NEW ISOTHIAZOLOQUINOLONES AND RELATED COMPOUNDS AS ANTI-INFECTIVE AGENTS

The invention provides certain compounds and salts of Formula I and Formula II: which possess antimicrobial activity. The invention also provides novel synthetic intermediates useful in making compounds of Formula I and Formula II. The variables A1, R2, R3, R5, R6, R7, A8 and R9 are defined herein. Certain compounds of Formula I and Formula II disclosed herein are potent and selective inhibitors of bacterial DNA synthesis and bacterial replication. The invention also provides antimicrobial compositions, including pharmaceutical compositions, containing one or more compounds of Formula I or Formula II and one or more carriers, excipients, or diluents. Such compositions may contain a compound of Formula I or Formula II as the only active agent or may contain a combination of a compound of Formula I or Formula II and one or more other active agents. The invention also provides methods for treating microbial infections in animals.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS REFERENCE TO RELATED APPLICATION

This application claims priority from U.S. Provisional Application No. 60/653,434 filed Feb. 16, 2005, which is hereby incorporated by reference in its entirety.

FIELD OF THE INVENTION

The present invention provides isothiazolo[5,4-b]quinolones and related compounds, which possess antimicrobial activity. Certain compounds provided herein possess potent antibacterial, antiprotozoal, or antifungal activity. Particular compounds provided herein are also potent and/or selective inhibitors of prokaryotic DNA synthesis and prokaryotic reproduction. The invention provides anti-microbial compositions, including pharmaceutical compositions, containing one or more carrier, diluents, or excipients. The invention provides pharmaceutical compositions containing an isothiazolo[5,4-b]quinoline or related compound as the only active agent or containing an isothiazolo[5,4-b]quinoline or related compound in combination with one or more other active agent, such as one or more other antimicrobial or antifungal agent. The invention provides methods for treating or preventing microbial infections in eukaryotes, preferably animals, by administering an effective amount of a isothiazolo[5,4-b]quinoline or related compound to a eukaryote suffering from or susceptible to microbial infection. The invention also provides methods of inhibiting microbial growth and survival by applying an effective amount of a isothiazolo[5,4-b]quinoline or related compound.

The invention also provides novel intermediates useful for the for the synthesis of isothiazolo[5,4-b]quinolones and related compounds. The invention also provides methods of synthesis for isothiazolo[5,4-b]quinolones and related compounds.

BACKGROUND OF THE INVENTION

Antimicrobial compounds are compounds capable of destroying or suppressing the growth or reproduction of microorganisms, such as bacteria, protozoa, mycoplasma, yeast, and fungi. The mechanisms by which antimicrobial compounds act vary. However, they are generally believed to function in one or more of the following ways: by inhibiting cell wall synthesis or repair; by altering cell wall permeability; by inhibiting protein synthesis; or by inhibiting synthesis of nucleic acids. For example, beta-lactam antibacterials inhibit the essential penicillin binding proteins (PBPs) in bacteria, which are responsible for cell wall synthesis. Quinolones act, at least in part, by inhibiting synthesis of DNA, thus preventing the cell from replicating.

Many attempts to produce improved antimicrobials yield equivocal results. Indeed, few antimicrobials are produced that are truly clinically acceptable in terms of their spectrum of antimicrobial activity, avoidance of microbial resistance, and pharmacology. Thus there is a continuing need for broad-spectrum antimicrobials, and a particular need for antimicrobials effective against resistant microbes.

Pathogenic bacteria are known to acquire resistance via several distinct mechanisms including inactivation of the antibiotic by bacterial enzymes (e.g., beta-lactamases that hydrolyze penicillin and cephalosporins); removal of the antibiotic using efflux pumps; modification of the target of the antibiotic via mutation and genetic recombination (e.g., penicillin-resistance in Neiserria gonorrhea); and acquisition of a readily transferable gene from an external source to create a resistant target (e.g., methicillin-resistance in Staphylococcus aureus). There are certain Gram-positive pathogens, such as vancomycin-resistant Enterococcus faecium, which are resistant to virtually all commercially available antibiotics.

Resistant organisms of particular note include methicillin-resistant and vancomycin- resistant Staphylococcus aureus, penicillin-resistant Streptococcus pneumoniae, vancomycin-resistant enterococci, fluoroquinolone-resistant E. coli, cephalosporin-resistant aerobic gram-negative rods and imipenem- resistant Pseudomonas aeruginosa. These organisms are significant causes of nosocomial infections and are clearly associated with increasing morbidity and mortality. The increasing numbers of elderly and immunocompromised patients are particularly at risk for infection with these pathogens. Therefore, there is a large unmet medical need for the development of new antimicrobial agents.

SUMMARY OF THE INVENTION

The invention provides compounds of Formula I and Formula II (shown below) and includes isothiazolo[5,4-b]quinolines and related compounds, which possess antimicrobial activity. The invention provides compounds of Formula I and Formula II that possess potent and/or selective antibacterial, antiprotozoal, or antifungal activity. The invention also provides anti-bacterial compositions containing one or more compounds of Formula I or Formula II, or a salt, solvate, or acylated prodrug of such a compound, and one or more carriers, excipients, or diluents.

The invention further comprises methods of treating and preventing microbial infections, particularly bacterial and protozoal infections by administering and effective amount of a compound of Formula I or Formula II to a eukaryote suffering from or susceptible to a microbial infection. These microbial infections include bacterial infections, for example E. coli infections, Staphylococcus infections, Salmonella infections and protozoal infections, for example Chlamydia infections. The invention is particularly includes methods of preventing or treating microbial infections in mammals, including humans, but also encompasses methods of preventing or treating microbial infections in other animals, including fish, birds, reptiles, and amphibians.

Methods of treatment include administering a compound of Formula I or Formula II alone as the single active agent or administering a compound of Formula I in combination with one or more other therapeutic agent, such as an antibacterial, an antifungal, an antiviral, an interferon, an efflux-pump inhibitor, a beta-lactamase inhibitor, or another compound of Formula I or Formula II.

The invention also provides methods of inhibiting microbial growth and survival by applying an effective amount of an isothiazolo[5,4-b]quinoline or related compound. The invention includes, for example, methods of inhibiting microbial growth and survival on medical instruments or on surfaces used for food preparation by applying a composition containing a compound of Formula I or Formula II.

Thus, the invention include compounds of Formula I and Formula II

A compound of Formula I

and the pharmaceutically acceptable salts thereof, wherein:

A1 is S, O, SO, or SO2.

R2 is hydrogen.

Or, R2 is C1-C8alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C7cycloalkyl(C0-C4carbohydryl), C4-C7cycloalkenyl(C0-C4carbohydryl), aryl(C0-C4carbohydryl), C2-C6heterocycloalkyl(C0-C4carbohydryl) each of which is substituted with 0 to 5 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, C1-C4alkyl, C1-C4alkoxy, C1-C2haloalkyl, C1-C2haloalkoxy, mono- and di-C1-C4alkylamino, C2-C4alkanoyl, C1-C4alkylthio, ═NOR10, ═NR10, —O(C═O)R10, —(C═O)NR10R11, —O(C═O)NR10R11, —(C═O)OR10, —(C═O)NR10OR11, —NR10(C═O)R11, —NR10(C═O)OR11, —NR10(C═O)NR11R12, —NR10(C═S)NR11R12, —NR10NR11R12, —SO3R10, —(S═O)OR10, —SO2R13, —SO2NR10R11, and —NR10SO2R13; where R10, R11, and R12 are independently hydrogen, C1-C4alkyl, or aryl, and R13 is C1-C4alkyl or aryl.

R3 is hydrogen, C1-C6alkyl, C1-C6alkanoyl, mono- or di-C1-C6alkylcarbamate, or C1-C6alkylsulfonate; each of which is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, C1-C4alkoxy, mono- and di-C1-C4alkylamino, C1-C2haloalkyl, and C1-C2haloalkoxy.

R5 is hydrogen, halogen, hydroxy, amino, cyano, nitro, or —NHNH2.

Or, R5 is C1-C4alkyl, C1-C4alkoxy, mono- or di-(C1-C4)alkylamino, mono-, di- or tri-C1-C4 alkylhydrazinyl, C2-C4alkanoyl, C1-C4alkylester, C1-C2haloalkyl, or C1-C2haloalkoxy; each of which is substituted with 0 to 3 substituents independently chosen from hydroxy, amino, halogen, oxo, C1-C4alkoxy, C1-C2haloalkyl, C1-C2haloalkoxy, and mono- and di-C1-C4alkylamino.

R6 is hydrogen, halogen, hydroxy, amino, cyano, C1-C4alkyl, C1-C4alkoxy, mono- or di-(C1-C4)alkylamino, —SO3R10, —SO2R10, or —SO2NR10R11; where R10 and R11 carry the definitions set forth above.

R7 is bromo, iodo, —O(SO2)CF3, or —N2BF4, or R7 is XRA.

Where, X is absent, —CH2—CH2—, —CH═CH—, —(C═O)—, —(C═O)NH—, or —C≡C—.

RA is C3-C6alkyl, C4-C7cycloalkyl, C4-C7cycloalkenyl, a 7-10 membered bicyclic saturated, partially unsaturated, or aromatic carbocyclic group, a 5-6 membered saturated, partially unsaturated, or aromatic heterocylic group bound via a carbon atom when X is absent or —CH2—CH2—, or bound via a carbon or nitrogen atom when X is —CH═CH— or —C≡C— or a RA is a 7-10 membered bicyclic saturated, partially unsaturated, or aromatic heterocylic group bound via a carbon atom when X is absent or —CH2—CH2—, or bound via a carbon or nitrogen atom when X is —CH═CH— or —C≡C—; each of which RA is substituted with 0 to 5 substituents independently chosen from (i), (ii), and (iii).

Or, R7 is XRB, where RB is phenyl substituted with 1 to 5 substituents independently chosen from (i), (ii), and (iii).

Or, R7 is XRC, where RC is cyclopropyl with 0 to 5 substituents independently chosen from (i), (ii), and (iii), with the proviso that RC is not substituted with amino, or mono- or di-(C1-C4)alkylamino.

Or, R7 is XRD where RD is phenyl fused to a 5- or 6-membered heterocycloalkyl ring containing 1 or 2 nitrogen or oxygen atoms, where RD is substituted with 0 to 3 substitutents chosen from (i), (ii), and (iii).

Where,

    • (i) is chosen from halogen, hydroxy, amino, cyano, and nitro,
    • (ii) is chosen from C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C1-C6alkoxy(C0-C4alkyl), mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C3-C7cycloalkyl(C0-C4carbohydryl), C3-C7cycloalkyl(C0-C4carbohydryl-O—), C4-C7cycloalkenyl(C0-C4carbohydryl), aryl(C0-C6carbohydryl), aryl(C1-C4alkoxy), C2-C6heterocycloalkyl(C0-C4carbohydryl), heteroaryl(C0-C6carbohydryl), C1-C6alkylthio, ═NOR10, ═NR10, —(C0-C4alkyl)(C═O)R10, —(C0-C4alkyl)O(C═O)R10, —(C0-C4alkyl)(C═O)NR10R11, —(C0-C4alkyl)O(C═O)NR10R11, —(C0-C4alkyl)(C═O)OR10, —(C0-C4alkyl)NR10(C═O)R11, —(C0-C4alkyl)NR10(C═O)OR11, —(C0-C4alkyl)NR10(C═O)NR11R12, —(C0-C4alkyl)NR10(C═O)(C1-C4alkyl)NR11(C═O)O—R12, —(C0-C4alkyl)NR10(C═S)NR11R12, —(C0-C4alkyl)NR10NR11R12, —(C0-C4alkyl)N═NR13, —(C0-C4alkyl)SO3R10, —(C0-C4alkyl)(S═O)OR10, —(C0-C4alkyl)SO2R13, —(C0-C4alkyl)SO2NR10R11, and —(C0-C4alkyl)NR10SO2R13; and
    • (iii) is chosen from —ORD, —(C═O)RD, —SO2RD, —SO3RD, —NR10SO2RD, where RD is C1-C4alkyl, C3-C1cycloalkyl(C0-C2alkyl), C2-C6heterocycloalkyl(C0-C2alkyl), aryl(C0-C2alkyl), or heteroaryl(C0-C2alkyl).

Where each of (ii) and (iii) is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, oxo, —COOH, —CONH2, C1-C4alkyl, C2-C4alkenyl, C2-C4alkynyl, C1-C4alkoxy, C1-C4alkoxycarbonyl, C3-C7cycloalkyl(C0-C4carbohydryl), C3-C7cycloalkyl(C0-C4alkoxy), mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C2-C4alkanoyl and phenyl.

A8 is nitrogen or CR8.

Wherein, R8 is hydrogen, halogen, hydroxy, amino, cyano, nitro, or —NHNH2, or

R8 is C1-C4alkyl, C1-C4alkoxy, mono- or di-(C1-C4)alkylamino, mono-, di-, or tri-C1-C4 alkylhydrazinyl, C2-C4alkanoyl, C1-C4alkylester, C1-C2haloalkyl, or C1-C2haloalkoxy; each of which is substituted with 0 to 3 substituents independently chosen from hydroxy, amino, halogen, oxo, C1-C4alkoxy, C1-C2haloalkyl, C1-C2haloalkoxy, and mono- and di-C1-C4alkylamino.

R9 is C1-C8alkyl, C3-C7cycloalkyl(C0-C4alkyl), or phenyl, each of which is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, —COOH, —CONH2, C1-C4alkyl, C2-C4alkenyl, C2-C4alkynyl, C1-C4alkoxy, C3-C7cycloalkyl(C0-C4alkyl), C3-C7cycloalkyl(C0-C4alkoxy), mono- and di-(C1-C4)alkylamino C1-C2haloalkyl, C1-C2haloalkoxy, and C2-C4alkanoyl.

The invention includes novel intermediates useful for the synthesis of antimicrobial compounds of Formula I and Formula II. These intermediates are compounds of Formula I and Formula II in which R7 is bromo, iodo, —O(SO2)CF3, or —N2BF4. The invention provides methods of synthesizing compounds of Formula I and Formula II comprising coupling an intermediate of the invention to an appropriate aryl or heteroaryl boronic acid, aryl or heteroaryl boronic acid ester, or compounds substituted with Li, Mg, B, Al, Si, Zn, Cu, Zr, or Sn at the point of coupling.

DETAILED DESCRIPTION OF THE INVENTION Chemical Description and Terminology

Prior to setting forth the invention in detail, it may be helpful to provide definitions of certain terms to be used herein. Compounds of the present invention are generally described using standard nomenclature.

In certain situations, the compounds of Formula I and Formula II may contain one or more asymmetric elements such as stereogenic centers, stereogenic axes and the like, e.g. asymmetric carbon atoms, so that the compounds can exist in different stereoisomeric forms. These compounds can be, for example, racemates or optically active forms. For compounds with two or more asymmetric elements, these compounds can additionally be mixtures of diastereomers. For compounds having asymmetric centers, it should be understood that all of the optical isomers and mixtures thereof are encompassed. In addition, compounds with carbon-carbon double bonds may occur in Z- and E-forms, with all isomeric forms of the compounds being included in the present invention. In these situations, the single enantiomers, i.e., optically active forms can be obtained by asymmetric synthesis, synthesis from optically pure precursors, or by resolution of the racemates. Resolution of the racemates can also be accomplished, for example, by conventional methods such as crystallization in the presence of a resolving agent, or chromatography, using, for example a chiral HPLC column.

Where a compound exists in various tautomeric forms, the invention is not limited to any one of the specific tautomers, but rather includes all tautomeric forms.

The present invention is intended to include all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example, and without limitation, isotopes of hydrogen include tritium and deuterium and isotopes of carbon include 11C, 13C, and 14C.

Certain compounds are described herein using a general formula that includes variables, e.g. A1, R2, R3, R5, R6, R7, A8, and R9. Unless otherwise specified, each variable within such a formula is defined independently of other variables. Thus, if a group is said to be substituted, e.g. with 0-2 R*, then said group may be substituted with up to two R* groups and R* at each occurrence is selected independently from the definition of R*. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.

The term “substituted”, as used herein, means that any one or more hydrogens on the designated atom or group is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded. When a substituent is oxo (i.e., ═O), imine (e.g. ═NHR), or oxime (e.g. ═NOR) then 2 hydrogens on the atom are replaced. An “oxo,” imine, or oxime substituent on an aromatic group or heteroaromatic group destroys the aromatic character of that group, e.g. a pyridyl substituted with oxo is pyridone. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds or useful synthetic intermediates. A stable compound or stable structure is meant to imply a compound that is sufficiently robust to survive isolation from a reaction mixture, and subsequent formulation into an effective therapeutic agent. Unless otherwise specified substituents are named into the core structure. For example, it is to be understood that when cycloalkyl(alkyl) is listed as a possible substituent the point of attachment of this substituent to the core structure is in the alkyl portion.

A dash (“—”) that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, —CONH2 is attached through the carbon atom.

As used herein, “alkyl” is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups, having the specified number of carbon atoms. Thus, the term C1-C6 alkyl as used herein includes alkyl groups having from 1 to about 6 carbon atoms. When C0-Cn alkyl is used herein in conjunction with another group, for example, arylC0-C4alkyl, the indicated group, in this case aryl, is either directly bound by a single covalent bond (C0), or attached by an alkyl chain having the specified number of carbon atoms, in this case from 1 to about 4 carbon atoms. Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, n-pentyl, and sec-pentyl. Preferred alkyl groups are lower alkyl groups, those alkyl groups having from 1 to about 8 carbon atoms, e.g. C1-C8 and C1-C6alkyl groups.

“Alkenyl” as used herein, indicates a hydrocarbon chain of either a straight or branched configuration having one or more carbon-carbon double bond bonds, which may occur at any stable point along the chain. Examples of alkenyl groups include ethenyl and propenyl.

“Alkynyl” as used herein, indicates a hydrocarbon chain of either a straight or branched configuration having one or more triple carbon-carbon bonds that may occur in any stable point along the chain, such as ethynyl and propynyl.

“Carbohydryl” as used herein, includes both branched and straight-chain hydrocarbon groups, which are saturated or unsaturated, having the specified number of carbon atoms. When C0-Cn carbohydrylis used herein in conjunction with another group, for example, arylC0-C4carbohydryl, the indicated group, in this case aryl, is either directly bound by a single covalent bond (C0), or attached by an carbohydryl chain, such as an alkyl chain, having the specified number of carbon atoms, in this case from 1 to about 4 carbon atoms. Examples include C1-C6alkyl, such as methyl, or 5-butyl, C2-C6alkynyl such and hexynyl, and C2-C6 alkenyl, such as 1-propenyl.

“Alkoxy” represents an alkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge. Examples of alkoxy include, but are not limited to, methoxy, ethoxy, n-propoxy, propoxy, n-butoxy,2-butoxy, t-butoxy, n-pentoxy, 2-pentoxy, 3-pentoxy, isopentoxy, neopentoxy, n-hexoxy, 2-hexoxy, 3-hexoxy, and 3-methylpentoxy.

“Alkanoyl” indicates an alkyl group as defined above, attached through a keto (—(C═O)—) bridge. Alkanoyl groups have the indicated number of carbon atoms, with the carbon of the keto group being included in the numbered carbon atoms. For example a C2alkanoyl group is an acetyl group having the formula CH3(C═O)—.

As used herein, the terms “mono- or di-alkylamino” or “mono- and di-alkylamino” indicate secondary or tertiary alkyl amino groups, wherein the alkyl groups are as defined above and have the indicated number of carbon atoms. The point of attachment of the alkylamino group is on the nitrogen. Examples of mono- and di-alkylamino groups include ethylamino, dimethylamino, and methyl-propyl-amino.

The term “mono- or di-alkylcarbamate” indicates 1 or 2 independently chosen alkyl groups, as define above, attached through a carbamate (—O(C═O)NRR) linkage where R represents the alkyl groups. Mono-alkylcarbamate groups have the formula (—O(C═O)NHR.

The term “alkylester” indicates and alkyl group as define above attached through an ester linkage, i.e. a group of the formula —O(C═O)alkyl.

The term “mono-, di-, or tri-alkylhydrazinyl” indicates from 1 to 3 independently chosen alkyl group as defined above attached through a single-bonded nitrogen-nitrogen linkage. At least one of the alkyl groups is attached to the terminal nitrogen (the nitrogen not bound to the core structure). When the term mono- or di-alkylhydrazinyl is used only the terminal nitrogen is alkyl substituted. Examples of alkylhydrazinyl groups include 2-butyl-1-hydrazinyl, 2-butyl-2-methyl-1-hydrazinyl, and 1,2-dimethyl-2-propyl-1-hydrazinyl.

The term “alkylthio” indicates an alkyl group as defined above attached through a sulfur linkage, i.e. a group of the formula alkyl-S—. Examples include ethylthio and pentylthio.

As used herein, the term “aryl” indicates aromatic groups containing only carbon in the aromatic ring or rings. Typical aryl groups contain 1 to 3 separate, fused, or pendant rings and from 6 to about 18 ring atoms, without heteroatoms as ring members. When indicated, such aryl groups may be further substituted with carbon or non-carbon atoms or groups. Such substitution may include fusion to a 5 to 7-membered saturated cyclic group that optionally contains 1 or 2 heteroatoms independently chosen from N, O, and S, to form, for example, a 3,4-methylenedioxy-phenyl group. Aryl groups include, for example, phenyl, naphthyl, including 1-naphthyl and 2-naphthyl, and bi-phenyl.

In the term “aryl(alkyl)”, aryl and alkyl are as defined above, and the point of attachment is on the alkyl group. This term encompasses, but is not limited to, benzyl, phenylethyl, and piperonyl. Likewise, in the term aryl(carbohydryl), aryl and carbohydryl are as defined above and the point of attachment is on the carbohydryl group, for example a phenylpropen-1-yl group.

“Cycloalkyl” as used herein, indicates saturated hydrocarbon ring groups, having the specified number of carbon atoms, usually from 3 to about 8 ring carbon atoms, or from 3 to about 7 carbon atoms. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl as well as bridged or caged saturated ring groups such as norborane or adamantane.

“Cycloalkenyl” as used herein, indicates an unsaturated, but not aromatic, hydrocarbon ring having at least one carbon-carbon double bond. Cycloalkenyl groups contain from 4 to about 8 carbon atoms, usually from 4 to about 7 carbon atoms. Examples include cyclohexenyl and cyclobutenyl.

In the terms “cycloalkyl(alkyl),” “cycloalkyl(carbohydryl),” and “cycloalkyl(alkoxy)” the terms cycloalkyl, alkyl, carbohydryl, and alkoxy are as defined above, and the point of attachment is on the alkyl, carbohydryl, or alkoxy group respectively. These terms include examples such as cyclopropylmethyl, cyclohexylmethyl, cyclohexylpropenyl, and cyclopentylethyoxy.

In the terms “cycloalkenyl(alkyl)” “cycloalkenyl(carbohydryl)” and the terms cycloalkenyl, alkyl, and carbohydryl are as defined above, and the point of attachment is on the alkyl or carbohydryl group respectively. These terms include examples such as cyclobutenylmethyl, cyclohexenylmethyl, and cyclohexylpropenyl.

“Haloalkyl” indicates both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more halogen atoms, generally up to the maximum allowable number of halogen atoms. Examples of haloalkyl include, but are not limited to, trifluoromethyl, difluoromethyl, 2-fluoroethyl, and penta-fluoroethyl.

“Haloalkoxy” indicates a haloalkyl group as defined above attached through an oxygen bridge.

“Halo” or “halogen” as used herein refers to fluoro, chloro, bromo, or iodo.

As used herein, “heteroaryl” indicates a stable 5- to 7-membered monocyclic or 7- to 10-membered bicyclic heterocyclic ring which contains at least 1 aromatic ring that contains from 1 to 4, or preferably from 1 to 3, heteroatoms chosen from N, O, and S, with remaining ring atoms being carbon. When the total number of S and O atoms in the heteroaryl group exceeds 1, these heteroatoms are not adjacent to one another. It is preferred that the total number of S and O atoms in the heteroaryl group is not more than 2. It is particularly preferred that the total number of S and O atoms in the aromatic heterocycle is not more than 1. A nitrogen atom in a heteroaryl group may optionally be quaternized. When indicated, such heteroaryl groups may be further substituted with carbon or non-carbon atoms or groups. Such substitution may include fusion to a 5 to 7-membered saturated cyclic group that optionally contains 1 or 2 heteroatoms independently chosen from N, O, and S, to form, for example, a [1,3]dioxolo[4,5-c]pyridyl group. Examples of heteroaryl groups include, but are not limited to, pyridyl, indolyl, pyrimidinyl, pyridizinyl, pyrazinyl, imidazolyl, oxazolyl, furanyl, thiophenyl, thiazolyl, triazolyl, tetrazolyl, isoxazolyl, quinolinyl, pyrrolyl, pyrazolyl, benz[b]thiophenyl, isoquinolinyl, quinazolinyl, quinoxalinyl, thienyl, isoindolyl, and 5,6,7,8-tetrahydroisoquinoline.

In the terms “heteroarylalkyl” and “heteroaryl(carbohydryl),” heteroaryl, alkyl, and carbohydryl are as defined above, and the point of attachment is on the alkyl or carbohydryl group respectively. These terms include such examples as pyridylmethyl, thiophenylmethyl, and pyrrolyl(1-ethyl).

The term “heterocycloalkyl” indicates a saturated cyclic group containing from 1 to about 3 heteroatoms chosen from N, O, and S, with remaining ring atoms being carbon. Heterocycloalkyl groups have from 3 to about 8 ring atoms, and more typically have from 5 to 7 ring atoms. A C2-C7heterocycloalkyl group contains from 2 to about 7 carbon ring atoms and at least one ring atom chosen from N, O, and S. Examples of heterocycloalkyl groups include morpholinyl, piperazinyl, piperidinyl, and pyrrolidinyl groups. A nitrogen in a heterocycloalkyl group may optionally be quaternized.

The term “heterocyclic group” indicates a 5-6 membered saturated, partially unsaturated, or aromatic ring containing from 1 to about 4 heteroatoms chosen from N, O, and S, with remaining ring atoms being carbon or a 7-10 membered bicyclic saturated, partially unsaturated, or aromatic heterocylic ring system containing at least 1 heteroatom in the two ring system chosen from N, O, and S and containing up to about 4 heteroatoms independently chosen from N, O, and S in each ring of the two ring system. Unless otherwise indicated, the heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. When indicated the heterocyclic rings described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable. A nitrogen atom in the heterocycle may optionally be quaternized. It is preferred that the total number of heteroatoms in a heterocyclic groups is not more than 4 and that the total number of S and O atoms in a heterocyclic group is not more than 2, more preferably not more than 1. Examples of heterocyclic groups include, pyridyl, indolyl, pyrimidinyl, pyridizinyl, pyrazinyl, imidazolyl, oxazolyl, furanyl, thiophenyl, thiazolyl, triazolyl, tetrazolyl, isoxazolyl, quinolinyl, pyrrolyl, pyrazolyl, benz[b]thiophenyl, isoquinolinyl, quinazolinyl, quinoxalinyl, thienyl, isoindolyl, dihydroisoindolyl, 5,6,7,8-tetrahydroisoquinoline, pyridinyl, pyrimidinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrrolidinyl, morpholinyl, piperazinyl, piperidinyl, and pyrrolidinyl.

Additional examples heterocyclic groups include, but are not limited to, phthalazinyl, oxazolyl, indolizinyl, indazolyl, benzothiazolyl, benzimidazolyl, benzofuranyl, benzoisoxolyl, dihydro-benzodioxinyl, oxadiazolyl, thiadiazolyl, triazolyl, tetrazolyl, oxazolopyridinyl, imidazopyridinyl, isothiazolyl, naphthyridinyl, cinnolinyl, carbazolyl, beta-carbolinyl, isochromanyl, chromanonyl, chromanyl, tetrahydroisoquinolinyl, isoindolinyl, isobenzotetrahydrofuranyl, isobenzotetrahydrothienyl, isobenzothienyl, benzoxazolyl, pyridopyridinyl, benzotetrahydrofuranyl, benzotetrahydrothienyl, purinyl, benzodioxolyl, triazinyl, phenoxazinyl, phenothiazinyl, pteridinyl, benzothiazolyl, imidazopyridinyl, imidazothiazolyl, dihydrobenzisoxazinyl, benzisoxazinyl, benzoxazinyl, dihydrobenzisothiazinyl, benzopyranyl, benzothiopyranyl, coumarinyl, isocoumarinyl, chromanyl, tetrahydroquinolinyl, dihydroquinolinyl, dihydroquinolinonyl, dihydroisoquinolinonyl, dihydrocoumarinyl, dihydroisocoumarinyl, isoindolinonyl, benzodioxanyl, benzoxazolinonyl, pyrrolyl N-oxide, pyrimidinyl N-oxide, pyridazinyl N-oxide, pyrazinyl N-oxide, quinolinyl N-oxide, indolyl N-oxide, indolinyl N oxide, isoquinolyl N-oxide, quinazolinyl N-oxide, quinoxalinyl N-oxide, phthalazinyl N-oxide, imidazolyl N-oxide, isoxazolyl N-oxide, oxazolyl N-oxide, thiazolyl N-oxide, indolizinyl N oxide, indazolyl N-oxide, benzothiazolyl N-oxide, benzimidazolyl N-oxide, pyrrolyl N-oxide, oxadiazolyl N-oxide, thiadiazolyl N-oxide, tetrazolyl N-oxide, benzothiopyranyl S-oxide, and benzothiopyranyl S,S-dioxide.

As used herein “Active agents” are compounds that have pharmaceutical utility, e.g. may be used to treat a patient suffering from a disease or condition, or may be used prophylacticly to prevent the onset of a disease or condition in a patient, or that may be used to enhance the pharmaceutical activity of other compounds.

“Salts” of the compounds of the present invention include inorganic and organic acid and base addition salts. The salts of the present compounds can be synthesized from a parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two. Generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred, where practicable. Salts of the present compounds further include solvates of the compounds and of the compound salts.

“Pharmaceutically acceptable salts” includes derivatives of the disclosed compounds wherein the parent compound is modified by making non-toxic acid or base salts thereof, and further refers to pharmaceutically acceptable solvates of such compounds and such salts. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts and the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, conventional non-toxic acid salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, mesylic, esylic, besylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, HOOC—(CH2)n—COOH where n is 0-4, and the like. Lists of additional suitable salts may be found, e.g., in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., p. 1418 (1985).

The term “prodrugs” includes any compounds that become compounds of Formula I when administered to a mammalian subject, e.g., upon metabolic processing of the prodrug. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate and like derivatives of functional groups (such as alcohol or amine groups) in the compounds of Formula I and Formula II.

The term “therapeutically effective amount” of a compound of this invention means an amount effective, when administered to a human or non-human patient, to provide a therapeutic benefit such as an amelioration of symptoms, e.g., an amount effective to decrease the symptoms of a microbial infection, and preferably an amount sufficient to reduce the symptoms of a bacterial, fungal, or protozoal infection. In certain circumstances a patient suffering from a microbial infection may not present symptoms of being infected. Thus a therapeutically effective amount of a compound is also an amount sufficient to prevent a significant increase or significantly reduce the detectable level of microorganism or antibodies against the microorganism in the patient's blood, serum, other bodily fluids, or tissues. The invention also includes using compounds of Formula I and Formula II in prophylactic therapies. In the context of prophylactic or preventative treatment a “therapeutically effective amount” is an amount sufficient to significantly decrease the treated animal's risk of contracting a microorganism infection. A significant reduction is any detectable negative change that is statistically significant in a standard parametric test of statistical significance such as Student's T-test, where p<0.05.

Antimicrobial Compounds

For the purposes of this document, the following numbering system will apply to the core 9H-isothiazolo[5,4-b]quinoline-3,4-dione (when A1=sulfur) structure or core 9H-isoxazolo[5,4-b]quinoline-3,4-dione (when A1=oxygen) structure. The numbers 1 through 9 refer specifically to positions within the tricyclic ring system whereas the letters A, B and C refer to the specific six (rings A and B) or five (ring C) member rings as shown below.

In addition to the compounds of Formula I and Formula II, described above the invention also includes compounds of Formula I and Formula II in which the variables (e.g. A1, R2, R3, R4, etc.) carry definitions other than those set forth above.

The A1 Variable

In certain embodiments, the invention includes compounds of Formula I and Formula II A1 is Sulfur.

In other embodiments A1 is SO.

The invention also includes compounds of Formula I and Formula II in which A1 is SO2.

In still other embodiments A1 is O.

The R2 Variable

The invention includes compounds of Formula I in which R2 is hydrogen or R2 is C1-C6alkyl or C3-C7cycloalkyl(C0-C4alkyl), each of which is substituted with at least one substituent chosen from hydroxy, amino, —COOH, —(C═O)NR10OR11, and —CONH2; and is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, —COOH, —CONH2, C1-C4alkyl, C1-C4alkoxy, C1-C2haloalkyl, C1-C2haloalkoxy, and mono- and di-C1-C4alkylamino, and C2-C4alkanoyl. Certain embodiments of the invention pertain to compounds of Formula I in which R2 is hydrogen.

The R3 Variable

The invention includes compounds and salts of Formula II in which R3 is C1-C6alkyl, C1-C6alkanoyl, mono- or di-C1-C6alkylcarbamate, or C1-C6alkylsulfonate; each of which is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, C1-C4alkoxy, mono- and di-C1-C4alkylamino, C1-C2haloalkyl, and C1-C2haloalkoxy.

The invention also includes compounds and salts of Formula II in which R3 is C1-C6alkyl or C1-C6alkanoyl, each of which is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, C1-C2alkoxy, mono- and di-C1-C2alkylamino, C1-C2haloalkyl, and C1-C2haloalkoxy.

The invention also includes compounds and salts of Formula II in which R3 is C1-C6alkyl or C1-C6alkanoyl.

The R5 Variable

Certain embodiments of the invention pertain to compounds and salts of Formula I and Formula II in which R5 is hydrogen, amino, C1-C2alkyl, C1-C2alkoxy, mono- or di-(C1-C4)alkylamino, or mono- or di-C1-C4 alkylhydrazinyl.

The invention also includes compounds and salts of Formula I and Formula II in which R5 hydrogen, amino, mono- or di-(C1-C2)alkylamino, or mono- or di-C1-C2 alkylhydrazinyl.

The invention includes compounds and salts of Formula I and Formula II in which R5 is hydrogen.

The R6 Variable

The invention includes compounds and salts of Formula I and Formula II in which R6 is hydrogen, halogen, or amino.

In certain embodiments the invention pertains to compounds and salts of Formula I and Formula II in which R6 is fluoro or hydrogen.

The invention includes compounds and salts of Formula I and Formula II in which R6 is halogen and R8 is C1-C2alkoxy.

The invention includes compounds and salts of Formula I and Formula II in which R6 is fluorine and R8 is methoxy.

The R7 Variable

The invention includes compounds and salts of Formula I and Formula II in which R7 is bromo, iodo, —O(SO2)CF3, or —N2BF4. These compounds are particularly useful as intermediates in the synthesis of antimicrobial compounds of Formula I and Formula II.

The invention includes compounds and salts of Formula I and Formula II in which R7 is XRA where X is absent, CH2—CH2—, —CH═CH—, or —C═C—; and RA is a 7-10 membered bicyclic saturated, partially unsaturated, or aromatic carbocyclic group, or RA is a 5-6 membered saturated, partially unsaturated, or aromatic heterocylic group bound via a carbon atom when X is absent or —CH2—CH2—, or bound via a carbon or nitrogen atom when X is —CH═CH— or —C≡C— or a RA is a 7-10 membered bicyclic saturated, partially unsaturated, or aromatic heterocylic group bound via a carbon atom when X is absent or —CH2—CH2—, or bound via a carbon or nitrogen atom when X is —CH═CH— or —C═C—; each of which RA is substituted with 0 to 5 substituents independently chosen from (i), (ii), and (iii). where:

    • (i) is chosen from halogen, hydroxy, amino, cyano, and nitro; and
    • (ii) is chosen from C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C3-C7cycloalkyl(C0-C4alkyl), C3-C7cycloalkyl(C0-C4alkoxy), C4-C7cycloalkenyl(C0-C4alkyl), aryl(C0-C6carbohydryl), aryl(C1-C4alkoxy), C2-C6heterocycloalkyl(C0-C4alkyl), heteroaryl(C0-C6carbohydryl), C1-C6alkylthio, ═NOR10, —(C0-C4alkyl)(C═O)R10, —(C0-C4alkyl)O(C═O)R10, —(C0-C4alkyl)(C═O)NR10R11, —(C0-C4alkyl)O(C═O)NR10R11, —(C0-C4alkyl) (C═O)OR10, —(C0-C4alkyl)NR10(C═O)R11, —(C0-C4alkyl)NR10(C═O)OR11, —(C0-C4alkyl)NR10(C═O)NR11R12, —(CO-C4alkyl)NR10(C═O)(C1-C4alkyl)NR11(C═O)O—R12, —(C0-C4alkyl)NR10(C═S)NR11R12, —(C0-C4alkyl)NR10NR11R12, —(C0-C4alkyl)N═NR13, —(C0-C4alkyl)SO3R10, —(C0-C4alkyl)(S═O)OR10, —(C0-C4alkyl)SO2R13, —(C0-C4alkyl)SO2NR10R11, and —(C0-C4alkyl)NR10SO2R13; and
    • (iii) is chosen from —ORD, —(C═O)RD, —SO2RD, —SO3RD, —NR10SO2RD, where RD is C1-C4alkyl, C3-C7cycloalkyl(C0-C2alkyl), C2-C6heterocycloalkyl(C0-C2alkyl), aryl(C0-C2alkyl), or heteroaryl(C0-C2alkyl).

Where each of (ii) and (iii) is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, oxo, —COOH, —CONH2, C2-C4alkenyl, C2-C4alkynyl, C1-C4alkoxy, C1-C4alkoxycarbonyl, C3-C7cycloalkyl(C0-C4alkyl), C3-C7cycloalkyl(C0-C4alkoxy), mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C2-C4alkanoyl, and phenyl.

The invention also pertains to compounds and salts of Formula I and Formula II in which

R7 is XRA, X is absent, CH2—CH2—, —CH═CH—, or —C≡—; and

RA is naphthyl, dihydronapthyl, tetrahydronapthyl, pyridyl, pyrimidinyl, pyrazinyl, furanyl, benz[b]thiophenyl, benzofuranyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, isoxazolyl, indolyl, dihydroindolyl, pyrrolyl, pyrazolyl, imidazolyl, thienyl, isoindolyl, dihydroisoindolyl, tetrahydropyridinyl, tetrahydroisoquinolinyl, or piperidin-4-yl group; each of which is substituted with 0 to 5 substituents independently chosen from (i), (ii), and (iii) which are as defined above.

The invention also includes compounds and salts of Formula I and Formula II in which

R7 is XRA, X is absent, CH2—CH2—, —CH═CH—, or —C≡C—; and

RA is naphthyl, dihydronapthyl, tetrahydronapthyl, pyridyl, pyrimidinyl, pyrazinyl, furanyl, benz[b]thiophenyl, benzofuranyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, isoxazolyl, indolyl, dihydroindolyl, pyrrolyl, pyrazolyl, imidazolyl, thienyl, isoindolyl, dihydroisoindolyl, tetrahydropyridinyl, tetrahydroisoquinolinyl, or piperidin-4-yl group; each of which is substituted with 0 to 5 substituents independently chosen from (i), (ii), and (iii). In this embodiment (i), (ii), and (iii) carry the following definitions:

    • (i) is chosen from halogen, hydroxy, amino, cyano, and nitro,
    • (ii) is chosen from C1-C4alkyl, C2-C4alkenyl, C1-C4alkoxy, mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C3-C7cycloalkyl(C0-C2alkyl), C3-C7cycloalkyl(C0-C2alkoxy), phenyl(C0-C2alkyl), phenyl(C0-C2alkoxy), pyrrolidinyl(C0-C2alkyl), piperidinyl(C0-C2alkyl), piperazinyl(C0-C2alkyl), morpholinyl(C0-C2alkyl), thiomorpholinyl(C0-C2alkyl), pyridyl, pyrimidinyl, pyrazinyl, furanyl, benzofuranyl, pyrrolyl, pyrazolyl, imidazolyl, thienyl, C1-C4alkylthio, ═NOR10, —(C0-C4alkyl)(C═O)R10, —(C0-C4alkyl)O(C═O)R10, —(C0-C4alkyl)(C═O)NR10R11, —(C0-C4alkyl)O(C═O)NR10R11, —(C0-C4alkyl) (C═O)OR10, —(C0-C4alkyl)NR10(C═O)R11, —(C0-C4alkyl)NR10(C═O)OR11, —(C0-C4alkyl)NR10(C═O)NR11R12, —(C0-C4alkyl)NR10(C═O)(C1-C4alkyl)NR11(C═O)O—R12, —(C0-C4alkyl)NR10(C═S)NR11R12, —(C0-C4alkyl)NR10NR11R12, —(C0-C4alkyl)N═NR13, —(C0-C4alkyl)SO3R10, —(C0-C4alkyl)(S═O)OR10, —(C0-C4alkyl)SO2R13, —(C0-C4alkyl)SO2NR10R11, and —(C0-C4alkyl)NR10SO2R13; and
    • (iii) is chosen from —ORD, —(C═O)RD, —SO2RD, —SO3RD, —NR10SO2RD, where RD is C1-C4alkyl, C3-C7cycloalkyl(C0-C2alkyl), pyrrolidinyl(C0-C2alkyl), piperidinyl(C0-C2alkyl), piperazinyl(C0-C2alkyl), morpholinyl(C0-C2alkyl), thiomorpholinyl(C0-C2alkyl), phenyl(C0-C2alkyl), naphthyl(C0-C2alkyl), pyridyl(C0-C2alkyl), pyrimidinyl(C0-C2alkyl), pyrazinyl(C0-C2alkyl), furanyl(C0-C2alkyl), benz[b]thiophenyl(C0-C2alkyl), benzofuranyl(C0-C2alkyl), quinolinyl(C0-C2alkyl), isoquinolinyl(C0-C2alkyl), quinazolinyl(C0-C2alkyl), isoxazolyl(C0-C2alkyl), indolyl(C0-C2alkyl), dihydroindolyl(C0-C2alkyl), pyrrolyl(C0-C2alkyl), pyrazolyl(C0-C2alkyl), imidazolyl(C0-C2alkyl), thienyl(C0-C2alkyl), isoindolyl(C0-C2alkyl), or dihydroisoindolyl(C0-C2alkyl).

Where each of (ii) and (iii) is substituted with 0 to 3 substitutents independently chosen from halogen, hydroxy, amino, cyano, nitro, oxo, —COOH, —CONH2, C1-C4alkyl, C2-C4alkenyl, C2-C4alkynyl, C1-C4alkoxy, C1-C4alkoxycarbonyl, C3-C7cycloalkyl(C0-C4alkyl), C3-C7cycloalkyl(C0-C4alkoxy), mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C2-C4alkanoyl, and phenyl.

The invention further includes compounds and salts of Formula I and Formula II in which R7 is XRA, X is absent, CH2—CH2—, —CH═CH—, or —C≡C—; and RA is a naphthyl, dihydronapthyl, tetrahydronapthyl, pyridyl, pyrimidinyl, pyrazinyl, furanyl, benz[b]thiophenyl, benzofuranyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, isoxazolyl, indolyl, dihydroindolyl, pyrrolyl, pyrazolyl, imidazolyl, thienyl, isoindolyl, dihydroisoindolyl, tetrahydropyridinyl, tetrahydroisoquinolinyl, or piperidin-4-yl group; each of which is substituted with 0 to 5 substituents independently chosen from (i), (ii), and (iii).

In this embodiment (i) is chosen from halogen, hydroxy, amino, cyano, and nitro, (ii) is chosen from C1-C4alkyl, C2-C4alkenyl, C1-C4alkoxy, mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C3-C7cycloalkyl(C0-C2alkyl), C3-C7cycloalkyl(C0-C2alkoxy), phenyl(C0-C2alkyl), phenyl(C0-C2alkoxy), pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, pyridyl, thienyl, C1-C4alkylthio, —(C═O)OR10, and —(C═O)NR10R11; and (iii) is chosen from —ORD, —(C═O)RD, —SO2RD, —SO3RD, and —NR10SO2RD, where RD is C1-C4alkyl, C3-C7cycloalkyl(C0-C2alkyl), piperidinyl, piperazinyl, phenyl, naphthyl, or pyridyl.

Each of (ii) and (iii) is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, oxo, —COOH, —CONH2, C1-C4alkyl, C1-C4alkoxy, C1-C4alkoxycarbonyl, C3-C7cycloalkyl(C0-C2alkyl), mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C2-C4alkanoyl, and phenyl.

Another embodiment of the invention pertains to compounds and salts of Formula I and Formula II in which R7 is XRA, X is absent, CH2—CH2—, —CH═CH—, or —C═C—; and RA is a naphthyl, dihydronapthyl, tetrahydronapthyl, pyridyl, pyrimidinyl, pyrazinyl, furanyl, benz[b]thiophenyl, benzofuranyl, quinolinyl, isoquinolinyl, quinazolinyl, quinoxalinyl, isoxazolyl, indolyl, dihydroindolyl, pyrrolyl, pyrazolyl, imidazolyl, thienyl, isoindolyl, dihydroisoindolyl, tetrahydropyridinyl tetrahydroisoquinolinyl, or piperidin-4-yl group; each of which is substituted with 0 to 5 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, C1-C4alkyl, C2-C4alkenyl, C1-C4alkoxy, mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C3-C7cycloalkyl(C0-C2alkyl), C3-C1cycloalkyl(C0-C2alkoxy), phenyl(C0-C2alkyl), phenyl(C0-C2alkoxy), pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, pyridyl, thienyl, C1-C4alkylthio, —(C═O)OR10, —(C═O)NR10R11; —ORD, —(C═O)RD, —SO2RD, —SO3RD, and —NR10SO2RD.

In this embodiment RD is C1-C4alkyl, piperidinyl, phenyl, naphthyl, or pyridyl; and each RD is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, C1-C2alkyl, C1-C2alkoxy, mono- and di-(C1-C2)alkylamino, C1-C2haloalkyl, and C1-C2haloalkoxy.

The invention includes certain compounds of Formula I and Formula II in which R7 is XRA and X is absent.

Other embodiments of the invention pertain to compounds and salts of Formula I and Formula II in which R7 is XRA, X is absent, CH2—CH2—, —CH═CH—, or —C≡C—; and RA is a pyridyl, pyrimidinyl, furanyl, quinolinyl, indolyl, pyrrolyl, isoindolyl, tetrahydroisoquinolinyl, or thienyl group; each of which is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, C1-C4alkyl, C1-C4alkoxy, mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, and C1-C2haloalkoxy.

The invention also includes certain compounds and salt of Formula I and Formula II in which RA is pyrid-3-yl, pyrid-4-yl, pyrimidin-5-yl, furan-3-yl, quinolin-3-yl, quinolin-5-yl, quinolin-6-yl, isoindol-5-yl, tetrahydroisoquinolin-5-yl, tetrahydroisoquinolin-6-yl, tetrahydroisoquinolin-7-yl, tetrahydroisoquinolin-8-yl, or indol-5-yl, each of which is substituted with 0 to 2 substituents independently chosen from halogen, hydroxy, amino, C1-C2alkyl, and C1-C2alkoxy.

Further included herein are certain compounds and salts of Formula I and Formula II in which RA is pyridyl-3-yl or pyrid-4-yl, each of which is substituted with 1 or 2 substituents independently chosen from fluoro, amino, hydroxy, cyano, and methyl.

The invention also provides compounds and salts of Formula I and Formula II in which RA is tetrahydroisoquinolin-5-yl, tetrahydroisoquinolin-6-yl, tetrahydroisoquinolin-7-yl, or tetrahydroisoquinolin-8-yl, each of which is substituted with 0 to 3 substituents independently chosen from C1-C3alkyl.

Also included are compounds and salts of Formula I and Formula II in which RA is tetrahydroisoquinolin-6-yl or tetrahydroisoquinolin-7-yl, each of which is substituted with 0 to 3 substituents independently chosen from C1-C3alkyl.

Compounds and salts of Formula I and Formula II are provided herein in which RA is tetrahydroisoquinolin-6-yl substituted at the 1, 2, and 3 positions with 0 to 3 methyl substituents.

Further included are compounds and salts of Formula I and Formula II in which

RA is isoindol-5-yl substituted with 0 to 3 independently chosen C1-C3alkyl substituents.

In still other embodiments the invention provides compounds and salts of Formula I and Formula II, in which RA is isoindol-5-yl substituted at the 1, 2, or 3 positions with 0 to 3 methyl substituents.

Still other embodiments of the invention include compounds and salts of Formula I and Formula II in which R7 is XRB where X is absent, CH2—CH2—, —CH═CH—, or —C═C—; and RB is phenyl; substituted with 1 to 5 substituents independently chosen from (i), (ii), and (iii), wherein

    • (i) is chosen from halogen, hydroxy, amino, cyano, and nitro,
    • (ii) is chosen from C1-C4alkyl, C2-C4alkenyl, C1-C4alkoxy, mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C3-C7cycloalkyl(C0-C2alkyl), C3-C7cycloalkyl(C0-C2alkoxy), phenyl(C0-C2alkyl), phenyl(C0-C2alkoxy), pyrrolidinyl(C0-C2alkyl), piperidinyl(C0-C2alkyl), piperazinyl(C0-C2alkyl), morpholinyl(C0-C2alkyl), thiomorpholinyl(C0-C2alkyl), pyridyl, pyrimidinyl, pyrazinyl, furanyl, benzofuranyl, pyrrolyl, pyrazolyl, imidazolyl, thienyl, C1-C4alkylthio, ═NOR10, —(C0-C4alkyl)(C═O)R10, —(C0-C4alkyl)O(C═O)R10, —(C0-C4alkyl)(C═O)NR10R11, —(C0-C4alkyl)O(C═O)NR10R11, —(C0-C4alkyl) (C═O)OR10, —(C0-C4alkyl)NR10(C═O)R11, —(C0-C4alkyl)NR10(C═O)OR11, —(C0-C4alkyl)NR10(C═O)NR11R12, —(C0C4alkyl)NR10(C═O)(C1-C4)NR11(C═O)OR12, —(C0-C4alkyl)NR10(C═S)NR11R12, —(C0-C4alkyl)NR10NR11R12, —(C0-C4alkyl)N═NR13, —(C0-C4alkyl)SO3R10, —(C0-C4alkyl)(S═O)OR10, —(C0-C4alkyl)SO2R13, —(C0-C4alkyl)SO2NR10R11, and —(C0-C4alkyl)NR10SO2R13; and
    • (iii) is chosen from —ORD, —(C═O)RD, —SO2RD, —SO3RD, —NR 10SO2RD, where RD is C4alkyl, C3-C7cycloalkyl(C0-C2alkyl), pyrrolidinyl(C0-C2alkyl), piperidinyl(C0-C2alkyl), piperazinyl(C0-C2alkyl), morpholinyl(C0-C2alkyl), thiomorpholinyl(C0-C2alkyl), phenyl(C0-C2alkyl), naphthyl(C0-C2alkyl), pyridyl(C0-C2alkyl), pyrimidinyl(C0-C2alkyl), pyrazinyl(C0-C2alkyl), furanyl(C0-C2alkyl), benz[b]thiophenyl(C0-C2alkyl), benzofuranyl(C0-C2alkyl), quinolinyl(C0-C2alkyl), isoquinolinyl(C0-C2alkyl), quinazolinyl(C0-C2alkyl), isoxazolyl(C0-C2alkyl), indolyl(C0-C2alkyl), dihydroindolyl(C0-C2alkyl), pyrrolyl(C0-C2alkyl), pyrazolyl(C0-C2alkyl), imidazolyl(C0-C2alkyl), thienyl(C0-C2alkyl), isoindolyl(C0-C2alkyl), and dihydroisoindolyl(C0-C2alkyl).

Each of (ii) and (iii) is substituted with 0 to 3 substitutents independently chosen from halogen, hydroxy, amino, cyano, nitro, oxo, —COOH, —CONH2, C1-C4alkyl, C2-C4alkenyl, C2-C4alkynyl, C1-C4alkoxy, C1-C4alkoxycarbonyl, C3-C7cycloalkyl(C0-C4alkyl), C3-C7cycloalkyl(C0-C4alkoxy), mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C2-C4alkanoyl, and phenyl.

The invention also includes compounds and salts of Formula I and Formula II in which R7 is XRB, where X is absent, CH2—CH2—, —CH═CH—, or —C≡C—; RB is phenyl; and

    • (i) is chosen from halogen, hydroxy, amino, cyano, and nitro,
    • (ii) is chosen from C1-C4alkyl, C2-C4alkenyl, C1-C4alkoxy, mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C3-C7cycloalkyl(C0-C2alkyl), C3-C7cycloalkyl(C0-C2alkoxy), phenyl(C0-C2alkyl), phenyl(C0-C2alkoxy), pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, pyridyl, thienyl, C1-C4alkylthio, —(C═O)OR10, and —(C═O)NR10R11; and
    • (iii) is chosen from —ORD, —(C═O)RD, —SO2RD, —SO3RD, and —NR10SO2RD, where RD is C1-C4alkyl, C3-C7cycloalkyl(C0-C2alkyl), piperidinyl, piperazinyl, phenyl, naphthyl, and pyridyl.

Each of (ii) and (iii) in this embodiment is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, oxo, —COOH, —CONH2, C1-C4alkoxy, C1-C4alkoxycarbonyl, C3-C7cycloalkyl(C0-C2alkyl), mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C2-C4alkanoyl, and phenyl.

The invention further includes compounds and salts of Formula I and Formula II in which R7 is XRB and X is absent, CH2—CH2—, —CH═CH—, or —C≡C-. RB is phenyl; substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, C1-C4alkyl, C2-C4alkenyl, C1-C4alkoxy, mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C3-C7cycloalkyl(C0-C2alkyl), C3-C7cycloalkyl(C0-C2alkoxy), phenyl(C0-C2alkyl), phenyl(C0-C2alkoxy), pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, pyridyl, thienyl, C1-C4alkylthio, —(C═O)OR10, and —(C═O)NR10R11; and

RB is substituted with 1 or 2 substituents independently chosen from —ORD, —(C═O)RD, —SO2RD, —SO3RD, and —NR10SO2RD, where RD is C1-C4alkyl, piperidinyl, phenyl, naphthyl, or pyridyl; and each RD is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, C1-C2alkyl, C1-C2alkoxy, mono- and di-(C1-C2)alkylamino, C1-C2haloalkyl, and C1-C2haloalkoxy.

The invention includes compounds and salts of Formula I and Formula II in which R7 is XRB and X is absent.

The invention pertains to certain compounds of Formula I and Formula II in which R7 is XRB and RB is phenyl substituted with 1 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, C1-C4alkyl, C1-C4alkoxy, mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, and C1-C2haloalkoxy.

Certain embodiments of the invention also pertain to compounds and salts of Formula I and Formula II in which R7 is XRB and RB is phenyl, substituted with 1 or 2 substituents independently chosen from halogen, hydroxy, amino, C1-C2alkyl, and C1-C2alkoxy.

Other embodiments of the invention pertain to compounds and salts of Formula I and Formula II in which phenyl substituted with 1 or 2 substituents independently chosen from fluoro, amino, hydroxy, cyano, and methyl.

In other embodiments the invention provides compounds and salts of Formula I and Formula II in which

R7 is XRD where RD is phenyl fused to a 5- or 6-membered heterocycloalkyl ring containing 1 or 2 nitrogen or oxygen atoms, where RD is substituted with 0 to 3 substitutents independently chosen from (i), (ii), and (iii).

    • (i) is chosen from halogen, hydroxy, amino, cyano, and nitro,
    • (ii) is chosen from C1-C4alkyl, C2-C4alkenyl, C1-C4alkoxy, mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C3-C7cycloalkyl(C0-C2alkyl), C3-C7cycloalkyl(C0-C2alkoxy), phenyl(C0-C2alkyl), phenyl(C0-C2alkoxy), pyrrolidinyl(C0-C2alkyl), piperidinyl(C0-C2alkyl), piperazinyl(C0-C2alkyl), morpholinyl(C0-C2alkyl), thiomorpholinyl(C0-C2alkyl), pyridyl, pyrimidinyl, pyrazinyl, furanyl, benzofuranyl, pyrrolyl, pyrazolyl, imidazolyl, thienyl, C1-C4alkylthio, ═NOR10, —(C0-C4alkyl)(C═O)R10, —(C0-C4alkyl)O(C═O)R10, —(C0-C4alkyl)(C═O)NR10R11, —(C0-C4alkyl)O(C═O)NR10R11, —(C0-C4alkyl) (C═O)OR10, —(C0-C4alkyl)NR10(C═O)R11, —(C0-C4alkyl)NR10(C═O)OR11, —(C0-C4alkyl)NR10(C═O)NR11R12, —(C0-C4alkyl)NR10(C═O)(C1-C4alkyl)NR11(C═O)O—R12, —(C0-Colkyl)NR10(C═S)NR11R12, —(C0-C4alkyl)NR10NR11R12, —(C0-C4alkyl)N═NR13, —(C0-C4alkyl)SO3R10, —(C0-C4alkyl)(S═O)OR10, —(C0-C4alkyl)SO2R13, —(C0-C4alkyl)SO2NR10R11, and —(C0-C4alkyl)NR10SO2R13; and
    • (iii) is chosen from —ORD, —(C═O)RD, —SO2RD, —SO3RD, —NR10SO2RD, where RD is C1-C4alkyl, C3-C7cycloalkyl(C0-C2alkyl), pyrrolidinyl(C0-C2alkyl), piperidinyl(C0-C2alkyl), piperazinyl(C0-C2alkyl), morpholinyl(C0-C2alkyl), thiomorpholinyl(C0-C2alkyl), phenyl(C0-C2alkyl), naphthyl(C0-C2alkyl), pyridyl(C0-C2alkyl), pyrimidinyl(C0-C2alkyl), pyrazinyl(C0-C2alkyl), furanyl(C0-C2alkyl), benz[b]thiophenyl(C0-C2alkyl), benzofuranyl(C0-C2alkyl), quinolinyl(C0-C2alkyl), isoquinolinyl(C0-C2alkyl), quinazolinyl(C0-C2alkyl), isoxazolyl(C0-C2alkyl), indolyl(C0-C2alkyl), dihydroindolyl(C0-C2alkyl), pyrrolyl(C0-C2alkyl), pyrazolyl(C0-C2alkyl), imidazolyl(C0-C2alkyl), thienyl(C0-C2alkyl), isoindolyl(C0-C2alkyl), or dihydroisoindolyl(C0-C2alkyl); where each of (ii) and (iii) is substituted with 0 to 3 substitutents independently chosen from halogen, hydroxy, amino, cyano, nitro, oxo, —COOH, —CONH2, C1-C4alkyl, C2-C4alkenyl, C2-C4alkynyl, C1-C4alkoxy, C1-C4alkoxycarbonyl, C3-C7cycloalkyl(C0-C4alkyl), C3-C7cycloalkyl(C0-C4alkoxy), mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C2-C4alkanoyl, and phenyl.

In certain embodiments provided herein R7 is XRD and X is absent.

In certain embodiments provided herein R7 is XRD and X is absent and RD is phenyl fused to a 5- or 6-membered heterocycloalkyl ring containing 1 or 2 nitrogen or oxygen atoms, where RD is substituted with 0 to 2 substitutents independently chosen from halogen, hydroxy, amino, C1-C2alkyl, and C1-C2alkoxy.

The A8 Variable

The invention includes compounds and salts of Formula I and Formula II in which A8 is nitrogen. Examples of such compounds include, but are not limited to, compounds of Formula III-Formula VI

The invention also includes compounds and salts of Formula I and Formula II in which A8 is CR8. Examples of such compounds include, but are not limited to, compounds of Formula VII-Formula X

The variables R2, R3, R4, R5, R6, R7, R8, and R9 shown in Formula III-Formula X carry any of the definitions set forth herein for these variables.

The invention includes compounds and salts of Formula I and Formula II in which R8 is hydrogen, halogen, C1-C2alkyl, C1-C2alkoxy, C1-C2haloalkyl, or C1-C2haloalkoxy.

Certain embodiments of the invention pertain to compounds and salts of Formula I and Formula II in which R8 is hydrogen or methoxy.

The R9 Variable

The invention includes compounds and salts of Formula I and Formula II in which R9 is C1-C4alkyl, cyclopropyl, or phenyl, each of which is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, C1-C2alkyl, C1-C2alkoxy, mono- and di-(C1-C2)alkylamino, C1-C2haloalkyl, and C1-C2haloalkoxy.

Other embodiments of the invention pertain to compounds and salts of Formula I and Formula II in which R9 is C1-C4alkyl or cyclopropyl, or R9 is phenyl substituted with 2 substituents chosen from halogen, hydroxy, amino, C1-C2alkyl, C1-C2alkoxy, mono- and di-(C1-C2)alkylamino, C1-C2haloalkyl, and C1-C2haloalkoxy.

Certain embodiments of the invention include compounds and salts of Formula I and Formula II in which R9 is ethyl, t-butyl, cyclopropyl or 2,4-difluorophenyl, and particularly include those compounds and salts in which R9 is cyclopropyl.

The invention includes compounds of Formula I and Formula II in which the variables A1, R2, R3, R5, R6, R7, A8 and R9 carry any combination of the definitions set forth for these variables above.

Certain compounds of Formula I and Formula II exhibit possess potent antibacterial, antifungal, and/or antiprotozoal activity. Particular compounds of the invention exhibit Minimum Inhibitory Concentrations (MIC) of 64 μg/ml or less against Staphyloccocus aureus and/or Eschericia coli in a standard assay for determining the MIC of a compound against these bacteria, such as the assay provided in Example 10 below. Preferred compounds of the Formula I and II exhibit MIC values of 10 μg/ml or less against Staphyloccocus aureus and/or Eschericia coli. More preferred compound of the Formula I and II exhibit MIC values of 4 μg/ml or less, or even more preferably 1 μg/ml or less, against Staphyloccocus aureus and/or Eschericia coli.

Certain compounds of Formula I and Formula II are selective antimicrobial agents; having the ability to kill or inhibit the growth or reproduction of microbial organisms, while having little or no effect on the cells of fish, amphibians, reptiles, birds, or mammals. The selectivity of compounds of Formula I and Formula II may be assessed by determining the CC50 (the concentration at which 50% of the cells are killed) for cultured cells of a higher animal, such as a fish, reptiles, amphibian, bird, or mammal. Certain compounds of the invention exhibit a CC50 of greater that 100 micromolar for mammalian cells. Certain compounds of the invention exhibit a CC50 of greater than 100 micromolar for cultured human hepatocytes, and also exhibit MIC values of 64 μg/ml or less, preferably 10 μg/ml or less, or more preferably 4 μg/ml or less, or still more preferably 1 μg/ml or less against Staphyloccocus aureus and/or Eschericia coli.

Without wishing to be bound to any particular theory it is believed that the antimicrobial properties of compounds of Formula I and Formula II are due to the ability to these compounds to inhibit the activity of microbial DNA gyrases while having little or no effect on the analogous enzyme, Topoisomerase II, present in higher organisms. Certain preferred compounds of the invention are 100-fold or more selective for bacterial DNA gyrases than for mammalian, particularly human, Topoisomerase II.

Synthetic Intermediates

The invention includes novel intermediates useful for the synthesis of antimicrobial compounds of Formula I and Formula II. Coupling reactions occur between R′—M and R″—Y in the presence of catalyst Q, where M is Li, Mg, B, Al, Si, Zn, Cu, Zr, or Sn; where Y is I, Br, Cl, —O(SO2)CF3, or —N2BF4; Q is Fe, Ni, Cu, Pd, or Rh, and R′ and R″ are the organic molecules to which M and Y are bound. In certain embodiments M is Boron, disubstituted with OH, OG, or G, where G is an optionally substituted straight, branched or cyclic alkyl group, or other suitable group; Y is Br, and where Q is Pd. A general review of this chemistry can be found in Tamao, K. and Miyaura, N. Topics in Current Chemistry 219: 1-9 (2002). A review of the use of coupling reagents in which M is Boron with a listing of potential boronates, palladium catalysts, and reaction conditions can be found in Miyaura, N. Topics in Current Chemistry 219: 11-59 (2002).

Thus the invention includes intermediates of Formula XI and Formula XII:

In which A1, A8, R2, R3, R5, R6, and R9 carry the definitions set forth above and Y is I, Br, Cl, —O(SO2)CF3, or —N2BF4. Here compounds of Formula XI and Formula XII play the role of R″—Y intermediate in the coupling reaction between R′—M and R″—Y. These intermediates are coupled to compounds of the Formula R′—M where R′ is a group R.

R is XRA where X is absent, CH2—CH2—, —CH═CH—, or —C≡C—, and RA is C3-C6alkyl, C4-C7cycloalkyl, C4-C7cycloalkenyl, a 5-6 membered saturated, partially unsaturated, or aromatic heterocylic group bound via a carbon atom when X is absent or —CH2—CH2—, or bound via a carbon or nitrogen atom when X is —CH═CH— or —C≡C— or a RA is a 7-10 membered bicyclic saturated, partially unsaturated, or aromatic heterocylic group bound via a carbon atom when X is absent or —CH2—CH2—, or bound via a carbon or nitrogen atom when X is —CH═CH— or —C≡C—; each of which RA is substituted with 0 to 5 substituents independently chosen from (i), (ii), and (iii); or

R is XRB, where RB is phenyl substituted with 1 to 5 substituents independently chosen from (i), (ii), and (iii); or

R is XRC, where RC is cyclopropyl with 0 to 5 substituents independently chosen from (i), (ii), and (iii), with the proviso that Rc is not substituted with amino, or mono- or di-(C1-C4)alkylamino.

Wherein (i) is chosen from halogen, hydroxy, amino, cyano, and nitro,

    • (ii) is chosen from C1-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, C3-C7cycloalkyl(C0-C4alkyl), C3-C7cycloalkyl(C0-C4alkoxy), C4-C7cycloalkenyl(C0-C4alkyl), aryl(C0-C6carbohydryl), aryl(C1-C4alkoxy), C2-C6heterocycloalkyl(C0-C4alkyl), heteroaryl(C0-C6carbohydryl), C1-C6alkylthio, -(C0-C4alkyl)O(C═O)R10, —(C0-C4alkyl)(C═O)NR10R11, —(C0-C4alkyl)O(C═O)NR10R11, —(C0-C4alkyl) (C═O)OR10, —(C0-C4alkyl)NR10(C═O)R11, —(C0-C4alkyl)NR10(C═O)OR11, —(C0-C4alkyl)NR10(C═O)NR11R12, —(C0-C4alkyl)NR10(C═S)NR11R12, —(C0-C4alkyl)NR10NR11R12, —(C0-C4alkyl)N═NR13, —(C0-C4alkyl)SO3R10, —(C0-C4alkyl)(S═O)OR10, —(C0-C4alkyl)SO2R13, —(C0-C4alkyl)SO2NR10R11, and —(C0-C4alkyl)NR10SO2R13; and
    • (iii) is chosen from —ORD, —(C═O)RD, —SO2RD, —SO3RD, —NR10SO2RD, where RD is C1-C4alkyl, C3-C7cycloalkyl(C0-C2alkyl), C2-C6heterocycloalkyl(C0-C2alkyl), aryl(C0-C2alkyl), or heteroaryl(C0-C2alkyl); where each of (ii) and (iii) is substituted with 0 to 3 substituents independently chosen from halogen, hydroxy, amino, cyano, nitro, —COOH, —(C═O)OCH3, —CONH2, C1-C4alkyl, C2-C4alkenyl, C2-C4alkynyl, C1-C4alkoxy, C3-C7cycloalkyl(C0-C4alkyl), C3-C7cycloalkyl(C0-C4alkoxy), mono- and di-(C1-C4)alkylamino, C1-C2haloalkyl, C1-C2haloalkoxy, and C2-C4alkanoyl.

M is Li, Mg, B, Al, Si, Zn, Cu, Zr, or Sn; or M is Boron, disubstituted with OH, OG, or G, where G is an optionally substituted straight, branched, or cyclic alkyl group, an optionally substituted aryl or arylalkyl group, or other suitable group.

The invention also includes intermediates of Formula XIII and Formula XIV in which

In which A1, A8, R2, R3, R5, R6, and R9\ carry the definitions set forth above and M Li, Mg, B, Al, Si, Zn, Cu, Zr, or Sn; or M is Boron, disubstituted with OH, OG, or G, where G is an optionally substituted straight, branched, or cyclic alkyl group, an optionally substituted aryl or arylalkyl group, or other suitable group. Here compounds of Formula XIII and Formula XIV play the role of R′—M in the coupling reaction. These intermediates are coupled to compounds of the Formula R″—Y where Y is I, Br, Cl, —O(SO2)CF3, or —N2BF4 and R″ carries the definition set forth above for R.

Pharmaceutical Preparations

Compounds and salts of Formula I and Formula II can be administered as the neat chemical, but are preferably administered as a pharmaceutical composition or formulation. Accordingly, the invention provides pharmaceutical formulations comprising a compound or pharmaceutically acceptable salt of Formula I or Formula II, together with one or more pharmaceutically acceptable carrier, excipients, adjuvant, diluent, excipient, or other ingredient.

Compounds of general Formula I and Formula II may be administered orally, topically, parenterally, by inhalation or spray, sublingually, transdermally, via buccal administration, rectally, as an ophthalmic solution, or by other means, in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, excipients, adjuvants, and vehicles.

A pharmaceutical composition comprising a compound or salt of Formula I or Formula II wherein the composition is formulated as an injectable fluid, an aerosol, a cream, a gel, a pill, a capsule, a tablet, a syrup, a transdermal patch, or an ophthalmic solution is provided herein.

In addition to the subject compound, the compositions of the invention may contain a pharmaceutically acceptable carrier, one or more compatible solid or liquid filler diluents or encapsulating substances, which are suitable for administration to an animal. Carriers must be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration to the animal being treated. The carrier can be inert or it can possess pharmaceutical benefits of its own. The amount of carrier employed in conjunction with the compound is sufficient to provide a practical quantity of material for administration per unit dose of the compound.

Exemplary pharmaceutically acceptable carriers or components thereof are sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and methyl cellulose; powdered tragacanth; malt; gelatin; talc; solid lubricants, such as stearic acid and magnesium stearate; calcium sulfate; vegetable oils, such as peanut oil, cottonseed oil, sesame oil, olive oil, and corn oil; polyols such as propylene glycol, glycerine, sorbitol, mannitol, and polyethylene glycol; alginic acid; emulsifiers, such as the TWEENS; wetting agents, such sodium lauryl sulfate; coloring agents; flavoring agents; tableting agents, stabilizers; antioxidants; preservatives; pyrogen-free water; isotonic saline; and phosphate buffer solutions.

In particular, pharmaceutically acceptable carriers for systemic administration include sugars, starches, cellulose and its derivatives, malt, gelatin, talc, calcium sulfate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffer solutions, emulsifiers, isotonic saline, and pyrogen-free water. Preferred carriers for parenteral administration include propylene glycol, ethyl oleate, pyrrolidone, ethanol, and sesame oil.

Optional active agents may be included in a pharmaceutical composition, which do not substantially interfere with the activity of the compound of the present invention.

Effective concentrations of one or more of the compounds of the invention including pharmaceutically acceptable salts, esters or other derivatives thereof, are mixed with one or more suitable pharmaceutical carrier, excipient, adjuvant, or vehicle. In instances in which the compounds exhibit insufficient solubility, methods for solubilizing compounds may be used. Such methods are known to those of skill in this art, and include, but are not limited to using cosolvents, such as dimethylsulfoxide (DMSO), using surfactants, such as Tween, or dissolution in aqueous sodium bicarbonate. Derivatives of the compounds, such as salts of the compounds or prodrugs of the compounds may also be used in formulating effective pharmaceutical compositions.

Upon mixing or addition of the compound(s) of Formula I and/or Formula II, the resulting mixture may be a solution, suspension, emulsion or the like. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the chosen carrier or vehicle. The effective concentration sufficient for ameliorating the symptoms of the disease, disorder or condition treated and may be empirically determined.

The pharmaceutical compositions containing compounds of general Formula I and/or Formula II may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents, such as sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide pharmaceutically elegant and palatable preparations.

Oral formulations contain between 0.1 and 99% of a compound of the invention and usually at least about 5% (weight %) of a compound of the present invention. Some embodiments contain from about 25% to about 50% or from 5% to 75% of a compound of invention.

Liquids Formulations

Compounds of the invention can be incorporated into oral liquid preparations such as aqueous or oily suspensions, solutions, emulsions, syrups, or elixirs, for example. Moreover, formulations containing these compounds can be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations can contain conventional additives, such as suspending agents (e.g., sorbitol syrup, methyl cellulose, glucose/sugar, syrup, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminum stearate gel, and hydrogenated edible fats), emulsifying agents (e.g., lecithin, sorbitan monsoleate, or acacia), non-aqueous vehicles, which can include edible oils (e.g., almond oil, fractionated coconut oil, silyl esters, propylene glycol and ethyl alcohol), and preservatives (e.g., methyl or propyl p-hydroxybenzoate and sorbic acid).

Orally administered compositions also include liquid solutions, emulsions, suspensions, powders, granules, elixirs, tinctures, syrups, and the like. The pharmaceutically acceptable carriers suitable for preparation of such compositions are well known in the art. Oral formulations may contain preservatives, flavoring agents, sweetening agents, such as sucrose or saccharin, taste-masking agents, and coloring agents.

Typical components of carriers for syrups, elixirs, emulsions and suspensions include ethanol, glycerol, propylene glycol, polyethylene glycol, liquid sucrose, sorbitol and water. Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent.

Suspensions

For a suspension, typical suspending agents include methylcellulose, sodium carboxymethyl cellulose, AVICEL RC-591, tragacanth and sodium alginate; typical wetting agents include lecithin and polysorbate 80; and typical preservatives include methyl paraben and sodium benzoate.

Aqueous suspensions contain the active material(s) in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents; naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol substitute, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan substitute. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n- propyl p-hydroxybenzoate.

Oily suspensions may be formulated by suspending the active ingredients in a vegetable oil, for example peanut oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide palatable oral preparations. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.

Emulsions

Pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, for example olive oil or peanut oil, or a mineral oil, for example liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monoleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monoleate.

Dispersible Powders

Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above.

Tablets and Capsules

Tablets typically comprise conventional pharmaceutically compatible adjuvants as inert diluents, such as calcium carbonate, sodium carbonate, mannitol, lactose and cellulose; binders such as starch, gelatin and sucrose; disintegrants such as starch, alginic acid and croscarmelose; lubricants such as magnesium stearate, stearic acid and talc. Glidants such as silicon dioxide can be used to improve flow characteristics of the powder mixture. Coloring agents, such as the FD&C dyes, can be added for appearance. Sweeteners and flavoring agents, such as aspartame, saccharin, menthol, peppermint, and fruit flavors, are useful adjuvants for chewable tablets. Capsules (including time release and sustained release formulations) typically comprise one or more solid diluents disclosed above. The selection of carrier components often depends on secondary considerations like taste, cost, and shelf stability.

Such compositions may also be coated by conventional methods, typically with pH or time-dependent coatings, such that the subject compound is released in the gastrointestinal tract in the vicinity of the desired topical application, or at various times to extend the desired action. Such dosage forms typically include, but are not limited to, one or more of cellulose acetate phthalate, polyvinylacetate phthalate, hydroxypropyl methylcellulose phthalate, ethyl cellulose, Eudragit coatings, waxes and shellac.

Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.

Injectable and Parenteral Formulations

Pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above. The sterile injectable preparation may also be sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Acceptable vehicles and solvents include water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are useful in the preparation of injectables.

Compounds of Formula I and Formula II may be administered parenterally in a sterile medium. Parenteral administration includes subcutaneous injections, intravenous, intramuscular, intrathecal injection, or infusion techniques. The drug, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle. Advantageously, adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle. In compositions for parenteral administration the carrier comprises at least about 90% by weight of the total composition.

Suppositories

Compounds of Formula I and Formula II may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are cocoa butter and polyethylene glycols.

Topical Formulations

Compounds of the invention may be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application. Topical compositions of the present invention may be in any form including, for example, solutions, creams, ointments, gels, lotions, milks, cleansers, moisturizers, sprays, skin patches, and the like.

Such solutions may be formulated as 0.01% -10% isotonic solutions, pH about 5-7, with appropriate salts. Compounds of the invention may also be formulated for transdermal administration as a transdermal patch.

Topical compositions containing the active compound can be admixed with a variety of carrier materials well known in the art, such as, for example, water, alcohols, aloe vera gel, allantoin, glycerine, vitamin A and E oils, mineral oil, propylene glycol, PPG-2 myristyl propionate, and the like.

Other materials suitable for use in topical carriers include, for example, emollients, solvents, humectants, thickeners and powders. Examples of each of these types of materials, which can be used singly or as mixtures of one or more materials, are as follows:

Emollients, such as stearyl alcohol, glyceryl monoricinoleate, glyceryl monostearate, propane-1,2-diol, butane-1,3-diol, mink oil, cetyl alcohol, iso-propyl isostearate, stearic acid, iso-butyl palmitate, isocetyl stearate, ( )ey' alcohol, isopropyl laurate, hexyl laurate, decyl oleate, octadecan-2-ol, isocetyl alcohol, cetyl palmitate, dimethylpolysiloxane, di-n-butyl sebacate, iso-propyl myristate, iso-propyl palmitate, iso-propyl stearate, butyl stearate, polyethylene glycol, triethylene glycol, lanolin, sesame oil, coconut oil, arachis oil, castor oil, acetylated lanolin alcohols, petroleum, mineral oil, butyl myristate, isostearic acid, palmitic acid, isopropyl linoleate, lauryl lactate, myristyl lactate, decyl oleate, and myristyl myristate; propellants, such as propane, butane, iso-butane, dimethyl ether, carbon dioxide, and nitrous oxide; solvents, such as ethyl alcohol, methylene chloride, iso-propanol, castor oil, ethylene glycol monoethyl ether, diethylene glycol monobutyl ether, diethylene glycol monoethyl ether, dimethyl sulphoxide, dimethyl formamide, tetrahydrofuran; humectants, such as glycerin, sorbitol, sodium 2-pyrrolidone-5-carboxylate, soluble collagen, dibutyl phthalate, and gelatin; and powders, such as chalk, talc, fullers earth, kaolin, starch, gums, colloidal silicon dioxide, sodium polyacrylate, tetra alkyl ammonium smectites, trialkyl aryl ammonium smectites, chemically modified magnesium aluminium silicate, organically modified montmorillonite clay, hydrated aluminium silicate, fumed silica, carboxyvinyl polymer, sodium carboxymethyl cellulose, and ethylene glycol monostearate.

The compounds of the invention may also be topically administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.

Other Formulations

Other compositions useful for attaining systemic delivery of the subject compounds include sublingual, buccal and nasal dosage forms. Such compositions typically comprise one or more of soluble filler substances such as sucrose, sorbitol and mannitol, and binders such as acacia, microcrystalline cellulose, carboxymethyl cellulose and hydroxypropyl methylcellulose. Glidants, lubricants, sweeteners, colorants, antioxidants and flavoring agents disclosed above may also be included.

Compositions for inhalation typically can be provided in the form of a solution, suspension or emulsion that can be administered as a dry powder or in the form of an aerosol using a conventional propellant (e.g., dichlorodifluoromethane or trichlorofluoromethane).

Additional Components

The compositions of the present invention may also optionally comprise an activity enhancer. The activity enhancer can be chosen from a wide variety of molecules that function in different ways to enhance antimicrobial effects of compounds of the present invention. Particular classes of activity enhancers include skin penetration enhancers and absorption enhancers.

Pharmaceutical compositions of the invention may also contain additional active agents chosen from a wide variety of molecules, which can function in different ways to enhance the antimicrobial or therapeutic effects of a compound of the present invention. These optional other active agents, when present, are typically employed in the compositions of the invention at a level ranging from about 0.01% to about 15%. Some embodiments contain from about 0.1% to about 10% by weight of the composition. Other embodiments contain from about 0.5% to about 5% by weight of the composition.

Packaged Formulations

The invention includes packaged pharmaceutical formulations. Such packaged formulations include a pharmaceutical composition containing one or more compounds or salts of Formula I or Formula II in a container and optionally include instructions for using the composition to treat an animal (typically a human patient) suffering from a microorganism infection or prevent a microorganism infection in an animal. In certain embodiments the instructions are instructions for using the composition to treat a patient suffering from a bacterial infection.

In all of the foregoing embodiments the compound of the invention can be administered alone or as mixtures, and the compositions may further include additional drugs or excipients as appropriate for the indication.

Methods of Treatment

The invention includes methods of preventing and treating microorganism infections, particularly bacterial and protozoal infections, by administering a therapeutically effective amount of one or more compounds of Formula I and of Formula II to an animal at risk for a microorganism infection or suffering from a microorganism infection. The animal may be a fish, amphibian, reptile or bird, but is preferably a mammal. Methods of treating and preventing microorganism infections in livestock animals, companion animals, and human patients are particularly preferred.

The compounds disclosed herein are useful for preventing and treating bacterial infections in animals. Furthermore compounds of the invention may be used to treat a variety of conditions not attributed to bacterial infections. These include diseases and disorders caused fungal infections, mycoplasma infections, protozoal infections, or other conditions involving infectious organisms.

In some circumstances an effective amount of a compound of Formula I or Formula II may be an amount sufficient to reduce the symptoms of the microorganism infection. Alternatively an effective amount of a Compound of Formula I may be an amount sufficient to significantly reduce the amount of microorganism or antibodies against the detectable in a patient's tissues or bodily fluids.

Methods of treatment also include inhibiting microorganism replication in vivo, in an animal at risk for a microorganism infection or suffering from such an infection, by administering a sufficient concentration of a compound of Formula I or Formula II to inhibit bacterial survival in vitro. By “sufficient concentration” of a compound administered to the patient is meant the concentration of the compound available in the animal's system to prevent or combat the infection. Such a concentration by be ascertained experimentally, for example by assaying blood concentration of the compound, or theoretically, by calculating bioavailability. The amount of a compound sufficient to inhibit bacterial survival in vitro may be determined with a conventional assay for bacterial survival such as the Minimum Inhibitory Concentration (MIC) Assay disclosed in Example 10, which follows.

The invention also includes using compounds of Formula I and Formula I in prophylactic therapies. In the context of prophylactic or preventative treatment an effective amount of a compound of the invention is an amount sufficient to significantly decrease the treated animal's risk of contracting a microorganism infection.

Compounds of the invention are particularly useful for treating and preventing infectious disorders. These include for example: ocular infections such as conjunctivitis; urinary tract and genital infections, such as complicated urinary tract infections, acute urinary tract and genital infections, such as pyelonephritis, cervical gonococcal infections, cystitis, urethral chlamydial infections, cervical chlamydial infections, urethral gonococcal infections, and prostatitis, respiratory infections, such as lower respiratory tract infections, acute sinusitis, acute exacerbations of chronic bronchitis, community-acquired pneumonia, and nosocomial pneumonia, skin infections, such as skin-structure infections, impetigo, folliculitis, boils, scalded skin syndrome, and cellulites, and other infections such as bone infections, joint infections, infectious diarrhea, typhoid fever, intra-abdominal infections, gynecologic infections, including toxic shock syndrome, pelvic infections, and post-surgical infections.

The disclosed compounds are useful for treating infections caused by the following microorganisms:

Aerobic Gram-positive Microorganisms: Including but not limited to Enterococcus faecalis, Enterococcus faecium, Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus saprophyticus, Streptococcus pneumoniae, Streptococcus pyogenes, Staphylococcus haemolyticus, and Staphylococcus hominis.

Aerobic Gram-negative Microorganisms: Including but not limited to Campylobacter jejuni, Citrobacter diversus, Citrobacter freundii, Enterobacter cloacae, Escherichia coli, Haemophilus influenzae, Haemophilus parainfluenzae, Klebsiella pneumoniae, Moraxella catarrhalis, Morganella morganii, Neisseria gonorrhoeae, Proteus mirabilis, Proteus vulgaris, Providencia rettgeri, Providencia stuartii, Pseudomonas aeruginosa, Stenotrophomonas maltophila, Salmonella typhi, Serratia marcescens, Shigella boydii, Shigella dysenteriae, Shigella flexneri, Shigella sonnei. Acinetobacter Iwoffi, Aeromonas hydrophila, Edwardsiella tarda, Enterobacter aerogenes, Klebsiella oxytoca, Legionella pneumophila, Pasteurella multocida, Salmonella enteritidis, Vibrio cholerae, Vibrio parahaemolyticus, Vibrio vulnificus, Yersinia enterocolitica, and H. Pylorii.

Non-bacterial microorganisms: Mycoplasma, Legionella and Chlamydia.

Dosage levels of the order of from about 0.1 mg to about 140 mg per kilogram of body weight per day are useful in the treatment of the above-indicated conditions (about 0.5 mg to about 7 g per patient per day). The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Dosage unit forms will generally contain between from about 1 mg to about 500 mg of an active ingredient.

Frequency of dosage may also vary depending on the compound used and the particular disease treated. However, for treatment of most infectious disorders, a dosage regimen of 4 times daily or less is preferred and a dosage regimen of 1 or 2 times daily is particularly preferred.

It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy.

Combination Administration

The compounds of the invention may also be useful in combination with other pharmaceutically active agents such as antibacterial agents, antiviral agents, antifungal agents, anti-inflammatories, interferon, efflux-pump inhibitors, and beta-lactamase inhibitors. Antibiotic agents include any molecule that tends to prevent, inhibit or destroy life and as such, includes anti-bacterial agents, anti-fungicides, anti-viral agents, and anti-parasitic agents.

A composition comprising a compound or salt of Formula I or Formula I in combination with another one or more antibacterial agent, antiprotozoal agent, antifungal agent, antiviral agent, interferon, efflux-pump inhibitor, or beta-lactamase inhibitor is provided herein.

Pharmaceutical compositions of the invention include single dosage forms containing of a compound of Formula I and/or Formula II and one or more other active agent, dosage forms containing more than one compound of Formula I and/or Formula II, and separate administration of a compound of Formula I and/or Formula II with another active agent.

The following active agents, which are useful in combinations of the invention, may be isolated from an organism that produces the agent or synthesized by methods known to those of ordinary skill in the art of medicinal chemistry or purchased from a commercial source.

Anti-bacterial antibiotic agents include, but are not limited to, penicillins, cephalosporins, carbacephems, cephamycins, carbapenems, monobactams, aminoglycosides, glycopeptides, quinolones, tetracyclines, macrolides, and fluoroquinolones (see Table below). Examples of antibiotic agents include, but are not limited to, Penicillin G (CAS Registry No.: 61-33-6); Methicillin (CAS Registry No.: 61-32-5); Nafcillin (CAS Registry No.: 147-52-4); Oxacillin (CAS Registry No.: 66-79-5); Cloxacillin (CAS Registry No.: 61-72-3); Dicloxacillin (CAS Registry No.: 3116-76-5); Ampicillin (CAS Registry No.: 69-53-4); Amoxicillin (CAS Registry No.: 26787-78-0); Ticarcillin (CAS Registry No.: 34787-01-4); Carbenicillin (CAS Registry No.: 4697-36-3); Mezlocillin (CAS Registry No.: 51481-65-3); Azlocillin (CAS Registry No.: 37091-66-0); Piperacillin (CAS Registry No.: 61477-96-1); Imipenem (CAS Registry No.: 74431-23-5); Aztreonam (CAS Registry No.: 78110-38-0); Cephalothin (CAS Registry No.: 153-61-7); Cefazolin (CAS Registry No.: 25953-19-9); Cefaclor (CAS Registry No.: 70356-03-5); Cefamandole formate sodium (CAS Registry No.: 42540-40-9); Cefoxitin (CAS Registry No.: 35607-66-0); Cefuroxime (CAS Registry No.: 55268-75-2); Cefonicid (CAS Registry No.: 61270-58-4); Cefmetazole (CAS Registry No.: 56796-20-4); Cefotetan (CAS Registry No.: 69712-56-7); Cefprozil (CAS Registry No.: 92665-29-7); Loracarbef (CAS Registry No.: 121961-22-6); Cefetamet (CAS Registry No.: 65052-63-3); Cefoperazone (CAS Registry No.: 62893-19-0); Cefotaxime (CAS Registry No.: 63527-52-6); Ceftizoxime (CAS Registry No.: 68401-81-0); Ceftriaxone (CAS Registry No.: 73384-59-5); Ceftazidime (CAS Registry No.: 72558-82-8); Cefepime (CAS Registry No.: 88040-23-7); Cefixime (CAS Registry No.: 79350-37-1); Cefpodoxime (CAS Registry No.: 80210-62-4); Cefsulodin (CAS Registry No.: 62587-73-9); Fleroxacin (CAS Registry No.: 79660-72-3); Nalidixic acid (CAS Registry No.: 389-08-2); Norfloxacin (CAS Registry No.: 70458-96-7); Ciprofloxacin (CAS Registry No.: 85721-33-1); Ofloxacin (CAS Registry No.: 82419-36-1); Enoxacin (CAS Registry No.: 74011-58-8); Lomefloxacin (CAS Registry No.: 98079-51-7); Cinoxacin (CAS Registry No.: 28657-80-9); Doxycycline (CAS Registry No.: 564-25-0); Minocycline (CAS Registry No.: 10118-90-8); Tetracycline (CAS Registry No.: 60-54-8); Amikacin (CAS Registry No.: 37517-28-5); Gentamicin (CAS Registry No.: 1403-66-3); Kanamycin (CAS Registry No.: 8063-07-8); Netilmicin (CAS Registry No.: 56391-56-1); Tobramycin (CAS Registry No.: 32986-56-4); Streptomycin (CAS Registry No.: 57-92-1); Azithromycin (CAS Registry No.: 83905-01-5); Clarithromycin (CAS Registry No.: 81103-11-9); Erythromycin (CAS Registry No.: 114-07-8); Erythromycin estolate (CAS Registry No.: 3521-62-8); Erythromycin ethyl succinate (CAS Registry No.: 41342-53-4); Erythromycin glucoheptonate (CAS Registry No.: 23067-13-2); Erythromycin lactobionate (CAS Registry No.: 3847-29-8); Erythromycin stearate (CAS Registry No.: 643-22-1); Vancomycin (CAS Registry No.: 1404-90-6); Teicoplanin (CAS Registry No.: 61036-64-4); Chloramphenicol (CAS Registry No.: 56-75-7); Clindamycin (CAS Registry No.: 18323-44-9); Trimethoprim (CAS Registry No.: 738-70-5); Sulfamethoxazole (CAS Registry No.: 723-46-6); Nitrofurantoin (CAS Registry No.: 67-20-9); Rifampin (CAS Registry No.: 13292-46-1); Mupirocin (CAS Registry No.: 12650-69-0); Metronidazole (CAS Registry No.: 443-48-1); Cephalexin (CAS Registry No.: 15686-71-2); Roxithromycin (CAS Registry No.: 80214-83-1); Co-amoxiclavuanate; combinations of Piperacillin and Tazobactam; and their various salts, acids, bases, and other derivatives.

Anti-fungals agents include but are not limited to Amphotericin B, Candicidin, Dermostatin, Filipin, Fungichromin, Hachimycin, Hamycin, Lucensomycin, Mepartricin, Natamycin, Nystatin, Pecilocin, Perimycin, Azaserine, Griseofulvin, Oligomycins, Neomycin, Pyrrolnitrin, Siccanin, Tubercidin, Viridin, Butenafine, Naftifine, Terbinafine, Bifonazole, Butoconazole, Chlordantoin, Chlormidazole, Cloconazole, Clotrimazole, Econazole, Enilconazole, Fenticonazole, Flutrimazole, Isoconazole, Ketoconazole, Lanoconazole, Miconazole, Omoconazole, Oxiconazole, Sertaconazole, Sulconazole, Tioconazole, Tolciclate, Tolindate, Tolnaftate, Fluconawle, Itraconazole, Saperconazole, Terconazole, Acrisorcin, Amorolfine, Biphenamine, Bromosalicylchloranilide, Buclosamide, Calcium Propionate, Chlorphenesin, Ciclopirox, Cloxyquin, Coparaffinate, Diamthazole, Exalamide, Flucytosine, Halethazole, Hexetidine, Loflucarban, Nifuratel, Potassium Iodide, Propionic Acid, Pyrithione, Salicylanilide, Sodium Propionate, Sulbentine, Tenonitrozole, Triacetin, Ujothion, Undecylenic Acid, and Zinc Propionate.

Antiviral agents include, but are not limited to, Acyclovir, Cidofovir, Cytarabine, Dideoxyadenosine, Didanosine, Edoxudine, Famciclovir, Floxuridine, Ganciclovir, Idoxuridine, Inosine Pranobex, Lamivudine, MADU, Penciclovir, Sorivudine, Stavudine, Trifluridine, Valacyclovir, Vidarabine, ZaIcitabine, Zidovudine, Acemannan, Acetylleucine, Amantadine, Amidinomycin, Delavirdine, Foscarnet, Indinavir, Interferon-alpha, Interferon-beta, Interferon-gamma, Kethoxal, Lysozyme, Methisazone, Moroxydine, Nevirapine, Podophyllotoxin, Ribavirin, Rimantadine, Ritonavir2, Saquinavir, Stailimycin, Statolon, Tromantadine, and Xenazoic Acid.

Antiinflammatory agents include, but are not limited to, Enfenamic Acid, Etofenamate, FIufenamic Acid, Isonixin, Meclofenamic Acid, Mefenamic Acid, Niflumic Acid, Talniflumate, Terofenamate, Tolfenamic Acid, Aceclofenac, Acemetacin, Alclofenac, Amfenac, Amtolmetin Guacil, Bromfenac, Bufexamac, Cinmetacin, Clopirac, Diclofenac, Etodolac, Felbinac, Fenclozic Acid, Fentiazac, Glucametacin, Ibufenac, Indomethacin, Isofezolac, Isoxepac, Lonazolac, Metiazinic Acid, Mofezolac, Oxametacine, Pirazolac, Proglumetacin, Sulindac, Tiaramide, Tolmetin, Tropesin, Zomepirac, Bumadizon, Butibufen, Fenbufen, Xenbucin, Clidanac, Ketorolac, Tinoridine, Alminoprofen, Benoxaprofen, Bermoprofen, Bucloxic Acid, Carprofen, Fenoprofen, Flunoxaprofen, Flurbiprofen, Ibuprofen, Ibuproxam, Indoprofen, Ketoprofen, Loxoprofen, Naproxen, Oxaprozin, Piketoprofen, Pirprofen, Pranoprofen, Protizinic Acid, Suprofen, Tiaprofenic Acid, Ximoprofen, Zaltoprofen, Difenamizole, Epirizole, Apazone, Benzpiperylon, Feprazone, Mofebutazone, Morazone, Oxyphenbutazone, Phenylbutazone, Pipebuzone, Propyphenazone, Ramifenazone, Suxibuzone, Thiazolinobutazone, Acetaminosalol, Aspirin, Benorylate, Bromosaligenin, Calcium Acetylsalicylate, Diflunisal, Etersalate, Fendosal, Gentisic Acid, Glycol Salicylate, Imidazole Salicylate, Lysine Acetylsalicylate, Mesalamine, Morpholine Salicylate, I-Naphthyl Salicylate, Olsalazine, Parsalmide, Phenyl Acetylsalicylate, Phenyl Salicylate, Salacetamide, Salicylamide O-Acetic Acid, Salicylsulfuric Acid, Salsalate, Sulfasalazine, Ampiroxicam, Droxicam, Isoxicam, Lornoxicam, Piroxicam, Tenoxicam, epsilon-Acetamidocaproic Acid, S-Adenosylmethionine, 3-Amino-4-hydroxybutyric Acid, Amixetrine, Bendazac, Benzydamine, alpha-Bisabolol, Bucolome, Difenpiramide, Ditazol, Emorfazone, Fepradinol, Guaiazulene, Nabumetone, Nimesulide, Oxaceprol, Paranyline, Perisoxal, Proquazone, Superoxide Dismutase, Tenidap, Zileuton, 21-Acetoxypregnenolone, Alclometasone, Algestone, Amcinonide, Beclomethasone, Betamethasone, Budesonide, Chloroprednisone, Clobetasol, Clobetasone, Clocortolone, Cloprednol, Corticosterone, Cortisone, Cortivazol, Deflazacort, Desonide, Desoximetasone, Dexamethasone, Diflorasone, Diflucortolone, Difluprednate, Enoxolone, Fluazacort, Flucloronide, Flumethasone, Flunisolide, Fluocinolone Acetonide, Fluocinonide, Fluocortin Butyl, Fluocortolone, Fluorometholone, Fluperolone Acetate, Fluprednidene Acetate, Fluprednisolone, Flurandrenolide, Fluticasone Propionate, Formocortal, Halcinonide, Halobetasol Propionate, Halometasone, Halopredone Acetale, Hydrocortamate, Hydrocortisone, Loteprednol Etabonale, Mazipredone, Medrysone, Meprednisone, Methylprednisolone, Mometasone Furoate, Paramethasone, Prednicarbate, Prednisolone, Prednisolone 25-Diethylamino-acetate, Prednisolone Sodium Phosphate, Prednisone, Prednival, Prednylidene, Rimexolone, Tixocortol, Triamcinolone, Triamcinolone Acetonide, Triamcinolone Benetonide, and Triamcinolone Hexacetonide.

Compounds of the invention may be combined with one or more Beta lactamase inhibitor when used in combination with a beta-lactam class antibiotic, such as penicillin or cephalosporins. Beta-lactamase inhibitors include, but are not limited to Clavulanic acid, Sulbactam, Sultamacillin, and Tazobactam.

Compounds of the invention may also be combined with one or more efflux pump inhibitor, such as a quinazolinone efflux pump inhibitors, d-ornithine-d-homophenylalanine-3-aminoquinoline, Phe-Arg-b-naphthylamide, propafenone, a phenothiazine or thioxanthene efflux pump inhibitor, 1-aza-9-oxafluorenes, N-[4-[2-(3,4-dihydro-6,7-dimethoxy-2(1H)-isoquinolinyl)ethyl]phenyl]-9,10-dihydro-5-methoxy-9-oxo-4-Acridinecarboxamide, reserpine, Milbemycin, Cinchonine, Verapamil, L-phenylalanyl-N-2-naphthalenyl-L-Argininamide (and analogs), 5′-methoxyhydnocarpin-D, methylxanthines, FK506, a cyclosporine efflux pump inhibitor, Nocardamine and other siderophores, Amiodarone, Cyclosporin A, Ro11-2933 (DMDP), Quinidine, and the optical isomers of Propranolol, Quinine (SQl) and Quinidine, Quinine-10,11-epoxide, Quercetin, Amitriptyline, Taxuspine C derivatives, Emodin, MC-002434; Agosterol A; Pheophorbide; pyridoquinolines such as 2,2′-[(2,8,10-trimethylpyrido[3,2-g]quinoline-4,6-diyl)bis(oxy)]bis[N,N-dimethyl-ethanamine, Gitonavir, and Gemfibrozil.

Synthesis of Compounds

The compounds of the invention are prepared according to methods well-known to those skilled in the art of organic chemical synthesis. The starting materials used in preparing the compounds of the invention are known, made by known methods, or are commercially available.

It is recognized that the skilled artisan in the art of organic chemistry can readily carry out standard manipulations of organic compounds without further direction. Examples of such manipulations are discussed in standard texts such as J. March, Advanced Organic Chemistry, John Wiley & Sons, 1992.

The skilled artisan will readily appreciate that certain reactions are best carried out when other functionalities are masked or protected in the compound, thus increasing the yield of the reaction and/or avoiding any undesirable side reactions. Often, the skilled artisan utilizes protecting groups to accomplish such increased yields or to avoid the undesired reactions. These reactions are found in the literature and are also well within the scope of the skilled artisan. Examples of many such manipulations can be found in, for example, T. Greene, Protecting Groups in Organic Synthesis, John Wiley & Sons, 1981.

The compounds of the invention may have one or more chiral center. As a result, one may selectively prepare one optical isomer, including diastereomers and enantiomers, over another, for example by chiral starting materials, catalysts or solvents, or may prepare both stereoisomers or both optical isomers, including diastereomers and enantiomers at once (a racemic mixture). Since the compounds of the invention may exist as racemic mixtures, mixtures of optical isomers, including diastereomers and enantiomers, or stereoisomers may be separated using known methods, such as through the use of, for example, chiral salts and chiral chromatography.

In addition, it is recognized that one optical isomer, including a diastereomer and enantiomer, or a stereoisomer, may have favorable properties over the other. When a racemic mixture is discussed herein, it is clearly contemplated to include both optical isomers, including diastereomers and enantiomers, or one stereoisomer substantially free of the other.

The invention also includes also includes all energetically accessible conformational and torsional isomers of the compounds disclosed.

When the substituent R7 in a compound of Formula I or Formula II is attached via an unsaturated aliphatic group, for example when R7 is phenyl (C2C6alkenyl), all geometric isomers of the compound are included.

This invention is further illustrated by the following examples that should not be construed as limiting. The contents of all references, patents and published patent applications cited throughout this application are incorporated herein by reference.

EXAMPLES Abbreviations

The following abbreviations are used in the reaction schemes and synthetic examples, which follow. This list in not meant to be an all-inclusive list of abbreviations used in the application as additional standard abbreviations, which are readily understood by those skilled in the art of organic synthesis, may also be used in the synthetic schemes and examples

(Boc)2O—Di-t-butyl dicarbonate
n-BuLi—n-Butyl lithium

DMAP—4-Dimethylaminopyridine DMF—N,N-Dimethylformamide DMSO—Dimethylsulfoxide

EtOAc—Ethyl acetate

NBS—N-bromosuccinamide NCS—N-chlorosuccinamide Pd(PPh3)4—Tetrakis(triphenylphosphine)palladium(0)

PTLC—Preparative thin layer chromatography

THF—Tetrahydrofuran

TLC—Thin-layer chromatography

General Methods

All nonaqueous reactions are performed under an atmosphere of dry argon gas (99.99%) using oven- or flame-dried glassware. Microwave-assisted syntheses are conducted in a commercial microwave reactor (Discover System, CEM Corporation). The progress of reactions is monitored using thin-layer chromatography on glass plates coated with Merck silica gel 60 (F254). Flash column chromatography is performed on Merck silica gel 60 (230-400 mesh). Melting points are recorded on an Electrothermal Model IA9100 digital melting point apparatus; the reported values are the average of three measurements. NMR spectra are recorded at ambient temperature using a Bruker Avance 300 spectrometer (1H at 300.1 MHz, 13C at 75.5 MHz, and 19F at 282.4 MHz). The chemical shifts for 1 H and 13C are reported in parts per million (δ) relative to external tetramethylsilane and are referenced to signals of residual protons in the deuterated solvent. The chemical shifts for 19F are reported in parts per million (δ) relative to external fluorotrichloromethane. Assignment of 1H and 13C NMR data is based on extensive two-dimensional correlation experiments (1H-1H COSY, 1H-13C HMQC, 1H-13C HMBC, and 1H-1H NOESY) and the usual principles of NMR spectroscopy (the magnitudes of coupling constants and chemical shifts). Analytical HPLC is performed using a YMC Pack Pro C18 50×4.6 mm 5 μm column with an isocratic elution of 0.24 min at 90:10 H2O:CH3CN containing 0.1% TFA followed by a 4-min linear gradient elution from 90:10 to 10:90 at a flow rate of 2.5 mL/min with UV detection at 254 nm. Preparative HPLC is performed using a YMC Pack Pro C18 150×20.0 mm 5 μum column with an isocratic elution of 0.24 min at 97:3 H2O:CH3CN containing 0.1% TFA followed by a 10-min linear gradient elution from 97:3 to 0:100 at a flow rate of 18.0 mL/min with UV detection at 254 nm. Low-resolution mass spectra are recorded on a Thermo Finnigan Surveyor MSQ instrument (operating in APCI mode) equipped with a Gilson liquid chromatograph. Unless noted otherwise, the quasi-molecular ions, [M+H]+, observed in the low-resolution mass spectra are the base peaks. High-resolution mass spectrometric analyses (ESI using sodium iodide as internal standard) are performed at the W. M. Keck Foundation Biotechnology Resource Laboratory (Yale University, New Haven, Conn.); the reported exact masses are the average of five measurements. Elemental analysis is performed at Prevalere Life Sciences, Inc. (Whitesboro, N.Y.).

Example 1 General Method for Tthe Preparation of 9-cyclopropyl-6-fluoro-7-phenyl-9H-isothiazolo[5,4-B]quinoline-3,4-diones (9)

9-cyclopropyl-6-fluoro-7-phenyl-9H-isothiazolo[5,4-b]quinoline-3,4-diones (9) are prepared in accordance with the synthetic scheme set forth below.

STEP A. Preparation of 4-bromo-2,5-difluorobenzoic acid (2)

Freshly titrated n-butyl lithium (27.0 ml, 1.39 M in hexanes) is added slowly (over about 30 minutes) to a −78° C. solution of diethyl ether (90 ml) containing 1,4-dibromo-2,5-difluorobenzene (1, 10.22 g, 0.038 mol). The resulting yellow solution is stirred at −78° C. for 2 hours to give a yellow suspension. Several pellets (˜10) of dry ice are added to the suspension, which is then allowed to warm slowly to room temperature as it degasses (approximately 40 minutes). The resulting suspension is acidified with a 1 M aqueous solution of hydrochloric acid (500 ml), and the product extracted with diethyl ether (5×200 ml). The combined organics are washed with water (4×100 ml) and filtered. The ether solution is concentrated to approximately 200 ml under reduced pressure, and the product extracted into a saturated aqueous solution of sodium bicarbonate (3×200 ml). The combined aqueous extracts are washed with methylene chloride (3×100 ml) and acidified with hydrochloric acid. The product is extracted with diethyl ether (3×200 ml), and the combined organic extracts washed with water (2×200 ml), dried over magnesium sulfate, and concentrated under reduced pressure to give (2) as a pale yellow solid. 1H NMR (300 MHz, DMSO-d6): δ7.74 (dd, JH-F=8.5 Hz, 6.5 Hz, 1H), 7.84 (dd, JH-F=10.0 Hz, 5.5 Hz, 1H), 13.7 (br, 1H, CO2H). 19F{1H} NMR (282 MHz, DMSO-d6): δ-114.0 (d, JF-F=17.0 Hz, 1F), −113.6 (d, JF-F=17.0 Hz, 1F). 13C{1H} NMR (75 MHz, DMSO-d6): δ113.6 (dd, JC-F=23.5 Hz, 10.0 Hz), 118.4 (dd, JC-F=26.5 Hz, 2.5 Hz, CH), 120.0 (dd, JC-F=19.0 Hz, 12.0 Hz), 122.2 (d, JC-F=28.0 Hz, CH), 154.4 (dd, JC- F=245.0 Hz, 5.5 Hz, CF), 156.8 (dd, JC-F=251.5 Hz, 4.0 Hz, CF), 163.4 (m, CO2H).

STEP B. Preparation of 3-(4-bromo-2,5-difluoro-phenyl)-3-oxo-propionic acid ethyl ester (3)

4-Bromo-2,5-difluorobenzoyl chloride is prepared from 2 as described previously. [Reuman, M.; et. al, J. Med. Chem. (1995) 38, 2531-2540]. Note that the addition of dimethylformamide is omitted from this procedure. This intermediate is used to prepare 3 as described previously [Wierenga, W.; Skulnick, H. I. J. Org. Chem. 1979, 44, 310-311]. 1H NMR (300 MHz, CDCl3): (enol, major) δ1.32 (t, JH-H=7.0 Hz, 3H, CO2CH2CH3), 4.26 (q, JH-H=7.0 Hz, 2H, CO2CH2CH3), 5.85 (s, 1H, CH3C(OH)═CH—CO2CH2CH3), 7.34 (dd, JH-F=10.5 Hz, 5.5 Hz, 1H, aromatic), 7.64 (dd, JH-F=9.0 Hz, 6.5 Hz, 1H, aromatic), 12.65 (s, 1H, OH). 19F{1H} NMR (282 MHz, CDCl3): δ−114.8 (d, JF-F=17.0 Hz, 1F), −112.6 (d, JF-F=17.0 Hz, 1F). 3 (keto, minor): 1H NMR (300 MHz, CDCl3): δ1.24 (t, JH-H=7.0 Hz, 3H, CO2CH2CH3), 3.93 (d, JH-F=4.0 Hz, 2H, CH2CO2CH2CH3), 4.19 (q, JH-H=7.0 Hz, 2H, CO2CH2CH3), 7.40 (dd, JH-F=9.5 Hz, 5.5 Hz, 1H, aromatic), 7.68 (dd, JH-F=8.5 Hz, 6.0 Hz, 1H, aromatic). 19F{1H} NMR (282 MHz, CDCl3): δ−114.3 (d, JF-F=17.0 Hz, 1F), −111.7 (d, JF-F=17.0 Hz, 1F).

STEP C. Preparation of 2-(4-bromo-2,5-difluoro-benzoyl)-3-cyclopropylamino-3-methylsulfanyl-acrylic acid ethyl ester (4)

Cyclopropyl thioisocyanate (0.57 ml, 6.15 mmole, 1.7 equiv.) is added to a stirred solution of 3-(4-Bromo-2,5-difluoro-phenyl)-3-oxo-propionic acid ethyl ester (3, 1.06 g, 3.5 mmole) in DMF (anhydrous, 10 ml) under argon at room temperature. The reaction mixture is cooled in an ice bath and NaH (150 mg, 60% in mineral oil, 3.7 mmole, 1.07 equiv.) is added portionwise at 0-5° C. under argon. After addition, the reaction mixture is allowed to warm to room temperature and stirred at room temperature until TLC indicates there is no remaining starting material. CH3I (0.38 ml, 5.6 mmole, 1.7 equiv.) is then added to the reaction mixture. The reaction is diluted with EtOAc and quenched with NH4Cl solution after stirring at room temperature for about 4 hours (TLC and LC MS are used to determine reaction completion). The organics are washed with brine, dried over Na2SO4, and concentrated. The resulting crude oil (4, 1.6 g) is purified by column chromatography (Silica gel, 40% EtOAc in hexanes, gradient, 40 minutes) to yield 4 as a yellow oil. 1H NMR (CDCl3) δ: 7.15 (m, 2H), 3.89 (q, 2H), 2.96 (m, 1H), 2.48 (s, 3H), 0.85 (m, 7H).

STEP D. Preparation of 7-bromo-1-cyclopropyl-6-fluoro-2-methylfulfanyl-4-oxo-1,4-dihydro-quinoline-3 -carboxylic acid ethyl ester (5)

NaH (345 mg, 60% in mineral oil, 8.6 mmole, 1.05 equiv.) is added to a stirred solution of 2-(4-bromo-2,5-difluoro-benzyl)-3-cyclopropylamino-3-methylsulfanyl-acrylic acid ethyl ester (4, 3.46 g, 8.2 mmole) in DMF (anhydrous, 100 ml). The reaction mixture is stirred at 75° C. for 18 hours (TLC is used to indicate reaction completion). The reaction mixture is cooled, diluted with NH4Cl solution, and extracted with EtOAc. The organics are washed with brine (4×30 ml), dried over Na2SO4, and concentrated in vacuo to give 5 as a light yellow solid. This intermediate is used without further purification 1H NMR (CDCl3) indicated >98% purity). 1HNMR (CDCl3) δ: 8.09 (d, 1H), 7.90 (d, 1H), 4.35 (q, 2H), 3.22 (m, 1H), 2.49 (s, 3H), 1.3-1.5 (m, 7H).

STEP E. Preparation of 7-bromo-1-cyclopropyl-6-fluoro-2-mercapto-4-oxo-1,4-dihydroquinoline-3-carboxylic acid ethyl ester. (6)

Sodium hydrogen sulfide (5 mg, 0.09 mmole, 1.5 equiv.) is added to a stirred solution of 7-Bromo-1-cyclopropyl-6-fluoro-2-methylsulfanyl-4-oxo-1,4-dihydroquinoline-3-carboxylic acid ethyl ester (5, 24 mg, 0.06 mmole) in THF (tetrahydrofuran, 2 ml) under argon at room temperature. The reaction is then stirred at 45° C. until TLC indicated completion. The reaction mixture is diluted with water and washed with diethyl ether. The aqueous layer is acidified by 1N HCl to a pH of approximately 2, and extracted with EtOAc. The resulting organics are washed with brine, dried over Na2SO4, and concentrated in vacuo. The crude product is purified by PTLC (20% CH3OH in CHCl3) to give 6. Alternatively, sodium hydrogen sulfide (20 mg, 0.36 mmole, 1.5 equiv.) is added to a stirred solution of 5 (crude, 96 mg, 0.24 mmole) in DMF (6 ml) under argon at room temperature. The reaction is then stirred at 40° C. until TLC indicates completion. The reaction mixture is diluted with water, acidified by 1N HCl to a pH of approximately 2, and extracted with EtOAc. The resulting organics are washed with brine, dried over Na2SO4, and concentrated. The crude is purified by PTLC (20% CH3OH in CHCl3) to yield 6. 1H NMR (CDCl3) δ: 8.40 (d, 1H), 8.06 (d, 1H), 4.71 (q, 2H), 3.46 (m, 1H), 1.68 (m, 7H).

STEP F. Preparation of 7-bromo-9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4-B]quinoline-3,4-dione (7)

A mixture of NaHCO3 solution (51 mg, 0.9 ml water) and hydroxylamine-O-sulfonic acid (27 mg, 0.24 mmole, 4.2 equiv.) is added to a stirred solution of 7-Bromo-1-cyclopropyl-6-fluoro-2-mercapto-4-oxo-1,4-dihydroquinoline-3-carboxylic acid ethyl ester (6, 22 mg, 0.057 mmole) in THF (0.7 ml). The reaction mixture is stirred at room temperature for approximately 3 hours until the reaction is complete. The reaction mixture is acidified by the addition of 0.5N HCl and filtered. The resulting solid is washed with water (3×) and dried yielding 7 as a white solid. 1H NMR (DMSO-d6) δ: 8.39 (d, 1H), 8.06 (d, 1H), 3.63 (m, 1H), 1.41 (m, 2H), 1.26 (m, 2H). 19F NMR (DMSO-d6) δ: 114.9 (s, 1H).

STEP G. Preparation of tert-butyl 7-bromo-9-cyclopropyl-6-fluoro-3,4-dioxoisothiazolo[5,4-B]quinoline-2(3H,4H,9H)-carboxylate (8)

4-Dimethylaminopyridine (DMAP, catalytic amount) and (Boc)2O (27 mg, 2 equivalents, 0.12 mmole) are added to a stirred solution of 7-Bromo-9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4-b]quinoline-3,4-dione (7, 22 mg, 0.062 mmole) in DMF (0.75 ml) under nitrogen. The reaction is stirred at room temperature for 18 hours. Water (1 ml) is added to the reaction mixture and the solid filtered, washed with water, and dried to yield 8 as a white solid. 1H NMR (DMSO-d6) δ: 8.30 (d, 1H), 7.91 (d, 1H), 3.58 (m, 1H), 1.57 (s, 9H), 1.41 (m, 2H), 1.27 (m, 2H). 19F NMR (DMSO-d6)) δ: 111.1 (s, 1H).

STEP H. Preparation of 9-cyclopropyl-6-fluoro-7-aryl-9H-isothiazolo[5,4-B]quinoline-3,4-dione (9)

Pd(PPh3)4 (6 to 10 mole %) is added to a stirred suspension of 7-Bromo-9-cyclopropyl-6-fluoro-3,4-dioxo-4,9-dihydro-3H-isothiazolo[5,4-b]quindine-2-carboxylic acid tert-butyl ester (8, 20 mg, 0.044 mmole) in DMF (1 ml), followed by addition of a boronic acid (2 equivalents, 0.088 mmole) and NaHCO3 solution (1M, 0.2 ml, 4.5 equiv.) under argon at room temperature. The reaction tube is sealed and then stirred in a microwave (100W, 130° C.) until completion (usually 10 minutes, though a longer reaction time may be needed for some boronic acids). The reaction mixture is monitored by LC MS until no starting material remains. The reaction mixture is then filtered and the filtrate concentrated in vacuo. The residue is washed with a mixture of MeOH: diethyl ether (approximately 5:95, 3×). The resulting light yellow solid product is dried and analyzed. Some products require HPLC purification.

Alternate Procedure for the Synthesis of 9-cyclopropyl-6-fluoro-7-aryl-9H-isothiazolo[5,4-B]quinoline-3,4-dione

Pd (PPh3)4 (3.1 mg, 6% mole), boronic acid or ester (2 equiv., 0.088 mmole) and NaHCO3 solution (1M, 0.2 mL, 4.5 equiv.) under argon at room temperature is added to a stirred solution of 7-bromo-9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4-b]quinoline-3,4-dione (20 mg, 0.044mmole) in DMF (1 mL). The reaction mixture is degassed by bubbling argon through for 10 minutes at room temperature. The reaction tube is sealed and then heated in a microwave (100W, 130° C.) until the reaction reaches completion (10-20 minutes). The reaction mixture is cooled to room temperature and then filtered. The filtrate is concentrated in vacuo. The residue is dissolved in a mixture of DMF:CHCl3:MeOH (0.5:3:0.5) (4 mL) and precipitated with diethyl ether. This dissolution and precipitation step is repeated 5 times. The resulting light yellow solid is washed with water (3 mL) and dried to afford the title compound.

Example 2 General Method for the Preparation of 9-cyclopropyl-8-methoxy-7-aryl-9H-isothiazolo[5,4-B]quinoline-3,4-diones

9-Cyclopropyl-8-methoxy-7-aryl-9H-isothiazolo[5,4-b]quinoline-3,4-diones (18) are prepared in accordance with the synthetic scheme set forth below.

STEP A. Preparation of 3-fluoro-2-methoxyphenylamine (11)

Potassium carbonate (59.25 g, 0.43 mol) is added slowly to a solution of dimethylformamide (200 mL) containing 2-fluoro-6-nitrophenol (10, 33.63 g, 0.21 mol) and dimethylsulfate (41.0 mL, 0.43 mol) at room temperature. The orange mixture is stirred at 80° C. for 6 h. The resulting yellow mixture is cooled to room temperature, diluted with water (500 mL), and extracted with hexanes (3×500 mL). The combined organic extracts are dried over magnesium sulfate and evaporated under reduced pressure to give 1-fluoro-2-methoxy-3-nitrobenzene as a yellow oil. This product is of sufficient purity (≧95% by NMR spectroscopy) to use directly in the next synthetic step. 1H NMR (CDCl3): δ4.08 (d, JH-F=2.0 Hz, 3H, OCH3), 7.13 (apparent t of d, JH-H=8.5 Hz, JH-F=5.0 Hz, 1H, H-5), 7.34 (d, JH-F=10.5 Hz, JH-H=8.5 Hz, JH-H=1.5 Hz, 1H, H-6), 7.58 (d of apparent t, JH-H=8.5 Hz, JH-H=1.5 Hz, JH-F=1.5 Hz, 1H, H-4). 19F{1H} NMR (CDCl3): δ−126.7 (s). 13C{1H} NMR (CDCl3): δ62.6 (d, JC-F=5.5 Hz, OCH3), 120.2 (d, JC-F=3.5 Hz, C-4), 121.1 (d, JC-F=19.5 Hz, C-6), 123.2 (d, JC-F=8.0 Hz, C-5), 142.2 (d, JC-F=14.5 Hz, C-2), 144.8 (br, C-3), 156.2 (d, JC-F=251.5 Hz, C-1). LCMS m/z calcd for C7H6FNO3 ([M]+) 171; found 183 ([M−CH2O+H+CH3CN]+, 26%), 183 ([M−CH2O +H]+, 100%).

A mixture containing 1-fluoro-2-methoxy-3-nitrobenzene (36.30 g, 0.21 mol), palladium on carbon (10% w/w, ˜8 g), and methanol (200 mL) is stirred under an atmosphere of hydrogen (1 atm) for 27 h. The mixture is filtered and the resulting solution is evaporated to dryness under reduced pressure to give 11 as a brown oil. This product is of sufficient purity (≧95% by NMR spectroscopy) to use directly in the next synthetic step. 1H NMR (CDCI3): δ3.75 (br, 2H, NH2), 3.91 (d, JH-F=1.5 Hz, 3H, OCH3), 6.46 (m, 2H, overlapping H-4 and H-6), 6.79 (apparent t of d, JH-H=8.0 Hz, JH-F=5.5 Hz, 1H, H-5). 19F{1H} NMR (CDCl3): δ−132.5 (s). 13C{1H} NMR (CDCl3): δ60.7 (d, JC-F=5.0 Hz, OCH3), 106.1 (d, JC-F=19.5 Hz, C-4), 110.9 (d, JC-F=2.5 Hz, C-6), 123.7 (d, JC-F=9.5 Hz, C-5), 134.9 (d, JC-F=13.0 Hz, C-2), 141.3 (d, JC-F=5.0 Hz, C-1), 154.4 (d, JC-F=244.0 Hz, C-3). LCMS m/z calcd for C7H8FNO ([M]+) 141; found 142 ([M+H]+).

STEP B. Preparation of 4-bromo-2-fluoro-3-methoxybenzoic acid (12).

(a) Hydrobromic acid (48% in water, 140 mL) is added slowly to an aliquot of 11 (14.33 g, 101.5 mmol) cooled to 0° C. The resulting solid is broken up with a glass rod and stirred vigorously at 0° C. for 10 min. A solution of sodium nitrite (7.40 g, 107.2 mmol) in water (50 mL) is added slowly (˜1.5 h) to the stirred slurry containing 3-fluoro-2-methoxyphenylamine and hydrobromic acid, maintaining the temperature of the reaction mixture below 5° C. A purple solution of cuprous bromide (9.62 g, 67.1 mmol) in hydrobromic acid (48% in water, 50 mL) is added dropwise to the reaction mixture, maintaining the temperature of the reaction mixture below 5° C. The resulting reaction mixture is heated at 60° C. until the evolution of gas ceases (˜2.5 h). The reaction mixture is cooled to room temperature, and the product extracted with diethyl ether (6×150 mL). The combined organic extracts are washed with brine (3×150 mL), dried over magnesium sulfate, and evaporated under reduced pressure to give 1-bromo-3-fluoro-2-methoxybenzene as a brown oil. This product is of sufficient purity (≧95% by NMR spectroscopy) to use directly in the next synthetic step. 1H NMR (CDCl3): δ3.95 (d, JH-F=1.5 Hz, 3H, OCH3), 6.88 (apparent t of d, JH-H=8.0 Hz, JH-F=5.5 Hz, 1H, H-5), 7.04 (d, JH-F=10.5 Hz, JH-H=8.0 Hz, JH-H=1.5 Hz, 1H, H-4), 7.30 (d of apparent t, JH-H=8.0 Hz, JH-H=1.5 Hz, JH-F=1.5 Hz, 1H, H-6). 19F{1H} NMR (CDCl3): δ−127.7 (s). 13C{1H} NMR (CDCl3): 61.4 (d, JC-F=5.0 Hz, OCH3), 116.2 (d, JC-F=19.5 Hz, C-4), 117.7 (d, JC-F=3.0 Hz, C-1), 124.5 (d, JC-F=8.0 Hz, C-5), 128.5 (d, JC-F=3.5 Hz, C-6), 145.7 (d, JC-F=12.5 Hz, C-2), 156.2 (d, JC-F=250.5 Hz, C-3).

(b) Lithium diisopropylamide (LDA) is formed by dropwise addition of n-butyllithium (1.6 M in hexanes, 56.0 mL, 89.6 mmol) to a stirred solution of diisopropylamine (13.7 mL, 96.9 mmol) in tetrahydrofuran (150 mL) at −78° C. The resulting solution is stirred at −78° C. for 5 min, 0° C. for 15 min, and then cooled again to −78° C. A solution of 1-bromo-3-fluoro-2-methoxybenzene (15.28 g, 74.5 mmol) in tetrahydrofuran (40 mL) is added dropwise to the previous solution over a period of 30 min. to give an amber solution. After stirring this solution at −78° C. for 1.5 h, dry ice (˜125 g) is added, and the resulting mixture is allowed to warm slowly (˜1 h) to room temperature with stirring as it degasses. The reaction mixture is acidified to pH ˜1 by addition of a 5% aqueous solution of hydrochloric acid (˜500 mL), and the product is extracted with diethyl ether (6×100 mL). The combined organic extracts are washed with brine (100 mL), and the product extracted with a saturated solution of aqueous sodium bicarbonate (3×100 mL). The combined aqueous extracts (pH ˜9) are washed with diethyl ether (3×100 mL) and acidified slowly to pH ˜1 by addition of a 37% aqueous solution of hydrochloric acid (˜50 mL). The product is extracted with diethyl ether (3×200 mL), and the combined organic extracts are washed with brine (100 mL), dried over anhydrous magnesium sulfate, and concentrated under reduced pressure to give 12 as an off-white solid. This product is of sufficient purity (≧95% by NMR spectroscopy) to use directly in the next synthetic step. mp 168-170° C. 1H NMR (CD3OD): δ3.92 (d, JH-F=1.0 Hz, 3H, OCH3), 7.44 (dd, JH-H=8.5 Hz, JH-F=1.5 Hz, 1H, H-5), 7.55 (dd, JH-H=8.5 Hz, JH-F=7.0 Hz, 1H, H-6). 19F {1H} NMR (CD3OD): δ−127.0 (s). 13C{1H} NMR (CD3OD): δ62.1 (d, JC-F=4.5 Hz, OCH3), 121.5 (d, JC-F=8.5 Hz, C-1), 123.6 (d, JC-F=2.0 Hz, C-4), 128.0 (s, C-6), 129.0 (d, JC-F=4.5 Hz, C-5), 147.7 (d, JC-F=13.5 Hz, C-3), 157.1 (d, JC-F=263.5 Hz, C-2), 166.3 (d, JC-F=3.0 Hz, CO2H). HRMS m/z calcd for C8H679BrFNaO3 270.9382 ([M+Na]+); found 270.9377.

STEP C. Preparation of ethyl 3-(4-bromo-2-fluoro-3-methoxyphenyl)-3-oxopropionate (13).

Compound 13 is prepared using the general two-step method of Wierenga and Skulnick. (Wierenga, W.; Skulnick, H. I. J. Org. Chem. 1979, 44, 310-311.)

(a) Dimethylformamide (5 drops) is added by Pasteur pipette to a mixture containing 12 (5.30 g, 21.3 mmol) and oxalyl chloride in methylene chloride (2.0 M, 21.3 mL, 42.6 mmol) at room temperature. The resulting mixture is stirred until an amber solution forms and the evolution of gas ceases (1 h). The solution is concentrated under reduced pressure to give the intermediate acid chloride as an off-white solid that is used directly in the following step.

(b) n-Butyllithium (1.6 M in hexanes) is added to a cooled (−78° C.) solution of tetrahydrofuran (50 mL) containing ethyl hydrogen malonate (5.62, 42.5 mmol) and 2,2′-bipyridyl (8.2 mg as indicator). The temperature of the reaction mixture is allowed to rise to ˜0° C. during the addition of n-butyllithium. Sufficient n-butyllithium (˜50 mL) is added until a pink color persists at −5° C. for 5-10 min. A solution of the acid chloride (vide supra) in methylene chloride (20 mL) is added in one portion to the reaction mixture that has been cooled to −78° C. The resulting mixture is allowed to warm to 10° C. (˜30 min), and quenched with an aqueous solution of hydrochloric acid (1 M, 100 mL). The reaction mixture is extracted with diethyl ether (3×100 mL). The combined organic extracts are washed with a saturated aqueous solution of sodium bicarbonate (3×100 mL), followed by brine (100 mL), dried over magnesium sulfate, and evaporated under reduced pressure to give the crude product. This material is purified by flash column chromatography (eluting with ethyl acetate/hexanes (1:6 v/v); Rf 0.43) to give pure 13 as a pale orange oil that solidified upon standing. mp 52-53° C. The title compound exists as a mixture of keto (major) and enol (minor) tautomers at room temperature in CDCl3, DMSO-d6, and CD3OD. 1H NMR (CDCl3): δ1.27 (t, JH-H=7.0 Hz, keto CO2CH2CH3), 1.34 (t, JH-H=7.0 Hz, enol CO2CH2CH3), 3.96 (m, overlapping keto OCH3, enol OCH3, and keto C(O)CH2CO2CH2CH3), 4.22 (q, JH-H=7.0 Hz, keto CO2CH2CH3), 4.27 (q, JH-H=7.0 Hz, enol CO2CH2CH3), 5.81 (d, JH-F=0.5 Hz, enol C(OH)═CHCO2CH2CH3), 7.39 (dd, JH-H=8.5 Hz, JH-F=1.5 Hz, enol aromatic H-5), 7.43 (dd, JH-H=8.5 Hz, JH-F=1.5 Hz, keto aromatic H-5), 7.47 (dd, JH-H=8.5 Hz, JH-F=7.0 Hz, enol aromatic H-6), 7.53 (dd, JH-H=8.5 Hz, JH-F=7.0 Hz, keto aromatic H-6), 12.67 (s, enol OH). 19F{1H} NMR (CDCl3): δ−126.3 (s, enol), −125.9 (s, keto). LCMS m/z calcd for C12H1279BrFO4 ([M]+) 318; found 319 ([M+H]+). HRMS m/z calcd for C12H1279BrFNaO4 340.9801 ([M+Na]+); found 340.9797.

STEP D. Preparation of ethyl 3-(4-bromo-2-fluoro-3-methoxyphenyl)-2-(cyclopropyliminomethylsulfanylmethyl)-3-hydroxyacrylate (14).

Sodium hydride (60% in mineral oil, 73.7 mg, 1.92 mmol) is added portionwise to a cooled (0° C.) solution containing 13 (569 mg, 1.78 mmol), cyclopropyl isothiocyanate (500 μL, 5.40 mmol), and dimethylformamide (5.0 mL). The resulting mixture was allowed to warm to room temperature with stirring overnight (18.5 h). Methyl iodide (700 μL, 11.22 mmol) is added to the resulting solution to give a precipitate within minutes. The mixture is stirred for an additional 24 h. The reaction mixture is quenched by addition of a saturated aqueous solution of ammonium chloride (50 mL) and extracted with ethyl acetate (3×100 mL). The combined organic extracts are washed with brine (200 mL), dried over magnesium sulfate, and evaporated under reduced pressure to give the crude product. This material is purified by flash column chromatography (eluting with 10% v/v ethyl acetate in methylene chloride; Rf0.59) to give 586.0 mg (76% yield) of 14 as a viscous yellow oil. 1H NMR (CDCl3): δ0.86 (m, 2H, c-Pr CH2), 0.89 (t, JH-H=7.0 Hz, CO2CH2CH3), 0.98 (m, 2H, c-Pr CH2), 2.52 (s, 3H, S—CH3), 3.01 (m, 1H, c-Pr CH), 3.90 (q, JH-H=7.0 Hz, 2H, CO2CH2CH3), 3.94 (d, JH-F=1.5 Hz, 3H, OCH3), 6.97 (dd, JH-H=8.5 Hz, JH-F=6.5 Hz, 1H, aromatic H-6), 7.30 (dd, JH-H=8.5 Hz, JH-F=1.5 Hz, 1H, aromatic H-5), 11.91 (br, 1H, OH). 19F{1H} NMR (CDCl3): δ−130.4 (s). 13C{1H} NMR (CDCl3): δ8.6 (c-Pr CH2), 13.5 (CO2CH2CH3), 18.1 (S—CH3), 28.5 (c-Pr CH), 60.3 (CO2CH2CH3), 61.4 (d, JC-F=5.0 Hz, OCH3), 104.2 (—C(OH)═C(CO2CH2CH3)—), 118.3 (d, JC-F=2.5 Hz, aromatic C-4), 123.4 (d, JC-F=3.5 Hz, aromatic C-6), 127.6 (d, JC-F=3.5 Hz, aromatic C-5), 131.9 (d, JC-F=14.5 Hz, aromatic C-1), 145.1 (d, JC-F=13.5 Hz, aromatic C-3), 152.6 (d, JC-F=253.0 Hz, C-2), 167.7 (CO2CH2CH3), 174.5 (—N═C(S—CH3)—), 185.5 (—C(OH)═C(CO2CH2CH3)—). LCMS m/z calcd for C17H1979BrFNO4S ([M]+) 431; found 432 ([M +H]+). HRMS m/z calcd for C17H2079BrFNO4S 432.0280 ([M+H]+); found 432.0276.

STEP E. Preparation of ethyl 7-bromo-1-cyclopropyl-8-methoxy-2-methylfulfanyl-4-oxo-1,4-dihydroquinoline-3-carboxylate (15).

Sodium hydride (60% in mineral oil, 51.9 mg, 1.30 mmol) is added portionwise to a solution of 14 (527.6 mg, 1.22 mmol) in dimethylformamide (5.0 mL) at room temperature. The reaction mixture is heated at 75° C. for 75 h, cooled to room temperature, and quenched by addition of a saturated aqueous solution of ammonium chloride (75 mL). The mixture is extracted with ethyl acetate (3×75 mL). The combined organic extracts are washed with brine (75 mL), dried over magnesium sulfate, and evaporated under reduced pressure to give crude 15 as a tan solid. This product is of sufficient purity (≧95% by NMR spectroscopy) to use directly in the next synthetic step. 1H NMR (CDCl3): δ0.70 (m, 2H, c-Pr CH2), 1.18 (m, 2H, c-Pr CH2), 1.39 (t, J=7.0 Hz, 3H, CO2CH2CH3), 2.63 (s, 3H, S—CH3), 3.68 (m, 1H, c-Pr CH), 3.80 (s, 3H, OCH3), 4.40 (q, J=7.0 Hz, 2H, CO2CH2CH3), 7.54 (d, J=8.5 Hz, 1H, aromatic H-6), 7.88 (d, J=8.5 Hz, 1H, aromatic H-5). 13C{1H} NMR (CDCl3): δ12.4 (br, c-Pr CH2), 14.2 (CO2CH2CH3), 18.4 (S—CH3), 37.0 (c-Pr CH), 60.8 (OCH3), 61.8 (CO2CH2CH3), 122.7 (CH, C-5), 123.1 (C—Br, C-7), 123.6 (C-3), 129.2 (C-4a), 129.3 (CH, C-6), 140.0 (C-8a), 147.9 (C—OCH3, C-8), 156.3 (C—S—CH3, C-2), 165.5 (CO2CH2CH3), 173.6 (C═O, C-4). LCMS m/z calcd for C17H1879BrNO4S ([M]+) 411; found 412 ([M+H]+). HRMS m/z calcd for C17H1879BrNNaO4S 434.0038 ([M+Na]+); found 434.0031.

STEP F. ethyl 7-bromo-1-cyclopropyl-2-methanesulfinyl-8-methoxy-4-oxo-1,4-dihydroquinoline-3-carboxylate (16)

m-Chloroperoxybenzoic acid (≦77%, 273.5 mg, 1.22 mmol) is added in one portion to a solution of crude ethyl 15 (from above ˜1.22 mmol) in methylene chloride (5.0 mL) at room temperature. The reaction mixture is stirred for 1 h, diluted with methylene chloride (10 mL), and washed with a saturated aqueous solution of sodium bicarbonate (25 mL). The organic layer is dried over magnesium sulfate and evaporated under reduced pressure to give the crude product. This material is purified by flash column chromatography (eluting with ethyl acetate; Rf0.37) to give 290.9 mg of pure ethyl 16 as a white solid. 1H NMR (CDCl3): δ0.54 (m, 1H, c-Pr CH2 (A)), 0.93 (m, 1H, c-Pr CH2 (B)), 1.12 (m, 1H, c-Pr CH2 (A)), 1.28 (m, 1H, c-Pr CH2 (B)), 1.38 (t, J=7.0 Hz, 3H, CO2CH2CH3), 3.26 (s, 3H, S(O)—CH3), 3.83 (s, 3H, OCH3), 3.92 (m, 1H, c-Pr CH), 4.40 (m, 2H, overlapping CO2CHHCH3), 7.58 (d, J=8.5 Hz, 1H, aromatic H-6), 7.87 (d, J=8.5 Hz, 1H, aromatic H-5). 13C{1H} NMR (CDCl3): δ10.8 (br, c-Pr CH2 (A)), 13.9 (br, c-Pr CH2 (B)), 14.1 (CO2CH2CH3), 35.1 (c-Pr CH), 41.4 (S(O)—CH3), 61.1 (OCH3), 62.1 (CO2CH2CH3), 118.9 (C-3), 122.8 (CH, C-5), 123.9 (C—Br, C-7), 129.5 (C-4a), 130.0 (CH, C-6), 138.2 (C-8a), 148.3 (C—OCH3, C-8), 164.0 (CO2CH2CH3), 164.1 (br, C-S(O)—CH3, C-2), 174.6 (C═O, C-4). LCMS m/z calcd for C17H1879BrNO5S ([M]+) 427; found 428 ([M+H]+). HRMS m/z calcd for C17H1879BrNNaO5S 449.9987 ([M+Na]+); found 449.9977.

STEP G. Preparation of 7-bromo-9-cyclopropyl-8-methoxy-9H-isothiazolo[5,4-B]quinoline-3,4-dione (17)
(a) Anhydrous sodium hydrogen sulfide (Alfa Aesar, 53.3 mg, 0.95 mmol) is added in one portion to a solution of dimethylformamide (4.0 mL) containing 16 (158.1 mg, 0.37 mmol) at room temperature. The resulting solution is heated at 50° C. for 1 h and allowed to cool to room temperature. The reaction mixture is quenched by addition of a 5% aqueous solution of hydrochloric acid (50 mL) and extracted with ethyl acetate (100 mL). The organic extract is washed with brine (50 mL) and evaporated to dryness under reduced pressure to give crude ethyl 7-bromo-1-cyclopropyl-2-mercapto-8-methoxy-4-oxo-1,4-dihydroquinoline-3-carboxylate. Rf˜0 (ethyl acetate). LCMS m/z calcd for C16H1679BrNO4S ([M]+) 397; found 398 ([M+H]+).

(b) A solution of sodium bicarbonate (316.9 mg, 3.77 mmol) in water (7.5 mL) is added to a solution of ethyl 7-bromo-1-cyclopropyl-2-mercapto-8-methoxy-4-oxo-1,4-dihydroquinoline-3-carboxylate (from above, ˜0.37 mmol) in tetrahydrofuran (7.5 mL) at room temperature. Hydroxylamine-O-sulfonic acid (214.7 mg, 1.90 mmol) is added as a solid and in one portion to this mixture. The resulting amber solution is stirred at room temperature for 2.5 h and quenched by addition of an aqueous solution of 5% hydrochloric acid (50 mL). The solid that formed is collected by filtration, washed with an aqueous solution of 5% hydrochloric acid (3×10 mL), washed with distilled water (3×10 mL), and dried in vacuo to give 17 as a tan solid. This product ids of sufficient purity (≧95% by 1H NMR spectroscopy) to use directly in the next synthetic step. 1H NMR (DMSO-d6): δ1.00 (m, 2H, c-Pr CH2), 1.20 (m, 2H, c-Pr CH2), 3.79 (s, 3H, OCH3), 3.85 (m, 1H, c-Pr CH), 7.66 (d, J=8.5 Hz, 1H, aromatic H-6), 7.93 (d, J=8.5 Hz, 1H, aromatic H-5), 11.67 (br, 1H). 13C{1H} NMR (DMSO-d6): 811.5 (c-Pr CH2), 35.1 (c-Pr CH), 61.9 (OCH3), 107.7, 122.9 (CH, C-5), 123.5 (C—Br, C-7), 127.9 (CH, C-6), 128.0 (C-4a), 136.5 (C-8a), 146.6 (C—OCH3), 164.5, 171.1 (C═O, C-4), 171.2 (br). LCMS m/z calcd for C14H1179BrN2O3S ([M]+) 366; found 367 ([M+H]+). HRMS m/z calcd for C14H1179BrN2NaO3S 388.9571 ([M+Na]); found 388.9577.

STEP H. General Method for the Preparation of 9-cyclopropyl-8-methoxy-7-aryl-9H-isothiazolo[5,4-b]quinoline-3,4-diones (18)

Compound 18 is prepared by Suzuki Cross-Coupling Reaction of 17 and aryl boronic acid (R═H) or aryl boronic ester (R=alkyl).

Under an atmosphere of argon, a reaction vessel is charged with 17 (0.1 mmol), dimethylformamide (2 mL), tetrahydrofuran (2 mL), tetrakis(triphenylphosphine)palladium(0) (0.01-0.02 mmol), the desired boronic acid or ester (0.3-0.4 mmol), and a 1 M aqueous solution of sodium bicarbonate (1-2 mmol). The resulting mixture is irradiated with microwaves at 130° C. for 10-20 min, allowed to cool, and evaporated to dryness under reduced pressure. The isolated residues are purified using preparative HPLC to give the desired products (95-99% purity). The purified products are isolated as TFA salts and converted to the corresponding hydrochloride salts by addition of a 5% aqueous solution of hydrochloric acid followed by evaporation; this process is repeated twice.

Example 3 Preparation of 9-cyclopropyl-7-(2,6-dimethyl-pyridin-4-yl)-6-fluoro-9H-isothiazolo[5,4-B]quinoline-3,4-dione (XXIII) (25)

9-Cyclopropyl-7-(2,6-dimethyl-pyridin-4-yl)-6-fluoro-9H-isothiazolo[5,4-b]quinoline-3,4-dione (25) is prepared in accordance with the synthetic scheme set forth below:

STEP A. Preparation of 2,6-lutidine 1-oxide (20)

A solution of 2,6-lutidine (19, 23 ml, 200 mmol) and 50% hydrogen peroxide (15 ml) in glacial acetic acid (100 ml) is refluxed at 110° C. for 3 hours. The solution is then concentrated in vacuo at 40° C. to approximately 60 ml. Water (20 ml) is added, and the concentration process is repeated three times. The concentrated solution is further dried by lyophlizer overnight, yielding 26 g of 2,6-Lutidine 1-oxide (20) that contains approximately 10% acetic acid. 1H NMR (300 MHz, CDCl3) δ2.52 (s, 6H), 7.15 (m, 3H). MS, m/z 124 (M+1), 247 (2M+1).

STEP B. Preparation of 4-nitro-2, 6-lutidine 1-oxide (21)

A mixture of 2,6-lutidine 1-oxide (20, 15 g, 110 mmol) and concentrated sulfuric acid (98%, 30 ml) and concentrated nitric acid (70%, 12 ml) is heated under reflux for 3 hours. The mixture is poured into a large excess of ice and extracted with chloroform (3×100 ml). The combined chloroform extracts are washed with aqueous sodium hydroxide and water and dried over magnesium sulfate. Removal of the solvent yields pure yellow solid 4-Nitro-2, 6-lutidine 1-oxide (21). 1H NMR (300 MHz, CDCl3) δ2.64 (s, 6H), 8.08 (s, 2H). MS, m/z 169 (M+1), 210 (M+MeCN).

STEP C. Preparation of 4-amino-2, 6-lutidine (22)

A mixture of 4-Nitro-2,6-lutidine 1-oxide (21, 5.1 g, 30 mmol), palladized charcoal (10% Pd, 1 g) and acetic acid (2 ml) in methanol (200 ml) is hydrogenated under pressure (40 psi) over 10 hours using a hydrogenation apparatus. The reaction is followed with LC-MS. After filtration, the filtrate is concentrated. The remaining oil is further dried by lyophilization, yielding 4.5 g of 4-amino-2, 6-lutidine (22) containing approximately 15% acetic acid. 1H NMR (300 MHz, CDCl3) δ2.3 (s, 6H), 7.2 (s, 2H). MS, m/z 123 (M+1), 243 (2M−1).

STEP D. Preparation of 4-bromo-2, 6-lutidine (23)

Bromine (4 g) is added, with stirring over 10 minutes, to a mixture of 4-amino-2, 6-lutidine (22, 1 g, approximately 6.5 mmol) in 48% HBr (12 ml) at −10° C., followed by cooling to −20° C. A solution of sodium nitrite (1.4 g) in water (4 ml) is added slowly. The mixture is stirred at −20° C. for 1 hour, and then warmed and left at room temperature for 3 hours. The mixture is distilled. The oil fraction of the distillate is extracted with chloroform (3×10 ml). The combined extracts are dried over magnesium sulfate. After filtration, the filtrate is neutralized in an ice-bath using 2M butyl lithium in hexanes until the pH reaches 7. A large amount of salt forms. After filtration, the filtrate is concentrated and dried, yielding 4-bromo-2,6-lutidine (23) oil. 1H NMR (300 MHz, CDCl3) δ2.82 (s, 6H), 7.5 (s, 2H). MS, m/z 186 and 188 (M+1).

STEP E. Preparation of 4-(2,6-dimethylpyridyl)boronic acid (24)

Butyl lithium (2M in hexanes, 0.6 ml, 1.2 mmol) is added to a solution of 4-bromo-2, 6-lutidine (23, 0.2 g, 1 mmol) and triisopropyl borate (0.28 ml, 1.2 mmol) in solution of toluene (1.6 ml) and THF (0.4 ml) over 10 min at −40° C. under helium. The reaction is stirred at −40° C. for 30 minutes and then warmed to −20° C. 2N HCl (1 ml) is added to quench the reaction. The mixture is warmed to room temperature. The aqueous layer is collected and neutralized using 5M NaOH. NaCl (approximately 0.4 g) is added. The aqueous layer is extracted with THF (3×10 ml), and the extracts are evaporated to dryness, yielding white solid 4-(2,6-dimethylpyridyl) boronic acid (24). 1H NMR (300 MHz, DMSO-d6) δ2.5 (s, 6H), 7.34 (s, 2H). MS, m/z 152 (M+1).

STEP F. Preparation of 9-cyclopropyl-7-(2,6-dimethyl-pyridin-4-yl)-6-fluoro-9H-isothiazolo[5,4-B]quinoline-3,4-dione (25)

A mixture of tert-butyl 7-bromo-9-cyclopropyl-6-fluoro-3,4-dioxoisothiazolo[5,4-b]quinoline-2(3H,4H,9H)-carboxylate (8, 22 mg, 0.048 mmol), 4-(2,6-Dimethylpyridyl)boronic acid (5, 23 mg, 0.12 mmol) and Pd (PPh3)4 (4 mg) in a solution of DMF (1 ml) and 1M NaHCO3 (0.22 ml) is sealed in a microwave reaction vessel with a stirrer. After filling with helium, the mixture is microwaved at 100W, 130° C. for 10 minutes. The reaction is followed by LC-MS. The reaction mixture is filtered and the filtrate evaporated to dryness. The residue is washed with a solution of methanol and ethyl ether (5:95) (3×3 ml), and dried in vacuum yielding pure product 25. 1H NMR (300 MHz, DMSO-d6) δ1.1-1.3 (m, 4H, —CH2—), 1.53 (s, 1H, —CH—), 2.47 (s, 6H, —CH3), 7.27 (s, 2H, Ar), 7.93, (d, 1H, Ar), 8.2 (d, 1H, Ar). 19F NMR (350 MHz, DMSO-d6) δ125 (s, 1F). MS, m/z 382 (M+1).

Example 4 Preparation of 7-(1,2,3,6-tetrahydro-pyridin-4-yl)-9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4B]-muinoline-3,4-dione (27)

STEP A. Preparation of 1-acetyl-4-tributylstannyl-1,2,3,6-tetrahydropyridine

Caution: organotin compounds are toxic (Buck, B.; Mascioni, A.; Que, L, Jr., Veglia, G. J. Am. Chem. Soc. 2003, 125, 13316-13317, and references cited therein). 1-(4-hydroxy-4-tributylstannanylpiperidin-1-yl)ethanone is prepared using the two-step procedure described previously (Kiely, J. S.; Lesheski, L. E.; Schroeder, M. C. Preparation of Certain 7-Substituted Quinolones. U.S. Pat. No. 4,945,160, Jul. 31, 1990). Because formation of 1-(4-hydroxy-4-tributylstannanylpiperidin-1-yl)ethanone is reversible, this compound is used immediately after isolation (without purification) to generate 1-acetyl-4-tributylstannyl-1,2,3,6-tetrahydropyridine. The isolated crude material is purified by flash column chromatography (eluting with 5% methanol in methylene chloride; Rf 0.30 (UV inactive)) to give the title product as a yellow oil. 1H NMR (CDCl3, 50° C.): δ0.89 (m, 15H, Bu), 1.33 (m, 6H, Bu), 1.50 (m, 6H, Bu), 2.07 (s, 3H, NC(O)CH3), 2.31 (m, 2H, H-2), 3.55 (m, 2H, H-3), 4.01 (m, 2H, H-6), 5.76 (m, 1H, H-5). 1H NMR spectroscopic data collected at room temperature matched those described in the literature, where two conformational isomers were detected. LCMS m/z calcd for C19H37NO120Sn ([M]+) 415; found 416 ([M+H]+).

STEP B. Preparation of 7-(1-acetyl-1,2,3,6-tetrahydro-pyridin-4-yl)-9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4-B]quinoline-3,4-dione (26).

A mixture containing 7-bromo-9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4-b]quinoline-3,4-dione (7, 100.0 mg, 0.28 mmol), 1-acetyl-4-tributylstannyl-1,2,3,6-tetrahydropyridine (190.0 mg, 0.46 mmol), tetrakis(triphenylphosphine)palladium(0) (16.0 mg, 0.014 mmol), and dimethylformamide (6.0 mL) Is sparged with argon gas and irradiated with microwaves (5×10 min irradiations at 130° C.). (For cross-coupling experiments described previously using C with conventional thermal heating, see: (a) Laborde, E.; Kiely, J. S.; Culbertson, T. P.; Lesheski, L. E. J. Med. Chem. 1993, 36, 1964-1970. (b) Kiely, J. S.; Laborde, E.; Lesheski, L. E.; Bucsh, R. A. J. Heterocyclic Chem. 1991, 28, 1581-1585. (c) Laborde, E.; Kiely, J. S.; Lesheski, L. E.; Schroeder, M. C. J. Heterocyclic Chem. 1990, 28, 191-198) The resulting yellow solution is evaporated to dryness under reduced pressure (˜6 mm Hg, 60° C.). The recovered solid is dissolved in a mixture containing 10% methanol in methylene chloride (10 mL), precipitated via addition of hexanes (100 mL), and collected by filtration. This process is repeated once. The product is purified further by flash column chromatography (eluting with 10% methanol in methylene chloride, Rf 0.50) to give pure 26 as a yellow solid (98% purity by HPLC). mp 243-244° C. 1H NMR (CDCl3/CD3OD (12:1 v/v), 50° C.): δ1.32 (m, 2H, c-Pr CH2), 1.45 (m, 2H, c-Pr CH2), 2.18 (s, 3H, NC(O)CH3), 2.65 (m, 2H, NCH2CH2), 3.45 (m, 1H, c-Pr CH), 3.73 (m, 1H, NCH2CH2), 3.87 (m, 1H, NCH2CH2), 4.23 (m, 1H, NCH2CH), 4.31 (m, 1H, NCH2CH), 6.19 (m, 1H, NCH2CH), 7.88 (d, JH-F=6.0 Hz, 1H, quinolone H-8), 8.08 (d, JH-F=11.0 Hz, 1H, quinolone H-5). 19F{1H} NMR (CDCl3/CD3OD (12:1 v/v), 50° C.): δ−119.4 (s). NMR spectroscopic data collected at room temperature indicated that two conformational isomers were present. LCMS m/z calcd for C20H18FN3O3S ([M]+) 399; found 400 ([M+H]+). HRMS m/z calcd for C20H18FN3NaO3S ([M+Na]+) 422.0951; found 422.0951.

STEP C. Preparation of 7-(1,2,3,6-tetrahydro-pyridin-4-yl)-9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4-B]quinoline-3,4-dione (27)

Compound 26 (11.1 mg, 0.028 mmol) is dissolved partially in an aqueous solution of hydrochloric acid (6 M, 3.0 mL) in air and heated at 90° C. to give an amber solution. After 22 h of heating, the solvent is removed under reduced pressure. The residue is dissolved in water (˜3 mL) and titrated to pH 7 with dilute sodium hydroxide. The precipitated solid is collected by filtration, washed with water (2×10 mL), and dried in vacuo to give 27 as a yellow solid (98% purity by HPLC). mp >241-242° C. dec. 1H NMR (DMSO-d6/acetic acid-d4 (5:1 v/v)): δ1.19 (m, 2H, c-Pr CH2), 1.29 (m, 2H, c-Pr CH2), 2.72 (m, 2H, NCH2CH2), 3.33 (m, 2H, NCH2CH2), 3.54 (m, 1H, c-Pr CH), 3.80 (m, 2H, NCH2CH), 6.21 (m, 1H, NCH2CH), 7.87 (br, 1H, aromatic), 7.91 (br, 1H, aromatic). 19F{1H} NMR (DMSO-d6/acetic acid-d4 (5:1 v/v)): δ−121.4 (s). LCMS m/z calcd for C18H16FN3O2S ([M]+) 357; found 358 ([M+H]+). FIRMS m/z calcd for C18H16FN3NaO2S ([M+Na]+) 380.0845; found 380.0847.

Example 5 Preparation of (RAC)-7-(6-amino-5,6,7,8-tetrahydronaphthalen-2-yl)-9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4-B]quinoline-3,4-dione (31)

STEP A. Preparation of (RAC)-tert-butyl(6-bromo-1,2,3,4-tetrahydronaphthalen-2-yl)carbamate (29).

(a) (rac)-6-Bromo-1,2,3,4-tetrahydronaphthalen-2-ylamine is prepared from 6-Bromo-3,4-dihydro-1H-naphthalen-2-one (28) via a general procedure described previously for the reductive amination of ketones using NaBH3CN as reducing agent. (Borch, R. F.; Bernstein, M. D.; Durst, H. D. J. Am. Chem. Soc. 1971, 93, 2897-2904.). The purity of this material (brown oil) was >95%, as determined by 1H NMR spectroscopy, and was used without further purification. 1H NMR (CDCl3): δ1.45 (br, 2H, NH2), 1.56 (m, 1H, H-3), 1.98 (m, 1H, H-3), 2.47 (dd, J=16.0 Hz, 9.5 Hz, 1H, H-1), 2.82 (m, 2H, H-4), 2.93 (dd, J=16.0 Hz, 4.5 Hz, 1H, H-1), 3.16 (m, 1H, H-2), 6.92 (d, J=8.0 Hz, 1H, H-8), 7.22 (m, 2H, overlapping H-5 and H-7). 13C NMR (CDCl3): δ27.8 (CH2, C-4), 32.5 (CH2, C-3), 38.9 (CH2, C-1), 47.0 (CH, C-2), 119.3 (C—Br, C-6), 128.7 (CH, C-7), 130.9 (CH, C-8), 131.4 (CH, C-5), 134.3 (C-8a), 138.2 (C-4a). LCMS m/z calcd for C10H12BrN ([M]+) 225; found 226 ([M+H]+).

(b) Di-tert-butyldicarbonate (575.7 mg, 2.64 mmol) in methylene chloride (5.0 mL) is added in one portion to a solution of methylene chloride (7.0 mL) at room temperature that contains (rac)-6-Bromo-1,2,3,4-tetrahydronaphthalen-2-ylamine (591.6 mg, 2.62 mmol) and triethylamine (1.1 mL, 7.89 mmol). After stirring at room temperature for 19 h in air, the resulting amber solution is diluted with methylene chloride (25 mL), washed with a saturated aqueous solution of sodium chloride (2×50 mL), dried over magnesium sulfate, and evaporated to dryness under reduced pressure to give the title compound as a pale yellow solid. The purity of isolated 29 was >95%, as determined by 1H NMR spectroscopy, and was used without further ° C. 1H NMR (CDCl3): δ1.45 (s, 9H, C(CH3)3), 1.70 (m, 1H, H-3), 2.04 (m, 1H, H-3), 2.55 (dd, J=16.5 Hz, 8.5 Hz, 1H, H-1), 2.84 (pseudo t, J=6.5 Hz, 2H, H-4), 3.05 (dd, J=16.5 Hz, 5.0 Hz, 1H, H-1), 3.94 (br, 1H, H-2), 4.58 (br, 1H, NH), 6.91 (d, J=8.0 Hz, 1H, H-8), 7.22 (m, 2H, overlapping H-5 and H-7). 13C NMR (CDCl3): δ27.1 (CH2, C-4), 28.4 (C(CH3)3), 28.7 (CH2, C-3), 35.6 (CH2, C-1), 46.0 (CH, C-2), 79.4 (C(CH3)3), 119.6 (C—Br, C-6), 128.9 (CH, C-7), 131.0 (CH, C-8), 131.5 (CH, C-5), 133.3 (C-8a), 137.8 (C-4a), 155.3 (NHCO2). LCMS m/z calcd for C15H20BrNO2 ([M]+) 325; found 311 ([M−C4H7+CH3CN]+, 22%), 270 ([M−C4H7]+, 81%), 267 ([M−C5H7O2+CH3CN]+, 43%), 226 ([M−C5H7O2]+, 100%), 209 ([M−C5H10NO2]+, 94%).

STEP B. Preparation of (RAC)-tert-butyl[6-(4,4,5,5-tetramethyl[1,3,2]dioxaborolan-2-yl)-1,2,3,4-tetrahydronaphthalen-2-yl]carbamate (30)

The title compound, 30, is prepared via palladium-catalyzed cross-coupling reaction of 29 with bis(pinicolato)diboron using known procedures. (Ishiyama, T.; Murata, M.; Miyaura, N. J. Org. Chem. 1995, 60, 7508-7510.) The crude product is purified by flash column chromatography (eluting with 2% (v/v) methanol in methylene chloride; Rf0.41) to give pure (rac)-tert-butyl[6-(4,4,5,5-tetramethyl[1,3,2]dioxaborolan-2-yl)-1,2,3,4-tetrahydronaphthalen-2-yl]carbamate as an off-white, crystalline solid. mp 53-54° C. 1H NMR (CDCl3): δ1.33 (s, 12H, OC(CH3)2C(CH3)2O), 1.45 (s, 9H, C(CH3)3), 1.73 (m, 1H, H-3), 2.06 (m, 1H, H-3), 2.64 (dd, J=16.5 Hz, 8.0 Hz, 1H, H-1), 2.88 (pseudo t, J=6.5 Hz, 2H, H-4), 3.12 (dd, J=16.5 Hz, 5.0 Hz, 1H, H-1), 3.97 (br, 1H, H-2), 4.58 (br, 1H, NH), 7.07 (d, J=8.0 Hz, 1H, H-8), 7.54 (d, J=8.0 Hz, 1H, H-7), 7.56 (s, 1H, H-5). 13C NMR (CDCl3): δ24.8 (OC(CH3)2C(CH3)20), 26.9 (br, CH2, C-4), 28.4 (C(CH3)3), 29.1 (br, CH2, C-3), 36.3 (br, CH2, C-1), 46.1 (br, CH, C-2), 79.3 (br, C(CH3)3), 83.7 (OC(CH3)2C(CH3)2O), 126.3 (br, C—B, C-6), 129.0 (CH, C-8), 132.1 (CH, C-7), 134.9 (C-4a), 135.4 (CH, C-5), 137.8 (C-8a), 155.3 (NHCO2). LCMS m/z calcd for C21H32BNO4 ([M]+) 373; found 359 ([M−C4H7+CH3CN]+, 21%), 318 ([M−C4147]+, 37%), 315 ([M−C5H7O2+CH3CN]+, 100%), 274 ([M−C5H7O2]+, 82%), 257 ([M−C5H10NO2]+, 72%).

STEP C. Preparation of (RAC)-7-(6-amino-5,6,7,8-tetrahydronaphthalen-2-yl)-9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4-B]quinoline-3,4-dione (31)

(a) A mixture containing 7-bromo-9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4-b]quinoline-3,4-dione (8) (31.8 mg, 0.090 mmol), 30 (68.0 mg, 0.182 mmol), tetrakis(triphenylphosphine)palladium(0) (7.2 mg, 0.006 mmol), dimethylformamide (1.5 mL), and a 1 M aqueous solution of sodium bicarbonate (360 μL, 0.360 mmol) is sparged with argon gas and irradiated with microwaves (5-min irradiation at 120° C.). The resulting green, gelatinous mixture was filtered and evaporated to dryness under reduced pressure (˜6 mm Hg, 40° C.). The recovered residue (orange oil) is washed with diethyl ether (10 mL) to give a yellow solid. This material is dissolved in a mixture containing 25% methanol in chloroform (2.0 mL), precipitated via addition of diethyl ether (10 mL), and collected by filtration; this process is repeated once. The product is washed further with water (2×10 mL) and dried in vacuo to give (rac)-tert-Butyl[6-(9-cyclopropyl-6-fluoro-3,4-dioxo-2,3,4,9-tetrahydro-isothiazolo[5,4-b]quinolin-7-yl)-1,2,3,4-tetrahydronaphthalen-2-yl]carbamate as a yellow solid (93% purity by HPLC; the remaining material is 9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4-b]quinoline-3,4-dione). 1H NMR (CDCl3/CD3OD (12:1 v/v), 50° C.): δ1.10 (m, 2H, c-Pr CH2), 1.21 (m, 2H, c-Pr CH2), 1.48 (s, 9H, C(CH3)3), 1.76 (m, 1H, H-3), 2.10 (m, 1H, H-3), 2.68 (m, 1H, H-1), 2.86 (m, 2H, H-4), 3.08 (m, 1H, c-Pr CH), 3.12 (m, 1H, H-1), 3.94 (m, 1H, H-2), 7.11 (m, 1H, aromatic), 7.20 (m, 2H, aromatic), 7.71 (m, 1H, aromatic), 7.92 (m, 1H, aromatic). 19F{1H} NMR (CDCl3/CD3OD (12:1 v/v), 50° C.): δ−123.8 (s). NMR spectra collected at room temperature contained broad, unresolved signals. LCMS m/z calcd for C28H28FN3O4S ([M]+) 521; found 522 ([M+H]+).

(b) In air, a solution of hydrogen chloride in acetic acid (1 M, 1.8 mL) is added at room temperature to a solution of methylene chloride (0.6 mL) containing (rac)-tert-Butyl[6-(9-cyclopropyl-6-fluoro-3,4-dioxo-2,3 ,4,9-tetrahydro-isothiazolo[5 ,4-b]quinolin-7-yl)-1,2,3,4-tetrahydronaphthalen-2-yl]carbamate (11.4 mg, 0.022 mmol). A yellow precipitate begins to appear within minutes of addition of the solution of hydrogen chloride. After stirring the mixture at room temperature for 18 h, additional methylene chloride is added (2 mL). The precipitate is collected by filtration, washed with methylene chloride (4×5 mL), and dried in vacuo to give pure 31 (97% purity by HPLC analysis) as a yellow powder. mp >257-258° C. dec. 1H NMR (DMSO-d6, 60° C.): δ1.21 (m, 2H, c-Pr CH2), 1.29 (m, 2H, c-Pr CH2), 1.80 (m, 1H, H-3), 2.12 (m, 1H, H-3), 2.83 (dd, J=17.0 Hz, 10.0 Hz, 1H, H-1), 2.90 (m, 2H, H-4), 3.13 (dd, J=17.0 Hz, 6.0 Hz, 1H, H-1), 3.46 (m, 1H, H-2), 3.57 (m, 1H, c-Pr CH), 7.25 (d, J=8.0 Hz, 1H, H-8), 7.37 (s, 1H, H-5), 7.39 (d, J=8.0 Hz, 1H, H-7), 7.93 (d, JH-F=10.5 Hz, 1H, ITQ H-5), 8.01 (d, JH-F=6.5 Hz, 1H, ITQ H-8). 19F{11H} NMR (DMSO-d6, 60° C.): δ−123.3 (s). NMR spectra collected at room temperature contained broad, unresolved signals. LCMS m/z calcd for C23H20FN3O2S ([M]+) 421; found 422 ([M+H]+). HRMS m/z calcd for C23H20FN3Na02S ([M+Na]+) 444.1158; found 444.1152.

Example 6 Preparation of 9-cyclopropyl-6-fluoro-7-(2-(Pyridin-2-yl)ethynyl) isothiazolo[5,4-B]Quinoline-3,4(2H,9H)-dione (Compound 60)

Pd (PPh3)4 (4.5 mg) is added to a stirred solution of 7-bromo-9-cyclopropyl-6-fluoro-9H-isothiazolo[5,4-b]quinoline-3,4-dione (23 mg, 0.065 mmole) in DMF (1mL), followed by the addition of 2-ethynylpyridine (2 equivalents, 0.13 mmole) and diisopropyl amine (0.15 mL.) under argon at room temperature. The reaction tube is sealed and then stirred in a microwave (100W, 90° C.) until complete as monitored by LC/MS. The reaction mixture is filtered and the filtrate concentrated in vacuo. The residue is dissolved in a mixture of DMF: CHCl3: MeOH (1:8:1) and precipitated by adding diethyl ether (1-2 ml). This process is repeated three to five times. The resulting solid, 9-cyclopropyl-6-fluoro-7-(2-(pyridin-2-yl)ethynyl) isothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione, is washed with water to remove salt. The product is dried and analyzed. HPLC may be needed to obtain adequately pure product.

Example 7 Preparation of 9-cyclopropyl-7-substituted-6-fluoro-9H-isoxazolo[5,4-B]quinoline-3,4-diones

A solution of a 1-cyclopropyl-7-substituted-6-fluoro-2-methanesulfinyl-4-oxo-1,4-dihydro-quinoline-3-carboxylic acid ethyl ester (X) (10 mg, 0.023 mmol), hydroxyurea (3 mg, 0.039 mmol) and 1,8-diazabicyclo[5,4,0]undec-7-ene (DBU) (6 μl, 0.04 mmol) in methanol (5 ml) is stirred overnight at room temperature. The mixture is evaporated to dryness in vacuo. The resulting residue was washed with 2% acetic acid. The remaining solid (9-Cyclopropyl-7-substituted-6-fluoro-9H-isoxazolo[5,4-b]quinoline-3,4-dione, (Y) is collected and dried in vacuo.

Example 8 Procedure for N- and O-Alkylation of Isothiazoloquinolones

Cesium carbonate (0.25 mmol) and akyl halide (0.10-0.50 mmol) are added sequentially to a solution of isothiazoloquinolone (A) (0.10 mmol) in dimethylformamide (20 mL) at room temperature. The resulting mixture is stirred for 18 h. The reaction mixture is quenched with water (100 mL), and the product extracted with ethyl acetate (3×150 mL). The combined organic extracts are washed with brine (100 mL), dried over magnesium sulfate, and evaporated under reduced pressure to give mixtures of the desired O- (B, major) and N-alkylated (C, minor) isothiazoquinolones. The mixture is separated into the individual O- and N-alkylated products by column chromatography.

Example 9 Additional compounds of Formula I and Formula II

The following compounds, shown in Table I, are made by the methods disclosed in Examples 1 to 8. Those of ordinary skill in the art will recognize that the procedures and starting materials may be varied in order to obtain the compounds disclosed herein.

TABLE I 19F- No. Structure Name MS 1H-NMR NMR 33 9-cyclopropyl-8- methoxy-7-(4- (piperidin-1- ylmethyl) phenyl) isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C26H27N3O3S [M+] 461.58; found ([M + H]+) 462.10 1H NMR (DMSO- d6): δ 1.07 (m, 2H), 1.22 (m, 2H), 1.80 (m, 6H), 2.87 (m, 2H), 3.29 (m, 2H), 3.35 (s, 3H), 3.85 (m, 1H), 7.42 (d, J = 8.4 Hz, 1H), 7.71 (d, J = 8.4 Hz, 2H), 7.76 (d, J = 8.4 Hz, 2H), 8.06 (d, J = 8.4 Hz, 1H) 34 9-cyclopropyl-6- fluoro- 8-methoxy- 7-(2- methylpyridin- 4-yl) isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C20H16FN3O3S [M+] 397.42; found ([M + H]+) 398.54 1H NMR (DMSO- d6): δ 1.01 (m, 2H), 1.10 (m, 2H), 2.50 (s, 3H), 3.37 (s, 3H), 3.76 (m, 1H), 7.28 (d, JH-H = 5.0 Hz, 1H), 7.35 (s, 1H), 7.75 (d, JH-F = 9.5 Hz, 1H), 8.55 (d, JH-H = 5.0 Hz, 1H) 19F NMR (DMSO- d6): δ −119.0 (s) 35 9-cyclopropyl-8- methoxy-7- (1,2,3,4- tetrahydro- isoquinolin-6- yl)isothiazolo [5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C23H21N3O3S [M+] 419.50; found ([M + H]+) 420.54 1H NMR (DMSO- d6): δ 1.06 (m, 2H), 1.22 (m, 2H), 3.10 (m, 2H), 3.36 (s, 3H), 3.43 (m, 2H), 3.85 (m, 1H), 4.35 (m, 2H), 7.36 (d, J = 8.0 Hz, 1H), 7.40 (d, J = 8.5 Hz, 1H), 7.50 (m, 1H), 7.56 (m, 1H), 8.05 (d, J = 8.5 Hz, 1H) 36 9-cyclopropyl- 6-fluoro- 8-methoxy- 7-(6- methylpyridin-3- yl)isothiazolo [5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C20H16FN3O3S [M+] 397.42; found ([M + H]+) 398.07 1H NMR (DMSO- d6): δ 1.15 (m, 4H), 2.74 (s, 3H), 3.43 (s, 3H), 3.84 (m, 1H), 7.86 (m, 2H), 8.36 (d, JH,F = 9.1 Hz, 1H), 8.80 (s, br, 1H) 19F NMR (DMSO- d6): δ −119.2 (s) 37 9-cyclopropyl- 6-fIuoro- 8-methoxy- 7-((R)-1- methylisoindolin- 5- yl)isothiazolo [5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C23H20FN3O3S [M+] 437.49; found ([M + H]+) 438.49 1H NMR (DMSO- d6): δ 1.06 (m, 2H), 1.18 (m, 2H), 1.65 (d, J = 7.0 Hz, 3H), 3.39 (s, 3H), 3.81 (m, 1H), 4.48-4.67 (m, 2H), 5.00 (m, 1H),7.55 (m, 3H), 7.79 (d, J = 9.5 Hz, 1H) 19F NMR (DMSO- d6): δ −118.7 (s) 38 9-cyclopropyl- 6-fluoro- 7-(isoindolin- 5-yl)-8- methoxy- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C22H18FN3O3S [M+] 423.20; found ([M + H]+) 424.17 1H NMR (DMSO- d6): δ 1.06 (m, 2H), 1.18 (m, 2H), 3.39 (s, 3H), 3.82 (m, 1H), 4.59 (m, 4H), 7.49-7.60 (m, 3H), 7.79 (d, J = 9.5 Hz, 1H) 19F NMR (DMSO- d6): δ −118.7 (s) 39 9-cyclopropyl- 6-fIuoro- 8-methoxy- 7-(1,2,3,4- tetrahydro- isoquinolin-6- yl)isothiazolo [5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C23H20FN3O3S ([M+]) 437; found 437 ([M + H]+) 1H NMR (DMSO- d6): δ 1.06 (m, 2H), 1.18 (m, 2H), 3.09 (m, 2H), 3.40 (s, 3H), 3.43 (m, 2H), 3.81 (m, 1H), 4.35 (m, 2H), 7.38 (m, 3H), 7.78 (d, J = 9.5 Hz, 1H) 19F NMR (DMSO- d6): δ −118.7 (s) 40 9-cyclopropyl-8- methoxy-7- (2-methyl- 1,2,3,4- tetrahydro- isoquinolin-6- yl)isothiazolo [5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C24H23N3O3S ([M]+) 433; found 434 ([M + H]+) 1H NMR (DMSO- d6 80° C.): δ 1.07 (m, 2H), 1.25 (m, 2H), 2.97 (s, 3H), 3.21 (m, 2H), 3.42 (s, 3H), 3.54 (m, 2H), 3.89 (m, 1H), 4.44 (m, 2H), 7.33 (m, 1H), 7.41 (m, 1H), 7.49-7.60 (m, 2H), 8.09 (m, 1H). 41 9-cyclopropyl- 6-fluoro- 8-methoxy-7- (2-methyl- 1,2,3,4- tetrahydro- isoquinolin-6- yl)isothiazolo [5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C24H22FN3O3S ([M]+) 451; found 452 ([M + H]+) 1H NMR (DMSO- d6, 80° C.): δ 1.08 (m, 2H), 1.22 (m, 2H), 2.98 (s, 3H), 3.20 (m, 2H), 3.45 (s, 3H), 3.55 (m, 2H), 3.85 (m, 1H), 4.46 (m, 2H), 7.34-7.45 (m, 3H), 7.81 (d, JH-F = 9.5 Hz, 1H). 19F NMR (DMSO- d6, 80° C.): δ −117.8 (s). 42 7-(3-amino-4- fluorophenyl)-9- cyclopropyl-8- methoxy- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C20H16FN3O3S [M+] 397.42; found ([M + H]+) 398.10 1H NMR (DMSO- d6): δ 1.12 (m, 4H), 3.37 (s, 3H), 3.84 (m, 1H), 6.83 (m, 1H), 7.12 (m, 2H), 7.33 (d, J = 8.2 Hz, 1H), 8.01 (d, J = 8.2 Hz, 1H) 19F NMR (DMSO- d6): δ −135.4 (s) 43 9-cyclopropyl-8- methoxy-7-(2- methylpyridin-4- yl)isothiazolo [5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C20H17N3O3S [M+] 379.43; found ([M + H]+) 380.08 1H NMR (DMSO- d6): δ 1.16 (m, 4H), 3.44 (s, 3H), 3.86 (m, 1H), 7.54 (d, J = 8.1 Hz, 1H), 7.96 (d, J = 5.9 Hz, 1H), 8.02 (s, br, 1H), 8.13 (d, J = 8.1 Hz, 1H), 8.81 (d, J = 5.9 Hz, 1H) 44 (E)-4-(9- cyclopropyl-6- fluoro-3,4- dioxo- 2,3,4,9- tetrahydro- isothiazolo[5, 4-b]quinolin-7- yl)picolinalde- hyde O- methyl oxime LCMS m/z calcd for C20H15FN4O3S ([M]+) 410; found 411 ([M + H]+) 1H NMR (DMSO- d6): δ 1.29 (m, 2H), 1.40 (m, 2H), 3.68 (m, 1H), 4.00 (s, 3H), 7.80 (m, 1H), 8.09 (m, 2H), 8.24 (d, J = 6.3 Hz, 1H), 8.32 (s, 1H), 8.83 ( d, J = 5.2 Hz, 1H). 19F NMR (DMSO- d6): δ −123.7 (s). 45 9-cyclopropyl- 6-fluoro- 7-(2- (hydroxymethyl) pyridin- 4-yl)isothiazolo [5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C19H14FN3O3S ([M]+) 383; found 384 ([M + H]+) 1H NMR (DMSO- d6): δ 1.21 (m, 2H), 1.31 (m, 2H), 3.59 (m, 1H), 4.73 (s, 2H), 4.36 (bs, 2H), 7.77 (d, J = 5.3 Hz, 1H), 7.91 (m, 1H), 8.01 (d, J = 10.7 Hz, 1H), 8.16 (d, J = 6.2 Hz, 1H), 8.72 (d, J = 5.3 Hz, 1H). 19F NMR (DMSO- d6): δ −123.5 (s). 46 9-cyclopropyl- 6-fluoro- 7-(6-fluoropyridin- 3-yl)- 8-methoxy- isothiazolo [5,4-b] quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C19H13F2N3O3S [M+] 401.39; found ([M + H]+) 402.03 1H NMR (DMSO- d6): δ 1.14 (m, 4H), 3.39 (s, 3H), 3.82 (m, 1H), 7.40 (dd, J1 = 8.3 Hz, J2 = 2.7 Hz, 1H), 7.83 (d, JH,F = 9.8 Hz, 1H), 8.21 (t, J = 8.3 Hz, 1H), 8.83 (s, br, 1H) 19F NMR (DMSO- d6): δ −119.3 (s), −69.14 (s) 47 9-cyclopropyl- 6-fluoro- 7-(2-fluoropyridin- 3-yl)- 8-methoxy- isothiazolo [5,4-b] quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C19H13F2N3O3S [M+] 401.39; found ([M + H]+) 402.02 1H NMR (DMSO- d6): δ 1.12 (m, 4H), 3.39 (s, 3H), 3.82 (m, 1H), 7.40 (dd, J1 = 8.2 Hz, J2 = 2.7 Hz, 1H), 7.83 (d, JH,F = 9.9 Hz, 1H), 8.21 (t, J = 8.2 Hz, 1H), 8.43 (s, br, 1H) 19F NMR (DMSO- d6): δ −119.0 (s), −70.1 (s) 48 9-cyclopropyl- 6-fIuoro- 7-(2-fluoropyridin- 4-yl)- 8-methoxy- isothiazolo [5,4-b] quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C19H13F2N3O3S [M+] 401.39; found ([M + H]+) 402.00 1H NMR (DMSO- d6): δ 1.14 (m, 4H), 3.45 (s, 3H), 3.82 (m, 1H), 7.43 (s, 1H), 7.55 (d, J = 5.1 Hz, 1H), 7.83 (d, JH,F = 9.6 Hz, 1H), 8.43 (d, J = 5.1 Hz, 1H) 19F NMR (DMSO- d6): δ −120.0 (s), −69.2 (s) 49 9-cyclopropyl- 7-(4- (hydroxymethyl) phenyl)- 8-methoxy- isothiazolo [5,4-b] quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C21H18N2O4S ([M]+) 394; found [M + H] :1H-NMR (DMSO- D6): δ 1.072-1.096 (m, 2H), 1.222- 1.269 (m, 2H), 3.343 (s, 3H), 3.828-3.899 (m, 1H), 4.586 (s, 2H), 7.423 (d, J = 8.1 Hz, 1H), 7.464 (d, J = 8.4 Hz, 2H), 7.626 (d, J = 8.1 Hz, 2H), 8.056 (d, J = 8.1 Hz, 1H) 50 9-cyclopropyl- 7-(3- hydroxy- phenyl)-8- methoxy- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C20H16N2O4S ([M]+) 380; found [M + H] 1H-NMR (DMSO- D6): δ 1.058- 1.068 (m, 2H), 1.213-1.234 (m, 2H), 3.372 (s, 3H), 3.842-3.899 (m, 1H), 6.828 (dd, J = 1.5, 0.9 Hz, 1H), 7.039- 7.062 (m, 2H), 7.311 (t, J = 8.1 Hz, 1H), 7.377 (d, 8.4 Hz, 1H), 8.036 (d, J = 8.1 Hz, 1H), 9.575 (brs, 1H) 51 7-(4-amino- phenyl)-9- cyclopropyl- 8- methoxy- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C20H17N3O3S ([M]+) 379; found [M + H] 1H-NMR (DMSO- D6): δ 1.031- 1.084 (m, 2H), 1.192-1.234 (m, 2H), 3.351 (s, 3H), 3.826-3.898 (m, 1H), 7.101 (brs, 2H), 7.405 (d, J = 8.4 Hz, 1H), 7.596(d, J = 7.5 Hz, 2H), 8.032 (d, J = 8.4 Hz, 1H). 52 7-(4-amino- 3,5- dimethylphenyl)- 9- cyclopropyl-6- fluoro-8- methoxy- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C22H20FN3O3S [M+] 425.48; found ([M + H]+) 426.00 1H NMR (DMSO- d6): δ 1.10 (m, 4H), 2.31 (s, 6H), 3.37 (s, 3H), 3.82 (m, 1H), 7.19 (s, 2H), 7.74 (d, JH,F = 10.0 Hz, 1H) 19F NMR (DMSO- d6): δ −118.4 (s) 53 7-(4-amino- 3,5- dimethylphenyl)- 9- cyclopropyl-6- fluoro- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS (APCI): m/z calcd for C21H18FN3O2S [M+] 395.46; found ([M + H]+) 397.20 1H NMR (DMSO- d6): δ 1.29 (m, 4H), 2.28 (s, 6H), 3.82 (m, 1H), 7.28 (s, 2H), 7.90 (d, JH,F = 11.3 Hz, 1H), 8.01 (d, J = 5.9 Hz, 1H) 19F NMR (DMSO- d6): δ −123.3 (s) 54 9-cyclopropyl- 6-fluoro- 7-(4- (hydroxymethyl) phenyl)-8- methoxy isothiazolo [5,4-b] quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C21H17FN2O4S ([M]+) 412; found [M + H] 1H-NMR (DMSO- D6): δ 1.068- 1.096 (m, 2H), 1.166-1.207 (m, 2H), 3.367 (s, 3H), 3.794-3.865 (m, 1H), 4.596 (s, 2H), 7.488 (s, 4H), 7.789 (d, J = 9.3 Hz, 1H) 19F (DMSO- D6): δ −118.79 55 9-cyclopropyl- 7-(2- fluoropyridin- 4-yl)-8- methoxy- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C19H14FN3O3S ([M]+) 383; found [M + H] 1H-NMR (DMSO- D6): δ 1.091- 1.115 (m, 2H), 1.202-1.244 (m, 2H), 3.745 (s, 3H), 3.854-3.879 (m, 1H), 7.472 (s, 1H), 7.528 (d, J = 8.1 Hz, 1H), 7.656 (dt, J = 5.1, 1.5 Hz, 1H), 8.108 (d, J = 8.4 Hz, 1H), 8.401 (d, J = 5.1 Hz, 1H) 19F (DMSO- D6): δ −69.01 56 9-cyclopropyl- 6-fluoro- 7-(2-methyl- pyridin-4- yl)isothiazolo [5,4- b][1,8] naphthyridine- 3,4(2H,9H)- dione LCMS m/z calcd for C18H13FN4O2S ([M]+) 368; found 369 ([M + H]+) 1H NMR (DMSO- d6): δ 1.21 (m, 2H), 1.30 (m, 2H), 2.75 (s, 3H), 3.48 (m, 1H), 8.25 (m, 1H), 8.29 (m, 1H), 8.51 (d, J = 10.5 Hz, 1H), 8.85 (d, J = 6.0 Hz, 1H) 19F NMR (DMSO- d6): δ −127.4 (s) 57 9-cyclopropyl- 7-(2,6- dimethylpyridin- 4-yl)-6- fluoro- isothiazolo[5,4- b][1,8] naphthyridine- 3,4(2H,9H)- dione LCMS m/z calcd for C19H15FN4O2S ([M]+) 382; found 383 ([M + H]+) 1H NMR (DMSO- d6): δ 1.21 (m, 2H), 1.31 (m, 2H), 2.73 (s, 6H), 3.47 (m, 1H), 8.15 (m, 2H), 8.52 (d, J = 10.5 Hz, 1H) 19F NMR (DMSO- d6): δ −127.1 (br) 58 9-cyclopropyl-8- methoxy-7- (pyridin-3- yl)isothiazolo [5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C19H15N3O3S ([M]+) 365; found M+ 1H-NMR (DMSO- D6): δ 1.112- 1.121 (m, 2H), 1.206-1.248 (m, 2H), 3.391 (s, 3H), 3.834-3.905 (m, 1H), 7.546 (d, J = 8.1 Hz, 1H), 7.825 (dd, J = 2.4, 5.1 Hz, 1H), 8.129 (d, J = 8.1 Hz, 1H), 8.391 (d, J = 7.8 Hz, 1H), 8.796 (d, J = 4.5 Hz, 1H), 8.999 (brs, 1H). 59 9-cyclopropyl-7- (isoquinolin- 6-yl)-8- methoxy- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C23H17N3O3S ([M]+) 415, found M+ 1H-NMR (DMSO- D6): δ 1.105- 1.158 (2H, m), 1.237-1.278 (2H, m), 3.369 (3H, s), 3.860- 3.918 (1H, m), 7.601 (1H, d, J = 8.1 Hz), 7.936 (1H, dd, J = 3.6, 4.8 Hz), 8.151 (1H, d, J = 8.4 Hz), 8.271 (1H, dd, J = 6.9, 1.8 Hz), 8.383 (1H, d, J = 8.7 Hz), 8.492 (1H, d, J = 1.8 Hz), 8.958 (1H, d, J = 8.1 Hz), 9.208 (1H, dd, J = 3.6, 1.2 Hz). 60 5-(9- cyclopropyl-6- fluoro-3,4- dioxo- 2,3,4,9- tetrahydro- isothiazolo[5, 4-b]quinolin-7- yl) nicotinonitrile LCMS (APCI): m/z calcd for C19H11FN4O2S [M+] 378.38; found ([M + H]+) 378.95 1H NMR (DMSO- d6): δ 1.31 (m, 4H), 3.61 (m, 1H), 8.02 (d, JH,F = 10.5 Hz, 1H), 8.24 (d, J = 5.2 Hz, 1H), 8.70 (s, br, 1H), 9.16 (s, br, 2H) 19 F NMR (DMSO- d6): δ −124.1 (s) 61 5-(9- cyclopropyl-8- methoxy-3,4- dioxo- 2,3,4,9- tetrahydro- isothiazolo[5, 4-b]quinolin-7- yl) nicotinonitrile LCMS (APCI): m/z calcd for C20H14N4O3S [M+] 390.42; found ([M + H]+) 392.09 1H NMR (DMSO- d6): δ 1.14 (m, 4H), 3.39 (s, 3H), 3.84 (m, 1H), 7.54 (d, J = 8.2 Hz, 1H), 8.10 (d, J = 8.2 Hz, 1H), 8.60 (m, 1H), 9.09 (d, J = 2.2 Hz, 1H), 9.12 (s, J = 2.2 Hz, 1H) 62 5-(9- cyclopropyl-6- fluoro-8- methoxy-3,4- dioxo-2,3,4,9- tetrahydro- isothiazolo[5, 4-b]quinolin-7- yl) nicotinonitrile LCMS (APCI): m/z calcd for C20H13FN4O3S [M+] 408.41; found ([M + H]+) 409.15 1H NMR (DMSO- d6): δ 1.16 (m, 4H), 3.39 (s, 3H), 3.84 (m, 1H), 7.86 (d, JH.F = 9.2 Hz, 1H), 8.61 (s, 1H), 9.04 (s, 1H), 9.15 (s, 1H) 19F NMR (DMSO- d6): δ −119.4 (s) 63 9-cyclopropyl- 7-(6- fluoro-2- methylpyridin- 3-yl)-8- methoxy isothiazolo [5,4-b] quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C20H16FN3O3S ([M]+) 397; found [M + H] 1H-NMR (DMSO- D6): δ 1.043- 1.051 (2H, m), 1.195-1.213 (2H, m), 2.375 (3H, s), 3.344 (3H, s), 3.813- 3.885 (1H, m), 7.156 (1H, dd, J = 5.1, 3.0 Hz), 7.327 (1H, d, J = 8.4 Hz), 7.950 (1H, t, J = 8.4 Hz), 8.093 (1H, d, J = 8.1 Hz) 19F (DMSO- D6): δ −70.31. 64 9-cyclopropyl- 6-fluoro- 7-(6-fluoro-2- methylpyridin-3- yl)isothiazolo [5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C19H13F2N3O2S ([M]+) 385; found M+ 1H-NMR (DMSO- D6): δ 1.246- 1.288 (2H, m), 1.333-1.353 (2H, m), 2.377 (3H, s), 3.395 (3H, s), 3.373- 3.746 (1H, m), 7.211 (1H, dd, J = 5.1, 3.0 Hz), 7.973-8.051 (3H, m). 19F (DMSO- D6): δ −68.94, −121.54. 65 9-cyclopropyl- 7-(2,6- dimethylpyridin- 3-yl)-8- methoxy- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C21H19N3O3S ([M]+) 393; found [M + H] 1H-NMR (DMSO- D6): δ 1.068- 1.078 (2H, m), 1.198-1.217 (2H, m), 2.608 (3H, s), 2.767 (3H, s), 3.378 (3H, s), 3.820- 3.877 (1H, m), 7.372 (1H, d, J = 9.0 Hz), 7.791 (1H, brs), 8.143 (1H, d, J = 8.7 Hz), 8.319 (1H, brs). 66 9-cyclopropyl- 6-fluoro- 7-(pyridin-4- yl)isothiazolo [5,4-b][1,8] naphthyridine- 3,4(2H,9H)- dione LCMS m/z calcd for C17H11FN4O2S ([M]+) 354; found 355 ([M + H]+) 1H NMR (DMSO- d6): δ 1.27 (m, 2H), 1.35 (m, 2H), 3.54 (m, 1H), 8.26 (m, 2H), 8.52 (d, J = 10.5 Hz, 1H), 8.93 (m, 2H) 19F NMR (DMSO- d6): δ −128.2 (s) 67 9-cyclopropyl- 6-fIuoro- 7-(6- methylpyridin-3- yl)isothiazolo [5,4-b][1,8] naphthyridine- 3,4(2H,9H)- dione LCMS m/z calcd for C18H13FN4O2S ([M]+) 368; found 369 ([M + H]+) 1H NMR (DMSO- d6): δ 1.20 (m, 2H), 1.27 (m, 2H), 2.68 (s, 3H), 3.48 (m, 1H), 7.83 (m, 1H), 8.45 (d, J = 10.5 Hz, 1H), 8.74 (m, 1H), 9.19 (m, 1H) 19F{1H} NMR (DMSO- d6): δ −129.1 (s) 68 9-cyclopropyl- 6-fluoro- 7-(pyridin-3- yl)isothiazolo [5,4-b][1,8] naphthyridine- 3,4(2H,9H)- dione LCMS m/z calcd for C17H11FN4O2S ([M]+) 354; found 355 ([M + H]+) 1H NMR (DMSO- d6): δ 1.22 (m, 2H), 1.28 (m, 2H), 3.48 (m, 1H), 7.70 (dd, J = 7.5 Hz, J = 5.0 Hz, 1H), 8.42 (d, J = 10.5 Hz, 1H), 8.55 (m, 1H), 8.76 (m, 1H), 9.27 (m, 1H) 19F{1H} NMR (DMSO- d6): δ −129.4 (s) 69 7-(4- (aminomethyl) phenyl)- 9-cyclopropyl- 6-fluoro- isothiazolo[5,4- b][1,8] naphthyridine- 3,4(2H,9H)- dione LCMS m/z calcd for C19H15FN4O2S ([M]+) 382; found 383 ([M + H]+, 70%), 366 (100%) 1H NMR (DMSO- d6): δ 1.15- 1.31 (m, 4H), 3.49 (m, 1H), 4.08 (q, J = 6.0 Hz, 2H), 7.65 (m, 2H), 8.11 (m, 2H), 8.36 (d, J = 10.5 Hz, 1H) 19F{1H} NMR (DMSO- d6): δ −129.3 (s) 70 9-cyclopropyl- 7-(2,6- difluoropyridin- 3-yl)-8- methoxy- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C19H13F2N3O3S ([M]+) 401, found [M + H] 1H-NMR (DMSO- D6): δ 1.046- 1.068 (2H, m), 1.173-1.215 (2H, m), 3.401 (3H, s), 3.822- 3.856 (1H, m), 7.380 (1H, dd, J = 8.1, 1.2 Hz), 7.450 (1H, d, J = 8.1 Hz), 8.107 (1H, d, J = 8.1 Hz), 8.363 (1H, q, J = 8.4 Hz) 19F (DMSO- D6): δ −70.13, −70.03 71 9-cyclopropyl- 6-fluoro- 7-(3-hydroxy- phenyl)-8- methoxy- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C20H15FN2O4S ([M]+) 398; found M+ 1H-NMR (DMSO- D6): δ 1.032- 1.091 (m, 2H), 1.162-1.203 (m, 2H), 3.372 (s, 3H), 3.808-3.842 (m, 1H), 6.856- 6.935 (m, 3H), 7.331 (t, J = 7.8 Hz, 1H), 7.769 (d, J = 9.3 Hz, 1H), 9.632 (brs, 1H) 19F (DMSO- D6): δ −118.57 72 7-(4-aminophenyl)- 9- cyclopropyl- 6-fluoro-8- methoxy- isothiazolo[5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C20H16FN3O3S ([M]+) 397, found M+ 1H-NMR (DMSO- D6): δ 1.042- 1.094 (2H, m), 1.165-1.206 (2H, m), 3.375 (3H, s), 3.797- 3.868 (1H, m), 7.254 (2H, d, J = 8.1 Hz), 7.514 (2H, d, J = 7.5 Hz), 7.783 (1H, d, J = 9.6 Hz); 19F: δ −118.97. 19F (DMSO- D6): δ −118.97 73 9-cyclopropyl- 6-fluoro- 8-methoxy- 7-(pyridin-3- yl)isothiazolo [5,4- b]quinoline- 3,4(2H,9H)- dione LCMS m/z calcd for C19H14FN3O3S ([M]+) 383, found M+ 1H-NMR (DMSO- D6): δ 0.965- 1.017 (2H, m), 1.049-1.177 (2H, m), 3.343 (3H, s), 3.739- 3.810 (1H, m), 7.728 (1H, dd, J = 2.7, 7.8 Hz), 7.795 (1H, d, J = 9.3 Hz), 8.178 (1H, d, J = 8.1 Hz), 8.731 (1H, d, J = 4.8 Hz), 8.819 (1H, brs) 19F (DMSO- D6): δ −119.37

Example 10 Antimicrobial Activity of Compounds of the Invention

The antimicrobial activity of the compounds of the invention may be evaluated by a number of methods, including the following visual minimum inhibitory concentration (MIC) assay. This assay determines the minimum concentration of compound required to inhibit growth of a bacterial strain.

Minimum Inhibitory Concentration (MIC) Assay

Whole-cell antibacterial activity is determined by broth microdilution using conditions recommended by the NCCLS (see National Committee for Clinical Laboratory Standards. 2001. Performance standards for antimicrobial susceptibility testing: 11th informational supplement. Vol. 21, no. 1, M100-S11. National Committee for Clinical Laboratory Standards, Wayne, Pa.). Test compounds are dissolved in DMSO and diluted 1:50 in Mueller-Hinton II broth (Becton-Dickinson) to produce a 256 μμg/ml stock solution. In a 96-well microtiter plate, the compound solution is serially two-fold diluted in Mueller-Hinton II broth. After the compounds are diluted, a 50 μl aliquot of the test organism (˜1×106 cfu/mL) is added to each well of the microtiter plate. The final test concentrations range from 0.125-128 μg/mL. Inoculated plates are incubated in ambient air at 37° C. for 18 to 24 hours. The organisms selected for testing included laboratory strains S. aureus ATCC 29213 and E. coli ATCC 25922 (strains purchased from American Type Culture Collection, Manassas, Va.). The minimum inhibitory concentration (MIC) was determined as the lowest concentration of compound that inhibited visible growth of the test organism.

Certain compounds shown in Table I have an MIC of 1 μg/ml or less against at least one of the S. aureaus and E. coli when tested in this assay. Certain compounds disclosed in Table I exhibit an MIC of 100 ng/ml or less against at least one of the S. aureaus and E. coli when tested in this assay.

Example 11 Cell Viability Staining with Alamar Blue

To determine whether the microcidal effect observed against S. aureus and E. coli is specific to bacterial cells, compounds are screened for cell viability effects on several human cell types.

Optimal cell density is first determined by plating cells in a 96-well plate standard sterile tissue culture plates in 100 μl media, 10% FBS at six cell densities from 500 cells/well to 15,000 cells/well. A cell free well containing only media is used as a control. Cells are incubated at 37° C. in a 5% CO2 incubator for 24 hours. 10% culture volume (10 ul) of Alamar Blue (Biosource, DAL1100, 100 mL) is then added. Cells are incubated at 37° C. in a 5% CO2 incubator and read in a Victor V plate reader, 544 nm excitation, 590 nm emission, at 3, 4, and 24 hours after the addition of Alamar Blue. The cell number vs. change in fluorescence is plotted to determine linearity of signal vs. cell number. The optimal density varies between 500-15,000 cells/well depending on the specific cell type. The optimal density is selected based on the highest number of cells that is still in the linear response range.

Determination of Compound Cytotoxicity

Cells are plated at optimal cell density in a standard sterile tissue culture 96 well plate, and incubated at 37° C. O/N in a 5% CO2 incubator. 12 to 48 hours post-plating media is removed. The cells are washed 1 or 2 times with 1×PBS and replaced with fresh media containing the test compound in 1% DMSO. 24 to 72 hours after addition of compound, the media is removed, and the cells washed 1 to 2 times with 1×PBS. Fresh media containing 1/10 volume of Alamar Blue is then added. Plates are incubated 4 hours at 37° C. in a 5% CO2 incubator and read in a Victor V plate reader, 544 nm excitation, 590 nm emission.

Compounds are diluted to 20 micromolar in 1% DMSO and media and screened in duplicate to obtain single concentration cytotoxicity data. Eight concentration points from 0.78 micromolar to 100 micromolar, run in duplicate, are used to determine cyctotoxicity CC50 values. Cells with 1% DMSO and media are used as a negative control, compounds having a known CC50 against a particular cell type are used as positive controls.

The change in fluorescence vs. concentration of test compound is plotted to determine the cytotoxicity of the compound.

Sample media conditions, optimal plating densities, and positive control compounds for two cell types screened are presented in Table II.

Certain compounds disclosed in Example 1 to 6 and Example 9 exhibit CC50 values greater than 10 uM against each of the cell lines listed below when tested in this assay. Other cell types that may be used include but are not limited to Balb/3TC, CEM-SS, HeLa, HepG2, HT-29, MRC-5, SK-N-SH, U-87 MG, 293T, and Huh-7.

TABLE II Plating Positive Cell Line Media Density Control CHO 1. F-12 Nutrient Mixture (Gibco 7,000 Terfenadine (Chinese #11765-054) containing cells/well CC50 = hamster 10% FBS, 1% Pen Strep, 4.3-6.5 μM ovary) 1.5 g/L Sodium Bicarbonate 2. McCoy's 5a medium, 10% FBS and PS/Gln Hep 2 Minimum Essential Medium - 7,000 Terfenadine (laryngeal Alpha Medium (Gibco cells/well CC50 = carcinoma) # 12571-063) containing 3-5 μM 10% FBS, 1% Pen Strep, 1.5 g/L Sodium Bicarbonate

Example 12 Pharmaceutical Formulations

Examples 12A through 12G are examples of pharmaceutical compositions containing the compounds of Formula I. The abbreviation ‘A.M.’ stands for an antimicrobial compound of the present invention.

Example 12A Oral Drops

5 grams of A.M. is dissolved in 5 ml of 2-hydroxypropanoic acid and 15 ml polyethylene glycol at about 60°-80° C. After cooling to about 30°-40° C., 350 ml polyethylene glycol is added and the mixture was stirred well. A solution of 17.5 g sodium saccharin in 25 ml purified water is then added. Flavor and polyethylene glycol q.s. (quantity sufficient) to a volume of 500 ml are added while stirring to provide an oral drop solution comprising 10 mg/ml of A.M.

Example 12B Capsules

20 grams of the A.M., 6 grams sodium lauryl sulfate, 56 grams starch, 56 grams lactose, 0.8 grams colloidal silicon dioxide, and 1.2 grams magnesium stearate are vigorously stirred together. The resulting mixture is subsequently filled into 1000 suitable hardened gelatin capsules, comprising each 20 mg of the active ingredient.

Example 12C Film-Coated Tablets

Preparation of tablet core: A mixture of 10 grams of the A.M., 57 grams lactose and 20 grams starch is mixed well and thereafter humidified with a solution of 0.5 grams sodium dodecyl sulfate, and 1.0 grams polyvinylpyrrolidone (KOLLIDON-K 90) in about 20 ml of water. The wet powder mixture is sieved, dried, and sieved again. Then 100 grams microcrystalline cellulose (AVICEL) and 15 grams hydrogenated vegetable oil (STEROTEX) are added. The whole is mixed well and compressed into tablets, giving 1000 tablets, each containing 10 mg of the active ingredient.

Coating: Ethyl cellulose (0.5 grams, ETHOCEL 22 CPS) in 15 ml of dichloromethane is added to a solution of 1.0 grams methyl cellulose (Methocel 60 HG™) in 7.5 ml of denatured ethanol. Then 7.5 ml of dichloromethane and 0.25 ml 1,2,3-propanetriol are added. Polyethylene glycol (1.0 grams) is melted and dissolved in 7.5 ml of dichloromethane and added to the cellulose-containing solution. Magnesium Octadecanoate (.25 grams), 0.5 grams polyvinylpyrrolidone, and 3.0 ml of concentrated color suspension (OPASPRAY K-1-2109) are added and the whole mixture homogenized. The tablet cores are coated with this mixture in a coating apparatus.

Example 12D Injectible Solutions

(i) 1.8 grams methyl 4-hydroxybenzoate and 0.2 grams propyl 4-hydroxybenzoate are dissolved in about 0.5 L of boiling water. After cooling to about 50° C., 4 grams lactic acid, 0.05 grams propylene glycol, and 4 grams of the A.M are added while stirring. The solution is cooled to room temperature and supplemented with water for injection q.s. giving a solution containing 4 mg/ml of A.M. The solution is sterilized by filtration and filled in sterile containers.

(ii) 100.0 g of an acid salt of an A.M. of the invention is dissolved in boiling water. After cooling to about 50° C., 37.5 grams lactic acid (90% by weight) are added while stirring. The solution is cooled to room temperature and water is added to 1 L. The solution is sterilized by filtration and filled in sterile containers.

(iii) 5.00 g of an acid salt of an A.M. of the invention is dissolved in boiling water. After cooling to about 50° C., 2.20 grams lactic acid (90% by weight) are added while stirring. The solution is cooled to room temperature and water is added to 100 ml.

Example 12E Gel

A compound or salt of the invention may be formed as a gel for topical application.

A gel is prepared by suspending A.M (0.2 g-5.0 g) in benzyl alcohol at room temperature. A mixture of hydroxypropyl cellulose (2.5) grams and demineralized water (q.s. 100 g) is added to the suspension with stirring.

Example 12F Cream

Phase I contains Sorbitan monostearate (2.0 g), Polyoxyethylene (20) sorbitan monostearate (1.5 g), Synthetic spermaceti (3.0 g) Cetyl stearyl alcohol (10.0 g) and 2-Octyldodecanol (13.5 g). The phase I mixture is heated to 75° C., stirred and mixed.

Phase II contains A.M. (1.0 g). Phase II is added to phase I, stirred and suspended.

Phase III contains Benzyl alcohol (1.0 g) and demineralized water (q.s. 100 g). Phase III is heated to 75° C. and added to phase II. The cream is mixed intensively and cooled slowly to room temperature, with further stirring. After cooling to room temperature the cream is homogenized.

Example 12G Sprays

The active compound solutions or suspensions prepared according to Example 12D can also be processed to sprays. For this purpose, for example, a 60 to 90% active compound solution is mixed with 20 to 40% of the usual propellants, for example N2, N2O, CO2, propane, butane, halogenohydrocarbons and the like.

Claims

1. A compound or pharmaceutically acceptable salt thereof, wherein the compound is:

9-cyclopropyl-6-fluoro-8-methoxy-7-(2-methylpyridin-4-yl)isothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-8-methoxy-7-(6-methylpyridin-3-yl)isothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-8-methoxy-7-((R)-1-methylisoindolin-5-yl)isothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
7-(3-amino-4-fluorophenyl)-9-cyclopropyl-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-8-methoxy-7-(2-methylpyridin-4-yl)isothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
(E)-4-(9-cyclopropyl-6-fluoro-3,4-dioxo-2,3,4,9-tetrahydroisothiazolo[5,4-b]quinolin-7-yl)picolinaldehyde O-methyl oxime;
9-cyclopropyl-6-fluoro-7-(2-(hydroxymethyl)pyridin-4-yl)isothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-7-(6-fluoropyridin-3-yl)-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-7-(2-fluoropyridin-3-yl)-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-7-(2-fluoropyridin-4-yl)-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-7-(4-(hydroxymethyl)phenyl)-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-7-(3-hydroxyphenyl)-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
7-(4-aminophenyl)-9-cyclopropyl-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
7-(4-amino-3,5-dimethylphenyl)-9-cyclopropyl-6-fluoro-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
7-(4-amino-3,5-dimethylphenyl)-9-cyclopropyl-6-fluoroisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-7-(4-(hydroxymethyl)phenyl)-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-7-(2-fluoropyridin-4-yl)-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-7-(2-methylpyridin-4-yl)isothiazolo[5,4-b][1,8]naphthyridine-3,4(2H,9H)-dione;
9-cyclopropyl-7-(2,6-dimethylpyridin-4-yl)-6-fluoroisothiazolo[5,4-b][1,8]naphthyridine-3,4(2H,9H)-dione;
9-cyclopropyl-8-methoxy-7-(pyridin-3-yl)isothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
5-(9-cyclopropyl-6-fluoro-3,4-dioxo-2,3,4,9-tetrahydroisothiazolo[5,4-b]quinolin-7-yl)nicotinonitrile;
5-(9-cyclopropyl-8-methoxy-3,4-dioxo-2,3,4,9-tetrahydroisothiazolo[5,4-b]quinolin-7-yl)nicotinonitrile;
5-(9-cyclopropyl-6-fluoro-8-methoxy-3,4-dioxo-2,3,4,9-tetrahydroisothiazolo[5,4-b]quinolin-7-yl)nicotinonitrile;
9-cyclopropyl-7-(6-fluoro-2-methylpyridin-3-yl)-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-7-(6-fluoro-2-methylpyridin-3-yl)isothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-7-(2,6-dimethylpyridin-3-yl)-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-7-(pyridin-4-yl)isothiazolo[5,4-b][1,8]naphthyridine-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-7-(6-methylpyridin-3-yl)isothiazolo[5,4-b][1,8]naphthyridine-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-7-(pyridin-3-yl)isothiazolo[5,4-b][1,8]naphthyridine-3,4(2H,9H)-dione;
7-(4-(aminomethyl)phenyl)-9-cyclopropyl-6-fluoroisothiazolo[5,4-b][1,8]naphthyridine-3,4(2H,9H)-dione;
9-cyclopropyl-7-(2,6-difluoropyridin-3-yl)-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
9-cyclopropyl-6-fluoro-7-(3-hydroxyphenyl)-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione;
7-(4-aminophenyl)-9-cyclopropyl-6-fluoro-8-methoxyisothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione; or 9-cyclopropyl-6-fluoro-8-methoxy-7-(pyridin-3-yl)isothiazolo[5,4-b]quinoline-3,4(2H,9H)-dione.

2. An anti-bacterial composition comprising a compound or salt of claim 1 together with a carrier, diluent, or excipient.

3. A pharmaceutical composition comprising a compound or salt of claim 1 together with a pharmaceutically acceptable carrier, diluent, or excipient.

4. A pharmaceutical composition of claim 3, wherein the composition is formulated as an injectable fluid, an aerosol, a cream, a gel, a pill, a capsule, a tablet, a syrup, a transdermal patch, or an ophthalmic solution.

5. A composition comprising a compound or salt of claim 1 in combination with another one or more other antibacterial agent, antiprotozoal agent, antifungal agent, antiviral agent, interferon, efflux-pump inhibitor, or beta-lactamase inhibitor.

6. A package comprising the pharmaceutical composition of claim 3 in a container and instructions for using the composition to treat a patient suffering from a microorganism infection.

7. The package of claim 6 wherein the instructions are instructions for using the composition to treat a patient suffering from a bacterial infection.

8. A method for treating a bacterial infection in an animal comprising administering to the animal a therapeutically effective amount of a compound or salt of claim 1.

9. The method of claim 8 wherein the bacterial or prtozoal infection is a urinary tract infection, pyelonephritis, lower respiratory tract infection, skin infection, skin-structure infection, urethral gonococcal infection, cervical gonococcal infection, urethral chlamydial infection, cervical chlamydial infection, bone infection, joint infection, gram-negative bacterial infection, infectious diarrhea, typhoid fever, prostatitis, acute sinusitis, acute exacerbation of chronic bronchitis, pneumonia, intra-abdominal infection, gynecologic infection, or pelvic infection.

10. The method of claim 8 wherein the animal is a fish, amphibian, reptile, bird, or mammal.

11. The method of claim 8 wherein the animal is a mammal.

12. The method of claim 11 wherein the mammal is a human.

Patent History
Publication number: 20120114601
Type: Application
Filed: Jan 12, 2012
Publication Date: May 10, 2012
Applicant: Achillion Pharmaceuticals, Inc. (New Haven, CT)
Inventors: Barton James Bradbury (Wallingford, CT), Godwin Pais (Hamden, CT), Quiping Wang (Bethany, CT), Milind Deshpande (Madison, CT), Michael Pucci (Kensington, CT), Yongsheng Song (Guilford, CT), Edlaine Lucien (New Haven, CT), Jason Allan Wiles (Hamden, CT), Akihiro Hashimoto (Branford, CT)
Application Number: 13/348,703
Classifications
Current U.S. Class: Interferon (424/85.4); At Least One Of The Ring Hetero Atoms Is Chalcogen (546/83); Three Or More Hetero Atoms In The Tricyclo Ring System (514/293)
International Classification: A61K 31/4745 (20060101); A61K 31/4375 (20060101); A61K 38/21 (20060101); A61P 31/12 (20060101); A61P 31/04 (20060101); A61P 33/02 (20060101); A61P 31/10 (20060101); A61P 17/00 (20060101); A61P 13/02 (20060101); A61P 11/00 (20060101); A61P 19/08 (20060101); A61P 19/02 (20060101); A61P 1/12 (20060101); C07D 513/14 (20060101); C07D 513/04 (20060101);