CA2303959A1 - Oxazolidinone derivatives and pharmaceutical compositions - Google Patents

Oxazolidinone derivatives and pharmaceutical compositions Download PDF

Info

Publication number
CA2303959A1
CA2303959A1 CA002303959A CA2303959A CA2303959A1 CA 2303959 A1 CA2303959 A1 CA 2303959A1 CA 002303959 A CA002303959 A CA 002303959A CA 2303959 A CA2303959 A CA 2303959A CA 2303959 A1 CA2303959 A1 CA 2303959A1
Authority
CA
Canada
Prior art keywords
compound
oxo
alkyl
phenyl
methyl
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
CA002303959A
Other languages
French (fr)
Inventor
Jackson B. Hester, Jr.
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pharmacia and Upjohn Co
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2303959A1 publication Critical patent/CA2303959A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing aromatic rings
    • 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

Abstract

A compound of Formula (I) or a pharmaceutically acceptable salt thereof, which is antimicrobial agents, effective against various human and veterinary pathogens, including gram positive aerobic organisms, gram negative organisms, and anaerobic organisms.

Description

OXAZOLIDINONE DERIVATIVES AND PHARMACEUTICAL COMPOSITIONS
BACKGROUND OF THE INVENTION
The present invention relates to novel oxazolidinone compounds or pharmaceutically acceptable salts thereof, and pharmaceutical agents that contain them as active ingredients for preventing or treating infectious diseases. The compounds are unique oxazolidinones having a hexahydro-1,4-diazepin-5-one substituent.
More specifically, novel oxazolidinone compounds of the present invention relates to useful antimicrobial agents, effective against various human and veterinary 1o pathogens, including gram positive aerobic organisms such as multiply-resistant staphylococci and streptococci, gram negative organisms such as H. in, fluenzae and M.
catarrhalis as well as anaerobic organisms such as bacteroides and clostridia species, and acid-resistant organisms such as Mycobacterium tuberculosis and Mycobacterium avium.
INFORMATION DISCLOSURE
International Publication No. 97/27188 discloses piperazine-3-one analogs which are homologs of the invention.
International Publication No. W093/23384 discloses oxazolidinones containing a 2o substituted diazine (piperazine) moiety and their uses as antimicrobials.
International Publication No. W093/09103 discloses substituted aryl and heteroaryl-phenyl-oxazolidinones useful as antimicrobials.
International Publication No. W090/02744 discloses 5'-indolinyl-5-amidomethyloxazolidinones, 3-(fused-ring substituted)phenyl-S-midomethyloxazolidinones, and 3-(nitrogen substituted)-phenyl-5-amidomethyloxazolidinones which are useful as antibacterial agents.
Other references disclosing various oxazolidinones include US Patents 5,547,950, 4,801,600, J. Med. Chem., 32, 1673-81 (1989); J. Med. Chem., 33, 2569-78 (1990);
Tetrahedron, 45, 1323-26 ( 1989); and J. Med. Chem., 35, 1156 ( 1992).
3o European Patent Publication 352,781 discloses phenyl and pyridyl substituted phenyl oxazolidinones.
European Patent Publication 316,594 discloses 3-substituted styryl oxazolidinones.

European Patent Publication 312,000 discloses phenylmethyl and pyridylmethyl substituted phenyl oxazolidinones.
SUMMARY OF THE INVENTION
s The present invention provides an oxazolidinone derivative represented by the general structural Formula I or a pharmaceutically acceptable salt thereof R~ O
O
~N ~ \ N ~O X
R-'N~

I
or a pharmaceutically acceptable salt thereof wherein:
R is H, C2_6 alkenyl, C2_~ alkynyl, Ci_6 alkyl, or C,_6 alkyl substituted with one or two of the following:
a) F, b) Cl, c) CF3, d) -OH, e) C~_a alkoxy, f) -CH2C(=O)C,_4 alkyl, 2o g) -OC(=O)N(R4)2.

h) C~_a alkyl S(O)n, (wherein n is 0 to 2), I) -CN, j) carboxy, k) -C,_4 alkoxycarbonyl, 1) -C(=O)N(R4)z, m) -N(R4)S02C,_4 alkyl, n) -N(R4)C(=O)C, _4 alkyl, o) -N(R4)C(-O)N(R4)2, p) -N(R4)C(=O)C,.a alkoxy, 3o q) aryl, or r) Het;

aryl is phenyl, optionally substituted with one or two of the following:

a) F, b) C1, c) Br, d) -CF3, e) CN, f) C,_3 alkoxy, or g) C,_3 alkylthio;

io Het is a 5- or 6-membered heteroaromatic moiety having 1-3 N , O or S atoms, optionally substituted with the following:

a) F, b) Cl, c) C,_3 alkoxy, t5 d) C,_3 alkylthio, or e) CN;

R, and R2 are independently H, F, or Cl;

R3 is a) C,_6 alkyl, optionally substituted with one to three F or one to two Cl, 2o b) C~_6 alkoxy, c) amino, d) C1_6 alkylamino, e) C,.6 dialkylamino f) C3.~ cycloalkyl, 25 g) C1_6 alkylthio, or -N~CH2) m h) V (wherein m is 0, 1, 2, 3 or 4);

R4 is 3o a) H, or b) C,_3 alkyl; and XisOorS.
The present invention also provides an antimicrobial agent or pharmaceutical composition that contains the oxazolidinone compound or a pharmaceutically acceptable salt thereof as an active ingredient. The antimicrobial agent containing the active ingredient of the present invention can be used for treatment or prevention of infectious diseases.
DETAILED DESCRIPTION OF THE INVENTION
The compounds of Formula I as structurally disclosed above are useful antimicrobials. Typically, as further explained below, the compounds can be administered 1o as antibacterial agents in a dosage range of from about 0.1 to 100 mg/kg or preferably from about 3.0 to about 50 mg/kg of body weight per day.
In the structural formula shown above the carbon content of various hydrocarbon containing moieties is indicated by a prefix designating the minimum and maximum number of carbon atoms in the moiety, i.e., the prefix Ci-Cj defines the number of carbon 15 atoms present from the integer "i" to the integer "j" inclusive.
The term "C~_6 alkyl" used herein refers to an alkyl group having one to six carbon atoms such as, for example, methyl, ethyl, propyl, butyl, pentyl, hexyl and isomeric forms thereof; preferably methyl, ethyl, propyl and isomeric forms thereof.
The term "C2_6 alkenyl" refers to at least one double bound alkenyl group having 2o two to six carbon atoms such as, for example, vinyl, 1-propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl, 2-butenyl, 1-pentenyl, 2-pentenyl, 1-hexenyl and isomeric forms thereof, preferably an alkenyl group having 2 to 6 carbon atoms, and more preferably an alkenyl group having 2 to 4 carbon atoms.
The term "C2_7 alkynyl" refers to at least one triple bond alkynyl group having two 25 to seven carbon atoms such as, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl and their isomeric forms thereof.
The term "C,_6 alkylamino" refers to an alkyl group having one to six carbon atoms attached to an amino moiety.
The term "C1_6 dialkylamino" refers to two alkyl groups having one to six carbon 3o atoms attached to an amino moiety.
The term "C1_4 alkoxy" refers to an alkyl group having one to four carbon atoms attached an oxygen atom of hydroxyl group such as, for example, methoxy, ethoxy, propoxy, butoxy and isomeric forms thereof, preferably an alkoxy group having 1 to 2 carbon atoms.
The term "C,_6 alkylthio" refers to an alkyl group having one to six carbon atoms attached a thio moiety such as, for example, methythio, ethylthio, propyithio and isomeric s forms thereof, preferably an alkylthio group having 1 to 2 carbon atoms.
The term "C3_6 cycloalkyl" refers to three to six carbon atoms forming cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and isomeric forms thereof.
The term "aryl" refers to an phenyl moiety optionally substituted with one or two F, Cl, Br, -CF3, -CN, -C,_3 alkoxy, or -C~_3 alkylthio;
1o The term "Het" refers to a 5- or 6-membered heteroaromatic moiety having one to three atoms selected from the group consisting of O, N or S atoms such as, for example, furan, thiophene, pyrrole, pyrazole, triazoles, oxazole, thiazole, isothiazole, oxadiazoles, oxathiazole, pyridine, pyridazine, pyrimidine, pyrazine, piperazine and triazines all of which can optionally be substituted with one substituent selected from the group 15 consisting of F, Cl, C1_3 alkoxy, C,_3 alkylthio or CN.
The compounds of the present invention can be converted to their salts according to conventional methods.
Pharmaceutically acceptable salts means acid addition salts useful for administering the compounds of this invention and these include hydrochloride, 20 hydrobromide, sulfate, phosphate, acetate, propionate, lactate, mesylate, maleate, succinate, tartrate, citrate, 2-hydroxyethyl sulfonate, fumarate and the like when a basic group is present. These salts may be in hydrated form. Some of the compounds of this invention may form metal salts such as sodium, potassium, calcium and magnesium salts and these are embraced by the term "pharmaceutically acceptable salts".
25 Due to the configuration at C-5 of the oxazolidinone ring of compounds as represented in the structure of Formula I the compounds of this invention may exist in geometric, optical and other isomeric forms and this invention embraces any of these isomers. The racemic mixture and enantiomers are all believed to be useful as an antibacterial. Regardless, the preferred absolute configuration at C-5 of the 30 oxazolidinone ring of compounds is as represented in the structure of Formula I. This absolute configuration is called (S) under the Cahn-Ingold-Prelog nomenclature system.
It is believed that a majority of the pharmacological activity resides in this (S}-enantiomer to produce the antibacterial effect.
Compounds of Formula I can be prepared as shown in Scheme I where P
represents an alcohol protecting group such as benzyl or tert-butyldimethylsilyl.
Structure 2 of this scheme are prepared according to the methods outlined in Example 1, Step 1 and 2. In Scheme I, the alcohols of 2 are protected as benzyl ethers.
In a suitable procedure for this reaction, a solution of the alcohol 2 in a solvent such as Et20 or THF is allowed to react first with sodium hydride at 0-25 °C and then with benzyl bromide and tetrabutylammonium iodide at 0-25 °C to give structure 3. The ethylene ketal of 3 can then be removed with an acidic catalyst such asp-toluenesulfonic acid in acetone (as described in Example 1, Step 2) to give structure 5 where P is benzyl.
Alternatively the 1o ketal of 2 can be removed, the resulting structure 4 is allowed to react with tert-butyldimethylsilyl chloride and imidazole in DMF or tert-butyldimethylsilyl chloride and triethyl amine in methylene chloride to give structure 5 where the alcohol protecting group (P) is tert-butyldimethylsilyl. Ketone 5 is then allowed to react with hydroxylamine hydrochloride and sodium acetate in methanol-methylene chloride to give oxime 6 (see ~5 Example l, Step 3). The Beckmann rearrangement of structure 6 is carried out with p-toluenesulfonyl chloride and sodium carbonate in aqueous acetone at 20-40 °C to give structure 7. For compounds of Formula I where R is not hydrogen, compounds 7 can be alkylated with R'Y where Y is Br, I, CH3S03 or p-CH3PhS03 and R' is an appropriate alkyl substituent. In one method for this alkylation compounds of structure 7 are allowed 2o to react with sodium hydride and R'Y in a solvent such as DMF at 0-25 °C to give 8.
Alternatively, structure 7 can react with R'Y, potassium hydroxide and tetrabutylammonium bromide in THF or acetonitrile at 20-50 °C to give 8. Deprotection of the alcohols 7 or 8 provide structure 9. When P is a benzyl ether, this can be accomplished by hydrogenolysis with hydrogen and a palladium catalyst in ethanol or 25 with ammonium formate and a palladium catalyst in methanol at 10-30 °C. The tert-butyldimethylsilyl protecting group can be removed under acidic conditions or with fluoride ion. This deprotection can be carried out, for example, with trifluoroacetic acid in methylene chloride at 25 °C or with tetrabutylammonium fluoride in THF at 25 °C to give alcohol 9. Transformation of the alcohol 9 to the amine 11 can be carried out as 3o described in Example 1, Step 1. Alternatively, the reaction of 9 with m-nitrobenzenesulfonyl chloride and triethylamine in methylene chloride at 5-25 °C will give the m-nitrobenzenesulfonate 10 which will react with ammonium hydroxide in THF
or acetonitrile -isopropanol at 30-60 °C to give the amine 11. The reaction of compound 11 with acyl halides, anhydrides, isocyanates, isothiocyanates or dithioesters provides compounds of Formula I.
Compounds of Formula I where R is hydrogen and X is oxygen are conveniently prepared by allowing compounds 12 to react with p-toluenesulfonyl chloride and sodium carbonate in aqueous acetone at 20-40 °C (see Example 1, Step 4).

Scheme I
R~ O R~ O
~O~N / \ N~O ~. ~O~N / \ N~O
O' \~ ",H O . "H
R2 ~OH R2 ~O-CH2Ph R~ O R~ O
O~N / \ N~O ---.r O~N / \ N~O
R ",H R ",H

R~ O R~ O
HN
HO N '-' N / \ N O ---~ O~N / \ N O
R ",H R ",H

._ _ R N N R/ \ N~O -. R N N R/ \ N~O
~",H O~ ~ ",H
R2 ~OP R2 ~OH

R~ O'I R~ O
O~N / \ N~O ~ ~~N / \ N~O
",H ~~--~ ",H
R2 ~ONos R2 ~NH2 HO R~ O
N '' ~~N / \ N~O
~.J ",H ~ ---~ Formula I

_g_ WO 99/24428 PC1'/US98/22639 The compounds of the invention are useful for the treatment of microbial infections in humans and other warm blooded animals by either parenteral, oral, or topical administration.
The term "treatment" as used herein means partial or total lessening of symptoms of a disease which a patient suffers from; the term "prevention" as used herein means partial or total avoidance of symptoms of a disease in a patient who, according to a doctor's diagnosis, may suffer from the disease or a related state unless the preventive measure is taken.
The pharmaceutical compositions of this invention may be prepared by combining 1o the compounds of Formula I of this invention with a solid or liquid pharmaceutically acceptable carrier and, optionally, with pharmaceutically acceptable adjuvants and excipients employing standard and conventional techniques. Solid form compositions include powders, tablets, dispersible granules, capsules and suppositories. A
solid carrier can be at least one substance which may also function as a diluent, flavoring agent, t5 solubilizer, lubricant, suspending agent, binder, tablet disintegrating agent, and encapsulating agent. Inert solid Garners include magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, cellulosic materials, low melting wax, cocoa butter, and the like. Liquid form compositions include solutions, suspensions and emulsions. For example, there may be provided solutions of the 2o compounds of this invention dissolved in water and water-propylene glycol and water-polyethylene glycol systems, optionally containing conventional coloring agents, flavoring agents, stabilizers and thickening agents.
Preferably, the pharmaceutical composition is made by employing conventional techniques in unit dosage form containing effective amounts of the active component, 25 that is, the compound of Formula I according to this invention.
The quantity of active component, that is the compound of Formula I, in the pharmaceutical composition and unit dosage form thereof may be varied or adjusted widely depending upon the particular application method, the potency of the particular compound and the desired concentration. Generally, the quantity of active component 3o will range between 0.5% to 90% by weight of the composition.
In therapeutic use for treating, or combatting bacterial infections in humans and other animals that have been diagnosed with bacterial infections, the compounds or pharmaceutical compositions thereof will be administered orally, parenterally and/or topically at a dosage to obtain and maintain a concentration, that is, an amount, or blood-level of active component in the animal undergoing treatment which will be antibacterially effective. Generally, such antibacterially effective amount of dosage of active component will be in the range of about 0.1 to about 100 mg/kg, more preferably about 3.0 to about 50 mg/kg of body weightlday. It is to be understood that the dosages may vary depending upon the requirements of the patient, the severity of the bacterial infection being treated, and the particular compound being used. Also, it is to be understood that the initial dosage administered may be increased beyond the above upper level in order to rapidly achieve the desired blood-level or the initial dosage may be 1o smaller than the optimum and the daily dosage may be progressively increased during the course of treatment depending on the particular situation. If desired, the daily dose may also be divided into multiple doses for administration, e.g., two to four times per day.
The compounds of Formula I are administered parenterally, i.e., by injection, for example, by intravenous injection or by other parenteral routes of administration.
Pharmaceutical compositions for parenteral administration will generally contain a pharmaceutically acceptable amount of the compound according to Formula I as a soluble salt (acid addition salt or base salt) dissolved in a pharmaceutically acceptable liquid carrier such as, for example, water-for-injection and a suitably buffered isotonic solution, for example, having a pH of about 3.5-6. Suitable buffering agents include, for example, 2o trisodium orthophosphate, sodium bicarbonate, sodium citrate, N-methylglucamine, L(+)-lysine and L(+)-arginine, to name a few. The compound according to Formula I
generally will be dissolved in the Garner in an amount sufficient to provide a pharmaceutically acceptable injectable concentration in the range of about 1 mg/ml to about 400 mg/ml.
The resulting liquid phan-naceutical composition will be administered so as to obtain the above-mentioned antibacterially effective amount of dosage. The compounds of Formula I according to this invention are advantageously administered orally in solid and liquid dosage forms.
As a topical treatment, an effective amount of a compound of Formula I is admixed in a pharmaceutically acceptable gel or cream vehicle that can be applied to the 3o patient's skin at the area of treatment. Preparation of such creams and gels is well known in the art and can include penetration enhancers.
The compounds of this invention are useful antimicrobial agents, effective against various human and veterinary pathogens, including gram positive aerobic organisms such - 1o -as multiply-resistant staphylococci and streptococci, gram negative organisms such as H.
in~luenzae and M. catarrhalis as well as anaerobic organisms such as bacteroides and clostridia species, and acid-resistant organisms such as Mycobacterium tuberculosis and Mycobacterium avium.
In order to more fully illustrate the nature of the invention and the manner of practice the same, the following experimental examples are presented, but they should j not be taken as limitations.
EXAMPLE 1 Preparation of (,S~-N-[(3-[3-fluoro-4-(1,2,3,4,6,7-hexahydro-5-oxo-1,4-t o diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyl]methyl) acetamide.
O
HN N / ~ N~O
O~ ~ ~~~H
~NHAc Step I: Preparation of (S)-N-[3-(3-fluoro-4-piperidin-I-yl-phenyl)-2-oxo-15 oxazolidin-5-ylmethyl)-acetamide Diisopropylethylamine (15.7 ml) and 3,4-difluoronitrobenzene (5.0 ml) are added successively to an ethyl acetate solution (70 ml) of piperidine (5.77 g) and the mixture is stirred at room temperature for 2 days. Water is added to the reaction solution and the separating ethyl acetate layer are washed with water and brine, dried over anhydrous 2o sodium sulfate. The solvent is evaporated to afford a vitro compound (10.1 g) in a yield of 100%. Palladium on carbon ( 10%, 1.0 g) is added to an ethyl acetate solution ( 101 ml) of the vitro compound ( 10.1 g) and the mixture is stirred at room temperature for 4 hours under hydrogen atmosphere. The palladium on carbon is filtered off and the filtrate is concentrated under vacuum to yield an amine (8.75 g, 100%). Sodium 25 hydrogencarbonate (5.0 g) and benzyloxycarbonyl chloride (8.4 ml) are added successively to a tetrahydrofuran (THF) solution (100 ml) of the amine (8.75 g), and the mixture is stirred at room temperature for 14 hours. Water is added to the reaction solution and the separating THF layer is washed with water and brine, dried over anhydrous sodium sulfate. The solvent is evaporated and the residue is purified by silica 3o gel column chromatography (solvent: ethyl acetate/hexane/chloroform =
I/6/4) to afford a benzyl carbamate ( 14.5 g) in a yield of 98%. Butyl lithium ( 1.6 M hexane solution: 5.2 WO 99!24428 PCT/US98/22639 ml) is added to a THF solution (24 ml) of the benzyl carbamate (2.75 g) at -78 C and the mixture is stirred for 5 min. At the same temperature, (R)-(-)-glycidyl butyrate { 1.25 ml) is added to the stirred solution and the mixture is stin:ed for 14 hours while the temperature is raised slowly to room temperature. Water is added to the reaction solution and the separating THF layer is washed with water and brine, dried over anhydrous sodium sulfate. The solvent is evaporated and the residue is purified by silica gel column chromatography (solvent: ethyl acetate/hexane - 3/1 ) to afford an alcohol (2.20 g) in a yield of 89%. Tosyl chloride (2.85 g) is added to a pyridine solution (8 ml) of the alcohol (2.20 g) and the mixture is stirred at room temperature for 6 h. Water (32 ml) is added to to the reaction solution and the mixture is stirred for 1 hour. The resulting precipitate is collected by filtration and washed with water, followed by drying under vacuum at room temperature to afford a tosylate (3.28 g) in a yield of 98%. Sodium azide (3.80 g) is added to a dimethylformamide (DMF) solution (23 mi) of the tosylate (3.28 g) at room temperature and the mixture is stirred at 65 C for S.S hours. After the reaction mixture is cooled to room temperature, water is added and the mixture is extracted with ethyl acetate; the organic layer is concentrated under vacuum. The resulting residue is dissolved in ethyl acetate and washed with water and brine, dried over anhydrous sodium sulfate. The solvent is evaporated and the residue is purified by silica gel column chromatography (solvent: ethyl acetate/hexane - 1/ 1 ) to afford an azide (2.20 g) in a yield of 94%. Acetic anhydride (0.65 ml) and pyridine ( 1.0 ml) are added to an ethyl acetate solution ( 19 ml) of the azide (2.20 g) at room temperature; after addition of palladium on carbon ( 10%, 0.22 g), the mixture is stirred at room temperature for 6 hours under 1 atm hydrogen atmosphere. The palladium on carbon is filtered off and the filtrate is washed with water and brine, dried over anhydrous sodium sulfate. The solvent is evaporated and the residue is purified by silica gel column chromatography (solvent:
acetone/hexane - 1 / 1 } to afford the title compound.
Step 2: Preparation of (S)-N-{3-[3-fluoro-4-(4-oxo-piperidin-1-yl)-phenyl]-2-oxo-oxazolidin-5-ylmethyl }-acetamide.
Using a commercially available 1,4-dioxo-8-aza-spiro[4.5]decane, (S)-N-{3-[4-( 1,4-dioxa-8-aza-spiro[4.5]dec-8-yl)-3-fluoro-phenyl]-2-oxo-oxazolidin-5-ylmethyl }-acetamide is synthesized by the same method as in Step 1. To an acetone solution (70 ml) of this compound (3.79 g), water (20 ml) and p-toluenesulfonic acid monohydrate (3.66 g) are added successively and the mixture is heated under reflux for 3 hours. After the reaction mixture is cooled to room temperature, acetone is distilled off and the aqueous layer is neutralized with triethylamine. The solution is extracted with methylene chloride and the organic layer is washed with brine, dried over anhydrous sodium sulfate.
The solvent is evaporated and the residue is purified by silica gel column chromatography s (solvent: chloroform/methanol a 50/1 - 25/1) to afford the title compound .
Step 3: Preparation of (S)-N-{3-[3-fluoro-4-(4-hydroxyimino-piperidin-1-yl)-phenyl]-2-oxo oxazolidin-5-ylmethyl }-acetamide.
Sodium acetate (517 mg) and hydroxylamine hydrochloride (219 mg) are successively added to a methanol-methylene chloride solution (10-10 ml) of 1.00 g of the 1o product of Step 2, and the mixture is stirred at room temperature for 2 days. The solvent is evaporated and the residue is dissolved in methanol, followed by addition of a silica gel (8 g). Methanol is evaporated and the residue is purified by silica gel column chromatography (solvent: chloroform/methanol = 50/ 1 - 25/ 1 ) to afford the title compound.
15 Step 4: Preparation of (,S~-N-[[3-[3-Fluoro-4-(1,2,3,4,6,7-hexahydro-5-oxo-1,4-diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyl]methyl]acetamide.
A stirred mixture of the compound of product of Step 3 (0.200 g, 0.549 mmol) in acetone (5.3 mL), under nitrogen, is treated first with 5% aqueous sodium carbonate (5.3 mL) and then, dropwise during 3minuteswith a solution of p-toluenesulfonyl chloride 20 (0.16 g, 0.82 mmol) in acetone (2.7 mL). Initially this mixture is a two phase solution;
however, after about 25minutesa precipitate began to form. It is kept at ambient temperature (23 °C) for 4 hours and filtered. The filtrate is concentrated under reduced pressure to remove acetone and the aqueous residue is extracted with CH2C12.
The extract is dried (MgS04) and concentrated to give a small amount of crude product. Most 25 of the product is in the aqueous layer which is concentrated in vacuo. The residue is combined with the crude product from the CH2Cl2 extract and chromatographed on silica gel with mixtures of MeOH-NH40H-CH2CI2 that continued 3-5% MeOH and 0.3-0.5%
NHaOH. The product is crystallized from MeOH-EtOAc to give the title compound.
mp 140-146 °C;
3o MS m/2 (relative intensity) 364 (M+, 96.1 ), 320 ( 100), 306 (6.7), 294 ( 10.9), 236 (41.8);
HRMS calcd for C17H2,FN4O4; 364.1547 (M+}; found 364.1545;

~H NMR [300 MHz, (CD3)2S0] b 1.81 (s, 3H), 2.57 (m, 2H), 3.07 (m, 4H), 3.24 (m, 2H), 3.38 (t, 2H}, 3.67 (d, d, 1H), 4.06 (t, 1H), 4.68 (m, 1H), 7.08 (t, 1H), 7.13 (d, d, 1 H), 7.45 (d, d, 1 H), 7.65 (t, 1 H), 8.21 (t, 1 H).
EXAMPLE 2: Preparation of (S~-N-([3-(3-fluoro-4-(1,2,3,4,6,7-hexahydro-5-oxo-1,4-diazepin-1-yl)phenyl] -2-oxo-5-oxazolidinyl]methyl] thioacetamide.
F
HN N , O
o~J ~
~H
'--~N~C~CH3 I I
S
to Step 1: Preparation of (5~-[3-[3-fluoro-4-{1,2,3,4,6,7-hexahydro-5-oxo-1,4-diazepin-1-yl)-phenyl]-2-oxo-5-oxazolidinyl]methyl tert-butyldimethylsilyl ether.
A stirred solution of 10.6 g (0.03 mol) of (S'~-[3-[4-{1,4-dioxo-8-azaspiro[4.5]dec-8-yl)-3-fluorophenyl]-2-oxo-5-oxazolidinyl]methanol, the intermediate of formula 2 15 (Scheme 1) for the preparation of (S~-N-{3-[3-fluoro-4-(4-oxopiperidin-1-yl)phenyl]-2-oxooxazolidin-5-ylmethyl}-acetamide (Example 1, Step 2), in acetone (230 mL) is treated with water (65 mL) and p-toluenesulfonic acid monohydrate ( 11.4 g, 0.06 mol), refluxed under nitrogen for 5 hours and kept at ambient temperature (24 °C) for 18 hours.
Tt is then concentrated in vacuo to remove acetone. The aqueous residue is neutralized 20 with sodium bicarbonate and extracted with ethyl acetate; the extract is washed with saturated sodium bicarbonate, water and dilute sodium chloride, dried (Na2S04) and concentrated to give the ketone, a compound of formula 4 (Scheme 1 ). A
stirred solution of the ketone and triethylamine ( 12.5 mL, 0.09 mol) in methylene chloride ( 100 mL) is treated with tert-butyldimethylsilyl chloride (6.03 g, 0.04 mol) and kept under nitrogen at 25 ambient temperature for 23 hours. Additional tert-butyldimethylsilyl chloride (3.0 g) is added and the mixture is kept at ambient temperature for an additional 20 hours.
Additional triethylamine (3.0 mL) and tert-butyldimethylsilyl chloride (3.0 g) are again added and the mixture is kept at ambient temperature for 4 days, diluted with methylene chloride, washed with water and dilute sodium chloride, dried (Na2S04) and 3o concentrated. Chromatography of the residue on silica gel with mixtures of acetone-heptane that contained 20-30% acetone gave 7.72 g of the tert-butyldimethylsilyl (TBDMS) ether, a compound of formula 5 (Scheme 1 ) where P is TBDMS. A stirred solution of the TBDMS ether (7.27 g, 17.2 mmol) in methanol ( 150 mL) is treated dropwise with a solution of hydroxylamine hydrochloride ( 1.44 g, 0.021 mol) and sodium acetate ( 1:72 g, 0.021 mol) in water ( 15 mL) and kept at ambient temperature for 20 hours. The mixture is concentrated under reduced pressure. A solution of the residue, a white solid, in methylene chloride is washed with water and dilute sodium chloride, dried (Na2S04) and concentrated to give 7.25 g of the oxime, a compound of formula 6 (Scheme 1 ). A stirred solution of the oxime in acetone ( 165 mL), under nitrogen, is to treated with 5% aqueous sodium carbonate (165 mL) and then dropwise during minutes with a solution of p-toluenesulfonyl chloride (4.92 g, 0.0258 mol) in acetone (80 mL). The mixture is kept at ambient temperature for 18 hours and then concentrated under reduced pressure. A solution of the residue in methylene chloride is washed with water and dilute sodium chloride, dried (Na2S04) and concentrated.
Chromatography of the residue on silica gel with 3% methanol - 0.3% ammonium hydroxide-methylene chloride gave 5.98 g of the titled compound.
Step 2: Preparation of (S~-[[3-[3-fluoro-4-(1,2,3,4,6,7-hexahydro-5-oxo-1,4-diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyl] methyl] amine.
An ice cold, stirred mixture of the product of Example 2, Step 1 (0.22 g, 0.50 2o mmol) in tetrahydrofuran (THF; 15 mL), under nitrogen, is treated dropwise during 2 minutes, with a 1 M solution of tetrabutylammonium fluoride in THF ( 1.5 mL). The mixture is kept in the ice bath for 10 minutes and at ambient temperature (24 °C) for 1 hour 25 minutes, diluted with ethyl acetate, washed with water and brine, dried (NazS04) and concentrated. Chromatography of the residue on silica gel with mixtures of methanol-methylene chloride containing 3-6% methanol gave 0.15 g of the alcohol, a compound of formula 9 (Scheme 1 ) where R is hydrogen: MS(ES) m/z 324 (M+H+).
A
stirred suspension of the alcohol (0.15 g, 0.46 mmol) in methylene chloride (15 mL) and THF (8 mL), under nitrogen, is treated with triethylamine (0.5 mL, 1.4 mmol) and then portionwise during 1 minute at ambient temperature, with 0.14 g (0.56 mmol) of m-3o nitrobenzenesulfonyl chloride. The mixture is stirred for 90 minutes, mixed with additional methylene chloride ( 10 mL) to give a solution and kept at ambient temperature for 1 hour. It is then kept for several days at -11 °C, diluted with methylene chloride, washed with saturated sodium bicarbonate, water and brine, dried (Na2S04) and WO 99/24428 PCT/US9$/22639 concentrated to give 0.21 g of the m-nitrobenzenesulfonate, a compound of formula 10 (Scheme 1 ). A stirred mixture of the m-nitrobenzenesulfonate (0.21 g, 0.44 mmol), acetonitrile ( 10 mL), 2-propanol ( 10 mL) and 29% ammonium hydroxide ( 10 mL) is warmed at 45-50 °C under a Dry Ice-acetone condenser for 4.5 hours and kept at ambient temperature for 18 hours. Additional ammonium hydroxide (5 mL) is added and the mixture is warmed at 45-50 °C for 4.5 hours, kept at ambient temperature for I hour, treated with 5 mL of ammonium hydroxide and kept at ambient temperature for 18 hours.
It is then concentrated to give a yellow solid which is chromatographed on silica gel with mixtures of methanol-methylene chloride containing 5-7.5% methanol followed by 8%
1o methanol-0.2% ammonium hydroxide-methylene chloride to give the titled product.
Step 3: Preparation of (S~-N-[[3-[3-fluoro-4-(1,2,3,4,6,7-hexahydro-5-oxo-1,4-diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyl]methyl]thioacetamide.
A stirred solution of 0.12 g of the product of Example 2, Step 2 and 0.40 mL
of triethylamine in a mixture of methylene chloride ( 10 mL) and methanol ( 10 mL), under t s nitrogen, is treated with ethyl dithioacetate (0.05 mL) and kept at ambient temperature for 145 hours. Additional 0.05 mL portions of ethyl dithioacetate are added after 24, 31 and 49 hours; additional triethylamine ( 1.0 mL) is also added after 49 hours. The mixture is concentrated to a small volume, diluted with ethyl acetate, washed with water and brine, dried (Na2S04) and concentrated. Chromatography of the residue on silica gel with 3.5%
2o methanol-methylene chloride gave 0.061 g of the titled product.
~H NMR [300 MHz, (CD3)2SOJ 8 2.42 (s, 3H), 2.56 (m, 2H), 3.07 (m, 4H), 3.24 (m, 2H), 3.76 (dd, 1 H), 3.87 (m, 2H), 4.11 (t, I H), 4.91 (m, 1 H), 7.12 (m, 2H), 7.46 (dd, 1 H}, 7.67 (broad s, 1 H), 10.35 (broad s, 1 H).
EXAMPLE 3: Preparation of (,S~-N-[[3-[3-fluoro-4-( 1,2,3,4,6,7-hexahydro-4-methyl-25 5-oxo-1,4-diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyl]methyl]-acetamide.
HsC~ F
N N / IA O

~H
~N~C~CH3 I I
O

Step 1: Preparation of (S}-[[3-[3-fluoro-4-(1,2,3,4,6,?-hexahydro-4-methyl-5-oxo-1,4-diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyl]methyl]amine.
A mixture of 0.63 g (1.4 mmol) of the product of Example 2, Step 2, methyl iodide (0.093 mL) and THF (40 mL) is added dropwise during 12 minutes, under nitrogen, to a stirred mixture of powdered potassium hydroxide (0.12 g) and tetrabutylammonium bromide (0.096 g) in THF ( 10 mL) and kept at ambient temperature for 20 hours. It is then diluted with ethyl acetate, washed with water and brine, dried (MgS04) and concentrated. Chromatography of the residue on silica gel with mixtures of acetone-methylene chloride that contained 10-40% acetone gave 0.46 g (71 %) of the to methylated product, a compound of formula 8 (Scheme I) where R' is methyl.
An ice cold, stirred mixture of this product (0.17 g, 0.38 mmol) and THF (12 mL), under nitrogen, is treated dropwise with a 1 M solution of tetrabutylammonium fluoride in THF
( 1.2 mL). It is kept in the ice bath for 15 minutes and at ambient temperature for 3 hours, mixed with ice water and extracted with ethyl acetate. The extract is washed with water t5 and brine, dried (MgS04) and concentrated to give 0.15 g of the alcohol, a compound of formula 9 (Scheme 1 ). A stirred, ice cold solution of the alcohol (0.52 g, 1.5 mmol) and triethylamine {0.60 mL} in methylene chloride (45 mL) is treated portionwise during 5 minutes with m-nitrobenzenesulfonyl chloride (0.42 g). The mixture is kept in the ice bath for 15 minutes and at ambient temperature for 3 hours, diluted with methylene 2o chloride, washed with saturated sodium bicarbonate, water and brine, dried (MgS04) and concentrated to give the m-nitrobenzenesulfonate, a compound of formula 10 (Scheme 1 ).
A stirred mixture of this product, acetonitrile (35 mL), 2-propanol (35 mL}
and concentrated ammonium hydroxide (35 mL) is kept at 45-50 °C under a Dry Ice-acetone condenser for 4.5 hours and at ambient temperature for 20 hours. Additional ammonium 25 hydroxide (6 mL) is added and the mixture is kept at 45-50 °C for 5.5 hours and at ambient temperature for 18 hours. The mixture is then concentrated under reduced pressure to remove the organic solvents and the aqueous residue is extracted first with ethyl acetate and then methylene chloride. The extracts are washed with water and brine, dried (MgSOa) and concentrated. Chromatography of the residue on silica gel with 3o mixtures of methanol-methylene chloride containing 7.5-10% methanol gave the titled compound.
Step 2: Preparation of (,S~-N-[[3-[3-fluoro-4-(1,2,3,4,6,7-hexahydro-4-methyl-oxo-I ,4-diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyljmethylJacetamide.
_ t~ .

A stirred, ice cold mixture of 0.10 g (0.30 mmol) of the product of Example 3, Step 1, and pyridine (1.74 mL), under nitrogen, is treated dropwise with acetic anhydride (0.57 mL, 6.04 mmol) and kept in the ice bath for 15 minutes and at ambient temperature for 3.5 hours. It is then concentrated in vacuo; the residue is mixed with ice water and s saturated sodium bicarbonate and extracted with ethyl acetate. The extract is washed with water and brine, dried (MgSOa) and concentrated. Crystallization of the residue from ethyl acetate-methanol gave 0.053 g of the titled compound.
mp 203-204 °C.
MS(ES) m/z 379 (M+H+), 401 (M+Na+).
Anal. calcd for C~gH~FN4O4: C, 57.13; H, 6.13; N, 14.81. Found: C, 57.05; H, 6.23; N, 14.85.
EXAMPLE 4 Preparation of (S~-N-[[3-[3-fluoro-4-(1,2,3,4,6,7-hexahydro-4-methyl-S-oxo-1,4-diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyl] methyl]-thioacetamide.
is N ~ I O
o~..J ~ JL
~H
~~~C~CH3 I I
S
2o An ice cold, stirred solution of 0.18 g (0.535 mmol) of the product of Example 3, Step 1 and triethylamine (0.21 mL) in THF (8 mL} and methylene chloride (10 mL) is treated with a solution of ethyl dithioacetate (0.074 mL, 0.64 mmol) in THF (2 mL). The mixture is kept at ambient temperature for 20 hours, treated with one drop of additional ethyl dithioacetate and kept at ambient temperature for 7 hours. It is then concentrated 25 under a stream of nitrogen. The residue is mixed with methylene chloride, washed with saturated sodium bicarbonate, water and brine, dried (Na2S04) and concentrated.
Chromatography of the residue on silica gel with mixtures of methanol-methylene chloride containing 2-4% methanol and crystallization of the product from ethyl acetate gave 0.13 g of the titled compound. mp 157-158 °C.
30 Anal. calcd for C,BH23FN4O3S: C, 54.81; H, 5.88; N, 14.20. Found: C, 54.83;
H, 5.93; N, 14.11.

EXAMPLE 5 MIC Test Method The in vitro MICs of test compounds are determined by a standard agar dilution method. A stock drug solution of each analog is prepared in the preferred solvent, usually DMSO:H20 (1:3}. Serial 2-fold dilutions of each sample are made using 1.0 ml aliquots of sterile distilled water. To each 1.0 ml aliquot of drug is added 9 ml of molten Mueller Hinton agar medium. The drug-supplemented agar is mixed, poured into 15 x l 100 mm petri dishes, and allowed to solidify and dry prior to inoculation.
Vials of each of the test organisms are maintained frozen in the vapor phase of a liquid nitrogen freezer. Test cultures are grown overnight at 35 C on the medium to appropriate for the organism. Colonies are harvested with a sterile swab, and cell suspensions are prepared in Trypticase Soy broth (TSB) to equal the turbidity of a 0.5 McFarland standard. A 1:20 dilution of each suspension is made in TSB. The plates containing the drug supplemented agar are inoculated with a 0.001 ml drop of the cell suspension using a Steers replicator, yielding approximately 104 to 105 cells per spot.
t 5 The plates are incubated overnight at 35 C.
Following incubation the Minimum Inhibitory Concentration (MIC pg/ml), the lowest concentration of drug that inhibits visible growth of the organism, is read and recorded. The data is shown in Table I.
TABLE I
Example SAURa SEPI EFAE' SPNE HIND MCAT' No. 9213 12084 9217 9912 30063 30610 MIC MIC MIC MIC MIC MIC

1 4 1 4 0.5 8 8 4 1 <0.5 1 <0.5 8 2 a) S. aureus, culture b) S. epidermidis, culture c) E. faecalis, culture d) S. pueumoniae, culture e) H. in, fluenzae, culture f) M. catarrhalis, culture

Claims (10)

1. A compound of Formula I:

or a pharmaceutically acceptable salt thereof wherein:

R is H, C2-6 alkenyl, C2-7 alkynyl, C1-6 alkyl, or C1-6 alkyl substituted with one or two of the following:

a) F, b) Cl, c) CF3, d) -OH, e) C1-4 alkoxy, f) -CH2C(=O)C1-4 alkyl, g) -OC(=O)N(R4)2, h) C1-4 alkyl S(O), (wherein n is 0, 1 or 2), i) -CN, j) carboxy, k) -C1-4 alkoxycarbonyl, l) -C(=O)N(R4)2, m) -N(R4)SO2C1-4 alkyl, n) -N(R4)C(=O)C1-4 alkyl, o) -N(R4)C(-O)N(R4)2, p) -N(R4)C(-O)C1-4 alkoxy, q) aryl, or r) Het;
aryl is phenyl, optionally substituted with one or two of the following:

a) F, b) Cl, c) Br, d) -CF3, e) CN, f) C1-3 alkoxy, or g) C1-3 alkylthio;
Het is a 5- or 6-membered heteroaromatic moiety having 1-3 N , O or S atoms, optionally substituted with the following:
a) F, b) Cl, c) C1-3 alkoxy, d) C1-3 allcylthio, or e) CN;
R1 and R2 are independently a) H, b) F, or c) Cl;
R3 is a) C1-6 alkyl, optionally substituted with one to three F or one to two Cl, b) C1-6 alkoxy, c) amino, d) C1-6 alkylamino, e) C1-6 dialkylamino f) C3-6 cycloalkyl, g) C1-6 alkylthio, or h) (wherein m is 0, 1, 2, 3 or 4);

R4 is a) H, or b) C1-3 alkyl; and X is O or S.
2. A compound of Claim 1 wherein X is O.
3. A compound of Claim 1 wherein X is S.
4. A compound of Claim 1 wherein R is H.
5. A compound of Claim 1 wherein R is C1-4 alkyl.
6. A compound of Claim 1 wherein R3 is C1-4 alkyl, optionally substituted with one to three F or one to two Cl.
7. The compound of Claim 1 wherein Formula I is the S-enantiomer.
8. A compound of Claim 1 which is (a) (S)-N-[[3-(3-fluoro-4-(1,2,3,4,6,7-hexahydro-5-oxo-1,4-diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyl]methyl]acetamide, (b) (S)-N-[[3-(3-fluoro-4-(1,2,3,4,6,7-hexahydro-5-oxo-1,4-diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyl]methyl]thioacetamide, (c) (S)-N-[[3-[3-fluoro-4-(1,2,3,4,6,7-hexahydro-4-methyl-5-oxo-1,4-diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyl]methyl]acetamide, or (d) (S)-N-[[3-[3-fluoro-4-(1,2,3,4,6,7-hexahydro-4-methyl-5-oxo-1,4-diazepin-1-yl)phenyl]-2-oxo-5-oxazolidinyl]methyl]thioacetamide.
9. A method for treating microbial infections in human comprising administering to a patient in need thereof an effective amound of a compound of Formula I as shown in Claim 1.
10. A pharmaceutical composition comprising a compound of Formula I as shown in Claim 1 and a pharmaceutically acceptable carrier.
CA002303959A 1997-11-12 1998-10-30 Oxazolidinone derivatives and pharmaceutical compositions Abandoned CA2303959A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US6537697P 1997-11-12 1997-11-12
US60/065,376 1997-11-12
PCT/US1998/022639 WO1999024428A1 (en) 1997-11-12 1998-10-30 Oxazolidinone derivatives and pharmaceutical compositions

Publications (1)

Publication Number Publication Date
CA2303959A1 true CA2303959A1 (en) 1999-05-20

Family

ID=22062294

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002303959A Abandoned CA2303959A1 (en) 1997-11-12 1998-10-30 Oxazolidinone derivatives and pharmaceutical compositions

Country Status (22)

Country Link
US (1) US5998406A (en)
EP (1) EP1030852B1 (en)
JP (1) JP2001522849A (en)
KR (1) KR20010031953A (en)
CN (1) CN1154644C (en)
AT (1) ATE250054T1 (en)
AU (1) AU739055B2 (en)
BR (1) BR9813985A (en)
CA (1) CA2303959A1 (en)
DE (1) DE69818294T2 (en)
DK (1) DK1030852T3 (en)
ES (1) ES2207010T3 (en)
HK (1) HK1030373A1 (en)
HU (1) HUP0100470A3 (en)
IL (1) IL136062A (en)
NO (1) NO317291B1 (en)
NZ (1) NZ504503A (en)
PT (1) PT1030852E (en)
RU (1) RU2215740C2 (en)
SK (1) SK284577B6 (en)
TR (1) TR200001330T2 (en)
WO (1) WO1999024428A1 (en)

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EE200000707A (en) 1998-06-05 2002-04-15 Astrazeneca Ab Oxazolidinone derivatives, process for their preparation and pharmaceutical compositions containing them
DE19962924A1 (en) 1999-12-24 2001-07-05 Bayer Ag Substituted oxazolidinones and their use
KR100355345B1 (en) * 2000-01-21 2002-10-12 한국과학기술연구원 Oxazolidinone derivatives containing thiophene ring, a preparative method thereof and composition
DE10129725A1 (en) 2001-06-20 2003-01-02 Bayer Ag Combination therapy of substituted oxazolidinones
EP1539745B1 (en) * 2002-08-12 2006-03-29 Pharmacia & Upjohn Company LLC N-aryl-2-oxazolidinones and their derivatives
DE10300111A1 (en) * 2003-01-07 2004-07-15 Bayer Healthcare Ag Process for the preparation of 5-chloro-N - ({(5S) -2-oxo-3- [4- (3-oxo-4-morpholinyl) phenyl] -1,3-oxazolidin-5-yl} methyl ) -2-thiophenecarboxamide
BRPI0413838A (en) * 2003-08-25 2006-10-24 Warner Lambert Co antimicrobial aryloxazolidinone compounds
DE10355461A1 (en) 2003-11-27 2005-06-23 Bayer Healthcare Ag Solid, high bioavailabilty oral formulations of N-substituted 5-chloro-2-thiophene-carboxamide derivative in hydrophilized form, useful for combating thrombo-embolic diseases
KR100854211B1 (en) 2003-12-18 2008-08-26 동아제약주식회사 Novel oxazolidinone derivatives, a process for the preparation thereof and pharmaceutical composition comprising the same for antibiotics
WO2006056877A2 (en) * 2004-11-29 2006-06-01 Pharmacia & Upjohn Company Llc Diazepine oxazolidinones as antibacterial agents
DE102004062475A1 (en) * 2004-12-24 2006-07-06 Bayer Healthcare Ag Solid, orally administrable, modified release pharmaceutical dosage forms
EP1685841A1 (en) * 2005-01-31 2006-08-02 Bayer Health Care Aktiengesellschaft Prevention and treatment of thromboembolic disorders
ES2335307T3 (en) 2005-06-29 2010-03-24 PHARMACIA &amp; UPJOHN COMPANY LLC HOMOMORFOLIN OXALIDINONAS AS ANTIBACTERIAL AGENTS.
DE102005045518A1 (en) 2005-09-23 2007-03-29 Bayer Healthcare Ag New 5-thienylaminocarbonylmethyl-oxazolidin-2-one derivatives, useful for treating and preventing thromboembolic disease, are selective inhibitors of coagulation factor Xa
RU2429236C2 (en) 2005-10-04 2011-09-20 Байер Шеринг Фарма Акциенгезельшафт Novel polymorphous form and amorphous form of 5-chloro-n-({(5s)-2-oxo-3-[4-(3-oxo-4-morpholinyl)-phenyl]-1,3-oxazolidin-5-yl}-methyl)-2-thiophene carboxamide
DE102005047561A1 (en) 2005-10-04 2007-04-05 Bayer Healthcare Ag Drug delivery system, useful to treat and/or prevent e.g. thromboembolic disease, comprises 5-chloro-N-(((5S)-2-oxo-3-(4-(3-oxo-4-morpholinyl)-phenyl)-1,3-oxazolidine-5-yl)-methyl)-2-thiophene carboxamide with fast release active substance
DE102005047558A1 (en) * 2005-10-04 2008-02-07 Bayer Healthcare Ag Combination therapy of substituted oxazolidinones for the prophylaxis and treatment of cerebral circulatory disorders
TW200804358A (en) 2006-03-31 2008-01-16 Res Found Itsuu Lab Novel compound having heterocycle
JPWO2009044777A1 (en) * 2007-10-02 2011-02-10 財団法人乙卯研究所 Oxazolidinone derivatives having a 7-membered heterocycle
WO2012059823A1 (en) 2010-11-03 2012-05-10 Wockhardt Limited Process for the preparation of phosphoric acid mono- (l-{4- [(s) -5- (acetylaminomethyl) - 2 - oxo - oxazolidin- 3 - yl] - 2, 6 - difluorophenyl} - 4 -methoxymethylpiperidin- 4 - yl) ester
WO2015173664A1 (en) 2014-05-14 2015-11-19 Wockhardt Limited Process for the preparation of (5s)-n-{3-[3,5-difluoro-4-(4-hydroxy-4-methoxymethyl-piperidin-1-yl)-phenyl]-2-oxo-oxazolidin-5-ylmethyl}-acetamide

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4801600A (en) * 1987-10-09 1989-01-31 E. I. Du Pont De Nemours And Company Aminomethyl oxooxazolidinyl cycloalkylbenzene derivatives useful as antibacterial agents
CA1320730C (en) * 1987-10-16 1993-07-27 The Du Pont Merck Pharmaceutical Company Aminomethyl oxooxazolidinyl aroylbenzene derivatives useful as antibacterial agents
DE3884563T2 (en) * 1987-10-21 1994-02-17 Du Pont Merck Pharma Aminomethyl oxo oxazolidinyl ethenyl benzene derivatives useful as an antibacterial agent.
US4948801A (en) * 1988-07-29 1990-08-14 E. I. Du Pont De Nemours And Company Aminomethyloxooxazolidinyl arylbenzene derivatives useful as antibacterial agents
DE68929303T2 (en) * 1988-09-15 2002-05-02 Upjohn Co 3- (nitrogen substituted) phenyl-5-beta-amidomethyloxazoliden-2-one
WO1993009103A1 (en) * 1991-11-01 1993-05-13 The Upjohn Company Substituted aryl- and heteroarylphenyloxazolidinones useful as antibacterial agents
SK283420B6 (en) * 1992-05-08 2003-07-01 Pharmacia & Upjohn Company Oxazolidinones containing a substituted diazine moiety and their use as antimicrobials
AU698699B2 (en) * 1993-11-22 1998-11-05 Pharmacia & Upjohn Company Esters of substituted-hydroxyacetyl piperazine phenyl oxazolidinones
JPH0873455A (en) * 1994-03-15 1996-03-19 Upjohn Co:The Oxazolidinone derivative and medicine composition containingit as effective component
GB9601666D0 (en) * 1996-01-27 1996-03-27 Zeneca Ltd Chemical compounds

Also Published As

Publication number Publication date
NO20002434L (en) 2000-05-11
BR9813985A (en) 2000-09-26
ES2207010T3 (en) 2004-05-16
SK284577B6 (en) 2005-07-01
DE69818294T2 (en) 2004-07-08
ATE250054T1 (en) 2003-10-15
CN1154644C (en) 2004-06-23
NZ504503A (en) 2002-10-25
PT1030852E (en) 2004-02-27
IL136062A (en) 2004-02-08
HUP0100470A2 (en) 2002-05-29
SK6182000A3 (en) 2000-12-11
IL136062A0 (en) 2001-05-20
KR20010031953A (en) 2001-04-16
HUP0100470A3 (en) 2002-08-28
WO1999024428A1 (en) 1999-05-20
NO20002434D0 (en) 2000-05-11
EP1030852A1 (en) 2000-08-30
TR200001330T2 (en) 2000-11-21
NO317291B1 (en) 2004-10-04
CN1271358A (en) 2000-10-25
DE69818294D1 (en) 2003-10-23
HK1030373A1 (en) 2001-05-04
US5998406A (en) 1999-12-07
JP2001522849A (en) 2001-11-20
RU2215740C2 (en) 2003-11-10
AU1277899A (en) 1999-05-31
DK1030852T3 (en) 2003-12-22
AU739055B2 (en) 2001-10-04
EP1030852B1 (en) 2003-09-17

Similar Documents

Publication Publication Date Title
EP1030852B1 (en) Oxazolidinone derivatives and pharmaceutical compositions
EP0750618B1 (en) Oxazolidinone derivatives and pharmaceutical compositions containing them
US6218413B1 (en) Oxazolidinone antibacterial agents having a thiocarbonyl functionality
EP0730591B1 (en) Esters of substituted-hydroxyacetyl piperazine phenyl oxazolidinones
AU703784B2 (en) 4-pyrimidinyl- or 4-pyrazinyl-piperazinyl-phenyl- oxazolidinone derivatives, their preparation and their use as anti-bacterial agents
US5668286A (en) Oxazolidinone derivatives and pharmaceutical compositions containing them
EP0874852B1 (en) Oxazolidinone antibacterial agent with tricyclic substituents
US20090318431A1 (en) Benzoxazinone and benzoxazepinone oxazolidinones as antibacterial agents
EP1036074B1 (en) S-oxide tetrahydrothiopyran phenyloxazolidinones
KR20020067557A (en) Oxazolidinones Having a Sulfoximine Functionality and Their Use as Antimicrobial Agent
US6281210B1 (en) Benzoic acid esters of oxazolidinones having a hydroxyacetylpiperazine substituent
US20040127530A1 (en) Antimibicrobial [3.1.0.] bicyclic oxazolidinone derivatives
MXPA02009317A (en) Novel benzosultam oxazolidinone antibacterial agents.
MXPA00004528A (en) Oxazolidinone derivatives and pharmaceutical compositions
US20040142939A1 (en) N-Aryl-2-oxazolidinone-5-carboxamides and their derivatives
US20040204463A1 (en) N-aryl-2-oxazolidinone-5-carboxamides and their derivatives
CZ20001687A3 (en) Oxazolidinone derivatives and pharmaceutical compositions based thereon
PL190431B1 (en) Derivatives of oxazolydinones and pharmacological compositions
US6348459B1 (en) Sultam and sultone derived oxazolidinones

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued