WO2005117831A1 - Injectable pharmaceutical suspension comprising posaconazole - Google Patents

Injectable pharmaceutical suspension comprising posaconazole

Info

Publication number
WO2005117831A1
WO2005117831A1 PCT/US2005/018945 US2005018945W WO2005117831A1 WO 2005117831 A1 WO2005117831 A1 WO 2005117831A1 US 2005018945 W US2005018945 W US 2005018945W WO 2005117831 A1 WO2005117831 A1 WO 2005117831A1
Authority
WO
WIPO (PCT)
Prior art keywords
formulation
posaconazole
mean
plasma
hour
Prior art date
Application number
PCT/US2005/018945
Other languages
French (fr)
Inventor
Leonore Witchey-Lakshmanan
Sydney Ugwu
Varda Sandweiss
Catherine Hardalo
Roberta S. Hare
Gopal Krishna
Zaiqi Wang
Marco Taglietti
Original Assignee
Schering Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=34971355&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2005117831(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Schering Corporation filed Critical Schering Corporation
Priority to BRPI0510417-3A priority Critical patent/BRPI0510417A/en
Priority to CA002567803A priority patent/CA2567803A1/en
Priority to AU2005249502A priority patent/AU2005249502A1/en
Priority to MXPA06013886A priority patent/MXPA06013886A/en
Priority to JP2007515456A priority patent/JP2008501034A/en
Priority to EP05755118A priority patent/EP1761247A1/en
Publication of WO2005117831A1 publication Critical patent/WO2005117831A1/en
Priority to IL179627A priority patent/IL179627A0/en
Priority to NO20066005A priority patent/NO20066005L/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • 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
    • A61K47/02Inorganic 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
    • A61K47/24Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing atoms other than carbon, hydrogen, oxygen, halogen, nitrogen or sulfur, e.g. cyclomethicone or phospholipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • 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

Definitions

  • the present invention relates to formulations useful for treating infections.
  • these formulations include the active pharmaceutical ingredient posaconazole in an injectable suspension that is stable when subjected to terminal steam sterilization, and throughout
  • Posaconazole an anti-fungal agent, represented by the following chemical
  • posaconazole other formulations of posaconazole have been disclosed.
  • a solid (capsule/tablet) of posaconazole is disclosed in U.S. Patent Nos. 5,972,381 and 5,834,472.
  • a topical form of posaconazole e.g., a lotion, cream, ointment, or "lacquer nail polish” is contemplated based on other similar formulations, e.g., U.S. Patent No. 4,957,730 (PENLAC® available from Demiik®).
  • U.S. Patent No. 5,858,410 discloses pharmaceutical compositions containing particles of active agents of average diameter less than 5 microns, having been conrminuted, without prior conversion into a melt, by using a piston-gap homogenizer.
  • U.S. Patent Application No. 10/440,368 discloses the use of a phospholipid surface active agent to stabilize rnicroparticles of solid fenofibrate in an orally ad ⁇ rju ⁇ istered pharmaceutical composition.
  • Patent No. 5,091, 188 discloses the use of phospholipids, to prevent coalescence of i crocrystalline active agents in injectable pharmaceutical
  • compositions examples include lecithin,
  • phosphatidic acid phosphatidyl ethanolamine
  • cholesterol steaiylamine
  • glycolipids glycolipids and mono-glycerides.
  • the present invention provides formulations of posaconazole that are stable when subjected to terminal steam sterilization. These formulations are useful for the treatment of infections.
  • an aqueous injectable suspension of posaconazole that is homogenously suspended in vehicle with the aid of a phospholipid.
  • a thermoprotectant agent is employed to reduce autoclave-induced particle size growth, as well as a buffer system to stabilize the phospholipid during autoclaving.
  • the formulations provided remain stable after 20 minutes of autoclaving at 121°C and after subsequent storage at 4°C to 40°C for at least 6 months.
  • the present invention provides formulations comprising a suspension of posaconazole, stabilized by a phospholipid, in a mixture comprising a thermoprotectant, and a buffer system.
  • the formulation has been sterilized by autoclaving or by irradiation.
  • the buffer system comprises sodium phosphate, which may be provided as sodium phosphate monobasic monohydrate,
  • sodium phosphate dibasic anhydrous or the combination of the two.
  • the buffer system comprises an organic buffer.
  • the buffer system comprises at least one of histidine, citric acid, glycine, sodium citrate, arnrnoniu ⁇ i sulfate, or acetic acid.
  • the buffer system maintains a pH of about 3.0 to about 9.0.
  • the buffer system mamtains a pH of about 6.0 to about 8.0.
  • the buffer system mamtains a pH of about 6.4 to
  • the phospholipid comprises a natural
  • the phospholipid comprises a synthetic
  • the phospholipid comprises a natural phospholipid and a synthetic phospholipid.
  • the phospholipid comprises l-palmitoyl-2-oleoyl- sn-glycero-3-phosphocholine (POPC) .
  • POPC l-palmitoyl-2-oleoyl- sn-glycero-3-phosphocholine
  • thermoprotectant comprises trehalose.
  • the phospholipid comprises l-palmitoyl-2-oleoyl- sn-glycero-3-phosphocholine (POPC)
  • the thermoprotectant comprises trehalose
  • the buffer system comprises sodium phosphate monobasic monohydrate, sodium phosphate dibasic anhydrous, or the combination of sodium phosphate monobasic monohydrate and sodium phosphate dibasic anhydrous.
  • the posaconazole has a particle size distribution whose median value is between about 1.0 and about 8.0 microns, with not more than about 3000 particles of 10 microns or greater size and not
  • the posaconazole has a particle size distribution whose median value is between about 1.0 and about 5.0 microns, with
  • the posaconazole has a particle size distribution
  • the formulation has ingredients comprising: Ingredient Concentration range Posaconazole about 50 mg/ml POPC about 40 mg/ml Sodium Phosphate, 0.345 mg/ml monobasic, monohydrate, USP Sodium Phosphate, dibasic, 1.065 anhydrous, USP Trehalose 250 mg/ml Water for Injection, USP q.s. 1 ml ad
  • the formulation has ingredients comprising: Ingredient Concentration range
  • POPC about 10 to about 60 mg/ml Sodium Phosphate, about 0.01 to about 0.6 monobasic, monohydrate, mg/ml
  • the formulation has ingredients comprising: Ingredient Concentration range Posaconazole about 40 to about 60 mg/ml POPC about 20 to about 50 mg/ml Trehalose about 100 to about 250 mg/ml Water for Injection, USP q.s. about 1 ml ad
  • the formulation has ingredients comprising: Ingredient Concentration Posaconazole 50 mg/ml
  • the formulation has ingredients further comprising an antioxidant.
  • the antioxidant comprises propyl gallate at a concentration of about 0.02 to about 0.005 mg/ml.
  • the antioxidant comprises butylated hydroxytoluene at a concentration of about 0.1 to about 0.02 mg/ml.
  • the antioxidant comprises alpha-D-tocopherol at a concentration of about 0.5 to about 0.01 mg/ml.
  • the formulation has ingredients comprising: Ingredient Concentration Posaconazole 50 mg/ml
  • the formulation has ingredients comprising: Ingredient Concentration Posaconazole 50 mg/ml POPC 40 mg/ml Histidine 3 mg/ml Citric acid monohydrate 0.24 mg/ml Alpha-D-tocopherol 0.05 mg/ml Trehalose 250 mg/ml Water q.s. ad 1 ml at a pH of about 6.5.
  • the formulation has a wt. ratio of phospholipid to posaconazole between about 60: 1 and about 1: 10. In some embodiments, the formulation has a wt. ratio of phospholipid to posaconazole between about 1: 1 and about 1:5.
  • the formulation has a wt. ratio of phospholipid to posaconazole between about 1: 1 and about 4:5.
  • the formulation has a the wt. ratio of thermoprotectant to posaconazole between about 300: 1 and about 1: 10.
  • the formulation has a wt. ratio of thermoprotectant to posaconazole between about 1 : 1 and about 6: 1.
  • the formulation has a wt. ratio of thermoprotectant to phospholipid between about 30: 1 and about 1 :6.
  • the formulation has a wt. ratio of thermoprotectant to phospholipid between about 5:4 and about 30:4.
  • the invention encompasses a method of treating or preventing an infection inan animal in need thereof which comprises
  • the anirnal is a mammal, a bird, a fish, or a reptile.
  • the animal is a mammal, including but not limited to a human.
  • the infection is caused by a fungus or a parasite.
  • the infection is selected from the group consisting of: oropharyngeal or esophageal candidiasis; refractory oropharyngeal and esophageal candidiasis; invasive aspergillosis, candidiasis, fusariosis, scedosporiosis, infections due to dimorphic fungi , zygomycosis, and invasive infections due to rare molds and yeasts; invasive mycoses in patients who are refractory to, or intolerant of, other therapies;
  • the invention encompasses a method wherein said formulation is ad ⁇ rinistered intravenously.
  • the invention encompasses a method wherein said formulation is administered intramuscularly, subcutaneously, ophthalmically, subconjuctivally, intraocularly, via anterior eye chamber injection, intravitreally, intraperitoneally, intrathecally, intracystically, intrapleurally, intranasally, topically, via wound irrigation, intradermally, intrabuccally, intra-abdominally, intra-articularly, intra-aurally, intrabronchially, intracapsularly, intra ⁇ ieningeally, intrapulrnonarilly, via inhalation, via endotracheal or endobronchial installation, via direct installation into pulmonary cavities, mtraspinally, intrasynovially, intrathoracically, via thoracostomy irrigation, vaginally, epidurally, rectally, intracisternally, intravascularly.intraventricularly, intraosseously, via irrigation of infected bone, or via application as part of any admixture with cement for pros
  • the formulation further comprises a second active
  • ingredient selected from one or more of the group consisting of:
  • antifungals such as azoles; amphotericin B; deoxycholate amphotericin
  • the invention encompasses a method further comprising administering a second active ingredient selected from one or
  • antifungals such as azoles
  • amphotericin a group consisting of: antifungals such as azoles; amphotericin
  • deoxycholate amphotericin B flucytosine; terbinafine; antibacterials; antivirals; steroids; nonsteroidal drugs ("NSAIDs”); chemotherapeutics; and, anti-emitics.
  • the formulation is further characterized by providing a mean maximum plasma concentration (Cmax) of posaconazole of at least about 467 ng/ml at steady state, and a mean plasma Area Under the Curve over 24 hours (AUC) value of posaconazole of at least
  • the formulation is further characterized by providing a mean maximum plasma concentration (Cmax) of posaconazole of at least about 852 ng/ml at steady state, and a mean plasma Area
  • the formulation is further characterized by
  • posaconazole at least one of: a mean plasma half-life in a range of about
  • the formulation is further characterized by providing a mean maximum plasma concentration (Cmax) of posaconazole of at least about 1480 ng/ml at steady state, and a mean plasma Area Under the Curve over 24 hours (AUC) value of posaconazole of at least
  • the formulation is further characterized as providing, after administration of a dosage of about 200 mg of said posaconazole, at least one of: a mean plasma half-life of about 18.7 to
  • the formulation is further characterized as
  • posaconazole at least one of: a mean plasma half-life of about 18.5 to about 51.4 hours; and a mean plasma steady state volume of distribution of about 200-500 L.
  • the formulation is further characterized as providing, after adriiinistration of a dosage of about 600 mg of said posaconazole, at least one of: a mean plasma half-life of about 27.2 to about 50.6 hours; and a mean plasma steady state volume of distribution of about 200-500 L.
  • the formulation is further characterized as providing a mean posaconazole blood concentration profile substantially similar to that of Figure 1 , when said formulation is infused over about 1 hour to deliver 25-600 mg of posaconazole.
  • the formulation is further characterized as providing a mean posaconazole plasma concentration profile substantially similar to that of Figure 2, when said formulation is infused over about 1 hour to deliver 25-600 mg of posaconazole.
  • the formulation is further characterized as
  • the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole
  • the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.9 and about 3.8, when a single dose of said formulation is infused over about 1 hour to deliver 50 mg of posaconazole.
  • the formulation is further characterized as providing a mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 2.2 and about 3.3, when a single dose of said formulation is infused over about 1 hour to deliver 100 mg of posaconazole.
  • the formulation is further characterized as
  • the formulation is further characterized as
  • said formulation is infused over about 1 hour to deliver 400 mg of posaconazole.
  • the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.9 and about 3.1, when a single dose of said formulation is infused over about 1 hour to deliver 600 mg of posaconazole.
  • the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cm x of between about 1.2 and about 2.5, at steady state when said formulation is infused over about 1 hour to deliver 25-600 mg of posaconazole, and repeated on a 24-hour basis.
  • the formulation is further characterized as
  • the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole
  • plasma Cmax of between about 1.5 and about 2.4 at steady state when said formulation is infused over about 1 hour to deliver 50 mg of posaconazole, and repeated on a 24-hour basis.
  • the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.7 and about 2.5, at steady state when said formulation is infused over about 1 hour to deliver 100 mg of posaconazole, and repeated on a 24-hour basis.
  • the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.2 and about 2.0, at steady state when said formulation is infused over about 1 hour to deliver 200 mg of posaconazole, and repeated on a 24-hour basis.
  • the formulation is further characterized as
  • the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole
  • plasma Cmax of between about 1.3 and about 1.7 at steady state when said formulation is infused over about 1 hour to deliver 600 mg of posaconazole, and repeated on a 24-hour basis.
  • the water in the formulation has been removed by lyophilization.
  • the anirnal treated is human, while in other embodiments the animal treated is non-human.
  • the formulation is one that is bioequivalent to a formulation disclosed herein.
  • the method further comprises ad ⁇ iimstering a bolus loading dose of said formulation and then admimstering an intravenous maintenance dose of said formulation.
  • the method comprises admimstering to said animal an effective amount of posaconazole to provide a mean maximum
  • Cmax plasma concentration of posaconazole of at least about 467 ng/ml
  • FIG. 1 shows posaconazole mean blood concentration-time profiles in healthy volunteers after 1 hr intravenous infusions of 25, 50, 100, 200,
  • FIG. 2 shows posaconazole mean plasma concentration-time profiles in healthy volunteers after 1 hr intravenous infusions of 25, 50, 100, 200,
  • FIG. 3 shows posaconazole mean plasma and blood concentration-time profiles in healthy volunteers after 1 hr intravenous infusion of 25 mg posaconazole.
  • FIG. 4 shows posaconazole mean plasma and blood concentration- time profiles in healthy volunteers after 1 hr intravenous infusion of 600 mg posaconazole.
  • the present invention encompasses formulations suitable for parenteral
  • adrrrinistration e.g., by injection, for treating an infection.
  • formulations comprise a suspension of posaconazole, stabilized by a phospholipid, in a mixture comprising water, a thermoprotectant, and a l
  • posaconazole Since posaconazole is minimally soluble in water, a suspension formulation is advantageous. Phospholipids have been found to be effective surfactants in forming stable suspensions of posaconazole
  • thermoprotectant is used to prevent agglomeration and crystal growth of the posaconazole particles during autoclaving.
  • Parenteral buffer systems are typically designed to be at physiological pH of about 7.4.
  • Phospholipids are known to be stable at a pH range of about 6 to about 7.
  • pH adjustment of injectable formulations can be necessary to achieve physiological compatibility, and thus, for example, to minimize injection-site irritation.
  • the rate of phospholipid hydrolysis can be temperature-sensitive.
  • the buffer systems are designed to meet physiological pH requirements, and to maintain the temperature/pH-
  • buffer systems were found to control degradation of POPC-contarning posaconazole formulations during autoclaving. For example, such formulations were found to be stable after 20 minutes of autoclaving at 121°C. In addition, these buffer systems stabilize such formulations during storage at 4°C to 25°C for at least 18 months following autoclaving.
  • other phospholipids that are similar to POPC could be used to stabilize the formulations disclosed herein.
  • unsaturated phospholipids with an acyl chain length ranging from C 1 2 to C20 wherein the degree of unsaturation of the acyl chain ranges from 1 to 4; as weE as saturated phospholipids with an acyl chain length ranging from C12 to Ci8 are useful according to the present invention.
  • useful unsaturated phospholipids include: 1 -palrmtoyl-2-oleoyl-sn-glycero-3-phosphocholine ("POPC") ,
  • DOPC 1,2-Dioleoyl-sn-Glycero-3-Phosphocholine
  • DOPE 1,2-Dioleoyl-sn-Glycero-3-Phosphoethanolamine
  • saturated phospholipids include:
  • DMPC 1,2-Di ⁇ yristoyl-sn-Glycero-3-Phosphocholine
  • the antioxidant comprises propyl gallate, preferably at a concentration of about 0.02 to about 0.005 mg/ml.
  • the antioxidant comprises butylated hydroxytoluene, preferably at a concentration of about 0.1 to about 0.02 mg/ml.
  • the antioxidant comprises propyl gallate, preferably at a concentration of about 0.02 to about 0.005 mg/ml, in combination with butylated hydroxytoluene, preferably at a concentration of about 0.1 to about 0.02 mg/ml.
  • the antioxidant comprises alpha-D-tocopherol, preferably at a concentration of about 0.5 to about 0.01 mg/ml.
  • phospholipid to posaconazole is preferably between about 1:0.1 and about
  • thermoprotectant to posaconazole is preferably between about 0.5: 1 and about 6: 1, more preferably, between about 2: 1 and about 6: 1.
  • thermoprotectant to phospholipid is preferably, between about
  • the formulations of the present invention comprise a suspension of solid particles of posaconazole of specific particle size distribution in an aqueous phase.
  • the particle size distribution displayed in the suspended particles is critical for physiological compatibility, syringeability, physical stability of the suspension, re-suspendability, and for pharmacokinetic characteristics and bio- distribution [le., sequestration within specific bodily tissues). Since these characteristics are critical to the formulation as delivered to the patient, it is important that processes that contribute to changes in particle size distribution after rnicronization are controlled.
  • Such processes can include agglomeration during autoclaving, and de- suspension due to temperature excursions and/or agitation experienced during shipping and storage. It is the particle size distribution in the
  • the inventors of the present invention have deterrnined that for injectable formulations of posaconazole, these characteristics are brought within
  • the particle size distributions display not more
  • pH adjustment system components that function in this way include
  • the present invention encompasses methods of prevention and treatment
  • infection is understood to include, but not be limited to, those having the following properties: characteristics: characteristics: characteristics: characteristics: characteristics: characteristics: characteristics: characteristics; and
  • Schizophyllum Crytococcus, Histoplasma, Blastomyces, Coccidioides, Fusarium, Exophiala, Zygomycocetes ⁇ e.g., Mucor, Rhizopus, and Rhizomucor), Kluyveromyces, Saccharomyces, Yarrowia, Pichia, Epidermophyton, Paracoccidioides, Scedosporium, Apophysomyces, Curvularia, Penicilliurn, Fonsecaea, Wangiella, Sporothrix, Pneumocystis, Trichosporon, Absidia, Cladophialophora, Rarnichloridiurn, Syncephalastrum, Madurella, Scytalidium, Leshmania, protozoa, bacteria, gram negatives, gram positives, anaerobes, including Legionella Borrelia, Mycoplasma, Treponema, Gardneralla, Trichomononas and Trypan
  • the present invention is intended to treat both opportunistic and non- opportunistic infections, where the term "opportunistic" as used herein denotes those infections caused by organisms capable of causing a disease only in a host whose resistance is lowered, e.g., by chemotherapy or H.I.V.
  • posaconazole is useful in the prevention and/or treatment
  • Initial (first line) treatment of oropharyngeal or esophageal candidiasis Salvage therapy of azole-refractory oropharyngeal and esophageal candidiasis (e.g., in patients who have failed oral fluconazole and/ or itraconazole);
  • Salvage therapy for invasive mycoses in patients who are refractory to or intolerant of other therapies e.g., amphotericin B, lipid formulations of amphotericin B, caspofungin, voriconazole and/or itraconazole;
  • Chagas disease (Trypanosorniasis due to T. cruzi) including acute and
  • Irr ⁇ nuno-suppressant therapy ⁇ e.g., chemotherapy, radiation therapy,
  • the present invention encompasses the administration of a posaconazole formulation adjunctive to imrnuno- suppressant therapy, wherein the posaconazole formulation functions prophylactically with regard to opportunistic infections including the
  • the present invention encompasses a variety of modes of administration to any part, organ, interstice or cavity ofan animal's body that is subject to an infection.
  • a non-lir ting set of examples of modes by which the posaconasole formulations of the present invention may be adrninistered includes: intravenously, intramuscularly, subcutaneously, ophthalmically, subconjuctivally, intraocularly, via anterior eye chamber injection, intravitreally, intraperitoneally, intrathecally, intracystically, intrapleurally, intranasally, topically, via wound irrigation, intradermally, intrabuccally, intra-abdominally, intra-articularly, intra-aurally,
  • inhalation via endotracheal or endobronchial installation, via direct installation into pulmonary cavities, intraspinally, rntrasynovially, intrathoracically, via thoracostomy irrigation, vaginally, epidurally, rectally, intracisternally, intravascularly.intraventricularly, intraosseously, via irrigation of infected bone, and via application as part
  • Co-formulations comprising combinations of posaconazole and at least one other active ingredient are also within the scope of the present invention.
  • active ingredients include: antifungals such as echinocandins (including caspofungin, ⁇ iicafungin, and anidulafungin) and azoles (including voriconazole, itraconazole, fluconazole, ketoconazole, ravuconazole); amphotericin B; deoxycholate amphotericin B; flucytosine; and terbinafine.
  • NSAIDs nonsteroidal anti-inflammatory drugs
  • chemotherapeutics chemotherapeutics
  • anti-emitics co-administradministered with at least one of the above active ingredients, aside from within a single formulation, is also within the scope of the present invention.
  • regimens each consisting of a frequency of dosing and a duration of administration.
  • Preferred frequencies of dosing include once every 12, 24,
  • bolus dosing at various rates and various doses, and combinations of a bolus loading dose, or several bolus loading doses, with an intravenous infusion
  • phospholipid refers to a lipid compound that yields on hydrolysis phosphoric acid, an alcohol, fatty acid and a nitrogenous base.
  • examples include natural and synthetic phoshpholipids, which include lecithin, cephalin, sphingornyelin and 1- pahnitoyl-2-oleoyl-sn-glycero-3-phosphocholine (“POPC”) .
  • POPC pahnitoyl-2-oleoyl-sn-glycero-3-phosphocholine
  • natural phospholipid refers to a phospholipid occurring in nature, or derived from a natural source.
  • Non- lirniting examples of natural phospholipids include egg phospholipids, soy
  • synthetic phospholipid refers to a man-made
  • Non-limiting examples of synthetic phospholipids include
  • POPC l-paln ⁇ itoyl-2-oleoyl-sn-glycero-3-phosphocholine
  • DOPC 1,2-oleoyl-sn- glycero-3-phosphocholine
  • DLPC DLPC
  • DLPC DLPC
  • DMPC l,2-Dipalmitoyl-sn-Glycero-3-Phosphocholine
  • DSPC 1,2- Stearoyl-sn-Glycero-3-Phosphocholine
  • buffer system refers to a buffer comprising one or more components that maintains a particular pH range.
  • suitable buffer systems include: phosphoric acid; glycine; sodium citrate; histidine; citric acid; acetic acid; tromethamine; ammonium sulfate; and combinations thereof.
  • the aforementioned components are understood to include the salts, hydrates and solvates thereof.
  • phosphoric acid includes the sodium phosphate or potassium phosphate salts, among other salts.
  • Preferred buffer systems include sodium phosphate monobasic, sodium phosphate dibasic, or a combination thereof. More preferred buffer systems include sodium phosphate monobasic monohydrate, sodium phosphate dibasic
  • organic buffer refers to a buffer comprising at least one organic compound.
  • suitable organic buffers include: glycine;. sodium citrate; histidine; citric acid; acetic acid; and combinations thereof.
  • antioxidant refers to an agent that hinders
  • antioxidants include propyl gallate, butylated hydroxytoluene, and alpha-D-tocopherol.
  • the phrase "median particle size” refers to the particle size present in the volume-weighted 50 th percentile, as ascertained by Malvern®, Sympatec®, or Horibe® laser diffraction particle size analysis. Particle sizes are measured throughout, and at the term ation of, the shelf life, typically up to 24 months after manufacture, when held at either refrigerated or room temperatures. Particle sizes are also measured and maintained when the formulation is diluted into large volume parenterals, e.g., 5% dextrose or water for injection.
  • the phrase "initial median particle size” refers to the particle size present within 1 week after a specified timepornt.
  • the initial median particle size after autoclaving refers to the median particle size present within 1 week after autoclaving has been
  • autoclaving refers to sterilization by the
  • terminal steam sterilization method For example, autoclaving for 20
  • thermoprotectant refers to an agent that
  • thermoprotectant is used to preserve the
  • Thermoprotectants are typically water soluble polyhydroxyl compounds.
  • trehalose is a thermoprotectant agent that may be used in conjunction with posaconazole.
  • Others include maltose, sorbitol, dextrose, sucrose, lactose and mannitol.
  • prodrug refers to a compound that is a drug precursor which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield posaconazole or a salt and/or solvate thereof.
  • solvate refers to a physical association between a compound with one or more solvent molecules. This physical association involves varying degrees of ionic and/or covalent bonding,
  • solvate encompasses both solution-phase and isolatable solvates.
  • suitable solvates include hydrates, ethanolates, and
  • injectable means adapted to parenteral ad ⁇ iinistration.
  • fungus means one of the diverse morphologic forms of yeasts and molds.
  • Fungi include Candida, dermatophytes, Dirnorphics, Dematiaceous ⁇ e.g., Alternaria and Bipolaris), Aspergillus, Acremonium, Basidiomycetes, Bjerkandera, Coprinus, Paecilomyces, Microsporum, Trichophyton, Pseudallescheria, Schizophyllurn, Crytococcus, Histoplasma, Blastomyces, Coccidioides, Fusarium, Exophiala, Zygomycocetes ⁇ e.g., Mucor, Rhizopus, and Rhizomucor), rvluyveromyces, Saccharomyces, Yarrowia, Pichia, Epidermophyton, Paracoccidioides, Scedosporium, Apophysomyces, Curvularia, Periicillium, Fonsecae
  • hyphae and includes as non-limiting examples Alternaria and Bipolaris.
  • Zygomycocete means a class of fungi characterized by sexual reproduction resulting in the formation of
  • zygospore and asexual reproduction by means of nonmotile spores called sporangiospores or conidia, and includes as non-liiniting examples Mucor, Rhizopus, and Rhizomucor.
  • anaerobe means a rnicroorganisrn that can live and grow in the absence of oxygen, and includes as non-limiting examples Legionella Borrelia, Mycoplasma, Treponema, Gardneralla, and Trichomononas .
  • parasite means an organism that lives on or in another and draws its nourishment therefrom. Parasites include Leshmania and Trypansoma, among others.
  • antifungal means an agent having activity against one or more fungi, and includes echinocandins such as caspofungin, micafungin, and anidulafungin.
  • azole means divtayleriimine, and includes voriconazole, itraconazole, fluconazole, ketoconazole, ravuconazole:
  • anirnal is understood to include humans, non- human mammals, fish, birds and reptiles.
  • bioequivalent is understood as having that meaning assigned to the term by the U.S. Food & Drug Administration. "Bioequivalence means the absence of a significant difference in the rate and extent to which the active ingredient or active moiety in pharmaceutical equivalents or pharmaceutical alternatives becomes available at the site of drug action when adrninistered at the same molar dose under similar conditions in an appropriately designed study.” 21 CFR 320.1(e). Methodologies for determiriing bioequivalence are given in "Guidance for Industry: Statistical Approaches to Establishing Bioequivalence," U.S. Department of Health and Human Services, Food and Drug Adrmnistration, Center for Drug Evaluation and Research
  • exemplary posaconazole formulations that include antioxidant are described in Examples 4-6.
  • the pH is 6.4 in Example 5.
  • the pH is 6.5 in Example 6.
  • Example 7 is a preferred embodiment of the present invention. Exam le 7
  • Example 7 The pH is 7.2 in Example 7.
  • the following is an exemplary placebo formulation wherein the pH is 6.4.
  • POS IV posaconazole intravenous drug product formulation
  • POS IV 50 mg/mL was diluted in 5% dextrose in water (D5W) in IV bags.
  • Subjects assigned to active drug received in a 100-mL volume one of the following single doses administered intravenously over 1 hour: Group 1 ,
  • Group 6 a 125-mL volume a single dose of 600 mg administered intravenously over 1 hour and 15 minutes.
  • EDTA ethylenediaminetetraacetate salt
  • posaconazole in plasma, the tube of blood (4 mL to 5 mL) was centrifuged within approximately 15 minutes of collection at approximately 4°C and IS Og for 10 minutes to completely separate red blood cells from plasma. All blood and plasma samples were immediately frozen to at least -20°C and maintained in the frozen state until assayed.
  • the blood and plasma concentrations of posaconazole were determined using validated high performance liquid chromatographic-mass spectrometric (LC-MS/MS) assays.
  • the lower limit of quantitation (LLOQ) of this assay was 5.0 ng/mL and the calbration range was 5 to 5000 ng/mL.
  • the terminal phase rate constant (k) was calculated as the negative of the slope of the log-linear terminal portion of the serum concentration-time curve using linear regression.
  • the terminal phase half-life, t ⁇ / 2 was calculated as 0.693/k.
  • AUC(tf) The area under the serum concentration-time curve from time 0 to the time of final quantifiable sample [AUC(tf)] was calculated using the linear trapezoidal rule. AUC(tf) was then extrapolated to irifinity (I) as follows:
  • AUC(I) AUC(tf) + Ces(tf)/k where Ces(tf) is the estimated concentration determined from linear regression at final measurable sampling time, tf.
  • Vdss CL x MRT where MRT is the mean residence time (adjusted for infusion duration)
  • posaconazole plasma concentrations declined unusually rapidly, and then, surprisingly, increased subsequently, followed by a slow declining terminal phase (see Figures 1-4).
  • This pharmacokinetic profile is believed to be atypical and unique among known azoles.
  • this pharmacokinetic pattern was also observed after the intravenous adrrunistration of posaconazole in animals. It is indicative of a rapid distribution of posaconazole to the liver and spleen and subsequent slow release from these tissues. Therefore, as noted in the literature with respect to another pharmaceutically active agent (Townsend RW, Zutshi A, Bekersky I., "Biodistribution of 4-
  • POS IV may be
  • RES reticuloendothelial system
  • Table 14 displays pharmacokinetic data resulting from such oral ad ⁇ iinistration, arranged by quartile based on the observed range of posaconazole plasma concentration values. For each quartile, the response rate for apergillosis is displayed.
  • the table shows that the target mean Cmax for a response rate of at least 50% should be in the range of 467 to 1480 ng/mL, or higher.
  • the pharmacokinetic modeling and steady-state projection based on the pharmacokinetic results of POS IV once-a-day (QD) dosing regimen show that the projected posaconazole mean Cmax at a 100 mg POS IV QD dose
  • posaconazole was slowly eliminated from plasma with an average terminal half-life of 21 to 39 hours.
  • the half-life was higher at the higher dose compared to that at lower dose groups (see Tables 15 and 16), in a range of about 15 hours (with a lOOmg dose) to about 51 hours (with a 400mg dose).
  • a long half- life is desirable as it provides the sustained and high plasma
  • Vdss extensive tissue distribution and penetration into the tissues, a characteristic that likely contributes to enhanced anti-infective activity.
  • the range in the data for Vdss was from 219 to 516L. This is consistent
  • volume distribution could have a range of 200 to 500 L.
  • the preferable ratios of blood to plasma posaconazole Cmax and AUC values are shown in Tables 18 and 19.
  • Overall posaconazole exposure (AUC) was higher in plasma compared to that in blood ⁇ see Tables 18 and 19 - AUC ratio).
  • the posaconazole concentrations were greater in blood than in plasma during the infusion and approximately up to 1 hr post-infusion (see Figures 3 and 4; Tables 18 and 19, Cmax ratio).
  • the coefficient of variation of the data suggests that the ratio of blood to plasma posaconazole Cmax could have a range of 1.8 to 3.5 for single dose infused over 1 hour to deliver 25-600 mg of posaconazole.
  • the coefficient of variation of the data suggests that
  • the ratio of blood to plasma posaconazole Cmax could have a range of 1.0 to 2.3 at steady state when posaconazole is infused over about 1 hour

Abstract

The present invention provides formulations useful for treating infections, in particular, formulations that include the active pharmaceutical ingredient posaconazole in an injectable suspension that is stable when subjected to terminal steam sterilization.

Description

INJECTABLE PHARMACEUTICAL SUSPENSION COMPRISING POSACONAZOLE
FIELD OF THE INVENTION
The present invention relates to formulations useful for treating infections. Specifically, these formulations include the active pharmaceutical ingredient posaconazole in an injectable suspension that is stable when subjected to terminal steam sterilization, and throughout
the shelf life of the product.
BACKGROUND OF THE INVENTION
Posaconazole, an anti-fungal agent, represented by the following chemical
structural formula
Figure imgf000003_0001
is being developed as an oral suspension (40 mg/ml) under the trademark NOXAFIL® by Schering Corporation, Kenilworth, NJ. See, for example, U.S. Patent No. 5,703,079, 5,661,151, WO 02/80678 published October 17, 2002, and EP 1 372 394 published January 2, 2004. In addition,
other formulations of posaconazole have been disclosed. A solid (capsule/tablet) of posaconazole is disclosed in U.S. Patent Nos. 5,972,381 and 5,834,472. Lastly, a topical form of posaconazole, e.g., a lotion, cream, ointment, or "lacquer nail polish" is contemplated based on other similar formulations, e.g., U.S. Patent No. 4,957,730 (PENLAC® available from Demiik®).
Certain aspects of stabilization of micronized particles in pharmaceutical compositions are addressed in the literature. For example, U.S. Patent No. 5,858,410 discloses pharmaceutical compositions containing particles of active agents of average diameter less than 5 microns, having been conrminuted, without prior conversion into a melt, by using a piston-gap homogenizer. U.S. Patent Application No. 10/440,368 discloses the use of a phospholipid surface active agent to stabilize rnicroparticles of solid fenofibrate in an orally adπrjuαistered pharmaceutical composition. U.S.
Patent No. 5,091, 188 discloses the use of phospholipids, to prevent coalescence of i crocrystalline active agents in injectable pharmaceutical
compositions. Examples of disclosed phospholipids include lecithin,
phosphatidic acid, phosphatidyl ethanolamine, cholesterol, steaiylamine, glycolipids and mono-glycerides.
None of the aforementioned references however, discloses an injectable suspension of posaconazole, that is stable when subjected to terminal steam sterilization and throughout the shelf life of the product. There is a need for such a formulation as it is desirable to ensure the physical
stability of the sterilized end product.
SUMMARY OF THE INVENTION
The present invention provides formulations of posaconazole that are stable when subjected to terminal steam sterilization. These formulations are useful for the treatment of infections. In particular, an aqueous injectable suspension of posaconazole that is homogenously suspended in vehicle with the aid of a phospholipid. In addition a thermoprotectant agent is employed to reduce autoclave-induced particle size growth, as well as a buffer system to stabilize the phospholipid during autoclaving. The formulations provided remain stable after 20 minutes of autoclaving at 121°C and after subsequent storage at 4°C to 40°C for at least 6 months.
The present invention provides formulations comprising a suspension of posaconazole, stabilized by a phospholipid, in a mixture comprising a thermoprotectant, and a buffer system.
In some embodiments, the formulation has been sterilized by autoclaving or by irradiation. In some embodiments, the buffer system comprises sodium phosphate, which may be provided as sodium phosphate monobasic monohydrate,
sodium phosphate dibasic anhydrous, or the combination of the two.
In some embodiments, the buffer system comprises an organic buffer.
In some embodiments, the buffer system comprises at least one of histidine, citric acid, glycine, sodium citrate, arnrnoniuπi sulfate, or acetic acid.
In some embodiments, the buffer system maintains a pH of about 3.0 to about 9.0.
In some embodiments, the buffer system mamtains a pH of about 6.0 to about 8.0.
In some embodiments, the buffer system mamtains a pH of about 6.4 to
about 7.6.
In some embodiments, the phospholipid comprises a natural
phospholipid.
In some embodiments, the phospholipid comprises a synthetic
phospholipid. In some embodiments, the phospholipid comprises a natural phospholipid and a synthetic phospholipid.
In some embodiments, the phospholipid comprises l-palmitoyl-2-oleoyl- sn-glycero-3-phosphocholine (POPC) .
In some embodiments, the thermoprotectant comprises trehalose.
In some embodiments, the phospholipid comprises l-palmitoyl-2-oleoyl- sn-glycero-3-phosphocholine (POPC), the thermoprotectant comprises trehalose, and the buffer system comprises sodium phosphate monobasic monohydrate, sodium phosphate dibasic anhydrous, or the combination of sodium phosphate monobasic monohydrate and sodium phosphate dibasic anhydrous.
In some embodiments, the posaconazole has a particle size distribution whose median value is between about 1.0 and about 8.0 microns, with not more than about 3000 particles of 10 microns or greater size and not
more than about 300 particles of 25 microns or greater size.
In some embodiments, the posaconazole has a particle size distribution whose median value is between about 1.0 and about 5.0 microns, with
not more than about 3000 particles of 10 microns or greater size and not more than about 300 particles of 25 microns or greater size. In some embodiments, the posaconazole has a particle size distribution
whose median value is between about 1.2 and about 4.5 microns, with
not more than about 3000 particles of 10 microns or greater size and not
more than about 300 particles of 25 microns or greater size.
In some embodiments, the formulation has ingredients comprising: Ingredient Concentration range Posaconazole about 50 mg/ml POPC about 40 mg/ml Sodium Phosphate, 0.345 mg/ml monobasic, monohydrate, USP Sodium Phosphate, dibasic, 1.065 anhydrous, USP Trehalose 250 mg/ml Water for Injection, USP q.s. 1 ml ad
In some embodiments, the formulation has ingredients comprising: Ingredient Concentration range
Posaconazole about 1 to about 100 mg/ml
POPC about 10 to about 60 mg/ml Sodium Phosphate, about 0.01 to about 0.6 monobasic, monohydrate, mg/ml
USP Sodium Phosphate, dibasic, about 0.04 to about 1.5 anhydrous, USP mg/ml Trehalose about 10 to about 300 rng/ml Water for Injection, USP q.s. about 1 ml ad
In some embodiments, the formulation has ingredients comprising: Ingredient Concentration range Posaconazole about 40 to about 60 mg/ml POPC about 20 to about 50 mg/ml Trehalose about 100 to about 250 mg/ml Water for Injection, USP q.s. about 1 ml ad
In some embodiments, the formulation has ingredients comprising: Ingredient Concentration Posaconazole 50 mg/ml
POPC 40 mg/ml Histidine 3 mg/ml Citric acid monohydrate 0.24 rng/ml Trehalose 250 mg/ml Water q.s. ad 1 ml at a pH of about 6.4.
In some embodiments, the formulation has ingredients further comprising an antioxidant.
In some embodiments, the antioxidant comprises propyl gallate at a concentration of about 0.02 to about 0.005 mg/ml.
In some embodiments, the antioxidant comprises butylated hydroxytoluene at a concentration of about 0.1 to about 0.02 mg/ml.
In some embodiments, the antioxidant comprises alpha-D-tocopherol at a concentration of about 0.5 to about 0.01 mg/ml.
In some embodiments, the formulation has ingredients comprising: Ingredient Concentration Posaconazole 50 mg/ml
POPC 40 mg/ml
Histidine 3 mg/ml Citric acid monohydrate 0.24 mg/ml Propyl gallate 0.01 mg/ml But lated hydroxytoluene 0.05 mg/ml Trehalose 250 mg/ml Water q.s. ad 1 ml at a pH of about 6.4.
In some embodiments, the formulation has ingredients comprising: Ingredient Concentration Posaconazole 50 mg/ml POPC 40 mg/ml Histidine 3 mg/ml Citric acid monohydrate 0.24 mg/ml Alpha-D-tocopherol 0.05 mg/ml Trehalose 250 mg/ml Water q.s. ad 1 ml at a pH of about 6.5.
In some embodiments, the formulation has a wt. ratio of phospholipid to posaconazole between about 60: 1 and about 1: 10. In some embodiments, the formulation has a wt. ratio of phospholipid to posaconazole between about 1: 1 and about 1:5.
In some embodiments, the formulation has a wt. ratio of phospholipid to posaconazole between about 1: 1 and about 4:5.
In some embodiments, the formulation has a the wt. ratio of thermoprotectant to posaconazole between about 300: 1 and about 1: 10.
In some embodiments, the formulation has a wt. ratio of thermoprotectant to posaconazole between about 1 : 1 and about 6: 1.
In some embodiments, the formulation has a wt. ratio of thermoprotectant to phospholipid between about 30: 1 and about 1 :6.
In some embodiments, the formulation has a wt. ratio of thermoprotectant to phospholipid between about 5:4 and about 30:4.
In some embodiments, the invention encompasses a method of treating or preventing an infection inan animal in need thereof which comprises
administering to said animal an effective amount of the formulation. In
some embodiments, the anirnal is a mammal, a bird, a fish, or a reptile. In some embodiments, the animal is a mammal, including but not limited to a human.
In some embodiments, the infection is caused by a fungus or a parasite.
In some embodiments, the infection is selected from the group consisting of: oropharyngeal or esophageal candidiasis; refractory oropharyngeal and esophageal candidiasis; invasive aspergillosis, candidiasis, fusariosis, scedosporiosis, infections due to dimorphic fungi , zygomycosis, and invasive infections due to rare molds and yeasts; invasive mycoses in patients who are refractory to, or intolerant of, other therapies;
Candidiasis, invasive mould infections in patients who have undergone intensive chemotherapy and/or radiation therapy for hematologic malignancies, bone marrow or peripheral stem cell transplant conditioning regimens, and patients receiving combination irnrnunosuppressive therapy for the treatment of acute or chronic graft-
versus-host disease or prevention of solid organ transplantation;
Chagas disease; and,
Leishmaniasis . In some embodiments, the invention encompasses a method wherein said formulation is adπrinistered intravenously.
In some embodiments, the invention encompasses a method wherein said formulation is administered intramuscularly, subcutaneously, ophthalmically, subconjuctivally, intraocularly, via anterior eye chamber injection, intravitreally, intraperitoneally, intrathecally, intracystically, intrapleurally, intranasally, topically, via wound irrigation, intradermally, intrabuccally, intra-abdominally, intra-articularly, intra-aurally, intrabronchially, intracapsularly, intraπieningeally, intrapulrnonarilly, via inhalation, via endotracheal or endobronchial installation, via direct installation into pulmonary cavities, mtraspinally, intrasynovially, intrathoracically, via thoracostomy irrigation, vaginally, epidurally, rectally, intracisternally, intravascularly.intraventricularly, intraosseously, via irrigation of infected bone, or via application as part of any admixture with cement for prosthetic devices.
In some embodiments, the formulation further comprises a second active
ingredient selected from one or more of the group consisting of:
antifungals such as azoles; amphotericin B; deoxycholate amphotericin
B; flucytosine; terbinafine; antibacterials; antivirals; steroids;
nonsteroidal
Figure imgf000014_0001
drugs ("NSAIDs"); chemotherapeutics; and anti-emitics. In some embodiments, the invention encompasses a method further comprising administering a second active ingredient selected from one or
more of the group consisting of: antifungals such as azoles; amphotericin
B; deoxycholate amphotericin B; flucytosine; terbinafine; antibacterials; antivirals; steroids; nonsteroidal
Figure imgf000015_0001
drugs ("NSAIDs"); chemotherapeutics; and, anti-emitics.
In some embodiments, the formulation is further characterized by providing a mean maximum plasma concentration (Cmax) of posaconazole of at least about 467 ng/ml at steady state, and a mean plasma Area Under the Curve over 24 hours (AUC) value of posaconazole of at least
about 9840 ng hr/ml at steady state, when said formulation is infused
over about 1 hour to deliver 100 mg of posaconazole, and repeated at an interval of about 24 hours.
In some embodiments, the formulation is further characterized by providing a mean maximum plasma concentration (Cmax) of posaconazole of at least about 852 ng/ml at steady state, and a mean plasma Area
Under the Curve over 24 hours (AUC) value of posaconazole of at least
about 24,600 ng hr/ml at steady state, when said formulation is infused
over about 1 hour to deliver 200 mg of posaconazole, and repeated at an interval of about 24 hours. In some embodiments, the formulation is further characterized by
providing, after administration of a dosage of about 100 mg of said
posaconazole, at least one of: a mean plasma half-life in a range of about
14.9 to about 38.4 hours; and a mean plasma steady state volume of distribution of about 200-500 L.
In some embodiments, the formulation is further characterized by providing a mean maximum plasma concentration (Cmax) of posaconazole of at least about 1480 ng/ml at steady state, and a mean plasma Area Under the Curve over 24 hours (AUC) value of posaconazole of at least
about 24,600 ng-hr/ml at steady state, when said formulation is infused
over about 1 hour to deliver at least 200 mg of posaconazole, and repeated at an interval of about 24 hours.
In some embodiments, the formulation is further characterized as providing, after administration of a dosage of about 200 mg of said posaconazole, at least one of: a mean plasma half-life of about 18.7 to
about 35.5 hours; and a mean plasma steady state volume of distribution of about 200-500 L.
In some embodiments, the formulation is further characterized as
providing, after adrrunistration of a dosage of about 400 mg of said
posaconazole, at least one of: a mean plasma half-life of about 18.5 to about 51.4 hours; and a mean plasma steady state volume of distribution of about 200-500 L.
In some embodiments, the formulation is further characterized as providing, after adriiinistration of a dosage of about 600 mg of said posaconazole, at least one of: a mean plasma half-life of about 27.2 to about 50.6 hours; and a mean plasma steady state volume of distribution of about 200-500 L.
In some embodiments, the formulation is further characterized as providing a mean posaconazole blood concentration profile substantially similar to that of Figure 1 , when said formulation is infused over about 1 hour to deliver 25-600 mg of posaconazole.
In some embodiments, the formulation is further characterized as providing a mean posaconazole plasma concentration profile substantially similar to that of Figure 2, when said formulation is infused over about 1 hour to deliver 25-600 mg of posaconazole.
In some embodiments, the formulation is further characterized as
providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.5 and about 3.8, when a single dose of said formulation is infused over about 1 hour to deliver 25-600 mg of
posaconazole. In some embodiments, the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole
plasma Cmax of between about 2.1 and about 3.3, when a single dose of said formulation is infused over about 1 hour to deliver 25 mg of posaconazole.
In some embodiments, the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.9 and about 3.8, when a single dose of said formulation is infused over about 1 hour to deliver 50 mg of posaconazole.
In some embodiments, the formulation is further characterized as providing a mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 2.2 and about 3.3, when a single dose of said formulation is infused over about 1 hour to deliver 100 mg of posaconazole.
In some embodiments, the formulation is further characterized as
providing a ratio of mean posaconazole blood Cmax to mean posaconazole
plasma Cma of between about 1.5 and about 3.2, when a single dose of said formulation is infused over about 1 hour to deliver 200 rng of posaconazole. In some embodiments, the formulation is further characterized as
providing a ratio of mean posaconazole blood Cmax to mean posaconazole
plasma Cmax of between about 1.7 and about 3.3, when a single dose of
said formulation is infused over about 1 hour to deliver 400 mg of posaconazole.
In some embodiments, the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.9 and about 3.1, when a single dose of said formulation is infused over about 1 hour to deliver 600 mg of posaconazole.
In some embodiments, the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cm x of between about 1.2 and about 2.5, at steady state when said formulation is infused over about 1 hour to deliver 25-600 mg of posaconazole, and repeated on a 24-hour basis.
In some embodiments, the formulation is further characterized as
providing a ratio of mean posaconazole blood Cmax to mean posaconazole
plasma Cmax of between about 1.5 and about 2.3, at steady state when said formulation is infused over about 1 hour to deliver 25 mg of
posaconazole, and repeated on a 24-hour basis. In some embodiments, the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole
plasma Cmax of between about 1.5 and about 2.4, at steady state when said formulation is infused over about 1 hour to deliver 50 mg of posaconazole, and repeated on a 24-hour basis.
In some embodiments, the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.7 and about 2.5, at steady state when said formulation is infused over about 1 hour to deliver 100 mg of posaconazole, and repeated on a 24-hour basis.
In some embodiments, the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.2 and about 2.0, at steady state when said formulation is infused over about 1 hour to deliver 200 mg of posaconazole, and repeated on a 24-hour basis.
In some embodiments, the formulation is further characterized as
providing a ratio of mean posaconazole blood Cmax to mean posaconazole
plasma Cmax of between about 1.2 and about 2.2, at steady state when said formulation is infused over about 1 hour to deliver 400 mg of
posaconazole, and repeated on a 24-hour basis. In some embodiments, the formulation is further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole
plasma Cmax of between about 1.3 and about 1.7 , at steady state when said formulation is infused over about 1 hour to deliver 600 mg of posaconazole, and repeated on a 24-hour basis.
In some embodiments, the water in the formulation has been removed by lyophilization.
In some embodiments, the anirnal treated is human, while in other embodiments the animal treated is non-human.
In some embodiments, the formulation is one that is bioequivalent to a formulation disclosed herein.
In some embodiments, the method further comprises adπiimstering a bolus loading dose of said formulation and then admimstering an intravenous maintenance dose of said formulation.
In some embodiments, the method comprises admimstering to said animal an effective amount of posaconazole to provide a mean maximum
plasma concentration (Cmax) of posaconazole of at least about 467 ng/ml
at steady state, and a mean plasma Area Under the Curve over 24 hours
(AUC) value of posaconazole of at least about 9840 ng-hr/ml at steady state, when said formulation is infused over about 1 hour to deliver 100
mg of posaconazole, and repeated at an interval of about 24 hours.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows posaconazole mean blood concentration-time profiles in healthy volunteers after 1 hr intravenous infusions of 25, 50, 100, 200,
400, and 600 mg posaconazole.
FIG. 2 shows posaconazole mean plasma concentration-time profiles in healthy volunteers after 1 hr intravenous infusions of 25, 50, 100, 200,
400, and 600 mg posaconazole.
FIG. 3 shows posaconazole mean plasma and blood concentration-time profiles in healthy volunteers after 1 hr intravenous infusion of 25 mg posaconazole.
FIG. 4 shows posaconazole mean plasma and blood concentration- time profiles in healthy volunteers after 1 hr intravenous infusion of 600 mg posaconazole.
DETAILED DESCRIPTION OF THE INVENTION
The present invention encompasses formulations suitable for parenteral
adrrrinistration, e.g., by injection, for treating an infection. These
formulations comprise a suspension of posaconazole, stabilized by a phospholipid, in a mixture comprising water, a thermoprotectant, and a l
buffer system. Since posaconazole is minimally soluble in water, a suspension formulation is advantageous. Phospholipids have been found to be effective surfactants in forming stable suspensions of posaconazole
in water or an aqueous medium.
These phospholipids can degrade when subjected to the temperature excursions experienced during teπninal sterilization [e.g., autoclavin ), a step which is necessary to assure the sterility of any injectable formulation. Thus, a thermoprotectant is used to prevent agglomeration and crystal growth of the posaconazole particles during autoclaving.
Parenteral buffer systems are typically designed to be at physiological pH of about 7.4. Phospholipids are known to be stable at a pH range of about 6 to about 7. Furthermore, pH adjustment of injectable formulations can be necessary to achieve physiological compatibility, and thus, for example, to minimize injection-site irritation. In addition, the rate of phospholipid hydrolysis can be temperature-sensitive. Thus, in
the present formulations, the buffer systems are designed to meet physiological pH requirements, and to maintain the temperature/pH-
dependent chemical stability of the phospholipid in the formulation
during high temperature excursions (such as experienced during
autoclaving), and throughout shelf life. In accordance with the above, it was found that POPC, an ingredient that
acts as a suspension stabilizer, was sensitive to autoclaving. Certain
buffer systems were found to control degradation of POPC-contarning posaconazole formulations during autoclaving. For example, such formulations were found to be stable after 20 minutes of autoclaving at 121°C. In addition, these buffer systems stabilize such formulations during storage at 4°C to 25°C for at least 18 months following autoclaving. Similarly, other phospholipids that are similar to POPC could be used to stabilize the formulations disclosed herein. For example, unsaturated phospholipids with an acyl chain length ranging from C12 to C20 wherein the degree of unsaturation of the acyl chain ranges from 1 to 4; as weE as saturated phospholipids with an acyl chain length ranging from C12 to Ci8 are useful according to the present invention. Examples of useful unsaturated phospholipids include: 1 -palrmtoyl-2-oleoyl-sn-glycero-3-phosphocholine ("POPC") ,
l,2-Myrtstoleoyl-sn-Glycero-3-Phosphocholine
Figure imgf000024_0001
1 , 2-Palmitoleoyl-sn-Glycero-3-Phosphocholine
Figure imgf000025_0001
1 ,2-Dioleoyl-sn-Glycero-3-Phosphocholine (DOPC)
Figure imgf000025_0002
1 ,2-Dioleoyl-sn-Glycero-3-Phosphoethanolamine (DOPE)
Figure imgf000025_0003
1 , 2-Linoleoyl-sn-Glycero-3-Phosphocholine
Figure imgf000025_0004
and l-01eoyl-2-Myristoyl-sn-Glycero-3-Phosphocholine
Figure imgf000026_0001
or combinations thereof.
Examples of saturated phospholipids include:
1 ,2-Dflauryl-sn-Glycero-3-Phosphocholine (DLPC)
Figure imgf000026_0002
1 ,2-Diπιyristoyl-sn-Glycero-3-Phosphocholine (DMPC)
Figure imgf000026_0003
1 ,2-Dipalrmtoyl-sn-Glycero-3-Phosphocholine (DPPC)
Figure imgf000026_0004
and
1 ,2-Stearoyl-sn-Glycero-3-Phosphocholine (DSPC)
Figure imgf000027_0001
or combinations thereof.
Unsaturated phospholipids are known be to prone to oxidation. To prevent such oxidation, an antioxidant can be employed. In some embodiments, the antioxidant comprises propyl gallate, preferably at a concentration of about 0.02 to about 0.005 mg/ml. In other embodiments, the antioxidant comprises butylated hydroxytoluene, preferably at a concentration of about 0.1 to about 0.02 mg/ml. In related embodiments, the antioxidant comprises propyl gallate, preferably at a concentration of about 0.02 to about 0.005 mg/ml, in combination with butylated hydroxytoluene, preferably at a concentration of about 0.1 to about 0.02 mg/ml. In yet other embodiments, the antioxidant comprises alpha-D-tocopherol, preferably at a concentration of about 0.5 to about 0.01 mg/ml.
The inventors have found certain ratios of components to result in advantageous formulations. For example, the weight ratio of
phospholipid to posaconazole is preferably between about 1:0.1 and about
1: 10, more preferably, between about 1: 1 and about 1:5, still more preferably, between about 1: 1 and about 4:5. The weight ratio of thermoprotectant to posaconazole is preferably between about 0.5: 1 and about 6: 1, more preferably, between about 2: 1 and about 6: 1. The weight
ratio of thermoprotectant to phospholipid is preferably, between about
20: 1 and about 5:4, more preferably, between about 20:4 and about 30:4.
The formulations of the present invention comprise a suspension of solid particles of posaconazole of specific particle size distribution in an aqueous phase. The particle size distribution displayed in the suspended particles is critical for physiological compatibility, syringeability, physical stability of the suspension, re-suspendability, and for pharmacokinetic characteristics and bio- distribution [le., sequestration within specific bodily tissues). Since these characteristics are critical to the formulation as delivered to the patient, it is important that processes that contribute to changes in particle size distribution after rnicronization are controlled.
Such processes can include agglomeration during autoclaving, and de- suspension due to temperature excursions and/or agitation experienced during shipping and storage. It is the particle size distribution in the
formulation as ready for administration to the patient that influences
pharmacokinetic characteristics and bio- distribution.
The inventors of the present invention have deterrnined that for injectable formulations of posaconazole, these characteristics are brought within
advantageous ranges with particle size distributions whose median values are between about 1.0 to about 8.0 microns, preferably, between about 1.0 to about 5.0 microns, more preferably between about 1.2 to about 4.5
microns. In each case, the particle size distributions display not more
than about 3000 particles of 10 microns or greater size and not more than about 300 particles of 25 microns or greater size.
In the injectable formulations of the present invention, which include POPC, it has been found useful to maintain a pH range of between about 3.0 and about 9.0, preferably between about 6.0 and about 8.0, and more preferably between about 6.4 and about 7.6.
The inventors have found that certain organic buffers, e.g., histidine and citric acid, are more advantageous in controlling the pH-related degradation of POPC in the formulation. Components used in pH adjustment systems can also function as components of the buffer system, after pH adjustment has been achieved. Non-limiting examples of pH adjustment system components that function in this way include
sodium hydroxide, hydrochloric acid, and phosphoric acid.
Anti-Infective Applications
The present invention encompasses methods of prevention and treatment
of a variety of infections caused by a broad spectrum of infectious agents. The term "infection" is understood to include, but not be limited to, those
disease states caused by molds, yeasts and other infectious agents, such as: Candida, dermatophytes, Dimorphics, Dematiaceous {e.g., Alternaria
and Bipolaris), Aspergillus, Acremonium, Basidiomycetes, Bjerkandera, Coprinus, Paecilomyces, Microsporum, Trichophyton, Pseudallescheria,
Schizophyllum, Crytococcus, Histoplasma, Blastomyces, Coccidioides, Fusarium, Exophiala, Zygomycocetes {e.g., Mucor, Rhizopus, and Rhizomucor), Kluyveromyces, Saccharomyces, Yarrowia, Pichia, Epidermophyton, Paracoccidioides, Scedosporium, Apophysomyces, Curvularia, Penicilliurn, Fonsecaea, Wangiella, Sporothrix, Pneumocystis, Trichosporon, Absidia, Cladophialophora, Rarnichloridiurn, Syncephalastrum, Madurella, Scytalidium, Leshmania, protozoa, bacteria, gram negatives, gram positives, anaerobes, including Legionella Borrelia, Mycoplasma, Treponema, Gardneralla, Trichomononas and Trypanosoma.
The present invention is intended to treat both opportunistic and non- opportunistic infections, where the term "opportunistic" as used herein denotes those infections caused by organisms capable of causing a disease only in a host whose resistance is lowered, e.g., by chemotherapy or H.I.V.
In particular, posaconazole is useful in the prevention and/or treatment
of the following disease states: Initial (first line) treatment of oropharyngeal or esophageal candidiasis; Salvage therapy of azole-refractory oropharyngeal and esophageal candidiasis (e.g., in patients who have failed oral fluconazole and/ or itraconazole);
Initial treatment of invasive aspergillosis, candidiasis, fusariosis, scedosporiosis, infections due to dimorphic fungi {e.g., ciyptococcosis, coccidioidomycosis, paracoccidioidomycosis, histoplasmosis, blastomycosis), zygomycosis, and invasive infections due to rare moulds and yeasts;
Salvage therapy for invasive mycoses in patients who are refractory to or intolerant of other therapies (e.g., amphotericin B, lipid formulations of amphotericin B, caspofungin, voriconazole and/or itraconazole);
Prevention of invasive Candidiasis, invasive mould infections (including zygomycosis and aspergillosis) in patients at high risk, including patients who have undergone intensive chemotherapy and/or radiation therapy for hematologic malignancies, bone marrow or peripheral stem cell transplant conditioning regimens, and patients receiving combination iirjmunosuppressive therapy for the treatment of acute or chronic graft-versus-host disease or prevention of solid organ
transplantation;
Chagas disease (Trypanosorniasis due to T. cruzi) including acute and
chronic forms; and,
Leishmaniasis, including visceral and localized forms. Administration
Irrπnuno-suppressant therapy {e.g., chemotherapy, radiation therapy,
myeloablative conditioning regimens) often results in one or more of the
above-referenced infections. The present invention encompasses the administration of a posaconazole formulation adjunctive to imrnuno- suppressant therapy, wherein the posaconazole formulation functions prophylactically with regard to opportunistic infections including the
above-referenced disease states.
The present invention encompasses a variety of modes of administration to any part, organ, interstice or cavity ofan animal's body that is subject to an infection. A non-lir ting set of examples of modes by which the posaconasole formulations of the present invention may be adrninistered includes: intravenously, intramuscularly, subcutaneously, ophthalmically, subconjuctivally, intraocularly, via anterior eye chamber injection, intravitreally, intraperitoneally, intrathecally, intracystically, intrapleurally, intranasally, topically, via wound irrigation, intradermally, intrabuccally, intra-abdominally, intra-articularly, intra-aurally,
intrabronchially, intracapsularly, intrarneningeally, intrapulrnonarilly, via
inhalation, via endotracheal or endobronchial installation, via direct installation into pulmonary cavities, intraspinally, rntrasynovially, intrathoracically, via thoracostomy irrigation, vaginally, epidurally, rectally, intracisternally, intravascularly.intraventricularly, intraosseously, via irrigation of infected bone, and via application as part
of any admixture with cement for prosthetic devices.
Co-formulations comprising combinations of posaconazole and at least one other active ingredient are also within the scope of the present invention. Non-lirniting examples of such active ingredients include: antifungals such as echinocandins (including caspofungin, πiicafungin, and anidulafungin) and azoles (including voriconazole, itraconazole, fluconazole, ketoconazole, ravuconazole); amphotericin B; deoxycholate amphotericin B; flucytosine; and terbinafine.
Also within the scope of this invention are combinations with an antibacterial, antiviral, steroid, or nonsteroidal anti-inflammatory drugs ("NSAIDs"), chemotherapeutics, and/or anti-emitics. Similarly, co- administration of posaconazole with at least one of the above active ingredients, aside from within a single formulation, is also within the scope of the present invention.
Also within the scope of the present invention are a variety of dosing
regimens, each consisting of a frequency of dosing and a duration of administration. Preferred frequencies of dosing include once every 12, 24,
36 and 48 hours. Preferred durations of administration are within the
range of 30 minutes to 4 hours, more preferably, 1 to 2 hours. Also included within the scope of preferred administration is bolus dosing, at various rates and various doses, and combinations of a bolus loading dose, or several bolus loading doses, with an intravenous infusion
maintenance dose that provides therapeutic plasma concentration ranges similar to or exceeding those described in Table 14 infra.
As used herein, the following terms shall have the definitions set forth below.
As used herein, the phrase "phospholipid" refers to a lipid compound that yields on hydrolysis phosphoric acid, an alcohol, fatty acid and a nitrogenous base. Examples include natural and synthetic phoshpholipids, which include lecithin, cephalin, sphingornyelin and 1- pahnitoyl-2-oleoyl-sn-glycero-3-phosphocholine ("POPC") .
As used herein, the phrase "natural phospholipid" refers to a phospholipid occurring in nature, or derived from a natural source. Non- lirniting examples of natural phospholipids include egg phospholipids, soy
phospholipids, and animal tissue phospholipids. Combinations of more than one natural phospholipid are within the scope of the present
invention.
As used herein, the phrase "synthetic phospholipid" refers to a man-made
phospholipid. Non-limiting examples of synthetic phospholipids include
l-palnιitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1 ,2-oleoyl-sn- glycero-3-phosphocholine (DOPC), l,2-Dilauryl-sn-Glycero-3- Phosphocholine (DLPC), l,2-Dinιyristoyl-sn-Glycero-3-Phosphocholine
(DMPC), l,2-Dipalmitoyl-sn-Glycero-3-Phosphocholine (DPPC) and 1,2- Stearoyl-sn-Glycero-3-Phosphocholine (DSPC). Combinations of more than one synthetic phospholipid are within the scope of the present invention.
As used herein, the phrase "buffer system" refers to a buffer comprising one or more components that maintains a particular pH range. Non- limiting examples of suitable buffer systems include: phosphoric acid; glycine; sodium citrate; histidine; citric acid; acetic acid; tromethamine; ammonium sulfate; and combinations thereof. The aforementioned components are understood to include the salts, hydrates and solvates thereof. Thus, for example, phosphoric acid includes the sodium phosphate or potassium phosphate salts, among other salts. Preferred buffer systems include sodium phosphate monobasic, sodium phosphate dibasic, or a combination thereof. More preferred buffer systems include sodium phosphate monobasic monohydrate, sodium phosphate dibasic
anhydrous, or a combination thereof. As used herein, the phrase "organic buffer" refers to a buffer comprising at least one organic compound. Non-
limiting examples of suitable organic buffers include: glycine;. sodium citrate; histidine; citric acid; acetic acid; and combinations thereof. As used herein, the term "antioxidant" refers to an agent that hinders
oxidation. Exemplary antioxidants include propyl gallate, butylated hydroxytoluene, and alpha-D-tocopherol.
As used herein, the phrase "median particle size" refers to the particle size present in the volume-weighted 50th percentile, as ascertained by Malvern®, Sympatec®, or Horibe® laser diffraction particle size analysis. Particle sizes are measured throughout, and at the term ation of, the shelf life, typically up to 24 months after manufacture, when held at either refrigerated or room temperatures. Particle sizes are also measured and maintained when the formulation is diluted into large volume parenterals, e.g., 5% dextrose or water for injection.
As used herein, the phrase "initial median particle size" refers to the particle size present within 1 week after a specified timepornt. For example, the initial median particle size after autoclaving refers to the median particle size present within 1 week after autoclaving has been
completed.
As used herein, the term "autoclaving" refers to sterilization by the
terminal steam sterilization method. For example, autoclaving for 20
minutes at 121°C suffices to sterilize the posaconazole formulations
disclosed herein. As used herein, the phrase "thermoprotectant" refers to an agent that
stabilizes the phospholipid during temperature excursions. In the
present invention, a thermoprotectant is used to preserve the
phospholipid, which is necessary to control crystal growth and aggolomeration of the posaconazole particles during autoclaving. Thermoprotectants are typically water soluble polyhydroxyl compounds. For example, trehalose is a thermoprotectant agent that may be used in conjunction with posaconazole. Others include maltose, sorbitol, dextrose, sucrose, lactose and mannitol.
As used herein, the term "prodrug" refers to a compound that is a drug precursor which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield posaconazole or a salt and/or solvate thereof.
As used herein, the term "solvate" refers to a physical association between a compound with one or more solvent molecules. This physical association involves varying degrees of ionic and/or covalent bonding,
including hydrogen bonding. In certain instances, the solvate will be
capable of isolation, for example, when one or more solvent molecules are
incorporated in the crystal lattice of the crystalline solid. The term "solvate" encompasses both solution-phase and isolatable solvates. Non-
limiting examples of suitable solvates include hydrates, ethanolates, and
methanolates. As used herein, the term "injectable" means adapted to parenteral adπiinistration.
As used herein, the term "fungus" means one of the diverse morphologic forms of yeasts and molds. Fungi include Candida, dermatophytes, Dirnorphics, Dematiaceous {e.g., Alternaria and Bipolaris), Aspergillus, Acremonium, Basidiomycetes, Bjerkandera, Coprinus, Paecilomyces, Microsporum, Trichophyton, Pseudallescheria, Schizophyllurn, Crytococcus, Histoplasma, Blastomyces, Coccidioides, Fusarium, Exophiala, Zygomycocetes {e.g., Mucor, Rhizopus, and Rhizomucor), rvluyveromyces, Saccharomyces, Yarrowia, Pichia, Epidermophyton, Paracoccidioides, Scedosporium, Apophysomyces, Curvularia, Periicillium, Fonsecaea, Wangiella, Sporothrix, Pneumocystis, Trichosporon, Absidia, Cladophialophora, Rarriichloridiurn, Syncephalastrum, Madurella, Scytalidium, l^shrnania, gram negatives, gram positives, Mycoplasma, Treponema, Gardneralla, and Trichomononas.
As used herein, the term "Dematiaceous" means dark conidia and/or
hyphae, and includes as non-limiting examples Alternaria and Bipolaris.
As used herein, the term "Zygomycocete" means a class of fungi characterized by sexual reproduction resulting in the formation of
zygospore, and asexual reproduction by means of nonmotile spores called sporangiospores or conidia, and includes as non-liiniting examples Mucor, Rhizopus, and Rhizomucor.
As used herein, the term "anaerobe" means a rnicroorganisrn that can live and grow in the absence of oxygen, and includes as non-limiting examples Legionella Borrelia, Mycoplasma, Treponema, Gardneralla, and Trichomononas .
As used herein, the term "parasite" means an organism that lives on or in another and draws its nourishment therefrom. Parasites include Leshmania and Trypansoma, among others.
As used herein, the term "antifungal" means an agent having activity against one or more fungi, and includes echinocandins such as caspofungin, micafungin, and anidulafungin.
As used herein, the term "azole" means divtayleriimine, and includes voriconazole, itraconazole, fluconazole, ketoconazole, ravuconazole:
As used herein, the term "mean maximum concentration (Cmax)" when followed by the term "at steady state" means that mean maximum
concentration value that occurs after administration of a sufficient
number of repeated doses of the formulation to generate maximum blood or plasma concentrations that are substantially equivalent to one another in value. Thus, the subsequent maximum concentration values are no
longer rising, but rather each peak achieves substantially the same maximum, value as the previous one and the next one.
As used herein, the term "anirnal" is understood to include humans, non- human mammals, fish, birds and reptiles.
As used herein, the term "bioequivalent" is understood as having that meaning assigned to the term by the U.S. Food & Drug Administration. "Bioequivalence means the absence of a significant difference in the rate and extent to which the active ingredient or active moiety in pharmaceutical equivalents or pharmaceutical alternatives becomes available at the site of drug action when adrninistered at the same molar dose under similar conditions in an appropriately designed study." 21 CFR 320.1(e). Methodologies for determiriing bioequivalence are given in "Guidance for Industry: Statistical Approaches to Establishing Bioequivalence," U.S. Department of Health and Human Services, Food and Drug Adrmnistration, Center for Drug Evaluation and Research
(CDER) June, 2001.
EXAMPLES
The following non-lirniting examples illustrate certain aspects of the
invention. Exemplary formulations of posaconazole in conjunction with POPC and trehalose using various buffer systems are detailed below in Tables 1-3.
These formulations provide ranges for buffer systems that maintain a
particular pH range.
Table 1. Re resentative osaconazole formulations at a H range of 6.4-7.4
Figure imgf000041_0001
Table 2. Re resentative osaconazole formulations at a H ran e of 6.4-6.6
Figure imgf000041_0002
Table 3. Re resentative osaconazole formulations at a H ran e of 6.6-6.8
Figure imgf000041_0003
An exemplary posaconazole formulation for each of the buffer systems described in Tables 1-3 is provided in Examples 1-3, respectively.
Figure imgf000042_0001
Of note, the pH is 7.4 in Example 1.
Figure imgf000042_0002
Of note, the pH is 6.4 in Example 2. Exam le 3
Figure imgf000042_0003
Of note, the pH is 6.6 in Example 3.
In addition, exemplary posaconazole formulations that include antioxidant are described in Examples 4-6.
Figure imgf000042_0004
Of note, the pH is 7.4 in Example 4.
Figure imgf000043_0001
The pH is 6.4 in Example 5.
Figure imgf000043_0002
The pH is 6.5 in Example 6.
Example 7 is a preferred embodiment of the present invention. Exam le 7
Figure imgf000043_0003
The pH is 7.2 in Example 7.
The following is an exemplary placebo formulation wherein the pH is 6.4.
This exemplary placebo formulation was utilized in the comparative
stability data study described below.
Figure imgf000043_0004
Figure imgf000044_0001
Comparative stability data study
The stability of POPC in formulation Examples 1-3 was compared with the aforementioned exemplary placebo both before and after autoclaving for 20 min at 121°C. In addition, posaconazole stability, particle size, pH, and a physical observation were ascertained for each formulation before and after autoclaving. Each formulation was also examined following an additional period of storage at 4°C, 25°C, and 40°C (le., 4°C ± 2°C at 60% ± 5% relative humidity; 25°C ± 2°C at 60% ± 5% relative humidity; and 40°C ± 2°C at ambient relative humidity, respectively) for 1 month, 3 months, and 6 months after autoclaving. Notably, particle size was determined using the Malvern laser diffraction particle size analysis technique. Particle sizes are characterized by values for median ("50th percentile") and maximum ("100th percentile"). The stability data from these comparative studies are compiled below for formulations reflected in Examples 1-6, shown in Tables 4-9, respectively.
Figure imgf000044_0002
Figure imgf000045_0001
Figure imgf000045_0002
Figure imgf000046_0001
Figure imgf000046_0002
Figure imgf000047_0001
Figure imgf000047_0002
Activities of posaconazole against a broad spectrum of infectious agents have been tested in vitro. Tables 10 and 11 display a subset of the results of this in vitro testing, showing some of those infectious agents against which posaconazole is most active.
Figure imgf000048_0001
Figure imgf000048_0002
Figure imgf000049_0001
Figure imgf000050_0001
Table 11. Geometric Mean MIC and MICF90] Values (μg/mL) for # Strains Tested (n) in Posaconazole (POS), Amphotericin (AMB) and Voriconazole (VOR)
Figure imgf000051_0001
Figure imgf000052_0001
Pharmacokinetics
The pharmacokinetic characteristics of the posaconazole formulation were
evaluated in a Phase- 1, single-site, randomized, evaluator-blinded (within dose level), placebo-controlled, rising-single-dose study, with up to six
groups of 12 healthy subjects. The purpose of the study was to evaluate the safety, tolerability, and phaπnacokinetics of the posaconazole intravenous drug product formulation (hereinafter referred to as "POS IV") when delivered intravenously. Table 12 shows the POS IV formulation, and Table 13 shows the physical characteristics of this formulation after sterilization, but before dilution in 5% dextrose.
Table 12. POS IV Formulation, 50 mg/mL
Figure imgf000053_0001
AN = As needed for pH adjustment
Table 13. Physical Characteristics of POS TV, 50 mg/mL
Figure imgf000053_0002
Figure imgf000054_0001
Within each dose group, subjects were randomized on Day 1 according to a computer-generated schedule provided by Schering-Plough Research Institute.
Healthy adult males or females 18 to 45 years of age having body mass indices (BMIs) of 19 to 27 were eligible for inclusion in Groups 1 to 4 of the study. Healthy adult males or females 18 to 45 years of age having
BMIs of 19 to 27 and having body weights of >60 kg were eligible for
inclusion in Groups 5 and 6 of the study.
POS IV (50 mg/mL) was diluted in 5% dextrose in water (D5W) in IV bags. Subjects assigned to active drug received in a 100-mL volume one of the following single doses administered intravenously over 1 hour: Group 1 ,
25 mg; Group 2, 50 mg; Group 3, 100 mg; Group 4, 200 mg; Group 5,
400 mg; Group 6, a 125-mL volume a single dose of 600 mg administered intravenously over 1 hour and 15 minutes.
Blood samples (10 mL each) for the deterrnination of posaconazole concentrations were collected immediately prior to dosing (0 hour), and at 0.25, 0.5, 1, 1.25, 1.5, 2, 3, 4, 6, 8, 10, 12, 14, 16, 24, 48, 72, 96, and 120 hours after the start of infusion, as well as on the follow-up visit on
Day 14. The blood samples were collected into two tubes containing
ethylenediaminetetraacetate salt (EDTA) with each tube containing 4 mL to 5 mL of blood, one tube for determination of posaconazole in whole
blood and other in plasma. For determination of posaconazole in plasma, the tube of blood (4 mL to 5 mL) was centrifuged within approximately 15 minutes of collection at approximately 4°C and IS Og for 10 minutes to completely separate red blood cells from plasma. All blood and plasma samples were immediately frozen to at least -20°C and maintained in the frozen state until assayed. The blood and plasma concentrations of posaconazole were determined using validated high performance liquid chromatographic-mass spectrometric (LC-MS/MS) assays. The lower limit of quantitation (LLOQ) of this assay was 5.0 ng/mL and the calbration range was 5 to 5000 ng/mL.
The following pharmacokinetic parameters were determined: maximum plasma concentration (Cmax); time of maximum plasma concentration (Tmax); the area under the plasma concentration versus time curve to infinity (AUC[I]); the area under the plasma concentration versus time
curve to the final measurable sampling time (AUC[tf]); terrninal phase
half-life (ty2); total body clearance (CL); and, volume of distribution at
steady-state (Vdss). Posaconazole blood and plasma concentrations above the LLOQ were used
for the non-compartmental pharmacokinetic analyses. Pharsight®
Knowledgebase Server®: version 2.0.1 (PKS) with WinNonlin version 4.0.1
(Pharsight Corporation, Gary, NC) was used to conduct the pharmacokinetic analysis. The Cmax and Tma were the observed values. The terminal phase rate constant (k) was calculated as the negative of the slope of the log-linear terminal portion of the serum concentration-time curve using linear regression. The terminal phase half-life, tι/2, was calculated as 0.693/k.
The area under the serum concentration-time curve from time 0 to the time of final quantifiable sample [AUC(tf)] was calculated using the linear trapezoidal rule. AUC(tf) was then extrapolated to irifinity (I) as follows:
AUC(I) = AUC(tf) + Ces(tf)/k where Ces(tf) is the estimated concentration determined from linear regression at final measurable sampling time, tf.
Total body clearance, CL, was calculated by the following equation:
CL = Dose/AUC(I)
The apparent volume of distribution at steady-state, Vdss, was calculated
as:
Vdss = CL x MRT where MRT is the mean residence time (adjusted for infusion duration)
determined from moment analysis. The observed single dose plasma concentrations were used for
pharmacokinetic modeling and simulation and to project steady-state concentrations for once-a-day (QD) dosing regimen. A nonparameteric
superposition method was used for the pharmacokinetic modeling and simulation under the assumption of linear pharmacokinetics {see Gibaldi M, Perrier D., Pharmacokinetics, 2nd ed., New York: Marcel Dekker, Inc., 1982:409-17).
After cessation of infusion of POS IV, posaconazole plasma concentrations declined unusually rapidly, and then, surprisingly, increased subsequently, followed by a slow declining terminal phase (see Figures 1-4). This pharmacokinetic profile is believed to be atypical and unique among known azoles. Moreover, this pharmacokinetic pattern was also observed after the intravenous adrrunistration of posaconazole in animals. It is indicative of a rapid distribution of posaconazole to the liver and spleen and subsequent slow release from these tissues. Therefore, as noted in the literature with respect to another pharmaceutically active agent (Townsend RW, Zutshi A, Bekersky I., "Biodistribution of 4-
[14C]cholesterol-Ambisome following a single intravenous administration
to rats", Drug Metabolism and Disposition. 2001 ;29:681 -5), POS IV may be
initially sequestrated in tissues, such as the liver and spleen, via uptake through the reticuloendothelial system ("RES"). Although not intended to
be limited to any single mechanism of action, it is believed that the resulting high concentrations of posaconazole in these tissues due to sequestration of the drug may contribute to enhanced anti-infective activity, since these tissues are often the sites of infection.
In order to determine the target dosing for intravenous administration, it was necessary to determine a target range for mean Cavg and mean Cmax. Previous studies on orally administered posaconazole are instructive in this regard. Table 14 displays pharmacokinetic data resulting from such oral adπiinistration, arranged by quartile based on the observed range of posaconazole plasma concentration values. For each quartile, the response rate for apergillosis is displayed.
Table 14. Pharmacokinetic Results of Orally Administered Posaconazole
Figure imgf000058_0001
The table shows that the target mean Cmax for a response rate of at least 50% should be in the range of 467 to 1480 ng/mL, or higher. The pharmacokinetic modeling and steady-state projection based on the pharmacokinetic results of POS IV once-a-day (QD) dosing regimen show that the projected posaconazole mean Cmax at a 100 mg POS IV QD dose
will be 714 ng/mL (see Table 17, 100 mg dose), which exceeds 467
ng/mL, the minimum clinically relevant mean plasma Cmax. The data in Table 17 suggest that there exists a dose between 50 and 100 mg which will result in the miriimum clinically relevant mean plasma Cmax of about 467. However, in terminal disease states, it is desirable to treat the patient with the maximum tolerated dose. Thus, having established that a dose of 100 mg is projected to achieve the rninimurn clinically relevant mean plasma Cmax, it may be desirable to dose at higher quantities, e.g., 200 mg, 400 mg, or 600 mg, subject to tolerability.
After intravenous adrninistration of POS IV formulation, posaconazole was slowly eliminated from plasma with an average terminal half-life of 21 to 39 hours. The half-life was higher at the higher dose compared to that at lower dose groups (see Tables 15 and 16), in a range of about 15 hours (with a lOOmg dose) to about 51 hours (with a 400mg dose). A long half- life is desirable as it provides the sustained and high plasma
concentration of antifungal agent over the entire dosing interval, likely contributing to better antifungal activity. The systemic clearance appeared
to decrease with increasing doses and ranged from 13 to 6 L/hr (see
Tables 15 and 16). The mean volume of distribution was large (326 to 408 L) exceeding total body water volume of about 40 L. This suggests
extensive tissue distribution and penetration into the tissues, a characteristic that likely contributes to enhanced anti-infective activity. The range in the data for Vdss was from 219 to 516L. This is consistent
with the coefficient of variation of the data, which suggests that the volume distribution could have a range of 200 to 500 L.
The preferable ratios of blood to plasma posaconazole Cmax and AUC values are shown in Tables 18 and 19. Overall posaconazole exposure (AUC) was higher in plasma compared to that in blood {see Tables 18 and 19 - AUC ratio). However, the posaconazole concentrations were greater in blood than in plasma during the infusion and approximately up to 1 hr post-infusion (see Figures 3 and 4; Tables 18 and 19, Cmax ratio). These unique differences between blood and plasma concentrations may contribute to the preferential sequestration of posaconazole in the liver and spleen, as previously noted. The coefficient of variation of the data suggests that the ratio of blood to plasma posaconazole Cmax could have a range of 1.8 to 3.5 for single dose infused over 1 hour to deliver 25-600 mg of posaconazole. The coefficient of variation of the data suggests that
the ratio of blood to plasma posaconazole Cmax could have a range of 1.0 to 2.3 at steady state when posaconazole is infused over about 1 hour,
and repeated on a 24-hour basis, to deliver 25-600 mg of posaconazole. A ratio different than that shown in Table 18 may provide different
distribution properties that could translate into differences in anti-
infective activity.
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Table 18. Ratio of Mean Blood and Plasma Posaconazole PK Parameters in Subjects receiving a Single Dose of POS IV infusion over 1-1.25 hr
Figure imgf000063_0002
Table 19. Ratio of Mean Projected Steady-state Blood and Plasma \ Posaconazole PK Parameters in Subjects receiving a Daily Dose of POS IV infusion over 1-1.25 hr
Figure imgf000064_0001
It is to be understood that all formulations that are bioequivalent to those disclosed herein are also within the scope of the present invention.
The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are intended to fall within the scope of the appended claims.
Various publications are cited herein, the disclosures of which are
incorporated by reference in their entireties.

Claims

What is claimed:
1. A formulation comprising a suspension of posaconazole, stabilized by a phospholipid, in a mixture comprising water, a thermoprotectant, and a buffer system.
2. The formulation of Claim 1 wherein said water has been removed by lyophilization.
3. The formulation of Claim 1 wherein said formulation has been sterilized by autoclaving.
4. The formulation of Claim 1 wherein said formulation has been sterilized by irradiation.
5. The formulation of Claim 1 wherein said buffer system comprises sodium phosphate.
6. The formulation of Claim 1 wherein said buffer system comprises an organic buffer.
7. The formulation of Claim 1 wherein said buffer system comprises at least one of histidine, citric acid, glycine, sodium citrate, arnrnoniurn sulfate, or acetic acid.
8. The formulation of Claim 1 wherein said buffer system maintains a pH of about 3.0 to about 9.0.
9. The formulation of Claim 1 wherein said buffer system maintains a pH of about 6.0 to about 8.0.
10. The formulation of Claim 1 wherein said buffer system maintains a pH of about 6.4 to about 7.6.
11. The formulation of Claim 1 wherein said phospholipid comprises a natural phospholipid.
12. The formulation of Claim 1 wherein said phospholipid comprises a synthetic phospholipid.
13. The formulation of Claim 1 wherein said phospholipid comprises a natural phospholipid and a synthetic phospholipid.
14. The formulation of Claim 1 wherein said phospholipid comprises 1- palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) .
15. The formulation of Claim 1 wherein said thermoprotectant comprises trehalose.
16. The formulation of Claim 1 wherein said phospholipid comprises 1- palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), said thermoprotectant comprises trehalose, and said buffer system comprises sodium phosphate.
17. The formulation of Claim 1 wherein said posaconazole has a particle size distribution whose median value is between about 1.0 and about 8.0 microns, with not more than about 3000 particles of 10 microns or greater size and not more than about 300 particles of 25
microns or greater size.
18. The formulation of Claim 1 wherein said posaconazole has a
particle size distribution whose median value is between about 1.0 and about 5.0 microns, with not more than about 3000 particles of 10
microns or greater size and not more than about 300 particles of 25
microns or greater size.
19. The formulation of Claim 1 wherein said posaconazole has a
particle size distribution whose median value is between about 1.2 and
about 4.5 microns, with not more than about 3000 particles of 10
microns or greater size and not more than about 300 particles of 25 microns or greater size.
20. The formulation of Claim 16 whose ingredients comprise: Ingredient Concentration range Posaconazole about 50 mg/ml POPC about 40 mg/ml Sodium Phosphate, 0.345 mg/ml monobasic, monohydrate, USP Sodium Phosphate, dibasic, 1.065 anhydrous, USP Trehalose 250 mg/ml Water for Injection, USP q.s. 1 ml ad
21. The formulation of Claim 16 whose ingredients comprise: Ingredient Concentration range
Posaconazole about 1 to about 100 mg/ml POPC about 10 to about 60 mg/ml Sodium Phosphate, about O.O1 to about 0.6 monobasic, monohydrate, mg/ml
USP Sodium Phosphate, dibasic, about O.O4 to about 1.5 anhydrous, USP mg/ml Trehalose about 10 to about 300 mg/ml Water for Injection, USP q.s. about 1 ml ad
22. The formulation of Claim 16 whose ingredients comprise: Ingredient Concentration range Posaconazole about 40 to about 60 mg/ml
POPC about 20 to about 50 mg/ml
Trehalose about 100 to about 250 mg/ml Water for Injection, USP q.s. about 1 ml ad
23. The formulation of Claim 7 whose ingredients comprise: Ingredient Concentration Posaconazole 50 mg/ml
POPC 40 mg/ml Histidine 3 mg/ml Citric acid monohydrate 0.24 mg/ml Trehalose 250 mg/ml Water q.s. ad 1 ml at a pH of about 6.4.
24. The formulation of Claim 1 further comprising an antioxidant.
25. The formulation of Claim 24, wherein the antioxidant comprises propyl gallate at a concentration of about 0.02 to about 0.005 mg/ml.
26. The formulation of Claim 24, wherein the antioxidant comprises butylated hydroxytoluene at a concentration of about 0.1 to about O.O2 mg/ml.
27. The formulation of Claim 24, wherein the antioxidant comprises alpha-D-tocopherol at a concentration of about 0.5 to about 0.01 mg/ml.
28. The formulation of Claim 24 whose ingredients comprise: Ingredient Concentration
Posaconazole 50 mg/ml POPC 40 mg/ml Histidine 3 mg/ml
Citric acid monohydrate 0.24 mg/ml Propyl gallate O.Ol mg/ml Butylated hydroxytoluene O.O5 mg/ml
Trehalose 250 mg/ml Water q.s. ad 1 ml at a pH of about 6.4.
29. The formulation of Claim 24 whose ingredients comprise: Ingredient Concentration Posaconazole 50 mg/ml POPC 40 mg/ml Histidine 3 mg/ml Citric acid monohydrate 0.24 mg/ml Alpha-D-tocopherol 0.05 mg/ml Trehalose 250 mg/ml Water q.s. ad 1 ml at a pH of about 6.5.
30. The formulation of Claim 1 wherein the wt. ratio of phospholipid to posaconazole is between about 60: 1 and about 1:10.
31. The formulation of Claim 1 wherein the wt. ratio of phospholipid to posaconazole is between about 1 : 1 and about 1:5.
32. The formulation of Claim 1 wherein the wt. ratio of phospholipid to posaconazole is between about 1: 1 and about 4:5.
33. The formulation of Claim 1 wherein the wt. ratio of
thermoprotectant to posaconazole is between about 300: 1 and about
1: 10.
34. The formulation of Claim 1 wherein the wt. ratio of thermoprotectant to posaconazole is between about 1 : 1 and about 6: 1.
35. The formulation of Claim 1 wherein the wt. ratio of thermoprotectant to phospholipid is between about 30: 1 and about 1 :6.
36. The formulation of Claim 1 wherein the wt. ratio of thermoprotectant to phospholipid is between about 5:4 and about 30:4.
37. A method of treating or preventing an infection in an animal in need thereof which comprises admimstering to said animal an effective amount of the formulation of Claim 20.
38. The method of Claim 37 wherein said infection is caused by a fungus or a parasite.
39. The method of Claim 37 wherein said infection is one or more selected from the group consisting of: oropharyngeal or esophageal candidiasis; refractory oropharyngeal and esophageal candidiasis; invasive aspergillosis, candidiasis, fusariosis, scedosporiosis, infections due to dimorphic fungi , zygomycosis, and invasive infections due to rare molds and yeasts; invasive mycoses in patients who are refractory to, or intolerant of, other therapies; Candidiasis, invasive mold infections in patients who have undergone intensive chemotherapy and/or radiation therapy for hematologic malignancies, bone marrow or peripheral stem cell transplant conditioning regimens, and patients receiving combination imrnunosuppressive therapy for the treatment of acute or chronic graft-versus-host disease or prevention of solid organ transplantation; Chagas disease; and, Leishmaniasis.
40. A method of treating or preventing an infection inan animal in need thereof which comprises administering to said animal an effective amount of the formulation of any of Claims 1, 21 , 22, 23, 28 and 29.
41. The method of Claim 40 wherein said infection is caused by a fungus or a parasite.
42. The method of Claim 40 wherein said infection is one or more selected from the group consisting of: oropharyngeal or esophageal candidiasis; refractory oropharyngeal and esophageal candidiasis; invasive aspergillosis, candidiasis, fusariosis, scedosporiosis, infections due to dimorphic fungi, zygomycosis, and invasive infections due to rare molds and yeasts; invasive mycoses in patients who are refractory to or intolerant of other therapies; Candidiasis, invasive mold infections in patients who have undergone intensive chemotherapy and/or radiation therapy for hematologic malignancies, bone marrow or peripheral stem cell transplant conditioning regimens, and patients receiving combination imrnunosuppressive therapy for the treatment of acute or chronic graft-versus-host disease or prevention of solid organ transplantation; Chagas disease; and, Leishmaniasis .
43. The method of Claim 37 wherein said formulation is adn inistered intravenously.
44. The method of Claim 37 wherein said formulation is adniinistered intramuscularly, subcutaneously, ophthalmically, subconjuctivally, intraocularly, via anterior eye chamber injection, intravitreally, intraperitoneally, intrathecally, intracystically, intrapleurally, intranasally, topically, via wound irrigation, intradermally, intrabuccally, intra-abdor inally, intra-articularly, intra- aurally, intrabronchially, intracapsularly, intrameningeally, intrapulmonarilly, via inhalation, via endotracheal or endobronchial installation, via direct installation into pulmonary cavities, intraspinally, intrasynovially, intrathoracically, via thoracostomy irrigation, vaginally, epidurally, rectally, intracisternally, mtravascularly.mtraventricularly, intraosseously, via irrigation of infected
bone, and via application as part of any admixture with cement for
prosthetic devices.
45. The method of Claim 40 wherein said formulation is adniinistered
intravenously.
46. The method of Claim 40 wherein said formulation is administered
intramuscularly, subcutaneous!/, ophthalmically, subconjuctivally,
intraocularly, via anterior eye chamber injection, intravitreally,
intraperitoneally, intrathecally, intracystically, intrapleurally, intranasally, topically, via wound irrigation, intraderrnally, intrabuccally, intra-abdorninally, intra-articularly, intra-aurally, intrabronchially, intracapsularly, intrarneningeally, intrapulrnonarilly, via inhalation, via endotracheal or endobronchial installation, via direct installation into pulmonary cavities, intraspinally, intrasynovially, intrathoracically, via thoracostomy irrigation, vaginally, epidurally, rectally, intracisternally, intravascularly, intraventricularly, intraosseously, via irrigation of infected bone, or via application as part of any admixture with cement for prosthetic devices.
47. The formulation of Claim 20, further comprising a second active ingredient selected from one or more of the group consisting of: antifungals; amphotericin B; deoxycholate amphotericin B; flucytosine; terbinafine; antibacterials; antivirals; steroids; nonsteroidal anti- inflammatory drugs ("NSAIDs"); chemotherapeutics; and anti-emitics.
48. The formulation of any of Claims 1, 21, 22, 23, 28 and 29 further
comprising a second active ingredient selected from one or more of the
group consisting of: antifungals; amphotericin B; deoxycholate amphotericin B; flucytosine; terbinafine; antibacterials; antivirals;
steroids; nonsteroidal anti-inflammatory drugs ("NSAIDs");
chemotherapeutics; and anti-emitics.
49. The method of Claim 37 further comprising administering a second
active ingredient selected from one or more of the group consisting of:
antifungals; amphotericin B; deoxycholate amphotericin B; flucytosine;
terbinafine; antibacterials; antivirals; steroids; nonsteroidal anti- irmammatory drugs ("NSAIDs"); chemotherapeutics; and anti-emitics.
50. The method of Claim 40 further comprising admimstering a second active ingredient selected from one or more of the group consisting of: antifungals; amphotericin B; deoxycholate amphotericin B; flucytosine; terbinafine; antibacterials; antivirals; steroids; nonsteroidal anti- ir ammatory drugs ("NSAIDs"); chemotherapeutics; and, anti-emitics.
51. The formulation of Claim 1 , further characterized by providing a mean maximum plasma concentration (Cmax) of posaconazole of at least about 467 ng/ml at steady state, and a mean plasma Area Under the Curve over 24 hours (AUC) value of posaconazole of at least about 9840
ng hr/ml at steady state, when said formulation is infused over about 1
hour to deliver 100 mg of posaconazole, and repeated at an interval of about 24 hours.
52. The formulation of Claim 1, further characterized by providing a
mean maximum plasma concentration (Cmax) of posaconazole of at least about 852 ng/ml at steady state, and a mean plasma Area Under the
Curve over 24 hours (AUC) value of posaconazole of at least about 24,600
ng-hr/ml at steady state, when said formulation is infused over about 1
hour to deliver 200 mg of posaconazole, and repeated at an interval of
about 24 hours.
53. The formulation of Claim 1, further characterized by providing a
mean maximum plasma concentration (Cmax) of posaconazole of at least
about 1480 ng/ml at steady state, and a mean plasma Area Under the
Curve over 24 hours (AUC) value of posaconazole of at least about 24,600
ng hr/ml at steady state, when said formulation is infused over about 1
hour to deliver at least 200 mg of posaconazole, and repeated at an interval of about 24 hours.
54. The formulation of Claim 1 , further characterized by providing, after adrninistration of a dosage of about 100 mg of said posaconazole, at least one of: a mean plasma half-life in a range of about 14.9 to about 38.4 hours; and a mean plasma steady state volume of distribution of about 2OO-5OO L.
55. The formulation of Claim 1, further characterized as providing, after administration of a dosage of about 200 mg of said posaconazole, at least one of: a mean plasma half-life of about 18.7 to about 35.5 hours; and a mean plasma steady state volume of distribution of about 2OO-5OO L.
56. The formulation of Claim 1 , further characterized as providing, after
administration of a dosage of about 400 mg of said posaconazole, at least
one of: a mean plasma half-life of about 18.5 to about 51.4 hours; and a
mean plasma steady state volume of distribution of about 2OO-5OO L.
57. The formulation of Claim 1, further characterized as providing, after
administration of a dosage of about 600 mg of said posaconazole, at least
one of: a mean plasma half-life of about 27.2 to about 50.6 hours; and a mean plasma steady state volume of distribution of about 2OO-5OO L.
58. The formulation of Claim 1, further characterized as providing a mean posaconazole blood concentration profile substantially similar to
that of Figure 1 , when said formulation is infused over about 1 hour to deliver 25-600 mg of posaconazole.
59. The formulation of Claim 1, further characterized as providing a mean posaconazole plasma concentration profile substantially similar to that of Figure 2, when said formulation is infused over about 1 hour to deliver 25-600 mg Of posaconazole.
60. The formulation of Claim 1 , further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.5 and about 3.8, when a single dose of said formulation is infused over about 1 hour to deliver 25-600 mg of posaconazole.
61. The formulation of Claim 1, further characterized as providing a ratio of mean posaconazole blood Cmaχ to mean posaconazole plasma Cmax of between about 2.1 and about 3.3, when a single dose of said formulation is infused over about 1 hour to deliver 25 mg of posaconazole.
62. The formulation of Claim 1, further characterized as providing a
ratio of mean posaconazole blood Cma to mean posaconazole plasma
Cmax of between about 1.9 and about 3.8, when a single dose of said formulation is infused over about 1 hour to deliver 50 mg of
posaconazole.
63. The formulation of Claim 1, further characterized as providing a
mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 2.2 and about 3.3, when a single dose of said formulation is infused over about 1 hour to deliver 1OO mg of posaconazole.
64. The formulation of Claim. 1 , further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.5 and about 3.2, when a single dose of said formulation is infused over about 1 hour to deliver 200 mg of posaconazole.
65. The formulation of Claim 1, further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.7 and about 3.3, when a single dose of said formulation is infused over about 1 hour to deliver 400 mg of posaconazole.
66. The formulation of Claim 1 , further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.9 and about 3.1, when a single dose of said formulation is infused over about 1 hour to deliver 600 mg of
posaconazole.
67. The formulation of Claim 1, further characterized as providing a
ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.2 and about 2.5, at steady state when said
formulation is infused over about 1 hour to deliver 25-600 mg of
posaconazole, and repeated on a 24-hour basis.
68. The formulation of Clai 1, further characterized as providing a
ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.5 and about 2.3, at steady state when said
formulation is infused over about 1 hour to deliver 25 mg of posaconazole, and repeated on a 24-hour basis.
69. The formulation of Claim 1, further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.5 and about 2.4, at steady state when said formulation is infused over about 1 hour to deliver 50 mg of posaconazole, and repeated on a 24-hour basis.
70. The formulation of Claim 1 , further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.7 and about 2.5, at steady state when said formulation is infused over about 1 hour to deliver 100 mg of posaconazole, and repeated on a 24-hour basis.
71. The formulation of Claim 1, further characterized as providing a ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.2 and about 2.0, at steady state when said
formulation is infused over about 1 hour to deliver 200 mg of
posaconazole, and repeated on a 24-hour basis.
72. The formulation of Claim 1, further characterized as providing a
ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cmax of between about 1.2 and about 2.2, at steady state when said formulation is infused over about 1 hour to deliver 400 mg of posaconazole, and repeated on a 24-hour basis.
73. The formulation of Claim 1, further characterized as providing a
ratio of mean posaconazole blood Cmax to mean posaconazole plasma Cm of between about 1.3 and about 1.7, at steady state when said formulation is infused over about 1 hour to deliver 600 mg of posaconazole, and repeated on a 24-hour basis.
74. The method of Claim 37, wherein said animal is a human.
75. The method of Claim 37, wherein said animal is a non-human.
76. The method of Claim. 40, wherein said animal is a human.
77. The method of Claim 40, wherein said animal is a non-human.
78. The formulation of Claim 1, further characterized as being bioequivalent to the formulation of any of Claims 1, 20, 21, 22, 23, 28 or 29.
79. The method of claim 37, further comprising admimstering a bolus loading dose of said formulation and then admiriistering an intravenous maintenance dose of said formulation.
80. A method of treating or preventing an infection in an ariimal in need
thereof which comprises administering to said animal an effective
amount of posaconazole to provide a mean maximum plasma
concentration (Cmax) of posaconazole of at least about 467 ng/ml at steady state, and a mean plasma Area Under the Curve over 24 hours (AUC)
value of posaconazole of at least about 9840 ng hr/ml at steady state, when said formulation is infused over about 1 hour to deliver 1OO mg of posaconazole, and repeated at an interval of about 24 hours.
PCT/US2005/018945 2004-05-28 2005-05-27 Injectable pharmaceutical suspension comprising posaconazole WO2005117831A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
BRPI0510417-3A BRPI0510417A (en) 2005-05-27 2005-05-27 posaconasol particulate-stabilized injectable pharmaceutical compositions
CA002567803A CA2567803A1 (en) 2004-05-28 2005-05-27 Injectable pharmaceutical suspension comprising posaconazole
AU2005249502A AU2005249502A1 (en) 2004-05-28 2005-05-27 Particulate-stabilized injectable pharmaceutical compositions of posaconazole
MXPA06013886A MXPA06013886A (en) 2004-05-28 2005-05-27 Injectable pharmaceutical suspension comprising posaconazole.
JP2007515456A JP2008501034A (en) 2004-05-28 2005-05-27 Injectable pharmaceutical suspension containing posaconazole
EP05755118A EP1761247A1 (en) 2004-05-28 2005-05-27 Injectable pharmaceutical suspension comprising posaconazole
IL179627A IL179627A0 (en) 2004-05-28 2006-11-27 Particulate-stabilized injectable pharmaceutical compositions of posaconasole
NO20066005A NO20066005L (en) 2004-05-28 2006-12-22 Injectable pharmaceutical suspension comprising posaconazole

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US57512604P 2004-05-28 2004-05-28
US60/575,126 2004-05-28

Publications (1)

Publication Number Publication Date
WO2005117831A1 true WO2005117831A1 (en) 2005-12-15

Family

ID=34971355

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/018945 WO2005117831A1 (en) 2004-05-28 2005-05-27 Injectable pharmaceutical suspension comprising posaconazole

Country Status (15)

Country Link
EP (1) EP1761247A1 (en)
JP (1) JP2008501034A (en)
KR (1) KR20070027564A (en)
CN (1) CN1988887A (en)
AR (1) AR049063A1 (en)
AU (1) AU2005249502A1 (en)
CA (1) CA2567803A1 (en)
IL (1) IL179627A0 (en)
MX (1) MXPA06013886A (en)
NO (1) NO20066005L (en)
PE (1) PE20060291A1 (en)
RU (1) RU2006146555A (en)
TW (1) TWI290051B (en)
WO (1) WO2005117831A1 (en)
ZA (1) ZA200609890B (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006130177A1 (en) * 2005-05-27 2006-12-07 Schering Corporation Particulate-stabilized injectable pharmaceutical compositions of posaconazole
WO2007131253A2 (en) * 2006-05-12 2007-11-22 Pharmacon-Forschung Und Beratung Gmbh Use of combination preparations comprising antifungal agents
EP2130540A1 (en) 2008-06-02 2009-12-09 Sandoz AG Pharmaceutical compositions containing a crystalline form of posaconazole
EP2141159A1 (en) 2008-07-03 2010-01-06 Sandoz AG A Crystalline form of posaconazole
WO2011003992A1 (en) 2009-07-09 2011-01-13 Sandoz Ag A crystalline form of posaconazole
WO2011144653A1 (en) 2010-05-19 2011-11-24 Sandoz Ag Process for the preparation of chiral triazolones
WO2012005973A1 (en) 2010-06-29 2012-01-12 Schering Corporation Posaconazole intravenous solution formulations stabilized by substituted beta-cyclodextrin
US9073904B2 (en) 2010-05-19 2015-07-07 Sandoz Ag Preparation of posaconazole intermediates
US9199919B2 (en) 2010-05-19 2015-12-01 Sandoz Ag Process for the preparation of chiral hydrazides
US9206146B2 (en) 2010-05-19 2015-12-08 Sandoz Ag Purification of posaconazole and of posaconazole intermediates
US9493428B2 (en) 2011-06-16 2016-11-15 Sandoz Ag Process for the preparation of a chiral compound
CN113933131A (en) * 2021-09-24 2022-01-14 合肥天一生物技术研究所有限责任公司 Vaginal microbial fluorescent staining solution

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103284959B (en) * 2012-02-22 2015-06-03 重庆圣华曦药业股份有限公司 Posaconazole freeze-dried powder injection and preparation method thereof
CN102755628B (en) * 2012-07-16 2013-10-23 中国科学院微生物研究所 Antifungal pharmaceutical composition
CN102805731B (en) * 2012-08-22 2014-04-30 北京莱瑞森医药科技有限公司 Posaconazole dry suspension and preparation method thereof
CN105030668B (en) * 2015-06-26 2018-03-27 济川药业集团有限公司 Oral posaconazole supensoid agent and preparation method thereof
CN106333925B (en) * 2015-07-10 2019-06-18 上海美悦生物科技发展有限公司 A kind of posaconazole liquid suspension and preparation method thereof
CN105997872B (en) * 2016-07-08 2019-02-19 河南省立眼科医院 A kind of antimycotic solution of ophthalmically acceptable nano-micelle containing posaconazole
CN110582264B (en) * 2017-03-26 2022-11-11 V·R·斯塔福德 Method for treating eyelid skin condition
WO2018191541A1 (en) * 2017-04-12 2018-10-18 Bhagwandin Vikash J Compositions, packaged pharmaceuticals, and methods of using posaconazole for the sensitization of resistant tumors
CN110507609B (en) * 2018-05-21 2021-09-17 上海医药工业研究院 Preparation method of posaconazole oral suspension
CN111514146A (en) * 2020-04-28 2020-08-11 山西振东泰盛制药有限公司 Pharmaceutical composition containing posaconazole

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5091188A (en) * 1990-04-26 1992-02-25 Haynes Duncan H Phospholipid-coated microcrystals: injectable formulations of water-insoluble drugs
WO2002080678A1 (en) * 2001-04-03 2002-10-17 Schering Corporation Antifungal composition with enhanced bioavailability

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5091188A (en) * 1990-04-26 1992-02-25 Haynes Duncan H Phospholipid-coated microcrystals: injectable formulations of water-insoluble drugs
WO2002080678A1 (en) * 2001-04-03 2002-10-17 Schering Corporation Antifungal composition with enhanced bioavailability
US20030055067A1 (en) * 2001-04-03 2003-03-20 Schering Corporation Antifungal composition with enhanced bioavailability

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
EZZET F ET AL: "THE PHARMACOKINETICS OF POSACONAZOLE IN NEUTROPENIC ONCOLOGY PATIENTS", INTERSCIENCE CONFERENCE ON ANTIMICROBIAL AGENTS & CHEMOTHERAPY, vol. 41, 22 September 2001 (2001-09-22), pages 4, XP001188159 *
MELLINGHOFF I ET AL: "USE OF POSACONAZOLE (SCH56592) FOR TREATMENT OF INVASIVE FUNGAL INFECTIONS REFRACTORY TO STANDARD ANTIFUNGAL THERAPY", BLOOD, W.B.SAUNDERS COMPANY, ORLANDO, FL, US, vol. 96, no. 11, PART 2, 16 November 2000 (2000-11-16), pages 341B, XP008028017, ISSN: 0006-4971 *
NOMEIR AMIN A ET AL: "Pharmacokinetics of SCH 56592, a new azole broad-spectrum antifungal agent, in mice, rats, rabbits, dogs, and cynomolgus monkeys", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, AMERICAN SOCIETY FOR MICROBIOLOGY, WASHINGTON, DC, US, vol. 44, no. 3, March 2000 (2000-03-01), pages 727 - 731, XP002204276, ISSN: 0066-4804 *

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006130177A1 (en) * 2005-05-27 2006-12-07 Schering Corporation Particulate-stabilized injectable pharmaceutical compositions of posaconazole
EP3192507A1 (en) 2006-05-12 2017-07-19 Christian Noe Use of combination preparations comprising antimycotic agents
US8815925B2 (en) 2006-05-12 2014-08-26 Christian Noe Use of combination preparations comprising antifungal agents
NO342103B1 (en) * 2006-05-12 2018-03-26 Christian Noe Combination drug containing an antimycotic agent and a non-steroidal anti-inflammatory agent for use in the topical treatment of Candida mycoses
WO2007131253A2 (en) * 2006-05-12 2007-11-22 Pharmacon-Forschung Und Beratung Gmbh Use of combination preparations comprising antifungal agents
WO2007131253A3 (en) * 2006-05-12 2008-11-27 Pharmacon Forschung & Beratung Gmbh Use of combination preparations comprising antifungal agents
US8552042B2 (en) 2006-05-12 2013-10-08 Christian Noe Use of combination preparations comprising antifungal agents
US8563555B2 (en) 2008-06-02 2013-10-22 Sandoz Ag Pharmaceutical compositions containing a crystalline form of posaconazole
WO2009147075A3 (en) * 2008-06-02 2010-01-28 Sandoz Ag Pharmaceutical compositions containing a crystalline form of posaconazole
CN102046172B (en) * 2008-06-02 2013-06-19 桑多斯股份公司 Pharmaceutical compositions containing a crystalline form of posaconazole
EP2130540A1 (en) 2008-06-02 2009-12-09 Sandoz AG Pharmaceutical compositions containing a crystalline form of posaconazole
EP2141159A1 (en) 2008-07-03 2010-01-06 Sandoz AG A Crystalline form of posaconazole
US8435998B2 (en) 2008-07-03 2013-05-07 Sandoz Ag Crystalline form of posaconazole
WO2011003992A1 (en) 2009-07-09 2011-01-13 Sandoz Ag A crystalline form of posaconazole
WO2011144653A1 (en) 2010-05-19 2011-11-24 Sandoz Ag Process for the preparation of chiral triazolones
US9073904B2 (en) 2010-05-19 2015-07-07 Sandoz Ag Preparation of posaconazole intermediates
US9199919B2 (en) 2010-05-19 2015-12-01 Sandoz Ag Process for the preparation of chiral hydrazides
US9206146B2 (en) 2010-05-19 2015-12-08 Sandoz Ag Purification of posaconazole and of posaconazole intermediates
US9040539B2 (en) 2010-05-19 2015-05-26 Sandoz Ag Process for the preparation of chiral triazolones
KR101834024B1 (en) * 2010-06-29 2018-03-02 머크 샤프 앤드 돔 코포레이션 Posaconazole intravenous solution formulations stabilized by substituted beta-cyclodextrin
US9358297B2 (en) 2010-06-29 2016-06-07 Merck Sharp & Dohme Corp. Posaconazole intravenous solution formulations stabilized by substituted β-cyclodextrin
EP2588116A1 (en) 2010-06-29 2013-05-08 Merck Sharp & Dohme Corp. Posaconazole intravenous solution formulations stabilized by substituted beta-cyclodextrin
WO2012005973A1 (en) 2010-06-29 2012-01-12 Schering Corporation Posaconazole intravenous solution formulations stabilized by substituted beta-cyclodextrin
EP3391890B1 (en) 2010-06-29 2021-08-25 Merck Sharp & Dohme Corp. Posaconazole intravenous solution formulations stabilized by substituted beta-cyclodextrin
EP3391890A1 (en) * 2010-06-29 2018-10-24 Merck Sharp & Dohme Corp. Posaconazole intravenous solution formulations stabilized by substituted beta-cyclodextrin
US9493428B2 (en) 2011-06-16 2016-11-15 Sandoz Ag Process for the preparation of a chiral compound
CN113933131A (en) * 2021-09-24 2022-01-14 合肥天一生物技术研究所有限责任公司 Vaginal microbial fluorescent staining solution
CN113933131B (en) * 2021-09-24 2024-01-26 合肥天一生物技术研究所有限责任公司 Vaginal microorganism fluorescent staining solution

Also Published As

Publication number Publication date
TW200609001A (en) 2006-03-16
AU2005249502A1 (en) 2005-12-15
JP2008501034A (en) 2008-01-17
IL179627A0 (en) 2007-05-15
CA2567803A1 (en) 2005-12-15
CN1988887A (en) 2007-06-27
ZA200609890B (en) 2008-09-25
TWI290051B (en) 2007-11-21
EP1761247A1 (en) 2007-03-14
RU2006146555A (en) 2008-07-20
PE20060291A1 (en) 2006-04-14
AR049063A1 (en) 2006-06-21
MXPA06013886A (en) 2007-01-26
NO20066005L (en) 2007-02-27
KR20070027564A (en) 2007-03-09

Similar Documents

Publication Publication Date Title
EP1761247A1 (en) Injectable pharmaceutical suspension comprising posaconazole
US20060009469A1 (en) Particulate-stabilized injectable pharmacutical compositions of posaconazole
US20060160823A1 (en) Particulate-stabilized injectable pharmaceutical compositions of Posaconazole
US11793755B2 (en) Pharmaceutical composition for intraarticular delivery
KR102284689B1 (en) Lyophilized liposomes
JP4972750B2 (en) Parenteral antifungal products
JP4959085B2 (en) Micelle
RU2618456C2 (en) Pharmaceutical azoles preparation for parenteral injection and methods of production and application for treatment of diseases, sensitive to azole compounds
US20190105268A1 (en) Viscoelastic Gel of Liraglutide Adapted for Once-Weekly or Once Bi-Weekly Administration
EP3593788A1 (en) Ophthalmic compositions containing a nitric oxide releasing prostamide
TW202024058A (en) Compounds and methods for treating fungal infections
ES2927051T3 (en) Injectable compositions of liposome-encapsulated triterpenoid antifungals
US20230049974A1 (en) Fingolimod extended release injectable suspension
CN107811969B (en) High-stability non-vesicular nanoparticles and application thereof in treating fungal infection
CA3220425A1 (en) Pharmaceutical composition, and aprepitant injection and freeze-dried powder injection
OA20177A (en) Compounds and methods for treating fungal infections.
EA045995B1 (en) COMPOUNDS AND METHODS FOR TREATING FUNGAL INFECTIONS

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2567803

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 179627

Country of ref document: IL

Ref document number: 2007515456

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: PA/a/2006/013886

Country of ref document: MX

Ref document number: 06120230

Country of ref document: CO

Ref document number: 4365/CHENP/2006

Country of ref document: IN

Ref document number: 2005249502

Country of ref document: AU

Ref document number: 1020067024996

Country of ref document: KR

Ref document number: 12006502382

Country of ref document: PH

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWE Wipo information: entry into national phase

Ref document number: 551777

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2005755118

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2006146555

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2005249502

Country of ref document: AU

Date of ref document: 20050527

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005249502

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200580024854.2

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 1020067024996

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2005755118

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0510417

Country of ref document: BR