US20220257584A1 - Methods of treating infections in overweight and obese patients using antibiotics - Google Patents

Methods of treating infections in overweight and obese patients using antibiotics Download PDF

Info

Publication number
US20220257584A1
US20220257584A1 US17/406,805 US202117406805A US2022257584A1 US 20220257584 A1 US20220257584 A1 US 20220257584A1 US 202117406805 A US202117406805 A US 202117406805A US 2022257584 A1 US2022257584 A1 US 2022257584A1
Authority
US
United States
Prior art keywords
group
overweight
obese
patient
present disclosure
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US17/406,805
Other languages
English (en)
Inventor
Erin M. Duffy
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Melinta Subsidiary Corp
Original Assignee
Melinta Subsidiary Corp
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=51581190&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20220257584(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Melinta Subsidiary Corp filed Critical Melinta Subsidiary Corp
Priority to US17/406,805 priority Critical patent/US20220257584A1/en
Publication of US20220257584A1 publication Critical patent/US20220257584A1/en
Assigned to SILICON VALLEY BANK reassignment SILICON VALLEY BANK AMENDED AND RESTATED INTELLECTUAL PROPERTY SECURITY AGREEMENT Assignors: MELINTA SUBSIDIARY CORP.
Priority to US18/324,754 priority patent/US20240165100A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/133Amines having hydroxy groups, e.g. sphingosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/40Cyclodextrins; Derivatives thereof

Definitions

  • the present disclosure relates generally to methods of treating infections in overweight and obese patients using antibiotics.
  • NCHS National Center for Health Statistics
  • Overweight and obese individuals are also at increased risk for many diseases and chronic health conditions, including hypertension, osteoarthritis, dyslipidemia, type 2 diabetes, heart disease, stroke, gallbladder disease, sleep apnea and respiratory problems and certain cancers (such as cancers of the pancreas, kidney, prostate, endometrium, breast and colon).
  • diseases and chronic health conditions including hypertension, osteoarthritis, dyslipidemia, type 2 diabetes, heart disease, stroke, gallbladder disease, sleep apnea and respiratory problems and certain cancers (such as cancers of the pancreas, kidney, prostate, endometrium, breast and colon).
  • renal function and some hepatic function are increased in overweight and obese patients, which can complicate the calculation of factors used to determine dosing for antibiotics. See, e.g., Griffin, K. A. et al. Am. J. Physiol. Renal. Physiol. 2008, 294(4), F685-F696.
  • Methods of calculating renal function e.g., glomerular filtration rate (GFR) or creatinine clearance rate
  • GFR glomerular filtration rate
  • creatinine clearance rate e.g., glomerular filtration rate (GFR) or creatinine clearance rate
  • antibiotic drugs such as allergies, rashes, hives, anaphylaxis, hypersensitivities, pruritus, infusion site pain, fatigue, gastrointestinal disorder, thrombocytopenia, phototoxicity, elevated liver enzymes, dysglycemia, QT prolongation, diarrhea, abdominal pain, nausea, vomiting, drug fever, serum sickness, vaginal candidiasis, renal toxicity, ototoxicity, dizziness, nystagmus, headache, liver toxicity, anorexia, hemolytic anemia, peripheral neuropathy, flushing, hypotension, itching, phlebitis, taste alteration, photosensitivity, tooth discoloration, lethargy, pseudomembranous colitis, jaundice and metallic taste.
  • FIG. 1 shows a comparison of cure and failure rates in patients treated for Acute Bacterial Skin and Skin Structure Infections (ABSSSI) with either delafloxacin or vancomycin for all patients (i.e. having any BMI).
  • ABSSSI Acute Bacterial Skin and Skin Structure Infections
  • FIG. 2 shows a comparison of cure and failure rates in patients treated for ABSSSI with either delafloxacin or vancomycin for patients having a BMI greater than or equal to 40 (morbidly obese and super obese).
  • FIG. 3 shows a comparison of cure and failure rates in patients treated for ABSSSI with either delafloxacin or vancomycin for patients having a BMI greater than or equal to 35 (severely obese, morbidly obese and super obese).
  • FIG. 4 shows a comparison of cure and failure rates in patients treated for ABSSSI with either delafloxacin or vancomycin for patients having a BMI greater than or equal to 30 (obese, severely obese, morbidly obese and super obese).
  • FIG. 5 shows a comparison of the cure/failure rates in patients treated for ABSSSI with either delafloxacin, linezolid or vancomycin for patients having a BMI greater than or equal to 30 (obese, severely obese, morbidly obese and super obese).
  • FIG. 6 shows a comparison of the cure/failure rates in patients treated for ABSSSI with either delafloxacin, linezolid or vancomycin for patients having a BMI greater than 25 and less than 30 (overweight).
  • FIG. 7 shows a comparison of the cure/failure rates in patients treated for ABSSSI with either delafloxacin, linezolid or vancomycin for patients having a BMI less than or equal to 25 (normal).
  • the present disclosure relates generally to methods of treating infections in overweight and obese patients using antibiotics.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is not based on the body mass index of the patient.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is independent of the body mass index of the patient.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is not determined by the body mass index of the patient.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is not calculated from the body mass index of the patient.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is not based on the weight or body surface area of the patient.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 10 mg/kg.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is from about 0.01 mg/kg to about 7 mg/kg.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is from about 10 mg/day to about 600 mg/day.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is about 1.5 to about 10-fold lower than a dose of the antibiotic compound that produces at least one side effect selected from the group consisting of allergies, rashes, hives, anaphylaxis, hypersensitivities, pruritus, infusion site pain, fatigue, gastrointestinal disorder, thrombocytopenia, phototoxicity, elevated liver enzymes, dysglycemia, QT prolongation, diarrhea, abdominal pain, nausea, vomiting, drug fever, serum sickness, vaginal candidiasis, renal toxicity, ototoxicity, dizziness, nystagmus, headache, liver toxicity, anorexia, hemolytic anemia, peripheral neuropathy, flushing, hypotension, itching, phlebitis, taste alteration, photosensitivity, tooth discoloration, lethargy, pseudomembranous
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is based on the therapeutically effective amount for a patient who is not overweight or obese.
  • the present disclosure results in part from the discovery that overweight and obese patients with infections were not being properly dosed with antibiotics, resulting in high incidence of adverse events and reduced efficacy.
  • the inventors have surprisingly found that bacterial infections can be treated in overweight and obese patients without the need to adjust the dosage of antibiotics for the patient's weight.
  • ABSSSI Bacterial Skin and Skin Structure Infections
  • MRaSA methicillin-resistant Staphylococcus aureus
  • the inventors have discovered that when delafloxacin was administered at a flat dosage of 300 mg BID to patients regardless of weight, cure rates were superior to those rates achieved with administration of a flat dosage of linezolid (600 mg BID) and vancomycin (1,000-2,000 mg BID), with no increase in adverse events.
  • delafloxacin has no specific limiting toxicity as do vancomycin (kidney) and linezolid (bone marrow).
  • the methods disclosed herein for treating infections in overweight and obese patients provide numerous advantages over the methods known in the art. For example, with traditional methods, as dosages increase with the weight of an overweight and obese patient, the potential for toxicity and adverse events also increases. In contrast, the present methods provide for consistently lower doses of antibiotics which reduce the toxicity and adverse event risks seen in prior methods. Further, because of the increased potential for toxicity and adverse events brought by prior methods, patients frequently require close monitoring. In contrast, the need to monitor overweight and obese patients treated with the present methods is significantly reduced. This also results in the possibility for outpatient administration or earlier release of patients, with later follow-up, reducing burdens on patients and health care providers. Compliance can also be expected to be improved because of the reduction in side effects.
  • the present methods reduce the burden on physicians and clinicians to calculate dosages for patients based on complex calculations of renal function, volume of distribution and other relevant factors. Instead, a consistent low dosage may be used without reference to the patient's weight. Still further, because overweight and obese patients are generally less healthy than non-overweight and obese patients, overweight and obese patients tend to be taking other medications, such as for hypertension, diabetes, etc. With the reduced dosage of antibiotics used in the present methods comes a reduced potential for drug-drug interactions with an overweight and obese patient's other medications.
  • compositions are described as having, including, or comprising specific components, it is contemplated that compositions also consist essentially of, or consist of, the recited components.
  • methods or processes are described as having, including, or comprising specific process steps, the processes also consist essentially of, or consist of, the recited processing steps.
  • order of steps or order for performing certain actions is immaterial so long as the method remains operable.
  • two or more steps or actions can be conducted simultaneously.
  • BMI Body Mass Index
  • normal as used herein with respect to body weight refers to a BMI less than or equal to 25.
  • weight refers to a BMI greater than 25 and less than 30.
  • severely obese refers to a BMI greater than or equal to 35 and less than 40.
  • morbidly obese refers to a BMI greater than or equal to 40 and less than 45.
  • the term “super obese” as used herein refers to a BMI greater than or equal to 45.
  • BMI Body Mass Index
  • terapéuticaally effective amount refers to the quantity of a compound which, when administered to a patient, results in a discernible physiological effect in the patient.
  • Cure or “Investigators' Global Assessment of Cure” as used herein refers to complete resolution of all signs and symptoms at the follow-up visit.
  • One or more disclosed compounds can be incorporated into a pharmaceutical composition or medicament.
  • the disclosed compounds can be administered by a variety of known methods, including, for example, orally, rectally, or by parenteral routes (e.g., intramuscular, intravenous, subcutaneous, nasal or topical).
  • parenteral routes e.g., intramuscular, intravenous, subcutaneous, nasal or topical.
  • the form in which the compounds are administered will be determined by the route of administration.
  • Such forms include, but are not limited to, capsular and tablet formulations (for oral and rectal administration), liquid formulations (for oral, intravenous, intramuscular, subcutaneous, ocular, intranasal, inhalation-based and transdermal administration) and slow releasing microcarriers (for rectal, intramuscular or intravenous administration).
  • the pharmaceutical composition or medicament can also contain a pharmaceutically acceptable vehicle, diluent, excipient or carrier and optional adjuvants, flavorings, colorants, wetting agents, emulsifying agents, pH buffering agents and preservatives.
  • a pharmaceutically acceptable vehicle include, for example, saline, sterile water, Ringer's solution and isotonic sodium chloride solutions.
  • the specific dosage level of active ingredient will depend upon a number of factors, including, for example, biological activity of the particular preparation, age, body weight, sex and general health of the patient being treated.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • side effect refers to the occurrence of an undesired physiological effect in a patient.
  • patient refers to a human to be treated by the presently disclosed methods.
  • the phrase “pharmaceutically acceptable” refers to those compounds, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines, alkali or organic salts of acidic residues such as carboxylic acids, and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include, but are not limited to, those derived from inorganic and organic acids selected from 2-acetoxybenzoic, 2-hydroxyethane sulfonic, acetic, ascorbic, benzene sulfonic, benzoic, bicarbonic, carbonic, citric, edetic, ethane disulfonic, ethane sulfonic, fumaric, glucoheptonic, gluconic, glutamic, glycolic, glycollyarsanilic, hexylresorcinic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxymaleic, hydroxynaphthoic, isethionic, lactic, lactobionic, lauryl sulfonic, maleic, malic, mandelic, methane sulfonic, napsylic, nitric, oxalic, pamoic, pantothenic, phenylacetic, phosphoric
  • the pharmaceutically acceptable salts disclosed herein can be synthesized from a parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two. In one embodiment, non-aqueous media, for example ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are useful for forming salts of the present compounds. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 18 th ed. (Mack Publishing Company, 1990). For example, salts can include, but are not limited to, the hydrochloride and acetate salts of the aliphatic amine-containing, hydroxyl amine-containing, and imine-containing compounds of the present disclosure.
  • the compounds disclosed herein can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules.
  • Nonlimiting examples of hydrates include monohydrates, dihydrates, etc.
  • Nonlimiting examples of solvates include ethanol solvates, acetone solvates, etc.
  • esters refer to derivatives of the disclosed compounds wherein the parent compound is modified by an alcohol ester of a carboxylic acid or a carboxylic acid ester of an alcohol.
  • the compounds disclosed herein can also be prepared as esters, for example pharmaceutically acceptable esters.
  • a carboxylic acid function group in a compound can be converted to its corresponding ester, e.g., a methyl, ethyl, or other ester.
  • an alcohol group in a compound can be converted to its corresponding ester, e.g., an acetate, propionate, or other ester.
  • unit dosage means a single dose of a pharmaceutical composition that is intended to be administered in its entirety.
  • a unit dosage is a convenient form for administering a premeasured amount of a drug active.
  • compositions of the present disclosure comprise a quinolone carboxylic acid compound, or a pharmaceutically acceptable salt, ester, or prodrug thereof, as an antimicrobial compound.
  • the quinolone carboxylic acid compound is alternatively known as a pyridone carboxylic acid or a pyridone carboxylic acid derivative.
  • the term “quinolone carboxylic acid compounds” includes quinolone carboxylic acid compounds, useful herein which are described, including their synthesis, formulation, and use, in U.S. Pat. No. 6,156,903, to Yazaki et al., issued Dec. 5, 2000 and its certificates of correction of Nov. 13, 2001 and Dec. 11, 2001; U.S. Pat. No.
  • quinolone carboxylic acid compounds disclosed herein include compounds corresponding to the following structure:
  • R 1 represents a hydrogen atom or a carboxyl protective group
  • R 2 represents a hydroxyl group, a lower alkoxy group, or a substituted or unsubstituted amino group
  • R 3 represents a hydrogen atom or a halogen atom
  • R 4 represents a hydrogen atom or a halogen atom
  • R 5 represents a halogen atom or an optionally substituted saturated cyclic amino group
  • R 6 represents a hydrogen atom, a halogen atom, a nitro group, or an optionally protected amino group
  • X, Y and Z may be the same or different and respectively represent a nitrogen atom, —CH ⁇ or —CR 7 ⁇ (wherein R 7 represents a lower alkyl group, a halogen atom, or a cyano group); and W represents a nitrogen atom or ⁇ CR 8 ⁇ (wherein R 8 represents a hydrogen atom, a halogen atom, or a lower alkyl group), or a
  • At least one of X, Y and Z represent a nitrogen atom.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , W, X, Y, and Z are defined for Formula 2, and not as defined for the cyclodextrins.
  • R 1 represents a hydrogen atom
  • R 2 represents an amino group
  • R 3 and R 4 represent a fluorine atom
  • R 6 represents a hydrogen atom
  • X represents a nitrogen atom
  • Y represents —CR 7 ⁇ (wherein R 7 represents a fluorine atom)
  • Z represents —CH ⁇
  • W is —CR 8 ⁇ (wherein R 8 represents a chlorine atom)
  • R 5 is not a 3-hydroxyazetidine-1-yl group.
  • compositions disclosed herein comprise a quinolone carboxylic acid derivative corresponding to the following compound (A),
  • compositions disclosed herein comprise the quinolone carboxylic acid derivative D-glucitol, 1-deoxy-1-(methylamino)-, 1-(6-amino-3,5-difluoro-2-pyridinyl)-8-chloro-6-fluoro-1,4-dihydro-7-(3-hydroxy-1-azetidinyl)-4-oxo-3-quinolinecarboxylate.
  • compositions disclosed herein comprise a crystalline form of the quinolone carboxylic acid D-glucitol, 1-deoxy-1-(methylamino)-, 1-(6-amino-3,5-difluoro-2-pyridinyl)-8-chloro-6-fluoro-1,4-dihydro-7-(3-hydroxy-1-azetidinyl)-4-oxo-3-quinolinecarboxylate.
  • compositions disclosed herein comprise a the quinolone carboxylic acid D-glucitol, 1-deoxy-1-(methylamino)-, 1-(6-amino-3,5-difluoro-2-pyridinyl)-8-chloro-6-fluoro-1,4-dihydro-7-(3-hydroxy-1-azetidinyl)-4-oxo-3-quinolinecarboxylate trihydrate.
  • compositions disclosed herein comprise a crystalline form of the quinolone carboxylic acid D-glucitol, 1-deoxy-1-(methylamino)-, 1-(6-amino-3,5-difluoro-2-pyridinyl)-8-chloro-6-fluoro-1,4-dihydro-7-(3-hydroxy-1-azetidinyl)-4-oxo-3-quinolinecarboxylate trihydrate.
  • compositions disclosed herein comprise from about 100 mg to about 500 mg of delafloxacin meglumine. In other embodiments, the compositions disclosed herein comprise about 300 mg of delafloxacin meglumine.
  • the present disclosure relates generally to methods of treating infections in overweight and obese patients using antibiotics.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is not based on the body mass index of the patient.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is independent of the body mass index of the patient.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is not determined by the body mass index of the patient.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is not calculated from the body mass index of the patient.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is not based on the weight or body surface area of the patient.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 10 mg/kg.
  • the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 9 mg/kg. In some embodiments, the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 8 mg/kg. In some embodiments, the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 7 mg/kg. In some embodiments, the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 6 mg/kg. In some embodiments, the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 5 mg/kg.
  • the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 4 mg/kg. In some embodiments, the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 3 mg/kg. In some embodiments, the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 2 mg/kg. In some embodiments, the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 1 mg/kg. In some embodiments, the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 0.75 mg/kg.
  • the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 0.5 mg/kg. In some embodiments, the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 0.25 mg/kg. In some embodiments, the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 0.1 mg/kg. In some embodiments, the therapeutically effective amount is less than the amount that would be administered to the patient on a basis of about 0.05 mg/kg.
  • the therapeutically effective amount of the antibiotic compound is administered on a mg/kg once daily basis. In other embodiments, the therapeutically effective amount of the antibiotic compound is administered on a mg/kg twice daily basis.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is from about 0.01 mg/kg to about 7 mg/kg.
  • the therapeutically effective amount is from about 0.1 mg/kg to about 5 mg/kg. In some embodiments, the therapeutically effective amount is from about 0.5 mg/kg to about 3 mg/kg. In some embodiments, the therapeutically effective amount is from about 1 mg/kg to about 2 mg/kg.
  • the therapeutically effective amount of the antibiotic compound is administered on a mg/kg once daily basis. In some embodiments, the therapeutically effective amount of the antibiotic compound is administered on a mg/kg twice daily basis.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is from about 10 mg/day to about 600 mg/day.
  • the therapeutically effective amount is from about 50 mg/day to about 500 mg/day. In some embodiments, the therapeutically effective amount is from about 100 mg/day to about 400 mg/day. In some embodiments, the therapeutically effective amount is from about 200 mg/day to about 300 mg/day.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is about 1.5 to about 10-fold lower than a dose of the antibiotic compound that produces at least one side effect selected from the group consisting of allergies, rashes, hives, anaphylaxis, hypersensitivities, pruritus, infusion site pain, fatigue, gastrointestinal disorder, thrombocytopenia, phototoxicity, elevated liver enzymes, dysglycemia, QT prolongation, diarrhea, abdominal pain, nausea, vomiting, drug fever, serum sickness, vaginal candidiasis, renal toxicity, ototoxicity, dizziness, nystagmus, headache, liver toxicity, anorexia, hemolytic anemia, peripheral neuropathy, flushing, hypotension, itching, phlebitis, taste alteration, photosensitivity, tooth discoloration, lethargy, pseudomembranous
  • the therapeutically effective amount is about 2- to about 8-fold lower. In some embodiments, the therapeutically effective amount is about 3- to about 6-fold lower. In some embodiments, the therapeutically effective amount is about 4- to about 5-fold lower.
  • a method of treating a bacterial infection in an overweight or obese patient comprising administering a therapeutically effective amount of an antibiotic compound to an overweight or obese patient in need thereof, wherein the therapeutically effective amount is based on the therapeutically effective amount for a patient who is not overweight or obese.
  • the antibiotic compound is selected from the group consisting of amikacin, gentamicin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem, cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cephalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefepime, ceftaroline fosamil, ceftobiprole, teicoplanin,
  • the antibiotic compound is a quinolone carboxylic acid antibiotic. In some embodiments, the compound is delafloxacin.
  • the dosage is between about 10 mg/day and about 600 mg/day. In some embodiments, the therapeutically effective amount is between about 50 mg/day to about 500 mg/day. In some embodiments, the therapeutically effective amount is between about 100 mg/day to about 400 mg/day. In some embodiments, the therapeutically effective amount is between about 200 mg/day to about 300 mg/day.
  • the overweight or obese patient has a body mass index greater than or equal to 25. In some embodiments, the overweight or obese patient has a body mass index greater than or equal to 30. In some embodiments, the overweight or obese patient has a body mass index greater than or equal to 35. In some embodiments, the overweight or obese patient has a body mass index greater than or equal to 40.
  • a Phase 2b clinical trial was designed to compare the efficacy of delafloxacin for the treatment of ABSSSI (including infections caused by MRSA) to Zyvox (linezolid), with and without aztreonam, and vancomycin, with and without aztreonam.
  • Delafloxacin met primary and secondary efficacy endpoints evaluated through end of 2011, including endpoints based on the draft guidance from the FDA in ABSSSI.
  • delafloxacin demonstrated numerical benefit over both Zyvox and vancomycin in the secondary endpoint, cessation of lesion spread and absence or resolution of fever at 48 to 72 hours, with cure rates of approximately 78%, 75%, and 73%, respectively.
  • delafloxacin showed that a greater percentage of patients experience a 30% or greater reduction in the size of the lesion at 48 to 72 hours than either comparator.
  • Phase 2b clinical trial a Phase 2a clinical trial had been conducted, the results of which supported our decision to design and conduct a Phase 2b clinical trial.
  • the Phase 2b study was a randomized, double-blind comparison of delafloxacin, Zyvox, with and without aztreonam, and vancomycin, with and without aztreonam, using objective efficacy measures to evaluate the relative clinical responses in subjects with ABSSSI; aztreonam was added by the investigator based on the believed or confirmed presence of Gram-negative bacteria.
  • the trial enrolled a total of 256 subjects across 34 centers in the United States. Subjects were randomized into three treatment arms to receive either delafloxacin, 300 mg intravenously every 12 hours, or the recommended dosing for Zyvox (600 mg every 12 hours), both with and without aztreonam, or vancomycin (1,000 to 2,000 mg every 12 hours), both with and without aztreonam.
  • the primary endpoint for the study was the Investigators' Global Assessment of Cure. Additionally, a key goal was to assess the utility, variability and measurement techniques of several objective measures of clinical efficacy for use in future clinical trials.
  • Efficacy was evaluated at multiple time points during the study, with a focus on the first five days of administration, through assessments of objective signs and symptoms of infection such as the extent/size of infection, fever, measurement of biochemical markers of inflammation and culture and susceptibility testing of bacterial isolates. Delafloxacin also demonstrated numerical benefit over both Zyvox and vancomycin in the microbiological intent-to-treat population, a subset of the intent-to-treat population, with respect to the Investigators' Global Assessment of Cure and the cessation of lesion spread and absence of fever at 48 and 72 hours. The types of infections treated included abscess, wound, cellulitis and burn related infections.
  • FIG. 1 shows cure/failure rates for treatment of ABSSSI with delafloxacin and vancomycin in all patients (i.e. having any BMI).
  • FIG. 2 shows cure/failure rates for treatment of ABSSSI with delafloxacin and vancomycin in patients having a BMI greater than or equal to 40 (super obese and morbidly obese).
  • FIG. 3 shows cure/failure rates for treatment of ABSSSI with delafloxacin and vancomycin in patients having a BMI greater than or equal to 35 (severely obese, morbidly obese and super obese).
  • FIG. 4 shows cure/failure rates for treatment of ABSSSI with delafloxacin and vancomycin in patients having a BMI greater than or equal to 30 (obese, severely obese, morbidly obese and super obese).
  • FIG. 5 shows a comparison of the cure/failure rates in patients treated for ABSSSI with either delafloxacin, linezolid or vancomycin for patients having a BMI greater than or equal to 30 (obese, severely obese, morbidly obese and super obese).
  • FIG. 6 shows a comparison of the cure/failure rates in patients treated for ABSSSI with either delafloxacin, linezolid or vancomycin for patients having a BMI greater than 25 and less than 30 (overweight).
  • FIG. 7 shows a comparison of the cure/failure rates in patients treated for ABSSSI with either delafloxacin, linezolid or vancomycin for patients having a BMI less than or equal to 25 (normal).
  • antimicrobial agents can be used in the methods disclosed herein. These antimicrobial agents can provide their therapeutic effect by a variety of biochemical or biophysical mechanisms.
  • agents useful in the present disclosure can include those which bind to or modulate ribosomal RNA, for example bacterial ribosomal RNA.
  • agents also useful in the present disclosure can include those which bind to or modulate the large ribosomal subunit, for example the large ribosomal subunit of a bacterial organism.
  • agents also useful in the present disclosure can include those which bind to or modulate DNA topoisomerases, for example bacterial DNA topoisomerases.
  • agents also useful in the present disclosure can include those which bind to or modulate bacterial DNA gyrase, for example bacterial DNA gyrase, i.e. gyrase being an example of a topoisomerase.
  • agents also useful in the present disclosure can include those which bind to or modulate bacterial topoisomerase IV.
  • Useful antimicrobial agents include antibacterial agents, antifungal agents, anti-viral agents, and anti-parasitic agents.
  • Useful chemical classes of compounds include those selected from oxazolidinones (e.g., linezolid, torezolid, tedizolid, eperezolid, N-[3-(2-fluoro-4′- ⁇ [(3H-[1,2,3]triazol-4-ylmethyl)-amino]-methyl ⁇ -biphenyl-4-yl)-2-oxo-oxazolidin-5-(S)-ylmethyl]-acetamide, and other oxazolidinones), macrolides, ketolides, streptogramin As, streptogramin Bs, chloramphenicol and chloramphenicol derivatives, fluorfenicol and fluorfenicol derivatives, glycopeptides, pleuromutilins, aminoglycosides, beta-lactams and carbapenems (including carb
  • the compounds useful in the present disclosure can include the pharmaceutically acceptable salts, esters, or prodrugs thereof.
  • the disclosure further provides methods for synthesizing any one of the compounds of the present disclosure.
  • the disclosure also provides pharmaceutical compositions comprising an effective amount of one or more of the compounds of the present disclosure and a pharmaceutically acceptable carrier.
  • the present disclosure further provides methods for making these compounds, carriers, and pharmaceutical compositions.
  • Oxazolidinones and their pharmaceutically acceptable salts, esters, and prodrugs thereof, can be used in the methods, compositions, and uses of the present disclosure.
  • Linezolid i.e. (N-[[(5S)-3-[3-fluoro-4-(4-morpholinyl) phenyl]-2-oxo-5-oxazolidinyl]methyl]acetamide), which is sold under the trade name or proprietary name Zyvox, is a commercially marketed oxazolidinone. See U.S. Pat. No. 6,559,305 B1, to Bergren, issued May 6, 2003; U.S. Pat. No. 5,688,792, to Barbachyn et al., issued Nov. 18, 1997; and M.
  • Nonlimiting examples of oxazolidinones include those selected from the group consisting of the following compounds
  • a salt would be the monohydrochloride salt of the foregoing oxazolidinones A, B, C, and D.
  • the triazole ring is a 5-membered heteroaromatic ring and that the location of the two double bonds drawn in most representations is an arbitrary depiction of one of the multiple structures that can be drawn, and is used for convenience and not intended as a limitation.
  • five different structures sometimes called tautomeric structures, can be drawn to depict a 1,2,3-triazole.
  • tautomeric structures can be indicated with double-headed arrows between each structure, indicating that the molecules so represented are in equilibrium with each other. For example, for Compound C, the following tautomeric structures can be drawn:
  • oxazolidinones useful herein and compounds such as oxazolidinones C and D are found in U.S. Pat. No. 6,969,726 B2, to Lou et al., issued Nov. 29, 2005, cited above.
  • Compound C is also known by the chemical name: Acetamide, N-[[(5S)-3-(2-Fluoro-4′-[[(1H-1,2,3-triazole-4-ylmethyl)-amino]methyl] [1,1′-biphenyll-4-yl]-2-oxo-5-oxazolidinyl]-methyl]-, and has the CAS registry number 869884-78-6.
  • the monohydrochloride salt of compound C is also known by the chemical name: Acetamide, N-[[(5S)-3-(2-Fluoro-4′-[[(1H-1,2,3-triazole-4-ylmethyl)-aminolmethyl] [1,1′-biphenyl]-4-yl]-2-oxo-5-oxazolidinyl]-methyl]-, monohydrochloride, and has the CAS registry number 869884-77-5.
  • Tedizolid is disclosed in U.S. Pat. No. 7,816,376, U.S. Patent Application Publication No. US 2009/0192197 and International Patent Application Publication No. WO 2004/058886, the contents of the foregoing of which are all incorporated herein by reference in their entireties.
  • A is selected from the group consisting of:
  • B is selected from the group consisting of:
  • M is selected from the group consisting of:
  • M-L is selected from the group consisting of:
  • X at each occurrence, independently is selected from the group consisting of:
  • L 1 is selected from the group consisting of:
  • L 2 is selected from the group consisting of:
  • n 0, 1, 2, 3, or 4;
  • n at each occurrence, independently is 0, 1, 2, 3, or 4;
  • A, B, L, M, R 1 , R 2 , R 3 , m, and n are defined as described above.
  • Particular compounds include those where A is selected from the group consisting of phenyl and pyridyl; B is selected from the group consisting of phenyl and pyridyl; m is 0, 1, or 2; and n is 0, 1, or 2.
  • A-B is:
  • A-B is:
  • A-B is:
  • R 3 is —NHC(O)R 4 .
  • Particular compounds according to these embodiments include those where R 4 is —CH3.
  • R 3 is:
  • A, B, L, M, R 1 , R 2 , m, and n are defined as described above.
  • M-L is M-L 1
  • L 1 is C 1-6 alkyl.
  • M-L 1 is M-CH2—.
  • M-L is M-L 1 -X-L 2
  • X is —NR 4 —.
  • X is —NH—, —N(O)—, or —N(OR 4 )—, where R 4 is H or C 1-6 alkyl.
  • Other compounds include those where X is
  • L 1 is C 1-6 alkyl
  • L 2 is C 1-6 alkyl
  • L 1 is —CH 2 —
  • L 2 is —CH2—
  • Particular examples of compounds according to these embodiments include those where M-L is M-CH 2 —NH—CH 2 — or
  • M-L is M-S—C—NR 4 -L 2 , wherein L 1 is C 1-6 alkyl, and L 2 is C 1-6 alkyl.
  • M-L is M-S—CH 2 CH 2 —NH—CH 2 —.
  • M is selected from the group consisting of:
  • A is phenyl, substituted phenyl, pyridyl, or substituted pyridyl.
  • M-L is not (imidazol-1-yl)methyl or (morpholin-4-yl)methyl.
  • B is phenyl or substituted phenyl. More preferably, B is substituted phenyl. In some embodiments, substituents include halogens.
  • M-L is selected from the group consisting of M-X, M-L 1 -X, M-L 1 -X-L 2 , M-X—X—, M-L 1 -X—X—, M-X—X-L 2 , and M-L 1 -X—X—L 2 .
  • M-L is selected from the group consisting of M-X, M-L 1 -X, M-L 1 -X-L 2 , M-L 1 -X-L 2 -X, M-X—X—, M-X—X-L 2 , and M-L 1 -X—X-L 2 .
  • Pyridonecarboxylic acid derivatives of the methods, compositions, and uses of the present disclosure include compounds corresponding to the following structure (Pyridonecarboxylic Acid Derivative 1)
  • R 1 represents a hydrogen atom or a carboxyl protective group
  • R 2 represents a hydroxyl group, a lower alkoxy group, or a substituted or unsubstituted amino group
  • R 3 represents a hydrogen atom or a halogen atom
  • R 4 represents a hydrogen atom or a halogen atom
  • R 5 represents a halogen atom or an optionally substituted saturated cyclic amino group
  • R 6 represents a hydrogen atom, a halogen atom, a nitro group, or an optionally protected amino group
  • X, Y and Z may be the same or different and respectively represent a nitrogen atom, CH or CR 7 (wherein R 7 represents a lower alkyl group, a halogen atom, or a cyano group), with the proviso that at least one of X, Y and Z represent a nitrogen atom, and W represents a nitrogen atom or CR 8 (wherein R 8 represents a hydrogen atom, a halogen
  • R 1 when R 1 is a carboxyl protective group, it may be any carboxylate ester residue which cleaves relatively easily to generate the corresponding free carboxyl group.
  • exemplary carboxyl protective groups include those which may be eliminated by hydrolysis, catalytic reduction, and other treatments under mild conditions such as lower alkyl groups such as methyl group, ethyl group, n-propyl group, ipropyl group, n-butyl group, i-butyl group, t-butyl group, pentyl group, hexyl group, and heptyl group; lower alkenyl groups such as vinyl group, allyl group, 1-propenyl group, butenyl group, pentenyl group, hexenyl group, and heptenyl group; aralkyl groups such as benzyl group; and aryl groups such as phenyl group and naphthyl group; and those which may be readily eliminated in the body such as lower
  • substituents R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , A, J 1 , J 2 , J 3 , W, X, Y, Z, e, f, and g are defined herein for convenience with respect to the chemical structure for the pyridonecarboxylic acid derivatives, e.g., Pyridonecarboxylic Acid Derivative 1, and do not refer to other substituents for other compounds disclosed herein.
  • the present disclosure relates to a method, composition, or use for a pyridonecarboxylic acid derivative of structure Pyridonecarboxylic Acid Derivative 1, wherein W is CR 8 , wherein R 8 represents a hydrogen atom, a halogen atom, or a lower alkyl group.
  • the present disclosure relates to a method, composition, or use for a pyridonecarboxylic acid derivative of structure Pyridonecarboxylic Acid Derivative 1, wherein R 5 is a group represented by the following formula (a) or (b):
  • A represents an oxygen atom, sulfur atom or NR 9 (wherein R 9 represents hydrogen atom or a lower alkyl group), e represents a number from 3 to 5, f represents a number from 1 to 3, g represents a number from 0 to 2, J 1 , J 2 and J 3 , which may be the same or different from one another, represent a hydrogen atom, hydroxyl group, lower alkyl group, amino lower alkyl group, amino group, lower alkylamino group, lower alkoxy group, or a halogen atom.
  • the present disclosure relates to a method, composition, or use for a pyridonecarboxylic acid derivative of structure Pyridonecarboxylic Acid Derivative 1, wherein R 5 is a group represented by formula (a).
  • the present disclosure relates to a method, composition, or use for a pyridonecarboxylic acid derivative of structure Pyridonecarboxylic Acid Derivative 1, wherein e in the formula (a) is 3 or 4.
  • the present disclosure relates to a method, composition, or use for a pyridonecarboxylic acid derivative of structure Pyridonecarboxylic Acid Derivative 1, wherein R 1 is a hydrogen atom; R 2 is an amino group, lower alkylamino group, or a di-lower alkylamino group; R 3 is a halogen atom; R 4 is a halogen atom; R 6 is hydrogen atom; X is a nitrogen atom; Y and Z are CH or CR 7 (wherein R 7 is a lower alkyl group or a halogen atom); and W is CR 8 (wherein R 8 is a halogen atom or a lower alkyl group).
  • the present disclosure relates to a method, composition, or use for a pyridonecarboxylic acid derivative of structure Pyridonecarboxylic Acid Derivative 1, wherein R 2 is amino group; R 3 is fluorine atom; R 4 is a fluorine atom; Y is CF; Z is CH; W is CR 8 (wherein R 8 is a chlorine atom, bromine atom or a methyl group), and e in formula (a) is 3.
  • the present disclosure relates to a method, composition, or use wherein said pyridonecarboxylic acid corresponds to the following structure:
  • pyridonecarboxylic acid is also known by the publicly disclosed code names ABT-492 and WQ 3034 and also by the chemical name 1-(6-amino-3,5-difluoro-2-pyridinyl)-8-chloro-6-fluoro-1,4-dihydro-7-(3-hydroxy-1-azetidinyl)-4-oxo-3-quinolinecarboxylic acid or 1-(6-amino-3,5-difluoro-2-pyridinyl)-8-chloro-6-fluoro-1,4-dihydro-7-(3-hydroxyazetidin-1-yl)-4-oxo-3-quinolinecarboxylic acid.
  • This carboxylic acid form of the compound corresponds to the CAS registry number 189279-58-1.
  • WO 2006/042034, cited above discloses the D-glucitol salt of this compound [D-glucitol 1-(6-amino-3,5-difluoro-2-pyridinyl)-8-chloro-6-fluoro-1,4-dihydro-7-(3-hydroxy-1-azetidinyl)-4-oxo-3-quinolinecarboxylate (salt)] and the trihydrate of the D-glucitol salt of this compound [D-glucitol 1-(6-amino-3,5-difluoro-2-pyridinyl)-8-chloro-6-fluoro-1,4-dihydro-7-(3-hydroxy-1-azetidinyl)-4-oxo-3-quinolinecarboxylate trihydrate (salt)].
  • the D-glucitol salt and the D-glucitol salt trihydrate correspond to the CAS registry numbers 352458-37-8 and 883105-02-0, respectively.
  • D-glucitol corresponds to the CAS registry number 6284-40-8.
  • WO 2006/042034 also discloses a crystalline form of the D-glucitol salt characterized when measured at about 25° C. with Cu—K ⁇ radiation, by the powder diffraction pattern shown in FIG. 1 of WO 2006/042034 and a crystalline form of the D-glucitol salt trihydrate when measured at about 25° C. with Cu—K ⁇ radiation, by the powder diffraction pattern shown in FIG. 2 of WO 2006/042034.
  • These D-glucitol salts are useful in the present disclosure. Also, see A. R. Haight et al., “Synthesis of the Quinolone ABT-492: Crystallizations for Optimal Processing”, Organic Process Research & Development (2006), 10(4), 751-756.
  • quinolone compounds useful herein include fluoroquinolones such asbalofloxacin, ciprofloxacin, clinafloxacin, enoxacin, fleroxacin, gatifloxacin, gemifloxacin, grepafloxacin, levofloxacin, lomefloxacin, moxifloxacin, nadifloxacin, norfloxacin, ofloxacin, pazufloxacin, pefloxacin, prulifloxacin, rufloxacin, sitafloxacin, sparfloxacin, temafloxacin, tosufloxacin, and trovafloxacin.
  • fluoroquinolones such asbalofloxacin, ciprofloxacin, clinafloxacin, enoxacin, fleroxacin, gatifloxacin, gemifloxacin, grepafloxacin, levofloxacin, lomeflox
  • Garenoxacin and its pharmaceutically acceptable salts, esters, and prodrugs thereof can be used in the methods, compositions, and uses of the present disclosure.
  • Garenoxacin is also known as 1-cyclopropyl -8-(difluoromethoxy)-7-(1R)-(1-methyl-2,3-dihydro-1H-5-isoinodyl)-4-oxo-1,4-dihydro-3-quinolinecarboxylic acid methanesulfonate monohydrate and by the publicly disclosed code names T-3811 and BM 284756. See M.
  • Gatifloxacin and its pharmaceutically acceptable salts, esters, and prodrugs thereof can be used in the methods, compositions, and uses of the present disclosure.
  • Gatifloxacin is sold under the trade name or proprietary Tequin. See U.S. Pat. No. 6,589,955 B2, to Raghavan et al., issued Jul. 8, 2003; U.S. Pat. No. 5,880,283, to Matsumoto et al., issued Mar. 9, 1999; and U.S. Pat. No. 4,980,470, to Masuzawa et al., issued Dec. 25, 1990 and its certificate of correction of Aug. 11, 1992.
  • Gemifloxacin and its pharmaceutically acceptable salts, esters, and prodrugs thereof can be used in the methods, compositions, and uses of the present disclosure.
  • Gemifloxacin is sold under the trade name or proprietary Factive. See U.S. Pat. No. 6,803,376 B1, to Appelbaum et al., issued Oct. 12, 2004; U.S. Pat. No. 6,723,734 B2, to Kim et al., issued Apr. 20, 2004; U.S. Pat. No. 6,455,540 B1, to Citron et al., issued Sep. 24, 2002; U.S. Pat. No. 6,340,689 B1, to Dubois et al., issued Jan. 22, 2002 and its certificate of correction of Jun. 18, 2002; U.S.
  • Levofloxacin and its pharmaceutically acceptable salts, esters, and prodrugs thereof can be used in the methods, compositions, and uses of the present disclosure.
  • Levofloxacin is sold under the trade name or proprietary Levaquin. See U.S. Pat. No. 5,053,407, to Hayakawa et al., issued Oct. 1, 1991 and its certificate of correction of Sep. 27, 1994.
  • Moxifloxacin and its pharmaceutically acceptable salts, esters, and prodrugs thereof can be used in the methods, compositions, and uses of the present disclosure.
  • Moxifloxacin is sold under the trade name or proprietary Avelox. See U.S. Pat. No. 5,849,752, to Grunenberg et al., issued Dec. 15, 1998; U.S. Pat. No. 5,607,942, to Petersen et al., issued Mar. 4, 1997; and U.S. Pat. No. 4,990,517, to Petersen et al., issued Feb. 5, 1991 and its certificate of correction of Apr. 25, 1995.
  • Benzoheterocyclic compounds useful herein are described, including their synthesis, formulation, and use, in U.S. Pat. No. 6,753,333 B2, to De Souza et al., issued Jun. 22, 2004; U.S. Pat. No. 6,750,224 B1, to Patel et al, issued Jun. 15, 2004 and its certificate of correction of Nov. 2, 2004; U.S. Pat. No. 6,664,267 B1, to de Souza et al., issued Dec. 16, 2003; U.S. Pat. No. 6,608,078 B2, to De Souza et al., issued Aug. 19, 2003; U.S. Pat. No. 6,514,986 B2 to De Souza et al., issued Feb. 4, 2003; U.S. Pat. No. 4,552,879 to Ishikawa et al., issued Nov. 12, 1985; and U.S. Pat. No. 4,399,134 to Ishikawa et al., issued Aug. 16, 1983.
  • Benzoheterocyclic compounds of the methods, compositions, and uses of the present disclosure include compounds corresponding to the following structure (Benzoheterocyclic Compound I)
  • R 1 represents a hydrogen atom or a lower alkyl group
  • R 2 represents a hydrogen atom or a halogen atom
  • R 3 represents a 1-pyrrolidinyl group which may be substituted with a hydroxymethyl group, a 1,2,5,6-tetrahydro-1-pyridyl group, or a group of the formula
  • R 4 represents a hydrogen atom, a lower alkyl group, a lower alkoxy group, a hydroxy group, a phenyl-lower alkyl group, a lower alkanoyloxy group, an amino group which may be substituted with a lower alkyl group or a lower alkanoyl group, an oxo group, or a carbamoyl group;
  • Z represents an oxygen atom, a sulfur atom or a methylene group; and m is 1 or 2; and n is an integer of 1 or 2; or a pharmaceutically acceptable salt ester or prodrug thereof
  • substituents R 1 , R 2 , R 3 , R 4 , Z, m, and n are defined herein for convenience with respect to the chemical structure for the benzoheterocyclic compounds, e.g., benzoheterocyclic compound (I) and do not refer to other substituents for other compounds of the present disclosure.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein n is 2.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein n is 1.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 3 represents a group of the formula
  • R 4 represents a hydrogen atom, a lower alkyl group, a lower alkoxy group, a hydroxy group, a phenyl-lower alkyl group, a lower alkanoyloxy group, an amino group which may be substituted with a lower alkyl group or a lower alkanoyl group, an oxo group, or a carbamoyl group;
  • Z represents an oxygen atom, a sulfur atom or a methylene group; and m is 1 or 2; and n is 1.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 3 represents a 1-pyrrolidinyl group which may be substituted with a hydroxymethyl group or a 1,2,5,6-tetrahydro-1-pyridyl group.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 4 represents a hydrogen atom, a hydroxy group or a lower alkanoyloxy group and the position at which the group of the formula
  • R 4 represents a hydrogen atom, a lower alkyl group, a lower alkoxy group, a hydroxy group, a phenyl-lower alkyl group, a lower alkanoyloxy group, an amino group which may be substituted with a lower alkyl group or a lower alkanoyl group, an oxo group, or a carbamoyl group;
  • Z represents an oxygen atom, a sulfur atom or a methylene group; and m is 1 or 2; and n is 1, is attached is the 8-position.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 4 represents a lower alkyl group, a lower alkoxy group, a phenyl-lower alkyl group, an amino group which may be substituted with a lower alkyl group or a lower alkanoyl group, an oxo group, a carbamoyl group, and the position at which the group of the formula
  • R 4 represents a hydrogen atom, a lower alkyl group, a lower alkoxy group, a hydroxy group, a phenyl-lower alkyl group, a lower alkanoyloxy group, an amino group which may be substituted with a lower alkyl group or a lower alkanoyl group, an oxo group, or a carbamoyl group;
  • Z represents an oxygen atom, a sulfur atom or a methylene group; and m is 1 or 2; and n is 1, is attached is the 8-position.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 2 represents a halogen atom.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 2 represents a hydrogen atom.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 2 represents a fluorine atom and the position at which the fluorine atom is attached is the 9-position.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 2 represents a chlorine atom and the position at which the fluorine atom is attached is the 9-position.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 1 represents a lower alkyl group.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 1 represents a methyl group.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 2 represents a fluorine atom attached to the 9-position and R 1 represents a methyl group.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 1 represents a methyl group, R 2 represents a fluorine atom attached to the 9-position and the position at which the group represented by R 3 is attached is the 8-position.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein the position at which R 3 is attached is the 9-position.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 1 represents a methyl group, R 2 represents a fluorine atom attached to the 8-position.
  • the present disclosure relates to a method, composition, or use for a benzoheterocyclic of structure Benzoheterocyclic Compound I, wherein R 1 represents a methyl group, R 2 represents a chlorine atom attached to the 8-position.
  • the present disclosure relates to a method, composition, or use wherein said benzoheterocyclic compound is 9-fluoro-8-(4-hydroxy-1-piperidyl)-5-methyl-6,7-dihydro-1-oxo-1H,5H-benzo[i,j]quinolizine-2-carboxylic acid or a pharmaceutically acceptable salt, ester, or prodrug thereof
  • the present disclosure relates to a method, composition, or use wherein said benzoheterocyclic compound is S-( ⁇ )-9-fluoro-6,7-dihydro-8-(4-hydroxypiperidin-1-yl)-5-methyl-1-oxo-1H,5H-benzo[i,j]quinolizine-2-carboxylic acid or a pharmaceutically acceptable salt, ester, or prodrug thereof.
  • said benzoheterocyclic compound is S-( ⁇ )-9-fluoro-6,7-dihydro-8-(4-hydroxypiperidin-1-yl)-5-methyl-1-oxo-1H,5H-benzo[i,j]quinolizine-2-carboxylic acid or a pharmaceutically acceptable salt, ester, or prodrug thereof.
  • the foregoing compound is also known by the chemical name nadifloxacin.
  • the present disclosure relates to a method, composition, or use wherein said benzoheterocyclic compound is S-( ⁇ )-9-fluoro-6,7-dihydro-8-(4-hydroxypiperidin-1-yl)-5-methyl-1-oxo-1H,5H-benzo[i,j]quinolizine-2-carboxylic acid arginine salt.
  • the present disclosure relates to a method, composition, or use wherein said benzoheterocyclic compound is a specific polymorph or crystalline form of S-( ⁇ )-9-fluoro-6,7-dihydro-8-(4-hydroxypiperidin-1-yl)-5-methyl-1-oxo-1H,5H-benzo[i,j]quinolizine-2-carboxylic acid arginine salt.
  • the present disclosure relates to a method, composition, or use wherein said benzoheterocyclic compound is S-( ⁇ )-9-fluoro-6,7-dihydro-8-(4-hydroxypiperidin-1-yl)-5-methyl-1-oxo-1H,5H-benzo[i,j]quinolizine-2-carboxylic acid arginine salt having the following X-ray diffraction data: (2 ⁇ ): 10.16, 11.78, 12,52, 16.00, 18.94, 19.66, 20.36, 21.28, 21.92, 22.52, 24.74, 25.28, 30.74.
  • the present disclosure relates to a method, composition, or use wherein said benzoheterocyclic compound is S-( ⁇ )-9-fluoro-6,7-dihydro-8-(4-hydroxypiperidin-1-yl)-5-methyl-1-oxo-1H,5H-benzo[i,j]quinolizine-2-carboxylic acid arginine salt having the following X-ray diffraction data: (2 ⁇ ): 18.28, 18.8, 19.8, 20.12, 20.62, 21.10, 21.44, 21.88, 22.6, 23.02.
  • the present disclosure relates to a method, composition, or use wherein said benzoheterocyclic compound is S-( ⁇ )-9-fluoro-6,7-dihydro-8-(4-hydroxypiperidin-1-yl)-5-methyl-1-oxo-1H,5H-benzo[i,j]quinolizine-2-carboxylic acid arginine salt having the following X-ray diffraction data: (2 ⁇ ): 14.02+0.2, 14.82+0.2, 19.28+0.2, 22.12+0.2, 22.96+0.2, 23.46+0.2, 28.36+0.2.
  • Beta-lactams for example carbapenems, examples of which are carbapenems with a 7-acylated imidazo[5-1,b]thiazole-2-yl group directly attached to the carbapenem moiety of the C-2 position, useful herein are described, including their synthesis, formulation, and use, in M. Kurazano et al., “In Vitro Activities of ME1036 (CP5609), a Novel Parenteral Carbapenem, against Methicillin-Resistant Staphylococci”, Antimicrobial Agents and Chemotherapy, vol. 48, no. 8, pp. 2831-2837 (August 2004); U.S. Patent Application Publication No.
  • Beta-lactam compounds of the methods, compositions, and uses of the present disclosure include compounds corresponding to the following structure (Beta-Lactam I)
  • R 2 and R 3 which may be the same or different, each represent a hydrogen atom; a halogen atom; lower alkyl optionally substituted by a halogen atom, cyano, hydroxyl, carbamoyl, amino, formylamino, lower alkylcarbonylamino, aminosulfonylamino, lower alkylthio, lower alkoxy, lower cycloalkyl, N,N-di-lower alkylamino, or N-carbamoyl lower alkyl-N,N-di-lower alkylammonino; lower cycloalkyl; lower alkylcarbonyl wherein the alkyl portion of lower alkylcarbonyl is optionally substituted by a halogen atom, cyano, hydroxyl, carbamoyl, amino, formylamino, lower alkylcarbonylamino, aminosulfonylamino,
  • substituents R 1 , R 2 , R 3 , Hy, and n are defined herein for convenience with respect to the chemical structure for the beta-lactams or carbapenems, e.g., Beta-Lactam I and Beta-Lactam II, and do not refer to other substituents for other compounds of the present disclosure.
  • the present disclosure relates to a method, composition, or use for a beta-lactam of structure Beta-Lactam I, wherein R 1 represents a hydrogen atom or methyl, R 2 and R 3 , which may be the same or different, each represent a hydrogen atom; a halogen atom; lower alkyl optionally substituted by a halogen atom, cyano, hydroxyl, carbamoyl, amino, formylamino, lower alkylcarbonylamino, aminosulfonylamino, or lower alkylthio; lower alkylcarbonyl wherein the alkyl portion of lower alkylcarbonyl is optionally substituted by a halogen atom, cyano, hydroxyl, carbamoyl, amino, formylamino, lower alkylcarbonylamino, aminosulfonylamino, or lower alkylthio; carbamoyl; aryl; or lower alkylthio wherein the al
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I wherein R 1 represents a hydrogen atom or methyl, R 2 and R 3 , which may be the same or different, each represent a hydrogen atom, a halogen atom, optionally substituted lower alkyl, lower cycloalkyl, lower alkylcarbonyl, carbamoyl, optionally substituted aryl, optionally substituted lower alkylthio, morpholinyl, lower alkylsulfonyl, or formyl, n is an integer of 0 to 2, and Hy represents a group selected from optionally substituted pyridinyl, optionally substituted pyridinium-yl, optionally substituted tetrahydropyridinyl, optionally substituted thiazolyl, optionally substituted pyrimidinyl, optionally substituted thienyl, optionally substituted quinolinyl, optionally substituted
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I wherein R 1 represents a hydrogen atom or methyl, R 2 and R 3 , which may be the same or different, each represent a hydrogen atom, a halogen atom, optionally substituted lower alkyl, optionally substituted lower alkylcarbonyl, carbamoyl, aryl, or optionally substituted lower alkylthio, n is an integer of 0 to 4, and Hy represents a group selected from optionally substituted pyridinyl, optionally substituted pyridinium-yl, optionally substituted tetrahydropyridinyl, optionally substituted thiazolyl, optionally substituted pyrimidinyl, optionally substituted thienyl, optionally substituted quinolinyl, optionally substituted quinolinium-yl, and optionally substituted pyrrolidinyl.
  • R 1 represents a hydrogen atom or
  • Beta-lactam compounds of the methods, compositions, and uses of the present disclosure include compounds corresponding to the following structure (Beta-Lactam II)
  • R 1 represents a hydrogen atom or methyl
  • R 2 and R 3 which may be the same or different, each represent a hydrogen atom; a halogen atom; lower alkyl optionally substituted by a halogen atom, cyano, hydroxyl, carbamoyl, amino, formylamino, lower alkylcarbonylamino, aminosulfonylamino, lower alkylthio, lower alkoxy, lower cycloalkyl, N,N-di-lower alkylamino, or N-carbamoyl lower alkyl-N,N-di-lower alkylammonino; lower cycloalkyl; lower alkylcarbonyl wherein the alkyl portion of lower alkylcarbonyl is optionally substituted by a halogen atom, cyano, hydroxyl, carbamoyl, amino, formylamino, lower alkylcarbonylamino, aminosulfonylamino
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam II, wherein R 1 represents a hydrogen atom or methyl, R 2 and R 3 , which may be the same or different, each represent a hydrogen atom, a halogen atom, lower alkyl optionally substituted by a halogen atom, cyano, hydroxyl, carbamoyl, amino, formylamino, lower alkylcarbonylamino, aminosulfonylamino, or lower alkylthio; lower alkylcarbonyl wherein the alkyl portion of lower alkylcarbonyl is optionally substituted by a halogen atom, cyano, hydroxyl, carbamoyl, amino, formylamino, lower alkylcarbonylamino, aminosulfonylamino, or lower alkylthio; carbamoyl; aryl; or lower alkylthio wherein the al
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein the substituent on the lower alkyl and lower alkylcarbonyl groups optionally represented by R 2 and R 3 is hydroxyl, lower alkoxy, N,N-di-lower alkylamino, or N-carbamoyl lower alkyl-N,N-di-lower alkylammonino, the substituent on the aryl group optionally represented by R 2 and R 3 is N,N-di-lower alkylamino, the substituent on the lower alkylthio group optionally represented by R 2 and R 3 is amino, hydroxyl, or azide, and the substituent on the saturated or unsaturated heterocyclic ring represented by Hy is lower alkyl optionally substituted by carboxylmethyl-substituted carbamoyl, carbamoyl, phenyl, aminophenyl,
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein R 1 represents a hydrogen atom or methyl, R 2 and R 3 represent a hydrogen atom, n is 0 (zero), and Hy represents pyridinium-yl having carbamoylmethyl at its 1-position.
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein n is 0 (zero).
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein R 1 represents methyl, and R 2 and R 3 represent a hydrogen atom.
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein R 1 represents methyl, R 2 and R 3 represent a hydrogen atom, n is 0 (zero), and Hy represents pyridinium-yl which optionally has carbamoyl lower alkyl, carboxyl lower alkyl, or aminosulfonylamino lower alkyl at its 1-position and amino lower alkylthio at other position than the 1-position.
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein R 1 represents methyl, R 2 and R 3 represent a hydrogen atom, n is 0 (zero), and Hy represents pyridin-3-yl.
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein R 1 represents methyl, R 2 and R 3 represent a hydrogen atom, n is 0 (zero), and Hy represents 1-carbamoylmethylpyridinium-3-yl.
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein R 1 , R 2 and R 3 represent a hydrogen atom, n is 0 (zero), and Hy represents 1-carbamoylmethylpyridinium-3-yl.
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein R 1 represents methyl, R 2 and R 3 represent a hydrogen atom, n is 0 (zero), and Hy represents 1-carbamoylmethyl-5-phenylpyridinium-3-yl.
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein R 1 represents methyl, R 2 and R 3 represent a hydrogen atom, n is 0 (zero), and Hy represents (2S)-pyrrolidin-2-yl.
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein R 1 represents methyl, R 2 and R 3 represent a hydrogen atom, n is 0 (zero), and Hy represents 1-carboxymethylpyridinium-3-yl.
  • the present disclosure relates to a method, composition, or use of a beta-lactam of structure Beta-Lactam I or Beta-Lactam II, wherein R 1 represents methyl, R 2 and R 3 represent a hydrogen atom, n is 0 (zero), and Hy represents 1-(2-aminosulfonylaminoethyl)pyridinium-3-yl.
  • the present disclosure relates to a method, composition, or use wherein said beta-lactam or carbapenem corresponds to the following structure:
  • beta-lactam or carbapenem is also known by the publicly disclosed code names ME1036 and CP5609.
  • Aminomethylcycline compounds such as 7-methylamino-9-(2,2-dimethyl-propyl)aminomethylcycline and their pharmaceutically acceptable salts, esters, and prodrugs thereof, can be used in the methods, compositions, and uses of the present disclosure.
  • the compound, 7-methylamino-9-(2,2-dimethyl-propyl)aminomethylcycline is also known by the publicly disclosed code names PTK 0796 and BAY 73-6944. See U.S. Pat. No. 6,846,939 B2, to Nelson et al., issued Jan. 25, 2005; U.S. Patent Application No. US 2005/0070510 A1, to Draper et al., published Mar. 31, 2005; U.S. Patent Application No.
  • Dalbavancin and its pharmaceutically acceptable salts, esters, and prodrugs thereof can be used in the methods, compositions, and uses of the present disclosure.
  • Dalbavancin which is a semisynthetic glycopeptide is also known by the publicly disclosed code names VER-001 and BI397. See G. Candiani et al., “In-Vitro and In-Vivo Antibacterial Activity of BI 397, a New Semi-Synthetic Glycopeptide Antibiotic”, J. Antimicrob. Chemotherapy, 44, pp. 179-192 (1999); U.S. Patent Application No. US 2005/0090433 Al, to Colombo et al., published Apr. 28, 2005; U.S. Patent Application No.
  • Daptomycin and its pharmaceutically acceptable salts, esters, and prodrugs thereof can be used in the methods, compositions, and uses of the present disclosure.
  • Daptomycin is sold under the trade name or proprietary Cubicin. See U.S. Pat. No. 6,852,689 B2, to Oleson, Jr. et al., issued Feb. 8, 2005; U.S. Pat. No. 6,468,967 B1, to Oleson, Jr. et al., issued Oct. 22, 2002; and U.S. Pat. No. 5,912,226, to Baker et al., issued Jun. 15, 1999; and PCT Application No. WO 00/18419, to Cubist Pharmaceuticals, Inc., published Apr. 6, 2000.
  • Oritavancin and its pharmaceutically acceptable salts, esters, and prodrugs thereof can be used in the methods, compositions, and uses of the present disclosure.
  • Oritavancin which is a glycopeptide
  • LY333328 is also known by the publicly disclosed code name LY333328. See R. C. Mercier et al., “Pharmacodynamic Evaluation of a New Glycopeptide, LY333328, and In Vitro Activity against Staphylococcus aureus and Enterococcus faecium ”, Antimicrobial Agents and Chemotherapy, vol. 41, no. 6, pp. 1307-1312 (June 1997); U.S. Pat. No. 5,998,581, to Berglund et al., issued Dec.
  • Televancin and its pharmaceutically acceptable salts, esters, and prodrugs thereof can be used in the methods, compositions, and uses of the present disclosure.
  • Televancin which is a peptidoglycan, can be prepared by the sequential reduction amination of vancomycin and reaction with aminomehtylphosphonic acid.
  • Televancin can also be prepared by the reductive alkylation of vancomycin with N-decyl-N-fluoroenyl-methyloxycarbonyl-2-aminoacetaldehyde via sodium cyano-borohydride and trifluoroacetic acid, and modification of the resorcinol position via Mannich aminomethylation.
  • Televancin can also be prepared from vancomycin or its analogues by the sequential reaction with a protected amino-aldehyde, an amine and then an aminoalkylphosphonic acid in the presence of formaldehyde.
  • a protected amino-aldehyde to Leadbetter et al., issued May 3, 2005
  • U.S. Pat. No. 6,878,686 B2 to Marquess et al., issued Apr. 12, 2005
  • U.S. Pat. No. 6,872,804 B2 to Mu, issued Mar. 29, 2005
  • U.S. Pat. No. 6,872,701 B2 to Leadbetter et al., issued Mar. 29, 2005; U.S. Pat. No.
  • DK-507k and its pharmaceutically acceptable salts, esters, and prodrugs thereof, can be used in the methods, compositions, and uses of the present disclosure.
  • DK-507k can be described as a fluoroquinolone.
  • DK-507k is also known by the chemical name ( ⁇ )-7-[(7S)-7-amino-5-azaspiro[2.4]heptan-5-yl]-6-fluoro-1-[(1R, 2S)-2-fluoro-1-cyclopropyl]-1,4-dihydro-8-methoxy-4-oxo-3-quinolinecarboxylic acid monohydrochloride monohydrate.
  • DK-507k can be represented by the following formula
  • the compound can also be obtained as crystals exhibiting characteristic peaks in the vicinity of angles of diffraction (20) of 6.9, 10.5, 14.4, 23.1, 26.9, and)27.8(° when subjected to powder X-ray diffractometry.
  • anhydrous free acid of the above compound as well as other salts, esters, and prodrugs, and also hydrates of the compounds can be prepared and used in the present disclosure. Also other crystal forms of the foregoing can be prepared and used in the present disclosure.
  • the methods of the present disclosure are useful for treating, preventing or reducing the risk of infection due to, e.g., a skin infection, nosocomial pneumonia, post-viral pneumonia, an abdominal infection, a urinary tract infection, bacteremia, septicemia, endocarditis, an atrio-ventricular shunt infection, a vascular access infection, meningitis, infection due to surgical or invasive medical procedures, a peritoneal infection, a bone infection, a joint infection, a methicillin-resistant Staphylococcus aureus infection, a vancomycin-resistant Enterococci infection, a linezolid-resistant organism infection, tuberculosis, a quinolone resistant Gram-positive infection, a ciprofloxacin resistant methicillin resistant (MRSA) infection, bronchitis, a complicated skin and skin structure infection (cSSSI), an uncomplicated skin and skin structure infection (uSSSI), a community respiratory-tract infection, and a
  • the dose of active compound and mode of administration e.g., injection, intravenous drip, etc. will depend upon the intended patient or subject and the targeted microorganism, e.g., the target bacterial organism.
  • Dosing strategies are disclosed in L. S. Goodman, et al., The Pharmacological Basis of Therapeutics, 201-26 (5th ed.1975), the entire contents of which is herein incorporated in its entirety.
  • compositions can be formulated in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the disclosure are dictated by and directly dependent on the unique characteristics of the active compound and the therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • administration can be by periodic injections of a bolus, or can be made more continuous by intravenous, intramuscular or intraperitoneal administration from an external reservoir (e.g., an intravenous bag).
  • the active compound can be used as part of a transplant procedure, it can be provided to the living tissue or organ to be transplanted prior to removal of tissue or organ from the donor.
  • the compound can be provided to the donor host.
  • the organ or living tissue can be placed in a preservation solution containing the active compound.
  • the active compound can be administered directly to the desired tissue, as by injection to the tissue, or it can be provided systemically, by parenteral administration, using any of the methods and formulations described herein and/or known in the art.
  • any commercially available preservation solution can be used to advantage.
  • useful solutions known in the art include Collins solution, Wisconsin solution, Belzer solution, Eurocollins solution and lactated Ringer's solution.
  • pharmacogenomics i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drug
  • Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drug.
  • a physician or clinician can consider applying knowledge obtained in relevant pharmacogenomics studies in determining whether to administer a drug as well as tailoring the dosage and/or therapeutic regimen of treatment with the drug.
  • the amount administered to a patient will likely depend on such variables as the overall health status of the patient, the relative biological efficacy of the compound delivered, the formulation of the drug, the presence and types of excipients in the formulation, the route of administration, and the infection to be treated, prevented, or reducing the risk of Also, it is to be understood that the initial dosage administered can be increased beyond the above upper level in order to rapidly achieve the desired blood-level or tissue level, or the initial dosage can be smaller than the optimum.
  • the dose of active compound comprises from about 0.1 to about 1500 mg of the compound per dose.
  • the dose of active compound is selected from about 25 mg, about 50 mg, about 75 mg, about 100 mg, about 125 mg, about 150 mg, about 175 mg, about 200 mg, about 225 mg, about 250 mg, about 275 mg, about 300 mg, about 325, about 350 mg, about 375 mg, about 400 mg, about 425 mg, about 450 mg, about 475 mg, about 500 mg, about 525 mg, about 550 mg, about 575 mg, about 600 mg, about 625 mg, about 650 mg, about 675 mg about 700 mg, about 725 mg, about 750 mg, about 775 mg, about 800 mg, about 825 mg, about 850 mg, about 875 mg, about 900 mg, about 925 mg, about 950 mg, about 975 mg, about 1000 mg, about 1025 mg, about 1050, mg, about 1075 mg, about 1100 mg, about 1125 mg, about 1150 mg, about 1175
  • the dosage is given on the basis of the parent or active moiety. Therefore, if a salt, hydrate, or another form of the parent or active moiety is used, a corresponding adjustment in the weight of the compound is made, although the dose is still referred to on the basis of the parent or active moiety delivered.
  • the parent or active moiety of interest is a monocarboxylic acid having a molecular weight of 250, and if the monosodium salt of the acid is desired to be delivered to be delivered at the same dosage, then an adjustment is made recognizing that the monosodium salt would have a molecular weight of approximately 272 (i.e.
  • a 250 mg dosage of the parent or active compound would correspond to about 272 mg of the monosodium salt, which would also deliver 250 mg of the parent or active compound. Said another way, about 272 mg of the monosodium salt would be equivalent to a 250 mg dosage of the parent or active compound.
  • Ingredients are identified by chemical, USP, or CTFA name.
  • formulations are useful for intravenous administration, either infusion or bolus, such as injection, to a patient for treating, preventing, or reducing the risk of a microbial infection, e.g., a skin infection, including uncomplicated skin infections, skin and soft tissue infections, complicated skin infections, pneumonia, including e.g., community acquired pneumonia, nosocomial (hospital acquired) pneumonia, hospital acquired community pneumonia, post-viral pneumonia, an abdominal infection, a urinary tract infection, bacteremia, septicemia, endocarditis, an atrio-ventricular shunt infection, a vascular access infection, meningitis, infection due to surgical or invasive medical procedures, a peritoneal infection, a bone infection, a joint infection, a methicillin-resistant Staphylococcus aureus infection, a vancomycin-resistant Enterococci infection, a linezolid-resistant organism infection, and tuberculosis. More specifically, this formulation is useful for reducing the risk of or preventing infection
  • Captisol beta-Cyclodextrin sulfobutyl ether sodium
  • composition is useful for intravenous administration to a patient for treating, preventing, or reducing the risk of a microbial infection.
  • Captisol beta-Cyclodextrin sulfobutyl ether sodium
  • the amount of EDTA solution add is increased to 0.15 mg/mL.
  • composition is useful for intravenous administration to a patient for treating, preventing, or reducing the risk of a microbial infection.
  • Formulations can also be prepared as lyophilisates.
  • the formulations of Examples 1 and 2, above can also be prepared as lyophiles. This is accomplished by sterile filtering the solutions into lyophile vials, and then freeze drying the vials using conventional freeze drying techniques.
  • Such formulations are reconstituted with water or another appropriate aqueous based solution. These lyophilisates are a compact and convenient form to store the formulation.
  • formulations can be made by replacing the microcrystalline cellulose with other fillers such as mannitol, lactose, xylitol, or other materials that can be used as fillers at an approximately equivalent or different weight.
  • the manufacturing process involves, but is not limited to as described immediately below.
  • the vitamin E TPGS was melted with mixing in a 55° C. oven to form a uniform melt and the appropriate amount was then weighed into a granulation beaker and held at 40-55° C.
  • the delafloxacin and the intragranular excipient(s), such as for example, arginine, PVP, microcrystalline cellulose, were weighed into a mixing vessel and mixed using a mechanical mixer.
  • This mixture was then mixed with the melted vitamin E TPGS and granulated to form a uniform granulation. If necessary, water can be added to help the granulation.
  • the resulting mixture was then dried at 30-35° C. until dried.
  • the material was then passed through a screen and collected in a mixing vessel.
  • This material was mixed with the extragranular excipients (such as magnesium stearate) in a mechanical mixer.
  • the resulting material was compressed into individual tablets, or alternatively loaded into standard gelatin capsules.
  • the resulting formulations are useful for oral administration, for treating, preventing, or reducing the risk of a microbial infection in a patient.
  • formulations can be made by replacing the microcrystalline cellulose with other fillers such as mannitol, lactose, xylitol, or other materials that can be used as fillers at an approximately equivalent or different weight.
  • the resulting formulation can be manufactured using the same or similar process described above in Example 4.
  • the resulting formulation is useful for oral administration, for treating, preventing, or reducing the risk of a microbial infection in a patient.
  • formulations can be made by replacing the microcrystalline cellulose with other fillers such as mannitol, lactose, xylitol, or other materials that can be used as fillers at an approximately equivalent or different weight.
  • the resulting formulation can be manufactured using the same or similar process described above in Example 1.
  • the resulting formulation is useful for oral administration, for treating, preventing, or reducing the risk of a microbial infection in a patient.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Dermatology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Inorganic Chemistry (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US17/406,805 2013-03-15 2021-08-19 Methods of treating infections in overweight and obese patients using antibiotics Abandoned US20220257584A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/406,805 US20220257584A1 (en) 2013-03-15 2021-08-19 Methods of treating infections in overweight and obese patients using antibiotics
US18/324,754 US20240165100A1 (en) 2013-03-15 2023-05-26 Methods of treating infections in overweight and obese patients using antibiotics

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361790179P 2013-03-15 2013-03-15
PCT/US2014/027220 WO2014152332A1 (en) 2013-03-15 2014-03-14 Methods of treating infections in overweight and obese patients using antibiotics
US201514775465A 2015-09-11 2015-09-11
US17/406,805 US20220257584A1 (en) 2013-03-15 2021-08-19 Methods of treating infections in overweight and obese patients using antibiotics

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2014/027220 Continuation WO2014152332A1 (en) 2013-03-15 2014-03-14 Methods of treating infections in overweight and obese patients using antibiotics
US14/775,465 Continuation US12036219B2 (en) 2014-03-14 Methods of treating infections in overweight and obese patients using antibiotics

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/324,754 Continuation US20240165100A1 (en) 2013-03-15 2023-05-26 Methods of treating infections in overweight and obese patients using antibiotics

Publications (1)

Publication Number Publication Date
US20220257584A1 true US20220257584A1 (en) 2022-08-18

Family

ID=51581190

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/406,805 Abandoned US20220257584A1 (en) 2013-03-15 2021-08-19 Methods of treating infections in overweight and obese patients using antibiotics
US18/324,754 Pending US20240165100A1 (en) 2013-03-15 2023-05-26 Methods of treating infections in overweight and obese patients using antibiotics

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/324,754 Pending US20240165100A1 (en) 2013-03-15 2023-05-26 Methods of treating infections in overweight and obese patients using antibiotics

Country Status (14)

Country Link
US (2) US20220257584A1 (zh)
EP (1) EP2969004A4 (zh)
JP (2) JP6617097B2 (zh)
KR (1) KR102237887B1 (zh)
AR (1) AR095594A1 (zh)
AU (2) AU2014239965A1 (zh)
BR (1) BR112015023044A8 (zh)
CA (1) CA2904387C (zh)
CL (1) CL2015002759A1 (zh)
HK (1) HK1219690A1 (zh)
IL (1) IL241113A0 (zh)
MX (1) MX2015011760A (zh)
TW (1) TWI641372B (zh)
WO (1) WO2014152332A1 (zh)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW201605447A (zh) * 2014-06-20 2016-02-16 美林塔治療學有限公司 處理感染的方法
CN107216353B (zh) * 2016-03-21 2020-05-19 山东诚创医药技术开发有限公司 一种头孢洛林酯咪唑盐的精制方法
US10624899B2 (en) 2016-07-14 2020-04-21 Achaogen, Inc. Combination products for the treatment of bacterial infections and methods of producing or dosing of same

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2373166A4 (en) * 2008-11-15 2014-11-05 Rib X Pharmaceuticals Inc ANTIMICROBIAL COMPOSITIONS

Also Published As

Publication number Publication date
WO2014152332A1 (en) 2014-09-25
HK1219690A1 (zh) 2017-04-13
BR112015023044A8 (pt) 2021-09-28
EP2969004A4 (en) 2016-09-21
EP2969004A1 (en) 2016-01-20
CA2904387A1 (en) 2014-09-25
TW201513862A (zh) 2015-04-16
CA2904387C (en) 2021-12-07
MX2015011760A (es) 2016-06-10
AU2014239965A1 (en) 2015-09-24
JP2019196401A (ja) 2019-11-14
BR112015023044A2 (pt) 2017-07-18
TWI641372B (zh) 2018-11-21
KR102237887B1 (ko) 2021-04-07
US20240165100A1 (en) 2024-05-23
AU2019200483A1 (en) 2019-02-14
IL241113A0 (en) 2015-11-30
JP2016513689A (ja) 2016-05-16
CL2015002759A1 (es) 2016-04-22
US20160058750A1 (en) 2016-03-03
KR20150133762A (ko) 2015-11-30
JP6617097B2 (ja) 2019-12-04
AR095594A1 (es) 2015-10-28

Similar Documents

Publication Publication Date Title
US20220257584A1 (en) Methods of treating infections in overweight and obese patients using antibiotics
EP2900244B1 (en) Tazobactam arginine antibiotic compositions
WO2008020904A2 (en) Method for reducing the risk of or preventing infection due to surgical or invasive medical procedures
KR20110091504A (ko) 퀴놀론 화합물의 제조 방법
US9409896B2 (en) Sustained release pharmaceutical compositions comprising an antibacterial agent
US20180029997A1 (en) Quinazolinone antibiotics
US12036219B2 (en) Methods of treating infections in overweight and obese patients using antibiotics
AU2021286284B2 (en) Antimicrobial compositions with effervescent agents
US20070238720A1 (en) Method for reducing the risk of or preventing infection due to surgical or invasive medical procedures
US20070238719A1 (en) Method for reducing the risk of or preventing infection due to surgical or invasive medical procedures
US8247400B2 (en) Cephem compounds useful for the treatment of bacterial infections

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: SILICON VALLEY BANK, CALIFORNIA

Free format text: AMENDED AND RESTATED INTELLECTUAL PROPERTY SECURITY AGREEMENT;ASSIGNOR:MELINTA SUBSIDIARY CORP.;REEL/FRAME:061314/0572

Effective date: 20220823

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION