NZ711823B2 - Ceftolozane antibiotic compositions - Google Patents

Ceftolozane antibiotic compositions Download PDF

Info

Publication number
NZ711823B2
NZ711823B2 NZ711823A NZ71182314A NZ711823B2 NZ 711823 B2 NZ711823 B2 NZ 711823B2 NZ 711823 A NZ711823 A NZ 711823A NZ 71182314 A NZ71182314 A NZ 71182314A NZ 711823 B2 NZ711823 B2 NZ 711823B2
Authority
NZ
New Zealand
Prior art keywords
ceftolozane
tazobactam
sodium chloride
composition
sodium
Prior art date
Application number
NZ711823A
Other versions
NZ711823A (en
Inventor
Nicole Damour
Giuseppe Alessandro Donadelli
Giovanni Focliato
Chun Jiang
Dario Resemini
Joseph Terraciano
Original Assignee
Merck Sharp & Dohme Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck Sharp & Dohme Corp filed Critical Merck Sharp & Dohme Corp
Priority claimed from NZ700372A external-priority patent/NZ700372A/en
Publication of NZ711823A publication Critical patent/NZ711823A/en
Publication of NZ711823B2 publication Critical patent/NZ711823B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • 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
    • 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
    • A61K31/433Thidiazoles
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • 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
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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

Abstract

This disclosure provides pharmaceutical compositions comprising ceftolozane, pharmaceutical compositions comprising ceftolozane and tazobactam, methods of preparing those compositions, and related methods and uses of these compositions. Specifically, ceftolozane is chemically unstable in certain lyophilized compositions, leading to the formation of degradation products. The methods of the present invention result in compositions having stabilized ceftolozane, thereby providing ceftolozane at a desired potency and with reduced impurities (degradation products). Stabilized compositions comprising ceftolozane and tazobactam can be prepared by lyophilizing ceftolozane in the absence of tazobactam and then blending the lyophilized ceftolozane with a solid tazobactam composition. philized compositions, leading to the formation of degradation products. The methods of the present invention result in compositions having stabilized ceftolozane, thereby providing ceftolozane at a desired potency and with reduced impurities (degradation products). Stabilized compositions comprising ceftolozane and tazobactam can be prepared by lyophilizing ceftolozane in the absence of tazobactam and then blending the lyophilized ceftolozane with a solid tazobactam composition.

Description

CEFTOLOZANE ANTIBIOTIC COMPOSITIONS CROSS-REFERENCE TO RELATED APPLICATIONS This application is a divisional application of New Zealand Application No. 700372, filed on 14 March 2014, and is d to International Patent Application No. , filed on 14 March 2014 and claims priority from U.S. ional Patent Application Nos. 61/882,936, filed on 26 September 2013; 61/792,092, filed on 15 March 2013; 61/793,007, filed on 15 March 2013 and 61/893,436, filed on 21 October 2013; each of which is incorporated herein by reference in its entirety.
CAL FIELD This disclosure relates to pharmaceutical compositions comprising ceftolozane, pharmaceutical compositions comprising tazobactam and ceftolozane, s of preparing those compositions, and related methods and uses thereof.
BACKGROUND Ceftolozane is a cephalosporin antibacterial agent. The antibacterial activity of ceftolozane is believed to result from its interaction with penicillin binding proteins (PBPs) to inhibit the biosynthesis of the bacterial cell wall which acts to stop bacterial replication.
Ceftolozane is also referred to as "CXA-101", FR264205, (6R,7R)[(5-amino{[(2- thyl)carbamoyl]amino}methyl-1H-pyrazoliumyl)methyl]({(2Z)(5-aminol ,2,4-thiadiazolyl)[(l-carboxy-l-methylethoxy)imino]acetyl}amino)oxothia-l- azabicyclo[4.2.0]octenecarboxylate, or (6R,7R)[5-Amino[3-(2-aminoethyl)ureido]- yl-lH-pyrazoliumylmethyl][2-(5-amino-1,2,4-thiadiazolyl)[(Z)-l-carboxyl-methylethoxyimino ]acetamido]cephemcarboxylic acid). As used herein, the term "ceftolozane" means (6R,7R)[(5-amino{[(2-aminoethyl)carbamoyl]amino}methyl-1H- pyrazoliumyl)methyl]({(2Z)(5-amino-l,2,4-thiadiazolyl)[(l-carboxy-l- methylethoxy)imino]acetyl}amino)oxothia-l-azabicyclo[4.2.0]octenecarboxylate or (6R,7R)[5-Amino[3-(2-aminoethyl)ureido]-l-methyl-lH-pyrazoliumylmethyl][2- (5-amino-1,2,4-thiadiazolyl)[(Z)-l-carboxy-l-methylethoxyimino]acetamido]cephem carboxylic acid in its free-base or salt form, including a sulfate form. Unless ise indicated, the term "CXA-101" as used herein can refer to ceftolozane in any pharmaceutically acceptable form, e.g., ozane in its ase or salt form, including a ceftolozane sulfate salt form.
Ceftolozane sulfate is a pharmaceutically acceptable salt of ceftolozane that can be ed with sodium chloride and other components to obtain an antibiotic composition suitable for administration by injection or infusion.
Antibacterial pharmaceutical compositions can include ceftolozane as a pharmaceutically acceptable salt formulated for intravenous stration. Ceftolozane sulfate is a pharmaceutically acceptable ceftolozane salt of formula (I) that can be formulated for intravenous administration or infusion.
H C+COZH H804' INHZ ,0 HN "l H HN’Q S O H2Naw)?/ | \ NH S’N DOMAIN 2 COZH CH3 US. Patent No. 7,129,232 discloses ceftolozane and various ceftolozane salts. For example, a ceftolozane hydrogen sulfate salt is disclosed among ceftolozane salts that can be formed "with a base or an acid addition salt such as a salt with an nic base, for example, an alkali metal salt [e.g., sodium salt, potassium salt, etc.], an alkaline earth metal salt [e.g., calcium salt, magnesium salt, etc.], an ammonium salt; a salt with an organic base, for example, an organic amine salt [e.g., hylamine salt, triethylamine salt, pyridine salt, picoline salt, ethanolamine salt, triethanolamine salt, dicyclohexylamine salt, N,N'- dibenzylethylenediamine salt, etc.]; an inorganic acid addition salt [e.g., hydrochloride, romide, sulfate, hydrogen sulfate, phosphate, etc.]; an organic carboxylic or sulfonic acid on salt [e.g., formate, acetate, trifluoroacetate, maleate, tartrate, citrate, te, methanesulfonate, benzenesulfonate, toluenesulfonate, etc.]; and a salt with a basic or acidic amino acid [e.g., arginine, aspartic acid, glutamic acid, etc.]." Antibiotic pharmaceutical compositions comprising a beta-lactam antibiotic compound (e.g., a osporin) (i.e., an antibiotic compound possessing one or more beta- lactam es) can be administered with a beta-lactamase tor (BLI) compound. For example, beta-lactam otic nds such as ceftolozane or other cephalosporin antibiotic compounds can be formulated with, and/or administered in combination with beta- lactamase inhibiting compounds (e.g., tazobactam and salts thereof) in order to mitigate the effects of bacterial beta-lactamase enzymes that can lead to bacterial resistance to antibiotic therapy. Tazobactam is a BLI compound approved for use in fixed dose combination with piperacillin in an injectable antibacterial product available under commercial names ZOSYN (U.S.) and TAZOCIN (e.g., in Canada, and the United Kingdom). Tazobactam sodium, a derivative of the penicillin nucleus, is a penicillanic acid sulfone having the chemical name sodium (25,3S,5R)—3-methyl—7-oxo(1H-1,2,3-triazol—1-ylmethyl)thia- cyclo[3.2.0]heptanecarboxylate-4,4-dioxide. The chemical formula is C10H11N4NaOsS and the molecular weight is 322.3. The al structure of tazobactam sodium COONa OmmN/N‘N ,s, (3st H00 Ceftolozane can be formulated with tazobactam in antibiotic compositions called CXA-201 (ceftolozane/tazobactam for injection), comprising ceftolozane and tazobactam in a 2:1 weight ratio between the amount of ceftolozane active and the amount of tazob actam acid, regardless of the salt forms of these compositions (e. g., 1,000 mg of ozane active can be included in about 1,147 mg of ozane sulfate). CXA-201 compositions include an amount of tazobactam in a ceutically acceptable form providing 500 mg of tazobactam acid per 1,000 mg of ozane active as acomposition formulated for ion, or for titution prior to parenteral administration. In one product presentation, CXA- 201 can be provided in a single container comprising ceftolozane sulfate and tazob actam sodium, administered by reconstituting a container-unit dosage form container of solid CXA- 201 to form a reconstituted injectable formulation. In one presentation (e.g., for treatment of certain urinary tract infections and/or certain intr-abdominal infections), each unit dosage form container of CXA-201 can contain 1000 mg of ceftolozane active (free base lent weight, e. g., provided as a ceutically able salt such as ozane sulfate) and sterile tazobactam sodium at a quantity equivalent of 500 mg of tazobactam free acid, in a solid form. In another presentation (e. g., for treatment of hospital acquired/ventilator- associated bacterial pneumonia (HABP/VABP)), a CXA-201 t can include a unit dosage form container providing 2,000 mg of ceftolozane active (e. g., as an equivalent amount of ceftolozane sulfate) and 1,000 mg of tazobactam acid (e. g., as an equivalent amount of tazobactam sodium). CXA-201 compositions display potent antibacterial activity against various egative infections such as, for e, complicated intra-abdominal infection (cIAI), complicated urinary tract infection (cUTI), or hospital acquired/ventilator- associated bacterial pneumonia (HABP/VABP).
As disclosed herein, ceftolozane was initially found to be chemically unstable in certain lyophilized compositions evaluated during the pment of CXA-101 and CXA- 201 pharmaceutical compositions. For example, ceftolozane had a al rate of about 51% in the absence of a stabilizing agent during both a 3 day stability test at 70 degrees C, indicating loss of almost half of the ceftolozane during the test (Example 2, Table 2 control sample), and a 5.88% reduction in ceftolozane purity during a 7 day stability test at 60 degrees C in the absence of a stabilizing agent (Example 2, Table 2a control sample).
Second, the formation of a number of additional ceftolozane ation products formed during the preparation of initial compositions was observed by additional peaks using high performance liquid tography (HPLC) during ity tests of ceftolozane alone (e. g., Peak P12 in Table 4 of Example 4, and the RT63 peak in Table 15 of Example 8), and testing of compositions with tazobactam and ceftolozane formed by co-lyophilization of ceftolozane and tazobactam (e.g., 2 peak in Tables 12 and 13 of Example 7). Accordingly, there remains an unmet need to identify formulations and manufacturing methods that ively stabilize ceftolozane both in a solid and liquid form to provide suitably stable pharmaceutical compositions comprising ozane and ctam (both in a powder form for reconstitution and in a reconstituted form for parenteral delivery). These formulations should address the need to provide ceutical compositions having desired levels of ceftolozane and tazobactam potency, as well as levels of impurities that are therapeutically acceptable for parenteral administration.
SUMMARY As provided herein, ceftolozane can be stabilized in pharmaceutical composition comprising ceftolozane and a stabilizing effective amount of a stabilizing agent selected from the group consisting of: sodium chloride, dextran 40, lactose, maltose, trehalose and sucrose.
The ceutical compositions provided herein are based in part on the surprising discovery that ceftolozane pharmaceutical compositions comprising these stabilizing agents demonstrate improved ceftolozane residual rates (e. g., % ozane remaining after 3 days at 70 0C as measured by HPLC) and/or chemical stability (e. g., lower reduction in ceftolozane purity measured by HPLC after 7 days at 60 0C in a stability test) ed l samples comprising ozane without a stabilizing agent.
Accordingly, preferred pharmaceutical antibiotic compositions can include ceftolozane sulfate and a stabilizing agent (e.g., 300 to 500 mg of a stabilizing agent per 1,000 mg ceftolozane active) in a lyophilized unit dosage form (e. g., powder in a container).
The unit dosage form can be dissolved with a pharmaceutically acceptable carrier (e. g., 0.9% sodium chloride aqueous isotonic saline and/or water for injection), and then intravenously administered. In certain ceftolozane compositions, the stabilizing agent can be selected from the group consisting of: sodium chloride, lactose, maltose and dextran 40, and/or selected from the group consisting of: sodium chloride, trehalose and sucrose.
In addition, the present disclosure es ceftolozane pharmaceutical compositions based in part on the surprising discovery that ceftolozane pharmaceutical compositions comprising about1000 mg of ceftolozane active per 189 mg sodium from sodium chloride demonstrate improved chemical stability and purity compared with pharmaceutical compositions comprising ceftolozane with comparatively less sodium chloride. For example, the invention is based in part on the discovery of the absence of the "RT63 Impurity" (also referred to herein as "Formula III") in HPLC is of ceutical compositions comprising about 1,000 mg of ceftolozane and 189 mg sodium from sodium chloride. By comparison, reducing the amount of sodium chloride relative to ceftolozane in tested compositions ed in at least 1.5-fold greater impurity at RT: 63 minutes (observed by HPLC using a sil column ODS-UG-5 ; 5 micrometers; 250 X 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C). The ceftolozane formulations with reduced levels of sodium were not as stable as the ceftolozane ation ning ,000 mg of ceftolozane per 189 mg sodium from sodium de per. Ceftolozane ations ning about 1,000 mg of ceftolozane effective per stabilizing-effective amount of sodium from sodium chloride maintained the level of RT63 Impurity below the detection limit (e. g., 0.03%) measured by HPLC using a sil column ODS-UG-5; 5 micrometers; 250 X 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 3O mL/min flow rate and oven temperature of 45 0C.
In a further embodiment provided herein, ceftolozane sulfate is stabilized in pharmaceutical compositions by incorporation of an effective amount of an inorganic salt stabilizing agent, in particular 125 to 500 mg (e. g., 480 to 500 mg) of sodium chloride per gram of ceftolozane active. This is based in part on the surprising discovery that ceftolozane pharmaceutical compositions comprising 125 to 500 mg (e. g., 480 to 500 mg) of sodium chloride per 1000 mg of ceftolozane active demonstrate ed ceftolozane purity and chemical stability compared to pharmaceutical compositions comprising ceftolozane with comparatively less sodium chloride. For example, the disclosed pharmaceutical itions have an improved stability as a decrease in the rate of ceftolozane purity and/or a se in the rate of formation of substances characterized by HPLC peaks 1 and 7 identified during a 7-day stability study in Example 5. The disclosed ceftolozane pharmaceutical compositions comprise a izing amount of sodium chloride (e. g., 125 to 500 mg of sodium chloride [more specifically, 480 to 500 mg] per 1000 mg of ceftolozane active). Certain preferred compositions demonstrate improved ceftolozane purity (e. g., Table 6) and chemical stability (e. g., with respect to the ition of HPLC peak 1 in Table 7) compared with pharmaceutical compositions comprising ceftolozane with comparatively less sodium chloride. For example, the sed pharmaceutical compositions lly comprise less than about 4% total impurity after being stored for seven days at 60 0C, as determined by HPLC using a Develosil column ODS-UG-5 ; 5 micrometers; 250 x 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C. Alternatively, the sed ceutical compositions comprise less than about 2% of the impurity represented by Peak 1 after being stored for seven days at 60 0C, as determined by HPLC using a Develosil column ODS-UG-5 ; 5 eters; 250 x 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90: 10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C, where Peak 1 has a retention time relative to ceftolozane of 0.1.
In an embodiment, pharmaceutical antibiotic compositions provided herein can include ceftolozane sulfate and stabilizing amount of sodium chloride (e. g., 125 to 500 mg more specifically 480 to 500 mg of sodium chloride and 1,000 mg ceftolozane active) in a unit dosage form (e. g., powder in a container). The unit dosage form can be dissolved with a pharmaceutically acceptable carrier, and then intravenously administered.
In another aspect, provided herein is a pharmaceutical ition comprising 125 mg to 500 mg sodium de per 1,000 mg of ceftolozane active, n the decrease in ceftolozane total purity is not greater than about 4% after storing the pharmaceutical composition for seven days in a sealed container at 60 0C, as determined by HPLC using a Develosil column ODS-UG-5; 5 micrometers; 250 x 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C.
In another aspect, provided herein is a pharmaceutical composition comprising 125 mg to 500 mg sodium chloride per 1,000 mg of ceftolozane active, wherein the increase in the amount of the impurity represented by Peak 1 is not greater than about 2% after storing the pharmaceutical composition for seven days at 60 0C, as determined by HPLC using a Develosil column -5; 5 micrometers; 250 X 4.6 mm, a mobile phase of sodium perchlorate buffer on (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C, where Peak 1 has a retention time relative to ceftolozane of about 0.1.
In embodiments of these aspects, the pharmaceutical composition further comprises L-arginine, or citric acid. In other ments, the pharmaceutical composition is formulated for parenteral administration. In another embodiment, the itions can be in a unit dosage form comprising 125 mg to 500 mg sodium chloride, 1,000 mg of ceftolozane in the form of ceftolozane sulfate, L-arginine and citric acid.
In other embodiments of these aspects, the pharmaceutical ition is lyophilized.
In another embodiment, the ozane is ceftolozane sulfate.
In another aspect, provided herein is a unit dosage form injectable pharmaceutical composition comprising 125 mg to 500 mg sodium de and 1,000 mg of ceftolozane active present as a composition of formula (I) H3CjD/C02H Hsor INHg (I).
In another aspect, provided herein is a pharmaceutical composition sing 125 mg to 500 mg sodium chloride per 1,000 mg of ceftolozane active present as ceftolozane sulfate, wherein the ozane total purity is at least about 94% after storing the pharmaceutical ition for three days at 60 0C, as determined by HPLC using a Develosil column ODS-UG-5; 5 micrometers; 250 X 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C.
Applicants have further discovered pharmaceutical compositions sing ceftolozane and tazob actam with reduced and even undetectable amounts of the compound RRT 1.22, and methods of manufacturing these compositions. This is based in part on the discovery that the formation of RRT 1.22 can be reduced if not completely suppressed by lyophilizing ceftolozane in the absence of tazobactam and then blending the lyophilized ceftolozane with a dry tazobactam composition, such as a tazobactam composition lized in the absence of ceftolozane (See Example 10 and the results reported in Tables 23 and 24 ). Based on these s, pharmaceutical compositions sing ceftolozane and tazobactam, and pharmaceutical compositions prepared using ceftolozane and tazobactam are provided herein. In particular, these pharmaceutical compositions can include ceftolozane and/or tazobactam with reduced or even undetectable amounts of the compound RRT 1.22: W Ki 8 "fi‘\g<,§w,//w.tfl\ Np‘tt" 7:)" '0 gm 0 O/.- N gs: ix 1 \, \.:',./ \\--" \ NH2 I N. 0.1;; \‘OH In one embodiment, a pharmaceutical composition can include ceftolozane and tazobactam with less than 0.15%, 0.10%, 0.05% or 0.03% by weight; or from 0.03-0.05%, 0.03-0.1% or 0.03-0.15% by HPLC or even undectable amounts of RRT 1.22 (e.g., less than about 0.03% of the compound RRT 1.22 measured by HPLC). These pharmaceutical compositions can be obtained by a s comprising the steps of (a) lizing ceftolozane in the e of tazobactam to obtain a lized ozane composition; and (b) combining the lyophilized ceftolozane with ctam under conditions suitable to obtain said ceutical composition with the aforementioned purity levels. The combination of the lyophilized ceftolozane composition with tazobactam can include blending the lyophilized ceftolozane composition with lyophilized or crystalline tazobactam material.
Also provided herein is a pharmaceutical composition comprising a blend of separately lyophilized tazobactam and ceftolozane sulfate in an amount ing 1,000 mg of ceftolozane active per 500 mg of tazobactam active, further comprising less than 0.15 %, 0.10%, 0.05% or 0.03% by weight; from 0.03-0.05%, 0.03-0.1% or 0.03-0.15% by HPLC; or even undectable amounts (e.g., less than about 0.03% by HPLC) of a compound of formula (III) detectable at a ion time relative to ceftolozane of 1.22 by high performance liquid chromatography (HPLC) using a Develosil column ODS-UG-5; 5 micrometers; 250 X 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90: 10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C (hereinafter referred to as the "method of Example 1").
CXA-201 compositions comprising less than about 0.15%, 0.10%, 0.05% or 0.03% by weight; or from .05%, 0.03-0.1% or 0.03-0.15% by HPLC of the compound of formula (III) can be obtained by a process comprising the steps of: (a) forming a first aqueous solution comprising ceftolozane (e. g., in a pharmaceutically acceptable salt such as formula (I)), (b) lyophilizing the first aqueous solution to obtain a lyophilized ceftolozane composition, and (c) blending the lized ceftolozane composition with a tazobactam composition (e. g., tazobactam acid lyophilized in the absence of ceftolozane) in an amount that provides a 2:1 weight ratio between the amount of ceftolozane active and ctam active.
In yet another aspect, provided herein is a method for the treatment of a bacterial infection in a mammal, comprising administering to said mammal a therapeutically effective amount of any one of the pharmaceutical compositions provided . In an embodiment, the ial infection is caused by the bacterial ion is caused by bacteria selected from the group consisting of: Staphylococcus aureus, Escherichia coli, Acinetobacter baumam'i, Haemophilus influenzae, Klebsiella pneumonia, and Pseudomonas osa. In another embodiment, the bacterial infection is ed from the group consisting of nosocomial nia, complicated intra-abdominal infection and complicated y tract infection.
In yet another aspect, any of the pharmaceutical compositions provided herein may be used for the manufacture of a medicament for the treatment of complicated intra-abdominal infection (cIAI), complicated urinary tract infection (cUTI), or hospital acquired/ventilator- associated bacterial pneumonia (HABP/VABP).
In still another aspect ed herein, an otic pharmaceutical composition comprises ceftolozane (or a pharmaceutically acceptable salt thereof) and tazobactam (or a ceutically acceptable salt thereof) in a fixed dose combination of 1,000 mg of ceftolozane active per 500 mg of tazobactam active, and a ceftolozane-stabilizing amount of 125 mg to 500 mg sodium chloride per 1,000 mg of ceftolozane active.
In a further aspect disclosed herein, a pharmaceutical composition comprising stabilized ceftolozane sulfate is obtained by a process comprising lyophilizing an aqueous solution comprising 125 mg to 500 mg sodium chloride with an amount of ceftolozane sulfate providing 1,000 mg of ceftolozane active, to obtain the lized stabilized ceftolozane sulfate composition.
Yet another aspect ed herein discloses an antibacterial pharmaceutical composition comprising ceftolozane sulfate and tazobactam in a ratio of 1,000 mg ceftolozane active per 500 mg of tazobactam active, the ceutical composition obtained by a process comprising the steps of: a) lyophilizing a first aqueous solution in the absence of ctam, the first aqueous solution comprising ceftolozane sulfate prior to lyophilization to obtain a first lyophilized ceftolozane composition; and b) ng the first lyophilized ceftolozane ition with tazobactam to obtain an antibacterial composition comprising less than 0.13% by HPLC of a compound of formula (III) detectable at a retention time relative to ceftolozane of 1.22 by high mance liquid chromatography using a Develosil column ODS-UG-5 ; 5 micrometers; 250 X 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C HQCJ'T" ‘0" E H ‘3‘ l3 "M" f..- N N‘7‘ \ \ g, V K." ‘\, '3‘? _ .0: HEN \SJ - \L- \ N , \,..- 0 NH? 0’ OH 0"" "‘0 '" (III) BRIEF DESCRIPTION OF THE FIGURES Figure 1 is a flowchart showing the steps for preparing a CXA-201 composition sing ozane (referred to as CXA-101) and tazobactam using a ng process, wherein the ceftolozane and tazobactam are lyophilized separately prior to blending as described herein.
Figure 2 is a rt showing the steps for ing a CXA-201 ition sing ceftolozane (referred to as CXA-101) and tazobactam using a co-lyophilization process, as described herein.
Figure 3 is a reference HPLC chromatogram showing the peaks of ceftolozane (CXA- 101) and related composition peaks.
Figure 4 is a plot of the data points from Table 6, showing the purity of the ceftolozane in CXA-101 compositions at 60°C on day 0, day 1, day 3, and day 7, as measured by HPLC, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride.
Figure 5 is a plot of the data points from Table 7, showing the peak area of the composition peak 1 in CXA-101 compositions at 60 0C on day 0, day 1, day 3, and day 7, as measured by HPLC, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride.
Figure 6 is a plot of the data points from Table 8, showing the total peak area of the composition with a RRT of 0.43 and the composition peak 3 in CXA-101 compositions at 60 0C on day 0, day 1, day 3, and day 7, as measured by HPLC, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride.
Figure 7 is a plot of the data points from Table 9, showing the peak area of the ition peak 7 in CXA-101 compositions at 60 0C on day 0, day 1, day 3, and day 7, as ed by HPLC, wherein the CXA-101 compositions comprise ceftolozane and sodium chloride.
Figure 8 is a plot of the data points from Table 17, showing the purity of ceftolozane in CXA-201 itions at 60 0C on day 0, day 1, day 3, and day 7, as measured by HPLC, wherein the CXA-201 compositions comprise ceftolozane, tazobactam, and sodium chloride.
Figure 9 is a plot of the data points from Table 18, showing the peak area of the composition peak 1 in CXA-201 compositions at 60 0C on day 0, day 1, day 3, and day 7, as measured by HPLC, wherein the 1 compositions comprise ceftolozane, tazobactam, and sodium chloride.
Figure 10 is a plot of the data points from Table 19, showing the total peak area of the composition with a RRT of 0.43 and the composition peak 3 in CXA-201 compositions at 60 0C on day 0, day 1, day 3, and day 7, as measured by HPLC, wherein the CXA-201 compositions se ceftolozane, tazobactam, and sodium chloride.
Figure 11 is a plot of the data points from Table 20, showing the peak area of the composition peak 7 in CXA-201 compositions at 60 0C on day 0, day 1, day 3, and day 7, as ed by HPLC, wherein the CXA-201 compositions comprise ceftolozane, tazobactam, and sodium chloride.
Figure 12 is a flowchart showing the manufacturing process for a ceftolozane/tazobactam composition Via co-filling.
Figure 13a is a flowchart showing the process for preparing a CXA-201 composition sing ceftolozane (referred to as CXA-lOl) and tazobactam using a ng process in a ted production area according to FDA ce.
Figure 13b is a flowchart showing the process for ing a ceftolozane/tazobactam composition Via co-filling in a dedicated production area according to FDA Guidance.
Figure 14 shows the mass spectra obtained for the RRT 1.22 compound.
Figure 15 shows the al structures for certain peaks in the spectra in Figure 14.
DETAILED DESCRIPTION I. Stabilizing Ceftolozane Ceftolozane can be stabilized in a pharmaceutical ition comprising ceftolozane and a stabilizing effective amount of a stabilizing agent selected from the group consisting of: sodium chloride, dextran 40, lactose, maltose, trehalose and sucrose. The stabilizing agent and the stabilizing effective amount of the stabilizing agent for combination with ceftolozane were determined by high performance liquid chromatography (HPLC) analysis, for example by detecting the ratio of peak areas obtained for ceftolozane compared to peaks for other substances.
Preferred stabilized ceftolozane compositions have a ceftolozane residual rate of greater than the residual rate measured for a comparable ceftolozane composition without the izing agent. Unless otherwise indicated, the al rate is ed by detecting the amount of ceftolozane in a sample before and after a stability test using HPLC, and determining the percentage of ceftolozane last during the stability test.
Referring to Example 2 (including Table 2), the residual rate of ceftolozane in the l sample without a stabilizing agent (i.e., 100 mg of ceftolozane) after 3 days at 70 degrees C was 51.2%, g that the HPLC peak area after the stability test for ceftolozane was about 51.2% of the HPLC peak area for ceftolozane at the start of the stability test (i.e., 3 days at 70 s C). Sodium chloride, dextran 40, lactose and maltose all showed higher ceftolozane residual rates than the control in Example 2, while ceftolozane was less stable than the control when combined with fructose, xylitol, sorbitol and glucose (e. g., as evidenced by a al rate lower than that of the control). In one embodiment, stabilized ceftolozane compositions comprise ceftolozane (e. g., ceftolozane sulfate) and a stabilizing effective amount of a stabilizing agent selected from the group consisting of: sodium chloride, dextran 40, lactose and maltose, where the stabilizing effective amount es a residual rate of at least 51.2% for the ozane in the stabilized ceftolozane composition after 3 days at 70 degrees C. Preferably, the stabilized ceftolozane ceutical compositions after 3 days at 70 degrees C can comprise at least about 70% of an initial amount of the stabilized ceftolozane in the pharmaceutical composition (i.e., a residual rate of about 70% or greater, as shown in Example 2), where the % of ceftolozane is ed by high performance liquid chromatography (HPLC) according to Example 1.
Referring to Example 2 (Table 2a), stabilized ceftolozane compositions are characterized by a ion in ceftolozane of less than about 5% after 7 days at 60 degrees C, where the % ion of ceftolozane is measured by HPLC according to Example 1. The stabilized ozane pharmaceutical composition comprising ceftolozane and a stabilizing agent selected from the group consisting of: sodium de, trehalose and sucrose can lose less than 5% of the amount of ceftolozane after 7 days at 60 degrees C, where the % loss of ozane is measured by HPLC according to Example 1. Sodium chloride, trehalose and sucrose all showed reduced reductions in ceftolozane purity after a 7 day stability test at 60 s C (as measured by the % HPLC peak ponding to ceftolozane before and after the stability test). In one embodiment, stabilized ceftolozane compositions comprise ceftolozane (e.g., ceftolozane sulfate) and a stabilizing effective amount of a stabilizing agent selected from the group consisting of: sodium chloride, trehalose and sucrose, where the stabilizing effective amount provides a ion in ceftolozane purity of not more than about % (e.g, not more than about 4%) for the ceftolozane in the stabilized ceftolozane composition after 3 days at 70 degrees C.
Accordingly, in one aspect, provided herein is a pharmaceutical composition comprising stabilized ceftolozane obtained by a process comprising lyophilizing a composition including ceftolozane and a stabilizing agent selected from the group consisting of: sodium chloride, dextran 40, lactose, maltose, tehalose and sucrose, to obtain a 3O lized stabilized ceftolozane pharmaceutical composition. In an embodiment, the stabilizing agent is selected from the group consisting of: sodium chloride, trehalose and sucrose. In another aspect, provided herein is a pharmaceutical composition comprising stabilized ceftolozane and a stabilizing agent selected from the group consisting of: sodium chloride, n 40, e, maltose, tehalose and sucrose, wherein the pharmaceutical composition after 3 days at 70 degrees C comprises at least about 70% of an initial amount of the stabilized ozane in the pharmaceutical composition.
In another aspect, provided herein is a container containing a unit dosage form of a pharmaceutical composition formulated for parenteral administration for the treatment of complicated intra-abdominal ions or complicated urinary tract infections, the pharmaceutical composition comprising 1,000 mg of ceftolozane active, nine, citric acid and about 300-500 mg of a stabilizing agent selected from the group consisting of: sodium chloride, ose, and sucrose, wherein the pharmaceutical composition after 3 days at 70 degrees C comprises at least about 70% of an initial amount of the ceftolozane active in the pharmaceutical composition.
Various ozane compositions are described herein. One stabilized ceftolozane composition comprises ozane (e. g., ceftolozane sulfate), L-arginine, citric acid, and a stabilizing agent. Preferably, the stabilized ceftolozane composition comprises 1,000 mg of ceftolozane active, L-arginine and stabilizing-effective amount of the stabilizing agent. The stabilizing effective amount can be readily determined using HPLC and a stability test as disclosed herein. The stabilizing-effective amount can be effective to provide: (1) a residual rate measured by HPLC of ozane of at least about 51.2% (including, e. g., at least about 70%, and at least about 80%) after 3 days at 70 degrees C and/or (2) a reduction in ceftolozane purity measured by HPLC of not more than about 5.11% (including, e. g., reductions of not more than about 5%, or 4%) after 7 days at 60 degrees C. Examples of stabilizing ive s include 100mg — 500 mg of the stabilizing agent per 1,000 mg of the ozane active, more preferably about 300-500 mg of the stabilizing agent per 1,000 mg of the ceftolozane active.
In the screening of ceftolozane izing agents, it has been found that, surprisingly, a preferred amount of sodium chloride can improve the stability of ceftolozane, including ozane in the ceftolozane sulfate form. For example, in one experiment, a ceftolozane composition comprising about 100 mg (about 1.71 mmol) sodium chloride per 100 mg (about 0.15 mmol) of ceftolozane was more stable compared to many ozane compositions 3O comprising known stabilizing sugars, such as fructose, xylitol, sorbitol, glucose, and D- mannitol, and as stable as other ceftolozane compositions comprising the same amount of certain sugars, such as dextran 40, lactose, and maltose (see Example 2). Interestingly, additional ments trated that the use of maltose in a ceftolozane composition resulted in a significant amount of additional compounds (see Example 3).
Surprisingly, pharmaceutical compositions comprising ceftolozane and 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane have been ob served to t better chemical stability over the course of time and/or in the presence of heat, and fewer additional compounds than those pharmaceutical compositions comprising ceftolozane and less sodium chloride (i.e., less than 125 mg sodium chloride per 1000 mg of ceftolozane) (see, e. g., Example 5). In particular embodiments described herein, the pharmaceutical compositions comprising ceftolozane and 125 to 500 mg sodium chloride per 1000 mg of ceftolozane have been found to be more stable than the compositions comprising ceftolozane and less than 125 mg sodium chloride per 1000 mg of ceftolozane.
Ceftolozane compositions having 5 0-481 mg of sodium de per 1,000 mg ceftolozane active were prepared as described in Table 5 and tested for stability as described in Example 5. Ceftolozane was more stable in compositions containing at least 125 mg of sodium chloride per 1,000 mg of ceftolozane active, as measured by high performance liquid chromatography (HPLC) is by detecting the ratio of peak areas obtained for ceftolozane ed to peaks for other nces. s otherwise indicated, HPLC measurements reported herein are obtained using a Develosil column -5 ; 5 micrometers; 250 x 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90: 10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C.) During the stability test of Example 5, ozane samples containing 125 mg, 190 mg and 481 mg of sodium chloride per 1,000 mg of ceftolozane active showed a decrease in ceftolozane total purity measured by HPLC that was at least about 35% less than reductions in ceftolozane total purity observed for formulations containing 50 mg or 75 mg sodium chloride per 1,000 mg ceftolozane active. Thus, ceftolozane compositions having at least 125 mg or more sodium chloride relative to the fixed amount of ceftolozane were about 35-90% more stable than comparable ceftolozane compositions having less than 125 mg sodium chloride (e. g., the % se in ceftolozane for the sample containing 75 mg sodium chloride was about 35% greater than the comparable % decrease in ceftolozane for the sample containing 190 mg sodium chloride). In on, samples obtained from ceftolozane compositions containing 125 mg, 190 mg and 481 mg of sodium chloride per 1,000 mg of ceftolozane active showed a decrease in ceftolozane that was up to about 90% less than reductions in ceftolozane ed for formulations containing 50 mg or 75 mg sodium chloride per (e. g., the % decrease in ceftolozane for the sample containing 50 mg sodium chloride was about 90% greater than the comparable % decrease in ceftolozane for the sample containing 481 mg sodium chloride).
The ceftolozane sodium-stabilized compositions having 125 mg or more sodium chloride relative to the fixed amount of 1,000 mg ceftolozane active also had lower ties of onal substances identified by peaks 1 and 7 having characteristic retention times measured by HPLC (see Table 1, ting retention times of about 0.1 for peak 1 and about 1.3 for peak 7 relative to ozane measured according to the HPLC method of Example 1). In particular, these sodium chloride stabilized ceftolozane compositions were terized by about 37-94% less of the material of peak 1 and about 38-306% less of the material of peak 7 (measured by corresponding HPLC peak areas) than comparable ceftolozane compositions having less than 125 mg sodium chloride (e. g., see 7-day stability study in Example 5). Referring to the data in Table 7 (Figure 5 ), the amount of the composition of peak 1 (measured by HPLC according to Example 1) was measured by the % increase in the peak 1 HPLC peak during the 7-day stability test of Example 5.
In particular, samples containing 125 mg, 190 mg and 481 mg of sodium chloride per 1,000 mg of ceftolozane active showed at least a 37% reduction in the amount of the peak 1 composition observed for these formulations containing at least 125 mg sodium chloride per 1,000 mg ceftolozane , compared to the compositions with 50 mg or 75 mg sodium chloride per 1,000 mg of ceftolozane active (e.g., the % increase in peak 1 for the sample containing 75 mg sodium chloride was about 37% greater than the comparable % decrease in ozane for the sample containing 190 mg sodium chloride). In addition, compositions containing 125 mg, 190 mg and 481 mg of sodium chloride per 1,000 mg of ceftolozane active showed up to a 94% reduction in the amount of the peak 1 composition ob served for these formulations containing at least 125 mg sodium chloride per 1,000 mg ceftolozane active, compared to the compositions with 50 mg or 75 mg sodium chloride per 1,000 mg of ozane active (e. g., the % se in peak 1 for the sample containing 50 mg sodium chloride was about 94% greater than the able % decrease in ceftolozane for the sample containing 481 mg sodium de). 3O The formulation of pharmaceutical itions can be selected to minimize decomposition of the constituent drug substances and to produce a composition that is stable under a variety of storage conditions.
Provided herein are pharmaceutical compositions useful for the treatment of bacterial infections sing ceftolozane and sodium chloride, wherein the sodium chloride is present in an amount sufficient to ize the ceftolozane. Also provided herein are pharmaceutical compositions sing ceftolozane, tazobactam, and sodium chloride, wherein the sodium chloride is present in an amount sufficient to stabilize the ceftolozane.
Advantageously, these ceutical compositions have fewer additional compounds and are more chemically stable, and can therefore be stored for longer periods of time.
In one embodiment, provided herein is a pharmaceutical ition comprising ceftolozane and 125 mg sodium chloride per 1000 mg of ceftolozane, e. g., 125 to 500 mg sodium chloride per 1000 mg of ceftolozane, 0 mg sodium chloride per 1000 mg of ceftolozane, 300-500 mg sodium chloride per 1000 mg of ceftolozane, 0 mg sodium chloride per 1000 mg of ceftolozane, 0 mg sodium chloride per 1000 mg of ceftolozane, 460-500 mg sodium chloride per 1000 mg of ceftolozane, or about 476 mg sodium chloride per 1000 mg of ceftolozane, wherein the purity of the ceftolozane in the composition is 75% or greater after 3 days at 70 0C. In another embodiment, provided herein is a pharmaceutical composition comprising ceftolozane and about 487 mg sodium chloride per 1000 mg of ozane, wherein the purity of the ceftolozane in the composition is 75% or greater after 3 days at 70 0C. In certain embodiments, the purity of the ceftolozane in the composition is 80% or greater, 85% or greater, 90% or greater, 95% or greater, 97% or greater, or 99% or greater after 3 days at 70 0C.
In another embodiment, provided herein is a pharmaceutical composition comprising ceftolozane and 125 mg sodium chloride per 1000 mg of ceftolozane, e. g., 125 to 500 mg sodium chloride per 1000 mg of ceftolozane, 200-500 mg sodium chloride per 1000 mg of ceftolozane, 0 mg sodium chloride per 1000 mg of ceftolozane, 0 mg sodium chloride per 1000 mg of ceftolozane, 450-500 mg sodium chloride per 1000 mg of ceftolozane, 460-500 mg sodium chloride per 1000 mg of ceftolozane, or about 476 mg sodium chloride per 1000 mg of ceftolozane, wherein the purity of the ceftolozane in the composition is 94.8% or greater after 3 days at 60°C. In another ment, provided herein is a pharmaceutical composition comprising ceftolozane and about 487 mg sodium chloride per 1000 mg of ozane, wherein the purity of the ceftolozane in the composition is 94.8% or r after 3 days at 60°C. In certain embodiments, the purity of the ceftolozane in the composition is 95% or greater, 96% or greater, 97% or greater, 98% or greater, or 99% or greater after 3 days at 60 0C.
In still another embodiment, provided herein is a pharmaceutical composition comprising ceftolozane and 125 mg sodium chloride per 1000 mg of ceftolozane, e. g., 125 to 500 mg sodium chloride per 1000 mg of ceftolozane, 200-500 mg sodium chloride per 1000 mg of ozane, 300-500 mg sodium chloride per 1000 mg of ceftolozane, 0 mg sodium chloride per 1000 mg of ceftolozane, 0 mg sodium chloride per 1000 mg of ceftolozane, 460-500 mg sodium chloride per 1000 mg of ceftolozane, or about 476 mg sodium chloride per 1000 mg of ceftolozane, wherein the purity of the ceftolozane in the composition decreases by 3.1% or less after 3 days at 60°C. In another ment, provided herein is a pharmaceutical ition comprising ceftolozane and about 487 mg sodium chloride per 1000 mg of ozane, wherein the purity of the ceftolozane in the composition decreases by 3.1% or less after 3 days at 60°C. In certain embodiments, the purity of the ceftolozane in the composition decreases by 3.0% or less, 2.5% or less, 2.0% or less, 1.5% or less, or 1% or less after 3 days at 60°C.
In another , provided herein is a ceutical composition comprising about 1,000 mg of ceftolozane active per 189 mg sodium from sodium chloride, and not more than 0.03% by high performance liquid chromatography (HPLC) of a RT63 Impurity at a retention time of about 63 minutes observed by HPLC using a Develosil column ODS-UG-5; 5 micrometers; 250 X 4.6 mm, a mobile phase of sodium perchlorate buffer on (pH 2.5)/CH3CN 90: 10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 °C., the pharmaceutical composition obtained by a process comprising the step of lyophilizing an aqueous solution comprising 189 mg sodium from sodium chloride per 1,000 mg of ceftolozane active in the form of ceftolozane sulfate to obtain a lyophilized ceftolozane composition, and formulating the pharmaceutical composition from the lyophilized ceftolozane ition.
In one embodiment, the pharmaceutical composition comprises a total of 1,000 mg of ceftolozane active. In another aspect, provided herein is a pharmaceutical composition obtained by a process comprising the step of lyophilizing an aqueous solution comprising 189 mg sodium from sodium de per 1,000 mg of ozane in the form of ceftolozane sulfate to obtain a lyophilized ceftolozane composition.
In one embodiment, the pH of the aqueous solution is 5.0 to 7.0, e.g., 6.0 to 7.0, and the aqueous solution further comprises L-arginine. In another embodiment, the pharmaceutical composition is formulated for parenteral administration and further comprises citric acid. In another embodiment, the composition is a unit dosage form in a container comprising tazobactam and 189 mg sodium from sodium chloride per 1,000 mg of ceftolozane active in the form of ceftolozane sulfate.
In another embodiment, the aqueous solution further comprises L-arginine and citric acid; the pH of the s solution is 6.0 to 7.0 prior to lyophilization; and the ceutical composition further comprises tazobactam blended with the lyophilized ceftolozane composition.
In still another aspect, provided herein is a container containing a unit do sage form of a pharmaceutical composition formulated for parenteral administration for the treatment of complicated intra-abdominal infections or complicated urinary tract infections, the pharmaceutical composition comprising 189 mg sodium from sodium chloride, and 1,000 mg ceftolozane active in the form of ceftolozane sulfate.
In one embodiment, the container comprises the ceftolozane sulfate, tazobactam and the sodium de and not more than 0.03% by high performance liquid chromatography (HPLC) of a RT63 Impurity at a ion time of about 63 s observed by HPLC using a Develosil column ODS-UG-S; 5 micrometers; 250 X 4.6 mm, a mobile phase of sodium orate buffer solution (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C.
Typically, antibiotic compositions do not contain sodium chloride or contain only a small amount of sodium chloride. For example, Maxipime®, which is approved for pneumonia, c therapy for febrile neutropenic ts, uncomplicated and complicated urinary tract infections, uncomplicated skin and skin structure infections, and complicated intra-abdominal ions, is a dry mixture of cefepime hydrochloride and L-arginine, wherein the mixture does not contain sodium chloride. lin® for injection, which is approved for respiratory tract infections, urinary tract infections, skin and skin ure ions, y tract infections, bone and joint infections, l infections, septicemia, and endocarditis and perioperative prophylaxis, comprises lyophilized cefazolin sodium that does not contain additional sodium salt. Furthermore, Rocephin®, which is approved for lower respiratory tract infections, acute bacterial otitis media, skin and skin structure infections, urinary tract infections, licated gonorrhea, pelvic inflammatory disease, bacterial septicemia, bone and joint infections, intra-abdominal infections, meningitis, and surgical prophylaxis, comprises ceftriaxone sodium that only comprises 13.5 mg of free sodium per 1000 mg of ceftriaxone sodium, which equals about 34 mg sodium chloride per 1000 mg of ceftriaxone sodium if the free sodium is in sodium chloride form. In contrast, the pharmaceutical compositions provided herein (compositions comprising ceftolozane and sodium chloride, and compositions comprising ceftolozane, ctam, and sodium de), have high amounts of sodium chloride, e.g., 125 -1000 mg sodium chloride per 1000 mg of ceftolozane.
Ceftolozane The compound 5-amino{ [(2-aminoethyl)carbamoyl]amino}{[(6R,7R)({(2Z)- 2-(5-amino-l,2,4- thiadiazol—3-yl)[(l-carboxy-l-methylethoxy)imino]acetyl}amino) carboxy—8-oxo-5 - thia- l -azabicyclo[4.2.0]octenyl]methyl } - 1 l- azolium monosulfate (also known also as ceftolozane sulfate, FR264205, "CXA-lOl") is a cephalosporin compound (shown below), the synthesis of which is described in US. Patent No. 7,129,232, wherein the compound is also named 7B-[(Z)(5-amino-l,2,4-thiadiazol—3- yl)(l -carboxy— l -methylethoxyimino)acetamido]—3- {3 -amino [3-(2-aminoethyl)ureido]— 2-methyl—l-pyrazolio}methylcephemcarboxylate. Ceftolozane has the al formula below and CAS registry number 6892933. "Ceftolozane" can be provided as the salt, ceftolozane sulfate.
Ceftolozane sulfate Unless otherwise indicated herein, the phrase "1000 mg ceftolozane" or "1 g ceftolozane" refers to an amount of ceftolozane containing the free base equivalent weight of ceftolozane provided in the free base form or any suitable salt form, as appropriate. For example, a composition containing 1000 mg of ceftolozane in the ceftolozane sulfate solid form will include greater than 1000 mg of material (e. g., due to at least the additional weight of the sulfate counter ion). ably, the ceftolozane is present as ozane sulfate. If a ceftolozane sulfate composition contains "1000 mg of ceftolozane" then it includes an amount of ceftolozane sulfate comprising 1000 mg of the ceftolozane molecule in free base equivalent form. For example, as shown in Table 29, 1147 mg ceftolozane sulfate ponds to 1000 mg of ceftolozane free base.
In another embodiment, "1000 mg ceftolozane" refers to an amount of ceftolozane that is considered a bioequivalent by the United States Food and Drug Administration (FDA), i.e. for which 90% CI of the relative mean Cmax, AUC(0-t) and AUC(0-oo) is within 80.00% to 125.00% of the reference formulation in the fasting state (see: "Guidance for Industry: Bioavailability and Bioequivalence Studies for Orally Administered Drug Products — General Considerations". Center for Drug Evaluation and Research, United States Food and Drug Administration, 2003).
"Ceftolozane active" refers to the active n of a salt form of ceftolozane, i.e., the free base form of ceftolozane.
As used herein, "125 to 1000 mg sodium chloride per 1000 mg of ceftolozane" refers to a ratio of sodium chloride to ceftolozane free base equivalent. For example, "125 to 1000 mg sodium de per 1000 mg of ceftolozane" includes, for example, 62.5 to 500 mg sodium chloride per 500 mg of ceftolozane, as well as, for example, 25 to 200 mg sodium chloride per 200 mg ceftolozane, etc.
In r aspect, provided herein is a method for the treatment of bacterial infections in a , comprising administering to said mammal a eutically effective amount of a pharmaceutical composition sing ceftolozane as described herein.
II. Ceftolozane in the Presence of ctam It has also been observed that pharmaceutical compositions sing ceftolozane, tazobactam, and 125 to 1000 mg sodium chloride per gram of ceftolozane exhibit better chemical stability and fewer additional compounds than those pharmaceutical compositions comprising ceftolozane and tazobactam, but less sodium chloride (see, e. g., Example 8). In ular embodiments described herein, the pharmaceutical compositions comprising ozane, tazobactam, and 125 to 500 mg sodium de per 1000 mg of ceftolozane have been found to be more stable than the compositions comprising ceftolozane, tazobactam, and less than 125 mg sodium chloride per gram of ceftolozane.
Adding high amounts of sodium chloride to CXA-201 compositions (e. g., 125-1000 mg sodium chloride per 1000 mg of ceftolozane, 125-500 mg sodium chloride per 1000 mg of ceftolozane, 200-500 mg sodium chloride per 1000 mg of ceftolozane, 300-500 mg sodium chloride per 1000 mg of ceftolozane, 400-500 mg sodium de per 1000 mg of ceftolozane, 450-500 mg sodium chloride per 1000 mg of ceftolozane, 460-500 mg sodium chloride per 1000 mg of ceftolozane, or about 476 mg sodium chloride per 1000 mg of ceftolozane) also inhibits the formation of certain additional nds. Adding about 487 mg sodium chloride per 1000 mg of ceftolozane to CXA-201 ition can also inhibit the formation of certain additional nds. For example, in one experiment, CXA-201 itions comprising 125 -481 mg sodium chloride per 1000 mg ceftolozane developed a reduced amount of a composition having a retention time of 63 minutes ("RT 63’") after three months at 25 0C (see the HPLC measurements shown in Example 8A).
Accordingly, in one aspect, provided herein is a pharmaceutical composition comprising ceftolozane, tazobactam, and 00 mg sodium chloride per 1000 mg of ceftolozane, e. g., 125-500 mg sodium de per 1000 mg of ceftolozane, 200-500 mg sodium chloride per 1000 mg of ceftolozane, 300-500 mg sodium chloride per 1000 mg of ceftolozane, 400-500 mg sodium chloride per 1000 mg of ceftolozane, 450-500 mg sodium chloride per 1000 mg of ceftolozane, 460-500 mg sodium chloride per 1000 mg of ozane, or about 476 mg sodium chloride per 1000 mg of ceftolozane. In another embodiment, provided herein is a pharmaceutical composition comprising ceftolozane, ctam, and about 487 mg sodium chloride per 1000 mg of ceftolozane. In another embodiment, provided herein is a pharmaceutical ition comprising ceftolozane, tazobactam, and 125-1000 mg sodium chloride per 1000 mg of ceftolozane, e.g., 125 -500 mg 3O sodium de per 1000 mg of ceftolozane, 200-500 mg sodium chloride per 1000 mg of ceftolozane, 300-500 mg sodium chloride per 1000 mg of ceftolozane, 400-500 mg sodium chloride per 1000 mg of ceftolozane, 450-500 mg sodium chloride per 1000 mg of ceftolozane, 460-500 mg sodium chloride per 1000 mg of ceftolozane, or about 476 mg sodium chloride per 1000 mg of ceftolozane, n the purity of the ceftolozane in the composition is 94.9% or greater after 3 days at 60°C. In another embodiment, provided herein is a pharmaceutical composition comprising ceftolozane, tazobactam, and about 487 mg sodium chloride per 1000 mg of ceftolozane, wherein the purity of the ceftolozane in the composition is 94.9% or greater after 3 days at 60°C. In certain embodiments, the purity of the ceftolozane in the composition is 95% or greater, 96% or r, 97% or greater, 98% or greater, or 99% or greater after 3 days at 60°C.
Tazobactam The compound (28,3S,5R)methyloxo(l H- l ,2,3 -triazol- l -ylmethyl)thia- l - azabicyclo[3.2.0]heptane-2—carboxylic acid 4,4-dioxide (also known as ctam) is a B- ase inhibitor of the following structure: As used herein, tazobactam can be a free acid, a sodium salt, an arginine salt, or a hydrate or solvate thereof. The phrases "250-750 mg tazobactam", "250-700 mg tazobactam," "300-700 mg tazobactam", "300-650 mg ctam", "350-650 mg tazobactam", "350-600 mg tazobactam", "400-600 mg tazobactam", "400-550 mg ctam", "450-550 mg tazobactam" or "about 500 mg tazobactam" refer to an amount of tazobactam containing the free acid equivalent weight of tazobactam ed in the free acid form or any suitable salt form. For e, a composition containing 500 mg of tazobactam in the tazobactam sodium solid form will include greater than 500 mg of material (e.g., due to at least the additional weight of the sodium counter ion). For example, as shown in Table 29, 537 mg tazobactam sodium corresponds to 500 mg of tazobactam free acid. Preferably, the tazobactam is present as tazobactam sodium. If a tazobactam sodium composition contains "500 mg of tazobactam" then it includes an amount of tazobactam sodium comprising 500 mg of the tazobactam molecule in free acid equivalent form.
In an embodiment, the tazobactam is tazobactam sodium e . In yet a further embodiment, the tazobactam sodium sterile powder is generated by neutralizing ctam acid with sodium bicarbonate followed by lyophilization.
As used herein, the term "tazobactam active" refers to the active portion of a salt form of ctam, i.e., tazobactam free acid.
In certain embodiments, the pharmaceutical compositions further comprise ctam sodium at a quantity equivalent of 500 mg of tazobactam free acid in a lyophilized powder form per 1000 mg of ceftolozane (anhydrous, free base equivalent).
III. Liquid Pharmaceutical Compositions or Formulations In another aspect, provided herein is a liquid pharmaceutical composition (e. g., an enous infusion solution) comprising ceftolozane and tazobactam, wherein the composition is suitable for intravenous administration. In one embodiment, the composition further comprises 125-1000 mg sodium chloride per 1000 mg of ceftolozane. In another embodiment, the composition further comprises 125 -500 mg sodium chloride per 1000 mg of ceftolozane. In an embodiment, the liquid pharmaceutical composition (e. g., an enous infusion solution) is prepared by titution of a ceftolozane and tazobactam composition with sterile water and/or normal sterile saline, followed by dilution with sterile water and/or normal sterile saline. In an embodiment, the liquid pharmaceutical composition (e. g., an intravenous infusion solution) is prepared by reconstitution of a ceftolozane and tazobactam ition with normal sterile , followed by dilution with normal sterile saline. In another embodiment, the liquid pharmaceutical composition (e. g., an intravenous infusion solution) has an osmolality n about 300 mOsm/kg and 900 mOsm/kg, ing injectable formulations with an osmolality of 0 mOsm/kg to 350-800 mOsm/kg, 400- 500 mOsm/kg and 500-600 mOsm/kg. In a further embodiment, the liquid pharmaceutical composition (e. g., an intravenous infusion solution) comprising 1,000 mg ceftolozane active and 500 mg of ctam active (as pharmaceutically acceptable salts thereof) has an osmolality that is between about 400 mOsm/kg and 500 mOsm/kg (e. g,, 446-478 mOsm/kg, 0 mOsm/kg, 420-490 mOsm/kg). In a further embodiment, the liquid pharmaceutical composition (e. g., an enous infusion solution) comprising 2,000 mg ceftolozane active and 1000 mg of tazobactam active (as pharmaceutically able salts thereof) has an osmolality that is between about 500 mOsm/kg and 650 mOsm/kg. In yet a further embodiment, the liquid pharmaceutical ition (e. g., an intravenous infusion solution) has an osmolality that is less than about 600 mOsm/kg (e. g,, 290-610 mOsm/kg, 350-605 mOsm/kg, 550-605 mOsm/kg, 589-604 mOsm/kg). In another embodiment, the ceftolozane and tazobactam of the liquid pharmaceutical composition (e.g., an enous infusion on) are controlled to pH 5 to 7. In a r embodiment, the ceftolozane and tazobactam of the liquid pharmaceutical ition (e. g., an intravenous infusion solution) are controlled to about pH 6.
In one embodiment, the methods further comprise reconstituting the lyophilized mixture in an s solvent, such that the resulting solution is suitable for infusion. The mixture can be reconstituted in saline and/or sterile water for injection.
Methods of Preparing Pharmaceutical Compositions Comprising Ceftolozane and Sodium Chloride Pharmaceutical compositions comprising ceftolozane and stabilizing-effective amount of a stabilizing agent can be obtained by lization. As is known to those skilled in the art, lyophilization is a process of freeze-drying in which water is sublimed from a frozen solution of one or more solutes. Specific methods of lization are described in ton’s Pharmaceutical Sciences, Chapter 84, page 1565, Eighteenth Edition, A. R.
Gennaro, (Mack Publishing Co., Easton, Pa., 1990). A pharmaceutical composition sing ceftolozane can be prepared by adding a stabilizing amount of sodium de in a fixed ratio to ceftolozane in an aqueous solution prior to lyophilization, then lyophilizing the solution to obtain a lyophilized composition comprising sodium chloride and ceftolozane.
In particular, the pharmaceutical antibiotic compositions can include stabilized ceftolozane sulfate obtained by a process sing the steps of lyophilizing an aqueous solution containing ceftolozane and a stabilizing-effective amount of a stabilizing agent, where the stabilizing-effective amount of the stabilizing agent is about 100 to 500 mg (preferably 300-500 mg) of the stabilizing agent per 1,000 mg ceftolozane active in the aqueous solution prior to lyophilization. A eutically effective amount of ceftolozane (e. g., ceftolozane sulfate) and a stabilizing-effective amount of the stabilizing agent can ved in an aqueous solution that can be lyophilized to obtain a stabilized ceftolozane pharmaceutical composition.
The method can further comprise the steps of: (1) forming a solution comprising sodium chloride and ceftolozane or a salt thereof followed by lyophilizing the solution; and (2) combining the lyophilized ceftolozane with other components (e. g., a B-lactamase inhibitor, such as tazobactam, or a lyophilized B-lactamase inhibitor, such as a lyophilized tazobactam) to obtain the pharmaceutical composition. The ing pharmaceutical composition can be a powder for reconstitution to obtain an injectable pharmaceutical ition that can be intravenously administered to a patient. In yet a further embodiment, the method ses adding 189 mg sodium from sodium chloride per 1000 mg of ceftolozane active in an aqueous solution, then lyophilizing the solution to obtain a lyophilized material comprising sodium chloride and ceftolozane sulfate in a ratio effective to provide a product with less than 0.03% of the RT63 Impurity as detected by HPLC in Example 2.
A. Blending In other embodiments, ceutical compositions sing ceftolozane can be obtained by methods that include the steps of: (1) adding a stabilizing amount of sodium chloride to ceftolozane ally ed by co-lyophiliZing or spray drying the ceftolozane and sodium de; and (2) combining the product of step (1) with other components. For e, the product of step (1) can be combined with a B-lactamase inhibitor, such as tazobactam (CAS#: 04-9), avibactam (CAS# 11925004), Sulbactam (CAS# 683738) and/or clavulanate (CAS# 580018). The beta lactamase inhibitor can be included in a crystalline or amorpous form, such as a lyophilized tazobactam or crystalline tazobactam (e. g., US Patent Nos. 8,476,425 and 603) to obtain the pharmaceutical composition.
Pharmaceutical compositions comprising ceftolozane and tazobactam with reduced or even undectable levels of the compound of RRT 1.22 (e.g., including levels of RRT 1.22 that are not detectable by HPLC according to Example 1 and/or comprise less than 0.15%, 0.10%, 0.05% or 0.03% by weight; or from 0.03-0.05%, 0.03-0.1% or 0.03-0.15% by HPLC according to Example 1) can be obtained by ng a first composition comprising a therapeutically effective amount of ceftolozane in the absence of tazobactam with a second composition comprising a therapeutically effective amount of tazob actam in the absence of ceftolozane to form a blended pharmaceutical composition.
Without being bound by theory, the compound RRT 1.22 can be formed by a reaction between ceftolozane and formylacetic acid, a by-product of tazobactam as illustrated in Marunaka et al. (Chem. Pharm. Bull. 1988, Vol. 36 (11), pp. 4478-4487.
Figure 1 is a flowchart showing the steps for preparing a CXA-201 composition 3O comprising ceftolozane (referred to as 1) and tazobactam using a blending process, wherein the ozane and tazobactam are lyophilized separately prior to ng as described herein.
The (first) ceftolozane composition can be prepared in the absence of tazob actam by forming a first aqueous solution comprising ceftolozane sulfate and other components including excipients, stabilizers, pH adjusting additives (e.g., buffers) and the like. Non- lirniting examples of these ves include sodium chloride, citric acid and L-arginine. For e, the use of sodium chloride results in greater stability; L-arginine is used to adjust pH of the aqueous solution (e.g., to pH 6-7) and to increase the solubility of ozane; and citric acid is used to prevent discoloration of the product, due to its ability to chelate metal ions. Preferably, the first aqueous solution ses about 125 mg — 500 mg sodium de per 1,000 mg of ceftolozane active. The ceftolozane can be included as an amount of ceftolozane sulfate of formula (1) containing at least about 1,000 mg ozane active.
The (first) aqueous on is then lized to form a first lyophilized ceftolozane composition, which is combined with tazobactam, e. g., the lyophilized tazobactam (e.g., lyophilized tazobactam sodium) or crystalline tazobactam.
The (second) ctam composition can be prepared in the absence of ozane by forming a second solution comprising tazobactam. The tazobactam can be included in an amount providing about 500 mg of tazobactam active per 1,000 mg ceftolozane active (i.e., a 1:2 weight ratio of tazobactam active to ceftolozane active). Unless otherwise indicated, tazobactam can be a free acid, a sodium salt, an arginine salt, or a hydrate or solvate thereof.
In one embodiment, the tazobactam in the d) tazobactam composition is ctam acid and the second composition further comprises sodium bicarbonate or sodium hydroxide.
Lyophilizing tazobactam in the presence of sodium bicarbonate or sodium hydroxide forms a lyophilized tazobactam sodium, which can then be further blended with the (first) lyophilized ceftolozane composition.
Pharmaceutical compositions with reduced or able amounts of the compound of RRT 1.22 can be obtained by lyophilizing ceftolozane without formylacetic acid and/or tazobactam under conditions that prevent formation of RRT 1.22 (e.g., Example 9). The presence of RRT 1.22 can be detected by HPLC (e.g., Examples 1, 6 and 7). Specific methods of lyophilization are bed in Remington’s Pharmaceutical es, Chapter 84, page 1565, Eighteenth Edition, A. R. Gennaro, (Mack Publishing Co., Easton, Pa., 1990).
The formation of the compound of formula (111) can be avoided by preventing the on of ceftolozane and formylacetic acid. In one embodiment, the compound of formula (111) can be suppressed by separately lyophliZing ceftolozane e and tazobactam in separate solutions, and then blending the lyophilized compositions to form a pharmaceutical composition.
In one aspect, antibiotic pharmaceutical compositions comprising ceftolozane and tazobactam with less than about 0.15%, 0.10%, 0.05% or 0.03% by weight; or from 0.03- 0.05%, .l % or 0.03-0.15% by HPLC of the compound of formula (III) are obtained by a process comprising the steps of: (a) lyophilizing ceftolozane in the absence of tazobactam to obtain a lyophilized ceftolozane composition, and (b) blending the lyophilized ceftolozane composition with a composition comprising tazobactam under conditions suitable for attaining the aforementioned purity levels, e.g., by blending with crystalline ctam or lized tazobactam.
In r aspect, otic pharmaceutical compositions comprising ceftolozane and tazobactam and less than about 0.15%, 0.10%, 0.05% or 0.03% by weight; or from 0.03- 0.05%, 0.03-0.l % or 0.03-0.15% by HPLC of the compound of a (III) are obtained by a s comprising the steps of: (a) lyophilizing tazobactam in the absence of ceftolozane to obtain a lyophilized tazobactam composition, and (b) blending the lyophilized tazob actam composition with a composition comprising ceftolozane (e.g., lyophilized ceftolozane sulfate).
In a third aspect, antibiotic pharmaceutical compositions sing ceftolozane and tazobactam and less than about 0.15%, 0.10%, 0.05% or 0.03% by weight; or from 0.03- 0.05%, .l % or .15% by HPLC of the compound of formula (III) are obtained by a s comprising the steps of: (a) lyophilizing tazobactam in the absence of ceftolozane to obtain a lyophilized ctam composition, (b) lyophilizing ceftolozane in the absence of tazobactam to obtain a lyophilized ceftolozane composition, and (c) blending the lyophilized tazobactam composition with the lyophilized ceftolozane composition.
Pharmaceutical compositions comprising the compound of formula (III), ceftolozane and tazobactam can be formulated to treat infections by parenteral administration (including subcutaneous, intramuscular, and intravenous) administration. Pharmaceutical compositions may additionally comprise excipients, stabilizers, pH adjusting additives (e.g., s) and the like. Non-limiting examples of these additives include sodium chloride, citric acid and L- arginine. For example, the use of sodium de results in greater stability; L-arginine is used to adjust pH and to increase the solubility of ceftolozane; and citric acid is used to prevent discoloration of the product, due to its ability to chelate metal ions. In one particular embodiment, the pharmaceutical compositions described herein are formulated for administration by intravenous injection or on.
Other pharmaceutical antibiotic compositions can include ceftolozane sulfate and the compound of formula (III). For example, pharmaceutical compositions comprising 0.13%, 0.15%, 0.30%, 0.38%, 0.74% or 0.97% of the compound of formula (III) are herein. The pharmaceutical antibiotic compositions can be provided in a unit do sage form (e. g., in a container). The unit do sage form can be dissolved with a pharmaceutically acceptable carrier, and then intravenously administered. The unit dosage form comprises 1000 mg of ceftolozane active and 500 mg tazobactam, typically 1000 mg ceftolozane active as ceftolozane sulfate and 500 mg of tazobactam active as tazobactam sodium, nate or free acid. The unit dosage forms are commonly stored in containers.
In another aspect, provided herein is a unit dosage form of a pharmaceutical composition comprising 1,000 mg ceftolozane and 500 mg tazobactam, the pharmaceutical composition ated for parenteral administration for the treatment of complicated intra- abdominal infections or complicated urinary tract infections, the pharmaceutical composition comprising ceftolozane sulfate and tazob actam, obtained by a process comprising the steps of: lyophlizing an s solution to obtain a lyophilized ozane composition, wherein the aqueous solution comprises water, ceftolozane sulfate, 125 -500 mg sodium chloride per 1,000 mg ceftolozane active in the aqueous solution, an amount of L-arginine to provide a pH of 6-7 in the solution prior to lyophilization; and blending the lyophilized ceftolozane composition with a lyophilized tazob actam composition in an amount ing the ratio of about 500 mg tazobactam free acid per 1,000 mg of ceftolozane active to obtain the unit dosage form.
Another embodiment of the invention is a container containing a unit dosage form of a pharmaceutical ition formulated for parenteral administration for the treatment of complicated intra-abdominal infections or complicated y tract ions. The container can be obtained by a s sing the steps of: a) lyophilizing an aqueous solution comprising 189 mg sodium from sodium chloride per 1000 mg ozane active in the form of ceftolozane sulfate and further comprising citric acid, and L-arginine to obtain a 3O lyophilized ozane composition; and b) filling a sufficient quantity of the lyophilized composition into a container to obtain a unit dosage form comprising 189 mg sodium from sodium chloride and 1,000 mg of ceftolozane active in the form of ozane sulfate. In one aspect, the pH of the aqueous solution is 6.0 to 7.0. In another aspect the pharmaceutical composition is formulated for parenteral administration by reconstituting the pharmaceutical composition in the container (e.g., with 10 mL of diluent such as water for injection or isotonic saline) followed by addition of the reconstituted pharmaceutical composition to a r for injection (e.g., about 100 mL of isotonic saline or other pharmaceutically acceptable carrier for enous administration). Optionally, the container is also filled with tazobactam (e.g., a lyophilized tazobactam such as tazobactam sodium). In yet another , the pharmaceutical composition is a liquid ition comprising 189 mg sodium from sodium chloride, 1,000 mg of ceftolozane active and tazobactam in an amount providing about 500 mg tazobactam acid equivalent per 1,000 mg of ceftolozane active, formulated for eral administration and the pH of the aqueous solution is 6.0 to 7.0.
The ceutical composition in the container can also be a Ceftolozane/Tazobactam for Injection Drug t, 1000 mg/500 mg. It is presented as a combination of two sterile active powders in a single container intended for titution and intravenous infusion. In an embodiment, the drug product is prepared by converting ceftolozane sulfate to a sterile drug product intermediate (composition) powder with excipients citric acid, sodium chloride and L-arginine. This is can done by lyophilization, as described herein. Tazobactam sodium drug nce can be presented as a sterile powder without any excipients. The tazobactam sodium drug substance can be lyophilized, spray dried or provided as a crystalline material. The drug product is then prepared by cally filling the two powders (e.g., the two separately lyophilized drug powders) tially into a single container.
In an embodiment, the container of ceftolozane/tazobactam for injection contains approximately 2255 mg ceftolozane sterile composition powder that contains 1147 mg ceftolozane sulfate, which is lent to 1000 mg ceftolozane free base, as well as approximately 537 mg ctam sodium e drug substance, equivalent to 500 mg tazobactam free acid. At the time of administration, the container is reconstituted with 10 mL vehicle, sterile 5% Dextrose Injection USP, Water for Injection or 0.9% Sodium de Injection USP, then the container contents further diluted in an infusion bag of 0.9% Sodium Chloride Injection USP or 5% Dextrose Injection USP, for administration. The constituents 3O are shown in Table 29.
A pharmaceutical composition can include ceftolozane sulfate and tazobactam in an amount providing 1,000 mg of ceftolozane active per 500 mg of tazobactam active, and 0.03% to 0.15% by HPLC of a compound of formula (III) detectable at a retention time relative to ceftolozane of 1.22 by high performance liquid chromatography using a Develosil column -5 ; 5 micrometers; 250 X 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C. Optionally, the pharmaceutical composition can further include 125 mg to 500 mg of sodium chloride per 1,000 mg of ceftolozane active, and L-arginine. The tazobactam in the composition can be tazobactam sodium.
In one embodiment of these methods of preparing, 125-500 mg sodium chloride per 1000 mg of ceftolozane is combined. In another embodiment of these methods of preparing, the amount of the sodium chloride ed is 200-500 mg sodium de per 1000 mg of ceftolozane, 300-500 mg sodium chloride per 1000 mg of ceftolozane, 400-500 mg sodium chloride per 1000 mg of ceftolozane, 450-500 mg sodium chloride per 1000 mg of ceftolozane, 0 mg sodium de per 1000 mg of ozane, or about 476 mg sodium chloride per 1000 mg of ceftolozane. In another embodiment of these methods of preparing, the amount of the sodium chloride combined is about 487 mg sodium chloride per 1000 mg of ceftolozane.
In another embodiment of these methods of preparing, the method further comprises lyophilizing the ceftolozane in the absence of the tazobactam. In yet another embodiment, the method can further comprise lyophilizing the tazobactam in the absence of the ceftolozane.
Accordingly, in one aspect, provided herein is a ceutical composition comprising ceftolozane and tazobactam, wherein the composition comprises less than 0.5%, 0.4% 0.3%, 0.25%, 0.2%, 0.15%, 0.1%, or 0.05% by weight of the compound RRT 1.22. In another aspect, provided herein is a pharmaceutical composition comprising ceftolozane and ctam, wherein the composition comprises less than 0.1% by weight of the compound RRT 1.22. In one embodiment, the pharmaceutical composition ses less than 0.05% by weight of the compound RRT 1.22. In another embodiment, the pharmaceutical ition comprises less than 0.15% by weight of the compound RRT 1.22. In yet r ment, the pharmaceutical composition comprises no detectable amount of the compound RRT 1.22 as measured by HPLC.
In contrast, a greater amount of nd RRT 1.22 was found in compositions of ceftolozane and tazob actam, wherein the compositions were formed through co- lyophilization, i.e., the ceftolozane and tazobactam were combined and co-lyophilized together, as d to being individually lyophilized and blended together (see, e. g., Examples 7 and 10).
In one aspect, provided herein is an antibacterial pharmaceutical composition comprising ceftolozane sulfate and tazob actam in a ratio of 1,000 mg ceftolozane active per 500 mg of tazobactam active, the pharmaceutical composition obtained by a s comprising the steps of: lyophiliZing a first aqueous solution in the absence of tazobactam, the first aqueous solution comprising ceftolozane sulfate prior to lyophilization to obtain a first lyophilized ceftolozane composition; and ng the first lyophilized ceftolozane composition with tazob actam to obtain an cterial composition comprising less than 0.13% by HPLC of a compound of formula (III) (compound RRT 1.22) detectable at a retention time relative to ceftolozane of 1.22 by high performance liquid chromatography using a Develosil column ODS-UG-S; 5 micrometers; 250 X 4.6 mm, a mobile phase of sodium perchlorate buffer on (pH 2.5)/CH3CN 90:10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 0C.
In one embodiment, the antibacterial composition comprises less than 0.03% of the nd of formula (III) detected by HPLC. In another embodiment, the first aqueous solution further comprises L-arginine in an amount effective to adjust the pH of the first aqueous solution to 6-7 prior to lization to obtain a first lyophilized ozane composition.
In another embodiment, the antibacterial pharmaceutical composition is obtained by a process further comprising the steps of: lyophilizing a second solution comprising ctam in the absence of ceftolozane to form a second lyophilized tazobactam composition; and blending the first lized ceftolozane ition and the second lized tazobactam composition to obtain the antibacterial composition.
In another embodiment, the tazob actam in the second solution is ctam acid, and wherein the tazob actam acid in the second solution is lyophilized in the presence of sodium bicarbonate to form the second lyophilized tazobactam solution.
In another embodiment, the first aqueous solution comprises nine in an amount effective to provide a pH of about 5-7, e. g., 6-7. In another embodiment, the first aqueous solution comprises 125 mg to 500 mg of sodium chloride per 1,000 mg of ceftolozane active.
In another embodiment, the first aqueous solution further comprises citric acid. In another embodiment, the first aqueous solution consists of ceftolozane e, citric acid, sodium chloride, L-arginine, and water.
In another aspect, provided herein is a unit dosage form of a pharmaceutical composition formulated for parenteral administration for the treatment of complicated intra- abdominal infections or complicated urinary tract infections, the pharmaceutical composition comprising ceftolozane sulfate and tazob actam in a ratio of 1,000 mg ceftolozane active per 500 mg of tazobactam active, the pharmaceutical composition obtained by a s comprising the steps of: lyophilizing a first aqueous on in the absence of tazobactam, the first aqueous solution comprising ceftolozane sulfate, 125 mg to 500 mg of sodium de per 1,000 mg of ozane active, at a pH of 6-7 prior to lyophilization to obtain a first lyophilized ceftolozane composition; lyophilizing a second solution comprising tazobactam in the absence of ceftolozane to form a second lyophilized tazobactam composition; and blending the first lyophilized ozane composition and the second lyophilized tazobactam composition to obtain the antibacterial composition.
In another embodiment, the unit do sage form comprises a total of not more than 0.03% by HPLC of a compound of formula (III) detectable at a retention time relative to ceftolozane of 1.22 by high performance liquid chromatography using a Develosil column ODS-UG-5; 5 micrometers; 250 X 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90: 10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 H30; 1L H3131, 00" lam/""--‘ Kl S fi‘w , g ’1‘3 N___3\,,. \ l; H a], N, ‘1‘, \x, l 1;: _ ‘5: 0 ,‘N 1 H2" ‘3" 0‘ 1 \ ‘N N": 0"" OH 0'1"," "‘0 " (III) In another embodiment, the unit dosage form ses a total of 1,000 mg of ceftolozane active and a total of 500 mg of tazobactam active.
In another ment, the unit do sage form comprises a total of not more than 0.03% by HPLC of a compound of formula (III) detectable at a ion time relative to ozane of 1.22 by high performance liquid chromatography using a Develosil column ODS-UG-5; 5 eters; 250 X 4.6 mm, a mobile phase of sodium perchlorate buffer solution (pH 2.5)/CH3CN 90: 10 (v/v) at a 1.0 mL/min flow rate and oven temperature of 45 (III) In another aspect, provided herein is a compound of a (III): (III); or a ceutically acceptable salt thereof.
In still another aspect, provided herein is a pharmaceutical composition comprising a compound of formula (III): H36, "1 H30"? "OH (III); or a pharmaceutically able salt .
In one embodiment, the pharmaceutical composition further comprises ceftolozane sulfate. In another embodiment, the pharmaceutical composition further comprises tazobactam.
In another embodiment, the compound of formula (III) is ed by a process comprising the step of reacting ceftolozane and formylacetic acid to obtain the compound of formula (III). In another embodiment, the compound of formula (III) is obtained by a process sing the step of reacting ceftolozane and tazobactam acid to obtain the nd of formula (III).
In another embodiment, the compound of formula (III) is obtained by a process comprising the steps of: forming an aqueous solution comprising ceftolozane and tazobactam acid; and lyophilizing the aqueous solution to obtain a lyophilized composition comprising the compound of a (III). In another embodiment, the aqueous solution ses ceftolozane sulfate and tazobactam acid in a 2:1 weight ratio between the amount of ceftolozane active and the amount of tazob actam acid. In another embodiment, the aqueous solution comprises sodium chloride, ceftolozane sulfate, tazobactam acid and L-arginine.
In another embodiment, the aqueous solution has a pH of about 5.0 to 7.0, e. g., 6.0 to 7.0. In r embodiment, the pharmaceutical composition is formulated for parenteral administration. In another embodiment, the compound of formula (III) is obtained by a process r comprising the step of performing high performance liquid chromatography (HPLC) on the lyophilized composition to isolate the compound of formula (III).
In another embodiment, the pharmaceutical composition ses about 0.l3-0.97% of the compound of a (III). In another embodiment, the pharmaceutical composition comprises up to about 0.15% of the compound of formula (III). In another aspect, provided herein is a pharmaceutical composition sing a compound of formula (III), "Q, ll H,C--"‘T "‘0" (III), the compound of formula (III) obtained by a process comprising the steps of : forming an aqueous solution sing tazobactam acid and ceftolozane sulfate in an amount providing 1,000 mg of ceftolozane active per 500 mg of tazobactam acid in the aqueous solution; lyophilizing the aqueous solution of step (a) to obtain a lyophilized composition sing a compound of formula (III); and ating the lyophilized composition as a pharmaceutical composition for eral delivery.
In one embodiment, the pH of the aqueous solution is 5.0 to 7.0, e.g., 6.0 to 7.0. In another ment, the pharmaceutical composition is formulated for parenteral administration.
In another aspect, provided herein is a pharmaceutical composition ated for parenteral administration for the treatment of cated intra—abdominal infections or complicated urinary tract infections, the ceutical composition comprising a compound of formula (III) in a lyophilized composition obtained by lyophilizing an aqueous solution comprising tazobactam and an amount of ceftolozane sulfate containing 1,000 mg of ceftolozane active per 500 mg of tazobactam acid.
B. Co-Lyophilization In one aspect, ed herein is a method of preparing a composition comprising ozane and sodium chloride, comprising combining sodium chloride with ceftolozane, wherein 125-1000 mg sodium chloride per 1000 mg of ceftolozane is combined, followed by lization of the sodium de ceftolozane mixture. The process is referred to herein as "co-lyophilization". In another aspect, provided herein is a method of preparing a composition comprising sodium de, tazobactam, and ceftolozane, comprising combining sodium chloride, tazobactam, and ceftolozane, wherein 125-1000 mg sodium de per 1000 mg of ceftolozane is combined, followed by lyophilization of the mixture of sodium chloride, tazobactam, and ceftolozane.
Also provided herein is a method of ing a pharmaceutical composition comprising sodium chloride, ceftolozane, and tazobactam, comprising combining sodium chloride, tazobactam, and ceftolozane, followed by spray-drying the mixture of sodium chloride, ceftolozane, and tazobactam.
Figure 2 is a rt showing the steps for preparing a CXA-201 composition comprising ceftolozane (referred to as CXA-lOl) and tazobactam using a co-lyophilization process, as bed .
In another aspect, provided herein is a ceutical composition comprising stabilized ceftolozane sulfate obtained by a process comprising lyophilizing an aqueous solution comprising 125 mg to 500 mg sodium chloride with an amount of ceftolozane sulfate providing 1,000 mg of ceftolozane active, to obtain the lyophilized stabilized ceftolozane sulfate composition.
In one embodiment, the stabilized ozane is ed by lizing the sodium chloride and ceftolozane sulfate with L-arginine. In another embodiment, the stabilized ceftolozane is obtained by lyophilizing an aqueous solution having a pH of about 5.0 to 7.0, e.g., 6.0 to 7.0.
In another embodiment, the stabilized ceftolozane is obtained by lyophilizing the sodium chloride and ceftolozane sulfate with L-arginine and citric acid. In another embodiment, the pharmaceutical composition is formulated for parenteral administration. In another embodiment, the composition is a unit dosage form in a ner comprising 125 mg to 500 mg sodium chloride, 1,000 mg of ceftolozane in the form of ceftolozane sulfate, and L-arginine. In another embodiment, the pharmaceutical composition is formulated for parenteral administration. In another embodiment, the pH of the aqueous solution is 6.0 to 7.0.
In another aspect, provided herein is a container sing a pharmaceutical composition of stabilized ceftolozane sulfate, obtained by a process comprising the step of: lyophilizing an aqueous solution comprising 125 mg to 500 mg sodium chloride with an amount of ceftolozane sulfate providing 1,000 mg of ceftolozane active, to obtain the lyophilized ized ozane sulfate composition; filling the lyophilized stabilized ceftolozane composition into a container.
IV. Manufacturing For the Prevention of Cross-Contamination Recent FDA manufacturing guidance (published in April 2013) states that manufacturing facilities dedicated to manufacturing a sensitizing non-penicillin beta-lactam compound should be "completely and comprehensively ted" from areas in the facility in which any class of sensitizing beta-lactam is manufactured. See US. Department of Health and Human Services Food and Drug Administration, Center for Drug tion and Research, Non-Penicillin Beta-Lactam Drugs: A CGMP Framework for Preventing Cross- ination (April 2013) ("FDA Guidance"). The FDA also considers separation of production ties for llins to be good cturing practice. The FDA Guidance can be understood to require the use of a dedicated facility to cture antibiotic compounds comprising a non-penicillin beta-lactam nd (e.g., a cephalosporin) and a BLI compound with a beta-lactam ring (e. g., tazobactam). Accordingly, a facility that manufactures a product containing both cephalosporin and a beta-lactam containing BLI such as tazobactam for sale in the United States cannot be subsequently used to manufacture any other products containing beta-lactam ring,other than additional combinations of other cephalosporins with the same BLI compound (e. g., other non-penicillin beta-lactam compounds ing other cephalosporin antibitoics cannot be subsequently manufactured in the facility).
Beta-lactam antibiotics, ing penicillin and the non-penicillin classes, share a basic chemical structure that includes a three-carbon, one-nitrogen cyclic amine structure known as the beta-lactam ring. The side chain associated with the beta-lactam ring is a variable group attached to the core ure by a peptide bond; the side chain variability contributes to antibacterial activity. As of the date of this publication, FDA has approved over 34 beta-lactam compounds as active ingredients in drugs for human use. (see, e.g., FDA’s Approved Drug Products with eutic lence Evaluations, generally known as the Orange Book) Beta-lactam otics include the following five classes: penicillins (e. g., ampicillin, oxacillin); cephalosporins (e. g., cephaleXin, cefaclor); penems (e. g., imipenem, meropenem); carbacephems (e.g., loracarbef); and monobactams (e. g., aZtreonam). (Yao, JDC, and RC Moellering, Jr., Antibacterial agents, in Manual of al Microbiology, 9th edition, edited by PR Murray et al., Washington DC, ASM Press, 2007.) Under the FDA Guidance, a manufacturing facility handling a product for sale in the United States containing both a cephalosporin (e.g, ceftolozane) and a penicillin nucleus (e. g., tazobactam) cannot be subsequently used in the manufacture of any other class of beta- lactam products, including all other penicillins, cephalosporins, penems, carbacephems and ctams or in the manufacture of other finished pharmaceuticals or active ceutical ingredients. The FDA Guidance states that (non-penicillin) cephalosporin beta-lactam compounds (e. g., such as ceftolozane) for sale in the United States must be "completely and comprehensively separated from" manufacturing areas that handle any other class of beta-lactam compound (e. g., compounds in the penicillin class).
A t containing ceftolozane and tazobactam includes both a nicillin beta- lactam cephalosporin (ceftolozane) and a beta-lactamase inhibitor with a beta-lactam moiety (tazobactam). Under the FDA Guidance, these two compounds must be "completely and comprehensively separated." Accordingly, there is a need for methods of manufacturing antibiotic itions comprising ceftolozane and tazobactam for sale in the United States in ance with the FDA Guidance, as well as antibiotic itions manufactured in accordance with the FDA Guidance without affecting the purity, stability, and safety of the resulting composition.
Provided herein are methods of manufacturing or preparing pharmaceutical compositions containing two or more beta-lactam compounds in accordance with FDA ce, as well as pharmaceutical compositions manufactured in compliance with FDA Guidance. Specifically, certain cturing methods are ed herein that conform to standards recommended by FDA Guidance for the avoidance of cross-contamination of non- penicillin beta-lactam drugs.
In one aspect, provided herein is an antibacterial pharmaceutical ition formulated for parenteral administration for the treatment of infections, the pharmaceutical composition comprising a therapeutically effective amount of ceftolozane sulfate and tazobactam in a ratio of 1,000 mg ceftolozane active per 500 mg of tazobactam active, the pharmaceutical composition obtained by a process comprising the steps of: a. lyophilizing a first aqueous solution in the absence of tazobactam, the first aqueous solution comprising ceftolozane sulfate, to obtain a first lyophilized ceftolozane composition; b. blending the lyophilized ceftolozane composition with a tazobactam composition comprising tazobactam prepared and provided in the absence of ceftolozane; wherein the process is completed in the absence of other non-cephalosporin beta- lactam compounds.
In another aspect, ed herein is a unit dosage form of a pharmaceutical composition ated for parenteral administration for the treatment of complicated intra- abdominal infections or complicated urinary tract infections, the pharmaceutical composition comprising ceftolozane sulfate and tazob actam in a ratio of 1,000 mg ceftolozane active per 500 mg of tazobactam active, the pharmaceutical ition obtained by a process comprising the steps of a. lyophilizing a first aqueous on in the absence of tazobactam, the first aqueous solution comprising ceftolozane sulfate, 125 mg to 500 mg of sodium chloride per 1,000 mg of ceftolozane active, to obtain a first lized ozane ition, b. lyophilizing a second solution comprising tazobactam in the absence of ozane to form a second lyophilized tazob actam composition; and c. blending the first lyophilized ozane composition and the second lyophilized tazobactam composition to obtain the antibacterial composition; wherein the process is completed in the absence of other non-cephalosporin beta- lactam compounds.
V. Methods of Treatment Pharmaceutical compositions comprising ceftolozane/tazobactam are being developed as an enous (IV) formulation for the treatment of cated urinary tract infections (cUTIs) and complicated intra-abdominal infections (cIAIs). ozane/tazobactam is an antibacterial composition including ceftolozane, a cephalosporin with potent eudomonal cephalo sporinactivity, in combination with tazobactam, a beta (B)-lactamase inhibitor (BLI). Like other members of the cephalosporin class, ceftolozane is believed to exert its bactericidal activity by inhibiting essential penicillin-binding proteins (PBPs), resulting in inhibition of cell wall synthesis and subsequent cell death. Ceftolozane has activity against Pseudomonas aeruginosa including strains that are resistant to carb s, cephalosporins, fluoroquinolones, and aminoglycosides, and other common Gram-negative pathogens, ing most extended- spectrum B-lactamase (ESBL)-producing Enterobacteriaceae. Tazobactam ts chromosomal- and plasmid-mediated bacterial class A and C B lactamases. Tazobactam is believed to protect ceftolozane from hydrolysis by covalently binding these enzymes, and broadens coverage to include most ESBL-producing Escherichia coli, Klebsiella pneumoniae, and other bacteriaceae, ing some Enterobacteriaceae overexpressing AmpC. ctam inhibits or decreases the activity of actamases (e. g., bacterial beta-lactamases), and can be combined with actam nds (e. g., antibiotics), thereby broadening the um of the beta-lactam compound and increasing the beta-lactam compound's efficacy against organisms that produce beta-lactamase. A compound or a composition possesses efficacy against an organism if it kills or weakens the organism, or inhibits or ts reproduction the organism.
The CXA-ZOl product (ceftolozane/tazobactam for injection) is undergoing regulatory review for the treatment of complicated intra-abdominal infections (cIAI) caused by tible isolates of the Gram-negative and Gram-positive microorganisms such as: Citerohacterfreimdii, Escherichia coli, Enterobacter cloacae, Klehsiella pneumonia, Klebsiella oxytoca, Proteus mirabilis, and Pseudomonas aeruginosa. In patients who are at risk of mixed aerobic-anaerobic infection, concurrent therapy with an anti-anaerobic agent can also be used.
The CXA-ZOl product (ceftolozane/tazobactam for injection) is oing regulatory review for the treatment of complicated urinary tract infections (cUTI), including pyelonephritis caused by susceptible isolates of the following Gram-negative microorganisms: Enterob acter spp, Escherichia coli, Klebsiella pneumonia, Proteus mirabilis andPseudomonas aeruginosa. In one embodiment, a pharmaceutical ition comprising a CXA-ZOl product (e.g, the unit dosage container of Table 29 below) is reconstituted in a ceutically able carrier (e. g., a total volume of about 90-150 mL, preferably about 110 mL, of 0.9% aqueous sodium de for injection or in initial volume of 10-20 mL of water for injection or 0.9% aqueous sodium chloride for injection, followed by dilution of this solution into a 100 mL volume of 0.9% aqueous sodium chloride for injection). The resulting pharmaceutical composition can be infused into a patient in need thereof for treatment of a complicated intra-abdominal infection (e. g, using 1 hour infusion times) three times per day (e.g., once every 8 hours) for a recommended duration of treatment (e. g. 4-10 days).
The preferred dosage of (ceftolozane/tazobactam for ion) for cUTI and cIAI is 1.5 g administered every 8 hours by intravenous (IV) infusion over 1 hour in patients 218 years of age. The duration of therapy should be guided by the severity and site of infection and the patient’s clinical and bacteriological progress. In one embodiment, a pharmaceutical composition comprising a CXA-ZOl product (e.g, the unit dosage container of Table 29 below) is reconstituted in a pharmaceutically acceptable carrier (e.g., a total volume of about 90-150 mL, preferably about 110 mL, of 0.9% aqueous sodium chloride for injection or in initial volume of 10-20 mL of water for injection or 0.9% aqueous sodium chloride for injection, followed by dilution of this on into a 100 mL volume of 0.9% aqueous sodium chloride for ion). The resulting pharmaceutical composition can be infused into a patient in need thereof for treatment of a Complicated Urinary Tract Infections (cUTI), Including ephritis (e. g, using 1 hour infusion times) three times per day (e. g., once every 8 hours) for a recommended duration of treatment (e.g. 7 days).
Ceftolozane/tazobactam displays potent antibacterial ty t common Gram- negative organisms, including Enterobacteriaceae and Pseudomonas aeruginosa; select Gram- ve organisms, including streptococci; the majority of pathogenic enteric i and select ositive anaerobic species, thus making ceftolozane/tazobactam a potentially practical choice for pathogens involved in gastro tinal, urinary and community acquired as well as nosocomial respiratory infections. In general, the Gram-positive and Gram- negative spectrum of ceftolozane is similar to ceftazidime, but its antipseudomonal activity is the mo st potent among all currently available B-lactams, including the cephalosporins and carbapenems. Most importantly, ceftolozane has been shown to be active against strains of P. aerugino sa that are resistant to carb apenems, cephalosporins, fluoroquinolones, and aminoglycosides, including the majority of multi-drug resistant isolates. Indeed, the minimum inhibitory concentration (MIC) required to inhibit the growth of 90% of sms (MIC90) for P. aeruginosa (MIC90 S 2 ug/mL) is the lowest among all systemically administered antipseudomonal otics.
In vitro studies have trated that ozane/tazobactam has a broad spectrum of activity against Gram-negative bacteria. The in vitro activity of ceftolozane and ceftolozane/tazobactam was evaluated against a broad range of Gram-positive and Gram- negative bacteria. It was observed that tazobactam potentiated the activity of ceftolozane against Acinetobacter spp. and common species of Enterobacteriaceae, including Citrobacter spp., Enterobacter e, E. coli, K. pneumoniae, Proteus mirabilis, and Serratia marcescens. These surveillance data demonstrate that 88% to 100% of these Enterobacteriaceae species are inhibited at < 8 ug/mL.
In one aspect, provided herein is a method for the treatment of ial infections in a mammal, comprising administering to said mammal a eutically effective amount of a pharmaceutical composition prepared according to the methods described herein. In another aspect, provided herein is a method for the treatment of bacterial infections in a mammal, comprising administering to said mammal a therapeutically effective amount of tazobactam and ceftolozane. In n ments of the above methods, the bacterial infection is caused by an extended-spectrum beta-lactamase-producing sm. In certain embodiments, the bacterial ion is caused by an antibiotic-resistant sm. In yet another aspect, the invention is a method for the treatment of bacterial infections in a mammal, comprising administering to said mammal a therapeutically effective amount of a pharmaceutical ition comprising both tazobactam and ceftolozane. In certain embodiments of the above methods, the ial infection is caused by an extended- spectrum beta-lactamase-producing organism. In certain embodiments, the bacterial infection is caused by an antibiotic-resistant organism.
In another , provided herein is a method for the ent of bacterial infections in a , comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising tazobactam, ceftolozane, and less than 0.1% by weight of the compound RRT 1.22. In r embodiment of the treatment , the pharmaceutical composition comprises tazobactam, ceftolozane, and less than 0.05% by weight of the compound RRT 1.22.
In n embodiments of the treatment methods, the pharmaceutical composition further comprises 125 to 1000 mg sodium chloride per 1000 mg of ceftolozane, e.g., 125 to 500 mg sodium chloride per 1000 mg of ceftolozane, 0 mg sodium chloride per 1000 mg of ceftolozane, 300-500 mg sodium chloride per 1000 mg of ceftolozane, 400-500 mg sodium chloride per 1000 mg of ceftolozane, 450-500 mg sodium chloride per 1000 mg of ceftolozane, 460-500 mg sodium chloride per 1000 mg of ceftolozane, or about 476 mg sodium chloride per 1000 mg of ozane. In one specific embodiment of the ent s, the ceutical composition further comprises about 487 mg sodium chloride per 1000 mg of ceftolozane.
In other embodiments of the treatment methods, the pharmaceutical composition comprises 250-750 mg tazobactam per 1000 mg of ceftolozane, e. g., 250-700 mg tazobactam per 1000 mg of ceftolozane, 300-700 mg tazobactam per 1000 mg of ceftolozane, 300-650 mg ctam per 1000 mg of ceftolozane, 350-650 mg tazobactam per 1000 mg of ceftolozane, 350-600 mg tazobactam per 1000 mg of ceftolozane, 400-600 mg tazobactam per 1000 mg of ceftolozane, 400-550 mg tazobactam per 1000 mg of ceftolozane, 450-550 mg tazobactam per 1000 mg of ceftolozane, or about 500 mg tazobactam per 1000 mg of ceftolozane.
Non-limiting examples of the bacterial ions that can be treated by the methods of the invention include infections caused by: aerobic and facultative gram-positive microorganisms (e. g., Staphylococcus aureus, Enterococcusfaecalis, Staphylococcus epidermidis, Streptococcus agalactiae, Streptococcus pneumonia, Streptococcus es, Viridans group streptococci), aerobic and facultative gram-negative microorganisms (e. g., Acinetohacter haumanii, Escherichia coli, Haemophilus influenza, Klehsiella pneumonia, Pseudomonas aeruginosa, Citrobacter koseri, Moraxella catarrhalis, Morganella morganii, Neisseria hoeae, Proteus mirahilis, Proteus vulgaris, Serratia marcescens, Providencia ii, Providencia rettgeri, Salmonella ca), ositive anaerobes (Clostridium perfringens), and gram-negative anaerobes (e. g., Bacteroidesfragilis group (e.g., B. fragilis, B. ovatus, B. thetaiotaomicron, and B. vulgates), Bacteroides distasonis, Prevotella melaninogenica).
In certain embodiments of the methods described herein, the bacterial infections ing from beta-lactamase-producing organisms are treated or controlled. miting examples of beta-lactamase-producing organisms include: (1) ESBL (extended-spectrum beta-lactamase)-producing sms selected from the group consisting of bacteriaceae spp.: Escherichia coli, Klebsiella spp. ding K. pneumoniae and K. oxytoca), Proteus mirahilis, Proteus vulgaris, Enterohacter spp., Serratia spp., Citrobacter spp., Pseudomonas spp., Acinetobacter spp.) and Bacteroides spp.; (2) CSBL (conventional-spectrum beta-lactamase)-producing sms, known to those of skill in the art; and (3) Inducible-AmpC—type beta-lactamases, such as Citrobacter spp., Serratia spp., Morganella morganii, Proteus vulgaris, and Enterohacter cloacae.
In certain embodiments of the methods described herein, the bacterial infection is ated with one or more of the following conditions: Appendicitis (complicated by rupture or abscess) and peritonitis caused by piperacillin—resistant actamase producing strains of Escherichia coli or the ing s of the Bacteroides fragilis group: B. is, B. ovatus, B. otaomicron, or B. vulgates; Uncomplicated and complicated skin and skin structure infections, including cellulitis, cutaneous abscesses, and ischemic/diabetic foot infections caused by piperacillin- resistant, beta-lactamase producing strains of Staphylococcus aureus; Postpartum endometritis or pelVic inflammatory e caused by piperacillin- resistant, beta-lactamase producing strains of Escherichia coli; Community-acquired pneumonia (moderate severity only) caused by piperacillin- resistant, beta-lactamase producing strains of Haemophilus influenza; Nosocomial pneumonia (moderate to severe) caused by piperacillin-resistant, beta- lactamase producing strains of Staphylococcus aureus and by Acinetobacter baumanii, Haemophilus influenzae, Klehsiella pneumoniae, and Pseudomonas aeruginosa. Nosocomial pneumonia is also known as al acquired/ventilator-associated bacterial pneumonia (HABP/VABP); Complicated intra-abdominal infections (cIAI); Complicated urinary tract infections (cUTIs); Acute Pyelonephritis; and Systemic Inflammatory Response Syndrome (SIRS).
Also ed herein is the use of tazobactam, and hydrates and solvates thereof, in combination with ceftolozane, for the preparation of a medicament for the treatment of bacterial infections. The bacterial ions can result from either gram-negative or gram- positive organisms.
The itions provided herein can be used in the treatment of infections caused by Pseudomonas aeruginosa, Serratia marcescens, Escherichia coli, Klebsiella niae, Haemophilus influenzae, or Streptococcus pneumonia.
In one embodiment of the treatment methods, the bacterial ions are Gram- negative bacterial infections. In one embodiment, the gram-negative infections are complicated y Tract Infections (cUTI) and cated intra-abdominal infections (cIAI). In another embodiment, the gram-negative bacterial infections are caused by Pseudomonas aeruginosa, E. coli, and/or Klebsiella pneumonia.
In a r embodiment, provided herein is a method for the treatment of gram- negative bacterial infections in a mammal, comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising ceftolozane, about 500 mg ctam free acid per 1000 mg of ceftolozane, about 476 mg sodium chloride per 1000 mg of ozane, about 587 mg L-arginine per 1000 mg of ozane, and about 21 mg anhydrous citric acid per 1000 mg of ceftolozane. In one embodiment, the gram-negative bacterial infections are selected from the group consisting of complicated Urinary Tract Infections (cUTI) and complicated intra-abdominal ions (cIAI). In another embodiment, the gram negative bacterial infection is nosocomial pneumonia.
In another specific embodiment, provided herein is a method for the ent of gram-negative bacterial infections in a mammal, comprising administering to said mammal a therapeutically effective amount of a ceutical composition comprising ceftolozane, about 500 mg tazobactam free acid equivalents per 1000 mg of ceftolozane, about 487 mg sodium chloride per 1000 mg of ceftolozane, about 600 mg L-arginine per 1000 mg of ceftolozane, and about 21 mg anhydrous citric acid per 1000 mg of ceftolozane. In one embodiment, the gram-negative bacterial infections are selected from the group consisting of complicated Urinary Tract Infections (cUTI) and complicated intra-abdominal infections (cIAI). In another embodiment, the gram negative bacterial infection is nosocomial pneumonia.
In one embodiment, provided herein is a method for the ent of an infection in a mammal, wherein the infection is caused by Pseudomonas aeruginosa, Serratia marcescens, Escherichia coli, Klebsiella pneumoniae, Haemophilus influenzae, or Streptococcus pneumoniae comprising administering to said mammal a eutically effective amount of a ceutical composition comprising ceftolozane, corresponding to approximately lOOOmg of the free base form of ceftolozane; tazobactam, corresponding to approximately 500mg of the tazobactam acid form; and 400-500mg sodium chloride. In one embodiment, the pharmaceutical composition r comprises 500-650 mg L-arginine and 15-30 mg anhydrous citric acid.
In another embodiment, provided herein is a method for the treatment of urinary tract infection, intra-abdominal infection, or nosocomial nia in a mammal, comprising administering to said mammal a therapeutically effective amount of a ceutical composition comprising ceftolozane, corresponding to approximately lOOOmg of the free base form of ceftolozane; tazobactam, ponding to approximately 500mg of the tazobactam acid form; and 400-500mg sodium chloride. In an embodiment, the pharmaceutical composition comprises 487 mg sodium chloride. In one embodiment, the pharmaceutical composition further comprises 500-650 mg L-arginine and 15-30 mg anhydrous citric acid.
In one embodiment, provided herein is a method for the ent of an infection in a , wherein the infection is caused by monas aeruginosa, Serratia marcescens, Escherichia coli, Klebsiella pneumoniae, Haemophilus influenzae, or Streptococcus pneumoniae comprising administering to said mammal a eutically effective amount of a pharmaceutical composition comprising approximately ll47mg ceftolozane sulfate; imately 537mg tazobactam sodium; and 400-500 mg sodium chloride. In one embodiment, the pharmaceutical composition further comprises 500-650 mg L-arginine and -30 mg ous citric acid.
In another embodiment, provided herein is a method for the ent of urinary tract infection, intra-abdominal ion, or nosocomial nia in a mammal, comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising approximately 1147 mg ceftolozane sulfate; approximately 537 mg tazobactam sodium; and 400-500 mg sodium chloride. In an embodiment, the 3O pharmaceutical composition comprises 487mg sodium chloride. In one embodiment, the pharmaceutical composition further comprises 500-650 mg L-arginine and 15-30 mg anhydrous citric acid.
In one embodiment, provided herein is a method for the treatment of an infection in a mammal, wherein the infection is caused by Pseudomonas aeruginosa, Serratia cens, Escherichia coli, Klebsiella pneumoniae, Haemophilus influenzae, or Streptococcus pneumoniae comprising administering to said mammal a eutically effective amount of a pharmaceutical composition comprising approximately 1147 mg ceftolozane sulfate; approximately 537 mg tazobactam sodium; imately 487 mg sodium chloride; and approximately 600 mg nine. In one embodiment, the pharmaceutical composition further comprises 15-30 mg anhydrous citric acid.
As used herein, ing", "treat" or "treatment" describes the management and care of a t for the purpose of ing a disease, condition, or disorder and includes the administration of a pharmaceutical composition of the present invention to alleviate the symptoms or complications of a disease, condition or disorder, or to eliminate the e, condition or disorder. The term "treat" can also include treatment of a cell in vitro or an animal model.
By a "therapeutically effective amount" of a compound of the invention is meant a sufficient amount of the compound to treat the disorder (e. g., ial infection). The specific therapeutically effective amount that is required for the treatment of any particular patient or organism (e.g., a mammal) will depend upon a variety of factors including the er being treated and the severity of the disorder; the activity of the specific compound or composition employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in ation or coincidental with the specific compound employed; and like factors well known in the l arts (see, for example, Goodman and Gilman’s, "The Pharmacological Basis of Therapeutics", Tenth Edition, A. Gilman, J.Hardman and L. Limbird, eds., McGraw- Hill Press, 3, 2001, which is incorporated herein by reference in its entirety). The therapeutically effective amount for a given ion can be readily determined by routine experimentation and is within the skill and judgment of the ordinary clinician.
As used herein, "189 mg sodium from sodium chloride per 1000 mg of ceftolozane" refers to a ratio of sodium from the sodium chloride to ceftolozane active. For example, "189 mg sodium from sodium de per 1000 mg of ceftolozane" includes, for example, 94.5 mg sodium from sodium de per 500 mg of ceftolozane, as well as, for example, 47.25 mg sodium from sodium chloride per 250 mg ceftolozane. In addition, "1,000 mg of ceftolozane as ceftolozane sulfate" refers to an amount of ozane sulfate effective to provide 1,000 mg of ceftolozane. "189 mg sodium from sodium chloride" refers to the amount of sodium chloride (e.g., 480 mg) effective to provide 189 mg of sodium. The amount of sodium from sodium chloride per gram of ceftolozane activity in a pharmaceutical composition containing ceftolozane sulfate, chloride and sodium chloride can be calculated using the relevant molecular weights of ceftolozane, ozane sulfate, sodium chloride and sodium. For example, a composition comprising about 1,147 mg ceftolozane sulfate and 189 mg sodium from sodium chloride contains 480 mg sodium chloride per 1,000 mg ceftolozane active.
Unless otherwise indicated, as used herein, the term "Related Substances" with respect to HPLC detection refers to all the ceftolozane related s impurities and degradation products other than ceftolozane ted and detected by HPLC ing to Example 1. Unless otherwise indicated, as used herein, the term "% Related Substances" refers to the % of the total HPLC peak area obtained by Example 1 attributed to all the ozane related process impurities and degradation products other than ceftolozane.
EXAMPLES Example I .' HPLC analysis of Compositions Comprising Ceftolozane .
The purity of ceftolozane in the pharmaceutical compositions was measured using the analytical HPLC method described below.
The HPLC methodologies bed herein were used to acquire the data provided in Examples 5 and 8.
Analflical HPLC Method A. Operative Conditions 3O Column Develosil ODS-UG-S ; 5 um, 250 x 4.6 mm (Nomura Chemical, Japan) Mobile phase Sodium orate buffer solution (PH 2.5)/CH3CN 90 : 10 (v/v) Flow rate 1.0 mL/min Wavelength 254 nm ion volume 10 uL Oven Temperature 45 0C Run Time 85 minutes Gradient Profile: B. Mobile Phase Preparation.
Sodium perchlorate buffer solution was made by ving 14.05 g of sodium perchlorate monohydrate in 1000.0 mL of water followed by adjusting pH to 2.5 with diluted perchloric acid (1 in 20).
Mobile phase was then made by mixing sodium perchlorate buffer on (pH 2.5) and acetonitrile in the ratio 90: 10 (v/v).
Sodium acetate buffer solution pH 5.5 (diluent) was made by dissolving 1.36 g of sodium acetate trihydrate in 1000.0 mL of water followed by adjusting to pH 5 .5 with diluted acetic acid (1 in 10).
C. Sample ation.
Sample solution: dissolve 20.0 mg, exactly weighed, of the Sample, in 20.0 mL of water (Prepare just before injection into HPLC system).
System suitability solution (1 %): take 1.0 mL of the sample solution (the first sample if more are present) and transfer into a 100.0 mL volumetric flask, dilute with water to volume and mix.
D. HPLC Analysis ure 1. Inject blank ) 2. Inject system suitability solution and check for tailing factor and tical plate number for the CXA-101 peak: 0 The g factor must not be greater than 1.5 0 Theoretical plates number must not be less than 10000 3. Inject sample solution 4. Inject system suitability solution and check for tailing factor and theoretical plate number for the CXA- 101 peak. 0 The tailing factor must not be greater than 1.5 0 Theoretical plates number must not be less than 10000 . Identify the peaks of related substances in the sample chromatogram based on the reference chromatogram reported in Figure 3 or, alternatively, on the basis of the following RRT values listed in Table 1.
Table 1. Identities and RRTs of the Products Related to Ceftolozane Compound RRT Proposed Structure Source Peak 1 Degradation 3—side chain product and process product Process product Process product Degradation 7—Epimer product and type process product Degradation A3 Isomer product and type process product Process product and Degradation Anti—Isomer . t type E. Calculations I. Report for each related substance its amount as expressed by area percent.
A X 100 C1 2 At +2 A1 wherein: Ci 2 Amount of related nce i in the sample, area % A1 2 Peak area of related substance i in the sample chromatogram At 2 Area of CXA-lOl peak in the sample chromatogram A, + 2 A1: Total peaks area in the sample chromatogram Stabilizing Sodium chloride Fructose Xylitol Sorbitol Dextran 40 agent Consider as any unspecified compound, each peak in the chromatogram except CXA- lOl, peaks from 1 to 11 and every peak present in the blank chromatogram and report the largest. 11. Report the total composition content as expressed by the following formula: A, X 100 CT = A, +2 A, n: CT = total composition content in the sample, area% A, 2 area of l peak in the sample chromatogram Z A, = total peak areas of composition in the sample chromatogram Figure 3 is a reference HPLC chromatogram g the peaks of ceftolozane (CXA- 101) and related compo sition peaks. e 2.‘ Screening ofStabilizing Agents Nine stabilizing agents were screened, including sodium chloride, fructose, xylitol, sorbitol, dextran 40, lactose, glucose, maltose, and D-mannitol. The purity of the ceftolozane in a composition comprising 100 mg ceftolozane and 100 mg of one of the stabilizing agents after 3 days at 70 0C was compared to a composition sing 100 mg ceftolozane but no stabilizing agent.
As shown in Table 2, the ceftolozane compositions comprising sodium de, dextran 40, e, or maltose were demonstrated to be more stable than the other ceftolozane compositions comprising the other stabilizing agents, or no stabilizing agent.
Sodium chloride and maltose were selected for further investigation.
Storage Appearance Color and clarity Reconstitution time(s) No stabilizing Stabilizing agent agent (Control) Storage Appearance Color and Reconstitution Table 2. Screenin of Stabilizin A ents The stabilizing effect of other non-reducing sugars such as sucrose and trehalose, as well as polyvinylpyrrolidone (PVP), was also ted in a ceftolozane formulation.
Five samples were prepared, the components of which are shown in Table 2a below.
Each sample ned 1000 mg of ceftolozane active, 40 mg citric acid monohydrate (equivalent of 36 mg citric acid anhydrous), and the same amount of L-arginine. Stabilizing reagents in four samples are 480 mg sodium chloride, 300 mg of trehalose, 300 mg of sucrose, and 300 mg of PVP, respectively. One sample was a control that contained no stabilizing reagent. The s were in lyophilized form and stored at 60 0C for 7 days.
The purities of the samples were monitored by HPLC on day 0, day 1, day 3 and day 7.
Table 2a. Com arison between stabilizin rea ents 1000 1000 1000 1000 1000 98.42 98.09 98.14 97.89 97.94 d 97.85 96.73 96.97 96.05 96.15 60 OC/3d 97.21 95.36 95.81 94.57 94.53 As shown in Table 2a, the sample containing sodium chloride exhibited the best stability. The purity of ceftolozane in the sample containing sodium chloride had the slightest purity drop over 7 days. This experiment further supports the discovery that sodium chloride provides surprisingly better stabilizing effect than the other reagents.
Example 3: ity Study of Ceftolozane Compositions Comprising Sodium Chloride, or Maltose, or No Stabilizing Agent Three ceftolozane compositions were prepared, the components of which are shown in Table 3. These compositions were put in a stressed ity study at 70 0C for 3 days and 6 days. The purity of the ozane in the compositions was analyzed using the HPLC method described in Example 1.
Table 3. Ceftolozane Compositions 200 mg Citric acid CEF/maltose 9.5 g active Ceftolozane .7 g L-Arginine 200 mg Citric acid g Maltose H20 .7 g L-Arginine CEF/sodium chloride 4.6 g Sodium Chloride The results are shown in Table 4 where only the most significant composition peaks (P1, P7, and P12) are shown. It was found that the composition sing maltose (CEF/maltose) contained a icantly large amount of the composition P12 peak, which was identified as having the following formula: ,COOH N’ O | H §( N S NH A\N o )7? HEN / 8/ N51\ 0 $3 NH2 0— 0 H30 In addition, the presence of maltose produced a particularly aggregated powder after lization, which has a potentially negative impact to manufacturing ceftolozane compositions.
In contrast, the ceftolozane ition sing sodium chloride (CEF/sodium chloride) was much more stable than the ceftolozane composition comprising maltose or the ceftolozane composition comprising no stabilizing agent. Therefore, sodium chloride was, unexpectedly, a better stabilizing agent for ceftolozane compositions.
Table 4. it Stud of Ceftolozane Com ositions Com risin Sodium Chloride or Maltose, or No Stabilizing Agent CEF/no izer Time (days) CEF/maltose P12 Total Time (days) 0.65 0.15 1.91 1.02 3.44 10.08 3.45 1.12 4.01 11.65 CEF/sodium P1 P7 P12 Total chloride Time (days) Example 4a: Manufacturing Procedure ofMono Productfor Injection 4a.1. Preparation of the Compound Solution of CXA-101 Lyophilized Product 1) Weigh 30 kg of water for injection into the compounding vessel; 2) Add 100 g of citric acid, anhydrous and 150 g of sodium bicarbonate into the compounding vessel and dissolve them with mixing; 3) Weigh 5,000 g potency of CXA-101 drug nce and suspend it with mixing.
(Note any generation of carbon dioxide.) 4) Slowly add 1,100 g of sodium bicarbonate and dissolve CXA-101 with .
(Again, note any generation of carbon dioxide..) ) Add 1,146 g of sodium chloride and 10,000 g of maltose, dissolve with mixing. 6) Purge dissolved carbon dioxide in the solution with nitrogen until the pH of the solution does not change. 7) Adjust the pH of the solution to 6.0 i 0.1 with 5%-sodium bicarbonate solution. 8) Adjust the total weight to 56,850 g (D20 2 1.137) with water for injection. 9) Confirm the pH of the compounded solution within the range 6.0 i 0.1. 4a.2. Prefiltration and Sterile-Filtration ) te the compounded solution with a sterile tilter-set which consists of a 0.2 um nylidene fluoride membrane filter (Durapore®, Millipore) and a 0.1 urn polyVinylidene fluoride membrane filter (Durapore®, Millipore) connected in tandem.
Confirm the integrity of each filter before and after the filtration. Take approximately 100 mL of the filtrate in order to check bioburden. 11) Filter the prefiltered compounded on through a sterile filter-set which ts of a 0.2 um polyVinylidene fluoride membrane filter and a 0.1 urn polyVinylidene fluoride membrane filter connected in tandem, and uce the final filtrate into an aseptic room. Confirm the integrity of each filter before and after the filtration. 4a.3. Processing of container, Stopper and Flip-off Cap 12) Wash a sufficient ty of 28 mL containers with water for injection and sterilize the washed containers by a dry-heat sterilizer. Then transfer the sterilized containers into a Grade A area located in an c room. 13) Wash a sufficient quantity of stoppers with, water for injection. Sterilize and dry the washed stoppers by steam sterilizer. Then transfer the ized stoppers into a Grade A area located in an aseptic room. 14) Sterilize a sufficient quantity of flip-off caps by steam sterilizer. Then transfer the sterilized flip-off caps into a Grade A or B area located in an aseptic room. 4a.4. Filling and Partially Stoppering ) Adjust the fill weight of the filtered compounded on to 11.37 g (corresponds 3O to mL of the compounded solution), then start filling operation. Check the filled weight in sufficient ncy and confirm it is in target range (11.37 g i 1%, 11.26 to 11.43 g). When deviation from the control range (11.37 g i 2%, 11.14 to 11.59 g) is ed, re-adjust the filling weight. 16) Immediately after a container is filled, partially stopper the container with a sterilized stopper. Load the filled and partially stoppered containers onto the shelves of a lyophilizer aseptically. 4a.5. Lyophilization to Crimping, Visual Inspection, Labeling and Packaging 17) After all filled and partially red containers are loaded into a lyophilizer, start the lyophilization m shown in Figure 4. Freeze the loaded containers at -40 0C and keep until all containers freeze. Forward the program to y drying step (shelf ature; -20°C, chamber pressure; 100 to 150 . y drying time should be determined by monitoring the product temperature. Forward the program to secondary drying step (shelf temperature; 30°C, chamber pressure; not more than 10 mTorr) after completion of the primary drying step. After all containers are dried completely, return the chamber pressure to atmospheric pressure with sterilized en. Then stopper containers completely.
Example 4.‘ Manufacturing Procedure ofBulk (Tray) Lyophilized Ceftolozane There are four main steps in the manufacture of a CXA-101 pharmaceutical composition: dissolution, sterile filtration, bulk lyophilization, and packaging into Sterbags®.
These four main steps are ed of a total of 20 minor steps. The flowchart of the manufacturing process is described below.
I. Dissolution 1. A prescribed amount of WFI (e.g., 81 kg WFI) is charged into a dissolution reactor. 2. A prescribed amount of citric acid (e.g., 20.7 mg anhydrous citric acid per ceftolozane active ) is added. 3. The solution is cooled to 5°C to 10°C. 4. A prescribed amount of CXA-101 drug substance (e.g., referenced to 1000 mg ceftolozane active) is added to the solution. 3O 5. A prescribed amount of L-arginine (e. g., 587 mg L-arginine per 1000 mg ozane active) is slowly added to the solution. 6. A check for complete dissolution is performed. Solution pH is verified to be in the target range of 6.5 to 7.0. 7. A prescribed amount of sodium chloride (e. g., 476 mg sodium chloride per 1000 mg ceftolozane active) is added to the solution. 8. A check for complete dissolution is performed. Solution pH is verified to be in the target range of 6.0 to 7.0. If the pH is out of this range, adjust with either L-Arginine or citric acid. 9. WFI is added to bring the net weight to 13.1 g and the solution is mixed well.
. Samples are withdrawn for testing of final pH. 11. Sterile filtration 11. The solution is passed through a filter (pore size 0.45 um) followed by two more filters (pore size 0.22 um) onto a shelf on the Criofarma lyophilizer. 12. The line is washed with WFI. 13. The washing solution from Step 12 is passed through e filtration.
III. Bulk lyophilization 14. The washing solution is loaded onto a separate shelf on the lyophilizer (and later discarded).
. The solution is lyophilized until dry. 16. The product shelf is cooled to 20°C 1 5 0C.
IV. Packaging into Sterbags® 17. The lyophilized pharmaceutical ition is milled. 18. The milled powder is sieved. 19. The sieved powder is blended for 30 minutes.
. The powder is then discharged into Sterbags® Example 5 .' Stabilizing Eflect ofSodium Chloride in CXA-I 01 Compositions A. ement in the Purity of the Ceftolozane in CXA-101 Pharmaceutical Compositions with Varying Amounts of Sodium Chloride A stability study was carried out at 30 0C and 60 0C and analyzed by HPLC. The sodium de content in the 1 compositions is bed in Table 5. The HPLC data are summarized in Tables 6-9. The data are also plotted in Figures 4-7 to show the 3O trends of the purity, and the s of the composition peak 1, the composition with a RRT of 0.43 and the composition peak 3, and the composition peak 7 in the 1 compositions with respect to NaCl.
Table 5. Sodium Chloride Content in the CXA-101 Compositions NaCl content 481.0 mg NaCl per 1000 mg of ceftolozane 190.0 mg NaCl per 1000 mg of ceftolozane 125.0 mg NaCl per 1000 mg of ceftolozane Table 6. Purit of ozane in CXA-101 Com ositions with Var in Amounts of Sodium Chloride 3days/600C 94.9 95.7 94.8 93.9 93.6 (A03) (2.3) (3.9) (4.1) Total A /60 °C 3.07 3.7 5.83 Total A130 "C 0.3 0.6 . 0.7 Table 7. HPLC Peak Area of Com osition Peak 1 in CXA-lOl Com ositions with Var in Amounts of Sodium de t0/600C - - tO/SOOC °C 1 0.86 lday/3OOC 3days/600C 3day/30°C 7days/600C 7day/30°C INCREASE % /60 °C INCREASE % /30 °C 0 15 0.27 0 30 0 33 Table 8. HPLC Peak Area of the Com osition with a RRT of 0.43 and Com osition Peak 3 1n CXA-lOl Com ositions with Var in Amounts of Sodium Chloride 3day/30°C 3 0.17 7day/3OOC 7 .
INCREASE %/60 0C 0.76 Table 9. The HPLC Peak Area of Com osition Peak 7 in CXA-101 Com ositions with Varying Amounts of Sodium Chloride —Da -—-— t0/300C 1.00 1.01 1.02 1day/30°C - . 1.02 60°C - 1.46 3day/30°C - 1.05 7day/30°C INCREASE % /60 0C I 0.39 0.54 0.55 INCREASE % /30 0C 0.01 0.02 0.05 Conclusion: The stability test demonstrates that high sodium chloride content enhances stability of CXA-101 Compositions.
The HPLC measurements on day 3 were used to analyze the stability of the CXA-101 compositions.
CXA-101 compositions comprising high s of sodium chloride (e.g., 125-1000 mg sodium de per 1000 mg of ceftolozane) were found to be more chemically stable than CXA-101 compositions sing low amounts of sodium chloride (e. g., less than 125 mg sodium chloride per 1000 mg of ceftolozane). Table 6 shows that, by day 3 of heating at 60 0C, sample A1, which has the highest salt concentration, is most stable, i.e., has the lowest A[0_[3 of all samples. By day 3, the sample with the lowest salt concentration, A5, has the highest A[0_[3 indicating the most degradation. Overall, A5 has degraded 141% more than A1.
Further, Table 6 shows that, by day 3 of heating at 60 0C, sample A3, which contains a lower salt concentration within the limits of the invention at 125 mg, is still significantly more stable than A4, a composition ning 75.0 mg of the salt. A3 has a A[0_[3 of 3.1, while A4 has a A[0_[3 of 3.9, g that A4 has degraded 26% more than A3.
B. Long-term Stability Study of CXA-101 Pharmaceutical Compositions with Varying Amounts of Sodium Chloride r stability study was carried out at 5 0C and 25 0C. The sodium chloride content in the CXA-101 compositions is bed in Table 9a. The amounts of citric acid and L-arginine in each composition were the same. These samples were in lyophilized form and were placed on long-term (24-36 months), real time stability programs.
The composition peak 1 is considered "diagnostic" for formulation failure because it is the first peak to go out of trend or specification (1.5%). Thus, the stability of these CXA- 101 compositions was also measured by the length of storage until formulation failure as indicated by the composition peak 1. The data in Table 9a were extrapolated from data collected after 4 months. Clearly, based on the amount of the composition peak 1 in the compositions, the ition with about 480 mg sodium chloride per 1 gram ceftolozane active was significantly more stable than the itions containing 125 mg or 62.5 mg sodium chloride per 1 gram of active ceftolozane (i.e., stability of ceftolozane compositions: 480>>125 5 mg).
Table 9a. The Peak 1 Failure Points of CXA-101 Com ositions with Var in s of Sodium Chloride oint at 5 OC noint at 25 0C 245 months 15 months 125 m; NaCl 70 months 5 months 62.5 m- NaCl *Results at 3 months = 1.34%, 4 months = 1.15% e 6: Manufacturing Process ofa CXA-201 Composition sing Tazobactam and CXA-IOI/Ceftolozane by Co-lyophilization The manufacturing process of a CXA-201 composition comprising tazobactam and ceftolozane by co-lyophilization is shown in Figure 2. Non-sterile bulk tazobactam and bulk ceftolozane were mixed, ed by dissolution and e filtration. The filtrate was then tray-lyophilized to obtain the CXA-201 composition. The CXA-201 composition can be container-filled as a final drug product. The components of a CXA-201 composition prepared by co-lyophilization are shown in Table 10.
Table 10. Components of a CXA-201 Composition Prepared by Co ilization ent Function Amount (mg/ ner) Ceftolozane Active pharmaceutical ingredient 1000 (potency) nine Alkalization reagent — Citric acid (anhydrous) Buffer Sodium chloride Stabilizer Tazobactam (free acid) Active pharmaceutical ingredient— Sodium bicarbonate Alkalization reagent Quantity sufficient1 for pH 4.8 to 7.0 Water Dissolution solvent Not more than 4% by weight Nitrogen Inert gas Sufficient quantity 1. Sodium content is approximately 78 mg/g of tazobactam in drug product after lyophilization. 2. Water is removed during the lization process and is controlled at no more than 4% by weight.
Example 7.‘ Assessment of philized Combo Drug Product (i. e., a CXA-ZOI Composition) A. Preparation of the Co-Lyophilized Combo Drug Product tie. the CXA-201 Composition) The components of the co-lyophilized CXA-201 composition are shown in Table 11.
This composition was prepared, as described above in Example 6.
Table 11: Components of the CXA-201 Composition Prepared by Co-Lyophilization CXA-201 Comp.
Citric acid NaCl B. Stressed Stability Test Stability studies of this CXA-201 composition prepared by co -lyophilization were carried out at 25 OC and 40 oC. The ition was analyzed using HPLC. The following Tables 12 and 13 are summaries of the HPLC ements at time zero, after one month (T1), and after three months (T2).
Table 12: Stabilit Data of Co-L o hilized CXA-201 Com osition at 25 oC/RH=60% Test items Spec. D.P. T1 25°C T2 25°C 0.31% 0.54% 0.71% 0.07% 0.07% 0.09% g 0.30% <0.03% <0.03% <0.03% 3 0.80% 0.08% 0.08% 0.09% g 1.00% 0.26% 0.29% <0.03% <0.03% <0.03% 0.64% 0.65% 0.66% -Peak8 g 0.15% <0.03% <0.03% <0.03% -Peak9 g 0.60% 0.10% -Peak10,11 g 0.15% each 0.04% -Peak12 g 2.00% <0.03% Others (RRT 0.43) g 0.15% 0.04% Others (RRT 1.22) g 0.15% 0.38% Others (RRT 2.18) g 0.15% 0.05% Others (RRT 2.77) g 0.15% 0.03% Sing. Unk. g 0.15% 0.05% Total 5 5.00% 2.77% pH report value -- 4.83 Table 13: Stabilit Data of Co-L o hilized CXA-201 Com osition at 40 oC/RH=75% Test items T1 40°C T2 40°C Related Substances 3 1.50% 2.22% g 0.40% 0.16% g 0.30% 0.06% g 0.80% 0.09% g 1.00% 0.30% <0.03% <0.03% <0.03% 0.64% 0.69% 0.78% -Peak8 30.15% <0.03% <0.03% 0.10% -Peak9 g 0.60% 0.05% 0.09% 0.09% -Peak10,11 g 0.15% each 0.04% 0.04% 0.05% -Peak12 g 2.00% <0.03% <0.03% <0.03% Others (RRT 0.43) g 0.15% <0.03% 0.09% 0.15% Others (RRT 1.22) g 0.15% 0 13% 0.74% Others (RRT 2.18) 0 03% <0.03% Others (RRT 2.77) <0.03% <0.03% 0.05% 0.11% 1.67% 4.49% C. sion: A new compound having RRT=1.22 was observed in the co-lyophilized CXA-201 compositions. While not wishing to be bound by theory, the nd RRT 1.22 was identified as a compound formed by a reaction between ceftolozane and formylacetic acid, which was a duct of tazobactam as illustrated in Marunaka et al. (Chem. Pharm. Bull. 1988, Vol. 36 (11), pp. 4478-4487).The stability data at 25 OC and at 40 0C have confirmed the continued formation of the compound RRT 1.22 over the course of time.
Exam 16 7a: Identi in the com ound 0 ormula III The Co-Lyophilized Combo Drug Product was prepared as described above in e 6. The formulation composition of the Co-Lyophilized Combo drug product is shown in Table 11 (Example 7). This sample maintained at 25 oC/RH=60% and 40 oC/RH=75% after one month (T1) and three months (T2). Samples were analyzed using a HPLC method as described in Example 1. The data for is of the samples by HPLC is shown in Example 10 in Table 23 (Stability data of Co-Lyo Combo Drug Product at 25 oC) and Table 24 (Stability data Co-Lyo Combo Drug t at 40 OC). The presence of the compound of Formula (111) was identified has having a retention time of about 1.22 as measured by HPLC (see Example 2). RRT=1.22 was observed in co-lyophilized drug product. The compound of formula (III) is ed to be formed by a reaction between ozane and formylacetic acid, which was a degradation product of tazobactam. The amount of the compound of formula (III) in a composition sing ozane and tazobactam can be increased over time at 25 OC and at 40 oC.
The material obtained from the RRT 1.22 peak was analyzed by LC/MS, providing the spectra shown in Figure 14. Figure 15 (below) is the corresponding structures for the peaks shown in Figure 14.
A test sample prepared from ceftolozane and tazobactam acid co-compounding on containing RRT 1.22 impurity was used on the LC/MS experiment. Liquid chromatography separation was med on a Zorbax SB CS, 3.5um, 3.0mm x 150mm column, using gradient elution with 20mM ammonium formate containing 0.1% Heptofluorobutyric acid pH 3.2 as mobile phase A and 0.1% Heptofluorobutyric acid in acetonitrile as mobile phase B. The gradient starts from 3% (initial) to 15% mobile phase B in 20 minutes (with RRT 1.22 g at about 10.7 minutes). Mass detection was performed using electrospray ionization technique under positive mode. The column effluent was also monitored at 254 nm using a photodiode-array detector. MS/MS fragmentation was performed on m/z 737.3 positive ion using nitrogen as collision gas, with collision energy set at 35V.
Example 8.‘ Stabilizing Eflect ofSodium Chloride in CXA-ZOI Compositions A. ion of the Composition at RT=63 minutes in 1 Compositions A stability study was carried out at 25°C and analyzed by HPLC. CXA-201 compositions comprise ceftolozane and tazobactam, further comprising high, mid, or low s of sodium chloride (480, 125, or 62.5 mg NaCl per 1000 mg of ceftolozane, respectively). Comparison of the compositions are listed in Table 14. The amounts of the composition RT 63’, as measured by the HPLC method, are summarized in Table 15.
Table 14. Comparison of the CXA-201 Compositions Lot CXA-lOl NaCl Tazobactam Table 15. RT 63’ Peak Area at t: 3 months, 25 oC/60% RH storage --lSt data collection 2nd data tion 3rd data collection High salt+TazoNa 63.90 63.30 62.49 "I." Conclusion: At the three month time point, the reduced salt formulations were observed to be not as stable as the full salt formulation; and trends indicate that reduction in salt causes at least 1.5-fold greater composition at RT: 63 minutes, as measured by HPLC.
The compositions comprising 480 mg NaCl per 1000 mg of ceftolozane had the least amount of the composition RT 63’ after 3 months at 25 0C. The amount of the composition RT 63’ in the compositions comprising 125 mg NaCl per 1000 mg of ceftolozane was 1.5 -fold or greater than the amount of the composition 63’ in the compositions sing 480 mg NaCl per 1000 mg of ceftolozane. The amount of the composition RT 63’ in the compositions comprising 62.5 mg NaCl per 1000 mg of ceftolozane was 2-fold or greater than the amount of the composition RT 63’ in the itions sing 480 mg NaCl per 1000 mg of ceftolozane. Thus, the CXA-201 itions comprising high amounts of sodium chloride (e.g., 125-1000 mg sodium chloride per 1000 mg of ceftolozane) were more chemically stable than the compositions comprising low amounts of sodium chloride (e.g., less than 125 mg sodium chloride per 1000 mg of ceftolozane).
B. Improvement in the Purity of Ceftolozane in CXA-201 Pharmaceutical Compositions with g s of Sodium Chloride A stability study was carried out at 30 0C and 60 0C analyzed by HPLC. The sodium chloride t in the CXA-201 compositions is described in Table 16. The HPLC data at 60 0C are summarized in Tables 17-20. The data are also plotted in Figures 8-11 to show the trends of the purity, and the amounts of the composition peak 1, the composition with a RRT of 0.43 and the composition peak 3, and the composition peak 7 in the CXA-201 compositions with respect to NaCl.
Table 16. The Sodium Chloride Content in the CXA-201 Compositions 481.0 mg sodium chloride per 1000 mg of ceftolozane 125 .0 mg sodium chloride per 1000 mg of ceftolozane 75.0 mg sodium chloride per 1000 mg of ceftolozane 50.0 mg sodium chloride per 1000 mg of ceftolozane Table 17. The Purit of Ceftolozane in CXA-201 Com ositions with Var in Amounts of Sodium Chloride 1day/60°C 1day/30°C 60°C (Aw—:3) 3day/300C 7days/60°C 7 Table 18. The HPLC Peak Area of ition Peak 1 in CXA-201 Compositions with Var in Amounts of Sodium Chloride 1day/60°C 1day/30°C 3days/60°C U) 3day/300C 7days/60°C 7day/30°C INCREASE% 2’60 °C INCREASE% /30 °C Table 19. The Total HPLC Peak Area of the Composition with a RRT of 0.43 and Composition Peak 3 in CXA-201 Com ositions with Var in Amounts of Sodium Chloride lday/30°C 3days/60°C 3days/30°C 7days/60°C 7days/30°C INCREASE% 2’60 oC INCREASE% 2’30 oC Table 20. The HPLC Peak Area of Composition Peak 7 in CXA-201 Compositions with Var in Amounts of Sodium de lday/60°C lday/30°C 3days/60°C °C 7days/60°C 7days/30°C INCREASE% /60 0C INCREASE% 130 °C Conclusion: The stability data shows that high sodium chloride t enhances stability of CXA-201 compositions.
Similarly to CXA-lOl compositions, CXA-201 compositions comprising high amounts of sodium chloride (e.g., 125-1000 mg sodium chloride per 1000 mg of ceftolozane) were found to be more chemically stable than CXA-201 compositions comprising low amounts of sodium chloride (e. g., less than 125 mg sodium chloride per 1000 mg of ceftolozane). Table 17 shows that, by day 3 of heating at 60 0C, sample B1 containing the highest salt concentration is most stable, i. e., has the lowest A[0_[3 of all samples. By day 3, the sample with the lowest salt concentration, B4, has the highest A[0_[3 ting the most degradation. Overall, B4 has degraded 15% more than B1.
Example 9.‘ Manufacturing Process ofa CXA-201 Composition (Comprising tazobactam and ceftolozane) by Blending A. e Dry Blending of Bulk Lyophilized Ceftolozane and Bulk Lyophilized Tazob actam A low energy drum blender that agitates the material by tumbling and also moving the bed up and down is used. A representative process of blending is described as s, also shown in Figure 1. The blender was charged with 23.4 kg of CXA-101 bulk product, and 5.4 kg of tazobactam bulk product. Both the CXA-101 and tazobactam were dually lyophilized hand. The material was blended for 180 s. In-process tests of content assay for both CXA-101 and tazob actam were performed to assess the homogeneity using the samples of blend materials taken from three places. The relative standard deviation (RSD) for each of CXA-101 and tazobactam content assay was no greater than 2% and the RSD for the ratio of CXA-101/ ctam was no greater than 2% (See Table 21).
Table 21. In-Process g of Blending Samples of a CXA-201 Composition at Three Places Results Acceptance Limits Samp 111%1' (expected value) minute minute 34.24 34.07 34.42 34.62 34.21 34.66 Content: Ceftolozane1 30.4%-37.2% 34.71 34.60 34.85 Mean . 34.64 RSD% . . 0.63 .2%-18.6% 17.96 18.20 17.12 1.87 2.01 1.87 2.10 Ratio of Content (w/w) 2.04 2.05 ozane/tazobactam 2.05 RSD = relative standard deviation Theoretical value: 33.96% Acceptance limits are 90% - 110% of the theoretical value. 2 Theoretical value: 16.99% Acceptance limits are 90% - 110% of the theoretical value. 3 Three samples are taken at each time point at three places to measure the percentage by weight of ceftolozane and tazobactam. The "Mean" is the average of the percentages or the weight ratios of Ceftolozane/tazobactam.
Acceptance limits were established based on batch history.
B. ing into Sterbags® The blended powder is then discharged into Sterbags®.
C. Finished CXA-201 Drug Product A fill and finish process is utilized for the final drug product, which is a pharmaceutical composition sing CXA-101 and tazobactam at a ratio of 1000 mg/ 500 mg. Glass containers are washed with WFI and depyrogenated in a Class 100 depyrogenation tunnel at a temperature of 320°C. Pre-washed and pre-siliconized stoppers are autoclaved for 40 minutes at 121°C. The bulk drug product is packaged in a Sterbag® system comprised of three bags. The outer bag is cleaned with disinfectant in a Class 10,000 clean room. The bag system is placed in a pass-through UV box where it is subjected to UV radiation (> 20 uW/cmz) for 20 minutes to sterilize the surface of the outer bag. The outer bag is d and left in the UV box. The middle bag is placed in a Class A laminar airflow (LAF) hood. The sterile middle bag is removed under LAF. The sterile, bottle- shaped inner bag is then placed in a sterile stainless steel carrier and attached to the filling machine.
Sterile bulk CXA-lOl/ tazobactam drug product is filled under a en blanket into -mL, Type I clear glass containers. The sterile drug product is graVity-fed into the filling machine under LAF. container fill s are ically checked throughout the filling ion to ensure proper ion of the filling line. Filling and stoppering operations are performed under Class 100 LAF conditions. Capping and container washing are done in the Class 10,000 clean room.
Example 10: Assessment ofBlend Combination Drug Product A. Preparation of the Blend Combination Drug Product (CXA-201 Composition) The blend drug product was prepared, as bed above in Example 9, on lab scale using a small blender. The components of the blend composition are shown in Table 22.
Table 22: Components of the Blend Composition Component Composition Quantity as active components Ceftolozane 10.8 g 1 for Injection Bulk (25 g) L-Arginine 6.7 g CXA-201 Comp.
Citric acid 233 mg Sodium chloride 5.2 g Tazobactam sodium sterile Bulk 5.4 g (as Tazo free (6 acid) B. Stressed Stability Test Stability studies of this CXA-201 composition prepared by the ng process were carried out at 25 OC and 40 oC. The composition was analyzed using HPLC method described in Example 1. The following Tables 23 and 24 are summaries of the HPLC measurements at time zero, after one month (T1), and after three months (T2).
Table 23: Stabilit Data of Blend CXA-201 Com osition at 25 oC/RH=60% g 1.50% 0.61% 0.93% 1.08% -Peak2 -Peak3 -Peak4 -Peak5 -Peak6 -Peak7 -Peak8 -Peak9 -Peak10,11 < 0.30% <0.03% 0.04% 0.05% Sing. Unk.
Total Assay 1 Teor. %=32.6% 32.5% n.a. n. a.
Assay Tazobactam Teor. %=17.4% 18.2% n.a. n. a.
Tazobactam Related g 4.0% 0.07% 0.12% 014% Compound A TE < 4.0% 2.6% n.a. n. a. .0-7.0 6.0 5.6 5.1 Table 24: Stability Data of Blend CXA-201 Composition at 40 oC/RH=75% Specifications T0 T1 40°C T2 40°C Related Substances 3 1.50% 0.61% 1.66% 2.28% -Peak2 -Peak3 g 0.80% 0.03% 0.04% 0.05% g 1.00% 0.09% 0.13% 0.14% .15% <0.03% <0.03% <0.03% Sing. Unk. g 0.15% 0.13% Total 5 5.00% 2.49% Assay CXA-101 Teor. %=32.6% 32.5% n.a.
Assay Tazobactam Teor. %=17.4% 18.2% n.a.
Tazobactam d 5 4.0% 0.07% 0.35% Compound A pH 0 6.0 5.0 4.4 C. Conclusion The data at both 25 OC and at 40 0C have shown that the blending process tely inhibits formation of the compound RRT=1.22.
Example I I .' Alkalizing Agent Selection Compositions for enous administration should be formulated to resemble the and pH of human blood to reduce vascular complications. The recommended pH is between 5 and 9 (ideal pH is as close to 7.4 as possible). Departing from this recommended pH ranges of an intravenously administered composition can result in the development of complications such as phlebitis, or inflammation of the veins. Marc Stranz, A Review ofpH and Osmolarity, 6 Int’l J. of Pharm. nding 216, 218 (May/June 2002). Unfortunately, few drug infusions are stable at a suitable pH for intravenous administration. Depending on the molecular structure, a drug is most stable or has the best solubility at a particular pH range (e. g., pH< 6) and divergence from this pH range may lead to increased drug decomposition.
It is thus challenging to find a balance between the safe limits of pH and optimum drug ity in compositions for enous administration. Marc Stranz, The Implications of Osmolality, Osmolarity andpH in Infusion Therapy, INS Annual Conference (May 2005).
A formulation close to physiologic pH was targeted. This necessitates an alkalizing agent due to intrinsic pH 1.92 of ceftolozane in solution (2%). The initial study of alkalizing agents included sodium hydroxide, L-arginine, tris, sodium bicarbonate, meglumine, diethanolamine, and triethanolamine. Samples containing 100 mg ceftolozane sulfate, 22.9 mg sodium chloride, 200 mg maltose, and 2 mg citric acid anhydrous were prepared and adjusted to ~ pH 4. The samples were lyophilized and powders stored at 70 0C for 3 days, 60 0C for 3, 6 and 9 days and at 40 0C for one month. The stored s were then analyzed for ozane content. Results are ed below in Table 25: Table 25: Effect of Alkalizin A ent on Ceftolozane Recover Storage i a: a bicarbonate a: arginine lamine o I .51 0dium l riethanolamine W .4 H m 2 b H —-----m Ceftolozane recovery was consistently above 90% in the presence of sodium hydroxide, nine, or sodium bicarbonate. Although sodium hydroxide performed well, as a strong base, it could promote base hydrolysis of the active more readily during scale up and be more difficult to dry during lyophilization than other alkalizing agents. Accordingly sodium hydroxide was not considered for further formulation development. L-arginine was thus chosen as the alkalizing agent for the formulation.
To ensure suitability of L-arginine as an alkalizing agent, a study was conducted to compare L-arginine against sodium bicarbonate. In this study, solutions were prepared to contain ceftolozane in the presence of sodium chloride and citric acid adjusted to imately pH 6 with either L-arginine or sodium bicarbonate. The solutions were then lized and samples distributed for accelerated and regular storage. A summary of the total additional compounds and pH for the various ions after 1 month is presented in Table 26.
Table 26: Effect of L-Ar inine and Sodium Bicarbonate on Ceftolozane Related Substances during Storage, pH 6 Bulk solution composition per 632 mg L-arginine 288 mg sodium bicarbonate 1000 mg ceftolozane free base 485 mg sodium chloride 481 mg sodium chloride 21 mg citric acid 21 mg citric acid Storage condition Total Total Related Related Substances Substances 40 OC,1month 2.32% 5.93% As seen in the table the bicarbonate-adjusted sample showed a larger se in related substances and a less stable pH profile. Accordingly, it was decided to maintain L- ne as the alkalizing agent in the formulation. e 12: Components ofa CXA-ZOI composition An example of a batch formulae for ceftolozane ition (compounding of ceftolozane substance with excipients such as citric acid, sodium chloride, and nine followed by sterile lyophilization) is found below in Table 27.
Table 27: Batch Formula for Ceftolozane composition Component Target Composition Amount per Batch (kg) mg/g — Ceftolozane Sulfatei) 172.1 114.0 202.6 L-Arginine, USP ~90 59.7 106.0 QS to achieve target pH Water for Injection, USP QS to 1000 Total Batch SizeW 1175 1) Ceftolozane sulfate is charged based on its measured potency to obtain 150 mg free base/g solution. 2) L-arginine is added as needed to obtain pH 6.5 i 0.5 in the bulk solution; 90 mg per gram solution is ered a representative amount.
An example of a batch formula for the ceftolozane/tazobactam drug t is presented in Table 28 below.
Table 28: Batch Formula Ceftolozane/Tazobactam Drug Product Component Amount per Amount per Batch, ner, mg kg Ceftolozane 2255 1 12.8 compositionl) compositionj) The target fill for ceftolozane is 1000 mg free base, added to the container as the composition. The amount 2255 mg is based on 100% theoretical potency of the composition. Actual weight will vary based on ition measured potency. ii)The target fill for tazobactam is 500 mg free acid, added to the container as its sodium salt form. The amount 537 mg is based on 100% theoretical y. iii) Nitrogen is used as a processing aid to blanket containers after powder filling and prior to insertion of stopper.
The unit composition of a dosage for reconstitution is described in Table 29.
Table 29: Unit Com ositions of Ceftolozane/Tazobactam for In'ection 1000 m / 500 m Component Function Nominal Composition mg per container Ceftolozane Ceftolozane Active 1 147 ition" Sulfate Citric Acid, Chelating Agent 21 Anhydrous Sodium Stabilizing Agent 487 L-Arginine Alkalizing Agent 600 Q.S. for pH adjustment Tazobactam Sodium Total Weiht 2792 1) Actual amount of ceftolozane composition will vary based on the measured potency. Ceftolozane sulfate, 1147 mg, corresponds to 1000 mg ceftolozane free base. 2) L—arginine is added as needed to achieve pH 6.5 i 0.5; 600 mg per container is considered a representative total amount. 3) Actual weight of tazobactam sodium will vary based on the measured potency. Tazobactam sodium 537 mg, ponds to 500 mg tazobactam free acid 4) Nitrogen blanket is applied after powders are dispensed to the container and prior to insertion of stopper.
Example 12a: pment and Implementation ofa System to Prevent Cross-Contamination in Accordance with FDA Guidance A ly published (April 2013) Food and Drug stration Guidance for Industry Non-Penicillin Beta-Lactam Drugs: A CGMP Frameworkfor Preventing Cross- Contamination provides direction on prevention of cross-contamination for facilities that manufacture non-penicillin beta-lactam drugs. Provided herein are steps for the development and entation of a system to t cross-contamination due to the introduction of both sterile ceftolozane drug t intermediate and tazob actam sodium into a facility that is in conformance with FDA Guidance.
Segregation steps to conform with FDA Guidance can include, but are not limited to: Relocation all other drug products to other sites Separating the ceftolozane/tazobactam product filling line and the nary cephapirin product filling line Creating separate HVAC systems ishing separate warehouse areas Formalizing separate material, waste and personnel flows Constructing temporary facilities for gowning and entrance to the line used for the ceftolozane/tazobactam drug product.
Constructing new walls, ing and reinforcing existing walls Equipping the existing emergency egress with alarms and gaskets to completely separate both lines throughout all the floors of the building Creating the permanent separation of locker, rest and break rooms for both lines of the facility: Dedicated maintenance and operations personnel for each part of the facility including different uniform colors for each part of the facility Dedicated equipment and tools for each part of the ty An ncy Recovery plan Example 13: Physicochemical and Biological Properties Ceftolozane/Tazobactamfor Injection, 1000 mg/ 500 mg As a product intended for intravenous use, several properties are ant for physiological compatibility. These include particulate matter, sterility, endotoxin limit, pH, and osmolality. Particulate matter and sterility are controlled at the point of manufacture.
The drug product is processed aseptically throughout the entire cturing process, inclusive of ceftolozane, tazobactam sodium, and ceftolozane/tazob actam in- container drug product.
The ceftolozane/tazobactam drug product is controlled to approximately pH 6, to provide physiological t, while still assuring adequate stability for the drug substances.
The ceftolozane drug product intermediate is controlled during compounding to pH 6.5 i 0.5 and is controlled at release to pH 5 to 7. The tazobactam sodium is controlled at release to pH to 7.
Ceftolozane/tazobactam ing reconstitution with normal saline and dilution for on also in normal saline (10 mg/mL ceftolozane; 5 mg/mL tazobactam) is slightly hypertonic, with osmolality approximately 500 mOsm/kg. However, slightly hypertonic intravenous infusion solutions are not uncommon as drug products are commonly prepared and diluted with already-isotonic solutions, such as normal saline. The lly accepted m upper limit for peripheral intravenous administration is approximately 900 mOsm/kg, though admixtures 600 to 900 mOsm/kg are lly administered through a central line. Therefore, to be within the limits of this range, the infusion product is less than 600 mOsm/kg.
Example [4: Determining Osmolality of CXA-ZOI Compositions CXA-lOl and Tazobactam Sodium samples (#1 - #3) were reconstituted as follows: Sample#l: Weighed 0.103g of Tazobactam Sodium and 0.462g of CXA-lOldissolved in 4mL of WFI Water and 6mL of USP Normal Saline.
Sample#2: d 0.103g of ctam Sodium and 0.462g of CXA-lOldissolved in 4mL of WFI Water added lOmL of USP Normal Saline.
Sample#3: Weighed 0.103g of Tazobactam Sodium dissolved in lmL of WFI Water and 0.462g of CXA-lOldissolved in lmL of WFI Water then mixed er added lOmL of USP Normal .
Tazobactam Sodium (Potency: 97.5%) CXA-lOl (Potency: 43.3%) WFI Water USP Normal Saline The osmolality of l and Tazobactam Sodium samples (#1 - #3) was then determined using a freezing point depression Osmometer (available from Advanced Instruments, Inc.).
Table 30: Osmolality of Reconstituted Solutions (2 XA-101 Tazobgctam Sample}? Cone; 833:2" WFI Water Saline fig???) (mgImLi m(mi.
A unit dosage form composition of Table 29 was reconstituted with lOmL of Sterile WFI or USP Normal Saline then added into lOOmL 5% Dextrose Injection (D5W) or 0.9% Sodium Chloride (NS) bags and the lity of the resulting bag solution was determined as shown in table Table 30a below.
Table 30a: Osmolarit of Ceftolozane Ba Solution mOsm/k sWFI — D5W NS — D5W sWFI — NS In Table 30a, data for osmolality of the following product reconstitution scenarios was determined using the composition from Table 29 0 5% Dextrose Injection USP, lOOmL Bag (Baxter) 0 0.9% Sodium de Injection USP, lOOmL Bag (Baxter) 0 sWFI — D5W: reconstituted with Sterile WFI then added into 5% Dextrose Injection bag 0 NS — D5W: reconstituted with USP Normal Saline then added into 5% se Injection bag 0 sWFI — NS: reconstituted with Sterile WFI then added into 0.9% Sodium Chloride Injection Bag 0 NS — NS: reconstituted with USP Normal Saline then added into 0.9% Sodium Chloride Injection bag Exam le 15: Exci ients in Ce tolo ane Dru t Intermediate The excipients in exemplary ceftolozane compositionswere chosen to ensure stability and processability of the ceftolozane drug nce into the drug product. The specific excipients, their quantities and functions are provided in Table 31. All ents are compendial and typical for sterile pharmaceutical do sage forms, requiring no additional treatment prior to use in the ation. The excipients are used in levels within the range established in other FDA approved products as described in the Inactive Ingredients Database (IID).
Table 31: Excipients Used in Ceftolozane Composition Component Function , Concentration ale for Inactive mg/ in Infusion ion Ingredients container Solution, % Database (IID) Range Citric acid Chelating Used to prevent 0.0025 to 50% agent discoloration and degradation Sodium Stabilizing Used as a stabilizing 0.187 to 45% de agent agent for ceftolozane sulfate L-arginine Alkalizing 600" Used to adjust 0.29 to 88% agent Q.S. for pH ceftolozane solution adjustment pH L—arginine is added as needed to achieve pH 6.5 i 0.5; 600 mg per container is considered a representative total amount.
Example 16: cturing Process ofa CXA-ZO] Composition (Comprising tazobactam and ceftolozane) by ling The ceftolozane /tazobactam finished drug t is a sterile powder fill of lyophilized active ingredients ceftolozane drug product intermediate (composition) and tazobactam sodium together into a e single container. The lyophilized form of the sterile tazobactam sodium contains no excipients. Ceftolozane sulfate drug substance is converted first into a sterile drug product intermediate, composition, by formulation with citric acid, sodium chloride and L-arginine, followed by lyophilization.
The full manufacturing process includes unit operations typical of an aseptic lyophilization process and aseptic powder filling process. The overall process can be outlined in two stages, as presented in the manufacturing flow chart of Figure 12. The first stage is the manufacturing of the sterile ceftolozane composition. The second stage is the g of the sterile drug powders into ners for the final drug product. The major s steps are: Preparation of the sterile ceftolozane composition comprises compounding the bulk solution for lyophilization; sterile filtering the bulk solution; lyophilizing the bulk solution into bulk powder; grinding and sieVing of the sterile bulk powder; and c packaging of the sterile bulk powder in Sterbags ®.
Filling of the sterile bulk powders comprises receipt of ceftolozane and ctam sterile powders at site; aseptic filling both sterile powders into the container sequentially; blanketing the container with a nitrogen headspace; stoppering and crimping the container; and inspecting the container prior to secondary packaging. [text continues on next page] Definitions of specific embodiments of the invention as claimed herein follow.
According to a first embodiment of the invention, there is provided an antibiotic pharmaceutical composition sing ceftolozane sulfate (or a pharmaceutically acceptable salt thereof) and tazobactam sodium (or a pharmaceutically acceptable salt thereof) in a fixed dose combination of 1,000 mg of ozane active per 500 mg of tazobactam active, and a ceftolozane-stabilizing amount of 125 mg to 500 mg sodium chloride per 1,000 mg of ceftolozane active wherein the ceftolozane sulfate is obtained by a process comprising lyophilizing an aqueous solution comprising 125 mg to 500 mg sodium chloride with an amount of ceftolozane sulfate providing 1,000 mg of ceftolozane active.
According to a second embodiment of the invention, there is provided a pharmaceutical composition comprising stabilized ceftolozane sulfate obtained by a process comprising lyophilizing an aqueous solution sing 125 mg to 500 mg sodium chloride with an amount of ceftolozane sulfate providing 1,000 mg of ceftolozane active, to obtain the lyophilized stabilized ceftolozane sulfate composition.
According to a third embodiment of the invention, there is provided a container sing a pharmaceutical composition of the second embodiment, n the process further comprises a step of filling the lyophilized stabilized ceftolozane composition into a container.
According to a fourth embodiment of the invention, there is provided a unit dosage form container comprising the antibacterial ition of the first embodiment comprising a pharmaceutical ition containing 1,000 mg of ceftolozane active and 500 mg of ctam active.
According to a fifth embodiment of the invention, there is provided a unit dosage form container sing the cterial composition of the first embodiment comprising a pharmaceutical composition containing 2,000 mg of ceftolozane active and 1,000 mg of tazobactam active.
In the t specification and claims, the word ‘comprising’ and its derivatives including ‘comprises’ and ‘comprise’ include each of the stated integers but does not exclude the inclusion of one or more r integers.
The reference to any prior art in this specification is not, and should not be taken as an acknowledgement or any form of suggestion that the prior art forms part of the common general knowledge.

Claims (16)

What is claimed is:
1. An antibiotic pharmaceutical composition comprising ceftolozane sulfate (or a pharmaceutically acceptable salt f) and tazobactam sodium (or a pharmaceutically acceptable salt thereof) in a fixed dose combination of 1,000 mg of ceftolozane active per 500 mg of tazobactam active, and a ceftolozane-stabilizing amount of 125 mg to 500 mg sodium chloride per 1,000 mg of ceftolozane active, wherein the ceftolozane sulfate is obtained by a process comprising lyophilizing an aqueous solution sing 125 mg to 500 mg sodium chloride with an amount of ozane sulfate providing 1,000 mg of ceftolozane active.
2. The pharmaceutical composition of claim 1, further comprising L-arginine.
3. The pharmaceutical composition of claim 1 or 2, further comprising citric acid.
4. The pharmaceutical composition of any one of claims 1-3, wherein the ceutical composition is lyophilized.
5. A pharmaceutical composition comprising stabilized ceftolozane sulfate obtained by a process comprising lyophilizing an aqueous solution comprising 125 mg to 500 mg sodium de with an amount of ozane sulfate providing 1,000 mg of ozane active, to obtain the lyophilized stabilized ceftolozane sulfate composition.
6. The pharmaceutical composition of claim 5, wherein the stabilized ceftolozane is obtained by lyophilizing the sodium chloride and ceftolozane sulfate with L-arginine.
7. The ceutical composition of claims 5 or 6, n the stabilized ceftolozane is obtained by lyophilizing an aqueous solution having a pH of about 6.0 to 7.0.
8. The pharmaceutical ition of any one of claims 5-7, wherein the stabilized ceftolozane is obtained by lyophilizing the sodium chloride and ceftolozane e with L- arginine and citric acid.
9. The pharmaceutical composition of any one of claims 1-3 and 5-8, wherein the pharmaceutical composition is formulated for parenteral administration.
10. The pharmaceutical composition of any one of claims 1-3 and 7-9, wherein the composition is a unit dosage form in a container comprising 125 mg to 500 mg sodium chloride, 1,000 mg of ceftolozane in the form of ozane sulfate, and L-arginine.
11. A container comprising a pharmaceutical composition of any one of claims 5-10, wherein the process further comprises a step of filling the lyophilized ized ozane composition into a container.
12. The container of claim 11, further comprising tazobactam or a pharmaceutically acceptable salt thereof.
13. A unit dosage form container comprising the cterial composition of any one of claims 1-10 comprising a pharmaceutical composition containing 1,000 mg of ceftolozane active and 500 mg of tazobactam .
14. The unit dosage form of claim 13 for the treatment of intra-abdominal infections or urinary tract infections.
15. The unit dosage form of claims 13 or 14, wherein the unit dosage form container is a vial or bag enclosing the ceftolozane and the tazobactam.
16. A unit dosage form container comprising the cterial composition of any one of claims 1-10 comprising a pharmaceutical composition containing 2,000 mg of ceftolozane active and 1,000 mg of tazobactam active.
NZ711823A 2013-03-15 2014-03-14 Ceftolozane antibiotic compositions NZ711823B2 (en)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US201361792092P 2013-03-15 2013-03-15
US201361793007P 2013-03-15 2013-03-15
US61/792,092 2013-03-15
US61/793,007 2013-03-15
US201361882936P 2013-09-26 2013-09-26
US61/882,936 2013-09-26
US201361893436P 2013-10-21 2013-10-21
US61/893,436 2013-10-21
NZ700372A NZ700372A (en) 2013-03-15 2014-03-14 Ceftolozane antibiotic compositions

Publications (2)

Publication Number Publication Date
NZ711823A NZ711823A (en) 2021-05-28
NZ711823B2 true NZ711823B2 (en) 2021-08-31

Family

ID=

Similar Documents

Publication Publication Date Title
JP7177314B2 (en) Ceftolozane antibiotic composition
US11278622B2 (en) Ceftolozane antibiotic compositions
US9925196B2 (en) Ceftolozane-tazobactam pharmaceutical compositions
AU2015200599B2 (en) Ceftolozane Antibiotic Compositions
NZ711823B2 (en) Ceftolozane antibiotic compositions