US20140274996A1 - Tazobactam and ceftolozane antibiotic compositions - Google Patents

Tazobactam and ceftolozane antibiotic compositions Download PDF

Info

Publication number
US20140274996A1
US20140274996A1 US14/214,367 US201414214367A US2014274996A1 US 20140274996 A1 US20140274996 A1 US 20140274996A1 US 201414214367 A US201414214367 A US 201414214367A US 2014274996 A1 US2014274996 A1 US 2014274996A1
Authority
US
United States
Prior art keywords
ceftolozane
sodium chloride
tazobactam
cxa
day
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
US14/214,367
Inventor
Nicole Miller Damour
Joseph Terracciano
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.)
Merck Sharp and Dohme LLC
Original Assignee
Calixa Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Calixa Therapeutics Inc filed Critical Calixa Therapeutics Inc
Priority to US14/214,367 priority Critical patent/US20140274996A1/en
Assigned to CUBIST PHARMACEUTICALS, INC. reassignment CUBIST PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DAMOUR, NICOLE MILLER, TERRACCIANO, JOSEPH
Publication of US20140274996A1 publication Critical patent/US20140274996A1/en
Priority to US14/522,893 priority patent/US20150045336A1/en
Assigned to CALIXA THERAPEUTICS, INC. reassignment CALIXA THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CUBIST PHARMACEUTICALS, INC.
Assigned to MERCK SHARP & DOHME CORP. reassignment MERCK SHARP & DOHME CORP. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CALIXA THERAPEUTICS, INC.
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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/542Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/545Compounds containing 5-thia-1-azabicyclo [4.2.0] octane ring systems, i.e. compounds containing a ring system of the formula:, e.g. cephalosporins, cefaclor, or cephalexine
    • A61K31/546Compounds containing 5-thia-1-azabicyclo [4.2.0] octane ring systems, i.e. compounds containing a ring system of the formula:, e.g. cephalosporins, cefaclor, or cephalexine containing further heterocyclic rings, e.g. cephalothin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/429Thiazoles condensed with heterocyclic ring systems
    • A61K31/43Compounds containing 4-thia-1-azabicyclo [3.2.0] heptane ring systems, i.e. compounds containing a ring system of the formula, e.g. penicillins, penems
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/429Thiazoles condensed with heterocyclic ring systems
    • A61K31/43Compounds containing 4-thia-1-azabicyclo [3.2.0] heptane ring systems, i.e. compounds containing a ring system of the formula, e.g. penicillins, penems
    • A61K31/431Compounds containing 4-thia-1-azabicyclo [3.2.0] heptane ring systems, i.e. compounds containing a ring system of the formula, e.g. penicillins, penems containing further heterocyclic rings, e.g. ticarcillin, azlocillin, oxacillin
    • 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
    • A61K47/02Inorganic compounds
    • 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
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/542Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/545Compounds containing 5-thia-1-azabicyclo [4.2.0] octane ring systems, i.e. compounds containing a ring system of the formula:, e.g. cephalosporins, cefaclor, or cephalexine

Definitions

  • compositions comprising ceftolozane
  • pharmaceutical compositions comprising tazobactam and ceftolozane
  • methods of making those compositions and related methods and uses thereof.
  • cephalosporin (6R,7R)-3-[5-amino-4- ⁇ [(2-aminoethyl)carbamoyl]amino ⁇ -1-methyl-1H-pyrazol-2-ium-2-yl)methyl]-7-( ⁇ (2Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-[(1-carboxy-1-methylethoxy)imino]acetyl ⁇ amino)-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylate (also referred to as ceftolozane, “CXA-101” or (6R,7R)-3-[5-Amino-4-[3-(2-aminoethyl)ureido]-1-methyl-1H-pyrazol-2-ium-2-ylmethyl]-7-[2-(5-amino-1,2,4-thiadiazol-3-yl)-2-[(
  • ceftolozane The antibacterial activity of ceftolozane is believed to result from its interaction with penicillin binding proteins (PBPs) to inhibit the biosynthesis of the bacterial cell wall which acts to stop bacterial replication.
  • PBPs penicillin binding proteins
  • Ceftolozane can be combined (e.g., mixed) with a ⁇ -lactamase inhibitor (“BLI”), such as tazobactam.
  • BLI ⁇ -lactamase inhibitor
  • Tazobactam is a BLI against Class A and some Class C ⁇ -lactamases, with well-established in vitro and in vivo efficacy in combination with active ⁇ -lactam antibiotics.
  • Antibiotic pharmaceutical compositions can include a beta-lactam compound having antibiotic properties (i.e., an antibiotic compound possessing one or more beta-lactam moieties) and a BLI, such as tazobactam.
  • Beta-lactam compounds can be formulated with and/or administered in combination with, beta-lactamase inhibiting compounds (e.g., tazobactam and salts thereof) in order to mitigate the effects of bacterial beta-lactamases.
  • beta-lactamase inhibiting compounds e.g., tazobactam and salts thereof
  • the combination of ceftolozane, and tazobactam in a 2:1 weight ratio is an antibiotic pharmaceutical composition (“CXA-201”) formulated for parenteral administration.
  • CXA-201 displays potent antibacterial activity in vitro against common Gram-negative and selected Gram-positive organisms.
  • CXA-201 is a combination antibacterial with activity against many Gram-negative pathogens known to cause intrapulmonary infections, including nosocomial pneumonia caused by P. aeruginosa.
  • compositions comprising ceftolozane, methods of making the compositions, and pharmaceutical compositions prepared using ceftolozane.
  • pharmaceutical compositions comprising ceftolozane and tazobactam, methods of making those compositions, and pharmaceutical compositions prepared using ceftolozane and tazobactam. Methods of making and related uses of these combinations are also provided.
  • composition comprising sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000 to 500:1000.
  • the sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate are at a weight ratio of 476:1000.
  • the sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate are at a weight ratio of 480:1000.
  • the sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate are at a weight ratio of 481:1000 to 500:1000.
  • the sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate are at a weight ratio of 481:1000.
  • the sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate are at a weight ratio of 485:1000.
  • the pharmaceutical compositions comprise less than 4% by weight of water.
  • a pharmaceutical composition comprising sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a molar ratio of 8.14:1 to 8.56:1.
  • a pharmaceutical composition comprising sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a molar ratio of 8.23:1 to 8.56:1.
  • a vial containing an antibiotic composition for treating an infection wherein the antibiotic composition comprises sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000 to 500:1000.
  • the antibiotic composition comprises sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000.
  • the antibiotic composition comprises sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 480:1000.
  • the antibiotic composition comprises sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 481:1000 to 500:1000.
  • the antibiotic composition comprises sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 481:1000.
  • the antibiotic composition comprises sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 485:1000.
  • the antibiotic compositions comprise less than 4% by weight of water.
  • the infections are caused by bacteria selected from the group consisting of: Staphylococcus aureus, Escherichia coli, Acinetobacter baumanii, Haemophilus influenzae, Klebsiella pneumoniae , and Pseudomonas aeruginosa.
  • the infections are selected from the group consisting of nosocomial pneumonia, complicated intra-abdominal infections and complicated urinary tract infections.
  • a method of preparing a composition comprising sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate, wherein the method comprises combining sodium chloride with 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000 to 500:
  • the method comprises combining sodium chloride with 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 481:1000 to 500:1000 to form a mixture, followed by lyophilization of the mixture.
  • a method of preparing a composition comprising sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate, wherein the method comprises combining sodium chloride with 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a molar ratio of 8.14:1
  • the method comprises combining sodium chloride with 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a molar ratio of 8.23:1 to 8.56:1 to form a mixture, followed by lyophilization of the mixture.
  • composition formed by any one of the methods provided herein.
  • provided herein is a method for the treatment of a bacterial infection in a mammal, wherein the method comprises administering to said mammal a therapeutically effective amount of a pharmaceutical composition provided herein.
  • the bacterial infection is caused by bacteria selected from the group consisting of: Staphylococcus aureus, Escherichia coli, Acinetobacter baumanii, Haemophilus influenzae, Klebsiella pneumonia , and Pseudomonas aeruginosa.
  • the bacterial infection is selected from the group consisting of nosocomial pneumonia, complicated intra-abdominal infections and complicated urinary tract infections.
  • FIG. 1 is a graph showing the lyophilization program used in the manufacturing of the monoproduct for injection.
  • FIG. 2 is a flowchart showing the manufacturing process for a CXA-101 composition for injection.
  • FIG. 3 is a flowchart showing the manufacturing steps for a pharmaceutical composition comprising ceftolozane and sodium chloride.
  • FIG. 4 is a flowchart showing the manufacturing process for preparing a CXA-201 composition comprising ceftolozane (referred to as CXA-101), tazobactam, and sodium chloride.
  • FIG. 5 is a reference HPLC chromatogram showing the peaks of ceftolozane (also referred to as CXA-101) and related impurities.
  • FIG. 6 is a plot of the data points from Table 5, showing the purity of the ceftolozane in CXA-101 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride.
  • the amount of the sodium chloride in the CXA-101 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (* marks), 190.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (X marks).
  • FIG. 7 is a plot of the data points from Table 6, showing the peak area of the impurity peak 1 in CXA-101 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride.
  • the amount of the sodium chloride in the CXA-101 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (* marks), 190.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (X marks).
  • FIG. 8 is a plot of the data points from Table 7, showing the total peak area of the impurity with a RRT of 0.43 and the impurity peak 3 in CXA-101 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride.
  • the amount of the sodium chloride in the CXA-101 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (* marks), 190.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (X marks).
  • FIG. 9 is a plot of the data points from Table 8, showing the peak area of the impurity peak 7 in CXA-101 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride.
  • the amount of the sodium chloride in the CXA-101 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (* marks), 190.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (X marks).
  • FIG. 10 is a plot of the data points from Table 10, showing the purity of ceftolozane in CXA-201 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-201 compositions comprise ceftolozane, tazobactam, and sodium chloride.
  • the amount of the sodium chloride in the CXA-201 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (X marks), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles).
  • FIG. 11 is a plot of the data points from Table 11, showing the peak area of the impurity peak 1 in CXA-201 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-201 compositions comprise ceftolozane, tazobactam, and sodium chloride.
  • the amount of the sodium chloride in the CXA-201 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (X marks), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles).
  • FIG. 12 is a plot of the data points from Table 12, showing the total peak area of the impurity with a RRT of 0.43 and the impurity peak 3 in CXA-201 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-201 compositions comprise ceftolozane, tazobactam, and sodium chloride.
  • the amount of the sodium chloride in the CXA-201 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (X marks), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles).
  • FIG. 13 is a plot of the data points from Table 13, showing the peak area of the impurity peak 7 in CXA-201 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-201 compositions comprise ceftolozane, tazobactam, and sodium chloride.
  • the amount of the sodium chloride in the CXA-201 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (X marks), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles).
  • FIG. 14 is a manufacturing flowchart showing CXA-201 development using a co-lyophilization process, as described herein.
  • FIG. 15 is the formula of the impurity RRT 1.22, which has been identified to be a degradation product formed by a reaction between ceftolozane and formylacetic acid, a degradation product of tazobactam as illustrated in Marunaka et al. (Chem. Pharm. Bull. 1988, Vol. 36 (11), pp. 4478-4487).
  • compositions comprising one or more drug substances or excipients can be prepared in a variety of ways, including, for example, blending and lyophilization (also known as “co-lyophilization”).
  • lyophilization is a process of freeze-drying in which water is sublimed from a frozen solution of one or more solutes. Specific methods of lyophilization are described in Remington's Pharmaceutical Sciences, Chapter 84, page 1565, Eighteenth Edition, A. R. Gennaro, (Mack Publishing Co., Easton, Pa., 1990).
  • the formulation of pharmaceutical compositions can be selected to minimize decomposition of the constituent drug substances and to produce a composition that is stable under a variety of storage conditions.
  • compositions comprising ceftolozane and 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane have been observed to exhibit better chemical stability over the course of time and/or in the presence of heat, and less impurities than those pharmaceutical compositions comprising ceftolozane and less sodium chloride (i.e., less than 125 mg sodium chloride per 1000 mg of ceftolozane).
  • the pharmaceutical compositions comprising ceftolozane and 125 to 500 mg sodium chloride per 1000 mg of ceftolozane have been found to be more stable than the compositions comprising ceftolozane and less than 125 mg sodium chloride per 1000 mg of ceftolozane.
  • compositions comprising ceftolozane, tazobactam, and 125 to 1000 mg sodium chloride per gram of ceftolozane exhibit better chemical stability and less impurities than those pharmaceutical compositions comprising ceftolozane and tazobactam, but less sodium chloride.
  • the pharmaceutical compositions comprising ceftolozane, tazobactam, and 125 to 500 mg sodium chloride per 1000 mg of ceftolozane have been found to be more stable than the compositions comprising ceftolozane, tazobactam, and less than 125 mg sodium chloride per gram of ceftolozane.
  • a pharmaceutical composition comprising ceftolozane and 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane.
  • the invention is a pharmaceutical composition comprising ceftolozane and tazobactam, further comprising 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane.
  • the pharmaceutical composition comprises 125 to 500 mg sodium chloride per 1000 mg of ceftolozane.
  • 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane refers to a ratio of sodium chloride to ceftolozane.
  • “125 to 1000 mg sodium chloride per 1000 mg of ceftolozane” includes, for example, 62.5 to 500 mg sodium chloride per 500 mg of ceftolozane, as well as, for example, 25 to 200 mg sodium chloride per 200 mg ceftolozane, etc.
  • compositions comprising ceftolozane and tazobactam prepared by blending these two compounds, wherein the ceftolozane and tazobactam are individually lyophilized prior to blending, have been observed to exhibit beneficial properties, including reduced levels of impurities.
  • the pharmaceutical composition prepared by blending ceftolozane and tazobactam, wherein the ceftolozane and tazobactam were individually lyophilized prior to blending lead to a much lower concentration of the following degradation product:
  • This degradation product having a Relative Retention Time (RRT) of 1.22 (relative to ceftolozane using the HPLC method described in Example 4), which is also referred to as “the Impurity RRT 1.22”, has been identified to be a degradation product formed by a reaction between ceftolozane and formylacetic acid, a degradation product of tazobactam as illustrated in Marunaka et al. (Chem. Pharm. Bull. 1988, Vol. 36 (11), pp. 4478-4487).
  • RRT Relative Retention Time
  • compositions of ceftolozane and tazobactam wherein the compositions were formed through co-lyophilization, i.e., the ceftolozane and tazobactam were combined and lyophilized together, as opposed to being individually lyophilized (see, e.g., Example 7).
  • composition comprising ceftolozane and tazobactam, wherein the composition has less than 0.1% by weight of the following compound:
  • the pharmaceutical composition has less than 0.05% by weight of the following compound:
  • composition includes preparations suitable for administration to mammals, e.g., humans.
  • the compounds of the present invention are administered as pharmaceuticals to mammals, e.g., humans, they can be given per se or as a pharmaceutical composition containing, for example, 0.1% to 99.9% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • compositions described herein can be formulated to have any concentration desired (i.e., any concentration of tazobactam, or a hydrate or solvate thereof, and any concentration of ceftolozane).
  • the composition is formulated such that it comprises at least a therapeutically effective amount of both compounds (i.e., a therapeutically effective amount of the combination of tazobactam, or a hydrate or solvate thereof, and ceftolozane).
  • compositions include those suitable for parenteral (including intravenous) administration, although the most suitable route will depend on the nature and severity of the condition being treated.
  • the compositions may be conveniently presented in unit dosage form, and prepared by any of the methods well known in the art of pharmacy.
  • compositions may additionally comprise excipients, stabilizers, pH adjusting additives (e.g., buffers) and the like.
  • pH adjusting additives e.g., buffers
  • Non-limiting examples of these additives include sodium chloride, citric acid and L-arginine.
  • sodium chloride results in greater stability
  • L-arginine is used to adjust pH and to increase the solubility of ceftolozane
  • citric acid is used to prevent discoloration of the product, due to its ability to chelate metal ions.
  • compositions disclosed herein can be prepared via blending.
  • blending refers to a process comprising physically combining ceftolozane and tazobactam, wherein each of ceftolozane and tazobactam have been individually lyophilized (i.e., lyophilized in the absence of one another) prior to being combined.
  • the pharmaceutical compositions described herein are formulated for parenteral administration. In another particular embodiment, the pharmaceutical compositions described herein are formulated for administration by intravenous injection or infusion.
  • a pharmaceutical composition comprising tazobactam and ceftolozane.
  • compositions prepared according to the following methods.
  • a pharmaceutical composition comprising sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000 to 500:1000, or at a molar ratio of 8.14:1 to 8.56:1.
  • a pharmaceutical composition comprising sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 481:1000 to 500:1000, or at a molar ratio of 8.23:1 to 8.56:1.
  • compositions comprising sodium chloride and 7 ⁇ -[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3- ⁇ 3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio ⁇ methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000, or 480:1000, or 481:1000, or 485:1000.
  • the pharmaceutical compositions further comprise tazobactam sodium at a quantity equivalent of 500 mg of tazobactam free acid in a lyophilized powder form per 1000 mg of ceftolozane (anhydrous, free base equivalent).
  • the pharmaceutical compositions comprise less than 4% by weight of water.
  • the pharmaceutical compositions are reconstituted with sterile saline and/or sterile water for injection.
  • a vial containing one of the pharmaceutical compositions described above for treating an infection is provided herein.
  • the infections are caused by bacteria selected from the group consisting of: Staphylococcus aureus, Escherichia coli, Acinetobacter baumanii, Haemophilus influenzae, Klebsiella pneumoniae , and Pseudomonas aeruginosa .
  • the infections are selected from the group consisting of nosocomial pneumonia, complicated intra-abdominal infections and complicated urinary tract infections.
  • a method of preparing a composition comprising ceftolozane and sodium chloride comprising combining sodium chloride with ceftolozane, wherein 125-1000 mg sodium chloride per 1000 mg of ceftolozane is combined, followed by lyophilization of the sodium chloride ceftolozane mixture.
  • a method of preparing a composition comprising sodium chloride, tazobactam, and ceftolozane comprising combining sodium chloride, tazobactam, and ceftolozane, wherein 125-1000 mg sodium chloride per 1000 mg of ceftolozane is combined, followed by lyophilization of the mixture of sodium chloride, tazobactam, and ceftolozane.
  • 125-500 mg sodium chloride per 1000 mg of ceftolozane is combined.
  • the method further comprises lyophilizing ceftolozane in the absence of tazobactam. In yet another embodiment of the methods described above, the method can further comprise lyophilizing tazobactam in the absence of ceftolozane.
  • the method can comprise the steps of: (1) adding 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane followed by lyophilizing ceftolozane; (2) lyophilizing tazobactam; and (3) combining the separately lyophilized ceftolozane and tazobactam to obtain said pharmaceutical composition.
  • the method comprises adding 125 to 500 mg sodium chloride per 1000 mg of ceftolozane.
  • a method of making a pharmaceutical composition comprising combining tazobactam and ceftolozane.
  • the method further comprises lyophilizing ceftolozane in the absence of tazobactam.
  • the method further comprises lyophilizing tazobactam in the absence of ceftolozane.
  • the method comprises the steps of: (1) lyophilizing ceftolozane; (2) lyophilizing tazobactam; and (3) combining the separately lyophilized ceftolozane and tazobactam to obtain said pharmaceutical composition.
  • the method further comprises packaging the blended powder into Sterbags®.
  • the molar ratio of tazobactam to ceftolozane in the mixture is in the range of 1:3 to 3:1. In another embodiment, the molar ratio of tazobactam to ceftolozane in the mixture is in the range of 1:2 to 2:1. In another embodiment, the molar ratio of tazobactam to ceftolozane in the mixture is in the range of 1:0.9 to 0.9:1. In a particular embodiment, the ratio of tazobactam to ceftolozane in the mixture is about 0.9:1. In another particular embodiment, the ratio of tazobactam to ceftolozane in the mixture is about 1:2.
  • Tazobactam inhibits or decreases the activity of beta-lactamases (e.g., bacterial beta-lactamases), and can be combined with beta-lactam compounds (e.g., antibiotics), thereby broadening the spectrum of the beta-lactam compound and increasing the beta-lactam compound's efficacy against organisms that produce beta-lactamase.
  • beta-lactamases e.g., bacterial beta-lactamases
  • beta-lactam compounds e.g., antibiotics
  • a method for the treatment of bacterial infections in a mammal comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition prepared according to the methods described herein.
  • a method for the treatment of bacterial infections in a mammal comprising administering to said mammal a therapeutically effective amount of tazobactam and ceftolozane.
  • the bacterial infection is caused by an extended-spectrum beta-lactamase-producing organism.
  • the bacterial infection is caused by an antibiotic-resistant organism.
  • a method for the treatment of bacterial infections in a mammal comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising ceftolozane.
  • the invention is a method for the treatment of bacterial infections in a mammal, comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising both tazobactam and ceftolozane.
  • the bacterial infection is caused by an extended-spectrum beta-lactamase-producing organism.
  • the bacterial infection is caused by an antibiotic-resistant organism.
  • the pharmaceutical composition further comprises 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane. In some other embodiments, the pharmaceutical composition comprises 125 to 500 mg sodium chloride per 1000 mg of ceftolozane.
  • a method for the treatment of bacterial infections in a mammal comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising tazobactam, ceftolozane, and less than 0.1% by weight of the following compound:
  • the pharmaceutical composition comprises tazobactam, ceftolozane, and less than 0.05% by weight of the following compound:
  • Non-limiting examples of bacterial infections that can be treated by the methods of the invention include infections caused by: aerobic and facultative gram-positive microorganisms (e.g., Staphylococcus aureus, Enterococcus faecalis, Staphylococcus epidermidis, Streptococcus agalactiae, Streptococcus pneumonia, Streptococcus pyogenes , Viridans group streptococci), aerobic and facultative gram-negative microorganisms (e.g., Acinetobacter baumanii, Escherichia coli, Haemophilus influenza, Klebsiella pneumonia, Pseudomonas aeruginosa, Citrobacter koseri, Moraxella catarrhalis, Morganella morganii, Neisseria gonorrhoeae, Proteus mirabilis, Proteus vulgaris, Serratia marcescens, Providencia stuartii
  • the bacterial infection resulting from beta-lactamase-producing organisms are treated or controlled.
  • beta-lactamase-producing organisms include:
  • ESBL extended-spectrum beta-lactamase-producing organisms selected from the group consisting of Enterobacteriaceae spp.: Escherichia coli, Klebsiella spp. (including K. pneumoniae and K. oxytoca ), Proteus mirabilis, Proteus vulgaris, Enterobacter spp., Serratia spp., Citrobacter spp., Pseudomonas spp., Acinetobacter spp.) and Bacteroides spp.;
  • CSBL conventional-spectrum beta-lactamase
  • Inducible-AmpC-type beta-lactamases such as Citrobacter spp., Serratia spp., Morganella morganii, Proteus vulgaris , and Enterobacter cloacae.
  • bacterial infection is associated with one or more of the following conditions:
  • Nosocomial pneumonia caused by piperacillin-resistant, beta-lactamase producing strains of Staphylococcus aureus and by Acinetobacter baumanii, Haemophilus influenzae, Klebsiella pneumoniae , and Pseudomonas aeruginosa.
  • tazobactam and hydrates and solvates thereof, in combination with ceftolozane, for the preparation of a medicament for the treatment of bacterial infection.
  • the bacterial infection can result from either gram-negative or gram-positive organisms.
  • treating describes the management and care of a patient for the purpose of combating a disease, condition, or disorder and includes the administration of a pharmaceutical composition of the present invention to alleviate the symptoms or complications of a disease, condition or disorder, or to eliminate the disease, condition or disorder.
  • the term “treat” can also include treatment of a cell in vitro or an animal model.
  • a “therapeutically effective amount” of a compound of the invention is meant a sufficient amount of the compound to treat the disorder (e.g., bacterial infection).
  • the specific therapeutically effective amount that is required for the treatment of any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound or composition employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts (see, for example, Goodman and Gilman's, “The Pharmacological Basis of Therapeutics”, Tenth Edition, A.
  • a sterile tilter-set which consists of a 0.2 um polyvinylidene fluoride membrane filter (Durapore®, Millipore) and a 0.1 urn polyvinylidene fluoride membrane filter (Durapore®, Millipore) connected in tandem. Confirm the integrity of each filter before and after the filtration. Take approximately 100 mL of the filtrate in order to check bioburden.
  • CXA-101 bulk drug product manufacturing process There are four main steps in the manufacture of CXA-101 bulk drug product: dissolution, sterile filtration, bulk lyophilization, and packaging into Sterbags®. These four main steps are composed of a total of 20 minor steps.
  • the flowchart of the CXA-101 bulk drug product manufacturing process is presented in FIG. 3 , and described below.
  • Solution pH is verified to be in the target range of 6.5 to 7.0.
  • Solution pH is verified to be in the target range of 6.0 to 7.0. If the pH is out of this range adjust with either L-Arginine or citric acid.
  • the solution is passed through the filter (pore size 0.45 ⁇ m) followed by double filters (pore size 0.22 ⁇ m) onto a shelf on the Criofarma lyophilizer.
  • the washing solution is passed from Step 12 through sterile filtration.
  • the washing solution is loaded onto a separate shelf in the lyophilizer (and later discarded).
  • the solution is lyophilized until dry.
  • the product shelf is cooled to 20° C. ⁇ 5° C.
  • the lyophilized bulk drug product powder is milled.
  • the milled powder is sieved.
  • the sieved powder is blended for 30 minutes.
  • a low energy drum blender that agitates the material by tumbling and also moving the bed up and down is used.
  • a representative process of blending is described as follows.
  • the blender was charged with 23.4 kg of CXA-101 bulk product, and 5.4 kg of tazobactam bulk product. Both the CXA-101 and tazobactam were individually lyophilized beforehand. The material was blended for 180 minutes.
  • In-process tests of content assay for both CXA-101 and tazobactam were performed to assess the homogeneity using the samples of blend materials taken from three places.
  • the RSD for each of CXA-101 and tazobactam content assay was no greater than 2% and the RSD for the ratio of CXA-101/tazobactam was no greater than 2% (See Table 1).
  • the flow chart of the fill and finish process for the final drug product, CXA-101/tazobactam for injection, 1000 mg/500 mg, is presented in FIG. 4 .
  • Glass vials are washed with WFI and depyrogenated in a Class 100 depyrogenation tunnel at a temperature of 320° C.
  • Pre-washed and pre-siliconized stoppers are autoclaved for 40 minutes at 121° C.
  • the bulk drug product is packaged in a Sterbag® system comprised of three bags.
  • the outer bag is cleaned with disinfectant in a Class 10,000 clean room.
  • the bag system is placed in a pass-through UV box where it is subjected to UV radiation (>20 ⁇ W/cm 2 ) for 20 minutes to sterilize the surface of the outer bag.
  • the outer bag is removed and left in the UV box.
  • the middle bag is placed in a Class A laminar airflow (LAF) hood.
  • LAF laminar airflow
  • the sterile middle bag is removed under LAF.
  • the sterile, bottle-shaped inner bag is then placed in a sterile stainless steel carrier and attached to the filling machine.
  • Sterile bulk CXA-101/tazobactam drug product is filled under a nitrogen blanket into 30-mL, Type I clear glass vials.
  • the sterile drug product is gravity-fed into the filling machine under LAF. Vial fill weights are periodically checked throughout the filling operation to ensure proper operation of the filling line. Filling and stoppering operations are performed under Class 100 LAF conditions. Capping and vial washing are done in the Class 10,000 clean room.
  • Sodium perchlorate buffer solution was made by dissolving 14.05 g of sodium perchlorate monohydrate in 1000.0 mL of water followed by adjusting pH to 2.5 with diluted perchloric acid (1 in 20).
  • Sodium acetate buffer solution pH 5.5 (diluent) was made by dissolving 1.36 g of sodium acetate trihydrate in 1000.0 mL of water followed by adjusting to pH 5.5 with diluted acetic acid (1 in 10).
  • Sample solution dissolve 20.0 mg, exactly weighed, of a sample, in 20.0 mL of water (prepare just before injection into HPLC system).
  • System suitability solution (1%) take 1.0 mL of the sample solution (use first sample if more are present) and transfer into a 100.0 mL volumetric flask, dilute with water to volume and mix.
  • Peak 1 Peak 1 (P1) ⁇ 0.1 Peak 2 (P2) ⁇ 0.2 Peak 3 (P3) ⁇ 0.4 Peak 4 (P4) ⁇ 0.6 Peak 5 (P5) ⁇ 0.9 CXA-101 1.0 Peak 6 (P6) ⁇ 1.1 Peak 7 (P7) ⁇ 1.3 Peak 8 (P8) ⁇ 1.4 Peak 9 (P9) ⁇ 1.7 Peaks 10, 11 (P10, 11) ⁇ 2.3
  • a t area of CXA-101 peak in the sample chromatogram
  • ⁇ A i total peak areas of impurities in the sample chromatogram
  • the co-lyophilization process is illustrated in FIG. 14 .
  • the components of a CXA-201 composition prepared by co-lyophilization are described in Table 14 below.
  • the Co-Lyo Combo Drug Product was prepared, as described above in Example 6.
  • the formulation composition of the Combo drug product is shown in Table 15.
  • the impurity was identified as a degradation product, shown in FIG. 15 , which was formed by a reaction between ceftolozane and formylacetic acid, which was a degradation product of tazobactam.
  • the stability data at 25° C. and at 40° C. have shown that the impurity increases over time.
  • the blend drug product was prepared, as described above in Example 3, on lab scale by use of a small blender.
  • the composition of the blend drug product is shown in Table 18.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Inorganic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This disclosure provides pharmaceutical compositions comprising ceftolozane, pharmaceutical compositions comprising ceftolozane and tazobactam, methods of making those compositions, and related methods and uses of these compositions.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Application No. 61/792,092, filed Mar. 15, 2013, and U.S. Provisional Application No. 61/793,007, filed Mar. 15, 2013. The contents of these applications are incorporated hereby by reference in their entireties.
  • TECHNICAL FIELD
  • This disclosure relates to pharmaceutical compositions comprising ceftolozane, pharmaceutical compositions comprising tazobactam and ceftolozane, methods of making those compositions, and related methods and uses thereof.
  • BACKGROUND
  • The cephalosporin (6R,7R)-3-[5-amino-4-{[(2-aminoethyl)carbamoyl]amino}-1-methyl-1H-pyrazol-2-ium-2-yl)methyl]-7-({(2Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-[(1-carboxy-1-methylethoxy)imino]acetyl}amino)-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylate (also referred to as ceftolozane, “CXA-101” or (6R,7R)-3-[5-Amino-4-[3-(2-aminoethyl)ureido]-1-methyl-1H-pyrazol-2-ium-2-ylmethyl]-7-[2-(5-amino-1,2,4-thiadiazol-3-yl)-2-[(Z)-1-carboxy-1-methylethoxyimino]acetamido]-3-cephem-4-carboxylic acid) is an antibacterial agent. The antibacterial activity of ceftolozane is believed to result from its interaction with penicillin binding proteins (PBPs) to inhibit the biosynthesis of the bacterial cell wall which acts to stop bacterial replication. Ceftolozane can be combined (e.g., mixed) with a β-lactamase inhibitor (“BLI”), such as tazobactam. Tazobactam is a BLI against Class A and some Class C β-lactamases, with well-established in vitro and in vivo efficacy in combination with active β-lactam antibiotics.
  • Antibiotic pharmaceutical compositions can include a beta-lactam compound having antibiotic properties (i.e., an antibiotic compound possessing one or more beta-lactam moieties) and a BLI, such as tazobactam. Beta-lactam compounds can be formulated with and/or administered in combination with, beta-lactamase inhibiting compounds (e.g., tazobactam and salts thereof) in order to mitigate the effects of bacterial beta-lactamases. For example, the combination of ceftolozane, and tazobactam in a 2:1 weight ratio is an antibiotic pharmaceutical composition (“CXA-201”) formulated for parenteral administration. CXA-201 displays potent antibacterial activity in vitro against common Gram-negative and selected Gram-positive organisms. CXA-201 is a broad-spectrum antibacterial with in vitro activity against Enterobacteriaceae including strains expressing extended spectrum β-lactamases-resistant (MIC90=1 μg/mL), as well as Pseudomonas aeruginosa (P. aeruginosa) including multi-drug resistant strains (MIC90=2 μg/mL). CXA-201 is a combination antibacterial with activity against many Gram-negative pathogens known to cause intrapulmonary infections, including nosocomial pneumonia caused by P. aeruginosa.
  • SUMMARY OF THE INVENTION
  • Provided herein are pharmaceutical compositions comprising ceftolozane, methods of making the compositions, and pharmaceutical compositions prepared using ceftolozane. Also provided herein are pharmaceutical compositions comprising ceftolozane and tazobactam, methods of making those compositions, and pharmaceutical compositions prepared using ceftolozane and tazobactam. Methods of making and related uses of these combinations are also provided.
  • In one aspect, provided herein is pharmaceutical composition comprising sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000 to 500:1000.
  • In one particular embodiment, the sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate are at a weight ratio of 476:1000.
  • In another particular embodiment, the sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate are at a weight ratio of 480:1000.
  • In a further embodiment, the sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate are at a weight ratio of 481:1000 to 500:1000.
  • In one particular embodiment, the sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate are at a weight ratio of 481:1000.
  • In yet another particular embodiment, the sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate are at a weight ratio of 485:1000.
  • In certain embodiments, the pharmaceutical compositions comprise less than 4% by weight of water.
  • In another aspect, provided herein is a pharmaceutical composition comprising sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a molar ratio of 8.14:1 to 8.56:1.
  • In one embodiment, provided herein is a pharmaceutical composition comprising sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a molar ratio of 8.23:1 to 8.56:1.
  • In another aspect, provided herein is a vial containing an antibiotic composition for treating an infection, wherein the antibiotic composition comprises sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000 to 500:1000.
  • In one particular embodiment of the vial, the antibiotic composition comprises sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000.
  • In another particular embodiment of the vial, the antibiotic composition comprises sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 480:1000.
  • In a further embodiment of the vial, the antibiotic composition comprises sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 481:1000 to 500:1000.
  • In one particular embodiment of the vial, the antibiotic composition comprises sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 481:1000.
  • In yet another particular embodiment of the vial, the antibiotic composition comprises sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 485:1000.
  • In certain embodiments of the vials described above, the antibiotic compositions comprise less than 4% by weight of water.
  • In other embodiments of the vials, the infections are caused by bacteria selected from the group consisting of: Staphylococcus aureus, Escherichia coli, Acinetobacter baumanii, Haemophilus influenzae, Klebsiella pneumoniae, and Pseudomonas aeruginosa.
  • In other embodiments of the vials, the infections are selected from the group consisting of nosocomial pneumonia, complicated intra-abdominal infections and complicated urinary tract infections.
  • In still another aspect, provided herein is a method of preparing a composition comprising sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate, wherein the method comprises combining sodium chloride with 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000 to 500:1000 to form a mixture, followed by lyophilization of the mixture.
  • In one embodiment, the method comprises combining sodium chloride with 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 481:1000 to 500:1000 to form a mixture, followed by lyophilization of the mixture.
  • In a further aspect, provided herein is a method of preparing a composition comprising sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate, wherein the method comprises combining sodium chloride with 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a molar ratio of 8.14:1 to 8.56:1 to form a mixture, followed by lyophilization of the mixture.
  • In one embodiment, the method comprises combining sodium chloride with 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a molar ratio of 8.23:1 to 8.56:1 to form a mixture, followed by lyophilization of the mixture.
  • In another aspect, provided herein is a composition formed by any one of the methods provided herein.
  • In yet another aspect, provided herein is a method for the treatment of a bacterial infection in a mammal, wherein the method comprises administering to said mammal a therapeutically effective amount of a pharmaceutical composition provided herein.
  • In one embodiment of the treatment method, the bacterial infection is caused by bacteria selected from the group consisting of: Staphylococcus aureus, Escherichia coli, Acinetobacter baumanii, Haemophilus influenzae, Klebsiella pneumonia, and Pseudomonas aeruginosa.
  • In one embodiment of the treatment method, the bacterial infection is selected from the group consisting of nosocomial pneumonia, complicated intra-abdominal infections and complicated urinary tract infections.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 is a graph showing the lyophilization program used in the manufacturing of the monoproduct for injection.
  • FIG. 2 is a flowchart showing the manufacturing process for a CXA-101 composition for injection.
  • FIG. 3 is a flowchart showing the manufacturing steps for a pharmaceutical composition comprising ceftolozane and sodium chloride.
  • FIG. 4 is a flowchart showing the manufacturing process for preparing a CXA-201 composition comprising ceftolozane (referred to as CXA-101), tazobactam, and sodium chloride.
  • FIG. 5 is a reference HPLC chromatogram showing the peaks of ceftolozane (also referred to as CXA-101) and related impurities.
  • FIG. 6 is a plot of the data points from Table 5, showing the purity of the ceftolozane in CXA-101 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride. The amount of the sodium chloride in the CXA-101 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (* marks), 190.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (X marks).
  • FIG. 7 is a plot of the data points from Table 6, showing the peak area of the impurity peak 1 in CXA-101 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride. The amount of the sodium chloride in the CXA-101 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (* marks), 190.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (X marks).
  • FIG. 8 is a plot of the data points from Table 7, showing the total peak area of the impurity with a RRT of 0.43 and the impurity peak 3 in CXA-101 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride. The amount of the sodium chloride in the CXA-101 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (* marks), 190.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (X marks).
  • FIG. 9 is a plot of the data points from Table 8, showing the peak area of the impurity peak 7 in CXA-101 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride. The amount of the sodium chloride in the CXA-101 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (* marks), 190.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (X marks).
  • FIG. 10 is a plot of the data points from Table 10, showing the purity of ceftolozane in CXA-201 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-201 compositions comprise ceftolozane, tazobactam, and sodium chloride. The amount of the sodium chloride in the CXA-201 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (X marks), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles).
  • FIG. 11 is a plot of the data points from Table 11, showing the peak area of the impurity peak 1 in CXA-201 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-201 compositions comprise ceftolozane, tazobactam, and sodium chloride. The amount of the sodium chloride in the CXA-201 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (X marks), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles).
  • FIG. 12 is a plot of the data points from Table 12, showing the total peak area of the impurity with a RRT of 0.43 and the impurity peak 3 in CXA-201 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-201 compositions comprise ceftolozane, tazobactam, and sodium chloride. The amount of the sodium chloride in the CXA-201 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (X marks), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles).
  • FIG. 13 is a plot of the data points from Table 13, showing the peak area of the impurity peak 7 in CXA-201 compositions at 60° C. on day 0, day 1, day 3, and day 7, as measured by the HPLC method described in Example 4, wherein the CXA-201 compositions comprise ceftolozane, tazobactam, and sodium chloride. The amount of the sodium chloride in the CXA-201 compositions is 481.0 mg sodium chloride per 1000 mg of ceftolozane (X marks), 125.0 mg sodium chloride per 1000 mg of ceftolozane (filled diamonds), 75.0 mg sodium chloride per 1000 mg of ceftolozane (filled squares), and 50.0 mg sodium chloride per 1000 mg of ceftolozane (filled triangles).
  • FIG. 14 is a manufacturing flowchart showing CXA-201 development using a co-lyophilization process, as described herein.
  • FIG. 15 is the formula of the impurity RRT 1.22, which has been identified to be a degradation product formed by a reaction between ceftolozane and formylacetic acid, a degradation product of tazobactam as illustrated in Marunaka et al. (Chem. Pharm. Bull. 1988, Vol. 36 (11), pp. 4478-4487).
  • DETAILED DESCRIPTION OF THE INVENTION
  • Pharmaceutical compositions comprising one or more drug substances or excipients can be prepared in a variety of ways, including, for example, blending and lyophilization (also known as “co-lyophilization”). As is known to those skilled in the art, lyophilization is a process of freeze-drying in which water is sublimed from a frozen solution of one or more solutes. Specific methods of lyophilization are described in Remington's Pharmaceutical Sciences, Chapter 84, page 1565, Eighteenth Edition, A. R. Gennaro, (Mack Publishing Co., Easton, Pa., 1990).
  • The formulation of pharmaceutical compositions can be selected to minimize decomposition of the constituent drug substances and to produce a composition that is stable under a variety of storage conditions.
  • Surprisingly, pharmaceutical compositions comprising ceftolozane and 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane have been observed to exhibit better chemical stability over the course of time and/or in the presence of heat, and less impurities than those pharmaceutical compositions comprising ceftolozane and less sodium chloride (i.e., less than 125 mg sodium chloride per 1000 mg of ceftolozane). In particular embodiments described herein (see, e.g., Example 5), the pharmaceutical compositions comprising ceftolozane and 125 to 500 mg sodium chloride per 1000 mg of ceftolozane have been found to be more stable than the compositions comprising ceftolozane and less than 125 mg sodium chloride per 1000 mg of ceftolozane.
  • It has also been observed that pharmaceutical compositions comprising ceftolozane, tazobactam, and 125 to 1000 mg sodium chloride per gram of ceftolozane exhibit better chemical stability and less impurities than those pharmaceutical compositions comprising ceftolozane and tazobactam, but less sodium chloride. In particular embodiments described herein (see, e.g., Example 5), the pharmaceutical compositions comprising ceftolozane, tazobactam, and 125 to 500 mg sodium chloride per 1000 mg of ceftolozane have been found to be more stable than the compositions comprising ceftolozane, tazobactam, and less than 125 mg sodium chloride per gram of ceftolozane. Accordingly, in one aspect, provided herein is a pharmaceutical composition comprising ceftolozane and 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane. In another aspect, the invention is a pharmaceutical composition comprising ceftolozane and tazobactam, further comprising 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane. In certain embodiments of both of these aspects, the pharmaceutical composition comprises 125 to 500 mg sodium chloride per 1000 mg of ceftolozane.
  • As used herein, “125 to 1000 mg sodium chloride per 1000 mg of ceftolozane” refers to a ratio of sodium chloride to ceftolozane. For example, “125 to 1000 mg sodium chloride per 1000 mg of ceftolozane” includes, for example, 62.5 to 500 mg sodium chloride per 500 mg of ceftolozane, as well as, for example, 25 to 200 mg sodium chloride per 200 mg ceftolozane, etc.
  • In addition, surprisingly, pharmaceutical compositions comprising ceftolozane and tazobactam prepared by blending these two compounds, wherein the ceftolozane and tazobactam are individually lyophilized prior to blending, have been observed to exhibit beneficial properties, including reduced levels of impurities. In a particular embodiment described herein (see, e.g., Example 3), the pharmaceutical composition prepared by blending ceftolozane and tazobactam, wherein the ceftolozane and tazobactam were individually lyophilized prior to blending, lead to a much lower concentration of the following degradation product:
  • Figure US20140274996A1-20140918-C00001
  • This degradation product, having a Relative Retention Time (RRT) of 1.22 (relative to ceftolozane using the HPLC method described in Example 4), which is also referred to as “the Impurity RRT 1.22”, has been identified to be a degradation product formed by a reaction between ceftolozane and formylacetic acid, a degradation product of tazobactam as illustrated in Marunaka et al. (Chem. Pharm. Bull. 1988, Vol. 36 (11), pp. 4478-4487).
  • In contrast, a much greater amount of this impurity was found in compositions of ceftolozane and tazobactam, wherein the compositions were formed through co-lyophilization, i.e., the ceftolozane and tazobactam were combined and lyophilized together, as opposed to being individually lyophilized (see, e.g., Example 7).
  • Accordingly, in one aspect, provided herein is a pharmaceutical composition comprising ceftolozane and tazobactam, wherein the composition has less than 0.1% by weight of the following compound:
  • Figure US20140274996A1-20140918-C00002
  • In an embodiment, the pharmaceutical composition has less than 0.05% by weight of the following compound:
  • Figure US20140274996A1-20140918-C00003
  • Ceftolozane
  • The compound 5-amino-4-{[(2-aminoethyl)carbamoyl]amino}-2-{[(6R,7R)-7-({(2Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-[(1-carboxy-1-methylethoxy)imino]acetyl}amino)-2-carboxy-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-en-3-yl]methyl}-1-methyl-1H-pyrazolium monosulfate (also known also as ceftolozane sulfate, FR264205, “CXA-101”) is a cephalosporin compound (shown below), the synthesis of which is described in U.S. Pat. No. 7,129,232, wherein the compound is also named 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate. As used herein, the term “ceftolozane” may also refer to “ceftolozane sulfate”.
  • Figure US20140274996A1-20140918-C00004
  • Pharmaceutical Compositions
  • The term “pharmaceutical composition” includes preparations suitable for administration to mammals, e.g., humans. When the compounds of the present invention are administered as pharmaceuticals to mammals, e.g., humans, they can be given per se or as a pharmaceutical composition containing, for example, 0.1% to 99.9% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • The pharmaceutical compositions described herein can be formulated to have any concentration desired (i.e., any concentration of tazobactam, or a hydrate or solvate thereof, and any concentration of ceftolozane). In some embodiments, the composition is formulated such that it comprises at least a therapeutically effective amount of both compounds (i.e., a therapeutically effective amount of the combination of tazobactam, or a hydrate or solvate thereof, and ceftolozane).
  • Pharmaceutical compositions include those suitable for parenteral (including intravenous) administration, although the most suitable route will depend on the nature and severity of the condition being treated. The compositions may be conveniently presented in unit dosage form, and prepared by any of the methods well known in the art of pharmacy.
  • Pharmaceutical compositions may additionally comprise excipients, stabilizers, pH adjusting additives (e.g., buffers) and the like. Non-limiting examples of these additives include sodium chloride, citric acid and L-arginine. For example, the use of sodium chloride results in greater stability; L-arginine is used to adjust pH and to increase the solubility of ceftolozane; and citric acid is used to prevent discoloration of the product, due to its ability to chelate metal ions.
  • The pharmaceutical compositions disclosed herein can be prepared via blending. As used herein, blending refers to a process comprising physically combining ceftolozane and tazobactam, wherein each of ceftolozane and tazobactam have been individually lyophilized (i.e., lyophilized in the absence of one another) prior to being combined.
  • In a particular embodiment, the pharmaceutical compositions described herein are formulated for parenteral administration. In another particular embodiment, the pharmaceutical compositions described herein are formulated for administration by intravenous injection or infusion.
  • In one aspect, provided herein is a pharmaceutical composition comprising tazobactam and ceftolozane.
  • In another aspect, provided herein are pharmaceutical compositions prepared according to the following methods.
  • In one embodiment, provided herein is a pharmaceutical composition comprising sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000 to 500:1000, or at a molar ratio of 8.14:1 to 8.56:1.
  • In another embodiment, provided herein is a pharmaceutical composition comprising sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 481:1000 to 500:1000, or at a molar ratio of 8.23:1 to 8.56:1.
  • In certain particular embodiments, provided herein are pharmaceutical compositions comprising sodium chloride and 7β-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-2-(1-carboxy-1-methylethoxyimino)acetamido]-3-{3-amino-4-[3-(2-aminoethyl)ureido]-2-methyl-1-pyrazolio}methyl-3-cephem-4-carboxylate at a weight ratio of 476:1000, or 480:1000, or 481:1000, or 485:1000.
  • In certain embodiments, the pharmaceutical compositions further comprise tazobactam sodium at a quantity equivalent of 500 mg of tazobactam free acid in a lyophilized powder form per 1000 mg of ceftolozane (anhydrous, free base equivalent).
  • In certain embodiments, the pharmaceutical compositions comprise less than 4% by weight of water.
  • In other embodiments, the pharmaceutical compositions are reconstituted with sterile saline and/or sterile water for injection.
  • In a further embodiment, provided herein is a vial containing one of the pharmaceutical compositions described above for treating an infection. In certain embodiments, the infections are caused by bacteria selected from the group consisting of: Staphylococcus aureus, Escherichia coli, Acinetobacter baumanii, Haemophilus influenzae, Klebsiella pneumoniae, and Pseudomonas aeruginosa. In other embodiments, the infections are selected from the group consisting of nosocomial pneumonia, complicated intra-abdominal infections and complicated urinary tract infections.
  • Methods of Making Pharmaceutical Compositions
  • In one aspect, provided herein is a method of preparing a composition comprising ceftolozane and sodium chloride, comprising combining sodium chloride with ceftolozane, wherein 125-1000 mg sodium chloride per 1000 mg of ceftolozane is combined, followed by lyophilization of the sodium chloride ceftolozane mixture.
  • In another aspect, provided herein is a method of preparing a composition comprising sodium chloride, tazobactam, and ceftolozane, comprising combining sodium chloride, tazobactam, and ceftolozane, wherein 125-1000 mg sodium chloride per 1000 mg of ceftolozane is combined, followed by lyophilization of the mixture of sodium chloride, tazobactam, and ceftolozane.
  • In one embodiment of these methods, 125-500 mg sodium chloride per 1000 mg of ceftolozane is combined.
  • In another embodiment of these methods, the method further comprises lyophilizing ceftolozane in the absence of tazobactam. In yet another embodiment of the methods described above, the method can further comprise lyophilizing tazobactam in the absence of ceftolozane.
  • In a further embodiment of these methods, the method can comprise the steps of: (1) adding 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane followed by lyophilizing ceftolozane; (2) lyophilizing tazobactam; and (3) combining the separately lyophilized ceftolozane and tazobactam to obtain said pharmaceutical composition. In yet a further embodiment, the method comprises adding 125 to 500 mg sodium chloride per 1000 mg of ceftolozane.
  • In another aspect, provided herein is a method of making a pharmaceutical composition, comprising combining tazobactam and ceftolozane. In one embodiment, the method further comprises lyophilizing ceftolozane in the absence of tazobactam. In another embodiment, the method further comprises lyophilizing tazobactam in the absence of ceftolozane.
  • In a further embodiment, the method comprises the steps of: (1) lyophilizing ceftolozane; (2) lyophilizing tazobactam; and (3) combining the separately lyophilized ceftolozane and tazobactam to obtain said pharmaceutical composition. In one embodiment, the method further comprises packaging the blended powder into Sterbags®.
  • In one embodiment of the method, and above embodiments, the molar ratio of tazobactam to ceftolozane in the mixture is in the range of 1:3 to 3:1. In another embodiment, the molar ratio of tazobactam to ceftolozane in the mixture is in the range of 1:2 to 2:1. In another embodiment, the molar ratio of tazobactam to ceftolozane in the mixture is in the range of 1:0.9 to 0.9:1. In a particular embodiment, the ratio of tazobactam to ceftolozane in the mixture is about 0.9:1. In another particular embodiment, the ratio of tazobactam to ceftolozane in the mixture is about 1:2.
  • Methods of Treatment
  • Tazobactam inhibits or decreases the activity of beta-lactamases (e.g., bacterial beta-lactamases), and can be combined with beta-lactam compounds (e.g., antibiotics), thereby broadening the spectrum of the beta-lactam compound and increasing the beta-lactam compound's efficacy against organisms that produce beta-lactamase. A compound or a composition possesses efficacy against an organism if it kills or weakens the organism, or inhibits or prevents reproduction the organism.
  • In one aspect, provided herein is a method for the treatment of bacterial infections in a mammal, comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition prepared according to the methods described herein. In another aspect, provided herein is a method for the treatment of bacterial infections in a mammal, comprising administering to said mammal a therapeutically effective amount of tazobactam and ceftolozane. In certain embodiments of the above methods, the bacterial infection is caused by an extended-spectrum beta-lactamase-producing organism. In certain embodiments, the bacterial infection is caused by an antibiotic-resistant organism.
  • In another aspect, provided herein is a method for the treatment of bacterial infections in a mammal, comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising ceftolozane. In yet another aspect, the invention is a method for the treatment of bacterial infections in a mammal, comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising both tazobactam and ceftolozane. In certain embodiments of the above methods, the bacterial infection is caused by an extended-spectrum beta-lactamase-producing organism. In certain embodiments, the bacterial infection is caused by an antibiotic-resistant organism.
  • In certain embodiments of both aspects, the pharmaceutical composition further comprises 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane. In some other embodiments, the pharmaceutical composition comprises 125 to 500 mg sodium chloride per 1000 mg of ceftolozane.
  • In another aspect, provided herein is a method for the treatment of bacterial infections in a mammal, comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising tazobactam, ceftolozane, and less than 0.1% by weight of the following compound:
  • Figure US20140274996A1-20140918-C00005
  • In one embodiment of the treatment method, the pharmaceutical composition comprises tazobactam, ceftolozane, and less than 0.05% by weight of the following compound:
  • Figure US20140274996A1-20140918-C00006
  • Non-limiting examples of bacterial infections that can be treated by the methods of the invention include infections caused by: aerobic and facultative gram-positive microorganisms (e.g., Staphylococcus aureus, Enterococcus faecalis, Staphylococcus epidermidis, Streptococcus agalactiae, Streptococcus pneumonia, Streptococcus pyogenes, Viridans group streptococci), aerobic and facultative gram-negative microorganisms (e.g., Acinetobacter baumanii, Escherichia coli, Haemophilus influenza, Klebsiella pneumonia, Pseudomonas aeruginosa, Citrobacter koseri, Moraxella catarrhalis, Morganella morganii, Neisseria gonorrhoeae, Proteus mirabilis, Proteus vulgaris, Serratia marcescens, Providencia stuartii, Providencia rettgeri, Salmonella enterica), gram-positive anaerobes (Clostridium perfringens), and gram-negative anaerobes (e.g., Bacteroides fragilis group (e.g., B. fragilis, B. ovatus, B. thetaiotaomicron, and B. vulgates), Bacteroides distasonis, Prevotella melaninogenica).
  • In certain embodiments of the methods described herein, the bacterial infection resulting from beta-lactamase-producing organisms are treated or controlled. Non-limiting examples of beta-lactamase-producing organisms include:
  • (1) ESBL (extended-spectrum beta-lactamase)-producing organisms selected from the group consisting of Enterobacteriaceae spp.: Escherichia coli, Klebsiella spp. (including K. pneumoniae and K. oxytoca), Proteus mirabilis, Proteus vulgaris, Enterobacter spp., Serratia spp., Citrobacter spp., Pseudomonas spp., Acinetobacter spp.) and Bacteroides spp.;
  • (2) CSBL (conventional-spectrum beta-lactamase)-producing organisms, known to those of skill in the art; and
  • (3) Inducible-AmpC-type beta-lactamases, such as Citrobacter spp., Serratia spp., Morganella morganii, Proteus vulgaris, and Enterobacter cloacae.
  • In certain embodiments of the methods described herein, bacterial infection is associated with one or more of the following conditions:
  • Community-acquired pneumonia (moderate severity only) caused by piperacillin-resistant, beta-lactamase producing strains of Haemophilus influenza;
  • Nosocomial pneumonia (moderate to severe) caused by piperacillin-resistant, beta-lactamase producing strains of Staphylococcus aureus and by Acinetobacter baumanii, Haemophilus influenzae, Klebsiella pneumoniae, and Pseudomonas aeruginosa.
  • Complicated intra-abdominal infections; Complicated urinary tract infections (cUTIs); Acute Pyelonephritis; Systemic Inflammatory Response Syndrome (SIRS).
  • Also provided herein is the use of tazobactam, and hydrates and solvates thereof, in combination with ceftolozane, for the preparation of a medicament for the treatment of bacterial infection. The bacterial infection can result from either gram-negative or gram-positive organisms.
  • As used herein, “treating”, “treat” or “treatment” describes the management and care of a patient for the purpose of combating a disease, condition, or disorder and includes the administration of a pharmaceutical composition of the present invention to alleviate the symptoms or complications of a disease, condition or disorder, or to eliminate the disease, condition or disorder. The term “treat” can also include treatment of a cell in vitro or an animal model.
  • By a “therapeutically effective amount” of a compound of the invention is meant a sufficient amount of the compound to treat the disorder (e.g., bacterial infection). The specific therapeutically effective amount that is required for the treatment of any particular patient or organism (e.g., a mammal) will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound or composition employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts (see, for example, Goodman and Gilman's, “The Pharmacological Basis of Therapeutics”, Tenth Edition, A. Gilman, J. Hardman and L. Limbird, eds., McGraw-Hill Press, 155-173, 2001, which is incorporated herein by reference in its entirety). The therapeutically effective amount for a given situation can be readily determined by routine experimentation and is within the skill and judgment of the ordinary clinician.
  • EXAMPLES Example 1 Manufacturing Procedure of Mono Product for Injection 1.1. Preparation of the Compound Solution of CXA-101 Lyophilized Product
  • 1) Weigh 30 kg of water for injection into the compounding vessel;
  • 2) Add 100 g of citric acid, anhydrous and 150 g of sodium bicarbonate into the compounding vessel and dissolve them with mixing;
  • 3) Weigh 5,000 g potency of CXA-101 drug substance and suspend it with mixing. (Note any generation of carbon dioxide.)
  • 4) Slowly add 1,100 g of sodium bicarbonate and dissolve CXA-101 with mixing. (Again, note any generation of carbon dioxide.)
  • 5) Add 1,146 g of sodium chloride and 10,000 g of maltose, dissolve with mixing.
  • 6) Purge dissolved carbon dioxide in the solution with nitrogen until the pH of the solution does not change.
  • 7) Adjust the pH of the solution to 6.0±0.1 with 5%-sodium bicarbonate solution.
  • 8) Adjust the total weight to 56,850 g (D20=1.137) with water for injection.
  • 9) Confirm the pH of the compounded solution within the range 6.0±0.1.
  • 1.2. Prefiltration and Sterile-Filtration
  • 10) Filtrate the compounded solution with a sterile tilter-set which consists of a 0.2 um polyvinylidene fluoride membrane filter (Durapore®, Millipore) and a 0.1 urn polyvinylidene fluoride membrane filter (Durapore®, Millipore) connected in tandem. Confirm the integrity of each filter before and after the filtration. Take approximately 100 mL of the filtrate in order to check bioburden.
  • 11) Filter the prefiltered compounded solution through a sterile filter-set which consists of a 0.2 um polyvinylidene fluoride membrane filter and a 0.1 urn polyvinylidene fluoride membrane filter connected in tandem, and introduce the final filtrate into an aseptic room. Confirm the integrity of each filter before and after the filtration.
  • 1.3. Processing of Vial, Stopper and Flip-off Cap
  • 12) Wash a sufficient quantity of 28 mL vials with water for injection and sterilize the washed vials by a dry-heat sterilizer. Then transfer the sterilized vials into a Grade A area located in an aseptic room.
  • 13) Wash a sufficient quantity of stoppers with water for injection. Sterilize and dry the washed stoppers by steam sterilizer. Then transfer the sterilized stoppers into a Grade A area located in an aseptic room.
  • 14) Sterilize a sufficient quantity of flip-off caps by steam sterilizer. Then transfer the sterilized flip-off caps into a Grade A or B area located in an aseptic room.
  • 1.4. Filling and Partially Stoppering
  • 15) Adjust the fill weight of the filtered compounded solution to 11.37 g (corresponds to 10 mL of the compounded solution), then start filling operation. Check the filled weight in sufficient frequency and confirm it is in target range (11.37 g±1%, 11.26 to 11.43 g). When deviation from the control range (11.37 g±2%, 11.14 to 11.59 g) is occurred, re-adjust the filling weight.
  • 16) Immediately after a vial is filled, partially stopper the vial with a sterilized stopper. Load the filled and partially stoppered vials onto the shelves of a lyophilizer aseptically.
  • 1.5. Lyophilization to Crimping, Visual Inspection, Labeling and Packaging
  • 17) After all filled and partially stoppered vials are loaded into a lyophilizer, start the lyophilization program shown in FIG. 1. Freeze the loaded vials at −40° C. and keep until all vials freeze. Forward the program to primary drying step (shelf temperature; −20° C., chamber pressure; 100 to 150 mTorr). Primary drying time should be determined by monitoring the product temperature. Forward the program to secondary drying step (shelf temperature; 30° C., chamber pressure; not more than 10 mTorr) after completion of the primary drying step. After all vials are dried completely, return the chamber pressure to atmospheric pressure with sterilized nitrogen. Then stopper vials completely.
  • 18) Unload the lyophilized vials from the chamber and crimp with sterilized flip-off caps.
  • 19) Subject all crimped vials to visual inspection and label and package all passed vials.
  • Example 2 Manufacturing Procedure of Bulk (Tray) Lyophilized Ceftolozane
  • There are four main steps in the manufacture of CXA-101 bulk drug product: dissolution, sterile filtration, bulk lyophilization, and packaging into Sterbags®. These four main steps are composed of a total of 20 minor steps. The flowchart of the CXA-101 bulk drug product manufacturing process is presented in FIG. 3, and described below.
  • I. Dissolution
  • 1. The prescribed amount of WFI is charged into the dissolution reactor.
  • 2. A prescribed amount of citric acid is added.
  • 3. The solution is cooled at 5° C. to 10° C.
  • 4. A prescribed amount of CXA-101 drug substance is added to the solution.
  • 5. A prescribed amount of L-arginine is slowly added to the solution.
  • 6. A check for complete dissolution is performed. Solution pH is verified to be in the target range of 6.5 to 7.0.
  • 7. A prescribed amount of sodium chloride is added to the solution.
  • 8. A check for complete dissolution is performed. Solution pH is verified to be in the target range of 6.0 to 7.0. If the pH is out of this range adjust with either L-Arginine or citric acid.
  • 9. WFI is added to bring the net weight to 124.4 kg and the solution is mixed well.
  • 10. Samples are withdrawn for testing of final pH.
  • II. Sterile Filtration
  • 11. The solution is passed through the filter (pore size 0.45 μm) followed by double filters (pore size 0.22 μm) onto a shelf on the Criofarma lyophilizer.
  • 12. The line is washed with WFI.
  • 13. The washing solution is passed from Step 12 through sterile filtration.
  • III. Bulk Lyophilization
  • 14. The washing solution is loaded onto a separate shelf in the lyophilizer (and later discarded).
  • 15. The solution is lyophilized until dry.
  • 16. The product shelf is cooled to 20° C.±5° C.
  • IV. Packaging into Sterbags®
  • 17. The lyophilized bulk drug product powder is milled.
  • 18. The milled powder is sieved.
  • 19. The sieved powder is blended for 30 minutes.
  • 20. The powder is then discharged into Sterbags®
  • Example 3 Manufacturing of Combination Product (Tazobactam and CXA-101) by Blending A. Sterile Dry Blending of Bulk Lyophilized Ceftolozane and Bulk Lyophilized Tazobactam
  • A low energy drum blender that agitates the material by tumbling and also moving the bed up and down is used. A representative process of blending is described as follows. For CXA-101/tazobactam for injection, the blender was charged with 23.4 kg of CXA-101 bulk product, and 5.4 kg of tazobactam bulk product. Both the CXA-101 and tazobactam were individually lyophilized beforehand. The material was blended for 180 minutes. In-process tests of content assay for both CXA-101 and tazobactam were performed to assess the homogeneity using the samples of blend materials taken from three places. The RSD for each of CXA-101 and tazobactam content assay was no greater than 2% and the RSD for the ratio of CXA-101/tazobactam was no greater than 2% (See Table 1).
  • TABLE 1
    In-Process Testing of Blending Samples of
    a CXA-201 Composition at Three Places
    Acceptance Results
    Limits Sam- 60 120 180
    Test (expected value) pling minute minute minute
    Content: 30.4%-37.2% 1 34.24 34.07 34.42
    Ceftolozane 1 2 34.62 34.21 34.66
    3 34.71 34.60 34.85
    Mean 3 34.52 34.30 34.64
    RSD % 0.72 0.80 0.63
    Content: 15.2%-18.6% 1 17.96 18.20 17.12
    Tazobactam 2 2 16.90 18.26 16.51
    3 17.27 16.93 17.02
    Mean 3 17.38 17.80 16.89
    RSD % 3.10 4.22 1.96
    Ratio of 2.004 1 1.91 1.87 2.01
    Content 2 2.05 1.87 2.10
    (w/w) 3 2.01 2.04 2.05
    ceftolozane/ Mean 3 1.99 1.93 2.05
    tazobactam RSD % 3.69 5.12 2.2
    RSD = relative standard deviation
    1Theoretical value: 33.96% Acceptance limits are 90%-110% of the theoretical value.
    2Theoretical value: 16.99% Acceptance limits are 90%-110% of the theoretical value.
    3 Three samples are taken at each time point at three places to measure the percentage by weight of ceftolozane and tazobactam. The “Mean” is the average of the percentages or the weight ratios of Ceftolozane/tazobactam.
    4Acceptance limits were established based on batch history.

    B. Packaging into Sterbags®
    The blended powder is then discharged into Sterbags®.
  • C. Finished Drug Product
  • The flow chart of the fill and finish process for the final drug product, CXA-101/tazobactam for injection, 1000 mg/500 mg, is presented in FIG. 4. Glass vials are washed with WFI and depyrogenated in a Class 100 depyrogenation tunnel at a temperature of 320° C. Pre-washed and pre-siliconized stoppers are autoclaved for 40 minutes at 121° C. The bulk drug product is packaged in a Sterbag® system comprised of three bags. The outer bag is cleaned with disinfectant in a Class 10,000 clean room. The bag system is placed in a pass-through UV box where it is subjected to UV radiation (>20 μW/cm2) for 20 minutes to sterilize the surface of the outer bag. The outer bag is removed and left in the UV box. The middle bag is placed in a Class A laminar airflow (LAF) hood. The sterile middle bag is removed under LAF. The sterile, bottle-shaped inner bag is then placed in a sterile stainless steel carrier and attached to the filling machine.
  • Sterile bulk CXA-101/tazobactam drug product is filled under a nitrogen blanket into 30-mL, Type I clear glass vials. The sterile drug product is gravity-fed into the filling machine under LAF. Vial fill weights are periodically checked throughout the filling operation to ensure proper operation of the filling line. Filling and stoppering operations are performed under Class 100 LAF conditions. Capping and vial washing are done in the Class 10,000 clean room.
  • Example 4 Analytical HPLC Method A. Operative Conditions
  • Column Develosil ODS-UG-5; 5 μm, 250 × 4.6 mm
    (Nomura Chemical, Japan)
    Mobile phase Sodium Perchlorate Buffer Solution
    (PH 2.5)/CH3CN 90:10 (vlv)
    Flow rate 1.0 mL/min
    Wavelength 254 nm
    Injection volume
    10 μL
    Oven Temperature 45° C.
    Run Time 85 minutes
  • Gradient Profile:
  • Time (min) A % B %
    0 75 25
    30 70 30
    60 0 100
    85 0 100
    85.1 75 25
    110 75 25
  • B. Mobile Phase Preparation.
  • Sodium perchlorate buffer solution was made by dissolving 14.05 g of sodium perchlorate monohydrate in 1000.0 mL of water followed by adjusting pH to 2.5 with diluted perchloric acid (1 in 20).
  • Mobile phase was then made by mixing sodium perchlorate buffer solution (pH 2.5) and acetonitrile in the ratio 90:10 (v/v).
  • Sodium acetate buffer solution pH 5.5 (diluent) was made by dissolving 1.36 g of sodium acetate trihydrate in 1000.0 mL of water followed by adjusting to pH 5.5 with diluted acetic acid (1 in 10).
  • C. Sample Preparation
  • Sample solution: dissolve 20.0 mg, exactly weighed, of a sample, in 20.0 mL of water (prepare just before injection into HPLC system).
  • System suitability solution (1%): take 1.0 mL of the sample solution (use first sample if more are present) and transfer into a 100.0 mL volumetric flask, dilute with water to volume and mix.
  • D. HPLC Analysis Procedure
  • 1. Inject blank (water)
    2. Inject system suitability solution and check for tailing factor and theoretical plate number for CAA-101 peak:
      • The tailing factor must not be greater than 1.5
      • Theoretical plates number must not be less than 10000
        3. Inject sample solution
        4. Inject system suitability solution and check for tailing factor and theoretical plate number for CXA-101 peak.
      • The tailing factor must not be greater than 1.5
      • Theoretical plates number must not be less than 10000
        5. Identify the peaks of Related Substances in the Sample chromatogram based on the reference chromatogram reported in FIG. 5 or, alternatively, on the basis of the following RRT values:
  • Impurity RRT
    Peak 1 (P1) ~0.1
    Peak 2 (P2) ~0.2
    Peak 3 (P3) ~0.4
    Peak 4 (P4) ~0.6
    Peak 5 (P5) ~0.9
    CXA-101 1.0
    Peak 6 (P6) ~1.1
    Peak 7 (P7) ~1.3
    Peak 8 (P8) ~1.4
    Peak 9 (P9) ~1.7
    Peaks 10, 11 (P10, 11) ~2.3
  • E. Calculations I. Report for Each Related Substance its Amount as Expressed by Area Percent.
  • C i = A i × 100 A t + A i
  • wherein:
  • Ci=Amount of related substance i in the Sample, area %
  • Ai=Peak area of related substance i in the Sample chromatogram
  • At=Area of CXA-101 peak in the Sample chromatogram
  • At+ΣAi=Total peaks area in the Sample chromatogram
  • Consider as any Unspecified Impurity, each peak in the chromatogram except CXA-101, peaks from 1 to 11 and every peak present in the blank chromatogram and report the largest.
  • II. Report the Total Impurities Content as Expressed by the Following Formula:
  • C T = A i × 100 A t + A i
  • wherein:
  • CT=total impurities content in the Sample, area %
  • At=area of CXA-101 peak in the sample chromatogram
  • ΣAi=total peak areas of impurities in the sample chromatogram
  • Example 5 Stabilizing Effect of Sodium Chloride A. Reduction of the Impurity at RT=63 Minutes
  • A stability study was carried out at 25° C., and samples were analyzed by HPLC. High, mid, and low salt formulations contained 480, 125, and 62.5 mg NaCl per 1000 mg of ceftolozane, respectively. Compositions of blend Drug Product are listed in Table 2. Test results are summarized in Table 3.
  • TABLE 2
    Comparison of the CXA-201 Compositions
    Lot CXA-101 NaCl Tazobactam
    C1
    10% High Na
    C2 20% Mid Na
    C3 20% Low Na
    C4 20% Mid Arginate
    C5 20% Low Arginate
  • TABLE 3
    RT 63′ Peak Area at t = 3 months, 25° C./60% RH storage
    1st data 2nd data 3rd data
    collection collection collection
    Sam- Area Area Area
    ple Summary RT % RT % RT %
    C1 High salt + Tazo Na 63.90 0.03 63.30 0.08 62.49 0.14
    C2 Mid salt + Tazo Na 63.78 0.06 63.12 0.12 62.45 0.28
    C3 Low salt + Tazo Na 63.75 0.12 63.11 0.14 62.46 0.29
    C4 Mid salt + Tazo Arg 63.76 0.10 63.16 0.13 62.44 0.28
    C5 Low salt + Tazo Arg 63.72 0.08 63.14 0.16 62.46 0.33
  • Conclusion: at the three month time point, the reduced salt formulations were observed to be not as stable as the full salt formulation; and trends indicate that reduction in salt causes at least 1.5-fold greater impurity at RT=63 minutes (HPLC).
  • B. CXA-101 Peak Trends with NaCl
  • A stability study was carried out at 30° C. and 60° C., and samples were analyzed by HPLC. Sodium chloride content in test samples is described in Table 4. Stability data are summarized in Tables 5-8. The data are also plotted in FIGS. 6-9 to show trends of total purity, peak 1, RRT 0.43+peak 3, and peak 7 with respect to NaCl.
  • TABLE 4
    Sodium Chloride Content in the CXA-101 Compositions
    Samples NaCl content
    A1 481.0 mg NaCl per 1000 mg of ceftolozane
    A2 190.0 mg NaCl per 1000 mg of ceftolozane
    A3 125.0 mg NaCl per 1000 mg of ceftolozane
    A4 75.0 mg NaCl per 1000 mg of ceftolozane
    A5 50.0 mg NaCl per 1000 mg of ceftolozane
  • TABLE 5
    The Purity of Ceftolozane in CXA-101 Compositions
    with Varying Amounts of Sodium Chloride
    Day A1 A2 A3 A4 A5
    t0/60° C. 0 96.6 98.0 97.9 97.8 97.7
    t0/30° C. 0 98.1 97.8 97.8 97.7
    1 day/60° C. 1 95.9 96.9 96.5 95.7 95.5
    1 day/30° C. 1 98.2 97.7 97.7 97.6
    3 days/60° C. 3 94.9 95.7 94.8 93.9 93.6
    t0-t3) (1.7) (2.3) (3.1) (3.9) (4.1)
    3 day/30° C. 3 98.0 97.5 97.5 97.3
    7 days/60° C. 7 93.6 94.0 94.2 92.3 91.9
    7 day/30° C. 7 97.8 97.2 97.1 97.0
    Total Δ/60° C. 3.07 4.06 3.7 5.48 5.83
    Total Δ/30° C. 0.3 0.6 0.7 0.7
  • TABLE 6
    The HPLC Peak Area of Impurity Peak 1 in CXA-101 Compositions
    with Varying Amounts of Sodium Chloride
    Day A1 A2 A3 A4 A5
    t0/60° C. 0 0.95 0.31 0.3 0.36 0.39
    t0/30° C. 0 0.47 0.36 0.36 0.39
    1 day/60° C. 1 1.36 0.86 0.94 1.36 1.39
    1 day/30° C. 1 0.48 0.40 0.42 0.48
    3 days/60° C. 3 1.71 1.31 1.73 2.06 2.1
    3 day/30° C. 3 0.53 0.50 0.52 0.58
    7 days/60° C. 7 2.26 2.14 2.07 2.86 2.93
    7 day/30° C. 7 0.62 0.63 0.66 0.72
    INCREASE %/60° C. 1.31 1.83 1.77 2.5 2.54
    INCREASE %/30° C. 0.15 0.27 0.30 0.33
  • TABLE 7
    The Total HPLC Peak Area of the Impurity with a
    RRT of 0.43 and Impurity peak 3 in CXA-101 Compositions
    with Varying Amounts of Sodium Chloride
    Day A1 A2 A3 A4 A5
    t0/60° C. 0 0.28 0.10 0.09 0.10 0.11
    t0/30° C. 0 0.15 0.10 0.10 0.11
    1 day/60° C. 1 0.37 0.13 0.16 0.35 0.36
    1 day/30° C. 1 0.13 0.09 0.09 0.10
    3 days/60° C. 3 0.68 0.21 0.31 0.71 0.71
    3 day/30° C. 3 0.17 0.13 0.13 0.14
    7 days/60° C. 7 1.04 0.36 0.30 0.81 0.81
    7 day/30° C. 7 0.19 0.16 0.16 0.17
    INCREASE %/60° C. 0.76 0.26 0.21 0.71 0.7
    INCREASE %/30° C. 0.04 0.06 0.06 0.06
  • TABLE 8
    The HPLC Peak Area of Impurity Peak 7 in CXA-101 Compositions
    with Varying Amounts of Sodium Chloride
    Day A1 A2 A3 A4 A5
    t0/60° C. 0 1.31 0.95 0.96 1.01 1.02
    t0/30° C. 0 0.69 1.00 1.01 1.02
    1 day/60° C. 1 1.37 1.10 1.10 1.23 1.29
    1 day/30° C. 1 0.68 0.99 1.01 1.02
    3 days/60° C. 3 1.43 1.19 1.27 1.41 1.46
    3 day/30° C. 3 0.68 1.03 1.01 1.05
    7 days/60° C. 7 1.49 1.31 1.35 1.55 1.57
    7 day/30° C. 7 0.68 1.01 1.03 1.07
    INCREASE %/60° C. 0.18 0.36 0.39 0.54 0.55
    INCREASE %/30° C. NC 0.01 0.02 0.05
  • Conclusion: the stability test demonstrates that high sodium chloride content enhances stability of Mono product CXA-101.
  • C. CXA-201 Peak Trends with NaCl
  • A stability study was carried out at 30° C. and 60° C., and samples were analyzed by HPLC. Sodium chloride content is described in Table 9. Stability data at 60° C. are summarized in Tables 10-13. The data are also plotted in FIGS. 10-13 to show trends of total purity, peak 1, RRT 0.43+peak 3, and peak 7 with respect to NaCl.
  • TABLE 9
    The Sodium Chloride Content in the CXA-201 Compositions
    Samples NaCl content
    B1 481.0 mg sodium chloride per 1000 mg of ceftolozane
    B2 125.0 mg sodium chloride per 1000 mg of ceftolozane
    B3 75.0 mg sodium chloride per 1000 mg of ceftolozane
    B4 50.0 mg sodium chloride per 1000 mg of ceftolozane
  • TABLE 10
    The Purity of Ceftolozane in CXA-201 Compositions
    with Varying Amounts of Sodium Chloride
    Day B1 B2 B3 B4
    t0
    0 98.1 97.8 97.8 97.7
    1 day/60° C. 1 97.2 96.3 96.2 96.0
    1 day/30° C. 1 98.2 97.7 97.6 97.6
    3 days/60° C. 3 95.4 (2.7) 94.9 (2.9) 94.7 (3.1) 94.6 (3.1)
    t0-t3)
    3 day/30° C. 3 98.0 97.5 97.4 97.3
    7 days/60° C. 7 92.7 93.8 93.6 93.4
    7 day/30° C. 7 97.8 97.2 97.0 96.9
    Total Δ/60° C. 5.3 4.0 4.2 4.3
    Total Δ/30° C. 0.3 0.6 0.8 0.8
  • TABLE 11
    The HPLC Peak Area of Impurity Peak 1 of Ceftolozane in CXA-
    201 Compositions with Varying Amounts of Sodium Chloride
    Day B1 B2 B3 B4
    t0
    0 0.47 0.38 0.38 0.41
    1 day/60° C. 1 1 1.08 1.09 1.14
    1 day/30° C. 1 0.48 0.44 0.45 0.49
    3 days/60° C. 3 1.85 1.64 1.66 1.71
    3 day/30° C. 3 0.53 0.53 0.56 0.61
    7 days/60° C. 7 3.3 2.28 2.25 2.29
    7 day/30° C. 7 0.62 0.67 0.71 0.77
    INCREASE %/60° C. 2.83 1.9 1.87 1.88
    INCREASE %/30° C. 0.15 0.29 0.33 0.36
  • TABLE 12
    The Total HPLC Peak Area of the Impurity with a RRT of
    0.43 and Impurity Peak 3 of Ceftolozane in CXA-201
    Compositions with Varying Amounts of Sodium Chloride
    Day B1 B2 B3 B4
    t0
    0 0.15 0.12 0.12 0.12
    1 day/60° C. 1 0.36 0.35 0.31 0.32
    1 day/30° C. 1 0.13 0.12 0.13 0.12
    3 days/60° C. 3 0.92 0.67 0.65 0.62
    3 days/30° C. 3 0.17 0.16 0.17 0.16
    7 days/60° C. 7 1.29 0.78 0.75 0.71
    7 days/30° C. 7 0.19 0.19 0.20 0.20
    INCREASE %/60° C. 1.14 0.66 0.63 0.59
    INCREASE %/30° C. 0.04 0.07 0.08 0.08
  • TABLE 13
    The HPLC Peak Area of Impurity Peak 7 of Ceftolozane in CXA-
    201 Compositions with Varying Amounts of Sodium Chloride
    Day B1 B2 B3 B4
    t0
    0 0.69 1.01 1.01 1.01
    1 day/60° C. 1 0.73 1.12 1.15 1.18
    1 day/30° C. 1 0.68 1.00 0.99 0.95
    3 days/60° C. 3 0.8 1.24 1.27 1.27
    3 days/30° C. 3 0.68 1.00 1.01 1.03
    7 days/60° C. 7 0.94 1.32 1.35 1.4
    7 days/30° C. 7 0.68 1.02 1.05 1.06
    INCREASE %/60° C. 0.25 0.31 0.34 0.39
    INCREASE %/30° C. NC 0.01 0.04 0.05

    Conclusion: the stability data shows that high sodium chloride content enhances stability of Combination product CXA-201.
  • Example 6 Manufacturing of Combination Product (Tazobactam and CXA-101) by Co-Lyophilization
  • The co-lyophilization process is illustrated in FIG. 14. The components of a CXA-201 composition prepared by co-lyophilization are described in Table 14 below.
  • TABLE 14
    Components of a CXA-201 Composition
    Prepared by Co-lyophilization
    Component Function Amount (mg/container)
    ceftolozane Active pharmaceutical 1000 (potency)
    ingredient
    L-arginine Alkalization reagent 587
    Citric acid Buffer 21
    (anhydrous)
    Sodium chloride Stabilizer 476
    Tazobactam Active pharmaceutical 500
    (free acid) ingredient
    Sodium Alkalization reagent Quantity sufficient1 for
    bicarbonate pH 4.8 to 7.0
    water Dissolution solvent Not more than 4% by weight2
    Nitrogen Inert gas Sufficient quantity
    1Sodium content is approximately 78 mg/g of tazobactam in drug product after lyophilization.
    2Water is removed during the lyophilization process and is controlled at no more than 4% by weight.
  • Example 7 Assessment of Co-Lyophilized Combo Drug Product A. Preparation of Co-Lyophilized Combo Drug Product
  • The Co-Lyo Combo Drug Product was prepared, as described above in Example 6. The formulation composition of the Combo drug product is shown in Table 15.
  • TABLE 15
    Components of the CXA-201 Composition
    Prepared by the Co-Lyophilization
    CXA-201 16.3 g active ceftolozane
    Comp. 8.1 g active Tazobactam free ac.
    15.5 g L-Arginine
    350 mg Citric acid
    7.9 g NaCl
    6.1 pH compounded solution
  • B. Stress Test
  • This sample was put into stability study. The following Tables 16 and 17 are representative examples that summarize the results at 25° C./RH=60% and 40° C./RH=75% after one month (T1) and three months (T2). Samples were analyzed using the HPLC method as described in Example 4.
  • TABLE 16
    Stability Data of Co-Lyophilized CXA-
    201 Composition at 25° C./RH = 60%
    Test items Spec. D.P. T0 T1 25° C. T2 25° C.
    Related Substances
    Peak1 ≦1.50% 0.31% 0.54% 0.71%
    Peak2 ≦0.40% 0.07% 0.07% 0.09%
    Peak3 ≦0.30% <0.03% <0.03% <0.03%
    Peak4 ≦0.80% 0.08% 0.08% 0.09%
    Peak5 ≦1.00% 0.27% 0.26% 0.29%
    Peak6 ≦0.15% <0.03% <0.03% <0.03%
    Peak7 ≦2.00% 0.64% 0.65% 0.66%
    Peak8 ≦0.15% <0.03% <0.03% <0.03%
    Peak9 ≦0.60% 0.05% 0.11% 0.10%
    Peak10, 11 ≦0.15% each 0.04% 0.04% 0.04%
    Peak12 ≦2.00% <0.03% <0.03% <0.03%
    Others (RRT 0.43) ≦0.15% <0.03% <0.03% 0.04%
    Others (RRT 1.22) ≦0.15% 0.13% 0.30% 0.38%
    Others (RRT 2.18) ≦0.15% 0.03% <0.03% 0.05%
    Others (RRT 2.77) ≦0.15% <0.03% 0.03% 0.03%
    Sing. Unk. ≦0.15% 0.05% 0.07% 0.05%
    Total ≦5.00% 1.67% 2.19% 2.77%
    pH report value 5.5 4.83
  • TABLE 17
    Stability Data Co-Lyophilized CXA-
    201 Composition at 40° C./RH = 75%
    Test items Spec. D.P. T0 T1 40° C. T2 40° C.
    Related Substances
    Peak1 ≦1.50% 0.31% 1.77% 2.22%
    Peak2 ≦0.40% 0.07% 0.10% 0.16%
    Peak3 ≦0.30% <0.03% <0.03% 0.06%
    Peak4 ≦0.80% 0.08% 0.09% 0.09%
    Peak5 ≦1.00% 0.27% 0.27% 0.30%
    Peak6 ≦0.15% <0.03% <0.03% <0.03%
    Peak7 ≦2.00% 0.64% 0.69% 0.78%
    Peak8 ≦0.15% <0.03% <0.03% 0.10%
    Peak9 ≦0.60% 0.05% 0.09% 0.09%
    Peak10, 11 ≦0.15% each 0.04% 0.04% 0.05%
    Peak12 ≦2.00% <0.03% <0.03% <0.03%
    Others (RRT 0.43) ≦0.15% <0.03% 0.09% 0.15%
    Others (RRT 1.22) ≦0.15% 0.13% 0.74% 0.97%
    Others (RRT 2.18) ≦0.15% 0.03% <0.03% 0.08%
    Others (RRT 2.77) ≦0.15% <0.03% <0.03% 0.04%
    Sing. Unk. ≦0.15% 0.05% 0.11% 0.25%
    Total ≦5.00% 1.67% 4.49% 6.32%
    pH report value 5.5 4.09
  • C. Conclusion:
  • A new impurity having RRT=1.22 was observed in the co-lyophilized drug product. The impurity was identified as a degradation product, shown in FIG. 15, which was formed by a reaction between ceftolozane and formylacetic acid, which was a degradation product of tazobactam. The stability data at 25° C. and at 40° C. have shown that the impurity increases over time.
  • Example 8 Assessment of Blend Combination Drug Product A. Preparation of Blend Combination Drug Product
  • The blend drug product was prepared, as described above in Example 3, on lab scale by use of a small blender. The composition of the blend drug product is shown in Table 18.
  • TABLE 18
    Components of the Blend Composition
    Quantity as active
    Component Composition components
    CXA-201 CXA-101 Ceftolozane 10.8 g
    Comp. for Injection L-Arginine 6.7 g
    Bulk (25 g) Citric acid 233 mg
    Sodium chloride 5.2 g
    Tazobactam sodium 5.4 g (as Tazo
    sterile Bulk(6 g) free acid)
  • B. Stress Test
  • This sample was put into stability study. The following Tables 19 and 20 are representative examples that summarizes the results at 25° C./RH=60% and 40° C./RH=75% after one month (T1) and three months (T2). Samples were analyzed using the HPLC method as described in Example 4.
  • TABLE 19
    Stability Data of Blend CXA-201
    Composition at 25° C./RH = 60%
    Test items Specifications T0 T1 25° C. T2 25° C.
    Related Substances
    Peak1 ≦1.50% 0.61% 0.93% 1.08%
    Peak2 ≦0.40% <0.03% <0.03% <0.03%
    Peak3 ≦0.30% <0.03% <0.03% <0.03%
    Peak4 ≦0.80% 0.03% 0.03% 0.04%
    Peak5 ≦1.00% 0.09% 0.12% 0.13%
    Peak6 ≦0.15% <0.03% <0.03% <0.03%
    Peak7 ≦2.00% 1.28% 1.34% 1.35%
    Peak8 ≦0.15% <0.03% <0.03% <0.03%
    Peak9 ≦0.60% 0.03% <0.03% 0.03%
    Peak10, 11 ≦0.30% <0.03% 0.04% 0.05%
    Sing. Unk. ≦0.15% 0.13% 0.13% 0.14%
    Total ≦5.00% 2.49% 3.03% 3.28%
    Assay CXA-101 Teor. 32.5% n.a. n.a.
    % = 32.6%
    Assay Tazobactam Teor. 18.2% n.a. n.a.
    % = 17.4%
    Tazobactam Related ≦4.0% 0.07% 0.12% 0.14%
    Compound A
    K.F. ≦4.0% 2.6% n.a. n.a.
    pH 5.0-7.0 6.0 5.6 5.1
  • TABLE 20
    Stability Data of Blend CXA-201
    Composition at 40° C./RH = 75%
    Test items Specifications T0 T1 40° C. T2 40° C.
    Related Substances
    Peak1 ≦1.50% 0.61% 1.66% 2.28%
    Peak2 ≦0.40% <0.03% <0.03% <0.03%
    Peak3 ≦0.30% <0.03% <0.03% 0.04%
    Peak4 ≦0.80% 0.03% 0.04% 0.05%
    Peak5 ≦1.00% 0.09% 0.13% 0.14%
    Peak6 ≦0.15% <0.03% <0.03% <0.03%
    Peak7 ≦2.00% 1.28% 1.41% 1.46%
    Peak8 ≦0.15% <0.03% <0.03% <0.03%
    Peak9 ≦0.60% 0.03% <0.03% 0.03%
    Peak10, 11 ≦0.30% <0.03% 0.08% 0.09%
    Sing. Unk. ≦0.15% 0.13% 0.14% 0.13%
    Total ≦5.00% 2.49% 4.21% 5.27%
    Assay CXA-101 Teor. 32.5% n.a. n.a.
    % = 32.6%
    Assay Tazobactam Teor. 18.2% n.a. n.a
    % = 17.4%
    Tazobactam Related ≦4.0% 0.07% 0.35% 0.54%
    Compound A
    K.F. ≦4.0% 2.6% n.a. n.a.
    pH 5.0-7.0 6.0 5.0 4.4
  • C. Conclusion
  • The data at both 25° C. and at 40° C. have shown that the blending process completely inhibits formation of the impurity RRT=1.22.

Claims (2)

1. A method of lyophilizing a CXA-101 pharmaceutical composition comprising the steps of:
a. combining in a vessel chamber water, citric acid, maltose, sodium bicarbonate and CXA-101 to form a solution;
b. adjusting the pH of the solution to about 6;
c. changing the temperature of the vessel to about −40° C. and maintaining until contents of the vessel freeze;
d. changing the temperature of the vessel to −20° C.;
e. changing the chamber pressure from 100 to 150 mTorr;
f. monitoring the temperature of the contents of the vessel;
g. changing the vessel temperature to 30° C.;
h. changing the chamber pressure to not more than 10 mTorr;
i. maintaining the contents of the chamber for a time sufficient to complete lyophilization of the contents of the chamber;
j. returning the chamber pressure to atmospheric pressure with sterilized nitrogen; and
k. stoppering the vessels completely.
2. The method of claim 1, practiced according to the program of FIG. 1.
US14/214,367 2013-03-15 2014-03-14 Tazobactam and ceftolozane antibiotic compositions Abandoned US20140274996A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/214,367 US20140274996A1 (en) 2013-03-15 2014-03-14 Tazobactam and ceftolozane antibiotic compositions
US14/522,893 US20150045336A1 (en) 2013-03-15 2014-10-24 Tazobactam and ceftolozane antibiotic compositions

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361792092P 2013-03-15 2013-03-15
US201361793007P 2013-03-15 2013-03-15
US14/214,367 US20140274996A1 (en) 2013-03-15 2014-03-14 Tazobactam and ceftolozane antibiotic compositions

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/522,893 Continuation US20150045336A1 (en) 2013-03-15 2014-10-24 Tazobactam and ceftolozane antibiotic compositions

Publications (1)

Publication Number Publication Date
US20140274996A1 true US20140274996A1 (en) 2014-09-18

Family

ID=51522651

Family Applications (12)

Application Number Title Priority Date Filing Date
US14/214,417 Abandoned US20140274997A1 (en) 2013-03-15 2014-03-14 Cephalosporin pharmaceutical compositions
US14/213,532 Abandoned US20140274991A1 (en) 2013-03-15 2014-03-14 Ceftolozane pharmaceutical compositions
US14/214,367 Abandoned US20140274996A1 (en) 2013-03-15 2014-03-14 Tazobactam and ceftolozane antibiotic compositions
US14/214,212 Abandoned US20140274993A1 (en) 2013-03-15 2014-03-14 Ceftolozane-tazobactam pharmaceutical compositions
US14/212,781 Abandoned US20140274990A1 (en) 2013-03-15 2014-03-14 Ceftolozane pharmaceutical compositions
US14/214,221 Active US9320740B2 (en) 2013-03-15 2014-03-14 Ceftolozane-tazobactam pharmaceutical compositions
US14/212,590 Abandoned US20140275000A1 (en) 2013-03-15 2014-03-14 Ceftolozane pharmaceutical compositions
US14/285,185 Active US8968753B2 (en) 2013-03-15 2014-05-22 Ceftolozane-tazobactam pharmaceutical compositions
US14/522,893 Abandoned US20150045336A1 (en) 2013-03-15 2014-10-24 Tazobactam and ceftolozane antibiotic compositions
US14/531,352 Abandoned US20150150883A1 (en) 2013-03-15 2014-11-03 Ceftolozane-tazobactam pharmaceutical compositions
US15/071,530 Active US9925196B2 (en) 2013-03-15 2016-03-16 Ceftolozane-tazobactam pharmaceutical compositions
US15/895,279 Abandoned US20180169106A1 (en) 2013-03-15 2018-02-13 Ceftolozane-tazobactam pharmaceutical compositions

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US14/214,417 Abandoned US20140274997A1 (en) 2013-03-15 2014-03-14 Cephalosporin pharmaceutical compositions
US14/213,532 Abandoned US20140274991A1 (en) 2013-03-15 2014-03-14 Ceftolozane pharmaceutical compositions

Family Applications After (9)

Application Number Title Priority Date Filing Date
US14/214,212 Abandoned US20140274993A1 (en) 2013-03-15 2014-03-14 Ceftolozane-tazobactam pharmaceutical compositions
US14/212,781 Abandoned US20140274990A1 (en) 2013-03-15 2014-03-14 Ceftolozane pharmaceutical compositions
US14/214,221 Active US9320740B2 (en) 2013-03-15 2014-03-14 Ceftolozane-tazobactam pharmaceutical compositions
US14/212,590 Abandoned US20140275000A1 (en) 2013-03-15 2014-03-14 Ceftolozane pharmaceutical compositions
US14/285,185 Active US8968753B2 (en) 2013-03-15 2014-05-22 Ceftolozane-tazobactam pharmaceutical compositions
US14/522,893 Abandoned US20150045336A1 (en) 2013-03-15 2014-10-24 Tazobactam and ceftolozane antibiotic compositions
US14/531,352 Abandoned US20150150883A1 (en) 2013-03-15 2014-11-03 Ceftolozane-tazobactam pharmaceutical compositions
US15/071,530 Active US9925196B2 (en) 2013-03-15 2016-03-16 Ceftolozane-tazobactam pharmaceutical compositions
US15/895,279 Abandoned US20180169106A1 (en) 2013-03-15 2018-02-13 Ceftolozane-tazobactam pharmaceutical compositions

Country Status (1)

Country Link
US (12) US20140274997A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8968753B2 (en) 2013-03-15 2015-03-03 Calixa Therapeutics, Inc. Ceftolozane-tazobactam pharmaceutical compositions
US9044485B2 (en) 2013-03-15 2015-06-02 Calixa Therapeutics, Inc. Ceftolozane antibiotic compositions
US9724353B2 (en) 2011-09-09 2017-08-08 Merck Sharp & Dohme Corp. Methods for treating intrapulmonary infections
US9872906B2 (en) 2013-03-15 2018-01-23 Merck Sharp & Dohme Corp. Ceftolozane antibiotic compositions
US10376496B2 (en) 2013-09-09 2019-08-13 Merck, Sharp & Dohme Corp. Treating infections with ceftolozane/tazobactam in subjects having impaired renal function

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2684150C (en) 2007-05-14 2016-10-04 Research Foundation Of State University Of New York Decenoic acid dispersion inducers in the treatment of biofilms
RU2671485C2 (en) * 2012-09-27 2018-11-01 Мерк Шарп И Доум Корп. Tazobactam arginine antibiotic compositions
CN110314163A (en) * 2018-03-30 2019-10-11 杭州森泽医药科技有限公司 A kind of latamoxef sodium pharmaceutical composition and application
US11541105B2 (en) 2018-06-01 2023-01-03 The Research Foundation For The State University Of New York Compositions and methods for disrupting biofilm formation and maintenance

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8906898B1 (en) * 2013-09-27 2014-12-09 Calixa Therapeutics, Inc. Solid forms of ceftolozane

Family Cites Families (265)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL37879A (en) 1970-10-27 1974-12-31 Ciba Geigy Ag 3-unsubstituted cephalosporin derivatives,process for their manufacture and compositions containing them
US4299829A (en) 1976-03-12 1981-11-10 Fujisawa Pharmaceutical Co., Ltd. 2-Lower alkyl-7-substituted-2 or 3-cephem 4-carboxylic acid compounds
US4409217A (en) 1977-03-14 1983-10-11 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4496562A (en) 1977-03-14 1985-01-29 Fujisawa Pharmaceutical Co., Ltd. 7-Substituted-3-cephem-4-carboxylic acid esters
PH17188A (en) 1977-03-14 1984-06-14 Fujisawa Pharmaceutical Co New cephem and cepham compounds and their pharmaceutical compositions and method of use
US4464369A (en) 1977-03-14 1984-08-07 Fujisawa Pharmaceutical Co., Ltd. 7-Acylamino-3-cephem-4-carboxylic acid derivatives and pharmaceutical compositions
JPS543087A (en) 1977-06-03 1979-01-11 Fujisawa Pharmaceut Co Ltd Preparation of cephalosporin compound
GB1604738A (en) 1977-07-28 1981-12-16 Yamanouchi Pharma Co Ltd 1,3-dithietane-2-carboxylic acid derivatives and the preparation thereof
JPS609719B2 (en) 1977-08-06 1985-03-12 武田薬品工業株式会社 Cephalosporin derivatives and their production method
US4370326A (en) 1977-09-13 1983-01-25 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds and composition
IT1192287B (en) 1977-11-14 1988-03-31 Fujisawa Pharmaceutical Co PHARMACEUTICAL ACTION DERIVATIVES OF CEPHALOSPORANIC ACID AND RELATED PREPARATION PROCEDURE
US4363807A (en) 1978-04-06 1982-12-14 Fujisawa Pharmaceutical Company, Limited Cepham compounds
SE7804231L (en) 1978-04-14 1979-10-15 Haessle Ab Gastric acid secretion
AR229883A1 (en) 1978-05-26 1983-12-30 Glaxo Group Ltd PROCEDURE FOR THE PREPARATION OF ANTIBIOTIC (6R, 7R) -7 - ((Z) -2- (2-AMINOTIAZOL-4-IL) -2- (2-CARBOXIPROP-2-OXIIMINO) -ACETAMIDO) -3- (1 -PIRIDINOMETIL) -CEF-3-EM-4-CARBOXYLATE
US4264597A (en) 1978-06-06 1981-04-28 Masashi Hashimoto Cephalosporin analogues and processes for the preparation thereof
US4268509A (en) 1978-07-10 1981-05-19 Fujisawa Pharmaceutical Co., Ltd. New cephem compounds and processes for preparation thereof
US4284631A (en) 1978-07-31 1981-08-18 Fujisawa Pharmaceutical Co., Ltd. 7-Substituted cephem compounds and pharmaceutical antibacterial compositions containing them
US4305937A (en) 1978-08-17 1981-12-15 Fujisawa Pharmaceutical Co., Ltd. 2-Lower alkyl-7-substituted-2 or 3-cephem-4-carboxylic acid compounds and antibacterial pharmaceutical compositions containing them
US4703046A (en) 1978-09-08 1987-10-27 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds and processes for preparation thereof
EP0048504B1 (en) 1978-09-12 1988-08-17 Fujisawa Pharmaceutical Co., Ltd. Intermediate compounds for preparing cephem compounds; processes for their preparation and processes for preparing cephem compounds
US4327093A (en) 1978-10-24 1982-04-27 Fujisawa Pharmaceutical Co., Ltd. 3,7-Disubstituted-2 or 3-cephem-4-carboxylic acid compounds
DE2945248A1 (en) 1978-11-13 1980-05-22 Fujisawa Pharmaceutical Co CEPHEM COMPOUNDS, METHOD FOR THEIR PRODUCTION AND ANTIBACTERIAL PHARMACEUTICAL AGENTS CONTAINING THE SAME
US4390534A (en) 1978-12-29 1983-06-28 Fujisawa Pharmaceutical Co., Ltd. Cephem and cepham compounds
US4332798A (en) 1978-12-29 1982-06-01 Fujisawa Pharmaceutical Co., Ltd. 7-Amino-thia-diazole oxyimino derivatives of cephem and cephem compounds
AU536842B2 (en) 1978-12-29 1984-05-24 Fujisawa Pharmaceutical Co., Ltd. Cephalosporin antibiotics
US4291031A (en) 1979-02-19 1981-09-22 Fujisawa Pharmaceutical Co., Ltd. 3-Phosphonocephalosporanic acid derivatives, and pharmaceutical composition comprising the same
US4339449A (en) 1979-03-27 1982-07-13 Fujisawa Pharmaceutical Company, Limited Analogous compounds of cephalosporins, and pharmaceutical composition comprising the same
FR2462439A1 (en) 1979-07-26 1981-02-13 Roussel Uclaf NOVEL PROCESS FOR THE PREPARATION OF PRODUCTS DERIVED FROM 7 - / (2-ARYL) 2-HYDROXYIMINO ACETAMIDO / CEPHALOSPORANIC ACID
EP0088454B1 (en) 1979-09-03 1986-03-05 Fujisawa Pharmaceutical Co., Ltd. New starting compounds for preparing cephem compounds and processes for their preparation
US4381299A (en) 1980-03-07 1983-04-26 Fujisawa Pharmaceutical Co., Ltd. 7-Amino-thiadiazole oxyimino derivatives of cephem and cepham compounds
US4332800A (en) 1979-10-12 1982-06-01 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4338313A (en) 1979-10-12 1982-07-06 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4409214A (en) 1979-11-19 1983-10-11 Fujisawa Pharmaceutical, Co., Ltd. 7-Acylamino-3-vinylcephalosporanic acid derivatives and processes for the preparation thereof
US4409215A (en) 1979-11-19 1983-10-11 Fujisawa Pharmaceutical Co., Ltd. 7-Acylamino-3-substituted cephalosporanic acid derivatives and processes for the preparation thereof
US4420477A (en) 1979-11-30 1983-12-13 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4443443A (en) 1979-12-17 1984-04-17 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4405617A (en) 1980-02-11 1983-09-20 Fujisawa Pharmaceutical Co., Ltd. 3-(Propynyltetrazol)thiomethyl-3-cephems
JPS56125392A (en) 1980-03-06 1981-10-01 Fujisawa Pharmaceut Co Ltd Cepham and cephem compound and preparation thereof
US4470980A (en) 1980-03-07 1984-09-11 Interx Research Corp. Method of increasing oral absorption of β-lactam antibiotics
JPS5711909A (en) 1980-06-23 1982-01-21 Shionogi & Co Ltd Stable freeze-dried preparation of beta-lactam
DE3173613D1 (en) 1980-07-04 1986-03-13 Fujisawa Pharmaceutical Co 7-oxo-cephalosporins and 6-oxo-penicillins, their analogues and process for their preparation
US4443444A (en) 1980-08-11 1984-04-17 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
EP0156118A1 (en) 1980-08-29 1985-10-02 Fujisawa Pharmaceutical Co., Ltd. New starting compounds for the preparation of cephem compounds and processes for preparation thereof
US4416879A (en) 1980-09-08 1983-11-22 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
GR78245B (en) 1980-09-12 1984-09-26 Ciba Geigy Ag
US4367228A (en) 1980-10-29 1983-01-04 Fujisawa Pharmaceutical Co., Ltd. Cephem compound and composition
US4431642A (en) 1980-12-01 1984-02-14 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
ES507942A0 (en) 1980-12-15 1983-02-01 Fujisawa Pharmaceutical Co A PROCEDURE FOR PREPARING DERIVATIVES OF 7-ACYLAMINOCEFHALOSPORANIC ACID.
US4427677A (en) 1980-12-31 1984-01-24 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
DE3177090D1 (en) 1980-12-31 1989-09-28 Fujisawa Pharmaceutical Co 7-acylaminocephalosporanic acid derivatives and processes for the preparation thereof
GR76342B (en) 1981-02-02 1984-08-06 Fujisawa Pharmaceutical Co
US4336253A (en) 1981-03-11 1982-06-22 Eli Lilly And Company Cephalosporin antibiotics
JPS6011917B2 (en) 1981-04-09 1985-03-28 山之内製薬株式会社 Novel cephalosporin compounds
DE3118732A1 (en) 1981-05-12 1982-12-02 Hoechst Ag, 6000 Frankfurt CEPHALOSPORINE DERIVATIVES AND METHOD FOR THEIR PRODUCTION
JPS57193489A (en) 1981-05-21 1982-11-27 Fujisawa Pharmaceut Co Ltd Syn-isomer of 7-substituted-3-cephem-4-carboxylic acid ester and its preparation
GR75487B (en) 1981-06-22 1984-07-23 Fujisawa Pharmaceutical Co
IE53429B1 (en) 1981-08-03 1988-11-09 Fujisawa Pharmaceutical Co New cephem compounds and processes for preparation thereof
US4430499A (en) 1981-09-08 1984-02-07 Eli Lilly And Company 7-[2-(2-Aminooxazol-4-yl)-2-(oximino)acetamido]cephalosporin antibiotics
US4577014A (en) 1981-09-08 1986-03-18 Eli Lilly And Company Thieno and furopyridinium-substituted cephalosporins
US4436912A (en) 1981-09-08 1984-03-13 Eli Lilly And Company 7-[2-(2-Aminooxazol-4-yl)-2-(oximino)acetamido cephalosporin antibiotics and intermediates therefor
US4521413A (en) 1981-09-14 1985-06-04 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
JPS5859991A (en) 1981-09-14 1983-04-09 Fujisawa Pharmaceut Co Ltd Novel cephem compound and its preparation
US4402955A (en) 1981-10-02 1983-09-06 Eli Lilly And Company Dioximino cephalosporin antibiotics
US4450270A (en) 1981-10-02 1984-05-22 Eli Lilly And Company Dioximino cephalosporin antibiotics
US4501739A (en) 1982-01-19 1985-02-26 Eli Lilly And Company Thieno and furopyridinium-substituted cephalosporins
DE3207840A1 (en) 1982-03-04 1983-09-15 Hoechst Ag, 6230 Frankfurt "CEPHALOSPORINE DERIVATIVES AND METHOD FOR THE PRODUCTION THEREOF"
JPS58154547A (en) 1982-03-09 1983-09-14 Nippon Shinyaku Co Ltd Stabilization of azulene derivative
US4640915A (en) 1982-03-29 1987-02-03 Fujisawa Pharmaceutical Co., Ltd. 1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid derivatives
AU541028B2 (en) 1982-06-21 1984-12-13 Taiho Pharmaceutical Co., Ltd. 6-unsubstituted penicillin derivatives
JPS58225091A (en) 1982-06-21 1983-12-27 Taiho Yakuhin Kogyo Kk Penicillin derivative and its preparation
US4563449A (en) 1982-07-19 1986-01-07 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4546101A (en) 1982-09-10 1985-10-08 Fujisawa Pharmaceutical Co., Ltd. New cephem compounds useful for treating infectious diseases in human being and animals and processes for preparation thereof
GB8323034D0 (en) 1983-08-26 1983-09-28 Fujisawo Pharmaceutical Co Ltd 7-substituted-3-vinyl-3-cephem compounds
US4609730A (en) 1982-11-22 1986-09-02 Fujisawa Pharmaceutical Co., Ltd. 7-[substituted imino-2-(2-aminothiazol-4-yl)-acetamido]-3(2,2-dihalovinyl or ethynyl)-3-cephem-4-carboxylic acid (syn isomers), having antimicrobial activities
GR79043B (en) 1982-12-06 1984-10-02 Fujisawa Pharmaceutical Co
US4608373A (en) 1982-12-13 1986-08-26 Yamanouchi Pharmaceutical Co., Ltd. Cephem compounds
US4463003A (en) 1982-12-22 1984-07-31 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4487768A (en) 1982-12-22 1984-12-11 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
DE3247613A1 (en) 1982-12-23 1984-07-05 Hoechst Ag, 6230 Frankfurt CEPHALOSPORINE DERIVATIVES AND METHOD FOR THEIR PRODUCTION
US4562073A (en) 1982-12-24 1985-12-31 Taiho Pharmaceutical Company Limited Penicillin derivatives
US4499088A (en) 1983-01-04 1985-02-12 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
DE3316798A1 (en) 1983-05-07 1984-11-08 Hoechst Ag, 6230 Frankfurt METHOD FOR PRODUCING CEPHEM COMPOUNDS
FR2550200B1 (en) 1983-08-01 1988-04-08 Fujisawa Pharmaceutical Co PROCESS FOR THE PREPARATION OF CEPHEM COMPOUNDS WITH ANTIMICROBIAL ACTIVITY AND NOVEL PRODUCTS THUS OBTAINED
JPS6045514A (en) 1983-08-22 1985-03-12 Shionogi & Co Ltd Stable antibacterial lyophilized pharmactical preparation
EP0137442A3 (en) 1983-10-08 1986-01-15 Hoechst Aktiengesellschaft Cephalosporin derivatives and process for their preparation
DE3409431A1 (en) 1983-10-08 1985-04-18 Hoechst Ag, 6230 Frankfurt CEPHALOSPORINE DERIVATIVES AND METHOD FOR THEIR PRODUCTION
US4690921A (en) 1983-10-11 1987-09-01 Yamanouchi Pharmaceutical Co., Ltd. Cephalosporin compounds and salts thereof
US4692443A (en) 1983-10-17 1987-09-08 Eli Lilly And Company 3-bicyclicpyridinium-methyl cephalosporins
EP0138552A3 (en) 1983-10-17 1986-03-19 Eli Lilly And Company Improvements on or relating to 3-bicyclicpyridinium-methyl cephalosporins
US4748172A (en) 1983-10-17 1988-05-31 Eli Lilly And Company 3-bicyclicpyridinium-methyl cephalosporins
GB8329030D0 (en) 1983-10-31 1983-11-30 Fujisawa Pharmaceutical Co Cephem compounds
GB8401093D0 (en) 1984-01-16 1984-02-15 Fujisawa Pharmaceutical Co Cephem compounds
JPS60169486A (en) 1984-02-10 1985-09-02 Yamanouchi Pharmaceut Co Ltd Preparation of 7-amino-3-substituted methyl-3-cephem-4- carboxylic acid and lower alkylsilyl derivative thereof
GB8406231D0 (en) 1984-03-09 1984-04-11 Fujisawa Pharmaceutical Co Cephem compounds
JPS60214792A (en) 1984-04-06 1985-10-28 Taiho Yakuhin Kogyo Kk Penamylacid ester derivative
US4705851A (en) 1984-09-28 1987-11-10 Fujisawa Pharmaceutical Co., Ltd. Process for the preparation of 3-phosphoniummethyl-3-cephem compounds
US4761410A (en) 1985-01-14 1988-08-02 Fujisawa Pharmaceutical Co., Ltd. Cephem Compounds
CA1277977C (en) 1985-01-21 1990-12-18 Shigeo Shimizu Beta-lactam antibiotics
GB8504072D0 (en) 1985-02-18 1985-03-20 Fujisawa Pharmaceutical Co Cephem compounds
JPS62103092A (en) 1985-07-18 1987-05-13 Sagami Chem Res Center Beta-lactam derivative
JPS6230789A (en) 1985-08-01 1987-02-09 Yamanouchi Pharmaceut Co Ltd 7-formylaminocephalosporin compound and production thereof
CN86107947A (en) 1985-11-22 1987-05-27 藤沢药品工业株式会社 New cephem compounds and preparation method thereof
US5194432A (en) 1985-11-22 1993-03-16 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4808617A (en) 1985-12-18 1989-02-28 Bristol-Myers Company Lyophilized or precipitated cephalosporin zwitterion and salt combination
JPS62158290A (en) 1985-12-28 1987-07-14 Banyu Pharmaceut Co Ltd Novel cephalosporin derivative
DE3789466T2 (en) 1986-03-17 1994-07-28 Fujisawa Pharmaceutical Co 3,7-disubstituted-3-cephem compounds and process for their preparation.
JP2690009B2 (en) 1986-07-10 1997-12-10 エーザイ 株式会社 Cefalosporin injection
US4833134A (en) 1986-08-19 1989-05-23 Takeda Chemical Industries, Ltd. Cephem compounds
JPS6351388A (en) 1986-08-22 1988-03-04 Teijin Ltd Cephalosporin derivative, production thereof and composition having antimicrobial activity
JPS6351389A (en) 1986-08-22 1988-03-04 Teijin Ltd Cephalosporin derivative, production thereof and composition having antimicrobial activity
US5162520A (en) 1986-09-22 1992-11-10 Fujisawa Pharmaceutical Co., Ltd. Intermediates for cephem compounds
CA1293719C (en) 1986-09-22 1991-12-31 Takao Takaya Cephem compounds and processes for preparation thereof
US4882434A (en) 1986-10-29 1989-11-21 Takeda Chemical Industries, Ltd. Gamma-lactonecarboxylic acid derivatives and their use as antibacterial agents or intermediates
DE3784147T2 (en) 1986-11-24 1993-06-03 Fujisawa Pharmaceutical Co 3-PYRROLIDINYLTHIO-1-AZABICYCLO (3.2.0) HEPT-2-EN-2-CARBONIC ACID DERIVATIVES.
KR880006244A (en) 1986-11-24 1988-07-22 후지사와 도모 기찌 로 3-Pyrrolidinylthio-1-azabibischloro [3.2.0] hapt2-ene-2-carboxylic acid compound and preparation method thereof
AU1630988A (en) 1987-05-30 1988-12-01 Kyoto Pharmaceutical Industries, Ltd. Cephalosporin compound and pharmaceutical composition thereof
IE61679B1 (en) 1987-08-10 1994-11-16 Fujisawa Pharmaceutical Co Water-soluble antibiotic composition and water-soluble salts of new cephem compounds
US5138066A (en) 1987-08-14 1992-08-11 Hoffmann-La Roche, Inc. Intermediates for cephalosporins with sulfur-containing oxyimino side chain
US5073550A (en) 1987-08-14 1991-12-17 Hoffmann-La Roche Inc. Cephalosphorins with sulfur-containing oxyimino side chain
EP0303172A3 (en) 1987-08-14 1991-05-15 F. Hoffmann-La Roche Ag Oxyimino-cephalosporins
ZA885709B (en) 1987-08-19 1989-04-26 Fujisawa Pharmaceutical Co Novel crystalline 7-(2-(2-aminothiazol-4-yl)-2-hydroxyiminoacetamido)-3-vinyl-3-cephem-4-carboxylic acid(syn isomer)
US5663163A (en) 1987-09-07 1997-09-02 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds and processes for preparation thereof
US5210080A (en) 1987-09-07 1993-05-11 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
IE63094B1 (en) 1987-09-14 1995-03-22 Fujisawa Pharmaceutical Co Cephem compound and a process for preparation thereof
DK637888A (en) 1987-11-24 1989-05-25 Hoffmann La Roche carboxylic esters
GB8804058D0 (en) 1988-02-22 1988-03-23 Fujisawa Pharmaceutical Co 3-alkenyl-1-azabicyclo(3 2 0)hept-2-ene-2-carboxylic acid compounds
JP2648750B2 (en) 1988-03-02 1997-09-03 大塚化学株式会社 Method for producing β-lactam derivative
US5173485A (en) 1988-03-09 1992-12-22 Fujisawa Pharmaceutical Company, Ltd. Cephem compounds
CS273349B2 (en) 1988-03-31 1991-03-12 Hoffmann La Roche Method of cephalosporin's new derivatives production
US5336768A (en) 1988-05-24 1994-08-09 Hoffmann-La Roche Inc. Antibacterial cephalosporin compounds
KR900006811B1 (en) 1988-05-11 1990-09-21 주식회사 럭 키 Cephalosphorin derivatives and its process
US4963544A (en) 1988-05-23 1990-10-16 Fujisawa Pharmaceutical Company, Ltd. 3-pyrrolidinylthio-1-azabicyclo[3.2.0]-hept-2-ene-2-carboxylic acid compounds
US5244890A (en) 1988-06-06 1993-09-14 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
EP0366189A3 (en) 1988-10-24 1992-01-02 Norwich Eaton Pharmaceuticals, Inc. Novel antimicrobial lactam-quinolones
JP2785195B2 (en) 1989-01-11 1998-08-13 ソニー株式会社 Optical encoder for disk drive
GB8905301D0 (en) 1989-03-08 1989-04-19 Fujisawa Pharmaceutical Co New cephem compound and a process for preparation thereof
JPH0347187A (en) 1989-04-12 1991-02-28 Yamanouchi Pharmaceut Co Ltd New cephalosporin derivative
US5102877A (en) 1989-04-28 1992-04-07 Fujisawa Pharmaceutical Co., Ltd. 1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid compounds
NO903360L (en) 1989-08-11 1991-02-12 Ici Pharma ANTIBIOTIC COMPOUNDS.
GB8923844D0 (en) 1989-10-23 1989-12-13 Fujisawa Pharmaceutical Co Carbapenem compounds
US5215982A (en) 1989-11-10 1993-06-01 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4982596A (en) 1990-01-26 1991-01-08 Buell Industries, Inc. Die for manufacturing a fastener blank
KR910015587A (en) 1990-02-27 1991-09-30 후지사와 토모키치로 Cefem compound
US5234920A (en) 1990-08-23 1993-08-10 Bristol-Myers Squibb Company Antibiotic C-7 catechol-substituted cephalosporin compounds, compositions, and method of use thereof
US5095012A (en) 1990-08-23 1992-03-10 Bristol-Myers Squibb Company Antibiotic c-7 catechol-substituted cephalosporin compounds, compositions, and method of use thereof
US5286721A (en) 1990-10-15 1994-02-15 Fujisawa Pharmaceutical Co., Ltd. 1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid compounds
GB9023479D0 (en) 1990-10-29 1990-12-12 Fujisawa Pharmaceutical Co New cephem compounds and processes for preparation thereof
US5281589A (en) 1991-06-15 1994-01-25 Cheil Foods & Chemicals, Inc. 3-fused pyridiniummethyl cephalosporins
US5523400A (en) 1993-04-16 1996-06-04 Hoffmann-La Roche Inc. Cephalosporin antibiotics
KR100194994B1 (en) 1993-06-05 1999-06-15 손경식 New cefem compound
AU679800B2 (en) 1993-11-06 1997-07-10 Taiho Pharmaceutical Co., Ltd. Crystalline penicillin derivative, and its production and use
EP0664117A1 (en) 1994-01-25 1995-07-26 F. Hoffmann-La Roche Ag Liposome solutions
TW293010B (en) 1994-04-20 1996-12-11 Hui-Po Wang Method for preparing cephalosporin derivatives
US5661144A (en) 1994-08-16 1997-08-26 Meiji Seika Kabushiki Kaisha Cephem derivatives with 3-substituted bis heterocycles
DE4440141A1 (en) 1994-11-10 1996-05-15 Hoechst Ag Novel crystalline cephem acid addition salts and process for their preparation
JPH09110877A (en) 1995-10-17 1997-04-28 Katayama Seiyakushiyo:Kk Cephem compound, its production and antibacterial agent containing the compound
ATE341554T1 (en) 1996-04-04 2006-10-15 Shionogi & Co CEPHEM COMPOUNDS AND MEDICATIONS CONTAINING THESE COMPOUNDS
AUPN955596A0 (en) 1996-04-30 1996-05-23 Fujisawa Pharmaceutical Co., Ltd. New compound
EP0970065A4 (en) 1997-11-29 2001-03-07 William L Truett Antibiotics and process for preparation
US6235311B1 (en) 1998-03-18 2001-05-22 Bristol-Myers Squibb Company Pharmaceutical composition containing a combination of a statin and aspirin and method
US6288234B1 (en) 1998-06-08 2001-09-11 Advanced Medicine, Inc. Multibinding inhibitors of microsomal triglyceride transferase protein
WO2000004915A1 (en) 1998-07-23 2000-02-03 Intrabiotics Pharmaceuticals, Inc. Compositions and methods for the treatment or prevention of pulmonary infections
TW526202B (en) 1998-11-27 2003-04-01 Shionogi & Amp Co Broad spectrum cephem having benzo[4,5-b]pyridium methyl group of antibiotic activity
CN1109688C (en) 1999-01-12 2003-05-28 中国药品生物制品检定所 Preparation and application of tazobactam semihydrate
US6207661B1 (en) 1999-02-22 2001-03-27 Baxter International Inc. Premixed formulation of piperacillin sodium and tazobactam sodium injection
TWI233805B (en) 1999-07-01 2005-06-11 Fujisawa Pharmaceutical Co Stabilized pharmaceutical composition in lyophilized form as antifungal agent
NZ517663A (en) 1999-08-16 2006-02-24 Revaax Pharmaceuticals Llc Neurotherapeutic clavulanate composition and method
DK1277471T3 (en) 2000-04-24 2008-08-11 Daiichi Sankyo Co Ltd Stable liquid preparation comprising sitafloxacin
JP3743822B2 (en) 2000-08-11 2006-02-08 大塚化学ホールディングス株式会社 Penicillin crystals and production method thereof
JP3743823B2 (en) 2000-08-11 2006-02-08 大塚化学ホールディングス株式会社 Penicillin crystals and production method thereof
US6599893B2 (en) 2000-08-29 2003-07-29 Essential Therapeutics, Inc. Cephalosporin antibiotics and prodrugs thereof
ES2254671T3 (en) 2001-05-01 2006-06-16 Astellas Pharma Inc. CEFEM COMPOUNDS.
JP3306473B1 (en) 2001-05-01 2002-07-24 大塚化学株式会社 Anhydrous crystal of β-lactam compound and method for producing the same
JP2002338578A (en) 2001-05-14 2002-11-27 Otsuka Chem Co Ltd HYDRATE CRYSTAL OF beta-LACTAM COMPOUND
WO2002102378A1 (en) 2001-06-18 2002-12-27 Revaax Pharmaceuticals, Llc Therapeutic treatment for sexual dysfunction
TWI335332B (en) 2001-10-12 2011-01-01 Theravance Inc Cross-linked vancomycin-cephalosporin antibiotics
US7378408B2 (en) 2001-11-30 2008-05-27 Pfizer Inc. Methods of treatment and formulations of cephalosporin
WO2003066053A1 (en) 2002-02-07 2003-08-14 Rutgers, The State University Antibiotic polymers
TW200305422A (en) 2002-03-18 2003-11-01 Shionogi & Co Broad spectrum cefem compounds
EP1507796B1 (en) 2002-05-24 2012-05-02 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
ES2274978T3 (en) 2002-06-07 2007-06-01 Orchid Chemicals And Pharmaceuticals Limited PROCEDURE FOR THE REPAIR OF PENAM DERIVATIVES FROM CEFAM DERIVATIVES.
AU2003260803A1 (en) 2002-08-30 2004-03-19 Orchid Chemicals And Pharmaceuticals Ltd. Sustained release pharmaceutical composition
US9211259B2 (en) 2002-11-29 2015-12-15 Foamix Pharmaceuticals Ltd. Antibiotic kit and composition and uses thereof
ES2290498T7 (en) 2002-10-30 2015-07-10 Astellas Pharma Inc. Cefem compounds
AU2003274518A1 (en) 2002-11-01 2004-05-25 Orchid Chemicals And Pharmaceuticals Ltd A process for the preparation of benzyl 2-oxo-4- (heteroaryl) dithio-alpha-isoprenyl-1- azetidineazetate derivatives
ATE540129T1 (en) 2002-11-22 2012-01-15 Univ Johns Hopkins TARGET FOR THE THERAPY OF COGNITIVE DISABILITIES
DE10304403A1 (en) 2003-01-28 2004-08-05 Röhm GmbH & Co. KG Process for the preparation of an oral dosage form with immediate disintegration and drug release
JP4288086B2 (en) 2003-02-25 2009-07-01 大日本印刷株式会社 Exposure equipment
AU2003230899A1 (en) 2003-04-14 2004-11-26 Wyeth Holdings Corporation Compositions containing piperacillin and tazobactam useful for injection
DE602004010862T2 (en) 2003-04-14 2009-01-02 Wyeth Holdings Corp. Compositions containing piperacillin and tazobactam for injection
AU2003902380A0 (en) 2003-05-16 2003-06-05 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
ES2335013T3 (en) 2003-05-23 2010-03-18 Theravance, Inc. GLUCOPEPTIDE-CEPHALOSPORIN ANTIBIOTICS RETICULATED.
ATE388153T1 (en) 2003-07-11 2008-03-15 Theravance Inc CROSS-LINKED GLYCOPEPTIDE CEPHALOSPORIN ANTIBIOTICS
US8173840B2 (en) 2003-07-29 2012-05-08 Signature R&D Holdings, Llc Compounds with high therapeutic index
US7589233B2 (en) 2003-07-29 2009-09-15 Signature R&D Holdings, Llc L-Threonine derivatives of high therapeutic index
US7273935B2 (en) 2003-08-21 2007-09-25 Orchid Chemicals & Pharmaceuticals, Ltd. Process for the preparation of 3-methylcepham derivatives
US7192943B2 (en) 2003-09-18 2007-03-20 Astellas Pharma Inc. Cephem compounds
FR2860235A1 (en) 2003-09-29 2005-04-01 Yang Ji Chemical Company Ltd USE OF A COMPOUND OF FORMULA (I) INHIBITOR OF AROMATASE FOR THERAPEUTIC PURPOSES AND COMPOUNDS OF FORMULA (I) AS SUCH
TW200523264A (en) 2003-10-09 2005-07-16 Otsuka Chemical Co Ltd CMPB crystal and method for producing the same
TW200519119A (en) 2003-10-10 2005-06-16 Otsuka Chemical Co Ltd PENAM crystal and process for producing the same
JP4535366B2 (en) 2003-12-03 2010-09-01 塩野義製薬株式会社 Method for producing cephem agent
WO2005074925A1 (en) 2004-01-30 2005-08-18 Wyeth Compositions substantially free of galactomannan containing piperacillin and tazobactam
WO2005078848A2 (en) 2004-02-11 2005-08-25 University Of Tennessee Research Foundation Inhibition of tumor growth and invasion by anti-matrix metalloproteinase dnazymes
KR20060135796A (en) 2004-03-05 2006-12-29 시오노기세이야쿠가부시키가이샤 3-pyridinium methylcephem compound
US7417143B2 (en) 2004-04-07 2008-08-26 Orchid Chemicals & Pharmaceuticals Limited Process for the preparation of Tazobactam in pure form
BRPI0516583A (en) 2004-10-14 2008-09-16 Wyeth Corp pharmaceutical composition and method of treating a bacterial infection
US20060099253A1 (en) 2004-10-20 2006-05-11 Wyeth Antibiotic product formulation
US20060173177A1 (en) 2005-01-28 2006-08-03 Gego Csaba L Process for preparation of penam derivatives
CN101080221A (en) 2005-02-14 2007-11-28 维纳斯药业有限公司 Parenteral combination therpy for infective conditions with drug resistant bacterium
WO2006088305A1 (en) 2005-02-15 2006-08-24 Chong Kun Dang Pharmaceutical Corp. Gastric-retentive controlled release mono-matrix tablet
EP1919449A2 (en) 2005-06-07 2008-05-14 Foamix Ltd. Antibiotic kit and composition and uses thereof
ITMI20051630A1 (en) 2005-09-02 2007-03-03 Acs Dobfar Spa INJECTABLE STERILE PHARMACEUTICAL FORMULATION CONTAINING AT LEAST TWO ACTIVE PRINCIPLES
JP2009510077A (en) 2005-09-29 2009-03-12 ネクター セラピューティックス Antibiotic formulations, unit doses, kits and methods
EP1787641A1 (en) 2005-11-22 2007-05-23 Helm AG Tazobactam-piperacillin lyophilisate
PL1959933T3 (en) 2005-12-05 2011-04-29 Sandoz Ag Process for the preparation of lyophilized piperacillin sodium in combination with tazobactam sodium, with improved stability after reconstitution
WO2007086011A1 (en) 2006-01-25 2007-08-02 Jegannathan Srinivas Formulation comprising cefepime, tazobactam and linezolid
WO2007086014A1 (en) 2006-01-25 2007-08-02 Jegannathan Srinivas Formulation comprising cefpirome, tazobactam and linezolid
WO2007086013A1 (en) 2006-01-25 2007-08-02 Jegannathan Srinivas Formulation comprising of ceftazidime, tazobactam and linezolid
WO2008075207A2 (en) 2006-04-04 2008-06-26 Foamix Ltd. Anti-infection augmentation foamable compositions and kit and uses thereof
WO2007129176A2 (en) 2006-04-28 2007-11-15 Wockhardt Ltd Improvements in therapy for treating resistant bacterial infections
US20070286818A1 (en) 2006-06-07 2007-12-13 Wyeth Treating cystic fibrosis with antibiotics via an aerosol drug
WO2007145866A1 (en) 2006-06-07 2007-12-21 Wyeth Treating cystic fibrosis with antibiotics via a swirler delivery
PE20080329A1 (en) 2006-06-07 2008-04-09 Wyeth Corp TREATMENT OF CYST FIBROSIS WITH ANTIBIOTICS VIA A DRUG IN AEROSOL
US20070286817A1 (en) 2006-06-07 2007-12-13 Wyeth Treating cystic fibrosis with antibiotics via a swirler delivery
DK2046802T3 (en) 2006-07-12 2013-09-16 Allecra Therapeutics Gmbh 2-substituted methylpenam derivatives
CN101129382B (en) 2006-08-25 2013-12-25 天津和美生物技术有限公司 Antibiotic compound containing beta-lactam antibiotic and buffering component
CN101129381B (en) 2006-08-25 2012-02-01 天津和美生物技术有限公司 Antibiotic compound containing beta-lactam antibiotic and ion chelating agent
CN101129383B (en) 2006-08-25 2014-04-02 天津和美生物技术有限公司 Antibiotic compound containing aminoglycoside antibiotic
AU2007293068C1 (en) 2006-09-07 2013-09-19 Boehringer Ingelheim Animal Health USA Inc. Soft chewable, tablet, and long-acting injectable veterinary antibiotic formulations
US20080103121A1 (en) 2006-10-30 2008-05-01 Gole Dilip J Cephalosporin derivative formulation
FI119678B (en) 2006-11-28 2009-02-13 Ipsat Therapies Oy Use of beta-lactamase
JP5324463B2 (en) 2006-12-10 2013-10-23 チョンシー ユー Transdermal delivery system for β-lactam antibiotics
DE102007009242A1 (en) 2007-02-22 2008-09-18 Evonik Röhm Gmbh Pellets with enteric-coated matix
CA2677670C (en) 2007-03-20 2010-08-03 Centre De Recherche Sur Les Biotechnologies Marines Compositions comprising polyunsaturated fatty acid monoglycerides or derivatives thereof and uses thereof
ITMI20070568A1 (en) 2007-03-22 2008-09-23 Acs Dobfar Spa INJECTABLE STERILE PHARMACEUTICAL COMOSIATION HAVING PIPERACILLIN SODIUM AND TAZOBACTAM SODIUM AS ACTIVE PRINCIPLES
US20090098088A1 (en) 2007-10-10 2009-04-16 The Procter & Gamble Company Methods And Kits For The Treatment Of Diverticular Conditions
WO2009105782A1 (en) 2008-02-21 2009-08-27 Sequoia Pharmaceuticals, Inc. Hiv protease inhibitor and cytochrome p450 inhibitor combinations
KR20100137439A (en) 2008-03-04 2010-12-30 엘란 파마 인터내셔널 리미티드 Stable liquid formulations of anti-infective agents and adjusted anti-infective agent dosing regimens
ITPI20080025A1 (en) 2008-03-31 2009-10-01 Italmed S R L COMPOSITION FOR DENTAL USE FOR THE TREATMENT OF PERIMPLANTS
RU2010143460A (en) 2008-05-01 2012-06-10 Дзе Проктер Энд Гэмбл Компани (US) METHODS AND KITS FOR THERAPY OF REDUCING INTESTINAL STATES
WO2010014285A1 (en) 2008-07-30 2010-02-04 Estabrook Pharmaceuticals, Inc. Compositions including clavulanic acid and related methods of use
CN101434610B (en) 2008-12-19 2011-07-20 齐鲁天和惠世制药有限公司 Penam iodide, preparation and use thereof
RU2016115906A (en) 2009-06-10 2018-11-28 Текфилдз Байокем Ко., Лтд. COMPOSITIONS OR MEDICATIONS OF ANTIMO-MICROBIAL AND RELATIVE ANTIMO-MICROBIAL COMPOUNDS Possessing a High Penetrating Ability
AU2010281439A1 (en) 2009-07-28 2012-03-15 Anacor Pharmaceuticals, Inc. Trisubstituted boron-containing molecules
CN101696212B (en) 2009-08-28 2012-01-11 海南美大制药有限公司 High-purity tazobactam sodium compound
EP2536408A1 (en) 2010-02-16 2012-12-26 Wockhardt Research Centre Efflux pump inhibitors
EP2544528A1 (en) 2010-03-09 2013-01-16 Merck Sharp & Dohme Corp. FtsZ INHIBITORS AS POTENTIATORS OF BETA-LACTAM ANTIBIOTICS AGAINST METHICILLIN-RESISTANT STAPHYLOCOCCUS
JP2013523830A (en) 2010-04-06 2013-06-17 プリサイエンス ラブス, エルエルシー Methods of treatment with 3-bromopyruvate and other selective inhibitors of ATP production
CN102020663B (en) 2010-11-24 2013-04-03 山东鑫泉医药有限公司 Tazobactam synthesis method
CN102382123A (en) 2011-03-10 2012-03-21 海南美好西林生物制药有限公司 Preparation method of tazobactam sodium
BR112013032770A2 (en) 2011-07-26 2017-02-07 Wockhardt Ltd pharmaceutical compositions comprising beta-lactam antibiotic, sulbactam and beta-lactamase inhibitor
CN103619843B (en) 2011-08-27 2017-01-11 沃克哈特有限公司 1,6-diazabicyclo[3,2,1]octan-7-one derivatives and use thereof in treating bacterial infections
US20130065874A1 (en) 2011-09-09 2013-03-14 Cubist Pharmaceuticals, Inc. Methods for treating intrapulmonary infections
TW201343645A (en) 2012-03-30 2013-11-01 Cubist Pharm Inc 1,3,4-oxadiazole and 1,3,4-thiadiazole β-lactamase inhibitors
EP2831069B1 (en) 2012-03-30 2017-07-12 Merck Sharp & Dohme Corp. Isoxazole beta-lactamase inhibitors
US8969570B2 (en) 2012-03-30 2015-03-03 Cubist Pharmaceuticals, Inc. Beta-lactamase inhibitors
US8916709B2 (en) 2012-03-30 2014-12-23 Cubist Pharmaceuticals, Inc. 1,2,4-oxadiazole and 1,2,4-thiadiazole β-lactamase inhibitors
US8809314B1 (en) 2012-09-07 2014-08-19 Cubist Pharmacueticals, Inc. Cephalosporin compound
US8476425B1 (en) 2012-09-27 2013-07-02 Cubist Pharmaceuticals, Inc. Tazobactam arginine compositions
RU2671485C2 (en) 2012-09-27 2018-11-01 Мерк Шарп И Доум Корп. Tazobactam arginine antibiotic compositions
US9872906B2 (en) * 2013-03-15 2018-01-23 Merck Sharp & Dohme Corp. Ceftolozane antibiotic compositions
CA2906151A1 (en) 2013-03-15 2014-09-18 Merck Sharp & Dohme Corp. Ceftolozane antibiotic compositions
US20140274997A1 (en) 2013-03-15 2014-09-18 Cubist Pharmaceuticals, Inc. Cephalosporin pharmaceutical compositions
US20150072968A1 (en) * 2013-09-09 2015-03-12 Calixa Therapeutics, Inc. Treating Infections with Ceftolozane/Tazobactam in Subjects Having Impaired Renal Function
US20160228448A1 (en) * 2013-09-27 2016-08-11 Merck Sharp & Dohme Corp. Solid Forms of Ceftolozane

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8906898B1 (en) * 2013-09-27 2014-12-09 Calixa Therapeutics, Inc. Solid forms of ceftolozane

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9724353B2 (en) 2011-09-09 2017-08-08 Merck Sharp & Dohme Corp. Methods for treating intrapulmonary infections
US10028963B2 (en) 2011-09-09 2018-07-24 Merck Sharp & Dohme Corp. Methods for treating intrapulmonary infections
US8968753B2 (en) 2013-03-15 2015-03-03 Calixa Therapeutics, Inc. Ceftolozane-tazobactam pharmaceutical compositions
US9044485B2 (en) 2013-03-15 2015-06-02 Calixa Therapeutics, Inc. Ceftolozane antibiotic compositions
US9320740B2 (en) 2013-03-15 2016-04-26 Merck Sharp & Dohme Corp. Ceftolozane-tazobactam pharmaceutical compositions
US9872906B2 (en) 2013-03-15 2018-01-23 Merck Sharp & Dohme Corp. Ceftolozane antibiotic compositions
US9925196B2 (en) 2013-03-15 2018-03-27 Merck Sharp & Dohme Corp. Ceftolozane-tazobactam pharmaceutical compositions
US10420841B2 (en) 2013-03-15 2019-09-24 Merck, Sharp & Dohme Corp. Ceftolozane antibiotic compositions
US11278622B2 (en) 2013-03-15 2022-03-22 Merck Sharp & Dohme Corp. Ceftolozane antibiotic compositions
US10376496B2 (en) 2013-09-09 2019-08-13 Merck, Sharp & Dohme Corp. Treating infections with ceftolozane/tazobactam in subjects having impaired renal function
US10933053B2 (en) 2013-09-09 2021-03-02 Merck Sharp & Dohme Corp. Treating infections with ceftolozane/tazobactam in subjects having impaired renal function

Also Published As

Publication number Publication date
US20140274990A1 (en) 2014-09-18
US20150045336A1 (en) 2015-02-12
US20150150883A1 (en) 2015-06-04
US8968753B2 (en) 2015-03-03
US20140274998A1 (en) 2014-09-18
US20180169106A1 (en) 2018-06-21
US9925196B2 (en) 2018-03-27
US20160193221A1 (en) 2016-07-07
US20140274997A1 (en) 2014-09-18
US20140274993A1 (en) 2014-09-18
US9320740B2 (en) 2016-04-26
US20140274991A1 (en) 2014-09-18
US20140275000A1 (en) 2014-09-18
US20140262868A1 (en) 2014-09-18

Similar Documents

Publication Publication Date Title
JP7177314B2 (en) Ceftolozane antibiotic composition
US11278622B2 (en) Ceftolozane antibiotic compositions
US20140274996A1 (en) Tazobactam and ceftolozane antibiotic compositions
AU2015200599B2 (en) Ceftolozane Antibiotic Compositions
NZ711823B2 (en) Ceftolozane antibiotic compositions

Legal Events

Date Code Title Description
AS Assignment

Owner name: CUBIST PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DAMOUR, NICOLE MILLER;TERRACCIANO, JOSEPH;REEL/FRAME:032884/0414

Effective date: 20140507

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: CALIXA THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CUBIST PHARMACEUTICALS, INC.;REEL/FRAME:037190/0908

Effective date: 20140603

AS Assignment

Owner name: MERCK SHARP & DOHME CORP., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CALIXA THERAPEUTICS, INC.;REEL/FRAME:037198/0658

Effective date: 20150610