WO2009124086A2 - Antibacterial drug for treatment of staphylococcus aureus and other gram-positive bacterial infections - Google Patents

Antibacterial drug for treatment of staphylococcus aureus and other gram-positive bacterial infections Download PDF

Info

Publication number
WO2009124086A2
WO2009124086A2 PCT/US2009/039023 US2009039023W WO2009124086A2 WO 2009124086 A2 WO2009124086 A2 WO 2009124086A2 US 2009039023 W US2009039023 W US 2009039023W WO 2009124086 A2 WO2009124086 A2 WO 2009124086A2
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
tetrahydro
benzo
cyano
substituted
Prior art date
Application number
PCT/US2009/039023
Other languages
French (fr)
Other versions
WO2009124086A3 (en
Inventor
Dongcheng Dai
Dennis E. Hruby
Tove C. Bolken
Rebecca L. Wilson
Kevin F. Jones
Robert Jordon
Original Assignee
Siga Technologies, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Siga Technologies, Inc. filed Critical Siga Technologies, Inc.
Publication of WO2009124086A2 publication Critical patent/WO2009124086A2/en
Publication of WO2009124086A3 publication Critical patent/WO2009124086A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/50Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D333/52Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes
    • C07D333/62Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the hetero ring
    • C07D333/68Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen

Definitions

  • This invention relates to the use of sulfonamide/thiophene derivatives and analogs, as well as compositions containing the same, for the treatment or prophylaxis of bacterial diseases associated with Gram- positive bacteria.
  • S. aureus are Gram-positive cocci that are frequently found on the skin or in the nose of healthy individuals. Under certain conditions however, such as when the skin barrier has been breached, S. aureus can cause a range of illnesses such as minor skin infections
  • S. aureus-related infections have been rising dramatically in the last decade. From 1999-2005 the estimated number of hospitalizations involving S. aureus-related infections increased from 294,570 to 411,921 cases (1). S. aureus infections are now the major cause of hospital-acquired surgical wound infections in the United States (2,3).
  • Antibiotic resistance is a continuously growing problem in the treatment of bacterial infectious diseases. This has occurred as a result of multifactorial processes, ' including over prescribing of medications by physicians, lack of patient compliance in completing dosing regimens, the indiscriminant use of antibiotics in farm animal feed, and the dissolution of geographical barriers due to increased travel (4). In addition, many of the new antibiotics coming to market are simply chemically altered versions of existing antibiotics, to which resistance is likely to occur in a very short period of time.
  • S. aureus infections can be particularly difficult to treat due to the ability of S. aureus to rapidly acquire multi-drug resistance.
  • penicillin-resistant S. aureus were identified in hospitals and then in the community (4) .
  • Within 10 years more than 80% of community- and nosocomially-acquired S. aureus were resistant to penicillin.
  • a similar pattern occurred after the introduction of methicillin in 1961, with methicillin-resistant S. aureus (MRSA) spreading rapidly into the community and across international borders (4) .
  • MRSA methicillin-resistant S. aureus
  • sortase A transpeptidase The enzyme responsible for the processing of many of these surface-expressed proteins is the sortase A transpeptidase. All sortase A substrates from numerous Gram-positive bacteria share common features (7) .
  • An N- terminal signal sequence directs the protein to the cell surface. After recognition and cleavage of the signal sequence, the surface protein is anchored in the membrane by a string of hydrophobic amino acids and a positively charged carboxy-terminal tail.
  • Membrane-associated sortase A recognizes the highly conserved LPXTG motif, cleaves the protein between the threonine and glycine, and catalyzes the formation of an amide bond with the peptidoglycan cross-bridges of the cell wall.
  • a high throughput screen was designed to identify inhibitors of S. aureus sortase A enzyme. Over 200,000 compounds were screened and several lead compounds were identified that exhibited potent inhibition of sortase enzyme both in vitro, and also decreased surface expression of several sortase-dependent virulence factors. Some of these compounds also severely inhibited the growth of aerobic and anaerobic gram positive bacteria.
  • Figure 1 shows an S. aureus sortase mutant having reduced binding to immobilized fibronectin.
  • Figure 2 shows S. aureus Protein A expression analyzed by flow cytometry after staining cells with goat anti-rabbit IgG-FITC. Numbers above the peaks indicate geometric means of fluorescence.
  • Figure 3 shows the loss of viability of Streptococcus pyogenes after growth in compound 21.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the following general Formula I or a pharmaceutically acceptable salt thereof :
  • R 1 , R 2 and R 3 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylamino, heterocycloalkylamino, arylamino, heteroarylamino, acylamino, arylacylamino, heteroarylacylamino, alkylsulfonylamino, arylsulfonylamino, acyl, arylacyl, heteroarylacylamin
  • the present invention also provides a compound having the following general Formula II or a pharmaceutically acceptable salt thereof:
  • R 6 , R 7 , R 8 , R 9 and R 10 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylamino, heterocycloalkylami.no, arylamino, heteroarylamino, acylamino, arylacylamino, heteroarylacylamino, alkylsulfonylamino, arylsulfonylamino, acyl,
  • the present invention further provides a method for the treatment or prophylaxis of a gram-positive bacterial infection or disease associated therewith, comprising administering in a therapeutically effective amount to a mammal in need thereof, a compound of Formula I below or a pharmaceutically acceptable salt thereof:
  • R 1 , R 2 and R 3 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylaraino, dialkylamino, • cycloalkylaraino, heterocycloalkylamino, arylamino, heteroarylamino, acylamino, arylacylamino, heteroarylacylamino, alkylsulfonylamino, arylsulfonyla ⁇ nino, acyl, arylacyl, heteroaryl
  • R 4 and R 5 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl and substituted carbamoyl; or R 4 and R 5 together with the nitrogen atom they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring.
  • R 1 , R 2 and R 3 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylamino, heterocycloalkylamino, arylamino, heteroarylaitu.no, acylamino, arylacylamino, heteroarylacylamino, alkylsulfonylamino, arylsulfonylamino, acyl, arylacyl, heteroaryl
  • R 4 and R 5 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl and substituted carbamoyl; or R 4 and R s together with the nitrogen atom they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring.
  • R 1 and R 2 together with the carbons they are attached to form a substituted ring.
  • R 3 is amino or nitro.
  • R 4 is hydrogen and R 5 is arylsulfonyl .
  • the compound of the present invention is selected from the group consisting of: 2- Acetylamino-4, 5-dihydro-naphtho [1,2-b] thiophene-3- carboxylic acid ethyl ester; 2-Amino-4-methyl-5-phenyl- thiophene-3-carboxylic acid ethyl ester; ethyl 2-amino- 4, 7-dihydro-5H-spiro [l-benzothiophene-6, 1 ' -cyclohexane] -3- carboxylate; 2- (4-Brorao-benzenesulfonylamino) -4,5,6,7,8,9- hexahydro-cycloocta[b]thiophene-3-carboxylic acid amide; 2-Amino-6-phenyl-4, 5, 6, 7-tetrahydro-benzo [b] thiophene-3- carboxylic acid methyl ester;
  • the compound of the present invention has the following general Formula II or a pharmaceutically acceptable salt thereof:
  • R 6 , R 7 , R 8 , R 9 and R 10 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylaraino, heterocycloalkylamino, arylamino, heteroarylamino, acylamino, arylacylamino, heteroarylacylainino, alkylsulfonylamino, arylsulfonylamino, acyl, aryl
  • each of R 7 and R B of the compound of Formula II is a halogen, more preferably a Cl. Also preferably, R 3 is Br.
  • the compound of Formula II is preferably selected from the group consisting of: 2-Bromo-N- (3-cyano- 6-methyl-4, 5, 6, 7-tetrahydro-benzo[b] thiophen-2-yl) - benzenesulfonamide; 3-Bromo-N- (3-cyano-6-methyl-4, 5, 6, 7- tetrahydro-benzo [b] thiophen-2-yl) -benzenesulfonamide; 4- Bromo-N- (3-cyano-6-methyl-4, 5, 6,7-tetrahydro- benzo[b] thiophen-2-yl) -benzenesulfonamide; 2-Chloro-N- ⁇ 3- cyano-6-methyl-4, 5, 6, 7-tetrahydro-benzo[b] thiophen-2-yl) - benzenesulfonamide; 2, 3, 4-Trichloro-N- (3-cyano-6-methyl- 4, 5, 6, 7-tetrahydro-
  • the method of the present invention is for the treatment or prophylaxis of a gram-positive bacterial infection or disease associated therewith, comprising administering in a therapeutically effective amount to a mammal in need thereof, a compound of Formula I described above .
  • the mammal is a human and the bacterial infection is a gram positive bacterial infection, more preferably belonging to a genus selected from the group consisting of Clostridium, Staphylococcus, Bacillus, and Listeria, also more preferably selected from the group consisting of a Staphylococcus aureus, Clostridium perfringens, C. difficile, Listeria monocytogenes, Bacillus anthracis, Streptococcus pneumoniae, Streptococcus pyogenes, E. faecalis infections, most preferably a Staphylococcus aureus infection.
  • the bacterial infection is associated with a condition selected from the group consisting of skin infections, pneumonia, meningitis, osteomyelitis, endocarditis, toxic shock syndrome, septicemia, and food poisoning.
  • the method of the present invention may also comprise co-administration of anti-bacterial agents such as antibiotics .
  • patient or “subject” is meant to include any mammal.
  • a “mammal,” for purposes of treatment, refers to any animal classified as a mammal, including but not limited to, humans, experimental animals including rats, mice, and guinea pigs, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, cats, cows, and the like.
  • Efficacy refers to the effectiveness of a particular treatment regime. Efficacy can be measured based on change of the course of the disease in response to an agent.
  • Successess refers to the effectiveness of a particular treatment regime. This includes a balance of efficacy, toxicity (e.g., side effects and patient tolerance of a formulation or dosage unit) , patient compliance, and the like. For a chronic administration regime to be considered “successful” it must balance different aspects of patient care and efficacy to produce a favorable patient outcome.
  • the terms "treating,” “treatment,” and the like are used herein to refer to obtaining a desired pharmacological and physiological effect.
  • the effect may be prophylactic in terms of preventing or partially preventing a disease, symptom, or condition thereof and/or may be therapeutic in terms of a partial or complete cure of a disease, condition, symptom, or adverse effect attributed to the disease.
  • treatment covers any treatment of a disease in a mammal, such as a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it, i.e., causing the clinical symptoms of the disease not to develop in a subject that may be predisposed to the disease but does not yet experience or display symptoms of the disease; ⁇ b) inhibiting the disease, i.e., arresting or reducing the development of the disease or its clinical symptoms; and (c) relieving the disease, i.e., causing regression of the disease and/or its symptoms or conditions.
  • Treating a patient's suffering from disease related to pathological inflammation is contemplated. Preventing, inhibiting, or relieving adverse effects attributed to pathological inflammation over long periods of time and/or are such caused by the physiological responses to inappropriate inflammation present in a biological system over long periods of time are also contemplated.
  • acyl refers to the groups H- C(O)-, alkyl-C(O)-, substituted alkyl-C(O)-, alkenyl-C(O) - , substituted alkenyl-C(O).-, alkynyl-C(O) -, substituted alkynyl-C (0) -, cycloalkyl-C(O) -, substituted cycloalkyl- C(O)-, aryl-C(O)-, substituted aryl-C(O)-, heteroaryl- C(O)-, substituted heteroaryl-C(O) -, heterocyclic-C(Q) -, and substituted heterocyclic-C(O)- wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl
  • Alkylamino refers to the group -NRR where each R is independently selected from the group consisting of ⁇ hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic and where each R is joined to form together with the nitrogen atom a heterocyclic or substituted heterocyclic ring wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.
  • alkenyl refers to alkenyl group preferably having from 2 to 10 carbon atoms and more preferably 2 to 6 carbon atoms and having at least 1 and preferably from 1-2 sites of alkenyl unsaturation.
  • Alkoxy refers to the group “alkyl-O-" which includes, by way of example, methoxy, ethoxy, ii-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1, 2-dimethylbutoxy, and the like.
  • Alkyl refers to linear or branched alkyl groups having from 1 to 10 carbon atoms, alternatively 1 to 6 carbon atoms. This term is exemplified by groups such as methyl, t-butyl, n-heptyl, octyl and the like.
  • Aryl or “Ar” refers to an unsaturated aromatic carbocyclic group of from 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl) which condensed rings may or may not be aromatic (e.g., 2-benzoxazolinone, 2H-1,4- benzoxazin-3 (4H) -one, and the like) provided that the point of attachment is through an aromatic ring atom.
  • a single ring e.g., phenyl
  • multiple condensed rings e.g., naphthyl or anthryl
  • aromatic e.g., 2-benzoxazolinone, 2H-1,4- benzoxazin-3 (4H) -one, and the like
  • Substituted aryl refers to aryl groups which are substituted with from 1 to 3 substituents selected from the group consisting of hydroxy, acyl, acylamino, thiocarbonylamino, acyloxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amidino, alkylamidino, thioamidino, amino, aminoacyl, aminocarbonyloxy, aminocarbonylamino, ' aminothiocarbonylamino, aryl, substituted aryl, aryloxy, substituted aryloxy, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, carboxyl, carboxylalkyl, carboxyl- substituted alkyl, carboxyl-cycloalkyl
  • Cycloalkyl refers to cyclic alkyl groups of from 3 to 8 carbon atoms having a single cyclic ring including, by way of example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclooctyl and the like. Excluded from this definition are multi-ring alkyl groups such as adamantanyl, etc.
  • Halo or "halogen” refers to fluoro, chloro, bromo and iodo.
  • Heteroaryl refers to an aromatic carbocyclic group of from 2 to 10 carbon atoms and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur within the ring or oxides thereof.
  • Such heteroaryl groups can have a single ring (e.g., pyridyl or furyl) or multiple condensed rings (e.g., indolizinyl or benzothienyl) wherein one or more of the condensed rings may or may not be aromatic provided that the point of attachment is through an aromatic ring atom.
  • the terra "heteroaryl having two nitrogen atoms in the heteroaryl, ring" refers to a heteroaryl group having two, and only two, nitrogen atoms in the heteroaryl ring and optionally containing 1 or 2 other heteroatoms in the heteroaryl ring, such as oxygen or sulfur.
  • Substituted heteroaryl refers to heteroaryl groups which are substituted with from 1 to 3 substituents selected from the group consisting of hydroxy, acyl, acylaraino, thiocarbonylamino, acyloxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amidino, alkylamidino, thioamidino, amino, aminoacyl, aminocarbonyloxy, aminocarbonylamino, aminothiocarbonylamino, aryl, substituted aryl, aryloxy, substituted aryloxy, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, carboxyl, carboxylalkyl, carboxyl-substituted alkyl, carboxyl-cycloalkyl,
  • “SuIfonyl” refers to the group -S(O) 2 R where R is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.
  • Optionally substituted means that the recited group may be unsubstituted or the recited group may be substituted.
  • “Pharmaceutically-acceptable carrier” means a carrier that is useful in preparing a pharmaceutical composition or formulation that is generally safe, nontoxic, and neither biologically nor otherwise undesirable, and includes a carrier that is acceptable for veterinary use as well as human pharmaceutical use.
  • “Pharmaceutically-acceptable cation” refers to the cation of a pharmaceutically-acceptable salt.
  • “Pharmaceutically-acceptable salt” refers to salts which retain the biological effectiveness and properties of compounds which are not biologically or otherwise undesirable.
  • Pharmaceutically-acceptable salts refer to pharmaceutically-acceptable salts of the compounds, which salts are derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like.
  • Salts derived from inorganic bases include, by way of example only, sodium, potassium, lithium, ammonium, calcium and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines, such as alkyl amines, dialkyl amines, trialkyl amines, substituted alkyl amines, di (substituted alkyl) amines, tri (substituted alkyl) amines, alkenyl amines, dialkenyl amines, trialkenyl amines, substituted alkenyl amines, di (substituted alkenyl) amines, tri (substituted alkenyl) amines, cycloalkyl amines, di (cycloalkyl) amines, tri (cycloalkyl) amines, substituted cyclo
  • Suitable amines include, by way of example only, isopropylamine, trimethyl amine, diethyl amine, tri (iso-propyl) amine, tri ⁇ n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, tromethamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, N-alkylglucamines, theobromine, purines, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like. It should also be understood that other carboxylic acid derivatives would be useful, for example, carboxylic acid amides, including carboxamides, lower alkyl carboxamides, dialkyl carboxamides, and the like.
  • Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mand ⁇ lic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene- sulfonic acid, salicylic acid, and the like.
  • a compound may act as a pro-drug.
  • Pro-drug means any compound which releases an active parent drug in vivo when such pro-drug is administered to a mammalian subject.
  • Pro-drugs are prepared by modifying functional groups present in such a way that the modifications may be cleaved in vivo to release the parent compound.
  • Pro-drugs include compounds wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that may be cleaved in vivo to regenerate the free hydroxyl, amino, or sulfhydryl group, respectively.
  • pro-drugs include, but are not limited to esters (e.g., acetate, formate, and benzoate derivatives), carbamates (e.g., N,N- dimethylamino-carbonyl) of hydroxy functional groups, and the like.
  • esters e.g., acetate, formate, and benzoate derivatives
  • carbamates e.g., N,N- dimethylamino-carbonyl
  • Treating" or “treatment” of a disease includes:
  • a “therapeutically-effective amount” means the amount of a compound that, when administered to a mammal for treating a disease, is sufficient to effect such treatment for the disease.
  • the “therapeutically-effective amount” will vary depending on the compound, the disease, and its severity and the age, weight, etc., of the mammal to be treated.
  • the compounds can be prepared from readily-available starting materials using the following general methods and procedures. It will be appreciated that where process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
  • the compounds may contain one or more chiral centers. Accordingly, if desired, such compounds can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-enriched mixtures. All such stereoisomers (and enriched mixtures) are included unless otherwise indicated. Pure stereoisomers (or enriched mixtures) may be prepared using, for example, optically-active starting materials or stereoselective reagents well-known in the art. Alternatively, racemic mixtures of such compounds can be separated using, for example, chiral column chromatography, chiral resolving agents, and the like.
  • the products contain chiral centers, they are a mixture of R, S enantiomers.
  • the chiral product can be obtained via purification techniques which separate enantiomers from a R, S mixture to provide for one or the other stereoisomer. Such techniques are known in the art.
  • the compounds can be provided as pro-drugs which convert (e.g., hydrolyze, metabolize, etc.) in vivo to a compound above .
  • compositions will be administered in a therapeutically-effective amount by any of the accepted modes of administration for these compounds.
  • the compounds can be administered by a variety of routes, including, but not limited to, oral, parenteral (e.g., subcutaneous, subdural, intravenous, intramuscular, intrathecal, intraperitoneal, intracerebral, intraarterial, or intralesional routes of administration) , topical, intranasal, localized (e.g., surgical application or surgical suppository), rectal, and pulmonary (e.g., aerosols, inhalation, or powder) . Accordingly, these compounds are effective as both injectable and oral compositions.
  • the compounds can be administered continuously by infusion or by bolus injection.
  • the actual amount of the compound i.e., the active ingredient, will depend on a number of factors, such as the severity of the disease, i.e., the condition or disease to be treated, age, and relative health of the subject, the potency of the compound used, the route and form of administration, and other factors.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population) .
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD5 0 /ED 50 .
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically-effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range which includes the IC 50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC 50 i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • compositions are administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications.
  • An amount adequate to accomplish this is defined as "therapeutically-effective dose.” Amounts effective for this use will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the inflammation, the age, weight, and general condition of the patient, and the like.
  • compositions administered to a patient are in the form of pharmaceutical compositions described supra. These compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
  • the active compound is effective over a wide dosage range and is generally administered in a pharmaceutically- or therapeutically-effective amount.
  • the therapeutic dosage of the compounds will vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician.
  • the dose will typically be in the range of about 0.5 mg to about 100 mg per kilogram body weight.
  • Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the clinician will administer the compound until a dosage is reached that achieves the desired effect.
  • the compounds are usually administered in the form of pharmaceutical compositions.
  • Pharmaceutical compositions contain as the active ingredient one or more of the compounds above, associated with one or more pharmaceutically-acceptable carriers or excipients.
  • the excipient employed is typically one suitable for administration to human subjects or other mammals.
  • the active ingredient is usually mixed with an excipient, diluted by an excipient, or enclosed within a carrier which can be in the form of a capsule, sachet, paper or other container.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium) , ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • the active compound In preparing a formulation, it may be necessary to mill the active compound to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it ordinarily is milled to a particle size of less than 200 mesh. If the' active compound is substantially water soluble, the particle size is normally adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • the compositions of the invention can be formulated so as to provide quick, sustained, or delayed- release of the active ingredient after administration to the patient by employing procedures known in the art.
  • unit dosage forms refers to physically-discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • the compound can be formulated for parenteral administration in a suitable inert carrier, such as a sterile physiological saline solution.
  • a suitable inert carrier such as a sterile physiological saline solution.
  • the dose administered will be determined by route of administration.
  • intravenous formulation should possess certain qualities aside from being just a composition in which the therapeutic agent is soluble.
  • the formulation should promote the overall stability of the active ingredient (s) , also, the manufacture of the formulation should be cost-effective. All of these factors ultimately determine the overall success and usefulness of an intravenous formulation.
  • solvents ethanol, glycerol, propylene glycol
  • stabilizers EDTA (ethylene diamine tetraacetic acid), citric acid
  • antimicrobial preservatives benzyl alcohol, methyl paraben, propyl paraben
  • buffering agents citric acid/sodium citrate, potassium hydrogen tartrate, sodium hydrogen tartrate, acetic acid/sodium acetate, maleic acid/sodium maleate, sodium hydrogen phthalate, phosphoric acid/potassium dihydrogen phosphate, phosphoric acid/disodium hydrogen phosphate
  • tonicity modifiers sodium chloride, mannitol, dextrose.
  • the presence of a buffer is necessary to maintain the aqueous pH in the range of from about 4 to about 8.
  • the buffer system is generally a mixture of a weak acid and a soluble salt thereof, e.g., sodium citrate/citric acid; or the raonocation or dication salt of a dibasic acid, e.g., potassium hydrogen tartrate; sodium hydrogen tartrate, phosphoric acid/potassium dihydrogen phosphate, and phosphoric acid/disodium hydrogen phosphate.
  • the amount of buffer system used is dependent on (1) the desired pH; and (2) the amount of drug. Generally, the amount o,f buffer used is able to maintain a formulation pH in the range of 4 to 8. Generally, a 1:1 to 10:1 mole ratio of buffer (where the moles of buffer are taken as the combined moles of the buffer ingredients, e.g., sodium citrate and citric acid) to drug is used.
  • a useful buffer is sodium citrate/citric acid in the range of 5 to 50 mg per ml. sodium citrate to 1 to 15 mg per ml. citric acid, sufficient to maintain an aqueous pH of 4-6 of the composition.
  • the buffer agent may also be present to prevent the precipitation of the drug through soluble metal complex formation with dissolved metal ions, e.g., Ca, Mg, Fe, Al, Ba, which may leach out of glass containers or rubber stoppers or be present in ordinary tap water.
  • the agent may act as a competitive complexing agent with the drug and produce a soluble metal complex leading to the presence of undesirable particulates.
  • the presence of an agent, e.g., sodium chloride in an amount of about of 1-8 r ⁇ g/ml, to adjust the tonicity to the same value of human blood may be required to avoid the swelling or shrinkage of erythrocytes upon administration of the intravenous formulation leading to undesirable side effects such as nausea or diarrhea and possibly to associated blood disorders.
  • the tonicity of the formulation matches that of human blood which is in the range of 282 to 288 m ⁇ sm/kg, and in general is 285 m ⁇ sm/kg, which is equivalent to the osmotic pressure corresponding to a 0.9% solution of sodium chloride.
  • An intravenous formulation can be administered by direct intravenous injection, i.v. bolus, or can be administered by infusion by addition to an appropriate infusion solution such as 0.9% sodium chloride injection or other compatible infusion solution.
  • compositions are preferably formulated in a unit dosage form, each dosage containing from about 5 to about 100 mg, more usually about 10 to about 30 mg, of the active ingredient.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • the active compound is effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood / however, that the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 2000 mg of the active ingredient.
  • the tablets or pills may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • liquid forms in which the novel compositions may be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • compositions for inhalation or insufflation include solutions and suspensions in pharraaceutically- acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically-acceptable excipients as described supra.
  • Compositions in pharmaceutically-acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device may be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered from devices which deliver the formulation in an appropriate manner.
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the compounds, which matrices are in the form of shaped articles, e.g., films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (e.g., poly(2- hydroxyethyl- methacrylate) as described by Langer et al., J. Biomed. Mater. Res. 15: 167-277 (1981) and Langer, Chem. Tech. 12: 98-105 (1982) or poly (vinyl alcohol)), polylactides (U.S. Patent No.
  • the compounds can be administered in a sustained-release form, for example a depot injection, implant preparation, or osmotic pump, which can be formulated in such a manner as to permit a sustained- release of the active ingredient.
  • Implants for sustained- release formulations are well-known in the art. Implants may be formulated as, including but not limited to, microspheres, slabs, with biodegradable or nonbiodegradable polymers. For example, polymers of lactic acid and/or glycolic acid form an erodible polymer that is well-tolerated by the host.
  • Transdermal delivery devices may also be employed. Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds in controlled amounts.
  • the construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Patent No. 5,023,252, issued June 11, 1991, herein incorporated by reference. Such patches may be constructed for continuous, pulsatile, or on-demand delivery of pharmaceutical agents.
  • Direct or indirect placement techniques may be used when it is desirable or necessary to introduce the pharmaceutical composition to the brain.
  • Direct techniques usually involve placement of a drug delivery catheter into the host's ventricular system to bypass the blood-brain barrier.
  • One such implantable delivery system used for the transport of biological factors to specific anatomical regions of the body is described in U.S. Patent No. 5,011,472, which is herein incorporated by reference.
  • Indirect techniques usually involve formulating the compositions to provide for drug latentiation by the conversion of hydrophilic drugs into lipid-soluble drugs .
  • Latentiation is generally achieved through blocking of the hydroxy, carbonyl, sulfate, and primary amine groups present on the drug to render the drug more lipid-soluble and amenable to transportation across the blood-brain barrier.
  • the delivery of hydrophilic drugs may be enhanced by intra-arterial infusion of hypertonic solutions which can transiently open the blo ⁇ d-brain barrier.
  • the compounds may be encapsulated, introduced into the lumen of liposomes, prepared as a colloid, or other conventional techniques may be employed which provide an extended serum half-life of the compounds.
  • a variety of methods are available for preparing liposomes, as described in, e.g., Szoka et al., U.S. Patent Nos. 4,235,871, 4,501,728 and 4,837,028 each of which is incorporated herein by reference.
  • compositions are suitable for use in a variety of drug delivery systems. Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences, Mace Publishing Company, Philadelphia, PA, 17th ed. (1985).
  • a tablet formula is prepared using the ingredients below:
  • a dry powder inhaler formulation is prepared containing the following components:
  • the active mixture is mixed with the lactose and the mixture is added to a dry powder inhaling appliance.
  • Capsules each containing 40 mg of medicament, are made as follows:
  • the active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of nominal 2.O g capacity and allowed to cool.
  • the medicament, sucrose, and xanthan gum are blended, passed through a NO. 10 mesh U.S. sieve, and then mixed with a previously made solution of the microcrystalline cellulose and sodium carboxymethyl cellulose in water.
  • the sodium benzoate, flavor, and color are diluted with some of the water and added with stirring. Sufficient water is then added to produce the required volume.
  • Hard gelatin tablets each containing 15 mg of active ingredient, are made as follows:
  • An intravenous formulation may be prepared as follows :
  • Therapeutic compound compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle or similar sharp instrument.
  • a topical formulation may be prepared as follows :
  • the white soft paraffin is heated until molten.
  • the liquid paraffin and emulsifying wax are incorporated and stirred until dissolved.
  • the active ingredient is added and stirring is continued until dispersed.
  • the mixture is then cooled until solid.
  • An aerosol formulation may be prepared as follows: ft solution of the candidate compound in 0.5% sodium bicarbonate/saline (w/v) at a concentration of 30.0 mg/ml ⁇ is prepared using the following procedure:
  • Example 13 A High throughput fluorescence quench biochemical assay to identify inhibitors of 5. aureus sortase A enzyme.
  • a high throughput screen was developed in order to identify compounds in the SIGA chemical compound library that inhibited the cleavage activity of S. aureus sortase A enzyme.
  • the N-terminal membrane anchor (a.a. 2- 25) of S. aureus sortase A was replaced with a six- histidine tag (SrtA ⁇ N ) and was cloned into an E. coli expression vector. Sortase A and was purified as previously described (11) .
  • sortase peptide (2-aminobenzoyl-LPTEGE-diaminopropionic acid- dinitrophenyl-NH 2 (Abz-LPETGE-Dap (DNE) -NH 2 )
  • Sortase buffer 40 mM Tris-Cl, pH 8.0, 150 mM NaCl, 1 mM TriGlycine, 5 mM CaCl 2
  • a total of 80 compounds were tested per 96-well plate.
  • Sortase inhibitors identified in the primary screen were retested in secondary assays to determine IC 50 (concentration at which compounds inhibit sortase activity 50%) and CC 50 (50% cytotoxic concentration in mammalian cells) values.
  • the sortase assay was carried out as described above except that half-log dilutions of compounds were added to the sortase reactions in triplicate at concentrations ranging from 25 ⁇ M to 0.008 ⁇ M.
  • 96-well plates were seeded with HEK-293 cells at subconfluent densities. For control wells, cells were diluted 1:2 and 1:4. Cells were incubated overnight at 37 0 C before adding compound in half-log dilutions starting from a 25 ⁇ M stock concentration. Plates were incubated at 37 0 C for 4 days. On the last day of incubation, alamarBlueTM (Invitrogen) was added to each well and plates incubated overnight at 37 0 C.
  • Metabolically active cells cause reduction of the REDOX indicator in the reagent alamarBlueTM to change from an oxidized (blue) form to a reduced (red) form. This color change can be detected by reading absorbance at A 570 and Asoo. Plates were read on an Envision spectrophotometer the next day.
  • Example 14 - Biological assays used to examine the effects of compounds on sortase A function in S. aureus.
  • Bioassays I and II Fibronectin and fibrinogen binding assays.
  • Fibronectin binding protein A and B are sortase-dependent surface proteins of S. aureus that mediate bacterial adhesion and invasion of epithelial cells, endothelial cells, fibroblasts and osteoblasts by forming a fibronectin bridge from the bacterial cell surface to a fibronectin-binding integrin on the host cell (12).
  • S. aureus that express FnBPA and FnBPB are able to bind to immobilized fibronectin.
  • An S. aureus srtA mutant does not express these proteins, and shows greatly reduced binding to fibronectin.
  • sortase-dependent surface proteins are clumping factors A and B (CIfA and CIfB) .
  • CIfA and CIfB are fibrinogen-binding proteins that mediate fibrinogen-dependent adhesion and clumping of S. aureus on blood clots and damaged tissue.
  • plate binding assays were developed.
  • aureus bacteria bound to the wells are able to convert the alamarBlueTM from a blue to a pink color at a faster rate than the S. aureus srtA mutant.
  • the sortase mutant showed an approximately 80% reduced ability to bind to fibronectin in this assay ( Figure 1) . Transformation of the sortase mutant with a plasmid expressing S. aureus sortase A partially restored the ability of the mutant to bind to fibronectin ⁇ ( Figure 1). Similar results were obtained in the fibrinogen-binding assay with the sortase mutant showing an 80% decrease in the ability to bind immobilized fibrinogen.
  • Bioassay III Surface Protein A detection by flow cytometry
  • Protein A is a highly-expressed sortase A- dependent surface protein that binds the Fc region of IgG molecules which disrupts opsonization and phagocytosis of S. aureus (13) .
  • Cultures of S. aureus RN4220 wild type, srtA. ⁇ mutant, complemented srtA mutant, or a protein A mutant (spa:ery) were grown overnight in CDM. Cultures were washed in FACS buffer and suspended in a 1:50 ' dilution of goat anti-rabbit IgG-FITC (Biorad) .
  • FIG. 1 shows that wild type S. aureus RN4220 exhibits an approximately 7- fold increased shift in fluorescence compared to the sortase mutant after incubation with antibody.
  • An S. aureus spa mutant which expresses no protein A, showed slightly lower fluorescence compared to the sortase mutant. This would be expected since a sortase mutant still expresses protein A, some of which remains associated with the cell wall.
  • Example 15 Growth inhibition assays, inhibition of sortase A function in biological assays, solubility and PK on compounds
  • S. aureus RN4220 was grown in CDM containing 1% DMSO or in compounds at 100 ⁇ M, 75 UM, 50 ⁇ M, 25 ⁇ M, 12.5 ⁇ M, and 6.2 ⁇ M concentrations (1% DMSO) in flat-bottom 96 well plates. Plates were incubated at 37 C C with shaking. The absorbance at O.D. ⁇ oo was read every hour for 7 hours and at 24 hours. For those compounds that showed severe growth inhibition, MIC assays using the broth microdilution method were performed as described (14) .
  • Time kill assays were performed to determine the ability of compound 21 to inhibit the growth of S. pyogenes. Cultures were grown overnight and diluted to 5.0 x 10 5 cfu/ml in Mueller-Hinton broth supplemented with 5% lysed horse blood. Compound 21 (at 7 ⁇ g/ml or vancomycin at 2 ⁇ g/ml, or DMSO as a negative control were added to the cultures. Cultures were grown at 35° C statically and 50 ⁇ l aliquots were taken and plated on brain-heart infusion agar. Plates were incubated at 37° C and colonies were enumerated after 48 hr. Figure 3 shows that after only 2 hours of growth of S. pyogenes in 7 ⁇ g/ml Compound 21, no viable colonies could be recovered.
  • Compounds 9-11 showed peak plasma concentrations (C max ) at 1.3, 2, and 2.3 hr, respectively, following administration. As shown in Table 3, each compound achieved significant oral exposure with a greater C max and Al)C( 0 - 24 ) observed for Compound 10. These plasma concentrations are well above the effective concentrations of Compounds 9 and 11 for inhibition of surface protein expression ( ⁇ 5 ⁇ g/ml) as well as the MIC value for growth inhibition of S. aureus (-13 ⁇ g/ml) . Plasma levels of Compound 10 are also above the effective concentration (9.8 ⁇ g/ml) and MIC (39 ⁇ g/ml) for S. aureus Newman as well as methicillin-resistant S. aureus 328 (see Tables 1- 3).

Abstract

Compounds, methods and pharmaceutical compositions for treating bacterial infections, by administering certain compounds in therapeutically effective amounts are disclosed. Methods for preparing the compounds and methods of using the compounds and pharmaceutical compositions thereof are also disclosed. In particular, the treatment and prophylaxis of bacterial infections such as caused by the Staphylococcus aureus family is disclosed.

Description

Antibacterial Drug for Treatment of Staphylococcus aureus and Other Gram-positive Bacterial Infections
CROSS REFERENCE TO RELATED APPLICATIONS [00001] This application claims priority to and benefit of U.S. Provisional Application No. 61/042,397, filed April 4, 2008, the contents of which are hereby incorporated by reference in their entirety.
FIELD OF THE INVENTION
[00002] This invention relates to the use of sulfonamide/thiophene derivatives and analogs, as well as compositions containing the same, for the treatment or prophylaxis of bacterial diseases associated with Gram- positive bacteria.
BACKGROUND OF THE INVENTION
[00003] S. aureus are Gram-positive cocci that are frequently found on the skin or in the nose of healthy individuals. Under certain conditions however, such as when the skin barrier has been breached, S. aureus can cause a range of illnesses such as minor skin infections
(impetigo, boils, scalded skin syndrome), pneumonia, meningitis, osteomyelitis, endocarditis, toxic shock syndrome, septicemia, and food poisoning. The number of S. aureus-related infections has been rising dramatically in the last decade. From 1999-2005 the estimated number of hospitalizations involving S. aureus-related infections increased from 294,570 to 411,921 cases (1). S. aureus infections are now the major cause of hospital-acquired surgical wound infections in the United States (2,3).
[00004] Antibiotic resistance is a continuously growing problem in the treatment of bacterial infectious diseases. This has occurred as a result of multifactorial processes,' including over prescribing of medications by physicians, lack of patient compliance in completing dosing regimens, the indiscriminant use of antibiotics in farm animal feed, and the dissolution of geographical barriers due to increased travel (4). In addition, many of the new antibiotics coming to market are simply chemically altered versions of existing antibiotics, to which resistance is likely to occur in a very short period of time.
[00005] S. aureus infections can be particularly difficult to treat due to the ability of S. aureus to rapidly acquire multi-drug resistance. For example, just two years after the introduction of penicillin in the 1940s, penicillin-resistant S. aureus were identified in hospitals and then in the community (4) . Within 10 years more than 80% of community- and nosocomially-acquired S. aureus were resistant to penicillin. A similar pattern occurred after the introduction of methicillin in 1961, with methicillin-resistant S. aureus (MRSA) spreading rapidly into the community and across international borders (4) . The rate of new instances of resistance is alarming. From 1999.to 2005, the number of MRSA-related hospitalizations more than doubled (1) . A recent study reports that 94,3_60 Americans developed serious MRSA infections in 2005 and about 18,650 of them died (5). Most of these cases were acquired during hospitalization or health care procedures while about 14 percent of the infections were community-acquired. Vancomycin is currently the antibiotic preferred for the treatment of most MRSA infections but the isolation of S. aureus with resistance to intermediate levels of vancomycin is increasingly being reported (6) . [00006] It is essential that new antibacterial targets be identified in order to combat the rapid spread of antibiotic-resistant S. aureus. To this end, SIGA is developing a new class of Gram-positive-specific antibiotics targeted at the processing of bacterial surface proteins. These surface proteins are important virulence determinants that are involved in adherence to tissue, internalization into host cell surfaces, and immune avoidance. Each of these is critical for staphylococcal pathogenesis.
[00007] The enzyme responsible for the processing of many of these surface-expressed proteins is the sortase A transpeptidase. All sortase A substrates from numerous Gram-positive bacteria share common features (7) . An N- terminal signal sequence directs the protein to the cell surface. After recognition and cleavage of the signal sequence, the surface protein is anchored in the membrane by a string of hydrophobic amino acids and a positively charged carboxy-terminal tail. Membrane-associated sortase A recognizes the highly conserved LPXTG motif, cleaves the protein between the threonine and glycine, and catalyzes the formation of an amide bond with the peptidoglycan cross-bridges of the cell wall.
[00008] The importance of sortase A in S. aureus virulence has been established in numerous animal models. Intravenous injection of C57BL/6, Swiss-Webster, or C3H/HeJ mice with a 5. aureus Newman srtA mutant SKM3 led to greatly reduced numbers of viable bacteria in kidney abscesses compared to infection with wild type Newman (8, 9) . Likewise, in a murine arthritis model the severity of arthritis exhibited as joint inflammation was decreased significantly in NMRI mice that had been infected with a srtA mutant (9, 10). Furthermore, studies by Weiss et al. report that a srtA mutant was also attenuated in its ability to establish colonization in rat cardiac vegetations (9) . From these studies and many others, it is apparent that the ability of S. aureus to interact with host cell tissues and proteins through its sortase- dependent surface-anchored proteins is crucial to its ability to establish infection and cause disease.
[00009] A high throughput screen was designed to identify inhibitors of S. aureus sortase A enzyme. Over 200,000 compounds were screened and several lead compounds were identified that exhibited potent inhibition of sortase enzyme both in vitro, and also decreased surface expression of several sortase-dependent virulence factors. Some of these compounds also severely inhibited the growth of aerobic and anaerobic gram positive bacteria.
BRIEF DESCRIPTION OF THE DRAWINGS
[000010] Figure 1 shows an S. aureus sortase mutant having reduced binding to immobilized fibronectin.
[000011] Figure 2 shows S. aureus Protein A expression analyzed by flow cytometry after staining cells with goat anti-rabbit IgG-FITC. Numbers above the peaks indicate geometric means of fluorescence.
[000012] Figure 3 shows the loss of viability of Streptococcus pyogenes after growth in compound 21.
SUMMARY OF THE INVENTION
[000013] The present invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the following general Formula I or a pharmaceutically acceptable salt thereof :
Figure imgf000006_0001
wherein R1, R2 and R3 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylamino, heterocycloalkylamino, arylamino, heteroarylamino, acylamino, arylacylamino, heteroarylacylamino, alkylsulfonylamino, arylsulfonylamino, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aπiinosulfonyl, substituted aminosulfonyl, carboxy, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl, substituted carbamoyl, halogen, cyano, isocyano and nitro; or R1 and R2 together with the carbons they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring and may be fused with an aromatic or aliphatic ring; and R4 and R5 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl and substituted carbamoyl; or R4 and R5 together with the nitrogen atom they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring.
[000014] The present invention also provides a compound having the following general Formula II or a pharmaceutically acceptable salt thereof:
Figure imgf000007_0001
wherein R6, R7, R8, R9 and R10 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylamino, heterocycloalkylami.no, arylamino, heteroarylamino, acylamino, arylacylamino, heteroarylacylamino, alkylsulfonylamino, arylsulfonylamino, acyl, arylacyl, heteroarylacylr alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, carboxy, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl, substituted carbamoyl, halogen, cyano, isocyano and nitro; and
optionally at least one of R6 and R7; R7 and R8; R8 and R9; and R9 and R10 together with the carbons they are attached to form a substituted or unsubstituted ring, which' optionally include one or more heteroatoms in the ring and optionally are fused with an aromatic or aliphatic ring, with a proviso that at least one of R6, R7, R8, R9 and R10 is halogen, and if R6, R7, R9 and R10 are H, R8 is not F or Cl, or if R7, R8, and R10 are H, R6 and R9 are not both Cl.
[000015] The present invention further provides a method for the treatment or prophylaxis of a gram-positive bacterial infection or disease associated therewith, comprising administering in a therapeutically effective amount to a mammal in need thereof, a compound of Formula I below or a pharmaceutically acceptable salt thereof:
Figure imgf000008_0001
Formula I
wherein R1, R2 and R3 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylaraino, dialkylamino, • cycloalkylaraino, heterocycloalkylamino, arylamino, heteroarylamino, acylamino, arylacylamino, heteroarylacylamino, alkylsulfonylamino, arylsulfonyla∑nino, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, carboxy, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl, substituted carbamoyl, halogen, cyano, isocyano and nitro; or R1 and R2 together with the carbons they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring and may be fused with an aromatic or aliphatic ring; and
[000016] R4 and R5 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl and substituted carbamoyl; or R4 and R5 together with the nitrogen atom they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring.
[000017] Other objects and advantages of the present invention will become apparent from the following description and appended claims.
DETAILED DESCRIPTION OF THE INVENTION
[000018] The compounds of the invention are of the following general Formula I:
Figure imgf000010_0001
wherein R1, R2 and R3 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylamino, heterocycloalkylamino, arylamino, heteroarylaitu.no, acylamino, arylacylamino, heteroarylacylamino, alkylsulfonylamino, arylsulfonylamino, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, carboxy, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl, substituted carbamoyl, halogen, cyano, isocyano and nitro; or R3 is as above and R1 and R2 together with the carbons they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring and may be fused with an aromatic or aliphatic ring; and
R4 and R5 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl and substituted carbamoyl; or R4 and Rs together with the nitrogen atom they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring.
[000019] Preferably, R1 and R2 together with the carbons they are attached to form a substituted ring. Also preferably, R3 is amino or nitro.
[000020] Preferably, R4 is hydrogen and R5 is arylsulfonyl .
[000021] Preferably, the compound of the present invention is selected from the group consisting of: 2- Acetylamino-4, 5-dihydro-naphtho [1,2-b] thiophene-3- carboxylic acid ethyl ester; 2-Amino-4-methyl-5-phenyl- thiophene-3-carboxylic acid ethyl ester; ethyl 2-amino- 4, 7-dihydro-5H-spiro [l-benzothiophene-6, 1 ' -cyclohexane] -3- carboxylate; 2- (4-Brorao-benzenesulfonylamino) -4,5,6,7,8,9- hexahydro-cycloocta[b]thiophene-3-carboxylic acid amide; 2-Amino-6-phenyl-4, 5, 6, 7-tetrahydro-benzo [b] thiophene-3- carboxylic acid methyl ester; 2-Amino-4- (4-sec-butyl- phenyl) -5-inethyl-thiophene-3-carboxylic acid methyl ester; 2- (3, 4-Dichloro-benzenesulfonylamino) -6-methyl-4, 5, 6, 7- tetrahydro-benzo[b] thiophene-3-carboxylic acid amide; 2- (4-Isopropyl-benzenesulfonylamino)-6-methyl-4,5, 6,7- tetrahydro-benzo[b] thiophene-3-carboxylic acid amide; 2- (4-Bromo-3-methyl-benzenesulfonylamino) -6-methyl-4, 5, 6, 7- tetrahydro-benzofb] thiophene-3-carboxylic acid amide; 2- (5-Bromo-2-methoxy-benzenesulfonylamino) -6-methyl-4, 5, 6, 7- tetrahydro-benzo[b] thiophene-3-carboxylic acid amide; 6- Methyl-2- {naphthalene-2-sulfonylamino) -4,5,6, 7-tetrahydro- benzo [b] thiophene-3-carboxylic acid amide; 2-(4-Chloro- benzenesulfonylamino) -6-methyl-4,5,6, 7-tetrahydro- benzo[b] thiophene-3-carboxylic acid amide; and 2-(3- Chloro-benzenesulfonylamino) -6-methyl-4, 5, 6, 7-tetrahydro- benzo [b] thiophene-3-carboxylic acid amide.
[000022] Also preferably, the compound of the present invention has the following general Formula II or a pharmaceutically acceptable salt thereof:
Figure imgf000012_0001
Formula II
wherein R6, R7, R8, R9 and R10 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylaraino, heterocycloalkylamino, arylamino, heteroarylamino, acylamino, arylacylamino, heteroarylacylainino, alkylsulfonylamino, arylsulfonylamino, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, carboxy, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl, substituted carbamoyl, halogen, cyano, isocyano and nitro; and
optionally at least one of R6 and R7; R7 and R8; R8 and R9; and R9 and R10 together with the carbons they are attached to form a substituted or unsubstituted ring, which optionally include one or more heteroatoms in the ring and optionally are fused with an aromatic or aliphatic ring, with a proviso that at least one of R6, R7, R8, R9 and R10 is halogen, and if R6, R7, R9 and R10 are H, Rβ is not F or Cl, or if R7, R8, and R10 are H, R6 and R9 are not both Cl.
[000023] Preferably, each of R7 and RB of the compound of Formula II is a halogen, more preferably a Cl. Also preferably, R3 is Br.
[000024] The compound of Formula II is preferably selected from the group consisting of: 2-Bromo-N- (3-cyano- 6-methyl-4, 5, 6, 7-tetrahydro-benzo[b] thiophen-2-yl) - benzenesulfonamide; 3-Bromo-N- (3-cyano-6-methyl-4, 5, 6, 7- tetrahydro-benzo [b] thiophen-2-yl) -benzenesulfonamide; 4- Bromo-N- (3-cyano-6-methyl-4, 5, 6,7-tetrahydro- benzo[b] thiophen-2-yl) -benzenesulfonamide; 2-Chloro-N-{3- cyano-6-methyl-4, 5, 6, 7-tetrahydro-benzo[b] thiophen-2-yl) - benzenesulfonamide; 2, 3, 4-Trichloro-N- (3-cyano-6-methyl- 4, 5, 6, 7-tetrahydro-benzo[b] thiophen-2-yl) - benzenesulfonamide; 2, 4, 5-Trichloro-N- (3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 3, 4-Dibromo-N-(3-cyano-β-methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2, 5-Dibromo-N- (3-cyano-6-methyl- 4, 5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2, 4-Dibromo-N-(3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2,4, 6-Tribromo-N- (3-cyano-6-methyl- 4,5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2,4, 5-Tribromo-N- (3-cyano-6-methyl- •4,5,6,7-tetrahydro-benzo[b]thiophen-2-yl)- benzenesulfonamide; 3, 4-Dichloro-N- (3-cyano-6-methyl- 4,5, 6,7-tetrahydro-benzo[b]thiophen-2-yl)- benzenesulfonamide; and 4-Bromo-N- (3-cyano-6-methyl- 4,5, 6,7-tetrahydro-benzo[b] thiophen-2-yl) -3-methyl- benzenesulfonamide.
[000025] The method of the present invention is for the treatment or prophylaxis of a gram-positive bacterial infection or disease associated therewith, comprising administering in a therapeutically effective amount to a mammal in need thereof, a compound of Formula I described above .
[000026] Preferably, the mammal is a human and the bacterial infection is a gram positive bacterial infection, more preferably belonging to a genus selected from the group consisting of Clostridium, Staphylococcus, Bacillus, and Listeria, also more preferably selected from the group consisting of a Staphylococcus aureus, Clostridium perfringens, C. difficile, Listeria monocytogenes, Bacillus anthracis, Streptococcus pneumoniae, Streptococcus pyogenes, E. faecalis infections, most preferably a Staphylococcus aureus infection.
[000027] Preferably, the bacterial infection is associated with a condition selected from the group consisting of skin infections, pneumonia, meningitis, osteomyelitis, endocarditis, toxic shock syndrome, septicemia, and food poisoning. [000028] The method of the present invention may also comprise co-administration of anti-bacterial agents such as antibiotics .
Definitions
[000029] In accordance with this detailed description, the following abbreviations and definitions apply. It must be noted that as used herein, the singular forms "a," wan," and "the" include plural referents unless the context clearly dictates otherwise.
[000030] The publications discussed herein are provided solely for their disclosure. Nothing herein is to be construed as an admission regarding antedating the publications. Further, the dates of publication provided may be different from the actual publication dates, which may need to be independently confirmed.
[000031] Where a range of values is provided, it is understood that each intervening value is encompassed. The upper and lower limits of these smaller ranges may independently be included in the smaller, subject to any specifically-excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either both of those included limits are also included in the invention. Also contemplated are any values that fall within the cited ranges.
[000032] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. Any methods and materials similar or equivalent to those described herein can also be used in practice or testing. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.
[000033] By "patient" or "subject" is meant to include any mammal. A "mammal," for purposes of treatment, refers to any animal classified as a mammal, including but not limited to, humans, experimental animals including rats, mice, and guinea pigs, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, cats, cows, and the like.
[000034] The term "efficacy" as used herein refers to the effectiveness of a particular treatment regime. Efficacy can be measured based on change of the course of the disease in response to an agent.
[000035] The term "success" as used herein in the context of a chronic treatment regime refers to the effectiveness of a particular treatment regime. This includes a balance of efficacy, toxicity (e.g., side effects and patient tolerance of a formulation or dosage unit) , patient compliance, and the like. For a chronic administration regime to be considered "successful" it must balance different aspects of patient care and efficacy to produce a favorable patient outcome.
[000036] The terms "treating," "treatment," and the like are used herein to refer to obtaining a desired pharmacological and physiological effect. The effect may be prophylactic in terms of preventing or partially preventing a disease, symptom, or condition thereof and/or may be therapeutic in terms of a partial or complete cure of a disease, condition, symptom, or adverse effect attributed to the disease. The term "treatment," as used herein, covers any treatment of a disease in a mammal, such as a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it, i.e., causing the clinical symptoms of the disease not to develop in a subject that may be predisposed to the disease but does not yet experience or display symptoms of the disease; {b) inhibiting the disease, i.e., arresting or reducing the development of the disease or its clinical symptoms; and (c) relieving the disease, i.e., causing regression of the disease and/or its symptoms or conditions. Treating a patient's suffering from disease related to pathological inflammation is contemplated. Preventing, inhibiting, or relieving adverse effects attributed to pathological inflammation over long periods of time and/or are such caused by the physiological responses to inappropriate inflammation present in a biological system over long periods of time are also contemplated.
[000037] As used herein, "acyl" refers to the groups H- C(O)-, alkyl-C(O)-, substituted alkyl-C(O)-, alkenyl-C(O) - , substituted alkenyl-C(O).-, alkynyl-C(O) -, substituted alkynyl-C (0) -, cycloalkyl-C(O) -, substituted cycloalkyl- C(O)-, aryl-C(O)-, substituted aryl-C(O)-, heteroaryl- C(O)-, substituted heteroaryl-C(O) -, heterocyclic-C(Q) -, and substituted heterocyclic-C(O)- wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.
[000038] "Alkylamino" refers to the group -NRR where each R is independently selected from the group consisting of ■ hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic and where each R is joined to form together with the nitrogen atom a heterocyclic or substituted heterocyclic ring wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.
[000039] "Alkenyl" refers to alkenyl group preferably having from 2 to 10 carbon atoms and more preferably 2 to 6 carbon atoms and having at least 1 and preferably from 1-2 sites of alkenyl unsaturation.
[000040] "Alkoxy" refers to the group "alkyl-O-" which includes, by way of example, methoxy, ethoxy, ii-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1, 2-dimethylbutoxy, and the like.
[000041] "Alkyl" refers to linear or branched alkyl groups having from 1 to 10 carbon atoms, alternatively 1 to 6 carbon atoms. This term is exemplified by groups such as methyl, t-butyl, n-heptyl, octyl and the like.
[000042] "Amino" refers to the group -NH2.
[000043] "Aryl" or "Ar" refers to an unsaturated aromatic carbocyclic group of from 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl) which condensed rings may or may not be aromatic (e.g., 2-benzoxazolinone, 2H-1,4- benzoxazin-3 (4H) -one, and the like) provided that the point of attachment is through an aromatic ring atom.
[000044] "Substituted aryl" refers to aryl groups which are substituted with from 1 to 3 substituents selected from the group consisting of hydroxy, acyl, acylamino, thiocarbonylamino, acyloxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amidino, alkylamidino, thioamidino, amino, aminoacyl, aminocarbonyloxy, aminocarbonylamino, ' aminothiocarbonylamino, aryl, substituted aryl, aryloxy, substituted aryloxy, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, carboxyl, carboxylalkyl, carboxyl- substituted alkyl, carboxyl-cycloalkyl, carboxyl- substituted cycloalkyl, carboxylaryl, carboxyl-substituted aryl, carboxylheteroaryl, carboxyl-substituted heteroaryl, carboxylheterocyclic, carboxyl-substicuted heterocyclic, carboxylamido, cyano, thiol, thioalkyl, substituted thioalkyl, thioaryl, substituted thioaryl, thioheteroaryl, substituted thioheteroaryl, thiocycloalkyl, substituted thiocycloalkyl, thioheterocyclic, substituted thioheterocyclic, cycloalkyl, substituted cycloalkyl, guanidino, guanidinosulfone, halo, nitro, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, oxycarbonylamino, oxythiocarbonylaraino, -S (O)2-alkyl, -S (O)2-substituted alkyl, -S (0)2-cycloalkyl, -S (0) 2-substituted cycloalkyl, - S(O)2-alkenyl, -S (O)2-substituted alkenyl, -S(O)2-aryl, - S (0>2-substituted aryl, -S (0) 2-heteroaryl, -S(O)2- substituted heteroaryl, -S(0)2-heterocyclic, -S(O)2- substituted heterocyclic, -OS (O)2-alkyl, -OS(O)2- substituted alkyl, -OS (0)2-aryl, -OS(O)2-substiruted aryl, -0S(0)2~heteroaryl, -OS (O)2-substituted heteroaryl, -OS(0)2-heterocyclic, -OS(O)2-substituted heterocyclic, - OS(O)2-NRR where R is hydrogen or alkyl, -NRS (O)2-alkyl, - NRS(O)2-substituted alkyl, -NRS (0)2-aryl, -NRS(O)2- substituted aryl, -NRS (0)2-heteroaryl, -NRS(O)2- substituted heteroaryl, -NRS(0)2-heterocyclic, -NRS(O)2- substituted heterocyclic, -NRS (0) 2-NR-alkyl, -NRS(O)2-NR- substituted alkyl, -NRS (0) 2-NR-aryl, -NRS(O)2-NR- substiruted aryl, -NRS (0) 2-NR-heteroaryl, -NRS(O)2-NR- substituted heteroaryl, -NRS (0) 2-NR-heterocyclic, - NRS (O)2-NR-substiruted heterocyclic where R is hydrogen or alkyl, mono- and di-alkylaraino, mono- and di- (substituted alkyl) amino, mono- and di-arylamino, mono- and di- substituted arylamino, mono- and di-heteroarylamino, mono- and di-sυbstituted heteroarylamino, mono- and di- heterocyclic amino, mono- and di-substituted heterocyclic amino, unsymmetric di-substituted amines having different substituents independently selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic and amino groups on the substituted aryl blocked by conventional blocking groups such as Boc, Cbz, formyl, and the like or substituted with -SO2NRR where R is hydrogen or alkyl.
[000045] "Cycloalkyl" refers to cyclic alkyl groups of from 3 to 8 carbon atoms having a single cyclic ring including, by way of example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclooctyl and the like. Excluded from this definition are multi-ring alkyl groups such as adamantanyl, etc.
[000046] "Halo" or "halogen" refers to fluoro, chloro, bromo and iodo.
[000047] "Heteroaryl" refers to an aromatic carbocyclic group of from 2 to 10 carbon atoms and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur within the ring or oxides thereof. Such heteroaryl groups can have a single ring (e.g., pyridyl or furyl) or multiple condensed rings (e.g., indolizinyl or benzothienyl) wherein one or more of the condensed rings may or may not be aromatic provided that the point of attachment is through an aromatic ring atom. Additionally, the heteroatoms of the heteroaryl group may be oxidized, i.e., to form pyridine N-oxides or 1, l-dioxo-1,2, 5- thiadiazoles and the like. Additionally, the carbon atoms of the ring may be substituted with an oxo (=0) . The terra "heteroaryl having two nitrogen atoms in the heteroaryl, ring" refers to a heteroaryl group having two, and only two, nitrogen atoms in the heteroaryl ring and optionally containing 1 or 2 other heteroatoms in the heteroaryl ring, such as oxygen or sulfur.
[000048] "Substituted heteroaryl" refers to heteroaryl groups which are substituted with from 1 to 3 substituents selected from the group consisting of hydroxy, acyl, acylaraino, thiocarbonylamino, acyloxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amidino, alkylamidino, thioamidino, amino, aminoacyl, aminocarbonyloxy, aminocarbonylamino, aminothiocarbonylamino, aryl, substituted aryl, aryloxy, substituted aryloxy, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, carboxyl, carboxylalkyl, carboxyl-substituted alkyl, carboxyl-cycloalkyl, carboxyl- substituted cycloalkyl, carboxylaryl, carboxyl-substituted aryl, carboxylheteroaryl, carboxyl-substituted heteroaryl, carboxylheterocyclic, carboxyl-substituted heterocyclic, carboxylamido, cyano, thiol, thioalkyl, substituted thioalkyl, thioaryl, substituted thioaryl, thioheteroaryl, substituted thioheteroaryl, thiocycloalkyl, substituted thiocycloalkyl, thioheterocyclic, substituted thioheterocyclic, cycloalkyl, substituted cycloalkyl, guanidino, guanidinosulfone, halo, nitro, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, oxycarbonylamino, oxythiocarbonylamino, -S (O)2-alkyl, -S (0)2-substituted alkyl, -S(0)2~cycloalkyl, -S (0) 2-substituted cycloalkyl, - S(0)2-alkenyl, -S (O)2-substituted alkenyl, -S(O)2-aryl, -S(O)2-substituted aryl, -S (Oh-heteroaryl, -S(O)2- substituted heteroaryl, -S (O)2-heterocyclic, -S(O)2- substituted heterocyclic, -OS (0)2~alkyl, -OS(O)2- substituted alkyl, -OS (0)2-aryl, -OS(O)2-substituted aryl, -0S(0)2-heteroaryl, -OS (O) 2-substituted heteroaryl, - OS(0)2-heterocyclic, -OS (O) 2-substituted heterocyclic, - OSO2-NRR where R is hydrogen or alkyl, -NRS (O)2-alkyl, - NRS (O) 2-substituted alkyl, -NRS(0)2-aryl, -NRS(O)2- substituted aryl, -NRS (0)2-heteroaryl, -NRS(Oh- substituted heteroaryl, -NRS (Oh-heterocyclic, -NRS(O)2- substituted heterocyclic, -NRS (O) 2-NR-alkyl, -NRS(O)2-NR- substiruted alkyl, -NRS (O)2-NR-aryl, -NRS(O)2-NR- substituted aryl, -NRS (0)2-NR-heteroaryl, -NRS(O)2-NR- substituced heteroaryl, -NRS (0)2-NR-heterocyclic, - NRS (O)2-NR-substituted heterocyclic where R is hydrogen or alkyl, mono- and di-alkylamino, mono- and di- (substituted alkyl) amino, mono- and di-arylamino, mono- and di- substituted arylamino, mono- and di-heteroarylamino, mono- and di-substituted heteroarylamino, mono- and di- heterocyclic amino, mono- and di-substituted heterocyclic amino, unsymmetric di-substituted amines having different substituents independently selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic and amino groups on the substituted aryl blocked by conventional blocking groups such as Boc, Cbz, formyl, and the like or substituted with -SO2NRR where R is hydrogen or alkyl. [000049] "SuIfonyl" refers to the group -S(O)2R where R is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein.
[000050] "Optionally substituted" means that the recited group may be unsubstituted or the recited group may be substituted.
[000051] "Pharmaceutically-acceptable carrier" means a carrier that is useful in preparing a pharmaceutical composition or formulation that is generally safe, nontoxic, and neither biologically nor otherwise undesirable, and includes a carrier that is acceptable for veterinary use as well as human pharmaceutical use.
[000052] "Pharmaceutically-acceptable cation" refers to the cation of a pharmaceutically-acceptable salt.
[000053] "Pharmaceutically-acceptable salt" refers to salts which retain the biological effectiveness and properties of compounds which are not biologically or otherwise undesirable. Pharmaceutically-acceptable salts refer to pharmaceutically-acceptable salts of the compounds, which salts are derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like.
[000054] Pharmaceutically-acceptable base addition salts can be prepared from inorganic and organic bases. Salts derived from inorganic bases include, by way of example only, sodium, potassium, lithium, ammonium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines, such as alkyl amines, dialkyl amines, trialkyl amines, substituted alkyl amines, di (substituted alkyl) amines, tri (substituted alkyl) amines, alkenyl amines, dialkenyl amines, trialkenyl amines, substituted alkenyl amines, di (substituted alkenyl) amines, tri (substituted alkenyl) amines, cycloalkyl amines, di (cycloalkyl) amines, tri (cycloalkyl) amines, substituted cycloalkyl amines, disubstituted cycloalkyl amine, trisubstituted cycloalkyl amines, cycloalkenyl amines, di (cycloalkenyl) amines, tri (cycloalkenyl) amines, substituted cycloalkenyl amines, disubstituted cycloalkenyl amine, trisubstituted cycloalkenyl amines, aryl amines, diaryl amines, triaryl amines, heteroaryl amines, diheteroaryl amines, triheteroaryl amines, heterocyclic amines, diheterocyclic amines, triheterocyclic amines, mixed di- and tri-amines where at least two of the substituents on the amine are different and are selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, and the like. Also included are amines where the two or three substituents , together with the amino nitrogen, form a heterocyclic or heteroaryl group.
[000055] Examples of suitable amines include, by way of example only, isopropylamine, trimethyl amine, diethyl amine, tri (iso-propyl) amine, tri {n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, tromethamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, N-alkylglucamines, theobromine, purines, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like. It should also be understood that other carboxylic acid derivatives would be useful, for example, carboxylic acid amides, including carboxamides, lower alkyl carboxamides, dialkyl carboxamides, and the like.
[000056] Pharmaceutically-acceptable acid addition salts may be prepared from inorganic and organic acids. Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandθlic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene- sulfonic acid, salicylic acid, and the like.
[000057] A compound may act as a pro-drug. Pro-drug means any compound which releases an active parent drug in vivo when such pro-drug is administered to a mammalian subject. Pro-drugs are prepared by modifying functional groups present in such a way that the modifications may be cleaved in vivo to release the parent compound. Pro-drugs include compounds wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that may be cleaved in vivo to regenerate the free hydroxyl, amino, or sulfhydryl group, respectively. Examples of pro-drugs include, but are not limited to esters (e.g., acetate, formate, and benzoate derivatives), carbamates (e.g., N,N- dimethylamino-carbonyl) of hydroxy functional groups, and the like.
[000058] "Treating" or "treatment" of a disease includes:
(1) preventing the disease, i.e. causing the clinical symptoms of the disease not to develop in a mammal that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease,
(2) inhibiting the disease, i.e., arresting or reducing the development of the disease or its clinical symptoms, or
(3) relieving the disease, i.e., causing regression of the disease or its clinical symptoms.
[000059] A "therapeutically-effective amount" means the amount of a compound that, when administered to a mammal for treating a disease, is sufficient to effect such treatment for the disease. The "therapeutically-effective amount" will vary depending on the compound, the disease, and its severity and the age, weight, etc., of the mammal to be treated.
Synthesis of Compounds
[000060] The compounds are readily prepared via several divergent synthetic routes with the particular route selected relative to the ease of compound preparation, the commercial availability of starting materials, and the like.
[000061] The compounds can be prepared from readily- available starting materials using the following general methods and procedures. It will be appreciated that where process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
[000062] Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. Suitable protecting groups for various functional groups as well as suitable conditions for protecting and deprotecting particular functional groups are well known in the art. For exampler numerous protecting groups are described in T. W. Greene and G. M. Wuts , Protecting Groups in Organic Synthesis, Second Edition, Wiley, New York, 1991, and references cited therein.
[000063] Furthermore, the compounds may contain one or more chiral centers. Accordingly, if desired, such compounds can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-enriched mixtures. All such stereoisomers (and enriched mixtures) are included unless otherwise indicated. Pure stereoisomers (or enriched mixtures) may be prepared using, for example, optically-active starting materials or stereoselective reagents well-known in the art. Alternatively, racemic mixtures of such compounds can be separated using, for example, chiral column chromatography, chiral resolving agents, and the like.
[000064] Unless otherwise indicated, if the products contain chiral centers, they are a mixture of R, S enantiomers. However, when a chiral product is desired, the chiral product can be obtained via purification techniques which separate enantiomers from a R, S mixture to provide for one or the other stereoisomer. Such techniques are known in the art.
[000065] The compounds can be provided as pro-drugs which convert (e.g., hydrolyze, metabolize, etc.) in vivo to a compound above .
Pharmaceutical Formulations of the Compounds [000066] In general, compounds will be administered in a therapeutically-effective amount by any of the accepted modes of administration for these compounds. The compounds can be administered by a variety of routes, including, but not limited to, oral, parenteral (e.g., subcutaneous, subdural, intravenous, intramuscular, intrathecal, intraperitoneal, intracerebral, intraarterial, or intralesional routes of administration) , topical, intranasal, localized (e.g., surgical application or surgical suppository), rectal, and pulmonary (e.g., aerosols, inhalation, or powder) . Accordingly, these compounds are effective as both injectable and oral compositions. The compounds can be administered continuously by infusion or by bolus injection.
[000067] The actual amount of the compound, i.e., the active ingredient, will depend on a number of factors, such as the severity of the disease, i.e., the condition or disease to be treated, age, and relative health of the subject, the potency of the compound used, the route and form of administration, and other factors.
[000068] Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population) . The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
[000069] The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used, the therapeutically-effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range which includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans . Levels in plasma may be measured, for example, by high performance liquid chromatography.
[000070] The amount of the pharmaceutical composition administered to the patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions are administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. An amount adequate to accomplish this is defined as "therapeutically-effective dose." Amounts effective for this use will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the inflammation, the age, weight, and general condition of the patient, and the like.
[000071] The compositions administered to a patient are in the form of pharmaceutical compositions described supra. These compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
[000072] The active compound is effective over a wide dosage range and is generally administered in a pharmaceutically- or therapeutically-effective amount. The therapeutic dosage of the compounds will vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. For example, for intravenous administration, the dose will typically be in the range of about 0.5 mg to about 100 mg per kilogram body weight. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems. Typically, the clinician will administer the compound until a dosage is reached that achieves the desired effect.
[000073] When employed as pharmaceuticals, the compounds are usually administered in the form of pharmaceutical compositions. Pharmaceutical compositions contain as the active ingredient one or more of the compounds above, associated with one or more pharmaceutically-acceptable carriers or excipients. The excipient employed is typically one suitable for administration to human subjects or other mammals. In making the compositions, the active ingredient is usually mixed with an excipient, diluted by an excipient, or enclosed within a carrier which can be in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium) , ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
[000074] In preparing a formulation, it may be necessary to mill the active compound to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it ordinarily is milled to a particle size of less than 200 mesh. If the' active compound is substantially water soluble, the particle size is normally adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.
[000075] Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions of the invention can be formulated so as to provide quick, sustained, or delayed- release of the active ingredient after administration to the patient by employing procedures known in the art.
[000076] The quantity of active compound in the pharmaceutical composition and unit dosage form thereof may be varied or adjusted widely depending upon the particular application, the manner or introduction, the potency of the particular compound, and the desired concentration. The term "unit dosage forms" refers to physically-discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
[000077] The compound can be formulated for parenteral administration in a suitable inert carrier, such as a sterile physiological saline solution. The dose administered will be determined by route of administration.
[000078] Administration of therapeutic agents by intravenous formulation is well known in the pharmaceutical industry. An intravenous formulation should possess certain qualities aside from being just a composition in which the therapeutic agent is soluble. For example, the formulation should promote the overall stability of the active ingredient (s) , also, the manufacture of the formulation should be cost-effective. All of these factors ultimately determine the overall success and usefulness of an intravenous formulation.
[000079] Other accessory additives that may be included in pharmaceutical formulations and compounds as follow: solvents: ethanol, glycerol, propylene glycol; stabilizers: EDTA (ethylene diamine tetraacetic acid), citric acid; antimicrobial preservatives: benzyl alcohol, methyl paraben, propyl paraben; buffering agents: citric acid/sodium citrate, potassium hydrogen tartrate, sodium hydrogen tartrate, acetic acid/sodium acetate, maleic acid/sodium maleate, sodium hydrogen phthalate, phosphoric acid/potassium dihydrogen phosphate, phosphoric acid/disodium hydrogen phosphate; and tonicity modifiers: sodium chloride, mannitol, dextrose.
[000080] The presence of a buffer is necessary to maintain the aqueous pH in the range of from about 4 to about 8. The buffer system is generally a mixture of a weak acid and a soluble salt thereof, e.g., sodium citrate/citric acid; or the raonocation or dication salt of a dibasic acid, e.g., potassium hydrogen tartrate; sodium hydrogen tartrate, phosphoric acid/potassium dihydrogen phosphate, and phosphoric acid/disodium hydrogen phosphate.
[000081] The amount of buffer system used is dependent on (1) the desired pH; and (2) the amount of drug. Generally, the amount o,f buffer used is able to maintain a formulation pH in the range of 4 to 8. Generally, a 1:1 to 10:1 mole ratio of buffer (where the moles of buffer are taken as the combined moles of the buffer ingredients, e.g., sodium citrate and citric acid) to drug is used.
[000082] A useful buffer is sodium citrate/citric acid in the range of 5 to 50 mg per ml. sodium citrate to 1 to 15 mg per ml. citric acid, sufficient to maintain an aqueous pH of 4-6 of the composition. [000083] The buffer agent may also be present to prevent the precipitation of the drug through soluble metal complex formation with dissolved metal ions, e.g., Ca, Mg, Fe, Al, Ba, which may leach out of glass containers or rubber stoppers or be present in ordinary tap water. The agent may act as a competitive complexing agent with the drug and produce a soluble metal complex leading to the presence of undesirable particulates.
[000084] In addition, the presence of an agent, e.g., sodium chloride in an amount of about of 1-8 rαg/ml, to adjust the tonicity to the same value of human blood may be required to avoid the swelling or shrinkage of erythrocytes upon administration of the intravenous formulation leading to undesirable side effects such as nausea or diarrhea and possibly to associated blood disorders. In general, the tonicity of the formulation matches that of human blood which is in the range of 282 to 288 mθsm/kg, and in general is 285 mθsm/kg, which is equivalent to the osmotic pressure corresponding to a 0.9% solution of sodium chloride.
[000085] An intravenous formulation can be administered by direct intravenous injection, i.v. bolus, or can be administered by infusion by addition to an appropriate infusion solution such as 0.9% sodium chloride injection or other compatible infusion solution.
[000086] The compositions are preferably formulated in a unit dosage form, each dosage containing from about 5 to about 100 mg, more usually about 10 to about 30 mg, of the active ingredient. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
[000087] The active compound is effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood/ however, that the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
[000088] For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 2000 mg of the active ingredient.
[000089] The tablets or pills may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
[000090] The liquid forms in which the novel compositions may be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
[000091] Compositions for inhalation or insufflation include solutions and suspensions in pharraaceutically- acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically-acceptable excipients as described supra. Compositions in pharmaceutically-acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device may be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered from devices which deliver the formulation in an appropriate manner.
[000092] The compounds can be administered in a sustained release form. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the compounds, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (e.g., poly(2- hydroxyethyl- methacrylate) as described by Langer et al., J. Biomed. Mater. Res. 15: 167-277 (1981) and Langer, Chem. Tech. 12: 98-105 (1982) or poly (vinyl alcohol)), polylactides (U.S. Patent No. 3,773,919), copolymers of L-glutamic acid and gamma ethyl-L-glutamate (Sidman et al., Biopolymers 22: 547-556, 1983), non-degradable ethylene-vinyl acetate (Langer et al., supra), degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT™ (i.e., injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D- (-) -3- hydroxybutyric acid (EP 133,988).
[000093] The compounds can be administered in a sustained-release form, for example a depot injection, implant preparation, or osmotic pump, which can be formulated in such a manner as to permit a sustained- release of the active ingredient. Implants for sustained- release formulations are well-known in the art. Implants may be formulated as, including but not limited to, microspheres, slabs, with biodegradable or nonbiodegradable polymers. For example, polymers of lactic acid and/or glycolic acid form an erodible polymer that is well-tolerated by the host.
[000094] Transdermal delivery devices ("patches") may also be employed. Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Patent No. 5,023,252, issued June 11, 1991, herein incorporated by reference. Such patches may be constructed for continuous, pulsatile, or on-demand delivery of pharmaceutical agents.
[000095] Direct or indirect placement techniques may be used when it is desirable or necessary to introduce the pharmaceutical composition to the brain. Direct techniques usually involve placement of a drug delivery catheter into the host's ventricular system to bypass the blood-brain barrier. One such implantable delivery system used for the transport of biological factors to specific anatomical regions of the body is described in U.S. Patent No. 5,011,472, which is herein incorporated by reference.
[000096] Indirect techniques usually involve formulating the compositions to provide for drug latentiation by the conversion of hydrophilic drugs into lipid-soluble drugs . Latentiation is generally achieved through blocking of the hydroxy, carbonyl, sulfate, and primary amine groups present on the drug to render the drug more lipid-soluble and amenable to transportation across the blood-brain barrier. Alternatively, the delivery of hydrophilic drugs may be enhanced by intra-arterial infusion of hypertonic solutions which can transiently open the bloόd-brain barrier.
[000097] In order to enhance serum half-life, the compounds may be encapsulated, introduced into the lumen of liposomes, prepared as a colloid, or other conventional techniques may be employed which provide an extended serum half-life of the compounds. A variety of methods are available for preparing liposomes, as described in, e.g., Szoka et al., U.S. Patent Nos. 4,235,871, 4,501,728 and 4,837,028 each of which is incorporated herein by reference.
[000098] Pharmaceutical compositions are suitable for use in a variety of drug delivery systems. Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences, Mace Publishing Company, Philadelphia, PA, 17th ed. (1985).
[000099] In the examples below, if an abbreviation is not defined above, it has its generally accepted meaning. Further, all temperatures are in degrees Celsius (unless otherwise indicated) . The following Methods were used to prepare the compounds set forth below as indicated.
Example 1 - General Synthetic Procedure
[0000100] The compounds of this invention were obtained from commercial sources or prepared following the procedures described in the literature. A sub-class of compounds was synthesized by the procedure summarized in the following synthetic scheme.
Figure imgf000042_0001
[0000101] 4-methyl-cyclohexanone reacting with 2-cyano- acetamide and sulfur yielded the intermediate, 2-amino-6- methyl-4, 5, 6, 7-tetrahydrobenzothiophene-3-carboxamide (L. D. Jennings, et al, Bioorganic & Medicinal Chemistry Letters 2005, 15 (21) , 4731-4735) . The intermediate was sulfonated with sulfonyl chlorides to give the final products, which may be 3-CONH2 or 3-CN sulfonamides, depending on the reaction conditions, such as base and solvent used in the reactions.
Example 2 - Formulation 1
[0000102] Hard gelatin capsules containing the following ingredients are prepared:
Figure imgf000043_0001
[0000103] The above ingredients are mixed and filled into hard gelatin capsules in 340 mg quantities.
Example 3- Formulation 2
[0000104] A tablet formula is prepared using the ingredients below:
Figure imgf000043_0002
[0000105] The components are blended and compressed to form tablets, each weighing 240 mg.
Example 4 - Formulation 3
[0000106] A dry powder inhaler formulation is prepared containing the following components:
Figure imgf000044_0001
[0000107] The active mixture is mixed with the lactose and the mixture is added to a dry powder inhaling appliance.
Example 5 - Formulation 4
[0000108] Tablets, each containing 30 mg of active ingredient, are prepared as follows:
Figure imgf000044_0002
[0000109] The active ingredient, starch, and cellulose I are passed through a No. 20 mesh U.S. sieve and mixed thoroughly. The solution of polyvinyl-pyrrolidone is mixed with the resultant powders, which are then passed through a 16 mesh U.S. sieve. The granules so produced are dried at 50° to 6O0C and passed through a 16 mesh U.S. sieve. The sodium carboxymethyl starch, magnesium stearate, and talc, previously passed through a No. 30 mesh U.S. sieve, are then added to the granules, which after mixing, are compressed on a tablet machine to yield tablets each weighing 150 mg.
Example 6 - Formulation 5
[0000110] Capsules, each containing 40 mg of medicament, are made as follows:
Figure imgf000045_0001
[0000111] The active ingredient, cellulose, starch, an magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 150 mg quantities.
Example 7 - Formulation 6
[0000112] Suppositories, each containing 25 mg of active ingredient, are made as follows:
Figure imgf000045_0002
[0000113] The active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of nominal 2.O g capacity and allowed to cool.
Example 8 - Formulation 7
[0000114] Suspensions, each containing 50 mg of medicament per 5.0 ml dose, are made as follows:
Figure imgf000046_0001
[0000115] The medicament, sucrose, and xanthan gum are blended, passed through a NO. 10 mesh U.S. sieve, and then mixed with a previously made solution of the microcrystalline cellulose and sodium carboxymethyl cellulose in water. The sodium benzoate, flavor, and color are diluted with some of the water and added with stirring. Sufficient water is then added to produce the required volume.
Example 9 - Formulation 8
[0000116] Hard gelatin tablets, each containing 15 mg of active ingredient, are made as follows:
Figure imgf000046_0002
Figure imgf000047_0001
[0000117] The active ingredient, cellulose, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 560 mg quantities.
Example 10 - Formulation 9
[0000118] An intravenous formulation may be prepared as follows :
Figure imgf000047_0002
[0000119] Therapeutic compound compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle or similar sharp instrument.
Example 11 - Formulation 10
[0000120] A topical formulation may be prepared as follows :
Figure imgf000047_0003
[0000121] The white soft paraffin is heated until molten. The liquid paraffin and emulsifying wax are incorporated and stirred until dissolved. The active ingredient is added and stirring is continued until dispersed. The mixture is then cooled until solid.
Example 12 - Formulation 11
[0000122] An aerosol formulation may be prepared as follows: ft solution of the candidate compound in 0.5% sodium bicarbonate/saline (w/v) at a concentration of 30.0 mg/mlι is prepared using the following procedure:
[0000123] Preparation of 0.5% Sodium Bicarbonate / Saline Stock Solution: 100. OmL
Figure imgf000048_0001
Procedure:
1. Add 0.5g sodium bicarbonate into a 100 mL volumetric flask.
2. Add approximately 90.0 ml saline and sonicate until dissolved.
3. Q.S. to 100.0 mL with saline and mix thoroughly.
[0000124] Preparation of 30.0 mg/mL Candidate Compound: 10.0 mL
Figure imgf000048_0002
Procedure:
1. Add 0.300 g of the candidate compound into a 10.0 mL volumetric flask.
2. Add approximately 9.7 mL of 0.5% sodium bicarbonate / saline stock solution.
3. Sonicate until the candidate compound is completely dissolved.
4. Q.S. to 10.0 mL with 0.5% sodium bicarbonate / saline stock solution and mix.
Example 13 - A High throughput fluorescence quench biochemical assay to identify inhibitors of 5. aureus sortase A enzyme.
Primary high throughput screening assay. [0000125] A high throughput screen was developed in order to identify compounds in the SIGA chemical compound library that inhibited the cleavage activity of S. aureus sortase A enzyme. The N-terminal membrane anchor (a.a. 2- 25) of S. aureus sortase A was replaced with a six- histidine tag (SrtAΛN) and was cloned into an E. coli expression vector. Sortase A and was purified as previously described (11) . Ninety six-well black solid polystyrene plates were seeded with 45.5 μM sortase peptide ( (2-aminobenzoyl-LPTEGE-diaminopropionic acid- dinitrophenyl-NH2 (Abz-LPETGE-Dap (DNE) -NH2) ) at a final concentration of 25 μM. Sortase buffer (40 mM Tris-Cl, pH 8.0, 150 mM NaCl, 1 mM TriGlycine, 5 mM CaCl2) was added to the peptide-only wells (column 12) . A total of 80 compounds were tested per 96-well plate. Compounds were diluted in DMSO and added robotically to all wells at a final concentration of 5 μM. Polyvinyl sulfone [500 μM] and berberine chloride [50 μM] were used as positive control drugs. Five μg 5. aureus SrtAΔM were added and plates incubated at room temperature overnight before reading in an Envision kinetic plate reader at excitation wavelength of 340 nm and emission 420 nm. Positive hits were those identified as having greater than 60% inhibition of fluorescence at 5 μM using the equation: % Inhibited= [ (Max-Result) / (Max-Min) ] x 100.-
Secondary Assays.
[0000126] Sortase inhibitors identified in the primary screen were retested in secondary assays to determine IC50 (concentration at which compounds inhibit sortase activity 50%) and CC50 (50% cytotoxic concentration in mammalian cells) values. The sortase assay was carried out as described above except that half-log dilutions of compounds were added to the sortase reactions in triplicate at concentrations ranging from 25 μM to 0.008 μM.
[0000127] To determine the CC50 values of the sortase inhibitors for mammalian cells, 96-well plates were seeded with HEK-293 cells at subconfluent densities. For control wells, cells were diluted 1:2 and 1:4. Cells were incubated overnight at 370C before adding compound in half-log dilutions starting from a 25 μM stock concentration. Plates were incubated at 370C for 4 days. On the last day of incubation, alamarBlue™ (Invitrogen) was added to each well and plates incubated overnight at 370C. Metabolically active cells cause reduction of the REDOX indicator in the reagent alamarBlue™ to change from an oxidized (blue) form to a reduced (red) form. This color change can be detected by reading absorbance at A570 and Asoo. Plates were read on an Envision spectrophotometer the next day. Example 14 - Biological assays used to examine the effects of compounds on sortase A function in S. aureus.
Bioassays I and II. Fibronectin and fibrinogen binding assays.
[0000128] Fibronectin binding protein A and B (FnBPA and B) are sortase-dependent surface proteins of S. aureus that mediate bacterial adhesion and invasion of epithelial cells, endothelial cells, fibroblasts and osteoblasts by forming a fibronectin bridge from the bacterial cell surface to a fibronectin-binding integrin on the host cell (12). S. aureus that express FnBPA and FnBPB are able to bind to immobilized fibronectin. An S. aureus srtA mutant does not express these proteins, and shows greatly reduced binding to fibronectin. Another set of sortase-dependent surface proteins are clumping factors A and B (CIfA and CIfB) . CIfA and CIfB are fibrinogen-binding proteins that mediate fibrinogen-dependent adhesion and clumping of S. aureus on blood clots and damaged tissue. In order to determine whether the sortase inhibitors identified in HTS would also decrease fibronectin or fibrinogen binding by S. aureus, plate binding assays were developed.
[0000129] Human fibrinogen (15 μg/ml), or fibronectin (12.5 μg/ml) in phosphate buffered saline (PBS) were added to a Nunc MaxiSorp 96 well flat-bottom Immuno plate and incubated overnight at 40C. The following day, plates were washed and blocked with 1% bovine serum albumin (in PBS) . Overnight cultures of 5. aureus RN4220 wild type or srtA mutant grown, in chemically defined medium (CDM) were washed in PBS and the Aβio were adjusted to approximately 1.0. Plates were washed before adding cells to the wells. Plates were incubated for 10 min at 370C and then washed 2 times before adding BHI media containing 10% alamarBlue™. Plates were incubated approximately 1.5 h at 370C, or until adequate color development had occurred (-1.5 hr) . Plates were read in a Wallac spectrophometer at As7o-A6io- Percent alamarBlue™ conversion was calculated according to manufacturer's instructions. Wild type S. aureus RN4220 that expresses fibronectin binding proteins is able to bind to the immobilized fibronectin in larger numbers than the sortase mutant. The increased number of wild type S. aureus bacteria bound to the wells are able to convert the alamarBlue™ from a blue to a pink color at a faster rate than the S. aureus srtA mutant. The sortase mutant showed an approximately 80% reduced ability to bind to fibronectin in this assay (Figure 1) . Transformation of the sortase mutant with a plasmid expressing S. aureus sortase A partially restored the ability of the mutant to bind to fibronectin ■ (Figure 1). Similar results were obtained in the fibrinogen-binding assay with the sortase mutant showing an 80% decrease in the ability to bind immobilized fibrinogen.
Bioassay III. Surface Protein A detection by flow cytometry
[0000130] Protein A is a highly-expressed sortase A- dependent surface protein that binds the Fc region of IgG molecules which disrupts opsonization and phagocytosis of S. aureus (13) . Cultures of S. aureus RN4220 wild type, srtA.Δ mutant, complemented srtA mutant, or a protein A mutant (spa:ery) were grown overnight in CDM. Cultures were washed in FACS buffer and suspended in a 1:50' dilution of goat anti-rabbit IgG-FITC (Biorad) . Cells were incubated covered in foil for 15 min before washing two times in FACS buffer (phosphate buffered saline containing 1% fetal bovine serum) . Cells were suspended in FACS buffer, fixed with 2% glutaraldehyde, and analyzed on a BD FACs Canto flow cytoraeter. Figure 2 shows that wild type S. aureus RN4220 exhibits an approximately 7- fold increased shift in fluorescence compared to the sortase mutant after incubation with antibody. An S. aureus spa mutant, which expresses no protein A, showed slightly lower fluorescence compared to the sortase mutant. This would be expected since a sortase mutant still expresses protein A, some of which remains associated with the cell wall. These results suggested that flow cytometry is a sensitive, quick assay for the detection of altered surface protein A expression by S, aureus after treatment with sortase inhibitors.
Example 15 - Growth inhibition assays, inhibition of sortase A function in biological assays, solubility and PK on compounds
S. aureus growth inhibition assays
[0000131] In order to test the compounds of the present invention in biological assays, it was first necessary to determine whether such compounds were inhibitory for growth of S. aureus in vitro. S. aureus RN4220 was grown in CDM containing 1% DMSO or in compounds at 100 μM, 75 UM, 50 μM, 25 μM, 12.5 μM, and 6.2 μM concentrations (1% DMSO) in flat-bottom 96 well plates. Plates were incubated at 37CC with shaking. The absorbance at O.D.βoo was read every hour for 7 hours and at 24 hours. For those compounds that showed severe growth inhibition, MIC assays using the broth microdilution method were performed as described (14) . Several compounds in the sulfonamide/thiophene series were found to be growth inhibitory for S. aureus (Tables 1 and 2) . In addition, several compounds were able to inhibit the growth of Bacillus subtilis, B. cereus and Listeria monocytogenes (Table 2) ,
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Table 2 - MIC values for growth inhibitory activities of sulfonamide/thiophene compounds
Figure imgf000058_0001
Growth inhibition of Streptococcus pyogenes.
[0000132] Time kill assays were performed to determine the ability of compound 21 to inhibit the growth of S. pyogenes. Cultures were grown overnight and diluted to 5.0 x 105 cfu/ml in Mueller-Hinton broth supplemented with 5% lysed horse blood. Compound 21 (at 7 μg/ml or vancomycin at 2 μg/ml, or DMSO as a negative control were added to the cultures. Cultures were grown at 35° C statically and 50 μl aliquots were taken and plated on brain-heart infusion agar. Plates were incubated at 37° C and colonies were enumerated after 48 hr. Figure 3 shows that after only 2 hours of growth of S. pyogenes in 7 μg/ml Compound 21, no viable colonies could be recovered.
Effects of inhibitors in sortase biological assays [0000133] Lead compounds identified as potent S. aureus sortase inhibitors in the in vitro assays were tested in the biological assays described above. S. aureus was grown overnight in CDM in compounds at concentrations that were sub-inhibitory to growth (12 μM-100 μM) . Fibrinogen and fibronectin binding and surface protein A expression assays were carried out as described above. The % inhibition values in the fibrinogen and fibronectin plate- binding assays are reported as a reduction in binding calculated as the percent of the wild type control wells. Table 3 summarizes the in vitro and biological assay data for the certain compounds of the present invention.
Table 3 - Summary of in vitro and S. aureus live cell assays for the sulfonamide/thiophcnc compounds.
Figure imgf000059_0001
Solubility and PK, and acute toxicity studies [0000134] To examine the pharmacologic properties of members of the sulfonaraide/thiophene series, compounds 9- 11 (see Table 1) were analyzed for solubility and oral bioavailability. The compounds were not soluble in simulated gastric fluid but all were highly soluble in water (Table 4) .
[0000135] The non-clinical pharmacokinetic profiles of Compounds 9-11 (see Table 1) were evaluated in healthy naϊve female CD-I mice following a single oral administration. Compounds at 100 mg/kg were administered separately as a 0.5% Tween 80® plus 0.5% hydroxypropyl methyl cellulose suspension. Mice were sacrificed at 1, 2, 3, 5, 7, or 24 hours and bled via heart puncture. No obvious toxicity was observed at any time point. Compound concentrations in plasma samples were analyzed by HPLC. Non-compartmental analysis was applied to the individual compounds 9-11 plasma concentration data using WinNonlin professional edition (Pharsight Corporation, Version 5.2, CA) . Compounds 9-11 showed peak plasma concentrations (Cmax) at 1.3, 2, and 2.3 hr, respectively, following administration. As shown in Table 3, each compound achieved significant oral exposure with a greater Cmax and Al)C(0-24) observed for Compound 10. These plasma concentrations are well above the effective concentrations of Compounds 9 and 11 for inhibition of surface protein expression (~5 μg/ml) as well as the MIC value for growth inhibition of S. aureus (-13 μg/ml) . Plasma levels of Compound 10 are also above the effective concentration (9.8 μg/ml) and MIC (39 μg/ml) for S. aureus Newman as well as methicillin-resistant S. aureus 328 (see Tables 1- 3).
Table 4 • Solubility and PK analysis
Figure imgf000060_0001
References
1. Klein, E., D. L. Smith, and R. Laxminarayan, Hospitalizations and deaths caused by methicillin- resistant Staphylococcus aureus, United States, 1999-2005. Emerg Infect Dis, 2007. 13(12): p. 1840-6.
2. Richards, M. J., et al., Nosocomial infections in medical intensive care units in the united States. National Nosocomial Infections Surveillance System. Crit Care Med, 1999. 27(5): p. 887-92.
3. Richards, M.J., et al., Nosocomial infections in pediatric intensive care units in the United States. National Nosocomial Infections Surveillance System. Pediatrics, 1999. 103(4): p. e39.
4. Lowy, F. D., Antimicrobial resistance: the example of Staphylococcus aureus. J Clin Invest, 2003. 111(9): p. 1265-73.
5. Klevens, R.M., et al . , Invasive methicillin-resistant Staphylococcus aureus infections in the United States. Jama, 2007. 298(15): p. 1763-71.
6. Casey, A.L., P.A. Lambert, and T.S. Elliott, Staphylococci. Int J Antimicrob Agents, 2007. 29 Suppl 3: p. S23-32.
7. Fischetti, V.A., V. Pancholi, and O. Schneewind, Conservation of a hexapeptide sequence in the anchor region of surface proteins from Gram-positive cocci. MoI Microbiol, 1990. 4(9): p. 1603-5. 8. Mazmanian, S.K., et al., Staphylococcus aureus sortase mutants defective in the display of surface proteins and in the pathogenesis of animal infections. Proc Natl Acad Sci U S A, 2000. 97 (10): p. 5510-5.
9. Weiss, W.J., et al., Effect of srtA and srfcB gene expression on the virulence of Staphylococcus aureus in animal models of infection. J Antimicrob Chemother, 2004. 53(3) : p. 480-6.
10. Jonsson, I.M., et al . , The role of Staphylococcus aureus sortase A and sortase B in murine arthritis. Microbes Infect, 2003. 5(9): p. 775-80.
11. Ton-That, H., et al., Purification and characterization of sortase, the transpeptidase that cleaves surface proteins of Staphylococcus aureus at the LPXTG motif. Proc Natl Acad Sci U SA, 1999. 96(22): p. 12424-9.
12. Palmqvist, N., et al . , Fibronectin-binding proteins and fibrinogen-binding clumping factors play distinct roles in staphylococcal arthritis and systemic inflammation. J Infect Dis, 2005. 191{5): p. 791-8.
13. Marraffini, L.A., A.C. Dedent, and O. Schneewind, Sortases and the art of anchoring proteins to the envelopes of Gram-positive bacteria. Microbiol MoI Biol Rev, 2006. 70(1) : p. 192-221.
14. Murray, P.R., Manual of Clinical Microbiology 7th edition, ed. E.J. Baron. 1999, Washington, DC: Americal Society of Microbiology: p.1541. [0000136] All references cited herein are .herein incorporated by reference in their entirety for all purposes.
[0000137] The invention has been described in terms of preferred embodiments thereof, but is more broadly applicable as will be understood by those skilled in the art. The scope of the invention is only limited by the following claims.

Claims

WH&T IS CLAIMED IS:
1. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound having the following general Formula I or a pharmaceutically acceptable salt thereof:
Figure imgf000064_0001
wherein R1, R2 and R3 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylamino, heterocycloalkylamino, arylamino, heteroarylamino, acylamino, arylacylamino, heteroarylacylamino, alkylsulfonylamino, arylsulfonylamino, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, carboxy, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl, substituted carbamoyl, halogen, cyano, isocyano and nitro; or R3 is as above and R1 and R2 together with the carbons they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring and may be fused with an aromatic or aliphatic ring; and
R4 and R5 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl and substituted carbamoyl; or R4 and R5 together with the nitrogen atom they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring.
2. The composition of claim 1, wherein R1 and R2 together with the carbons they are attached to form a substituted ring.
3. The composition of claim 1, wherein R3 is nitro.
4. The composition of claim 1, wherein said R3 is amino.
5. The composition of claim 1, wherein R4 is hydrogen and R5 is arylsulfonyl.
6. The composition of claim 1, wherein the compound of Formula I is selected from the group consisting of: 2- Acetylamino-4 , 5-dihydro-naphtho [1, 2-b] thiophene-3- carboxylic acid ethyl ester; 2-Amino-4-methyl-5-phenyl- thiophene-3-carboxylic acid ethyl ester; ethyl 2-amino- 4,7-dihydro-5H-spiro[l-benzothiophene-6,l'-cyclohexane]-3- carboxylate; 2- (4-Bromo-benzenesulfonylamino) -4,5, 6,7, 8, 9- hexahydro-cycloocta [b] thiophene-3-carboxylic acid amide; 2-Amino-6-phenyl-4 ,5, 6, 7-tetrahydro-benzo [b] thiophene-3- carboxylic acid methyl ester; 2-Amino-4-(4-sec-butyl- phenyl) -5-methyl-thiophene-3-carboxylic acid methyl ester; 2- (3, 4-Dichloro-benzenesulfonylamino) -6-methyl-4, 5, 6, 7- tetrahydro-benzo[b] thiophene-3-carboxylic acid amide; 2- (4-Isopropyl-benzenesulfonylamino) -6-methyl-4, 5, 6, 7- tetrahydro-benzofb] thiophene-3-carboxylic acid amide; 2- (4-Bromo-3-methyl-benzenesulfonylamino) -6-methyl-4, 5, 6,7- tetrahydro-benzo[b] thiophene-3-carboxylic acid amide; 2- (5-Bromo-2-methoxy-benzenesulfonylamino) -6-methyl-4, 5, 6,7- tetrahydro-benzo[b] thiophene-3-carboxylic acid amide; 6- Methyl-2- (naphthalene-2-sulfonylamino) -4, 5, 6, 7-tetrahydro- benzo [b] thiophene-3-carboxylic acid amide; 2-(4-Chloro- benzenesulfonylamino) -6-methyl-4, 5, 6, 7-tetrahydro- benzo [b] thiophene-3-carboxylic acid amide; 2-(3-Chloro- benzenesulfonylamino) -6-methyl-4, 5, 6, 7-tetrahydro- benzo [b] thiophene-3-carboxylic acid amide; 2-Bromo-N- (3- cyano-6-methyl-4, 5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 3-Bromo-N- (3-cyano-6-methyl-4, 5, 6, 1- tetrahydro-benzo[b] thiophen-2-yl) -benzenesulfonamide; 4- Bromo-N-(3-cyano-6-methyl-4, 5, 6, 7-tetrahydro- benzo [b] thiophen-2-yl) -benzenesulfonamide; 2-Chloro-N- (3- cyano-6-methyl-4, 5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2,3, 4-Trichloro-N- (3-cyano-6-methyl- 4,5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2,4, 5-Trichloro-N- {3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 3, 4-Dibromo-N- (3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2, 5-Dibromo-N- (3-cyano-6-methyl- 4,5, 6,7-tetrahydro-benzo[b] thiophen-2-yl)- benzenesulfonamide; 2, 4-Dibromo-N-(3-cyano-6-methyl- 4,5, 6, 7-tetrahydro-benzo[b] thiophen-2-yl) - benzenesulfonamide; 2, 4, 6-Tribromo-N- (3-cyano-6-methyl- 4, 5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2, 4, 5-Tribromo-N- (3-cyano-δ-methyl- 4, 5, 6, 7-tetrahydro-benzo[b] thiophen-2-yl) - benzenesulfonamide; 3, 4-Dichloro-N- (3-cyano-6-methyl- 4,5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; and 4-Bromo-N- (3-cyano-6-methyl- 4, 5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) -3-methyl- benzenesulfonamide .
7. A compound having the following general Formula II or a pharmaceutically acceptable salt thereof:
Figure imgf000067_0001
wherein R6, R7, R8, R9 and R10 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylamino, heterocycloalkylamino, arylamino, heteroarylamino, acylaπiino, arylacylamino, heteroarylacylami.no, alkylsulfonylamino, arylsulfonylamino, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, carboxy, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl, substituted carbamoyl, halogen, cyano, isocyano and nitro; and
optionally at least one of R6 and R7; R7 and R8; R8 and R9; and R9 and R10 together with the carbons they are attached to form a substituted or unsubstituted ring, which optionally include one or more heteroatoms in the ring and optionally are fused with an aromatic or aliphatic ring, with a proviso that at least one of R6, R7, R8, R9 and R10 is halogen, and if R6, R7, R9 and R10 are H, R8 is not F or Cl, or if R7, R8, and R10 are H, R6 and R9 are not both Cl.
8. The compound of claim 7, wherein each of R7 and R8 is a halogen.
9. The compound of claim 8, wherein R7 and R8 are Cl.
10. The compound of claim 7, wherein R8 is Br.
11. The compound of claim 7, wherein the compound of Formula II is selected from the group consisting of: 2- Bromo-N- (3-cyano-6-methyl-4, 5, 6, 7-tetrahydro- benzo [b] thiophen-2-yl) -benzenesulfonamide; 3-Bromo-N- (3- cyano-6-methyl-4 , 5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 4-Bromo-N- (3-cyano-6-methyl-4, 5, 6,7- tetrahydro-benzo[b]thiophen-2-yl) -benzenesulfonamide; 2- Chloro-N- (3-cyano-6-methyl-4, 5, 6, 7-tetrahydro- benzo[b]thiophen-2-yl) -benzenesulfonamide; 2, 3, 4- Trichloro-N- (3-cyano-6-methyl-4,5, 6,7-tetrahydro- benzo [b] thiophen-2-yl) -benzenesulfonamide; 2,4,5- Trichloro-N- (3-cyano-6-methyl-4, 5, 6, 7-tetrahydro- benzo[b]thiophen-2-yl) -benzenesulfonamide; 3, 4-Dibromo-N- (3-cyano-6-methyl-4 , 5, 6, 7-tetrahydro-benzo [b] thiophen-2- yl) -benzenesulfonamide; 2, 5-Dibromo-N- (3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2, 4-Dibromo-N- (3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2, 4, 6-Tribromo-N- (3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2, 4, 5-Tribromo-N-{3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 3, 4-Dichloro-N- (3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; and 4-Bromo-N- (3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) -3-methyl- benzenesulfonamide .
12. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and the compound of claim 7.
13. A method for the treatment or prophylaxis of a gram- positive bacterial infection or disease associated therewith, comprising administering in a therapeutically effective amount to a mammal in need thereof, a compound of Formula I below or a pharmaceutically acceptable salt thereof:
Figure imgf000070_0001
wherein R1, R2 and R3 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylamino, heterocycloalkylamino, arylamino, heteroarylamino, acylamino, arylacylamino, heteroarylacylamino, alkylsulfonylamino, arylsulfonylamino, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted arainosulfonyl, carboxy, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl, substituted carbamoyl, halogen, cyano, isocyano and nitro; or R3 is as above and R1 and R2 together with the carbons they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring and may be fused with an aromatic or aliphatic ring; and
R4 and R5 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl and substituted carbamoyl; or R4 and R5 together with the nitrogen atom they are attached to form a substituted or unsubstituted ring, which may include one or more heteroatoms in the ring.
14. The method of claim 13, wherein R1 and R2 together with the carbons they are attached to form a substituted ring.
15. The method of claim 13, wherein R3 is nitro.
16. The method of claim 13, wherein said R3 is amino.
17. The method of claim 13, wherein R4 is hydrogen and Rs is arylsulfonyl.
18. The method of claim 13, wherein the compound of Formula I is selected from the group consisting of: 2- Acetylamino-4, 5-dihydro-naphtho [1, 2-b] thiophene-3- carboxylic acid ethyl ester; 2-Amino-4-methyl-5-phenyl- thiophene-3-carboxylic acid ethyl ester; ethyl 2-amino-
4, 7-dihydro-5H-spiro [l-benzothiophene-6, 1 ' -cyclohexane] -3- carboxylate; 2- (4-Bromo-benzenesulfonylamino) -4,5, 6,7,8, 9- hexahydro-cycloocta [b] thiophene-3-carboxylic acid amide; 2-Amino-6-phenyl-4,5, 6, 7-tetrahydro-benzo[b] thiophene-3- carboxylic acid methyl ester; 2-Amino-4- (4-sec-butyl- phenyl)-5-methyl-thiophene-3-carboxylic acid methyl ester; 2- (3, 4-Dichloro-benzenesulfonylamino) -6-rαethyl-4, 5,6,7- tetrahydro-benzo(b) thiophene-3-carboxylic acid amide; 2- (4-Isopropyl-benzenesulfonylamino) -6-methyl-4, 5, 6, 7- tetrahydro-benzo[b] thiophene-3-carboxylic acid amide; 2- (4-Bromo-3-methyl-benzenesulfonylamino) -6-methyl-4, 5/6,7- tetrahydro-benzo[b] thiophene-3-carboxylic acid amide; 2- (5-Bromo-2-methoxy-benzenesulfonylamino)-6-methyl-4, 5, 6, 7- tetrahydro-benzo[b] thiophene-3-carboxylic acid amide; 6- Methyl-2- (naphthalene-2-sulfonylaraino) -4,5, 6, 7-tetrahydro- benzo[b] thiophene-3-carboxylic acid amide; 2-(4-Chloro- benzenesulfonylanu.no) -β-methyl-4, 5, 6, 7-tetrahydro- benzo[b] thiophene-3-carboxylic acid amide; 2-(3-Chloro- benzenesulfonylamino) -6-methyl-4, 5, 6, 7-tetrahydro- benzo[b] thiophene-3-carboxylic acid amide; 2-Bromo-N-(3- cyano-6-methyl-4, 5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 3-Bromo-N- (3-cyano-6-methyl-4, 5,6,7- tetrahydro-benzo [b] thiophen-2-yl) -benzenesulfonamide; 4- Bromo-N- (3-cyano-6-methyl-4, 5, 6, 7-tetrahydro- benzo [b] thioρhen-2-yl) -benzenesulfonamide; 2-Chloro-N- (3- cyano-6-methyl-4, 5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2,3, 4-Trichloro-N- (3-cyano-6-methyl- 4, 5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2,4, 5-Trichloro-N- (3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo[b] thiophen-2-yl) - benzenesulfonamide; 3, 4-Dibromo-N- (3-cyano-6™methyl- 4,5,6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2, 5-Dibromo-N- (3-cyano-β-methyl- 4, 5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2, 4-Dibromo-N- (3-cyano-6-methyl- 4, 5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2, 4, 6-Tribromo-N- (3-cyano-6-methyl- 4,5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 2, 4, 5-Tribromo-N- (3-cyano-6-methyl- 4, 5, 6, 7-tetrahydro-benzo [b] thiophen-2-yl) - benzenesulfonamide; 3, 4-Dichloro-N- (3-cyano-6-methyl- 4,5, 6, 7-tetrahydro-benzo[b] thiophen-2-yl) - benzenesulfonaraide; and 4-Bromo-N- (3-cyano-6-methyl- 4,5,6, 7-tetrahydro-benzo[b] thiophen-2-yl) -3-methyl- benzenesulfonamide .
19. The method of claim 13, wherein the mammal is a human.
20. The method of claim 13, wherein the bacterial infection is caused by Staphylococcus aureus or E. faecalis.
21. The method of claim 20, wherein the bacterial infection is caused by Staphylococcus aureus.
22. The method of claim 13, wherein the bacterial infection is caused by a bacterium belonging to a genus selected from the group consisting of Clostridium, Staphylococcus, Bacillus, and listeria.
23. The method of claim 13, wherein said bacterial infection is associated with a condition selected from the group consisting of skin infections, pneumonia, meningitis, osteomyelitis, endocarditis, toxic shock syndrome, septicemia, and food poisoning.
24. The method of claim 13, which further comprises coadministration of at least one antibiotic.
25. The method of claim 13, wherein the compound of Formula I is a compound having the following general Formula II:
Figure imgf000074_0001
wherein R6, R1, RB, R9 and R10 are independently selected from the group consisting of hydrogen, substituted or unsubstituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, arylalkyl, aryl, heteroaryl, hydroxy, alkyloxy, aryloxy, heteroaryloxy, acyloxy, arylacyloxy, heteroarylacyloxy, alkylsulfonyloxy, arylsulfonyloxy, thio, alkylthio, arylthio, amino, alkylamino, dialkylamino, cycloalkylamino, heterocycloalkylamino, arylamino, heteroarylamino, acylamino, arylacylamino, heteroarylacylamino, alkylsulfonylaitri.no, arylsulfonylamino, acyl, arylacyl, heteroarylacyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, hydroxysulfonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aminosulfonyl, substituted aminosulfonyl, carboxy, alkoxycarbonyl, cycloalkyloxycarbonyl, aryloxycarbonyl, carbamoyl, substituted carbamoyl, halogen, cyano, isocyano and nitro; and
optionally at least one of R6 and R7; R7 and R8; R8 and R9; and R9 and R10 together with the carbons they are attached to form a substituted or unsubstituted ring, which optionally include one or more heteroatoms in the ring and optionally are fused with an aromatic or aliphatic ring, with a proviso that at least one of R6, R7, R8, R9 and R10 is halogen, and if R6, R7, R9 and R10 are H, R8 is not F or
Cl, or if R7, R8, and R10 are H, R6 and R3 are not both Cl.
26. The method of claim 25, wherein each of R7 and Re is a halogen.
27. The method of claim 26, wherein R7 and R8 are Cl.
28. The method of claim 25, wherein R8 is Br.
PCT/US2009/039023 2008-04-04 2009-03-31 Antibacterial drug for treatment of staphylococcus aureus and other gram-positive bacterial infections WO2009124086A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4239708P 2008-04-04 2008-04-04
US61/042,397 2008-04-04

Publications (2)

Publication Number Publication Date
WO2009124086A2 true WO2009124086A2 (en) 2009-10-08
WO2009124086A3 WO2009124086A3 (en) 2010-02-25

Family

ID=41136091

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/039023 WO2009124086A2 (en) 2008-04-04 2009-03-31 Antibacterial drug for treatment of staphylococcus aureus and other gram-positive bacterial infections

Country Status (1)

Country Link
WO (1) WO2009124086A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012149452A1 (en) * 2011-04-29 2012-11-01 Regents Of The University Of Michigan Compounds, formulations, and methods of protein kinase c inhibition
US9588110B2 (en) 2011-07-28 2017-03-07 Cell Signaling Technology, Inc. Multi component antibody based detection technology
WO2023101556A1 (en) * 2021-12-02 2023-06-08 Rijksuniversiteit Groningen Novel inhibitors of aspartate transcarbamoylase (atcase) and compositions, methods and uses related thereto.

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5958910A (en) * 1996-07-19 1999-09-28 Hoffmann-La Roche Inc. Use of N-(4-aryl-thiazol-2-yl)-sulfonamides
US20070275962A1 (en) * 2003-09-10 2007-11-29 Gpc Biotech Ag Heterobicyclic Compounds as Pharmaceutically Active Agents

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5958910A (en) * 1996-07-19 1999-09-28 Hoffmann-La Roche Inc. Use of N-(4-aryl-thiazol-2-yl)-sulfonamides
US20070275962A1 (en) * 2003-09-10 2007-11-29 Gpc Biotech Ag Heterobicyclic Compounds as Pharmaceutically Active Agents

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
OCHSNER ET AL.: 'Mode of Action and Biochemical Characterization of REP8839, a Novel Inhibitor of Methionyl-tRNA Synthetase.' ANTIMICROB. AGENTS CHEMOTHER. vol. 49, no. 10, 2005, pages 4253 - 4262 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012149452A1 (en) * 2011-04-29 2012-11-01 Regents Of The University Of Michigan Compounds, formulations, and methods of protein kinase c inhibition
US8889672B2 (en) 2011-04-29 2014-11-18 The Regents Of The University Of Michigan Compounds, formulations, and methods of protein kinase C inhibition
US9588110B2 (en) 2011-07-28 2017-03-07 Cell Signaling Technology, Inc. Multi component antibody based detection technology
US10577640B2 (en) 2011-07-28 2020-03-03 Cell Signaling Technology, Inc. Multi component detection
WO2023101556A1 (en) * 2021-12-02 2023-06-08 Rijksuniversiteit Groningen Novel inhibitors of aspartate transcarbamoylase (atcase) and compositions, methods and uses related thereto.

Also Published As

Publication number Publication date
WO2009124086A3 (en) 2010-02-25

Similar Documents

Publication Publication Date Title
AU2009232276B2 (en) Antiviral drugs for treatment of arenavirus infection
US20070134729A1 (en) Novel antibacterial agents
US20100029597A1 (en) Reca inhibitors with antibiotic activity, compositions and methods of use
KR20150119007A (en) Methods of treating topical microbial infections
US8461188B2 (en) Therapeutic combination of daptomycin and protein synthesis inhibitor antibiotic, and methods of use
JP5782615B2 (en) Methods of treatment using a single dose of oritavancin
JP2018517770A (en) Halogenated salicylanilide for treating Clostridium infection
US20190274978A1 (en) Antibacterial compositions
CN110279698A (en) Cephalo spy coughs up a ceremonial jade-ladle, used in libation antibiotic composition
US11951148B1 (en) Pristinamycin compositions, LpxC compositions, their improvements, and combinations thereof
US10758529B2 (en) Pharmaceutical compositions containing azaquinone for inhibiting clostridium difficile activity
TW201806604A (en) 9-aminomethyl minocycline compounds and methods of use thereof in urinary tract infection (UTI) treatment
BR112020009973A2 (en) antimicrobial peptides and methods of using them
US20070196859A1 (en) Novel antibacterial agents
WO2009124086A2 (en) Antibacterial drug for treatment of staphylococcus aureus and other gram-positive bacterial infections
WRIGHT et al. The penicillins
CN102573882A (en) Methods of treating bacterial infections using oritavancin
Hassan et al. A patent review of pharmaceutical and therapeutic applications of oxadiazole derivatives for the treatment of chronic diseases (2013–2021)
CA3130739A1 (en) Compositions providing enhanced antibacterial activity against gram-positive bacteria and use thereof
WO2000056725A1 (en) N-adamant-1-yl-n'-[4-chlorobenzothiazol-2-yl] urea useful in the treatment of inflammation and as an anticancer radiosensitizing agent
KR101649675B1 (en) Composition for antibiotics against Staphylococcus aureus
JP6552648B2 (en) Antibacterial composition
US20080171766A1 (en) Novel Method
Standard Orchid Healthcare
US20190282592A1 (en) Drug Target For Treating Veterinary Infections And Methods Of Using Same

Legal Events

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

Ref document number: 09728282

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09728282

Country of ref document: EP

Kind code of ref document: A2