CA2464109A1 - Process for the preparation of oxazolidinones and method of use thereof - Google Patents

Process for the preparation of oxazolidinones and method of use thereof Download PDF

Info

Publication number
CA2464109A1
CA2464109A1 CA002464109A CA2464109A CA2464109A1 CA 2464109 A1 CA2464109 A1 CA 2464109A1 CA 002464109 A CA002464109 A CA 002464109A CA 2464109 A CA2464109 A CA 2464109A CA 2464109 A1 CA2464109 A1 CA 2464109A1
Authority
CA
Canada
Prior art keywords
substituted
oxazolidinones
group
aryl
oxazolidinone
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
CA002464109A
Other languages
French (fr)
Inventor
Rawle I. Hollingsworth
Guijun Wang
Raghavakaimal Padmakumar
Jianmin Mao
Huiping Zhang
Zongmin Dai
Kanakamma Puthuparampil
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.)
Michigan State University MSU
Original Assignee
Michigan State University
Rawle I. Hollingsworth
Guijun Wang
Raghavakaimal Padmakumar
Jianmin Mao
Huiping Zhang
Zongmin Dai
Kanakamma Puthuparampil
Synthon Chiragenics Corporation
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 Michigan State University, Rawle I. Hollingsworth, Guijun Wang, Raghavakaimal Padmakumar, Jianmin Mao, Huiping Zhang, Zongmin Dai, Kanakamma Puthuparampil, Synthon Chiragenics Corporation filed Critical Michigan State University
Publication of CA2464109A1 publication Critical patent/CA2464109A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/08Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D263/16Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/18Oxygen atoms
    • C07D263/20Oxygen atoms attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/08Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D263/16Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/18Oxygen atoms
    • C07D263/20Oxygen atoms attached in position 2
    • C07D263/24Oxygen atoms attached in position 2 with hydrocarbon radicals, substituted by oxygen atoms, attached to other ring carbon atoms
    • 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
    • C07D413/12Heterocyclic 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 chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/18Testing for antimicrobial activity of a material
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B30/00Methods of screening libraries
    • C40B30/04Methods of screening libraries by measuring the ability to specifically bind a target molecule, e.g. antibody-antigen binding, receptor-ligand binding
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B50/00Methods of creating libraries, e.g. combinatorial synthesis
    • C40B50/08Liquid phase synthesis, i.e. wherein all library building blocks are in liquid phase or in solution during library creation; Particular methods of cleavage from the liquid support
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/195Assays involving biological materials from specific organisms or of a specific nature from bacteria
    • G01N2333/305Assays involving biological materials from specific organisms or of a specific nature from bacteria from Micrococcaceae (F)
    • G01N2333/31Assays involving biological materials from specific organisms or of a specific nature from bacteria from Micrococcaceae (F) from Staphylococcus (G)

Abstract

A process for preparing N-(substituted)-C~- (substituted methyl)-oxazolidinones, C-(substituted methyl)-oxazolidinones, and N-(substituted)-C~-(substituted ethyl)-oxazolidinones, preferably chiral, from optically active C-(protected oxymethyl)~ oxazolidinones is described. The process can be used to produce combinatorial libraries of the above substituted oxazolidinones in a two or three step reaction comprising a plurality of reagents differing in numbers of carbons or particular substituted oxazolidinones. A number of substituted oxazolidinones produced using the above process have been discovered to have antimicrobial activity.

Description

PROCESS FOR THE PREPARATION OF OXAZOLIDINONES
AND METHOD OF USE THEREOF
CROSS-REFERENCE TO RELATED APPLICATION
This application claims priority to U.S.
Provisional Application No.'60/330,266 filed October 18, 2001, and U.S. Provisional Application No.
60/330,268 filed October 18, 2001.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR
DEVELOPMENT
Not applicable.
Reference to a "Computer Listing Appendix submitted on a Compact Disc"
Not Applicable.
BACKGROUND OF THE INVENTION
(1) Field of the Invention The present invention relates to a process for preparing N-(substituted)-C-(substituted methyl) oxazolidinones, C-(substituted methyl)-oxazolidinones, and N-(substituted)-C-(substituted methyl)-oxazolidinones, preferably chiral, from optically active C-(protected oxymethyl)-oxazolidinones. The process can be used to produce combinatorial libraries of the above substituted oxazolidinones in a two or three step reaction comprising a plurality of reagents differing in numbers of carbons or particular substituted oxazolidinones. A number of substituted oxazolidinones produced using the above process have been discovered to have antimicrobial activity.
(2) Description of Related Art Oxazolidinones, particularly substituted oxazolidinones such as 3-(substituted)-5 alkylaminomethyl- and 3-(substituted)-5 acylaminomethyl-2-oxazolidinones, are an important class of drug substances which are used for a wide variety of drug applications. These applications 1o include use as antibacterial agents and in therapies for treating behavior disorders (Bowersock et al., Antimicrob. Agents Chemotherp. 44: 1367-1369 (2000);
Skold, Acta Vet. Scand. Suppl. 93: 23-36 (2000);
Diekema and Jones, Drugs 59: 7-16 (2000); Genin et al., J. Med. Chem. 43: 953-970 (2000); Johnson et al., J. Antimicrob. Chemother. 45: 225-230 (2000); Schulin et al., Antimicrob. Agents Chemotherp. 43: 2873-2876 (1999); Cynamon et al., Antimicrob. Agents Chemotherp. 43: 1189-1191 (1999); Chen and Reamer, Organic Letts. 1: 293-294 (1999); Brenner et al., Clin Therapeut. 22: 411-419 (2000); Clemett and Markham, Drugs 59: 815-827 (2000); Brickner et al., J. Med.
Chem. 39: 673-679 (1996); Barry, Antimicrob. Agents Chemotherp. 32: 150-152 (1988); Slee et al., Antimicrob. Agents Chemotherp. 31: 1791-1797 (1987);
Manninen et al., Abs. Paps. Amer. Chem. Soc. 212: 389-ORGN, Part 2, (Aug. 25, 1996) ) .
There are several methods for making the oxazolidinone nucleus in 3-(substituted)-5 3o alkylaminomethyl- and 3-(substituted)-5 acylaminomethyl-2-oxazolidinones. The general structure of 3-(substituted)-5-(substituted methyl)-2-oxazolidinone is wherein R1 is alkyl, aryl, heteroalkyl, heteroaryl, or mixture thereof, or hydrogen or hydroxy, and R2 is alkyl, aryl, heteroalkyl, heteroaryl, or mixture thereof. The following disclose processes for preparing oxazolidinones and substituted oxazolidinones.
U.S. Patent No. 6,288,238 B1 to 1o Hollingsworth and Wang disclose a process for preparing 5-hydroxymethyl-2-oxazolidinones in one step from 3,4-boronic acid ester protected 3,4 dihydroxybutyramides.
U.S. Patent No. 6,288,239 B1 to Hollingsworth and Wang discloses a process for preparing 5-trityloxymethyl-2-oxazolidinones arid suggests a scheme for the alkylation of N-lithio-N
substituted carbamates with oxiranes such as glycidyl butyrate as shown in Scheme 1.

Scheme 1 O O
O
O ~-Ph ~ O O
(base) O OH-R- ~ O ~ R \~OH
R
O O
MsCl/Py R ~O NaN3 R ~ H2 cat _OMs -~ \~N3 O O
Ac20 R-~ NH2 ~ R-~ NHAc Glycidyl equivalents such as epichlorohydrin can be used instead of glycidyl butyrate.
Schaus and Jacobsen (Tetrahedron Letts. 37:
7937-7940 (1996)) teach using optically active N
oxiranylmethylacetamides to prepare chiral 3 (substituted)-5-acetamidomethyl-2-oxazolidinones in one step by the alkylation of N-lithio-N-aryl (or 1o alkyl) carbamates as shown in Scheme 2.
Scheme 2 p O O
~~Ph O ~ (base) O
R-NH ~ H R ~ N
However, the above processes do not allow for the rapid synthesis of a plurality of substituted oxazolidinones at the same time in the same reaction.
Thus, producing a plurality of substituted oxazolidinones for drug screening is slow and cumbersome which affects the rate in which new and useful drugs can be discovered. Therefore, there remains a need for a rapid and simple process that can produce a plurality of substituted oxazolidinones at the same time in the same reaction. Being able to produce a plurality of drug candidates in a short 1o period of time would accelerate the rate at which new and useful drugs and other compounds are discovered.
The present invention provides a simple and rapid process for synthesizing substituted oxazolidinones.
Strains of Gram positive bacteria resistant to the present repertoire of antibiotics have been increasing in prevalence over the past several decades (Skold, Acta Vet. Scand. Suppl. 93: 23-36 (2000)).
Resistant Gram positive that have been commonly encountered include among others those in the 2o staphylococci, streptococci, pneumococci, and enterococci families. Because of the increasing prevalence of these antibiotic resistant bacterial strains, there is a clear need for new antimicrobial agents.
Several species of substituted oxazolidinones have been discovered to be effective antimicrobial agents against particular antibiotic resistant strains of Gram positive bacteria.
Linezolid (Clemett and Markham, Drugs 59: 815-827 (2000); Johnson et al., J. Antimicrob. Chemother. 45:
225-230 (2000)) is a substituted oxazolidinone which has been approved for the treatment of microbial infections. The structure of linezolid is shown below.

~N ~ ~ ~O
O
H~
N
O
A number of other substituted oxazolidinones with varying degrees of antibacterial activity against Gram positive and in some cases Gram negative bacteria are also known (Barry, Antimicrob. Agents Chemotherp. 32:
150-152 (1988); Brickner et al., J. Med. Chem. 39:
673-679 (1996); Genin et al., J. Med. Chem. 43: 953=
970 (2000); Slee et al., Antimicrob. Agents 1o Chemotherp. 31: 1791-1797 (1987)).
Most, if not all, of the known substituted oxazolidinones which have been found to have antibacterial activity have the structure shown below wherein the R3 substituent is aryl and the relative stereochemistries of the groups on the chiral center (C-5) is as indicated.

f A comparison of the structures for all of the known substituted oxazolidinones which have 2o antimicrobial activity, the general consensus has arisen that there are at least three elements of these substituted oxazolidinones which are critical for biological activity. The first element is that when the oxazolidinone ring is oriented as shown below such that all the ring atoms are in one plane, the carbonyl oxygen points up, the ring nitrogen is to the left, and the 5-substituent is to the right, then of the two possible orientations for the 5-substituent (distal or proximal), the proximal substituent is required for biological activity.

5 ~~~~~distal proximal The second element is that the 3-substituent is an 1o aryl. The third element is that the 5-substituent is an alkylamino methyl or an acetamidomethyl group. No substituted oxazolidinone which has antibacterial activity has been found which does not have all three of the above elements.
Because microorganisms will eventually develop resistance to antibiotics, there is a continual need for new antibiotics. The present invention provides families of novel substituted oxazolidinones which have antimicrobial activity but 2o which have structures which do not conform to the consensus structure thought to be necessary for antimicrobial activity.
SUMMARY OF THE INVENTION
The present invention provides a process for preparing N-(substituted)-C-(substituted methyl)-oxazolidinones, C-(substituted methyl)-oxazolidinones, and N-(substituted)-C-(substituted methyl)-oxazolidinones, preferably chiral, from optically -active C-(protected oxymethyl)-oxazolidinones. The process can be used to produce combinatorial libraries of the above substituted oxazolidinones in a two or three step reaction comprising a plurality of reagents differing in numbers of carbons or particular substituted oxazolidinones.
Therefore, the present invention provides a process for producing a library of substituted oxazolidinones which comprises (a) reacting a C-(protected oxymethyl)- oxazolidinone in an anhydrous organic solvent containing a first reagent including a plurality of compounds having different numbers of carbons which are reactive with N in the C- (protected oxymethyl)-oxazolidinone under alkylation or Buchwald conditions in an inert atmosphere to produce a mixture of N-(substituted)-C-(protected oxymethyl)-oxazolidinones (I); and (b) reacting the mixture of (I) produced in step (a) in an aqueous organic solvent with a second reagent which removes the protecting 2o group and replaces it with another group from the second reagent to produce the library of substituted oxazolidinones.
In a further embodiment of the above process, the second reagent is a reducing agent which removes the protecting group of the N-(substituted)-C-(protected oxymethyl)-oxazolidinone to provide a mixture of N-(substituted)-C-hydroxymethyl-oxazolidinones (II) as the library of substituted oxazolidinones.
3o In a further embodiment of the above process, the mixture of (II) is further reacted with a third reagent containing a plurality of compounds reactive with the hydroxymethyl in an anhydrous - g -organic solvent to produce a mixture of N-(substituted)-C-(substituted methyl)-oxazolidinones (III) as the library of substituted oxazolidinones.
Preferably, the anhydrous organic solvent further includes pyridine. In particular embodiments, the third reagent produces a mixture of 3-(substituted)-5-(substituted methyl)-2-oxazolidinones or a mixture of 3-(substituted)-4-(substituted methyl)-2-oxazolidinones.
1o The present invention further provides a process for producing a library of substituted oxazolidinones which comprises (a) reacting a C-(protected oxymethyl)- oxazolidinone in an anhydrous organic solvent containing a first reagent including a plurality of compounds having different numbers of carbons which are reactive with N in the C- (protected oxymethyl)-oxazolidinone under alkylation or Buchwald conditions in an inert atmosphere to produce a mixture of N-(substituted)-C-(protected oxymethyl)-oxazolidinones (I); (b) reacting the mixture of (I) produced in step (a) in an aqueous organic solvent with a second reagent which removes the protecting group of the N-(substituted)-C-(protected oxymethyl)-oxazolidinones to produce a mixture of N-(substituted)-C-hydroxymethyl-oxazolidinones (II); and (c) reacting the mixture of (II) produced in step (b) in an anhydrous organic solvent with a third reagent containing a plurality of compounds reactive with the hydroxymethyl of the ,mixture of (II) to produce a 3o mixture of N-(substituted)-C-(substituted methyl)-oxazolidinones (III) as the library of substituted oxazolidinones. Preferably, the anhydrous organic solvent in step (c) further includes pyridine. In particular embodiments, the third reagent produces a mixture of 3-(substituted)-5-(substituted methyl)-2-oxazolidinones or a mixture of 3-(substituted)-4-(substituted methyl)-2-oxazolidinones.
s In a further embodiment of the above processes, substituted is selected from the group consisting of acyl, alkyl, aryl, aryl sulfonyl, heteroalkyl, heteroaryl, cycle, heterocycle, thio, and mixtures thereof.
1o In a further embodiment of the above processes, the substituted oxazolidinones in the library are separated chromatographically.
In a preferred embodiment of the above process, the protecting group is a trityl group.
1s In a further embodiment of the above processes, under the alkylation conditions in step (a) the anhydrous organic solvent further includes an alkali without substantial reducing activity, preferably, the alkali is an ionic hydride, most 2o preferably, the ionic hydride is sodium hydride, and under the Buchwald conditions in step (a) the anhydrous organic solvent further includes a palladium catalyst, preferably, the palladium catalyst is Pd (OAC) 2.
25 In a further embodiment of the above processes, the mixture of N-(substituted)-C-(protected oxymethyl)-oxazolidinones (I) produced in step (a) are purified by extracting the reaction mixture with the organic solvent, drying over a drying agent, and then 3o removing the solvent.
In a still further embodiment of the above processes, the N-(substituted)-C-hydroxymethyl-oxazolidinones (II) produced in step (b) are purified by removing the solvent.
In a still further embodiment of the above processes, the N-(substituted)-C-(substituted methyl)-oxazolidinones (III) produced in step (c) are purified by extracting the reaction mixture with the organic solvent, drying over a drying agent, and then removing the solvent.
In a further embodiment of the above processes, the present invention provides a process 1o for preparing a library of substituted oxazolidinones which comprises reacting a C-hydroxymethyl-oxazolidinone in an anhydrous organic solvent including pyridine with a reagent containing a plurality of compounds reactive with the hydroxy group to produce a mixture of substituted oxazolidinones as the library of substituted oxazolidinones.
In a further embodiment of the above processes, the reaction produces a mixture of 5-(substituted methyl)-2-oxazolidinones, a mixture of 4-(substituted methyl)-2-oxazolidinones, a mixture of N-(substituted)-C-(hydroxymethyl)-2-oxazolidinones, or a mixture of N-(substituted)-C-(substituted methyl)-2-oxazolidinones.
In a further embodiment of the above processes, substituted is selected from the group consisting of aryl, alkyl, aryl, aryl sulfonyl, heteroalkyl, heteroaryl, cycle, heterocycle, thio, and mixtures thereof.
In a further embodiment of the above 3o processes, the substituted oxazolidinones in the library are separated chromatographically.
The present invention further provides a library of substituted oxazolidinones selected from the group consisting of N-(substituted)-C-(substituted methyl)-oxazolidinones, N-(substituted)-C-hydroxymethyl-oxazolidinones, and C-(substituted methyl)-oxazolidinones.
In a further embodiment of the library, substituted in N-(substituted) includes at least 10 different individual groups.
In a further embodiment of the library, substituted in C-(substituted) includes at least 10 different individual groups.
In a further embodiment of the library, the library is a mixture of N-(substituted)-C-hydroxymethyl-2-oxazolidinones or a mixture selected from the group consisting of 3-(substituted)-5-(substituted methyl)-2-oxazolidinones and 3-(substituted)-4-(substituted methyl)-2-oxazolidinones.
In a further embodiment of the library, substituted is selected from the group consisting of acyl, alkyl, aryl, aryl sulfonyl, heteroalkyl, 2o heteroaryl, cycle, heterocycle, thio, and mixtures thereof.
The present invention further provides a method of screening substituted oxazolidinones for biological activity which comprises (a) providing a library of the substituted oxazolidinones wherein the substituted oxazolidinones are selected from the group consisting of N-(substituted)-C-(substituted methyl)-oxazolidinones, N-(substituted)-C-hydroxymethyl-oxazolidinones, and C-(substituted methyl)-oxazolidinones; (b) chromatographically separating the substituted oxazolidinones in the library; and (c) testing the separated substituted oxazolidinones for the biological activity.

In a further embodiment of the above method, substituted in N-(substituted) includes at least 10 different individual groups.
In a further embodiment of the above method, substituted in C-(substituted methyl) includes at least 10 different individual groups.
In a further embodiment of the above method, the substituted oxazolidinones is a mixture of N=
(substituted)-C-hydroxymethyl-2-oxazolidinones or a 1o mixture selected from the group consisting of 3 (substituted)-5-(substituted methyl)-2-oxazolidinones, 3-(substituted)-4-(substituted methyl)-2 oxazolidinones, 5-(substituted methyl)-2 oxazolidinones, and 4-(substituted methyl)-2 oxazolidinones.
In a further embodiment of the above method, substituted is selected from the group consisting of aryl, alkyl, aryl, aryl sulfonyl, heteroalkyl, heteroaryl, cycle, heterocycle, thin, and mixtures 2o thereof.
The present invention further provides a substituted oxazolidinone with biological activity obtained by the above method.
The present invention further provides a process for producing a substituted oxazolidinone which comprises (a) reacting a C-(protected oxymethyl)-oxazolidinone in an anhydrous organic solvent containing a first reagent including a compound which is reactive with N in the C- (protected oxymethyl)-oxazolidinone under alkylation or Buchwald conditions in an inert atmosphere to produce an N-(substituted)-C-(protected oxymethyl)-oxazolidinone;
(b) reacting the N-(substituted)-C-(protected oxymethyl)-oxazolidinone in an aqueous organic solvent with a second reagent with a second reagent which replaces the protecting group of the N-(substituted)-C-(protected oxymethyl)-oxazolidinone with a hydrogen to produce an N-(substituted)-C-hydroxymethyl-oxazolidinone; and (c) reacting the N-(substituted)-C-hydroxymethyl-oxazolidinone in an anhydrous organic solvent with a third reagent containing a compound reactive with the hydroxy group to produce N-(substituted)-C-(substituted methyl)-oxazolidinones as the substituted oxazolidinone.
In a further embodiment of the above process, the anhydrous organic solvent in step (c) further includes pyridine.
In a further embodiment of the above process, substituted is selected from the group consisting of acyl, alkyl, aryl, aryl sulfonyl, heteroalkyl, heteroaryl, cycle, heterocycle, thio, and mixtures thereof.
2o In a further embodiment of the above process, the protecting group is a trityl group.
In a further embodiment of the above process, the substituted oxazolidinone has the formula O
~R2 Rl~ Y~C
wherein R1 is selected from the group consisting of hydrogen, acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, phenacyl, aryl sulfonyl, thio, and mixture thereof, or a hydrogen; R2 is selected from the group consisting of acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, aryl sulfonyl, phenacyl, thio, and mixture thereof, or a hydrogen, wherein hetero is an atom selected from the group consisting of O, N, P, and S; and y is a heteroatom selected from the group consisting of 0, N, and S.
In a further embodiment of the above process, the substituted oxazolidinone has the formula O
~R2 O N
R1-HNy l0 wherein R1 is selected from the group consisting of alkyl sulfonyl, aryl sulfonyl, alkyl, acyl, aryl, and thio and R2 is selected from the group consisting of alkyl, acyl, aryl, and thiol or, the substituted oxazolidinone has the formula O
~R2 O N
R1 Oy wherein R1 is selected from the group consisting of 2o alkyl sulfonyl, aryl sulfonyl, alkyl, acyl, aryl, and thio and R2 is selected from the group consisting of alkyl, acyl, aryl, and thio~ or, the substituted oxazolidinone has the formula O
O N RZ
R1-HNy X
wherein R1 is selected from the group consisting of alkyl, acyl, thio, and aryl, RZ is selected from the group consisting of C-3, C-4, -and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from the group. consisting of F, N02, C1, alkyl, and aryl;
or, the substituted oxazolidinone has the formula X
wherein R1 is selected from the group consisting of alkyl, acyl, thin, and aryl, R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from the group consisting of F, N02, C1, alkyl, and aryl;
or, the substituted oxazolidinone has the formula X
wherein R1 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, R2 is selected from the group consisting of alkyl, aryl, acyl, thio, and heterocycle, and X is selected from the group consisting of F, N02, Cl, alkyl, and aryl; or, the substituted oxazolidinone has the formula O

X
wherein R1 is selected from the group consisting of alkyl, aryl, acyl, thio, or heterocycle, R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from the group consisting of F, N02, C1, 1o alkyl, and aryl; or, the substituted oxazolidinone has the formula O
0 N~NH-R2 wherein R1 is selected from the group consisting of alkyl, aryl, acyl, thio, and heterocycle and R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers or, the substituted oxazolidinone has the formula O
0 N~NH-R2 R1 Oy wherein R1 is selected from the group consisting of 2o alkyl, aryl, acyl, thio, and heterocycle and R2 is selected from the group consisting of C-3, C-4, with C-5 chiral synthons with 1, 2, or 3 chiral centers.
In a further embodiment of the above process, under the alkylation conditions in step (a) the anhydrous organic solvent further includes an alkali without substantial reducing activity, preferably, the alkali is an ionic hydride, most preferably, the ionic hydride is sodium hydride.
In a further embodiment of the above process, under the Buchwald conditions in step (a) the anhydrous organic solvent further includes a palladium catalyst, preferably, the palladium catalyst is l0 Pd (OAc) 2.
In a further embodiment of the above process, the mixture of N-(substituted)-C-(protected oxymethyl)-oxazolidinone produced in step (a) is purified by extracting the reaction mixture with the organic solvent, drying over a drying agent, and then removing the solvent.
In a further embodiment of the above process, the N-(substituted)-C-hydroxymethyl oxazolidinone produced in step (b) is purified by 2o removing the solvent.
In a further embodiment of the above process, the N-(substituted)-C-(substituted methyl)-oxazolidinone produced in step (c) is purified by extracting the reaction mixture with the organic solvent, drying over a drying agent, and then removing the solvent.
The present invention further provides a substituted oxazolidinone which has the formula O
~R2 RliYwC

wherein R1 is selected from the group consisting of hydrogen, acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, phenacyl, aryl sulfonyl, thio, and mixture thereof, or a hydrogen; RZ is selected from the group consisting of acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, aryl sulfonyl, phenacyl, thio, and mixture thereof, or a hydrogen, wherein hetero is an atom selected from the group consisting to of O, N, P, and S~ and y is a heteroatom selected from the group consisting of O, N, and S.
The present invention further provides a substituted oxazolidinone which has the formula O
~R2 O N

wherein R1 is selected.from the group consisting of alkyl sulfonyl, aryl sulfonyl, alkyl, acyl, aryl, and thio and R2 is selected from the group consisting of 2o alkyl, aryl, aryl, and thin; or, a substituted oxazolidinone which has the formula ~R2 wherein R1 is selected from the group consisting of alkyl sulfonyl, aryl sulfonyl, alkyl, acyl, aryl, and thio and RZ is selected from the group consisting of alkyl, acyl, aryl, and thio; or, a substituted oxazolidinone which has the formula O

R1-HN~
X
s wherein R1 is selected from the group consisting of alkyl, acyl, thio, and aryl, R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from 1o the group consisting of F, N02, C1, alkyl, and aryl;
or, a substituted oxazolidinone which has the formula X
wherein R1 is selected from the group consisting of is alkyl, acyl, thio, and aryl, R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from the group consisting of F, N02, C1, alkyl, and aryl;
or, a substituted oxazolidinone which has the formula R1-HNy X
wherein R1 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with l, 2, or 3 chiral centers, R2 is selected from the group consisting of alkyl, aryl, acyl, thio, and heterocycle, and X is selected from the group consisting of F, N02, C1, alkyl, and aryl; or, a substituted oxazolidinone which has the formula O

R 1-HNy X
wherein R1 is selected from the group consisting of alkyl, aryl, acyl, thio, or heterocycle, R2 is selected to from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from the group consisting of F, NOz, C1, alkyl, and aryl; or, a substituted oxazolidinone which has the formula O
O N~NH-R2 wherein R1 is selected from the group consisting of alkyl, aryl, acyl, thio, and heterocycle and R2 is selected from the group consisting of C-3, C-4, and C
5 chiral synthons with l, 2, or 3 chiral centers; or, 2o a substituted oxazolidinone which has the formula NH-R
O N~
R1 Oy wherein R1 is selected from the group consisting of alkyl, aryl, acyl, thio, or heterocycle and R2 is selected from the group consisting of C-3, C-4, with C-5 chiral synthons with 1, 2, or 3 chiral centers.
The present invention further provides an antimicrobial composition comprising a carrier and one or more substituted oxazolidinones of the formula O
~R2 Rl~ YwC
wherein R1 is selected from the group consisting of hydrogen, aryl, alkyl, aryl, heteroalkyl, heteroaryl, 1o heterocycle, phenacyl, aryl sulfonyl, thio, and mixture thereof, or a hydrogen R2 is selected from the group consisting of acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, aryl sulfonyl, phenacyl, thio, and mixture thereof, or a hydrogen, wherein hetero is an atom selected from the group consisting of 0, N, P, and S; and y is a heteroatom selected from the group consisting of 0, N, and S.
OBJECTS
2o Therefore, it is the object of the present invention to provide a process for producing substituted oxazolidinones which are substituted at the N-position or the C-position, or both.
It is a further object of the present invention to provide a process for producing a library of substituted oxazolidinones comprising a plurality of oxazolidinones substituted at the N-position, a plurality of oxazolidinones substituted at the C
position, or a plurality of oxazolidinones substituted at both the N-position and the C-position.
These and other objects of the present invention will become increasingly apparent with reference to the following drawings and preferred embodiments.
DESCRIPTION OF THE DRAWINGS
Figure 1A shows the conversion of 5 trityloxymethyl-2-oxazolidinone to 3-(2,5 to dimethoxyphenacyl)-5-trityloxymethyl-2-oxazolidinone.
Figure 1B shows the conversion of 3-(2,5-dimethoxyphenacyl)-5-trityloxymethyl-2-oxazolidinone to 3-(2,5-dimethoxyphenacyl)-5-hydroxymethyl-2-oxazolidinone.
Figure 1C shows the conversion of 3-(2,5-dimethoxyphenacyl)-5-hydroxymethyl-2-oxazolidinone to a library of ten 3-(2,5-dimethoxyphenacyl)-5-(substituted methyl)-2-oxazolidinones.
Figure 2 shows the ten chlorides used in the 2o O-functionalization.
Figure 3 shows an HPLC profile of the library of ten 3-(2,5-dimethoxyphenacyl)-5-(substituted methyl)-2-oxazolidinones prepared as shown in Figures 1A to 1C.
Figure 4 shows the structure of the ten 3-(2,5-dimethoxyphenacyl)-5-(substituted methyl)-2-oxazolidinones identified in Figure 3.
DETAILED DESCRIPTION OF THE INVENTION
3o All patents, patent applications, government publications, government regulations, and literature references cited in this specification are hereby incorporated herein by reference in their entirety.

In case of conflict, the present description, including definitions, will control.
The present invention provides a novel process for preparing collections or combinatorial libraries of substituted oxazolidinones. In particular, the present invention provides a process for preparing libraries of optically active or chiral N-(substituted)-C-(substituted methyl)-oxazolidinones, N-(substituted)-C-(methyl)-oxazolidinones, and C-io (substituted methyl)-oxazolidinones bearing alkyl or aryl substituents in the N-substituted position (3-position) and a methyl group substituted with a heteroatom such as 0, N, or S in the C-substituted position (4- or 5-position) and wherein the heteroatom is further substituted with hydrogen or acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, phenacyl, aryl sulfonyl, or mixture thereof.
In a preferred embodiment, the substituted oxazolidinones comprising the library are optically 2o active or chiral N-(substituted)-C-(substituted methyl)-2-oxazolidinones, N-(substituted)-C-(methyl)-2-oxazolidinones, and C-(substituted)-2-oxazolidinones bearing alkyl or aryl substituents in the N-substituted position (3-position) and a methyl group substituted with a heteroatom in the C-substituted position (4- or 5-position) and wherein the heteroatom is further substituted with hydrogen or acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, phenacyl, aryl sulfonyl, or mixture thereof.
3o As used herein, the term "substituted"
refers to groups other than hydrogen substituted at the N-position or the methyl at the C-position.
Preferably, the substituting group is an organic group. Therefore, when the N-position is substituted, it is substituted with a group such as acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, aryl sulfonyl, phenacyl, or mixture thereof. When N is not referred to as being "substituted", the N has a hydrogen at the N-position. When the C-position methyl is substituted, it is referred to as "substituted methyl" wherein "substituted" is a group such as acyl, alkyl, aryl, heteroalkyl, heteroaryl, to heterocycle, phenacyl, aryl sulfonyl, or mixture thereof.
The general structure of these substituted oxazolidinones is shown below ~R2 Rii Y~C
wherein R1 is an acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, phenacyl, aryl sulfonyl, thio, or mixture thereof, or a hydrogen, R2 is an acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, 2o aryl sulfonyl, phenacyl, thio, or mixture thereof, or a hydrogen (when N is not substituted by an organic group), and y is a heteroatom selected from the group consisting of 0, N, and S. The heteroatom comprising R1 or R2 can include one or more atoms selected from the group consisting of 0, P, S, N, A1, and Si.
The above genus comprises at least eight families of substituted oxazolidinones. The first family (Family I) comprises substituted oxazolidinones with the following general structure O
~R2 wherein R1 is alkyl sulfonyl, aryl sulfonyl, alkyl, acyl, aryl, or thio and R2 is alkyl, acyl, aryl, or thio.
The second family (Family II) comprises substituted oxazolidinones with the following general structure O
~R2 wherein R1 is alkyl sulfonyl, aryl sulfonyl, alkyl, acyl, aryl, or thio and R2 is alkyl, acyl, aryl, or thio.
The third family (Family III) comprises z5 substituted oxazolidinones with the following general structure wherein R1 is alkyl, acyl, thio, or aryl, RZ is a C-3, 2o C-4, or C-5 chiral synthon with l, 2, or 3 chiral centers, and X is F, N02, C1, Alkyl, or aryl.
The fourth family (Family IV) comprises substituted oxazolidinones with the general structure wherein Rl is alkyl, acyl, thio, or aryl, R2 is a C-3, C-4, or C-5 chiral synthon with 1, 2, or 3 chiral centers, and X is F, N02, C1, Alkyl, or aryl.
The fifth family (Family V) comprises substituted oxazolidinones with the general structure X
1o wherein R1 is a C-3, C-4, or C-5 chiral synthon with 1, 2, or 3 chiral centers, R2 is alkyl, aryl, acyl, thio, or heterocycle, and X is F, N02, C1, Alkyl, or aryl.
The sixth family (Family VI) comprises substituted oxazolidinones with the general structure z Rl-wherein R1 is alkyl, aryl, aryl, thio, or heterocycle, RZ is a C-3, C-4, or C-5 chiral synthon with l, 2, or 3 chiral centers, and X is F, N02, C1, Alkyl, or aryl.
The seventh family (Family VII) comprises 2o substituted oxazolidinones with the general structure O
O N~NH-R2 wherein R1 is alkyl, aryl, aryl, thio, or heterocycle and R2 is a C-3, C-4, or C-5 chiral synthon with 1, 2, or 3 chiral centers.
The eighth family (Family VIII) comprises substituted oxazolidinones with the general structure O N~NH-R2 wherein R1 is alkyl, aryl, acyl, thio, or heterocycle and RZ is a C-3, C-4, or C-5 chiral synthon with 1, 2, to or 3 chiral centers.
Examples of the C-3, C-4, and C-5 chiral synthons with l, 2, or 3 chiral centers include, but are not limited to, (R)-3-acetoxy-4-bromobutyric acid, (S)-3-acetoxy-4-bromobutyric acid, (R)-3-Acetoxy-4-bromobutiryl chloride, (S)-3-Acetoxy-4-bromobutiryl chloride, (R)-2-Acetoxy-1,4-dibromobutane, (S)-2-Acetoxy-1,4-dibromobutane, (R)-3-Acetoxy-gamma-butyrolactone, (S)-3-Acetoxy-gamma-butyrolactone, (R)-4-Acetoxy-2-thioxopyrrolidine, (S)-4-Acetoxy-2-2o thioxopyrrolidine, (R)-4-Acetylthio-2-pyrrolidinone, (S)-4-Acetylthio-2-pyrrolidinone, (R)-4-Amino-1,3-butanediol, (S)-4-Amino-1,3-butanediol, (R)-3-Amino-1,2-dihydroxypropane, hydrochloride, (S)-3-Amino-1,2-dihydroxypropane, hydrochloride, (R)-4-Amino-3-hydroxy-1-trityloxy-butane, (S)-4-Amino-3-hydroxy-1-trityloxy-butane, (R)-4-Amino-3-hydroxybutanoic acid, (S)-4-Amino-3-hydroxybutanoic acid, (S)-4-Aminomethyl-2,2-dimethyl-1,3-dioxolane, (R)-3-Amino-1,2-propanediol, (S)-3-Amino-1,2-propanediol, (S)-N-Benzyl-3,4-dihydroxybutyramide, (R)-1-Benzyl-4-hydroxy-2-pyrrolidinone, (S)-1-Benzyl-4-hydroxy-2-pyrrolidinone, (R)-1-Benzyl-3-mesyloxy pyrrolidine, (S)-1-Benzyl-3-mesyloxy pyrrolidine, (R)-1-Benzyl-3-pyrrolidinol, (S)-1-Benzyl-3-pyrrolidinol, (R)-3-Bromo-1-(bromomethyl)propyl-methoxymethyl, (S)-3-to Bromo-1-(bromomethyl)propyl-methoxymethyl ether, (R)-4-Bromo-1,3-butanediol, (S)-4-Bromo-1,3-butanediol, (R)-4-Bromo-1,3-diacetoxy-butane, (S)-4-Bromo-1,3-diacetoxy-butane, (R)-3-Bromo-1,2-dihydroxypropane, (S)-3-Bromo-1,2-dihydroxypropane, (R)-4-Bromo-1,2-epoxybutane, (S)-4-Bromo-1,2-epoxybutane, (R)-5-Bromo-4-(methoxymethoxy)-pentanenitrile, (S)-5-Bromo-4-(methoxymethoxy)-pentanenitrile, (4R)-4-Bromomethyl-2-phenyl-1,3-dioxane, (4S)-4-Bromomethyl-2-phenyl-1,3-dioxane, (R)-1,3-Butanediol, (S)-1,3-Butanediol, (R)-1, 2, 4-Butanetriol, (S) -1, 2, 4-Butanetriol, (R) -1, 2, 4-Butanetriol trimesylate, (S)-1,2,4-Butanetriol trimesylate, (R)-4-Cyano-1,2-epoxybutane, (S)-4-Cyano-1,2-epoxybutane, 1,3-Dehydro-2-deoxy-N-acetylneuraminic acid, (R)-1,4-Dibromo-2-butanol, (S)-1,4-Dibromo-2-butanol, (R)-3,4-Dihydroxybutyramide, (S)-3,4-Dihydroxybutyramide, (R)-2,2-Dimethyl-4-aminomethyl-1,3-dioxane, (S)-2,2-Dimethyl-4-aminomethyl-1,3-dioxane, (R)-2,2-Dimethyl-1,3-dioxolane-4-acetamide, (S)-2,2-Dimethyl-1,3-dioxolane-so 4-acetamide, (R)-2,2-Dimethyl-1,3-dioxolane-4-acetic acid, methyl ester, (S)-2,2-Dimethyl-1,3-dioxolane-4-acetic acid, methyl ester, (R)-2,2-Dimethyl-1,3-dioxolane-4-acetonitrile, (S)-2,2-Dimethyl-1,3-dioxolane-4-acetonitrile, (R)-2,2-Dimethyl-1,3-dioxolane-4-propanol, (S)-2,2-Dimethyl-1,3-dioxolane-4-propanol, (3R)-1,3-Dioxane-2-methyl-4-carboxylic acid, (3S)-1,3-Dioxane-2-methyl-4-carboxylic acid, (R)-1,4-Ditosyloxy-2-butanol, (S)-1,4-Ditosyloxy-2-butanol, (3R)-3-(1-Ethoxyethoxy)-gamma-butyrolactone, (3S)-3-(1-Ethoxyethoxy)-gamma-butyrolactone, (2R)-2-(1-Ethoxyethoxy)-1,4-butanediol, (2S)-2-(1-Ethoxyethoxy)-1,4-butanediol, Ethyl (R)-4-bromo-3-1o hydroxybutanoate, Ethyl (S)-4-bromo-3-hydroxybutanoate, Ethyl (R)-4-chloro-3-hydroxybutanoate, Ethyl (S)-4-chloro-3-hydroxybutanoate, (R)-4-cyano-3-hydroxybutanamide, (S)-4-cyano-3-hydroxybutanamide, Ethyl (R)-4-cyano-3-hydroxybutanoate, Ethyl (S)-4-cyano-3-hydroxybutanoate, Ethyl (R)-3,4-epoxybutanoate, Ethyl (S)-3,4-epoxybutanoate, Ethyl (R)-3-hydroxy-decanoate, Ethyl (S)-3-hydroxy-decanoate, Ethyl (R)-3-hydroxy-tetradecanoate, Ethyl (S)-3-hydroxy-tetradecanoate, 2o Ethyl (R)-4-iodo-3-hydroxybutanoate, Ethyl (S)-4-iodo-3-hydroxybutanoate, (R)-4-(4-Fluorophenoxy)methyl butyrolactone, (S)-4-(4-Fluorophenoxy)methyl butyrolactone, (1S,3R)-3-Hydroxy-cyclopentanecarboxylic acid, (1S,3S)-3-Hydroxy-cyclopentanecarboxylic acid, (R)-4-Hydroxy-1-cyclopentene-1-carboxylic acid, (S)-4-Hydroxy-1-cyclopentene-1-carboxylic acid, (R)-4-Hydroxy-2-pyrrolidinone, (S)-4-Hydroxy-2-pyrrolidinone, (4R)-4-(2-Hydroxyethyl)-2-phenyl-1,3-dioxolane, (4S)-4-(2-3o Hydroxyethyl)-2-phenyl-1,3-dioxolane, (R)-2-Hydroxy gamma-butyrolactone, (S)-2-H drox y y-gamma-butyrolactone, (R)-3-Hydroxy-gamma-butyrolactone, (S)-3-Hydroxy-gamma-butyrolactone, (R)-4-Hydroxymethyl butyrolactone, (S)-4-Hydroxymethyl butyrolactone, (R)-4-Hydroxy-2-pyrrolidinethione, (S)-4-Hydroxy-2-pyrrolidinethione, (R)-3-Hydroxytetrahydrofuran, (S)-3-Hydroxytetrahydrofuran, (R)-4-Mercapto-2-pyrrolidinone, (S)-4-Mercapto-2-pyrrolidinone, (R)-2-(Methoxy-1-methylethoxy)-butanediol, (S)-2-(1-Methoxy-1-methylethoxy)-butanediol, Methyl (R)-4,5-dihydroxyisopropylidenepentanoate, Methyl (S)-4,5-dihydroxyisopropylidenepentanoate, Methyl (R)-2-lo phenyl-1,3-dioxolane-4-acetate, Methyl (S)-2-phenyl-1,3-dioxolane-4-acetate, Methyl (R)-3-hydroxy-4-trityloxy-butanoate, Methyl (S)-3-hydroxy-4-trityloxy-butanoate, (R)-3-Pyrrolidinol, (S)-3-Pyrrolidinol, (R)-3-Chloro-1,2-propanediol, (S)-3-Chloro-1,2-propanediol, (2S)-(+)-glycidal tosylate, Benzyl (R)-glycidyl ether, (R)-3-chlorolactic acid, Ethyl (S)-4-chloro-3-hydroxybutanoate, (S)-3-Hydroxybutyrolactone, (R)-2-hydroxybutyrolactone, (S)-2-Hydroxybutyrolactone, (R)-2-Chlrobutyric acid, (R)-2-2o bromobutyric acid, (S)-1-iso-propylaminopropanediol, (S)-1-tert-Butylaminopropanediol, (R)-1-cyclohexyl-ethyl-amine, (R)-Ethyl-nipecotate, (S)-Ethyl-nipecotate, (R)-Glycerol-3-phosphate, alpha-Glycerophosphatidylcholine, alpha-glycerophosphatidylethanolamine, (R)-0-Isopropylidene glycerol, (S)-0-Isopropylidene glycerol, (R)-0-Isopropylidene glycerol mesylate, (S)-0-Isopropylidene glycerol mesylate, (R)-0-Isopropylidene glycerol tosylate, (S)-0-Isopropylidene glycerol tosylate, (R)-0-methyl-0-isopropylidene glycerate, (R)-2-tetrahydrofuroic acid, (R)-1-Tosyl-glycerol, and (S)-1-Tosyl-glycerol.
The process for synthesis of the substituted oxazolidinones preferably uses an optically active C-protected oxazolidinone as the starting material, preferably a 5-(protected hydroxymethyl)-oxazolidinone such as 5-trityloxymethyl-oxazolidinone wherein the trityl is triphenylmethyl. Most preferably, the protected oxazolidinone is a 5-(protected hydroxymethyl)-2-oxazolidinone which in a further preferred embodiment is a 5-trityloxymethyl-2-oxazolidinone. The synthesis of 5-trityloxymethyl-2-oxazolidinone and its use are disclosed in U.S. Patent No. 6,288,239 B1 to Hollingsworth and Wang. The structure of 5-trityloxymethyl-2-oxazolidinone is shown below.
The general process for producing a library of substituted oxazolidinones comprises the following steps. First, a C-(protected oxymethyl)-oxazolidinone is N-arylated with a mixture of compounds comprising a 2o plurality of different aryl acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, aryl sulfonyl, or phenacyl bromides under Buchwald conditions (Yin and Buchwald, Org. Letts 2: 1101-1104 (2000)) or by simple alkylation under nitrogen. This produces a plurality of N-(substituted)-C-(protected oxymethyl)-oxazolidinones (I).

For example, under simple alkylation conditions, a C-(protected oxymethyl)-oxazolidinone is dissolved in an organic solvent such as tetrahydrofuran (THF) containing a strong base (alkali) which preferably does not have substantial reducing activity or which has reducing activity which is suppressed at low temperatures. Hydrides are strong bases which are suitable for the reaction.
Preferably, the strong base is an ionic hydride such 1o as an alkali hydride. Most preferred is sodium hydride which is a powerful base without substantial reducing activity. Other strong bases which may be used include lithium hydride, potassium hydride, rubidium hydride, cesium hydride, sodium alcoholates, sodium amide, and metallic sodium. In general, about 1 equiv. of the strong base (alkali), preferably sodium hydride, is added to the solvent containing the C-(protected oxymethyl)-oxazolidinone. In the case of sodium hydride, which is insoluble in organic 2o solvents, the sodium hydride is provided as a suspension in an organic solvent such as hexane.
After allowing the mixture containing the protected oxazolidinone and strong base to incubate at about 0° C
for about 10 minutes with stirring under an inert atmosphere such as nitrogen, the mixture is warmed to room temperature and stirred for about two hours and a mixture of n different arylating reagents, preferably in a molar ratio of about 1 to 1 to 1 to 2 (C-(protected oxymethyl)-oxazolidinone to mixture), is 3o added. The reaction is incubated at room temperature with stirring for a time (about eight hours) sufficient to arylate the N at the 3-position with the n different arylating reagents to produce n N-(substituted)-C-(protected oxymethyl)-oxazolidinones.
The reaction is then quenched by adding an aqueous solution containing an acid such as NH9C1 and the N-(substituted)-C-protected oxymethyl)-oxazolidinones are recovered by extracting the quenched reaction with the organic solvent, drying the extract over a drying agent such as anhydrous Na2S04, and concentrating the extract under reduced pressure (in vacuo). The N
(substituted)-C-(protected oxymethyl)-oxazolidinones 1o are preferably purified by chromatography.
Under Buchwald conditions, the C-(protected oxymethyl)-oxazolidinone and about 1 to 2 equiv. of a mixture of n different arylating reagents are incubated with a Pd(OAc)2 catalyst in an organic solvent such as tetrahydrofuran (THF) under an inert atmosphere such as argon at a temperature between about 45° to 110° C for a time sufficient to arylate the N at the 3-position with the n different arylating reagents to produce n N-(substituted)-C-(protected oxymethyl)-oxazolidinones (in general, about eight hours as determined by gas chromatography). The reaction is then cooled to room temperature, diluted with an organic solvent such as dichloromethane, filtered, and concentrated under reduced pressure (in vacuo). The N-(substituted)-C-(protected oxymethyl)-oxazolidinones are preferably purified by chromatography.
Next, the N-arylated oxazolidinones (N
substituted) are deprotected in the usual fashion by 3o hydrogenolysis using H2 and a palladium catalyst or an acid such as to produce a library of n N-(substituted)-C-hydroxymethyl-oxazolidinones (II).
For example, the N-arylated oxazolidinones, preferably purified by chromatography or the like, are incubated in an aqueous solvent such as wet dichloromethane (CH2C12 ) ( about 8 :1 CH2C12 : H20 ) further containing an acid such as trifluoroacetic acid (CF3C02H) at room temperature for a time sufficient (about four hours) to deprotect the C-hydroxymethyl. Preferably, the C-protecting group in the above reaction is a triphenylmethyl group. The reaction is quenched by adding triethylamine or other quenching agent and the to deprotected oxazolidinone concentrated under reduced pressure (in vacuo). The concentrated deprotected oxazolidinone is preferably further purified by chromatography.
In a further step, the n N-(substituted)-C
hydroxymethyl-oxazolidinones (II) are O-functionalized with a mixture of n different alkylation, acylation, sulfonylation, halogenation, or other such species.
For example, a mixture containing a plurality of different acyl, alkyl, aryl, heteroalkyl, heteroacyl, 2o heteroaryl, heterocycle, phenacyl, aryl sulfonyl, amides thereof, thiols thereof, or other such species, or the 0 of the hydroxymethyl is replaced by N-aryl, N-sulfonyl, N-sulfide, or other N-species, or the O of the hydroxymethyl is replaced by a thioalkyl, thioaryl, or other thio-species. Methods for converting the hydroxyl group to a nitrogen containing function can be done by any of the methods which are known. These include mesylation or tosylation followed by displacement with ammonia, azide, 3o benzylamine, or other nitrogen nucleophiles as taught for example in U.S. Patent No. 6,288,239 B1 to Hollingsworth and Wang or U.S. Patent No. 5,837,870 to Pearlman et al. For example, n substituted oxazolidinones (II), preferably purified by chromatography or the like, are then incubated in an organic solvent such as dry dichloromethane containing about 1 equiv. pyridine and about 1 equiv. of a mixture of n different acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, phenacyl, aryl sulfonyl, thins and amides thereof, or other such species halides at room temperature for time sufficient (about 12 to 16 hours) to functionalize the C-hydroxymethyl 1o at the 4- or 5-position. Afterwards, the reaction is quenched by addition of ammonium chloride, extracting the organic layer with dichloromethane, drying the organic layer extract over a drying agent such as anhydrous NazS09, and concentrating under reduced pressure (in vacuo). The above process generates a library of n2 N-(substituted)-C-(substituted methyl)-oxazolidinones (III). For example, if ten different aryl bromides reagents are used in the first step and ten different halide reagents in the second step, 100 2o N-(substituted)-C-(substituted)-oxazolidinones (III) are obtained.
Each of the products (I, II, or III) produced above can be separated chromatographically and each separately evaluated as drug or antimicrobial candidates.
The process takes advantage of the ease of reaction of the nitrogen atom at the 3-position in C-(protected oxymethyl)-oxazolidinones which enables both arylation and alkylation sequences for 3o substituting the N to be used. A further advantage is that in one or two steps, the protecting group can be removed and the hydroxyl group functionalized. In the same scheme, the oxygen substituent at the C-position methyl can be replaced with halo, thio, phenoxy, azido, or substituted nitrogen groups under standard Mitsunobu conditions (Mitsunobu, Synthesis 1 (1981)).
Alternatively, the hydroxyl group can be first converted to a sulfonate, halo, or other such activating group.
Thus, the process involves essentially two steps, the first step is generating a first library of n N-(substituted)-C-hydroxymethyl-oxazolidinones from a C-protected oxazolidinone and the second step is O-functionalizing acylating the C-hydroxymethyl to generate a library of n2 N-(substituted)-C-(substituted methyl)-oxazolidinones.
In a preferred embodiment, the first library comprises at least 10 different N-(substituted)-C
hydroxymethyl-oxazolidinones prepared by reacting C
(protected oxymethyl)-oxazolidinones with at least 10 different N-arylating reagents and the second library comprises at least 100 different N-(substituted)-C
(substituted)-oxazolidinones prepared by reacting the ten N-(substituted)-C-hydroxymethyl-oxazolidinones with at least ten different O-functionalizing acylating reagents. In other embodiments of the library, the substituted oxazolidinones comprise a plurality of molecules with N-position substitutions and a single substitution group at the C-position of the molecules or a plurality of molecules with C-position substitutions and a single substitution group at the N-position of the molecules. A further so embodiment of the library can comprise substituted oxazolidinones with either N-position substitutions only (N-(substituted)-C-(methyl)-oxazolidinones) or C-position substitutions only (C-(substituted methyl)-oxazolidinones. The particular library embodiment chosen depends on the particular objectives of the drug or antimicrobial screening program.
In a preferred embodiment, the oxazolidinone is 2-oxazolidinone. In a further preferred embodiment as shown in Scheme 3 below, the C-(protected hydroxymethyl)-2-oxazolidinone is C-trityloxymethyl-2 oxazolidinone. The C-trityloxymethyl-2-oxazolidinone is N-arylated (N at position 3) with a mixture of n to different aryl acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, aryl sulfonyl, or phenacyl bromides, or other such bromides under Buchwald conditions (Yin and Buchwald, Org. Letts. 2: 1101-ll04 (2000)) or simple alkylation under nitrogen to produce i5 N-(substituted)-C-trityloxymethyl-2-oxazolidinones.
The trityl (Tr) group is then removed by hydrogenation to produce a library of n N-(substituted)-C-hydroxymethyl-2-oxazolidinones.

Scheme 3 O O
RlBr...RnBr O~NH (Buchwald arylation ~ ~Rl. . . Rn O N
or simp a a y a ion Tr0 under nitrogen) Tr0 O
H2 / Pd O~N~Rl . . . Rn R~ X . . . R' X
1 n or H+
HO~
O
O~N~R1. . . Rn R' 1X . . . R' n-Y
In a further step, the N-(substituted)-C-hydroxymethyl-2-oxazolidinones are O-functionalized acylated with a mixture of n different acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, phenacyl, aryl sulfonyl, amides thereof, or other such species halides, or the O of the hydroxymethyl is replaced by N-aryl, N-sulfonyl, N-sulfide, or other N-species, or 1o the 0 is replaced by a thioalkyl, thioaryl, or other thio-species. This generates a library of n2 N-(substituted)-C-(substituted methyl)-2-oxazolidinones.
For example, if ten aryl bromides reagents are used in the first step and ten halide species reagents in the second step, a total of 100 N-(substituted)-C-(substituted methyl)-2-oxazolidinones are made. In one embodiment, the starting oxazolidinone is 4-(protected oxymethyl)-2-oxazolidinone, preferably 4-trityloxymethyl-oxazolidinone, and the n library comprises 3-(substituted)-4-hydroxymethyl-2-oxazolidinone and the n2 library comprises 3-(substituted)-4-(substituted methyl)-2-oxazolidinone.
In a preferred embodiment, the starting oxazolidinone is 5-(protected oxymethyl)-2-oxazolidinone, preferably 5-trityloxymethyl-oxazolidinone, and the n library comprises 3-(substituted)-5-hydroxymethyl-2-oxazolidinone and the n2 library comprises 3-(substituted)-5-(substituted methyl)-2-oxazolidinone.
1o The novel process allows for the rapid synthesis of a plurality of substituted oxazolidinones including but not limited to 3-(substituted)-5-(substituted methyl)-oxazolidinones, 3-(substituted)-4-(substituted methyl)-oxazolidinones, 3-(substituted)-5-(substituted methyl)-2-oxazolidinones, 3-(substituted)-4-(substituted methyl)-2-oxazolidinones, 3-(substituted)-5-hydroxymethyl-oxazolidinones, 3-(substituted)-4-hydroxymethyl-oxazolidinones, 3-(substituted)-5-hydroxymethyl-2-oxazolidinones, and 3-(substituted)-4-hydroxymethyl-2-oxazolidinones. The number of substituted oxazolidinones synthesized by the novel process depends on the number of substituting reagents included in the process. Therefore, the use of substituting reagents such as sulfur, nitrogen, and oxygen nucleophiles on the primary hydroxyl group and the amine group of the oxazolidinones affords access to a plurality of families of optically active compounds in a single process which is fast and 3o simple.
The substituted oxazolidinones produced by the above process can be separated using standard chromatography methods and the separated substituted oxazolidinones screened for biological activity including antimicrobial activity or for usefulness as a drug or an intermediate for synthesizing a drug.
Technologies and methods for screening compounds in s combinatorial libraries are well known in the art.
For any particular substituted oxazolidinone which has useful characteristics, biological activities, or which is a useful intermediate for the synthesis of other compounds, the above process for 1o making the combinatorial library is modified to a process for making the particular substituted oxazolidinone. The modified process differs from the process for preparing the library in that the plurality of reagents shown in Scheme 3 and described 15 above is replaced with the particular reagents which will result in the synthesis of the particular substituted oxazolidinone. Thus, the general method involves the following steps: (1) substituting the N-group of optically active C-protected oxazolidinone, 2o preferably trityloxy-2-oxazolidione, by simple alkylation under nitrogen or N-arylating under Buchwald conditions to produce an N-substituted oxazolidinone, (2) removing the C-protecting group, and (3) substituting the hydrogen of the C-4 or C-5 25 hydroxymethyl with an alkylation, acylation, arylation, sulfonylation, or other such species halide, or substituting the hydroxy group with a thioalkyl, thioaryl, or other thio-group species to replace the O of the hydroxymethyl with S, or 3o substituting the hydroxy group under conditions with an N-acyl, N-sulfonyl, N-sulfide, or other N-group species to replace the O of the hydroxymethyl with N.
The above process enables particular substituted oxazolidinones of any one of the eight families (Families I to VIII) to be prepared.
When libraries comprising substituted oxazolidinones prepared according to the process of the present invention were tested for antimicrobial activity, many of the substituted oxazolidinones in the libraries with the genus structure were discovered to have antimicrobial activity against gram positive and Gram negative bacteria. In particular, many of to the substituted oxazolidinones were found to be active against Gram positive bacteria such as those of the genera Staphlococcus and Enterococcus and Gram negative bacteria such as those of the genera Escherichia with 90 to 100$ Minimum Inhibitory Concentrations (MIC9o-loo) of less than 10 ug/mL. The discovery that many of the novel substituted oxazolidinones had antibacterial activity was surprising since the novel substituted oxazolidinones do not contain all three elements considered necessary 2o for antibacterial activity. Thus, the novel substituted oxazolidinones represent an new class of antimicrobial agents which are active against a variety of bacteria, in particular, Gram positive bacteria such as Staphylococcus aureus, Pseudomonas aeriginose, pneumococci (Streptococcus pneumoniae), enterococci (Enterococcus faecium, Enterococcus faecalis, Enterococcus gallinarum), Groups A, B, C, and G streptococci, Streptococcus oralis, and Streptococcus sanguis and Gram negative bacteria such 3o as Escherichia coli.
Preferably, the substituted oxazolidinones with antimicrobial activity are embraced by the species of Families I to VIII. Tables 2 and 3 show examples of substituted oxazolidinones produced as disclosed herein which have been shown to have antimicrobial activity. Tables 1 and 2 show the results of analyses of the antimicrobial activity for several of the substituted oxazolidinones. The substituted oxazolidinones which are particularly useful antimicrobials have an MIC9o_loo against at least one gram positive bacteria of about 300 ug/mL or less, preferably, of about 100 ug/mL or less, most 1o preferably, of about 10 ug/mL or less. Because particular strains of these bacteria species have developed antibiotic resistance, the novel substituted oxazolidinones are particularly useful for use against the antibiotic resistant strains of bacteria such as those shown in Table 1.
To inhibit or prevent a bacterial infection from developing in a human or animal or to treat a bacterial infection in a human or animal patient, compositions comprising a carrier and one or more of 2o the novel substituted oxazolidinones disclosed herein can be administered to the human or animal intravenously; by injection; orally by tablet, capsule, or liquid suspension; or topically.
For intravenous administration, one or more of the novel substituted oxazolidinones is dissolved in dimethyl sulfoxide or other pharmaceutically acceptable organic solvent, which is then diluted to about 5~ (v/v) in a carrier which is a sterile isotonic solution. A suitable isotonic solution so includes sodium citrate, citric acid, and dextrose wherein the Na+ content is about 0.38 mg/mL (1.7 mEq/100 mL). Linezolid in the above isotonic solution has been approved for human use by the U.S. Food and Drug Administration. The intravenous solution can be applied as 15- to 20- minute infusions or by continuous infusion over an extended time period through a catheter surgically implanted through the patient's vein. In particular embodiments, the one or more novel substituted oxazolidinones is combined with one or more antibiotics or other antibacterial agents.
For injection, one or one or more of the novel substituted oxazolidinones is dissolved in to dimethyl sulfoxide or other pharmaceutically acceptable organic solvent, which is then diluted to about 5g (v/v) in a carrier which is a sterile isotonic solution or sterile distilled water. The solution can be administered subcutaneously, intramuscularly, or peritoneally. In particular embodiments, one or more the substituted oxazolidinones is combined with one or more antibiotics or other antibacterial agents.
For oral administration, one or more of the 2o novel substituted oxazolidinones is mixed with a pharmaceutically acceptable carrier and the mixture compressed into a tablet, which can be film coated, or encapsulated within a pharmaceutically acceptable capsule. For example, one or more of the novel oxazolidinones are admixed with a carrier which includes as the inactive ingredients: corn starch, microcrystalline cellulose, hydroxy propylcellulose, sodium starch glycolate, magnesium stearate, hydroxypropyl methylcellulose, polyethylene glycol, so titanium dioxide, and carnauba wax. The admixture is formed into tablets or encapsulated in capsules. Each tablet or capsule contains about 0.1 mEq Na+.
Linezolid in a carrier which includes the above inactive ingredients has been approved for human use by the U.S. Food and Drug Administration. In particular embodiments, one or more of the substituted oxazolidinones is combined with one or more antibiotics or other antibacterial agents.
Alternatively, the novel substituted oxazolidinones are administered orally as a suspension. In this embodiment, one or more of the novel substituted oxazolidinones is provided in a to pharmaceutically acceptable flavored granule or powder carrier for constitution into a suspension for oral administration. For example, one or more of the novel substituted oxazolidinones are admixed with a granule or powder which includes as the inactive ingredients:
sucrose, citric acid, sodium citrate, microcrystalline cellulose, carboxy methyl cellulose sodium, aspartame, xanthan gum, mannitol, sodium benzoate, colloidal silicon dioxide, sodium chloride, and flavors.
Linezolid in a granule or powder containing the above 2o inactive ingredients has been approved for human use by the U.S. Food and Drug Administration. In particular embodiments, one or more of the substituted oxazolidinones is combined with one or more antibiotics or other antibacterial agents.
For topical administration, one or more of the substituted oxazolidinones can be provided in an ointment, a lotion, a cream, or a gel. In particular embodiments, one or more of the substituted oxazolidinones is combined with one or more steroids, 3o one or more antibiotics or other antibacterial agents, or both.
The following examples are intended to promote a further understanding of the present invention.

This example illustrates the preparation of (S)-5-trityloxymethyl-2-oxazolidinone using the process disclosed in U.S. Patent No. 6,288,239 B1 to Hollingsworth and Wang.
In a flask, (S)-3,4-dihydroxybutyramide (11.9 g, 0.10 moles) was dissolved in 50 mL of 1o tetrahydrofuran (THF) to which 50 mL of dimethylformamide and 10 mL pyridine was added followed by 30.6 g (0.11 moles) of trityl chloride. A
drying tube filled with calcium chloride was used to exclude moisture. The reaction mixture was stirred for 36 hours at room temperature. Afterwards, the reaction mixture was filtered to remove the solids.
The liquid was concentrated under reduced pressure to remove most of the solvent. The concentrate was poured into ice water, stirred for about half an hour, 2o and then the water layer was removed from the organic layer containing the 3-Hydroxy-4-trityloxy butyramide.
The product was a semi-crystalline liquid which was dried in vacuo. Afterwards, the excess trityl chloride was washed away by tituration with hexane.
The 3-hydroxy-4-trityloxy butyramide (3.61 g, 0.01 moles) was dissolved in 30 mL THF. Fifteen mL
of a 13~ sodium hypochlorite solution was added and the mixture was stirred vigorously. Next, 1.6 g of sodium hydroxide dissolved in 10 mL of water was 3o added. The reaction was stirred at 55-60° C for eight hours after which time the conversion to 5-trityloxymethyl-2-oxazolidinone was completed as indicated by TLC and 1H-NMR spectroscopy. The organic layer was separated from the aqueous layer and saved.
The aqueous layer was extracted three times with THF.
The saved organic layer and the THF extracts were combined and then concentrated to remove the solvent.
The residue was taken up in dichloromethane and the solution dried over sodium sulfate. Afterwards, the solution was concentrated to remove the solvent and the oxazolidinone was obtained as a white crystalline product (3.4 g, yield 95~). Normally, this crude 1o product did not need further purification.

This comparative example illustrates the N
arylation of 5-trityloxymethyl-2-oxazolidinone to produce (S)-3-(2-nitro)phenyl-5-trityloxymethy)-2 oxazolidinone using the procedure disclosed in Shakespeare, Tetrahedron Lett. 40: 2035-2038 (1999).
To 36 mg of 5-trityloxymethyl-2-oxazolidinone, 30 mg (1.5 equivs) 1-bromo-2 2o nitrobenzene, 2.4 mg (0.1 equivs) palladium (II) acetate, 5.5 mg (0.1 equivs) 1,1' bis(diphenylphosphino)-ferrocene, 16 mg (0.15 equivs) potassium t-butoxide, and 1 mL toluene were added under a nitrogen atmosphere. The mixture was heated at 110° C for 14 hours after which time the mixture was resolved by thin-layer chromatography (TLC) comprising silica with dichloromethane as the eluant.
The TLC indicated complete conversion to a single product: (S)-3-(2-nitro)phenyl-5 3o trityloxymethyl-2-oxazolidinone. The mixture was cooled and diluted with dichloromethane. The dark brown organic solution was washed with 5~ sodium carbonate, concentrated, and chromatographed on silica gel using dichloromethane as the eluant. The product (47 mg, 98$.) was obtained as a pale yellow solid which crystallized from chloroform: methanol as off-white crystals with a melting point of 236-237° C. The product was analyzed by 1H-NMR, 13C NMR, IR, MS, and HRMS.
1H-NMR (300 MHz, CDC13) b 8.03 (dd, 1H, J--8.0, 2.1 Hz), 7.65 (td, 1H, J--8.0, 2.1 Hz), 7.46-7.53 (m, 6H), 7.44 (td, 1H, J--8.0, 2.1 Hz), 7.20-7.36 to (m, 10H), 4.83 (m, 1H), 4.07 (t, 1H, J--8.5 Hz), 3.89 (t, 1H, J--8 Hz), 3.58 (dd, 1H, J--11.8, 4.5 Hz), 3.36 (dd, 1H, J--11.8, 4.5 Hz) . 13C NMR (75 MHz, CDC13) b 176.4, 143.5, 134.1, 131.7, 127.6, 87.5, 73.4, 64.0, 49.4. IR cm 1 3057, 2924, 1760, 1607, 1532, 1489, 1449, 1411, 1355. MS (electron impact) m/z 57, 71, 91, 105, 131, 165, 243, 259, 403, 463, 480 (M+) . HRMS
(electron impact) analyzed for C29H24N205: theoretical MW 480.1685, observed MW 480.1683.

This example illustrates the preparation of a library of substituted 2-oxazolidinones, which are members of Family II, according to the process of the present invention. In this example, (S)-5-trityloxymethyl-2-oxazolidinone is N-acylated with 2,5-dimethoxyphenacyl bromide, detritylated, and then acylated with ten different acyl halides or anhydrides to produce a library of n=10 3-(2,5 dimethoxyphenacyl)-5-(substituted methyl)-2 oxazolidinones.
In the first step (Figure 1A), (S)-3-(2,5-dimethocyphenacyl)-5-trityloxymethyl-2-oxazolidinone was produced in a reaction comprising (S)-5-trityloxymethyl-2-oxazolidinone and the aryl bromide:
2,5-dimethoxyphenacyl bromide. To a solution of 3.59 g (10 mmoles) of(S)-5-trityloxymethyl-2-oxazolidinone (MW 359.2) in 40 mL THF at 4° C, 400 mg (10 mmoles) NaH
(MW 24) as a 60g suspension in hexane was added. The reaction mixture was stirred for about 10 minutes under nitrogen at 0° C and then warmed up to room temperature and stirred for an additional two hours.
Then, 2.59 g (10 mmoles) of 2-bromo-4'dimethoxyacetophenone (MW 259.1) was added and the reaction mixture stirred at room temperature for about eight hours. Afterwards, the reaction was quenched by adding 20 mL 20$ NHqCl. The organic layer was removed and saved. The aqueous layer was extracted two times with 40 mL aliquots of THF. The THF extracts were combined with the saved organic layer and the mixture dried with 2.5 g anhydrous Na2S09. The mixture was then concentrated in vacuo to provide a crude product.
The crude product was purified by flash column 2o chromatography using 40o EtOAc:Hexane followed by 60~
EtOAc:Hexane. This produced 2.73 g (51~ yield) of the (5)-3-(2,5-dimethocyphenacyl)-5-trityloxymethyl-2-oxazolidinone (product)(MW 537.6). The product was compared to the starting material by TLC using 40~
EtOAC/Hexane as the solvent. The Rf of the starting material was 0.2 and the Rf of the product was 0.4.
In the second step (Figure 1B), the trityl group was removed from the ( S ) -3- ( 2, 5-dimethocyphenacyl)-5-trityloxymethyl-2-oxazolidinone.
3o To 1. 07 g (2. 0 mmoles) of (S) -3- (2, 5-dimethocyphenacyl)-5-trityloxymethyl-2-oxazolidinone in wet CH2C12 (8 mL CH2C12, 1 mL H20) , 0.14 mL CF3COZH
(210 mg, 1.8 mmoles) (MW 114.02) was added and the reaction mixture stirred for about four hours.
Afterwards, the reaction was quenched by adding 0.2 mL
triethylamine and the reaction mixture concentrated in vacuo. The residue was purified by flash chromatography to produce 472 mg (80$ yield) of (S)-3-(2,5-dimethocyphenacyl)-5-hydroxymethyl-2-oxazolidinone (product) (MW 295.29). The product was compared to the starting material by TLC using 800 EtOAC:Hexane as the solvent. The Rf of the starting 1o material was 0.7 and the Rf of the product was 0.1.
In the third step (Figure 1C), the library of ten substituted 2-oxazolidinones was produced in a reaction comprising the (S)-3-(2,5-dimethocyphenacyl)-5-hydroxymethyl-2-oxazolidinone and the ten different acetyl chlorides shown in Figure 2. To about 295 mg (1.0 mmoles) of (S)-3-(2,5-dimethocyphenacyl)-5-hydroxymethyl-2-oxazolidinone in dry CH2C12 (8 mL
CHZC12 ) , 1. 0 equiv. ( 1.1 mmoles ) of pyridine was added and the reaction mixture stirred at room temperature.
2o To this reaction mixture was added 1.0 equiv. of a mixture of ten different acetyl chlorides. The reaction was stirred overnight at room temperature.
Afterwards, TLC of an aliquot indicated that complete conversion of the (S)-3-(2,5-dimethocyphenacyl)-5-hydroxymethyl-2-oxazolidinone to (S)-3-(2,5-dimethocyphenacyl)-5-(substituted methyl)-2-oxazolidinone had occurred. Therefore, about 3 mL of 20o NH9C1 was added to the reaction mixture and the organic layer removed and saved. The aqueous layer 3o was extracted two times with 40 mL aliquots of CH2C12.
The CHZC12 extracts were combined with the saved organic layer and the mixture dried with 2.5 g anhydrous NaZS09. The mixture was then concentrated in vacuo to provide a crude product. The crude product was analyzed by 1H-NMR, 13C NMR, HPLC, and TLC using a EtOAc:hexane (2:1) solvent system.
An HPLC profile of the (S)-3-(2,5 dimethocyphenacyl)-5-(substituted methyl)-2 oxazolidinone products made is shown in Figure 3. The products represented by the peaks in the HPLC are shown in Figure 4. This example illustrates the principle of the present invention. As shown by this 1o example, providing n=10 acetyl halides in a single reaction produces 10 (S)-3-(2,5-dimethocyphenacyl)-5-(substituted methyl)-2-oxazolidinone products. If n=10 aryl bromides had been used as well to arylate the N at the 3-position, the process would have generated 100 (S)-3-(substituted)-5-(substituted methyl)-2-oxazolidinone products.

The substituted oxazolidinone (S)-3-(3,3-2o dimethyl-2-butone)-5-(4-nitro-benzenesulfonyloxymethyl)-2-oxazolidinone is prepared as follows.
In the first step, (S)-3-(3,3-dimethyl-2-butone)-5-trityloxymethyl-2-oxazolidinone is produced in a reaction comprising (S)-5-trityloxymethyl-2-oxazolidinone and BrCH2COC(CH3)3. To a solution of about 10 mmoles of (S)-5-trityloxymethyl-2-oxazolidinone in 40 mL tetrahydrofuran (THF) at 4° C, mmoles NaH as a 60$ suspension in hexane is added.
The reaction mixture is stirred for about 10 minutes under nitrogen at 0° C and then warmed up to room 5 temperature and stirred for an additional two hours.
Then, about 10 mmoles of BrCH2COC (CH3) 3 is added and the reaction mixture stirred at room temperature for about eight hours. Afterwards, the reaction is quenched by adding 20 mL 20$ NH9C1. The organic layer 1o is removed and saved. The aqueous layer is extracted two times with 40 mL aliquots of THF. The THF
extracts are combined with the saved organic layer and the mixture dried with 2.5 g anhydrous Na2S09. The mixture is then concentrated in vacuo to provide (S) 1s 3-(3,3-dimethyl-2-butone)-5-trityloxymethyl-2 oxazolidinone as a crude product. The crude product is purified by flash column chromatography using 40$
EtOAc:Hexane followed by 60$ EtOAc:Hexane. The product is compared to the starting material by TLC
2o using 40$ EtOAC/Hexane as the solvent.
In the second step, the trityl group is removed from the (S)-3-(3,3-dimethyl-2-butone)-5-trityloxymethyl-2-oxazolidinone. To about 2.0 mmoles of the crude product in wet CH2C12 ( 8 mL CH2C12, 1 mL
2s H20) , 0 .14 mL CF3COZH ( 1. 8 mmoles ) is added and the reaction mixture stirred for about four hours.
Afterwards, the reaction is quenched by adding 0.2 mL
triethylamine and the reaction mixture concentrated in vacuo. The residue is purified by flash 3o chromatography to produce (S)-3-(3,3-dimethyl-2-butanone)-5-hydroxymethyl-2-oxazolidinone. The product can be compared to the starting material by TLC using 80$ EtOAC:Hexane as the solvent to determine the yield.
In the third step, the (S)-3-(3,3-dimethyl-2-butone)-5-(4-vitro-benzenesulfonyloxymethyl)-2-oxazolidinone is produced in a reaction comprising the (S)-3-(3,3-dimethyl-2-butanone)-5-hydroxymethyl-2-oxazolidinone and nitrobenzenesulfonyl chloride. To about 1.0 mmoles of the ((S)-3-(3,3-dimethyl-2-butanone)-5-hydroxymethyl-2-oxazolidinone in dry CH2C12 (8 mL CH2C12) , 1.0 equiv. (1.1 mmoles) of pyridine is to added and the reaction mixture stirred at room temperature. To this reaction mixture is added 1.0 equiv. of compound nitrobenzenesulfonyl chloride. The reaction is stirred overnight at room temperature.
Afterwards, an aliquot of the reaction is analyzed by TLC to determine whether complete .conversion of the (S)-3-(3,3-dimethyl-2-butanone)-5-hydroxymethyl-2-oxazolidinone to the substituted oxazolidinone has occurred. Thereafter, about 3 mL of 20~ NH9C1 is added to the reaction mixture and the organic layer is 2o removed and saved. The aqueous layer is extracted two times with 40 mL aliquots of CHzCl2. The CH2C12 extracts are combined with the saved organic layer and the mixture is dried with 2.5 g anhydrous Na2S09. The mixture is then concentrated in vacuo to provide a crude product of the substituted oxazolidinone. The crude product is analyzed by 1H-NMR, 13C NMR, HPLC, and TLC using a EtOAc:hexane (2:1) solvent system and is further purified by standard chromatography methods.

The substituted oxazolidinone (S)-3-(3,3-dimethyl-2-butone)-5-(4-isocyanobenzenesulfonyloxymethyl)-2-oxazolidinone N=C=0 O
is prepared as follows.
(S)-3-(3,3-dimethyl-2-butone)-5-hydroxymethyl-2-oxazolidinone is prepared as in Example 4. Then the (S)-3-(3,3-dimethyl-2-butone)-5-(4-isocyanobenzenesulfonyloxymethyl)-2-oxazolidinone is produced in a reaction comprising the (S)-3-(3,3-dimethyl-2-butanone)-5-hydroxymethyl-2-oxazolidinone and isocyanobenzenesulfonyl chloride as follows.
1o To about 1. 0 mmoles of the ( (S) -3- (3, 3-dimethyl-2-butanone)-5-hydroxymethyl-2-oxazolidinone in dry CH2C12 (8 mL CHZC12), 1.0 equiv. (1.1 mmoles) of pyridine is added and the reaction mixture stirred at room temperature. To this reaction mixture is added 1..0 equiv. of isocyanobenzenesulfonyl chloride. The reaction is stirred overnight at room temperature.
Afterwards, an aliquot of the reaction is analyzed by TLC to determine whether complete conversion of the (S)-3-(3,3-dimethyl-2-butanone)-5-hydroxymethyl-2-oxazolidinone to the substituted oxazolidinone has occurred. Thereafter, about 3 mL of 20$ NH9C1 is added to the reaction mixture and the organic layer is removed and saved. The aqueous layer is extracted two times with 40 mL aliquots of CH2C12. The CH2C12 extracts are combined with the saved organic layer and the mixture is dried with 2.5 g anhydrous Na2S09. The mixture is then concentrated in vacuo to provide a crude product of the substituted oxazolidinone. The crude product is analyzed by 1H-NMR, 13C NMR, HPLC, and TLC using a EtOAc:hexane (2:1) solvent system and is further purified by standard chromatography methods.

s The substituted oxazolidinone (S)-3-(3,3-dimethyl-2-butone)-5-(7-chloro-2,1,3-benzoxadiazole-4-sulfonyloxymethyl)-2-oxazolidinone (34) ~O~
is prepared as follows.
(S)-3-(3,3-dimethyl-2-butone)-5-hydroxymethyl-2-oxazolidinone is prepared as in Example 4. The substituted oxazolidinone 34 is then produced in a reaction comprising the (S)-3-(3,3-dimethyl-2-butanone)-5-hydroxymethyl-2-oxazolidinone and 7-chloro-2,1,3-benzoxadiazole-4-sulfonyl chloride as follows.
To about 1. 0 mmoles of the ( (S) -3- (3, 3-dimethyl-2-butanone)-5-hydroxymethyl-2-oxazolidinone in dry CH2C12 (8 mL CH2C12), 1.0 equiv. (1.l mmoles) of 2o pyridine is added and the reaction mixture stirred at room temperature. To this reaction mixture is added 1.0 equiv. of compound 7-chloro-2,1,3-benzoxadiazole-4-sulfonyl chloride. The reaction is stirred overnight at room temperature. Afterwards, an aliquot of the reaction is analyzed by TLC to determine whether complete conversion of the (S)-3-(3,3-dimethyl-2-butanone)-5-hydroxymethyl-2-oxazolidinone to the substituted oxazolidinone 34 has occurred.

Thereafter, about 3 mL of 20~ NH9C1 is added to the reaction mixture and the organic layer is removed and saved. The aqueous layer is extracted two times with 40 mL aliquots of CH2C12. The CH2C12 extracts are combined with the saved organic layer and the mixture is dried with 2.5 g anhydrous Na2S04. The mixture is then concentrated in vacuo to provide a crude product of the substituted oxazolidinone 34. The crude product is analyzed by 1H-NMR, 13C NMR, HPLC, and TLC
1o using a EtOAc:hexane (2:1) solvent system and is further purified by standard chromatography methods.

The substituted oxazolidinone (S)-3-(3-butene-2-one)-5-(7-chloro-2,1,3-benzoxadiazole-4-sulfonyloxymethyl)-2-oxazolidinone is prepared as follows.
In the first step, (S)-3-(3-butene-2-one)-5 2o trityloxymethyl-2-oxazolidinone is produced in a reaction comprising (S)-5-trityloxymethyl-2 oxazolidinone and BrCHCHCOCH3 . To a solution of about 10 mmoles of (S)-5-trityloxymethyl-2-oxazolidinone in 40 mL tetrahydrofuran (THF) at 4° C, 10 mmoles NaH as a 60o suspension in hexane is added. The reaction mixture was stirred for about 10 minutes under nitrogen at 0° C and then warmed up to room temperature and stirred for an additional two hours. Then, about mmoles of BrCHCHCOCH3 is added and the reaction mixture stirred at room temperature for about eight hours. Afterwards, the reaction is quenched by adding mL 20~ NHQC1. The organic layer is removed and 5 saved. The aqueous layer is extracted two times with 40 mL aliquots of THF. The THF extracts are combined with the saved organic layer and the mixture dried with 2.5 g anhydrous Na2S0q. The mixture is then concentrated in vacuo to provide (S)-3-(3-butene-2-10 one)-5-trityloxymethyl-2-oxazolidinone as a crude product. The crude product is purified by flash column chromatography using 40~ EtOAc:Hexane followed by 60~ EtOAc:Hexane. The product is compared to the starting material by TLC using 40$ EtOAC/Hexane as the 15 solvent.
In the second step, the trityl group is removed from the (S)-3-(3-butene-2-one)-5 trityloxymethyl-2-oxazolidinone as in Example 4 to produce (S)-3-(3-butene-2-one)-5-hydroxymethyl-2 20 oxazolidinone.
In the third step, the substituted oxazolidinone is produced in a reaction comprising the (S)-3-(3-butene-2-one)-5-hydroxymethyl-2-oxazolidinone and 7-chloro-2,1,3-benzoxadiazole-4-sulfonyl chloride.
To about 1.0 mmoles of the ((S)-3-(3-butene-2-one)-5-hydroxymethyl-2-oxazolidinone in dry CH2C12 (8 mL
CHZC12) , 1. 0 equiv. (1.1 mmoles) of pyridine is added and the reaction mixture stirred at room temperature.
To this reaction mixture is added 1.0 equiv. of 7-3o chloro-2,1,3-benzoxadiazole-4-sulfonyl chloride. The reaction is stirred overnight at room temperature.
Afterwards, an aliquot of the reaction is analyzed by TLC to determine whether complete conversion of the (S)-3-(3-butene-2-one)-5-hydroxymethyl-2-oxazolidinone to the substituted oxazolidinone has occurred.
Thereafter, about 3 mL of 20$ NHqCl is added to the reaction mixture and the organic layer is removed and saved. The aqueous layer is extracted two times with 40 mL aliquots of CH2C12. The CH2C12 extracts are combined with the saved organic layer and the mixture is dried with 2.5 g anhydrous Na2S04. The mixture is then concentrated in vacuo to provide a crude product to of the substituted oxazolidinone. The crude product is analyzed by 1H-NMR, 13C NMR, HPLC, and TLC using an EtOAc:hexane (2:1) solvent system and is further purified by standard chromatography methods.

The substituted oxazolidinones were tested for antimicrobial activity as follows.
The following American Type Culture Collection (ATCC, 10801 University Boulevard, 2o Manassas, Virginia) quality control strains were selected for the initial screening of the antimicrobial properties of the substituted oxazolidinones as suggested by the NCCLS:
Enterococcus faecalis 29212, Escherichia coli 25922, Pseudomonas aeruginosa 27853, and Staphylococcus aureus 29213. See, NCCLS document M7-A5. Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard-Fifth Edition. Volume 20, Number 2. January 2000, (ISBN 1-56238-394-9) and Lorian V. Antibiotics.
Laboratory Medicine. 4th ed. Baltimore: Williams and Wilkins, pp. 52-111 (1996). For subsequent testing, the following strains have been used: Staphylococcus aureus NRS4 (992; HIP5836; New Jersey) (Smith et al., New Engl. J. Med. 340: 493-501 (1999); Tenover et al., J. Clin. Microbiol. 36: 1020-1027 (1998)), Staphylococcus aureus NRS3 (963sm; HIP5827; Michigan) (Smith et al., ibid.; Tenover et al., ibid.), Staphylococcus aureus NRS103 (Becker) (Karakawa and Vann, Sem. Infect. Dis. 4: 285 (1982), Staphylococcus aureus NR5102 (Reynolds) Karakawa and Vann, ibid;
McMurray et al., JID 162: 759-762 (1990)), 1o Staphylococcus epi dermidis NRS101 (ATCC 35984), Streptococcus pneumoniae (ATCC 49619), Enterococcus faecalis (ATCC 51299) , and Staphylococcus aureus (ATCC
43300.
Stock cultures of these strains were obtained by seeding colonies from overnight streak plates (Tryptic Soy Agar II (TSAII) and 5$ sheep's blood (SB) agar plates: Becton Dickinson, CA#2211261) into sterile Mueller-Hinton Broth (MHB)(Becton Dickinson, CA#211443) and growing the suspensions to 2o mid-late log phase in 13 mL screw-cap tubes. Glycerol (Sigma, CA#G-6279) was sterilized by autoclaving for 15 minutes at 121° C in 6 mL volumes, and then stored at 2-8° C. This was diluted to 20$ in dH20, and then added 1:1 (250 uL:250 uL, 10$ final glycerol concentration) to the logarithmically growing bacterial suspension of each strain. Tubes were frozen and stored at -70° C. Purity of the stock cultures was tested by thawing one tube of each strain in a water bath at 37° C and plating them on TSAII with 5$ SB. Plates were incubated overnight at 37° C and colonies were examined for morphology. Growth curves and approximate CFU/mL were also obtained.

DMSO susceptibility determinations were performed as follows. DMSO (Alfa Aesar, CA#22914) was diluted to 2x the final starting concentration of 20$
in MHB, pH 7.36 (this was the consistent pH value of MHB)(should be between 7.2-7.4 according to NCCLS).
Two-fold serial dilutions were performed in 15 mL
conical tubes and poured into sterile reservoirs.
Using an 8-channel micropipetman, 50 uL from each reservoir was transferred to every well in the 1o corresponding column (1-11) of a sterile 96-well, U-bottom microplate (Nalge Nunc, Intl., CA#262162). As a positive growth control, 50 uL of MHB alone was added to each well of Column 12. Bacteria were grown overnight on TSAII + 5~SB, and 3-4 colonies were seeded into 6 mL of sterile MHB in 13 mL screw cap tubes. Tubes were grown at 35° C to mid-log phase, and were diluted to an optical density of 0.12 at 625nm (or approx. 1x108 CFU/mL), using 0.9~ sterile saline.
This solution was further diluted 1:100 with 0.9$
2o sterile saline (1x106 CFU/mL), and 50 uL was added to each well for a final inoculum of 1x105 CFU/mL. As a negative growth control, well H12 was inoculated only with 0.9~ sterile saline. The plate was tightly fitted with sealing tape (Corning Costar, CA#3095) and was incubated for a period of 18 hours at 35° C, after which growth was observed. 2.5o DMSO was determined to be the smallest concentration of DMSO to exhibit no visual effects on bacterial growth as compared with the positive controls for all strains tested. This 3o was confirmed by performing colony counts to assess cell viability in the presence of DMSO. For each strain, 10 uL was removed from one of the inoculated wells containing 2.5~, 0.15, and 0~ DMSO (after mixing). This was diluted 1:100000 in sterile 0.9~
DMSO, plated on TSAII + S~SB, and grown overnight at 35° C. Plates were then observed for differences in the number of viable colonies (theoretically, each colony arises from a single cell) based on the varying concentrations of DMSO. No differences were observed.
High purity substituted oxazolidinones prepared according to the method of the present invention and a ZYVOX standard (ZYVOX is a trade name 1o for linezolid available from Pharmacia Corporation) were provided by Synthon Corporation, Monmouth Junction, NJ. Compounds were dissolved at 10 mg/mL in DMSO, as after a dilution of approx. 39.0 to reach the desired final starting concentration of 256ug/ml, the concentration of DMSO is approximately 2.5~.
Compounds were then stored at room temperature (25° C) in the dark.
Antimicrobial susceptibility screening was as follows. All compounds were initially screened for 2o activity in duplicate at 256 ug/mL (2.5~ DMSO), including ZYVOX, the positive control for antimicrobial activity. A single well of bacterially inoculated 2.5g DMSO served as a positive control for bacterial growth, while a well of DMSO inoculated with , of 50 uL of sterile 0.9~ saline served as a negative growth control. Controls were prepared on every microplate, so that 46 was the maximum number of compounds that were screened per plate. Broth was pipetted into sterile microcentrifuge tubes, to which 3o the compounds were then added (1:19.53 dilution or 2x final concentration) . Each tube was vortexed, and 50 uL was immediately transferred to the microplate wells for each strain. Solubility was assessed by visual observation. MB (medium broth solubility) was recorded if the solution appeared only slightly cloudy. LB (low broth solubility) was recorded if the solution was extremely cloudy, and especially if larger, clumpy precipitates formed. Bacteria were grown as described above, although absorbance was measured at 650 nm and bacteria were diluted to an initial OD of 0.12-0.15. Bacteria were then further diluted 1:100 with 0.9~ sterile saline. Within 15 1o minutes of this final dilution, 50 uL of this suspension was added to each well for a final volume of O.1 L, and a final bacterial concentration of 1x105 CFU/mL (except for the negative control well). Plates were grown as above and all observations were recorded.
Compounds exhibiting activity at this concentration were then screened for their MICs (minimum inhibitory concentration) in duplicate, at concentrations ranging from 256-0.25 ug/mL (columns 1-11) using the broth microdilution method. Two-fold dilutions were obtained using an 8-channel micropipetman and tips were changed between each column transfer, after mixing 10 times and expelling the maximum amount of fluid. The MIC was defined as the lowest concentration of test compound that inhibited visible growth after a period of 18 hours incubation at 35° C . Bacteria were prepared, and the microwell plates were incubated as described above.
In addition, bacteria were routinely sampled before 3o addition to the wells. From the pre-well concentration, samples were diluted 1:200 with 0.90 sterile saline. At this concentration, 100 uL was plated and the remaining sample was diluted 1:10. 100 uL of this was then also spread-plated onto TSAII + 5$
SB and grown overnight at 35° C. Colonies were counted and multiplied by the dilution factor to obtain starting CFU/mL. As a positive MIC control, ZYVOX was tested in parallel for each strain, and the dilution of 5$ (2x final concentration) DMSO served as a positive growth control. To control for variation between the volume transferred by each tip of the 8-channel micropipetman, 5~ (2x final concentration) 1o DMSO was added to column 12 using the micropipetman.
All wells, but that of the negative control (H12), were inoculated with the same bacterial suspension resulting in a final starting concentration of 1x105 CFU/mL. The negative control was inoculated with 0.90 sterile saline.
At this time, a total of 1625 substituted oxazolidinones have been successfully screened for activity at 256 ug/mL (Compounds 1422, 1474, 147$, 1595 were absent). Of these, 71 were tested against 2o Staphylococcus aureus (Gram positive), 60 were tested against Enterococcus faecalis (Gram positive), and one compound tested against Escherichia coli (Gram negative) have proven effective with MICs at or below 256 ug/mL.
Tables 1 and 2 show the antimicrobial activity for several of the substituted oxazolidinones. Table 1 further shows that several were also able to inhibit the growth of myeloid, erythroid, and megakaryocytic cells. Table 3 shows 3o several substituted oxazolidinones which have been found to be particularly antimicrobial. In general, many of the substituted oxazolidinones were as effective as ZYVOX. Thus, the results show that many of the substituted oxazolidinones prepared according to the process herein have antimicrobial applications, in particular, as antimicrobial agents against drug resistant strains of gram positive bacteria.

U

O

aoao o y~o o 0 0 o O ' ''1 t0 . .. . ~I

H O O O O01M ~ M w ?G ~
,~ U

rt N

'~ W
~

-1 r~
W
j ~ -~
a N m a o ~U ~ o p r 0 0o N o ~
U

H . N ~

U J-1 ~ O O OO O ~O r1 U

W ri O

r-Ila i~

U

b ~ '~ -.-1 W

N M N ~N O M V'N

N

,~., ~, W

-~1 W

M d'OD~ d'O d'~ ri P4 00 t0 M w O

sa ,~ .~1 a OGo0oM H 1 N ~ M M Mr1 .,1 ~ .1 ~n ' ~' a ~
~a o a ~ r n ~n .R rs U ~o a 'E-~ W o o 0 00~o o ~
H C'C'DOV' O N~l'l N Or1U
M .,..) U

a rir1N A.'"1 U
M O

N

ODODN N N NN N N~ ~i G' v H o a ~1 ~ -r-i v aototo~o toto U ~ N N MM MV'N 1N u7N u7u~N ~

V V'N NN N N

-rl a ~, N ' ~ ' 1 ~ N M MC MV V 'N ~N u MN i~
'T ' J , .. VN NN N N (n ~ r~

h -.d m .

N
M f"1 w V ' ' ' '0 00 0 0 o ,. ~
-~
N

W E N V VN VN N dInIn1nIn N ~N
N NN N ~i N

N N

.,..1 U

?~
a U ~I~I 'lalH '10l .r1 N N N N crN V' V' N O
~ a a'a ~ov'~ N ~ v' '-i rn ~c U
.-1 -I

N . ~
.
, .. p ,U
, 1 rt 'd r-I

-.~i~h I(1I(10001CDInGDr1N1pn M r1O ~
' N

f0 OODO e1O OI~O NN ~N O N t~
1 t0~ ~

1 1 ~CDCDN V ~O v-1,..1N N 01 ~ H H v-1eWiN N Ne~lr1 V O N fn U r1 Table 2 MIC~~~~~(ug/mL) MICI~.i~~(uglmL) COMPOUND COMPOUND # SA-ATCC29213 EF-ATCC 29212 3a zso CI N N ~-O~N
O
b 108 62.5 ~N' /
~0 OSO \ / NOz 0 110 31.3 ~N ~0 NOz 0 a 126 15.6 Ø
N' N O O~~~N
\ / S-O.r"' CI
0 ,0~ 235 62.5 ~N~O N~ /N
O OSO ~ / Ci 0 236 31.3 ~.--N ~0 ~0 OSO \ / NOz 0 ~ 250 250 0 0 ~cN
ozN \ / o-o.
O 253 15.6 O J( 0zN ~ / S_O~N
O
254 125 _ H~CO
~S~
0 ,0~ 255 62.5 N\ ~N
-N
O \ / ~f 250 2~ 250 CI / \ 's-o~~

Hb~N
266 15.6 N _ ob \ / No, o p OzN \ / ~-O~N
O

~N'~l NP N
0~~ ~ 1 28S 62.5 C~Hp N~ \
'--LO-5 ~ ~ I
0"0 250 CI ~ ~ ~-O~N~O~
N ~N O O
b 0 294 31.3 ~N
~O.~S~ ~ ~ 0:
O O

p ,O~ 323 2S0 H9CO~N~p N\ /N
~J ~--~O-S ~ ~ I
° ° 125 0 324 62.5 , 0 O O~N
a ~N'~~~ O~'C~CH
" 250 334 62.5 _ o o-C° o OZN ~ ~ S-0~ IN ~

q,Nu~'T5~
F Exit Afac~ 469.01 O B2-Hp-KIO
31.3 o E5t.lp-Ia Ct,H uCIFN, F Exact Maa,: 9.01 62.5 _ 0 O
xN
OpN ~ ~ S-O~, O

O=N ~ / 1~~ N

0 669 62.5 _ o o- ~~
OZN ~ ~ S~~N~CH~

N~ N O O

CI ~ ~ 5-O~N~CH~

O ~ ~" 695 62.5 O O~N~OEt ci i ~ 'S~°y O 771 15.6 02N / \

_ 0 H J( 02N ~ ~ S-N J~,N

o N ~o ~ 870 16 OzN / \ ~~~N
O
NOZ

N$'N
0 0 N-~4Ha \ /

02N ~ / ~-0, ~ N.,i~'~

0_~ N ~
C I \ / ~'-O ~
O O~ 929 64 02N \ / 'S'-0~N~
O

_ 0 J(N
02N ~ ~ S-0~, ~

_ 0 J( 02N ~ ~ S-O.~rN '~

0,, p"N
o,r,~8-o _ 0 0- ~
p2N ~ ~ S~~N~NMe2 N~ N 0 0 O O- II
CI \ / S-0~N~NMe=
O

CI ~ ~ S-O~N~CH~

OZN

0 O--I~ O
02N ~ ~ S_O~N

N\0 N O O

CI ~ ~ S-O~N
O

cl ~ ~ ~-o~,N

O O -~
O ~~N
i O

=N / \ ~1 _o ~Et Exact Mass: 400.0 ~0 02N \ / ~-O~N~
O

CI ~ ~ S-0 OxN~
n ~0 Nb N 128 p 1210 256 0 J( CI ~ ~ S-O~j ,N~

OZN
O ~0~ 1411 64 CHs N\~/ N\ ~N
~O OSO ~ ~ 1 C ~N(CHa)a Exact Mala: 302.00 CI

~Q[/~~ /C ~N(CHa)a / Exla1 Maaa: 101.V1f1O
P~~B

aN ~ ~ ~-0~
~N(GNS)a Exec Mess:399.0 U117 a 180s s 0=N ~ ~ N ~p ~O-S ~ ~ NO=

2H ~
~0~ ~ / NOT

o,N ~ ~
~o ~\
y0 H

OZN ~ ~ N- 'p ~O w N
I0I 1985 $
I .NN

CI ~ ~ ~-O~H
O ~ HO= 2017 16 o~N
~pN ~ . ~y Enact M aas: 405.06 o e, 2019 16 y~
O=N-~S E:act M acs: 437.99 at 2020 64 IN
~Esact M eee: d50.01 o~N ~~
y o OZN ~ EzactMass:338.06 32 0t1 2025 64 O~N~OEt J--' o OZN ~ Enact Mass: 368.07 32 Ta~.e 3 FactemaIIDinternaIIDStructure CC 001 26 N~

. ~
o 0 s / \
~ \ Nip SCC 002 207 , ~o ~o 0 Nb,N

~

SCC 003 253 o N

F

SCC 005 1021 N~N~o~'s~'~
/ \ No2 o 0 0~( CC 006 192 o2N \ / s-oJ~,N~

O
SCC 007 1687 OZN ~ ~ S-N~,N-SCC 008 1705 02N ~ ~ S-N'jL, JAN

SCC 009 1715 02N ~ \ S-N~,N~

O
SCC010 1808 / \ ~o-s / \ N~

SCC 011 1809 / \

O
SCC 012 2278 / \ ~~S

O
SCC 013 2405 oZN ~ ~ s-o~,N~

O

SCC 014 2428 02N ~ ~ s-N~,N

O

SCC 015 2570 pZN ~ ~ S-N~N-StandardZyvox ~N ~ ~ N~H o . F N

This example shows the synthesis of various examples of the substituted oxazolidinones.
A. Sulphonates and esters 1. Synthesis of Precursors N-Ethyl 5-hydroxymethyl-2-oxazolidinone O
O
O' -NH ~ ~ ~ ~u~, THF, RT O~N~
HO J--~
Ph3COy 2. TFA-H20, RT
to To a solution containing the oxazolidinone (5.0 g, 13.9 mmol) in dry THF (50 mL) was added potassium-t-butoxide (2.03 g, 18.1 mmol, 1.3 equiv) at RT and stirred under NZ atm for 0.5 h. To this solution was added ethyl iodide (3.25 g, 20.8 mmol, 1.5 equiv) and stirred for 2h after which TLC (1:1 hex-EtOAc) showed completion of reaction. The reaction was quenched by adding satd. NHQC1 solution.
THF was removed on rotovap and residue diluted with 2o CH2C12 (100 mL). Organic layer washed with brine and dried (MgS04). Removal of solvent gave a light yellow oil. This crude product was taken in CHZC12 (50 mL) .
Added trifluroacetic acid (4.75 g, 41.7 mmol, 3.0 equiv)-water (1.0 g, 55.5 mmol, 4 equiv) dropwise and stirred for 2h. The solvent was removed on rotovap and residue purified by flash column chromatography (silica gel, EtOAc) to get the product as a colorless oil (1.3 g, 66$ in two steps).
1H NMR(CDC13, 200 MHz) : b 4.6 (m, 1H), 4.1 (s, 1H), 3.88 (dd, 1H)3.6 (m, 5H), 1.2 (t, 3H). 13C
NMR (CDC13, 50 MHz): S 157.86, 73.45, 62.69, 45.01, 38.59, 12.22.
A similar procedure was followed for the alkylation of the oxazolidinone with B12 (R&S), B38(R&S), B39, E11(R&S), E16(R&S), E81(R&S), G3(R&S), G4-oxa-C4, W14, W15, W17, W19 and W23.
O O ~ O
HO~N~'~u%~O~ B12 HO~N~~~~ G4 /\ IIO
O
O~N O ~ / ~ B 38 HO~N~ G12 HO~
O O HO~N~ G12-oxa-C4 O~N ~ / N~ B 39 O
HO~
HO~N~ W15 O O O
HO~N~ E11 ' O
O O HO~N~~\j~ W17 O O
HO~'\/N ~ ~ E16 O

O~N
O~O HO~, ~~/\~ W23 HO~,N- G3 O
O HO.J~N~ W14 HON G3-oxa-C4 O
1o N-isopropyl-5-trityloxymethyl-2-oxazolidinone TrO~j~,NH KBu~ T~O~N
To a solution containing the oxazolidinone (15.0g, 41.7 mmol) in dry THF (150 mL) was added potassium-t-butoxide (8.4 g, 75.0 mmol, 1.8 equiv) at RT and stirred under N2 atm for 0.5h. To this solution was added isopropyl iodide (7.8g, 45.9 mmol, 1.1 equiv) and stirred at 70° C for 15 h. The reaction was quenched by adding satd. NH9C1 solution. THF was removed on rotovap and residue diluted with CH2C12 (200 mL). Organic layer washed with brine and dried (MgSOq) . Removal of solvent gave a light yellow oil.
The residue was purified by flash column 1o chromatography (silica gel, hexane-EtOAc, 4:1) to get the product as a colorless solid (15.0 g, 90~).
1H NMR (CDC13, 200 MHz) : b 7. 3 (m, 15H) , 4 . 56 (m, 1H) , 4 .1 (m, 1H) , 3.3 (m, 4H) , 1.1 (m, 6H) . 13C
NMR (CDC13, 50 MHz) : b 157.0, 143.4, 128.6, 127.9, 127.2, 86.8, 71.9, 64.0, 44.6, 41.6, 19.8, 19.6.
Synthesis of ethyl (2-bromo)-t-butyl ketone (class G4):
OTMS O
Br~O~ SnBrl, CH2C12 Br RT
To a suspension of SnBr2 (242 mg, 5 mold) in CH2C12 (5.0 mL) was added pinacolone trimethylsilyl enol ether (3.0 g, 17.4 mmol) in CH2C12 (2.0 mL) followed by bromomethyl methylether (3.26 g, 26.1 mmol, 2.1 mL, 1.5 equiv) in CH2C12 (2.0 mL) . Stirred at RT for 3.5 h after which TLC showed complete conversion. The solvent was removed on rotovap to get an orange yellow liquid. The crude product was passed 3o through a short pad of silica packed in hexane and the product was eluted with 5$ EtOAc-hexanes as a pale yellow liquid (2.65 g, 79~).

1H NMR (CDC13, 200 MHz) : b 3.54 (t, 2H) , 3.06 (t, 2H) , 1.12 (s, 9H) . 13C NMR (CDC13, 50 MHz) : ~
212.4, 43.9, 39.6, 26Ø
Homologation of t-butylacetyl chloride CI~~~ 1. Me3SiCHN2, CH3CN-THF Br'~~
O 2. HB I ~ 'r O
t-Butylacetyl chloride (4.0 g, 29.7 mmol) 1o was added dropwise to a solution of trimethylsilyldiazomethane (37.1 mL, 74.3 mmol) in CH3CN-THF (100 mL, 1:1) at 0° C added the t-butylacetyl chloride (4.0 g, 29.7 mmol) dropwise and then refrigerated for 40 h. Solvent was removed on rotovap and residue diluted with CH2C12(100 mL). Washed with satd. NaHC03(50 mL) solution followed by brine, dried (MgS09) and concentrated to an yellow liquid (4.5 g).
This product was taken in THF (25 mL) and cooled to 0°
C and added HBr (48~) (8.4g, 104.0 mmol, 3.5 equiv) 2o dropwise. After the addition, reaction mixture stirred at that temp for 30 min. Diluted with CH2C12 (75 mL) and washed with satd. NaHC03(50 mL) followed by brine, dried (MgSOq ) and concentrated to an yellow liquid. Purified by column chromatography (silica gel) and the product was eluted with 2-4g EtOAc-hexane.
Pale yellow liquid (3.3 g, 82.50 .
1H NMR (CDC13, 200 MHz) : b 3.84 (s, 2H) , 2.48 (s, 2H) , 0.98 (s, 9H) . 13C NMR (CDC13, 50 MHz) : b 200.8, 51.7, 36.2, 31.1, 29.4.
3o Following a similar procedure W14, W15, W19, W23 were prepared from corresponding acid chlorides.

O O

O O
O O

O O
Synthesis of C4-Oxazolidinone 4,4-dibenzylamino-1,3-(S)butanediol OH O OH
Bn2N~ LiBr,~ Bn2N
OEt ~F~ 55oC OH
Mol. Wt.: 327.42 Mol. Vllt.: 285.38 R.B charged with Liar (14.8 g, 170.8 mmol, 4.0 equiv), NaBH9(6.37 g, 170.8 mmol, 4.0 equiv) and to THF (125 mL) and stirred at 50° C for 2h. Added the ester (14.0 g, 42.7 mmol, 1.0 equiv) in THF (25 mL) slowly and stirring continued for 2h (TLC no SM).
Reaction mixture cooled to RT and added satd. NH9C1 dropwise (cool in ice-bath) till no gas evolution.
z5 Most of the THF was removed on rotovap and residue diluted with EtOAc (250 mL). Washed with brine, dried (MgS09) and concentrated to a colorless oil and purified by column chromatography (70g EA-hexane).
Product obtained as a colorless syrup (yield: 9.2 g, 20 77$) .
1H NMR (CDC13, 200 MHz) : b 7 .2 (m, 10H) , 3. 8 (m, 6H) , 3. 4 (d, 2H) , 2 . 4 (m, 2H) , 1 . 6 (m, 2H) . 13C NMR
(CDC13, 50 MHz): b 138.3, 128.9, 128.3, 127.2, 66.8, 60.7, 59.5, 58.4, 36.3.
4-Amino-1,3-(S)butanediol _ 77 _ OH Pd(OH)2 OH
Bn2N ~ A
OH HZN ~OH
Mol. Wt.: 285.38 Mol. Wt.: 105.14 THF-MeOH (30+40 mL) suspension containing the diol ( 9. 0 g, 31. 5 mmol) and wet Pd (OH) 2 ( 10$, 3. 5g) was hydrogenated under 40 psi (2.81 kgf/cm2) for 20h.
The solution warmed and the hot solution filtered through a short pad of celite. Washed several times with methanol (towards the end few drops of TEA
added) . Removal of solvent gave a colorless oil (3.3 g, quantitative).
4-(Benzyloxycarbonyl)-amino-1,3-(S)butanediol OH H OH
H2N~ CbzCl, Na2C03 ~ ~ O N
OH OH

Mol. Wt.: 105.14 Mol. Wt.: 239.27 R. B. charged with the amine (3.3 g, 31.3 mmol, 1.0 equiv) and THF-H20 (20+40 mL) . Added Na2C03 (4.0 g, 37.8 mmol, 1.2 equiv) and cooled to 5° C.
Added CbzCl (6.458, 37.8 mmol, 5.4 mL, 1.2 equiv) dropwise keeping temp. below 5° C and stirred at that 2o temp. for 3h. Diluted with water (100 mL) and extracted into EtOAc (3x100 mL). Washed with brine, dried (MgS04) and concentrated to a colorless oil which solidified on keeping (9.0 g).
1H NMR(CDC13, 200 MHz) : b 7.2 (s, 5H), 5.6(s, 1H) , 5. 0 (s, 2H) , 3. 5 (m, 7H) , 1. 6 (m, 2H) . 13C NMR
(CDC13, 50 MHz): S 157.1, 136.2, 128.4, 128.0, 127.9, 70.2, 66.8, 60.4, 46.9, 35.7.
_ 78 -4-(Benzyloxycarbonyl)-amino-1(O-trityl)-3 (S)butanediol H OH
O~N~OH TrChTEA / ~ O NJ H
O CH2CI2 ~~ ~ ~OCPh3 O
Mol. Wt: 239.27 Mol. Wt: 481.58 Reaction mix. containing the crude diol (~9.0 g, 31.5 mmol, 1.0 equiv), TrCl (10.5 g, 37.6 mmol, 1.2 equiv) and TEA 2.5 (7.96 g, 78.6 mmol, equiv) in CH2C12 (100 mL) stirred at RT for 21 h. The to reaction with water, brine and ried mixture washed d (MgS04) and concentrated g).
to a pale yellow oil (20.0 5-trityloxyethyl-2-oxazolidinone H OH O
/ ~ O\ /N J~OCPh KtBuO, THF, RT HN~O
J( 3 1 , O ~oCPh3 Mol. Wk: 481.58 Mol. Wt.: 373.44 The above crude product (20 g, 31.5 mmol based on purity) taken in anhydrous THF (150 mL) and treated with KtBuO (7.84 g, 70 mmol, 2.2 equiv) and 2o stirred at RT for 7 h. Diluted with water (100 mL) , bulk of the THF removed on rotovap. Residue extracted with EtOAc (3x100 mL), washed with brine, dried (MgS04) and concentrated to a light brown oil.
Purified by column chromatography (40$ EtOAc-hexane) to get a pale yellow foamy oil which solidifies (10.7 g, 91$ in three steps).
1H NMR (CDC13, 200 MHz) : b 7 .25 (m, 15H) , 5.75(s, 1H), 4.8(m, 1H), 3.6(t, 1H), 3.2(t, 3H), _ 79 -2.0 (m, 2H) . 13C NMR (CDC13, 50 MHz) : b 159. 6, 143.8, 128.5, 127.8, 127.0, 86.9, 75.0, 59.3, 46.0, 35.2.
N-(4-nitrophenyl)-5-trityloxymethyl-2-oxazolidinone O
O
Pd(OAc)2, Cat O~N / ~ NOZ
O NH +Br ~ / NOZ ~ Ph3C0~
Ph3C0~ NatOBu, Tol.
The reaction mixture containing the oxazolidinone (5.0 g, 13.9 mmol), 4-bromonitrobenzene (4.2 g, 20.8 mmol, 1.5 equiv), 1,1' bis(diphenylphosphinoferrocene) (0.77 g, 1.39 mmol, 0.1 equiv) , Pd (OAc) 2 (0. 31g, 1.39 mmol, 0.1 equiv) and sodium-t-butoxide (2.0 g, 20.8 mmol, 1.5 equiv) in dry toluene (130 mL) was stirred under N2 atm at 110° C for 8 h. The solvent was removed on rotovap and the dark residue was chromatographed on silica gel using 300 EtOAc-hexane to get the product as a dark yellow foamy solid (2.7 g, 41$) .
1H NMR (CDC13, 200 MHz) : b 8.2 (d, 2H) , 7 . 6 (d, 2H), 7.2(m, 15H), 4.8(m, 1H), 4.2(t, 1H), 3.9(m, 1H), 3. 8 (dd, 1H) , 3.4 (dd, 1H) . 13C NMR (CDC13, 50 MHz) : b 155, 145, 144, 129.5, 129.3, 129, 126, 118, 87, 72, 64, 47.
2. Synthesis of library-sulphonates and esters O
O
O R~-C_O~N-R
O~( RCOCUR~S02CI
N-R
HO~, CH2C12,TEA, RT O OR O
R2-S-O.J~N_R
a O

0.1 mM solution of the oxazolidinone in CH2C12 (25 mL) was prepared. From the above std. soln.
syringed out 1.0 mL each (0.10 mmol) into 3 mL capped vials. Added triethylamine, 28 uL/vial (0.2 mmol, 2.0 equiv). Added 1.0 mL (0.10 mmol) of the stock solution (0.10 mM) of acid/sulphonyl chlorides into respective vials. The vial capped, the solution mixed well and kept aside at RT (20 h). All compounds purified by prep. TLC. (EtOAc-hexane). Silica gel band containing the product was taken in CH3CN (15.0 mL) . Filtered and washed with more CH3CN (3 mL) , and solvent removed on rotovap. The product obtained was transferred to small vials using CH2C12, all samples air dried and finally dried in vacuo. All samples were analyzed by LCMS:
Oxazolidinones and sulphonyl/acid chlorides used:
B12: K2, K4, K5, K8, K9, K10, K11, E112 B12(R): K2, K10, K21, K22, K23, K83, E112 B38: K00, K0, K1, K2, K3, K4, K5, K6, K7, K8, K9, K10, K11, E0, E1, E4, E7, E8, E9, E10, E11, E15, E16, E112 B38(R): K10, E112, E117, E120, E124, E136, E154, B39: K00, K0, K1, K2, K3, K4, K5, K6, K7, K8, K9, K10, K11, E0, E1, E4, E7, E8, E9, E10, E11, E15, E16, E112 E11: K2, K4, K5, K8, K9, K10, K11, E112 E11 (R) : K2, K10, K21, K22, K23, K83, E112 E16: K00, K0, K1, K2, K3, K4, K5, K6, K7, K8, K9, K10, K11, E14, E0, E1, E4, E7, E8, E9, E10, E11, E15, E16, E16 (R) : K2, K10, K21, K22, K23, K83, E8, E112 E81: K4, K5, K8, K9, K10, K11, E112 E81 (R) : K2, K10, K21, K22, K23, K83, E112 l0 G4: K2, K4, K5, K8, K10, K11, K21, K22, K23, K27, K30, K52, K54, K55, K56, K59, K60, K66, K83, K90, K91, K92, K93, K94, K95, K96, K97, E4, E11, E81, E82, E90, E107, E112, E113, E117, E159, E164, E168 G4 (R) : K10, K21, K22, K23, K83, G5: K00, K0, K1, K2, K3, K4, K5, K8, K9, K10, K11, K12, K21, K22, K23, K27, K30, K52, K54, K55, K56, K59, K60, K66, K83, K90, K91, K92, K93, K94, K95, K96, K97, K98, K99, K100, K101, K102, K117, E8, E11, E81, E82, E90, E107, E112, E113, E117, E120, E124, E136, E154, E159, E164, E168, E183, E184 G5 (R) : K10, K11, K12, K21, K22, K23, K83 G9 (R) : K2, K90, K91, K92, K93, K94, K95, K96, K97, K98, K99, K100, K101, K102, K117, G12: K00, K0, K1, K2, K3, K4, K5, K8, K9, K10, K11, 3o K12, K21, K22, K23, K27, K30, K52, K54, K55, K56, K59, K60, K66, K83, K90, K91, K92, K93, K94, K95, K96, K97, K98, K99, K100, K101, K102, K104, K105, K106, K107, K109, K110, K112, K113, K114, K115, K117, G12 (R) : K2, K101, K102, K117, E112, E183, E184 G13 (R) : K00, K0, K1, K2, K3, K4, K5, K8, K9, K10, K11, K12, K21, K22, K23, K27, K30, K52, K54, K55, K56, K59, K60, K66, K83, K90, K91, K92, K93, K94, K95, K96, K97, K98, K99, K100, K101, K102, K104, K105, K106, K107, K109, K110, K112, K113, K114, K115, E112, E183 W14: K101, K102, K104, K105, K106, K107, K109, K110, K112, K113, Kil4, K115, E112, E183 W15: K10, K21, K22, K23, K83, E8, E11, E81, E82, E90, E107, E112, E113, E117, E120, E124, E136, E154, E159, E164, E168 W17: K2, K4, K9, K10, K11, K12, K21, , K27, K29, K30, K31, K52, K53, K54, K55, K56, K59, K60, K61, K66, K70 E8, E11, E81, E82, E90, E107, E112, E113, E159, E164, 2o E168 W19: K10, K11, K12, K21, K22, K23, K83 E8, E11, E81, E82, E90, E107, E112, E113, E117, E120, E124, E136, E154, E157, E159, E164, E168 W23: K2, K4, K9, K10, K11, K21, K22, K23, K27, K29, K30, K31, K52, K53, K54, K55, K56, K59, K60, K61, K66, K70, K83, E8, E11, E81, E82, E90, E107, E112, E113, E117, E120, E124, E136, E154, E157, E159, E164, E168 G12-oxa-C4: K2, K10, K11, K93, K95, K96, K97, K100, K101, K102, K104, K105, K106, K107, K109, K110, K112, K115, K117, E183, E184 G3-oxa-C4: K2, K10, K11, K93, K95, K96, K97, K100, K101, K102, K104, K105, K106, K107, K109, K110, K112, K115, K117, E183, E184 Amines A. Amides and Sulphonamides 1. Synthesis of precursors (5R)-methanesulphonyloxymethyl-3-[(1R)-phenylethyl oxazolidine-2-one) O O
O- _N ~ MsCI, TEA p~N
HO
CH2C12 Ms0 To an ice-cooled solution of the oxazolidinone (5.0 g, 22.6 mmol, 1.0 equiv) in CH2C12 ' (50 mL) was added TEA (4.57 g, 45.1 mmol, 2.0 equiv) followed by MsCl (3.36 g, 29.3 mmol, 1.3 equiv) 2o dropwise and then stirred for 2 h. Diluted with CH2C12 (50 mL), washed with water (25 mL), brine, dried (MgS09) and concentrated to get an oil which solidified on keeping ( 6 . 5 g, 97 $ ) .
1H NMR (CDC13, 200 MHz) : b 7. 4 (s, 5H) , 5.25 (m, 1H), 4.7 (m, 1H), 4.4 (m, 2H), 3.4 (m, 2H), 3.15 (s, 3H) , 1. 65 (d, 3H) . 13C NMR (CDC13, 50 MHz) : b 156.1, 138.9, 128.5, 127.8, 126.7, 69.8, 68.8, 51.5, 41.3, 37.4, 16Ø
(5R)-Azidomethyl-3-[(1R)-phenylethyl-oxazolidine-2-one) O O
O_ _N ~ NaN3, DMSO O~N
MsO~ I i 80oC N3 Reaction mixture containing the mesylate (6.5 g, 21.7 mmol) and NaN3 (2.12 g, 32.6 mmol, 1.5 equiv) in DMSO (60 mL) was stirred at 80° C for 3 h under N2 atm. Then cooled to RT, diluted with water (100 mL) and CH2C12 (150 mL). Organic layer washed with brine, dried (MgS04) and concentrated to a pale 1o yellow liquid. Crude product filtered through a short pad of silica using 40~ EtOAc-hexane. Colorless oil which crystallized on keeping (5.0 g, 94$).
1H NMR (CDC13, 200 MHz) : b 7 .4 (s, 5H) , 5.3 (m, 1H), 4.6 (m, 1H), 3.4 (m, 4H), 1.66 (d, 3H). 13C
NMR (CDC13, 50 MHz) : b 156.4, 139.0, 128.6, 127.8, 126.8, 71.1, 53.1, 51.4, 42.3, 16.0 (5R)-Aminomethyl-3-[(1R)-phenylethyl-oxazolidine-2 one ) O O
Pd-C, EtOH O- _N
O~N
N ~ ~ ' H2N

H2, RT
R.B charged with Pd-C (10g, 500 mg) and ethanol (10 mL). Added oxazolidinone (2.0 g) in ethanol (10 mL). Flushed with HZ three times and stirred under H2 overnight (17h). Filtered through a short celite pad and washed with methanol. Solvent removed on rotovap to a light orange oil. Purified by silica gel column (20-50~ MeOH in EtOAc) to get a light orange oil which solidifies on keeping (1.1 g, 62~) .
1H NMR (CDC13, 200 MHz) : b 7. 4 (s, 5H) , 5.3 (m, 1H), 4.6 (m, 1H), 3.4 (m, 2H), 3.1 (t, 2H), 2.8 (s, 2H) , 1. 66 (d, 3H) . 13C NMR (CDC13, 50 MHz) : b 157.1, 139.4, 128.6, 127.8, 126.9, 73.7, 58.0, 51.4, 42.6, 16.2.
Amines belonging to classes G3, G5, G9, G12, B38 (both isomers), G3-oxa-C4, G12-oxa-C4 were 1o prepared in a similar manner.
O O
H2N. ~ N ~ / NOZ G9 H2N ~,N- G3 O O O
H2N~N~ G5 H N O~N ~ / NEt2 B38 2 ~/
O
H2N ~N~ G12 O
O~N- G3-oxa-C4 H2N~
O
O~N-~ G12-oxa-C4 H2N~
2. Library-synthesis O O O O
/ NEt2 RS02CUR1COC1 R_X NON ~ / NEt2 H2N.~,N TEA, CH2CI2 X=S,C
Y=O
Z=O,0 O.lmM Solution of the oxazolidinone in CH2C12 (25 mL) was prepared. From the above std. soln.

syringed out 1.0 mL each (0.10 mmol) into 3 mL capped vials. Added triethylamine, 28 uL/vial (0.2 mmol, 2.0 equiv). Added 1.0 mL (0.10 mmol) of the stock solution (0.10 mM) of acid/sulphonyl chlorides into respective vials. The vial capped, the solution mixed well and kept aside at RT (20 h). All compounds purified by prep. TLC. (EtOAc-hexane). Silica gel band containing the product was taken in CH3CN (15.0 mL). Filtered and washed with more CH3CN (3 mL), and solvent removed on 1o rotovap. The product obtained was transferred to small vials using CH2C12, all samples air dried and finally dried in vacuo. All samples were analyzed by LCMS.
Other Amines used for the library:
G3 (R) , G5 (R) , GS (S) , G12 (R) , G9 (R) , G9 (S) , G13 (S) .
O O
O~
HZN~N- G3 H2N~,N ~ ~ N02 G9 O O
H2N~N~ GS 0I _N ~ G13 O

H2N.~rN ~
O
O~N- G3-oxa-C4 H2N ~
O
O~N~ G12-oxa-C4 HZN~
Sulphonyl/acid chlorides used:
838: K1, K9, K10, K11, K21, K22, K23, K83, E112 G12-oxa-C4: K2, K93, K95, K96, K97, K99, K100, K101, K102, K117, E183, E184 _ 87 _ G12: K2, K93, K95, K96, K97, K99, K100, K101, K102, K117, E183, E184 G9 (R) : E183, E184 G9 (S) : E183, E184 G5: K00, K0, Kl, K2, K3, K4, K5, K8, K10, K11, K12, K21, K22, K23, K83, K90, K91, K92, K93, K94, K95, K96, K97, K98, K99, K100, K101, K102, K117,E183, E112, E184 l0 G5 (R) : K2, K93, K95, K96, K97, K99, K100, K101, K102, K117, E183, E184 G3 (R) : K2, K93, K95, K96, K97, K100, K101, K102, K117, E183, E184 G13 (R) : K00, K0, K1, K2, K5, K9, K10, K11, K12, K21, K22, K23, K52, K60, K83, K90, K91, K92, K93, K94, K95, K96, K97, K98, K99, K100, K101, K102, E183, B. Urea type compounds O O
H
HZN~N~ R~ RHN~N.~rN~

Oxazolidinone (0.1 mmol, l4mg) in CH2C12 (1.0 mL) treated with the respective isocyante (0.1 mmol).
In cases where the solution was not homogeneous 0.5 mL
THF was also added. Reaction mixture kept at RT for 16 h and purified by prep. TLC (hexane-EtOAc) . All products were analyzed by LCMS.
_ 88 _ Other amines used for the library: G12(R), G12(S), G5(S), G9(R), G9(S), O O
O JL OJ( H2N.~N~ HzN.J~rN~ H2N.~N-O O
OJ( OJ(N~
H2N..J~N ~ ~ NOz H2N~1~, G9 G 12-oxa-C4 Isocyanates used for the library:
G12 (R) : DD2, DD3, DD4, DDS, DD6, DD7 G12: DD2, DD3, DD4, DDS, DD6, DD7 G5: DD2, DD3, DD4, DDS, DD6, DD7 G5 (R) : DD2, DD3, DD4, DDS, DD6, DD7 G12-oxa-C4: DD2, DD3, DD4, DDS, DD6, DD7 G9 (R) : DD2, DD3, DD4, DDS, DD6, DD7 G9 (S) : DD2, DD3, DD4, DDS, DD6, DD7 G3: DD2, DD3, DD4, DDS, DD6, DD7 C. Sulphenyl compounds O.lmM solution of the oxazolidinone in CH2C12 (5 mL) was prepared. From the above std. soln.
2o syringed out 1.0 mL each (0.10 mmol) into 3 mL capped vials. Added triethylamine, 28 uL/vial (0.2 mmol, 2.0 equiv). Added 1.0 mL (0.10 mmol) of the stock solution (0.10 mM) of sulphenyl chlorides into respective vials. The vial capped, the solution mixed well and kept aside at RT (20h). All compounds purified by prep. TLC. (EtOAc-hexane). Silica gel band containing the product was taken in CH3CN(15.0 mL) . Filtered and washed with more CH3CN (3 mL) , and solvent removed on rotovap. The product obtained was transferred to small vials using CH2C12, all samples air dried and finally dried in vacuo. All samples were analyzed by LCMS.
Amines used for the library: G5, G12, G9, G9(R), G12-oxa-C4 O O
HZN~N~ G5 H2N~N ~ ~ NOz G9 O O
O/( ~ ~~0-KN~ G12-oxa-C4 H2N~,N G12 H2N~r l0 Oxazolidinones and sulphenyl chlorides used:
G5: BB3, BBS, BB7, BB9 G5(R): BB3, BBS, BB7, BB9 G12: BB3, BBS, BB7, BB9 G12 (.R) : BB3, BBS, BB7, BB9 G9: BB3, BBS, BB7, BB9 G9(R): BB3, BBS, BB7, BB9 G12-oxa-C4: BB3, BBS, BB7, BB9 D. Substituted aryl amines-Buchwald coupling O O
Arl, Cul, K3P04 A~N~N
HZN.~,N~ isopropanol ethylene glycol 80oC
The reaction mixture containing the aminooxazolidinone (50 mg, 0.35 mmol), CuI (3.3 mg, 5 mold) , K3P09 (148. 6 mg, 0.70 mmol, 2 equiv) , ethyleneglycol (43.4 mg, 0.70 mmol) and the aryl iodide (0.52 mmol, 1.5 equiv) in isopropanol (1.5 mL) was stirred at 70° C for 24 h. Reaction mixture was cooled and filtered. Filtrate purified by prep. TLC.
Products were analyzed by LCMS.
Other amines used for library: G5(R), G9(R), G9 (S ) , G12 (R) O O O
H2N.~LN~ H2N.J~rN~ H2N.J~N-O
H2N.~N ~ ~ NO2 to Oxazolidinones and aryl iodides used:
G9 (S) : AA6, AA7, AA9, AAll, AA12 AA16, AA17, AA18, AA26, AA35 G9 (R) : AA6, AA.7, AA9, AA16, AA17, AA18, AA2 6, AA35 G12 (R) : AA6, AA7, AA9, AA16, AA17, AA18, AA26, AA27, 2o G12 (S) : AA6, AA7, AA9, AA16, AA17, AA18, AA26, AA35 G5(R): AA6, AA7, AA9, AA16, AA17, AA18, AA26, AA35 Ether Derivatives 5-Tosyloxymethyl-N-isopropyl-2-oxazolidinone O
p~N~ TsCh TsO~.JI,N
HO~

CH2C12 (25 mL) solution containing the oxazolidinone (2.3 g, 14.4 mmol, 1.0 equiv) was cooled in ice-bath. Added TEA (3.65 g, 36.1 mmol, 2.5 equiv) followed by TsCl (4.1 g, 21.5 mmol, 1.5 equiv) in small portions. Stirring continued at 0° C-RT (7 h).
Diluted with more CH2C12 (50 mL), washed with 1N HC1 (50 mL), water, brine, dried (MgS09) and concentrated l0 to a brown liquid. Crude product passed through a silica gel column (60$ EtOAc-hexane) to get the product as a colorless solid (4.3g, 96~).
1H NMR (CDC13, 200 MHz ) : b 7 . 5 (ABq, 4H) , 4 . 6 (m, 1H), 4.0 (m, 3H), 3.5 (t, 1H), 3.3 (dd, 1H), 2.39 (s, 3H) , 1.2 (m, 6H) . 13C NMR (CDC13, 50 MHz) : b 155.8, 145.2, 131.9, 129.9, 127.7, 69.5, 68.7, 44.7, 40.9, 21.4, 19.4, 19.3.
Library Synthesis 2o Method 1 O
O _ O~N
O~ ~ 1. KBuOt, THF
TsO~,N --~ \ /
3.0 mL vial charged with 1-naphthol (12.7 mg, 0.09 mmol, 1.1 equiv) and THF (1.0 mL). Added KtBuO (13.4 mg, 0.12 mmol, 1.5 equiv). Stirred for 30 min. at RT. Added the tosylate (25 mg, 0.08 mmol, 1.0 equiv) in THF (1 mL). Stirred at RT for 2.0 h.
Purified by prep. TLC (EtOAc-hexane) to get the pure product. Analyzed by LCMS.
Other libraries synthesized O O
O~N \ / N
RO

O O
O~N \ / NEt2 RO

to Method 2 O O
ROH, DIAD/DEAD, PPh3-styrene O~
HO~i~,N'-\ RO~,N
THF
To a solution containing the oxazolidinone i5 (14.4 mg, 0.1 mmol), PPh3-polystyrene (120 mg, l.2 equiv, loading 1.0 mmol/g), phenol (0.1 mmol, 1.0 equiv) and CH2C12 (l.OmL). Added DIAD/DEAD (1.2 equiv) in THF (1.0 mL) slowly. Stirred gently for 24 h. The crude product was purified by prep. TLC (hexane 2o EtOAc) .
Other libraries synthesized: G9, G5 O O
O~N~ HON \ / N02 HO~, 2s Alcohols/phenols used:

B38: M1, M2, M3, M4, M5, M6, M14, M37, M38, B39: M3, M5, M35, M38 G5: M39, M42, M43, M44, M45, M46, M47 G12 (S) : M1, M2, M3, M4, M5, M6, M11, M13, M14, M24, M30, M34, M35, M37, M38, M39, M40, M41, M42, M43, M44, M45, M46, M47 G12 (R) : M40, M41 G9(R): M1, M2, M3, M4, M5, M6, M11, M13, M14, M24, M30, M34, M35, M37, M38, M39, M40, M41, Sulphonylchlorides (K):

0 _ 0 _ O S_CI _ O
\ / S-cl I ~ o c1 \ / s-cl °2N \ / o-cl o ~ o H
_ O _ O O O ,I
- N ~S-CI O~ N
O \ / O-CI O=C=N \ / O CI ( S S-CI ~H~S ~ HN
~S~CI
O

Br .O
° O N~ ~N O F F / I
/ \ ~~ N- S-G ~~ Nr\~ ,O
CI~S-CI ~ / " G S-CI F~ oS~
N O O \ / ~~ O O Cl O

O
/ ' ° c1 ° / \ ~ ' ~ I ~ ° s-cl 'N o ~ ~ s-cl / \ s_cl ~o-cl ° / o c1 p / o / \ S-CI N O N 0 N ~N O ~ S~N CF3 O
0 'O

O ~0 rN O,N S O
~ ii O , O I' N S O O \ S S-G F C ' I % S-CI
O S_CI O / O-CI S ~ I / S-CI ~ I / a 3 O
/ O O O
/ \
CI

S ,N CI ~~
p ~N \ S O N N S O ~ S O
INI ~ ~ ~ S-CI F3C ~ I / S-CI S'/ I / S-CI S_N I / OS-CI

K55 K56=K113 K58 K59 O O
0 ~ \ S O ti3CO2C N
/ \ I S s-G ~N ~ ~ s-G ~ / s-G N _~ 's-cl -N ~ o S- 0 0 0 N~ S-CI \N \ O-G ' , NOZ OZN I ~ S_G U N / ,SO,-G
Ph'N~O N~ O HN ~ S CI HN
O

s-cl F / \ s-G Br / \ s-G / \ s-G /_\ s-G
N / ., .. .. 0 0 S~~O O O F F
N F

O O O
G / \ s-G o N- ~/ \ -s-G G / \ s-cl , F3c / \ s-cl / \ s-cl ~0 2 ~0 0 0 0 F / \ F S-CI / \ S-CI / \ S-G NC / \ S-CI Br / \ S-CI
O ~O \ O O O
F ~F NOZ NOZ
Kgg K100 K101 K102 K103 F
c1 CI
G O O
° ° / \ ~ F /-\ s-G / \ s-G
F / \ S-CI F3C / \ S-CI S-CI " ..

O O
O O CI F3c~0 FaC ~ / \ v ,N F F
/ \ O N S O / \
s-G / \ S-G ~\ ~ ~ s-G F~-s-G
p O F3C p O

Klpg K110 K 111=K56 K 112 CI
O O O
\ N-- ~~ ~- S-CI V ~ ~ S-CI ~ ~ ~ S-CI AcNH ~ ~ S-CI
'O' N O O O O

_ O
S-CI
OzN O

Acid chlorides (E):

O ~ CI Me0 CF3 F3C OMe I ~ G ~CI I / ~ ~ CI ~ C
/ I O I / ' O
O /

O
O 'I
O O O
S~I F ~ CI ~ CI O O ~ CI
I / / ~ CI I / O
F I / F F I F_ I /
F F

/ CI O O CI O
~~CI O
CI ~( ~ CI
CI ~ HCI I , HCI
N~ ~ O I , N
O N

CI O
O ~ ~ CI
CI ~ CI N~ 1 CI /O ' G ~O
I ~ O
F N F
E14. E15 E16 E81 E82 O CI
S 0 CI ~ O / ~ O Ph /, p ~ I ~/ C
CI I / O~G p I / CI / / CI S
CI CI IO F O

CI
O N~ ~ - 0 /
\ N / I CI ~ I CI ~ '~ CI \ ~ CF3 CI ~ N ~ I CI
O.~ N \
Ph O O N~O O

OZN
O O N CI - C
I \rN p Ph ~ ,N ~ CI ~ ~ ~ 02N \ / \ / 0 F3C N ~~I / \
O

Phenols (M):

Br / \ OH CI / \ OH F / ~ OH NC / \ OH F3C / \ OH

O
-N
O
/ \ OH OHC / \ OH HZNOC / \ OH ~ / \ OH HZN-S / \ O
O

CI NO~ CN CONHZ O
~NH
/ N OH / N OH N \ OH / \ OH N \ OH
-N ~ / \ OH
N

OH OH OH OH
HO
w w w w ~ ~ N~ ~ ~ N
off I , , I , ~ I , J C ~J I ~ ~ \ /
N ~ F3C N N N N
OH

OH OH OH
OH ~OH
HO ~ N ~ No2 ~
I I ~ I , OzN/v ~ i i J NOZ
M39 M,~ M41 M42 M43 OH SH
OH OH
H3C0 ~ ~ OH ~ ~ No2 w w CH3 w w I w I ~ i I~

Aryl iodides(AA):
_ 99 _ \ I \ I I I \ I
\O ~ / ( NOz / OZN / OzN / NO
AA 5 AA 6 NOz AA 9 I NHz 02N \ I ~ N I I I
I ~N
N~ / I
NOz H N F3C
AA 11 Ap 12 AA 16 AA 17 I \ I
I
I ~ ~ /
F
AA 18 ~ 26 F CF3 Sulphenyl chlorides (BB), Isocyantes (DD):

S-CI ~ O"CI ~ S-C) ~ S-CI
ON ~ ~ O
OZN v 2 OZN v ~NOZ N02 NCO ~ NCO ~ NCO ~ NCO
~ / /
/ / p2N ~CFa NOZ
NOZ

NCO
NCO

Scheme 1 O
O o~ 0 0 PhaCO~NH ~ ~a~~N R ~ HO~N~R

O OI' R~O~N~R

~ OMe R = -OMe -OBn ~
v 'CI
a b c d a _ O ~--~ N--NHMe -NMey -NN~ - VN~ ~/N~N~
f 9 h i 1 Experimental 1. Preparation of the precursors of libraries.
General Methods. Column chromatography was performed on silica gel. TLC was performed on silica gel GF254. NMR spectra were recorded with an Varian VXR-200 NMR spectrometer 1H NMR and 13C NMR spectra were recorded at either 200 MHz or 50 MHz. LC-MS was carried out at PE-Sciex AP150EX single quadrapole instrument.
1o Typical Reactions for preparation of the alcohols (3):
Method A:
A solution of t-BuOK (50 mL, 50 mmol, 1 M) in THF was dropped into a solution of starting material 1 ( 18 g, 50 mmol ) in dry THF ( 120 mL) under nitrogen at rt in 5 min. The mixture was stirred for 10 min at rt. Methyl bromoacetate (7.65 g, 50 mmol) was dropped into the flask in 5 min. The mixture was stirred for 1 h at rt. 10 ~ NH9C1 (20 mL) and hexanes (40 mL) were added, respectively. The organic phase is separated. The solvents were evaporated to give a crude product, without purification for next step.
The crude product was dissolve into DCM (100 mL).
Water (1.8 mL, 100 mmol) and TFA (8.55 g, 75 mmol) were added to the flask. The mixture was stirred for 2 h at rt: Removal of volatile materials gave a residue, which was co-evaporated with CH3CN (2 X 60 mL) to remove the trace water. Column chromatography purification (1:1 ethyl acetate/hexanes, then ethyl 3o acetate) afforded a pure product (3a) (5.2 g, 55 $).
NMR: DH 3.4-4.1 (7H, m), 3.66 (3H, s), 4.5-4.7 (1H, m) ppm, ~~: 44.98, 46.10, 52.17, 62.61, 74.08, 158.20 (C=O), 168.92 (C=O, ester) ppm.

3b: yield: 36 ~, NMR: bH 2.9 (1H, br,s), 3.46-3.86 (4H, m), 4.02 (2H, s,), 4.54-4.70 (1H, m), 5.14 (2H, s), 7.2-7.3 (5H, ppm.
m) 3e: yield: 8 .1 ~ . aH 2 .
5 ( 1H, br) , 3. 51- 3 . 94 (4H, m), 3.79 (3H, s), 4.67 (2H, s), 4.5-4.7 (1H, m), 7.1-7.5 (4H, m) ppm.

Method B:

NaH (0.4 g, 10 mmol, 60 ~) was added in portions into a solution of starting material 1 (3.59 1o g, 10 mmol ) in dry THF ( 40 mL) under nitrogen at rt in min. The mixture was stirred for 1 h at rt.
Bromopinacolone (1.79 g, 10 mL) was dropped into the flask in 10 min. The mixture was stirred overnight at rt. 10 ~ NH4C1 (10 mL) and hexanes (20 mL) were added, respectively. The organic phase is separated.
The solvents were evaporated to give a crude product, without purification for next step. The crude product was dissolve into DCM (30 mL). Water (0.36 mL, 20 mmol) and TFA (1.71 g, 15 mmol) were added to the 2o flask. The mixture was stirred for 1 h at rt.
Removal of volatile materials gave a residue, which was co-evaporated with CH3CN (2 X 30 mL) to remove the trace water. Column chromatography purification (3:1 ethyl acetate/hexanes, then ethyl acetate) afforded a pure product (3c) ( 1. 68 g, 7.8 $ ) . NMR: sH 1.14 ( 9H, s), 3.36-3.9(6H, m), 4.18 (2H, s), 4.54-4.7 (1H, m) ppm, b~: 26.08, 43.10, 46.30, 48.21, 63.11, 74.05, 158.45 (C=O), 209.60 (C=0, ketone) ppm.
3d: yield: 8.1 ~. bH 2.5 (1H, br), 3.56 3.9(4H, m), 4.66 (2H, s), 4.5-4.7 (1H, m) 7.43, 7.47, 7.84, 7.88 (4H, AB) ppm.
Method C:
A solution of t-BuOK (10 mL, 10 mmol, 1 M) in THF was dropped into a solution of starting material 1 (3.59 g, 10 mmol) in dry THF (40 mL) under nitrogen at rt in 5 min. The mixture was stirred for 1 h at rt . A solution of methyl bromoacetamide ( 1 . 52 g, 10 mL) in THF was dropped into the flask in 10 min.
The mixture was stirred overnight at rt. Con. NH9C1 (5 mL) and brine (5 mL) were added, respectively. The organic phase is separated. The solvents were evaporated to give a crude product. Flash column 1o chromatography (hexanes/ethyl acetate 1 . 1) gave a pure product. To a solution of the pure material in DCM (30 mL) was added TFA (1.71 g, 15 mmol) and water (0.36 g). The mixture was stirred for 1 h at rt.
Removal of volatile materials gave a residue, which was partitioned in water (50 mL) and t-BuOMe (20 mL) .
The separated aqueous layer was washed with t-BuOMe (20 mL). Water was evaporated to give a residue, which was co-evaporated with CH3CN (2 X 30 mL) to remove the trace water. A product (3f) (0.91 g, 48 obtained, without further purification for next step.
3g: yield: 99~. bH 2.91, 2.96 (6H, 2 s), 3. 5-3. 9 (4H, m) , 3. 92-4.16 (2H, AB) , 4 .41 (1H, br) , 4.5-4.7 (1H, m) ppm.
3h: yield: 74~. bH 3.25-3.61 (13H, m), 4.04 (2H, s), 4.5-4.7 (1H, m) ppm.
3i: yield: 990.
3j: yield: 81~.
2. Preparation of libraries:
3o A. parallel synthesis:
Typical reaction procedures (Reaction scales might be various accordingly):
Library 4-esters: To a solution of an acyl chloride (E) (0.1 mmol) in dry DCM (1 mL) were add a solution of 3 (0.1 mmol, 0.1 M) in DCM and triethyl amine (20.2 mg, 0.2 mmol). The mixture was standing overnight at rt. The reaction was completed. The product was purified with preparative TLC.
Library 4-sulfonates: To a solution of a sulfonyl chloride (K) (0.1 mmol) in dry DCM (1 mL) were add a solution of 3 (0.1 mmol, 0.1 M) in DCM and triethyl amine (20.2 mg, 0.2 mmol). The mixture was 1o standing overnight at rt. The reaction was completed.
The product was purified with preparative TLC.
Library 4-ethers: To a mixture of 3 (0.1 mmol), a phenol (M) (0.1 mmol) and Ph3P-polystyrene (0.1 g, 0.1 mmol Ph3P) in dry DCM (1 mL) was add a solution of DEAD (1 mL, 0.1 M). The mixture was standing at rt for three days. The reaction was completed. The product was purified with preparative TLC.
2o Libraries:
(1) 4a-esters:
Entry MW mMol EO 140.57 0.1 E1 182.65 0.1 E2 252.62 0.1 E3 252.62 0.1 E4 186.66 0.1 E5 212.53 0.1 E6 194.54 0.1 E7 198.60 0.1 E8 212.63 0.1 E9 256.08 0.1 E12 178.02 0.1 E13 178.02 0.1 E14 211.98 0.1 E15 131.52 0.1 E16 130.53 0.1 E17 130.53 0.1 E37 274.08 0.1 E40 154.60 0.1 E49 190.63 0.1 E55 175.03 0.1 1o E90 181.04 0.1 E92 215.48 0.1 E99 216.73 0.1 E113 229.06 0.1 E117 223.68 0.1 E120 207.61 0.1 E124 176.00 0.1 E136 200.67 0.1 E154 309.07 0.1 E157 226.66 0.1 2o E159 293.67 0.1 E164 319.76 0.1 E168 192.60 0.1 (2) 4a-sulfonates:
Entry MW mMol KO 190.65 0.1 K00 190.65 0.1 K1 211.07 0.1 K2 221.62 0.1 3o K3 206.65 0.1 K5 182.65 0.1 K6 254.71 0.1 K8 290.65 0.1 K9 195.62 0.1 K10 253.06 0.1 K11 327.71 0.1 K12 227.67 0.1 K16 438.33 0.1 K17 180.62 0.1 K21 218.62 0.1 K23 234.69 0.1 K29 238.65 0.1 1o K30 252.67 0.1 K31 363.21 0.1 K52 279.79 0.1 K53 249.70 0.1 K54 317.69 0.1 K55 249.70 0.1 K56 330.74 0.1 K59 301.12 0.1 K60 259.74 0.1 K61 306.82 0.1 2o K66 237.66 0.1 K70 256.71 0.1 K76 262.72 0.1 K83 234,69 0.1 K90 194.61 0.1 K91 255.52 0.1 K92 232.73 0.1 K93 244.62 0.1 K94 245.51 0.1 K96 256.06 0.1 3o K97 289.62 0.1 K98 302.86 0.1 K100 235.65 0.1 K101 217.63 0.1 K102 201.63 0.1 K104 229.06 0.1 K105 279.07 0.1 K106 245.51 0.1 K107 212.60 0.1 K109 312.62 0.1 K110 240.71 0.1 K111 330.74 0.1 K112 230.59 0.1 1o K113 242.69 0.1 K114 262.72 0.1 K115 243.67 0.1 (3) 4a-ethers:

Materials MW mMol M1 173 0.1 M2 128.55 0.1 M3 112.10 0.1 M5 162.11 0.1 2o M6 137.18 0.1 M11 95.1 0.1 M14 148.16 0.1 M24 161.16 0.1 M25 349.23 0.1 M30 160.17 0.1 M34 213.15 0.1 M35 146.14 0.1 M37 211.21 0.1 M38 189.25 0.1 (4) 4b-sulfonates:
Entry MW mMol KO 190.65 0.1 K00 190.65 0.1 K1 211.07 0.1 K2 221.62 0.1 K3 206.65 0.1 K5 182.65 0.1 K6 254.71 0.1 K8 290.65 0.1 K10 253.06 0.1 K11 327.71 0.1 1o K12 227.67 0.1 K21 218.62 0.1 K23 234.69 0.1 K56 330.74 0.1 K70 256.71 0.1 K76 262.72 0.1 K83 234,69 0.1 K90 194.61 0.1 K91 255.52 0.1 K92 232.73 0.1 2o K93 244.62 0.1 K94 245.51 0.1 K96 256.06 0.1 K97 289.62 0.1 K98 302.86 0.1 K100 235.65 0.1 K101 221.62 0.1 K102 201.63 0.1 K104 229.06 0.1 K105 279.07 0.1 3o K106 245.51 0.1 K107 212.60 0.1 K109 312.62 0.1 K110 240.71 0.1 K111 330.74 0.1 K112 230.59 0.1 K113 242.69 0.1 K114 262.72 0.1 K115 243.67 0.1 (5) 4c-esters:

Entry MW mMol EO 140.57 0.25 1o E00 120.58 0.25 E1 182.65 0.25 E2 252.62 0.20 E4 186.66 0.25 E5 212.53 0.25 E6 194.54 0.25 E7 198.60 0.25 E8 212.63 0.25 E10 108.52 0.25 E11 104.53 0.25 2o E12 178.02 0.25 E13 178.02 0.25 E14 211.98 0.25 E15 131.52 0.25 E16 130.53 0.25 (6) 4c-sulfonates:

Entry MW mMol KO 190.65 0.15 K00 190.65 0.15 K1 211.07 0.15 K2 221.62 0.15 K3 206.65 0.15 K4 217.63 0.15 K5 182.65 0.15 K6 254.72 0.15 K8 290.65 0.15 K9 195.62 0.15 K10 253.06 0.15 K11 327.71 0.15 K12 227.67 0.15 (7) 4d-esters:

1o Entry MW mMol E7 198.60 0.09 E10 108.52 0.09 Ell 104.53 0.09 E15 131.52 0.09 (8) 4d-sulfonates:

Entr y MW mMol K5 182.65 0.09 K8 290.65 0.09 2o K10 253.06 0.09 K11 327.71 0.09 (9) 4e-esters:

Entr y MW mMol EO 140.57 0.075 E1 182.65 0.075 E4 186.66 0.075 E8 212.63 0.075 E7 198.60 0.075 3o E10 108.52 0.075 E11 104.53 0.075 E15 131.52 0.075 E16 130.53 0.075 (10) 4e-sulfonates:

Entry mMol MW

KO 190.65 0.075 K00 190.65 0.075 K1 211.07 0.075 K2 221.62 0.075 K3 206.65 0.075 K4 217.63 0.075 1o K5 182.65 0.075 K6 254.72 0.075 K8 290.65 0.075 K9 195.62 0.075 K10 253.06 0.075 K11 327.71 0.075 K12 227.67 0.075 (11) 4f-esters:

Entr y MW Wt/V mMol 2o E183 185.56 0.1 E184 230.56 0.1 (12) 4f-sulfonates:

Entr y MW Wt/V mMol K2 221.62 0.1 K96 256.06 0.1 K101 221.62 0.1 K106 245.51 0.1 K117 221.62 0.1 (13) 4g-sulfonates:
Entry MW Wt/V mMol KO 190.65 0.1 K00 190.65 0.1 K1 211.07 0.1 K2 221.62 0.1 K4 217.63 0.1 K5 182.65 0.1 K6 254.71 0.1 K8 290.65 0.1 K10 253.06 0.1 K11 327.71 0.1 1o K12 227.67 0.1 K19 229.09 0.1 K21 218.62 0.1 K22 253.07 0.1 K23 234.69 0.1 K30 252.67 0.1 K52 279.79 0.1 K54 317.69 0.1 K55 249.70 0.1 K56 330.74 0.1 2o K59 301.12 0.1 K60 259.74 . 0.1 K66 237.66 0.1 K69 181.60 0.1 K70 256.71 0.1 K76 262.72 0.1 K83 234,69 0.1 K90 194.61 0.1 K91 255.52 0.1 K92 232.73 0.1 3o K93 244.62 0.1 K94 245.51 0.1 K95 266.62 0.1 K96 256.06 0.1 K97 289.62 0.1 K98 302.86 0.1 K99 266.57 0.1 K100 235.65 0.1 K101 217.63 0.1 K104 229.06 0.1 K105 279.07 0.1 K106 245.51 0.1 K107 212.60 0.1 1o K109 312.62 0.1 K110 240.71 0.1 K111 330.74 0.1 K112 230.59 0.1 K113 242.69 0.1 K115 243.67 0.1 (14) 4h-esters:

Entry MW mMol EO 140.57 0.1 E1 182.65 0.1 E2 252.62 0.1 E3 252.62 O.I

E4 186.66 0.1 E5 212.53 0.1 E6 194.54 0.1 E7 198.60 0.1 E8 212.63 0.1 E9 256.08 0.1 E12 178.02 0.1 3o E13 178.02 0.1 E14 211.98 0.1 E15 131.52 0.1 E16 130.53 0.1 E17 130.53 0.1 E37 274.08 0.1 E40 154.60 0.1 E49 190.63 0.1 E55 175.03 0.1 E90 181.04 0.1 E92 215.48 0.1 E99 216.73 0.1 E113 229.06 0.1 to E117 223.68 0.1 E120 207.61 0.1 E124 176.00 0.1 E136 200.67 0.1 E154 309.07 0.1 E157 226.66 0.1 E159 293.67 0.1 E164 319.76 0.1 E168 192.60 0.1 (15) 4h-sulfonates:

Entr y MW Wt/V mMol KO 190.65 0.1 K00 190.65 0.1 K1 211.07 0.1 K2 221.62 0.1 K3 206.65 0.1 K4 217.63 0.1 K5 182.65 0.1 K6 254.71 0.1 3o K8 290.65 0.1 K9 195.62 0.1 K10 253.06 0.1 K11 327.71 0.1 K12 227.67 0.1 K19 229.09 0.1 K21 218.62 0.1 K23 234.69 0.1 K27 247.95 0.1 K30 252.67 0.1 K52 279.79 0.1 K53 249.70 0.1 K54 317.69 0.1 1o K55 249.70 0.1 K56 330.74 0.1 K59 301.12 0.1 K60 259.74 0.1 K61 306.82 0.1 K66 237.66 0.1 K69 181.60 0.1 K70 256.71 0.1 K76 262.72 0.1 K83 234, 69 0:1 (16) 4h-ethers:

Materials MW Wt/V mMol M1 173 0.1 M2 128.55 0.1 M3 112.10 0.1 M5 162.11 0.1 M6 137.18 0.1 M14 148.16 0.1 M24 161.16 0.1 3o M34 213.15 0.1 M35 146.14 0.1 (17) 4i-sulfonates:

Entry MW Wt/V mMol KO 190.65 0.1 K00 190.65 0.1 K1 211.07 0.1 K2 221.62 0.1 K3 206.65 0.1 K5 182.65 0.1 K6 254.71 0.1 K8 290.65 0.1 1o K10 253.06 0.1 K11 327.71 0.1 K12 227.67 0.1 K16 438.33 0.1 K19 229.09 0.1 K21 218.62 0.1 K22 253.07 0.1 K23 234.69 0.1 K52 279.79 0.1 K60 259.74 0.1 2o K70 256.71 0.1 K76 262.72 0.1 K83 234,69 0.1 K90 194.61 0.1 K91 255.52 0.1 K92 232.73 0.1 K93 244.62 0.1 K94 245.51 0.1 K96 256.06 0.1 K97 289.62 0.1 3o K98 302.86 0.1 K100 235.65 0.1 K101 221.62 0.1 K102 201.63 0.1 K104 229.06 0.1 K105 279.07 0.1 K106 245.51 0.1 K107 212.60 0.1 K109 312.62 0.1 K110 240.71 0.1 K111 330.74 0.1 K112 230.59 0.1 K113 242.69 0.1 to K115 243.67 . 0.1 (18) 4j-sulfonates:

Entr y MW Wt/V mMol KO 190.65 0.1 K1 211.07 0.1 K2 221.62 0.1 K3 206.65 0.1 K8 290.65 0.1 K21 218.62 0.1 2o K60 259.74 0.1 K70 256.71 0.1 K83 234,69 0.1 K90 194.61 0.1 K91 255.52 0.1 K93 244.62 0.1 K94 245.51 0.1 K97 289.62 0.1 K98 302.86 0.1 K100 235.65 0.1 K101 221.62 0.1 K102 201.63 0.1 B. Combinatorial synthesis.

Procedure: To a solution of 3c (4.0 mmol), DMAP (9.8 mg, 0.08 mmol), pyridine (576.8 mg. 8.0 mmol) in CHZC12 (15 mL) was drop a solution of the acyl chlorides in CH2C12 (5 mL) into the flask in 5 min.
s The mixture was stirred at rt. for 24 h, washed with 1 N NaHC03 and dried over Na2S09. Removal of the solvents gave a crude product (1.35 g). The products were separated with HPLC (4.6 X 25 cm, C-18 Column;
f low rate : 1. 0 mL/min; 0 min : H20 ( 7 0 ) , CH3CN ( 12 ) , CH30H
( 18 ) ; 2 0 min : H20 ( 5 0 ) , CH3CN ( 2 0 ) , CH30H ( 3 0 ) ; 2 2 min H20 ( 50 ) , CH3CN ( 0 ) , CH30H ( 50 ) ; 55 min : H20 ( 17 ) , CH3CN(0), CH30H (83); post run; 10 min).
(19) 4c-esters:

Entry MW mMol EO 140.57 0.25 mmol E00 120.58 0.25 mmol E1 182.65 0.25 mmol E2 252.62 0.25 mmol 2o E4 186.66 0.25 mmol E5 212.53 0.25 mmol E6 194.54 0.25 mmol E7 198.60 0.25 mmol E8 212.63 0.25 mmol E10 108.52 0.25 mmol E11 104.53 0.25 mmol E12 178.02 0.25 mmol E13 178.02 0.25 mmol E14 211.98 0.25 mmol 3o E15 131.52 0.25 mmol E16 130.53 0.25 mmol Scheme 2 //O //O
o~ O o~ OI' HO~ 3~ N3~N~ .--- ~5 O O
O O O-~ O
HZN~N ~ RHN~N

1. Preparation of precursors of libraries:
Azide 5: To a solution of 3c (0.52 g, 2.4 mmol ) and Ph3P ( 0 . 7 9 g, 3 . 0 mmol ) in THF was dropped DEAD (0.56 g, 3.2 mmol) and DPPA (0.83 g, 3.0 mmol) at 0° C, respectively. The mixture was allowed to warm to rt. The mixture was stirred at rt for 2 h. Removal of volatile materials gave a residue, which was to purified by column chromatography to afford a pure product (5) (0.5 g, 87 ~).
Amine 6: To a solution of 5 (0.48 g, 2.0 mmol ) in THF was added Ph3P ( 0 . 63 g, 2 . 4 mmol ) at rt .
The mixture was stirred at rt overnight. Removal of volatile materials gave a residue. 90 $ MeOH (20 mL) was added to the flask. The solution was stirred at rt for 2 h. Removal of the solvents gave a residue, which was purified by column chromatography to afford a pure product ( 6 ) ( 0 . 2 g, 4 7 ~ ) . bH 1 .17 ( 9H, s ) , 1.41 (2H, br) , 2 . 8-3.2 (2H, m) , 3 . 2-3. 6 (2H, m) , 4 . 07, 4.16, 4.22, 4.31 (2H, AB), 4.48-4.64 (1H, m) ppm.
2. Preparation of libraries (parallel synthesis):
Procedure: To a solution of a sulfonyl chloride (K) (0.1 mmol) in dry DCM (1 mL) were add a solution of 6 (0.1 mmol, 0.1 M) in DCM and triethyl amine (20.2 mg, 0.2 mmol). The mixture was standing at rt for 6 h. The reaction was completed. The product was purified with preparative TLC.
(20) Library 7-sulfonamides:

Entr y Materials MW Wt/V mMol K2 221.62 mg 0.1 K4 217.63 mg 0.1 K5 182.65 ing 0.1 K8 290.65 mg 0.1 K9 195.62 mg 0.1 to K10 253.06 mg 0.1 K11 327.71 mg 0.1 K12 227.67 mg 0.1 K21 218.62 mg 0.1 K22 253.06 mg 0.1 K23 234.68 mg 0.1 K83 234.68 mg 0.1 Scheme 3 -~( 0 O NH ~ Ph3CO~N~CN
Ph3C0~

O
HON~CN -~ RO~N~CN

1. Preparation of precursors of libraries:
Alcohol 9: A solution of t-BuOK (10 mL, 10 mmol, 1 M) in THF was dropped into a solution of starting material 1 (3.59 g, 10 mmol) in dry THF (40 mL) under nitrogen at rt in 5 min. The mixture was stirred for 10 min at rt. Bromoacetonitrile (1.2 g, 10 mmol) was dropped into the flask in 5 min. The mixture was stirred for 1 h at rt. 10 o NH9C1 (20 mL) and hexanes (40 mL) were added, respectively. The organic phase is separated. The solvents were evaporated to give a crude product, without purification for next step. The crude product was dissolve into DCM (30 mL). Water (0.36 mL, 20 mmol) and TFA (1.71 g, 15 mmol) were added to the flask.
The mixture was stirred for 2 h at rt. Removal of volatile materials gave a residue, which was co-to evaporated with CH3CN (2 X 30 mL) to remove the trace water. Column chromatography purification (1:1 ethyl acetate/hexanes, then ethyl acetate) afforded a pure product (9) (0.59 g, 38 ~) . bH 2.49 (1H, br) , 3.35-3.66 (4H, m), 4.36 (1H, s), 4.5-4.7 (1H, m) ppm.
2. Preparation of libraries (parallel synthesis):
Procedure: (1) Esters: To a solution of an acyl chloride (E) (0.1 mmol) in dry DCM (1 mL) were add a solution of 9 (0.1 mmol, 0.1 M) in N
2o methylmorpholine and triethyl amine (20.2 mg, 0.2 mmol). The mixture was standing overnight at rt. The reaction was completed. The product was purified with preparative TLC.
(21) Library 10 -esters:
Entry MW Wt/V mMol E183 185.56 0.1 E184 230.56 0.1 (2) Sulfonates: To a solution of a sulfonyl chloride (K) (0.1 mmol) in dry DCM (1 mL) were add a solution of 9 (0.1 mmol, 0.1 M) in N-methylmorpholine and triethyl amine (20.2 mg, 0.2 mmol). The mixture was standing at rt for 6 h. The reaction was completed.
The product was purified with preparative TLC.
(22) Library 10-sulfonates:

Entr y MW Wt/V mMol K2 221.62 0.1 K96 256.06 0.1 K101 221.62 0.1 to K106 245.51 0.1 K117 221.62 0.1 Scheme 4 l/0 //
O~ O~O O O
Ph3CO~NH ~ Ph3CO~N~

O
O p O
HO~N~ ~ RO~N

1. Preparation of precursors of libraries:
Compound 11: NaH (0.44 g, 11 mmol, 60 0) was added in portions into a solution of starting material 1 (3.59 g, 10 mmol) in dry HMPA (30 mL) under nitrogen 2o at rt in 10 min. The mixture was stirred for 1 h at rt. (Me0) 2CCH3CH2Br (1 .83 g, 10 mL) was dropped into the flask in 10 min. The mixture was stirred overnight at rt, then heated to 80° C for two days.
The reaction was cooled down to rt. Con. NHqCl (10 mL) and brine (10 mL) were added. The mixture was extracted by t-BuOMe (2 X 30 mL). The organic phase was dried over Na2504. was separated. Removal of volatile materials gave a residue, which was purified by column chromatography (4:1 ethyl acetate/hexanes) to afford a pure product (11) (2.0 g, 45 ~) . sH 1.25 (3H, s), 3.1-3.7 (6H, m), 3.18 (6H, s), 4.5-4.7 (1H, m) ppm.
Compound 12: To a solution 11 (1.8 g, 3.9 mmol) in DCM (20 mL) was added TFA (1.71 g, 15 mmol) and water ( 0. 36 g) . The mixture was stirred for 1 h at rt. Removal of volatile materials gave a residue, which was partitioned in water (30 mL) and t-BuOMe (20 1o mL). The separated aqueous layer was washed with t-BuOMe (20 mL). Water was evaporated to give a residue, which was co-evaporated with CH3CN (2 X 30 mL) to remove the trace water. A product (12) (0.43 g, 64 obtained, without further purification for next step. b" 2.15 (3H, s), 3.47-3.90 (5H, m), 4.06 (1H, s), 4.59-4.72 (1H, m) ppm.
2. Preparation of libraries (parallel synthesis):
Sulfonates: To a solution of a sulfonyl 2o chloride (K) (0.1 mmol) in dry DCM (1 mL) were add a solution of 12 (0.1 mmol, 0.1 M) in DCM and triethyl amine (20.2 mg, 0.2 mmol). The mixture was standing overnight at rt. The reaction was completed. The product was purified with preparative TLC.
(23) Library 13-sulfonates:

Entr y MW Wt/V mMol K2 221.62 0.1 K5 182.65 0.1 3o K6 254.71 0.1 K10 253.06 0.1 K11 327.71 0.1 K12 227.67 0.1 K19 229.09 0.1 K21 218.62 0.1 K22 253.07 0.1 K23 234.69 0.1 K54 317.69 0.1 K60 259.74 0.1 K83 234, 69 0.1 K90 194.61 0.1 K91 255.52 0.1 l0 K92 232.73 0.1 K93 244.62 0.1 K94 245.51 0.1 K96 256.06 0.1 K97 289.62 0.1 K98 302.86 0.1 K99 266.57 0.1 K100 235.65 0.1 K101 217.63 0.1 K102 201.63 0.1 Reaction Procedure:
Amine compound synthesis:
Method 1:
a). Alkylation:
~o O Ph Ph O CH I Ph-C-O N
~ ~

H -~ h Ph-C
-O
\~

1 Cz~H~N03 Ph Exact Mass: 373.17 Ct3HmN~ Mol. Wt.: 373.44 Mol. IM.: 359.42 Materials d MW Wt/V mMol SM 359.42 14.8 g 41 60o NaH 24 2.0 g 50 G3 2.28 141.94 3.6 mL 61.5 THF 140 mL
Procedure:
1. To a solution of oxazolidinone and THF, sodium hydride power was added under N2 protection, and ice bath.
2. The mixture was stirred for half hour at 0° C, then let it warm up to room temperature.
3. G3 was added into the solution slowly, and the reaction was stirred for overnight.
4. The reaction was quenched with water and extracted with Ethyl acetate/hexane mixture. The combined organic layer was washed with NH9C1, 1s brine and dried over Na2S09, 5. The organic solvents were removed by water Rota-vap and the crude residue was carried on next step without purification.
2o b). Deprotection:

Ph C O
O~ ~

Ph- H
O TFAlCH2G2/H20 P ~~'~N
~

h C24HzsN~ ~ C5H9N~

Exact Mass: 373.17 Exact Mass: 131.06 Mol.1M.: 373.44 Mol.1M.: 131.13 Materials d MW Wt/V mMol equiv 25 SM 373.17 crude 41 TFA 1.48 114.02 4.7 mL 62 1.5 water 18 1.5 mL 82 2.0 CH2C12 10 ml 3o Procedure:

1. The mixture was stirred for 3 h at room temperature.
2. The reaction was quenched by three drops of triethyl amine and dried over Na2S09, 3. The solvents were removed by Rota-vap and the residue was purified by column chromatography. The elute solvents: 2/1 - hexane /EtOAc to 1/2 - hexane /EtOAc, then use pure EtOAc.
1o c). Tosylation and azidelation:
p 1). MsG/Et3NICH2Cl2 O C6H~~N05S
-~ Exact Mass: 209.04 HO~Ni ~ Ms-O~Ni Mol. Wt.: 209.22 ~ ~/s Exact Mass: 131.06 Mol. Wt.: 131.13 2). NaN3/ DMSO
O
N3 , CsHaN40z s Exact Mass: 156.06 Mol. Wt.: 156.14 Materials d MW Wt/V mMol Equiv SM 131.13 3.93 30 1 MsCl 1.48 114.55 3.01 39 1.3 Et3N 101 5.8 mL 42 1.4 THF

DMSO

NaN3 65 3.1 g 48 1.6 Procedure:
1). Starting material was treated with methanesulfonyl chloride in the presence of triethylamine in methylene chloride.
2) The reaction mixture was stirred at ice bath for 3 hours.
3). The reaction was washed with water and the organic layer was dried over Na2S09, 4). The organic solvent was removed to give the residue, which was treated with sodium azide in DMSO, 5) The result solution was heated up to 80° C for two hours, then diluted with water and extracted with methylene chloride.
6). The organic layer was dried over Na2S0q, l0 7). The solvent was removed and the crude was purified by flash column chromatography to afford azide compound.
d). Hydrogenation O
N3 Ni Pd/C in EtOH O

-~ HzN~N~
l -\~~ff ~~

~sHaNeOz C
s Ht oN202 Exact Mass: 156.06 Exact Mass: 130.07 Mol. Wt: 156.14 Moi. V1R.: 130.15 Oxazolidinone: 3.4 g Pd-C (10$): 800 mg EtOH: 30 mL
1. Hydrogenation bottle was charged with azide compound and EtOH.
2. Flushed with N2 3. Pd-C was deactivated with two drops of water then added into the reaction mixture.
4. Reaction was run for overnight under hydrogenater with 30 Psi (2 atmosphere) 5. TLC showed complete conversion and the reaction 3o mixture was filtered under water pump.

6. The residue (2.46 g) was obtained and used to carry on next step without purification.
Library Design:
Oxazolidinones: RC2, SP40 (0.08 nM) in Acid chlorides: E0, E2, E8, E92, E124, E154, E157, E159, E117, E120, E164, E136 Parallel synthesis procedure:
1). Oxazolidinones (0.16 nM) were made and transferred into small vials.
2). To those vials Et3N (1.5 equiv) was added.
3). After 20 mins, acid chlorides or sufonyl chlorides were added into the reaction vials.
4). The compounds were isolated by CombiFlash, sq 16x open access purification system.
Library:
Oxazolidinones: RC2, SP40 (0.08 nM) in Sufonyl chlorides: K2, K3, K4, K10, K21, K22, K23, K83, K90, K91, K92, K93, K94, K95, K96, K97, K98, K99, K100 Parallel synthesis procedure:
1). Oxazolidinones (0.16 nM) were made and transferred into small vials.
2). To those vials Et3N (1.5 equiv) was added.
3). After 20 mins, acid chlorides or sufonyl chlorides 3o were added into the reaction vials.
4). The compounds were isolated by para-TLC (2/1 -EtOAC/ Hexane).
Nitrogen linkage library compound synthesis O
HZN Ni Et3N/CH2CI2 RHN ~~,,(~/
~~N
s ~H~oNzOz Exact Mass: 130.07 Mol.1M.: 130.15 Library Design:
Oxazolidinone: amine Acid Chloride: E0, E2, E8, E92, E124, E154, E157, E159, E117, E120, E164, E136 Sufonyl chlorides: K2, K3, K4, K10, K21, K22, K23, K83, K90, K91, K92, K93, K94, K95, K96, K97, K98, K100 Parallel synthesis procedure:
1). Oxazolidinones (O.OlnM) were made and transferred into small vials.
2). To those vials Et3N (1.5 equiv) was added.
3). After 20 mins, acid chlorides or sufonyl chlorides were added into the reaction vials.
4). The compounds were isolated by para-TLC (2/1 -EtOAC/ Hexane).
Library:

HO~N~R~ Et3NlCH2Cl2 R~ ~ ~R~
~Jv ~ ~~N
s Oxazolidinones: SG3, SC3, and.SC5 (0.08 nM) in CHZC12 Acid chlorides: E0, E2, E8, E92, E124, E154, E157, E159, E117, E120, E164, E136 Parallel synthesis procedure:
1). Oxazolidinones (0.16 nM) were made and transferred into small vials.
2). To those vials Et3N (1.5 equiv) was added.
3). After 20 mins, acid chlorides or sufonyl chlorides were added into the reaction vials.
4). The compounds were isolated by para-TLC (2/1 -EtOAC/ Hexane).
io Ether type of Linkages:
o ~o O~ M N-HO~~N_ s CSHeN03 Enact Mass: 131.06 Mol. Wt.: 131.13 O
O
O--~ _ MO
HC~~N~ -C7H» ~s Exac3 Mass: 157.07 Md. Wt.: 157.17 Library Design:
Oxazolidinones: SG3, SC5 M compounds: M1, M2, M3, M4, M5, M6, M6, M11, M14, M14, M24, M30 M34, M35, M37, M38 DEAD = 0.10 nM in THF (MW 174 ) 2o Ph3P-polystyrene 1 mmol/g 100 mg for each compound 0.1 mMol THF 1 mL
Procedure:
l.The vials were charged with starting material, THF, CH2C12 and Ph3P-polystrene .

2. A solution of DEAD was added into the reaction mixture.
3. The reactions were stand for overnight.
4. Separated by pre-TLC.
N-Aryl linkage:
Using Buchwald Reaction:
_ o O Ph Ph ~ ~ ~ I ~ Ph-C-O~N I ~ NOz Ph-C O NH -~ Ph s \~

Ph Pd(OAC)2, CzsH24Nz~
K OtBu C~Hz~N03 C Exact Mass:480.17 H4BrNOz Exact Mass: s Mol. Wt.: 480.51 359. 15 Exact Mass: 200.94 Mol. Wt.: 359.42Mol. Wt.:
202.01 Materials d MW Wt/V mMol eyv SM 359.42 1.0 g 2.7 1 Bromo compound 202.01 0.76 g 3.7 1.3 Palladium(II) acetate 224.49 82mg 0.36 0.13 sodium t-butoxide 96.11 0.4 g 4.16 1.5 Ferrocene 554.40 155 mg 0.27 0.1 Toluene 140 mL
Procedure:
1. A 100 mL flask loaded with oxazolidinone, bromo compound, palladium(II) acetate, 1,1' bis(diphenylphosphino)-ferrocene and sodium t butoxide and flashed by N2 protection for 10 mins.
2. Toluene was added and heated up to 110° C for overnight and then diluted with dichloromethane after it cooled down to room temperature.

Buchwald Reaction:
_ o O NC ~ ~ Br Ph-C-O~N / ~ N
---~ ~

Ph-C-O~\\~ ----~ h NH Pd(OAC)2, Ph K O~Bu ~aH2tN~ C~H4BrN ~oHz4NzOa Exact Mass: 35A.15 Exact Mass: ~~ ~~: x.18 180.95 Mol.1M.: 359.42 Mol. lfllt.:182.02Md. VVt.: 460.52 Materials d MW Wt/V mMol equiv SM 359.42 6.0 g 16.7 1 Bromo compound 182.12 3.65 g 20 1.2 Palladium(II) acetate 224.49 487 mg 2.2 0.13 1o sodium t-butoxide 96.11 2.4 g 25 1.5 Ferrocene 554.40 926 mg 1.67 0.1 Toluene 140 mL

Procedure:
1. A 100 mL Round flask was loaded with oxazolidinone, bromo compound, palladium(II) acetate, l,1'-bis(diphenylphosphino)-ferrocene and sodium t-butoxide and flashed by N2 protection for 10 mins.
2. Toluene was added and heated up to 110° C for overnight and then cool down to room temperature, diluted with dichloromethane.
3.filtered by celite.
4. Separated by column. EtOAc/Hexane - ~ elute solvent.
Synthesis of 3-trityloxy-2-hydroxy-propylamine:

O
LiOHfi-PrOH/ MeOH 0H
Ph3CO~NH p~CO ~S~ NH2 ren~nc C23H21 N~3 C22H23N~2 Mol. Wt: 359.42 Mol. Wt: 333.42 Procedure:
1. To a 500 mL Round flask was charged with 18g SM and 90 mL of isopropyl alcohol 10 mL of MeOH then 50 mL
of LiOH saturated solution.
2. The mixture was heated under reflux overnight at ~70° C.
3. Cool down to room temperature and solvents were Zo removed on Rota-vap.
4.Extract with EtOAc (1x50mL, and 1x50mL).
5. The combined EtOAc layers was washed with saturated NaCl, and dried with anhydrous Na2S0q.
6. The solid was filtered and solution was divided into three parts and concentrated them separately.
Total 18.05 g, 100 yield was obtained.
Amine Oxazolidinone formation:
Method 2:
2o a). Hydrazine formation:
OH OH
Ph~~sl O EtOH/ NH2NH2 Ph-~~s~ O
OEt ~~~~NHNH
Ph reflex for overnight Ph-~ 2 C20H25N~3 C~sHz~Na02 Exact Mass: 327.18 Exact Mass: 329.21 Mol. Wt.: 327.42 Mol. Wt.: 329.44 Materials d MW Wt/V mMol Equiv SM 327.18 5.7 g 17.42 1 EtOH 10 mL
hydrazine 32 3 mL 26 1.5 1. To a round flask was Loaded hydrazine, EtOH and ester.
2. The reaction was heated up to reflux for overnight.
3. The solvents were removed by water rota-vap.
4. NMR showed there is no ester.
b). Curtius Rearrangement:
O
OH O---y Ph~~S~ O Ph-~~S~~NH
Phi NHNH2 NaN02/H2S04 Ph--~
C~9HZ~N302 50 ° C fOf 2 h C~$HZpN202 Exact Mass: 329.21 Exact Mass: 296.15 Mol. VIIt.: 329.44 Mol. Wt: 296.36 Materials d MW Wt/V mMol Equiv SM 329.44 17.4 1 H2S04 98 2.04 g 20.8 1.2 NaN02 69 2.4 34.8 2 2o water 17 mL
Procedure:
1). The hydrazide compound was dissolved in water (17 mL ) .
2). To the reaction mixture, concentrated sulfuric acid (2.04 g) diluted in water (10 mL) was added into the stirred solution.
3). The mixture was cooled in the ice bath and then NaN02 was added.

4). The reaction mixture was stirred at 50° C for 2 hrs.
Buchwald Reaction:
Org. Lett., Vol.2,No.8,2000 Pd-Catalyzed Amination of Activated Aryl Halides:
~e~4~:
O Br~NOy and wss: zoo.er O
Md.lAh.: 202.01 N O~ H Ph N ~ \ / NOz Ph~ -Pd(OAC)2, Cs2C03 Phv XaMphos, 1,4-dioxane Materials d MW Wt/V mMol equiv SM 359.42 454 mg 1.26 1 Bromo compound 202 305 mg 1.5 1.2 Pd(OAC)2 224.49 2.9 mg 0.013 0.01 Cesium carbonate 325.82 575 mg 1.8 1.4 Xantphos 578.63 10 mg 0.018 .015 1,4-dioxane 3 mL

2o Procedure:
1. A 10 mL Round flask was loaded with oxazolidinone, bromo compound, palladium(II) acetate, Xantphos and Cesium carbonate.
2. The flask was back-filled N2 for 10 mins.
3. 1,4-dioxane was added and heated up to100° C for overnight and then cool down to room temperature, diluted with dichloromethane.
4.filtered by silicon gel.
5.
3o Oxazolidinone MW

SC2 169.18 B11 295.26 SG3 131.13 K2 221.62 K10 253.06 K6 254.72 1o Desired products:
SC2K2, SC3K2, SC5K2, SC5K10, SG2K10, B11K6, SG3K2 0.2 nmol of starting material were used in the presence of 3 equivalent of triethylamine as base in 1 mL of dichloromethane. The reactions were stirred for overnight.
Remake some of the library compounds for testing according to the result on March 14, 2002.
Oxazolidinone MW

2o SC3 184 SG3 131.13 K2 221.62 K10 253.06 K3 206.65 K23 234.68 K22 254.12 SG3-N 130.07 E8 212.63 Desired products:
SC3E8, SG3E8, SG3-N-E8, SG3-N-K2, SG3-N-K3, Procedure:

0.1 nmol of starting material were used in the presence of 1.5 equivalent of triethylamine as base in 1 mL of dichloromethane. The reactions were stirred for overnight.
E112-Oxazolidione Library compound synthesis:
O-linkage and N-linkage:
Oxazolidinone MW

1o SC2 169.18 B11 295.26 SG3 131.1 SG3-NH- 130.07 2o Desired products:
SC3E112, SG3-N-E112, SG3 -E112, SC1E112, B11E112.
0 0 _ ph, O~N \ / N~ Pd(OH)2lTHF/MeOH p~N ~ ~ NH

PhJ ~ ~ HZN
300 mg of ZD3-75-2 500 mg of Pd(OH)2 5 mL of THF
5 mL of MeOH
3o The reaction was stirred at room temperature 10 mins (see new spot and starting material on TLC, new spot is more polar) and 40 mins (see one new spot, which is less polar than starting material, and only one spot shown on TLC).
O CtoHtaNa~
Mol.1M.: 207.23 O N ~ ~ NH2 O O O
HZN~ Et3l~lH/CH2C12 ~N~N-~NH

(w ( O ._O CtsHtaFNaOs j'~/~~ CgHsCIF03 Mol. Wt.: 387.36 F ~ Mol. Wt.: 216.59 O o O O i-0 O', o, ~N.J~rN~NH~F
F
C~H~FZN308 Mol. Wt.: 567.49 Variety of E112-Oxazolidione library compound synthesis:
Oxazolidinone MW

RC2 169.18 SG3 131.1 CsH6CIF03 F I Moi.111It.:216.59 O-~
O O
C
Desired products:

RC5E112 RC1E112 ZD3-87-E112 (387.36 or 567.49), ZD3-88-1 (206.24), ZD3-88-2 (206.24), ZD3-88-3 (206.24) Exploring new Linkages:
1). Copper-Catalyzed Coupling of Alkylamines and Aryl Iodides CSHtoNzOz Mol. Wt.:130.15 5 ~I% Cul O
2 equiv K3P04 O
' n HOCH2CH20H
2 equ -NCO
NHZ
~

,, Isopropanol, ~NH

C,~HuN~

Mol. VYt.:
206.24 C~H51 Mot. Wt.: 204.01 Materials d MW Wt/V mMol equiv SM 130.15 100 mg 1 Iodo compound 204.0 170 mg 1.1 CuI 190.44 7.6 mg 0.05 K3pp4 212.5 322 mg 2 HO (CH2) 20H 1.13 62 . 07 0 .1 mL 2 Isopropanol 1 mL

2o Procedure:
To a schlenk tube, CuI and K3P09 were added then the tube was back-filled with nitrogen for 10 mins, and then rest of starting material were added, the reaction mixture was heated up to 80° C.
Results and discussion:
Desired product was obtained by para-TLC.
(2/1 = EtOAc/Hexane).

,.O
P _~\h Ph-C-0~N~N~
,O CNH ~h s P~h _~// C2eHaoN2oa Ph-C-0~NH Mol. Wt.: 442.55 i CH20, Ethanol, refl~c for 2 h Ph CzaH2~N~
Exact Mass: 359.15 Mol. Wt.: 359.42 Materials d MW Wt/V mMol equiv SM 359.42 0.5 g 2.78 1 Pyrrolidine 0.2 mL
37$ HCHO 0.2 mL
Ethanol 6 mL
Procedure:
1o A solution of SM, pyrrolidine and formaldehyde in ethanol (6 mL) was refluxed for 2 h.
The solvent was evaporated.
Copper-Catalyzed Coupling of Alkylamines and Aryl Iodides Organic Lett. 2002 Vol.4, No.4. page 581-584 ~H~oN2oz Mol. Wt.:130.15 5 ~% Cul 0 2 equiv K3P04 o ~NH2 2 equiv HOCH2CH20HNCO
-~soProPanot, ~---~NH~
~

C w C~~H~~N~

~HS~ Mol. Wt: 206.24 Mol. Wt.: 204.01 2o Materials mMol equiv d MW Wt/V

SM 130.15 100mg 1 Iodo compound 209.0 170mg 1.1 CuI 190.44 7.6 mg 0.05 K3P09 212.5 322 mg 2 HO(CH2)20H 1.13 62.07 0.1 mL 2 Isopropanol 1 mL
Procedure:
To a schlenk tube, CuI and K3POQ were added then the tube was back-filled with Nitrogen for 10 mins, and then rest of starting material were added, the reaction mixture was heated up to 80° C.
to Copper-Catalyzed Coupling of Alkylamines Aryl and Iodides Organic Lett. 2002 Vol.4, No.4. page 581-584 CsHtoNzOz Mol. Wt.:130.15 p 5 mol% Cul 2 equ'rv K3P04 C
' rv HOCH2CH20H ~~C OCH3 ~-~,.NH 2 equ Isopropanol, ~NH-~

OCH3 gp C

t C~zH~sNz~a Mot. Ylh.:
236.27 C~H~IO

Mol. Vlft.: 234.03 Materials d MW Wt/V mMol equiv SM 130.15 100 mg 0.76 1 Iodo compound 234.0 211 mg 0.84 1.1 CuI 190.44 7.6mg 0.05 2o K3P0q 212.5 322 mg 2 HO(CH2)20H 1.13 62.07 0.1 mL 2 Isopropanol 1 mL

Procedure:
To a schlenk tube, CuI and K3P04 were added then the tube was back-filled with Nitrogen for 10 mins, and then rest of starting material were added, the reaction mixture was heated up to 90° C.

Copper-Catalyzed Coupling of Alkylamines and Aryl Iodides Organic Left. 2002 Vol.4, No.4. page 581-584 CsH~oNzOz Mol.1M.:130.15 O 5 rtal% Cul -NCO 2 equiv K3P04 p ~N~ 2 equrv HOCH2CH20H_NJb kopropanol, o ~-~~,.NH

\
I

C~zHtsN20z ~H~I Mol. Wt.:
220.27 Mol. Wt.: 218.03 Materials d MW Wt/V mMol equiv SM 130.15 40 mg 1 1o Iodo compound 218.0 72 mg 1.1 CuI 190.44 3 mg 0.05 K3P09 212 . 5 12 8 2 mg HO(CH2)20H 1.13 62.07 0.1 mL 2 Isopropanol 1 mL

is Procedure:
To a schlenk tube, CuI and K3P09 were added then the tube was back-filled with Nitrogen for 10 mins, and then rest of starting material were added, 2o the reaction mixture was heated up to 90° C.
CAN. J.CHEM.Vo1.61, 411 (1983) O NH Ph O-O V Ph--C-O~N~;.
Ph O~ Ph \~NH
Ph-C O .~l~J~ CH20, Ethanol, reflux for 2 h ~8H30Nz04 Ph Exact Mass: 458.22 Mol. Wt.: 458.55 CzaH2~N~a Exact Mass: 359.15 Mol. Wt.: 359.42 Materials d MW Wt/V mMol equiv SM 359.42 0.68 g 2.78 1 Morpholine 87.12 0.4 mL
37 ~ HCHO 0.4 mL
Ethanol 8 mL
Procedure:
A solution of SM, morpholine and 1o formaldehyde in 8 mL in ethanol was refluxed for 4 h.
The solvent was evaporated.
CNH
_O
,.O
O ~O
HO~NH CEO, Ethanol, re8~c for 2 h HO~N~N
s Exact Mass:~117.04 ~H,eN~03 Exact Mass: 200.12 Mol. Wt.: 117.10 Mol.1M.: 200.24 Materials d MW Wt/V mMol equiv SM 117 0.47 g Pyrrolidine 0.2 mL
37 ~ HCHO 0.4 mL
Ethanol 3 mL
Procedure:
A solution of SM, morpholine and formaldehyde in 3 mL in ethanol was refluxed for 2 h.
The solvent was evaporated.
Deprotection:

O o Ph Ph--C-O~N~N~ TFA/ CH2Ci2/ H20 Hp~N~N
s Ph CsHtsNzpa CzsHaoNzOa Exact Mass: 200.12 Md.1M.: 442.55 Mol. Wt: 200.24 Materials d MW Wt/V mMol equiv SM 442.55 24 mg 0.05 1 TFA one drop H20 one drop CHZC12 1 mL
1o Stir for overnight.
CtoHtaNaOz p~N ~ ~ NHz Exact Mass: 207.10 Mol. Wt.: 207.23 H2N R) D~Iue~eIMeOH
CZZHa3NaOtz (s) Exact Mass: 531.21 O Mol. Wt.: 531.51 HO
HO (s) ~s) OH
OH
HO (s) CsHtzOs ~ HN~--Nip O
Mol. Wt.: 180.16 ~s) ~ ~NH H H
OH t~ HO
amine compound: 18 mg D-glucose: 140 mg 1s MeOH: 3 mL
Procedure:
To a schlenk tube was added Amine compound, D-glucose and MeOH. The reaction was heated up to 60°
2o C for overnight.
Copper-Catalyzed Coupling of Alkylamines and Aryl Iodides Organic Lett. 2002 Vol.4, No.4. page 581-584 CsH~oNzOz Mol. Vlft .: 130.15 O 5 mol%
Cut 2 equiv tCiP04 O

~NH 2 e9uN

_NJlO

Isopropanol, o ~NH~

t ~ OCH3 C

C~3H~N~

~H~Ip Exact Mass: 252.15 Exact Mass: 233.95 Mol. Wt.: 252.31 Moi. Wt: 234.03 Materials d MW Wt/V mMol equiv SM 130.15 40 mg 1 Iodo compound 218.0 72 mg 1.1 CuI 190.44 3 mg 0.05 K3P09 212.5 128 mg 2 HO(CH2)20H 1.13 62.07 0.1 mL 2 1o Isopropanol 1 mL

Procedure:
To a schlenk tube, CuI and K3P0q were added then the tube was back-filled with Nitrogen for 10 mins, and then rest of starting material were added, the reaction mixture was heated up to 95° C for overnight.
Copper-Catalyzed Coupling of Alkylamines and Aryl 2o Iodides Organic Lett. 2002 Vol.4, No.4. page 581-584 CeHtoNzdt Mol.1M. : 130.15 O 5 mol% Cul -NCO 2 equiv K3P04O

~NH 2 equiv HOCH2CH20H
2 ~NJIO

Isopropanol, NH
t 80 C

COOH
HOOC I

C~zHmNzOa C~H510z Exact Mass: 250.10 Exact Mass: 247.93 Mol. Wt.: 250.25 Mol. Wt: 248.02 Materials d MW Wt/V mMol equiv SM 130.15 40 mg 1 Iodo compound 248.0 72 mg 1.0 CuI 190.44 3 mg 0.05 K3P04 212.5 128 mg 2 HO(CH2)20H 1.13 62.07 0.1 mL 2 Isopropanol 1 mL

Procedure:
To a schlenk tube, CuI and K3P04 were added 1o then the tube was back-filled with Nitrogen for 10 mins, and then rest of starting material were added, the reaction mixture was heat ed up to 95 C for overnight.

O O
--N~O -N~O

I~NHz ~NH ~ ~ NOZ

CH3CN/ Hunig's I
base F

50 C Cf1H12FN3~4 I ~ ~ NOZ Exact Mass: 269.08 Mol. Wt.: 269.23 F

CsH3FIN02 Exact Mass: 266.92 Mol. Vltt.: 267.00 The solution of 4 equiv of Hunig's base and 1 equiv of amine starting material in CH3CN was stirred at 50° C for overnight.
Reduction of nitro group O
Ph~ O N ~ ~ NOZ HCOONH4 phl O"N ~ ~ NH2 (N ~ Pd-C, MeOH N
PhJ
Ph--~
C24H23N3~4 C24H25N3~2 Exact Mass: 417.17 Exact Mass: 387.19 Mol. Wt.: 417.46 Mol. Wt.: 387.47 Materials d MW Wt/V mMol equiv SM 417.46 70 mg 0.24 1 HCOONHq 63 56 0.88 3.6 Pd/C (Wt) 10 THF 0.4 mL
MeOH 0.4 mL
1. To a solution of oxazolidinone and THF-MeOH(1:1) was .Ammonium formate and Pd-C.
2. The reaction was stirred at room temperature for 2 h.
3. Diluted with THF.
4. Filtered and washed several times with THF.
5. Filtrate concentrated to a dark yellow solid.
Reduction of nitro group Pd(OH)2lfHF/MeOH
p --i 'N O _ I~NH ~ 7 N02 H2 I~'--~NH ~ ~ NHZ
F F
Ct~H~2FNa0~ Ct~H~4FNaOz Exact Mass: 269.08 Exact Mass: 239.11 Mol. Wt.: 269.23 Mol. Wt.: 239.25 Materials d MW Wt/V mMol equiv SM 269.23 120 0.45 1 Pd(OH)2 10$
THF 1 mL

MeOH 1 mL
The reaction was stirred at room temperature under H2 for 2 hrs .
Deprotection:
o _ ~o Ph-C-O~N~ ~O TFA/CH2Ci2/H20 HO O N~ ~
-. ~(s~/ N~
~aH3oN20a Enact Mass: 458.22 CsHtsN20a Mol.1M.: 458.5fi Exact Mass: 216.11 Mol. Vlft.: 216.23 Materials d MW Wt/V mMol equiv to SM 458.55 800 mg 0.57 1 TFA 0.3 mL
H20 0 .1 mL
CH2C12 3 mL
CsHtoN202 Mol. Wt.: 130.15 O 5 mol% Cui -N~ 2 equ~i K3P04 O
NHZ 2 equ'n HOCH2CH20H ,NCO
~NH-p,A
AA group I I
t , ~N i I ~ NOZ N~ C6H41N0z CsHaIN02 CsHaIN02 Exact Mass: 248.93 Exact Mass: 248.93 ~~ ~~: 248.93 Mol. Wt; 249.01 Mol. Wk: 249.01 Md. Wt.: 249.01 I CN'/I
JTN
02N~NOz CaH31N2 Exact Mass:
205.93 CsH3INZOa Mol. Wt.:
205.98 Exact Mass:
293.91 Mol. Wt.:
294.00 Materials d MW Wt/V mMol equiv SM 130.15 40 mg 0.31 1 CuI 190.44 3 mg 0.05 K3P09 212 . 5 12 8 mg 2 HO(CH2)20H 1.13 62.07 0.1 mL 2 Isopropanol 1 mL

AA Group: 0.31 mmol to AA6 77 mg AA7 77 mg AA9 91 mg AA12 65 mg Procedure:
To a test tube, CuI and K3P09 were added then the tube was back-filled with Nitrogen for 10 mins, and then rest of starting material were added, the 2o reaction mixture was heated up to 80° C for overnight.
CsH,oNz~z Mol. Wt.: 130.15 O 5 moi% Cul 2 equiv IG3P04 O
~NH 2 a9uiv HOCH2CH20H _N~O
z Isopropanol, ~ NH
80°C
1 NOz OZN I ~ Exact~Mass 251.09 Mol. Wt.: 251.24 CsH~INOz Exact Mass: 248.93 Mol. Wt.: 249.01 Materials d MW . Wt/V mMol equiv SM 130.15 200 mg CuI 190.44 15 mg K3POa 212.5 640 mg HO(CH2)20H 1.13 62.07 0.5 mL

Isopropanol 2 mL

AA8 249 385 mg Procedure:

To a t est tube, added then CuI and K3P04 were the tube was back-filled with Nitrogen for 10 mins, and then rest of starting materialwere added, the reaction mixture was heated C for up to 90 overnight.

C5H1oN2~2 Mol.1M.: 130.1 5 O 5 mol% Cul -NCO 2 equH lC3P04O

~NH 2 equiv HOCH2_CH20H_NJ'O
z panol, ~NH-AA

80 C ~'r AA grou p I
I 1 w I w I I w I 1 w I I w ~ I ~- F3C ~ ~ NHz F ~ COCH3 I w C I
~ , ~ COOH ~dN 3 OOH 07t'taFSlCeH~IO
COOH

Mol. Mpl, O~~ 0~~~ Mol Y11L: VYL: Y12.
272 219.02 272.01 01 Mot YK.
246.05 Mol Nrt.. 248.02 Mot .
Wt. 248.02 C71'ISIOi AA18 AA19 AA13 Mot. wc. z4a.o2 ~1 T

Materials d MW Wt/V mMol equiv SM 130.15 40 mg 0.31 1 CuI 190.44 3 mg 0.05 K3P04 212 . 5 12 8 2 mg HO(CH2)20H 1.13 62.07 0.1 mL 2 Isopropanol 1 mL

AA Group: 0.31 mmol 2o AA13, AA14, AA15 77 mg ~ AA16, 84 mg ; AA17 68 mg ; AA19 77 mg Procedure:
To a small vial, CuI and K3P04 were added then the tube was back-filled with Nitrogen for 10 mins, and then rest of starting material were added, the reaction mixture was heated up to 80° C for overnight.
CAN. J.CHEM.Vo1.61, 411 (1983) O NH
O '--i O
Ph Ph N ~H N ~ vN~O
~~ ~sJ~/ CH20, Etharal, refl~nc fog 2 h Ph C~aH2o~'hoz CzaHzsNa~
Mol. VIIt.: 296.36 Mol. Wt: 395.49 Materials d MW Wt/V mMol equiv SM 296 0.1 g 0.4 1 1o Morpholine 87.12 0.1 mL
37~ HCHO 0.1 mL
Ethanol 1.5 mL
Procedure:
s5 A solution of SM, morpholine and formaldehyde in 1.5 mL in ethanol was refluxed for 2 h. The solvent was evaporated.
CNH
Phi - \O Phi ' \O
~NH N~N~N
s CH20, Ethanol, reflmc for 2 h C~eH2oN20z GzathsNs02 Mol. Wt: 296.36 Mol. Wt: 379.50 2o Materials d MW Wt/V mMol equiv SM 296.36 0.1 g Pyrrolidine 0.1 mL
37$ HCHO 0.1 mL
Ethanol 1.5 mL
Procedure:

A solution of SM, morpholine and formaldehyde in 1.5 mL in ethanol was refluxed for 2 h. The solvent was evaporated.
Ether type of Linkages.:

O-~ M-O~N-HO~~N- ~ s ~/s CSHsN03 Exact Mass: 131.06 Mol. Wt: 131.13 Library Design:
Oxazolidinones: SG3, 130 mg in 5 mL of CHC12. 0.5 mL
1o was took for each reaction.
M compounds: M1, M4, M5, M11, M14, M24, M30, M35, M37, DIAD = 0.10 nM in THF (MW 202), 202 mg in 10 mL THF.
1 mL was for each reaction.
Ph3P-polystyrene 1 mmol/g 100 mg for each compound 0.1 mMol Procedure:
l.The vials were charged with SM and Ph3P-polystrene.
2. A solution of DIAD in THF was added into the reaction mixture.
3. The reactions were stirred for overnight.
Ether type of Linkages:
o ~o O-~ M-O N_ HO~~N-~/s CsHyN03 Exact Mass: 131.06 Moi. Wt.: 131.13 Library Design:
Oxazolidinones: SG3, 130 mg in 5 mL of CHC12. 0.5 mL
was took for each reaction.
M compounds: M9, M13, M20, M21, M22, M23, M23, M29, M32, M33 DIAD = 0.10 nM in THF (MW 202), 202 mg in 10 mL THF
and 1.5 mL of DMPU as co-solvent. 1.2 mL was took for each reaction.
1o Ph3P-polystyrene 1 mmol/g 100 mg for each compound 0.1 mMol Procedure:
1. The vials were charged with SM and Ph3P-polystrene.
2. A solution of DIAD in THF was added into the reaction mixture.
3. The reactions were stirred for overnight.
Acid hydrazide from ester OH
O OH
O N ~S/ EtOH/ N H2N H2 ~ S O
OEt O N
reflux for overnight ~ NHNH2 CloHlsNOa C8H17N3~3 Mol. Wt.: 217.26 Mol. Wt.: 203.24 Materials d MW Wt/V mMol Equiv SM 217.26 2 g 9.2 1 EtOH 5 mL
hydrazine 32 588 mg 18.4 2 1. Loaded hydrazine and EtOH with a round flask and ester was added slowly.

2. The reaction was heated up to reflux for overnight.
3. The solvents were removed by water rota-vap.
4.NMR showed there is no ester.
s Curtius Rearrangement:
O
OH O
O NaN02/H2S04 p ~S~ NH
NHNH2 50 ° C for 2 h C H N O
CgH~~N3O3 8 14 2 3 Moi. VI/t.: 203.24 Mol. Wt.: 186.21 1o Materials d MW Wt/V mMol Equiv SM 203 9.2 1 H2S09 98 1.0 g 11 1.2 NaN02 69 1.27 18.4 2 water 15 mL
is Procedure:
1). The hydrazide compound was dissolved in water (7 mL), and concentrated sulfuric acid (1.0 g) diluted in water (3 mL) and added into the stirred 2o solution. The mixture was cooled in the ice bath and then NaN02 (in 5 mL water) was added slowly.
2). The reaction mixture was stirred at 50° C
for overnight.
25 Library:
SG3-N- MW 130.15 SG3 MW 131.13 For SG3: K107, K96, K100, K114, K115,K76 For SG3-N: K76, K96, K100, K101, K112, K114, K115. K2 Procedure:
0.10 mol of S
0.15 mmol of Et3N
0.08 mmol of K compounds Acid hydrazide from ester OH
Phi O OH
N (s) EtOH/ NH2NH2 Ph-~ O
Phi OEt N (s) reflex for overnight Phi NHNH2 C21 H~NO3 ClsHz3Na02 Mol. Wt.: 343.46 Mol. Wt.: 313.39 Materials d MW Wt/V mMol Equiv SM 343.46 16 g 46.6 1 EtOH 30 mL
hydrazine 32 2.2 g 70 1.5 1. Loaded hydrazine and EtOH with a schlenk tube and ester.
2. The reaction was heated up to 80° C for overnight.
3. The solvents were removed by water rota-vap.
Ether type of Linkages:
o ~o O--~ M-~N-HO~\~N_ ~ s CSHgN03 Exact Mass: 131.06 Mol. Wt: 131.13 Library Design:

Oxazolidinones: SG3, 104 mg in 4 mL of CH2C12. 0.5 mL
was took for each reaction.
M compounds: M2, M3, M6, M34, M38, M39, M40, M41, DIAD = 0.10 nM in THF (MW 202), 160 mg in 8 mL THF.
1 mL was for each reaction.
Ph3P-polystyrene 1 mmol/g 100 mg for each compound 0.1 mMol Procedure:
l.The vials were charged with SM and Ph3P-polystrene.
2. A solution of DIAD in THF was added into the reaction mixture.
3. The reactions were stirred for overnight.
Nitro compounds with special linkages, ,.o o~l~( ~5 0 HO~\~N- ~ OzN / \ ~_~N-GsHsNO _~a Exact Mass: 131.06 CtzHtzNzg Md. Wk: 131.13 Mol.1M.: 296.23 ,,O
~O
OzN / \ S-O
s K117 CttHtzNzasS
Md. Wt.: 284.29 OZN ~ O
~N-O
CttHtzN2gS
O Mol. Wt.: 31629 ~N / \ O-O
N-Ct2HtzNzOs Mol. Vlft.: 280.23 SG3-N- MW 130.15 SG3 MW 131.13 For SG3 and SG3-N: K117, BB3, BBS, E183 Procedure:
0.1 mmol of Starting material 0.15 mmol of Et3N
0.08 mmol of K and B compounds Exploration of different linkages Alkylation:

O~ G16 O
Ph-C-O~\\~NH ~ Ph Ph ~~~ N~~
Ph HZ
C2aHZtNOa Mol. Wt.: 359.42 G~H24CIN03 Mol. Wt.: 421.92 ~~O
Ph p~ G14 O
Ph P-O~\~~NH ~ Ph-Ch O O
~CF3 CzaH2tN~ Ph Mol. Wt.: 359.42 C~H~F3N03 Mol.111k.: 441.44 For G16, Materials MW Wt/V mMol Equiv SM 359.42 1 g 2.78 1 G16 143.42 0.6 mL
t-BuOK 3 mL
2o THF 8 mL
For G14 Materials M Wt/V mMol Equiv SM 359.42 1 g 2.78 1 G14 (d 2.1) 209.94 0.4 mL 4.17 1.5 t-BuOK 3 mL
THF 8 mL
Alkylation: (By NaH) ,.o Ph -~\ G16 O
Ph-C-O~NH ~ Ph p ~h s Phi-O~\~N~~CI
Cz3H21Np3 h Hy Mol. Wt.: 359.42 C2~HZ~CIN03 Mol. Wt.: 421.92 O
Ph O~ G14 O
Ph-C-O~\\~NH ~ Ph O
Ph ~~ 5 N~F3 C~Hz~NOa Ph Mol. Vlft.: 359.42 C~H22p3N03 Mol. Wt.: 441.44 For G16, 1o Materials MW Wt/V mMol Equiv SM 359.42 1 g 2.78 1 G16 143.42 0.6 mL
NaH (60$) 24 166 mg 4.17 1.5 THF 8 mL
For G14 Materials MW Wt/V mMol Equiv SM 359.42 1 g 2.78 1 G14 (d 2.1) 209.94 0.4 mL 4.17 1.5 2o NaH (60$) 24 166 mg 4.17 1.5 THF 8 mL

Alkylation: (By NaH) Ph p~ G15 Ph p-//
Ph-C-C1~~NH ~ Ph-C-O~~N~
Ph S Ph CzsHztN~ czs~+n~
Mol.1M.: 359.42 ~~ ~.. 4ot.5o For G16, Materials MW Wt/V mMol Equiv SM 359.42 6.5 g 18.1 1 G15(d1.74) 170 2.5 mL
NaH (60$) 24 941.2 mg THF 50 mL
to Deprotection: (G15) Ph ~-C~N~ ~ HO~N~
P ~ l~' ~/ls~/J
d.~w~ ~l.so ~HtaN~
Mol. WL: 159.18 TFA 2 mL
H20 0.5 mL
CH2C12 5 mL
Deprotection: (combine 1 g of the reaction of G16 with BuOK and with NaH) O O
Ph Ph-C-O~ ~G p N~
~/~/ H
Ph CsHtoGN03 C2sHzaG~a Mol.1M.: 179.60 Mol. Vlft.: 437.96 T FA 1 mL
H20 0 . 3 mL
CH2C12 3 mL

Glycosylation:

-N~O _ (~NH ~ ~ ' NH2 F
CttH~aFNaOz Exact Mass: 239.11 O OH
Mol. Wt.: 239.25 D-Glucose/MeOH _ ~ (R) O
OH N~NH~-NH OH
(s) O 60 °C (s) F (s) (R) OH
HO ~ HO
HO (s) C1~H24FN30~
(s) O Exact Mass: 401.16 OH Mol. Wt.: 401.39 CsHt20s Mol. Wt.: 180.16 5 mg of starting material mg of sugar 1 mL of MeOH
l0 Glycosylation C24H25N3~2 PhNy / \ NH2 Exact Mass: 387.19 Moi. Wt.: 387.47 Ph-~ (R) D-GlucoselMeOH OH
O (R) (s) OH O 60 °C Ph~ OxN / \ NH O OH H
Ph N~ (s) (R) HO " HO
HO (s) (s) O C H N O
OH 30 3a s s Exact Mass: 532.24 CsH~20s Mol. Wt.: 180.16 Mol. VIIt.: 532.61 10 mg of starting material 18 mg of sugar 1 mL of MeOH

Deprotection:

Ph O~ ~ TFA/CH2Ct2JH20 O
Ph-C-O~N N
Ph ~ HO~N~N
~oHz4Nz~ CttHtoN2~
Exact Mass: 460.18 Exact Mass: 218.07 Mol. Wt.: 460.52 Mol. Wt.: 218.21 SM 700 mg TFA 1.2 mL
H20 0 . 3 mL
CHZC12 3 mL
1o The mixture was stirred for two hours. The TLC showed no starting material left.
Library:
SG3-N- MW 130.15 SG3 MW 131.13 SC3 MW 183.20 K100 (235.65), K101(221.62), K102(201.63), K103(339.51), K104 (229.06), K105 (279.06), K106 (245.51), K107 (212.60), K108 (372.67), K109 (312.62) 2o K110 (240.71), K111 (330.74) Procedure:
0.11 mol of S
0.15 mmol of Et3N
0.08 mmol of K compounds New library linkage:

HN~ rPh THFRBuOK R-Nip rPh N GorC ~N

~Ph ~S ~Ph CtaH2oN20z G2, G3, C1, C3.
C2, C5 Mol. Wt.: 296.36 Copper-Catalyzed Coupling of Alkylamines and Aryl Iodides Organic Lett. 2002 Vol.4, No.4. page 581-584 C.slitoN202 O
Mol. Wt.: 130.15 5 ~% Cul --NCO
2 equiv IG3P04 ~-~.NH~
-N O 2 equ'rv HOCH2CH20H Fa NHZ Isopropa~ CtzHtaFsNzOz Mol.1M.: 274.24 ~I

C~H4F31 I OH
Mol. Wt.: 272.01 ~N NH
F3C~ ~~ ~.CF

CteHtaFsNzO
Mol. Wt.: 392.34 Materials d MW Wt/V mMol equiv SM 131 100 mg 1 to Iodo compound 272.01 500 mg 1.8 CuI 190.44 19 mg 0.1 K3P04 212.5 322 mg 2 HO(CH2)20H 1.13 62.07 0.15 mL 2 Isopropanol 1 mL

Procedure:
To a small vial, CuI and K3P04 were added then the tube was back-filled with Nitrogen for 10 mins, and then rest of starting material were added, 2o the reaction mixture was heated up to 60° C for overnight, and Isolated by parp-TLC.
CAN. J.CHEM.Vo1.61, 411 (1983) O
n p O ~NH HO~N~NVO
O
HO ~ H CsH~sN20a ~s / CH20, Etharwl, refl~c for 2 h Mol. Wt: 216.23 C4H~N03 Mol.1M.: 117.10 Materials d MW Wt/V mMol equiv SM 359.42 0.1 g 2.78 1 Morpholine 87.12 0.1 mL
37~ HCHO 0.05 mL
Ethanol 1.5 mL
Procedure:
A solution of SM, morpholine and to formaldehyde in 1.5 mL in ethanol was heated it up to 60° C for overnight.
CAN. J.CHEM.Vo1.61, 411 (1983) O NH
O V HO~N~ ~O
O
HO ~ H CsH~sNzO~
~s / CH20, Ethanol, refl~nc for 2 h Mol. VVt.: 216.23 C~H~N03 Moi. Wt.: 117.10 Materials d MW Wt/V mMol equiv SM 117.10 456 mg 3.9 1 Morpholine 87.12 1 mL
37~ HCHO 0.4 mL
Ethanol 3 Procedure:
A solution of SM, morpholine and formaldehyde in 3 mL in ethanol was heated it up to 60°
C for overnight.

O CNH O
O'~
H HO~~vN
HO~~ CH20, Ethand, 60°C ''~C°9~H'~'sN~
C~H~N03 Md. Wt.: 200.24 Mol. Wt.: 117.10 Materials d MW Wt/V mMol equiv s SM 117 443 mg 3.78 1 pyrrolidine 1 mL
37$ HCHO 0.4 mL
Ethanol 3 mL
Procedure:
A solution of SM, morpholine and formaldehyde in 3 mL in ethanol was heated it up to 60°
C for overnight.
CsH~oN202 O
C

Md. Wt.: 130.15 5 md% -N
Cul O

O 2 equiv ~--~,,NH~
C iCiP04 ~

O equ F3 -N Isoprope~ C'12H13F3N2~2 NHy Mol.1M.: 274.24 [~ I

C~H~F31 I OH

Md.lM.:272.01 ~N NH
F3 C ~~ ~F

C~8H~8F6N20 1 s Md. VIIt.: 392.34 Materials d MW Wt/V mMol equiv SM 130.15 40 mg 0.31 1 CuI 190.44 3 mg 0.05 2 o K3P04 212 . 12 8 2 5 mg HO ( CHZ ) 20H 1.13 62 . 07 0 .1 2 mL

Isopropanol 1 mL

AA Group: 0.31 mmol AA2 0 , AA21, AA2 2 , AA2 3 , AA2 4 , AA2 5 , AA2 6 , AA2 7 , AA2 8 Procedure:
To a test tube, CuI and K3P04 were added then the tube was back-filled with Nitrogen for 10 mins, and then rest of starting material were added, the reaction mixture was heated up to 65° C for overnight.
Library linkage:
NCO K or E BB compounds J ~OH Et3N,~ . ~N-i ~OR
CsH~sNz04 Mol. V4t.: 216.23 Starting material: 22 mg for each reaction K2, K90, K91, K92, K95, K96, K97, K100, K101, K102, K114, K117, E183, E184, BB3, BB5 Procedure:
0.1 mmol of starting material 0.15 mmol of Et3N
0.1 mmol of K and BB or E compounds.
The reactions were stirred for overnight.
Buchwald Reaction:
Org. Zett., Vol.2,No.8,2000 Pd-Catalyzed Amination of Activated Ary Halides:
O _ C~H4F31 Mol. Wt.: 272.01 O
Ph-~ O-~ I ~ ~ F
N~NH ph~ O~ _ Phi C~aHzoNzOz Pd(OAC)2, Cs2C03 Phi ~/N ~ / CF3 Moi. Wt.: 296.36 ~ntPh°s, 1.4-dloxane C~H~F3N~
Mol. VVt.: 440.46 Materials d MW Wt/V mMol equiv SM 296.36 454 mg 1.6 1 Iodo compound 272 500 mg 1.8 1.2 Pd(OAC)2 224.49 10 mg 0.016 0.01 Cesium carbonate 325.82 730 mg 2.24 1.4 Xantphos 578.63 14 mg 0.024 0.015 1,4-dioxane 3 mL

1o Procedure:
1. A 10 mL Round flask was loaded with oxazolidinone, bromo compound, palladium(II) acetate, Xantphos and Cesium carbonate and flashed by N2 protection for 10 mins.
2. 1,4-dioxane was added and heated up to 70° C for overnight and then cool down to room temperature, diluted with dichloromethane.
Library linkage:
O o K orb ands N N N NJIO
OH Et3N,CH2Cl2, rt. ~OR
C~oHzoNz~
Md. Vlft.: 216.28 Starting material: 22 mg for each reaction K2, K90, K91, K92, K95, K96, K97, K100, K101, K102, K117, E183, BB3 Procedure:
0.2 mmol of starting material 0.15 mmol of Et3N
0.2 mmol of K and BB or E compounds.

The reactions were stirred for overnight.
Alkylation: (By NaH) Pn ~
c ~ o -o --~ ~~ ~ ~N~
Pn-~~NH

Ph S NaH

Mol. Wt.: 359.42 C.lsH2~N03 Mol. Wt.: 389.49 For G16, Materials Wt/V mMol Equiv MW

SM 359.42 50 g 139 1 1o G3 (d 2.28) 141.9 17 mL 278 2 NaH (60~) 24 10.3 g 257 1.8 THF 400 mL

NaH was added into the pre-cooled (by dry i5 ice and acetone) the THF and SM solution, then stirred for two hours. G3 were added after re-cooled the reaction mixture.
Deprotection: (G3) Ph p--~
-C-O~ O

Nr i Ph HO
~/~~/ N
P

h ~t4H23N03 C5H9N03 20 Mol. Vllt.: 373.44 Mol. Wt.: 131.13 TFA 14 mL
H20 4 mL
CH2C12 50 mL
Ether type of Linkages:

O
O-~ ~p M
HO~\~N N --CsH~sN20s Md. Wt.: 216.23 Library Design:
Oxazolidinones: 25 mg (1.17 equiv) for each reaction.
M compounds: M1, M2, M3, M4, M5, M6, M11, M14, M24, M30, M34, M35, M37, M38, M39, M40, M41(1 equiv) DIAD = 0.10 nM in THF (MW 202), 400 mg in 17 mL THF.
1 mL was taken for each reaction.
1o Ph3P-polystyrene 1 mmol/g 100 mg for each compound 0.1 mMol Procedure:
4. The vials were charged with SM and Ph3P-polystrene.
5. A solution of DIAD in THF was added into the reaction mixture.
6. The reactions were stirred for overnight.
CAN. J.CHEM.Vol.6l, 411 (1983) _ o O pzN / ~ NVNH Md~ Wt 3202 3 O n O HO~Nv VN / ~ Np2 NH
HO~
C4H~N03 CH20, Ethanol, refl~nc for 2 H C~SHZpN405 MOL Wt.: 117.10 Md.1111t.: 336.34 Materials d MW Wt/V mMol equiv SM 117.10 500 mg Nitro compound 207.23 884 mg 37~ HCHO 0.6 mL
Ethanol 6 mL

Procedure:
A solution of SM, nitro compound and formaldehyde in 3 mL in ethanol was heated it up to 65°
C for overnight.
p CNH ,,O
HO~~N~N
HO ~ H o s CH20, Ethanol, 60 C
CsH~sN~a C4H~N03 Mol. Wt.: 200.24 Mol.1M.: 117.10 Materials d MW Wt/V mMol equiv 1o SM 117 500 mg pyrrolidine 1 mL~
37~ HCHO 0.6 mL
Ethanol 3 mL
Procedure:
A solution of SM, morpholine, and formaldehyde in 3 mL in ethanol was heated it up to 60°
C for overnight.
2o Alkylation: (By NaH) Q I rBf Q
P //
h C ~~ h O

Ph- H ~ Ph-C
-O \~ N O
I H N

Ph a Ph ~H2t CzsH2sN~
NOa Mol. Mol. Wt.: 399.48 Wt.:
35A.42 For C18, Materials MW Wt/V mMol Equiv SM 359.42 0.5 g 1.39 1 C18 (d1.5) 120 0.2 mL 2.78 2 NaH (60~) 24 100 mg 2.57 1.8 THF 4 mL
NaH was added into the pre-cooled (by dry ice and acetone) the THF and SM solution, then stirred for two hours. G3 were added after re-cooled the reaction mixture.
alkylation: (By NaH) C6H9Br O Mol. Wt: 149.03 Ph Br O
Ph-C-O H -~ Ph--C-O O
Ph ~H Ph ~sH2t NOa czaHzsN~
Mol. VYt.: 359.42 Mol. Wt.: 427.53 For C18, Materials MW Wt/V mMol Equiv SM 359.42 0.5 g 1.39 1 C19 149 414 mg 2.78 2 NaH (60~) 24 100 mg 2.57 1.8 THF 4 mL

NaH was added into the pre-cooled ( by dry ice and acetone) the THF and SM solution, then stirred 2o for two hours. C19 were added after re-cooled the reaction mixture.
Curtius Rearrangement:
OH O
Ph-~~5~ O Ph-~~S~NH
Phi NHNH2 NaN02/H2S04 Ph-~
C~9HZ~N30z 50 ° C for 2 h C~8H2pNy02 Exact Mass: 329.21 Exact Mass: 296.15 Mol. Wt.: 329.44 Moi. V1R.: 296.36 Materials d MW Wt/V mMol Equiv SM 329.44 26 79 1 H2S09 98 9.2 mL 95 1.2 NaN02 69 8.2 g 118 1.5 water 100 mL

Procedure:
1). The hydrazide compound was dissolved in water (75 mL), and concentrated sulfuric acid (9.2 mL) 1o diluted in water (25 mL) and added into the stirred solution. The mixture was cooled in the ice bath and then NaN02 in water (20 mL) was added dropwise.
2). The reaction mixture was stirred at room temperature for overnight. Then was put on the water rota-vap without vacuum to shake for 6 hours at 50° C.
3). The reaction was neutralized by sodium carbonate and extracted with EtOAc three times, brine and dried over Na2S09.
4). The solvents were removed by water 2o rota-vap to afford residues.
5). Column chromatograph to isolate the desired compound.
O
OH O-V
Ph-~~S) O Ph~~S~~NH
PhJ NHNH2 NaN02/H2S04 PhJ
C~9HZ~N30Z 50 ° C for 2 h c~~~N2o2 Exact Mass: 329.21 Exact Mass: 296.15 Mol. Wt.: 329.44 Mol. Vlh.: 296.36 Materials d MW Wt/V mMol Equiv SM 329.44 4.5 g 1 H2S09 98 1.6 mL 1.2 NaN02 . 6 9 1. 4 g 1. 5 water 30 mL
Procedure:
1). The hydrazide compound was dissolved in water (25 mL), and concentrated sulfuric acid (1.6 mL) diluted in water (5 mL) and added into the stirred solution. The mixture was cooled in the ice bath and then NaN02 powder was added directly..
2). The reaction mixture was stirred at room temperature for overnight. Then was put on the water rota-vap without vacuum to shake for 6 hours at 50° C.
3). The reaction was neutralized by sodium carbonate and extracted with EtOAc three times, brine and dried over Na2S09.
4). The solvents were removed by water rota-vap to afford residues.
5). Column chromatographed to isolate the desired compound.
~1O~N ~ ~ Noz WOH)2/THF/MeOH O~N ~ ~ NHZ
J H2 HzN
Ph 300 mg of ZD3-75-2 500 mg of Pd(OH)2 5 mL of THF
5 mL of MeOH
The reaction was stirred at room temperature 10 mins (see new spot and starting material on TLC, 3o new spot is more polar) and 40 mins (see one new spot, which is less polar than starting material, and only one spot shown on TLC).

o o ~IO~NH Pd(OH)21THF/MeOH O~NH

HzN'fR!

C4HaNz~
C~aHzoNzOz Mol.1I11t.: 116.12 Mol. Wt.:
296.36 300 mg of ZD3-75-2 500 mg of Pd(OH)2 5 mL of THF
5 mL of MeOH
The reaction was stirred at room temperature 10 mins (see new spot and starting material on TLC, new spot is more polar) and 40 mins (see one new spot, which is less polar than starting material, and only one spot shown on TLC).
o O ~N~S_ ~ NHz G12G2/ Et3N/ BB3 O N NHz C~sH~sN~04S
H N \ / Mol. Wt.: 360.39 li~~ N-~NH-S-~-NOZ
OzN~-S-N S/
C2zHtsNaOsSz Mol. Wt.: 513.55 To a solution of starting material in THF, Et3N was added then BB3 was added. The reaction was stirred for overnight. Isolated by parp-TLC (3/1=
2o EtOAc/Hexane) Library:
Oxazolidinone derivatives: SC1, H, SG3-N, RG3, RC3 (0.1 nmol CH2C12 solution except H in THF) Nitrobenzene derivatives: BB3, BB7, BBB, BB9 ((0.1 nmol CH2C12 solution except BB7 in THF) Base: triethylamine (0.15 nmol) The reactions were set up in the usual way and kept for overnight. Isolated by parp-TLC (3/1 =
EtOAc/Hexane).

Library New M compounds:

Oxazolidinone derivatives: RG3, RCS, RC3 ( 0 .1 nmol CH2C12 solution) to Nitrobenzene derivatives: M 42, M43, M44, M45, M46, (0.1 nmol) DIAD = 0.10 nM in THF (MW 202), 1 equiv.

Ph3P-polystyrene 1 mmol/g 100 mg for each compound 0.1 mMol Procedure:

7. The vials were charged with SM and Ph3P-polystrene.

8. A solution of DIAD in THF was added into the reaction mixture.

The reactions were stirred for overnight.

The reactions were set up in the usual way and kept for overnight. Isolated by pare-TLC (3/1 =

EtOAc/Hexane).

Alkylation: (By t-BuOK) ~-er o Ph O ~
C O

Ph- Ph-C O N
-O
~~NH ~
~

Ph ~/tS Ph C23H21 N3 CzsHzsN~

Mol.1M.: 359.42 Mol. Vlft.: 399.48 For C18, Materials MW Wt/V mMol Equiv SM 359.42 0.5 g 1.39 1 C18 (d1.5) 120 0.2 mL 2.78 2 t-BuOK 3 mL 2.57 1.8 THF 5 mL

To a solution of SM and THF was added t-BuOK, then C19 was added. The reaction mixture was heated up to 60° C in sealed tube.
Alkylation: (By t-BuOK) C5H98r Mol.1/VL:
O 149.03 O
~

Ph Ph ~ O
Ph-C-O H Ph-C-O

Ph NaH ph cxtHz~N03 ~aHzsN~

Mol. Wt.: 359.42 Mol. Wt.: 427.53 For C19, Materials MW Wt/V mMol Equiv SM 359.42 0.5 g 1.39 1 C19 149 414 mg 2.78 2 t-BuOK 3 mL 2.57 1.8 THF 5 mL

To a solution of SM and THF was added t-BuOK, then C19 was added. The reaction mixture was heated up to 60° C in sealed tube.
Alkylation: (By t-BuOK) p ,_O
Ph, ~Br P ',.',~~~/n N H --~ N W~
Pn-) \~ t_s"otc J
~n CtsHu~'hpz C2t H2sNzpz Mol. Wt: 312.41 Mol. Wt.: 338.44 For C18, Materials MW Wt/V mMol Equiv SM 359.42 0.5 g 1.39 1 C18 (d1.5) 120 0.2 mL 2.78 2 t-BuOK 3 mL 2.57 1.8 THF 5 mL

to To a solution of SM and THF was added t-BuOK, then C19 was added. The reaction mixture was heated up to 60° C in sealed tube.
Library linkage:

NCO K or E BB compounds ~tJ N W~/N~O
~N ~ ~ ~ ~ ~p~.1 Et3N,CH2Cl2, rt. ~ ~ ~/ ~-pR
C15H20N4p5 Mol.1M.: 336.34 Starting material: 22 mg for each reaction K2, K91, K92, K95, K96, K97, K100, K101, K117, E183, 2o E184, BB3, BB7, BB9, BBS, K93, K98, K94, K106, AC2, AC3, ACS, AC7.
Procedure:
0.08 mol of starting material 0.15 mmol of Et3N
0.09 mmol of K and BB, AC or E compounds.
The reactions were stirred for weekend.

Library linkage:

NAC co~ ~N ~ ~ N N'/N~O
N VN~ 11-~~(( ((~~O''nn''~,uu~. Et3NlfHF,CH2Ci2, rt. ~ \.-/ ~.OR
C~sH2oN~~s Mol.1M.: 336.34 Starting material: 14 mg for each reaction.
AC1, AC4, ACB, AC9, AC10, AC11, AC12, MsCl 1o Procedure:
0.04 mmol of starting material 0.15 mmol of Et3N
0.06 mmol of AC compounds.
The reactions were stirred for overnight.
While the present invention is described herein with reference to illustrated embodiments, it should be understood that the invention is not limited hereto. Those having ordinary skill in the art and 2o access to the teachings herein will recognize additional modifications and embodiments within the scope thereof. Therefore, the present invention is limited only by the claims attached herein.

Claims (5)

  1. WE CLAIM:

    A process for producing a library of substituted oxazolidinones which comprises:
    (a) reacting a C-(protected oxymethyl)-oxazolidinone in an anhydrous organic solvent containing a first reagent including a plurality of compounds having different numbers of carbons which are reactive with N in the C-(protected oxymethyl)-oxazolidinone under alkylation or Buchwald conditions in an inert atmosphere to produce a mixture of N-(substituted)-C-(protected oxymethyl)-oxazolidinones (I); and (b) reacting the mixture of (I) produced in step (a) in an aqueous organic solvent with a second reagent which removes the protecting group and replaces it with another group from the second reagent to produce the library of substituted oxazolidinones.

    The process of Claim 1 wherein the second reagent is a reducing agent which removes the protecting group of the N-(substituted)-C-(protected oxymethyl)-oxazolidinone to provide a mixture of N-(substituted)-C-hydroxymethyl-oxazolidinones (II) as the library of substituted oxazolidinones.

    The process of Claim 2 wherein the mixture of (II) is further reacted with a third reagent containing a plurality of compounds reactive with the hydroxymethyl in an anhydrous organic solvent to produce a mixture of N-(substituted)-C-(substituted methyl)-oxazolidinones (III) as the library of substituted oxazolidinones.

    The process of Claim 3 wherein the anhydrous organic solvent further includes pyridine.

    The process of Claim 3 wherein the third reagent produces a mixture of 3-(substituted)-5-(substituted methyl)-2-oxazolidinones.

    The process of Claim 3 wherein the third reagent produces a mixture of 3-(substituted)-4-(substituted methyl)-2-oxazolidinones.

    The process of Claim 1 wherein substituted is selected from the group consisting of acyl, alkyl, aryl, aryl sulfonyl, heteroalkyl, heteroaryl, cycle, heterocycle, thio, and mixtures thereof.

    The process of Claim 1, 2, or 3 wherein the substituted oxazolidinones in the library are separated chromatographically.

    The process of Claim 1, 2, or 3 wherein the substituted oxazolidinones in the library are separated chromatographically and then the separated substituted oxazolidinones are each screened for biological activity.

    The process of Claim 1 wherein the protecting group is a trityl group.

    The process of Claim 1 wherein under the alkylation conditions in step (a) the anhydrous organic solvent further includes an alkali without substantial reducing activity.

    The process of Claim 11 wherein the alkali is an ionic hydride.

    The process of Claim 12 wherein the ionic hydride is sodium hydride.

    The process of Claim 1 wherein under the Buchwald conditions in step (a) the anhydrous organic solvent further includes a palladium catalyst.

    The process of Claim 14 wherein the palladium catalyst is Pd(OAc)2.

    The process of Claim 1 wherein the mixture of N-(substituted)-C-(protected oxymethyl)-oxazolidinones (I) produced in step (a) are purified by extracting the reaction mixture with the organic solvent, drying over a drying agent, and then removing the solvent and the substituted oxazolidinones produced in step (b) are purified by removing the solvent.

    The process of Claim 2 wherein the N-(substituted)-C-hydroxymethyl-oxazolidinones (II) are purified by removing the solvent.

    The process of Claim 3 wherein the N-(substituted)-C-(substituted methyl)-oxazolidinones (III) are purified by extracting the reaction mixture with the organic solvent, drying over a drying agent, and then removing the solvent.

    A process for producing a library of substituted oxazolidinones which comprises:
    (a) reacting a C-(protected oxymethyl)-oxazolidinone in an anhydrous organic solvent containing a first reagent including a plurality of compounds having different numbers of carbons which are reactive with N in the C-(protected oxymethyl)-oxazolidinone under alkylation or Buchwald conditions in an inert atmosphere to produce a mixture of N-(substituted)-C-(protected oxymethyl)-oxazolidinones (I);
    (b) reacting the mixture of (I) produced in step (a) in an aqueous organic solvent with a second reagent which removes the protecting group of the N-(substituted)-C-(protected oxymethyl)-oxazolidinones to produce a mixture of N-(substituted)-C-hydroxymethyl-oxazolidinones (II); and (c) reacting the mixture of (II) produced in step (b) in an anhydrous organic solvent with a third reagent containing a plurality of compounds reactive with the hydroxymethyl of the mixture of (II) to produce a mixture of N-(substituted)-C-(substituted methyl)-oxazolidinones (III) as the library of substituted oxazolidinones.

    The process of Claim 19 wherein the anhydrous organic solvent in step (c) further includes pyridine.

    The process of Claim 19 wherein the third reagent produces a mixture of 3-(substituted)-5-(substituted methyl)-2-oxazolidinones.

    The process of Claim 19 wherein the third reagent produces a mixture of 3-(substituted)-4-(substituted methyl)-2-oxazolidinones.

    The process of Claim 19 wherein substituted is selected from the group consisting of acyl, alkyl, aryl, aryl sulfonyl, heteroalkyl, heteroaryl, cycle, heterocycle, thio, and mixtures thereof.

    The process of Claim 19, 21, or 22 wherein the substituted oxazolidinones in the library are separated chromatographically.

    The process of Claim 19 wherein the protecting group is a trityl group.

    The process of Claim 19 wherein under the alkylation conditions in step (a) the anhydrous organic solvent further includes an alkali without substantial reducing activity.

    The process of Claim 26 wherein the alkali is an ionic hydride.

    The process of Claim 27 wherein the ionic hydride is sodium hydride.

    The process of Claim 19 wherein under the Buchwald conditions in step (a) the anhydrous organic solvent further includes a palladium catalyst.

    The process of Claim 29 wherein the palladium catalyst is Pd(OAc)2.

    The process of Claim 19 wherein the mixture of N-(substituted)-C-(protected oxymethyl)-oxazolidinones (I) produced in step (a) are purified by extracting the reaction mixture with the organic solvent, drying over a drying agent, and then removing the solvent.

    The process of Claim 19 wherein the N-(substituted)-C-hydroxymethyl-oxazolidinones (II) produced in step (b) are purified by removing the solvent.

    The process of Claim 19 wherein the N-(substituted)-C-(substituted methyl)-oxazolidinones (III) produced in step (c) are purified by extracting the reaction mixture with the organic solvent, drying over a drying agent, and then removing the solvent.

    A process for preparing a library of substituted oxazolidinones which comprises:
    reacting a C-hydroxymethyl-oxazolidinone in an anhydrous organic solvent including pyridine with a reagent containing a plurality of compounds reactive with the hydroxy group to produce a mixture of substituted oxazolidinones as the library of substituted oxazolidinones.

    The process of Claim 34 wherein the reaction produces a mixture of 5-(substituted methyl)-2-oxazolidinones.

    The process of Claim 34 wherein the reaction produces a mixture of 4-(substituted methyl)-2-oxazolidinones.

    The process of Claim 34 wherein the reaction produces a mixture of N-(substituted)-C-(hydroxymethyl)-2-oxazolidinones.

    The process of Claim 34 wherein the reaction produces a mixture of N-(substituted)-C-(substituted methyl)-2-oxazolidinones.

    The process of Claim 34 wherein substituted is selected from the group consisting of acyl, alkyl, aryl, aryl sulfonyl, heteroalkyl, heteroaryl, cycle, heterocycle, thio, and mixtures thereof.

    The process of Claim 34, 35, 36, 37, or 38 wherein the substituted oxazolidinones in the library are separated chromatographically.

    A library of substituted oxazolidinones selected from the group consisting of N-(substituted)-C-(substituted methyl)-oxazolidinones, N-(substituted)-C-hydroxymethyl-oxazolidinones, and C-(substituted methyl)-oxazolidinones.

    The library of Claim 41 wherein substituted in N-(substituted) includes at least 10 different individual groups.

    The library of Claim 41 or 42 wherein substituted in C-(substituted) includes at least 10 different individual groups.

    The library of Claim 41 wherein the library is a mixture of N-(substituted)-C-hydroxymethyl-2-oxazolidinones.

    The library of Claim 41 wherein the library is a mixture selected from the group consisting of 3-(substituted)-5-(substituted methyl)-2-oxazolidinones and 3-(substituted)-4-(substituted methyl)-2-oxazolidinones.

    The library of Claim 41 wherein substituted is selected from the group consisting of acyl, alkyl, aryl, aryl sulfonyl, heteroalkyl, heteroaryl, cycle, heterocycle, thio, and mixtures thereof.

    A method of screening substituted oxazolidinones for biological activity which comprises:
    (a) providing a library of the substituted oxazolidinones wherein the substituted oxazolidinones are selected from the group consisting of N-(substituted)-C-(substituted methyl)-oxazolidinones, N-(substituted)-C-hydroxymethyl-oxazolidinones, and C-(substituted methyl)-oxazolidinones:
    (b) chromatographically separating the substituted oxazolidinones in the library and (c) testing the separated substituted oxazolidinones for the biological activity.

    The method of Claim 47 wherein substituted in N-(substituted) includes at least 10 different individual groups.

    The method of Claim 47 or 48 wherein substituted in C-(substituted methyl) includes at least 10 different individual groups.

    The method of Claim 47 wherein the substituted oxazolidinones is a mixture of N-(substituted)-C-hydroxymethyl-2-oxazolidinones.

    The method of Claim 47 wherein the substituted oxazolidinones are a mixture selected from the group consisting of 3-(substituted)-5-(substituted methyl)-2-oxazolidinones, 3-(substituted)-4-(substituted methyl)-2-oxazolidinones, 5-(substituted methyl)-2-oxazolidinones, and 4-(substituted methyl)-
  2. 2-oxazolidinones.

    The method of Claim 47 wherein substituted is selected from the group consisting of acyl, alkyl, aryl, aryl sulfonyl, heteroalkyl, heteroaryl, cycle, heterocycle, thio, and mixtures thereof.

    A substituted oxazolidinone with biological activity obtained by the method of Claim 47.

    A process for producing a substituted oxazolidinone which comprises:
    (a) reacting a C-(protected oxymethyl)-oxazolidinone in an anhydrous organic solvent containing a first reagent including a compound which is reactive with N in the C-(protected oxymethyl)-oxazolidinone under alkylation or Buchwald conditions in an inert atmosphere to produce an N-(substituted)-C-(protected oxymethyl)-oxazolidinone;
    (b) reacting the N-(substituted)-C-(protected oxymethyl)-oxazolidinone in an aqueous organic solvent with a second reagent with a second reagent which replaces the protecting group of the N-(substituted)-C-(protected oxymethyl)-oxazolidinone with a hydrogen to produce an N-(substituted)-C-hydroxymethyl-oxazolidinone; and (c) reacting the N-(substituted)-C-hydroxymethyl-oxazolidinone in an anhydrous organic solvent with a third reagent containing a compound reactive with the hydroxy group to produce N-(substituted)-C-(substituted methyl)-oxazolidinones as the substituted oxazolidinone.

    The process of Claim 54 wherein the anhydrous organic solvent in step (c) further includes pyridine.

    The process of Claim 54 wherein substituted is selected from the group consisting of aryl, alkyl, aryl, aryl sulfonyl, heteroalkyl, heteroaryl, cycle, heterocycle, thio, and mixtures thereof.

    The process of Claim 54 wherein the protecting group is a trityl group.

    The process of Claim 54 wherein the substituted oxazolidinone has the formula wherein R1 is selected from the group consisting of hydrogen, aryl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, phenacyl, aryl sulfonyl, thio, and mixture thereof, or a hydrogen; R2 is selected from the group consisting of acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, aryl sulfonyl, phenacyl, thio, and mixture thereof, or a hydrogen, wherein hetero is an atom selected from the group consisting of O, N, P, and S; and y is a heteroatom selected from the group consisting of O, N, and S.

    The process of Claim 54 wherein the substituted oxazolidinone has the formula wherein R1 is selected from the group consisting of alkyl sulfonyl, aryl sulfonyl, alkyl, acyl, aryl, and thio and R2 is selected from the group consisting of alkyl, acyl, aryl, and thio.

    The process of Claim 54 wherein the substituted oxazolidinone has the formula wherein R1 is selected from the group consisting of alkyl sulfonyl, aryl sulfonyl, alkyl, aryl, aryl, and thio and R2 is selected from the group consisting of alkyl, aryl, aryl, and thio.

    The process of Claim 54 wherein the substituted oxazolidinone has the formula wherein R1 is selected from the group consisting of alkyl, acyl, thio, and aryl, R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from the group consisting of F, NO2, Cl, alkyl, and aryl.

    The process of Claim 54 wherein the substituted oxazolidinone has the formula wherein R1 is selected from the group consisting of alkyl, acyl, thio, and aryl, R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from the group consisting of F, NO2, Cl, alkyl, and aryl.

    The process of Claim 54 wherein the substituted oxazolidinone has the formula wherein R1 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, R2 is selected from the group consisting of alkyl, aryl, acyl, thio, and heterocycle, and X is selected from the group consisting of F, NO2, Cl, alkyl, and aryl.

    The process of Claim 54 wherein the substituted oxazolidinone has the formula wherein R1 is selected from the group consisting of alkyl, aryl, acyl, thio, or heterocycle, R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from the group consisting of F, NO2, Cl, alkyl, and aryl.

    The process of Claim 54 wherein the substituted oxazolidinone has the formula wherein R1 is selected from the group consisting of alkyl, aryl, acyl, thio, and heterocycle and R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers.

    The process of Claim 54 wherein the substituted oxazolidinone has the formula wherein R1 is selected from the group consisting of alkyl, aryl, acyl, thio, and heterocycle and R2 is selected from the group consisting of C-3, C-4, with C-5 chiral synthons with 1, 2, or 3 chiral centers.

    The process of Claim 54 wherein under the alkylation conditions in step (a) the anhydrous organic solvent further includes an alkali without substantial reducing activity.

    The process of Claim 67 wherein the alkali is an ionic hydride.

    The process of Claim 68 wherein the ionic hydride is sodium hydride.

    The process of Claim 54 wherein under the Buchwald conditions in step (a) the anhydrous organic solvent further includes a palladium catalyst.

    The process of Claim 70 wherein the palladium catalyst is Pd(OAc)2.

    The process of Claim 54 wherein the mixture of N-(substituted)-C-(protected oxymethyl)-oxazolidinone produced in step (a) is purified by extracting the reaction mixture with the organic solvent, drying over a drying agent, and then removing the solvent.

    The process of Claim 54 wherein the N-(substituted)-C-hydroxymethyl-oxazolidinone produced in step (b) is purified by removing the solvent.

    The process of Claim 54 wherein the N-(substituted)-C-(substituted methyl)-oxazolidinone produced in step (c) is purified by extracting the reaction mixture with the organic solvent, drying over a drying agent, and then removing the solvent.

    A substituted oxazolidinone which has the formula wherein R1 is selected from the group consisting of hydrogen, acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, phenacyl, aryl sulfonyl, thio, and mixture thereof, or a hydrogen R2 is selected from the group consisting of acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, aryl sulfonyl, phenacyl, thio, and mixture thereof, or a hydrogen, wherein hetero is an atom selected from the group consisting of O, N, P, and S; and y is a heteroatom selected from the group consisting of O, N, and S.

    A substituted oxazolidinone which has the formula wherein R1 is selected from the group consisting of alkyl sulfonyl, aryl sulfonyl, alkyl, acyl, aryl, and thio and R2 is selected from the group consisting of alkyl, acyl, aryl, and thio.

    A substituted oxazolidinone which has the formula wherein R1 is selected from the group consisting of alkyl sulfonyl, aryl sulfonyl, alkyl, acyl, aryl, and thio and R2 is selected from the group consisting of alkyl, aryl, aryl, and thio.

    A substituted oxazolidinone which has the formula wherein R1 is selected from the group consisting of alkyl, acyl, thio, and aryl, R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from the group consisting of F, NO2, Cl, alkyl, and aryl.

    A substituted oxazolidinone which has the formula wherein R1 is selected from the group consisting of alkyl, acyl, thio, and aryl, R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from the group consisting of F, NO2, Cl, alkyl, and aryl.

    A substituted oxazolidinone which has the formula wherein R1 is selected from the group consisting of C-
  3. 3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, R2 is selected from the group consisting of alkyl, aryl, acyl, thio, and heterocycle, and X is selected from the group consisting of F, NO2, Cl, alkyl, and aryl.
  4. A substituted oxazolidinone which has the formula wherein R1 is selected from the group consisting of alkyl, aryl, acyl, thio, or heterocycle, R2 is selected from the group consisting of C-3, C-4, and C-5 chiral synthons with 1, 2, or 3 chiral centers, and X is selected from the group consisting of F, NO2, Cl, alkyl, and aryl.

    A substituted oxazolidinone which has the formula wherein R1 is selected from the group consisting of alkyl, aryl, acyl, thio, and heterocycle and R2 is selected from the group consisting of C-3, C-4, and C-
  5. 5 chiral synthons with 1, 2, or 3 chiral centers.

    A substituted oxazolidinone which has the formula wherein R1 is selected from the group consisting of alkyl, aryl, acyl, thio, or heterocycle and R2 is selected from the group consisting of C-3, C-4, with C-5 chiral synthons with 1, 2, or 3 chiral centers.

    An antimicrobial composition comprising a carrier and one or more substituted oxazolidinones of the formula wherein R1 is selected from the group consisting of hydrogen, acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, phenacyl, aryl sulfonyl, thio, and mixture thereof, or a hydrogen; R2 is selected from the group consisting of acyl, alkyl, aryl, heteroalkyl, heteroaryl, heterocycle, aryl sulfonyl, phenacyl, thio, and mixture thereof, or a hydrogen, wherein hetero is an atom selected from the group consisting of O, N, P, and S; and y is a heteroatom selected from the group consisting of O, N, and S.
CA002464109A 2001-10-18 2002-10-17 Process for the preparation of oxazolidinones and method of use thereof Abandoned CA2464109A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US33026801P 2001-10-18 2001-10-18
US33026601P 2001-10-18 2001-10-18
US60/330,266 2001-10-18
US60/330,268 2001-10-18
PCT/US2002/033181 WO2003106413A2 (en) 2001-10-18 2002-10-17 Process for the preparation of oxazolidinones and method of use thereof

Publications (1)

Publication Number Publication Date
CA2464109A1 true CA2464109A1 (en) 2003-12-24

Family

ID=29739345

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002464109A Abandoned CA2464109A1 (en) 2001-10-18 2002-10-17 Process for the preparation of oxazolidinones and method of use thereof

Country Status (6)

Country Link
US (2) US7390825B1 (en)
EP (1) EP1578723A4 (en)
JP (1) JP2005519996A (en)
AU (1) AU2002367959A1 (en)
CA (1) CA2464109A1 (en)
WO (1) WO2003106413A2 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1937286B1 (en) 2005-09-12 2016-03-09 Abela Pharmaceuticals, Inc. Compositions comprising dimethyl sulfoxide (dmso)
US8480797B2 (en) 2005-09-12 2013-07-09 Abela Pharmaceuticals, Inc. Activated carbon systems for facilitating use of dimethyl sulfoxide (DMSO) by removal of same, related compounds, or associated odors
JP5399072B2 (en) 2005-09-12 2014-01-29 アベラ ファーマスーティカルズ インコーポレイテッド System for removing dimethyl sulfoxide (DMSO) or related compounds or odors associated therewith
US8435224B2 (en) 2005-09-12 2013-05-07 Abela Pharmaceuticals, Inc. Materials for facilitating administration of dimethyl sulfoxide (DMSO) and related compounds
US7700590B2 (en) 2006-02-09 2010-04-20 University Of New Orleans Research And Technology Foundation, Inc. Antibacterial agents
JP5492769B2 (en) * 2007-06-26 2014-05-14 サノフイ Regioselective metal-catalyzed synthesis of fused benzimidazoles and azabenzimidazoles
US20110091903A1 (en) * 2007-11-20 2011-04-21 Bommarito G Marco Method of analyzing a sample for a bacterium using diacetylene-containing polymer sensor
BRPI0921494A2 (en) 2008-11-03 2018-10-30 Prad Reasearch And Development Ltd method of planning a underground forming sampling operation, method of controlling a underground forming sampling operation, method of controlling a drilling operation for an underground formation, and method of sampling during the drilling operation.
AU2010313253B2 (en) 2009-10-30 2015-02-19 Abela Pharmaceuticals, Inc. Dimethyl sulfoxide (DMSO) and methylsulfonylmethane (MSM) formulations to treat osteoarthritis
CN115304500A (en) * 2021-05-06 2022-11-08 北京康派森医药科技有限公司 Method for synthesizing metoprolol impurity

Family Cites Families (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL133317C (en) * 1966-06-24 1900-01-01
CH635573A5 (en) * 1977-03-24 1983-04-15 Sandoz Ag Process for preparing novel 1,2-dihydroxypropane derivatives
US4340606A (en) * 1980-10-23 1982-07-20 E. I. Du Pont De Nemours And Company 3-(p-Alkylsulfonylphenyl)oxazolidinone derivatives as antibacterial agents
US4473501A (en) * 1981-05-04 1984-09-25 G. D. Searle & Co. Dihydro azino isoquinolines
EP0101076B1 (en) * 1982-08-13 1989-01-04 Kanegafuchi Kagaku Kogyo Kabushiki Kaisha Process for production of optically active oxazolidinone derivative
DE3317194A1 (en) * 1983-05-11 1984-11-15 Henkel KGaA, 4000 Düsseldorf POLYURETHANE PREPOLYMERS BASED ON OLEOCHEMICAL POLYOLS, THEIR PRODUCTION AND USE
US4518772A (en) * 1983-06-23 1985-05-21 The Proctor & Gamble Company Synthesis of higher polyol fatty acid polyesters using high soap:polyol ratios
US4772597A (en) * 1983-10-14 1988-09-20 Pfizer Inc. 2-azacycloalkylthiopenem derivatives
CA1260948A (en) * 1984-12-05 1989-09-26 E. I. Du Pont De Nemours And Company Aminomethyl oxooxazolidinyl benzene derivatives useful as antibacterial agents
NL8601904A (en) * 1986-07-23 1988-02-16 Unilever Nv PROCESS FOR THE PREPARATION OF POLYOL FATTY ACID POLYESTERS.
US4812533A (en) * 1987-03-25 1989-03-14 Caschem, Inc. Hydroxy acid esterified polyols
US5288514A (en) * 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US5721099A (en) * 1992-10-01 1998-02-24 Trustees Of Columbia University In The City Of New York Complex combinatorial chemical libraries encoded with tags
DE4400465A1 (en) * 1993-07-12 1995-01-19 Bayer Ag New mixtures and their use as binders for the production of composite materials
US5596085A (en) * 1995-04-11 1997-01-21 Kraft Foods, Inc. Method for preparing polyol fatty acid polyesters by transesterification
KR100530297B1 (en) * 1996-04-11 2005-11-22 파마시아 앤드 업존 캄파니 엘엘씨 Process to prepare oxazolidinones
US6617114B1 (en) * 1996-10-31 2003-09-09 Karo Bio Ab Identification of drug complementary combinatorial libraries
CA2318969A1 (en) * 1998-01-23 1999-07-29 Mikhail F. Gordeev Oxazolidinone combinatorial libraries, compositions and methods of preparation
US20020058774A1 (en) * 2000-09-06 2002-05-16 Kurth Thomas M. Transesterified polyol having selectable and increased functionality and urethane material products formed using the polyol
US6107433A (en) * 1998-11-06 2000-08-22 Pittsburg State University Process for the preparation of vegetable oil-based polyols and electroninsulating casting compounds created from vegetable oil-based polyols
WO2000044717A2 (en) * 1999-01-27 2000-08-03 Kosan Biosciences, Inc. Synthesis of oligoketides
US6407272B1 (en) * 1999-07-14 2002-06-18 Arizona Chemical Company Secondary alcohol esters of hydroxyacids and uses thereof
DE19962924A1 (en) 1999-12-24 2001-07-05 Bayer Ag Substituted oxazolidinones and their use
US6288238B1 (en) * 2000-09-19 2001-09-11 Board Of Trustees Operating Michigan State University Process for the preparation of 5-hydroxymethyl 2-oxazolidinone and novel intermediate
US6288239B1 (en) * 2000-09-19 2001-09-11 Board Of Trustees Operating Michigan State University 5-trityloxymethyl-oxazolidinones and process for the preparation thereof

Also Published As

Publication number Publication date
JP2005519996A (en) 2005-07-07
AU2002367959A1 (en) 2003-12-31
WO2003106413A2 (en) 2003-12-24
EP1578723A2 (en) 2005-09-28
US20080146458A1 (en) 2008-06-19
US7390825B1 (en) 2008-06-24
US20070265451A1 (en) 2007-11-15
EP1578723A4 (en) 2006-11-02
WO2003106413A3 (en) 2005-10-20
AU2002367959A8 (en) 2003-12-31

Similar Documents

Publication Publication Date Title
US20070265451A1 (en) Process for the preparation of oxazolidinones and method of use thereof
US6743811B2 (en) Oxazalidinone compounds and methods of preparation and use thereof
Wipf et al. From aziridines to oxazolines and thiazolines: The heterocyclic route to thiangazole
US7691889B2 (en) Antimycobacterial compounds
US6083967A (en) S-oxide and S,S-dioxide tetrahydrothiopyran phenyloxazolidinones
JP2012506908A (en) Synthetic mimics of host defense and their use
Singh et al. New Antibacterial tetrahydro-4 (2H)-thiopyran and thiomorpholine S-oxide and S, S-dioxide phenyloxazolidinones
US6660832B1 (en) Macrocyclic compounds and preparation methods thereof
JP2009503021A (en) Quinoline derivatives as antibacterial agents
Bharath et al. RETRACTED ARTICLE: Design, synthesis of novel oxazolidino-amides/sulfonamides conjugates and their impact on antibacterial activity
US7332611B2 (en) N-thiolated 2 oxazolidone antibiotics
WO2003011859A2 (en) Oxazolidinone derivatives as antibacterial agents
Nadaf et al. Design, synthesis and biological evaluation of novel N, 4-diphenylthiazol-2-amine derivatives
DE60013147T2 (en) CASPASE INHIBITORS
Aaramadaka et al. Synthesis and evaluation of urea and thiourea derivatives of oxazolidinones as antibacterial agents
WO2001021599A1 (en) Caspase inhibitor
Wu et al. Metal-free multicomponent cascade reactions of homopropargylic amines and acyl chlorides as well as potassium thiocyanate and diiodine: an access to thiazine imides
NO328933B1 (en) Peptide deformylaseinhibitorer
KR20110036603A (en) 3-(n-heterocyclyl)-pyrrolidinyl-phenyl-oxazolidinones as antibacterial agents
Ozdemır et al. Investigation of Biological Properties of New 1-(2, 6-Dimetilmorfolin-4-ylmetil)-3-alkyl (aryl)-4-[3-ethoxy-(4-benzenesulfonyloxy)-benzylidenamino]-4, 5-dihydro-1H-1, 2, 4-triazol-5-ones
WO2024069378A1 (en) Novel compounds for the treatment of mammalian infections
CA2453182A1 (en) Crystalline thiazine oxazolidinones
Xu Preparation of thioalkyl-substituted small ring nitrogen heterocycles and 5-disubstituted-ethyl-2-oxazolidinones for antibacterial and antiviral candidacy
CZ20001975A3 (en) S-oxide and S,S-dioxide tetrahydropyran phenyloxazolidinones
JP2002363165A (en) Antibacterial agent having pyrazole skeleton

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued