomers, polymorphs, pseudopolymorphs, pharmaceutically acceptable salts, hydrates, or biohydrolyzable esters, amides, or solvates of the fluoroquinolones of formula I and prodrugs of these compounds, hi addition, compositions incorporating the compounds of the invention, or using compounds of the invention as starting material are also contemplated in this invention.
  • the new compounds of the invention have increased potency and bactericidal activity that can be attributed to the combinations of the respective Ri, Y, R 5 , and Q substituents in the fluoroquinolone cores and the respective X, R 8 , R 8 >, and R 9 substituents on the 7-substituted piperidino moieties introduced in the cores.
  • the compounds of the invention thus belong to a new generation of dual-targeting, non-effluxed, diastereomeric, enantiomorphic antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives.
  • the compounds of the invention may be rightly called new generation triple- targeting, chiral, broad-spectrum antimicrobial agents.
  • the present invention encompasses certain compounds, dosage forms, and methods of administering the compounds, to a human or other animal subject.
  • Specific compounds and compositions to be used in the invention must, accordingly, be pharmaceutically acceptable.
  • a “pharmaceutically-acceptable” compound is one that is suitable for use with humans and/or animals without undue adverse side effects (such as toxicity, irritation, and allergic response) commensurate with a reasonable benefit/risk ratio.
  • Ri is C ⁇ - 5 alkyl being unsubstituted or substituted with from 1 to 3 fluoro atoms, C 3-6 cycloalkyl being unsubstituted or substituted with from 1 to 2 fluoro atoms, or aryl being unsubstituted or substituted with from 1 to 3 fluoro atoms; or when Q is CH and the nitrogen atom to which Ri is linked forms an optionally substituted 5-, 6- or 7-membered ring with the carbon atom of Q, the ring optionally containing one or more hetero atoms selected from nitrogen, oxygen or sulfur atoms, said heteroatom(s) represented by T, preferably Ri is CH 2 CH 2 -, CH 2 T-, CH 2 CH 2 CH 2 -, CH 2 CH 2 T-, CH 2 TCH 2 -, TCH 2 T-, TCH 2 CH 2 CH 2 CH 2 - CH 2 CH 2 T-, CH 2 TCH 2 CH 2- , or TCH 2 CH 2 T- where T represents NH, O, or
  • This 5- to 7- membered ring may be substituted with 1 or 2 of the same substituents as those defined above for R ls preferably by one C1-C5 alkyl group.
  • Y is OR 3 where R 3 is hydrogen; R 3 is C ⁇ -C 20 alkyl, such as straight chain or branched chain aliphatic residues such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl or their branched chain isomers;
  • R 3 is aralkyl such as benzyl, phenethyl, or phenylpropyl; R 3 is CH 2 CH(NH 2 )COOH;
  • R 3 is (CH 2 )n-CHR ⁇ 0 -OCOR ⁇ or (CH 2 ) n -CHR ⁇ 0 -OCO 2 R u wherein R 10 is H, or CH 3 ; n is 0-3 and Ru is C1-C20 alkyl as hereinbefore defined, or substituted C ⁇ -C 6 alkyl with substituents such as hydroxy, halogen, amino, or mercapto; or aralkyl such as benzyl; phenethyl, phenylpropyl or Rn is
  • R 3 is ⁇ -aminoalkanoyl such as ⁇ -aminopropionyl or R 3 is alkanoylalkyl group such as acetoxymethyl, acetoxyethyl, pivaloyloxy-methyl, or pivaloyloxyethyl group; or R 3 is
  • A is CH or N, and when A is CH, Z is NH or NCH 3 , and when A is N, Z is CH, O, NH, S, or NCH 3 ;
  • p is 0 - 2 ;
  • q is 0 - 2, preferably it is a group such as N-methylpiperidin-4-yl, pyrrolidin-2-yl-ethyl, piperidin-2-yl-ethyl, or morpholin-2-yl-ethyl; or
  • Y is NHR 2 , wherein R2 is H, C ⁇ -20 alkyl such as straight chain or branched chain aliphatic residues as defined above, C 3-6 cycloalkyl, substituted C 3-6 cycloalkyl wherein the substituent is C ⁇ - 2 alkyl such as methyl or ethyl or trifluoroalkyl such as trifluoromethyl or halogen such as fluorine, chlorine, bromine or R 2 is aryl such as unsubstituted or substituted phenyl wherein the substituent is C ⁇ -3 alkyl, C1.
  • heteroaryl such as pyridyl, pyrimidinyl, quinolinyl, isoquinolinyl, furyl, oxazolinyl, thiazolyl, or thiadiazolyl, all of which heteroaryl residues may be further substituted or unsubstituted, wherein the substituent is methyl or ethyl; or R 2 is an amino acid residue derived from one of the 20 naturally occurring amino acids viz.
  • R5 is H, C ⁇ -5 alkyl, Ci-s alkoxy, amino, C ⁇ -5 alkylamino such as-NHCH 3 , N(CH 3 ) 2 , and the like; or acylamino such as -NHCOCH3, -NHCOC(CH 3 ) 3 , and the like;
  • Q is -N-, -C(R 8 )- (R 8 being H, F, CI, bromo, methoxy, C ⁇ -4 alkyl, or unsubstituted or substituted C ⁇ -4 alkoxy, wherein when the alkoxy is substituted it is substituted by one or more halogen atoms such as F, CI, or Br), or when Q is CH and the nitrogen atom to which R x is linked forms an optionally substituted 5-, 6- or 7-membered ring with the carbon atom of Q, the ring optionally containing one or more hetero atoms selected from nitrogen, oxygen or sulfur atoms, said heteroatom(s) represented by T, preferably Rj is CH 2 CH 2 -, CH 2 T-, CH 2 CH 2 CH 2 -, CH 2 CH 2 T-, CH 2 TCH 2 -, TCH 2 T-, TCH 2 CH 2 CH 2 CH 2 - CH 2 CH 2 T-, CH 2 TCH 2 CH 2-) or TCH CH 2 T- where
  • the substituent is as defined above for R x .
  • This 5- to 7- membered ring may be substituted with 1 or 2 of the same substituents as those defined above for R l9 preferably by one C 1 -C 5 alkyl group.
  • X is OI t wherein R4 is hydrogen, C ⁇ -C 20 alkyl as hereinbefore defined, glycosyl, aralkyl such as benzyl; or Ci-C 6 alkanoyl such as acetyl, propionyl, pivaloyl, stearoyl, or nonadecanoyl or aminoalkanoyl such as aminoacetyl, aminopropionyl and the like or an amino acid residue derived from one of the 20 naturally occurring amino acids viz.
  • R4 is hydrogen, C ⁇ -C 20 alkyl as hereinbefore defined, glycosyl, aralkyl such as benzyl; or Ci-C 6 alkanoyl such as acetyl, propionyl, pivaloyl, stearoyl, or nonadecanoyl or aminoalkanoyl such as aminoacetyl, aminopropionyl and the like or an amino acid residue derived from one of the 20 naturally occurring amino acids viz.
  • R4 is 1- aminocyclohexylcarbonyl or COORn wherein R ⁇ is as hereinbefore defined or R 4 is -(CH2) n - CHRio-OCOOR ⁇ where Rio and Ru are as hereinbefore defined, or R 4 is C 6 HnO 6 , PO2(CH 3 )H, PO 3 H 2 , PO (OCH 3 )H or SO 3 H thus giving respectively the gluconic acid, phosphonic acid, phosphoric acid and sulfonic acid ester derivatives of the compounds; or X is NR 6 R
  • the amino acid residue is derived from alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine or valine.
  • the amino acid residue is derived from a single amino acid or from combinations of amino acids that form dipeptide, tripeptide or polypeptide amino acid unit residues wherein a terminal carboxy group is optionally protected by C ⁇ - alkyl or aralkyl groups and a terminal amino group is optionally protected by a l -Boc (teritarybutyloxycarbonyl), F-Moc (fluorenylmethoxycarbonyl) or Cbz (benzyloxycarbonyl) group or R 6 may also be COORn wherein Ru is as hereinbefore defined or R 6 is CeHnOo thus giving the gluconic acid ester derivative of the compounds.
  • R 7 is H, Ci-6 alkyl as hereinbefore defined, C 3 - 6 cycloalkyl, aralkyl such as benzyl, phenethyl, or phenylpropyl; C ⁇ -6 alkanoyl such as COCH 3 , COCH 2 CH 3 , COC(CE_ 3 ) 3 , aralkyloxycarbonyl such as benzyloxycarbonyl or amino (C ⁇ - 2 o)alkanoyl such as aminoacetyl, aminopropionyl, etc.; or an amino acid residue derived from one of the 20 naturally occurring amino acids or the optically active isomers thereof, or the racemic mixtures thereof.
  • the amino acid residue is derived from alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine or valine.
  • the amino acid residue is derived from a single amino acid or from combinations of amino acids that fonn dipeptide, tripeptide or polypeptide amino acid unit residues, wherein a terminal carboxy group is optionally protected by C ⁇ -4 alkyl or aralkyl groups and a terminal amino group is optionally protected by a l -Boc (teritarybutyloxycarbonyl), F-Moc (fluorenylmethoxycarbonyl) or Cbz (benzyloxycarbonyl) group or R 7 maybe CeHnO 6 thus giving the gluconic acid ester derivative of the compounds.
  • R 8 /R 8 ' are substituents at the 3/3-position of the piperidino ring and are the same or different and represent H, C ⁇ -6 alkyl, substituted Ci- ⁇ alkyl wherein the substituent is amino, hydroxy, halogen such as one or more fluorine, chlorine, or bromine atoms; alkylamino, or aralkyl such as benzyl.
  • R 9 is a substituent at the 4-position or 5-position of the piperidino ring and represents H, Ci- ⁇ alkyl, C 1 - 5 alkylamino, C 1 - 3 dialkylamino or aryl, aralkyl such as benzyl or phenethyl or a trihaloalkyl such as trifluoromethyl.
  • aryl is substituted or unsubstituted phenyl.
  • the phenyl, alkyl, and cycloalkyl group may be substituted at one or more positions by the usual aromatic substituents such as halogen namely F, CI, or Br; alkyl such as methyl, ethyl, trifluoromethyl, etc.
  • Substituted phenyl groups include such as halophenyl, trifluoromethylphenyl, monofluorophenyl, 2-fluorophenyl, 4- fluorophenyl, or 2,4 difluorophenyl.
  • This invention also includes optical isomers, diastereomers, enantiomers, polymorphs, pseudopolymorphs, pharmaceutically acceptable salts, hydrates, or biohydrolyzable esters, amides, imides, or solvates of the fluoroquinolones of formula I and prodrugs of these compounds.
  • a pseudopolymorph is a polymorph that differs from a true polymorph by the incorporation of solvent.
  • the compounds of this invention are effective antimicrobial agents which are a new generation of antibacterial agents, in particular a new generation of respiratory antibacterials, effective against multidrug- resistant pathogens with broad spectrum coverage of gram-positive and gram-negative microbes, such as sensitive and fluoroquinolone-resistant pneumococci, staphylococci, streptococci, anaerobes, enterococci and atypical pathogens, hi addition, the compounds of the invention have potent cidal action for fluoroquinolone-resistant strains.
  • the compounds of the invention have the preferced potential to address the unmet need for orally effective drugs for the treatment of multidrug-resistant pneumococcal infections like life-threatening pneumoniae and meningitis, to which pediatric and geriatric patients are vulnerable. They are unusually cidal for viridans streptococci, which are the causative groups of strains responsible for bacteremias, soft tissue infections, abscesses, sepsis and endocarditis. They are potential antitubercular agents against sensitive and resistant mycobacteria.
  • the above described physicochemical parameters also contribute to their unusually favourable drugability properties. They are orally effective with once-a-day potential. They have favourable penetration into tissues like the lung, liver, kidney and heart over serum thus enabling the targeting of organ-specific infections. They are relatively non-phototoxic, with favourable cytotoxicity and cardiotoxicity profiles which are usually the problem toxicities displayed by the fluoroquinolone class of compounds.
  • optically active compounds and diastereomeric isomers each having the substituent in a specific stereo and three-dimensional spatial orientation have both excellent antibacterial activity and high safety features.
  • the compounds of the invention are sufficiently basic to form acid addition salts.
  • the compounds are useful both in the free base fonn and the form of acid addition salts, and both forms are within the purview of the invention.
  • the acid addition salts are in some cases a more convenient form for use.
  • acids addition salts include, but are not limited to acetate, benzenesulfonate, fumarate, hydrochloride, hydrobromide, hydroiodide, hydrogensulfate, isethionate, lactate, malate, maleate, malonate, methanesulfonate, pamoate (embonate), phosphate/diphosphate, stearate, succinate, sulfate, tartrate, trifluoroacetate, trifluoromethanesulfonate, p-toluenesulfonate, and the like.
  • Preferred acid addition salts include halides, sulfonates, carboxylates, phosphates, and the like.
  • amino acid maybe selected from one of the 20 naturally occurring amino acids: alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine or valine or the optically active isomers thereof or the racemic mixtures thereof or dipeptides, tripeptides and polypeptides derived from the monoaminoacid units thereof.
  • alkali/base addition salts include the alkali metal salts (such as sodium and potassium), alkaline earth metal salts (such as magnesium and calcium), inorganic salts, such as ammonium, substituted ammonium, choline and organic base salts from basic amines such as diethanolamine, ethylenediamine, guanidine or heterocyclic amines such as piperidine, hydroxyethylpyrrolidine, hydroxyethylpiperidine, morpholine, piperazine, N-methyl piperazine and the like or basic amino acids such as optically pure and racemic isomers of arginine, lysine, histidine, tryptophan and the like.
  • Acids used to prepare acid addition salts include preferably those, which produce, when combined with the free base, pharmaceutically acceptable salts. These salts have anions that are relatively innocuous to the animal organism, such as a mammal, in pharmaceutical doses of the salts so that the beneficial property inherent in the free base are not vitiated by any side effects ascribable to the acid's anions.
  • the pharmaceutically acceptable acid addition salts of compounds of the formula I are prepared in a conventional manner by treating a solution or suspension of the free base of formula I with about one chemical equivalent of a pharmaceutically acceptable acid such as an inorganic acid or organic acid.
  • a pharmaceutically acceptable acid such as an inorganic acid or organic acid.
  • Conventional concentration and recrystallization techniques are employed in isolating the salts.
  • the free base can be dissolved in an aqueous alcohol solution containing the appropriate acid and the salt is isolated by evaporation of the solution.
  • they may be prepared by reacting the free base with an acid in an organic solvent so that the salt separates directly. Where separation of the salt is difficult, it can be precipitated with a second organic solvent, or can be obtained by concentration of the solution.
  • all acid addition salts are within the scope of the present invention. All acid addition salts are useful as sources of the free base form, even if the particular salt per se is desired only as an intermediate product. For example, when the salt is formed only for purposes of purification or identification, or when it is used as an intermediate in preparing a pharmaceutically acceptable salt by ion exchange procedures, these salts are clearly contemplated to be a part of this invention.
  • the amino moiety of piperidine is a potential point of formation for the subject compounds of a pharmaceutically acceptable anionic salt; such salts are included in the subject invention compounds.
  • Preferred salts are acid addition salts with, for example, HCl, CH 3 COOH, CH 3 SO 3 H, HCOOH, CF 3 COOH, gluconic acid, Ci- 20 straight chain or branched alkanoic acids or one of the 20 naturally occurring amino acids as hereinbefore defined or dipeptide, tripeptide or polypeptide derivatives of the monoaminoacid units thereof.
  • “Host” is a substrate capable of sustaining a microbe, preferably it is a living organism, and most preferably an animal, more preferably a mammal, and more preferably still a human.
  • Biohydrolyzable amides are aminoacyl, acylamino, or other amides of the compounds of the invention, where the amide does not essentially interfere, preferably does not interfere, with the activity of the compound, or where the amide is readily converted in vivo by a host to yield an active compound.
  • Biohydrolyzable imides are imides of the compounds of the invention, where the imide does not essentially interfere, preferably does not interfere, with the activity of the compound, or where the imide is readily converted in vivo by a host to yield an active compound.
  • Preferred imides are hydroxyimides.
  • Biohydrolyzable esters are esters of the compounds of the invention, where the ester does not essentially interfere, preferably does not interfere, with the antimicrobial activity of the compound, or where the ester is readily converted in a host to yield an active compound. Many such esters are known in the art.
  • esters include lower alkyl esters, lower acyloxy-alkyl esters (such as acetoxymethyl, acetoxyethyl, aminocarbonyloxymethyl, pivaloyloxylmethyl and pivaloyloxylethyl esters); lactonyl esters (such as phthalidyl and thiophthalidyl esters) lower alkoxyacyloxyalkyl esters (such as methoxycarbonyloxymethyl, ethoxycarbonyloxyethyl and isopropoxycarbonyloxyethyl esters), alkoxyalkyl esters, choline esters and alkylacylaminoalkyl esters (such as acetamidomethyl esters) and alkyl amino acid esters.
  • lower alkyl esters such as acetoxymethyl, acetoxyethyl, aminocarbonyloxymethyl, pivaloyloxylmethyl and pivaloyloxylethyl esters
  • Optical isomer "Optical isomer”, “stereoisomer”, “diastereomer” "polymorph” “pseudopolymorph”, “hydrates” and “solvates” as referred to herein have the standard art recognized meanings. Solvates are generally formed during the process of crystallization when molar or submolar amounts of the solvents get incorporated into the crystal structure of the compound.
  • the compounds of the invention may contain chiral center(s), thus any such compound includes and contemplates each optical isomer, diastereomer or enantiomer thereof, in purified or substantially purified form, and mixtures thereof, including racemic mixtures.
  • the compounds of the invention may have one or more chiral centers. As a result, one may selectively prepare one optical isomer, including diastereomer and enantiomer, over another, for example by use of chiral starting materials, catalysts or solvents, one may prepare both stereoisomers or both optical isomers, including diastereomers and enantiomers at once (a racemic mixture).
  • the compounds of the invention may exist as racemic mixtures, mixtures of optical isomers, including diastereomers and enantiomers, or stereoisomers, they may be separated using known methods, such as chiral resolution, chiral chromatography and the like.
  • one optical isomer, including diastereomer and enantiomer, or stereoisomer may have favorable properties over the other.
  • a racemic mixture when one racemic mixture is disclosed, it is clearly contemplated that both optical isomers, including diastereomers and enantiomers, or stereoisomers substantially free of the other are disclosed and claimed as well.
  • a quinolone derivative includes prodrugs of a quinolone.
  • the preferred compounds of the invention are those compounds of Formula I which are composed of on one hand the following core fluoroquinolone moieties displayed below minus the respective 7-amino substituent:
  • R 5 H, CH 3 , orNH 2 ;
  • R 8 H, or CH 3;
  • R 9 H, or CH 3; and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • R 5 H, CH , or H 2 ;
  • R 9 H, or CH 3; and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • R 5 H, CH 3 , orNH 2;
  • R 8 H, or CH 3;
  • R 9 H, or CH 3; and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • Q C-H, C-OCH 3 , C-F or N, and when Q is CH and the nitrogen atom to which Ri is linked forms substituted 6 membered ring with the carbon atom of Q and Ri are C-CH 2 CH 2 C*H(CH 3 ) or C-OCH 2 CH(CH 3 );
  • Ri C-C 3 H 5 , or C 6 H 3 F 2 (2,4), and when Q is CH and the nitrogen atom to which Ri is linked forms substituted 6 membered ring with the carbon atom of Q and Ri are C-CH 2 CH 2 C*H(CH 3 ) or C-OCH 2 CH(CH 3 );
  • R 5 H, CH 3 , or NH 2;
  • R 8 H, or CH 3;
  • R 9 - H or CH 3; and optical isomers, phannaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • R 5 H, CH 3 , orNH 2;
  • R 8 H, or CH 3;
  • R 9 H, or CH 3; and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • R 8 H,CH 3 ,orC 2 H 5 , and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • Q C-OCH 3 , or C-F, and when Q is CH and the nitrogen atom to which Ri is linked forms substituted 6 membered ring with the carbon atom of Q and Ri is C-OCH 2 CH(CH 3 );
  • Ri C-C 3 H 5 and when Q is CH and the nitrogen atom to which Ri is linked forms substituted 6 membered ring with the carbon atom of Q and Ri is C-OCH 2 CH(CE ⁇ 3 );
  • R 5 H, or NH 2
  • R 8 H, or CH 3; and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • R 5 H, or NH 2 ,
  • R 8 H, or CH 3 , and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • Q C-OCH 3 ;
  • R 5 H, orNH 2;
  • R 8 H, or CH 3; and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • R 5 H, or H 2
  • R 6 COCH 3) CO-(O-C 2 -C 4 alkyl), COOCH 2 C 6 H 5 , amino C M alkanoyl, carboxy amino C M alkanoyl, dipeptidoalkanoyl wherein the terminal amino group is unprotected or protected with a '-Boc-protecting group.
  • Y NH 2 , NHR 2 , wherein R 2 is H, C ⁇ -2 alkyl; or Y is OR 3 where R 3 is C M alkyl (branched or unbranched); CH 2 C 6 H 5; C 2 -C 3 carboxylic acid and their C ⁇ -C alkyl esters (unbranched or branched); or five or six member heterocycle (unsubstituted or substituted); or oc-aminoalkanoyl such as oc -aminopropionyl; and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • Q C-(C ⁇ -C 2 ) alkyl, C-OCH 3 , C-F or N, and when Q is CH and the nitrogen atom to which Ri is linked forms substituted 6 membered ring with the carbon atom of Q and Ri is C-
  • Ri C H 5 , C-C 3 H 5 , or C 6 H 3 F 2 (2,4), and when Q is CH and the nitrogen atom to which Ri is linked forms substituted 6 membered ring with the carbon atom of Q and Ri is C-
  • R 5 H, orNH 2 ,
  • R 8 H, C 3 -C alkyl (unbranched or branched), CH 2 C 6 H 5 , or CF 3 , and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • Q C-H, C-(C ⁇ -C 2 ) alkyl, C-OCH 3 , C-F or N, and when Q is CH and the nitrogen atom to which
  • Ri is linked forms substituted 6 membered ring with the carbon atom of Q and Ri are C-
  • Ri C 2 H 5 , C-C 3 H 5 , or C 6 H 3 F 2 (2,4), and when Q is CH and the nitrogen atom to which Ri is linked forms substituted 6 membered ring with the carbon atom of Q and Ri are C-
  • R 5 H, or NH 2
  • R 8 H, CH 3 , or C 2 H 5>
  • R 9 H, CH 3 , or C 2 H 5 , and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • Q C-H, C-(C ⁇ -C 2 ) alkyl, C-OCH 3 , C-F or N, and when Q is CH and the nitrogen atom to which
  • Ri is linked fonns substituted 6 membered ring with the carbon atom of Q and Ri is C-
  • Ri C ⁇ _ 2 alkyl, C-C 3 H 5 , or CeH 3 F 2 (2,4), and when Q is CH and the nitrogen atom to which Ri is linked forms substituted 6 membered ring with the carbon atom of Q and Ri is C-
  • R 5 H, orNH 2
  • R 9 H, or C 2 H 5; and optical isomers, pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • Ri C 2 H 5 , C-C3H5, or C- C 6 H 3 F 2 (2,4); and pharmaceutically acceptable salts, hydrates, biohydrolyzable esters, polymorphs and pseudomorphs thereof.
  • Particularly preferred compounds of the invention are those where Q is C-OCH 3 and C-CH 3 .
  • Q is C-OCH 3 and C-CH 3 .
  • a list of these preferred compounds are given below under the heading of specific compounds of the invention.
  • 150 5 -Amino- 1 -cyclopropyl-6-fluoro- 1 ,4-dihydro-8-methoxy-7-(4-hydroxy-4-methyl- 1 - piperidinyl)-4-oxo-quinoline-3-carboxylic acid and its isomers and its salts; 151. l-Cyclopropyl-6-fluoro-l,4-dihydro-8-methoxy-7-(4-hydroxy-4-trifluoro methyl-1- piperidinyl)-4-oxo-quinoline-3-carboxylic acid and its isomers and its salts;
  • 201 cis- 1 -(2,4-Difluorophenyl)-6-fluoro-8-ethyl-7-(4-hydroxy-3-methyl- 1 -piperidinyl)- 1 ,4- dihydro-4-oxo-quinoline-3-carboxylic acid and its isomers and its salts; 202. tians-l-(2,4-Difluorophenyl)-6-fluoro-8-ethyl-7-(4-hydroxy-3-methyl-l-piperidinyl)-l,4- dihydro-4-oxo-quinoline-3-carboxylic acid and its isomers and its salts;
  • Another embodiment of the invention encompasses a process to make the compounds of the invention, which comprises the following general methods.
  • General Methods In general, the fluoroquinolone compounds were prepared by heating the appropriate 6,7-dihalo fluoroquinolone core moiety (bearing either a free 3 carboxylic acid or a 3 carboxylic acid ester or an O 3 O 4 borane chelate) or the appropriate 8,9-dihalo fluoroquinolone core moiety (bearing an O-B-diacetoxy borane) with the appropriate, 4-substituted/unsubstituted amino/hydroxy-3 - substituted/unsubstituted piperidine moiety in an organic solvent, optionally in the presence of a base at 50° - 120°C, preferably 50 - 90°C for 4 - 72 hr preferably 16 - 24 hr and isolating the product.
  • a base at 50° - 120°C, preferably 50 - 90°C for 4
  • Suitable solvents include acetone, alcohol, acetonitrile, dimethyl sulphoxide, N,N- dimethylformamide, preferably acetonitrile or dimethyl sulphoxide.
  • Suitable bases include triethylamine, pyridine, l,5-diazabicyclo[4.3.0]non-5-ene (DBN), l,7-diazabicyclo[5.4.0]undec- 7-ene (DBU), preferably triethylamine.
  • the protecting group is removed by treatment with aqueous alkali or inorganic acid at 30 - 120 °C, preferably 30 - 80 °C for 4 — 12 hours, preferably 4 - 6 hours and isolating the product.
  • Method 1 l-Cyclopropyl-6-fluoro-7- ⁇ (4-amino/substituted amino /disubstituted amino/hydroxy-3 - alkyl/3,3-dialkyl/3,5-dialkyl)l-l-piperidinyl)-l,4-dihydro-4-oxo-quinoline-3-carboxylic acid and isomers were prepared by heating a mixture of l-cyclopropyl-6-fluoro-7-chloro-l,4-dihydro-4- oxo-quinoline-3-carboxylic acid with appropriate ⁇ 4-(amino/ substituted amino/disubstituted amino/hydroxy -3-alkyl/3,3-dialkyl/3,5-dialkyl/3,3,5-trialkyl) ⁇ piperidine in an organic solvent optionally in the presence of a base at 50° - 120°C, preferably 90°C for 4- 72 hr.
  • the solvent may be selected from acetone, alcohol, acetonitrile, dimethyl sulphoxide, N,N- dimethylformamide preferably acetonitrile or dimethyl sulphoxide.
  • the base may be selected from triethylamine, pyridine, l,5-diazabicyclo[4.3.0]non-5-ene (DBN), or 1,7- diazabicyclo[5.4.0]undec-7-ene (DBU), preferably triethylamine.
  • Method 2 l-Cyclopropyl-6-fluoro-8-methoxy-7- ⁇ (4-amino/substituted amino/ disubstituted amino/ hydroxy -3-alkyl/3,3-dialkyl/3,5-dialkyl/3,3,5-trialkyl)-l-piperidinyl)-l,4-dihydro-4-oxo- quinoline-3 -carboxylic acid and isomers were prepared by a procedure described in Method 1 by using [ 1 -cyclopropyl-6,7-difluoro-8-methoxy- 1 ,4-dihydro-4-oxo-quinoline-3-carboxylate-
  • Method 4 9-Fluoro-5-methyl-6,7-dihydro-8-(3/4/5-substituted-4-hydroxyl-l-pi ⁇ eridinyl)-l-oxo-lH,5H- benzo[i,j]quinolizine-2-carboxylic acid its isomers were prepared by a procedure described in Method 1 by using (0-B)-diacetoxy- ⁇ S-(-)-8,9-difluoro-5-methyl-6,7-dihydro-l-oxo-lH,5H- benzo[i,j] quinolizine-2-carboxy ⁇ borane and appropriate 3/4/5 -substituted-4-hydroxy piperidine and optionally hydrolyzing the obtained boron complex in the presence of a base.
  • Method 5 l-Cyclopropyl-6-fluoro-7- ⁇ (4-amino/substituted amino/ disubstituted amino/ hydroxy -3- alkyl/3 ,3 -dialkyl/3 ,5-dialkyl)l- 1 -piperidinyl)- 1 ,4-dihydro-4-oxo- 1 ,8-naphthyridone-3-carboxylic acid and isomers were prepared by a procedure described in Method 1 by using 1-cyclopropyl- 7-chloro-6-fluoro-l,4-dihydro-4-oxo-l,8-naphthyridone-3-carboxylic acid and appropriate ⁇ 4- amino/ substituted amino/disubstituted amino/hydroxy-3-alkyl/3,3-dialkyl/3,5- dialkyl) ⁇ piperidine.
  • Method 6 l-(2,4-Difluorophenyl)-6-fluoro-7- ⁇ (4-amino/substituted amino/ disubstituted amino/ hydroxy-3- alkyl/3,3-dialkyl/3,5-dialkyl)l-l-piperidinyl)-l,4-dihydro-4-oxo-l,8-naphthyridone-3-carboxylic acid and isomers were prepared by a procedure described in Method 1 by using ethyl-1- (difluorophenyl)-7-chloro-6-fluoro- 1 ,4-dihydro-4-oxo- 1 ,8 -naphthyridone-3 -carboxylate and appropriate ⁇ 4-amino/ substituted amino/disubstituted amino/hydroxy-3-alkyl/3,3-dialkyl/3,5- dialkyl ⁇ piperidine.
  • Method 8 l-Cyclopropyl-6-fluoro -8-methyl-7- ⁇ 4-amino/substituted amino/ disubstituted amino/ hydroxy - 3-alkyl/3,3-dialkyl-l-piperidinyl ⁇ - l,4-dihydro-4-oxo-quinoline-3 -carboxylic acid and isomers were prepared by a procedure described in Method 1 by using [l-cyclopropyl-6,7-difluoro-8- methyl-l,4-dihydro-4-oxo-quinoline-3-carboxylate-O 3 ,O ] difluoroboron chelate and appropriate ⁇ 4-amino/ substituted amino/disubstituted amino/hydroxy-3-alkyl/3,3- dialkyl ⁇ piperidine and optionally hydrolyzing the obtained boron complex in the presence of a base.
  • Method 9 l-Cyclopropyl-6-fluoro -8-ethyl-7- ⁇ 4-amino/substituted amino/ disubstituted amino/ hydroxy -3- alkyl/3,3-dialkyl-l-piperidinyl ⁇ - l,4-d_hydro-4-oxo-quinoline-3-carboxylic acid and isomers were prepared by a procedure described in Method 1 by using [l-cyclopropyl-6,7-difluoro-8- ethyl-l,4-dihydro-4-oxo-quinoline-3-carboxylate-O 3 ,O 4 ] difluoroboron chelate and appropriate ⁇ 4-amino/ substituted amino/disubstituted amino/ hydroxy -3-alkyl/3,3-dialkyl ⁇ piperidine and optionally hydrolyzing the obtained boron complex in the presence of a base.
  • Method 11 l-Ethyl-6-fluoro-8-methyl-7- ⁇ 4-amino/substituted amino/ disubstituted amino/ hydroxy -3- alkyl/3,3-dialkyl-l-piperidinyl ⁇ -l,4-dihydro-4-oxo-quinoline-3-carboxylic acid and isomers were prepared by a procedure described in Method 1 by using [l-ethyl-6,7-difluoro -8-methyl- l,4-dihydro-4-oxo-quinoline-3-carboxylate-O 3 ,O 4 ]difluoroboron chelate and appropriate ⁇ 4- atnino/ substituted amino/disubstituted amino/ hydroxy -3-alkyl/3,3-dialkyl ⁇ pi ⁇ eridine and optionally hydrolyzing the obtained boron complex in the presence of a base.
  • Method 12 l,8-Diethyl-6-fluoro-7- ⁇ 4-amino/substituted amino/ disubstituted amino/ hydroxy -3-alkyl/3,3- dialkyl-l-piperidinyl ⁇ -l,4-dihydro-4-oxo-quinoline-3-carboxylic acid and isomers were prepared by a procedure described in Method 1 by using [l,8-diethyl-6,7-difluoro -l,4-dihydro-4-oxo- quinoline-3-carboxylate-O 3 ,O 4 ]difluoroboron chelate and appropriate ⁇ 4-amino/ substituted amino/disubstituted amino/ hydroxy -3-alky_/3,3-dialkyl ⁇ piperidine and optionally hydrolyzing the obtained boron complex in the presence of a base.
  • Method 13 l-(2,4-Difluorophenyl)-6-fluoro-8-methyl-7- ⁇ 4-amino/substituted amino/ disubstituted amino/ hydroxy -3-alkyl/3,3-dialkyl-l-piperidinyl ⁇ -l,4-dihydro-4-oxo-quinoline-3-carboxylic acid and isomers were prepared by a procedure described in Method 1 by using [l-(2,4-Difluorophenyl)- 6,7-difluoro -8-methyl-l,4-dihydro-4-oxo-quinoline-3-carboxylate-O3,O4]difluoroboron chelate and appropriate ⁇ 4-amino/ substituted amino/disubstituted amino/ hydroxy -3-alkyl/3,3- dialkyl ⁇ piperidine and optionally hydrolyzing the obtained boron complex in the presence of a base.
  • Method 14 l-(2,4-Difluorophenyl)-6-fluoro-8-ethyl-7- ⁇ 4-amino/substituted amino/ disubstituted amino/ hydroxy -3-alkyl/3,3-dialkyl-l-piperidinyl ⁇ -l,4-dihydro-4-oxo-quinoline-3-carboxylic acid and isomers were prepared by a procedure described in Method 1 by using [l-(2,4-Difluorophenyl)- 6,7-difluoro-8-ethyl-l,4-dihydro-4-oxo-quinoline-3-carboxylate-O3,O4] difluoroboron chelate and appropriate ⁇ 4-amino/ substituted amino/disubstituted amino/ hydroxy -3-alkyl/3,3- dialkyl ⁇ iperidine and optionally hydrolyzing the obtained boron complex in the presence of a base.
  • (+)-4- t -Butyloxycarbonylamino -3,3-dimethylpiperidine (can be prepared according to
  • (+)-4- t -Butyloxycarbonylamino -3,3-dimethylpiperidine prepared according to Preparation 19, step 7
  • (-)-4- t -Butyloxycarbonylamino-3,3-dimethylpiperidine prepared according to Preparation 19, step 7
  • the invention also provides novel intermediate amines that can be used to prepare the amines listed above.
  • novel intermediate amines are listed below and the methods for their preparation are included in parenthesis alongside each one of them as follows:
  • the intermediates can be classified as (a) the unprotected dialkyl or trialkyl 4-piperidones, (b) the dialkyl or trialkyl 1-N protected 4-piperidones and (c) the dialkyl 1-N protected, 4-protected amino piperidines.
  • the unprotected dialkyl or trialkyl 4-piperidone intermediates can be used to prepare the corresponding amines by using the known art of transforming a carbonyl functionality to an amine functionality by using reagents such as ammonium acetate, methyl amine hydrocholide, ethyl amine hydrochloride, cyclopropyl amine etc to form an imino functionality which upon subsequent reduction can produce desired amine after reduction.
  • the reducing agents can be selected from sodium cyano borohydride or palladium on carbon in a solvent such as methanol, ethanol, ethyl acetate etc.
  • the dialkyl or trialkyl 1-N protected 4-piperidones are used to prepare the corresponding deprotected amines by using the known art of deprotection such as hydrogenolysis to deprotect a benzyloxycarbonyl moiety or a benzyl moiety by stirring the intermediate in the presence of catalysts and a hydrogen source.
  • the catalyst can be selected from palladium on carbon, palladium hydroxide on carbon and hydrogen source can be selected from hydrogen gas or ammonium formate, cyclohexene, in solvents such as methanol ethanol, ethyl acetate etc.
  • dialkyl 1-N protected, 4-protected amino piperidines can be used to prepare the corresponding deprotected amines by using the known art of deprotection as described in the previous paragraph to remove a benzyloxycarbonyl or a benzyl moiety and the known art of removing a butyloxycarbonyl group by stirring the intermediate, in the presence of inorganic acids such as concentrated hydrochloric acid or organic acids such as trifluoro acetic acid in solvents such as tetrahydrofuran, dioxane etc.
  • inorganic acids such as concentrated hydrochloric acid or organic acids such as trifluoro acetic acid in solvents such as tetrahydrofuran, dioxane etc.
  • the preparations 1 - 38 include examples of the specific preparative methods used of how to use the described amine intermediates for the preparation of the conesponding novel amines of the invention.
  • the pharmaceutically acceptable acid addition salts of compounds of the fonnula I are prepared in a conventional manner by treating a solution or suspension of the free base of the formula I with about one chemical equivalent of a pharmaceutically acceptable acid such as an inorganic acid or organic acid.
  • a pharmaceutically acceptable acid such as an inorganic acid or organic acid.
  • concentration and recrystallization techniques are employed in isolating the salts.
  • the free base can be dissolved in an aqueous alcohol solution containing the appropriate acid and the salt is isolated by evaporation of the solution.
  • they may be prepared by reacting the free base with an acid in an organic solvent so that the salt separates directly. Where separation of the salt is difficult, it can be precipitated with a second organic solvent, or can be obtained by concentration of the solution.
  • acids addition salts include, but are not limited to acetate, benzenesulfonate, fiunarate, hydrochloride, hydrobromide, hydroiodide, hydrogensulfate, isethionate, lactate, malate, maleate, malonate, methanesulfonate, pamoate (embonate), phosphate/diphosphate, stearate, succinate, sulfate, tartrate, trifluoroacetate, trifluoromethanesulfonate, p-toluenesulfonate, and the like.
  • Prefened acid addition salts include halides, sulfonates, carboxylates, phosphates, and the like.
  • amino acid may be selected from one of the 20 naturally occurring amino acids: alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine or valine or the optically active isomers thereof or the racemic mixtures thereof or dipeptides, tripeptides and polypeptides derived from the monoaminoacid units thereof.
  • alkali/base addition salts of compounds of formula I may be prepared by conventional methods from the conesponding acids e.g. by reaction with about one equimolar amount of an alkali/base.
  • Preferred alkali base addition salts include the alkali metal salts (such as sodium and potassium), alkaline earth metal salts (such as magnesium and calcium), inorganic salts, such as ammonium, substituted ammonium, choline and organic base salts from basic amines such as diethanolamine, N-methyl glucamine, ethylenediamine, guanidine or heterocyclic amines such as piperidine, hydroxyethylpyrrolidine, hydroxyethylpiperidine, morpholine, piperazine, N-methyl piperazine and the like or basic amino acids such as optically pure and racemic isomers of arginine, lysine, histidine, tryptophan and the like.
  • the hydrates, pseudopolymorphs, polymorphs and solvates of all the compounds of the invention are also included and are prepared by conventional methods.
  • the present invention also encompasses the process of making the intermediate amines, as illustrated in the detailed preparations that are used in the condensation with the fluoroquinolone nucleus.
  • the 3-substituted-4-aminopiperidine intermediates can exist as a mixture of cis and trans isomers and the mixture was prepared by the procedure described in Preparation 1.
  • the mixture of cis and trans isomers of 4-amino-3-methylpiperidine was prepared by a sequence as described in Preparation 34.
  • Each cis and trans isomer is a racemic mixture and can be resolved into optically active enantiomeric forms by the usual methods of optical resolution of amines known to those skilled in the art.
  • Other 3-substituted-4-aminopiperidines were synthesised using a similar method.
  • the 4-amino-3,5-dimethylpiperidine intermediate was obtained in the following manner.
  • 4- Amino-l-carbethoxy-3,5-dimethylpiperidine (a mixture of isomers) was prepared according to the procedure described in Preparation 25 and separated by silica gel column cliromatography into the two major mixtures of isomers, one mixture designated as upper mixture A and the other designated as lower mixture B of isomers. Conformations of these mixtures of isomers were not assigned.
  • some compounds of the invention can be prepared by condensing the respective mixture of isomers of 4-amino-3,5-dimethyl-piperidine with [l-cyclopropyl-6,7-difluoro-l,4-dihydro-8- methoxy-4-oxo-quinoline-3-carboxylate-O 3 ,O 4 ]difluoroboron chelate, as exemplified in the ' section on examples described later in this specification.
  • the present invention also encompasses an antiinfective composition for the treatment of humans and animals in need of prophylaxix and/or therapy for systemic or topical infections especially resistant gram-positive organism infections, gram-negative organism infections, mycobacterial infections and nosocomial pathogen infections, which composition comprises an amount of a compound of the invention, the derivatives, isomers, salts, polymorphs, pseudopolymorphs, and hydrates thereof, substantially sufficient to eradicate said infection, but not to cause any undue side effects.
  • Compounds and compositions of this invention can be administered to humans and animals who are at risk of being infected, for example a compound or composition of this invention can be administered to a patient prior to and/or after surgery.
  • the compounds of the invention have superior bactericidal activity against pneumococci and streptococci of various groups. Cidal features available in such molecules add to their clinical attractiveness as it would offer clinicians a valuable treatment option to treat a broader range of infections caused by staphylococci, MRSA, MRSE, pneumococci, streptococci, mycobacteria and diverse range of anaerobic bacteria of clinical importance in a situation such as patients allergic to ⁇ -lactam or possibility of infections due to macrolide resistant strains of streptococci and pneumococci or MRSA/QRSA.
  • the compounds of this invention are effective antimicrobial agents against a broad range of pathogenic microorganisms with advantages in low susceptibility to microbial resistance, reduced toxicity, and improved pharmacology.
  • the molecules of the invention also retain the other valuable features, of being bactericidal to fluoroquinolone resistant staphylococci (QRSA with resistant gyrase) and even to staphylococcal and pneumococcal isolates possessing Nor A efflux pumps and other efflux pumps.
  • the compounds of the invention also display efflux pump inhibitory activity. A combination of all these properties coupled with overall good safety and tolerability observed in a new molecule renders them worthy of therapeutic use in humans and animals. By virtue of such features, they have considerable advantages over existing fluoroquinolone antibacterials, in particular in the treatment of respiratory diseases and infections of skin and soft tissue.
  • Streptococci are implicated as one of the most common pathogens, in both the pediatric and adult population in diverse infections/diseases.
  • diseases which can thus be prevented, alleviated and/or cured by the formulations according to the invention include but are not limited to are meningitis, otitis externa, otitis media; pharyngitis; pneumonia; life-threatening bacteremia, peritonitis; pyelonephritis; cystitis; endocarditis; systemic infections; bronchitis; arthritis; local infections; and septic diseases.
  • Several molecules of the present inventions also exhibit spectacular gains in potency against Mycobacterium tuberculosis and therefore of potential value in the treatment of latent and recalcitrant mycobacterial infections such as tuberculosis.
  • the present invention encompasses certain compounds, dosage forms, and methods of administering the compounds to a human or other animal subject.
  • Specific compounds and compositions to be used in the invention must, accordingly, be pharmaceutically acceptable.
  • a “pharmaceutically acceptable” component is one that is suitable for use with humans and / or animals without undue adverse side effects (such as toxicity, irritation, and allergic response) commensurate with a reasonable benefit/risk ratio.
  • compositions are prepared according to conventional procedures used by persons skilled in the art to make stable and effective compositions.
  • an effective amount of the active compound or the active ingredient is any amount, which produces the desired results.
  • the pharmaceutical compositions may contain the active compounds of the invention, their derivatives, salts and hydrates thereof, in a form to be administered alone, but generally in a form to be administered in admixture with a pharmaceutical carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
  • Suitable carriers which can be used are, for example, diluents or excipients such as fillers, extenders, binders, emollients, wetting agents, disintegrants, surface active agents and lubricants which are usually employed to prepare such drugs depending on the type of dosage form. Any suitable route of administration may be employed for providing the patient with an effective dosage of the compound of the invention their derivatives, salts and hydrates thereof.
  • Dosage forms include (solutions, suspensions, etc) tablets, pills, powders, troches, dispersions, suspensions, emulsions, solutions, capsules, injectable preparations, patches, ointments, creams, lotions, shampoos and the like.
  • the prophylactic or therapeutic dose of the compounds of the invention, their derivatives, salts or hydrates thereof, in the acute or chronic management of disease will vary with the severity of condition to be treated, and the route of administiation. i addition, the dose, and perhaps the dose frequency, will also vary according to the age, body weight and response of the individual patient.
  • the total daily dose range, for the compounds of the invention, the derivatives, salts or hydrates thereof, for the conditions described herein is from about 200 mg to about 1500 mg, in single or divided doses.
  • a daily dose range should be between about 400 mg to 1200 mg, in single or divided dosage, while most preferably a daily dose range should be between about 500 mg to about 1000 mg in divided dosage.
  • intravenous administiation may be a single dose or up to 3 divided doses
  • intravenous administiation can include a continuous drip. It may be necessary to use dosages outside these ranges in some cases as will be apparent to those skilled in the art. Further, it is noted that the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with individual patient's response. The term "an amount sufficient to eradicate such infections but insufficient to cause undue side effects" is encompassed by the above - described dosage amount and dose frequency schedule.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets, or tablets, or aerosol sprays, each containing a predetermined amount of the active ingredient, as a powder or granules, or as a solution or a suspension in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion, or a water-in-oil liquid emulsion.
  • Such compositions may be prepared by any of the methods of pharmacy, but all methods include the step of bringing into association the active ingredient with the carrier, which constitutes one or more necessary ingredients.
  • compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation.
  • the compositions of the present invention include compositions such as suspensions, solutions, elixirs, aerosols, and solid dosage forms.
  • Carriers as described in general above are commonly used in the case of oral solid preparations (such as powders, capsules and tablets), with the oral solid preparations being preferred over the oral liquid preparations. The most preferced oral solid preparation is tablets.
  • tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are employed.
  • suitable carriers include excipients such as lactose, white sugar, sodium chloride, glucose solution, urea, starch, calcium carbonate, kaolin, crystalline cellulose and silicic acid, binders such as water, ethanol, propanol, simple syrup, glucose, starch solution, gelatin solution, carboxymethyl cellulose, shellac, methyl cellulose, potassium phosphate and polyvinyl pyrrolidone, disintegrants such as dried starch, sodium alginate, agar powder, laminaria powder, sodium hydrogen carbonate, calcium carbonate, Tween (fatty acid ester of polyoxyethylenesorbitan), sodium lauryl sulfate, stearic acid monoglyceride, starch, and lactose, disintegration inhibitors such as white sugar, stearic acid glyceryl ester, cacao butter and hydrogenated oils, absorption promoters such as
  • the tablet if desired, can be coated, and made into sugar-coated tablets, gelatin-coated tablets, enteric-coated tablets, film-coated tablets, or tablets comprising two or more layers.
  • tablets may be coated by standard aqueous or non-aqueous techniques.
  • a wide variety of conventional carriers known in the art can be used.
  • suitable carriers are excipients such as glucose, lactose, starch, cacao butter, hardened vegetable oils, kaolin and talc, binders such as gum arabic powder, tragacanth powder, gelatin, and ethanol, and disintegrants such as laminaria and agar.
  • suitable carriers include polyethylene glycol, cacao butter, higher alcohols, gelatin, and semi-synthetic glycerides.
  • a second preferred method is parenterally for intramuscular, intravenous or subcutaneous administration.
  • a third preferred route of administration is topically, for which creams, ointments, shampoos, lotions, dusting powders and the like are well suited.
  • an effective amount of the compound according to this invention in a topical form is from about 0.1% w/w to about 10 % w/w of the total composition.
  • the effective amount of the compound of the invention is 1% w/w of the total composition.
  • the compounds of the present invention may also be administered by controlled release means and/or delivery devices such as those described in U.S. Patent Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123 and 4,008,719; the disclosures of which are hereby incorporated by reference.
  • each tablet contains from about 200 mg to about 1500 mg of the active ingredient.
  • the tablet, cachet or capsule contains either one of three dosages, about 200 mg, about 400 mg, or about 600 mg of the active ingredient.
  • diluents customarily used in the art can be used.
  • suitable diluents are water, ethyl alcohol, polypropylene glycol, ethoxylated isostearyl alcohol, polyoxyethylene sorbitol, and sorbitan esters.
  • Sodium chloride, glucose or glycerol may be incorporated into a therapeutic agent.
  • the antimicrobial pharmaceutical composition may further contain ordinary dissolving aids, buffers, pain-alleviating agents, and preservatives, and optionally coloring agents, perfumes, flavors, sweeteners, and other drugs.
  • viscous to semi-solid or solid forms comprising a carrier compatible with topical application and having a dynamic viscosity preferably greater than water.
  • suitable formulations include but are not limited to solutions, suspensions, emulsions, creams, ointments, powders, liniments, salves, aerosols, etc., which are, if desired, sterilized or mixed with auxiliary agents, e.g. preservatives, antioxidants, stabilizers, wetting agents, buffers or salts for influencing osmotic pressure, etc.
  • auxiliary agents e.g. preservatives, antioxidants, stabilizers, wetting agents, buffers or salts for influencing osmotic pressure, etc.
  • sprayable aerosol preparations wherein the active ingredient preferably in combination with a solid or liquid inert carrier material.
  • a specific embodiment of the invention is the preparation of storage stable compositions of the compounds of the invention of formula I.
  • Such stable compositions can be advantageously made through the use of selective stabilizers.
  • Different stabilizers are known to those skilled in the art of making pharmaceutical compositions.
  • stabilizers such as disodium ethylenediaminetetraacetic acid (EDTA), tromethamine, cyclodextrins such as gamma- cyclodextrin, hydroxy-propyl-gainma-cyclodextrin have been found to be useful.
  • EDTA disodium ethylenediaminetetraacetic acid
  • cyclodextrins such as gamma- cyclodextrin, hydroxy-propyl-gainma-cyclodextrin have been found to be useful.
  • the pharmaceutical compositions contain an effective amount of the active compounds of the invention, its derivatives, salts or hydrates thereof described in this specification as hereinbefore described in admixture with a pharmaceutically acceptable carrier, diluent or excipients, and optionally other therapeutic ingredients.
  • a method for preparing a 3-alkyl substituted- l-benzyl-4-piperidone comprising the steps of:
  • alkyl halide under stirring at a temperature between 25°C - 65 °C, preferably 25°C - 40 °C for 1-12 hrs, preferably 2-3 hrs; and 3) heating with inorganic acid such as hydrochloric acid, suliuric acid, preferably hydrochloric acid, in the presence of solvent such as water, dioxane , N, N-dimethylformamide, preferably water or dioxane at 80 °C -120 °C, preferably 90 °C -110 °C for 5-36 hrs, preferably 24 - 36 hrs, and isolating the product.
  • solvent such as water, dioxane , N, N-dimethylformamide, preferably water or dioxane at 80 °C -120 °C, preferably 90 °C -110 °C for 5-36 hrs, preferably 24 - 36 hrs, and isolating the product.
  • a method for preparing a 3, 3 -dialkyl/3 ,5 -dialkyl/ 3,3,5-trialkyl substituted-l-benzyl-4- piperidone comprising the steps of:
  • alkyl halide selected from C1-C6 lower alkyl/ alkenyl iodides bromides such as methyl iodide, ethyl iodides, allyl bromide, propargyl bromide etc or C1-C6 aralkyl iodides/bromides/chlorides such as benzyl bromide, benzyl chloride etc under stirring at temperature a between -10 °C to 45 °C , preferably -10 °C to 35 °C for 12 - 24 hrs, preferably 12 - 16 hrs, and isolating the product.
  • alkyl halide selected from C1-C6 lower alkyl/ alkenyl iodides bromides such as methyl iodide, ethyl iodides, allyl bromide, propargyl bromide etc or C1-C6 aralkyl iodides/bromides/chlor
  • Step-1 4-Amino-l-carbethoxy-3-methylpiperidine:
  • the aqueous layer was basified with 1 M sodium hydroxide solution (pH -10) and extracted with ethyl acetate. Ethyl acetate extract was dried (Na 2 SO ) and concentrated to dryness to furnish 4-amino-l-carbethoxy-3-methylpiperidine.
  • Step-1 l-Carbethoxy-4-methylamino-3-methylpiperidine: Methylamine hydrochloride (10 g, 148 mmol) was added to the stined solution of 1-carbethoxy- 3-methyl-4-piperidone (7 g, 37.83 mmol) obtained as described in Preparation 1, in methanol (50 ml) followed by 8.3g (148 mmol) KOH. Stirring was continued for 3 hr at ambient temperature. The resulting mixture was cooled at 0°C and sodium cyanoborohydride (3.0 g, 46.0 mmol) was added to it. Cooling was removed after 10 min. and resulting mixture was stined for 12 hr at ambient temperature.
  • reaction mixture was concentrated to dryness, triturated with water, acidified with cone. HCl (pH 3 - 4) and extracted with ethyl acetate to remove impurities.
  • the aqueous layer was basified with 1 M sodium hydroxide solution (pH -10) and extracted with ethyl acetate.
  • Ethyl acetate extract was dried (Na 2 SO 4 ) and concentrated to dryness to give 1- carbethoxy-4-methylamino-3-methylpiperidine.
  • Step-2 4-Methylamino-3-methylpiperidine l-Carbethoxy-4-methylamino-3-methylpiperidine (4.0 g, 18.34 mmol) was suspended in 5 M NaOH solution (15 ml), stined at 110°C for 24 hr, cooled, extracted with ethyl acetate, dried (Na 2 SO 4 ) and concentrated to dryness to afford 4-methylamino-3-methyl-piperidine.
  • Step-1 l-Carbethoxy-4-ethylamino-3-methylpiperidine
  • Step-2 4-Ethylamino-3-methylpiperidine l-Carbethoxy-4-ethylamino-3 -methylpiperidine (2.3 g, 10.74 mmol) was suspended in 5 M NaOH solution (15 ml), stined at 110°C for 120 hr, cooled, extracted with ethyl acetate, dried (Na 2 SO ) and concentrated to dryness to afford 4-ethylamino-3-methylpiperidine. Yield 0.7 g (46 %), C 8 H 18 N 2 , m/z 143, (M+l).
  • Step-1 l-Carbethoxy-4-cyclopropylamino-3-methylpiperidine:
  • the aqueous layer was basified with 1 M sodium hydroxide solution (pH -10) and extracted with ethyl acetate. Ethyl acetate extract was dried (Na 2 SO 4 ) and concentrated to dryness to afford l-carbethoxy-4-cyclopropylamino-3- methylpiperidine.
  • Step-2 4-Cvclopropylamino-3-methylpiperidine: l-Carbethoxy-4-cyclopropylamino-3-methylpiperidine (6.0 g, 26.66 mmol) was suspended in 5
  • Step-1 l-Carbethoxy-4-dimethylamino-3 -methylpiperidine:
  • Paraformaldehyde (5.1g) was added to the stined solution of 4-amino-l-carbethoxy-3- methylpiperidine (3.5 g, 18.8 mmol) in methanol (100 ml) at 0°C and sodium cyanoborohydride (1.7 g, 27.0 mmol) was added to it.
  • acetic acid (1 ml) was added to the resulting mixture and stirring was continued for 96 hr at ambient temperature.
  • the reaction mixture was concentrated to dryness, triturated with water, acidified with cone.
  • Step-2 4-Dimethylamino-3-methylpiperidine l-Carbethoxy-4-dimethylamino-3-methylpiperidine (3.5 g, 16.3 mmol) was suspended in 5 M NaOH solution (20 ml), stined at 110°C for 48 hr, cooled, extracted with ethyl acetate, dried (Na 2 SO 4 ) and concentrated to dryness to afford 4-dimethylamino-3-methylpiperidine.
  • Methylamine hydrochloride (17 g, 252 mmol) was added to the stined solution of 1-carbethoxy- 3-ethyl-4- ⁇ iperidone (10 g, 50 mmol) in methanol (150 ml) followed by 14.1 g (252 mmol) KOH. Stining was continued for 3 hr at ambient temperature. The resulting mixture was cooled at 0°C and sodium cyanoborohydride (4.72 g, 75 mmol) was added to it. Cooling was removed after 10 min. and resulting mixture was stined for 12 hr at ambient temperature. The reaction mixture was concentrated to dryness, triturated with water, acidified with cone.
  • Cyclopropylamine 14.25 g, 250 mmol was added to the stined solution of l-carbethoxy-3- ethyl-4-piperidone (10 g, 50 mmol) in methanol (100 ml) and stining was continued for 12 hr at ambient temperature. The resulting mixture was cooled at 0°C and sodium cyanoborohydride (3.9 g, 62 mmol) was added to it. Cooling was removed after 10 min. and stirring was continued for 12 hr at ambient temperature. The reaction mixture was concentrated to dryness, triturated with water, acidified with cone.
  • Method-B l-Benzyl-4-piperidone (100 gm, 0.53 mol) diluted with 100 ml THF was added to a suspension of 60% NaH (42 g, 1.05 mol) in 700 ml THF at -10 to -5 °C. The mixture was stined for 1 hour and methyl iodide (150 gm, 1.06 mol) diluted in 50 ml THF was added, maintaining the temperature between -3 to -10 °C. The resultant mixture was stined. Ethyl acetate (800 ml) was added to the reaction mixture followed addition of 300 ml water. The organic layer was separated washed with 2 x 300 ml water and concentrated under vacuum to obtain a syrup.
  • the syrup was triturated with 200 ml hexane. the mass was filtered at room temperature over celite and the filtrate concentrated to afford a 121 gm of crude compound.
  • the crude compound upon high vacuum distillation afforded 85 g distillate.
  • To the distillate (50 gm) was charged 50 ml of- concentrated hydrochloric acid.
  • the suspension was stined for 15 minutes and hydrochloric acid was evaporated under vacuum to obtain a thick residue.
  • Isopropanol (50 ml) was added to this residue and was evaporated under vacuum to obtain a solid.
  • the solid was dissolved in 200 ml isopropanol under reflux and stirring, and cooled under stirring to effect crystallization.
  • the crystalline solid was filtered under suction at 20-30 °C to give a white crystalline compound as hydrochloride salt of 1 -benzyl- 3,3 -dimethyl-4- piperidone.
  • the solid was dissolved in 100 ml water and made alkaline with aqueous ammonia solution to pH 10-11. Alkaline aqueous phase was extracted with 50 ml chloroform thrice. Combined organic extract was washed with water, dried over sodium sulfate and then evaporated under vacuum to afford 28 g (80%) compound as an oil. m/z (M+l) 218.
  • Step-2 4-Amino-3,3-dimethylpiperidine:
  • Ammonium acetate (3.5 g, 45.45 mmol) was added to the stined solution of l-benzyl-3,3- dimethyl-4-piperidone (2.0 g, 9.2 mmol), in methanol (20 ml) and stining was continued for 3 hr at ambient temperature. The resulting mixture was cooled at 0°C and sodium cyanoborohydride (0.58 g, 9.2 mmol) was added. Cooling was removed after 10 min. and resulting mixture was stined for 6 hr at ambient temperature. The reaction mixture was concentrated to dryness, triturated with water, acidified with cone. HCl (pH 3 - 4) and extracted with ethyl acetate to remove impurities.
  • the aqueous layer was basified with 1 M sodium hydroxide solution (pH -10) and extracted with ethyl acetate. Ethyl acetate extract was dried (Na 2 SO 4 ) and concentrated to dryness to furnish 4-amino-l-benzyl-3,3-dimethyl piperidine.
  • Step-2 4-Amino l- ⁇ butyloxycarbonyl- 33-dimethylpiperidine:
  • Step-3 4-Benzyloxycarbonylamino- 1 - ⁇ utyloxycarbonyl-S -dimethylpiperidme:
  • Benzyl chloroformate 50% in toluene, 450 ml, 1.57 mol was added to a stined suspension of 4- amino-l-'-butyloxycarbonyl-S, 3 -dimethylpiperidine (365 g ) as prepared above and NaHCO 3 (150 g, 1.78 mol) in dry tetrahydrofuran (1.5 L).
  • the reaction mixture was stined for 20 hr at 35°C.
  • the reaction mixture was diluted with 3.5 L water and was extracted with 2.0 L X 2 ethyl acetate. Combined organic extract was washed with water, dried over Na 2 SO and concentrated to dryness.
  • Step-4 4-Benzyloxycarbonylamino-3.3-dimethylpiperidine: 6 N HCl (200 ml) was added to a stined solution of 4-benzyloxycarbonylamino-l- t - butyloxycarbonyl-3,3-dimethylpiperidine (350 g, 0.96 mol) in dioxane (200 ml). The resulting mixture was stined for 1 hr and concentrated to dryness. The resultant residue was treated with water (3.0 L) water and was extracted with 1.0 L X 2 ethyl acetate. The aqueous layer was basified with 2 M aqueous NaOH and extracted with 2.5 L X 2 dichloromethane.
  • Step-1 1- Benzyloxycarbonyl-3.3-dimethyl-4-piperidone:
  • Method B l-Benzyl-3,3-dimethyl-4-piperidone (255 g , 1.175 mol) was dissolved in 800 ml toluene. To the clear solution was charged 50% benzyl chloroformate in toluene (441 ml, 1.29 mol). The reaction mixture was stined at temperature between 80-85°C for 4-5 hrs. The reaction mixture was concentrated under vacuum. Crude product was purified by silica gel column chromatograhy to give 292 g (95%) titled compound, m/z (M+l) 262.
  • Step-2 4-Amino- l-benzyloxycarbonyl- 3.3-dimethylpiperidine: l-Benzyloxycarbonyl-3,3-dimethyl-4-piperidone (290 g, 1.11 mol) was dissolved in methanol (1.5 L) and under stirring addition of ammonium acetate (600 g, 7.80 mol ) was made at 30- 35°C. The reaction mixture was stined for 5 hrs. Sodium cyanoborohydride (35 g, 0.55 mol) was added portion wise to the suspension, over 0.5 hrs by maintaining temperature between 0- 10°C. The reaction mixture was stined for 6-7 hrs.
  • Resultant 'solid A' was treated separately to obtain (-) isomer of 4-amino- 1 -benzyloxycarbonyl- 3, 3 -dimethyl- piperidine as described in Step-4.
  • the filtrate was concentrated under vacuum to obtain a 'solid B'.
  • the 'solid B' was treated with aqueous K2CO3 solution made by dissolving 112 g K 2 CO 3 in 1.2 L water and was extracted with 750 ml chloroform thrice, Combined organic extract was washed with 200 ml water and dried over sodium sulfate. Evaporation of organic solvent afforded 71 g (71% ) as an oil.
  • the compound was subjected to a second resolution by dissolving it ( 70 g, 0.267 mol ⁇ in 840 ml 2-3 % aqueous ethyl alcohol (moisture content, 2.6 %>) under stirring and resultant solution was treated with (40 g, 0.267 mol) D-(-)-tartaric acid at 60-70°C.
  • the reaction mixture was agitated at 65-70 °C for half an hour.
  • the reaction mass was cooled and was filtered at 20-30°C.
  • the wet cake was washed with 210 ml additional ethyl alcohol to afford a crystalline salt.
  • Step-4 (-)- 4-Amino-l-benzyloxycarbonyl- 3,3-dimethylpiperidine
  • Step-3 was treated with aqueous K CO 3 solution made by dissolving 164 g K 2 CO 3 in 1.6 L water and was extracted with 750 ml chloroform thrice. Combined organic extract was washed with 200 ml water and dried over sodium sulfate. Evaporation of organic solvent afforded 85 g (85%) as an oil.
  • the compound was subjected to a second resolution by dissolving it (84 g, 0.32 mol ⁇ in 1.0 L 2-3 % aqueous ethyl alcohol (moisture content, 2.6 %) under stirring and resultant solution was treated with L-(+)-tartaric acid (46 g, 0.307 mol) at 60-70°C.
  • the reaction mixture was agitated at 65-70°C for half an hour.
  • the reaction mass was cooled and was filtered at 20-30°C.
  • the wet cake was washed with 250 ml additional ethyl alcohol to afford a crystalline salt.
  • the resultant white solid was treated with aqueous K 2 CO3 solution made by dissolving 93 g K 2 CO in 1.0 L water and was extracted with 750 ml X 3 chloroform. Combined organic extract was washed with 200 ml water and was dried over sodium sulfate. Evaporation of organic solvent afforded 71 g (83%) as an oil.
  • Step-5 (+ -!- Benzyloxycarbonyl-4- t' butyloxycarbonylamino-3,3-dimethylpiperidine:
  • Step-6 (-)-l -Benzyloxycarbonyl- -4- t' butyloxycarbonylamino-3,3-dimethylpiperidine
  • Step-7 (+)-4- t -Butyloxycarbonylamino -3.3-dimethylpiperidine:
  • (+)-l- Benzyloxycarbonyl-4- t" butyloxycarbonylamino-3,3-dimethylpiperidine (75 g, 0.207 mol) was dissolved in 500 ml methanol and the solution was transfened to a Pan reactor. Wet 10%> palladium on carbon (7.5 g) was added to the solution and was stined at 200 psi hydrogen pressure. Reaction progress was monitored on TLC and reaction was completed in 3-4 hrs. The reaction mixture was filtered as soon as it was completed. The residue was washed with 100 ml methanol. Filtrate was evaporated to dryness to afford the required product (47 g) in quantitative yield. m/z (M+l) 229.
  • Step-8 (-)-4- t -Butyloxycarbonylamino-3 ,3-dimethyf ⁇ iperidine
  • Ammonium acetate (7.85 g, 102 mmol) was added to the stined solution of l-benzyl-3, 3 -diethyl piperidin-4-one (5.0 g, 20.40 mmol) in methanol (100 ml) and stining was continued for 3 hr at ambient temperature. The resulting mixture was cooled at 0°C and sodium cyanoborohydride (1.3 g, 20.40 mmol) was added to it. Cooling was removed after 10 min. and resulting mixture was stined for 6 hr at ambient temperature. The reaction mixture was concentrated to dryness, triturated with water, acidified with cone.
  • Step-1 Mixture of isomers of 4-amino-l-carbethoxy-3,5-dimethylpiperidine:
  • the aqueous layer was basified with 1 M sodium hydroxide solution (pH -10) and extracted with ethyl acetate. Ethyl acetate extract was dried (Na 2 SO ) and concentrated to give 4- amino-l-carbethoxy-3,5-dimethylpiperidine.
  • the obtained 4-amino-l-carbethoxy-3,5- dimethylpiperidine was subjected to silica gel column cliromatography. Elution with 5% methanol in chloroform furnished a solid, which was a mixture of stereoisomers, conformational analysis of which was not obtained. Yield 3.5 g (77 %), m.p. 218-20°C, C ⁇ 0 H ⁇ 8 N 2 O 2 , m/z 201 (M+l).
  • Step-2 Separation of isomers of 4-amino-l-carbethoxy-3,5-dimethylpiperidine: 4-Amino-l-carbethoxy-3,5-dimethylpiperidine obtained as per procedure described in Step-1 was subjected to silica gel column chromatography. Elution with chloroform gave "upper" mixture of isomers of 4-amino-l-carbethoxy-3,5-dimethylpiperidine m.p.
  • Step-3 Mixture of isomers of 4-amino-3.5 -dimethylpiperidine (Mixtures A+B): A mixture of isomers of 4-amino-l-carbethoxy-3,5-dimethylpiperidine (10 g, 51 mmol) obtained by a procedure as described in step-1, was stined in 5 M NaOH solution (100 ml) at 100°C for 48 hr, cooled, extracted with ethyl acetate, dried (Na 2 SO 4 ) and concentrated to afford mixture of isomers of 4-amino-3,5-dimethylpiperidine as oil as "mixture A + B".
  • Step-4 "Upper" mixture of isomers of 4-amino-3.5-dimethylpiperidine (Mixture A of isomers): A "upper" mixture of isomers of 4-amino-l-carbethoxy-3,5-dimethylpi ⁇ eridine (1.5 g, 7.5 mmol) obtained as described in Step-2, was stined in 5 M NaOH solution (10 ml) at 100°C for 48 hr, cooled, extracted with ethyl acetate, dried (Na 2 SO 4 ) and concentrated to afford "upper" mixture of isomers of 4-amino-3,5-dimethyl piperidine as oil as mixture A of isomers, of which the confonnational analysis was not obtained.
  • Step-5 "Lower" mixture of isomers of 4-amino-3.5-dimethylpiperidine (Mixture B of isomers): A "lower” mixture of isomers of 4-amino- l-carbethoxy-3,5-dimethylpiperidine (1.0 g, 5.0 mmol) obtained by a procedure as described in step-2, was stined in 5 M NaOH solution (10 ml) at 100°C for 48 hr, cooled, extracted with ethyl acetate, dried (Na 2 SO 4 ) and concentrated to afford "lower" mixture of isomers of 4-amino-3,5-dimethylpiperidine as an oil as mixture B of isomers, of which the conformational analysis was not obtained. Yield 0.51 g (80 %), CyHi6N 2 , m z 129 (M+l).
  • Powdered KOH (0.92 g, 16.44 mmol) was added in portions to the stined solution of methylamine hydrochloride (1.11 g, 16.44 mmol) in methanol (20 ml) at 0-5°C and 1- carbethoxy-3,5-dimethyl-4-piperidinone (2.2 g, 1.1 mmol),obtained by a procedure as described in Example 25 (Step-1) , was added in portions to it. The resulting reaction mixture was stined for 45 min at 10°C and a solution of sodium cyanoboro-hydride (0.7 g, 1.1 mmol) in methanol (5 ml) was added dropwise to it.
  • Mixtures A+B of 4-methylamino-l-carbethoxy-3,5-dimethylpi ⁇ eridine (1.3 g, 0.6 mmol) was stined in a mixture of 10% NaOH solution (20 ml) and ethyl alcohol (10 ml) at 100 D C for 120 hr, cooled, extracted with ethyl acetate, dried (Na 2 SO 4 ) and concentiated to afford a Mixtures A+B of 4-methylamino-3,5-dimethylpiperidine. Yield 0.61 g (70 %), C 8 H ⁇ 8 N 2 ,m/z 143 (M+l).
  • Mixture A of isomers and Mixture B of isomers of 4-methylamino-3,5-dimethylpiperidme were prepared by separation technique at 4-methylamino-l-carbethoxy-3,5-dimethylpiperidme stage by using silica gel column chromatography similar as described in Preparation 25 (Step-2) followed by aqueous sodium hydroxide mediated hydrolysis.
  • Powdered KOH (2.8g, 50 mmol) was added in portions to the stined solution of ethylamine hydrochloride (4.0 g, 50 mmol) in methanol (50 ml) and l-carbethoxy-3,5-dimethyl-4- piperidinone (5.0 g, 25.12 mmol) obtained by a procedure as described in Example 25 (Step-1), was added to it.
  • the resulting reaction mixture was stined for 6 hr.
  • a solution of sodium cyanoborohydride (1.6 g, 25.12 mmol) in methanol (10 ml) was added dropwise to it and stirring was continued for 16 hr.
  • the reaction mixture was concentrated to dryness.
  • Mixture A+B of l-carbethoxy-4-cyclopropylamino-3,5- dimethylpiperidine was separated over silica gel column chromatography. Elute from 5% ethyl acetate in hexane gave Mixture A of isomers of l-carbethoxy-4-cyclopropyamino-3,5- dimethylpiperidine as oil.
  • Mixture B of isomers of l-carbethoxy-4-cyclopropylamino-3,5-dimethylpiperidine (2.8 g, 1.16 mmol) was stined in a mixture 5 N NaOH solution (20 ml) and ethyl alcohol (5 ml) at 100°C for 120 hr, cooled, extracted with ethyl acetate, dried (Na 2 SO 4 ) and concentrated to afford Mixture B of isomers of 4-cyclopropylamino-3,5-dimethylpiperidine as an oil.
  • Mixture A of isomers and Mixture B of isomers of 4-dimethylamino-3,5-dimethylpiperidine were prepared by separation technique at l-benzyl-4-dimethylamino-3,5-dimethylpiperidine stage by using silica gel column chromatography followed by debenzylation by using catalytic palladium hydroxide on carbon.
  • Preparation 30 4-Amino-3,5-diethylpiperidine Ammonium acetate (10 g, 130mmol) was added to the stined solution of l-benzyl-3,5-diethyl-4- piperidone (3.5 g, 14.3 mmol) in methanol (50 ml) and stirring was continued for 4 hr at ambient temperature.
  • the aqueous layer was basified with 1 M sodium hydroxide solution (pH -10) and extracted with ethyl acetate. Ethyl acetate extract was dried (Na 2 SO ) and concentrated to dryness to furnish 4-amino-3,3,5-tri ⁇ nethylpiperidine. Yield 0.45 g (54 %), C s H ⁇ 8 N 2 , m/z 143 (M+l).
  • reaction mixture was concentrated to dryness, triturated with water, acidified with cone. HCl (pH 2) and extracted with ethyl acetate to remove impurities.
  • the aqueous layer was basified with IM sodium hydroxide solution (pH 9) and extracted with ethyl acetate.
  • Ethyl acetate extract was dried (Na 2 SO 4 ) and concentrated to dryness to furnish 4-amino-l-benzyl-3,5-diethyl-3-methylpiperidine as an oil. Yield 2.1 g (84 %), C ⁇ 7 H 28 N 2 , m z 261 (M+l).
  • Ammonium acetate (2.5 g, 32.46mmol) was added to the stined solution of l-benzyl-3, 5- dimethyl-3-ethyl-4-piperidone (3.0 g, 12.25 mmol) in methanol (40 ml) and stining was continued for 20 hr at ambient temperature. The resulting mixture was cooled at 0°C and sodium cyanoborohydride (0.8 g, 12.7 mmol) was added to it. Cooling was removed after 10 min. and resulting mixture was stined for 20 hr at ambient temperature. The reaction mixture was concentrated to dryness, triturated with water, acidified with cone.
  • Preparation 34 cis or tra/._--4-t-Butyloxycarbonylamino- 1 -benzyl-3-methylpiperidine: Step-1: Ethyl-l-benzyl-3-methyl-4-oxo-piperidine-3-carboxylate: Ethyl -l-benzyl-4-oxo-piperidine -3 -carboxylate hydrochloride (150 g, 0.504 mol) was suspended in a solvent mixture of 750 ml THF and 750 ml DMF at room temperature. Addition of powdered KOH (56 g, 1.0 mol) was made in two equal lots keeping half an hour interval between two additions.
  • methyl iodide (78 g, 0.55 mol) was added over period of 10 minutes and it was stined for three hours at room temperature. The reaction was quenched by adding 4 ltr water followed by 1.5 ltr diethyl ether. Layers were separated. Organic layer was washed with water and dried over Na 2 SO 4 .
  • Step -2 l-Benzyl-3-methyl-4-piperidone: Ethyl-l-benzyl-3-methyl-4-oxo-piperidine-3-carboxylate (85 g, 0.31 mol) was dissolved in cone HCl (82 ml, 0.77 mol) and the reaction mixture was heated to 100 °C for 32 hrs. Solvent was removed under reduced pressure and the resulting solid was dissolved in the 300 ml CHC13 and 400 ml pet ether was added to this under stirring to provide a solid. The solid was filter and was dissolved in 400 ml ethyl acetate and organic layer was washed with 10% NaOH aqueous solution. Layers were separated and concentration of organic layer afforded 48 g ( 75%) oil as a titled product. mass (ES + ) 204, Molecular Formula C ⁇ 3 H ⁇ 7 NO,
  • Step-3 4-Amino- 1 -benzyl-3-methyl ⁇ i ⁇ eridine:
  • Ammonium acetate (178.3 g, 2.31 mol) was charged to a solution of l-benzyl-3 -methyl-4- piperidone (47 g, 0.232 mol) in 500 ml methanol. The suspension was stined for 4 hours at room temperature. Sodium cyanoborohydride (7.3 g, 0.116 mol) was added in lots at 10 °C and it was stined for 1 hour.
  • Solvent was removed under vacuum and resultant solid was suspended in 500 ml water and acidified with dilute aqueous HCl. It was extracted with 200ml X 2 chloroform and layers were separated. Aqueous layer was basified with ammonia solution to pH 9 and extracted with 500 ml X 3 CHCI3. Drying of organic layer over Na 2 SO and evaporation afforded 44 g (92%>) titled product as an oil, which was used directly for further reaction.
  • Step -4 cis or t7- ?t_?-4-t-Butyloxycarbonylamino-l-benzyl-3-methylpiperidine:
  • Di-tert-butyloxydicarbonate (45 g, 0.206 mol) was charged in lots to a solution of 4-amino-l- benzyl-3-methylpiperidine (42 g, 0.206 mol) and 300 ml CH 2 C1 2 followed by 5 ml triethylamine.
  • trans-4-t-Butyloxycarbonylamino-3-methylpiperidine was obtained as per procedure depicted in preparation 000 by using trans-4-t-Butyloxycarbonylamino-l-benzyl-3-methylpiperidine in the place of cis-4-t-Butyloxycarbonylamino- 1 -benzyl-3-methylpiperidine in 96%> yield.
  • the titled compound was prepared by the procedure described in Preparation 37, by using (+)- 4- amino-1 -benzyloxycarbonyl- 3,3-dimethylpiperidine (cf. Preparation 19, Step 3, 44 g, 168.0 mmol), to give 15.0 (70%) titled compound.
  • Example 1 1 -Cyclo p ropyl-6-fluoro- 1 ,4-dihvdro-8-methoxy-7-(4-amino-3-methyl- 1 - ⁇ iperidinyl)-4-oxo- guinoline-3 -carboxylic acid
  • the reaction mixture was concentrated to dryness.
  • the obtained residue was treated with triethylamine (3 ml) and ethanol (15 ml) and refluxed for 16 hr.
  • the resulting mixture was concentrated to dryness; the solid thus obtained was triturated with water (10 ml), filtered, washed with water, dried and purified by preparative HPLC to furnish the required product.
  • Example 2 tr n_?-l-Cyclopropyl-6-fluoro-l,4-dihvdro-8-methoxy-7-(4-amino-3-methyl-l-piperidinyl ' )-4- oxo-quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1, where tr ⁇ »s-4-amino-3- methylpiperidine was used in place of 4-amino-3-methylpiperidine. Yield 35%, m.p.240-42°C, C 20 H 2 FN 3 O 4 , m/z 390 (M+l).
  • Example 3 c._?-l-cvclopropyl-6-fluoro-l,4-dihydro-8-methoxy-7-(4-amino-3-methyl-l-pi ⁇ eridinyl)-4-oxo- quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1, where c ⁇ -4-amino-3- methylpiperidine was used in place of 4-amino-3 -methylpiperidine Yield 35%, m.p.246-50°C, C 2 oH 24 N 3 O 4 F, m z 390 (M+l).
  • Example 4 1 -Cyclopropyl -6-fluoro- 1.4-dihydro-8-methoxy-7-(4-methylamino-3 -methyl- 1 -piperidinyl)-4- oxo-quinoline-3 -carboxylic acid It was prepared in a similar manner as described in Example 1, where 4-methylamino-3- methylpiperidine was used in place of 4-amino-3-methylpiperidine. Yield 50 %>, m.p.240°C (decomp.), C 2 ⁇ H 26 FN 3 O 4 , m/z 404 (M+l).
  • Example 5 l-Cyclopropyl-6-fluoro-1.4-dihvdro-8-methoxy-7-(4-ethylamino-3-methyl-l-piperidinyl')-4-oxo- quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1, where 4-ethylamino-3- methylpiperidine was used in place of 4-amino-3 -methylpiperidine. Yield 52 %, m.p.l60-62°C, C 22 H 28 FN 3 O 4 , m/z 418 (M+l).
  • Example 7 l-Cvclopropyl-6-fluoro-1.4-dihydro-8-methoxy-7-f4-dimethylamino-3-methyl-l-piperidinyl)-4- oxo-quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1, where 4-dimethyl amino-3- methylpiperidine was used in place of 4-amino-3-methylpiperidine.
  • (+Vl-Cvclopropyl-6-fluoro-1.4-dihydro-8-methoxy-7-(4-benzyloxycarbonylamino-3,3- dimethyl-l- ⁇ iperidinyl)-4-oxo-quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1, where 4-(+)-benzyloxy carbonylamino-3,3-dimethylpiperidine was used in place of 4-amino-3 -methylpiperidine. Yield
  • Example- 10 ( ⁇ )- 1 -Cyclopropyl-6-fluoro- 1 ,4-dihydro-8-methoxy-7-(4-anrino-3,3- dimethyl- 1 -piperidinyl)-4- oxo-quinoline-3-carboxylic acid 4-Benzyloxycarbonylamino-3,3-dimethyl piperidine (100 g, 0.381 mol) was suspended in 200 ml acetonitrile under stining.
  • the solid separated in the reaction mixture was filtered and washed with 50 ml ethanol.
  • the filtered solid was stined with reflux at 100-110 °C in concentrated hydrochloric acid (250 ml) for 2 hr.
  • the resulting solution was taken to dryness by evaporating the acid under vacuum to obtain a residue.
  • To the residue was added 1 L acetone and the suspension stined for 1 hr.
  • the resulting solid was filtered and washed with acetone.
  • the residue was suspended in 600 ml chloroform and was refluxed for 30 minutes.
  • the suspension was filtered and the residue washed with chloroform.
  • the residue was suspended in methanol (600 ml) and was stined at 30-35°C for 30 minutes.
  • An alternate procedure to prepare this compound is by a method similar to that described in Example 1 where 4-amino-3,3-dimethylpiperidine was used in place of 4-amino-3- methylpiperidine.
  • a second alternate procedure to prepare this compound is by treating l-cyclopropyl-6-fluoro- l,4-dihydro-8-methoxy-7-(4-l-carbethoxyamino-3,3-dimethyl-l-piperidinyl)-4-oxo-quinoline-3- carboxylic acid [obtained from condensation of l-cyclopropyl-6,7-difluoro-l,4-dihydro-8- methoxy-4-oxo-quinoline-3-carboxylic acid difluoroborane chelate and 4-1-carbethoxyamino- 3,3-dimethylpiperidine] (2.0 g, 4.0 mmol) under reflux with aqueous NaOH (0.6M, 100 ml) for 4 hr
  • Example- 11 (+)- 1 -Cyclopropyl-6-fluoro- 1.4-dihydro-8-methoxy-7-(4-amino-3.3 -dimethyl- 1 -piperidinyl)-4- oxo-quinoline-3-carboxylic acid.
  • Hydrochloride The hydrochloride salt was obtained by modifying the procedure in Example 10 after obtaining the solid residue from filtration of the suspension in methanol before dissolving in water and adjusting the pH to 8.9 by adding 30% aqueous sodium hydroxide solution. The residue obtained from filtration of the suspension from methanol was dissolved in 2.0 ltr at reflux temperature. It was then filtered hot and concentrated to approximately one fourth of its volume and left overnight.
  • Example 14 (+ - l-Cyclopropyl-6-fluoro-1.4-dihvdro-8-methoxy-7-(4-amino-3.3-dimethyl-l-piperidinyl)-4- oxo-quinoline-3 -carboxylic acid hydrochloride (+)-3,3-Dimethyl-4- t -butyloxycarbonylamino piperidine (46 g, 0.201mol) was suspended in 200 ml acetonitrile under stirring.
  • the solid was suspended in 300 ml chloroform and was refluxed for 30 minutes. The suspension was filtered and was washed with chloroform. The wet solid was suspended in methanol 100 ml and was stined at 30- 35°C for 30 minutes. The reaction mixture was filtered. The residue was dissolved in 1,3 L methanol at reflux temperature. It was filtered hot and concentrated to approximately one fourth of its volume and was left overnight.
  • Example - 15 (-)-l-Cvclopropyl-6-fluoro-1.4-dihvdro-8-methoxy-7-(4-amino-3,3-dimethyl-l-piperidinyl)-4- oxo-quinoline-3-carboxylic acid hydrochloride
  • (+)- l-Cvclopropyl-6-fluoro-1.4-dihvdro-8-methoxy-7-(4-amino-3,3-dimethyl-l-piperidinyl)-4- oxo-quinoline-3 -carboxylic acid 8.50 Grams (19.34 mmol) of (+)- l-cyclopropyl-6-fluoro-l,4-dihydro-8-methoxy-7-(4-amino-
  • Example - 21 (-)- 1 -Cyclopropyl-6-fluoro- 1 ,4-dihvdro-8-methoxy-7-(4-amino-3 ,3-dimethyl- 1 - piperidinyl)- 4-oxo-quinoline-3-carboxylic acid gluconate Similarly, by using the procedure mentioned above, 2.20 g (54.60 mmol) of (-)- l-cyclopropyl-6- fluoro-l,4-dihydro-8-methoxy-7-(4-amino-3,3-dimethyl-l-piperidinyl)-4-oxo-quinoline-3- carboxylic acid was converted to 1.65 g (50%)) titled compound, mp 154-156°C.
  • Example 22 l-CvclopiOpyl-6-fluoro-1.4-dihydro-8-methoxy-7-(4-acetylamino-3.3-dimethyl-l-piperidinyl)-4- oxo-quinoline-3 -carboxylic acid It was prepared in a similar manner as described in Example 1, where 4-acylamino-3,3- dimethylpiperidine was used in place of 4-amino-3-methylpiperidine. Yield 20%, m.p,194-96°C, C 23 H 28 FN 3 O 5 , m/z 446 (M+l).
  • 4-oxo-quinoline-3 -carboxylic acid It was prepared in a similar manner as described in Example 1, where 4-ethylamino-3,3- dimethylpiperidine was used in place of 4-amino-3-methylpiperidine Yield 60 %, m.p.230-32°C, C 23 H 30 FN 3 O 4 , m z 432 (M+l).
  • Example 28 l-Cvclopropyl-6-fluoro-1.4-dihvdro-8-methoxy-7-(4-amino-3-ethyl-3-methyl-l-piperidinyl ⁇ -4- oxo-quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1, where 4-amino-3-ethyl-3- methylpiperidine was used in place of 4-amino-3-methylpiperidine. m.p.152-54 °C, C 22 H 28 FN 3 O 4 , m/z 418 (M+l).
  • Example 30 l-Cyclopropyl-6-fluoro-l,4-dihydro-8-methoxy-7-f4-cyclopropylamino-3-ethyl-3-methyl-l- piperidinyl l-4-oxo-quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1, where 4-cyclopropyl amino-3- ethyl-3 -methylpiperidine was used in place of 4-amino-3-methyl ⁇ iperidine m.p. 210-12°C, C 25 H 32 FN 3 O4, m/z 458 (M+l).
  • Example 34 Mixture B of isomers of l-cyclopropyl-6-fluoro-l,4-dihvdro-8-methoxy-7-(4-amino-3,5- dimethyl-l-piperidinyl>4-oxo-quinoline-3-carboxylic acid
  • Example 35 Mixtures A+B of lcyclopropyl-6-fluoro-l,4-dihydro-8-methoxy-7- ⁇ 4-methylamino-3,5- dimethyl-l-piperidinyl))-4-oxo-quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1, where 4-methylamino-3,5- dimethylpiperidine was used in place of 4-amino-3-methylpiperidine, m.p. 268-72°C, C 22 H 28 FN 3 O 4 , m/z 418 (M+l).
  • Example 36
  • Mixture A of isomers of l-cyclopropyl-6-fluoro-1.4-dihvdro-8-methoxy-7-f4-methylamino-3,5- dimethyl- 1 -piperidinyl)-4-oxo-quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1, where Mixture A of isomers of 4- methyl. ⁇ mino-3,5-dimethylpiperid_ne was used in place of 4-amino-3 -methylpiperidine, C 22 H 28 FN 3 O 4 , m/z 418 (M+l).
  • Example 37 Mixture B of isomers of l-cyclopropyl-6-fluoro-l,4-dihvdro-8-methoxy-7-(4-methylamino-3,5- dimethyl- 1 -piperidinvf)-4-oxo-quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1, where Mixture A of isomers of 4- methylamino-3,5-dimethylpiperidine was used in place of 4-amino-3-methylpi ⁇ eridine, C 22 H 28 FN 3 O 4 , m/z 418 (M+l).
  • Example 38 Example 38
  • Mixture B of isomers of l-cyclopropyl-6-fluoro-1.4-dihvdro-8-methoxy-7-(4-ethylamino-3.5- dimethyl- 1 -piperidinyl)-4-oxo-quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1 , where Mixture B of isomers of 4- methylamino-3,5-dimethylpiperidine was used in place of 4-amino-3-methylpiperidine, m.p.
  • Example 40 Mixture A of isomers of l-cvclopropyl-6-fluoro-l,4-dihydro-8-methoxy-7-(4-cvclo ⁇ ropylamino- 3.5-dimethyl-l-piperidinyl)-4-oxo-quinoline-3-carboxylic acid It was prepared in a similar ma ier as described in Example 1, where Mixture A of isomers of 4- cyclopropyl amino-3,5-dimethylpiperidine was used in place of 4-amino-3-methyl-piperidine, C 24 H 30 FN 3 O 4 , m/z 444 (M+l) Example 41
  • Example 42 1 -Cvclopropyl-6-fluoro- 1 ,4-dihvdro-8-methoxy-7-(4-amino-3.3 ,5-trimethyl- 1 -pi ⁇ eridinyl)-4- oxo-quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 1, where 4-amino-3,3,5- trimethylpiperidine was used in place of 4-amino-3-methylpiperidine, m.p. 218-20°C, C 22 H 28 FN 3 O 4 , m/z 418 (M+l).
  • Example 43 5-Amino- 1 -cvclopropyl-6-fluoro- 1.4-dihvdro-8-methoxy-7-(4-methylamino-3 ,3-dimethyl- 1 - piperidinyI)-4-oxo-quinoline-3 -carboxylic acid
  • a suspension of 5-amino-l-cyclopropyl-6,7-difluoro-l,4-dihydro-8-methoxy-4-oxo-quinoline-3- carboxylic acid (0.17 g, 0.55 mmol), 3,3-dimethyl-4-methylamino piperidine (0.15 g, 1.06 mmol) and triethylamine (lg, 10 mmol) in a mixture of dimethylsulfoxide (10 ml) and acetonitrile (10 ml) was heated at 70°C for 18 hr.
  • Acetonitrile was distilled off, filtered to remove suspended impurities, diluted with water (5 ml). The precipitate thus separated was filtered, washed with water, dried and purified by silicagel column chromatography. Elute from a mixture of ethyl acetate and methanol furnished the required product.
  • Example 50 5_- Amino- 1 -cycloprop yl-6-fluoro- 1 ,4-dihydro-8-methoxy-7-f4-ethylamino-3,3 -dimethyl- 1 - piperidinyl)) -4-oxo-quinoline-3-carboxylic acid
  • Example 53 Mixture B of isomers of 5-Amino- l-cvclopropyl-6-fluoro-l,4-dihydro-8-methoxy-7-(4-amino- 3.5-dimethyl-l-piperidinyl)-4-oxo-quinoline-3-carboxylic acid It was prepared in a similar maimer as described in Example 43, where Mixture B of isomers of 4-amino-3,5-dimethylpiperidine was used in place of 3,3-dimethyl-4-methylamino-piperidine, m.p. 248-50°C (decomp.), C 2 ⁇ H 27 FN 4 O 4 , m/z 419 (M+l),
  • Example 61 5-Amino-l-cvclopropyl-6-fluoro-1.4-dihvdro-8-methoxy-7-(4-amino-3-ethyl-3-methyl-l- piperidinyl)-4-oxo-quinoline-3-carboxylic acid It was prepared in a similar manner as described in Example 43, where 4-amino-3-ethyl-3- methylpiperidine was used in place of 3,3-dimethyl-4-methylaminopiperidine, m.p. 210 °C (decompose), C 22 H 29 FN 4 O 4 , m/z 433 (M+l).
  • Example 62 5-Amino-l-cyclopropyl-6-fluoro-l,4-dihvdro-8-methoxy-7-(4-methylamino-3-ethyl-3-methyl-l- piperidinyl) ) -4-oxo-quinoline-3 -carboxylic acid It was prepared in a similar manner as described in Example 43, where 4-methylamino-3-ethyl- 3-methylpiperidine was used in place of 3,3-dimethyl-4-methylaminopiperidine, m.p. 164-66 °C, C 23 H 3 ⁇ FN 4 O 4 , m/z 447 (M+l).
  • Example - 67 cis- 1 -Cyclopropyl-6-fluoro- 1 ,4-dihydro-8-methyl-7-(4-amino-3-methyl- 1 -piperidinyl)-4-oxo- quinoline-3 -carboxylic acid.
  • the compound was prepared by a procedure as described in Example 66, by using cis-4-amm.o- 3-methylpiperidine, m. p.
  • Example - 68 ( ⁇ )-l-Cvclopropyl-6-fluoro-l,4-dihvdro-8-methyl-7-(4-amino-3,3-dimethyl-l-piperidinyl)-4- oxo-quinoline-3-carboxylic acid.
  • the compound was prepared by a procedure as described in Example 66, by using (+)-4-amino- 3,3-dimethylpiperidine, m. p.
  • Example - 69 (-)-l -Cycloprop yl-6- fluoro- 1 ,4-dihvdro-8-methyl-7-(4-amino-3 ,3-dimethyl- 1 -piperidinyl)-4-oxo- quinoline-3-carboxylic acid.
  • the compound was prepared by a procedure as described in Example 66, by using (-)-4-amino- 3,3-dimethylpiperidine, m. p. 195-198 °C, C 2 ⁇ H 26 FN 3 O 3 , m/z 388 (M+l).
  • the compound was prepared by a procedure as described in Example 66, by using (+)-4-amino- 3,3-dimethylpiperidine, m. p. 195-198 °C, C 2 ⁇ H 26 FN 3 O 3 , m/z 388 (M+l).
  • the compound was prepared by a procedure as described in Example 66, by using 4- methylamino-3-methylpiperidine, m. p 233-235 °C, C 2 ⁇ H 2 6FN 3 O 3 , m/z 388 (M+l), PMR (CD3OD): 0.8-1.4 (m, 7H), 1.9-2.4 (m, 3H), 2.8 (s, 3H), 2.9 (s, 3H), 3.2-3.7 (m, 5H), 4.3 (m, 1H),7.9 (d,lH), 8.9 (s,lH).
  • Example - 72 1 -Cvclopropyl-6-fluoro- 1 ,4-dihvdro -8-methyl -7-(4-dimethylamino-3-methyl- 1 -p_peridinyl)-4- oxo-quinoline-3-carboxylic acid.
  • the compound was prepared by a procedure as described in Example 66, by using 4- dimethylamino-3-methyl ⁇ iperidine, m. p.
  • the compound was prepared by a procedure as described in Example 66, by using 4-ethylamino- 3,3-dimethylpiperidine, m. p.
  • the compound was prepared by a procedure as described in Example 66, by using 4- dimethylamino-3, 3 -dimethylpiperidine, m. p. 209-210 °C, C 23 H 29 FN 3 O 3 , m/z 416 (M+l), PMR
  • Example - 75 trans- 1 -Cyclopropyl-6-fluoro- 1 ,4-dihydro-8-ethyl -7-(4-amino-3-methyl- 1 -piperidinyl) -4-oxo- quinoline-3-carboxylic acid.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Oncology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
PCT/IN2002/000232 2001-12-13 2002-12-12 New generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments WO2003050107A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU2002361493A AU2002361493A1 (en) 2001-12-13 2002-12-12 New generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments
JP2003551131A JP2005511743A (ja) 2001-12-13 2002-12-12 新世代の三重標的化したキラルな広域スペクトラム抗菌性の7位置換ピペリジノ−キノロンカルボン酸誘導体、その調製方法、組成物、および医薬としての使用
EP02796957A EP1453824A1 (en) 2001-12-13 2002-12-12 New generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments
CA002468190A CA2468190A1 (en) 2001-12-13 2002-12-12 New generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US34116501P 2001-12-13 2001-12-13
US60/341,165 2001-12-13
US10/128,996 2002-04-23
US10/128,996 US6964966B2 (en) 2001-04-25 2002-04-23 Generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments
PCT/IN2002/000111 WO2002085886A2 (en) 2001-04-25 2002-04-24 Chiral, broad-spectrum antibacterial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation and compositions
INPCT/IN02/00111 2002-04-24

Publications (1)

Publication Number Publication Date
WO2003050107A1 true WO2003050107A1 (en) 2003-06-19

Family

ID=27271958

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2002/000232 WO2003050107A1 (en) 2001-12-13 2002-12-12 New generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments

Country Status (4)

Country Link
EP (1) EP1453824A1 (ja)
JP (1) JP2005511743A (ja)
AU (1) AU2002361493A1 (ja)
WO (1) WO2003050107A1 (ja)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005026145A2 (en) * 2003-09-12 2005-03-24 Warner-Lambert Company Llc Quinolone antibacterial agents
WO2005066154A2 (en) * 2003-11-20 2005-07-21 Wockhardt Limited Polymorphs of 1-cyclopropyl-6-fuoro-8-methoxy-7-(4-amino-3, 3-dimethylpiperidin-1-yl) -1, 4-dihydro-4-oxo-quinoline-3-carboxylic acid hydrochloride and mesalyte salts
WO2009046383A1 (en) * 2007-10-05 2009-04-09 Cylene Pharmaceuticals, Inc. Quinolone analogs and methods related thereto
US7569563B2 (en) 2005-06-30 2009-08-04 Aicuris Gmbh & Co. Hk Substituted quinolones II
US7867992B2 (en) 2004-07-21 2011-01-11 Aicuris Gmbh & Co. Kg Substituted quinolones
US7902227B2 (en) 2007-07-27 2011-03-08 Janssen Pharmaceutica Nv. C-7 isoxazolinyl quinolone / naphthyridine derivatives useful as antibacterial agents
US7977349B2 (en) 2006-02-09 2011-07-12 Aicuris Gmbh & Co. Kg Substituted quinolones III
RU2547835C1 (ru) * 2014-03-06 2015-04-10 Федеральное государственное бюджетное учреждение науки Институт органического синтеза им. И.Я. Постовского Уральского отделения Российской академии наук Азолоазиниевые соли фторхинолонов, обладающие антибактериальным и противовирусным действием
WO2018011017A1 (de) 2016-07-11 2018-01-18 Bayer Pharma Aktiengesellschaft 7-substituierte 1-pyridyl-naphthyridin-3-carbonsäureamide und ihre verwendung
EP3296298A1 (de) 2016-09-14 2018-03-21 Bayer Pharma Aktiengesellschaft 7-substituierte 1-aryl-naphthyridin-3-carbonsäureamide und ihre verwendung
WO2018050510A1 (de) 2016-09-14 2018-03-22 Bayer Aktiengesellschaft 7-substituierte 1-aryl-naphthyridin-3-carbonsäureamide und ihre verwendung
US9957282B2 (en) 2015-12-14 2018-05-01 Senhwa Biosciences, Inc. Crystalline forms of quinolone analogs and their salts
US10435403B2 (en) 2015-06-09 2019-10-08 Bayer Pharma Aktiengesellschaft Positive allosteric modulators of muscarinic M2 receptor
US10857156B2 (en) 2015-11-20 2020-12-08 Senhwa Biosciences, Inc. Combination therapy of tetracyclic quinolone analogs for treating cancer
US11524012B1 (en) 2018-02-15 2022-12-13 Senhwa Biosciences, Inc. Quinolone analogs and their salts, compositions, and method for their use

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009107571A1 (ja) * 2008-02-27 2009-09-03 住友化学株式会社 アルキルピペリジン-3-イルカーバメートの光学分割方法およびその中間体
JP2009256337A (ja) * 2008-03-26 2009-11-05 Sumitomo Chemical Co Ltd ピペリジン−3−イルカーバメート化合物の製造方法

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4399134A (en) * 1980-11-10 1983-08-16 Otsuka Pharmaceutical Co., Ltd. Pyrroloquinoline and benzoquinolizine compounds and antimicrobial compositions
US4894458A (en) * 1986-03-17 1990-01-16 Kyorin Pharmaceutical Co., Ltd. Quinolonecarboxylic acid derivatives and their preparation
WO1997031000A1 (en) * 1996-02-21 1997-08-28 Darwin Discovery Limited Quinolones and their therapeutic use
WO2000018404A1 (en) * 1998-09-30 2000-04-06 Alcon Laboratories, Inc. Antibiotic compositions for treatment of the eye, ear and nose
WO2000068229A2 (en) * 1999-05-07 2000-11-16 Wockhardt Limited (s)-benzoquinolizine carboxylic acids and their use as antibacterial agents
WO2001085728A2 (en) * 2000-05-08 2001-11-15 Wockhardt Limited Antibacterial chiral 8-(substituted piperidino)-benzo [i, j] quinolizines, processes, compositions and methods of treatment
WO2002009758A2 (en) * 2000-08-01 2002-02-07 Wockhardt Limited Inhibitors of cellular efflux pumps of microbes

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4399134A (en) * 1980-11-10 1983-08-16 Otsuka Pharmaceutical Co., Ltd. Pyrroloquinoline and benzoquinolizine compounds and antimicrobial compositions
US4894458A (en) * 1986-03-17 1990-01-16 Kyorin Pharmaceutical Co., Ltd. Quinolonecarboxylic acid derivatives and their preparation
WO1997031000A1 (en) * 1996-02-21 1997-08-28 Darwin Discovery Limited Quinolones and their therapeutic use
WO2000018404A1 (en) * 1998-09-30 2000-04-06 Alcon Laboratories, Inc. Antibiotic compositions for treatment of the eye, ear and nose
WO2000068229A2 (en) * 1999-05-07 2000-11-16 Wockhardt Limited (s)-benzoquinolizine carboxylic acids and their use as antibacterial agents
WO2001085728A2 (en) * 2000-05-08 2001-11-15 Wockhardt Limited Antibacterial chiral 8-(substituted piperidino)-benzo [i, j] quinolizines, processes, compositions and methods of treatment
WO2002009758A2 (en) * 2000-08-01 2002-02-07 Wockhardt Limited Inhibitors of cellular efflux pumps of microbes

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHU, DANIEL T. W. ET AL: "Synthesis and structure-activity relationships of novel arylfluoroquinolone antibacterial agents", J. MED. CHEM. (1985), 28(11), 1558-64, XP001061840 *
JAEN-OLTRA, JOSE ET AL: "Artificial neural network applied to prediction of fluorquinolone antibacterial activity by topological methods", JOURNAL OF MEDICINAL CHEMISTRY (2000), 43(6), 1143-1148, XP002212820 *
SUTO, MARK J. ET AL: "Fluoroquinolones: relationships between structural variations, mammalian cell cytotoxicity and antimicrobial activity", J. MED. CHEM. (1992), 35(25), 4745-50, XP001094135 *

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005026145A3 (en) * 2003-09-12 2005-06-16 Warner Lambert Co Quinolone antibacterial agents
WO2005026145A2 (en) * 2003-09-12 2005-03-24 Warner-Lambert Company Llc Quinolone antibacterial agents
WO2005066154A2 (en) * 2003-11-20 2005-07-21 Wockhardt Limited Polymorphs of 1-cyclopropyl-6-fuoro-8-methoxy-7-(4-amino-3, 3-dimethylpiperidin-1-yl) -1, 4-dihydro-4-oxo-quinoline-3-carboxylic acid hydrochloride and mesalyte salts
WO2005066154A3 (en) * 2003-11-20 2005-10-27 Prasad Keshav Deshpande Polymorphs of 1-cyclopropyl-6-fuoro-8-methoxy-7-(4-amino-3, 3-dimethylpiperidin-1-yl) -1, 4-dihydro-4-oxo-quinoline-3-carboxylic acid hydrochloride and mesalyte salts
US7538221B2 (en) 2003-11-20 2009-05-26 The Company Of Wockhardt Limited Polymorphs of racemic, dextrorotatory, and levorotatory enantioners of 1-cyclopropyl-6-fluoro-8-methoxy-7-(4-amino-3,3-dimethylpiperidin-1-yl)-1,4-dihydro-4-oxo-quinoline-3-carboxylic acid hydrochloride and mesylate salts
US7867992B2 (en) 2004-07-21 2011-01-11 Aicuris Gmbh & Co. Kg Substituted quinolones
US7569563B2 (en) 2005-06-30 2009-08-04 Aicuris Gmbh & Co. Hk Substituted quinolones II
US7977349B2 (en) 2006-02-09 2011-07-12 Aicuris Gmbh & Co. Kg Substituted quinolones III
US7902227B2 (en) 2007-07-27 2011-03-08 Janssen Pharmaceutica Nv. C-7 isoxazolinyl quinolone / naphthyridine derivatives useful as antibacterial agents
CN101888780B (zh) * 2007-10-05 2014-08-13 生华生物科技股份有限公司 喹诺酮类似物及其相关方法
US7928100B2 (en) 2007-10-05 2011-04-19 Cylene Pharmaceuticals, Inc. Quinolone analogs and methods related thereto
CN101888780A (zh) * 2007-10-05 2010-11-17 赛林药物股份有限公司 喹诺酮类似物及其相关方法
WO2009046383A1 (en) * 2007-10-05 2009-04-09 Cylene Pharmaceuticals, Inc. Quinolone analogs and methods related thereto
US8853234B2 (en) 2007-10-05 2014-10-07 Senhwa Biosciences, Inc. Quinolone analogs and methods related thereto
RU2547835C1 (ru) * 2014-03-06 2015-04-10 Федеральное государственное бюджетное учреждение науки Институт органического синтеза им. И.Я. Постовского Уральского отделения Российской академии наук Азолоазиниевые соли фторхинолонов, обладающие антибактериальным и противовирусным действием
US10435403B2 (en) 2015-06-09 2019-10-08 Bayer Pharma Aktiengesellschaft Positive allosteric modulators of muscarinic M2 receptor
US11229654B2 (en) 2015-11-20 2022-01-25 Senhwa Biosciences, Inc. Combination therapy of tetracyclic quinolone analogs for treating cancer
US10857156B2 (en) 2015-11-20 2020-12-08 Senhwa Biosciences, Inc. Combination therapy of tetracyclic quinolone analogs for treating cancer
US9957282B2 (en) 2015-12-14 2018-05-01 Senhwa Biosciences, Inc. Crystalline forms of quinolone analogs and their salts
US10519154B2 (en) 2016-07-11 2019-12-31 Bayer Pharma Aktiengesellschaft 7-substituted 1-pyridyl-naphthyridine-3-carboxylic acid amides and use thereof
WO2018011017A1 (de) 2016-07-11 2018-01-18 Bayer Pharma Aktiengesellschaft 7-substituierte 1-pyridyl-naphthyridin-3-carbonsäureamide und ihre verwendung
WO2018050510A1 (de) 2016-09-14 2018-03-22 Bayer Aktiengesellschaft 7-substituierte 1-aryl-naphthyridin-3-carbonsäureamide und ihre verwendung
EP3296298A1 (de) 2016-09-14 2018-03-21 Bayer Pharma Aktiengesellschaft 7-substituierte 1-aryl-naphthyridin-3-carbonsäureamide und ihre verwendung
US10927109B2 (en) 2016-09-14 2021-02-23 Bayer Aktiengesellschaft 7-substituted 1-aryl-naphthyridine-3-carboxylic acid amides and use thereof
US11472803B2 (en) 2016-09-14 2022-10-18 Bayer Aktiengesellschaft 7-substituted 1-aryl-naphthyridine-3-carboxylic acid amides and use thereof
US11524012B1 (en) 2018-02-15 2022-12-13 Senhwa Biosciences, Inc. Quinolone analogs and their salts, compositions, and method for their use

Also Published As

Publication number Publication date
AU2002361493A1 (en) 2003-06-23
JP2005511743A (ja) 2005-04-28
EP1453824A1 (en) 2004-09-08

Similar Documents

Publication Publication Date Title
US7626032B2 (en) Generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments
US6964966B2 (en) Generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments
EP1015445B1 (en) Antimicrobial quinolones, their compositions and uses
JP5153329B2 (ja) (r/s)リファマイシン誘導体、その調製、および医薬組成物
EP1453824A1 (en) New generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments
KR101490222B1 (ko) 항균성 퀴놀린 유도체
JP6552550B2 (ja) 抗菌性ピペリジニル置換3,4−ジヒドロ−1h−[1,8]−ナフチリジノン類
KR101307717B1 (ko) 트리―, 테트라― 치환―3―아미노피롤리딘 유도체
CA2417799A1 (en) Inhibitors of cellular efflux pumps of microbes
US7019143B2 (en) Antimicrobial quinolones, their compositions and uses
WO2018165614A1 (en) Bacterial efflux pump inhibitors
JP2017222646A (ja) 抗菌性ホモピペリジニル置換3,4−ジヒドロ−1h−[1,8]−ナフチリジノン類
WO2001085728A2 (en) Antibacterial chiral 8-(substituted piperidino)-benzo [i, j] quinolizines, processes, compositions and methods of treatment
US6509349B1 (en) Antimicrobial 2-pyridones, their compositions and uses
US6608078B2 (en) Antibacterial chiral 8-(substituted piperidino)-benzo [i,j] quinolizines, processes, compositions and methods of treatment
KR20160003244A (ko) 퀴놀론 유도체
IE65175B1 (en) 7-(2-methyl-4-aminopyrrolidin-1-yl) naphthyridine and quinoline compounds
CA2468190A1 (en) New generation triple-targeting, chiral, broad-spectrum antimicrobial 7-substituted piperidino-quinolone carboxylic acid derivatives, their preparation, compositions and use as medicaments
CA2869453A1 (en) Antibacterial quinoline derivatives
Bhawsar et al. WCK 1152, WCK 1153: Discovery and structure activity relationship for the treatment of resistant pneumococcal and staphylococcal respiratory infections
WO2005066154A2 (en) Polymorphs of 1-cyclopropyl-6-fuoro-8-methoxy-7-(4-amino-3, 3-dimethylpiperidin-1-yl) -1, 4-dihydro-4-oxo-quinoline-3-carboxylic acid hydrochloride and mesalyte salts
WO2005030752A1 (ja) 8−シアノキノロンカルボン酸誘導体

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 274/MUMNP/2004

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2002796957

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2468190

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2003551131

Country of ref document: JP

WWP Wipo information: published in national office

Ref document number: 2002796957

Country of ref document: EP