US20130065874A1 - Methods for treating intrapulmonary infections - Google Patents

Methods for treating intrapulmonary infections Download PDF

Info

Publication number
US20130065874A1
US20130065874A1 US13/607,138 US201213607138A US2013065874A1 US 20130065874 A1 US20130065874 A1 US 20130065874A1 US 201213607138 A US201213607138 A US 201213607138A US 2013065874 A1 US2013065874 A1 US 2013065874A1
Authority
US
United States
Prior art keywords
ceftolozane
tazobactam
infection
pharmaceutical composition
elf
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
US13/607,138
Inventor
Gurudatt A. Chandorkar
Jennifer A. Huntington
Tara Parsons
Obiamiwe C. Umeh
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.)
Calixa Therapeutics Inc
Original Assignee
Cubist Pharmaceuticals LLC
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 Cubist Pharmaceuticals LLC filed Critical Cubist Pharmaceuticals LLC
Priority to US13/607,138 priority Critical patent/US20130065874A1/en
Assigned to ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT reassignment ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT SECURITY AGREEMENT Assignors: ADOLOR CORPORATION, CALIXA THERAPEUTICS, INC., CUBIST PHARMACEUTICALS HOLDINGS, INC., CUBIST PHARMACEUTICALS U.S., CUBIST PHARMACEUTICALS, INC.
Publication of US20130065874A1 publication Critical patent/US20130065874A1/en
Assigned to CUBIST PHARMACEUTICALS, INC. reassignment CUBIST PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHANDORKAR, GURUDATT A., HUNTINGTON, JENNIFER A., PARSONS, Tara, UMEH, OBIAMIWE C.
Priority to US14/512,608 priority patent/US9724353B2/en
Assigned to CALIXA THERAPEUTICS, INC. reassignment CALIXA THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CUBIST PHARMACEUTICALS, INC.
Priority to US14/550,125 priority patent/US20150080360A1/en
Assigned to ADOLOR CORPORATION, CUBIST PHARMACEUTICALS, INC., CALIXA THERAPEUTICS, INC. reassignment ADOLOR CORPORATION RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT
Priority to US15/629,360 priority patent/US10028963B2/en
Priority to US16/016,524 priority patent/US20180296567A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic 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
    • A61K31/33Heterocyclic 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This disclosure relates to the treatment of intrapulmonary bacterial infections, including the treatment of nosocomial pneumonia infections, with a cephalosporin.
  • cephalosporin (6R,7R)-3-[5-Amino-4-[3-(2-aminoethyl)ureidol-1-methyl-1H-pyrazol-2-ium-2-ylmethyl]-7-[2-(5-amino-1,2,4-thiadiazol-3-yl)-2-RZ)-1-carboxy-1-methylethoxyimino]acetamido]-3-cephem-4-carboxylic acid (also referred to as “CXA-101” and previously designated FR264205) is an antibacterial agent.
  • CXA-101 can be provided as the compound shown in FIG. 1 .
  • CXA-101 The antibacterial activity of CXA-101 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.
  • CXA-101 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.
  • the combination of CXA-101 and tazobactam in a 2:1 weight ratio is an antibiotic pharmaceutical composition (“CXA-201”) 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.
  • Intrapulmonary infections such as nosocomial pneumonia, remain a major cause of morbidity and mortality, especially infections caused by drug resistant pathogens such as P. aeruginosa.
  • One challenge in treating intrapulmonary infections with systemic administration of an antibiotic is determining the antibiotic dose that will provide a therapeutically safe and effective concentration of the antibiotic at the site of an infection on the mucosal side of the bronchi in the lung (i.e., in the bronchial secretions).
  • Many antibiotics diffuse poorly from the bloodstream across the bronchi [e.g., Pennington, J.
  • ELF epithelial lining fluid
  • the concentration of antibiotics measured in the ELF of the lung has varied widely.
  • the reported penetration ratio of telavancin in healthy human volunteers ranges widely between 0.43 and 1.24 (Lodise, Gottokd, Drusano, 2008 Antimicrobial Agents and Chemotherapy).
  • predicting the penetration of a drug into the ELF a priori based on the structure, molecular weight, size and solubility is difficult due to the limited data available on the effect of physicochemical properties on the lung penetration of drugs.
  • the efficacy of a particular drug in treating intrapulmonary infections cannot be predicted solely on the basis of data, such as in vitro data relating to the activity of that drug against a particular bacterium, which does not give any indication as whether the drug will accumulate at a therapeutically safe and effective concentration at the site of an infection on the mucosal side of the bronchi in the lung (i.e., in the bronchial secretions).
  • data such as in vitro data relating to the activity of that drug against a particular bacterium, which does not give any indication as whether the drug will accumulate at a therapeutically safe and effective concentration at the site of an infection on the mucosal side of the bronchi in the lung (i.e., in the bronchial secretions).
  • tigicycline a glycylcycline antimicrobial
  • aeruginosa aeruginosa
  • it has been approved by the FDA for the treatment of complicated skin and skin structure infections, complicated intra-abdominal infections, and community acquired pneumonia.
  • tigicycline is not approved for the treatment of nosocomial pneumonia, in view of an increased mortality risk associated with the use of tigicycline compared to other drugs in patients treated for nosocomial pneumonia.
  • the present invention provides methods for treating intrapulmonary infections, including nosocomial pneumonia, with systemic administration of a pharmaceutical composition comprising ceftolozane.
  • the invention is based in part on results from a human clinical study designed to assess the ELF penetration of CXA-201 in comparison to piperacillin/tazobactam, indicated for the treatment of nosocomial pneumonia.
  • the study described herein quantified the penetration of CXA-201 into the lung, as measured by the ratio of area under the concentration-time curve (AUC) in epithelial lining fluid (ELF) to AUC in plasma (AUC(ELF)/AUC(plasma) ratio).
  • CXA-201 penetrated well into the ELF of healthy volunteers compared to piperacillin/tazobactam, an agent widely used for treatment of lower respiratory infections.
  • the intrapulmonary pharmacokinetics measured in the study supports the use of CXA-201 as a parenteral (e.g., intravenous) antibiotic for treatment of intrapulmonary infections, such as nosocomial pneumonia or other lower respiratory tract infections.
  • FIG. 1 is the chemical structure of a salt of ceftolozane hydrogen sulfate salt.
  • FIG. 2A is a graph showing the ELF Concentration vs. Time Profile for ceftolozane hydrogen sulfate salt (Median and Range) for CXA-201.
  • FIG. 2B is a graph showing the ELF Concentration vs. Time Profile for Tazobactam (Median and Range) for CXA-201.
  • FIG. 3A is a graph showing the (Comparative) ELF Concentration vs. Time Profile for Piperacillin (Median and Range) for a piperacillin/tazobactam comparator (ZOSYN®).
  • FIG. 3B is a graph showing the (Comparative) ELF Concentration vs. Time Profile for Tazobactam (Median and Range) for a piperacillin/tazobactam comparator (ZOSYN®).
  • FIGS. 4A and 4B are synthetic schemes for preparing ceftolozane hydrogen sulfate salt.
  • ceftolozane means CXA-101 in a free-base or salt form, preferably a hydrogen sulfate form (illustrated in FIG. 1 ).
  • ceftolozane is CXA-101 in its free-base form.
  • ceftolozane is CXA-101 in its salt form, preferably a hydrogen sulfate form.
  • ceftolozane (in free base or salt form, preferably hydrogen sulfate form) and tazobactam are in a 2:1 (ceftolozane:tazobactam) weight ratio.
  • methods of treating intrapulmonary infections, including nosocomial pneumonia with systemic administration of a pharmaceutical composition comprising ceftolozane hydrogen sulfate and tazobactam in a 2:1 weight ratio.
  • ceftolozane hydrogen sulfate and tazobactam in a 2:1 weight ratio.
  • the combination of ceftolozane hydrogen sulfate and tazobactam in a 2:1 weight ratio is referred to herein and in the examples as “CXA-201.”
  • the invention provides a method of treating an intrapulmonary infection comprising administering a therapeutically effective amount of a pharmaceutical composition comprising ceftolozane.
  • the method may comprise administering a pharmaceutical composition comprising ceftolozane in combination with tazobactam.
  • the invention provides a method of treating an intrapulmonary infection comprising the step of intravenously administering about every 8 hours to a subject in need thereof a pharmaceutical composition comprising 3.0 g of ceftolozane.
  • the method may comprise administering a pharmaceutical composition comprising ceftolozane in combination with tazobactam.
  • the method comprises administering CXA-201 and the infection comprises Gram-negative bacteria.
  • the invention provides a method of treating an intrapulmonary infection comprising the step of intravenously administering every 8 hours to a subject in need thereof a pharmaceutical composition comprising 3.0 g of ceftolozane.
  • the invention provides a method of providing tazobactam or ceftolozane in the epithelial lining fluid of a subject in an amount effective to treat an intrapulmonary infection, comprising the step of intravenously administering to the subject a pharmaceutical composition comprising ceftolozane.
  • the method may comprise administering a pharmaceutical composition further comprising tazobactam, optionally wherein the pharmaceutical composition is CXA-201.
  • the method may comprise administering about 1.5 g of ceftolozane and tazobactam in total every 8 hours.
  • the amount of the ceftolozane in the ELF of the subject effective to treat an intrapulmonary infection is at least about 8 ⁇ g/ml.
  • the ELF concentration of ceftolozane in the ELF may reach at least about 8 ⁇ g/ml after administration of the pharmaceutical composition.
  • the subject is typically a human having, or believed to be at risk of having, nosocomial pneumonia.
  • the subject (or patient) may, in some embodiments, have ventilator acquired pneumonia or hospital acquired pneumonia.
  • the invention provides the use of ceftolozane in the manufacture of a medicament for the treatment of an intrapulmonary infection comprising administering a therapeutically effective amount of a pharmaceutical composition comprising the ceftolozane.
  • the use may comprise administering the pharmaceutical composition comprising ceftolozane, in combination with tazobactam.
  • the invention provides the use of ceftolozane in the manufacture of a medicament for the treatment of an intrapulmonary infection comprising intravenously administering a pharmaceutical composition comprising 3.0 g of the ceftolozane every 8 hours to a subject in need thereof.
  • the use may comprise administering the pharmaceutical composition comprising ceftolozane in combination with tazobactam.
  • the use comprises administering ceftolozane and tazobactam and the infection comprises Gram-negative bacteria.
  • the invention provides the use of ceftolozane in the manufacture of a medicament for the treatment of an intrapulmonary infection comprising intravenously administering a pharmaceutical composition comprising the ceftolozane, wherein tazobactam or ceftolozane is provided in the epithelial lining fluid of a subject in an amount effective to treat the intrapulmonary infection.
  • the use may comprise administering a pharmaceutical composition further comprising tazobactam, optionally wherein the pharmaceutical composition is CXA-201.
  • the use may comprise administering about 1.5 g of ceftolozane and tazobactam every 8 hours.
  • the amount of the ceftolozane in the ELF of the subject effective to treat an intrapulmonary infection is at least about 8 ⁇ g/ml.
  • the ELF concentration of ceftolozane in the ELF may reach at least about 8 ⁇ g/ml after administration of the pharmaceutical composition.
  • the subject is typically a human having, or believed to be at risk of having, nosocomial pneumonia.
  • the subject (or patient) may, in some embodiments, have ventilator acquired pneumonia or hospital acquired pneumonia.
  • the pharmaceutical composition may be administered parenterally.
  • the pharmaceutical composition may be administered intravenously.
  • the pharmaceutical composition is intravenously administered about once every 8 hours as an infusion.
  • the pharmaceutical composition may be intravenously administered as a 60-minute infusion.
  • the intrapulmonary infection may be an infection in the lung.
  • the intrapulmonary infection may be pneumonia.
  • the intrapulmonary infection is nosocomial pneumonia.
  • the intrapulmonary infection may comprise Pseudomonas aeruginosa, Enterobacteriaceae, or a combination thereof.
  • the intrapulmonary infection comprises Pseudomonas aeruginosa.
  • the intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for CXA-201 of ⁇ 8 ⁇ g/ml.
  • the intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane of ⁇ 8 ⁇ g/ml.
  • the invention provides ceftolozane, for use in a method of treating an intrapulmonary infection.
  • the ceftolozane is parenterally administered.
  • the ceftolozane is intravenously administered.
  • the ceftolozane is administered about once every 8 hours as an infusion.
  • the ceftolozane is intravenously administered as a 60-minute infusion.
  • the ceftolozane is for use in a method of treating an intrapulmonary infection wherein the intrapulmonary infection comprises an infection in the lung.
  • the intrapulmonary infection may be pneumonia.
  • the ceftolozane is for use in a method of treating nosocomial pneumonia.
  • the intrapulmonary infection may comprise Pseudomonas aeruginosa, Enterobacteriaceae, or a combination thereof.
  • the intrapulmonary infection comprises Pseudomonas aeruginosa.
  • the intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane and tazobactam of ⁇ 8 ⁇ g/ml.
  • the intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane of ⁇ 8 ⁇ g/ml.
  • the invention also provides ceftolozane, for use in a method of treating an intrapulmonary infection, comprising administration of ceftolozane in combination with tazobactam.
  • the ceftolozane and/or tazobactam is parenterally administered.
  • the ceftolozane and/or tazobactam is intravenously administered.
  • the ceftolozane and/or tazobactam is administered about once every 8 hours as an infusion.
  • the ceftolozane and/or tazobactam is intravenously administered as a 60-minute infusion.
  • both the ceftolozane and tazobactam are parenterally administered. In another embodiment, both the ceftolozane and tazobactam are intravenously administered. In some embodiments, both the ceftolozane and tazobactam are administered about once every 8 hours as an infusion. In some embodiments, both the ceftolozane and tazobactam are intravenously administered as a 60-minute infusion. In one embodiment, the ceftolozane is for use in a method of treating an intrapulmonary infection wherein the intrapulmonary infection comprises an infection in the lung. The intrapulmonary infection may be pneumonia.
  • the ceftolozane is for use in a method of treating nosocomial pneumonia.
  • the intrapulmonary infection may comprise Pseudomonas aeruginosa, Enterobacteriaceae, or a combination thereof.
  • the intrapulmonary infection comprises Pseudomonas aeruginosa.
  • the intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane and tazobactam of ⁇ 8 ⁇ g/ml.
  • the intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane of ⁇ 8 ⁇ g/ml.
  • the invention provides tazobactam, for use in a method of treating an intrapulmonary infection, comprising administration of tazobactam in combination with ceftolozane.
  • the tazobactam and/or ceftolozane is parenterally administered.
  • the tazobactam and/or ceftolozane is intravenously administered.
  • the tazobactam and/or ceftolozane is administered about once every 8 hours as an infusion.
  • the tazobactam and/or ceftolozane is intravenously administered as a 60-minute infusion.
  • both the tazobactam and ceftolozane are parenterally administered. In another embodiment, both the tazobactam and ceftolozane are intravenously administered. In another embodiment, both the tazobactam and ceftolozane are administered about once every 8 hours as an infusion. In another embodiments, both the tazobactam and ceftolozane are intravenously administered as a 60-minute infusion.
  • the tazobactam is for use in a method of treating an intrapulmonary infection wherein the intrapulmonary infection comprises an infection in the lung.
  • the intrapulmonary infection may be pneumonia.
  • the tazobactam is for use in a method of treating nosocomial pneumonia.
  • the intrapulmonary infection may comprise Pseudomonas aeruginosa, Enterobacteriaceae, or a combination thereof.
  • the intrapulmonary infection comprises Pseudomonas aeruginosa.
  • the intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane and tazobactam of ⁇ 8 ⁇ g/ml.
  • the intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane of ⁇ 8 ⁇ g/ml.
  • the invention provides ceftolozane and tazobactam, as a combined preparation for simultaneous, separate or sequential use in a method of treating an intrapulmonary infection.
  • the ceftolozane and tazobactam are parenterally administered.
  • the ceftolozane and tazobactam are intravenously administered.
  • the ceftolozane and tazobactam are administered about once every 8 hours as an infusion.
  • the ceftolozane and tazobactam are intravenously administered as a 60-minute infusion.
  • the ceftolozane and tazobactam are for use in a method of treating an intrapulmonary infection wherein the intrapulmonary infection comprises an infection in the lung.
  • the intrapulmonary infection may be pneumonia.
  • the ceftolozane and tazobactam are for use in a method of treating nosocomial pneumonia.
  • the intrapulmonary infection may comprise Pseudomonas aeruginosa, Enterobacteriaceae, or a combination thereof.
  • the intrapulmonary infection comprises Pseudomonas aeruginosa.
  • the intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane and tazobactam of ⁇ 8 ⁇ g/ml.
  • the intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane of ⁇ 8 ⁇ g/ml.
  • the invention provides ceftolozane for use in a method of providing tazobactam or ceftolozane in the epithelial lining fluid of a subject in an amount effective to treat an intrapulmonary infection, comprising the step of intravenously administering ceftolozane.
  • ceftolozane is administered in combination with tazobactam.
  • CXA-201 is administered.
  • about 1.5 g of ceftolozane and tazobactam is administered every 8 hours.
  • the amount of the ceftolozane in the ELF of the subject effective to treat an intrapulmonary infection is at least about 8 ⁇ g/ml.
  • the ELF concentration of ceftolozane in the ELF may reach at least about 8 ⁇ g/ml after administration of the ceftolozane.
  • the subject is typically a human having, or believed to be at risk of having, nosocomial pneumonia.
  • the subject (or patient) may, in some embodiments, have ventilator acquired pneumonia or hospital acquired pneumonia.
  • the safe and effective treatment of intrapulmonary infection with CXA-201 includes administration of an amount of the CXA-201 selected to provide a therapeutically effective dose of the CXA-201 antibiotic in the epithelial lining fluid (ELF).
  • ELF epithelial lining fluid
  • the penetration of CXA-201 into the ELF compared to a piperacillin/tazobactam comparator was assessed in a Phase 1 clinical study in healthy adult volunteers.
  • the piperacillin/tazobactam comparator contained piperacillin/tazobactam in an 8:1 weight ratio with a total of 2.79 mEq of sodium per gram of piperacillin, FDA approved under the tradename ZOSYN® (“Zosyn”).
  • the study results evaluate the penetration of intravenously administered CXA-201 into healthy human lungs, as measured by the ratio of area under the concentration-time curve (AUC) in epithelial lining fluid (ELF) to AUC in plasma (AUC(ELF)/AUC(plasma)ratio).
  • AUC area under the concentration-time curve
  • a 4.5 g amount of piperacillin/tazobactam incorporates the same dose of tazobactam (0.5 g) as 1.5 g of CXA-201.
  • a multiple-dose regimen was used in this study to ensure that the concentrations of the analytes reached steady-state in both plasma and ELF prior to assessment. Healthy volunteers were chosen to standardize the subject population and minimize the variability associated with using actively ill patients. The objectives of the study included: (1) determination and comparison of the ELF to plasma concentration ratios of multiple-doses of intravenous CXA-201 compared to piperacillin/tazobactam in healthy adult volunteers, and (2) assessment of the safety and tolerability of multiple-doses of intravenous CXA-201 in healthy adult volunteers.
  • BAL bronchoalveolar lavage
  • Plasma and BAL datapoints were used to construct one concentration-time profile in the ELF using the mean concentrations at each time point.
  • a single ELF sample was obtained by bronchoalveolar lavage (BAL) from each healthy volunteer at one of 5 scheduled time points (5 subjects/time point/treatment group).
  • the ELF to plasma concentrations of multiple-doses was determined.
  • Serial plasma samples were collected pre- and post-treatment over a 6-hour (piperacillin/tazobactam) or 8-hour (CXA-201) time period.
  • Urea levels in the plasma and BAL were used to calculate the ELF drug concentrations (see Table 1).
  • Pharmacokinetic parameters for ELF were calculated by non-compartmental analysis using the mean concentrations at each time point.
  • the intrapulmonary penetration of CXA-201 into the ELF was determined by dividing the ELF AUC 0 -t by mean plasma AUC 0 -t.
  • the concentration of CXA-201 and piperacillin/tazobactam in ELF were estimated from the concentration of drug in BAL fluid, the volume of BAL fluid collected, and the ratio of urea concentration in BAL fluid to that in plasma. Calculation of ELF volume was determined by the urea dilution method, using urea as an endogenous marker of ELF recovered by BAL. Concentration of CXA-201 and piperacillin/tazobactam in ELF was estimated from the concentration of drug in BAL fluid, the volume of BAL fluid collected, and the ratio of urea concentration in BAL fluid to that in plasma. The following formulas represent these calculations:
  • CXA-201 (CXA/T) [CXA/T] BAL ⁇ V BAL /V ELF
  • [CXAM BAL is the concentration of CXA-201 in BAL fluid; V BAL is the volume of aspirated BAL fluid (total); V ELF is V BAL ⁇ [urea] BAL /[urea] plasma , where [urea] BAL is the concentration of urea in the BAL fluid (supernatant) and [urea] plasma is the concentration of urea in the plasma specimens.
  • [PIP/T] BAL is the concentration of piperacillin/tazobactam in BAL fluid
  • V BAL is the volume of aspirated BAL fluid (total)
  • V ELF is V BAL ⁇ [urea] BAL /[urea] plasma , where [urea] BAL is the concentration of urea in the BAL fluid (supernatant) and [urea] plasma is the concentration of urea in the plasma specimens.
  • the ELF concentration vs. time profiles for ceftolozane and tazobactam components of CXA-201 are shown in FIGS. 2A and 2B , respectively.
  • Comparative data showing the ELF concentration vs. time profiles for piperacillin and tazobactam components of the comparator drug are shown in FIGS. 3A and 3B , respectively.
  • the ELF to plasma penetration ratios are shown in Table 2.
  • the PK parameters were determined by non-compartmental PK analysis. PHOENIX® WinNonlin v 6.1 (PHARSIGHT®, Mountain View, Calif.) was used for the derivation of all PK individual measures for each subject.
  • the PK parameters for ELF were calculated by taking the mean concentrations of the 5 subjects at each time point and constructing a single profile over the duration of sampling. In the event that the urea concentrations determined in plasma or ELF were below quantifiable limits, thereby providing only an estimate of concentration, those values were not used in the calculation of mean concentration at that time point.
  • the ceftolozane, piperacillin, and tazobactam PK parameters that were computed in plasma and ELF were:
  • the ELF/plasma AUC ratio for the ceftolozane component of CXA-201 was 0.48, compared to 0.26 for the piperacillin component of the comparator drug (piperacillin/tazobactam).
  • the ELF/plasma AUC ratio for tazobactam was 0.44 and 0.54 when given as part of CXA-201 and piperacillin/tazobactam, respectively.
  • the ELF concentrations of ceftolozane exceeded 8 ⁇ g/mL for 60% of the 8-hour dosing interval.
  • the plasma and ELF concentrations of tazobactam when given as piperacillin/tazobactam was approximately 2-fold higher than when an equivalent dose was given as CXA-201.
  • ceftolozane and tazobactam i.e., administered as CXA-201 penetrated well into the ELF of healthy volunteers compared to piperacillin/tazobactam, an agent widely used for treatment of lower respiratory infections.
  • CXA-201's intrapulmonary pharmacokinetics support use of CXA-201 as a parenteral (e.g., intravenous) antibiotic for treatment of lower respiratory tract infections, including infections caused by pathogens with minimum inhibitory concentrations of ⁇ 8 ⁇ g/ml.
  • the concentrations of ceftolozane in ELF exceeded 8 ⁇ g/mL, a concentration that inhibits 99% of P. aeruginosa, for approximately 60% of the 8-hour dosing interval for the CXA-201 regimen of 1.5 grams every eight hours as a 60 minute infusion.
  • TEAEs treatment-emergent AEs
  • a Monte Carlo simulation was performed based on clinical trial data to predict an effective CXA-201 dose for treating nosocomial pneumonia using PHOENIX® NLME (PHARSIGHT®, Mountain View, Calif.) software, a tool for data processing and modeling for population PK/PD analysis.
  • a population pharmacokinetic (PK) model was developed using the CXA-201 plasma concentration versus time data from a previously conducted Phase 2 study in patients with complicated intra abdominal infections. Estimates of clearance and volume of distribution along with the associated inter-individual variability were obtained from these analyses.
  • the outputs from the PK population model served as inputs for a clinical trial simulation performed using PHARSIGHT® Trial Simulator (PHARSIGHT®) software, a tool for defining and testing interactive drug models, exploring and communicating study design attributes, and performing statistical and sensitivity analysis through graphical and statistical summaries.
  • PHARSIGHT® PHARSIGHT® Trial Simulator
  • an ELF/Plasma AUC ratio of 0.48 for ceftolozane (modeled as a numerical range of 0.25-0.65) calculated from the ceftolozane ELF study mentioned above was used to generate a random/Plasma AUC ratio from the range 0.25-0.65 for each simulated patient. This range reflects a conservative estimate of the potential distribution in a patient population.
  • the model simulated plasma and ELF concentration of CXA-201 versus time profiles for 1,000 hypothetical clinical trial patients with nosocomial pneumonia.
  • the model evaluated the probability of clinical success of the 3.0 g every 8 hour (q8h) dose of CXA-201 against three key pathogens in nosocomial pneumonia.
  • the MIC distribution for these pathogens was imputed from 2008 United States surveillance data.
  • Clinical success was defined as the achievement of an ELF or plasma concentration of ceftolozane higher than the MIC(s) of the lower respiratory pathogen(s) for a given patient.
  • the relavent PK/PD driver for CXA-201 is the percentage of time above MIC during the dosing interval.
  • the target is to achieve concentrations that exceed the MIC of the pathogen for 45-50% of the time between each q8H dose.
  • a threshold of 50% time above the minimum inhibitory concentration [T>MIC] on Day 7 of treatment was used.
  • Plasma and ELF concentrations were estimated at 15 time-points post-administration on Day 7 when dosed every 8 hours. The results of these simulations are shown in Table 5.
  • CXA-201 can be prepared by combining ceftolozane and tazobactam in a 2:1 weight ratio.
  • CXA-201 can be obtained using methods described in U.S. Pat. No. 7,129,232 and Toda et al., “Synthesis and SAR of novel parenteral anti-pseudomonal cephalosporins: Discovery of FR264205, ” Bioorganic & Medicinal Chemistry Letters, 18, 4849-4852 (2008), incorporated herein by reference in its entirety.
  • ceftolozane can be obtained by the synthetic schemes of FIGS. 4A and 4B . Referring to FIGS. 4A and 4B .
  • ceftolozane can be performed via activation of the thiadiazolyl-oximinoacetic acid derivative (I) with methanesulfonyl chloride and K 2 CO 3 in DMA at 10° C., followed by coupling with the 7-aminocephem (II) by means of Et 3 N in cold EtOAc/H 2 O, affords amide (III) (1).
  • the antibacterial activity of the CXA-201 or other compounds can be measured by the minimum inhibitory concentrations (MIC) of the compounds against various bacteria measured by using the broth microdilution method performed according to Clinical and Laboratory Standards Institute (CLSI) guidelines with modifications as described below (CLSI guidelines can be derived from the CLSI document M7-A8 published in January 2009: “Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard-Eighth Edition”).
  • MIC minimum inhibitory concentrations
  • individual colonies can be isolated by streaking frozen glycerol material containing Staphylococcus or Pseudomonas spp. onto rich, non-selective, tryptic soy agar containing 5% sheep's blood (TSAB), and incubated at 37° C. for 18-24 hrs.
  • TSAB sheep's blood
  • CAMHB that contained compound concentrations ranging from 64-0.06 ⁇ g/mL in two-fold dilutions can also be added to the broth microdilution assay plates for a final volume 100 ⁇ L per well, therefore final culture OD 600 was approximately 0.001 and the final NaCl concentration for the MRSA strain was 2%.
  • Plates can be incubated for 18-20 hours at 37° C. with aeration (200 rpm). Following incubation, growth can be confirmed visually placing plates over a viewing apparatus (stand with a minor underneath) and then OD 600 can be measured using a SpectraMax 340PC384 plate reader (Molecular Devices, Sunnyvale, Calif.). Growth was defined as turbidity that could be detected with the naked eye or achieving minimum OD 600 of 0.1. MIC values were defined as the lowest concentration producing no visible turbidity.

Abstract

This disclosure relates to the treatment of intrapulmonary bacterial infections, including treatment of nosocomial pneumonia lung infections with pharmaceutical compositions containing the cephalosporin ceftolozane.

Description

    RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Application No. 61/532,914, filed Sep. 9, 2011, titled “Methods for Treating Intrapulmonary Infections,” and U.S. Provisional Application No. 61/657,386, filed Jun. 8, 2012, titled “Methods for Treating Intrapulmonary Infections.” The contents of any patents, patent applications, and references cited throughout this specification are hereby incorporated by reference in their entireties.
  • TECHNICAL FIELD
  • This disclosure relates to the treatment of intrapulmonary bacterial infections, including the treatment of nosocomial pneumonia infections, with a cephalosporin.
  • BACKGROUND
  • The cephalosporin (6R,7R)-3-[5-Amino-4-[3-(2-aminoethyl)ureidol-1-methyl-1H-pyrazol-2-ium-2-ylmethyl]-7-[2-(5-amino-1,2,4-thiadiazol-3-yl)-2-RZ)-1-carboxy-1-methylethoxyimino]acetamido]-3-cephem-4-carboxylic acid (also referred to as “CXA-101” and previously designated FR264205) is an antibacterial agent. CXA-101 can be provided as the compound shown in FIG. 1. The antibacterial activity of CXA-101 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. CXA-101 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. The combination of CXA-101 and tazobactam in a 2:1 weight ratio is an antibiotic pharmaceutical composition (“CXA-201”) 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.
  • Intrapulmonary infections, such as nosocomial pneumonia, remain a major cause of morbidity and mortality, especially infections caused by drug resistant pathogens such as P. aeruginosa. One challenge in treating intrapulmonary infections with systemic administration of an antibiotic is determining the antibiotic dose that will provide a therapeutically safe and effective concentration of the antibiotic at the site of an infection on the mucosal side of the bronchi in the lung (i.e., in the bronchial secretions). Many antibiotics diffuse poorly from the bloodstream across the bronchi [e.g., Pennington, J. E., “Penetration of antibiotics into respiratory secretions,” Rev Infect Dis 3(1):67-73 (1981)1, which can result in the administration of higher doses of antibiotic than would be prescribed for a truly systemic infection. Furthermore, the purulent sputum that characterizes infected patients tends to compromise the potency of many antibiotics (See e.g., Levy, J., et al., “Bioactivity of gentamicin in purulent sputum from patients with cystic fibrosis or bronchiectasis: comparison with activity in serum,” J Infect Dis 148(6):1069-76 (1983)). In some cases, the result is the prescription of large amounts of a systemically administered antibiotic to treat an intrapulmonary infection.
  • The efficacy of an antibiotic depends in part on the concentration of the drug at the site of action. Efficacy of antimicrobial therapy requires adequate antibiotic concentrations at the site of bacterial infection, and some authorities believe that epithelial lining fluid (ELF) concentrations are a reasonable surrogate for predicting effective concentrations for treating intrapulmonary infections such as pneumonia. For many antibiotics, clinical data correlating ELF concentrations to clinical outcome is unavailable and the clinical significance of differences in pulmonary penetration of antibiotics is unknown or poorly characterized. Few studies have quantified the penetration of β-lactam agents into the lung, as measured by the ratio of area under the concentration-time curve (AUC) in ELF to AUC in plasma (AUC(ELF)/AUC(plasma) ratio). For some published studies, the concentration of antibiotics measured in the ELF of the lung has varied widely. For example, the reported penetration ratio of telavancin in healthy human volunteers ranges widely between 0.43 and 1.24 (Lodise, Gottfreid, Drusano, 2008 Antimicrobial Agents and Chemotherapy). Thus, predicting the penetration of a drug into the ELF a priori, based on the structure, molecular weight, size and solubility is difficult due to the limited data available on the effect of physicochemical properties on the lung penetration of drugs.
  • Accordingly, the efficacy of a particular drug in treating intrapulmonary infections, in particular nosocomial pneumonia, cannot be predicted solely on the basis of data, such as in vitro data relating to the activity of that drug against a particular bacterium, which does not give any indication as whether the drug will accumulate at a therapeutically safe and effective concentration at the site of an infection on the mucosal side of the bronchi in the lung (i.e., in the bronchial secretions). For instance, tigicycline, a glycylcycline antimicrobial, has in vitro activity against many species of Gram-positive and Gram-negative bacteria, including P. aeruginosa, and it has been approved by the FDA for the treatment of complicated skin and skin structure infections, complicated intra-abdominal infections, and community acquired pneumonia. However, tigicycline is not approved for the treatment of nosocomial pneumonia, in view of an increased mortality risk associated with the use of tigicycline compared to other drugs in patients treated for nosocomial pneumonia.
  • SUMMARY
  • The present invention provides methods for treating intrapulmonary infections, including nosocomial pneumonia, with systemic administration of a pharmaceutical composition comprising ceftolozane. The invention is based in part on results from a human clinical study designed to assess the ELF penetration of CXA-201 in comparison to piperacillin/tazobactam, indicated for the treatment of nosocomial pneumonia. The study described herein quantified the penetration of CXA-201 into the lung, as measured by the ratio of area under the concentration-time curve (AUC) in epithelial lining fluid (ELF) to AUC in plasma (AUC(ELF)/AUC(plasma) ratio). The results of the study indicate that CXA-201 penetrated into the ELF of human patients, with a ceftolozane ELF/plasma AUC ratio of 0.48. The measured ELF concentrations of ceftolozane exceeded 8 μg/mL for 60% of the 8-hour dosing interval, a concentration that is predicted to inhibit 99% of Pseudomonas aeruginosa based on current surveillance data.
  • The study showed that CXA-201 penetrated well into the ELF of healthy volunteers compared to piperacillin/tazobactam, an agent widely used for treatment of lower respiratory infections. The intrapulmonary pharmacokinetics measured in the study supports the use of CXA-201 as a parenteral (e.g., intravenous) antibiotic for treatment of intrapulmonary infections, such as nosocomial pneumonia or other lower respiratory tract infections.
  • BRIEF DESCRIPTION OF DRAWINGS
  • FIG. 1 is the chemical structure of a salt of ceftolozane hydrogen sulfate salt.
  • FIG. 2A is a graph showing the ELF Concentration vs. Time Profile for ceftolozane hydrogen sulfate salt (Median and Range) for CXA-201.
  • FIG. 2B is a graph showing the ELF Concentration vs. Time Profile for Tazobactam (Median and Range) for CXA-201.
  • FIG. 3A is a graph showing the (Comparative) ELF Concentration vs. Time Profile for Piperacillin (Median and Range) for a piperacillin/tazobactam comparator (ZOSYN®).
  • FIG. 3B is a graph showing the (Comparative) ELF Concentration vs. Time Profile for Tazobactam (Median and Range) for a piperacillin/tazobactam comparator (ZOSYN®).
  • FIGS. 4A and 4B are synthetic schemes for preparing ceftolozane hydrogen sulfate salt.
  • DETAILED DESCRIPTION
  • The present disclosure relates to the treatment of intrapulmonary infections, including nosocomial pneumonia, with systemic administration of a pharmaceutical composition comprising ceftolozane, including the parenteral administration of a therapeutically effective amount of a pharmaceutical composition comprising ceftolozane and tazobactam. As used herein, the term “ceftolozane” means CXA-101 in a free-base or salt form, preferably a hydrogen sulfate form (illustrated in FIG. 1). In one embodiment, ceftolozane is CXA-101 in its free-base form. In another embodiment, ceftolozane is CXA-101 in its salt form, preferably a hydrogen sulfate form.
  • In a preferred embodiment, ceftolozane (in free base or salt form, preferably hydrogen sulfate form) and tazobactam are in a 2:1 (ceftolozane:tazobactam) weight ratio. In a particular embodiment, provided herein are methods of treating intrapulmonary infections, including nosocomial pneumonia, with systemic administration of a pharmaceutical composition comprising ceftolozane hydrogen sulfate and tazobactam in a 2:1 weight ratio. The combination of ceftolozane hydrogen sulfate and tazobactam in a 2:1 weight ratio is referred to herein and in the examples as “CXA-201.”
  • In one aspect, the invention provides a method of treating an intrapulmonary infection comprising administering a therapeutically effective amount of a pharmaceutical composition comprising ceftolozane. The method may comprise administering a pharmaceutical composition comprising ceftolozane in combination with tazobactam.
  • In another aspect, the invention provides a method of treating an intrapulmonary infection comprising the step of intravenously administering about every 8 hours to a subject in need thereof a pharmaceutical composition comprising 3.0 g of ceftolozane. The method may comprise administering a pharmaceutical composition comprising ceftolozane in combination with tazobactam. In one embodiment, the method comprises administering CXA-201 and the infection comprises Gram-negative bacteria. In another aspect, the invention provides a method of treating an intrapulmonary infection comprising the step of intravenously administering every 8 hours to a subject in need thereof a pharmaceutical composition comprising 3.0 g of ceftolozane.
  • In another aspect, the invention provides a method of providing tazobactam or ceftolozane in the epithelial lining fluid of a subject in an amount effective to treat an intrapulmonary infection, comprising the step of intravenously administering to the subject a pharmaceutical composition comprising ceftolozane. The method may comprise administering a pharmaceutical composition further comprising tazobactam, optionally wherein the pharmaceutical composition is CXA-201. The method may comprise administering about 1.5 g of ceftolozane and tazobactam in total every 8 hours. In one embodiment, the amount of the ceftolozane in the ELF of the subject effective to treat an intrapulmonary infection is at least about 8 μg/ml. The ELF concentration of ceftolozane in the ELF may reach at least about 8 μg/ml after administration of the pharmaceutical composition. The subject is typically a human having, or believed to be at risk of having, nosocomial pneumonia. The subject (or patient) may, in some embodiments, have ventilator acquired pneumonia or hospital acquired pneumonia.
  • In another aspect, the invention provides the use of ceftolozane in the manufacture of a medicament for the treatment of an intrapulmonary infection comprising administering a therapeutically effective amount of a pharmaceutical composition comprising the ceftolozane. The use may comprise administering the pharmaceutical composition comprising ceftolozane, in combination with tazobactam.
  • In another aspect, the invention provides the use of ceftolozane in the manufacture of a medicament for the treatment of an intrapulmonary infection comprising intravenously administering a pharmaceutical composition comprising 3.0 g of the ceftolozane every 8 hours to a subject in need thereof. The use may comprise administering the pharmaceutical composition comprising ceftolozane in combination with tazobactam. In one embodiment, the use comprises administering ceftolozane and tazobactam and the infection comprises Gram-negative bacteria.
  • In another aspect, the invention provides the use of ceftolozane in the manufacture of a medicament for the treatment of an intrapulmonary infection comprising intravenously administering a pharmaceutical composition comprising the ceftolozane, wherein tazobactam or ceftolozane is provided in the epithelial lining fluid of a subject in an amount effective to treat the intrapulmonary infection. The use may comprise administering a pharmaceutical composition further comprising tazobactam, optionally wherein the pharmaceutical composition is CXA-201. The use may comprise administering about 1.5 g of ceftolozane and tazobactam every 8 hours. In one embodiment, the amount of the ceftolozane in the ELF of the subject effective to treat an intrapulmonary infection is at least about 8 μg/ml. The ELF concentration of ceftolozane in the ELF may reach at least about 8 μg/ml after administration of the pharmaceutical composition. The subject is typically a human having, or believed to be at risk of having, nosocomial pneumonia. The subject (or patient) may, in some embodiments, have ventilator acquired pneumonia or hospital acquired pneumonia. In the methods and uses of the invention, the pharmaceutical composition may be administered parenterally. The pharmaceutical composition may be administered intravenously. In some embodiments, the pharmaceutical composition is intravenously administered about once every 8 hours as an infusion. The pharmaceutical composition may be intravenously administered as a 60-minute infusion.
  • In the methods and uses of the invention, the intrapulmonary infection may be an infection in the lung. The intrapulmonary infection may be pneumonia. In a preferred embodiment, the intrapulmonary infection is nosocomial pneumonia. The intrapulmonary infection may comprise Pseudomonas aeruginosa, Enterobacteriaceae, or a combination thereof. Typically, the intrapulmonary infection comprises Pseudomonas aeruginosa. The intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for CXA-201 of ≦8 μg/ml. The intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane of ≦8 μg/ml.
  • In another aspect, the invention provides ceftolozane, for use in a method of treating an intrapulmonary infection. In one embodiment, the ceftolozane is parenterally administered. Typically, the ceftolozane is intravenously administered. In some embodiments, the ceftolozane is administered about once every 8 hours as an infusion. In some embodiments, the ceftolozane is intravenously administered as a 60-minute infusion.
  • In one embodiment, the ceftolozane is for use in a method of treating an intrapulmonary infection wherein the intrapulmonary infection comprises an infection in the lung. The intrapulmonary infection may be pneumonia. In a preferred embodiment, the ceftolozane is for use in a method of treating nosocomial pneumonia. The intrapulmonary infection may comprise Pseudomonas aeruginosa, Enterobacteriaceae, or a combination thereof. Typically, the intrapulmonary infection comprises Pseudomonas aeruginosa. The intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane and tazobactam of ≦8 μg/ml. The intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane of ≦8 μg/ml.
  • The invention also provides ceftolozane, for use in a method of treating an intrapulmonary infection, comprising administration of ceftolozane in combination with tazobactam. In one embodiment, the ceftolozane and/or tazobactam is parenterally administered. Typically, the ceftolozane and/or tazobactam is intravenously administered. In some embodiments, the ceftolozane and/or tazobactam is administered about once every 8 hours as an infusion. In some embodiments, the ceftolozane and/or tazobactam is intravenously administered as a 60-minute infusion. In one embodiment, both the ceftolozane and tazobactam are parenterally administered. In another embodiment, both the ceftolozane and tazobactam are intravenously administered. In some embodiments, both the ceftolozane and tazobactam are administered about once every 8 hours as an infusion. In some embodiments, both the ceftolozane and tazobactam are intravenously administered as a 60-minute infusion. In one embodiment, the ceftolozane is for use in a method of treating an intrapulmonary infection wherein the intrapulmonary infection comprises an infection in the lung. The intrapulmonary infection may be pneumonia. In a preferred embodiment, the ceftolozane is for use in a method of treating nosocomial pneumonia. The intrapulmonary infection may comprise Pseudomonas aeruginosa, Enterobacteriaceae, or a combination thereof. Typically, the intrapulmonary infection comprises Pseudomonas aeruginosa. The intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane and tazobactam of ≦8 μg/ml. The intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane of ≦8 μg/ml.
  • In another aspect, the invention provides tazobactam, for use in a method of treating an intrapulmonary infection, comprising administration of tazobactam in combination with ceftolozane. In one embodiment, the tazobactam and/or ceftolozane is parenterally administered. Typically, the tazobactam and/or ceftolozane is intravenously administered. In some embodiments, the tazobactam and/or ceftolozane is administered about once every 8 hours as an infusion. In some embodiments, the tazobactam and/or ceftolozane is intravenously administered as a 60-minute infusion. In one embodiment, both the tazobactam and ceftolozane are parenterally administered. In another embodiment, both the tazobactam and ceftolozane are intravenously administered. In another embodiment, both the tazobactam and ceftolozane are administered about once every 8 hours as an infusion. In another embodiments, both the tazobactam and ceftolozane are intravenously administered as a 60-minute infusion.
  • In one embodiment, the tazobactam is for use in a method of treating an intrapulmonary infection wherein the intrapulmonary infection comprises an infection in the lung. The intrapulmonary infection may be pneumonia. In a preferred embodiment, the tazobactam is for use in a method of treating nosocomial pneumonia. The intrapulmonary infection may comprise Pseudomonas aeruginosa, Enterobacteriaceae, or a combination thereof. Typically, the intrapulmonary infection comprises Pseudomonas aeruginosa. The intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane and tazobactam of ≦8 μg/ml. The intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane of ≦8 μg/ml.
  • In another aspect, the invention provides ceftolozane and tazobactam, as a combined preparation for simultaneous, separate or sequential use in a method of treating an intrapulmonary infection. In one embodiment, the ceftolozane and tazobactam are parenterally administered. Typically, the ceftolozane and tazobactam are intravenously administered. In some embodiments, the ceftolozane and tazobactam are administered about once every 8 hours as an infusion. In some embodiments, the ceftolozane and tazobactam, are intravenously administered as a 60-minute infusion.
  • In one embodiment, the ceftolozane and tazobactam are for use in a method of treating an intrapulmonary infection wherein the intrapulmonary infection comprises an infection in the lung. The intrapulmonary infection may be pneumonia. In a preferred embodiment, the ceftolozane and tazobactam are for use in a method of treating nosocomial pneumonia. The intrapulmonary infection may comprise Pseudomonas aeruginosa, Enterobacteriaceae, or a combination thereof. Typically, the intrapulmonary infection comprises Pseudomonas aeruginosa. The intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane and tazobactam of ≦8 μg/ml. The intrapulmonary infection may comprise a pathogen with minimum inhibitory concentration for ceftolozane of ≦8 μg/ml.
  • In another aspect, the invention provides ceftolozane for use in a method of providing tazobactam or ceftolozane in the epithelial lining fluid of a subject in an amount effective to treat an intrapulmonary infection, comprising the step of intravenously administering ceftolozane. In some embodiments, ceftolozane is administered in combination with tazobactam. Preferably, CXA-201 is administered. In preferred embodiments, about 1.5 g of ceftolozane and tazobactam is administered every 8 hours. In one embodiment, the amount of the ceftolozane in the ELF of the subject effective to treat an intrapulmonary infection is at least about 8 μg/ml. The ELF concentration of ceftolozane in the ELF may reach at least about 8 μg/ml after administration of the ceftolozane. The subject is typically a human having, or believed to be at risk of having, nosocomial pneumonia. The subject (or patient) may, in some embodiments, have ventilator acquired pneumonia or hospital acquired pneumonia.
  • The safe and effective treatment of intrapulmonary infection with CXA-201 includes administration of an amount of the CXA-201 selected to provide a therapeutically effective dose of the CXA-201 antibiotic in the epithelial lining fluid (ELF). The penetration of CXA-201 into the ELF compared to a piperacillin/tazobactam comparator was assessed in a Phase 1 clinical study in healthy adult volunteers. The piperacillin/tazobactam comparator contained piperacillin/tazobactam in an 8:1 weight ratio with a total of 2.79 mEq of sodium per gram of piperacillin, FDA approved under the tradename ZOSYN® (“Zosyn”). The study results evaluate the penetration of intravenously administered CXA-201 into healthy human lungs, as measured by the ratio of area under the concentration-time curve (AUC) in epithelial lining fluid (ELF) to AUC in plasma (AUC(ELF)/AUC(plasma)ratio).
  • In the study, a 4.5 g amount of piperacillin/tazobactam incorporates the same dose of tazobactam (0.5 g) as 1.5 g of CXA-201. A multiple-dose regimen was used in this study to ensure that the concentrations of the analytes reached steady-state in both plasma and ELF prior to assessment. Healthy volunteers were chosen to standardize the subject population and minimize the variability associated with using actively ill patients. The objectives of the study included: (1) determination and comparison of the ELF to plasma concentration ratios of multiple-doses of intravenous CXA-201 compared to piperacillin/tazobactam in healthy adult volunteers, and (2) assessment of the safety and tolerability of multiple-doses of intravenous CXA-201 in healthy adult volunteers.
  • The study was a Phase 1 prospective, randomized (1:1), comparator controlled, open-label study of 50 healthy adult volunteers. Each healthy volunteer received 3 doses of either CXA-201(1.5 grams every 8 hours as a 60-minute infusion) or piperacillin/tazobactam (4.5 grams every 6 hours as a 30-minute infusion). Subjects received 3 doses of a study drug, underwent serial blood draws at planned plasma sampling timepoints, and underwent a single bronchoalveolar lavage (BAL) procedure at one of the scheduled timepoints (Table 1).
  • TABLE 1
    Plasma Sampling and BAL Timepoints
    Plasma Sampling Timepoints BAL Timepoints
    Intensive plasma sampling 5 subjects per timepoint per
    from all 25 subjects for treatment group; in hours from
    one dosing interval start of the third infusion
    CXA-201
    0 (pre-PK dose trough), 1, 2, 1, 2, 4, 6, 8 hours post start
    4, 6, 8 hours post start of of infusion of the third dose of
    infusion of the third dose of CXA 201
    CXA 201
    Piperacillin/tazobactam
    0 (pre-PK dose trough), 0.5, 1, 0.5, 1, 2, 4, 6 hours post start
    2, 4, 6 hours post start of of infusion of the third dose of
    infusion of the third dose of piperacillin/tazobactam
    piperacillin/tazobactam
  • A total of 51 subjects were enrolled; 25 in the CXA-201 group and 26 in the piperacillin/tazobactam group. Key Inclusion Criteria for the study were: (1) healthy adult male or non-pregnant females between 18 and 50 years, inclusive; (2) body mass index between 18.5 and 30; and (3) forced Expiratory Volume in 1 second (FEV1) ≧80%. Key Exclusion Criteria for the study were: (1) pregnancy or lactation; (2) clinically significant systemic disease or the existence of any surgical or medical condition that may have interfered with the distribution, metabolism, or excretion of CXA-201; (3) history of asthma or any restrictive or obstructive lung disease; (4) history of smoking or abuse of narcotics or alcohol; (5) positive test for human immunodeficiency virus, Hepatitis B surface antigen, or Hepatitis C antibodies; (6) any condition or situation where bronchoscopy was not advisable; and (7) impairment of renal function (CrCl<90 mL/min)
  • Determination of the ELF to Plasma Concentration Ratios of Multiple-Doses of Intravenous CXA-201 Compared to Piperacillin/Tazobactam in Healthy Adult Volunteers.
  • Plasma and BAL datapoints were used to construct one concentration-time profile in the ELF using the mean concentrations at each time point. After dosing, a single ELF sample was obtained by bronchoalveolar lavage (BAL) from each healthy volunteer at one of 5 scheduled time points (5 subjects/time point/treatment group). The ELF to plasma concentrations of multiple-doses was determined. Serial plasma samples were collected pre- and post-treatment over a 6-hour (piperacillin/tazobactam) or 8-hour (CXA-201) time period. Urea levels in the plasma and BAL were used to calculate the ELF drug concentrations (see Table 1). Pharmacokinetic parameters for ELF were calculated by non-compartmental analysis using the mean concentrations at each time point. The intrapulmonary penetration of CXA-201 into the ELF was determined by dividing the ELF AUC0-t by mean plasma AUC0-t.
  • The concentration of CXA-201 and piperacillin/tazobactam in ELF were estimated from the concentration of drug in BAL fluid, the volume of BAL fluid collected, and the ratio of urea concentration in BAL fluid to that in plasma. Calculation of ELF volume was determined by the urea dilution method, using urea as an endogenous marker of ELF recovered by BAL. Concentration of CXA-201 and piperacillin/tazobactam in ELF was estimated from the concentration of drug in BAL fluid, the volume of BAL fluid collected, and the ratio of urea concentration in BAL fluid to that in plasma. The following formulas represent these calculations:

  • CXA-201 (CXA/T)=[CXA/T]BAL ×V BAL /V ELF
  • [CXAMBAL is the concentration of CXA-201 in BAL fluid; VBAL is the volume of aspirated BAL fluid (total); VELF is VBAL×[urea]BAL/[urea]plasma, where [urea]BAL is the concentration of urea in the BAL fluid (supernatant) and [urea]plasma is the concentration of urea in the plasma specimens.

  • Piperacillin/tazobactam=[PIP/T]BAL ×V BAL /V ELF
  • [PIP/T]BAL is the concentration of piperacillin/tazobactam in BAL fluid; VBAL is the volume of aspirated BAL fluid (total); VELF is VBAL×[urea]BAL/[urea]plasma, where [urea]BAL is the concentration of urea in the BAL fluid (supernatant) and [urea]plasma is the concentration of urea in the plasma specimens.
  • No oral antibiotic therapy was permitted. Safety was monitored through the review of vital signs, laboratory and physical examinations and the occurrence of adverse events (AEs). Subjects who received three doses of study medication and had both BAL and plasma samples collected were included in the pharmacokinetic (PK) analysis population. All randomized subjects who received any dose (including partial doses) of study medication were included in the safety analysis population.
  • The results of the study (Table 2) indicate that CXA-201 penetrated well into ELF. The ceftolozane component of CXA-201 ELF/plasma AUC ratio was 0.48, compared to 0.26 for the piperacillin component of piperacillin/tazobactam. The ELF concentrations of ceftolozane exceeded 8 μg/mL for 60% of the 8-hour dosing interval. The plasma concentrations for ceftolozane were consistent with those seen previously at this dose.
  • The ELF concentration vs. time profiles for ceftolozane and tazobactam components of CXA-201 are shown in FIGS. 2A and 2B, respectively. Comparative data showing the ELF concentration vs. time profiles for piperacillin and tazobactam components of the comparator drug are shown in FIGS. 3A and 3B, respectively. The ELF to plasma penetration ratios are shown in Table 2.
  • The PK parameters were determined by non-compartmental PK analysis. PHOENIX® WinNonlin v 6.1 (PHARSIGHT®, Mountain View, Calif.) was used for the derivation of all PK individual measures for each subject. The PK parameters for ELF were calculated by taking the mean concentrations of the 5 subjects at each time point and constructing a single profile over the duration of sampling. In the event that the urea concentrations determined in plasma or ELF were below quantifiable limits, thereby providing only an estimate of concentration, those values were not used in the calculation of mean concentration at that time point. The ceftolozane, piperacillin, and tazobactam PK parameters that were computed in plasma and ELF were:
      • Cmax (μg/mL): Maximum plasma and ELF concentration over the entire sampling phase directly obtained from the experimental plasma concentration time data, without interpolation.
      • Tmax (hr): Sampling time at which Cmax occurred, obtained directly from the experimental plasma and ELF concentration time data, without interpolation.
      • Clast (μg/mL): Plasma or ELF concentration when last quantifiable concentration was observed, relative to the end of infusion.
      • Tlast (hr): Time when the last quantifiable concentration was observed.
      • AUC0-t (μg*hr/mL): An area under the concentration time curve from the time of the dose to the end of the dosing interval.
      • Percent penetration into ELF: Calculated as the ratio of AUC0-tELF and mean AUC0-tPlasma.
  • TABLE 2
    Summary of ELF to Plasma Penetration Ratios
    Mean Plasma ELF ELF
    AUC0-τ AUC0-τ Penetration
    Analyte (μg*hr/mL) (μg*hr/mL) Ratio
    ceftolozane 158.5 75.1 0.48
    (in CXA-201)
    Tazobactam 19.3 8.5 0.44
    (in CXA-201)
    Piperacillin 357.3 94.5 0.26
    (in piperacillin/tazobactam)
    Tazobactam 46.1 24.7 0.54
    (in piperacillin/tazobactam)
  • The ELF/plasma AUC ratio for the ceftolozane component of CXA-201 was 0.48, compared to 0.26 for the piperacillin component of the comparator drug (piperacillin/tazobactam). The ELF/plasma AUC ratio for tazobactam was 0.44 and 0.54 when given as part of CXA-201 and piperacillin/tazobactam, respectively. The ELF concentrations of ceftolozane exceeded 8 μg/mL for 60% of the 8-hour dosing interval. The plasma and ELF concentrations of tazobactam when given as piperacillin/tazobactam was approximately 2-fold higher than when an equivalent dose was given as CXA-201.
  • The results show that ceftolozane and tazobactam (i.e., administered as CXA-201) penetrated well into the ELF of healthy volunteers compared to piperacillin/tazobactam, an agent widely used for treatment of lower respiratory infections. CXA-201's intrapulmonary pharmacokinetics support use of CXA-201 as a parenteral (e.g., intravenous) antibiotic for treatment of lower respiratory tract infections, including infections caused by pathogens with minimum inhibitory concentrations of ≦8 μg/ml. The concentrations of ceftolozane in ELF exceeded 8 μg/mL, a concentration that inhibits 99% of P. aeruginosa, for approximately 60% of the 8-hour dosing interval for the CXA-201 regimen of 1.5 grams every eight hours as a 60 minute infusion.
  • Assessment of the safety and tolerability of multiple-doses of intravenous CXA-201 in healthy adult volunteers.
  • Among the subjects, 50 of the 51 (98%) subjects received all 3 doses of study medication and completed the BAL procedure. One subject prematurely discontinued piperacillin/tazobactam and terminated their participation in the study due to an AE of hypersensitivity that occurred during administration of the first dose. Demographics and baseline characteristics are summarized in Table 3, the two treatment arms were well balanced.
  • TABLE 3
    Demographics and Baseline Characteristics (Safety Population)
    Piperacillin/
    CXA-201 tazobactam
    1.5 grams 4.5 grams
    (N = 25) (N = 26)
    Sex, n (%)
    Female 11 (44.0) 11 (42.3)
    Male 14 (56.0) 15 (57.7)
    Age, years
    Mean (SD) 32.6 (7.8) 34.2 (8.5)
    Minimum, Maximum 21, 47 22, 49
    Race, n (%)
    White 20 (80.0) 21 (80.8)
    Black or African American 2 (8.0) 2 (7.7)
    Asian 1 (4.0) 0 (0.0)
    American Indian or Alaska Native 0 (0.0) 1 (3.8)
    Native Hawaiian or Pacific Islander 1 (4.0) 0 (0.0)
    Other 1 (4.0) 2 (7.7)
    BMI, kg/m2
    Mean (SD) 26.21 (2.6) 23.23 (2.4)
    Minimum, Maximum 22.3, 30.0 20.6, 29.9
  • During the study, treatment-emergent AEs (TEAEs) occurred in 20.0% (5/25) of subjects receiving CXA-201 and 23.1% (6/26) of subjects receiving piperacillin/tazobactam. No serious AEs were reported in either treatment group. All AEs were mild in severity. The incidence and pattern of AEs were generally similar in the 2 treatment groups, Table 4.
  • TABLE 4
    TEAEs by Preferred Term (Safety Population)
    Subjects with at least 1 TEAE 5 (20.0) 6 (23.1)
    Diarrhea 1 (4.0) 3 (11.5)
    Viral Upper Respiratory Infection 1 (4.0) 0 (0)
    Musculoskeletal Chest Pain 1 (4.0) 0 (0)
    Somnolence 1 (4.0) 0 (0)
    Hematuria 1 (4.0) 0 (0)
    Cough 1 (4.0) 0 (0)
    Type I Hypersensitivity 0 (0) 1 (3.8)
    Alanine Aminotransferase Increased 0 (0) 1 (3.8)
    Aspartate Aminotransferase Increased 0 (0) 1 (3.8)
    Blood Creatine Phosphokinase Increased 0 (0) 1 (3.8)
    Hyperkalemia 0 (0) 1 (3.8)
  • Eight subjects had TEAEs assessed as related to study drug; two in the CXA-201 group (diarrhea and somnolence in 1 subject each) and six in the piperacillin/tazobactam group (diarrhea in 3 subjects, type I hypersensitivity in 1 subject, blood creatine phosphokinase increased in 1 subject, and alanine aminotransferase increased, aspartate aminotransferase increased, and hyperkalaemia all in the same 1 subject). One piperacillin/tazobactam-treated subject discontinued study drug due to an adverse event, type I hypersensitivity. There were no clinically significant changes in safety laboratory assessments or vital signs.
  • CXA-201 appeared safe and well tolerated in this group of healthy adult subjects.
  • Determining Appropriate Dose
  • A Monte Carlo simulation was performed based on clinical trial data to predict an effective CXA-201 dose for treating nosocomial pneumonia using PHOENIX® NLME (PHARSIGHT®, Mountain View, Calif.) software, a tool for data processing and modeling for population PK/PD analysis. A population pharmacokinetic (PK) model was developed using the CXA-201 plasma concentration versus time data from a previously conducted Phase 2 study in patients with complicated intra abdominal infections. Estimates of clearance and volume of distribution along with the associated inter-individual variability were obtained from these analyses. The outputs from the PK population model served as inputs for a clinical trial simulation performed using PHARSIGHT® Trial Simulator (PHARSIGHT®) software, a tool for defining and testing interactive drug models, exploring and communicating study design attributes, and performing statistical and sensitivity analysis through graphical and statistical summaries. Based on the mean ELF penetration data, an ELF/Plasma AUC ratio of 0.48 for ceftolozane (modeled as a numerical range of 0.25-0.65) calculated from the ceftolozane ELF study mentioned above was used to generate a random/Plasma AUC ratio from the range 0.25-0.65 for each simulated patient. This range reflects a conservative estimate of the potential distribution in a patient population. Using the results from the PK population model and the ELF/Plasma AUC ratio, the model simulated plasma and ELF concentration of CXA-201 versus time profiles for 1,000 hypothetical clinical trial patients with nosocomial pneumonia. The model evaluated the probability of clinical success of the 3.0 g every 8 hour (q8h) dose of CXA-201 against three key pathogens in nosocomial pneumonia. The MIC distribution for these pathogens was imputed from 2008 United States surveillance data. Clinical success was defined as the achievement of an ELF or plasma concentration of ceftolozane higher than the MIC(s) of the lower respiratory pathogen(s) for a given patient. In vivo models have demonstrated that, as for typical cephlaosporins, the relavent PK/PD driver for CXA-201 is the percentage of time above MIC during the dosing interval. The target is to achieve concentrations that exceed the MIC of the pathogen for 45-50% of the time between each q8H dose. Thus, a threshold of 50% time above the minimum inhibitory concentration [T>MIC] on Day 7 of treatment was used. Plasma and ELF concentrations were estimated at 15 time-points post-administration on Day 7 when dosed every 8 hours. The results of these simulations are shown in Table 5.
  • TABLE 5
    Probability of Target Attainment versus Key Pathogens
    in Nosocomial Pneumonia Using the Simulated 3.0 g
    versus the 1.5 g Dose of Ceftolozane/tazobactam
    50% 50%
    T > MIC T > MIC
    Pathogen Dosing Regimen in Plasma in ELF
    P. aeruginosa 1.5 g q8 h 98.2 94.6
    3.0 g q8 h 99.4 98.5
    E. coli 1.5 g q8 h 96.3 94.2
    3.0 g q8 h 98.8 95.5
    K. pneumoniae 1.5 g q8 h 90.2 87.3
    3.0 g q8 h 92.6 89.3
    Abbreviation: T > MIC = Time above minimum inhibitory concentration.
  • These simulations demonstrate that the 3.0 g dose of CXA-201 administered every 8 hours is expected to provide adequate concentrations for treatment of the vast majority of lower respiratory infections caused by these pathogens.
  • Following these simulations, the safety and tolerability of a 10 day course of CXA-201 3.0 g IV q8h was evaluated in healthy human volunteers. Subjects were randomized to receive either 3.0 g (2.0/1.0 g) CXA-201 (n=8), 1.5 g (1.0/0.5 g) CXA-201 (n=4), or placebo (n=4). The data showed that CXA-201 was generally safe and well tolerated in this study. There were no serious adverse events or deaths reported in this study.
  • In conclusion, given the pharmacokinetic simulations conducted, the favorable data from the intrapulmonary PK study and demonstrated safety and tolerability of the higher dose of CXA-201 in the Phase 1 study mentioned above, the data provide justification for the use of 3.0 g CXA-201 IV q8h for the treatment of patients with nosocomial pneumonia caused by Gram-negative pathogens.
  • Preparing CXA-201
  • CXA-201 can be prepared by combining ceftolozane and tazobactam in a 2:1 weight ratio. CXA-201 can be obtained using methods described in U.S. Pat. No. 7,129,232 and Toda et al., “Synthesis and SAR of novel parenteral anti-pseudomonal cephalosporins: Discovery of FR264205,” Bioorganic & Medicinal Chemistry Letters, 18, 4849-4852 (2008), incorporated herein by reference in its entirety.
  • According to the method disclosed in Toda et al., “Synthesis and SAR of novel parenteral anti-pseudomonal cephalosporins: Discovery of FR264205,” Bioorganic & Medicinal Chemistry Letters, 18, 4849-4852 (2008), ceftolozane can be obtained by the synthetic schemes of FIGS. 4A and 4B. Referring to FIGS. 4A and 4B, synthesis of ceftolozane can be performed via activation of the thiadiazolyl-oximinoacetic acid derivative (I) with methanesulfonyl chloride and K2CO3 in DMA at 10° C., followed by coupling with the 7-aminocephem (II) by means of Et3N in cold EtOAc/H2O, affords amide (III) (1). Substitution of the allylic chloride of compound (III) with 4-[(N-Boc-aminoethyl)carbamoylamino]-1-methyl-5-tritylaminopyrazole (IV) in the presence of 1,3-bis(trimethylsilyl)urea (BSU) and KI in DMF then affords the protected pyrazolium adduct (V), which, after full deprotection with trifluoroacetic acid in anisole/CH2Cl2, can be isolated as the hydrogensulfate salt by treatment with H2SO4 in i-PrOH/H2O (1, 2). Scheme 1. The pyrazolyl urea intermediate (IV) can be prepared as follows. Treatment of 5-amino-1-methylpyrazole (VI) with NaNO2/HCl in water at 5° C. gives the 4-nitrosopyrazole derivative (VII), which can be reduced to the diaminopyrazole (VIII) by catalytic hydrogenation over Pd/C in the presence of H2SO4. Selective acylation of the 4-amino group of compound (VIII) with phenyl chloroformate in the presence of NaOH in H2O/dioxane at 10° C. then yields the phenyl carbamate (IX). After protection of the free amine group of carbamate (IX) with chlorotriphenylmethane in the presence of Et3N in THF, the resulting N-trityl derivative (X)can be coupled with N-Boc-ethylenediamine (XI) in the presence of Et3N in DMF to afford pyrazolyl urea (IV).
  • Biological Activity Assay
  • The antibacterial activity of the CXA-201 or other compounds can be measured by the minimum inhibitory concentrations (MIC) of the compounds against various bacteria measured by using the broth microdilution method performed according to Clinical and Laboratory Standards Institute (CLSI) guidelines with modifications as described below (CLSI guidelines can be derived from the CLSI document M7-A8 published in January 2009: “Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard-Eighth Edition”).
  • To prepare for MIC testing, individual colonies can be isolated by streaking frozen glycerol material containing Staphylococcus or Pseudomonas spp. onto rich, non-selective, tryptic soy agar containing 5% sheep's blood (TSAB), and incubated at 37° C. for 18-24 hrs.
  • On the day of testing, primary cultures can be started by scraping off 5-10 colonies from the TSAB plates. The material can be suspended in ˜5 mL of cation adjusted Mueller Hinton Broth (CAMHB) in 14 mL culture tubes and can be incubated at 37° C. with aeration (200 rpm) for ˜2 hrs until the OD600 was ≧0.1.
  • Inoculum cultures can be prepared by standardizing the primary cultures to OD600=0.1 and then adding 20 μL of the adjusted primary culture per 1 mL CAMHB for Pseudomonas and CAMHB plus 4% NaCl for MRSA so that the final inoculum density was ˜105 colony forming units per milliliter. Diluted inoculum cultures can be used to inoculate 50 μL per well in 96 well broth microdilution assay plates. 50 μL of CAMHB that contained compound concentrations ranging from 64-0.06 μg/mL in two-fold dilutions can also be added to the broth microdilution assay plates for a final volume 100 μL per well, therefore final culture OD600 was approximately 0.001 and the final NaCl concentration for the MRSA strain was 2%.
  • Plates can be incubated for 18-20 hours at 37° C. with aeration (200 rpm). Following incubation, growth can be confirmed visually placing plates over a viewing apparatus (stand with a minor underneath) and then OD600 can be measured using a SpectraMax 340PC384 plate reader (Molecular Devices, Sunnyvale, Calif.). Growth was defined as turbidity that could be detected with the naked eye or achieving minimum OD600 of 0.1. MIC values were defined as the lowest concentration producing no visible turbidity.
  • The examples and illustrative embodiments described herein are provided by way of illustration, and do not constitute additional limitations on the scope of the claims. While some embodiments have been shown and described in the instant specification, the specification as ready by one of ordinary skill in the relevant arts also discloses various modifications and substitutions of embodiments explicitly disclosed herein. The exemplary embodiments from the specification are not provided to read additional limitations into the claims.

Claims (23)

1. A method of treating an intrapulmonary infection comprising the step of intravenously administering about every 8 hours to a subject in need thereof a pharmaceutical composition comprising 3.0 g of ceftolozane.
2. The method of claim 1, wherein the pharmaceutical composition further comprises tazobactam.
3. The method of claim 2, wherein the pharmaceutical composition comprises ceftolozane and tazobactam and the infection comprises Gram-negative bacteria.
4. A method of treating an intrapulmonary infection comprising the step of administering a therapeutically effective amount of a pharmaceutical composition comprising ceftolozane.
5. The method of claim 1, wherein the intrapulmonary infection includes an infection in the lung.
6. The method of claim 1, wherein the intrapulmonary infection is pneumonia.
7. The method of claim 1, wherein the intrapulmonary infection is nosocomial pneumonia.
8. The method of claim 1, wherein the pharmaceutical composition is parenterally administered.
9. The method of claim 1, wherein the pharmaceutical composition is intravenously administered.
10. The method of claim 1, wherein the pharmaceutical composition is intravenously administered about once every 8 hours as an infusion.
11. The method of claim 10, wherein the pharmaceutical composition is intravenously administered as a 60-minute infusion.
12. The method of claim 1, wherein the infection comprises Pseudomonas aeruginosa, Enterobacteriaceae, or a combination thereof.
13. The method of claim 1, wherein the infection comprises Pseudomonas aeruginosa.
14. The method of claim 1, wherein the infection comprises a pathogen with minimum inhibitory concentration for ceftolozane and tazobactam of ≦8 μg/ml.
15. The method of claim 1, wherein the infection comprises a pathogen with minimum inhibitory concentration for ceftolozane of ≦8 μg/ml.
16. A method of providing tazobactam or ceftolozane in the epithelial lining fluid of a subject in an amount effective to treat an intrapulmonary infection, comprising the step of intravenously administering to the subject a pharmaceutical composition comprising ceftolozane.
17. The method of claim 16, wherein the pharmaceutical composition further comprises tazobactam and the pharmaceutical composition is CXA-201.
18. The method of claim 1, wherein the method comprises administering about 1.5 g of ceftolozane and tazobactam every 8 hours.
19. The method of claim 1, wherein the amount of the ceftolozane in the epithelial lining fluid of the subject effective to treat an intrapulmonary infection is at least about 8 μg/ml.
20. The method of claim 1, wherein the ELF concentration of ceftolozane in the ELF reaches at least about 8 μg/ml after administration of the pharmaceutical composition.
21. The method of claim 1, wherein the subject is a human having, or believed to be at risk of having, nosocomial pneumonia.
22. The method of claim 21, wherein the patient has ventilator acquired pneumonia or hospital acquired pneumonia.
23. The method of claim 1, wherein the treatment comprises administering ceftolozane every 8 hours.
US13/607,138 2011-09-09 2012-09-07 Methods for treating intrapulmonary infections Abandoned US20130065874A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US13/607,138 US20130065874A1 (en) 2011-09-09 2012-09-07 Methods for treating intrapulmonary infections
US14/512,608 US9724353B2 (en) 2011-09-09 2014-10-13 Methods for treating intrapulmonary infections
US14/550,125 US20150080360A1 (en) 2011-09-09 2014-11-21 Methods for treating intrapulmonary infections
US15/629,360 US10028963B2 (en) 2011-09-09 2017-06-21 Methods for treating intrapulmonary infections
US16/016,524 US20180296567A1 (en) 2011-09-09 2018-06-22 Methods for treating intrapulmonary infections

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161532914P 2011-09-09 2011-09-09
US201261657386P 2012-06-08 2012-06-08
US13/607,138 US20130065874A1 (en) 2011-09-09 2012-09-07 Methods for treating intrapulmonary infections

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/512,608 Continuation US9724353B2 (en) 2011-09-09 2014-10-13 Methods for treating intrapulmonary infections

Publications (1)

Publication Number Publication Date
US20130065874A1 true US20130065874A1 (en) 2013-03-14

Family

ID=47830390

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/607,138 Abandoned US20130065874A1 (en) 2011-09-09 2012-09-07 Methods for treating intrapulmonary infections
US14/512,608 Active US9724353B2 (en) 2011-09-09 2014-10-13 Methods for treating intrapulmonary infections
US14/550,125 Abandoned US20150080360A1 (en) 2011-09-09 2014-11-21 Methods for treating intrapulmonary infections
US15/629,360 Active US10028963B2 (en) 2011-09-09 2017-06-21 Methods for treating intrapulmonary infections
US16/016,524 Abandoned US20180296567A1 (en) 2011-09-09 2018-06-22 Methods for treating intrapulmonary infections

Family Applications After (4)

Application Number Title Priority Date Filing Date
US14/512,608 Active US9724353B2 (en) 2011-09-09 2014-10-13 Methods for treating intrapulmonary infections
US14/550,125 Abandoned US20150080360A1 (en) 2011-09-09 2014-11-21 Methods for treating intrapulmonary infections
US15/629,360 Active US10028963B2 (en) 2011-09-09 2017-06-21 Methods for treating intrapulmonary infections
US16/016,524 Abandoned US20180296567A1 (en) 2011-09-09 2018-06-22 Methods for treating intrapulmonary infections

Country Status (9)

Country Link
US (5) US20130065874A1 (en)
EP (3) EP2862569A1 (en)
JP (1) JP6151257B2 (en)
CN (1) CN103945842A (en)
CA (1) CA2848012A1 (en)
EA (1) EA028342B1 (en)
HK (1) HK1200096A1 (en)
MX (1) MX352760B (en)
WO (1) WO2013036783A2 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8685957B1 (en) 2012-09-27 2014-04-01 Cubist Pharmaceuticals, Inc. Tazobactam arginine compositions
US8809314B1 (en) 2012-09-07 2014-08-19 Cubist Pharmacueticals, Inc. Cephalosporin compound
US20140303136A1 (en) * 2013-03-14 2014-10-09 Calixa Therapeutics, Inc. Ceftolozane antibiotic compositions
US8906898B1 (en) 2013-09-27 2014-12-09 Calixa Therapeutics, Inc. Solid forms of ceftolozane
US8968753B2 (en) 2013-03-15 2015-03-03 Calixa Therapeutics, Inc. Ceftolozane-tazobactam pharmaceutical compositions
WO2015196077A1 (en) * 2014-06-20 2015-12-23 Merck Sharp & Dohme Corp. Reactions of thiadiazolyl-oximinoacetic acid derivative compounds
WO2015035376A3 (en) * 2013-09-09 2015-12-30 Merck Sharp & Dohme Corp. Treating infections with ceftolozane/tazobactam in subjects having impaired renal function
US20160000921A1 (en) * 2013-03-15 2016-01-07 Merck Sharp & Dohme Corp. Ceftolozane antibiotic compositions
WO2016028670A1 (en) * 2014-08-18 2016-02-25 Merck Sharp & Dohme Corp. Salt forms of ceftolozane
US10214543B2 (en) 2014-12-30 2019-02-26 Merck Sharp & Dohme Corp. Synthesis of cephalosporin compounds

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8513305B2 (en) 2007-05-14 2013-08-20 Research Foundation Of State University Of New York Induction of a physiological dispersion response in bacterial cells in a biofilm
CA2848012A1 (en) 2011-09-09 2013-03-14 Cubist Pharmaceuticals, Inc. Methods for treating intrapulmonary infections
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

Family Cites Families (196)

* 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
PH17188A (en) 1977-03-14 1984-06-14 Fujisawa Pharmaceutical Co New cephem and cepham compounds and their pharmaceutical compositions and method of use
US4496562A (en) 1977-03-14 1985-01-29 Fujisawa Pharmaceutical Co., Ltd. 7-Substituted-3-cephem-4-carboxylic acid esters
US4409217A (en) 1977-03-14 1983-10-11 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
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
AU536842B2 (en) 1978-12-29 1984-05-24 Fujisawa Pharmaceutical Co., Ltd. Cephalosporin antibiotics
US4332798A (en) 1978-12-29 1982-06-01 Fujisawa Pharmaceutical Co., Ltd. 7-Amino-thia-diazole oxyimino derivatives of cephem and cephem compounds
US4390534A (en) 1978-12-29 1983-06-28 Fujisawa Pharmaceutical Co., Ltd. Cephem and cepham compounds
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
EP0025199B1 (en) 1979-09-03 1984-10-31 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds, processes for their preparation and pharmaceutical compositions containing them
US4338313A (en) 1979-10-12 1982-07-06 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4332800A (en) 1979-10-12 1982-06-01 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4381299A (en) 1980-03-07 1983-04-26 Fujisawa Pharmaceutical Co., Ltd. 7-Amino-thiadiazole oxyimino derivatives of cephem and cepham compounds
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
US4409214A (en) 1979-11-19 1983-10-11 Fujisawa Pharmaceutical, Co., Ltd. 7-Acylamino-3-vinylcephalosporanic 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
EP0043546B1 (en) 1980-07-04 1986-01-29 Fujisawa Pharmaceutical Co., Ltd. 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
IT1142096B (en) 1980-12-15 1986-10-08 Fujisawa Pharmaceutical Co 7-ACHYLAMINE CEPHALOSPORANIC ACID DERIVATIVES AND PROCEDURES FOR THEIR PREPARATION
DE3177090D1 (en) 1980-12-31 1989-09-28 Fujisawa Pharmaceutical Co 7-acylaminocephalosporanic acid derivatives and processes for the preparation thereof
US4427677A (en) 1980-12-31 1984-01-24 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
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"
US4640915A (en) 1982-03-29 1987-02-03 Fujisawa Pharmaceutical Co., Ltd. 1-azabicyclo[3.2.0]hept-2-ene-2-carboxylic acid derivatives
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
US4487768A (en) 1982-12-22 1984-12-11 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
US4463003A (en) 1982-12-22 1984-07-31 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
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
EP0137442A3 (en) 1983-10-08 1986-01-15 Hoechst Aktiengesellschaft Cephalosporin derivatives and process for their preparation
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
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
US4751295A (en) 1984-04-09 1988-06-14 Bristol-Myers Company Cephalosporin derivatives
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
AU586215B2 (en) 1985-01-21 1989-07-06 Nippon Pharmaceutical Development Institute Company Limited Novel ```-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
JPS62158290A (en) 1985-12-28 1987-07-14 Banyu Pharmaceut Co Ltd Novel cephalosporin derivative
EP0237735B1 (en) 1986-03-17 1994-03-30 Fujisawa Pharmaceutical Co., Ltd. 3,7-Disubstituted-3-cephem compounds and process for production of the same
US4833134A (en) 1986-08-19 1989-05-23 Takeda Chemical Industries, Ltd. Cephem compounds
JPS6351389A (en) 1986-08-22 1988-03-04 Teijin Ltd Cephalosporin derivative, production thereof and composition having antimicrobial activity
JPS6351388A (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
EP0303172A3 (en) 1987-08-14 1991-05-15 F. Hoffmann-La Roche Ag Oxyimino-cephalosporins
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
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
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
GB8817653D0 (en) 1988-07-25 1988-09-01 Fujisawa Pharmaceutical Co New cephem compounds & processes for preparation thereof
JPH0288582A (en) 1988-09-24 1990-03-28 Katayama Seiyakushiyo:Kk Novel cephem compound and production thereof
EP0366189A3 (en) 1988-10-24 1992-01-02 Norwich Eaton Pharmaceuticals, Inc. Novel antimicrobial lactam-quinolones
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
JPH05222058A (en) 1992-02-17 1993-08-31 Fujisawa Pharmaceut Co Ltd New cephem compound and its salt
JPH0656848A (en) 1992-02-17 1994-03-01 Fujisawa Pharmaceut Co Ltd New cephem compound and its salt
JPH06128268A (en) 1992-10-16 1994-05-10 Takeda Chem Ind Ltd Cephem compound, its production and antibacterial composition
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
KR0180798B1 (en) 1993-11-06 1999-03-20 고바야시 요시오 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
KR100248851B1 (en) 1994-08-16 2000-04-01 이치로 키타사토 A novel cephem derivative
DE4440141A1 (en) 1994-11-10 1996-05-15 Hoechst Ag Novel crystalline cephem acid addition salts and process for their preparation
EP0893446B1 (en) 1996-04-04 2006-10-04 Shionogi & Co., Ltd. Cephem compounds and drugs containing the compounds
WO1999028308A1 (en) 1997-11-29 1999-06-10 Truett William L Antibiotics and process for preparation
WO1999063929A2 (en) 1998-06-08 1999-12-16 Advanced Medicine, Inc. Multibinding inhibitors of microsomal triglyceride transferase protein
AU5006499A (en) * 1998-07-23 2000-02-14 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
US6207661B1 (en) 1999-02-22 2001-03-27 Baxter International Inc. Premixed formulation of piperacillin sodium and tazobactam sodium injection
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.
TWI335332B (en) 2001-10-12 2011-01-01 Theravance Inc Cross-linked vancomycin-cephalosporin antibiotics
TW200305422A (en) 2002-03-18 2003-11-01 Shionogi & Co Broad spectrum cefem compounds
ATE556088T1 (en) 2002-05-24 2012-05-15 Theravance Inc CONJUGATE CONTAINING CEPHALOSPORIN AND GLYCOPEPTIDE ANTIBIOTIC
WO2003104241A1 (en) 2002-06-07 2003-12-18 Orchid Chemicals & Pharmaceuticals Limited Process for preparation of penam derivatives from cepham derivatives
AU2002952355A0 (en) * 2002-10-30 2002-11-14 Fujisawa Pharmaceutical Co., Ltd. Cephem compounds
EP1556389B9 (en) 2002-10-30 2015-09-09 Astellas Pharma Inc. Cephem compounds
AU2003302472B2 (en) 2002-11-22 2010-10-21 The Johns Hopkins University Target for therapy of cognitive impairment
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
EP1633759B1 (en) 2003-05-23 2009-12-02 Theravance, Inc. Cross-linked glycopeptide-cephalosporin antibiotics
DE602004012269T2 (en) 2003-07-11 2009-04-30 Theravance, Inc., South San Francisco 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
TW200524943A (en) 2003-09-18 2005-08-01 Fujisawa Pharmaceutical Co Cephem compounds
JP4535366B2 (en) 2003-12-03 2010-09-01 塩野義製薬株式会社 Method for producing cephem agent
JP2007519747A (en) 2004-01-30 2007-07-19 ワイス A composition comprising piperacillin and tazobactam and substantially free of galactomannan
US20060019914A1 (en) 2004-02-11 2006-01-26 University Of Tennessee Research Foundation Inhibition of tumor growth and invasion by anti-matrix metalloproteinase DNAzymes
AU2005219718A1 (en) 2004-03-05 2005-09-15 Shionogi & Co., Ltd. 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
AU2005295644A1 (en) 2004-10-14 2006-04-27 Wyeth Compositions containing piperacillin, tazobactam and a aminocarboxilic acid in a sodium lactate diluent
EP1919449A2 (en) 2005-06-07 2008-05-14 Foamix Ltd. Antibiotic kit and composition and uses thereof
CN101299995B (en) 2005-12-05 2013-04-24 桑多斯股份公司 Process for the preparation of lyophilized piperacilline sodium with improved stability after reconstitution
WO2007129176A2 (en) 2006-04-28 2007-11-15 Wockhardt Ltd Improvements in therapy for treating resistant bacterial infections
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
US20080160067A1 (en) 2006-09-07 2008-07-03 Albert Boeckh Novel soft chewable, tablet, and long-acting injectable veterinary antibiotic formulations
FI119678B (en) 2006-11-28 2009-02-13 Ipsat Therapies Oy Use of beta-lactamase
DE102007009242A1 (en) 2007-02-22 2008-09-18 Evonik Röhm Gmbh Pellets with enteric-coated matix
WO2008113177A1 (en) 2007-03-20 2008-09-25 Centre De Recherche Sur Les Biotechnologies Marines Compositions comprising polyunsaturated fatty acid monoglycerides or derivatives thereof and uses thereof
US10580459B2 (en) 2007-08-23 2020-03-03 Sony Interactive Entertainment America Llc Dynamic media interaction using time-based metadata
CA2702257C (en) 2007-10-09 2016-07-12 Sopharmia, Inc. Broad spectrum beta-lactamase inhibitors
US20090098088A1 (en) 2007-10-10 2009-04-16 The Procter & Gamble Company Methods And Kits For The Treatment Of Diverticular Conditions
EP2285216A4 (en) 2008-02-21 2012-09-19 Sequoia Pharmaceuticals Inc Hiv protease inhibitor and cytochrome p450 inhibitor combinations
EP2280713A1 (en) 2008-05-01 2011-02-09 The Procter & Gamble Company Methods and kits for the treatment of inflammatory bowel disorder conditions
KR102102972B1 (en) 2009-06-10 2020-04-22 테크필즈 바이오켐 코., 엘티디. High penetration compositions or prodrugs of antimicrobials and antimicrobial-related compounds
RU2012107163A (en) * 2009-07-28 2013-09-10 Анакор Фармасьютикалз, Инк. TRIZED SUBSTITUTE BORN-CONTAINING MOLECULES
CA2848012A1 (en) 2011-09-09 2013-03-14 Cubist Pharmaceuticals, Inc. Methods for treating intrapulmonary infections
WO2013141279A1 (en) 2012-03-21 2013-09-26 新神戸電機株式会社 Electrode plate for rectangular nonaqueous electrolyte storage cell
BR112015023523B8 (en) 2013-03-15 2023-03-07 Merck Sharp & Dohme PHARMACEUTICAL COMPOSITIONS AND SINGLE DOSAGE FORM CONTAINER
US20140274990A1 (en) 2013-03-15 2014-09-18 Cubist Pharmaceuticals, Inc. Ceftolozane pharmaceutical compositions
JP6128268B2 (en) 2016-06-17 2017-05-17 株式会社デンソー Rotating electric machine stator

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Cubist (Cubist Annoucnes Positive Results from Two Phase 2 Trials, CSA-201 and CDAD Program, June 2011, pages 1-5). *
Sader (Antimicrobial Activity of CSA-101, a Novel Cephalosporin Tested in Combinationwith Tazobactam against Enterobacteriaceae, Pseudomonas aeruginosa and Bacterodies fragilis Strains Having Various Resistance Phenotypes, Antimicrobial Agents and Chemotherapy, May 2011, pages 2390-2394) *
Takeda (In Vitro and In Vivo Activities of a new Cephalosporin, FT264205, against Pseudomonas aeruginosa, Antimicrobial Agents and Chemotherapy, Mar 2007, pages 826-830) *

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8809314B1 (en) 2012-09-07 2014-08-19 Cubist Pharmacueticals, Inc. Cephalosporin compound
US8685957B1 (en) 2012-09-27 2014-04-01 Cubist Pharmaceuticals, Inc. Tazobactam arginine compositions
US20140303136A1 (en) * 2013-03-14 2014-10-09 Calixa Therapeutics, Inc. Ceftolozane antibiotic compositions
US20160279140A1 (en) * 2013-03-15 2016-09-29 Merck Sharp & Dohme Corp Ceftolozane antibiotic compositions
US9872906B2 (en) * 2013-03-15 2018-01-23 Merck Sharp & Dohme Corp. Ceftolozane antibiotic compositions
US11278622B2 (en) 2013-03-15 2022-03-22 Merck Sharp & Dohme Corp. Ceftolozane antibiotic compositions
US9044485B2 (en) * 2013-03-15 2015-06-02 Calixa Therapeutics, Inc. Ceftolozane antibiotic compositions
US10420841B2 (en) 2013-03-15 2019-09-24 Merck, Sharp & Dohme Corp. Ceftolozane antibiotic compositions
US8968753B2 (en) 2013-03-15 2015-03-03 Calixa Therapeutics, Inc. Ceftolozane-tazobactam pharmaceutical compositions
US20160000921A1 (en) * 2013-03-15 2016-01-07 Merck Sharp & Dohme Corp. Ceftolozane antibiotic compositions
US9925196B2 (en) 2013-03-15 2018-03-27 Merck Sharp & Dohme Corp. Ceftolozane-tazobactam pharmaceutical compositions
US9320740B2 (en) 2013-03-15 2016-04-26 Merck Sharp & Dohme Corp. Ceftolozane-tazobactam pharmaceutical compositions
US10376496B2 (en) 2013-09-09 2019-08-13 Merck, Sharp & Dohme Corp. Treating infections with ceftolozane/tazobactam in subjects having impaired renal function
EP3043797A4 (en) * 2013-09-09 2017-04-19 Merck Sharp & Dohme Corp. Treating infections with ceftolozane/tazobactam in subjects having impaired renal function
WO2015035376A3 (en) * 2013-09-09 2015-12-30 Merck Sharp & Dohme Corp. Treating infections with ceftolozane/tazobactam in subjects having impaired renal function
AU2017279686B2 (en) * 2013-09-09 2020-01-30 Merck Sharp & Dohme Llc 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
AU2020202756B2 (en) * 2013-09-09 2021-12-23 Merck Sharp & Dohme Llc Treating infections with Ceftolozane/Tazobactam in subjects having impaired renal function
US8906898B1 (en) 2013-09-27 2014-12-09 Calixa Therapeutics, Inc. Solid forms of ceftolozane
WO2015048217A1 (en) * 2013-09-27 2015-04-02 Calixa Therapeutics, Inc. Solid forms of ceftolozane
US9695196B2 (en) 2014-06-20 2017-07-04 Merck Sharp & Dohme Corp. Reactions of thiadiazolyl-oximinoacetic acid derivative compounds
WO2015196077A1 (en) * 2014-06-20 2015-12-23 Merck Sharp & Dohme Corp. Reactions of thiadiazolyl-oximinoacetic acid derivative compounds
WO2016028670A1 (en) * 2014-08-18 2016-02-25 Merck Sharp & Dohme Corp. Salt forms of ceftolozane
US10000510B2 (en) 2014-08-18 2018-06-19 Merck Sharp & Dohme Corp. Salt forms of ceftolozane
US10214543B2 (en) 2014-12-30 2019-02-26 Merck Sharp & Dohme Corp. Synthesis of cephalosporin compounds

Also Published As

Publication number Publication date
JP6151257B2 (en) 2017-06-21
US20180296567A1 (en) 2018-10-18
EP3616695A1 (en) 2020-03-04
EP2753326A4 (en) 2015-04-15
EA028342B1 (en) 2017-11-30
MX2014002758A (en) 2014-10-17
EP2862569A1 (en) 2015-04-22
US20180064724A1 (en) 2018-03-08
HK1200096A1 (en) 2015-07-31
MX352760B (en) 2017-12-07
CA2848012A1 (en) 2013-03-14
WO2013036783A2 (en) 2013-03-14
US20150045338A1 (en) 2015-02-12
CN103945842A (en) 2014-07-23
US9724353B2 (en) 2017-08-08
JP2014526468A (en) 2014-10-06
US10028963B2 (en) 2018-07-24
US20150080360A1 (en) 2015-03-19
EP2753326A2 (en) 2014-07-16
EA201490590A1 (en) 2015-04-30
WO2013036783A3 (en) 2013-05-02

Similar Documents

Publication Publication Date Title
US10028963B2 (en) Methods for treating intrapulmonary infections
JP7245289B2 (en) Methods of treating bacterial infections
JP6870029B2 (en) Ceftrozan antibiotic composition
TWI690317B (en) Combination therapy for treatment of resistant bacterial infections
US11278622B2 (en) Ceftolozane antibiotic compositions
KR20190092566A (en) Combination therapy using amidine substituted β-lactam compounds and β-lactamase inhibitors for infection by antibiotic resistant bacterial strains
US20100197650A1 (en) Compositions and methods of treatment comprising ceftaroline
BR112013030282B1 (en) COMPOSITIONS UNDERSTANDING CEFEPIMA AND TAZOBACTAMA
AU2020202756B2 (en) Treating infections with Ceftolozane/Tazobactam in subjects having impaired renal function
Solomkin et al. Principles of antibiotic therapy
KR20120083344A (en) Compositions and methods for treating bacterial infections using ceftaroline
EP3919059B1 (en) Composition for treating carbapenem antibiotic-resistant acinetobacter baumannii infection
James et al. Antimicrobial therapy
Schriever et al. Antimicrobial Agents
US20180303853A1 (en) Compositions and methods for treating bacterial infections using ceftaroline

Legal Events

Date Code Title Description
AS Assignment

Owner name: ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT, CAN

Free format text: SECURITY AGREEMENT;ASSIGNORS:CUBIST PHARMACEUTICALS, INC.;ADOLOR CORPORATION;CALIXA THERAPEUTICS, INC.;AND OTHERS;REEL/FRAME:029339/0669

Effective date: 20121120

AS Assignment

Owner name: CUBIST PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PARSONS, TARA;CHANDORKAR, GURUDATT A.;UMEH, OBIAMIWE C.;AND OTHERS;REEL/FRAME:030080/0035

Effective date: 20130115

AS Assignment

Owner name: CALIXA THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CUBIST PHARMACEUTICALS, INC.;REEL/FRAME:034180/0472

Effective date: 20141113

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: CUBIST PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT;REEL/FRAME:036180/0070

Effective date: 20150707

Owner name: ADOLOR CORPORATION, PENNSYLVANIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT;REEL/FRAME:036180/0070

Effective date: 20150707

Owner name: CALIXA THERAPEUTICS, INC., MASSACHUSETTS

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:ROYAL BANK OF CANADA, AS ADMINISTRATIVE AGENT;REEL/FRAME:036180/0070

Effective date: 20150707