WO2017199265A1 - Heterocyclic compounds useful as anti-bacterial agents and method for production - Google Patents

Heterocyclic compounds useful as anti-bacterial agents and method for production Download PDF

Info

Publication number
WO2017199265A1
WO2017199265A1 PCT/IN2017/050188 IN2017050188W WO2017199265A1 WO 2017199265 A1 WO2017199265 A1 WO 2017199265A1 IN 2017050188 W IN2017050188 W IN 2017050188W WO 2017199265 A1 WO2017199265 A1 WO 2017199265A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
amino
alkyl
oxooxazolidin
ethyl
Prior art date
Application number
PCT/IN2017/050188
Other languages
French (fr)
Inventor
Shahul Hameed Peer Mohamed
Nagakumar Bharatham
Nainesh Katagihallimath
Sreevalli SHARMA
Radha NANDISHAIAH
Original Assignee
Bugworks Research India Pvt Ltd
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 to EP17737888.2A priority Critical patent/EP3458452B1/en
Priority to BR112018073743-7A priority patent/BR112018073743A2/en
Priority to CA3024824A priority patent/CA3024824A1/en
Priority to AU2017267631A priority patent/AU2017267631A1/en
Priority to CN201780029570.5A priority patent/CN109476652B/en
Priority to RU2018142830A priority patent/RU2018142830A/en
Application filed by Bugworks Research India Pvt Ltd filed Critical Bugworks Research India Pvt Ltd
Priority to MX2018014165A priority patent/MX2018014165A/en
Priority to JP2019513506A priority patent/JP6671541B2/en
Priority to US16/301,887 priority patent/US10711011B2/en
Priority to KR1020187036214A priority patent/KR20190034154A/en
Publication of WO2017199265A1 publication Critical patent/WO2017199265A1/en
Priority to ZA2018/08531A priority patent/ZA201808531B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/5381,4-Oxazines, e.g. morpholine ortho- or peri-condensed with carbocyclic ring systems
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53831,4-Oxazines, e.g. morpholine ortho- or peri-condensed with heterocyclic ring systems
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53861,4-Oxazines, e.g. morpholine spiro-condensed or forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present invention relates to the field of medicinal chemistry and more particularly to the development of antimicrobial compounds effective against bacteria, virus, fungi and protozoa including spectrum of Gram-negative and Gram-positive pathogens.
  • the present disclosure relates to compounds of Formula I, its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof and pharmaceutical compositions containing them as the active ingredient.
  • the present disclosure further relates to the synthesis and characterization of aforementioned compounds to exhibit high antimicrobial activity.
  • the compounds of the present disclosure are useful as medicaments and their use in the manufacture of medicaments for treatment, prevention or suppression of diseases, and conditions mediated by microbes.
  • the present invention also provides evidence for treating infection caused by microbes.
  • bacterial pathogens may be classified as either Gram- positive or Gram-negative pathogens.
  • Antibiotic compounds with effective activity against both Gram-positive and Gram-negative pathogens are regarded as having a broad spectrum of activity.
  • Current antibacterial drugs used to treat and prevent bacterial infection have been found to have limited effect.
  • new compounds with potent antibacterial activity with reduced potential for developing resistance, which possess improved efficacy against bacterial infections that resist treatment with currently available antibiotics, or which possess selectivity against target microorganisms.
  • the present disclosure is based on the surprising discovery that compounds of Formula I (see below) exhibits advantageous antimicrobial properties.
  • the present disclosure provides a compound of Formula I
  • Ri is selected from the group consisting of hydrogen, Ci- 6 alkyl, C 2 - 6 alkenyl, and C3-6 cycloalkyl, each of which is unsubstituted or substituted with 1 to 3 groups independently selected from halogen, hydroxy, amino, oxetane, -OCi_ 6 alkyl, C 3 - 6 cycloalkylamino, Ci- 6 alkylamino, or di(Ci- 6 alkyl)amino;
  • R 2 is selected from the group consisting of hydrogen, hydroxyl, and amino
  • X is -NH, -NCi-6 alkyl, O, or CR 3 R4;
  • R 3 and R4 are independently selected from the group consisting of hydrogen, halogen, and
  • R5 is selected from the group consisting of hydrogen, halogen, cyano, -OCi- 6 alkyl, -OQ- 6 haloalkyl, and Ci- 6 alkyl;
  • Y 2 is N or CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, halogen, cyano, -OCi- 6 alkyl, and - OCi-6 haloalkyl;
  • Zi is NH when Z 2 is C 1-6 alkylene; or Zi is C 1-6 alkylene when Z 2 is NH;
  • n 1 or 2;
  • W is CH 2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
  • Ring A is selected from the group consisting of a 3-10 membered substituted or unsubstituted heteroaryl ring system which is unsaturated or partially unsaturated optionally having up to three heteroatom independently selected from O, N, NH or S.
  • the present disclosure further relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in killing or inhibiting the growth of a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
  • the present disclosure further relates to use of a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in killing or inhibiting the growth of a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
  • the present disclosure further relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in treating a disease or condition in a patient wherein said disease or condition is caused by a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
  • the present disclosure further relates to use of a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in treating disease or condition in a patient, wherein said disease or condition is caused by a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
  • the patient is a typically a mammal, preferably a human.
  • the present disclosure further relates to a method of treating a disease or condition in a patent, said method comprising administering to a patient a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein said disease or condition is caused by microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
  • the present disclosure relates to a composition
  • a composition comprising a compound of
  • the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, together with a pharmaceutically acceptable carrier, optionally in combination with one or more other pharmaceutical compositions.
  • the present disclosure relates to a process of preparation of compound of
  • Formula I or its stereoisomers pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof.
  • the present disclosure relates to a process of preparation of a composition
  • a composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof together with a carrier.
  • the present disclosure relates to a process of preparation of pharmaceutical composition
  • a process of preparation of pharmaceutical composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, together with a pharmaceutically acceptable carrier, optionally in combination with one or more other pharmaceutical compositions.
  • Figure 1 illustrates the MIC 90 of Example 14 against clinical strains of four major gram negative bacterial species, in accordance with an embodiment of the present disclosure.
  • Figure 2 illustrates the in vivo efficacy of Example 13 in mice thigh infection model, in accordance with an embodiment of the present disclosure.
  • C x _ y indicates the numerical range of carbon atoms that are present in the group; for example, Ci_ 6 alkyl includes Ci alkyl (methyl), C 2 alkyl (ethyl), C 3 alkyl (propyl and isopropyl) and C 4 alkyl (butyl, 1 -methylpropyl, 2- methylpropyl, and i-butyl).
  • the bonding atom of a group may be any suitable atom of that group; for example, propyl includes prop-l-yl and prop-2-yl.
  • alkyl refers to a monoradical branched or unbranched saturated hydrocarbon chain having from 1 to 10 carbon atoms. This term is exemplified by groups such as n-butyl, iso-butyl, t-butyl, n-hexyl, n-decyl, and the like. The groups may be optionally substituted.
  • alkyl ene refers to a diradical branched or unbranched saturated hydrocarbon chain having from 1 to 6 carbon atoms. This term is exemplified by groups such as methylene, ethylene, propylene, butylene, hexylene, and the like. The groups may be optionally substituted. Representative substituted alkylene groups include hydroxyl substituted alkylenes.
  • alkenyl refers to a monoradical of a branched or unbranched unsaturated hydrocarbon group preferably having from 2, 3, 4, 5, or 6 carbon atoms and having 1, 2, or 3, double bond (vinyl), preferably 1 double bond.
  • the groups may be optionally substituted.
  • cycloalkyl refers to carbocyclic groups of from 3 to 12 carbon atoms having a single cyclic ring or multiple condensed rings which may be partially unsaturated.
  • Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, and the like, or multiple ring structures or carbocyclic groups to which is fused an aryl group, for example indane, and the like.
  • the groups may be optionally substituted.
  • alkoxyl refers to an alkyl group, as defined above, having an oxygen radical attached thereto.
  • Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like.
  • An "ether” is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as can be represented by one of -O-alkyl, - O- alkenyl, -O-alkynyl.
  • Halo or "Halogen”, alone or in combination with any other term means halogens such as chloro (CI), fluoro (F), bromo (Br), and iodo (I).
  • heteroaryl refers to an heteroaromatic carbocyclic group of 3 to 10 carbon atoms having a single ring (e.g. pyridine) or multiple rings (e.g. isoquinoline), or multiple condensed (fused) rings.
  • Preferred heteroaryls include fhiophene, pyrazole, thiazole, pyridine and the like. The groups may be optionally substituted.
  • the term "substituted" is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds.
  • Illustrative substituents include, for example, those described herein above.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
  • the term "effective amount” means an amount of a compound or composition which is sufficient enough to significantly and positively modify the symptoms and/or conditions to be treated (e.g., provide a positive clinical response).
  • the effective amount of an active ingredient for use in a pharmaceutical composition will vary with the particular condition being treated, the severity of the condition, the duration of the treatment, the nature of concurrent therapy, the particular active ingredient(s) being employed, the particular pharmaceutically-acceptable excipient(s)/carrier(s) utilized, the route of administration, and like factors within the knowledge and expertise of the attending physician
  • the compounds described herein may contain one or more chiral centers and/or double bonds and therefore, may exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers), regioisomers, enantiomers or diastereomers. Accordingly, the chemical structures depicted herein encompass all possible enantiomers and stereoisomers of the illustrated or identified compounds including the stereoisomerically pure form (e.g., geometrically pure, enantiomerically pure or diastereomerically pure) and enantiomeric and stereoisomeric mixtures.
  • stereoisomers such as double-bond isomers (i.e., geometric isomers), regioisomers, enantiomers or diastereomers.
  • Enantiomeric and stereoisomeric mixtures can be resolved into their component enantiomers or stereoisomers using separation techniques or chiral synthesis techniques well known to the person skilled in the art.
  • the compounds may also exist in several tautomeric forms including the enol form, the keto form and mixtures thereof. Accordingly, the chemical structures depicted herein encompass all possible tautomeric forms of the illustrated or identified compounds.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable salt” embraces salts with a pharmaceutically acceptable acid or base.
  • Pharmaceutically acceptable acids include both inorganic acids, for example hydrochloric, sulphuric, phosphoric, diphosphoric, hydrobromic, hydroiodic and nitric acid and organic acids, for example citric, fumaric, maleic, malic, mandelic, ascorbic, oxalic, succinic, tartaric, benzoic, acetic, methanesulphonic, ethanesulphonic, benzenesulphonic or p-toluenesulphonic acid.
  • Pharmaceutically acceptable bases include alkali metal (e.g. sodium or potassium) and alkali earth metal (e.g. calcium or magnesium) hydroxides and organic bases, for example alkyl amines, arylalkyl amines and heterocyclic amines.
  • polymorphs refers to crystal forms of the same molecule, and different polymorphs may have different physical properties such as, for example, melting temperatures, heats of fusion, solubilities, dissolution rates and/or vibrational spectra as a result of the arrangement or conformation of the molecules in the crystal lattice.
  • solvate refers to a crystal form of a substance which contains solvent.
  • hydrate refers to a solvate wherein the solvent is water.
  • drug sensitive bacterium is a bacterium which is not able to survive exposure to at least one drug.
  • Ri is selected from the group consisting of hydrogen, Ci- 6 alkyl, C 2 - 6 alkenyl, and C3-C6 cycloalkyl, each of which is unsubstituted or substituted with 1 to 3 groups independently selected from halogen, hydroxy, amino, oxetane, -OCi_ 6 alkyl, C 3 - 6 cycloalkylamino, Ci- 6 alkylamino, or di(Ci- 6 alkyl)amino;
  • R 2 is selected from the group consisting of hydrogen, hydroxyl, and amino
  • X is -NH, -NCi-6 alkyl, O, or CR 3 R4;
  • R 3 and R4 are independently selected from the group consisting of hydrogen, halogen, and
  • Yi is N or CR 5 ;
  • R5 is selected from the group consisting of hydrogen, halogen, cyano, -OCi- 6 alkyl, -OCi- 6 haloalkyl, and Ci- 6 alkyl;
  • Y 2 is N or CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, halogen, cyano, -OCi- 6 alkyl, Ci- 6 alkyl, and -OCi- 6 haloalkyl;
  • Zi is NH when Z 2 is Ci_ 6 alkylene; or Zi is Ci_ 6 alkylene when Z 2 is NH;
  • n 1 or 2;
  • W is CH 2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
  • Ring A is selected from the group consisting of
  • Ri is selected from the group consisting of hydrogen, Ci- 6 alkyl, C 2 - 6 alkenyl, and C 3 - 6 cycloalkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, hydroxy, amino, oxetane, -OCi alkyl, C 3 - 6 cycloalkylamino, Q- 2 alkylamino, or di(Ci- 2 alkyl)amino;
  • R 2 is selected from the group consisting of hydrogen, hydroxyl, and amino
  • X is -NH, -NCi-6 alkyl, O, or CR 3 R4;
  • R 3 and R4 are independently selected from the group consisting of hydrogen, halogen, and
  • Yi is N or CR 5 ;
  • R5 is selected from the group consisting of hydrogen, halogen, cyano, - -OCi- 6 alkyl, - OCi-6 haloalkyl, and Ci- 6 alkyl;
  • Y 2 is N or CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCi- 6 alkyl, -Ci- 6 alkyl, and -OCi- 6 haloalkyl;
  • Zi is NH when Z 2 is C 1-6 alkylene; or Zi is C 1-6 alkylene when Z 2 is NH;
  • n 1 or 2;
  • W is CH 2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Zi is NH when Z 2 is C 1-6 alkylene; or Zi is Ci-6 alkylene or C 1-6 hydroxy alkylene when Z 2 is NH.
  • the present disclosure relates to a compound of Formula I
  • Ri is selected from the group consisting of hydrogen, Ci- 6 alkyl, C 2 - 6 alkenyl, and C 3 - 6 cycloalkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, hydroxy, amino, oxetane, -OCi alkyl, C 3 - 6 cycloalkylamino, d- 2 alkylamino, or di(Ci- 2 alkyl)amino;
  • R 2 is selected from the group consisting of hydrogen, hydroxyl, and amino
  • X is -NH, -NCi alkyl, O, or CR 3 R4;
  • R 3 and R4 are independently selected from the group consisting of hydrogen, fluorine, and Ci alkyl;
  • Yi is N or CR 5 ;
  • R5 is selected from the group consisting of hydrogen, fluorine, chlorine, cyano, -OCH 3 , - OCF 3 , OCHF 2 , and Ci alkyl;
  • Y 2 is N or CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH3, - OCHF 2 , and -OCF 3 ;
  • Zi is NH when Z 2 is CH 2 ; or Zi is CH 2 , CH 2 CH 2 or CH(OH)CH 2 when Z 2 is NH;
  • n 1 or 2
  • W is O when dotted line (— ) represents either a bond or no bond;
  • Ring A is selected from the group consisting of
  • the present disclosure relates to a compound
  • Ri is selected from the group consisting of hydrogen, Ci- 6 alkyl, C 2 - 6 alkenyl, and C 3 - 6 cycloalkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from halogen, hydroxy, amino, oxetane, -OCi_ 6 alkyl, C 3 - 6 cycloalkylamino, Ci- 6 alkylamino, or di(Ci- 6 alkyl)amino;
  • R 2 is selected from the group consisting of hydrogen, hydroxyl, and amino
  • X is -NH, -NCi-6 alkyl, O, or CR 3 R4;
  • R 3 and R4 are independently selected from the group consisting of hydrogen, halogen, and
  • Yi is N or CR 5 ;
  • R5 is selected from the group consisting of hydrogen, halogen, cyano, -OCi_ 6 alkyl, -OCi_ 6 haloalkyl, and Ci_ 6 alkyl;
  • Y 2 is N or CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, halogen, cyano, Ci_ 6 alkyl, -OCi_ 6 alkyl, and -OCi_ 6 haloalkyl;
  • Zi is NH when Z 2 is Ci_ 6 alkylene; or Zi is Ci_ 6 alkylene when Z 2 is NH;
  • n 1 or 2;
  • W is CH 2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
  • Ring A is selected from the group consisting of
  • Ri is selected from the group consisting of hydrogen, Ci- 6 alkyl, C 2 - 6 alkenyl, and C 3 -C 6 cycloalkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, hydroxy, amino, oxetane, -OCi alkyl, C 3 -C 6 cycloalkylamino, d- 2 alkylamino, or di(Ci- 2 alkyl)amino;
  • R 2 is selected from the group consisting of hydrogen, hydroxyl, and amino;
  • X is -NH, -NCi-6 alkyl, O, or CR 3 R 4 ;
  • R and R4 are independently selected from the group consisting of hydrogen, fluorine, and
  • Yi is N or CR 5 ;
  • R5 is selected from the group consisting of hydrogen, fluorine, chlorine, cyano, -OCH3, - OCF 3 , OCHF 2 , and Ci alkyl;
  • Y 2 is N or CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH 3 , and -OCF 3 ;
  • Zi is NH when Z 2 is CH 2 ; or Zi is CH 2 , CH 2 CH 2 or CH(OH)CH 2 when Z 2 is NH;
  • n 1 or 2;
  • W is CH 2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
  • Ring A is selected from the group consisting of
  • the present disclosure relates to a compound
  • Ri is selected from the group consisting of hydrogen, C 1 -4 alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, amino, hydroxy, oxetane, C3-C6 cycloalkylamino, C 1 -2 alkylamino, or di(Ci-2 alkyl) amino;
  • R 2 is selected from the group consisting of hydrogen, amino, and hydroxyl
  • X is O, -NCialkyl, or CR 3 R 4 ;
  • R 3 and R 4 are independently selected from the group consisting of hydrogen, fluorine, and Ci alkyl;
  • Yi is N or CR 5 ;
  • R5 is selected from the group consisting of hydrogen, fluorine, chlorine, cyano, -OCi alkyl, -OCF 3 , OCHF 2 , and Ci alkyl;
  • Y 2 is N or CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCi alkyl, and - OCF 3 ;
  • Zi is NH when Z 2 is CH 2 ; or Z x is CH 2 , CH 2 CH 2 or CH(OH)CH 2 when Z 2 is NH;
  • n 1 or 2;
  • W is CH 2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
  • Ring A is selected from the group consisting of:
  • Ri is selected from the group consisting of hydrogen, Q- 4 alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, hydroxy, amino, oxetane, C 3 - 6 cycloalkylamino, d- 2 alkylamino, or di(Ci- 2 alkyl) amino;
  • R 2 is selected from the group consisting of hydrogen, amino, and hydroxyl;
  • R 3 and R 4 are independently selected from the group consisting of hydrogen, and Ci alkyl;
  • Yi is N or CR 5 ;
  • R 5 is selected from the group consisting of hydrogen, fluorine, chlorine, cyano, -OCH 3 , - CF 3 , OCHF 2 , and Ci alkyl;
  • Y 2 is N or CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH3, -OCHF 2 , and -OCF 3 ;
  • Zi is NH when Z 2 is CH 2 ; or Z x is CH 2 , CH 2 CH 2 or CH(OH)CH 2 when Z 2 is NH;
  • n 1 or 2;
  • W is CH 2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
  • Ring A is selected from the group consisting of
  • the present disclosure relates to a compound of Formula I
  • Ri isCi alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, hydroxy, amino, oxetane, Ci- 2 alkylamino, or di(Ci- 2 alkyl) amino;
  • R 2 is hydrogen
  • X is CR 3 R 4 ;
  • R 3 and R 4 are independently selected from the group consisting of hydrogen, and Ci alkyl;
  • Yi is N or CR 5 ;
  • R5 is hydrogen, Ci alkyl, or cyano
  • Y 2 is CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH 3 , and -OCF 3 ;
  • Zi is NH when Z 2 is CH 2 ; or
  • Z x is CH 2 , CH 2 CH 2 or CH(OH)CH 2 when Z 2 is NH;
  • n 1 or 2;
  • W is CH 2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
  • Ring A is selected from the group consisting of
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein
  • Ri is C 2 alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, amino, hydroxyl, d- 2 alkylamino, or di(Ci- 2 alkyl) amino;
  • R 2 is hydrogen
  • X is O or CR 3 R 4 ;
  • R 3 and R 4 are independently selected from the group consisting of hydrogen, and Ci alkyl;
  • Yi is N or CR 5 ;
  • R5 is selected from the group consisting of hydrogen, Ci alkyl, and cyano
  • Y 2 is CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH 3 , OCHF 2 and-OCF 3 ;
  • Zi is NH when Z 2 is CH 2 ; or Zi is CH 2 , CH 2 CH 2 or CH(OH)CH 2 when Z 2 is NH;
  • n 1 or 2;
  • W is O when dotted line (— ) represents either a bond or no bond; ing A is selected from the group consisting of
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein
  • Ri is Ci alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, amino, hydroxyl, d- 2 alkylamino, or di(Ci-2 alkyl) amino;
  • R 2 is hydrogen
  • X is O
  • R 3 and R4 are independently selected from the group consisting of hydrogen, and Q alkyl
  • Yi is N or CR 5 ;
  • R5 is selected from the group consisting of hydrogen, Ci alkyl, and cyano
  • Y 2 is CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH 3 , OCHF 2 and-OCF 3 ;
  • Zi is NH when Z 2 is CH 2 ; or Zi is CH 2 , CH 2 CH 2 or CH(OH)CH 2 when Z 2 is NH;
  • Ring A is selected from the group consisting of
  • the present disclosure relates to a compound of Formula I
  • Ri is Ci_4 alkyl
  • R 2 is hydrogen
  • R 3 and R4 are independently selected from the group consisting of hydrogen, and Q alkyl
  • Yi is N or CR 5 ;
  • R5 is hydrogen
  • Y 2 is CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH 3 , and -OCF 3 ;
  • Zi is NH when Z 2 is CH 2 ; or
  • Z x is CH 2 , CH 2 CH 2 or CH(OH)CH 2 when Z 2 is NH;
  • n 1 or 2;
  • W is O when dotted line (— ) represents either a bond or no bond;
  • Ring A is selected from the roup consisting of
  • the present disclosure relates to a compound of Formula I
  • R 2 is hydrogen
  • X is CR 3 R 4 ;
  • R 3 and R 4 are independently selected from the group consisting of hydrogen and Ci alkyl; Yi is N or CR 5 ;
  • R 5 is hydrogen, CH 3 , CN
  • Y 2 is CH
  • Y 3 is CR 6 ;
  • R 6 is hydrogen, fluorine, cyano, -OCH 3 , and -OCF 3 ;
  • Zi is NH when Z 2 is CH 2 ; or Zi is CH 2 CH 2 or CH(OH)CH 2 when Z 2 is NH;
  • n 1 or 2;
  • W is O when dotted line (— ) represents either a bond or no bond;
  • Ring A is selected from the group consisting of
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein
  • Ri is selected from the group consisting of Ci- 3 alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, amino, hydroxy, Ci- 2 alkylamino, or di(Ci- 2 alkyl)amino;
  • R 2 is hydrogen
  • X is CR 3 R4;
  • R 3 and R 4 are independently selected from the group consisting of hydrogen and Ci alkyl; Yi is N or CR 5 ; R5 is selected from the group consisting of hydrogen, CH 3 , and CN;
  • Y 2 is CH
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH 3 , and -OCF 3 ;
  • Zi is NH when Z 2 is CH 2 ; or Zi is CH 2 , CH 2 CH 2 or CH(OH)CH 2 when Z 2 is NH;
  • n 1 or 2;
  • W is O when dotted line (— ) represents either a bond or no bond;
  • Ring A is selected from the group consisting of
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof,
  • Ri is selected from the group consisting of hydrogen, Ci- 6 alkyl, C 2 - 6 alkenyl, and C 3 -C 6 cycloalkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, hydroxy, amino, oxetane, -OCi alkyl, C 3 -C 6 cycloalkylamino, Q- 2 alkylamino, or di(Ci- 2 alkyl)amino;
  • R 2 is selected from the group consisting of hydrogen, hydroxyl, and amino
  • X is -NH, -NCi-6 alkyl, O, or CR 3 R4;
  • R 3 and R4 are independently selected from the group consisting of hydrogen, halogen, and
  • Yi is N or CR 5 ;
  • R5 is selected from the group consisting of hydrogen, halogen, cyano, -OCi- 6 alkyl, -OCi- 6 haloalkyl, and Ci- 6 alkyl;
  • Y 2 is N or CH;
  • Y 3 is N or CR 6 ;
  • R 6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCi- 6 alkyl alkyl, and -OCi- 6 haloalkyl;
  • Zi is NH when Z 2 is C 1-6 alkylene; or Zi is Ci- 6 alkylene when Z 2 is NH;
  • n 1 or 2
  • W is CH 2 wherein dotted line (— ) represents no bond; W is O when dotted line (— represents either a bond or no bond;
  • Ring A is selected from the group consisting of
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein X is NH, -N(CH 3 ), O, CH 2 , CH-F, CH-CH 3 , CF 2 or C(CH 3 ) 2 .
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein X is O, -CH, C(F) 2 , or C-(CH 3 ) 2 .
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein X is -CH 2 , or C- (CH 3 ) 2 .
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein R 2 is selected from H, hydroxy or amino.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein R 2 is H.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Zi and Z 2 is independently selected from -NH-, -CH 2 , or CH 2 CH 2 .
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Y 2 is N or CH.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Y 2 is CH.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Yi is N, CH, CF, CC1, C-CN, C- OCH 3 , C-OCF 3, C-OCHF 2 , or C-CH 3 .
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Yi is CH.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Y 3 is N, -CH, -CF, -C(CN), - C(OCH 3 ), -C(OCF 3 ), or -C(OCHF 2 ).
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Y 3 is -CH, or -CF.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein n is 1 or 2
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, which is selected from a group consisting of: 6-(5-(2-((( 1 -Methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3 ,2-b] [ 1 ,4]oxazin-3(4H)-one,
  • the present disclosure relates to a process of preparation of compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in killing or inhibiting the growth of a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in killing or inhibiting the growth of Gram-positive and Gram-negative bacteria.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in killing or inhibiting the growth of drug sensitive and drug resistance bacterium selected from a group consisting of Escherichia coli, Salmonella typhi, Salmonella paratyphi, Salmonella typhimurium, Salmonella enteritidis, Shigella dysenteriae, Shigella flexneri, Shigella boydii, Shigella sonnei, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pneumoniae, Streptococcus pygenes, Stenotrophomonas maltophilia, Haemophilus influenza, Klebsiella pneumoniae, Legionella pneumophila, Mycoplasma pneumoniae, Acine
  • the present disclosure relates to use of a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in killing or inhibiting the growth of a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in treating a disease or condition in a patient wherein said disease or condition is caused by a microorganism selected from the group consisting of Gran negative and Gram positive pathogens.
  • the present disclosure relates to use of a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in treating disease or condition in a patient, wherein said disease or condition is caused by a microorganism selected from the group consisting of Gran negative and Gram positive pathogens.
  • the patient is a typically a mammal, preferably a human.
  • the present disclosure relates to a method of treating a disease or condition in a patent, said method comprising administering to a patient a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein said disease or condition is caused by microorganism selected from the group consisting of Gram negative and Gram positive pathogens.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use as a medicament.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in the preparation of medicaments for inhibiting microbial growth.
  • the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in the preparation of medicaments for inhibiting bacterial growth.
  • the present disclosure relates to medicaments that include a compound of Formula I, or an addition salt of the compound of formula I with a pharmaceutically acceptable acid or base.
  • medicaments find their use in therapeutics, especially in the treatment of bacterial infection caused by both drug sensitive and drug resistance bacterium including quinolone resistance belonging to Gram positive and Gram negative species; especially of those caused by Escherichia coli, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Clostridium difficili, Neisseria gonorrhoeae, and Enterococcus faecalis .
  • the present disclosure relates to the use of a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in the manufacture of a medicament for the treatment of an infection caused by bacterial species in a warmblooded animal, such as man.
  • the present disclosure relates to the use of a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in the manufacture of a medicament for the production of an antibacterial effect in a warm-blooded animal such as man.
  • the present disclosure relates to a method for treating bacterial infections caused by bacterial species in a warm-blooded animal such as man, said method including administering to said animal an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.
  • the present disclosure relates to a method for producing an antibacterial effect in a warm-blooded animal such as man, said method including administering to said animal an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.
  • the present disclosure relates to a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prophylaxis of bacterial infections in a warm-blooded animal, such as man.
  • the present disclosure relates to a compound of Formula I, or a pharmaceutically acceptable salt thereof, for the therapeutic and prophylactic treatment of mammals including humans, in particular in treating bacterial infections caused by bacterial species, is normally formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition.
  • the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, and at least one pharmaceutically acceptable carrier, diluent, or excipient.
  • the present disclosure relates to the use of a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in the manufacture of a medicament for the treatment of a bacterial infection caused by bacterial species in a warm-blooded animal such as man.
  • the present disclosure relates to the use of a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in the manufacture of a medicament for the production of an antibacterial effect in a warm-blooded animal such as man.
  • the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof together with a pharmaceutically acceptable carrier, and in combination with at least one antibiotic.
  • the present disclosure relates to a method for treating infection caused by bacterial species in a warm-blooded animal such as man, said method including administering to said animal an effective amount of a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof.
  • a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof.
  • the present disclosure relates to a method for producing an antibacterial effect in a warm-blooded animal such as man, said method including administering to said animal an effective amount of a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof.
  • a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof.
  • the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, together with a pharmaceutically acceptable carrier, optionally in combination with one or more other pharmaceutical compositions.
  • the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, and a pharmaceutically acceptable diluent or carrier.
  • the present disclosure relates to a composition
  • a composition comprising a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, and a carrier.
  • pharmaceutically acceptable includes compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the compounds of Formula I may form stable pharmaceutically acceptable acid or base salts, and in such cases administration of a compound as a salt may be appropriate.
  • acid addition salts include acetate, adipate, ascorbate, benzoate, benzenesulfonate, bicarbonate, bisulfate, butyrate, camphorate, camphorsulfonate, choline, citrate, cyclohexyl sulfamate, diethylenediamine, ethanesulfonate, fumarate, glutamate, glycolate, hemisulfate, 2-hydroxyethylsulfonate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, hydroxymaleate, lactate, malate, maleate, methanesulfonate, meglumine, 2-naphthalenesulfonate, nitrate, oxalate, pamoate, persulfate
  • base salts include ammonium salts; alkali metal salts such as sodium, lithium and potassium salts; alkaline earth metal salts such as aluminum, calcium and magnesium salts; salts with organic bases such as dicyclohexylamine salts and N10 methyl-D-glucamine; and salts with amino acids such as arginine, lysine, ornithine, and so forth.
  • basic nitrogen- containing groups may be quaternized with such agents as: lower alkyl halides, such as methyl, ethyl, propyl, and butyl halides; dialkyl sulfates such as dimethyl, diethyl, dibutyl; diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl halides; arylalkyl halides such as benzyl bromide and others.
  • Non-toxic physiologically acceptable salts are preferred, although other salts may be useful, such as in isolating or purifying the product.
  • the salts may be formed by conventional means, such as by reacting the free base form of the product with one or more equivalents of the appropriate acid in a solvent or medium in which the salt is insoluble, or in a solvent such as water, which is removed in vacuo or by freeze drying or by exchanging the anions of an existing salt for another anion ona suitable ion-exchange resin.
  • compositions of the disclosure may be in a form suitable for oral use (for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions), for administration by inhalation (for example as a finely divided powder or a liquid aerosol), for administration by insufflation (for example as a finely divided powder) or for parenteral administration (for example as a sterile aqueous or oily solution for intravenous, subcutaneous, intramuscular or intramuscular dosing or as a suppository for rectal dosing).
  • oral use for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixir
  • the present disclosure relates to a process of preparation of a composition
  • a composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof together with a carrier.
  • compositions of the present disclosure relate to a process of preparation of pharmaceutical composition
  • a process of preparation of pharmaceutical composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, together with a pharmaceutically acceptable carrier, optionally in combination with one or more other pharmaceutical compositions.
  • compositions of the present disclosure may be obtained by conventional procedures using conventional pharmaceutical excipients well known in the art.
  • compositions intended for oral use may contain, for example, one or more colouring, sweetening, flavouring and/or preservative agents
  • Suitable pharmaceutically acceptable excipients for a tablet formulation include, for example, inert diluents such as lactose, sodium carbonate, calcium phosphate or calcium carbonate; granulating and disintegrating agents such as corn starch or algenic acid; binding agents such as starch; lubricating agents such as magnesium stearate, stearic acid or talc; preservative agents such as ethyl or propyl j?-hydroxybenzoate; and anti- oxidants, such as ascorbic acid.
  • Tablet formulations may be uncoated or coated either to modify their disintegration and the subsequent absorption of the active ingredient within the gastrointestinal tract, or to improve their stability and/or appearance, in either case, using conventional coating agents or procedures well known in the art.
  • compositions for oral use may be in the form of hard gelatin capsules in which the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules in which the active ingredient is mixed with water or oil such as peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • soft gelatin capsules in which the active ingredient is mixed with water or oil such as peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions generally contain the active ingredient in finely powdered form or in the form of nano or micronized particles together with one or more suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents such as lecithin or condensation products of an alkylene oxide with fatty acids (for example polyoxethylene stearate), or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexito
  • the aqueous suspensions may also contain one or more preservatives such as ethyl or propyl p-hydroxybenzoate; anti-oxidants such as ascorbic acid); coloring agents; flavoring agents; and/or sweetening agents such as sucrose, saccharine or aspartame.
  • preservatives such as ethyl or propyl p-hydroxybenzoate
  • anti-oxidants such as ascorbic acid
  • coloring agents such as ascorbic acid
  • flavoring agents such as ascorbic acid
  • sweetening agents such as sucrose, saccharine or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such as arachis oil, olive oil, sesame oil or coconut oil or in a mineral oil such as liquid paraffin.
  • the oily suspensions may also contain a thickening agent such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set out above, and flavoring agents may be added to provide a palatable oral preparation.
  • These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water generally contain the active ingredient together with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients such as sweetening, flavoring and coloring agents, may also be present.
  • the pharmaceutical compositions of the disclosure may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil, such as olive oil or arachis oil, or a mineral oil, such as for example liquid paraffin or a mixture of any of these.
  • Suitable emulsifying agents may be, for example, naturally-occurring gums such as gum acacia or gum tragacanth, naturally-occurring phosphatides such as soya bean, lecithin, an esters or partial esters derived from fatty acids and hexitol anhydrides (for example sorbitan monooleate) and condensation products of the said partial esters with ethylene oxide such as polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening, flavoring and preservative agents.
  • Syrups and elixirs may be formulated with sweetening agents such as glycerol, propylene glycol, sorbitol, aspartame or sucrose, and may also contain a demulcent, preservative, flavoring and/or coloring agent.
  • compositions may also be in the form of a sterile injectable aqueous or oily suspension, which may be formulated according to known procedures using one or more of the appropriate dispersing or wetting agents and suspending agents, which have been mentioned above.
  • a sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example a solution in 1,3-butanediol.
  • compositions for administration by inhalation may be in the form of a conventional pressurized aerosol arranged to dispense the active ingredient either as an aerosol containing finely divided solid or liquid droplets.
  • Conventional aerosol propellants such as volatile fluorinated hydrocarbons or hydrocarbons may be used and the aerosol device is conveniently arranged to dispense a metered quantity of active ingredient.
  • compositions for administration may also be formulated as a liposome preparation.
  • the liposome preparation can comprise liposomes which penetrate the cells of interest or stratum corneum, and fuse with the cell membrane, resulting in delivery of the contents of the liposome into the cell.
  • Other suitable formulations can employ niosomes.
  • Niosomes are lipid vesicles similar to liposomes, with membrane consisting largely of nonoinic lipids, some forms of which are effective for transporting compounds across the stratum corneum.
  • compositions for administration may also be formulated as a depot preparation, which may be administered by implantation or by intramuscular injection.
  • the compositions may be formulated with suitable polymeric or hydrophobic material (as an emulsion in acceptable oil), ion exchange resins, or sparingly soluble derivatives.
  • the compound of the present disclosure can also be administered in sustained release forms or from sustained release drug delivery systems.
  • the amount of active ingredient that is combined with one or more excipients to produce a single dosage form will necessarily vary depending upon the host treated and the particular route of administration.
  • a formulation intended for oral administration to humans will generally contain, for example, from 0.5 mg to 4 g of active agent compounded with an appropriate and convenient amount of excipients which may vary from about 5 to about 98 percent by weight of the total composition.
  • Dosage unit forms will generally contain about 1 mg to about 500 mg of an active ingredient.
  • the size of the dose required for the therapeutic or prophylactic treatment of a particular disease state will necessarily be varied depending on the host treated, the route of administration and the severity of the illness being treated.
  • a daily dose in the range of 1 -25 mg kg is employed. Accordingly, the optimum dosage may be determined by the practitioner who is treating any particular patient.
  • the compounds disclosed herein may be applied as a sole therapy or may involve, in addition to a compound of the disclosure, one or more other substances and/or treatments. Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate administration of the individual components of the treatment. Where the administration is sequential or separate, the delay in administering the second component should not be such as to lose the beneficial effect of the combination.
  • Suitable classes and substances may be selected from one or more of the following: i) other antibacterial agents for example macrolides e.g. erythromycin, azithromycin or clarithromycin; quinolones e.g. ciprofloxacin or levofloxacin; ⁇ lactams e.g. penicillins e.g.
  • amoxicillin or piperacillin cephalosporins e.g. ceftriaxone or ceftazidime
  • carbapenems e.g. meropenem or imipenem etc
  • aminoglycosides e.g. gentamicin or tobramycin; or oxazolidinones
  • anti-infective agents for example, an antifungal triazole e.g.
  • biological protein therapeutics for example antibodies, cytokines, bactericidal/ permeability increasing protein (BPI) products; and/or iv) one or more antibacterial agents useful in the treatment of Mycobacterium tuberculosis such as one or more of rifampicin, isoniazid, pyrizinamide, ethambutol, quinolones e.g. moxifloxacin or gatifloxacin, streptomycin and/or v) efflux pump inhibitors.
  • biological protein therapeutics for example antibodies, cytokines, bactericidal/ permeability increasing protein (BPI) products
  • BPI permeability increasing protein
  • antibacterial agents useful in the treatment of Mycobacterium tuberculosis such as one or more of rifampicin, isoniazid, pyrizinamide, ethambutol, quinolones e.g. moxifloxacin or gatifloxacin, streptomycin and/or
  • the present disclosure relates to a compound of the Formula I, or a pharmaceutically acceptable salt thereof and a chemotherapeutic agent selected from: i) one or more additional antibacterial agents; and/or ii) one or more anti-infective agents; and/or iii) biological protein therapeutics for example antibodies, cytokines, bactericidal/permeability increasing protein (BPI) products; iv) one or more antibacterial agents useful in the treatment of pulmonary tuberculosis, extra-pulmonary tuberculosis, avium infections, buruli ulcers and/or v) one or more efflux pump inhibitors.
  • a chemotherapeutic agent selected from: i) one or more additional antibacterial agents; and/or ii) one or more anti-infective agents; and/or iii) biological protein therapeutics for example antibodies, cytokines, bactericidal/permeability increasing protein (BPI) products; iv) one or more antibacterial agents useful in the treatment of pulmonary tub
  • MeOD -deuterated methanol i.e. D 3 COD
  • Boc denotes t-butoxycarbonyl
  • Cbz denotes benzyloxycarbonyl
  • Atm denotes atmospheric pressure
  • rt denotes room temperature
  • mg denotes milligram
  • g denotes gram
  • ⁇ L ⁇ denotes microliter
  • mL denotes milliliter
  • L denotes liter
  • denotes micromolar
  • mM denotes millimolar
  • M denotes molar
  • N denotes normal
  • nm denotes nanometer
  • FAB mass spectral data were generally obtained using a Platform spectrometer (supplied by Micromass) run in electrospray and, where appropriate, either positive ion data or negative ion data were collected or using Agilent 1100 series LC/MS equipped with Sedex 75ELSD, and where appropriate, either positive ion data or negative ion data were collected.
  • the lowest mass major ion is reported for molecules where isotope splitting results in multiple mass spectral peaks (for example when chlorine is present).
  • Reverse Phase HPLC was carried out using YMC Pack ODS AQ (100x20 mmID, S 5A particle size, 12 nm pore size) on Agilent instruments; each intermediate was purified to the standard required for the subsequent stage and was characterized in sufficient detail to confirm that the assigned structure was correct; purity was assessed by HPLC, TLC, or NMR and identity was determined by infrared spectroscopy (IR), mass spectroscopy or NMR spectroscopy as appropriate.
  • IR infrared spectroscopy
  • Step 1 Synthesis of 6-bromo-2-nitropyridin-3-ol (la): To a solution of 4,6-dichloro-5-metho 2-nitropyridin-3-ol (20g, 0.142mol) in DMF(400ml), N- bromosuccinimide (32.52g, 0.187mol) was added portion wise over a period of 5 hours at 0°C. The reaction mixture was stirred for room temperature for 12h. After completion of the reaction, the reaction mixture was concentrated in vacuo. The residue was taken up in the Ether and the mixture was stirred for 30min.
  • Step 2 Synthesis of ethyl 2-((6-bromo-2-nitropyridin-3-yl)oxy)acetate (lb): To solution of 6-bromo-2-nitropyridin-3-ol, la (40g, 0.182mol) in Acetone (400ml), cooled to 0°C, potassium carbonate (50.4 lg, 0.365mol), was added and stirred for 5min. Then, Ethyl bromoacetate (39.7g, 0.237mol) was added slowly and refluxed at 65 °C for 12 h. After completion of the reaction, it was filtered and the filtrate was concentrated in vacuo.
  • Step 3 Synthesis of 6-bromo-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (I): To a stirred solution of ethyl 2-((6-bromo-2-nitropyridin-3-yl)oxy)acetate, lb (21g, 0.0687mol) in glacial acetic acid (400ml), Iron powder(l 1.51g, 0.2063mol) was added and heated to 100°C for 6 hours. After completion of the reaction, reaction mixture was filtered through celite bed using ethyl acetate, 10% Methanol and concentrated in vacuo.
  • Step 1 Synthesis of (but-3-en-l-yloxy)(tert-butyl)dimethylsilane, Ila:
  • Step 2 Synthesis of tert-butyldimethyl(2-(oxiran-2-yl)ethoxy)silane, lib: To a stirred solution of (but-3-en- l-yloxy)(tert-butyl)dimethylsilane, Ila (24g, 0.1287mol) in dichloromethane (480ml), cooled to 0°C, m-CPBA(63.49g, 0.2575mol), was added portion wise and stirred at 25°C for 7 hours. After completion of the reaction, reaction mixture was filtered to remove inorganics. Then, it was quenched with 10% sodium thio sulphate and extracted with dichloromethane.
  • Step 3 Synthesis of l-amino-4-((tert-butyldimethylsilyl)oxy)butan-2- ol, lie: To a stirred solution of tert-butyldimethyl(2-(oxiran-2-yl)ethoxy)silane, lib (22g, 0.1087mol) in methanol (100ml), were added NH 3 in methanol (250ml) and aqueous ammonia (100ml) and stirred at 25°C for 14 hours.
  • Step 4 Synthesis of 5-(2-((tert- butyldimethylsilyl)oxy)ethyl)oxazolidin-2-one, lid: To a stirred solution of l-amino-4- ((tert-butyldimethylsilyl)oxy)butan-2-ol, lie ( 20g, 0.0912mol) in THF (300ml), cooled to 0°C, CDI (22.5g, 0.1368mol), was added portion wise and heated at 50°C for 14 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with ethyl acetate.
  • Step 5 Synthesis of 6-(5-(2-((tert-butyldimethylsilyl)oxy)ethyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, He: To a stirred solution of 6-bromo-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, I (0.3g, 0.0013mol) and 5-(2-((tert- butyldimethylsilyl)oxy)ethyl)oxazolidin-2-one, lid ( 0.385g, 0.0015mol) in Aldrich dry THF(lOml), were added i-butyl-X-Phos mesyl chloride complex (0.051g, 0.00006mol) and sodium tert-butoxide (0.187g, 0.0019mol) and degassed for 20 mins.
  • reaction mixture was concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 21-25% ethyl acetate in pet ether to obtain 6- (5-(2-((tert-butyldimethylsilyl)oxy)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2- b][l,4]oxazin-3(4H)-one, He (0.2g, 40%).
  • Step 6 Synthesis of 6-(5-(2-hydroxyethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one,
  • Ilf To a stirred solution of 6-(5-(2-((tert- butyldimethylsilyl)oxy)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)- one, He (lg, 0.0025mol) in THF (15ml), cooled to 0°C, Tert-butyl ammonium fluoride (1.99g, 0.0076mol) was added drop wise and stirred at 25°C for 3 hours.
  • reaction mixture was quenched with water and extracted with ethyl acetate.
  • the separated organic layer was washed with brine solution, dried over Na 2 S0 4 , filtered and concentrated in vacuo to obtain colourless gummy solid of 6-(5-(2- hydroxyethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Ilf (0.5g, 71.63%).
  • the crude material was taken for the next step without any purification.
  • Step 7 Synthesis of 2-(2-oxo-3-(3-oxo-3,4-dihydro-2H-pyrido[3,2- b][l,4]oxazin-6-yl)oxazolidin-5-yl)ethyl methanesulfonate, Ilg: To a stirred solution of 6-(5-(2-hydroxyethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Ilf (0.5g, 0.0017mol) in dichloromethane, cooled to 0°C, triethylamine (0.543g, 0.0053mol) and mesyl chloride (0.155g, 0.0013mol) were added and stirred at 25°C for 2 hours.
  • reaction mixture was quenched with water and extracted with dichloromethane.
  • the separated organic layer was washed with water, brine solution, dried over Na 2 S0 4 , filtered and concentrated in vacuo to obtain pale brown gummy solid of 2-(2-oxo-3-(3-oxo-3,4-dihydro-2H-pyrido[3,2-b][l,4]oxazin-6-yl)oxazolidin-5- yl)ethyl methanesulfonate, Ilg (0.5g, 82.50%).
  • the crude material was taken for the next step without any purification.
  • Step 8 Synthesis of 6-(5-(2-azidoethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, Ilh: To a stirred solution of 2-(2-oxo-3-(3-oxo- 3,4-dihydro-2H-pyrido[3,2-b][l,4]oxazin-6-yl)oxazolidin-5-yl)ethyl methanesulfonate, Ilg (0.5g, 0.0013mol) in DMF (10ml), cooled to 0°C, sodium azide (0.181g, 0.0027mol) was added and heated at 60°C for 3 hours.
  • reaction mixture was quenched with water and extracted with ethyl acetate.
  • the separated organic layer was washed with water, brine solution, dried over Na 2 S0 4 , filtered and concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 50-60% ethyl acetate in pet ether to obtain colourless solid of 6-(5-(2- azidoethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Ilh (0.32g, 81.01%).
  • Step9 Synthesis of 6-(5-(2-aminoethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one (Intermediate II): To a stirred solution of 6-(5-(2- azidoethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, 12 (0.32g, O.OOlOmol) in THF: MeOH (1 : 1) (10ml), 10% palladium on carbon (30mg) was added and stirred at 25°C under H 2 bladder pressure for 3 hours.
  • reaction mixture was filtered through celite bed using THF and MeOH and concentrated in vacuo to obtain colourless solid of 6-(5-(2-aminoethyl)-2-oxooxazolidin- 3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, II (0.25g, 89.92%).
  • the crude material was taken for the next step without any purification.
  • Step-1 Synthesis of3-(2-oxo-3-(3-oxo-3,4-dihydro-2H-pyrido[3,2- b][l,4]oxazin-6-yl)oxazolidin-5-yl)propanenitrile(IIIa): To a stirred solution of starting material Ilg (0.7 g, 1.96 mmol) in DMSO (5 mL) was added sodium cyanide (0.38 g, 7.84 mmol), and the resulting reaction mixture was heated to 50°C for 3h. The reaction was monitored by LCMS. After completion of the reaction, the reaction mixture was poured into water (15ml) and extracted with Ethyl acetate (2 x 30 mL).
  • Step-2 Synthesis of tert-butyl (3-(2-oxo-3-(3-oxo-3,4-dihydro-2H- pyrido[3,2-b][l,4]oxazin-6-yl)oxazolidin-5-yl)propyl)carbamate (Illb): To a stirred solution of starting material, Ilia (0.1 g,0.346 mmol) in Methanol (5 mL) was added Cobalt(II)chloride hexahydrate (0.41 g, 1.734 mmol) and Boc-anhydride (0.138 g, 0.692 mmol) at 0°C.
  • Step-3 Synthesis of 6-(5-(3-Aminopropyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one(III)
  • Illb 50 mg,
  • dichloromethane 5 mL
  • trifluoroacetic acid 0.5 mL
  • the reaction mixture was concentrated in vaccuo, and washed with pet ether to afford title compound (40 mg) as light brown solid.
  • Step 1 Synthesis of (pent-3-en-l-yloxy)(tert-butyl)dimethylsilane,
  • Step 2 Synthesis of tert-butyldimethyl(2-(oxiran-2-yl)ethoxy)silane, Hid: A stirred solution of (pent-3-en-l-yloxy)(tert-butyl)dimethylsilane, IIIc (25 g, 0.125 mol) in dichloromethane (500 ml) was cooled to 0°C and m-CPBA(63.49g, 0.2575mol), was added portion wise over 20 mins. After the completion of addition, reaction was warmed to 25°C and stirred for 7 hours. Reaction mixture was filtered to remove inorganics.
  • Step 4 6-(5-(2-((tert-butyldimethylsilyl)oxy)propyl)-2-oxooxazolidin- 3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Illf: To a stirred solution of 6-bromo- 2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, I (0.300 g, 1.310 mmol) and 5-(2-((tert- butyldimethylsilyl)oxy)propyl)oxazolidin-2-one, Ille ( 0.407 g, 1.572 mmol) in Aldrich dry THF(12ml), were added t-butyl-X-Phos mesyl chloride complex (0.052g, 0.0655 mmol) and sodium tert-butoxide (0.189 g, 1.965 mmol) and degassed
  • Step 5 Synthesis of 6-(5-(3-hydroxypropyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, Illg: To a stirred solution of 6-(5-(2-((tert- butyldimethylsilyl)oxy)propyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin- 3(4H)-one, Illf (4.3g, 0.0105mol) in THF (45ml), cooled to 0°C, Tert-butyl ammonium fluoride (5.64g, 0.0216mol) was added drop wise and stirred at 25°C for 3 hours.
  • reaction mixture was quenched with water and extracted with ethyl acetate.
  • the separated organic layer was washed with brine solution, dried over Na 2 S0 4 , filtered and concentrated in vacuo to obtain colourless gummy solid of 6-(5-(3- hydroxypropyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Illg (2.95g, 95.81%).
  • the crude material was taken for the next step without any purification.
  • Step 6 Synthesis of 3-(2-oxo-3-(3-oxo-3,4-dihydro-2H-pyrido[3,2- b][l,4]oxazin-6-yl)oxazolidin-5-yl)propyl methanesulfonate, Illh: To a stirred solution of 6-(5-(3-hydroxypropyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin- 3(4H)-one, IIIg(2.95g, O.OlOmol) in dichloromethane (30ml), cooled to 0°C, triethylamine (3.05g, 0.030mol) and mesyl chloride (1.14g, O.OlOmol) were added and stirred at 25 °C for 2 hours.
  • reaction mixture was quenched with water and extracted with dichloromethane.
  • the separated organic layer was washed with water, brine solution, dried over Na 2 S0 4 , filtered and concentrated in vacuo to obtain pale brown gummy solid of 3-(2-oxo-3-(3-oxo-3,4-dihydro-2H- pyrido[3,2-b][l,4]oxazin-6-yl)oxazolidin-5-yl)propyl methanesulfonate, Illh (2.8g, 75.47%).
  • the crude material was taken for the next step without any purification.
  • Step 7 Synthesis of 6-(5-(3-azidopropyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, Illi: To a stirred solution of 3-(2-oxo-3-(3-oxo- 3,4-dihydro-2H-pyrido[3,2-b][l,4]oxazin-6-yl)oxazolidin-5-yl)propyl methanesulfonate, Illh (2.8g, 0.0075mol) in DMF (30ml), cooled to 0°C, sodium azide (0.98g, 0.0150mol) was added and heated at 60°C for 3 hours.
  • reaction mixture was quenched with water and extracted with ethyl acetate.
  • the separated organic layer was washed with water, brine solution, dried over Na 2 S0 4 , filtered and concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 38-43% ethyl acetate in pet ether to obtain colourless solid of 6-(5-(3- azidopropyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Illi (1.5g, 63.02%).
  • Step 8 Synthesis of 6-(5-(3-aminopropyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, III: To a stirred solution of 6-(5-(3-azidopropyl)- 2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one,IIIi (1.3g, O.OOlOmol) in THF: MeOH (1 : 1) (10ml), 10% palladium on carbon (0.6g) was added and stirred at 25°C under H 2 bladder pressure for 3 hours.
  • reaction mixture was filtered through celite bed using THF and MeOH and concentrated in vacuo to obtain colourless solid of 6-(5-(3-aminopropyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2- b][l,4]oxazin-3(4H)-one, III (0.8g, 67.22%).
  • the crude material was taken for the next step without any purification.
  • Step 1 Synthesis of 5-((4-((tert-butyldimethylsilyl)oxy)-2- hydroxybutyl)amino)-2-methylnicotinonitrile, Va: To a stirred solution of 5-bromo-2- methylnicotinonitrile (850mg, 4.31mmol) and 5-(2-((tert- butyldimethylsilyl)oxy)ethyl)oxazolidin-2-one, lid ( 1268mg, 5.17mmol) in Aldrich dry THF(25ml), were added i-butyl-X-Phos mesyl chloride complex (171mg, 0.21mmol) and sodium tert-butoxide (621mg, 6.47mmol) and degassed for 20 min.
  • reaction mixture was concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 26-30% ethyl acetate in pet ether to obtain 5- ((4-((tert-butyldimethylsilyl)oxy)-2-hydroxybutyl)amino)-2-methylnicotinonitrile, Va (500mg, 35%).
  • Step 2 Synthesis of 5-(5-(2-((tert-butyldimethylsilyl)oxy)ethyl)-2- oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vb: To a stirred solution of 5-((4-((tert- butyldimethylsilyl)oxy)-2-hydroxybutyl)amino)-2-methylnicotinonitrile, Va (500mg, 1.49mmol) in dry Dichlormethane (5ml), cooled to 0°C, triefhylamine (754mg, 7.54mmol) and phosgene (1170mg, 11.92mmol) were added and stirred at 25°C for 2h.
  • Vb To a stirred solution of 5-((4-((tert- butyldimethylsilyl)oxy)-2-hydroxybutyl)amino)-2-methylnicotinonitrile, Va (500mg, 1.49
  • reaction mixture was quenched with water and extracted with ethyl acetate.
  • the separated organic layer was washed with brine solution, dried over Na 2 S0 4 , filtered and concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 25-27% ethyl acetate in pet ether to obtain 5-(5-(2-((tert-butyldimethylsilyl)oxy)ethyl)-2- oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vb (340mg, 64%).
  • Step 3 Synthesis of 5-(5-(2-hydroxyethyl)-2-oxooxazolidin-3-yl)-2- methylnicotinonitrile, Vc: To a stirred solution of 5-(5-(2-((tert- butyldimethylsilyl)oxy)ethyl)-2-oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vb (340mg, 0.94mmol) in THF (8ml), cooled to 0°C, Tert-butyl ammonium fluoride (737mg, 2.82mmol) was added drop wise and stirred at 25°C for 3 hours.
  • Vc To a stirred solution of 5-(5-(2-((tert- butyldimethylsilyl)oxy)ethyl)-2-oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vb (340mg, 0.94mmol) in THF (8m
  • reaction mixture was quenched with water and extracted with ethyl acetate.
  • the separated organic layer was washed with brine solution, dried over Na 2 S0 4 , filtered and concentrated in vacuo to obtain colourless gummy solid of 5-(5-(2-hydroxyethyl)-2- oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vc (200mg, 86.2%).
  • the crude material was taken for the next step without any purification.
  • Step 4 Synthesis of 2-(3-(5-cyano-6-methylpyridin-3-yl)-2- oxooxazolidin-5-yl)ethyl methanesulfonate, Vd: To a stirred solution 5-(5-(2- hydroxyethyl)-2-oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vc (200mg, 0.80mmol) in dichlorome thane, cooled to 0°C, triethylamine (245mg, 2.42mmol) and mesyl chloride (91mg, 0.80mmol) were added and stirred at 25°C for 2 hours.
  • Vd To a stirred solution 5-(5-(2- hydroxyethyl)-2-oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vc (200mg, 0.80mmol) in dichlorome thane, cooled to 0°C, trieth
  • reaction mixture was quenched with water and extracted with dichlorome thane.
  • the separated organic layer was washed with water, brine solution, dried over Na 2 S0 4 , filtered and concentrated in vacuo to obtain pale brown gummy solid of 2-(3-(5-cyano-6- methylpyridin-3-yl)-2-oxooxazolidin-5-yl)ethyl methanesulfonate, Vd (200mg, 77%).
  • the crude material was taken for the next step without any purification.
  • Step 5 Synthesis of 5-(5-(2-azidoethyl)-2-oxooxazolidin-3-yl)-2- methylnicotinonitrile, Ve: To a stirred solution of 2-(3-(5-cyano-6-methylpyridin-3-yl)- 2-oxooxazolidin-5-yl)ethyl methanesulfonate, Vd (200mg, 0.614mmol) in DMF (4ml), cooled to 0°C, sodium azide (59mg, 0.92mmol) was added and heated at 60°C for 3 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with ethyl acetate.
  • Step 6 Synthesis of 5-(5-(2-aminoethyl)-2-oxooxazolidin-3-yl)-2- methylnicotinonitrile, V: To a stirred solution of 5-(5-(2-azidoethyl)-2-oxooxazolidin-3- yl)-2-methylnicotinonitrile, Ve (50mg, 0.183mmol) in Methanol (5ml), triethylamine (74mg, 0.734mmol) and 1,3-propane dithiol (79mg, 0.734mmol) were added and stirred at 25°C for 12 hours.
  • V To a stirred solution of 5-(5-(2-azidoethyl)-2-oxooxazolidin-3- yl)-2-methylnicotinonitrile, Ve (50mg, 0.183mmol) in Methanol (5ml), triethylamine (74mg, 0.734mmol)
  • reaction mixture was filtered to remove inorganics and concentrated in vacuo to obtain crude pale green colour of gummy liquid of 5-(5-(2-aminoethyl)-2-oxooxazolidin-3-yl)-2-methylnicotinonitrile, V (43mg, 95%).
  • the crude material was taken for the next step without any purification.
  • Step-1 Synthesis of Via: To a solution of chloral hydrate (5.28 g, 31.9 mmol) in deionized water (90 mL), sodium sulphate (53.6 g, 377.9 mmol), 2- bromoaniline (5 g, 29.11 mmol), were added sulphuric acid (20 mL) followed by hydroxylamine hydrochloride (6.66 g, 87.2 mmol) and the whole mixture was heated to 130 C for 30 min. The mixture was cooled to room temperature, poured on ice-water mixture and extracted with ethylacetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. Crude obtained was taken for the next step without any purification.
  • Step-2 Sulphuric acid (50 mL) was added to the crude obtained in stepl and the mixture was heated to 70 °C for lh. After the complete consumption of starting material, reaction mixture was poured over ice, and extracted with ethyl acetate (2 x 200 mL). Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. Crude was purified by silica gel column chromatography by using 20% ethyl acetate in pet ether for elution to get 2.5 g of Via.
  • Step-3 Synthesis of VIb: To a solution of Via (2.5 g, 11.061 mmol) in ethanol (50 mL), hydrazine hydrate (99%, 0.5 mL) was added and the mixture was refluxed under nitrogen atmosphere for 30 min. Formed yellow precipitate was isolated by filtration, and suspended in anhydrous ethanol. Potassium tertiary butoxide (4.03 g, 35.90 mmol) was added to the above suspension. The mixture was refluxed for 2h. After the completion of reaction, mixture was poured over ice-water, acidified with dil. HC1 to pH 2. Mixture was extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude of VIb (0.89 g) which was taken for the next step without any purification.
  • Step-4 Preparation of Vic: To the suspension of VIb (200mg, 0.94 mmol) in water (5 mL) and sodium hydroxide (56mg, 1.41 mmol) was added dimethyl sulphate (178mg, 1.41 mmol) and the mixture was heated at 100 oC for lh. After the consumption of starting material, reaction mixture was cooled to 0 oC, added water and extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. So obtained crude was purified by column chromatography by using 10% ethyl acetate in pet-ether to get pure Vic (180 mg).
  • Step-5 Preparation of VId: To a solution of Vic (180mg, 0.79 mmol) in DMF (4 mL) was added tributylvinyltin (0.26 mL, 2.25 mmol) followed by palladium tetrakis-triphenylphosphine (45 mg, 0.103 mmol) and the mixture was degassed for 30 min by purging with argon. Reaction was heated to 100 °C for 3h. After the completion of reaction, mixture was cooled to 0 °C, added water and extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. So, obtained crude was purified by column chromatography using 10% ethyl acetate in pet-ether to pure VId (100 mg)
  • Step-6 Preparation of VI: Olefin VId ( lOOmg, 0.52 mmol) was dissolved in 4 mL methanol and purged with oxygen for 10 min. Mixture was than cooled to -78 °C and ozone was continuously passed through reaction mixture for 30 min. After the complete consumption of VId, reaction mass was quenched with dimethylsulphide (0.5 mL) and evaporated under reduced pressure to dryness. Residue obtained was purified by column chromatography with eluent 15% ethyl acetate in pet ether to obtain pale yellow solid (30mg).
  • Step-1 Synthesis of Vila: To a solution of chloral hydrate (9.55 g, 57.89 mmol) in deionized water (150 mL), sodium sulphate (100 g, 684.19 mmol), 3-bromo-2- fluoroaniline (10 g, 52.63 mmol), were added sulphuric acid (40 mL) followed by hydroxylamine hydrochloride (10.97 g, 157.89 mmol) and the whole mixture was heated to 130 °C for 30 min. The mixture was cooled to room temperature, powered on ice-water mixture and extracted with ethylacetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. Crude obtained was taken for the next step without any purification.
  • Step-2 Sulphuric acid (100 mL) was added to the crude obtained in step 1 and the mixture was heated to 70 °C for lh. After the complete consumption of starting material, reaction mixture was poured over ice, and extracted with ethyl acetate (2 x 200 mL). Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. Crude was purified by silica gel column chromatography by using 20% ethyl acetate in pet ether for elution to get 7.5 g of Vila.
  • Step-3 Preparation of Vllb: To a solution of Vila (2.8 g, 11.48 mmol) in ethanol (50 mL), hydrazine hydrate (99%, 0.5 mL) was added and the mixture was refluxed under nitrogen atmosphere for 30 min. Formed yellow precipitate was isolated by filteration, and suspended in anhydrous ethanol. Potassium tertiary butoxide (4.03 g, 35.90 mmol) was added to the above suspension. The mixture was refluxed for 2h. After the completion of reaction, mixture was poured over ice-water, acidified with dil. HC1 to pH 2. Mixture was extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude of Vllb (2.0 g) which was taken for the next step without any purification.
  • Step-4 Preparation of VIIc: To the suspension of Vllb (lg, 4.34 mmol) in water (30 mL) and sodium hydroxide (0.397 g, 13.04 mmol) was added dimethyl sulphate (1.4 mL, 13.04 mmol) and the mixture was heated at 100 °C for 2h. After the consumption of starting material, reaction mixture was cooled to 0 °C, added water and extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. So, obtained crude was purified by column chromatography by using 21% ethyl acetate in pet-ether to get pure VIIc (0.500 g)
  • Step-5 Preparation of Vlld: To a solution of VIIc (0.500g, 2.04 mmol) in DMF (10 mL) was added tributylvinyltin (0.65 mL, 2.25 mmol) followed by palladium tetrakis-triphenylphosphine (0.118 g, 0.103 mmol) and the mixture was degassed for 30 min by purging with argon. Reaction was heated to 100 °C for 3h. After the completion of reaction, mixture was cooled to 0 °C, added water and extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. So, obtained crude was purified by column chromatography by using 40% dichloromethane in pet-ether to pure Vlld (0.300 g).
  • Step-6 Preparation of VII: Olefin Vlld (0.300g, 1.57 mmol) was dissolved in 10 mL methanol purged with oxygen for 10 min. Mixture was than cooled to -40 °C and ozone was continuously passed through reaction mixture for 30 min. After the complete consumption of Vlld, reaction mass was quenched with dimethylsulphide (0.5 mL) and evaporated under reduced pressure to dryness. Residue obtained was purified by column chromatography with eluent 40% ethyl acetate in pet ether to obtain white fluffy solid (0.050g).
  • Step-1 Preparation of Villa: To the solution of Vllb (0.200 g, 0.869 mmol) in DMF (5mL) was added sodium hydride (60%, 0.140 g, 3.478 mmol), at 0 °C. After stirring at same temperature for 5 min., methyl iodide (2mL, 3.478 mmol) was introduced slowly, reaction temperature was gradually raised to room temperature and stirred for 2h. After the completion of reaction, whole reaction mixture was poured slowly on ice-cold water and extracted with ethyl acetate. Crude obtained after solvent evaporation was purified by column chromatography using 10% ethyl acetate in hexane to yield 0.180 g of Villa
  • Step-2 Preparation of Vlllb: To a solution of Villa (0.180g, 0.66 mmol) in DMF (5 mL) was added tributylvinyltin (0.2 mL, 0.727 mmol) followed by palladium tetrakis-triphenylphosphine (0.038 g, 0.033 mmol) and the mixture was degassed for 30 min by purging with argon. Reaction was heated to 100 °C for 3h. After the completion of reaction, mixture was cooled to 0 °C, added water and extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. So, obtained crude was purified by column chromatography by using 40% dichloromethane in pet-ether to pure VHIb (0.080 g)
  • Step-3 Preparation of VIII: Olefin VHIb (0.080 g, 3.65 mmol) was dissolved in 5 mL methanol purged with oxygen for 10 min. Mixture was than cooled to - 40 °C and ozone was continuously passed through reaction mixture for 30 min. After the complete consumption of VHIb, reaction mass was quenched with dimethylsulphide (0.5 mL) and evaporated under reduced pressure to dryness. Residue obtained was purified by column chromatography with eluent 40% ethyl acetate in pet ether to obtain white fluffy solid (0.038g).
  • Step-1 Preparation of IXa: To the solution of isatin intermediate Vila (6.6g, 27.04 mmol) in DMF (66 mL), were sequentially added K 2 C0 3 (5.4g, 39.08 mmol) and ethyl iodide (5.8 mL, 72.51 mmol) at room temperature, under nitrogen atmosphere. After stirring at rt for 2h, reaction mixture was cooled in ice -bath and water (100 mL) was added. Whole mixture was extracted with ethyl acetate. Organic layer was washed successively with cold water followed by brine and dried over sodium sulphate before evaporating on rotatory evaporator. Obtained crude residue was suspended in n-hexane and stirred for 20 mins. Solid was isolated by filtration to get the intermediate IXa (4.63 g)
  • Step-4 Preparation of IX: Olefin IXc (0.300g, 1.57 mmol) was dissolved in 10 mL methanol purged with oxygen for 10 min. Mixture was than cooled to -40 °C and ozone was continuously passed through reaction mixture for 30 min. After the complete consumption of, reaction mass was quenched with dimethylsulphide (0.5 mL) and evaporated under reduced pressure to dryness. Residue obtained was purified by column chromatography with eluent 40% ethyl acetate in pet ether to obtain white fluffy solid of aldehyde IX (0.090g).
  • Step 1 Synthesis of 7-allyl-6-fluoro-l-methylindolin-2-one (Xa)
  • Step 2 Synthesis of 2-(6-fluoro-l-methyl-2-oxoindolin-7- vDacetaldehyde (X)
  • Step 5 Synthesis of 4-allyl-5-fluoro-3-methylbenzord1oxazol-2(3H)- one (XHe)
  • Step 6 Synthesis of 2-(5-fluoro-3-methyl-2-oxo-2,3- dihydrobenzo[d1oxazol-4-yl)acetaldehyde (XII)
  • Step 1 Synthesis of 6-fluoro-l-methyl-lH-pyrrolor3,2-blpyridine (Xllla)
  • Step 2 Synthesis of 6-fluoro-l-methyl-lH-pyrrolor3,2-b]pyridine 4- oxide (XHIb) [000213] To a solution of XHIa (6 g, 39.95 mmol) in dry DCM (120 mL), was added mCPBA (13.80 g, 79.91 mmol) at 0°C in portions, then the reaction mixture was stirred at room temperature for 16h. After the consumption of starting material, reaction mixture was cooled to 0°C, added 10% aq NaHC03 (100 mL) and extracted with DCM (2X 100 mL)). The combined organic layers were washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude.
  • Step 3 Synthesis of 7-chloro-6-fluoro-l-methyl-lH-pyrrolor3,2- blpyridine (XIII c)
  • Step 4 Synthesis of 6-fluoro-l-methyl-lH-pyrrolo[3,2-b]pyridine-7- carbonitrile (XIII d)
  • Step 5 Synthesis of 3,3-dibromo-6-fluoro-l-methyl-2-oxo-2,3-dihvdro- lH-pyrrolor3,2-blpyridine-7-carbonitrile (XHIe)
  • Step 6 Synthesis of 6-fluoro-l-methyl-2-oxo-2,3-dihvdro-lH- Pyrrolo[3,2-b1pyridine-7-carbonitrile (XHIf)
  • Example 1 6-(5-(2-(((l-Methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
  • reaction mixture was quenched by water and concentrated in vacuo to remove methanol. Residual mixture was extracted with ethyl acetate. The combined organic layer was washed with water followed by brine, dried over sodium sulphate and concentrated in vacuo.
  • Example 4 Synthesis of 6-(5-(2-(((6-fluoro-l,3,3-trimethyl-2-oxoindolin-7- yl)methyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)- one
  • reaction mixture was filtered through celite and filtrate was concentrated under reduced pressure. Resulting residue was dissolved in methanol, cooled to 0°C, and sodium borohydride (0.012g, 0.00033mol) was added. After the completion of addition, reaction was gradually warmed to room temperature and stirred for 1 hour. After completion of reaction, the reaction mixture was quenched by water and concentrated in vacuo to remove methanol. It was extracted with ethyl acetate and water. The separated organic layer was washed with brine, dried over sodium sulphate and concentrated in vacuo.
  • Example 6 6-(5-(3-(((6-fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
  • Example 7 6-(5-(2-(((l-Ethyl-6-fluoro-2-oxoindolin-7-yl)methyl)amino)ethyl)-2- oxooxazolidin-3- l)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
  • Example 8 6-(5-(3-(((l-Ethyl-6-fluoro-2-oxoindolin-7-yl)methyl)amino)propyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
  • reaction mixture was quenched by water and concentrated in vacuo to remove methanol. Residual mixture was extracted with ethyl acetate. The combined organic layer was washed with water followed by brine, dried over sodium sulphate and concentrated in vacuo.
  • Example 9 Chiral 6-(5-(3-(((6-fluoro-l-methyl-2-oxoindolin-7- yl)methyl)amino)propyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)- one (Enantiomer 1)
  • Example 10 Chiral 6-(5-(3-(((6-fluoro-l-methyl-2-oxoindolin-7- yl)methyl)amino)propyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-
  • Enantiomer 1 (Example 9): 0.750 gm, Chiral HPLC retention time: 5.74 min.
  • Enantiomer 2 (Example 10): 0.650 gm, Chiral HPLC retention time: 7.38 min.
  • Example 11 6-(5-(2-((2-(6-fluoro-l-methyl-2-oxoindolin-7-yl)ethyl)amino)ethyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
  • Example 11 [000237] To a stirred solution of amine II (0.13 g, 0.467 mmol) and aldehyde X (0.125 g, 0.603 mmol) in MeOH (4 mL), glacial acetic acid (0.2 mL) was added and stirred at 25°C for 15 min. The solution was then, cooled to 0°C followed by addition of sodium cyanoborohydride resin (0.125 g) and stirred at 25 °C for 2 h. After completion of reaction, the reaction mixture was filtered to remove the resin and the filtrate was concentrated in vacuo. It was purified by reverse phase preparative HPLC to get Examplell as a pale yellow solid (21 mg).
  • Example 12 6-(5-(3-(((6-fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2- oxooxazolidin-3-yl)-2H-benzo[b][l,4]oxazin-3(4H)-one
  • Example 13 Chiral 6-(5-(2-((2-(6-fluoro-l-methyl-2-oxoindolin-7- yl)ethyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomerl)
  • Example 14 Chiral 6-(5-(2-((2-(6-fluoro-l-methyl-2-oxoindolin-7- yl)ethyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomer2)
  • Enantiomer 1 (Example 13): 0.200 gm, Chiral HPLC retention time: 3.7 min
  • Stepl 6-fluoro-l-methyl-7-(oxiran-2-yl)indolin-2-one( 15a) [000242] To a solution of Vlld (2 g, 10.45 mmol) in DCM (60 mL) and 5% NaHC0 3 aqueous solution (20 mL) was added meta chloroperbenzoic acid (5.41 g 31.38 mmol) at 0°C.The reaction mass was stirred at room temperature for 16 h. After completion of the reaction, added 5% NaHC0 3 aqueous solution (50 mL) and extracted with ethyl acetate (2X 100 mL).
  • Step2 6-(5-(2-((2-(6-fluoro-l-methyl-2-oxoindolin-7-yl)-2-hydroxyethyl) amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
  • Example 16 6-(5-(2-((2-(5-Fluoro-3-methyl-2-oxo-2,3-dihydrobenzo[d]oxazol-4- yl)ethyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
  • Examplel6 was purified by preparative HPLC to afford Examplel6 as an off- white solid (0.026 g, 10 %).
  • Example 17 6-(5-(3-(((6-Fluoro-l-methyl-2-oxo-2,3-dihydro-lH-pyrrolo[3,2- b]pyridin-7-yl)methyl)amino)propyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2- b][l,4]oxazin-3(4H)-one
  • Step2 To a mixture of XIII (0.20g, 1.02 mmol) and aldehyde 17a (0.298 g, 1.02 mmol) in dry MeOH (6 mL) and DCM (6 mL) was added AcOH (0.20 mL) and allowed to stir for 16 h. To this was added Sodium cyanoborohydride resin (0.843 g, 2.04 mmol) and stirred for 15 min. The reaction mixture was filtered and concentrated. Crude was purified by column chromatography (230/400 mesh, 7% DCM in MeOH) to get desire compound Examplel7 as white solid (40 mg, 12 %). LC_MS Calc. for Calc.
  • the compounds of Formula I are of interest due to their potent antibacterial effects.
  • the ability of the invention compounds disclosed herein to achieve an antibacterial effect may be evaluated with regard to their ability to inhibit the growth of bacterial species like Escherichia coli ATCC 25922, Staphylococcus aureus ATCC 29213, Klebsiella pneumoniae ATCC 13883, Acinetobacter baumannii ATCC 19606, Pseudomonas aurigenosa ATCC 27853, Enterococcus faecalis ATCC 29212 and Enterococcus faecalis ATCC 29212 using an assay based on the following Minimum Inhibitory Concentration (MIC) protocol:
  • MIC Minimum Inhibitory Concentration
  • test bacteria are grown in Luria Bertani Broth (HIMEDIA M1245), 25 grams of the powder is dissolved in 1000 ml distilled water and sterilized by autoclaving at 15 lbs pressure (121°C) for 20 minutes. The medium sterility is checked by incubating at 37°C for a period of 48 h.
  • Bacterial cultures that are stored as glycerol stocks at -80°C are sub cultured on LB agar plates to obtain isolated colonies. A single colony of each strain is cultured in LB broth. The cultures are incubated at 37° C, 200 rpm till they reach an optical density (OD at 600nm) of 0.8 to 1. This log phase culture is diluted in LB broth to a cell number of 5-8* 10 5 CFU/mL to be used as inoculum for MIC experiments. Test compounds are dissolved in dimethyl sulfoxide (DMSO) to a stock concentration of 4 mg/ml.
  • DMSO dimethyl sulfoxide
  • a twofold dilution series of this DMSO stock is prepared in a 96 well V bottom microtitre plate from rows A to H. A 3 ⁇ L ⁇ volume of these dilutions are transferred to a 96-well flat bottom microtitre assay plate. Controls to monitor the effects of DMSO and media sterility are included. Each well is inoculated with 150 ⁇ of the above diluted culture. The plates are incubated at 37°C overnight in a humidified incubator. The following morning, the plates are read using a Spectrophotometer at 600 nM wavelength. Minimum Inhibitory Concentration (MIC) is defined as the lowest drug concentration containing well which shows no turbidity. The antibacterial activity (MIC) determined against representative Gram positive (S.
  • the present invention also provides evidence for treating infection caused both Gram positive and Gram negative bacteria through the inhibition of bacterial topoisomerases using E. coli DNA gyrase enzyme.
  • E. coli gyrase supercoiling and its inhibition was assayed using a kit procured from Inpiralis (K0001) and the protocol (PMID: 2172086) was adapted with necessary modifications.
  • the compounds to be tested are incubated for 10 minutes with 2.5 nM of E. coli DNA gyrase in a 30 ⁇ volume reaction and 3.2% DMSO.
  • the reactions are then started with the addition of 60 ng relaxed pBR322 plasmid DNA and continued for 45min at 37 °C.
  • the reaction mixture contains 35 mM Tris.HCl (pH 7.5), 24 mM KC1, 1.8 mM spermidine, 4 mM MgCl 2 , 2 mM DTT, 6.5% (w/v) glycerol, 0.1 mg/mL BSA, and 1 mM ATP.
  • the reaction is then stopped by addition of 0.75 of Proteinase K (20 mg/mL) and 3 ⁇ of 2% SDS and further incubated at 37 °C for 30min.
  • STEB 40 % (w/v) sucrose, 100 mM Tris-HCl pH8, 1 mM EDTA, 0.5 mg/ml Bromophenol Blue
  • the 1 % agarose gels are run for 3 h at 4V/cm in 1 x TAE (40 mM Tris, 20 mM Acetic acid, 1 mM EDTA).
  • IC 50 S are determined by quantifying the supercoiled and relaxed DNA in each of the reactions from a gel image by a densitometric method using the Quantity One Software (Bio-rad).
  • E. coli topoisomerase IV decatenation activity and its inhibition was assayed using a kit procured from Inpiralis (D4002) and the kit protocol was adapted with necessary modifications similar to the gyrase supercoiling assays.
  • the compounds to be tested were incubated for 10 minutes with 5 nM of E. coli topoisomerase IV in a 30 ⁇ volume reaction and 3.2% DMSO.
  • the reactions were started with the addition of 60 ng of kDNA and continued for 40min at 37 °C.
  • the final reaction mixture contains 40 mM Tris.HCl (pH 7.6), 100 mM potassium glutamate, 10 mM magnesium acetate, 10 mM DTT, 1 mM ATP, and 50 ⁇ g/ml albumin.
  • the reactions were stopped by addition of 0.75 ⁇ of Proteinase K (20 mg/mL) and 3 ⁇ of 2% SDS and further incubated at 37 °C for 30min. This was followed by the addition of 4 ⁇ of STEB (40 % (w/v) sucrose, 100 mM Tris-HCl pH8, 1 mM EDTA, 0.5 mg/ml Bromophenol Blue) and the kDNA /minicircles forms were separated by agarose gel electrophoresis.
  • the 1 % agarose gels were run for 3 h at 4V/cm in IX TAE (40 mM Tris, 20 mM Acetic acid, 1 mM EDTA). To visualize the DNA, the gels were stained for 10 min with 0.7 ⁇ g/mL ethidium bromide and excess dye was removed by several washes with water. IC 50 S were determined by quantifying the Kinetoplast DNA band inside the gel well and decatenated minicircles that migrate into the gel in each of the reactions from a gel image by a densitometric method using the Quantity One Software (Bio-rad).
  • PK pharmacokinetics
  • BALB/c mice mice following single intravenous (i.v.) dose of 2 mg/kg and single escalating oral doses of 2, 10, 50 and 100 mg/kg, respectively.
  • This study was performed following all ethical practices as laid down in the guidelines for animal care (Registration number No. 1852/PO/Rc/S/16/CPCSEA). The study was approved by the Institutional Animals Ethics Committee (IAEC) of the test facility.
  • IAEC Institutional Animals Ethics Committee
  • IVPK for IVPK in BALB/c mice, the formulation used was DMSO: PEG400: Ethanol: Water (10:20: 10:60 v/v) and Example 13 administered as solution.
  • Examplel3 For POPK, the formulation used was 0.1 % Tween80 in 0.25% CMC and Examplel3 administered as suspension through an oral gavage. The plasma (mice) samples were analyzed by LC-MS/MS. PK parameters were estimated by non-compartmental analysis in Phoenix WinNonlin 6.4 The results of the per-oral and intravenous PK study of Examplel3 are presented in Table 4 and 5
  • V ss (ml/kg) 13339
  • Example 13 Following a single i.v. dose of 2.0 mg/kg in BALB/c mice, Example 13 showed high systemic clearance [5.7 L/h/kg], a very high volume of distribution at steady state [ 13.3 L/kg], and a moderate ti /2 of 1.7 h.
  • the AUCo- ⁇ ranged between 0.07 to 4.82 ⁇ g.h/ml
  • C max ranged between 0.004 to 0.072 ⁇ g/ml
  • t ranged between 1.4 to 5.6 h.
  • the oral bioavailability was moderate and ranged between 19-27 %.
  • Cmax also increased linearly with dose between 10 and 100 mg/kg [C max /Dose range 0.011 to 0.024].
  • Examplel3 was tested in BALB/c mice thigh infection model following per oral doses of 100 mg/kg once, 50 mg/kg once and twice, 33 mg/kg thrice and 25 mg/kg four times over a 24 hour period post infection. This study was performed following all ethical practices as laid down in the guidelines for animal care (Registration number No. 1852/PO/Rc/S/16/CPCSEA). The study was approved by the Institutional Animals Ethics Committee (IAEC) of the test facility. The formulation used was 0.1% Tween80 in 0.25% CMC and Examplel3 administered as suspension through an oral gavage.
  • IAEC Institutional Animals Ethics Committee
  • mice were infected with [ ⁇ 1 x 10 6 CFU/mouse] E.coli [ATCC 25922] by intramuscular route. Two hours post infection, mice were treated orally with total doses of 100, 50, 50, 33 and 25 mg/kg of Examplel3 as one, two, three and four equally divided doses over a 24 h period. Animals were sacrificed 24 hr post infection and thigh tissues were harvested to enumerate the bacterial CFU count. Thigh muscles were placed in 1 ml of sterile LB broth and homogenized using a homogenizer (Omni Tip (220 volt hand held)).
  • the compounds of the present disclosure show high antibacterial activity against various pathogens including Gram-positive and Gram-negative bacteria through the inhibition of bacterial topoisomerase via a novel mechanism.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Virology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

The present disclosure relates to compounds of Formula I, its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms thereof and pharmaceutical compositions containing them as the active ingredient which can be used as medicaments. The aforementioned substances can also be used in the manufacture of medicaments for treatment, prevention or suppression of diseases, and conditions mediated by microbes. The present disclosure also relates to the synthesis.and characterization of aforementioned substances.

Description

Heterocyclic compounds useful as anti-bacterial agents and method for production
[0001] The present invention relates to the field of medicinal chemistry and more particularly to the development of antimicrobial compounds effective against bacteria, virus, fungi and protozoa including spectrum of Gram-negative and Gram-positive pathogens. The present disclosure relates to compounds of Formula I, its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof and pharmaceutical compositions containing them as the active ingredient. The present disclosure further relates to the synthesis and characterization of aforementioned compounds to exhibit high antimicrobial activity.
[0002] The compounds of the present disclosure are useful as medicaments and their use in the manufacture of medicaments for treatment, prevention or suppression of diseases, and conditions mediated by microbes. The present invention also provides evidence for treating infection caused by microbes.
BACKGROUND
[0003] There is an alarming worldwide concern regarding the rapid evolution of antibiotic resistance, which could result in strains against which there are no effective antibacterial agents. In general, bacterial pathogens may be classified as either Gram- positive or Gram-negative pathogens. Antibiotic compounds with effective activity against both Gram-positive and Gram-negative pathogens are regarded as having a broad spectrum of activity. Current antibacterial drugs used to treat and prevent bacterial infection have been found to have limited effect. Further, there is a continuing need to identify new compounds with potent antibacterial activity with reduced potential for developing resistance, which possess improved efficacy against bacterial infections that resist treatment with currently available antibiotics, or which possess selectivity against target microorganisms.
[0004] From the foregoing, it is clear that compounds used in the state of the art to treat and prevent bacterial infection have been found to have limited effect. Further, there is a continuing need to identify new compounds with improved antibacterial activity, which have less potential for developing resistance, which possess improved effectiveness against bacterial infections that resist treatment with currently available antibiotics, or which possess unexpected selectivity against target microorganisms.
SUMMARY
[0005] The present disclosure is based on the surprising discovery that compounds of Formula I (see below) exhibits advantageous antimicrobial properties. Thus, the present disclosure provides a compound of Formula I
Figure imgf000003_0001
Formula I
or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof,
wherein Ri is selected from the group consisting of hydrogen, Ci-6 alkyl, C2-6 alkenyl, and C3-6 cycloalkyl, each of which is unsubstituted or substituted with 1 to 3 groups independently selected from halogen, hydroxy, amino, oxetane, -OCi_6 alkyl, C3-6 cycloalkylamino, Ci-6 alkylamino, or di(Ci-6 alkyl)amino;
R2 is selected from the group consisting of hydrogen, hydroxyl, and amino;
X is -NH, -NCi-6 alkyl, O, or CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen, halogen, and
Ci-6 alkyl;
Yi is N or CR5; R5 is selected from the group consisting of hydrogen, halogen, cyano, -OCi-6 alkyl, -OQ- 6 haloalkyl, and Ci-6 alkyl;
Y2 is N or CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, halogen, cyano, -OCi-6 alkyl, and - OCi-6 haloalkyl;
Zi is NH when Z2 is C1-6 alkylene; or Zi is C1-6 alkylene when Z2 is NH;
n is 1 or 2;
W is CH2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of a 3-10 membered substituted or unsubstituted heteroaryl ring system which is unsaturated or partially unsaturated optionally having up to three heteroatom independently selected from O, N, NH or S.
[0006] The present disclosure further relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in killing or inhibiting the growth of a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
[0007] The present disclosure further relates to use of a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in killing or inhibiting the growth of a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
[0008] The present disclosure further relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in treating a disease or condition in a patient wherein said disease or condition is caused by a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa. [0009] The present disclosure further relates to use of a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in treating disease or condition in a patient, wherein said disease or condition is caused by a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa. The patient is a typically a mammal, preferably a human.
[00010] The present disclosure further relates to a method of treating a disease or condition in a patent, said method comprising administering to a patient a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein said disease or condition is caused by microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
[00011] The present disclosure relates to a composition comprising a compound of
Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof together with a carrier.
[00012] The present disclosure relates to a pharmaceutical composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, together with a pharmaceutically acceptable carrier, optionally in combination with one or more other pharmaceutical compositions.
[00013] The present disclosure relates to a process of preparation of compound of
Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof.
[00014] The present disclosure relates to a process of preparation of a composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof together with a carrier.
[00015] The present disclosure relates to a process of preparation of pharmaceutical composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, together with a pharmaceutically acceptable carrier, optionally in combination with one or more other pharmaceutical compositions.
[00016] These and other features, aspects, and advantages of the present subject matter will become better understood with reference to the following description. This summary is provided to introduce a selection of concepts in a simplified form. This summary is not intended to identify key features or essential features of the disclosure, nor is it intended to be used to limit the scope of the subject matter.
BRIEF DESCRIPTION OF THE DRAWINGS
[00017] The detailed description is described with reference to the accompanying figures. In the figures, the left-most digit(s) of a reference number identifies the figure in which the reference number first appears. The same numbers are used throughout the drawings to reference like features and components.
[00018] Figure 1 illustrates the MIC90 of Example 14 against clinical strains of four major gram negative bacterial species, in accordance with an embodiment of the present disclosure.
[00019] Figure 2 illustrates the in vivo efficacy of Example 13 in mice thigh infection model, in accordance with an embodiment of the present disclosure.
DETAILED DESCRIPTION
[00020] Those skilled in the art will be aware that the present disclosure is subject to variations and modifications other than those specifically described. It is to be understood that the present disclosure includes all such variations and modifications. The disclosure also includes all such steps, features, compositions and compounds referred to or indicated in this specification, individually or collectively, and any and all combinations of any or more of such steps or features.
Definitions
[00021] For convenience, before further description of the present disclosure, certain terms employed in the specification, and examples are collected here. These definitions should be read in the light of the remainder of the disclosure and understood as by a person of skill in the art. The terms used herein have the meanings recognized and known to those of skill in the art, however, for convenience and completeness, particular terms and their meanings are set forth below.
[00022] The articles "a", "an" and "the" are used to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article.
[00023] The terms "comprise" and "comprising" are used in the inclusive, open sense, meaning that additional elements may be included. Throughout this specification, unless the context requires otherwise the word "comprise", and variations, such as "comprises" and "comprising", will be understood to imply the inclusion of a stated element or step or group of element or steps but not the exclusion of any other element or step or group of element or steps.
[00024] The term "including" is used to mean "including but not limited to".
"Including" and "including but not limited to" are used interchangeably.
[00025] In the structural formulae given herein and throughout the present disclosure, the following terms have been indicated meaning, unless specifically stated otherwise.
[00026] In this specification, the prefix Cx_y as used in terms such as Cx_yalkyl and the like (where x and y are integers) indicates the numerical range of carbon atoms that are present in the group; for example, Ci_6 alkyl includes Ci alkyl (methyl), C2 alkyl (ethyl), C3 alkyl (propyl and isopropyl) and C4 alkyl (butyl, 1 -methylpropyl, 2- methylpropyl, and i-butyl). Unless specifically stated, the bonding atom of a group may be any suitable atom of that group; for example, propyl includes prop-l-yl and prop-2-yl. [00027] The term "alkyl" refers to a monoradical branched or unbranched saturated hydrocarbon chain having from 1 to 10 carbon atoms. This term is exemplified by groups such as n-butyl, iso-butyl, t-butyl, n-hexyl, n-decyl, and the like. The groups may be optionally substituted.
[00028] The term " alkyl ene" refers to a diradical branched or unbranched saturated hydrocarbon chain having from 1 to 6 carbon atoms. This term is exemplified by groups such as methylene, ethylene, propylene, butylene, hexylene, and the like. The groups may be optionally substituted. Representative substituted alkylene groups include hydroxyl substituted alkylenes.
[00029] The term "alkenyl" refers to a monoradical of a branched or unbranched unsaturated hydrocarbon group preferably having from 2, 3, 4, 5, or 6 carbon atoms and having 1, 2, or 3, double bond (vinyl), preferably 1 double bond. The groups may be optionally substituted.
[00030] The term "cycloalkyl" refers to carbocyclic groups of from 3 to 12 carbon atoms having a single cyclic ring or multiple condensed rings which may be partially unsaturated. Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, and the like, or multiple ring structures or carbocyclic groups to which is fused an aryl group, for example indane, and the like. The groups may be optionally substituted.
[00031] The terms "alkoxyl" or "alkoxy" refers to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like. An "ether" is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as can be represented by one of -O-alkyl, - O- alkenyl, -O-alkynyl.
[00032] "Halo" or "Halogen", alone or in combination with any other term means halogens such as chloro (CI), fluoro (F), bromo (Br), and iodo (I).
[00033] The term "heteroaryl" refers to an heteroaromatic carbocyclic group of 3 to 10 carbon atoms having a single ring (e.g. pyridine) or multiple rings (e.g. isoquinoline), or multiple condensed (fused) rings. Preferred heteroaryls include fhiophene, pyrazole, thiazole, pyridine and the like. The groups may be optionally substituted.
[00034] As used herein, the term "substituted" is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described herein above. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this disclosure, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
[00035] The term "effective amount" means an amount of a compound or composition which is sufficient enough to significantly and positively modify the symptoms and/or conditions to be treated (e.g., provide a positive clinical response). The effective amount of an active ingredient for use in a pharmaceutical composition will vary with the particular condition being treated, the severity of the condition, the duration of the treatment, the nature of concurrent therapy, the particular active ingredient(s) being employed, the particular pharmaceutically-acceptable excipient(s)/carrier(s) utilized, the route of administration, and like factors within the knowledge and expertise of the attending physician
[00036] The compounds described herein may contain one or more chiral centers and/or double bonds and therefore, may exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers), regioisomers, enantiomers or diastereomers. Accordingly, the chemical structures depicted herein encompass all possible enantiomers and stereoisomers of the illustrated or identified compounds including the stereoisomerically pure form (e.g., geometrically pure, enantiomerically pure or diastereomerically pure) and enantiomeric and stereoisomeric mixtures. Enantiomeric and stereoisomeric mixtures can be resolved into their component enantiomers or stereoisomers using separation techniques or chiral synthesis techniques well known to the person skilled in the art. The compounds may also exist in several tautomeric forms including the enol form, the keto form and mixtures thereof. Accordingly, the chemical structures depicted herein encompass all possible tautomeric forms of the illustrated or identified compounds.
[00037] The term "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[00038] "Pharmaceutically acceptable salt" embraces salts with a pharmaceutically acceptable acid or base. Pharmaceutically acceptable acids include both inorganic acids, for example hydrochloric, sulphuric, phosphoric, diphosphoric, hydrobromic, hydroiodic and nitric acid and organic acids, for example citric, fumaric, maleic, malic, mandelic, ascorbic, oxalic, succinic, tartaric, benzoic, acetic, methanesulphonic, ethanesulphonic, benzenesulphonic or p-toluenesulphonic acid. Pharmaceutically acceptable bases include alkali metal (e.g. sodium or potassium) and alkali earth metal (e.g. calcium or magnesium) hydroxides and organic bases, for example alkyl amines, arylalkyl amines and heterocyclic amines.
[00039] The compounds discussed herein in many instances may have been named and/or checked with ACD/Name by ACD/Labs® and/or Electronic Lab Notebook by CambridgeSoft®.
[00040] The term "polymorphs" refers to crystal forms of the same molecule, and different polymorphs may have different physical properties such as, for example, melting temperatures, heats of fusion, solubilities, dissolution rates and/or vibrational spectra as a result of the arrangement or conformation of the molecules in the crystal lattice.
[00041] The term "solvate", as used herein, refers to a crystal form of a substance which contains solvent. [00042] The term "hydrate" refers to a solvate wherein the solvent is water.
[00043] The term "drug sensitive bacterium" as used herein is a bacterium which is not able to survive exposure to at least one drug.
[00044] The resent disclosure relates to a compound of Formula I
Figure imgf000011_0001
Formula I
or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof,
wherein
Ri is selected from the group consisting of hydrogen, Ci-6 alkyl, C2-6 alkenyl, and C3-C6 cycloalkyl, each of which is unsubstituted or substituted with 1 to 3 groups independently selected from halogen, hydroxy, amino, oxetane, -OCi_6 alkyl, C3-6 cycloalkylamino, Ci-6 alkylamino, or di(Ci-6 alkyl)amino;
R2 is selected from the group consisting of hydrogen, hydroxyl, and amino;
X is -NH, -NCi-6 alkyl, O, or CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen, halogen, and
Ci_6 alkyl;
Yi is N or CR5;
R5 is selected from the group consisting of hydrogen, halogen, cyano, -OCi-6 alkyl, -OCi- 6 haloalkyl, and Ci-6 alkyl;
Y2 is N or CH;
Y3 is N or CR6; R6 is selected from the group consisting of hydrogen, halogen, cyano, -OCi-6 alkyl, Ci-6 alkyl, and -OCi-6 haloalkyl;
Zi is NH when Z2 is Ci_6 alkylene; or Zi is Ci_6 alkylene when Z2 is NH;
n is 1 or 2;
W is CH2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000012_0001
Formula I
or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof,
wherein Ri is selected from the group consisting of hydrogen, Ci-6 alkyl, C2-6 alkenyl, and C3-6 cycloalkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, hydroxy, amino, oxetane, -OCi alkyl, C3-6 cycloalkylamino, Q-2 alkylamino, or di(Ci-2 alkyl)amino;
R2 is selected from the group consisting of hydrogen, hydroxyl, and amino;
X is -NH, -NCi-6 alkyl, O, or CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen, halogen, and
Ci_6 alkyl;
Yi is N or CR5;
R5 is selected from the group consisting of hydrogen, halogen, cyano, - -OCi-6 alkyl, - OCi-6 haloalkyl, and Ci-6 alkyl;
Y2 is N or CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCi-6 alkyl, -Ci-6 alkyl, and -OCi-6 haloalkyl;
Zi is NH when Z2 is C1-6 alkylene; or Zi is C1-6 alkylene when Z2 is NH;
n is 1 or 2;
W is CH2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
Figure imgf000013_0001
[00046] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Zi is NH when Z2 is C1-6 alkylene; or Zi is Ci-6 alkylene or C1-6 hydroxy alkylene when Z2 is NH.
[00047] According to an embodiment, the present disclosure relates to a compound of Formula I
Figure imgf000014_0001
Formula I
or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof,
wherein
Ri is selected from the group consisting of hydrogen, Ci-6 alkyl, C2-6 alkenyl, and C3-6 cycloalkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, hydroxy, amino, oxetane, -OCi alkyl, C3-6 cycloalkylamino, d-2 alkylamino, or di(Ci-2 alkyl)amino;
R2 is selected from the group consisting of hydrogen, hydroxyl, and amino;
X is -NH, -NCi alkyl, O, or CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen, fluorine, and Ci alkyl;
Yi is N or CR5;
R5 is selected from the group consisting of hydrogen, fluorine, chlorine, cyano, -OCH3, - OCF3, OCHF2, and Ci alkyl;
Y2 is N or CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH3, - OCHF2, and -OCF3;
Zi is NH when Z2 is CH2; or Zi is CH2, CH2CH2 or CH(OH)CH2when Z2 is NH;
n is 1 or 2
W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000015_0001
According to an embodiment, the present disclosure relates to a compound
Figure imgf000015_0002
or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof,
wherein
Ri is selected from the group consisting of hydrogen, Ci-6 alkyl, C2-6 alkenyl, and C3-6 cycloalkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from halogen, hydroxy, amino, oxetane, -OCi_6 alkyl, C3-6 cycloalkylamino, Ci-6 alkylamino, or di(Ci-6 alkyl)amino;
R2 is selected from the group consisting of hydrogen, hydroxyl, and amino;
X is -NH, -NCi-6 alkyl, O, or CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen, halogen, and
Ci_6 alkyl;
Yi is N or CR5;
R5 is selected from the group consisting of hydrogen, halogen, cyano, -OCi_6 alkyl, -OCi_ 6 haloalkyl, and Ci_6 alkyl;
Y2 is N or CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, halogen, cyano, Ci_6 alkyl, -OCi_6 alkyl, and -OCi_6 haloalkyl;
Zi is NH when Z2 is Ci_6 alkylene; or Zi is Ci_6 alkylene when Z2 is NH;
n is 1 or 2;
W is CH2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000017_0001
Figure imgf000017_0002
Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof,
wherein
Ri is selected from the group consisting of hydrogen, Ci-6 alkyl, C2-6 alkenyl, and C3-C6 cycloalkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, hydroxy, amino, oxetane, -OCi alkyl, C3-C6 cycloalkylamino, d-2 alkylamino, or di(Ci-2 alkyl)amino;
R2 is selected from the group consisting of hydrogen, hydroxyl, and amino; X is -NH, -NCi-6 alkyl, O, or CR3R4;
R and R4 are independently selected from the group consisting of hydrogen, fluorine, and
Ci alkyl;
Yi is N or CR5;
R5 is selected from the group consisting of hydrogen, fluorine, chlorine, cyano, -OCH3, - OCF3, OCHF2, and Ci alkyl;
Y2 is N or CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH3, and -OCF3; Zi is NH when Z2 is CH2; or Zi is CH2, CH2CH2 or CH(OH)CH2 when Z2 is NH;
n is 1 or 2;
W is CH2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000018_0001
According to an embodiment, the present disclosure relates to a compound
Figure imgf000019_0001
Formula I
or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof,
wherein
Ri is selected from the group consisting of hydrogen, C1-4 alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, amino, hydroxy, oxetane, C3-C6 cycloalkylamino, C1-2 alkylamino, or di(Ci-2 alkyl) amino;
R2 is selected from the group consisting of hydrogen, amino, and hydroxyl;
X is O, -NCialkyl, or CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen, fluorine, and Ci alkyl;
Yi is N or CR5;
R5 is selected from the group consisting of hydrogen, fluorine, chlorine, cyano, -OCi alkyl, -OCF3, OCHF2, and Ci alkyl;
Y2 is N or CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCi alkyl, and - OCF3;
Zi is NH when Z2 is CH2; or Zx is CH2, CH2CH2 or CH(OH)CH2 when Z2 is NH;
n is 1 or 2; W is CH2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of:
Figure imgf000020_0001
Formula I
or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein
Ri is selected from the group consisting of hydrogen, Q-4 alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, hydroxy, amino, oxetane, C3-6 cycloalkylamino, d-2 alkylamino, or di(Ci-2 alkyl) amino; R2 is selected from the group consisting of hydrogen, amino, and hydroxyl;
R3 and R4 are independently selected from the group consisting of hydrogen, and Ci alkyl;
Yi is N or CR5;
R5 is selected from the group consisting of hydrogen, fluorine, chlorine, cyano, -OCH3, - CF3, OCHF2, and Ci alkyl;
Y2 is N or CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH3, -OCHF2, and -OCF3;
Zi is NH when Z2 is CH2; or Zx is CH2 , CH2CH2 or CH(OH)CH2 when Z2 is NH;
n is 1 or 2;
W is CH2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000021_0001
[00052] According to an embodiment, the present disclosure relates to a compound of Formula I
Figure imgf000022_0001
Formula I
or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein
Ri isCi alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, hydroxy, amino, oxetane, Ci-2 alkylamino, or di(Ci-2 alkyl) amino;
R2 is hydrogen;
X is CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen, and Ci alkyl;
Yi is N or CR5;
R5 is hydrogen, Ci alkyl, or cyano;
Y2 is CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH3, and -OCF3; Zi is NH when Z2 is CH2; or Zx is CH2, CH2CH2 or CH(OH)CH2 when Z2 is NH;
n is 1 or 2;
W is CH2 wherein dotted line (— ) represents no bond; W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000023_0001
[00053] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein
Ri is C2 alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, amino, hydroxyl, d-2 alkylamino, or di(Ci-2 alkyl) amino;
R2 is hydrogen;
X is O or CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen, and Ci alkyl;
Yi is N or CR5;
R5 is selected from the group consisting of hydrogen, Ci alkyl, and cyano;
Y2 is CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH3, OCHF2 and-OCF3;
Zi is NH when Z2 is CH2; or Zi is CH2, CH2CH2 or CH(OH)CH2 when Z2 is NH;
n is 1 or 2;
W is O when dotted line (— ) represents either a bond or no bond; ing A is selected from the group consisting of
Figure imgf000024_0001
[00054] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein
Ri is Ci alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, amino, hydroxyl, d-2 alkylamino, or di(Ci-2 alkyl) amino;
R2 is hydrogen;
X is O;
R3 and R4 are independently selected from the group consisting of hydrogen, and Q alkyl;
Yi is N or CR5;
R5 is selected from the group consisting of hydrogen, Ci alkyl, and cyano;
Y2 is CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH3, OCHF2 and-OCF3;
Zi is NH when Z2 is CH2; or Zi is CH2, CH2CH2 or CH(OH)CH2 when Z2 is NH;
n is 1 or 2; W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000025_0001
[00055] According to an embodiment, the present disclosure relates to a compound of Formula I
Figure imgf000025_0002
Formula I
or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof,
wherein
Ri is Ci_4 alkyl;
R2 is hydrogen;
Figure imgf000025_0003
R3 and R4 are independently selected from the group consisting of hydrogen, and Q alkyl;
Yi is N or CR5;
R5 is hydrogen;
Y2 is CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH3, and -OCF3; Zi is NH when Z2 is CH2; or Zx is CH2, CH2CH2 or CH(OH)CH2 when Z2 is NH;
n is 1 or 2;
W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the roup consisting of
Figure imgf000026_0001
[00056] According to an embodiment, the present disclosure relates to a compound of Formula I
Figure imgf000026_0002
Formula I
or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Ri is selected from the group consisting of methyl, ethyl, isopropyl;
R2 is hydrogen;
X is CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen and Ci alkyl; Yi is N or CR5;
R5 is hydrogen, CH3, CN;
Y2 is CH;
Y3 is CR6;
R6 is hydrogen, fluorine, cyano, -OCH3, and -OCF3;
Zi is NH when Z2 is CH2; or Zi is CH2CH2 or CH(OH)CH2 when Z2 is NH;
n is 1 or 2;
W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000027_0001
[00057] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein
Ri is selected from the group consisting of Ci-3 alkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, -OCi alkyl, amino, hydroxy, Ci-2 alkylamino, or di(Ci-2 alkyl)amino;
R2 is hydrogen;
X is CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen and Ci alkyl; Yi is N or CR5; R5 is selected from the group consisting of hydrogen, CH3, and CN;
Y2 is CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH3, and -OCF3; Zi is NH when Z2 is CH2; or Zi is CH2, CH2CH2 or CH(OH)CH2 when Z2 is NH;
n is 1 or 2;
W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000028_0001
[00058] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof,
wherein
Ri is selected from the group consisting of hydrogen, Ci-6 alkyl, C2-6 alkenyl, and C3-C6 cycloalkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, hydroxy, amino, oxetane, -OCi alkyl, C3-C6 cycloalkylamino, Q-2 alkylamino, or di(Ci-2 alkyl)amino;
R2 is selected from the group consisting of hydrogen, hydroxyl, and amino;
X is -NH, -NCi-6 alkyl, O, or CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen, halogen, and
Ci_6 alkyl;
Yi is N or CR5;
R5 is selected from the group consisting of hydrogen, halogen, cyano, -OCi-6 alkyl, -OCi- 6 haloalkyl, and Ci-6 alkyl; Y2 is N or CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCi-6 alkyl alkyl, and -OCi-6 haloalkyl;
Zi is NH when Z2 is C1-6 alkylene; or Zi is Ci-6alkylene when Z2 is NH;
n is 1 or 2
W is CH2 wherein dotted line (— ) represents no bond; W is O when dotted line (— represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000029_0001
[00059] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein X is NH, -N(CH3), O, CH2, CH-F, CH-CH3, CF2 or C(CH3)2.
[00060] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein X is O, -CH, C(F)2, or C-(CH3)2.
[00061] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein X is -CH2, or C- (CH3)2. [00062] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein R2 is selected from H, hydroxy or amino.
[00063] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein R2 is H.
[00064] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Zi and Z2 is independently selected from -NH-, -CH2, or CH2CH2.
[00065] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Y2 is N or CH.
[00066] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Y2 is CH.
[00067] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Yi is N, CH, CF, CC1, C-CN, C- OCH3, C-OCF3, C-OCHF2, or C-CH3.
[00068] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Yi is CH.
[00069] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Y3 is N, -CH, -CF, -C(CN), - C(OCH3), -C(OCF3), or -C(OCHF2).
[00070] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein Y3 is -CH, or -CF.
[00071] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein n is 1 or 2
[00072] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, which is selected from a group consisting of: 6-(5-(2-((( 1 -Methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3 ,2-b] [ 1 ,4]oxazin-3(4H)-one,
6-(5-(2-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-pyrido [3 ,2-b] [ 1 ,4]oxazin-3 (4H)-one,
6-(5-(2-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-benzo[b][l,4]oxazin-3(4H)-one,
6-(5-(2-(((6-Fluoro-l,3,3-trimethyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one,
5-(5-(2-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2-methylnicotinonitrile, 6-(5-(3-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2-oxooxazolidin- 3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one,
6-(5-(2-(((l-Ethyl-6-fluoro-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-pyrido [3 ,2-b] [ 1 ,4]oxazin-3 (4H)-one,
6-(5-(3-(((l-Ethyl-6-fluoro-2-oxoindolin-7-yl)methyl)amino)propyl)-2-oxooxazolidin-3- yl)-2H-pyrido [3 ,2-b] [ 1 ,4]oxazin-3 (4H)-one,
6-(5-(3-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2-oxooxazolidin- 3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomerl),
6-(5-(3-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2-oxooxazolidin- 3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomer2),
6-(5-(2-((2-(6-Fluoro-l-methyl-2-oxoindolin-7-yl)ethyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-pyrido [3 ,2-b] [ 1 ,4]oxazin-3 (4H)-one,
6-(5-(3-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2-oxooxazolidin- 3-yl)-2H-benzo[b] [ 1 ,4]oxazin-3(4H)-one,
6-(5-(2-((2-(6-Fluoro-l-methyl-2-oxoindolin-7-yl)ethyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomerl),
6-(5-(2-((2-(6-Fluoro-l-methyl-2-oxoindolin-7-yl)ethyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomer2),
6-(5-(2-((2-(6-Fluoro- 1 -methyl-2-oxoindolin-7-yl)-2-hydroxyethyl) amino)ethyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one,
6-(5-(2-((2-(5-Fluoro-3-methyl-2-oxo-2,3-dihydrobenzo[d]oxazol-4- yl)ethyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, and
6-(5-(3-(((6-Fluoro-l-methyl-2-oxo-2,3-dihydro-lH-pyrrolo[3,2-b]pyridin-7- yl)methyl)amino)propyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one.
[00073] According to an embodiment, the present disclosure relates to a process of preparation of compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof. [00074] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in killing or inhibiting the growth of a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
[00075] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in killing or inhibiting the growth of Gram-positive and Gram-negative bacteria.
[00076] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in killing or inhibiting the growth of drug sensitive and drug resistance bacterium selected from a group consisting of Escherichia coli, Salmonella typhi, Salmonella paratyphi, Salmonella typhimurium, Salmonella enteritidis, Shigella dysenteriae, Shigella flexneri, Shigella boydii, Shigella sonnei, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pneumoniae, Streptococcus pygenes, Stenotrophomonas maltophilia, Haemophilus influenza, Klebsiella pneumoniae, Legionella pneumophila, Mycoplasma pneumoniae, Acinetobacter baumannii, Acinetobacter haemolyticus, Acinetobacter junii, Acinetobacter Iwoffi, Burkholderia cepacia, Chlamydophila pneumoniae, Clostridium difficili, Pseudomonas aeruginosa, Enterobacter aerogenes, Enterobacter cloacae, Moraxella catarrhalis, Enterococcus faecium, Enterococcus faecalis, Enterococcus faeciu, Neisseria gonorrhoeae, Neisseria meningitides, or any combinations thereof.
[00077] According to an embodiment, the present disclosure relates to use of a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in killing or inhibiting the growth of a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
[00078] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in treating a disease or condition in a patient wherein said disease or condition is caused by a microorganism selected from the group consisting of Gran negative and Gram positive pathogens.
[00079] According to an embodiment, the present disclosure relates to use of a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in treating disease or condition in a patient, wherein said disease or condition is caused by a microorganism selected from the group consisting of Gran negative and Gram positive pathogens. The patient is a typically a mammal, preferably a human.
[00080] According to an embodiment, the present disclosure relates to a method of treating a disease or condition in a patent, said method comprising administering to a patient a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein said disease or condition is caused by microorganism selected from the group consisting of Gram negative and Gram positive pathogens.
[00081] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use as a medicament.
[00082] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in the preparation of medicaments for inhibiting microbial growth.
[00083] According to an embodiment, the present disclosure relates to a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in the preparation of medicaments for inhibiting bacterial growth.
[00084] According to an embodiment, the present disclosure relates to medicaments that include a compound of Formula I, or an addition salt of the compound of formula I with a pharmaceutically acceptable acid or base. These medicaments find their use in therapeutics, especially in the treatment of bacterial infection caused by both drug sensitive and drug resistance bacterium including quinolone resistance belonging to Gram positive and Gram negative species; especially of those caused by Escherichia coli, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Clostridium difficili, Neisseria gonorrhoeae, and Enterococcus faecalis .
[00085] According to an embodiment, the present disclosure relates to the use of a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in the manufacture of a medicament for the treatment of an infection caused by bacterial species in a warmblooded animal, such as man.
[00086] According to an embodiment, the present disclosure relates to the use of a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in the manufacture of a medicament for the production of an antibacterial effect in a warm-blooded animal such as man.
[00087] According to an embodiment, the present disclosure relates to a method for treating bacterial infections caused by bacterial species in a warm-blooded animal such as man, said method including administering to said animal an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.
[00088] According to an embodiment, the present disclosure relates to a method for producing an antibacterial effect in a warm-blooded animal such as man, said method including administering to said animal an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.
[00089] According to an embodiment, the present disclosure relates to a compound of Formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment and/or prophylaxis of bacterial infections in a warm-blooded animal, such as man.
[00090] According to an embodiment, the present disclosure relates to a compound of Formula I, or a pharmaceutically acceptable salt thereof, for the therapeutic and prophylactic treatment of mammals including humans, in particular in treating bacterial infections caused by bacterial species, is normally formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition.
[00091] According to an embodiment, the present disclosure relates to a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, and at least one pharmaceutically acceptable carrier, diluent, or excipient.
[00092] According to an embodiment, the present disclosure relates to the use of a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in the manufacture of a medicament for the treatment of a bacterial infection caused by bacterial species in a warm-blooded animal such as man.
[00093] According to an embodiment, the present disclosure relates to the use of a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in the manufacture of a medicament for the production of an antibacterial effect in a warm-blooded animal such as man.
[00094] According to an embodiment, the present disclosure relates to a pharmaceutical composition comprising a compound of formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof together with a pharmaceutically acceptable carrier, and in combination with at least one antibiotic.
[00095] According to an embodiment, the present disclosure relates to a method for treating infection caused by bacterial species in a warm-blooded animal such as man, said method including administering to said animal an effective amount of a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof.
[00096] According to an embodiment, the present disclosure relates to a method for producing an antibacterial effect in a warm-blooded animal such as man, said method including administering to said animal an effective amount of a pharmaceutical composition including a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof.
[00097] According to an embodiment, the present disclosure relates to a pharmaceutical composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, together with a pharmaceutically acceptable carrier, optionally in combination with one or more other pharmaceutical compositions.
[00098] According to an embodiment, the present disclosure relates to a pharmaceutical composition comprising a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, and a pharmaceutically acceptable diluent or carrier.
[00099] According to an embodiment, the present disclosure relates to a composition comprising a compound of Formula I, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, and a carrier.
[000100] The language "pharmaceutically acceptable" includes compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[000101] The compounds of Formula I may form stable pharmaceutically acceptable acid or base salts, and in such cases administration of a compound as a salt may be appropriate. Examples of acid addition salts include acetate, adipate, ascorbate, benzoate, benzenesulfonate, bicarbonate, bisulfate, butyrate, camphorate, camphorsulfonate, choline, citrate, cyclohexyl sulfamate, diethylenediamine, ethanesulfonate, fumarate, glutamate, glycolate, hemisulfate, 2-hydroxyethylsulfonate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, hydroxymaleate, lactate, malate, maleate, methanesulfonate, meglumine, 2-naphthalenesulfonate, nitrate, oxalate, pamoate, persulfate, phenylacetate, phosphate, diphosphate, picrate, pivalate, propionate, quinate, salicylate, stearate, succinate, sulfamate, sulfanilate, sulfate, tartrate, tosylate (p-toluenesulfonate), trifluoroacetate, and undecanoate. Examples of base salts include ammonium salts; alkali metal salts such as sodium, lithium and potassium salts; alkaline earth metal salts such as aluminum, calcium and magnesium salts; salts with organic bases such as dicyclohexylamine salts and N10 methyl-D-glucamine; and salts with amino acids such as arginine, lysine, ornithine, and so forth. Also, basic nitrogen- containing groups may be quaternized with such agents as: lower alkyl halides, such as methyl, ethyl, propyl, and butyl halides; dialkyl sulfates such as dimethyl, diethyl, dibutyl; diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl halides; arylalkyl halides such as benzyl bromide and others. Non-toxic physiologically acceptable salts are preferred, although other salts may be useful, such as in isolating or purifying the product.
[000102] The salts may be formed by conventional means, such as by reacting the free base form of the product with one or more equivalents of the appropriate acid in a solvent or medium in which the salt is insoluble, or in a solvent such as water, which is removed in vacuo or by freeze drying or by exchanging the anions of an existing salt for another anion ona suitable ion-exchange resin.
[000103] The compositions of the disclosure may be in a form suitable for oral use (for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions), for administration by inhalation (for example as a finely divided powder or a liquid aerosol), for administration by insufflation (for example as a finely divided powder) or for parenteral administration (for example as a sterile aqueous or oily solution for intravenous, subcutaneous, intramuscular or intramuscular dosing or as a suppository for rectal dosing).
[000104] The present disclosure relates to a process of preparation of a composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof together with a carrier.
[000105] The present disclosure relates to a process of preparation of pharmaceutical composition comprising a compound of Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, together with a pharmaceutically acceptable carrier, optionally in combination with one or more other pharmaceutical compositions. [000106] The compositions of the present disclosure may be obtained by conventional procedures using conventional pharmaceutical excipients well known in the art. Thus, compositions intended for oral use may contain, for example, one or more colouring, sweetening, flavouring and/or preservative agents
[000107] Suitable pharmaceutically acceptable excipients for a tablet formulation include, for example, inert diluents such as lactose, sodium carbonate, calcium phosphate or calcium carbonate; granulating and disintegrating agents such as corn starch or algenic acid; binding agents such as starch; lubricating agents such as magnesium stearate, stearic acid or talc; preservative agents such as ethyl or propyl j?-hydroxybenzoate; and anti- oxidants, such as ascorbic acid. Tablet formulations may be uncoated or coated either to modify their disintegration and the subsequent absorption of the active ingredient within the gastrointestinal tract, or to improve their stability and/or appearance, in either case, using conventional coating agents or procedures well known in the art.
[000108] Compositions for oral use may be in the form of hard gelatin capsules in which the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules in which the active ingredient is mixed with water or oil such as peanut oil, liquid paraffin, or olive oil.
[000109] Aqueous suspensions generally contain the active ingredient in finely powdered form or in the form of nano or micronized particles together with one or more suspending agents, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents such as lecithin or condensation products of an alkylene oxide with fatty acids (for example polyoxethylene stearate), or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives such as ethyl or propyl p-hydroxybenzoate; anti-oxidants such as ascorbic acid); coloring agents; flavoring agents; and/or sweetening agents such as sucrose, saccharine or aspartame.
[000110] Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such as arachis oil, olive oil, sesame oil or coconut oil or in a mineral oil such as liquid paraffin. The oily suspensions may also contain a thickening agent such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set out above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
[000111] Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water generally contain the active ingredient together with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients such as sweetening, flavoring and coloring agents, may also be present.
[000112] The pharmaceutical compositions of the disclosure may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, or a mineral oil, such as for example liquid paraffin or a mixture of any of these. Suitable emulsifying agents may be, for example, naturally-occurring gums such as gum acacia or gum tragacanth, naturally-occurring phosphatides such as soya bean, lecithin, an esters or partial esters derived from fatty acids and hexitol anhydrides (for example sorbitan monooleate) and condensation products of the said partial esters with ethylene oxide such as polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening, flavoring and preservative agents. [000113] Syrups and elixirs may be formulated with sweetening agents such as glycerol, propylene glycol, sorbitol, aspartame or sucrose, and may also contain a demulcent, preservative, flavoring and/or coloring agent.
[000114] The pharmaceutical compositions may also be in the form of a sterile injectable aqueous or oily suspension, which may be formulated according to known procedures using one or more of the appropriate dispersing or wetting agents and suspending agents, which have been mentioned above. A sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example a solution in 1,3-butanediol.
[000115] Compositions for administration by inhalation may be in the form of a conventional pressurized aerosol arranged to dispense the active ingredient either as an aerosol containing finely divided solid or liquid droplets. Conventional aerosol propellants such as volatile fluorinated hydrocarbons or hydrocarbons may be used and the aerosol device is conveniently arranged to dispense a metered quantity of active ingredient.
[000116] Compositions for administration may also be formulated as a liposome preparation. The liposome preparation can comprise liposomes which penetrate the cells of interest or stratum corneum, and fuse with the cell membrane, resulting in delivery of the contents of the liposome into the cell. Other suitable formulations can employ niosomes. Niosomes are lipid vesicles similar to liposomes, with membrane consisting largely of nonoinic lipids, some forms of which are effective for transporting compounds across the stratum corneum.
[000117] Compositions for administration may also be formulated as a depot preparation, which may be administered by implantation or by intramuscular injection. The compositions may be formulated with suitable polymeric or hydrophobic material (as an emulsion in acceptable oil), ion exchange resins, or sparingly soluble derivatives.
[000118] The compound of the present disclosure can also be administered in sustained release forms or from sustained release drug delivery systems.
[000119] For further information on formulation, drug delivery as well as processing techniques the reader is referred to Remington's Pharmaceutical Sciences (21 s Edition, 2005, University of the sciences in Philadelphia, Lippincott William & Wilkins)
[000120] The amount of active ingredient that is combined with one or more excipients to produce a single dosage form will necessarily vary depending upon the host treated and the particular route of administration. For example, a formulation intended for oral administration to humans will generally contain, for example, from 0.5 mg to 4 g of active agent compounded with an appropriate and convenient amount of excipients which may vary from about 5 to about 98 percent by weight of the total composition. Dosage unit forms will generally contain about 1 mg to about 500 mg of an active ingredient. For further information on Routes of Administration and Dosage Regimes the reader is referred to Chapter 25.3 in Volume 5 of Comprehensive Medicinal Chemistry (Corwin Hansch; Chairman of Editorial Board), Pergamon Press 1990 and Remington's Pharmaceutical Sciences (21st Edition, 2005, University of the sciences in Philadelphia, Lippincott William & Wilkins).
[000121] As stated above the size of the dose required for the therapeutic or prophylactic treatment of a particular disease state will necessarily be varied depending on the host treated, the route of administration and the severity of the illness being treated. Preferably a daily dose in the range of 1 -25 mg kg is employed. Accordingly, the optimum dosage may be determined by the practitioner who is treating any particular patient.
[000122] In any of the pharmaceutical compositions, processes, methods, uses, medicaments, and manufacturing features mentioned herein, any of the alternate aspects of the compounds of the disclosure described herein also apply.
[000123] The compounds disclosed herein may be applied as a sole therapy or may involve, in addition to a compound of the disclosure, one or more other substances and/or treatments. Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate administration of the individual components of the treatment. Where the administration is sequential or separate, the delay in administering the second component should not be such as to lose the beneficial effect of the combination. Suitable classes and substances may be selected from one or more of the following: i) other antibacterial agents for example macrolides e.g. erythromycin, azithromycin or clarithromycin; quinolones e.g. ciprofloxacin or levofloxacin; β lactams e.g. penicillins e.g. amoxicillin or piperacillin; cephalosporins e.g. ceftriaxone or ceftazidime; carbapenems, e.g. meropenem or imipenem etc; aminoglycosides e.g. gentamicin or tobramycin; or oxazolidinones; and/or ii) anti-infective agents for example, an antifungal triazole e.g. or amphotericin; and/or iii) biological protein therapeutics for example antibodies, cytokines, bactericidal/ permeability increasing protein (BPI) products; and/or iv) one or more antibacterial agents useful in the treatment of Mycobacterium tuberculosis such as one or more of rifampicin, isoniazid, pyrizinamide, ethambutol, quinolones e.g. moxifloxacin or gatifloxacin, streptomycin and/or v) efflux pump inhibitors.
[000124] According to an embodiment, the present disclosure relates to a compound of the Formula I, or a pharmaceutically acceptable salt thereof and a chemotherapeutic agent selected from: i) one or more additional antibacterial agents; and/or ii) one or more anti-infective agents; and/or iii) biological protein therapeutics for example antibodies, cytokines, bactericidal/permeability increasing protein (BPI) products; iv) one or more antibacterial agents useful in the treatment of pulmonary tuberculosis, extra-pulmonary tuberculosis, avium infections, buruli ulcers and/or v) one or more efflux pump inhibitors.
[000125] If not commercially available, the necessary starting materials for the procedures such as those described herein may be made by procedures which are selected from standard organic chemical techniques, techniques which are analogous to the synthesis of known, structurally similar compounds, or techniques which are analogous to the described procedure or the procedures described in the Examples.
[000126] It is noted that many of the starting materials for synthetic methods as described herein are commercially available and/or widely reported in the scientific literature, or could be made from commercially available compounds using adaptations of processes reported in the scientific literature. The reader is further referred to Advanced Organic Chemistry, 5th Edition, by Jerry March and Michael Smith, published by John Wiley & Sons 2001, for general guidance on reaction conditions and reagents.
[000127] It will also be appreciated that in some of the reactions mentioned herein it may be necessary/desirable to protect any sensitive groups in compounds. The instances where protection is necessary or desirable are known to those skilled in the art, as are suitable methods for such protection. Conventional protecting groups may be used in accordance with standard practice (for illustration see T.W. Greene, Protective Groups in Organic Synthesis, published by John Wiley and Sons, 1991) and as described hereinabove.
Abbreviations
[000128] The following abbreviations are employed in the examples and elsewhere herein:
TLC- thin layer chromatography;
HPLC -high pressure liquid chromatography;
MPLC - medium pressure liquid chromatography;
NMR - nuclear magnetic resonance spectroscopy;
DMSO - dimethylsulfoxide;
CDCI3 - deuterated chloroform;
MeOD -deuterated methanol, i.e. D3COD;
MS - mass spectroscopy; ESP (or ES) - electrospray; EI - electron impact; APCI - atmospheric pressure chemical ionization;
THF - tetrahydrofuran;
DCM - dichlorome thane;
MeOH - methanol;
DMF -dimethylformamide;
EtOAc - ethyl acetate;
LC/MS - liquid chromatography/mass spectrometry;
h - hour(s); min is minute(s);
d - day(s); MTBD - N-methyl-l,5,7-triazabicyclo[4.4.0]dec-5-ene;
TFA - trifluoroacetic acid; v/v - ratio of volume/volume;
Boc denotes t-butoxycarbonyl;
Cbz denotes benzyloxycarbonyl;
Bz denotes benzoyl;
atm denotes atmospheric pressure;
rt denotes room temperature;
mg denotes milligram; g denotes gram;
μL· denotes microliter;
mL denotes milliliter;
L denotes liter;
μΜ denotes micromolar;
mM denotes millimolar; M denotes molar;
N denotes normal; and
nm denotes nanometer.
EXAMPLES
[000129] The following examples provide the details about the synthesis, activities and applications of the compounds of the present disclosure. It should be understood the following is representative only, and that the invention is not limited by the details set forth in these examples.
Materials and methods:
[000130] Evaporations were carried out by rotary evaporation in vacuo and work up procedures were carried out after removal of residual solids by filtration; temperatures are quoted as °C; operations were carried out at room temperature, that is typically in the range 18 to 26 °C and without the exclusion of air unless otherwise stated, or unless the skilled person would otherwise work under an inert atmosphere; column chromatography (by the flash procedure) was used to purify compounds and was performed on Merck Kieselgel silica (Art. 9385) unless otherwise stated; in general, the course of reactions was followed by TLC, HPLC, or LC/MS and reaction times are given for illustration only; yields are given for illustration only and are not necessarily the maximum attainable; the structure of the end products of the invention was generally confirmed by NMR and mass spectral techniques. Proton magnetic resonance spectra were generally determined in DMSO d6 unless otherwise stated, using a Bruker DRX 300 spectrometer or a Bruker DRX-400 spectrometer, operating at a field strength of 300 MHz or 400 MHz, respectively. In cases where the NMR spectrum is complex, only diagnostic signals are reported. Chemical shifts are reported in parts per million downfield from tetramethylsilane as an external standard (* scale) and peak multiplicities are shown thus: s, singlet; d, doublet; dd, doublet of doublets; dt, doublet of triplets; dm, doublet of multiplets; t, triplet, m, multiplet; br, broad. Fast atom bombardment (FAB) mass spectral data were generally obtained using a Platform spectrometer (supplied by Micromass) run in electrospray and, where appropriate, either positive ion data or negative ion data were collected or using Agilent 1100 series LC/MS equipped with Sedex 75ELSD, and where appropriate, either positive ion data or negative ion data were collected. The lowest mass major ion is reported for molecules where isotope splitting results in multiple mass spectral peaks (for example when chlorine is present). Reverse Phase HPLC was carried out using YMC Pack ODS AQ (100x20 mmID, S 5A particle size, 12 nm pore size) on Agilent instruments; each intermediate was purified to the standard required for the subsequent stage and was characterized in sufficient detail to confirm that the assigned structure was correct; purity was assessed by HPLC, TLC, or NMR and identity was determined by infrared spectroscopy (IR), mass spectroscopy or NMR spectroscopy as appropriate.
[000131] Synthesis of intermediates
Synthesis of 6-Bromo-2H-pyrido[3, 2-b][l,4]oxazin-3(4H)-one (Intermediate I)
Figure imgf000047_0001
[000132] Step 1: Synthesis of 6-bromo-2-nitropyridin-3-ol (la): To a solution of 4,6-dichloro-5-metho 2-nitropyridin-3-ol (20g, 0.142mol) in DMF(400ml), N- bromosuccinimide (32.52g, 0.187mol) was added portion wise over a period of 5 hours at 0°C. The reaction mixture was stirred for room temperature for 12h. After completion of the reaction, the reaction mixture was concentrated in vacuo. The residue was taken up in the Ether and the mixture was stirred for 30min. The precipitate was removed by filtration and the filtrate was concentrated in vacuo to get 6-bromo-2-nitropyridin-3-ol, la (40g, 46%) as a mixture of mono and di bromo compound. The crude LCMS showed 46% of expected mono-bromo compound, this material was used as such for next step without further purification. LCMS Calculated for C5H3BrN203, 218.99, Observed = 219.2.
[000133] Step 2: Synthesis of ethyl 2-((6-bromo-2-nitropyridin-3-yl)oxy)acetate (lb): To solution of 6-bromo-2-nitropyridin-3-ol, la (40g, 0.182mol) in Acetone (400ml), cooled to 0°C, potassium carbonate (50.4 lg, 0.365mol), was added and stirred for 5min. Then, Ethyl bromoacetate (39.7g, 0.237mol) was added slowly and refluxed at 65 °C for 12 h. After completion of the reaction, it was filtered and the filtrate was concentrated in vacuo. The crude was diluted with water and extracted with Ethyl acetate (2* 600 mL). The combined organic layers were washed with brine solution, dried over Na2S04, filtered and concentrated in vacuo. It was purified by column chromatography on silica gel with gradient elution of 20-22% of Ethyl acetate in pet ether to obtain ethyl 2-((6- bromo-2-nitropyridin-3-yl)oxy)acetate, lb (21g, 75.32 %) as a pale yellow solid. LCMS = Calculated for C9H9BrN205, 305.38, Observed = 306.2.
[000134] Step 3: Synthesis of 6-bromo-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (I): To a stirred solution of ethyl 2-((6-bromo-2-nitropyridin-3-yl)oxy)acetate, lb (21g, 0.0687mol) in glacial acetic acid (400ml), Iron powder(l 1.51g, 0.2063mol) was added and heated to 100°C for 6 hours. After completion of the reaction, reaction mixture was filtered through celite bed using ethyl acetate, 10% Methanol and concentrated in vacuo. It was washed with Methanol to obtain pure 6-bromo-2H-pyrido[3,2-b][l,4]oxazin- 3(4H)-one, I (12g, 76.28% ). LCMS = Calculated for C7H5BrN202, 229.03, Observed = 230.2.
[000135] Synthesis of 6-(5-(2-Aminoethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2- b][l,4]oxazin-3(4H)-one (Intermediate II)
Figure imgf000049_0001
[000136] Step 1: Synthesis of (but-3-en-l-yloxy)(tert-butyl)dimethylsilane, Ila:
To a stirred solution of but-3-en-l-ol (lOg, 0.1386 mol) in Dichloromethane (150 ml), cooled to 0°C, triethylamine ( 19.64g, 0.1941 mol), was added and stirred for 5min. Then, tert-butyldimethylsilyl chloride (25.08g, 0.1664mol) and DMAP (a catalytic amount) in dichloromethane was slowly added to the reaction mixture and stirred at 25 °C for 4 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with MTBE. The combined organic layers were washed with brine solution, dried over Na2S04, filtered and concentrated in vacuo at 40°C to obtain (but-3-en-l- yloxy)(tert-butyl)dimethylsilane, Ila ( 24g, 93% ). The crude material was taken for the next step without any purification based on NMR monitoring of the reaction.
[000137] Step 2: Synthesis of tert-butyldimethyl(2-(oxiran-2-yl)ethoxy)silane, lib: To a stirred solution of (but-3-en- l-yloxy)(tert-butyl)dimethylsilane, Ila (24g, 0.1287mol) in dichloromethane (480ml), cooled to 0°C, m-CPBA(63.49g, 0.2575mol), was added portion wise and stirred at 25°C for 7 hours. After completion of the reaction, reaction mixture was filtered to remove inorganics. Then, it was quenched with 10% sodium thio sulphate and extracted with dichloromethane. The separated organic layer was washed with 10% sodium bicarbonate, brine solution, dried over Na2S04, filtered and concentrated in vacuo at 40°C to obtain tert-butyldimethyl(2-(oxiran-2- yl)ethoxy)silane, lib (22g, 85%). The crude material was taken for the next step without any purification.
[000138] Step 3: Synthesis of l-amino-4-((tert-butyldimethylsilyl)oxy)butan-2- ol, lie: To a stirred solution of tert-butyldimethyl(2-(oxiran-2-yl)ethoxy)silane, lib (22g, 0.1087mol) in methanol (100ml), were added NH3 in methanol (250ml) and aqueous ammonia (100ml) and stirred at 25°C for 14 hours. After completion of the reaction, reaction mixture was concentrated in vacuo to obtain 1 -amino-4-((tert- butyldimethylsilyl)oxy)butan-2-ol, He ( 20g, 84%). The crude material was taken for the next step without any purification. LCMS = Calculated for C10H25NO2S1, 219.4, Observed = 220.2.
[000139] Step 4: Synthesis of 5-(2-((tert- butyldimethylsilyl)oxy)ethyl)oxazolidin-2-one, lid: To a stirred solution of l-amino-4- ((tert-butyldimethylsilyl)oxy)butan-2-ol, lie ( 20g, 0.0912mol) in THF (300ml), cooled to 0°C, CDI (22.5g, 0.1368mol), was added portion wise and heated at 50°C for 14 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with ethyl acetate. The separated organic layer was washed with water, brine solution, dried over Na2S04, filtered and concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 35-40% ethyl acetate in pet ether to obtain 5-(2-((tert-butyldimethylsilyl)oxy)ethyl)oxazolidin-2-one, lid (lOg, 45%). LCMS = Calculated for C11H23N03Si, 245.39, Observed = 246.4.
[000140] Step 5: Synthesis of 6-(5-(2-((tert-butyldimethylsilyl)oxy)ethyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, He: To a stirred solution of 6-bromo-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, I (0.3g, 0.0013mol) and 5-(2-((tert- butyldimethylsilyl)oxy)ethyl)oxazolidin-2-one, lid ( 0.385g, 0.0015mol) in Aldrich dry THF(lOml), were added i-butyl-X-Phos mesyl chloride complex (0.051g, 0.00006mol) and sodium tert-butoxide (0.187g, 0.0019mol) and degassed for 20 mins. Then, it was irradiated in microwave at 100°C for 25mins. After completion of the reaction, reaction mixture was concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 21-25% ethyl acetate in pet ether to obtain 6- (5-(2-((tert-butyldimethylsilyl)oxy)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2- b][l,4]oxazin-3(4H)-one, He (0.2g, 40%). LCMS = Calculated for Ci8H27N305Si, 393.52, Observed = 394.5.
[000141] Step 6: Synthesis of 6-(5-(2-hydroxyethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, Ilf: To a stirred solution of 6-(5-(2-((tert- butyldimethylsilyl)oxy)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)- one, He (lg, 0.0025mol) in THF (15ml), cooled to 0°C, Tert-butyl ammonium fluoride (1.99g, 0.0076mol) was added drop wise and stirred at 25°C for 3 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with ethyl acetate. The separated organic layer was washed with brine solution, dried over Na2S04, filtered and concentrated in vacuo to obtain colourless gummy solid of 6-(5-(2- hydroxyethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Ilf (0.5g, 71.63%). The crude material was taken for the next step without any purification. LCMS = Calculated for ^Η13Ν305, 279.24, Observed = 280.2.
[000142] Step 7: Synthesis of 2-(2-oxo-3-(3-oxo-3,4-dihydro-2H-pyrido[3,2- b][l,4]oxazin-6-yl)oxazolidin-5-yl)ethyl methanesulfonate, Ilg: To a stirred solution of 6-(5-(2-hydroxyethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Ilf (0.5g, 0.0017mol) in dichloromethane, cooled to 0°C, triethylamine (0.543g, 0.0053mol) and mesyl chloride (0.155g, 0.0013mol) were added and stirred at 25°C for 2 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with dichloromethane. The separated organic layer was washed with water, brine solution, dried over Na2S04, filtered and concentrated in vacuo to obtain pale brown gummy solid of 2-(2-oxo-3-(3-oxo-3,4-dihydro-2H-pyrido[3,2-b][l,4]oxazin-6-yl)oxazolidin-5- yl)ethyl methanesulfonate, Ilg (0.5g, 82.50%). The crude material was taken for the next step without any purification. LCMS = Calculated for C13H15N3O7S, 357.34, Observed = 358.4. [000143] Step 8: Synthesis of 6-(5-(2-azidoethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, Ilh: To a stirred solution of 2-(2-oxo-3-(3-oxo- 3,4-dihydro-2H-pyrido[3,2-b][l,4]oxazin-6-yl)oxazolidin-5-yl)ethyl methanesulfonate, Ilg (0.5g, 0.0013mol) in DMF (10ml), cooled to 0°C, sodium azide (0.181g, 0.0027mol) was added and heated at 60°C for 3 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with ethyl acetate. The separated organic layer was washed with water, brine solution, dried over Na2S04, filtered and concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 50-60% ethyl acetate in pet ether to obtain colourless solid of 6-(5-(2- azidoethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Ilh (0.32g, 81.01%). LCMS = Calculated for Ci2Hi2N604, 304.27, Observed = 305.5.
[000144] Step9: Synthesis of 6-(5-(2-aminoethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one (Intermediate II): To a stirred solution of 6-(5-(2- azidoethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, 12 (0.32g, O.OOlOmol) in THF: MeOH (1 : 1) (10ml), 10% palladium on carbon (30mg) was added and stirred at 25°C under H2 bladder pressure for 3 hours. After completion of the reaction, reaction mixture was filtered through celite bed using THF and MeOH and concentrated in vacuo to obtain colourless solid of 6-(5-(2-aminoethyl)-2-oxooxazolidin- 3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, II (0.25g, 89.92%). The crude material was taken for the next step without any purification. 1H NMR (400 MHz, DMSO-d6) δ 7.60 (d, J = 8.6 Hz, 1H), 7.44 (d, J = 8.6 Hz, 1H), 4.79-4.80 (m, 1H), 4.62 (s, 2H), 4.21- 4.25 (m, 1H), 3.73-3.77 (m, 1H), 2.87-2.90 (m, 2H), 1.75-1.88 (m, 2H), 1.36 (s, 2H); LCMS calculated for Ci2H14N404, 278.27 Observed = 279
[000145] Synthesis of 6-(5-(3-Aminopropyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one (Intermediate III)
Figure imgf000053_0001
[000146] Step-1: Synthesis of3-(2-oxo-3-(3-oxo-3,4-dihydro-2H-pyrido[3,2- b][l,4]oxazin-6-yl)oxazolidin-5-yl)propanenitrile(IIIa): To a stirred solution of starting material Ilg (0.7 g, 1.96 mmol) in DMSO (5 mL) was added sodium cyanide (0.38 g, 7.84 mmol), and the resulting reaction mixture was heated to 50°C for 3h. The reaction was monitored by LCMS. After completion of the reaction, the reaction mixture was poured into water (15ml) and extracted with Ethyl acetate (2 x 30 mL). The combined organic layers were washed with brine solution (20mL) and concentrated under reduced pressure to obtain crude compound, which was purified by column chromatography eluted with 40% EtOAC in pet ether to afford title compound (O.lg) as an off white solid.
[000147] Step-2: Synthesis of tert-butyl (3-(2-oxo-3-(3-oxo-3,4-dihydro-2H- pyrido[3,2-b][l,4]oxazin-6-yl)oxazolidin-5-yl)propyl)carbamate (Illb): To a stirred solution of starting material, Ilia (0.1 g,0.346 mmol) in Methanol (5 mL) was added Cobalt(II)chloride hexahydrate (0.41 g, 1.734 mmol) and Boc-anhydride (0.138 g, 0.692 mmol) at 0°C. After 5 min stirring, sodium borohydride (40 mg, 1.038 mmol) was added, reaction was warmed to room temperature and stirred for 4h. After the complete consumption of starting material, the reaction mixture was filtered through celite bed, and filtrate was concentrated on rotatory evaporator. Obtained residue was diluted with water, and extracted with ethyl acetate (2 x 20 mL). The combined organic layers were washed with brine solution (15 mL) and concentrated to obtain a crude residue, which was purified by column chromatography by using 35% EtOAC in pet ether to afford title compound (50 mg) as a pale yellow solid. [000148] Step-3: Synthesis of 6-(5-(3-Aminopropyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one(III) To a stirred solution of starting material, Illb (50 mg,) in dichloromethane (5 mL), was added trifluoroacetic acid (0.5 mL) at 0°C and stirred at RT for 3 h. After completion of the reaction, the reaction mixture was concentrated in vaccuo, and washed with pet ether to afford title compound (40 mg) as light brown solid. XH NMR (400 MHz, DMSO-d6) δ 7.71 (bs, 1H), δ 7.59 (d, / = 8.6 Hz, 1H), 7.44 (d, J = 8.6 Hz, 1H), 4.68-4.75 (m, 1H), 4.61 (s, 2H), 4.22 (dd, / = 8.4, 10 Hz, 1H), 3.70 (dd, / = 6.6 Hz, 10 Hz, 1H), 2.85 (t, = 8.0 Hz, 2H), 1.73-1.81 (m, 2H), 1.61- 1.71 (m, 2H); LCMS calculated for d3H16N404, 292.30 Observed = 293.2.
[000149] Alternate route for the synthesis of 6-(5-(3-Aminopropyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Intermediate III)
Figure imgf000054_0001
[000150] Step 1: Synthesis of (pent-3-en-l-yloxy)(tert-butyl)dimethylsilane,
IIIc: The solution of pent-3-en-l-ol (20g, 0.232 mol) in dichloromethane (200 ml) was cooled to 0°C, and triethylamine (45.01 mL, 0.325 mol), followed by tert- butyldimethylsilyl chloride (50.3 g, 0.0.278 mol) and DMAP (0.56 g, 4.644 mmol) were added to the reaction mixture and stirred at 25°C for 4 hours. After completion of the reaction, reaction mixture was diluted with dichloromethane (100 mL) and washed successively with water and brine solution. Organic layer was dried over Na2S04, and concentrated in vacuo at 40°C to obtain (pent-3-en-l-yloxy)(tert-butyl)dimethylsilane, IIIc ( 42.06g ). The crude material was taken for the next step without any purification. 1HNMR (DMSO-d6) δ 5.76 - 5.86 (m, 1H), 4.93-5.04 (m, 2H), 3.58 (t, = 8.4 hz, 2H), 2.02-2.10 (m, 2H), 1.49-1.58 (m, 2H), 0.93 (s, 9H), 0.09 (s, 6H)
[000151] Step 2: Synthesis of tert-butyldimethyl(2-(oxiran-2-yl)ethoxy)silane, Hid: A stirred solution of (pent-3-en-l-yloxy)(tert-butyl)dimethylsilane, IIIc (25 g, 0.125 mol) in dichloromethane (500 ml) was cooled to 0°C and m-CPBA(63.49g, 0.2575mol), was added portion wise over 20 mins. After the completion of addition, reaction was warmed to 25°C and stirred for 7 hours. Reaction mixture was filtered to remove inorganics. Then, it was quenched with 10% sodium thio sulphate and extracted with dichloromethane. The separated organic layer was washed with 10% sodium bicarbonate, brine solution, dried over Na2S04, filtered and concentrated in vacuo at 40°C to obtain tert-butyldimethyl(2-(oxiran-2-yl)ethoxy)silane, Hid (21g). The crude material was taken for the next step without any purification.
[000152] Step 3: Synthesis of intermediate Ille: To the mixture of epoxide Hid (9 g, 0.039 mol) and ethylcarbamate (3.5 g, 0.039 mol) in a sealed tube, was added triethylamine (5 mL, 0.039 mol) and the mixture was heated at 145 °C for overnight. After the completion of reaction, reaction mixture was diluted with water and extracted with ethyl acetate. Organic layer was washed with water, dried over sodium sulphate and evaporated under reduced pressure. Crude residue was purified by silica gel column chromatography (25% ethyl acetate in pet-ether) to get intermediate Ille (4.0 g).LCMS calculated for Ci2H25N03Si 259.42, observed = 260.0
[000153] Step 4: 6-(5-(2-((tert-butyldimethylsilyl)oxy)propyl)-2-oxooxazolidin- 3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Illf: To a stirred solution of 6-bromo- 2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, I (0.300 g, 1.310 mmol) and 5-(2-((tert- butyldimethylsilyl)oxy)propyl)oxazolidin-2-one, Ille ( 0.407 g, 1.572 mmol) in Aldrich dry THF(12ml), were added t-butyl-X-Phos mesyl chloride complex (0.052g, 0.0655 mmol) and sodium tert-butoxide (0.189 g, 1.965 mmol) and degassed for 20 mins. Then, it was irradiated in microwave at 100°C for 40 mins. After completion of the reaction, reaction mixture was concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 21-25% ethyl acetate in pet ether to obtain 6-(5-(2-((tert-butyldimethylsilyl)oxy)propyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, Illf (0.231g). LCMS calculated for Ci9H29N305Si 407.42, Observed = 408
[000154] Step 5: Synthesis of 6-(5-(3-hydroxypropyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, Illg: To a stirred solution of 6-(5-(2-((tert- butyldimethylsilyl)oxy)propyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin- 3(4H)-one, Illf (4.3g, 0.0105mol) in THF (45ml), cooled to 0°C, Tert-butyl ammonium fluoride (5.64g, 0.0216mol) was added drop wise and stirred at 25°C for 3 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with ethyl acetate. The separated organic layer was washed with brine solution, dried over Na2S04, filtered and concentrated in vacuo to obtain colourless gummy solid of 6-(5-(3- hydroxypropyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Illg (2.95g, 95.81%). The crude material was taken for the next step without any purification. LCMS = Calculated for C13H15N3O5, 293.28, Observed = 293.8.
[000155] Step 6 Synthesis of 3-(2-oxo-3-(3-oxo-3,4-dihydro-2H-pyrido[3,2- b][l,4]oxazin-6-yl)oxazolidin-5-yl)propyl methanesulfonate, Illh: To a stirred solution of 6-(5-(3-hydroxypropyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin- 3(4H)-one, IIIg(2.95g, O.OlOmol) in dichloromethane (30ml), cooled to 0°C, triethylamine (3.05g, 0.030mol) and mesyl chloride (1.14g, O.OlOmol) were added and stirred at 25 °C for 2 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with dichloromethane. The separated organic layer was washed with water, brine solution, dried over Na2S04, filtered and concentrated in vacuo to obtain pale brown gummy solid of 3-(2-oxo-3-(3-oxo-3,4-dihydro-2H- pyrido[3,2-b][l,4]oxazin-6-yl)oxazolidin-5-yl)propyl methanesulfonate, Illh (2.8g, 75.47%). The crude material was taken for the next step without any purification. LCMS = Calculated for Ci4Hi7N307S, 371.36, Observed = 371.8.
[000156] Step 7 Synthesis of 6-(5-(3-azidopropyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, Illi: To a stirred solution of 3-(2-oxo-3-(3-oxo- 3,4-dihydro-2H-pyrido[3,2-b][l,4]oxazin-6-yl)oxazolidin-5-yl)propyl methanesulfonate, Illh (2.8g, 0.0075mol) in DMF (30ml), cooled to 0°C, sodium azide (0.98g, 0.0150mol) was added and heated at 60°C for 3 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with ethyl acetate. The separated organic layer was washed with water, brine solution, dried over Na2S04, filtered and concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 38-43% ethyl acetate in pet ether to obtain colourless solid of 6-(5-(3- azidopropyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one, Illi (1.5g, 63.02%). LCMS = Calculated for ¾Η14Ν6θ4, 318.29, Observed = 318.8.
[000157] Step 8 Synthesis of 6-(5-(3-aminopropyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, III: To a stirred solution of 6-(5-(3-azidopropyl)- 2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one,IIIi (1.3g, O.OOlOmol) in THF: MeOH (1 : 1) (10ml), 10% palladium on carbon (0.6g) was added and stirred at 25°C under H2 bladder pressure for 3 hours. After completion of the reaction, reaction mixture was filtered through celite bed using THF and MeOH and concentrated in vacuo to obtain colourless solid of 6-(5-(3-aminopropyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2- b][l,4]oxazin-3(4H)-one, III (0.8g, 67.22%). The crude material was taken for the next step without any purification. 1H NMR (400 MHz, DMSO-d6) δ 7.71 (bs, 1H), δ 7.59 (d, J = 8.6 Hz, 1H), 7.44 (d, = 8.6 Hz, 1H), 4.68-4.75 (m, 1H), 4.61 (s, 2H), 4.22 (dd, = 8.4, 10 Hz, 1H), 3.70 (dd, = 6.6 Hz, 10 Hz, 1H), 2.85 (t, = 8.0 Hz, 2H), 1.73-1.81 (m, 2H), 1.61-1.71 (m, 2H); LCMS calculated for C13H16N4O4, 292.30 Observed = 293.2.S
[000158] Synthesis of 6-(5-(2-Aminoethyl)-2-oxooxazolidin-3-yl)-2H- benzo[b][l,4]oxazin-3(4H)-
Figure imgf000057_0001
[000159] Intermediate IV, 6-(5-(2-Aminoethyl)-2-oxooxazolidin-3-yl)-2H- benzo[b][l,4]oxazin-3(4H)-one (CAS number 1072793-84-0 ) was synthesized as reported earlier in WO2008126024. [000160] Synthesis of 5-(5-(2-aminoethyl)-2-oxooxazolidin-3-yl)-2- methylnicotinonitrile (Inter
Figure imgf000058_0001
Step-l Step-2
Figure imgf000058_0002
[000161] Step 1: Synthesis of 5-((4-((tert-butyldimethylsilyl)oxy)-2- hydroxybutyl)amino)-2-methylnicotinonitrile, Va: To a stirred solution of 5-bromo-2- methylnicotinonitrile (850mg, 4.31mmol) and 5-(2-((tert- butyldimethylsilyl)oxy)ethyl)oxazolidin-2-one, lid ( 1268mg, 5.17mmol) in Aldrich dry THF(25ml), were added i-butyl-X-Phos mesyl chloride complex (171mg, 0.21mmol) and sodium tert-butoxide (621mg, 6.47mmol) and degassed for 20 min. Then, it was irradiated in microwave at 100°C for 20mins. After completion of the reaction, reaction mixture was concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 26-30% ethyl acetate in pet ether to obtain 5- ((4-((tert-butyldimethylsilyl)oxy)-2-hydroxybutyl)amino)-2-methylnicotinonitrile, Va (500mg, 35%). LCMS = Calculated for C17H29N3O2S1, 335.52, Observed = 336.2.
[000162] Step 2: Synthesis of 5-(5-(2-((tert-butyldimethylsilyl)oxy)ethyl)-2- oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vb: To a stirred solution of 5-((4-((tert- butyldimethylsilyl)oxy)-2-hydroxybutyl)amino)-2-methylnicotinonitrile, Va (500mg, 1.49mmol) in dry Dichlormethane (5ml), cooled to 0°C, triefhylamine (754mg, 7.54mmol) and phosgene (1170mg, 11.92mmol) were added and stirred at 25°C for 2h. After completion of the reaction, reaction mixture was quenched with water and extracted with ethyl acetate. The separated organic layer was washed with brine solution, dried over Na2S04, filtered and concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 25-27% ethyl acetate in pet ether to obtain 5-(5-(2-((tert-butyldimethylsilyl)oxy)ethyl)-2- oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vb (340mg, 64%). LCMS = Calculated for Ci8H27N303Si, 361.52, Observed = 362.
[000163] Step 3: Synthesis of 5-(5-(2-hydroxyethyl)-2-oxooxazolidin-3-yl)-2- methylnicotinonitrile, Vc: To a stirred solution of 5-(5-(2-((tert- butyldimethylsilyl)oxy)ethyl)-2-oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vb (340mg, 0.94mmol) in THF (8ml), cooled to 0°C, Tert-butyl ammonium fluoride (737mg, 2.82mmol) was added drop wise and stirred at 25°C for 3 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with ethyl acetate. The separated organic layer was washed with brine solution, dried over Na2S04, filtered and concentrated in vacuo to obtain colourless gummy solid of 5-(5-(2-hydroxyethyl)-2- oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vc (200mg, 86.2%). The crude material was taken for the next step without any purification. LCMS = Calculated for Ci2Hi3N303, 247.25, Observed = 247.8.
[000164] Step 4: Synthesis of 2-(3-(5-cyano-6-methylpyridin-3-yl)-2- oxooxazolidin-5-yl)ethyl methanesulfonate, Vd: To a stirred solution 5-(5-(2- hydroxyethyl)-2-oxooxazolidin-3-yl)-2-methylnicotinonitrile, Vc (200mg, 0.80mmol) in dichlorome thane, cooled to 0°C, triethylamine (245mg, 2.42mmol) and mesyl chloride (91mg, 0.80mmol) were added and stirred at 25°C for 2 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with dichlorome thane. The separated organic layer was washed with water, brine solution, dried over Na2S04, filtered and concentrated in vacuo to obtain pale brown gummy solid of 2-(3-(5-cyano-6- methylpyridin-3-yl)-2-oxooxazolidin-5-yl)ethyl methanesulfonate, Vd (200mg, 77%). The crude material was taken for the next step without any purification. LCMS = Calculated for C13H15N3O5S, 325.34, Observed = 326.
[000165] Step 5: Synthesis of 5-(5-(2-azidoethyl)-2-oxooxazolidin-3-yl)-2- methylnicotinonitrile, Ve: To a stirred solution of 2-(3-(5-cyano-6-methylpyridin-3-yl)- 2-oxooxazolidin-5-yl)ethyl methanesulfonate, Vd (200mg, 0.614mmol) in DMF (4ml), cooled to 0°C, sodium azide (59mg, 0.92mmol) was added and heated at 60°C for 3 hours. After completion of the reaction, reaction mixture was quenched with water and extracted with ethyl acetate. The separated organic layer was washed with water, brine solution, dried over Na2S04, filtered and concentrated in vacuo. It was purified by column chromatography on silica gel (230-400mesh) with gradient elution of 48-52% ethyl acetate in pet ether to obtain colourless gummy solid of 5-(5-(2-azidoethyl)-2- oxooxazolidin-3-yl)-2-methylnicotinonitrile, Ve (lOOmg, 60%). LCMS = Calculated for Ci2Hi2N602, 272.27, Observed = 273.5.
[000166] Step 6: Synthesis of 5-(5-(2-aminoethyl)-2-oxooxazolidin-3-yl)-2- methylnicotinonitrile, V: To a stirred solution of 5-(5-(2-azidoethyl)-2-oxooxazolidin-3- yl)-2-methylnicotinonitrile, Ve (50mg, 0.183mmol) in Methanol (5ml), triethylamine (74mg, 0.734mmol) and 1,3-propane dithiol (79mg, 0.734mmol) were added and stirred at 25°C for 12 hours. After completion of the reaction, reaction mixture was filtered to remove inorganics and concentrated in vacuo to obtain crude pale green colour of gummy liquid of 5-(5-(2-aminoethyl)-2-oxooxazolidin-3-yl)-2-methylnicotinonitrile, V (43mg, 95%). The crude material was taken for the next step without any purification. LCMS = Calculated for Ci2Hi4N402, 246.27, Observed = 247.0.
[000167] Synthesis of l-methyl-2-oxoindoline-7-carbaldehvde. Intermediate VI
Figure imgf000061_0001
Steps 1& 2 Step 4
M
Figure imgf000061_0002
Mol. Wt: 175.19 Mol. Wt: 173.21
[000168] Step-1: Synthesis of Via: To a solution of chloral hydrate (5.28 g, 31.9 mmol) in deionized water (90 mL), sodium sulphate (53.6 g, 377.9 mmol), 2- bromoaniline (5 g, 29.11 mmol), were added sulphuric acid (20 mL) followed by hydroxylamine hydrochloride (6.66 g, 87.2 mmol) and the whole mixture was heated to 130 C for 30 min. The mixture was cooled to room temperature, poured on ice-water mixture and extracted with ethylacetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. Crude obtained was taken for the next step without any purification.
[000169] Step-2: Sulphuric acid (50 mL) was added to the crude obtained in stepl and the mixture was heated to 70 °C for lh. After the complete consumption of starting material, reaction mixture was poured over ice, and extracted with ethyl acetate (2 x 200 mL). Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. Crude was purified by silica gel column chromatography by using 20% ethyl acetate in pet ether for elution to get 2.5 g of Via.
[000170] Step-3: Synthesis of VIb: To a solution of Via (2.5 g, 11.061 mmol) in ethanol (50 mL), hydrazine hydrate (99%, 0.5 mL) was added and the mixture was refluxed under nitrogen atmosphere for 30 min. Formed yellow precipitate was isolated by filtration, and suspended in anhydrous ethanol. Potassium tertiary butoxide (4.03 g, 35.90 mmol) was added to the above suspension. The mixture was refluxed for 2h. After the completion of reaction, mixture was poured over ice-water, acidified with dil. HC1 to pH 2. Mixture was extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude of VIb (0.89 g) which was taken for the next step without any purification.
[000171] Step-4: Preparation of Vic: To the suspension of VIb (200mg, 0.94 mmol) in water (5 mL) and sodium hydroxide (56mg, 1.41 mmol) was added dimethyl sulphate (178mg, 1.41 mmol) and the mixture was heated at 100 oC for lh. After the consumption of starting material, reaction mixture was cooled to 0 oC, added water and extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. So obtained crude was purified by column chromatography by using 10% ethyl acetate in pet-ether to get pure Vic (180 mg).
[000172] Step-5: Preparation of VId: To a solution of Vic (180mg, 0.79 mmol) in DMF (4 mL) was added tributylvinyltin (0.26 mL, 2.25 mmol) followed by palladium tetrakis-triphenylphosphine (45 mg, 0.103 mmol) and the mixture was degassed for 30 min by purging with argon. Reaction was heated to 100 °C for 3h. After the completion of reaction, mixture was cooled to 0 °C, added water and extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. So, obtained crude was purified by column chromatography using 10% ethyl acetate in pet-ether to pure VId (100 mg)
[000173] Step-6 Preparation of VI: Olefin VId ( lOOmg, 0.52 mmol) was dissolved in 4 mL methanol and purged with oxygen for 10 min. Mixture was than cooled to -78 °C and ozone was continuously passed through reaction mixture for 30 min. After the complete consumption of VId, reaction mass was quenched with dimethylsulphide (0.5 mL) and evaporated under reduced pressure to dryness. Residue obtained was purified by column chromatography with eluent 15% ethyl acetate in pet ether to obtain pale yellow solid (30mg). 1HNMR (400MHz, CDC13) 510.45 (s, 1H), 7.81 (d, J = 7.6 Hz, 1H), 7.45 (dd, J = 1.2, 7.6 Hz, 1H), 7.13 (t, J = 7.6 Hz, 1H), 3.60 (s, 2H), 3.55 (s, 3H); UPLCMS calculated for Ci0H9NO2 175.19, observed = 175.8
-methyl-2-oxoindoline-7-carbaldehyde. Intermediate VII
Figure imgf000063_0001
Steps 1& 2 Step 3 Step 4
Figure imgf000063_0002
Mol. Wt: 193.18 Mol. Wt: 191.21
[000175] Step-1: Synthesis of Vila: To a solution of chloral hydrate (9.55 g, 57.89 mmol) in deionized water (150 mL), sodium sulphate (100 g, 684.19 mmol), 3-bromo-2- fluoroaniline (10 g, 52.63 mmol), were added sulphuric acid (40 mL) followed by hydroxylamine hydrochloride (10.97 g, 157.89 mmol) and the whole mixture was heated to 130 °C for 30 min. The mixture was cooled to room temperature, powered on ice-water mixture and extracted with ethylacetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. Crude obtained was taken for the next step without any purification.
[000176] Step-2: Sulphuric acid (100 mL) was added to the crude obtained in step 1 and the mixture was heated to 70 °C for lh. After the complete consumption of starting material, reaction mixture was poured over ice, and extracted with ethyl acetate (2 x 200 mL). Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. Crude was purified by silica gel column chromatography by using 20% ethyl acetate in pet ether for elution to get 7.5 g of Vila.
[000177] Step-3:Preparation of Vllb: To a solution of Vila (2.8 g, 11.48 mmol) in ethanol (50 mL), hydrazine hydrate (99%, 0.5 mL) was added and the mixture was refluxed under nitrogen atmosphere for 30 min. Formed yellow precipitate was isolated by filteration, and suspended in anhydrous ethanol. Potassium tertiary butoxide (4.03 g, 35.90 mmol) was added to the above suspension. The mixture was refluxed for 2h. After the completion of reaction, mixture was poured over ice-water, acidified with dil. HC1 to pH 2. Mixture was extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude of Vllb (2.0 g) which was taken for the next step without any purification.
[000178] Step-4:Preparation of VIIc: To the suspension of Vllb (lg, 4.34 mmol) in water (30 mL) and sodium hydroxide (0.397 g, 13.04 mmol) was added dimethyl sulphate (1.4 mL, 13.04 mmol) and the mixture was heated at 100 °C for 2h. After the consumption of starting material, reaction mixture was cooled to 0 °C, added water and extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. So, obtained crude was purified by column chromatography by using 21% ethyl acetate in pet-ether to get pure VIIc (0.500 g)
[000179] Step-5: Preparation of Vlld: To a solution of VIIc (0.500g, 2.04 mmol) in DMF (10 mL) was added tributylvinyltin (0.65 mL, 2.25 mmol) followed by palladium tetrakis-triphenylphosphine (0.118 g, 0.103 mmol) and the mixture was degassed for 30 min by purging with argon. Reaction was heated to 100 °C for 3h. After the completion of reaction, mixture was cooled to 0 °C, added water and extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. So, obtained crude was purified by column chromatography by using 40% dichloromethane in pet-ether to pure Vlld (0.300 g).
[000180] Step-6:Preparation of VII: Olefin Vlld (0.300g, 1.57 mmol) was dissolved in 10 mL methanol purged with oxygen for 10 min. Mixture was than cooled to -40 °C and ozone was continuously passed through reaction mixture for 30 min. After the complete consumption of Vlld, reaction mass was quenched with dimethylsulphide (0.5 mL) and evaporated under reduced pressure to dryness. Residue obtained was purified by column chromatography with eluent 40% ethyl acetate in pet ether to obtain white fluffy solid (0.050g). 1HNMR (400MHz, CDC13) δ 10.47 (s, 1H), 7.34-7.37 (m, 1H), 6.78-6.83 (m, 1H), 3.53 (s, 2H), 3.35 (s, 3H); UPLCMS calculated for Ci0H8FNO2 193.18, observed = 194.1
[000181] Synthesis of 6-fluoro-l,3,3-trimethyl-2-oxoindoline-7-carbaldehvde, Intermediate VIII
Figure imgf000065_0001
[000182] Step-1: Preparation of Villa: To the solution of Vllb (0.200 g, 0.869 mmol) in DMF (5mL) was added sodium hydride (60%, 0.140 g, 3.478 mmol), at 0 °C. After stirring at same temperature for 5 min., methyl iodide (2mL, 3.478 mmol) was introduced slowly, reaction temperature was gradually raised to room temperature and stirred for 2h. After the completion of reaction, whole reaction mixture was poured slowly on ice-cold water and extracted with ethyl acetate. Crude obtained after solvent evaporation was purified by column chromatography using 10% ethyl acetate in hexane to yield 0.180 g of Villa
[000183] Step-2: Preparation of Vlllb: To a solution of Villa (0.180g, 0.66 mmol) in DMF (5 mL) was added tributylvinyltin (0.2 mL, 0.727 mmol) followed by palladium tetrakis-triphenylphosphine (0.038 g, 0.033 mmol) and the mixture was degassed for 30 min by purging with argon. Reaction was heated to 100 °C for 3h. After the completion of reaction, mixture was cooled to 0 °C, added water and extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. So, obtained crude was purified by column chromatography by using 40% dichloromethane in pet-ether to pure VHIb (0.080 g)
[000184] Step-3: Preparation of VIII: Olefin VHIb (0.080 g, 3.65 mmol) was dissolved in 5 mL methanol purged with oxygen for 10 min. Mixture was than cooled to - 40 °C and ozone was continuously passed through reaction mixture for 30 min. After the complete consumption of VHIb, reaction mass was quenched with dimethylsulphide (0.5 mL) and evaporated under reduced pressure to dryness. Residue obtained was purified by column chromatography with eluent 40% ethyl acetate in pet ether to obtain white fluffy solid (0.038g). Structure was confirmed by NMR.1HNMR (400MHz, CDC13) 510.48 (s, 1H), 7.27-7.32 (m, 1H), 6.79-6.84 (m, 1H), 3.36 (s, 3H), 1.38 (s, 6H).
[000185] Synthesis of l-ethyl-6-fluoro-2-oxoindoline-7-carbaldehyde.
Intermediate IX
Figure imgf000066_0001
Figure imgf000067_0001
Step 3 t-butyl vinyl tin, Pd(PPh3)4, DMF, 100 °C, 3 h
Figure imgf000067_0002
IX IXc
[000186] Step-1: Preparation of IXa: To the solution of isatin intermediate Vila (6.6g, 27.04 mmol) in DMF (66 mL), were sequentially added K2C03 (5.4g, 39.08 mmol) and ethyl iodide (5.8 mL, 72.51 mmol) at room temperature, under nitrogen atmosphere. After stirring at rt for 2h, reaction mixture was cooled in ice -bath and water (100 mL) was added. Whole mixture was extracted with ethyl acetate. Organic layer was washed successively with cold water followed by brine and dried over sodium sulphate before evaporating on rotatory evaporator. Obtained crude residue was suspended in n-hexane and stirred for 20 mins. Solid was isolated by filtration to get the intermediate IXa (4.63 g)
[000187] Step-2: Preparation of IXb: Hydrazine hydrate ( 0.5 mL) was added to the solution of intermediate IXa (0.300 g )in ethanol and the mixture was heated in sealed tube to 130 °C. After 2 hours, usual aqueous work up was performed to isolate crude solid which was purified by column chromatography with eluent 30% ethylacetate in pet- ether to get 140 mg of compound IXb. UPLC calculated for Ci0H9BrFNO; 258.09, Observed = 258.0. [000188] Step-3: Preparation of IXc: To a solution of IXb (1.0 g, 3.815 mmol) in DMF (20 mL) was added tributylvinyltin (1.3mL, 4.263 mmol) followed by palladium tetrakis-triphenylphosphine (0.223 g, 0.193 mmol) and the mixture was degassed for 30 min by purging with argon. Reaction was heated to 100 °C for 3h. After the completion of reaction, mixture was cooled to 0 °C, added water and extracted with ethyl acetate. Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. So, obtained crude was purified by column chromatography by using 40% dichloromethane in pet-ether to pure IXc (0.370 g) LCMS Calculated for Ci2H12FNO 205.09, Observed = 205.8
[000189] Step-4:Preparation of IX: Olefin IXc (0.300g, 1.57 mmol) was dissolved in 10 mL methanol purged with oxygen for 10 min. Mixture was than cooled to -40 °C and ozone was continuously passed through reaction mixture for 30 min. After the complete consumption of, reaction mass was quenched with dimethylsulphide (0.5 mL) and evaporated under reduced pressure to dryness. Residue obtained was purified by column chromatography with eluent 40% ethyl acetate in pet ether to obtain white fluffy solid of aldehyde IX (0.090g). 1HNMR (400MHz, CDC13) δ 10.46 (s, 1H), 7.34-7.37 (m, 1H), 6.78-6.83 (m, 1H), 4.13, 4.10 (ABq, J = 7.0 Hz, 2H), 3.52 (s, 2H), 1.12 (t, J = 7.0 Hz, 1H); UPLCMS calculated for CnHi0FNO2 207.20, observed = 208
[000190] Synthesis of 2-(6-fluoro-l-methyl-2-oxoindolin-7-yl)acetaldehvde, IntermediateX
Figure imgf000068_0001
[000191] Step 1: Synthesis of 7-allyl-6-fluoro-l-methylindolin-2-one (Xa)
[000192] To a solution of VIIc (0.5 g, 2.049 mmol) in DMF (8 mL) was added allyltributylltin (0.45 mL, 1.45 mmol) followed by tetrakis(triphenylphosphine)palladium (0) (0.15 g, 0.13 mmol) and the mixture was degassed for 30 min by purging with argon. Reaction was heated to 100°C for 4 h. After the completion of reaction, the mixture was cooled to 0°C, diluted with water and extracted with EtOAc (100 mL). Organic layer was washed with brine, dried over sodium sulphate and evaporated under reduced pressure. The crude was purified by column chromatography on silica gel (230-400 mesh, 15% EtOAc in pet-ether) to pure Xa (0.255 g). UPLC_MS Calc. for Ci2Hi2FNO, 205.23; Obs. 206.5 [M++H].
[000193] Step 2: Synthesis of 2-(6-fluoro-l-methyl-2-oxoindolin-7- vDacetaldehyde (X)
[000194] Intermediate Xa (0.15 g, 0.731 mmol) was dissolved in 5 mL of DCM and cooled to -78°C. Ozone was continuously passed through reaction mixture for 30 min. After the complete consumption of Xa, reaction mass was quenched with dimethylsulphide (0.2 mL) and evaporated under reduced pressure to dryness. The residue obtained was purified by column chromatography on silica gel (230-400 mesh, 10% ethyl acetate in pet ether) to obtain X (0.125 g). This was characterized by GCMS Calc. for CnH10FNO2, 207.20; Obs. 207.0 [M+].
[000195] Intermediate XI, 6-(5-(3-aminopropyl)-2-oxooxazolidin-3-yl)-2H- benzo[b][l,4]oxazin-3(4H)-one (CAS number 1072800-19-1) was synthesized as reported earlier in WO2008126024
[000196] Synthesis of 2-(5-fluoro-3-methvl-2-oxo-2,3-dihydrobenzo[d1oxazol-4- yl)acetaldehyde (XII)
Figure imgf000069_0001
Xlle [000197] Stepl: Synthesis of 3-bromo-4-fluoro-2-nitrophenol (Xlla)
[000198] To a suspension of 3-bromo-4-fluoro phenol (8.00 g , 42.0 mmol) and ammonium nickel-(II)sulfate hexahydrate (8.00 g, 20.5 mmol) in 100 mL of dichloromethane was added 6 mL of nitric acid over 10 minutes while maintaining the internal temperature below 25 °C with an ice bath. The resulting mixture was allowed to stir for 20 minutes and poured into 250 g of crushed ice. The layers were separated and the aqueous phase was extracted with dichloromethane (2 x lOOmL). The organic layers were combined, washed with brine solution (100 mL), dried over anhydrous Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography using silica gel (60 - 120 mesh) eluting with 70 % dichloromethane in hexane give the product Xlla as a yellow solid. Yield: (3.00 g, 31%); 1H NMR (400 MHz, CDC13): δ 10.38 (s, 1H), 7.90 (d, = 7.6 Hz, 1H), 7.48 (d, = 6.0 Hz, 1H; ); UPLC-MS: Calc. for C6H3BrFN03 234.93; Obs. 235.9 [M+H]+.
[000199] Step2: Synthesis of 2-amino-3-bromo-4-fluorophenol (Xllb)
[000200] To a stirring solution of Xlla (3.00 g, 12.7 mmol) in methanol (50 mL) was added NiCl2 (3.20 g, 25.4 mmol) and NaBH4 (1.44 g, 38.0 mmol) and allowed to stir at room temperature for 10 minutes. The reaction mixture was filtered and washed with ethyl acetate (200 mL). The filtrate was concentrated to get the crude product Xllb (2.0 gm). The crude was used for the next step without any further purification.
[000201] Step3: Synthesis of 4-bromo-5-fluorobenzo[d1oxazol-2(3H)-one (XIIc)
[000202] To a stirring solution of Xllb (2.00 g, 9.70 mmol) in THF (30 mL) was added CDI (2.04 g, 12.6 mmol) and heated to 70 °C for 12 hours. After completion, the reaction mixture was allowed to cool to room temperature and diluted with ethyl acetate (50 mL) and washed with water (2 x 30 mL). The organic layer was dried over Na2S04 and concentrated under reduced pressure to get the crude product. The crude was purified by column chromatography using silica gel (60 - 120 mesh) eluting with 25 % EtOAc in hexane to afford product XIIc ( 2.00 g, 67%).1H NMR (400 MHz, DMSO-d6): δ 12.02 (brs, 1H), 7.73 (d, = 6.0 Hz, 1H), 7.22 (d, = 8.0 Hz, 1H).LC_MS: Calc. for C7H3BrFN02 230.93; Obs. 232.0 [M+H]+. [000203] Step 4: Synthesis of 4-bromo-5-fluoro-3-methylbenzordloxazol-2(3H)- one (Xlld)
[000204] To a stirring solution of XIIc (1.50 g, 6.50 mmol) in acetonitrile (20 mL) was added K2C03 (2.69 g, 19.5 mmol) and Me2S04 (1.70 mL, 19.5 mmol) and the mixture was heated to reflux for 1 hour. The reaction mixture was cooled to room temperature and diluted with ethyl acetate (50 mL) and washed with water (2 x 30 mL). The organic layer was dried over Na2S04 and concentrated under reduced pressure to get the crude product, which was purified by column chromatography using silica gel (60 - 120 mesh) eluting with 25 % ethyl acetate in hexane to afford product Xlld (1.00 g, 62 %); 1H NMR (400 MHz, CDC13): δ 7.40 (d, = 8.0 Hz, 1H), 6.81 (d, = 8.0 Hz, 1H), 3.40 (s, 1H).
[000205] Step 5: Synthesis of 4-allyl-5-fluoro-3-methylbenzord1oxazol-2(3H)- one (XHe)
[000206] To a solution of Xlld (1.00 g, 4.06 mmol) in DMF (20 mL) was added tributylallyltin (1.5 mL, 4.87 mmol) and the mixture was degassed for 30 minutes by purging with nitrogen, followed by added Pd(PPh3)4 (0.23 g, 0.20 mmol). The reaction mixture was heated to 100 °C for 8 hours. After completion, the reaction mixture was cooled to 0 C, quenched with water and extracted with ethyl acetate (2 x 50 mL). The combined organic layers were washed with water (50 mL), brine (50 mL), dried over Na2S04 and evaporated under reduced pressure to get crude product, which was purified by column chromatography using silica gel (60 - 120 mesh) eluting with 20 % ethyl acetate in petroleum ether to get pure product XHe (0.45 gm, 53%).
[000207] Step 6: Synthesis of 2-(5-fluoro-3-methyl-2-oxo-2,3- dihydrobenzo[d1oxazol-4-yl)acetaldehyde (XII)
[000208] To a solution of XHe (0.30 g, 1.44 mmol) in methanol (10 mL) was purged 03 at - 78 °C for 10 minutes. The reaction mixture was quenched with dimethylsulphide (1 mL) and allowed to stir for 30 minutes. The reaction mixture was concentrated and the residue was diluted with ethyl acetate (2 x 25 mL) and washed with water (20 mL), brine solution (20 mL), dried over Na2S04 and evaporated under reduced pressure to get crude product XII (0.13 g, crude). The crude was used for the next step without any further purification
[000209] Synthesis of 7-(aminomethvl)-6-fluoro-l-methyl-l,3-dihvdro-2H- pyrrolo[3,2-b]pyridin-2-one (XIII)
Zn(CN)2, Pd2dba3
Figure imgf000072_0001
[000210] Step 1: Synthesis of 6-fluoro-l-methyl-lH-pyrrolor3,2-blpyridine (Xllla)
[000211] To a solution of 6-Fluoro-lH-pyrrolo[3,2-b]pyridine (CAS: 1190320-33-2 , 6.5 g, 47.74 mmol) in dry DMF (65 mL), was added NaH(3.82 g, 95.49 mmol) at 0°C in portions, then the reaction mixture was stirred at room temperature for lh. Methyl iodide (5.30 mL, 95.49 mmol) was added at 0°C, then stirred at room temperature for 2h. After the consumption of starting material, reaction mixture was cooled to 0°C, added water and extracted with ethyl acetate (2X 100 mL)). The combined organic layers were washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude. Crude was purified by column chromatography (230/400 mesh, 30% ethyl acetate in pet-ether) to get desire compound Xllla as white solid (6 g, 85%). LC_MS Calc. for C8H7FN2 150.16; obtained: 151.11H NMR (400 MHz, DMSO-D6): δ 8.33 (s, 1H), 7.89-7.86 (m, 1H), 7.64 (d, 1H, J=2.8 Hz), 6.58 (s, 1H), 3.80 (s, 3H).
[000212] Step 2: Synthesis of 6-fluoro-l-methyl-lH-pyrrolor3,2-b]pyridine 4- oxide (XHIb) [000213] To a solution of XHIa (6 g, 39.95 mmol) in dry DCM (120 mL), was added mCPBA (13.80 g, 79.91 mmol) at 0°C in portions, then the reaction mixture was stirred at room temperature for 16h. After the consumption of starting material, reaction mixture was cooled to 0°C, added 10% aq NaHC03 (100 mL) and extracted with DCM (2X 100 mL)). The combined organic layers were washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude. Crude was washed with n- Hexane (2X 100 mL), then dried to get desire compound XHIb as white solid (4 g, 62%).LC_MS Calc. for C8H7FN20 166.16; obtained: 167.1 ; 1H NMR (400 MHz, DMSO-D6): δ 8.34-8.33 (m, 1H), 7.71-7.69 (m, 1H), 7.58-7.53 (m, 1H), 6.65 (d, 1H, J=3.2 Hz), 3.81 (s, 3H).
[000214] Step 3: Synthesis of 7-chloro-6-fluoro-l-methyl-lH-pyrrolor3,2- blpyridine (XIII c)
[000215] POCl3 (40 mL), was added to a compound XHIb (4 g, 24.07 mmol) drop wise at 0°C, then the reaction mixture was heated at 100 °C for 2h. After the consumption of starting material, reaction mixture was cooled to 0°C, added 10% aq NaHC03 (100 mL) in drop wise and extracted with ethyl acetate (2X 100 mL)). The combined organic layers were washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude. Crude was purified by column chromatography (230/400 mesh, 30% ethyl acetate in pet-ether) to get desire compound XIIIc as white solid (2 g, 45%).LC_MS Calc. for C8H6C1FN2 184.60; obtained: 185.2; *H NMR (400 MHz, DMSO-D6): δ 8.43 (s, 1H), 7.71 (s, 1H), 6.63 (d, 1H, J=3.6 Hz), 4.09 (s, 3H).
[000216] Step 4: Synthesis of 6-fluoro-l-methyl-lH-pyrrolo[3,2-b]pyridine-7- carbonitrile (XIII d)
[000217] To a solution of XIIIc (2 g, 10.83 mmol) in NMP (40 mL), was added zinc dust (14 mg, 0.21 mmol), Pd2dba3 (0.20 g, 0.21 mmol) and DPPF (0.24 g, 0.43 mmol) at 0°C, then the reaction mixture was purged with N2 forl5 min, then Zinc cyanide (2.54 g, 21.66 mmol) was added and the reaction mixture was heated at 130°C for 24h. After the consumption of starting material, reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (2X 100 mL)). The combined organic layers were washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude. Crude was purified by column chromatography (230/400 mesh, 30% ethyl acetate in pet-ether) to get desire compound XHId as white solid (1 g, 57%). LC_MS Calc. for C9H6FN3 175.17; obtained: 176.2; 1H NMR (400 MHz, DMSO-D6): δ 8.60 (s, 1H), 7.87-7.86 (m, 1H), 6.78-6.76 (m, 1H), 4.05 (s, 3H).
[000218] Step 5: Synthesis of 3,3-dibromo-6-fluoro-l-methyl-2-oxo-2,3-dihvdro- lH-pyrrolor3,2-blpyridine-7-carbonitrile (XHIe)
[000219] To a solution of XHId ( 1 g, 5.70 mmol) in tert-Butanol (30 mL) and water (30 mL) was added N-Bromo succinimide (2.03 g, 11.40 mmol) in portions. Then the reaction mixture was stirred at room temperature for lh. After the consumption of starting material, reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (2X 100 mL)). The combined organic layers were washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude. The resultant crude was washed with n-hexane to get pure compound XHIe as pale yellow solid (1 g, 57%).LC_MS Calc. for C9H4Br2FN30 348.96; obtained: 349.7;1H NMR (400 MHz, DMSO-D6): δ 8.49 (s, 1H), 3.52 (s, 3H).
[000220] Step 6: Synthesis of 6-fluoro-l-methyl-2-oxo-2,3-dihvdro-lH- Pyrrolo[3,2-b1pyridine-7-carbonitrile (XHIf)
[000221] To a solution of XHIe (1 g, 2.86 mmol) in dry THF (10 mL) and aq.sat. Ammonium chloride solution (10 mL) was added Zinc dust (3.74 g, 57.31 mmol) in portions. Then the reaction mixture was stirred at room temperature for 16 h. After the consumption of starting material, reaction mixture was filtered through celite, diluted with water (100 mL) and extracted with ethyl acetate (2X 100 mL)). The combined organic layers were washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude. The resultant crude was washed with n-hexane to get pure compound XHIf as pale yellow oil (0.5 g, 91%).LC_MS Calc. for C9H6FN30 191.17; obtained: 192.0; 1H NMR (400 MHz, DMSO-D6): δ 8.22 (s, 1H), 3.71 (s, 2H), 3.44 (s, 3H). [000222] Step 7 :Svnthesis of tert-butyl ((6-fluoro-l-methyl-2-oxo-2,3-dihvdro- lH-pyrrolor3,2-blpyridin-7-yl)methyl)carbamate (XHIg)
[000223] To a solution of XHIf (0.50 g, 2.61 mmol) in dry Methanol (10 mL) was added Boc anhydride (0.7 mL, 3.13 mmol) and Nickel chloride (0.680 g, 5.23 mmol), then the reaction mixture was cooled to 0°C, NaBH4 (0.297 g, 7.84 mmol) in portions. Then the reaction mixture was stirred at room temperature for 30 min. After the consumption of starting material, reaction mixture was filtered through celite, diluted with water (20 mL) and extracted with ethyl acetate (2X 50 mL)). The combined organic layers were washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude. Crude was purified by column chromatography (230/400 mesh, 30% ethyl acetate in pet-ether) to get desire compound XHIg as white solid (0.35 g, 50%).LC_MS Calc. for Ci4Hi8FN303 295.31 ; obtained: 296.2; 1H NMR (400 MHz, DMSO-D6): δ 11.06 (s, 2H), 8.08-8.06 (m, 1H), 7.45-7.40 (m, 1H), 4.38 (s, 2H), 3.65- 3.64 (m, 2H), 2.56 (s, 9H).
[000224] Step 8:Svnthesis of 7-(aminomethyl)-6-fluoro-l-methyl-1.3-dihvdro- 2H-pyrrolo[3,2-b]pyridin-2-one (XIII)
[000225] To a solution of XHIf (0.35 g, 1.18 mmol) in dry DCM (7 mL) was added 4M HCl in 1,4-dioxane (2 mL) at 0°C, then the reaction mixture was stirred at room temperature for 1 h. After the consumption of starting material, reaction mixture was concentrate to dryness. The crude was dissolved in dry Methanol (10 mL) neutralized by adding Amberlyst A26 resin at 0°C, filtered and evaporated under reduced pressure to get crude. The resultant crude was washed with n-hexane to get XIII as pale yellow oil (0.20 g, 90%) and the product is taken next without further characterization.
Example 1 : 6-(5-(2-(((l-Methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
Figure imgf000076_0001
[000226] To a stirred solution of 6-(5-(2-aminoethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one II (45mg, 0.161mmol) and 1 -mefhyl-2-oxoindoline- 7-carbaldehyde VI (29mg, 0.00018mol) in Dichlormethane : methanol (9: 1= 5ml), molecular sieves (O.lg) was added and stirred at 25°C for 2 hours. After confirmation of imine generation by 1H NMR, it was filtered and concentrated in vacuo. Residue obtained was dissolved in methanol, cooled to 0°C, and sodium borohydride (12mg, 0.322mmol) was added. Reaction was gradually warmed to 25 °C and stirred for 2 hours. After completion of reaction, the reaction mixture was quenched by water and concentrated in vacuo to remove methanol. Residual mixture was extracted with ethyl acetate. The combined organic layer was washed with water followed by brine, dried over sodium sulphate and concentrated in vacuo. The crude was purified by mass based preparative purification to afford title compound (3mg) as a white solid. 1H NMR (400MHz, MeOD) δ 8.52 (bs, 1H), 7.70 (d, 8.8 Hz, 1H), 7.367(dd, 1H, J = 8.8 Hz, 0.8 Hz, 1H), 7.28 (d, J = 7.6 Hz, 2H), 7.07 (t, J = 7.6 Hz, 1H), 4.82 (m, 1H), 4.64 (s, 2H), 4.32 (t, J = 9.6 Hz, 1H), 3.89-3.93 (m, 1H), 3.57 (s, 3H), 3.03-3.09 (m, 1H), 2.18 (s, 2H), 2.01-2.08 (m, 2H). LCMS = Calculated for C22H23N5O5, 437.56, Observed = 438, HPLC = 89.04% (HPLC Column: ATLANTIS dC18(250x4.6)mm,5^ Mobile Phase A: 0.1% TFA in water, Mobile Phase B: Acetonitrile.
Example 2: 6-(5-(2-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-on
Figure imgf000077_0001
[000227] To a stirred solution of 6-(5-(2-aminoethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, II (0.26g, 0.0009mol) and 6-fluoro-l-methyl-2- oxoindoline-7-carbaldehyde, VII (0.198g, O.OOlOmol) in methanol (5ml), glacial acetic acid (0.2ml) and stirred at 25 °C for 2 hours. Then, cooled to 0°C, sodium cyano borohydride resin (0.001 lmol) was added and stirred at 25°C for 2 hours. After completion of reaction, the reaction mixture was filtered to remove the resin and the filtrate was concentrated in vacuo. It was purified by reverse phase preparative HPLC to get pure compound colourless solid of 6-(5-(2-(((5-fluoro-l-methyl-2-oxoindolin-4- yl)methyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (90mg, 21.27%). XH NMR (400MHz, MeOD) δ = 7.71 - 7.73 (m, 1Η),7.38-7.42 (m, 2H), 6.95-7.00 (m, IH), 4.82-4.89 (m, IH), 4.64-4.67 (m, 4H), 4.38-4.43 (m, IH), 3.91-3.96 (m, IH), 3.54 (s, 2H), 3.61 (s, 3H), 3.42-3.49 (m, 2H), 2.23-2.29 (m, 2H). LCMS calculated for C22H22FN5O5 455.16, observed = 456.01, HPLC = 99.69% (HPLC Column: ATLANTIS dC18(250x4.6)mm,5^ Mobile Phase A: 0.1% TFA in water, Mobile Phase B: Acetonitrile.
Example 3: 6-(5-(2-(((6-fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2- oxooxazolidin-3-yl)-2H-benzo[b][l,4]oxazin-3(4H)-one
Figure imgf000077_0002
[000228] To a stirred solution of IV (40 mg, 0.144 mmol) and oxoindole VII (29mg, 0.00018mol) in dichlormethane : methanol (9: 1= 5ml), molecular sieves (O.lg) was added and stirred at 25 °C for 2 hours. After confirmation of imine generation by XH NMR, it was filtered and concentrated in vacuo. Residue obtained was dissolved in methanol, cooled to 0°C, and sodium borohydride (12mg, 0.322mmol) was added. Reaction was gradually warmed to 25 °C and stirred for 2 hours. After completion of reaction, the reaction mixture was quenched by water and concentrated in vacuo to remove methanol. Residual mixture was extracted with ethyl acetate. The combined organic layer was washed with water followed by brine, dried over sodium sulphate and concentrated in vacuo. The crude was purified by mass based preparative purification to afford title compound (13 mg) as a white solid.1H NMR (400MHz, MeOD) 7.36-7.43 (m, 1H), 7.36-7.37 (m, 1H), 6.95-7.03 (m, 3H), 4.83 - 4.86 (m, 1H), 4.66 (s, 2H), 4.57 (s, 2H), 4.24 (t, J = 8.8 Hz, 1H), 3.79-3.83 (m, 1H), 3.70 (s, 2H), 3.54 (s, 3H), 3.42-3.48 (m, 2H), 2.26-2.31 (m, 2H); LCMS calculated for C23H23FN4O5, 454.6 observed 455.
Example 4: Synthesis of 6-(5-(2-(((6-fluoro-l,3,3-trimethyl-2-oxoindolin-7- yl)methyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)- one
Figure imgf000078_0001
[000229] To a stirred solution of 6-(5-(2-aminoethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, II (0.045g, 0.00016mol) and 6-fluoro- 1,3,3- trimethyl-2-oxoindoline-7-carbaldehyde,VIII (0.035g, 0.00016mol) in Dichlormethane : methanol (9: 1= 5ml), molecular sieves (O. lg) was added and stirred at 25°C for 2 hours. After confirmation of imine generation by 1H NMR, reaction mixture was filtered through celite and filtrate was concentrated under reduced pressure. Resulting residue was dissolved in methanol, cooled to 0°C, and sodium borohydride (0.012g, 0.00033mol) was added. After the completion of addition, reaction was gradually warmed to room temperature and stirred for 1 hour. After completion of reaction, the reaction mixture was quenched by water and concentrated in vacuo to remove methanol. It was extracted with ethyl acetate and water. The separated organic layer was washed with brine, dried over sodium sulphate and concentrated in vacuo. It was purified by column chromatography on silica gel with gradient elution of 4 -5% methanol in dichloromethane to obtain white solid of 6-(5-(2-(((6-fluoro-l,3,3-trimethyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (21mg, 21.27%). 1H NMR (400MHz, MeOD) δ 7.58-7.69 (m, 1H), 7.35-7.37 (m, 1H), 7.21-7.24 (m, 1H), 4.79-4.80 (m, 1H), 4.633 (s, 2H), 4.293-4.297 (m, 1H), 4.27 (s, 2H), 3.89-3.92 (m, 1H), 3.37 (s, 3H), 2.88-2.91 (m, 2H), 2.00-2.04 (m, 2H), 1.30 (s 6H). LCMS Calculated for C24H26FN5O5, 483.50, Observed = 484.5HPLC = 92.99% (HPLC Column: ATLANTIS dC18(250x4.6) ηττη,5μ, Mobile Phase A: 0.1% TFA in water, Mobile Phase B: Acetonitrile.
Example 5: Synthesis of 5-(5-(2-(((6-Fluoro-l-methyl-2-oxoindolin-7- yl)methyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2-methylnicotinonitrile
Figure imgf000079_0001
2hr
[000230] To a stirred solution of 6-(5-(2-aminoethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one, V (43mg, 0.174mmol) and 6-fluoro-l-methyl-2- oxoindoline-7-carbaldehyde ( VII; 35mg, 0.183mmol) in Dichlormethane : methanol (9: 1= 5ml), molecular sieves (O. lg) was added and stirred at 25°C for 2 hours. After confirmation of imine generation by XH NMR, it was filtered and concentrated in vacuo. It was dissolved in methanol, cooled to 0°C, sodium borohydride (12mg, 0.348mmol) was added and stirred at 25°C for 1.5 hours. After completion of reaction, the reaction mixture was quenched by water and concentrated in vacuo to remove methanol. It was extracted with ethyl acetate and water. The separated organic layer was washed with brine, dried over sodium sulphate and concentrated in vacuo. It was purified by reverse phase preparative HPLC to get pure compound colourless solid of 5-(5-(2-(((6-fluoro-l- methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2- methylnicotinonitrile, BWC0136 ( 6mg, 8%). purification. 1H NMR (400MHz, MeOD) δ = 8.89 (d, J = 2.8 Hz, 1H), 8.36 (d, J = 2.8 Hz, 1H), 7.25-7.29 (m, 1H), 6.84-6.88 (m, 1H), 4.488-4.492 (m, 1H), 4.22-4.26 (m, 3H), 3.85-3.89 (m, 1H), 3.52-3.60 (m, 4H), 3.06-3.11 (m, 2H), 2.72 (s, 3H), 2.12-2.17 (m, 2H); LCMS = Calculated for C22H22FN5O3, 423.45, Observed = 424. HPLC = 94.92% (HPLC Column: ATLANTIS dC18(250x4.6) ηττη,5μ, Mobile Phase A: 0.1% TFA in water, Mobile Phase B: Acetonitrile).
Example 6: 6-(5-(3-(((6-fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
Figure imgf000080_0001
[000231] To a stirred solution of starting material, III (0.02 g, 0.068 mmol) in DCM/MeOH (9/1, 5 ml) was added aldehyde, VII (13.15 mg, 0.068 mmol), followed by molecular sieves (50mg), and triethyl amine (few drops). The resulting reaction mixture was stirred at RT for 3h. After confirming the formation of imine by 1H-NMR, the reaction mixture was filtered and concentrated under reduced pressure. The residue was dissolved in methanol, cooled to 0°C and sodium cyanoborohydride (5 mg, 0.136 mmol) was added. After stirring at RT for 2h, the reaction mixture was concentrated on rotary evaporator. Obtained residue was diluted with water and extracted with ethyl acetate. The crude was purified by mass based preparative purification to afford title compound (5 mg) as a pale yellow liquid. XH NMR (400MHz, MeOD) δ 8.44 (bs, 1H), 7.70 (d, J = 8.6 Hz, 1H), 7.36 (d, J = 8.6 Hz, 1H), 7.30-7.34 (m, 1H), 6.87-6.92 (m, 1H), 4.74-4.76 (m, 1H), 4.63 (s, 2H), 4.39 (s, 2H), 4.31-4.36 (m, 1H), 3.84-3.88 (m, 1H), 3.67-3.70 (m, 2H), 3.54-3.58 9m, 5H), 3.08-3.10 (m, 2H), 1.84-1.90 (m, 4H); LCMS calculated for C23H24FN5O5, 469.47 Observed = 470.0; HPLC = 87.33% (Zorbax Eclipse plus C18 RRHD(50X2.1) ηττη,1.8μ; Mobile phase A:0.1%TFA in WaterB:Acetonitrile)
Example 7: 6-(5-(2-(((l-Ethyl-6-fluoro-2-oxoindolin-7-yl)methyl)amino)ethyl)-2- oxooxazolidin-3- l)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
Figure imgf000081_0001
[000232] To a stirred solution of II (30 mg, 0.108 mmol) and oxoindole IX (23mg, 0.113mol) in dichlorme thane: methanol (9: 1= 5ml), molecular sieves (O.lg) was added and stirred at 25°C for 2 hours. After confirmation of imine generation by XH NMR, it was filtered and concentrated in vacuo. Residue obtained was dissolved in methanol, cooled to 0°C, and sodium borohydride (8mg, 0.2162mmol) was added. Reaction was gradually warmed to 25 °C and stirred for 2 hours. After completion of reaction, the reaction mixture was quenched by water and concentrated in vacuo to remove methanol. Residual mixture was extracted with ethyl acetate. The combined organic layer was washed with water followed by brine, dried over sodium sulphate and concentrated in vacuo. The crude was purified by mass based preparative purification to afford title compound, (7.2 mg) as a white solid.1H NMR (400MHz, MeOD) 8.35 (bs, 1H), 7.69 (d, J = 8.6 Hz, 1H), 7.36 (d, J = 8.6Hz, 1H), 7.33-7.30 (m, 1H), 6.92-6.87 (m, 1H), 4.84-4.82 (m, 1H), 4.33 (t, J = 8.8 Hz, 1H), 4.23 (s, 2H), 4.02-4.08 (m, 2H), 3.89-3.93 (m, 1H), 3.53-3.70 (m, 2H), 3.13-3.16 (m, 2H), 2.68 (s, 1H), 2.12-2.14 (d, 2H), 1.27-1.31 (m, 2H); LCMS calculated for C24H25FN4O5, 469.47 observed 470.
Example 8: 6-(5-(3-(((l-Ethyl-6-fluoro-2-oxoindolin-7-yl)methyl)amino)propyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
Figure imgf000082_0001
NaBH4, MeOH, 3h Example s
[000233] To a stirred solution of III (20 mg, 0.0684 mmol) and oxoindole IX (15mg, 0.0724mol) in dichlormethane: methanol (9: 1= 5ml), molecular sieves (O.lg) was added and stirred at 25 °C for 2 hours. After confirmation of imine generation by 1H NMR, it was filtered and concentrated in vacuo. Residue obtained was dissolved in methanol, cooled to 0°C, and sodium borohydride (8mg, 0.2162 mmol) was added. Reaction was gradually warmed to 25°C and stirred for 2 hours. After completion of reaction, the reaction mixture was quenched by water and concentrated in vacuo to remove methanol. Residual mixture was extracted with ethyl acetate. The combined organic layer was washed with water followed by brine, dried over sodium sulphate and concentrated in vacuo. The crude was purified by mass based preparative purification to afford title compound, (3.3 mg) as a white solid.1H NMR (400MHz, MeOD) 8.28 (bs, 1H), 7.71 (d, J = 8.8 Hz, 1H), 736-7.41 (m, 2H), 6.93-6.98 (m, 1H), 4.75-4.78 (m, 1H), 4.64 (s, 2H), 4.40 (s, 2H), 4.33-4.37 (m, 1H), 3.98-4.03 (m, 2H), 3.85-3.89 (m, 1H), 3.56- 3.59 (m, 1H), 3.22 (t, J = 6.8 Hz, 2H); LCMS calculated for C25H27FN4O5, 483.5 observed 484.
Example 9: Chiral 6-(5-(3-(((6-fluoro-l-methyl-2-oxoindolin-7- yl)methyl)amino)propyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)- one (Enantiomer 1) Example 10: Chiral 6-(5-(3-(((6-fluoro-l-methyl-2-oxoindolin-7- yl)methyl)amino)propyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-
Figure imgf000083_0001
[000234] To a stirred solution of III (4.95 g, 0.025 mol) in MeOH (370 mL), glacial acetic acid (10 mL) was added was added aldehyde, VII (4.825, 0.025 mol) and stirred at 25°C for 15 min. The solution was then, cooled to 0°C followed by addition of sodium cyanoborohydride resin (20 g, 0.050 mol) and stirred at 25°C for 6 h. After completion of reaction, the reaction mixture was filtered to remove the resin and the filtrate was concentrated in vacuo. It was purified by flash column chromatography on silica gel (230-400 mesh, 8% MeOH in DCM to get the racemic Example 6 as a pale yellow solid (2.52 g, 21.48%). 1H NMR (400 MHz, CD3OD). δ 7.71 (d, = 8.40 Hz, 1H), 7.37-7.43 (m, 2H), 7.0-6.94 (m, 1H), 4.87 (m, 1H), 4.79-4.7 (m, 1H), 4.7-4.6 (m, 4H), 4.34-4.39 (m, 1H), 3.9-3.8 (m, 1H), 3.61 (s, 2H), 3.52 (s, 3H), 1.91-1.97 (m, 4H). LC_MS calculated for C23H24FN505 469.47, observed = 470.01. HPLC = 96.84% (HPLC Column: ATLANTIS dCl 8(250x4.6)™,5μ, Mobile Phase A: 0.1% TFA in water, Mobile Phase B: Acetonitrile.
[000235] The Part of racemic compound (Example 6, 2gm) was separated into its enantiomers (Example 9 and Example 10) by Chiral supercritical fluid chromatography
(SFC) using the following SCF conditions
Column : Lux C3 (250*30) mm
Mobile phase : C02: 20mm Ammonia in Methanol (65:35)
Total flow : lOOg/min Injection volume : 0.5ml
Back Pressure : lOObar
Wave length : 212 nm
Cycle time : 11.0 min
[000236] Two pure fractions were collected and their solvents were evaporated under vacuo afforded enantiomerl and 2 as off white solid.
Enantiomer 1 (Example 9): 0.750 gm, Chiral HPLC retention time: 5.74 min.
Enantiomer 2 (Example 10): 0.650 gm, Chiral HPLC retention time: 7.38 min.
Example 11: 6-(5-(2-((2-(6-fluoro-l-methyl-2-oxoindolin-7-yl)ethyl)amino)ethyl)-2- oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
Figure imgf000084_0001
Example 11 [000237] To a stirred solution of amine II (0.13 g, 0.467 mmol) and aldehyde X (0.125 g, 0.603 mmol) in MeOH (4 mL), glacial acetic acid (0.2 mL) was added and stirred at 25°C for 15 min. The solution was then, cooled to 0°C followed by addition of sodium cyanoborohydride resin (0.125 g) and stirred at 25 °C for 2 h. After completion of reaction, the reaction mixture was filtered to remove the resin and the filtrate was concentrated in vacuo. It was purified by reverse phase preparative HPLC to get Examplell as a pale yellow solid (21 mg). 1H NMR (400 MHz, DMSO-J6). δ 11.22 (s, 1H), 8.77 (brs, 2H), 7.61 (d, 1H, = 8.40 Hz), 7.45 (d, 1H = 8.40 Hz), 7.23-7.18 (m, 1H), 6.9-6.84 (m, 1H), 4.81-4.77 (m, 1H), 4.77 (s, 2H), 4.26 (t, 1H, = 6 Hz), 3.60 (s, 2H), 3.42 (s, 3H), 3.23-3.15 (m, 6H), 2.15-2.10 (m, 2H). LC_MS calculated for C23H24FN5O5 469.47; observed 470.0. Example 12: 6-(5-(3-(((6-fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2- oxooxazolidin-3-yl)-2H-benzo[b][l,4]oxazin-3(4H)-one
Figure imgf000085_0001
Example 12
[000238] To a stirred solution of XI (0.1 g, 0.34 mmol) and 6-fluoro-l-methyl-2- oxoindoline-7-carbaldehyde, VII (0.066 g, 0.34 mmol) in methanol (5 ml), glacial acetic acid (0.2 ml) and stirred at 25°C for 15 min. Then, cooled to 0°C, sodium cyanoborohydride resin (0.295 g, 0.68 mmol) was added and stirred at 25°C for 6 h. After completion of reaction, the reaction mixture was filtered to remove the resin and the filtrate was concentrated under reduced pressure. It was purified by reverse phase preparative HPLC to obtain example 12 (0.04 g, 25%). 1H NMR (400 MHz, DMSO-J6): δ 10.76 (s, 1H), 8.79 (brs, 2H), 7.4-7.3 (m, 2H), 7.0-6.9 (m, 3H), 4.75-4.7 (m, 1H), 4.55 (s, 2H), 4.5-4.4 (m, 2H), 3.7-3.63 (m, 3H), 3.41 (s, 3H), 3.2-3.16 (m, 2H), 1.85-1.7 (m, 4H). LCMS calculated for C24H25FN4O5 468.49, observed 469.0.
Example 13: Chiral 6-(5-(2-((2-(6-fluoro-l-methyl-2-oxoindolin-7- yl)ethyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomerl)
Example 14: Chiral 6-(5-(2-((2-(6-fluoro-l-methyl-2-oxoindolin-7- yl)ethyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomer2)
Figure imgf000086_0001
[000239] To a mixture of X (lg, 4.82 mmol) and amine II (1.60 g, 5.79 mmol) in dry MeOH (20 mL) and DCM (20 mL) was added AcOH (1 mL) and allowed to stir for 16 h. To this was added Sodium cyanoborohydride resin (3.97 g, 9.65 mmol) and stirred for 15 min. The reaction mixture was filtered and concentrated. Crude was purified by column chromatography (230/400 mesh, 7% DCM in MeOH) to get the racemic mixture of compound as an Off-white solid (Example 11, 0.5 g, 26 %)
[000240] The Part of racemic compound (Example 11, 0.5gm) was separated into its enantiomers (Example 13 and Example 14) by Chiral supercritical fluid chromatography (SFC) using the following SCF conditions
Column : YMC Cellulose
Mobile phase : C02: 20mm Ammonia in Methanol (65:35)
flow Rate : 4 mL/min
Injection volume : 0.5ml
Back Pressure : lOObar
Wave length : 212 nm
Cycle time : 11.0 min
The separated pure fractions were collected and their solvents were evaporated under vacuo afforded enantiomerl and 2 as off white solid.
Enantiomer 1 (Example 13): 0.200 gm, Chiral HPLC retention time: 3.7 min
LC_MS Calc. for Calc. for C23H24FN5O5, 469.47; Obs 470.0; [M++H] ; HPLC Purity = 95.07% (HPLC Column: Atlantis dC18 (250*4.6) mm 5μπι, Mobile Phase A: 0.1% TFA in water, Mobile Phase B: Acetonitrile.);1!! NMR (400 MHz, DMSO-D6): δ 11.23 (s, 1H), 7.59 (d, 1H, J=8.4 Hz), 7.44 (d, 1H, J=8.8 Hz), 7.18-7.15 (m, 1H), 6.86-6.82 (m, 1H), 4.79-4.76 (m, 1H), 4.62 (s, 2H), 4.26-4.22 (m, 1H), 3.79-3.75 (m, 1H), 3.53 (s, 2H), 3.12-3.10 (m, 2H), 2.96 (s, br, 4H), 2.03 (s, br, 2H).
[000241] Enantiomer 2 (Example 14): 0.200 gm Chiral HPLC retention time: 4.5 min LC_MS Calc. for Calc. for C23H24FN5O5, 469.47; Obs 470.0; [M++H]; HPLC Purity = 97.02% (HPLC Column: Atlantis dC18 (250*4.6) mm 5μπι, Mobile Phase A: 0.1% TFA in water, Mobile Phase B: Acetonitrile.^H NMR (400 MHz, DMSO-D6): δ 11.20 (s, 1H), 7.59 (d, 1H, J=8.8 Hz), 7.43 (d, 1H, J=8.8 Hz), 7.13-7.09 (m, 1H), 6.82-6.78 (m, 1H), 4.76-4.73 (m, 1H), 4.61 (s, 2H), 4.23-4.18 (m, 1H), 3.76-3.72 (m, 1H), 3.50 (s, 2H), 3.47 (s, 3H), 2.99-2.96 (m, 2H), 2.72-2.65 (m, 4H), 1.90-1.83 (m, 2H).
Example 15: 6-(5-(2-((2-(6-fluoro-l-methyl-2-oxoindolin-7-yl)-2-hydroxyethyl) amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
Figure imgf000087_0001
Examplel5 ( dlastereomerl)
a&b= configuration unknown
Stepl: 6-fluoro-l-methyl-7-(oxiran-2-yl)indolin-2-one( 15a) [000242] To a solution of Vlld (2 g, 10.45 mmol) in DCM (60 mL) and 5% NaHC03 aqueous solution (20 mL) was added meta chloroperbenzoic acid (5.41 g 31.38 mmol) at 0°C.The reaction mass was stirred at room temperature for 16 h. After completion of the reaction, added 5% NaHC03 aqueous solution (50 mL) and extracted with ethyl acetate (2X 100 mL). The combined organic layers were washed with brine, dried over sodium sulphate and evaporated under reduced pressure to get crude to afford compound 15a (1.3 g, 60 % yield) as off-white solid.LC_MS Calc. for Calc. for CiiH10FNO2, 207.20; Obs 208.1 [M++H]1H NMR (400 MHz, DMSO-D6): δ 7.90 (s, 1H), 7.72-7.52 (m, 1H), 7.26-7.21 (m, 1H), 6.85-6.78 (m, 1H), 4.25 (s, 1H), 3.54 (s, 2H), 3.33 (s, 3H). Step2: 6-(5-(2-((2-(6-fluoro-l-methyl-2-oxoindolin-7-yl)-2-hydroxyethyl) amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
[000243] To a mixture of 15a (1.3 g, 6.27 mmol) and amine II (2.09 g, 7.52 mmol) in ethanol (52 mL) and water (13mL) was heated at 100°C in a sealed tube for 24h. After completion, the reaction mixture was concentrated to get the crude product. The crude was purified by reverse phase grace column chromatography to afford desired product (Example 15) as an off white solid (0.3g) and the only one diastereomer was obtained from the epoxide 15a opening reaction. LC_MS Calc. for Calc. for C23H24FN5O6, 485.47; Obs 486.2; [M++H]; HPLC Purity = 88.67% (HPLC Column: Atlantis dC18 (250*4.6) mm 5μπι, Mobile Phase A: 0.1% TFA in water, Mobile Phase B: Acetonitrile.); 1H NMR (400 MHz, DMSO-D6): δ 11.22 (s, 1H), 8.78 (s, 1H), 7.59 (d, 1H, J=8.4 Hz), 7.45 (d, 1H, J=8.8 Hz), 7.25 (s, br, 1H), 6.31 (s, 1H), 4.79-4.78 (m, 1H), 4.62 (s, 1H), 4.28-4.23 (m, 2H), 3.61 (s, 3H), 3.46 (s, 3H), 3.17 (s, 2H), 2.16-2.09 (m, 2H). Example 16: 6-(5-(2-((2-(5-Fluoro-3-methyl-2-oxo-2,3-dihydrobenzo[d]oxazol-4- yl)ethyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
Figure imgf000088_0001
Example 16
[000244] To a mixture of XII (0.12 g, 0.57 mmol) and amine (II, 0.19 g, 0.68 mmol) in dry methanol (10 mL) and dichloromethane (10 mL) was added AcOH (0.10 mL) and allowed to stir for 16 hours at room temperature. To this was added MP- cyanoborohydride resin (0.46 g, 1.14 mmol) and stirred for another 15 minutes at room temperature. The reaction mixture was filtered and concentrated to remove methanol. The residue was diluted with dichloromethane (50 mL) and washed with 10 % aqueous NaHC03. The organic layer was dried over Na2S04 and concentrated to get the crude product. The crude was purified by preparative HPLC to afford Examplel6 as an off- white solid (0.026 g, 10 %). LC_MS: Calc. for C22H22FN5O6 471.45; Obs. 472.0 [M+H] +; 1H NMR (400 MHz, DMSO-D6): δ 11.22 (brs, 1H), 8.67 (brs, 2H), 7.60 (d, = 8.4 Hz, 1H), 7.45 (d, = 8.8 Hz, 1H), 7.37 - 7.32 (m, 2H), 4.80 - 4.78 (m, 1H), 4.63 (s, 2H), 4.28 - 4.24 (m, 1H), 3.79 - 3.75 (m, 1H), 3.31 (s, 3H), 3.20 - 3.14 (m, 4H), 2.98 - 2.96 (m, 2H), 2.12 - 2.10 (m, 2H). Example 17: 6-(5-(3-(((6-Fluoro-l-methyl-2-oxo-2,3-dihydro-lH-pyrrolo[3,2- b]pyridin-7-yl)methyl)amino)propyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2- b][l,4]oxazin-3(4H)-one
Figure imgf000089_0001
[000245] Stepl: To a stirred solution of Illg (1 g, 3.40 mmol) in DCM (20 mL), cooled to 0°C, was added Dess-martin periodinane (2.16 g, 5.11 mmmol) and allowed to stir at rt for 2h. The reaction mixture was carefully quenched with NaHC03 solution (10 mL). The organic layer was separated and washed with sodiumthiosulphate solution (2x25 mL) and over Na2S04, filtered and concentrated in vacuum to obtain colorless gummy solid of 17a. This was used in the next step without further purification (0.5 g, crude). LC_MS: Calculated for Ci3Hi3N305, 291.26, Observed = 292.0
[000246] Step2: To a mixture of XIII (0.20g, 1.02 mmol) and aldehyde 17a (0.298 g, 1.02 mmol) in dry MeOH (6 mL) and DCM (6 mL) was added AcOH (0.20 mL) and allowed to stir for 16 h. To this was added Sodium cyanoborohydride resin (0.843 g, 2.04 mmol) and stirred for 15 min. The reaction mixture was filtered and concentrated. Crude was purified by column chromatography (230/400 mesh, 7% DCM in MeOH) to get desire compound Examplel7 as white solid (40 mg, 12 %). LC_MS Calc. for Calc. for C22H23FN6O5, 470.46; Obs 471.2; [M++H]; HPLC Purity = 94.20% (HPLC Column: Phenomenex Gemini-N x C18 (150*4.6) mm, 3μπι, Mobile Phase A: lOmM NH40Ac in water, Mobile Phase B: Acetonitrile.);1!! NMR (400 MHz, DMSO-D6): δ 11.21 (s, 1H), 8.96 (s, br, 2H), 8.23 (s, 1H), 7.59 (d, 1H, J=8.8 Hz), 7.44 (d, 1H, J=8.4 Hz), 7.24-6.98 (m, 1H), 4.76 (s, br, 1H), 4.62 (s, 2H), 4.45 (s, 2H), 4.25-4.20 (m, 1H), 3.76-3.70 (m, 3H), 3.40 (s, 3H), 3.17-3.13 (m, 2H), 1.81-1.75 (m, 4H).
Example 18
Biological Activity
1. Antibacterial activity:
[000247] The compounds of Formula I are of interest due to their potent antibacterial effects. The ability of the invention compounds disclosed herein to achieve an antibacterial effect may be evaluated with regard to their ability to inhibit the growth of bacterial species like Escherichia coli ATCC 25922, Staphylococcus aureus ATCC 29213, Klebsiella pneumoniae ATCC 13883, Acinetobacter baumannii ATCC 19606, Pseudomonas aurigenosa ATCC 27853, Enterococcus faecalis ATCC 29212 and Enterococcus faecalis ATCC 29212 using an assay based on the following Minimum Inhibitory Concentration (MIC) protocol:
[000248] The test bacteria are grown in Luria Bertani Broth (HIMEDIA M1245), 25 grams of the powder is dissolved in 1000 ml distilled water and sterilized by autoclaving at 15 lbs pressure (121°C) for 20 minutes. The medium sterility is checked by incubating at 37°C for a period of 48 h.
[000249] Bacterial cultures that are stored as glycerol stocks at -80°C are sub cultured on LB agar plates to obtain isolated colonies. A single colony of each strain is cultured in LB broth. The cultures are incubated at 37° C, 200 rpm till they reach an optical density (OD at 600nm) of 0.8 to 1. This log phase culture is diluted in LB broth to a cell number of 5-8* 105 CFU/mL to be used as inoculum for MIC experiments. Test compounds are dissolved in dimethyl sulfoxide (DMSO) to a stock concentration of 4 mg/ml. A twofold dilution series of this DMSO stock is prepared in a 96 well V bottom microtitre plate from rows A to H. A 3 μL· volume of these dilutions are transferred to a 96-well flat bottom microtitre assay plate. Controls to monitor the effects of DMSO and media sterility are included. Each well is inoculated with 150 μΕ of the above diluted culture. The plates are incubated at 37°C overnight in a humidified incubator. The following morning, the plates are read using a Spectrophotometer at 600 nM wavelength. Minimum Inhibitory Concentration (MIC) is defined as the lowest drug concentration containing well which shows no turbidity. The antibacterial activity (MIC) determined against representative Gram positive (S. aureus, E.faecalis) and Gram negative (E.coli, P.aurigenosa, K. pneumoniae and A.baumannii) pathogen were reported Table 1. The exemplified compounds belonging to Formula I demonstrated potent antibacterial activity both Gram positive and Gram negative pathogens.
Figure imgf000091_0001
12 0.25 0.5 0.25 2 0.5 0.13
13 0.03 0.25 0.06 0.5 0.125 0.06
14 0.03 0.25 0.03 0.5 0.125 0.03
15 1 2 1 2 2 4
16 4 4 0.5 4 1 0.5
17 1 4 1 >16 2 0.5
Ciprofloxacin 0.4 0.4 0.012 0.1 0.05 0.4
2. Enzyme inhibition assay: Determination IC50 against E.coli Gyrase supercoiling
[000250] The compounds belonging to Formula I or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in killing or inhibiting the growth of Gram-positive and Gram-negative bacteria through inhibition of bacterial Type II topoisomerases namely, DNA gyrase and Topo IV.
[000251] The present invention also provides evidence for treating infection caused both Gram positive and Gram negative bacteria through the inhibition of bacterial topoisomerases using E. coli DNA gyrase enzyme.
Procedure for E. coli DNA gyrase supercoiling assay
[000252] E. coli gyrase supercoiling and its inhibition was assayed using a kit procured from Inpiralis (K0001) and the protocol (PMID: 2172086) was adapted with necessary modifications. The compounds to be tested are incubated for 10 minutes with 2.5 nM of E. coli DNA gyrase in a 30 μΐ volume reaction and 3.2% DMSO. The reactions are then started with the addition of 60 ng relaxed pBR322 plasmid DNA and continued for 45min at 37 °C. The reaction mixture contains 35 mM Tris.HCl (pH 7.5), 24 mM KC1, 1.8 mM spermidine, 4 mM MgCl2, 2 mM DTT, 6.5% (w/v) glycerol, 0.1 mg/mL BSA, and 1 mM ATP. The reaction is then stopped by addition of 0.75
Figure imgf000092_0001
of Proteinase K (20 mg/mL) and 3 μΤ of 2% SDS and further incubated at 37 °C for 30min. This was followed by the addition of 4 uL of STEB (40 % (w/v) sucrose, 100 mM Tris-HCl pH8, 1 mM EDTA, 0.5 mg/ml Bromophenol Blue) and the supercoiled/relaxed forms of plasmid DNA were separated by agarose gel electrophoresis. The 1 % agarose gels are run for 3 h at 4V/cm in 1 x TAE (40 mM Tris, 20 mM Acetic acid, 1 mM EDTA). To visualize the DNA the gels are stained for 10 min with 0.7 μg/mL ethidium bromide and excess dye is removed by several washes with water. IC50S are determined by quantifying the supercoiled and relaxed DNA in each of the reactions from a gel image by a densitometric method using the Quantity One Software (Bio-rad).
Procedure for E. coli topoisomerase IV decatenation assay
[000253] E. coli topoisomerase IV decatenation activity and its inhibition was assayed using a kit procured from Inpiralis (D4002) and the kit protocol was adapted with necessary modifications similar to the gyrase supercoiling assays. The compounds to be tested were incubated for 10 minutes with 5 nM of E. coli topoisomerase IV in a 30 μΐ volume reaction and 3.2% DMSO. The reactions were started with the addition of 60 ng of kDNA and continued for 40min at 37 °C. The final reaction mixture contains 40 mM Tris.HCl (pH 7.6), 100 mM potassium glutamate, 10 mM magnesium acetate, 10 mM DTT, 1 mM ATP, and 50 μg/ml albumin. The reactions were stopped by addition of 0.75 μΤ of Proteinase K (20 mg/mL) and 3 μΤ of 2% SDS and further incubated at 37 °C for 30min. This was followed by the addition of 4 μΤ of STEB (40 % (w/v) sucrose, 100 mM Tris-HCl pH8, 1 mM EDTA, 0.5 mg/ml Bromophenol Blue) and the kDNA /minicircles forms were separated by agarose gel electrophoresis. The 1 % agarose gels were run for 3 h at 4V/cm in IX TAE (40 mM Tris, 20 mM Acetic acid, 1 mM EDTA). To visualize the DNA, the gels were stained for 10 min with 0.7 μg/mL ethidium bromide and excess dye was removed by several washes with water. IC50S were determined by quantifying the Kinetoplast DNA band inside the gel well and decatenated minicircles that migrate into the gel in each of the reactions from a gel image by a densitometric method using the Quantity One Software (Bio-rad).
[000254] Representing examples belonging to Formula I were evaluated against of E. coli DNA gyrase and Topo IV enzyme using gel based supercoiling assay for gyrase inhibition and decatenation assay for Topo IV inhibition. The results of bacterial Type II Topo isomerases (Gyrase and Topo IV) were presented in the table 2. The results presented in the Table 2 indicates that compounds belonging to Formula I exerts its' antibacterial activity through inhibition bacterial type II topoisomerase activity and signifies the dual mode of inhibition for observed antibacterial activity of the compounds.
Figure imgf000094_0001
ND-Not determined, NA-Not applicable
MIC50 and MI90 studies using clinical strains
[000255] To test if the compounds from the series are able to retain the antibacterial activity against clinical strains of bacteria, antibacterial susceptibility studies (MIC50 and MIC90 determination) were carried for a representative compound (Examplel4) from the series using clinical strains of four gram negative bacterial species (E.coli, P.aurigenosa, K. pneumoniae and A.baumannii) according the standard CLSI guidelines and the results are presented in Figure 1 and Table 3. The standard drugs ciprofloxacin and meropenem were as positive control in the study.
Table 3: Results of MIC50 and MIC90 studies
Figure imgf000095_0001
Figure imgf000096_0001
[000256] The results presented in the Table 3 and Figure 1 indicates that compounds belonging to formula I works against both drug sensitive and resistant clinical strains of gram negative bacteria and retain the antibacterial activity. The MIC 90 values of Examplel4 are 0.5 to 4 μg/ml range for 4 bacterial species and it is found to be superior in comparison to standard drugs used in the study.
Mouse in-vivo pharmacokinetic profile of Example 13
[000257] To assess the Oral and Intravenous pharmacokinetic profile of Examplel3, pharmacokinetics (PK) of Example 13 was characterized in BALB/c mice following single intravenous (i.v.) dose of 2 mg/kg and single escalating oral doses of 2, 10, 50 and 100 mg/kg, respectively. This study was performed following all ethical practices as laid down in the guidelines for animal care (Registration number No. 1852/PO/Rc/S/16/CPCSEA). The study was approved by the Institutional Animals Ethics Committee (IAEC) of the test facility. For IVPK in BALB/c mice, the formulation used was DMSO: PEG400: Ethanol: Water (10:20: 10:60 v/v) and Example 13 administered as solution. For POPK, the formulation used was 0.1 % Tween80 in 0.25% CMC and Examplel3 administered as suspension through an oral gavage. The plasma (mice) samples were analyzed by LC-MS/MS. PK parameters were estimated by non-compartmental analysis in Phoenix WinNonlin 6.4 The results of the per-oral and intravenous PK study of Examplel3 are presented in Table 4 and 5
Table 4: NCA PK parameters of Examplel3 in mice [i.v., 2 mg/kg]
Figure imgf000096_0002
Parameter Estimate
AUC (% Extrap) 0.002
CL (ml/h/kg) 5688
Vss (ml/kg) 13339
Table 5: NCA PK parameters of example 13 in mice following single oral doses
Figure imgf000097_0001
[000258] Following a single i.v. dose of 2.0 mg/kg in BALB/c mice, Example 13 showed high systemic clearance [5.7 L/h/kg], a very high volume of distribution at steady state [ 13.3 L/kg], and a moderate ti/2 of 1.7 h. Following single oral doses of 2, 10, 50 and 100 mg/kg in BALB/c mice, the AUCo- ranged between 0.07 to 4.82 μg.h/ml, Cmax ranged between 0.004 to 0.072 μg/ml and t ranged between 1.4 to 5.6 h. The oral bioavailability was moderate and ranged between 19-27 %.
The AUCo-oo increased linearly [r2=0.9914] with dose [AUC/Dose range 0.036 to 0.048]. Cmax also increased linearly with dose between 10 and 100 mg/kg [Cmax/Dose range 0.011 to 0.024].
In vivo efficacy in the mouse E.coli thigh Model:
[000259] To assess the Oral efficacy, Examplel3 was tested in BALB/c mice thigh infection model following per oral doses of 100 mg/kg once, 50 mg/kg once and twice, 33 mg/kg thrice and 25 mg/kg four times over a 24 hour period post infection. This study was performed following all ethical practices as laid down in the guidelines for animal care (Registration number No. 1852/PO/Rc/S/16/CPCSEA). The study was approved by the Institutional Animals Ethics Committee (IAEC) of the test facility. The formulation used was 0.1% Tween80 in 0.25% CMC and Examplel3 administered as suspension through an oral gavage. Neutropenic mice were infected with [~1 x 106CFU/mouse] E.coli [ATCC 25922] by intramuscular route. Two hours post infection, mice were treated orally with total doses of 100, 50, 50, 33 and 25 mg/kg of Examplel3 as one, two, three and four equally divided doses over a 24 h period. Animals were sacrificed 24 hr post infection and thigh tissues were harvested to enumerate the bacterial CFU count. Thigh muscles were placed in 1 ml of sterile LB broth and homogenized using a homogenizer (Omni Tip (220 volt hand held)). Serial ten-fold dilutions of the thigh homogenates were prepared in sterile LB broth, 0.05 ml of four dilutions for each thigh was plated onto Lactose agar plates. Bacterial colonies were enumerated on each plate following -20 hours of incubation at 37°C to determine the density of bacteria per thigh. The Mean ± SD bacterial density Log 10 CFU/thigh was estimated for each group. The means of the treated groups were compared with the corresponding vehicle groups using a one way ANOVA at a 95% confidence levels. A P<0.05 was considered significant. The mean Log 10 CFU/thigh reduction at 24 h post treatment relative to the 2 h infection control was estimated for each group. The results of the efficacy study is presented in Figure 2. [000260] Examplel3 showed significant efficacy when dosed with 100 mg kg single, two, three and four equally divided doses, whereas single dose of 50mg/kg was found to be ineffective.
ADVANTAGES
[000261] The above mentioned implementation examples as described on this subject matter and its equivalent thereof have many advantages, including those which are described.
[000262] The compounds of the present disclosure show high antibacterial activity against various pathogens including Gram-positive and Gram-negative bacteria through the inhibition of bacterial topoisomerase via a novel mechanism.
[000263] Although the subject matter has been described in considerable detail with reference to certain embodiments thereof, other embodiments are possible. As such, the spirit and scope of the invention should not be limited to the description of the embodiments contained herein.

Claims

I/We Claim
1. A compound of Formula I
Figure imgf000100_0001
Formula I
or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, wherein
Ri is selected from the group consisting of hydrogen, Ci-6 alkyl, C2-6 alkenyl, and C3-C6 cycloalkyl, each of which is unsubstituted or substituted with 1 to 3 groups independently selected from halogen, amino, hydroxy, oxetane, -OCi-6 alkyl, C3-6 cycloalkylamino, Ci-6 alkylamino, or di(Ci-6 alkyl)amino;
R2 is selected from the group consisting of hydrogen, hydroxyl, and amino;
X is -NH, -NCi-6 alkyl, O, or CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen, halogen, and
Ci-6 alkyl;
Yi is N or CR5;
R5 is selected from the group consisting of hydrogen, halogen, cyano, -OCi-6 alkyl, -OQ- 6 haloalkyl, and Ci-6 alkyl;
Y2 is N or CH;
Y3 is N or CR6; R6 is selected from the group consisting of hydrogen, halogen, cyano, Ci_6 alkyl, -OC alkyl, and -OCi-6 haloalkyl;
Zi is NH when Z2 is Ci_6 alkylene; or Zi is Ci_6 alkylene when Z2 is NH;
n is 1 or 2;
W is CH2 wherein dotted line (— ) represents no bond; W is O when dotted line (— represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000101_0001
2. A compound as claimed in claim 1, or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof
wherein
Ri is selected from the group consisting of hydrogen, Ci-6 alkyl, C2-6 alkenyl, and C3-6 cycloalkyl, wherein alkyl is optionally substituted with 1 to 3 groups independently selected from fluorine, hydroxy, amino, oxetane, -OCi alkyl, C3-6 cycloalkylamino, d-2 alkylamino, or di(Ci-2 alkyl)amino;
R2 is selected from the group consisting of hydrogen, hydroxyl, and amino;
X is -NH, -NCi alkyl, O, or CR3R4;
R3 and R4 are independently selected from the group consisting of hydrogen, fluorine, and Ci alkyl;
Yi is N or CR5; R5 is selected from the group consisting of hydrogen, fluorine, chlorine, cyano, -OCH3 OCF3, OCHF2, and Ci alkyl;
Y2 is N or CH;
Y3 is N or CR6;
R6 is selected from the group consisting of hydrogen, fluorine, cyano, -OCH3, - OCHF and -OCF3;
Zi is NH when Z2 is CH2; or Zi is CH2, CH2CH2 or CH(OH)CH2 when Z2 is NH;
n is 1 or 2
W is O when dotted line (— ) represents either a bond or no bond;
Ring A is selected from the group consisting of
Figure imgf000102_0001
3. The compound of formula I as claimed in claim 1 or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, which is selected from a group consisting of:
Figure imgf000102_0002
6-(5-(2-(((l-Methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H- pyrido[3 ,2-b] [ 1 ,4]oxazin-3(4H)-one
Figure imgf000103_0001
6-(5-(2-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-pyrido [3 ,2-b] [ 1 ,4]oxazin- (4H)-one,
Figure imgf000103_0002
6-(5-(2-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-benzo[b] [1,4] oxazin-3 (4H)-one,
Figure imgf000103_0003
6-(5-(2-(((6-Fluoro-l,3,3-trimethyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2- oxooxazolidin-3-yl)-2H-pyrido -b][l,4]oxazin-3(4H)-one,
Figure imgf000103_0004
5-(5-(2-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2-methylnicotinonitrile,
Figure imgf000104_0001
6-(5-(3-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2-oxooxazolidin- 3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one,
Figure imgf000104_0002
6-(5-(2-((( 1 -Ethyl-6-fluoro-2-oxoindolin-7-yl)methyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-pyrido[3,2-b][l,4]ox
Figure imgf000104_0003
6-(5-(3-(((l-Ethyl-6-fluoro-2-oxoindolin-7-yl)methyl)amino)propyl)-2-oxooxazolidin-3- yl)-2H-pyrido[3,2-b][l,4]oxazin-3 4H)-one,
Figure imgf000104_0004
6-(5-(3-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2-oxooxazolidin- 3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomerl),
Figure imgf000105_0001
Eriantfomerl
6-(5-(3-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2-oxooxazolidin- 3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomer2),
Figure imgf000105_0002
6-(5-(2-((2-(6-Fluoro-l-methyl-2-oxoindolin-7-yl)ethyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-pyrido [3 ,2-b] [ 1 ,4]oxazin-3 (4H)-one,
Figure imgf000105_0003
6-(5-(3-(((6-Fluoro-l-methyl-2-oxoindolin-7-yl)methyl)amino)propyl)-2-oxooxazolidin- 3-yl)-2H-benzo[b] [ 1 ,4]oxazin-3(4H)-one,
Figure imgf000106_0001
a= Absolute configuration unkown
6-(5-(2-((2-(6-Fluoro-l-methyl-2-oxoindolin-7-yl)ethyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomerl),
Figure imgf000106_0002
a= Absolute configuration unkown
6-(5-(2-((2-(6-Fluoro-l-methyl-2-oxoindolin-7-yl)ethyl)amino)ethyl)-2-oxooxazolidin-3- yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one (Enantiomer2),
Figure imgf000106_0003
a&b= configuration unknown
6-(5-(2-((2-(6-Fluoro-l-methyl-2-oxoindolin-7-yl)-2-hydroxyethyl) amino)ethyl)-2- oxooxazolidin-3-yl)-2H- rido[3,2-b][l,4]oxazin-3(4H)-one,
Figure imgf000106_0004
6-(5-(2-((2-(5-Fluoro-3-methyl-2-oxo-2,3-dihydrobenzo[d]oxazol-4- yl)ethyl)amino)ethyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin- and
Figure imgf000107_0001
6-(5-(3-(((6-Fluoro-l-methyl-2-oxo-2,3-dihydro-lH-pyrrolo[3,2-b]pyridin-7- yl)methyl)amino)propyl)-2-oxooxazolidin-3-yl)-2H-pyrido[3,2-b][l,4]oxazin-3(4H)-one
4. A process of preparation of compound of Formula I as claimed in any of claims 1 to 3 or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof.
5. A compound as claimed in any of claims 1 to 3 or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof for use as a medicament.
6. A compound as claimed in any of claims 1 to 3 or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof for use in killing or inhibiting the growth of a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
7. A compound as claimed in any of claims 1 to 3 or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, for use in treatment of a bacterial infection caused by a Gram-positive bacterium or a Gram-negative bacterium.
8. A method for treatment of bacterial infection in a subject comprising: administering to the subject an effective amount of the compound of any of claims 1 to 3.
9. The method as claimed in claim 8, wherein the bacterial infection is caused by a Gram-positive bacterium or a Gram-negative pathogens.
10. The method as claimed in claim 9, wherein the bacterial infection is caused by E. coli, P. aeruginasa, K. pneumonia, A. baumannii, S. aureus, E. faecalis, or E. faecium.
11. A compound as claimed in any of claims 1 to 3 or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof for use in treating a disease or condition in a patient wherein said disease or condition is caused by a microorganism selected from the group consisting of Gram positive and Gram negative pathogens.
12. Use of a compound of Formula I in any of claims 1 to 3 or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof, in killing or inhibiting the growth of a microorganism selected from the group consisting of bacteria, virus, fungi and protozoa.
13. A pharmaceutical composition comprising a compound of Formula I in any of claims 1 to 3 or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof together with a pharmaceutically acceptable carrier.
14. A pharmaceutical composition comprising a compound of Formula I in any of claims 1 to 3 or its stereoisomers, pharmaceutically acceptable salts, complexes, hydrates, solvates, tautomers, polymorphs, racemic mixtures, optically active forms and pharmaceutically active derivative thereof together with a pharmaceutically acceptable carrier, and in combination with at least one antibiotic.
PCT/IN2017/050188 2016-05-20 2017-05-19 Heterocyclic compounds useful as anti-bacterial agents and method for production WO2017199265A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
BR112018073743-7A BR112018073743A2 (en) 2016-05-20 2017-05-19 compound, process, method for treating bacterial infection in a subject, use of a compound, and pharmaceutical compositions
CA3024824A CA3024824A1 (en) 2016-05-20 2017-05-19 Heterocyclic compounds useful as anti-bacterial agents and method for production
AU2017267631A AU2017267631A1 (en) 2016-05-20 2017-05-19 Heterocyclic compounds useful as anti-bacterial agents and method for production
CN201780029570.5A CN109476652B (en) 2016-05-20 2017-05-19 Heterocyclic compounds useful as antibacterial agents and process for their production
RU2018142830A RU2018142830A (en) 2016-05-20 2017-05-19 Heterocyclic compounds applicable as antibacterial agents and method for their preparation
EP17737888.2A EP3458452B1 (en) 2016-05-20 2017-05-19 Heterocyclic compounds useful as anti-bacterial agents and method for production
MX2018014165A MX2018014165A (en) 2016-05-20 2017-05-19 Heterocyclic compounds useful as anti-bacterial agents and method for production.
JP2019513506A JP6671541B2 (en) 2016-05-20 2017-05-19 Heterocyclic compounds useful as antibacterial agents and methods for their production
US16/301,887 US10711011B2 (en) 2016-05-20 2017-05-19 Substituted oxazolidines as anti-bacterial agents
KR1020187036214A KR20190034154A (en) 2016-05-20 2017-05-19 Heterocyclic compounds useful as antibacterial agents and production methods
ZA2018/08531A ZA201808531B (en) 2016-05-20 2018-12-18 Heterocyclic compounds useful as anti-bacterial agents and method for production

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN201641017526 2016-05-20
IN201641017526 2016-05-20

Publications (1)

Publication Number Publication Date
WO2017199265A1 true WO2017199265A1 (en) 2017-11-23

Family

ID=57139944

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2017/050188 WO2017199265A1 (en) 2016-05-20 2017-05-19 Heterocyclic compounds useful as anti-bacterial agents and method for production

Country Status (13)

Country Link
US (1) US10711011B2 (en)
EP (1) EP3458452B1 (en)
JP (1) JP6671541B2 (en)
KR (1) KR20190034154A (en)
CN (1) CN109476652B (en)
AU (1) AU2017267631A1 (en)
BR (1) BR112018073743A2 (en)
CA (1) CA3024824A1 (en)
GB (1) GB2550437B (en)
MX (1) MX2018014165A (en)
RU (1) RU2018142830A (en)
WO (1) WO2017199265A1 (en)
ZA (1) ZA201808531B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018225097A1 (en) 2017-06-08 2018-12-13 Bugworks Research India Pvt Ltd Heterocyclic compounds useful as anti-bacterial agents and method for production thereof
US20190292199A1 (en) * 2016-05-20 2019-09-26 Bugworks Research, Inc. Heterocyclic compounds useful as anti-bacterial agents and method for production
WO2019186590A1 (en) 2018-03-28 2019-10-03 Bugworks Research India Pvt Ltd Oxazolidinone antibiotic compounds and process of preparation

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008126024A2 (en) 2007-04-11 2008-10-23 Actelion Pharmaceuticals Ltd Oxazolidinone antibiotic derivatives
WO2010041219A1 (en) * 2008-10-10 2010-04-15 Actelion Pharmaceuticals Ltd 2-benzothiophenyl-and 2-naphthyl-0xaz0lidin0nes and their azaisostere analogues as antibacterial agents
WO2014170821A1 (en) * 2013-04-16 2014-10-23 Actelion Pharmaceuticals Ltd Antibacterial biaromatic derivatives

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
YU52403A (en) * 2000-12-26 2006-03-03 Dr.Reddy's Research Foundation Heterocyclic compounds having antibacterial activity, process for their preparation and pharmaceutical compositions containing them
JP2009526036A (en) * 2006-02-08 2009-07-16 ファイザー・プロダクツ・インク Oxazolidinones containing oxindole as antibacterial agents
CA2642406A1 (en) * 2006-02-14 2007-08-23 Pfizer Products Inc. Benzoxazinone and benzoxazepinone oxazolidinones as antibacterial agents
PL2794600T3 (en) * 2011-12-22 2018-06-29 Novartis Ag 2,3-Dihydro-benzo[1,4]oxazine derivatives and related compounds as phosphoinositide-3 kinase (PI3K) inhibitors for the treatment of e.g. rheumatoid arthritis
AR102256A1 (en) * 2014-10-15 2017-02-15 Actelion Pharmaceuticals Ltd ANTIBACTERIAL BASIC BIAROMATIC DERIVATIVES WITH REPLACEMENT OF AMINOALCOXI
GB2550437B (en) * 2016-05-20 2021-06-23 Bugworks Res Inc Heterocyclic compounds useful as anti-bacterial agents and method for production

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008126024A2 (en) 2007-04-11 2008-10-23 Actelion Pharmaceuticals Ltd Oxazolidinone antibiotic derivatives
WO2010041219A1 (en) * 2008-10-10 2010-04-15 Actelion Pharmaceuticals Ltd 2-benzothiophenyl-and 2-naphthyl-0xaz0lidin0nes and their azaisostere analogues as antibacterial agents
WO2014170821A1 (en) * 2013-04-16 2014-10-23 Actelion Pharmaceuticals Ltd Antibacterial biaromatic derivatives

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical -` Sciences", 2005, LIPPINCOTT WILLIAM & WILKINS
"Remington's Pharmaceutical Sciences", 2005, LIPPINCOTT WILLIAM & WILKINS
CORWIN HANSCH: "Comprehensive Medicinal Chemistry", vol. 5, 1990, PERGAMON PRESS
JERRY MARCH; MICHAEL SMITH: "Advanced Organic Chemistry", 2001, JOHN WILEY & SONS
T.W. GREENE: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY AND SONS

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190292199A1 (en) * 2016-05-20 2019-09-26 Bugworks Research, Inc. Heterocyclic compounds useful as anti-bacterial agents and method for production
US10711011B2 (en) 2016-05-20 2020-07-14 Bugworks Research, Inc. Substituted oxazolidines as anti-bacterial agents
WO2018225097A1 (en) 2017-06-08 2018-12-13 Bugworks Research India Pvt Ltd Heterocyclic compounds useful as anti-bacterial agents and method for production thereof
WO2019186590A1 (en) 2018-03-28 2019-10-03 Bugworks Research India Pvt Ltd Oxazolidinone antibiotic compounds and process of preparation
CN112218862A (en) * 2018-03-28 2021-01-12 巴格沃克斯研究有限公司 Oxazolidinone antibiotic compounds and process for their preparation
JP2021519314A (en) * 2018-03-28 2021-08-10 バグワークス・リサーチ・インコーポレイテッド Oxazolidinone antibiotic compound and preparation method
US11590142B2 (en) 2018-03-28 2023-02-28 Bugworks Research, Inc. Oxazolidinone antibiotic compounds and process of preparation
CN112218862B (en) * 2018-03-28 2024-01-02 巴格沃克斯研究有限公司 Oxazolidinone antibiotic compounds and process for preparing same
JP7473204B2 (en) 2018-03-28 2024-04-23 バグワークス・リサーチ・インコーポレイテッド Oxazolidinone antibiotic compounds and methods of preparation

Also Published As

Publication number Publication date
US20190292199A1 (en) 2019-09-26
ZA201808531B (en) 2020-02-26
MX2018014165A (en) 2019-08-21
EP3458452B1 (en) 2023-08-30
EP3458452A1 (en) 2019-03-27
BR112018073743A2 (en) 2019-02-26
JP6671541B2 (en) 2020-03-25
US10711011B2 (en) 2020-07-14
RU2018142830A3 (en) 2020-06-22
RU2018142830A (en) 2020-06-22
CN109476652B (en) 2022-07-15
AU2017267631A1 (en) 2018-12-06
JP2019519605A (en) 2019-07-11
CN109476652A (en) 2019-03-15
GB2550437B (en) 2021-06-23
GB201614801D0 (en) 2016-10-19
GB2550437A (en) 2017-11-22
CA3024824A1 (en) 2017-11-23
KR20190034154A (en) 2019-04-01

Similar Documents

Publication Publication Date Title
US11897900B2 (en) Inhibitors of KEAP1-Nrf2 protein-protein interaction
EP3634968B1 (en) Heterocyclic compounds useful as anti-bacterial agents and method for production thereof
EP3458452B1 (en) Heterocyclic compounds useful as anti-bacterial agents and method for production
AU2007331247A1 (en) 2-quinolinone and 2-quinoxalinone- derivatives and their use as antibacterial agents
JP5662940B2 (en) New antimicrobial agents
US11590142B2 (en) Oxazolidinone antibiotic compounds and process of preparation
EP3717490B1 (en) Anti-bacterial heterocyclic compounds and their synthesis
WO2023017549A1 (en) Antibiotic pyrazinothiazine derivatives and process of preparation thereof

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 2019513506

Country of ref document: JP

Kind code of ref document: A

Ref document number: 3024824

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112018073743

Country of ref document: BR

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

Ref document number: 17737888

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2017267631

Country of ref document: AU

Date of ref document: 20170519

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20187036214

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017737888

Country of ref document: EP

Effective date: 20181220

ENP Entry into the national phase

Ref document number: 112018073743

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20181119